Next Article in Journal
microRNAs Mediated Regulation of the Ribosomal Proteins and its Consequences on the Global Translation of Proteins
Next Article in Special Issue
Sialic Acids and Their Influence on Human NK Cell Function
Previous Article in Journal
Tumor-Associated Antigen xCT and Mutant-p53 as Molecular Targets for New Combinatorial Antitumor Strategies
Previous Article in Special Issue
Characterization of the Polysialylation Status in Ovaries of the Salmonid Fish Coregonus maraena and the Percid Fish Sander lucioperca
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Glycosylation of Cancer Extracellular Vesicles: Capture Strategies, Functional Roles and Potential Clinical Applications

by
Álvaro M. Martins
1,2,3,†,
Cátia C. Ramos
1,2,4,†,
Daniela Freitas
1,2,* and
Celso A. Reis
1,2,3,5,*
1
Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
2
Institute of Molecular Pathology and Immunology (IPATIMUP), University of Porto, 4200-135 Porto, Portugal
3
Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
4
Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
5
Faculty of Medicine of the University of Porto (FMUP), 4200-319 Porto, Portugal
*
Authors to whom correspondence should be addressed.
These authors contributed equally to the work.
Cells 2021, 10(1), 109; https://doi.org/10.3390/cells10010109
Submission received: 9 December 2020 / Revised: 29 December 2020 / Accepted: 4 January 2021 / Published: 8 January 2021
(This article belongs to the Special Issue Sugars on Cell Surfaces and Their Biological Purposes)

Abstract

:
Glycans are major constituents of extracellular vesicles (EVs). Alterations in the glycosylation pathway are a common feature of cancer cells, which gives rise to de novo or increased synthesis of particular glycans. Therefore, glycans and glycoproteins have been widely used in the clinic as both stratification and prognosis cancer biomarkers. Interestingly, several of the known tumor-associated glycans have already been identified in cancer EVs, highlighting EV glycosylation as a potential source of circulating cancer biomarkers. These particles are crucial vehicles of cell–cell communication, being able to transfer molecular information and to modulate the recipient cell behavior. The presence of particular glycoconjugates has been described to be important for EV protein sorting, uptake and organ-tropism. Furthermore, specific EV glycans or glycoproteins have been described to be able to distinguish tumor EVs from benign EVs. In this review, the application of EV glycosylation in the development of novel EV detection and capture methodologies is discussed. In addition, we highlight the potential of EV glycosylation in the clinical setting for both cancer biomarker discovery and EV therapeutic delivery strategies.

1. Introduction

Extracellular vesicles (EVs) are small nano-sized particles, secreted by all cell types and capable of encapsulating and transporting several molecules to a target delivery site [1]. EVs can be found in various biological fluids and can be harvested in relatively non-invasive ways. Therefore, these particles are attractive systems for targeted drug delivery approaches and valuable sources of circulating cancer biomarkers.
Alterations of the glycosylation pathway are a common feature of malignant cell transformation [2,3,4]. These carbohydrates are capable of modulating several processes during cancer progression, including activation of oncogenic signaling pathways, interference with cell–cell and cell–extracellular matrix (ECM) adhesion and mediate cancer cell metastasis [3,4]. In addition, alteration in the glycosylation pattern of a cell has also been associated with content sorting processes [5,6,7], and with the capacity of cells to interact and uptake certain EVs [8,9,10,11]. Interestingly, some of the tumor-associated glycan alterations have already been identified as enriched in cancer EVs (Figure 1), which may constitute important biomarkers with the potential to be used in the clinical setting.
Despite technological advances, the structural characterization of glycans remains quite challenging. The diversity and complexity of these carbohydrates, together with methodological limitations, makes it challenging to deeply analyze the EV glycome [12,13]. The presence of specific glycosylation profiles in tumor EVs highlights its potential to be used not only to develop novel cancer EV detection and isolation methods but also as a source of novel circulating biomarkers. In this review, after a brief description of the main types of changes in glycosylation found in cancer and their impact on different pathological processes, we pointed out the challenges faced by the currently available methods used in the analysis of the EVs glycome. In addition, we summarized the glycosylation patterns already identified in tumor-EVs and discussed their known function in cancer and how they have been used to develop additional EV detection and isolation technologies. Finally, we highlight the potential clinical application of the EV glycosylation in anti-cancer therapy and in the discovery of new biomarkers for the prognosis and early diagnosis of different types of cancer.

2. The Impact of Glycosylation in Cancer Progression

Glycans are carbohydrate structures that modify both proteins and lipids through a biosynthetic pathway finely regulated by glycosyltransferases and sugar transporters. Glycans can be mainly found at the surface of the cellular membrane forming the commonly known glycocalyx, and they are essential mediators of cell–cell communication and cell–matrix interaction [3,14,15]. The major types of glycosylation that can be affected in cancer include (I) N-glycans, characterized by an N-linkage to an Asn residue in an Asn-X-Ser/Thr sequon, where X can be any amino acid except proline. N-glycans have a defined core structure, and can be classified depending on their structures and branches in high-mannose, complex or hybrid [16]; (II) O-GalNAc glycans, also known as mucin-type O-glycans, are carbohydrate chains initiated by a GalNAc sugar covalently linked by an oxygen atom to a Ser/Thr residue. They often appear as long ramified structures, and have multiple core structures, the major includes core-1 to core-4 [17]; and (III) glycosaminoglycans (GAGs), long and non-ramified carbohydrate chains consisting of repeating disaccharide units [18]. The glycosylation pathway is highly regulated by numerous players, including the expression, localization and activity of both glycosyltransferases and glycosidases, and the availability of nucleotide sugar donors [19]. Therefore, variations in the expression levels of specific glycosyltransferases [20] or its mislocalization in the endoplasmic reticulum (ER) and Golgi apparatus (GA) [21], dysregulation of chaperone activity [22] or alterations in nucleotide sugar transporter availability and cofactors [19] will result in the synthesis of aberrant glycosylation in cancer.
These macromolecules play pivotal roles in several physio- and pathological processes either by functioning as structural scaffolds, recognition cues or modulators of other important biomolecules [14]. Genetic and epigenetic alterations that disturb the glycosylation machinery often arise during malignant transformation, which results in loss or increased expression of certain glycans and the appearance of novel glycans [2,3]. The presence of aberrant glycosylation in cancer cells can impact several biological processes including tumor cell proliferation [23], angiogenesis [24], invasion [25] and metastasis [26]. Alterations in the glycosylation pathway provide multiple adaptive advantages, including receptor tyrosine kinase (RTK) activation [25,27,28,29], regulation of adhesion-related proteins [30,31,32] and immune response modulation [15,33], which significantly contribute to cancer progression. All these aspects will be briefly discussed, as they have already been extensively reviewed [3,4,34].

2.1. Receptor Tyrosine Kinase Activation

RTKs are a family of receptors that control several cellular signaling pathways involved in numerous biological processes, such as cell proliferation, growth, motility and differentiation [35]. Dysregulation of RTK activity is highly associated with cancer, which will impact the progression of the disease [36,37]. RTKs carry several glycosylation sites, and their hyperactivated status in malignant conditions is often associated with aberrant glycosylation. Namely, the increase in tumor-associated N-glycan branched structures promotes activation of different RTKs associated with cancer progression, such as the epidermal growth factor receptor (EGFR) [38,39,40], human epidermal growth factor receptor 2 (HER2) [41,42,43] and vascular endothelial growth factor receptor (VEGFR) [44,45]. The presence of aberrant glycans in cancer cells can directly regulate RTK–ligand binding and downstream signaling, and also stimulate receptor oligomerization through galectin-receptor binding, which increases the permanence of the receptor at the cell membrane [46]. Additionally, heparan sulfate (HS) chains can form cell membrane docking sites for ligands, including the hepatocyte growth factor (HGF) [47] and the vascular endothelial growth factor (VEGF) [47,48], resulting in enhanced binding and activation of MET and VEGF receptors, respectively.

2.2. Cell Adhesion Molecules

Integrins and cadherins are important cell receptor proteins responsible for the interactions between cells and the ECM [49]. However, their function can also be affected in cancer due to the presence of aberrant glycosylation, which consequently disturbs cell–cell and cell–ECM interactions [31]. These perturbations can be mechanically induced, when the glycocalyx of cancer cells promotes integrin clustering, thus increasing integrin signaling and adhesion [50]. Additionally, aberrant glycosylation can modulate the kinetic activity of this type of proteins, for example, the increase of α2,6 sialylation in breast cancer cell lines reduces the affinity of α5β1 and α2β1 integrins towards collagen IV and fibronectin [51]. Furthermore, overexpression of sialyl-Tn (STn) hinders the downstream signaling of α5β1 integrin causing significant morphological alterations in the cell [52]. Regarding the cadherins, N-glycan branching structures have been described to destabilize E-cadherin at the cell surface, and thus promote cell proliferation and tumor progression [53].
Selectins are another important type of cell adhesion molecule. These include E-selectin, P-selectin and L-selectin which are expressed in the vascular endothelium, platelets and leukocytes, respectively [54]. These lectins bind to terminal sialofucosylated structures, for example, sialyl-Lewisx (SLex) and sialyl-Lewisa (SLea), present in either N-glycans, O-glycans or glycolipids. The interaction between selectins and these terminal structures is crucial to the leukocyte “rolling” and extravasation process during inflammation [55]. By expressing SLex and SLea, cancer cells are able to hijack this mechanism to interact with the selectins present in the vascular endothelium, traveling through the circulatory system and thus, potentiate their metastatic behavior [56,57].

2.3. Immune Response Regulation

Furthermore, glycans are crucial regulators of the immune response. Aberrant glycosylation can modulate the interaction between tumor cells and selectins, galectins or sialic acid-binding immunoglobulin-type lectins (Siglecs), which are the lectins involved in the immune response (reviewed in [15]). For instance, siglec-7 and siglec-9 are two inhibitor receptors expressed in natural killer (NK) cells that recognize and bind to sialic acids [58]. As a consequence, the increased sialylation in cancer cells induces inhibitory responses, hampering the ability of NK cells to effectively target and kill cancer cells [58,59,60]. Galectins are another important type of lectin in the immune regulation process, whose ligands are β-galactoside sugar residues. Interestingly, it was described that Galectin-9 can interact with Dectin-1, a C-type lectin, present in macrophages and program them into a tolerogenic state in pancreatic ductal carcinoma [61]. Additionally, galectin-3 has been reported to suppress CD8+ T cell responses towards tumor cells and causing T-cell anergy. This effect could be reverted through the depletion of galectin [62,63]. An additional example of relevant lectins of the immune system is the dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), which is present in macrophages. The interaction between this lectin and fucosylated glycans promotes tumor evasion from the immune response in breast cancer [64].

3. Shortcomings of Glycomic Approaches and Their Application in Extracellular Vesicle Glycome Analysis

In the past years, the field of glycomics has substantially evolved. Nevertheless, this evolution has been slower when compared to other-omics. The fact that glycosylation is not a template-based process, together with its complex structural diversity has slowed the development of tools and methods to accurately characterize glycans structurally [65,66]. Undoubtedly, the development of specific antibodies and plant-derived lectins for carbohydrate antigen detection has significantly boosted our ability to identify the presence of specific glycosylated structures. Lectins are proteins with binding specificity towards determined glycan structures, which can be immobilized in a microarray format and then be used to assess the overall glycome of a given sample [67]. Due to their high sensitivity, these lectins can potentially be used to probe the glycome of EVs from any source, such as cells or biofluids. Nevertheless, some lectins have overlapping binding affinities towards certain glycan structures. Therefore, these tools alone do not serve to provide a clean and comprehensive glycosylation profiling of a given sample. For a more extensive glycosylation analysis, advanced mass spectrometry techniques are required as they provide an in-depth glycosylation characterization of a given sample [68]. Indeed, the study of EV glycosylation has been rapidly growing in recent years. However, the yield and purity of current EV isolation approaches constitute limitations for the application of high-resolution EV glycomic methodologies [69]. Therefore, lectin-based approaches have generally been used to identify major glycan structures present in EVs from different sources [8,70,71,72,73], as we carefully detail in the following chapters. The majority of the studies approaching the EV glycome using mass spectrometry have been mainly focused on N-glycosylation. This is partially a consequence of the increased difficulty of decoding O-glycosylation [74]. To apply mass spectrometry methods for glycomic analysis, glycans must first be released from the sample, often through chemoenzymatic reactions. Although there is a specific enzymatic reaction to isolate N-glycans using PNGase F, there are no reliable enzymatic digestion protocols to isolate O-glycan chains [75,76]. It is possible to remove O-glycans using laborious chemical protocols, such as β-elimination. However, this chemical reaction may induce artifacts due to sample “peeling”, which is a phenomenon characterized by the isomerization and degradation of the innermost monosaccharides from the O-glycan chain [77]. Different conditions have been tested and successfully applied to reduce the “peeling” phenomenon, albeit not eliminating it completely [78]. In addition, O-glycosylation sites do not have a defined amino acid sequence, in contrast to N-glycosylation. Any Ser/Thr residue in a protein may or may not be modified with an O-glycan chain, making it more difficult to accurately predict O-glycosylation sites [17,79]. As a side remark, there are online resources, the NetOGlyc and the NetNGlyc, that predict putative O- and N-glycosylation sites, respectively, based on the provided protein/peptide sequence.
To overcome the challenge of O-glycosylation site disclosure, a genetic strategy targeting the COSMC gene, which codifies a key molecular chaperone that together with the C1GalT1 glycosyltransferase is responsible for the O-glycan elongation, was developed [80]. The COSMC gene knockout will lead to the shift of the O-glycosylation pathway towards the expression of simple forms of O-glycans, namely Tn and sialyl-Tn. The glycoengineered cell line was named “SimpleCell” (SC). By the targeted purification of the O-glycopeptides and further mass spectrometry characterization, this approach allowed the identification of the protein carriers of these structures and the disclosure of the respective O-glycosylation sites [80,81]. For instance, gastric SC models allowed the identification of novel important O-glycoproteins and their O-glycosites with biomarker potential for gastric cancer [82]. More importantly, this strategy has proved its usefulness in identifying novel biomarkers for cancer detection [83].
Different high-resolution quantitative mass spectrometry methods previously established to elucidate the N-glycome and O-glycome of cell lines have been adapted to the study of EV glycosylation. For example, the N-glycome of EVs isolated from glioma [71], melanoma [84] and ovarian carcinoma cell lines [72] has already been characterized. Additionally, the N-glycome of urinary EVs from prostate cancer patients has also been assessed using other high-resolution glycomic methods [85]. Regarding EV O-glycosylation, our group has demonstrated the presence of the tumor-associated antigen, STn by specific antibody recognition in gastric cancer EVs [86]. Other groups have also identified the presence of Tn and STn in cervical cancer cell line models using an innovative lectin-based approach [87]. Interestingly, also identified was the presence of proteins modified with O-GlcNAcylation, a type of intracellular O-glycosylation, in metastatic colorectal cancer EVs [88]. Outside of the cancer context, both the N- and O-glycosylation profile of EVs from healthy individual urinary samples was also disclosed using high-resolution glycomic techniques, such as matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) and liquid chromatography/tandem mass spectrometry (LC/MS-MS) [89].
Given the fundamental impact of aberrant cellular glycosylation in cancer progression and its association with patients’ poor-survival [3,4,25,90], we believe that a full characterization of cancer EV glycans by high-resolution approaches will disclose important cancer circulating biomarkers and set the ground to further explore its role in cancer biology.

4. Cancer Extracellular Vesicles Glycosylation

EVs are small nano-sized particles that are released into the extracellular space by all types of cells. These vesicles exert a broad array of biological functions, being important mediators of intercellular communication [1]. EV diameter typically ranges from 35–5000 nm, and therefore are quite smaller than cells, but much larger than proteins [91,92]. The general term EVs comprises three main types of vesicles which are classified, according to their size and biogenesis mechanism, into exosomes, microvesicles (ectosomes or microparticles) and apoptotic bodies. Exosomes have an endocytic origin and are produced by the inward budding of the plasma membrane of the cell. This invagination of the cell membrane leads to the formation of multivesicular bodies (MVBs) that can later either fuse with lysosomes for content degradation or fuse with the cellular membrane to be secreted in the form of exosomes. The formation and release of exosomes can be regulated by the endosomal sorting complex required for transport (ESCRT) [93,94]. In this type of vesicles, it is expected to find ESCRT proteins and accessory proteins for this complex, such as the ALG-2-interacting protein X (Alix), the tumor susceptibility gene 101 (TSG101) and the chaperones HSP70, Hsc70 and HSP90β, independently of the type of cell origin [92,95,96]. Another mechanism, independent of the ESCRT complex, can be used for exosome release [97]. In the absence of this complex, the endosome pathway may be regulated by the type II neutral sphingomyelase and the tetraspanin family proteins [98]. Therefore, exosomes will contain high levels of tetraspanins such as CD9, CD63 and CD81 [99]. On the other hand, microvesicles are released into the extracellular space by direct shedding of the plasma membrane [93,94]. Therefore, microvesicles can carry both cytosolic and plasma membrane proteins, including the same tetraspanins found on exosomes [100]. Microvesicles can also contain cytoskeletal and heat shock proteins and integrins [101,102]. Finally, apoptotic bodies are formed during the cellular apoptotic process. The process of cell apoptosis is characterized by the condensation of chromatin followed by the degradation of the internal structure of a cell [93,94]. The disintegrated cellular content will be part of the apoptotic bodies’ cargo. Therefore, this type of vesicles can contain proteins associated with several organelles such as histones (nucleus), the heat shock protein HSP60 (mitochondria) and the chaperone GRP78 (endoplasmic reticulum) [103,104,105].
EVs are composed of a phospholipid bilayer that provides protection to their cargo against degradation by the proteases and nucleases present in the external environment [106,107]. EVs encapsulate several molecules, including cytosolic and cytoskeletal proteins as well as enzymes and nucleic acids (mRNA, miRNA, tRNA, rRNA, DNA) [108,109,110]. The surface of the EVs is composed of lipids (ceramide, cholesterol, phosphatidylserine and sphingomyelin) and proteins (transmembrane proteins, antigen presenters and adhesion molecules) [1]. In addition, glycans are also relevant constituents of the EV composition surface [6,86,91,111].
Cancer EVs are able to mediate communication between cells locally and at a distance, and their cargo can influence the behavior of the recipient cell [112]. Importantly, tumor microenvironment stressors, such as hypoxia [113,114], acidosis [115], starvation [116,117], oxidative stress [118,119], radiation [114] and anti-cancer therapies [120], are important regulators of not only EV secretion and trafficking, but also of its molecular composition (as reviewed in [121]). EV cargo is mainly similar to the composition of the parental cell [1]. Although, they still have unique molecular profiles resultant from specific sorting mechanisms during the EV biogenesis process. Particularly, specific patterns of glycans were found enriched in EVs [5,6,122]. In cancer, important modifications occurring in surface glycans, both at cellular and EV level, may constitute important markers for EV detection, isolation and, importantly, for tumoral and non-tumoral EV distinction.

4.1. Extracellular Vesicle Surface Glycans as Relevant Markers for Its Detection and Isolation

The unique presence of certain EV surface glycans holds the potential to be targetable for the development of novel EV detection and isolation methodologies. Cancer EVs are rich in high mannose and complex type N-glycans, polylactosamine, as well as sialylated glycans, namely α2,6-linked sialic acids [6,123] (Figure 1). Detailed analysis of EV glycosylation can provide useful information to facilitate the distinction between different populations of EVs. So far, several strategies have been developed to characterize the glycan profile of EVs. Nevertheless, the identification of certain EV glycans can often fail due to technical difficulties. Importantly, an in situ rolling circle amplification strategy to enhance the detection capacity of the glycans present in cancer EV samples has been reported [87]. Another fluorescent-based strategy, the evanescent-field fluorescence-assisted (EFF) lectin array system, has also proved to successfully profile the glycans present in exosomes derived from mesenchymal stem cells [124]. These methods were able to quantify the detected glycans present in the EV samples due to the fluorescence emitted signals at the time of the glycan recognition. Therefore, these approaches hold the potential to distinguish the type and amount of glycans carried by EVs from different sources and/or to differentiate between the glycans present in EVs and their parent cells.
Different subpopulations of EVs may share size and biochemical characteristics, which makes it impossible to differentiate or specifically isolate them from a given sample [125,126]. In fact, most of the currently available methods for EV isolation will enrich different sub-populations of EVs carrying diverse molecular compositions [127,128,129,130]. Our group previously showed that different EV isolation techniques resulted in an enrichment of diverse EV glycosylated profiles, depending on the capacity of each method to separate cancer EVs from the non-EV content [86]. Furthermore, if EVs from in vitro cell cultures are used, culture conditions are critical for downstream EV glycosylation analysis [86]. Interestingly, asymmetric-flow field-flow fractionation (AF4) technology proved to efficiently isolate distinct EV sub-populations, including exomeres, carrying a specific glycan and protein profile, from a heterogeneous EV source [91]. Similar results were obtained when different EV populations were separated based on their tetraspanin profile and specific glycan signatures could be distinguished according to the host-cell type and each EV subpopulation [131].
Traditional isolation methods are based on EV size and buoyant density, namely, ultracentrifugation-based methods [132,133], microfiltration [134,135] and size exclusion chromatography [136,137]. Others explore the fact that EVs change their solubility and/or aggregate in different solutions, namely, by precipitation [137,138,139]. More recently, isolation methods were developed based on highly specific interactions with the exposed components on the EV surface, called immunoaffinity, or microfluidic technologies [140,141,142,143]. Nevertheless, there is no gold standard EV isolation method and each approach has its own advantages and disadvantages. As glycans comprise a major molecular component of EV surface, they constitute a valuable source of targets to capture and isolate EVs from a heterogeneous sample. Indeed, the use of the lectins peanut agglutinin (PNA) and Artocarpus integrifolia (AIA or Jacalin), that specifically bind to T-antigen, and the Maackia amurensis lectin I (MAL-I), that has a high affinity for Gal or GalNAc residues with an α(2,3)-linked sialic acid, allowed to isolate different sized urinary EVs from healthy samples based on their surface glycosylation profile, with increased yields and higher purity when compared to CD9/CD81/CD63 antibody-based isolation [144]. In addition, the STL lectin, recognizing the N-acetylglucosamine and lactosamine residues, also showed high affinity and specificity when isolating EVs from healthy urine samples [145]. One of the main challenges when using urine samples is the co-purification of the highly glycosylated Tamm–Horsfall protein (THP). This glycoprotein is able to form aggregates and capture EVs, hampering further EV biomarker downstream analysis [146]. However, the use of lectin microarrays allowed the distinction between EV glycosylation and THP glycosylation, which potentiated the isolation of urine EVs with minimal interference of this glycoprotein [70]. A similar approach was also applied to tumor EVs. By coupling a high mannose-type glycan-specific lectin to beads it was possible to capture small EVs from melanoma, glioblastoma, lung and colon cancer cells [147]. Interestingly, CD109, integrin α6 and ADAM10 present on melanoma small EVs were apparently responsible for the identified EV-lectin interaction, as they carry high mannose glycans [147]. These results demonstrated the potential of lectin-conjugated beads to detect and isolate different sub-populations of EVs within a sample, based on its glycosylation profile. In addition, a nanoparticle-based time-resolved fluorescence immunoassay (NP-TRFIA) showed to be able to capture EVs from urine samples and cell supernatants based on the interaction with the tetraspanins and glycan antigens present at the EV surface [148]. This approach also provided a general EV surface glycan profiling, which revealed a differential expression pattern of tumor-associated proteins on more aggressive versus less aggressive prostate cancer cell line-derived EVs [148].
Apart from the affinity-based isolation methods, a commercially available precipitation kit, the ExoGAG, was also developed taking into consideration the glycosylation profile of EVs. This method precipitates EVs out of the solution due to the presence of negatively charged GAG at the EV surface [149]. Interestingly, ExoGAG has proved to effectively isolate EVs from liquid biopsy patient samples with higher yields and purity when compared with UC and allowed the identification of Annexin A2 as an EV marker associated with endometrial cancer staging and recurrence [149].
Despite the technological advances in the field, the study of EVs is still technically challenging. Particularly, innovative and less laborious detection and isolation methods are an urgent need to facilitate an in-depth study of the different EV cargos as a reliable source of biomarkers. The presence of specific patterns of glycans at the EV surface constitute valuable sources to potentiate the development of more sensitive and specific EV detection and isolation methodologies, with the potential to be translated into the clinical setting.

4.2. The Functional Roles of Extracellular Vesicle Glycosylation in Cancer

Up to date, little is still known regarding the role of EV glycosylation in cancer. Nevertheless, the role and impact of glycosylation in EV biodistribution, uptake and protein cargo sorting have already been proven (Table 1).

4.2.1. EV Biodistribution and Uptake

It has been described that alterations at the EV terminal sialylation resulted in a re-direction of the EVs to particular organs in mouse models [150]. In this work, the authors radiolabeled and treated EVs isolated from mouse liver progenitor cells with neuraminidase, a glycosidase that cleaves terminal sialic acid residues, and observed an increased accumulation of neuraminidase treated EVs in the lungs and axillary lymph nodes when compared to the non-treated EVs [150]. As increased sialylation is a common feature of cancer cells, exploring and understanding the role of terminal sialic acids in EV organ tropism may open new avenues to block cancer metastasis. Interestingly, EV sialylation also seems to play a role during the EV uptake by recipient cells [8]. The glycosylation profile of EVs isolated from murine hepatic cell lines was characterized and further manipulated by different glycosidases. For most of the cell lines tested, desialylated EVs had higher uptake efficiency. In other cases, cleavage of EV N-glycans by PNGase F digestion proved to be more efficient during EV uptake than neuraminidase treatment [8]. In a cancer context, it was also demonstrated that removal of the terminal sialylation through neuraminidase treatment also led to a small increase in EV uptake by ovarian cancer cells, although non-significant [152]. It is possible that the decrease in negative charge at EV surface, after sialic acid removal, could facilitate the interaction between EVs and recipient cells, due to the exposure of other carbohydrate ligands at the EV surface that allows for specific lectins to bind and facilitate the EV uptake [152]. In addition, it is established that the proteoglycans present at the cell membrane play major roles during EV-uptake [157]. For instance, the glypican and syndecan, two known heparan sulfate proteoglycans (HSPGs) at the cell surface, mediate the uptake of EVs in glioblastoma cells [9]. Nevertheless, the HSPGs present at the cell membrane are important mediators of EV internalization, but not vital, since perturbation of HSPGs synthesis does not completely inhibit EV uptake, suggesting the existence of HSPG-independent internalization mechanisms [9]. Altogether, these results interestingly suggest that EV and cell glycosylation can impact EV uptake and, therefore, drive specific EV organ biodistribution tropism.
Considering that the presence of certain glycans in cancer cells can regulate the immune response against tumor cells [3], it is possible that cancer EV glycosylation can also affect the immune system. For example, macrophages expressing siglec-1 use this receptor to capture sialylated pathogens [158]. It has been shown in vivo that these macrophages also incorporate highly sialylated EVs, especially enriched in α2,3-sialylation. This effect could be attenuated in siglec-1-deficient mice or when treating EVs with neuraminidase [159]. Considering the high prevalence of α2,3-sialylation in cancer, we can speculate that the presence of this type of glycans in tumor-derived EVs can mediate a communication axis between immune and cancer cells. Although some of the studies mentioned above were performed using non-cancerous cells, the results provided new insights into the potential roles that EV glycosylation modifications might play in a cancer context.

4.2.2. Protein Sorting

Proteins are sorted into EVs through a variety of mechanisms depending on a multitude of factors, such as the class of EV and the protein to be sorted [93,160]. Proteins destined for extracellular vesicle secretion are clustered at the site of EV assembly, prior to their formation. From these proteins, a large portion will carry important glycan modifications [73].
There has been growing evidence supporting the importance of glycosylation in regulating EV protein sorting. Changes in the glycosylation profile acquired during malignant transformation may interfere with EV protein sorting. It was suggested that the conservation of specific glycan structures, such as increased high mannose, polylactosamine, α-2,6 sialylation and complex N-linked glycans and loss of terminal blood group A and B antigens is due to sorting mechanisms of glycoproteins and glycolipids into the EVs [6]. Additionally, it was also demonstrated that the sorting of EWI-2 into the EVs is regulated by its glycosylation, more specifically by complex N-glycans [161]. Both inhibition and site-directed abrogation of these complex N-glycans in EWI-2 hindered its recruitment to the EVs without altering its whole cellular localization [161]. Recently, it was reported that altered expression of the glycosyltransferase FUT8, responsible for the addition of fucose residues to the N-glycan chains, altered the proteome of secreted EVs in prostate cancer [162]. This study suggests that the aberrant expression of fucosylation in prostate cancer cell models leads to a decrease of endosomal sorting proteins in the EVs, such as the ESCRT and other clathrin-mediated endocytosis components, which results in altered protein cargo profiles in the secreted EVs [162]. Furthermore, aberrant O-glycosylation has also been reported as capable of affecting the sorting of some proteins into EVs, including the CD44 protein [163]. This heavily O-glycosylated transmembrane protein has been previously observed to be expressed in EVs carrying truncated O-glycan chains, namely STn [86]. It was observed in colorectal cancer cell line models expressing truncated O-glycans that CD44 is increasingly secreted via EVs when compared to the EVs of their normally glycosylated counterparts [163]. Interestingly, the role of ST6Gal1, a glycosyltransferase that catalyzes the transfer of sialic acids to galactose residues present in N-glycans in an α2,6 position [164], in EV protein cargo sorting has been addressed. Knocking-out ST6Gal1 in the SW620 colorectal cancer cell line model contributed to an increased tumor cell adhesion and migration, through the decreased expression of KAI1 in EVs [165]. This protein, a member of the tetraspanin superfamily, is described to inhibit several relevant signaling pathways during metastasis of cancer cells [166]. On the other hand, the sorting of some proteins into EVs was not affected by its glycosylation status. For instance, Lipocalin 2 (LCN2), a highly conserved protein that is secreted in EVs, has one N-glycosylation site which was speculated to be important in the trafficking of LCN2 into the EV [167]. However, it has been verified that this was not the case, since when treating several cell lines with tunicamycin, an inhibitor of N-glycosylation, LCN2 was still being sorted into EVs [167]. Considering all the studies mentioned above, the role of glycosylation in EV cargo sorting may be dependent on the type of cells and/or the protein to be sorted. Nevertheless, deciphering the role of glycosylation in cancer EV sorting might elucidate novel mechanisms to target cancer progression. It is known that EVs have the ability to reprogram recipient cells and regulate physiological processes both in health and disease conditions, due to their ability to carry and transfer functional cargo [168,169].

4.2.3. Cell Behavior Modulation

Glycans, being part of the EV functional cargo, have been described to play important roles in the cell reprogramming process. EVs derived from breast cancer were able to induce invasion of the recipient cancer cells due to the transfer of the extracellular matrix metalloproteinase inducer (EMMPRIN) [170]. This effect was observed to be dependent on the N-glycosylation status of the EMMPRIN, more precisely on the Asn 160 and Asn 268 glycosylation sites [170]. In another study, treating ovarian cancer cell lines with EVs enriched in CD82 inhibited their cell adhesion. However, when treating the same cell line with EVs enriched with CD82 lacking the N-glycosylation site at Asn 157, no inhibition of cell adhesion in the recipient cells was observed [171]. EVs may also transport active enzymes that could modulate the recipient phenotypes [168]. For instance, Zhang Q et al. reported that cancer EVs carrying the ST6Gal1 enzyme induced an increase in the total α2,6 sialylation of the recipient cell [172]. Taking into account that α2,6 sialylation is a major alteration associated with increased cancer aggressiveness [3], this study suggests a possible mechanism by which EVs can induce cell tumorigenicity.
Considering the critical roles glycans play both in health and cancer situations, further deepening our understanding of the functional roles of EV glycosylation in cancer will open avenues to develop new tools for cancer diagnosis and therapy.

4.3. The Potential Clinical Application of Cancer Extracellular Vesicles Glycosylation

EVs are capable of carrying several bioactive molecules and, depending on their surface composition, hold the potential to be used as natural vehicles for localized drug delivery [173,174]. In addition, as cancer EVs can be found in several biofluids [148] and their cargo partially reflects the content of parental cells [1], these vesicles are also considered promising sources of circulating cancer biomarkers.

4.3.1. EV Glycosylation as Therapeutic Delivery Tools

EVs can be re-engineered to carry specific molecules, either by manipulating their parental cells or by direct functionalization of the EVs [175,176].
Several studies have shown the possibility of using glycosylphosphatidylinositol (GPI) as an anchor to attach specific antigens to the membrane of EVs for target therapy purposes. For example, the HER-2 remained stable after its fusion with GPI and incorporation into murine breast cancer EVs, which induced strong HER-2-specific antibody responses when injected into mice [177]. In addition, fusing anti-EGFR nanobodies to the GPI anchor of neuroblastoma EVs resulted in a significantly increased capacity of these EVs to bind tumor cells that overexpress the EGFR [178]. Furthermore, the incorporation of the GPI-anchored immune-stimulatory molecule interleukin 12 (IL-12) in EVs isolated from different tumor cell lines resulted in increased in vitro T cell proliferation [179]. These studies highlight the potential of modifying the EV surface to successfully transport antigens to their destination site (Figure 2). To explore the real potential of EVs for cancer drug delivery purposes, a reliable system capable of tracking both in vitro and in vivo interactions of these natural nanoparticles is required. Interestingly, a new method of natural particle labeling based on glycan trafficking was recently reported. In this study, azido-sugars were metabolically incorporated into the cellular glycans and further packaged into the EVs, which allowed these EVs to be traceable in vivo [180].
After reaching their target destination, EVs can be incorporated by recipient cells and release their content. As discussed before, the presence of HSPGs on recipient cells has proved to act as receptors for cancer-derived EVs [9]. The structure and function of these HSPGs can be regulated by heparanase [181]. Interestingly, the use of heparanase inhibitors showed to block tumor progression by reducing exosome uptake by receptor cells [11,182,183]. Currently, there are some heparanase inhibitors, such as modified heparins or HS mimetics, whose potential use in the clinic is being tested [184,185,186,187,188]. Examples include chemically modified N-desulfated, N-acetylated and glycol-split heparin derivatives [189] and a heparanase inhibitor [190], that alone or together with lapatinib, resulted in inhibition of the tumor growth in patients with myeloma [186] or in brain metastatic breast cancer, respectively [191].
Interestingly, it was demonstrated that a specific EV glycosylation coating per se can induce a host immunogenic response. In melanoma, the modification of apoptotic EVs surface towards overexpression of high mannose type glycans, a natural ligand of DC-SIGN, increased the uptake of these EVs by monocyte-derived dendritic cells, leading to an increase in CD8+ T cell response [153]. In addition, the enzymatic removal of sialic acids and insertion of palmitoyl-LeY in glioblastoma EVs led to an enhanced EV uptake by dendritic cells in a DC-SIGN dependent manner, a receptor involved in the activation of CD8+ and CD4+ T cell responses [192]. Therefore, these studies showed that modifications of the EV glycan surface hold potential as a vaccination strategy to potentiate an anti-tumor immune response. The capacity of certain EV glycans to naturally stimulate the immune system should be further explored for the development of novel potential immune-related therapies.
A different strategy with the potential to be applied for cancer therapy involves the hemofiltration of the patient circulating exosomes using the Aethlon ADAPT™ system (adaptive dialysis-like affinity platform technology) [193]. This system aims to capture tumoral EVs through interaction with their surface proteins or glycans. The efficacy of the ADAPT™ system was evaluated in patients with end-stage renal disease. It was possible to reduce the circulating hepatitis C virus by targeting the high mannose glycans present on the viral particles [194]. Although there is no concrete data on its usage in removing EVs from the circulation of cancer patients, the ADAPT™ system is a promising strategy to capture tumor EVs based on their glycosylation profile.
The role of EVs in modulating therapeutic resistance has already been reported (as reviewed in [195]). EVs can be used by tumor cells as resistance mechanisms through the packaging and release of drugs by these vesicles [196,197,198]. Furthermore, the transfer of proteins, such as the multidrug resistance P-glycoprotein [199,200,201,202], or specific microRNAs [203,204,205] from drug-resistant cells to drug-sensitive cells can lead to the modulation of gene expression and the acquisition of resistance in recipient cells. Since glycans present on the EV surface are important mediators of the interaction and uptake of these vesicles by the recipient cells [8,9,10,11], changes in the EV glycosylation will alter the intercellular communication between resistant and sensitive cells. EVs are also capable of modulating the immune response. Through the delivery of specific cargo, such as immune-stimulatory or immune-suppressive molecules, EVs can regulate the activity of immune cells (as reviewed in [206,207]). As previously addressed in this review, glycosylation is an important modulator of the immune response, and cancer cells use specific glycan profiles to escape immunosurveillance. Thus, it is possible that tumor-derived EVs carrying these glycan signatures will also suppress the immune response.

4.3.2. Cancer Biomarker Discovery

Besides their potential application for cancer therapy, EVs also represent a valuable source of circulating biomarkers (Table 1). High EV concentrations have been found in several body fluids, including blood, urine, saliva, cerebrospinal fluid, lymph, pleural effusions, semen, bronchoalveolar lavage, bile, synovial fluid, nasal secretions, breast milk, ocular effluent and ascites (as reviewed in [168]). As alterations in cell glycosylation are a common feature of cancer progression, and glycans are highly present in cancer EVs [73,208], the disclosure of cancer EV glycosylation holds a tremendous potential to identify novel reliable cancer EV biomarkers. In fact, most of the currently available cancer biomarkers are based on the detection of glycans, including the sialyl Lewis A antigen (CA19-9) and the STn (CA72-4), or glycoproteins, such as the alpha-fetoprotein (AFP), the prostate-specific antigen (PSA), mucin 16 (CA125), mucin 1 (CA15-3) and the carcinoembryonic antigen (CEA), which are used to follow both patient treatment response and tumor recurrence in several types of cancers (as reviewed in [3,4,209]).
Interestingly, the carbohydrate antigen CA125 was identified in serum-derived exosomes from patients with ovarian cancer and the detected levels were significantly higher in the exosomes when compared to the levels detected directly in the serum of these patients [210]. Yokose and his collaborators also studied the glycan profiles of serum EVs and revealed a significant increase in O-glycosylated EVs in pancreatic cancer patients in the early stages of the disease, even when the patient samples were negative for the CA19-9 antigen [211]. Interestingly, elevated levels of CA19-9 were detected in exosomes from pancreatic cancer patients when compared to healthy samples. The analysis of CA19-9 in exosomes proved to be more sensitive than its direct measurement from the serum, which allowed to identify CA19-9 positive exosomes in patients thought to be negative for the presence of this antigen [212]. In addition, a highly glycosylated form of the CD133 glycoprotein carrying increased levels of sialic acids was found in exosomes from pancreatic cancer patient’s ascites and was also associated with patient survival. Although further studies are needed, these results demonstrate the prognosis potential of CD133-specific glycosylation in pancreatic cancer [213]. In addition to glycoproteins, the glycosphingolipids abundantly present on the surface of prostate cancer EVs have also been reported as promising biomarkers for this type of cancer [214].
The overexpression or de novo synthesis of particular glycans or glycoconjugates during cancer progression holds the potential to differentiate tumor EVs from benign EVs. Indeed, the proteoglycan glypican-1 (GPC1) and the tumor antigen chondroitin sulfate proteoglycan 4 (CSPG4) were detected in tumor exosomes from heterogeneous samples of pancreatic cancer [155] or melanoma [156], respectively. In both cases, these glycoproteins were able to differentiate tumor-derived EVs from non-malignant particles. In a recent study, the proteoglycan versican (VCAN) and the glycoprotein tenascin C (TNC) also proved to be able to distinguish tumor from non-tumor tissues with high sensitivity and specificity, pointing to their use as cancer EVs markers [215]. In the same study, the galactoside-binding soluble 3 binding protein (LGALS3BP) was identified in most of the EV samples [215], which is in line with the previous reports of the presence of LGALS3BP in uveal melanoma [123] and ovarian cancer EVs [72,111]. Interestingly, the LGALS3BP protein was also found to be strongly enriched in the recently discovered cancer exomere particles [91]. Moreover, the glycoprotein basigin (CD147 or EMMPRIN) and the proteoglycan biglycan (BGN) were found enriched in pancreatic tumor EVs when compared to EVs secreted by non-tumor adjacent tissues [215]. In accordance, highly glycosylated variants of EMMPRIN were predominantly detected on cancer patient-derived microvesicles and were positively correlated with poor survival in several types of cancer [216].
In addition, the O-GlcNAc glycosylation has also been found elevated in breast [154] and colorectal cancer EVs [88], when compared to normal conditions. In particular, the O-GlcNAc modification of the transitional endoplasmic reticulum ATPase (TER ATPase) and RuVB-like1 proteins was identified in colorectal metastatic EVs [88]. Elevated levels of O-GlcNAc were also detected in TER ATPase as well as in 70 kDa heat-shock protein (HSP70) proteins present in breast cancer EVs, which may act to protect cytosolic and nuclear proteins against degradation [154]. The elevated presence of this type of glycosylation modification conjugated with specific proteins identified in tumoral EVs when compared to normal conditions suggests its potential use as a biomarker for both breast cancer and metastatic colorectal cancer.
Despite the high potential of EV glycosylation in the discovery of novel cancer biomarkers, studies addressing the glycan profile of blood circulating EVs are still quite scarce. Nevertheless, the N-glycome of exosomes from hepatocellular carcinoma patient samples was characterized using a reverse capture strategy, and the majority of the N-glycans found in EVs from patients with HCC were modified with sialic acids or fucoses, in contrast to the N-glycans identified in EV from healthy samples [151]. Walker et al. also reported significant differences between the glycan profiles identified directly in the plasma or the plasma-derived EVs from the same individuals [217]. Interestingly, a new integrated analytical platform, termed the integrated magnetic analysis of glycans in extracellular vesicles (iMAGE), was developed to directly analyze the EV glycosylation profile in biological samples. This platform aims to facilitate the EV glycome analysis taking advantage of the magnetic nanotechnologies [218]. The effectiveness of this strategy was evaluated by spiking kidney and brain cancer-EVs into urine and serum EV-depleted samples, respectively, and analyzing the glycan signatures. This strategy proved to be efficient in detecting EVs. Subsequently, when analyzing the glycan profile of ascites samples from patients with gastric and colorectal cancer, it was possible to distinguish patients based on their prognosis only by the glycans present at the EV surface. The distinction of these patients was possible by the increased signal of different lectins associated with a poor prognosis, including the Jacalin, ConA, RCA120, PHA-E, STA, LEL, WGA, DSL and LCA lectins. Although further studies are needed to prove the iMAGE platform’s robustness, this new method of profiling glycans may prove to be very useful in the search for novel biomarkers in cancer research [218]. Although several techniques can be used to analyze glycans, their study faces several technical challenges. The most commonly used methods only provide relative and not absolute quantification of the glycans present in a sample and are often based on a targeted search for specific patterns of cancer-associated glycans [219]. In addition, the variability of the results obtained when the same samples are analyzed in different laboratories, in which different methods were used, demonstrates the difficulty in choosing the best methodology, and the need for reference standards that support that choice [220]. Nevertheless, the presence of different glycosylation profiles under normal and cancer conditions highlights the potential of studying EV-specific glycosylation for the identification of novel cancer circulating biomarkers. Indeed, the studies addressing EV glycosylation denote a high potential of EV glycans to distinguish from normal vs. tumoral EVs. Therefore, we believe further in-depth studies of EV glycosylation will bring several benefits for the future of cancer patients.

5. Discussion

Glycosylation is a post-translational modification that plays a variety of roles in both physiological and pathological contexts [221]. Interestingly, some of the aberrant glycosylation signatures acquired during cancer progression have been identified in cancer EVs [86]. These discoveries have boosted a worldwide effort to characterize the glycome of tumor-derived EVs and use this knowledge to develop innovative methods for both EV detection and isolation based on specific glycosylation signatures. Challenges stemming from the structural complexity of glycans and the low yield of current EV isolation methods have hindered our ability to fully elucidate and understand their biological role during disease progression. Nevertheless, different studies already gave insights into the biological roles of altered glycosylation during EV uptake, biodistribution and protein cargo sorting. Indeed, these biological processes were reported to be relevant in cancer progression [169]. Furthermore, EVs constitute an accessible source of glycans and specific glycans/glycoproteins signatures with the potential to predict their origin, and therefore to potentially distinguish EVs released from healthy or cancer tissues or even to identify the type of cancer of origin. Thus, studies characterizing EV glycosylation profiles of healthy individuals and cancer patients resorting to high-resolution methods are a need to identify potential novel cancer biomarkers. As an important remark, the standardization of current EV isolation/characterization protocols across the world remains a milestone to uniformize and ensure the reproducibility of studies conducted in the field, especially those aiming to be translated into the clinical setting [222].
There is still a large number of questions to be answered regarding the functional role of EV glycosylation in cancer as well as challenges on how to address these questions. Future studies aiming to provide these answers will allow us to discover potential EV glycan-based biomarkers for cancer diagnosis/prognosis/patient stratification and develop innovative therapeutic strategies that can be applied for cancer treatment.

Author Contributions

Conceptualization, D.F. and C.A.R.; writing—original draft preparation Á.M.M., C.C.R. and D.F.; writing—review and editing, Á.M.M., C.C.R., D.F. and C.A.R.; supervision, D.F. and C.A.R. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by FEDER funds through the Operational Programme for Competitiveness Factors COMPETE 2020 (POCI-01-0145-FEDER-016585; POCI-01-0145-FEDER-007274) and national funds through the Foundation for Science and Technology (FCT), under the projects: PTDC/BBB-EBI/0567/2014 to C.A.R and UID/BIM/04293/2013; and the project NORTE-01-0145-FEDER-000029, supported by Norte Portugal Regional Programme (NORTE 2020), under the PORTUGAL 2020 Partnership Agreement, through the European Regional Development Fund (ERDF).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

D.F. acknowledges the Foundation for Science and Technology (FCT) under the program Stimulus of Scientific Employment, Individual Support (CEECIND)—3rd Edition (2020.04384.CEECIND). A.M.M. acknowledges the financial support of the Portuguese Cancer League (LPCC) “Bolsa de Investigação em Oncologia LPCC-NRN 2020” and the FCT for the Ph.D. research scholarship (2020.05359.BD).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AF4asymmetric-flow field-flow fractionation
AFPalpha-fetoprotein
BGNbiglycan
CEAcarcinoembryonic antigen
CSPG4tumor antigen chondroitin sulfate proteoglycan 4
DC-SIGNdendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin
ECMextracellular matrix
EFFevanescent-field fluorescence-assisted
EGFRepidermal growth factor receptor
EMMPRINextracellular matrix metalloproteinase inducer
ERendoplasmic reticulum
ESCRTendosomal sorting complex required for transport
EVsextracellular vesicles
FasLfas ligand
GAgolgi apparatus
GAGsglycosaminoglycans
GPC1glypican-1
GPIglycosylphosphatidylinositol
HCChepatocellular carcinoma
HER2human epidermal growth factor receptor 2
HGFhepatocyte growth factor
HSheparan Sulfate
HSPGsheparan sulfate proteoglycans
IL-12interleukin 12
iMAGEintegrated magnetic analysis of glycans in extracellular vesicles
LC/MS-MSliquid chromatography/tandem mass spectrometry
LCN2lipocalin 2
LGALS3BPgalactoside-binding soluble 3 binding protein
MALDI TOFmatrix-assisted laser desorption ionization time-of-flight
MAL-IMaackia amurensis lectin I
MAPKmitogen-activated protein kinase
MVBsmultivesicular bodies
NKNatural Killer
NKG2DNatural Killer group 2D
NP-TRFIAnanoparticle-based time-resolved fluorescence immunoassay
PD-L1programmed death-ligand 1
PNApeanut agglutinin
PSAprostate-specific antigen
RTKreceptor tyrosine kinase
SCsimple-cell
Siglecssialic acid binding immunoglobulin-type lectins
STnSialyl-Tn
TER ATPasetransitional endoplasmic reticulum ATPase
TGF-βtransforming growth factor-β
THPTamm–Horsfall protein
TMZalkylating agent temozolomide
TRAILTNF-related apoptosis-inducing ligand
VEGFvascular endothelial growth factor
VEGFRvascular endothelial growth factor receptor

References

  1. 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. [Google Scholar] [CrossRef] [PubMed]
  2. Christiansen, M.N.; Chik, J.; Lee, L.; Anugraham, M.; Abrahams, J.L.; Packer, N.H. Cell Surface Protein Glycosylation in Cancer. Proteomics 2014, 14, 525–546. [Google Scholar] [CrossRef] [PubMed]
  3. Pinho, S.S.; Reis, C.A. Glycosylation in Cancer: Mechanisms and Clinical Implications. Nat. Rev. Cancer 2015, 15, 540–555. [Google Scholar] [CrossRef] [PubMed]
  4. Mereiter, S.; Balmaña, M.; Campos, D.; Gomes, J.; Reis, C.A. Glycosylation in the Era of Cancer-Targeted Therapy: Where Are We Heading? Cancer Cell 2019, 36, 6–16. [Google Scholar] [CrossRef] [PubMed]
  5. Surman, M.; Hoja-Łukowicz, D.; Szwed, S.; Drożdż, A.; Stępień, E.; Przybyło, M. Human Melanoma-Derived Ectosomes Are Enriched with Specific Glycan Epitopes. Life Sci. 2018, 207, 395–411. [Google Scholar] [CrossRef] [PubMed]
  6. Batista, B.S.; Eng, W.S.; Pilobello, K.T.; Hendricks-Muñoz, K.D.; Mahal, L.K. Identification of a Conserved Glycan Signature for Microvesicles. J. Proteome Res. 2011, 10, 4624–4633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Zhang, X.; Wang, Y. Glycosylation Quality Control by the Golgi Structure. J. Mol. Biol. 2016, 428, 3183–3193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Williams, C.; Pazos, R.; Royo, F.; González, E.; Roura-Ferrer, M.; Martinez, A.; Gamiz, J.; Reichardt, N.-C.; Falcón-Pérez, J.M. Assessing the Role of Surface Glycans of Extracellular Vesicles on Cellular Uptake. Sci. Rep. 2019, 9, 11920. [Google Scholar] [CrossRef] [Green Version]
  9. Christianson, H.C.; Svensson, K.J.; van Kuppevelt, T.H.; Li, J.-P.; Belting, M. Cancer Cell Exosomes Depend on Cell-Surface Heparan Sulfate Proteoglycans for Their Internalization and Functional Activity. Proc. Natl. Acad. Sci. USA 2013, 110, 17380–17385. [Google Scholar] [CrossRef] [Green Version]
  10. Chen, L.; Brigstock, D.R. Integrins and Heparan Sulfate Proteoglycans on Hepatic Stellate Cells (HSC) Are Novel Receptors for HSC-Derived Exosomes. FEBS Lett. 2016, 590, 4263–4274. [Google Scholar] [CrossRef]
  11. Franzen, C.A.; Simms, P.E.; Van Huis, A.F.; Foreman, K.E.; Kuo, P.C.; Gupta, G.N. Characterization of Uptake and Internalization of Exosomes by Bladder Cancer Cells. Biomed. Res. Int. 2014, 2014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Cummings, R.D.; Pierce, J.M. The Challenge and Promise of Glycomics. Chem. Biol. 2014, 21, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Mereiter, S.; Balmaña, M.; Gomes, J.; Magalhães, A.; Reis, C.A. Glycomic Approaches for the Discovery of Targets in Gastrointestinal Cancer. Front. Oncol. 2016, 6, 55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Varki, A. Biological Roles of Glycans. Glycobiology 2017, 27, 3–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Rodrigues, J.G.; Balmaña, M.; Macedo, J.A.; Poças, J.; Fernandes, Â.; de-Freitas-Junior, J.C.M.; Pinho, S.S.; Gomes, J.; Magalhães, A.; Gomes, C.; et al. Glycosylation in Cancer: Selected Roles in Tumour Progression, Immune Modulation and Metastasis. Cell Immunol. 2018, 333, 46–57. [Google Scholar] [CrossRef] [Green Version]
  16. Nagae, M.; Yamaguchi, Y. Function and 3D Structure of the N-Glycans on Glycoproteins. Int. J. Mol. Sci. 2012, 13, 8398–8429. [Google Scholar] [CrossRef] [Green Version]
  17. Gill, D.J.; Clausen, H.; Bard, F. Location, Location, Location: New Insights into O-GalNAc Protein Glycosylation. Trends Cell Biol. 2011, 21, 149–158. [Google Scholar] [CrossRef]
  18. Prabhakar, V.; Capila, I.; Sasisekharan, R. The Structural Elucidation of Glycosaminoglycans. Methods Mol. Biol. 2009, 534, 147–156. [Google Scholar] [CrossRef] [Green Version]
  19. Kumamoto, K.; Goto, Y.; Sekikawa, K.; Takenoshita, S.; Ishida, N.; Kawakita, M.; Kannagi, R. Increased Expression of UDP-Galactose Transporter Messenger RNA in Human Colon Cancer Tissues and Its Implication in Synthesis of Thomsen-Friedenreich Antigen and Sialyl Lewis A/X Determinants. Cancer Res. 2001, 61, 4620–4627. [Google Scholar]
  20. Marcos, N.T.; Bennett, E.P.; Gomes, J.; Magalhaes, A.; Gomes, C.; David, L.; Dar, I.; Jeanneau, C.; DeFrees, S.; Krustrup, D.; et al. ST6GalNAc-I Controls Expression of Sialyl-Tn Antigen in Gastrointestinal Tissues. Front. Biosci. (Elite Ed.) 2011, 3, 1443–1455. [Google Scholar] [CrossRef]
  21. Gill, D.J.; Chia, J.; Senewiratne, J.; Bard, F. Regulation of O-Glycosylation through Golgi-to-ER Relocation of Initiation Enzymes. J. Cell. Biol. 2010, 189, 843–858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Wang, Y.; Ju, T.; Ding, X.; Xia, B.; Wang, W.; Xia, L.; He, M.; Cummings, R.D. Cosmc Is an Essential Chaperone for Correct Protein O-Glycosylation. Proc. Natl. Acad. Sci. USA 2010, 107, 9228–9233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Lau, K.S.; Partridge, E.A.; Grigorian, A.; Silvescu, C.I.; Reinhold, V.N.; Demetriou, M.; Dennis, J.W. Complex N-Glycan Number and Degree of Branching Cooperate to Regulate Cell Proliferation and Differentiation. Cell 2007, 129, 123–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Croci, D.O.; Cerliani, J.P.; Pinto, N.A.; Morosi, L.G.; Rabinovich, G.A. Regulatory Role of Glycans in the Control of Hypoxia-Driven Angiogenesis and Sensitivity to Anti-Angiogenic Treatment. Glycobiology 2014, 24, 1283–1290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Freitas, D.; Campos, D.; Gomes, J.; Pinto, F.; Macedo, J.A.; Matos, R.; Mereiter, S.; Pinto, M.T.; Polónia, A.; Gartner, F.; et al. O-Glycans Truncation Modulates Gastric Cancer Cell Signaling and Transcription Leading to a More Aggressive Phenotype. EBioMedicine 2019, 40, 349–362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Häuselmann, I.; Borsig, L. Altered Tumor-Cell Glycosylation Promotes Metastasis. Front. Oncol. 2014, 4, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Contessa, J.N.; Bhojani, M.S.; Freeze, H.H.; Rehemtulla, A.; Lawrence, T.S. Inhibition of N-Linked Glycosylation Disrupts Receptor Tyrosine Kinase Signaling in Tumor Cells. Cancer Res. 2008, 68, 3803–3809. [Google Scholar] [CrossRef] [Green Version]
  28. Gomes, C.; Osório, H.; Pinto, M.T.; Campos, D.; Oliveira, M.J.; Reis, C.A. Expression of ST3GAL4 Leads to SLe(x) Expression and Induces c-Met Activation and an Invasive Phenotype in Gastric Carcinoma Cells. PLoS One 2013, 8, e66737. [Google Scholar] [CrossRef]
  29. Mereiter, S.; Magalhães, A.; Adamczyk, B.; Jin, C.; Almeida, A.; Drici, L.; Ibáñez-Vea, M.; Gomes, C.; Ferreira, J.A.; Afonso, L.P.; et al. Glycomic Analysis of Gastric Carcinoma Cells Discloses Glycans as Modulators of RON Receptor Tyrosine Kinase Activation in Cancer. Biochim. Biophys. Acta 2016, 1860, 1795–1808. [Google Scholar] [CrossRef]
  30. Langer, M.D.; Guo, H.; Shashikanth, N.; Pierce, J.M.; Leckband, D.E. N-Glycosylation Alters Cadherin-Mediated Intercellular Binding Kinetics. J. Cell Sci. 2012, 125, 2478–2485. [Google Scholar] [CrossRef] [Green Version]
  31. Marsico, G.; Russo, L.; Quondamatteo, F.; Pandit, A. Glycosylation and Integrin Regulation in Cancer. Trends Cancer 2018, 4, 537–552. [Google Scholar] [CrossRef] [PubMed]
  32. Hang, Q.; Isaji, T.; Hou, S.; Wang, Y.; Fukuda, T.; Gu, J. A Key Regulator of Cell Adhesion: Identification and Characterization of Important N-Glycosylation Sites on Integrin A5 for Cell Migration. Mol. Cell Biol. 2017, 37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Läubli, H.; Borsig, L. Altered Cell Adhesion and Glycosylation Promote Cancer Immune Suppression and Metastasis. Front. Immunol. 2019, 10, 2120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Stowell, S.R.; Ju, T.; Cummings, R.D. Protein Glycosylation in Cancer. Annu. Rev. Pathol. 2015, 10, 473–510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Lemmon, M.A.; Schlessinger, J. Cell Signaling by Receptor Tyrosine Kinases. Cell 2010, 141, 1117–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Du, Z.; Lovly, C.M. Mechanisms of Receptor Tyrosine Kinase Activation in Cancer. Mol. Cancer 2018, 17, 58. [Google Scholar] [CrossRef] [PubMed]
  37. Butti, R.; Das, S.; Gunasekaran, V.P.; Yadav, A.S.; Kumar, D.; Kundu, G.C. Receptor Tyrosine Kinases (RTKs) in Breast Cancer: Signaling, Therapeutic Implications and Challenges. Mol. Cancer 2018, 17, 34. [Google Scholar] [CrossRef] [Green Version]
  38. Wang, C.; Yang, Y.; Yang, Z.; Liu, M.; Li, Z.; Sun, L.; Mei, C.; Chen, H.; Chen, L.; Wang, L.; et al. EGF-Mediated Migration Signaling Activated by N-Acetylglucosaminyltransferase-V via Receptor Protein Tyrosine Phosphatase Kappa. Arch. Biochem. Biophys. 2009, 486, 64–72. [Google Scholar] [CrossRef]
  39. Hang, Q.; Isaji, T.; Hou, S.; Zhou, Y.; Fukuda, T.; Gu, J. N-Glycosylation of Integrin A5 Acts as a Switch for EGFR-Mediated Complex Formation of Integrin A5β1 to A6β4. Sci. Rep. 2016, 6, 33507. [Google Scholar] [CrossRef] [Green Version]
  40. Britain, C.M.; Holdbrooks, A.T.; Anderson, J.C.; Willey, C.D.; Bellis, S.L. Sialylation of EGFR by the ST6Gal-I Sialyltransferase Promotes EGFR Activation and Resistance to Gefitinib-Mediated Cell Death. J. Ovarian Res. 2018, 11, 12. [Google Scholar] [CrossRef] [Green Version]
  41. Guo, H.-B.; Johnson, H.; Randolph, M.; Nagy, T.; Blalock, R.; Pierce, M. Specific Posttranslational Modification Regulates Early Events in Mammary Carcinoma Formation. Proc. Natl. Acad. Sci. USA 2010, 107, 21116–21121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Scott, D.A.; Casadonte, R.; Cardinali, B.; Spruill, L.; Mehta, A.S.; Carli, F.; Simone, N.; Kriegsmann, M.; Del Mastro, L.; Kriegsmann, J.; et al. Increases in Tumor N-Glycan Polylactosamines Associated with Advanced HER2-Positive and Triple-Negative Breast Cancer Tissues. Proteom. Clin. Appl. 2019, 13, e1800014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Duarte, H.O.; Balmaña, M.; Mereiter, S.; Osório, H.; Gomes, J.; Reis, C.A. Gastric Cancer Cell Glycosylation as a Modulator of the ErbB2 Oncogenic Receptor. Int. J. Mol. Sci. 2017, 18, 2262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Markowska, A.I.; Jefferies, K.C.; Panjwani, N. Galectin-3 Protein Modulates Cell Surface Expression and Activation of Vascular Endothelial Growth Factor Receptor 2 in Human Endothelial Cells. J. Biol. Chem. 2011, 286, 29913–29921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Croci, D.O.; Cerliani, J.P.; Dalotto-Moreno, T.; Méndez-Huergo, S.P.; Mascanfroni, I.D.; Dergan-Dylon, S.; Toscano, M.A.; Caramelo, J.J.; García-Vallejo, J.J.; Ouyang, J.; et al. Glycosylation-Dependent Lectin-Receptor Interactions Preserve Angiogenesis in Anti-VEGF Refractory Tumors. Cell 2014, 156, 744–758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Ferreira, I.G.; Pucci, M.; Venturi, G.; Malagolini, N.; Chiricolo, M.; Dall’Olio, F. Glycosylation as a Main Regulator of Growth and Death Factor Receptors Signaling. Int. J. Mol. Sci. 2018, 19, 580. [Google Scholar] [CrossRef] [Green Version]
  47. Cecchi, F.; Pajalunga, D.; Fowler, C.A.; Uren, A.; Rabe, D.C.; Peruzzi, B.; Macdonald, N.J.; Blackman, D.K.; Stahl, S.J.; Byrd, R.A.; et al. Targeted Disruption of Heparan Sulfate Interaction with Hepatocyte and Vascular Endothelial Growth Factors Blocks Normal and Oncogenic Signaling. Cancer Cell 2012, 22, 250–262. [Google Scholar] [CrossRef] [Green Version]
  48. Shintani, Y.; Takashima, S.; Asano, Y.; Kato, H.; Liao, Y.; Yamazaki, S.; Tsukamoto, O.; Seguchi, O.; Yamamoto, H.; Fukushima, T.; et al. Glycosaminoglycan Modification of Neuropilin-1 Modulates VEGFR2 Signaling. EMBO J. 2006, 25, 3045–3055. [Google Scholar] [CrossRef] [Green Version]
  49. Weber, G.F.; Bjerke, M.A.; DeSimone, D.W. Integrins and Cadherins Join Forces to Form Adhesive Networks. J. Cell Sci. 2011, 124, 1183–1193. [Google Scholar] [CrossRef] [Green Version]
  50. Paszek, M.J.; DuFort, C.C.; Rossier, O.; Bainer, R.; Mouw, J.K.; Godula, K.; Hudak, J.E.; Lakins, J.N.; Wijekoon, A.C.; Cassereau, L.; et al. The Cancer Glycocalyx Mechanically Primes Integrin-Mediated Growth and Survival. Nature 2014, 511, 319–325. [Google Scholar] [CrossRef] [Green Version]
  51. Yuan, Y.; Wu, L.; Shen, S.; Wu, S.; Burdick, M.M. Effect of Alpha 2,6 Sialylation on Integrin-Mediated Adhesion of Breast Cancer Cells to Fibronectin and Collagen IV. Life Sci. 2016, 149, 138–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Clément, M.; Rocher, J.; Loirand, G.; Le Pendu, J. Expression of Sialyl-Tn Epitopes on Beta1 Integrin Alters Epithelial Cell Phenotype, Proliferation and Haptotaxis. J. Cell Sci. 2004, 117, 5059–5069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Pinho, S.S.; Figueiredo, J.; Cabral, J.; Carvalho, S.; Dourado, J.; Magalhães, A.; Gärtner, F.; Mendonfa, A.M.; Isaji, T.; Gu, J.; et al. E-Cadherin and Adherens-Junctions Stability in Gastric Carcinoma: Functional Implications of Glycosyltransferases Involving N-Glycan Branching Biosynthesis, N-Acetylglucosaminyltransferases III and V. Biochim. Biophys. Acta 2013, 1830, 2690–2700. [Google Scholar] [CrossRef] [PubMed]
  54. Borsig, L. Selectins in Cancer Immunity. Glycobiology 2018, 28, 648–655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. McEver, R.P. Selectins: Initiators of Leucocyte Adhesion and Signalling at the Vascular Wall. Cardiovasc. Res. 2015, 107, 331–339. [Google Scholar] [CrossRef] [Green Version]
  56. Ding, D.; Yao, Y.; Zhang, S.; Su, C.; Zhang, Y. C-Type Lectins Facilitate Tumor Metastasis. Oncol. Lett. 2017, 13, 13–21. [Google Scholar] [CrossRef] [PubMed]
  57. Esposito, M.; Mondal, N.; Greco, T.M.; Wei, Y.; Spadazzi, C.; Lin, S.-C.; Zheng, H.; Cheung, C.; Magnani, J.L.; Lin, S.-H.; et al. Bone Vascular Niche E-Selectin Induces Mesenchymal-Epithelial Transition and Wnt Activation in Cancer Cells to Promote Bone Metastasis. Nat. Cell Biol. 2019, 21, 627–639. [Google Scholar] [CrossRef]
  58. Jandus, C.; Boligan, K.F.; Chijioke, O.; Liu, H.; Dahlhaus, M.; Démoulins, T.; Schneider, C.; Wehrli, M.; Hunger, R.E.; Baerlocher, G.M.; et al. Interactions between Siglec-7/9 Receptors and Ligands Influence NK Cell-Dependent Tumor Immunosurveillance. J. Clin. Investig. 2014, 124, 1810–1820. [Google Scholar] [CrossRef] [Green Version]
  59. Hudak, J.E.; Canham, S.M.; Bertozzi, C.R. Glycocalyx Engineering Reveals a Siglec-Based Mechanism for NK Cell Immunoevasion. Nat. Chem. Biol. 2014, 10, 69–75. [Google Scholar] [CrossRef] [Green Version]
  60. Macauley, M.S.; Crocker, P.R.; Paulson, J.C. Siglec-Mediated Regulation of Immune Cell Function in Disease. Nat. Rev. Immunol. 2014, 14, 653–666. [Google Scholar] [CrossRef] [Green Version]
  61. Daley, D.; Mani, V.R.; Mohan, N.; Akkad, N.; Ochi, A.; Heindel, D.W.; Lee, K.B.; Zambirinis, C.P.; Pandian, G.S.B.; Savadkar, S.; et al. Dectin 1 Activation on Macrophages by Galectin 9 Promotes Pancreatic Carcinoma and Peritumoral Immune Tolerance. Nat. Med. 2017, 23, 556–567. [Google Scholar] [CrossRef] [PubMed]
  62. Kouo, T.; Huang, L.; Pucsek, A.B.; Cao, M.; Solt, S.; Armstrong, T.; Jaffee, E. Galectin-3 Shapes Antitumor Immune Responses by Suppressing CD8+ T Cells via LAG-3 and Inhibiting Expansion of Plasmacytoid Dendritic Cells. Cancer Immunol. Res. 2015, 3, 412–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Demotte, N.; Wieërs, G.; Van Der Smissen, P.; Moser, M.; Schmidt, C.; Thielemans, K.; Squifflet, J.-L.; Weynand, B.; Carrasco, J.; Lurquin, C.; et al. A Galectin-3 Ligand Corrects the Impaired Function of Human CD4 and CD8 Tumor-Infiltrating Lymphocytes and Favors Tumor Rejection in Mice. Cancer Res. 2010, 70, 7476–7488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Merlotti, A.; Malizia, A.L.; Michea, P.; Bonte, P.-E.; Goudot, C.; Carregal, M.S.; Nuñez, N.; Sedlik, C.; Ceballos, A.; Soumelis, V.; et al. Aberrant Fucosylation Enables Breast Cancer Clusterin to Interact with Dendritic Cell-Specific ICAM-Grabbing Non-Integrin (DC-SIGN). Oncoimmunology 2019, 8, e1629257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kunej, T. Rise of Systems Glycobiology and Personalized Glycomedicine: Why and How to Integrate Glycomics with Multiomics Science? OMICS 2019, 23, 615–622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Rudd, P.; Karlsson, N.G.; Khoo, K.-H.; Packer, N.H. Glycomics and Glycoproteomics, 3rd ed.; Cold Spring Harbor Laboratory Press: New York, NY, USA, 2015–2017; Chapter 51.
  67. Hirabayashi, J.; Kuno, A.; Tateno, H. Development and Applications of the Lectin Microarray. Top. Curr. Chem. 2015, 367, 105–124. [Google Scholar] [CrossRef]
  68. Wuhrer, M. Glycomics Using Mass Spectrometry. Glycoconj. J. 2013, 30, 11–22. [Google Scholar] [CrossRef] [Green Version]
  69. Gerlach, J.Q.; Griffin, M.D. Getting to Know the Extracellular Vesicle Glycome. Mol. Biosyst. 2016, 12, 1071–1081. [Google Scholar] [CrossRef]
  70. Gerlach, J.Q.; Krüger, A.; Gallogly, S.; Hanley, S.A.; Hogan, M.C.; Ward, C.J.; Joshi, L.; Griffin, M.D. Surface Glycosylation Profiles of Urine Extracellular Vesicles. PLoS ONE 2013, 8, e74801. [Google Scholar] [CrossRef]
  71. Costa, J.; Gatermann, M.; Nimtz, M.; Kandzia, S.; Glatzel, M.; Conradt, H.S. N-Glycosylation of Extracellular Vesicles from HEK-293 and Glioma Cell Lines. Anal. Chem. 2018, 90, 7871–7879. [Google Scholar] [CrossRef]
  72. Gomes, J.; Gomes-Alves, P.; Carvalho, S.B.; Peixoto, C.; Alves, P.M.; Altevogt, P.; Costa, J. Extracellular Vesicles from Ovarian Carcinoma Cells Display Specific Glycosignatures. Biomolecules 2015, 5, 1741–1761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Williams, C.; Royo, F.; Aizpurua-Olaizola, O.; Pazos, R.; Boons, G.-J.; Reichardt, N.-C.; Falcon-Perez, J.M. Glycosylation of Extracellular Vesicles: Current Knowledge, Tools and Clinical Perspectives. J. Extracell. Vesicles 2018, 7, 1442985. [Google Scholar] [CrossRef] [PubMed]
  74. North, S.J.; Hitchen, P.G.; Haslam, S.M.; Dell, A. Mass Spectrometry in the Analysis of N-Linked and O-Linked Glycans. Curr. Opin. Struct. Biol. 2009, 19, 498–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Schulz, B.L.; Packer, N.H.; Karlsson, N.G. Small-Scale Analysis of O-Linked Oligosaccharides from Glycoproteins and Mucins Separated by Gel Electrophoresis. Anal. Chem. 2002, 74, 6088–6097. [Google Scholar] [CrossRef] [PubMed]
  76. Geyer, H.; Geyer, R. Strategies for Analysis of Glycoprotein Glycosylation. Biochim. Biophys. Acta 2006, 1764, 1853–1869. [Google Scholar] [CrossRef]
  77. Merry, A.H.; Neville, D.C.A.; Royle, L.; Matthews, B.; Harvey, D.J.; Dwek, R.A.; Rudd, P.M. Recovery of Intact 2-Aminobenzamide-Labeled O-Glycans Released from Glycoproteins by Hydrazinolysis. Anal. Biochem. 2002, 304, 91–99. [Google Scholar] [CrossRef]
  78. Kozak, R.P.; Royle, L.; Gardner, R.A.; Fernandes, D.L.; Wuhrer, M. Suppression of Peeling during the Release of O-Glycans by Hydrazinolysis. Anal. Biochem. 2012, 423, 119–128. [Google Scholar] [CrossRef]
  79. King, S.L.; Joshi, H.J.; Schjoldager, K.T.; Halim, A.; Madsen, T.D.; Dziegiel, M.H.; Woetmann, A.; Vakhrushev, S.Y.; Wandall, H.H. Characterizing the O-Glycosylation Landscape of Human Plasma, Platelets, and Endothelial Cells. Blood Adv. 2017, 1, 429–442. [Google Scholar] [CrossRef] [Green Version]
  80. Steentoft, C.; Vakhrushev, S.Y.; Joshi, H.J.; Kong, Y.; Vester-Christensen, M.B.; Schjoldager, K.T.-B.G.; Lavrsen, K.; Dabelsteen, S.; Pedersen, N.B.; Marcos-Silva, L.; et al. Precision Mapping of the Human O-GalNAc Glycoproteome through SimpleCell Technology. EMBO J. 2013, 32, 1478–1488. [Google Scholar] [CrossRef] [Green Version]
  81. Ye, Z.; Mao, Y.; Clausen, H.; Vakhrushev, S.Y. Glyco-DIA: A Method for Quantitative O-Glycoproteomics with in Silico-Boosted Glycopeptide Libraries. Nat. Methods 2019, 16, 902–910. [Google Scholar] [CrossRef]
  82. Campos, D.; Freitas, D.; Gomes, J.; Magalhães, A.; Steentoft, C.; Gomes, C.; Vester-Christensen, M.B.; Ferreira, J.A.; Afonso, L.P.; Santos, L.L.; et al. Probing the O-Glycoproteome of Gastric Cancer Cell Lines for Biomarker Discovery. Mol. Cell. Proteom. 2015, 14, 1616–1629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Campos, D.; Freitas, D.; Gomes, J.; Reis, C.A. Glycoengineered Cell Models for the Characterization of Cancer O-Glycoproteome: An Innovative Strategy for Biomarker Discovery. Expert. Rev. Proteom. 2015, 12, 337–342. [Google Scholar] [CrossRef] [PubMed]
  84. Harada, Y.; Kizuka, Y.; Tokoro, Y.; Kondo, K.; Yagi, H.; Kato, K.; Inoue, H.; Taniguchi, N.; Maruyama, I. N-Glycome Inheritance from Cells to Extracellular Vesicles in B16 Melanomas. FEBS Lett. 2019, 593, 942–951. [Google Scholar] [CrossRef] [PubMed]
  85. Vermassen, T.; D’Herde, K.; Jacobus, D.; Van Praet, C.; Poelaert, F.; Lumen, N.; Callewaert, N.; Decaestecker, K.; Villeirs, G.; Hoebeke, P.; et al. Release of Urinary Extracellular Vesicles in Prostate Cancer Is Associated with Altered Urinary N-Glycosylation Profile. J. Clin. Pathol. 2017, 70, 838–846. [Google Scholar] [CrossRef] [PubMed]
  86. Freitas, D.; Balmaña, M.; Poças, J.; Campos, D.; Osório, H.; Konstantinidi, A.; Vakhrushev, S.Y.; Magalhães, A.; Reis, C.A. Different Isolation Approaches Lead to Diverse Glycosylated Extracellular Vesicle Populations. J. Extracell. Vesicles 2019, 8, 1621131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Feng, Y.; Guo, Y.; Li, Y.; Tao, J.; Ding, L.; Wu, J.; Ju, H. Lectin-Mediated in Situ Rolling Circle Amplification on Exosomes for Probing Cancer-Related Glycan Pattern. Anal. Chim. Acta 2018, 1039, 108–115. [Google Scholar] [CrossRef]
  88. Chaiyawat, P.; Weeraphan, C.; Netsirisawan, P.; Chokchaichamnankit, D.; Srisomsap, C.; Svasti, J.; Champattanachai, V. Elevated O-GlcNAcylation of Extracellular Vesicle Proteins Derived from Metastatic Colorectal Cancer Cells. Cancer Genom. Proteom. 2016, 13, 387–398. [Google Scholar]
  89. Zou, G.; Benktander, J.D.; Gizaw, S.T.; Gaunitz, S.; Novotny, M.V. Comprehensive Analytical Approach toward Glycomic Characterization and Profiling in Urinary Exosomes. Anal. Chem. 2017, 89, 5364–5372. [Google Scholar] [CrossRef]
  90. Peixoto, A.; Relvas-Santos, M.; Azevedo, R.; Santos, L.L.; Ferreira, J.A. Protein Glycosylation and Tumor Microenvironment Alterations Driving Cancer Hallmarks. Front. Oncol. 2019, 9. [Google Scholar] [CrossRef] [Green Version]
  91. Zhang, H.; Freitas, D.; Kim, H.S.; Fabijanic, K.; Li, Z.; Chen, H.; Mark, M.T.; Molina, H.; Martin, A.B.; Bojmar, L.; et al. Identification of Distinct Nanoparticles and Subsets of Extracellular Vesicles by Asymmetric Flow Field-Flow Fractionation. Nat. Cell. Biol. 2018, 20, 332–343. [Google Scholar] [CrossRef]
  92. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding Light on the Cell Biology of Extracellular Vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef] [PubMed]
  94. Bebelman, M.P.; Smit, M.J.; Pegtel, D.M.; Baglio, S.R. Biogenesis and Function of Extracellular Vesicles in Cancer. Pharmacol. Ther. 2018, 188, 1–11. [Google Scholar] [CrossRef] [PubMed]
  95. Willms, E.; Johansson, H.J.; Mäger, I.; Lee, Y.; Blomberg, K.E.M.; Sadik, M.; Alaarg, A.; Smith, C.I.E.; Lehtiö, J.; EL Andaloussi, S.; et al. Cells Release Subpopulations of Exosomes with Distinct Molecular and Biological Properties. Sci. Rep. 2016, 6, 22519. [Google Scholar] [CrossRef] [PubMed]
  96. Géminard, C.; De Gassart, A.; Blanc, L.; Vidal, M. Degradation of AP2 during Reticulocyte Maturation Enhances Binding of Hsc70 and Alix to a Common Site on TFR for Sorting into Exosomes. Traffic 2004, 5, 181–193. [Google Scholar] [CrossRef] [PubMed]
  97. Stuffers, S.; Sem Wegner, C.; Stenmark, H.; Brech, A. Multivesicular Endosome Biogenesis in the Absence of ESCRTs. Traffic 2009, 10, 925–937. [Google Scholar] [CrossRef]
  98. Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brügger, B.; Simons, M. Ceramide Triggers Budding of Exosome Vesicles into Multivesicular Endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef] [PubMed]
  99. Andreu, Z.; Yáñez-Mó, M. Tetraspanins in Extracellular Vesicle Formation and Function. Front. Immunol. 2014, 5. [Google Scholar] [CrossRef] [Green Version]
  100. Escola, J.M.; Kleijmeer, M.J.; Stoorvogel, W.; Griffith, J.M.; Yoshie, O.; Geuze, H.J. Selective Enrichment of Tetraspan Proteins on the Internal Vesicles of Multivesicular Endosomes and on Exosomes Secreted by Human B-Lymphocytes. J. Biol. Chem. 1998, 273, 20121–20127. [Google Scholar] [CrossRef] [Green Version]
  101. Østergaard, O.; Nielsen, C.T.; Iversen, L.V.; Jacobsen, S.; Tanassi, J.T.; Heegaard, N.H.H. Quantitative Proteome Profiling of Normal Human Circulating Microparticles. J. Proteome. Res. 2012, 11, 2154–2163. [Google Scholar] [CrossRef]
  102. Heijnen, H.F.; Schiel, A.E.; Fijnheer, R.; Geuze, H.J.; Sixma, J.J. Activated Platelets Release Two Types of Membrane Vesicles: Microvesicles by Surface Shedding and Exosomes Derived from Exocytosis of Multivesicular Bodies and Alpha-Granules. Blood 1999, 94, 3791–3799. [Google Scholar] [CrossRef] [PubMed]
  103. Théry, C.; Boussac, M.; Véron, P.; Ricciardi-Castagnoli, P.; Raposo, G.; Garin, J.; Amigorena, S. Proteomic Analysis of Dendritic Cell-Derived Exosomes: A Secreted Subcellular Compartment Distinct from Apoptotic Vesicles. J. Immunol. 2001, 166, 7309–7318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Vig, S.; Buitinga, M.; Rondas, D.; Crèvecoeur, I.; van Zandvoort, M.; Waelkens, E.; Eizirik, D.L.; Gysemans, C.; Baatsen, P.; Mathieu, C.; et al. Cytokine-Induced Translocation of GRP78 to the Plasma Membrane Triggers a pro-Apoptotic Feedback Loop in Pancreatic Beta Cells. Cell Death Dis. 2019, 10, 309. [Google Scholar] [CrossRef] [PubMed]
  105. Campanella, C.; D’Anneo, A.; Gammazza, A.M.; Bavisotto, C.C.; Barone, R.; Emanuele, S.; Lo Cascio, F.; Mocciaro, E.; Fais, S.; De Macario, E.C.; et al. The Histone Deacetylase Inhibitor SAHA Induces HSP60 Nitration and Its Extracellular Release by Exosomal Vesicles in Human Lung-Derived Carcinoma Cells. Oncotarget 2015, 7, 28849–28867. [Google Scholar] [CrossRef] [PubMed]
  106. Kusuma, G.D.; Barabadi, M.; Tan, J.L.; Morton, D.A.V.; Frith, J.E.; Lim, R. To Protect and to Preserve: Novel Preservation Strategies for Extracellular Vesicles. Front. Pharmacol. 2018, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Kalra, H.; Drummen, G.P.C.; Mathivanan, S. Focus on Extracellular Vesicles: Introducing the Next Small Big Thing. Int. J. Mol. Sci. 2016, 17, 170. [Google Scholar] [CrossRef] [Green Version]
  108. O’Brien, K.; Breyne, K.; Ughetto, S.; Laurent, L.C.; Breakefield, X.O. RNA Delivery by Extracellular Vesicles in Mammalian Cells and Its Applications. Nat. Rev. Mol. Cell Biol. 2020, 21, 585–606. [Google Scholar] [CrossRef]
  109. Momen-Heravi, F.; Getting, S.J.; Moschos, S.A. Extracellular Vesicles and Their Nucleic Acids for Biomarker Discovery. Pharmacol. Ther. 2018, 192, 170–187. [Google Scholar] [CrossRef] [Green Version]
  110. Massaro, C.; Sgueglia, G.; Frattolillo, V.; Baglio, S.R.; Altucci, L.; Dell’Aversana, C. Extracellular Vesicle-Based Nucleic Acid Delivery: Current Advances and Future Perspectives in Cancer Therapeutic Strategies. Pharmaceutics 2020, 12, 980. [Google Scholar] [CrossRef]
  111. Escrevente, C.; Grammel, N.; Kandzia, S.; Zeiser, J.; Tranfield, E.M.; Conradt, H.S.; Costa, J. Sialoglycoproteins and N-Glycans from Secreted Exosomes of Ovarian Carcinoma Cells. PLoS ONE 2013, 8, e78631. [Google Scholar] [CrossRef] [Green Version]
  112. Becker, A.; Thakur, B.K.; Weiss, J.M.; Kim, H.S.; Peinado, H.; Lyden, D. Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis. Cancer Cell 2016, 30, 836–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. King, H.W.; Michael, M.Z.; Gleadle, J.M. Hypoxic Enhancement of Exosome Release by Breast Cancer Cells. BMC Cancer 2012, 12, 421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Wysoczynski, M.; Ratajczak, M.Z. Lung Cancer Secreted Microvesicles: Underappreciated Modulators of Microenvironment in Expanding Tumors. Int. J. Cancer 2009, 125, 1595–1603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Parolini, I.; Federici, C.; Raggi, C.; Lugini, L.; Palleschi, S.; De Milito, A.; Coscia, C.; Iessi, E.; Logozzi, M.; Molinari, A.; et al. Microenvironmental PH Is a Key Factor for Exosome Traffic in Tumor Cells. J. Biol. Chem. 2009, 284, 34211–34222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Haraszti, R.A.; Miller, R.; Dubuke, M.L.; Rockwell, H.E.; Coles, A.H.; Sapp, E.; Didiot, M.-C.; Echeverria, D.; Stoppato, M.; Sere, Y.Y.; et al. Serum Deprivation of Mesenchymal Stem Cells Improves Exosome Activity and Alters Lipid and Protein Composition. iScience 2019, 16, 230–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Garcia, N.A.; Ontoria-Oviedo, I.; González-King, H.; Diez-Juan, A.; Sepúlveda, P. Glucose Starvation in Cardiomyocytes Enhances Exosome Secretion and Promotes Angiogenesis in Endothelial Cells. PLoS ONE 2015, 10, e0138849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Hedlund, M.; Nagaeva, O.; Kargl, D.; Baranov, V.; Mincheva-Nilsson, L. Thermal- and Oxidative Stress Causes Enhanced Release of NKG2D Ligand-Bearing Immunosuppressive Exosomes in Leukemia/Lymphoma T and B Cells. PLoS ONE 2011, 6, e16899. [Google Scholar] [CrossRef]
  119. Eldh, M.; Ekström, K.; Valadi, H.; Sjöstrand, M.; Olsson, B.; Jernås, M.; Lötvall, J. Exosomes Communicate Protective Messages during Oxidative Stress; Possible Role of Exosomal Shuttle RNA. PLoS ONE 2010, 5, e15353. [Google Scholar] [CrossRef] [Green Version]
  120. Lv, L.-H.; Wan, Y.-L.; Lin, Y.; Zhang, W.; Yang, M.; Li, G.-L.; Lin, H.-M.; Shang, C.-Z.; Chen, Y.-J.; Min, J. 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. J. Biol. Chem. 2012, 287, 15874–15885. [Google Scholar] [CrossRef] [Green Version]
  121. Kucharzewska, P.; Belting, M. Emerging Roles of Extracellular Vesicles in the Adaptive Response of Tumour Cells to Microenvironmental Stress. J. Extracell. Vesicles 2013, 2. [Google Scholar] [CrossRef]
  122. Guo, Y.; Tao, J.; Li, Y.; Feng, Y.; Ju, H.; Wang, Z.; Ding, L. Quantitative Localized Analysis Reveals Distinct Exosomal Protein-Specific Glycosignatures: Implications in Cancer Cell Subtyping, Exosome Biogenesis, and Function. J. Am. Chem. Soc. 2020, 142, 7404–7412. [Google Scholar] [CrossRef] [PubMed]
  123. Surman, M.; Hoja-Łukowicz, D.; Szwed, S.; Kędracka-Krok, S.; Jankowska, U.; Kurtyka, M.; Drożdż, A.; Lityńska, A.; Stępień, E.; Przybyło, M. An Insight into the Proteome of Uveal Melanoma-Derived Ectosomes Reveals the Presence of Potentially Useful Biomarkers. Int. J. Mol. Sci. 2019, 20, 3789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Shimoda, A.; Tahara, Y.; Sawada, S.-I.; Sasaki, Y.; Akiyoshi, K. Glycan Profiling Analysis Using Evanescent-Field Fluorescence-Assisted Lectin Array: Importance of Sugar Recognition for Cellular Uptake of Exosomes from Mesenchymal Stem Cells. Biochem. Biophys. Res. Commun. 2017, 491, 701–707. [Google Scholar] [CrossRef] [PubMed]
  125. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Ramirez, M.I.; Amorim, M.G.; Gadelha, C.; Milic, I.; Welsh, J.A.; Freitas, V.M.; Nawaz, M.; Akbar, N.; Couch, Y.; Makin, L.; et al. Technical Challenges of Working with Extracellular Vesicles. Nanoscale 2018, 10, 881–906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Williams, C.; Palviainen, M.; Reichardt, N.-C.; Siljander, P.R.-M.; Falcón-Pérez, J.M. Metabolomics Applied to the Study of Extracellular Vesicles. Metabolites 2019, 9, 276. [Google Scholar] [CrossRef] [Green Version]
  128. Tang, Y.-T.; Huang, Y.-Y.; Zheng, L.; Qin, S.-H.; Xu, X.-P.; An, T.-X.; Xu, Y.; Wu, Y.-S.; Hu, X.-M.; Ping, B.-H.; et al. Comparison of Isolation Methods of Exosomes and Exosomal RNA from Cell Culture Medium and Serum. Int. J. Mol. Med. 2017, 40, 834–844. [Google Scholar] [CrossRef] [Green Version]
  129. Royo, F.; Zuñiga-Garcia, P.; Sanchez-Mosquera, P.; Egia, A.; Perez, A.; Loizaga, A.; Arceo, R.; Lacasa, I.; Rabade, A.; Arrieta, E.; et al. Different EV Enrichment Methods Suitable for Clinical Settings Yield Different Subpopulations of Urinary Extracellular Vesicles from Human Samples. J. Extracell. Vesicles 2016, 5, 29497. [Google Scholar] [CrossRef]
  130. Van Deun, J.; Mestdagh, P.; Sormunen, R.; Cocquyt, V.; Vermaelen, K.; Vandesompele, J.; Bracke, M.; De Wever, O.; Hendrix, A. The Impact of Disparate Isolation Methods for Extracellular Vesicles on Downstream RNA Profiling. J. Extracell. Vesicles 2014, 3. [Google Scholar] [CrossRef] [Green Version]
  131. Matsuda, A.; Kuno, A.; Yoshida, M.; Wagatsuma, T.; Sato, T.; Miyagishi, M.; Zhao, J.; Suematsu, M.; Kabe, Y.; Narimatsu, H. Comparative Glycomic Analysis of Exosome Subpopulations Derived from Pancreatic Cancer Cell Lines. J. Proteome Res. 2020, 19, 2516–2524. [Google Scholar] [CrossRef]
  132. Tauro, B.J.; Greening, D.W.; Mathias, R.A.; Ji, H.; Mathivanan, S.; Scott, A.M.; Simpson, R.J. Comparison of Ultracentrifugation, Density Gradient Separation, and Immunoaffinity Capture Methods for Isolating Human Colon Cancer Cell Line LIM1863-Derived Exosomes. Methods 2012, 56, 293–304. [Google Scholar] [CrossRef] [PubMed]
  133. Théry, C.; Amigorena, S.; Raposo, G.; Clayton, A. Isolation and Characterization of Exosomes from Cell Culture Supernatants and Biological Fluids. Curr. Protoc. Cell Biol. 2006, 30, 3–22. [Google Scholar] [CrossRef] [PubMed]
  134. Salih, M.; Zietse, R.; Hoorn, E.J. Urinary Extracellular Vesicles and the Kidney: Biomarkers and Beyond. Am. J. Physiol. Renal. Physiol. 2014, 306, F1251–F1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Taylor, D.D.; Shah, S. Methods of Isolating Extracellular Vesicles Impact Down-Stream Analyses of Their Cargoes. Methods 2015, 87, 3–10. [Google Scholar] [CrossRef] [PubMed]
  136. Taylor, D.D.; Zacharias, W.; Gercel-Taylor, C. Exosome Isolation for Proteomic Analyses and RNA Profiling. Methods Mol. Biol. 2011, 728, 235–246. [Google Scholar] [CrossRef]
  137. Gámez-Valero, A.; Monguió-Tortajada, M.; Carreras-Planella, L.; la Franquesa, M.; Beyer, K.; Borràs, F.E. Size-Exclusion Chromatography-Based Isolation Minimally Alters Extracellular Vesicles’ Characteristics Compared to Precipitating Agents. Sci. Rep. 2016, 6, 33641. [Google Scholar] [CrossRef] [Green Version]
  138. Karttunen, J.; Heiskanen, M.; Navarro-Ferrandis, V.; Das Gupta, S.; Lipponen, A.; Puhakka, N.; Rilla, K.; Koistinen, A.; Pitkänen, A. Precipitation-Based Extracellular Vesicle Isolation from Rat Plasma Co-Precipitate Vesicle-Free MicroRNAs. J. Extracell. Vesicles 2018, 8. [Google Scholar] [CrossRef]
  139. Bickmore, D.C.; Miklavcic, J.J. Characterization of Extracellular Vesicles Isolated From Human Milk Using a Precipitation-Based Method. Front. Nutr. 2020, 7. [Google Scholar] [CrossRef] [Green Version]
  140. He, M.; Crow, J.; Roth, M.; Zeng, Y.; Godwin, A.K. Integrated Immunoisolation and Protein Analysis of Circulating Exosomes Using Microfluidic Technology. Lab. Chip. 2014, 14, 3773–3780. [Google Scholar] [CrossRef] [Green Version]
  141. Kanwar, S.S.; Dunlay, C.J.; Simeone, D.M.; Nagrath, S. Microfluidic Device (ExoChip) for on-Chip Isolation, Quantification and Characterization of Circulating Exosomes. Lab. Chip. 2014, 14, 1891–1900. [Google Scholar] [CrossRef]
  142. Greening, D.W.; Xu, R.; Ji, H.; Tauro, B.J.; Simpson, R.J. A Protocol for Exosome Isolation and Characterization: Evaluation of Ultracentrifugation, Density-Gradient Separation, and Immunoaffinity Capture Methods. Methods Mol. Biol. 2015, 1295, 179–209. [Google Scholar] [CrossRef] [PubMed]
  143. Oksvold, M.P.; Neurauter, A.; Pedersen, K.W. Magnetic Bead-Based Isolation of Exosomes. Methods Mol. Biol. 2015, 1218, 465–481. [Google Scholar] [CrossRef] [PubMed]
  144. Gerlach, J.Q.; Maguire, C.M.; Krüger, A.; Joshi, L.; Prina-Mello, A.; Griffin, M.D. Urinary Nanovesicles Captured by Lectins or Antibodies Demonstrate Variations in Size and Surface Glycosylation Profile. Nanomedicine (Lond.) 2017, 12, 1217–1229. [Google Scholar] [CrossRef] [PubMed]
  145. Echevarria, J.; Royo, F.; Pazos, R.; Salazar, L.; Falcon-Perez, J.M.; Reichardt, N.-C. Microarray-Based Identification of Lectins for the Purification of Human Urinary Extracellular Vesicles Directly from Urine Samples. Chembiochem 2014, 15, 1621–1626. [Google Scholar] [CrossRef] [PubMed]
  146. Wachalska, M.; Koppers-Lalic, D.; van Eijndhoven, M.; Pegtel, M.; Geldof, A.A.; Lipinska, A.D.; van Moorselaar, R.J.; Bijnsdorp, I.V. Protein Complexes in Urine Interfere with Extracellular Vesicle Biomarker Studies. J. Circ. Biomark. 2016, 5, 4. [Google Scholar] [CrossRef] [PubMed]
  147. Yamamoto, M.; Harada, Y.; Suzuki, T.; Fukushige, T.; Yamakuchi, M.; Kanekura, T.; Dohmae, N.; Hori, K.; Maruyama, I. Application of High-Mannose-Type Glycan-Specific Lectin from Oscillatoria Agardhii for Affinity Isolation of Tumor-Derived Extracellular Vesicles. Anal. Biochem. 2019, 580, 21–29. [Google Scholar] [CrossRef]
  148. Islam, M.K.; Syed, P.; Lehtinen, L.; Leivo, J.; Gidwani, K.; Wittfooth, S.; Pettersson, K.; Lamminmäki, U. A Nanoparticle-Based Approach for the Detection of Extracellular Vesicles. Sci. Rep. 2019, 9, 10038. [Google Scholar] [CrossRef]
  149. Herrero, C.; de la Fuente, A.; Casas-Arozamena, C.; Sebastian, V.; Prieto, M.; Arruebo, M.; Abalo, A.; Colás, E.; Moreno-Bueno, G.; Gil-Moreno, A.; et al. Extracellular Vesicles-Based Biomarkers Represent a Promising Liquid Biopsy in Endometrial Cancer. Cancers (Basel) 2019, 11, 2000. [Google Scholar] [CrossRef] [Green Version]
  150. Royo, F.; Cossío, U.; Ruiz de Angulo, A.; Llop, J.; Falcon-Perez, J.M. Modification of the Glycosylation of Extracellular Vesicles Alters Their Biodistribution in Mice. Nanoscale 2019, 11, 1531–1537. [Google Scholar] [CrossRef] [Green Version]
  151. Lv, J.; Wang, Z.; Li, F.; Zhang, Y.; Lu, H. Reverse Capture for Selectively and Sensitively Revealing the N-Glycome of Serum Exosomes. Chem. Commun. (Camb.) 2019, 55, 14339–14342. [Google Scholar] [CrossRef]
  152. Escrevente, C.; Keller, S.; Altevogt, P.; Costa, J. Interaction and Uptake of Exosomes by Ovarian Cancer Cells. BMC Cancer 2011, 11, 108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Horrevorts, S.K.; Stolk, D.A.; van de Ven, R.; Hulst, M.; van Het Hof, B.; Duinkerken, S.; Heineke, M.H.; Ma, W.; Dusoswa, S.A.; Nieuwland, R.; et al. Glycan-Modified Apoptotic Melanoma-Derived Extracellular Vesicles as Antigen Source for Anti-Tumor Vaccination. Cancers (Basel) 2019, 11, 1266. [Google Scholar] [CrossRef] [Green Version]
  154. Netsirisawan, P.; Chokchaichamnankit, D.; Srisomsap, C.; Svasti, J.; Champattanachai, V. Proteomic Analysis Reveals Aberrant O-GlcNAcylation of Extracellular Proteins from Breast Cancer Cell Secretion. Cancer Genom. Proteom. 2015, 12, 201–209. [Google Scholar]
  155. Melo, S.A.; Luecke, L.B.; Kahlert, C.; Fernandez, A.F.; Gammon, S.T.; Kaye, J.; LeBleu, V.S.; Mittendorf, E.A.; Weitz, J.; Rahbari, N.; et al. Glypican-1 Identifies Cancer Exosomes and Detects Early Pancreatic Cancer. Nature 2015, 523, 177–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Sharma, P.; Ludwig, S.; Muller, L.; Hong, C.S.; Kirkwood, J.M.; Ferrone, S.; Whiteside, T.L. Immunoaffinity-Based Isolation of Melanoma Cell-Derived Exosomes from Plasma of Patients with Melanoma. J. Extracell. Vesicles 2018, 7, 1435138. [Google Scholar] [CrossRef] [PubMed]
  157. Mulcahy, L.A.; Pink, R.C.; Carter, D.R.F. Routes and Mechanisms of Extracellular Vesicle Uptake. J. Extracell. Vesicles 2014, 3. [Google Scholar] [CrossRef] [Green Version]
  158. Klaas, M.; Crocker, P.R. Sialoadhesin in Recognition of Self and Non-Self. Semin. Immunopathol. 2012, 34, 353–364. [Google Scholar] [CrossRef]
  159. Saunderson, S.C.; Dunn, A.C.; Crocker, P.R.; McLellan, A.D. CD169 Mediates the Capture of Exosomes in Spleen and Lymph Node. Blood 2014, 123, 208–216. [Google Scholar] [CrossRef]
  160. Ko, S.Y.; Naora, H. Extracellular Vesicle Membrane-Associated Proteins: Emerging Roles in Tumor Angiogenesis and Anti-Angiogenesis Therapy Resistance. Int. J. Mol. Sci. 2020, 21, 5418. [Google Scholar] [CrossRef]
  161. Liang, Y.; Eng, W.S.; Colquhoun, D.R.; Dinglasan, R.R.; Graham, D.R.; Mahal, L.K. Complex N-Linked Glycans Serve as a Determinant for Exosome/Microvesicle Cargo Recruitment. J. Biol. Chem. 2014, 289, 32526–32537. [Google Scholar] [CrossRef] [Green Version]
  162. Clark, D.J.; Schnaubelt, M.; Hoti, N.; Hu, Y.; Zhou, Y.; Gooya, M.; Zhang, H. Impact of Increased FUT8 Expression on the Extracellular Vesicle Proteome in Prostate Cancer Cells. J. Proteome Res. 2020, 19, 2195–2205. [Google Scholar] [CrossRef] [PubMed]
  163. Gao, T.; Wen, T.; Ge, Y.; Liu, J.; Yang, L.; Jiang, Y.; Dong, X.; Liu, H.; Yao, J.; An, G. Disruption of Core 1-Mediated O-Glycosylation Oppositely Regulates CD44 Expression in Human Colon Cancer Cells and Tumor-Derived Exosomes. Biochem. Biophys. Res. Commun. 2020, 521, 514–520. [Google Scholar] [CrossRef] [PubMed]
  164. Dall’Olio, F. The Sialyl-Alpha2,6-Lactosaminyl-Structure: Biosynthesis and Functional Role. Glycoconj. J. 2000, 17, 669–676. [Google Scholar] [CrossRef]
  165. Jung, Y.R.; Park, J.-J.; Jin, Y.B.; Cao, Y.J.; Park, M.-J.; Kim, E.J.; Lee, M. Silencing of ST6Gal I Enhances Colorectal Cancer Metastasis by Down-Regulating KAI1 via Exosome-Mediated Exportation and Thereby Rescues Integrin Signaling. Carcinogenesis 2016, 37, 1089–1097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Zöller, M. Tetraspanins: Push and Pull in Suppressing and Promoting Metastasis. Nat. Rev. Cancer 2009, 9, 40–55. [Google Scholar] [CrossRef] [PubMed]
  167. Borkham-Kamphorst, E.; Van de Leur, E.; Meurer, S.K.; Buhl, E.M.; Weiskirchen, R. N-Glycosylation of Lipocalin 2 Is Not Required for Secretion or Exosome Targeting. Front. Pharmacol. 2018, 9, 426. [Google Scholar] [CrossRef] [Green Version]
  168. Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological Properties of Extracellular Vesicles and Their Physiological Functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Xu, R.; Rai, A.; Chen, M.; Suwakulsiri, W.; Greening, D.W.; Simpson, R.J. Extracellular Vesicles in Cancer—Implications for Future Improvements in Cancer Care. Nat. Rev. Clin. Oncol. 2018, 15, 617–638. [Google Scholar] [CrossRef]
  170. Menck, K.; Scharf, C.; Bleckmann, A.; Dyck, L.; Rost, U.; Wenzel, D.; Dhople, V.M.; Siam, L.; Pukrop, T.; Binder, C.; et al. Tumor-Derived Microvesicles Mediate Human Breast Cancer Invasion through Differentially Glycosylated EMMPRIN. J. Mol. Cell. Biol. 2015, 7, 143–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Li, J.; Xu, J.; Li, L.; Ianni, A.; Kumari, P.; Liu, S.; Sun, P.; Braun, T.; Tan, X.; Xiang, R.; et al. MGAT3-Mediated Glycosylation of Tetraspanin CD82 at Asparagine 157 Suppresses Ovarian Cancer Metastasis by Inhibiting the Integrin Signaling Pathway. Theranostics 2020, 10, 6467–6482. [Google Scholar] [CrossRef]
  172. Zhang, Q.; Higginbotham, J.N.; Jeppesen, D.K.; Yang, Y.-P.; Li, W.; McKinley, E.T.; Graves-Deal, R.; Ping, J.; Britain, C.M.; Dorsett, K.A.; et al. Transfer of Functional Cargo in Exomeres. Cell Rep. 2019, 27, 940–954.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Tang, K.; Zhang, Y.; Zhang, H.; Xu, P.; Liu, J.; Ma, J.; Lv, M.; Li, D.; Katirai, F.; Shen, G.-X.; et al. Delivery of Chemotherapeutic Drugs in Tumour Cell-Derived Microparticles. Nat. Commun. 2012, 3, 1282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Sun, D.; Zhuang, X.; Xiang, X.; Liu, Y.; Zhang, S.; Liu, C.; Barnes, S.; Grizzle, W.; Miller, D.; Zhang, H.-G. A Novel Nanoparticle Drug Delivery System: The Anti-Inflammatory Activity of Curcumin Is Enhanced When Encapsulated in Exosomes. Mol. Ther. 2010, 18, 1606–1614. [Google Scholar] [CrossRef] [PubMed]
  175. Armstrong, J.P.K.; Holme, M.N.; Stevens, M.M. Re-Engineering Extracellular Vesicles as Smart Nanoscale Therapeutics. ACS Nano 2017, 11, 69–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Villata, S.; Canta, M.; Cauda, V. EVs and Bioengineering: From Cellular Products to Engineered Nanomachines. Int. J. Mol. Sci. 2020, 21, 6048. [Google Scholar] [CrossRef] [PubMed]
  177. Patel, J.M.; Vartabedian, V.F.; Bozeman, E.N.; Caoyonan, B.E.; Srivatsan, S.; Pack, C.D.; Dey, P.; D’Souza, M.J.; Yang, L.; Selvaraj, P. Plasma Membrane Vesicles Decorated with Glycolipid-Anchored Antigens and Adjuvants via Protein Transfer as an Antigen Delivery Platform for Inhibition of Tumor Growth. Biomaterials 2016, 74, 231–244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Kooijmans, S.A.A.; Aleza, C.G.; Roffler, S.R.; van Solinge, W.W.; Vader, P.; Schiffelers, R.M. Display of GPI-Anchored Anti-EGFR Nanobodies on Extracellular Vesicles Promotes Tumour Cell Targeting. J. Extracell. Vesicles 2016, 5, 31053. [Google Scholar] [CrossRef] [PubMed]
  179. Nagarajan, S.; Selvaraj, P. Human Tumor Membrane Vesicles Modified to Express Glycolipid-Anchored IL-12 by Protein Transfer Induce T Cell Proliferation in Vitro: A Potential Approach for Local Delivery of Cytokines during Vaccination. Vaccine 2006, 24, 2264–2274. [Google Scholar] [CrossRef]
  180. Lee, T.S.; Kim, Y.; Zhang, W.; Song, I.H.; Tung, C.-H. Facile Metabolic Glycan Labeling Strategy for Exosome Tracking. Biochim. Biophys. Acta. Gen. Subj. 2018, 1862, 1091–1100. [Google Scholar] [CrossRef]
  181. Hammond, E.; Khurana, A.; Shridhar, V.; Dredge, K. The Role of Heparanase and Sulfatases in the Modification of Heparan Sulfate Proteoglycans within the Tumor Microenvironment and Opportunities for Novel Cancer Therapeutics. Front. Oncol. 2014, 4, 195. [Google Scholar] [CrossRef]
  182. Atai, N.A.; Balaj, L.; van Veen, H.; Breakefield, X.O.; Jarzyna, P.A.; Van Noorden, C.J.F.; Skog, J.; Maguire, C.A. Heparin Blocks Transfer of Extracellular Vesicles between Donor and Recipient Cells. J. Neurooncol. 2013, 115, 343–351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Sento, S.; Sasabe, E.; Yamamoto, T. Application of a Persistent Heparin Treatment Inhibits the Malignant Potential of Oral Squamous Carcinoma Cells Induced by Tumor Cell-Derived Exosomes. PLoS ONE 2016, 11, e0148454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Dredge, K.; Hammond, E.; Handley, P.; Gonda, T.J.; Smith, M.T.; Vincent, C.; Brandt, R.; Ferro, V.; Bytheway, I. PG545, a Dual Heparanase and Angiogenesis Inhibitor, Induces Potent Anti-Tumour and Anti-Metastatic Efficacy in Preclinical Models. Br. J. Cancer 2011, 104, 635–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Zhou, H.; Roy, S.; Cochran, E.; Zouaoui, R.; Chu, C.L.; Duffner, J.; Zhao, G.; Smith, S.; Galcheva-Gargova, Z.; Karlgren, J.; et al. M402, a Novel Heparan Sulfate Mimetic, Targets Multiple Pathways Implicated in Tumor Progression and Metastasis. PLoS ONE 2011, 6, e21106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Ritchie, J.P.; Ramani, V.C.; Ren, Y.; Naggi, A.; Torri, G.; Casu, B.; Penco, S.; Pisano, C.; Carminati, P.; Tortoreto, M.; et al. SST0001, a Chemically Modified Heparin, Inhibits Myeloma Growth and Angiogenesis via Disruption of the Heparanase/Syndecan-1 Axis. Clin. Cancer Res. 2011, 17, 1382–1393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Basche, M.; Gustafson, D.L.; Holden, S.N.; O’Bryant, C.L.; Gore, L.; Witta, S.; Schultz, M.K.; Morrow, M.; Levin, A.; Creese, B.R.; et al. A Phase I Biological and Pharmacologic Study of the Heparanase Inhibitor PI-88 in Patients with Advanced Solid Tumors. Clin. Cancer Res. 2006, 12, 5471–5480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Liu, C.-J.; Lee, P.-H.; Lin, D.-Y.; Wu, C.-C.; Jeng, L.-B.; Lin, P.-W.; Mok, K.-T.; Lee, W.-C.; Yeh, H.-Z.; Ho, M.-C.; et al. Heparanase Inhibitor PI-88 as Adjuvant Therapy for Hepatocellular Carcinoma after Curative Resection: A Randomized Phase II Trial for Safety and Optimal Dosage. J. Hepatol. 2009, 50, 958–968. [Google Scholar] [CrossRef]
  189. Naggi, A.; Casu, B.; Perez, M.; Torri, G.; Cassinelli, G.; Penco, S.; Pisano, C.; Giannini, G.; Ishai-Michaeli, R.; Vlodavsky, I. Modulation of the Heparanase-Inhibiting Activity of Heparin through Selective Desulfation, Graded N-Acetylation, and Glycol Splitting. J. Biol. Chem. 2005, 280, 12103–12113. [Google Scholar] [CrossRef] [Green Version]
  190. Pala, D.; Rivara, S.; Mor, M.; Milazzo, F.M.; Roscilli, G.; Pavoni, E.; Giannini, G. Kinetic Analysis and Molecular Modeling of the Inhibition Mechanism of Roneparstat (SST0001) on Human Heparanase. Glycobiology 2016, 26, 640–654. [Google Scholar] [CrossRef] [Green Version]
  191. Zhang, L.; Ngo, J.A.; Wetzel, M.D.; Marchetti, D. Heparanase Mediates a Novel Mechanism in Lapatinib-Resistant Brain Metastatic Breast Cancer. Neoplasia 2015, 17, 101–113. [Google Scholar] [CrossRef] [Green Version]
  192. Dusoswa, S.A.; Horrevorts, S.K.; Ambrosini, M.; Kalay, H.; Paauw, N.J.; Nieuwland, R.; Pegtel, M.D.; Würdinger, T.; Van Kooyk, Y.; Garcia-Vallejo, J.J. Glycan Modification of Glioblastoma-Derived Extracellular Vesicles Enhances Receptor-Mediated Targeting of Dendritic Cells. J. Extracell. Vesicles 2019, 8, 1648995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Marleau, A.M.; Chen, C.-S.; Joyce, J.A.; Tullis, R.H. Exosome Removal as a Therapeutic Adjuvant in Cancer. J. Transl. Med. 2012, 10, 134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Tullis, R.H.; Duffin, R.P.; Handley, H.H.; Sodhi, P.; Menon, J.; Joyce, J.A.; Kher, V. Reduction of Hepatitis C Virus Using Lectin Affinity Plasmapheresis in Dialysis Patients. Blood Purif. 2009, 27, 64–69. [Google Scholar] [CrossRef] [PubMed]
  195. Samuel, P.; Fabbri, M.; Carter, D.R.F. Mechanisms of Drug Resistance in Cancer: The Role of Extracellular Vesicles. Proteomics 2017, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Ifergan, I.; Scheffer, G.L.; Assaraf, Y.G. Novel Extracellular Vesicles Mediate an ABCG2-Dependent Anticancer Drug Sequestration and Resistance. Cancer Res. 2005, 65, 10952–10958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Shedden, K.; Xie, X.T.; Chandaroy, P.; Chang, Y.T.; Rosania, G.R. Expulsion of Small Molecules in Vesicles Shed by Cancer Cells: Association with Gene Expression and Chemosensitivity Profiles. Cancer Res. 2003, 63, 4331–4337. [Google Scholar] [PubMed]
  198. Muralidharan-Chari, V.; Kohan, H.G.; Asimakopoulos, A.G.; Sudha, T.; Sell, S.; Kannan, K.; Boroujerdi, M.; Davis, P.J.; Mousa, S.A. Microvesicle Removal of Anticancer Drugs Contributes to Drug Resistance in Human Pancreatic Cancer Cells. Oncotarget 2016, 7, 50365–50379. [Google Scholar] [CrossRef]
  199. Corcoran, C.; Rani, S.; O’Brien, K.; O’Neill, A.; Prencipe, M.; Sheikh, R.; Webb, G.; McDermott, R.; Watson, W.; Crown, J.; et al. Docetaxel-Resistance in Prostate Cancer: Evaluating Associated Phenotypic Changes and Potential for Resistance Transfer via Exosomes. PLoS ONE 2012, 7, e50999. [Google Scholar] [CrossRef]
  200. Zhang, F.; Zhu, Y.; Zhao, Q.; Yang, D.; Dong, Y.; Jiang, L.; Xing, W.; Li, X.; Xing, H.; Shi, M.; et al. Microvesicles Mediate Transfer of P-Glycoprotein to Paclitaxel-Sensitive A2780 Human Ovarian Cancer Cells, Conferring Paclitaxel-Resistance. Eur. J. Pharmacol. 2014, 738, 83–90. [Google Scholar] [CrossRef] [PubMed]
  201. Bebawy, M.; Combes, V.; Lee, E.; Jaiswal, R.; Gong, J.; Bonhoure, A.; Grau, G.E.R. Membrane Microparticles Mediate Transfer of P-Glycoprotein to Drug Sensitive Cancer Cells. Leukemia 2009, 23, 1643–1649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Lv, M.; Zhu, X.; Chen, W.; Zhong, S.; Hu, Q.; Ma, T.; Zhang, J.; Chen, L.; Tang, J.; Zhao, J. Exosomes Mediate Drug Resistance Transfer in MCF-7 Breast Cancer Cells and a Probable Mechanism Is Delivery of P-Glycoprotein. Tumour Biol. 2014, 35, 10773–10779. [Google Scholar] [CrossRef] [PubMed]
  203. Yu, D.-D.; Wu, Y.; Zhang, X.-H.; Lv, M.-M.; Chen, W.-X.; Chen, X.; Yang, S.-J.; Shen, H.; Zhong, S.-L.; Tang, J.-H.; et al. Exosomes from Adriamycin-Resistant Breast Cancer Cells Transmit Drug Resistance Partly by Delivering MiR-222. Tumour Biol. 2016, 37, 3227–3235. [Google Scholar] [CrossRef] [PubMed]
  204. Mikamori, M.; Yamada, D.; Eguchi, H.; Hasegawa, S.; Kishimoto, T.; Tomimaru, Y.; Asaoka, T.; Noda, T.; Wada, H.; Kawamoto, K.; et al. MicroRNA-155 Controls Exosome Synthesis and Promotes Gemcitabine Resistance in Pancreatic Ductal Adenocarcinoma. Sci. Rep. 2017, 7, 42339. [Google Scholar] [CrossRef] [PubMed]
  205. Wu, H.; Zhou, J.; Mei, S.; Wu, D.; Mu, Z.; Chen, B.; Xie, Y.; Ye, Y.; Liu, J. Circulating Exosomal MicroRNA-96 Promotes Cell Proliferation, Migration and Drug Resistance by Targeting LMO7. J. Cell. Mol. Med. 2017, 21, 1228–1236. [Google Scholar] [CrossRef] [PubMed]
  206. Mittal, S.; Gupta, P.; Chaluvally-Raghavan, P.; Pradeep, S. Emerging Role of Extracellular Vesicles in Immune Regulation and Cancer Progression. Cancers (Basel) 2020, 12, 3563. [Google Scholar] [CrossRef]
  207. Xie, F.; Zhou, X.; Fang, M.; Li, H.; Su, P.; Tu, Y.; Zhang, L.; Zhou, F. Extracellular Vesicles in Cancer Immune Microenvironment and Cancer Immunotherapy. Adv. Sci. (Weinh) 2019, 6. [Google Scholar] [CrossRef]
  208. Costa, J. Glycoconjugates from Extracellular Vesicles: Structures, Functions and Emerging Potential as Cancer Biomarkers. Biochim. Biophys. Acta Rev. Cancer 2017, 1868, 157–166. [Google Scholar] [CrossRef]
  209. Reis, C.A.; Osorio, H.; Silva, L.; Gomes, C.; David, L. Alterations in Glycosylation as Biomarkers for Cancer Detection. J. Clin. Pathol. 2010, 63, 322–329. [Google Scholar] [CrossRef]
  210. Chen, Z.; Liang, Q.; Zeng, H.; Zhao, Q.; Guo, Z.; Zhong, R.; Xie, M.; Cai, X.; Su, J.; He, Z.; et al. Exosomal CA125 as A Promising Biomarker for Ovarian Cancer Diagnosis. J. Cancer 2020, 11, 6445–6453. [Google Scholar] [CrossRef]
  211. Yokose, T.; Kabe, Y.; Matsuda, A.; Kitago, M.; Matsuda, S.; Hirai, M.; Nakagawa, T.; Masugi, Y.; Hishiki, T.; Nakamura, Y.; et al. O-Glycan-Altered Extracellular Vesicles: A Specific Serum Marker Elevated in Pancreatic Cancer. Cancers (Basel) 2020, 12, 2469. [Google Scholar] [CrossRef]
  212. Sancho-Albero, M.; Sebastián, V.; Sesé, J.; Pazo-Cid, R.; Mendoza, G.; Arruebo, M.; Martín-Duque, P.; Santamaría, J. Isolation of Exosomes from Whole Blood by a New Microfluidic Device: Proof of Concept Application in the Diagnosis and Monitoring of Pancreatic Cancer. J. Nanobiotechnol. 2020, 18, 150. [Google Scholar] [CrossRef] [PubMed]
  213. Sakaue, T.; Koga, H.; Iwamoto, H.; Nakamura, T.; Ikezono, Y.; Abe, M.; Wada, F.; Masuda, A.; Tanaka, T.; Fukahori, M.; et al. Glycosylation of Ascites-Derived Exosomal CD133: A Potential Prognostic Biomarker in Patients with Advanced Pancreatic Cancer. Med. Mol. Morphol. 2019, 52, 198–208. [Google Scholar] [CrossRef] [PubMed]
  214. Llorente, A.; Skotland, T.; Sylvänne, T.; Kauhanen, D.; Róg, T.; Orłowski, A.; Vattulainen, I.; Ekroos, K.; Sandvig, K. Molecular Lipidomics of Exosomes Released by PC-3 Prostate Cancer Cells. Biochim. Biophys. Acta 2013, 1831, 1302–1309. [Google Scholar] [CrossRef] [PubMed]
  215. Hoshino, A.; Kim, H.S.; Bojmar, L.; Gyan, K.E.; Cioffi, M.; Hernandez, J.; Zambirinis, C.P.; Rodrigues, G.; Molina, H.; Heissel, S.; et al. Extracellular Vesicle and Particle Biomarkers Define Multiple Human Cancers. Cell 2020, 182, 1044–1061.e18. [Google Scholar] [CrossRef] [PubMed]
  216. Menck, K.; Bleckmann, A.; Wachter, A.; Hennies, B.; Ries, L.; Schulz, M.; Balkenhol, M.; Pukrop, T.; Schatlo, B.; Rost, U.; et al. Characterisation of Tumour-Derived Microvesicles in Cancer Patients’ Blood and Correlation with Clinical Outcome. J. Extracell. Vesicles 2017, 6, 1340745. [Google Scholar] [CrossRef] [Green Version]
  217. Walker, S.A.; Aguilar Díaz De León, J.S.; Busatto, S.; Wurtz, G.A.; Zubair, A.C.; Borges, C.R.; Wolfram, J. Glycan Node Analysis of Plasma-Derived Extracellular Vesicles. Cells 2020, 9, 1946. [Google Scholar] [CrossRef] [PubMed]
  218. Wang, Z.; Sun, X.; Natalia, A.; Tang, C.S.L.; Ang, C.B.T.; Ong, C.-A.J.; Teo, M.C.C.; So, J.B.Y.; Shao, H. Dual-Selective Magnetic Analysis of Extracellular Vesicle Glycans. Matter 2020, 2, 150–166. [Google Scholar] [CrossRef] [Green Version]
  219. Etxebarria, J.; Reichardt, N.-C. Methods for the Absolute Quantification of N-Glycan Biomarkers. Biochim. Biophys. Acta 2016, 1860, 1676–1687. [Google Scholar] [CrossRef]
  220. Thobhani, S.; Yuen, C.-T.; Bailey, M.J.A.; Jones, C. Identification and Quantification of N-Linked Oligosaccharides Released from Glycoproteins: An Inter-Laboratory Study. Glycobiology 2009, 19, 201–211. [Google Scholar] [CrossRef] [Green Version]
  221. Schjoldager, K.T.; Narimatsu, Y.; Joshi, H.J.; Clausen, H. Global View of Human Protein Glycosylation Pathways and Functions. Nat. Rev. Mol. Cell. Biol. 2020. [Google Scholar] [CrossRef]
  222. Soekmadji, C.; Li, B.; Huang, Y.; Wang, H.; An, T.; Liu, C.; Pan, W.; Chen, J.; Cheung, L.; Falcon-Perez, J.M.; et al. The Future of Extracellular Vesicles as Theranostics—An ISEV Meeting Report. J. Extracell. Vesicles 2020, 9, 1809766. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of an extracellular vesicle (EV) and its functional cargo. EVs carry a wide variety of functional molecules, including glycoproteins and glycolipids. The major common classes of glycoconjugates found in human cells are depicted on the left. Aberrant tumor-associated glycosylation already identified in cancer EVs are depicted on the right. The glycostructures were represented at the expected EV localization (intern or at the EV membrane) considering the knowledge from the cell glycans. Nevertheless, the specific localization of some of these structures is not yet fully elucidated.
Figure 1. Schematic representation of an extracellular vesicle (EV) and its functional cargo. EVs carry a wide variety of functional molecules, including glycoproteins and glycolipids. The major common classes of glycoconjugates found in human cells are depicted on the left. Aberrant tumor-associated glycosylation already identified in cancer EVs are depicted on the right. The glycostructures were represented at the expected EV localization (intern or at the EV membrane) considering the knowledge from the cell glycans. Nevertheless, the specific localization of some of these structures is not yet fully elucidated.
Cells 10 00109 g001
Figure 2. Potential clinical applications of extracellular vesicle (EV) glycosylation. These include (A) development of glycan-based EV detection and capture methodologies; (B) EV biomarker discovery for cancer diagnostic, prognostic and/or patient stratification based on EV glycomic and glycoproteomic profiling; (C) development of potential novel cancer therapy strategies through the manipulation of EV glycosylation surface.
Figure 2. Potential clinical applications of extracellular vesicle (EV) glycosylation. These include (A) development of glycan-based EV detection and capture methodologies; (B) EV biomarker discovery for cancer diagnostic, prognostic and/or patient stratification based on EV glycomic and glycoproteomic profiling; (C) development of potential novel cancer therapy strategies through the manipulation of EV glycosylation surface.
Cells 10 00109 g002
Table 1. List of the main structures of glycans identified in different cancer extracellular vesicles (EVs) samples and their relevance and potential impact in the EV field.
Table 1. List of the main structures of glycans identified in different cancer extracellular vesicles (EVs) samples and their relevance and potential impact in the EV field.
Glycan StructureCancer TypeSampleRelevance/Potential ImpactReferences
GalNAcα-Thr/Ser (Tn antigen)CervicalCell linesEV biomarker potential[87]
Neu5Acα(2,6)-GalNAcα-Thr/Ser (STn antigen)Gastric, cervicalCell linesEV biomarker potential[86,87]
Gal-β(1,3)-GalNAcα-Thr/Ser (T antigen)Ovarian, cervical Cell linesEV detection
EV biomarker potential
[72,87]
Sialylation (α(2,6)- or α(2,3)-linked)Hepatic, melanoma, cervical, pancreatic, ovarian, colorectal, glioma, breast, gastricCell lines, human serum, healthy individuals’ urine EV detection and capture
EV biomarker potential
EV uptake
EV biodistribution
[5,6,8,70,71,72,84,86,87,89,91,111,122,123,131,144,148,150,151,152]
Core fucosylated N-glycansHepatocellular, breast, glioma, gastric
melanoma
Cell lines, human serumEV detection
EV biomarker potential
[71,84,86,87,91,151]
Terminal fucosylationMelanoma, breastCell linesEV detection[5,91]
Complex N-glycansMelanoma, colorectal, hepatocellular Cell lines, human serum, healthy individuals’ urineEV detection
EV biomarker potential
[6,70,84,89,151]
Branched N-glycansPancreatic, melanoma, breast, ovarian, gastricCell linesEV detection
EV biomarker potential
[5,84,86,91,111,123,131]
Bisected N-glycansMelanoma, pancreatic, ovarian, gastricCell lines, healthy individuals’ urineEV detection
EV biomarker potential
[5,70,72,86,91,123]
High mannose N-glycansMelanoma, glioblastoma, lung, colorectal, ovarian, hepatocellularCell lines, human serum, healthy individuals’ urine EV detection and capture
EV therapy potential
EV biomarker potential
EV uptake
[6,70,72,89,111,123,144,151,153]
PolylactosamineColorectal, melanomaCell linesEV detection[6]
O-GlcNAcBreast, colorectalCell linesEV biomarker potential[88,154]
GAGsEndometrial, ovarian, breastCell linesEV capture[149]
ProteoglycansPancreatic, melanoma, breastCell lines, human and mice serum, human plasmaEV capture
EV biomarker potential
[155,156]
EV, extracellular vesicles; GalNAc, N-Acetylgalactosamine; Thr, threonine; Ser, serine; Neu5Ac, N-Acetylneuraminic acid; STn, sialyl-Tn; Gal, galactose; O-GlcNAc, O-GlcNAcylation; GAGs, glycosaminoglycans.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Martins, Á.M.; Ramos, C.C.; Freitas, D.; Reis, C.A. Glycosylation of Cancer Extracellular Vesicles: Capture Strategies, Functional Roles and Potential Clinical Applications. Cells 2021, 10, 109. https://doi.org/10.3390/cells10010109

AMA Style

Martins ÁM, Ramos CC, Freitas D, Reis CA. Glycosylation of Cancer Extracellular Vesicles: Capture Strategies, Functional Roles and Potential Clinical Applications. Cells. 2021; 10(1):109. https://doi.org/10.3390/cells10010109

Chicago/Turabian Style

Martins, Álvaro M., Cátia C. Ramos, Daniela Freitas, and Celso A. Reis. 2021. "Glycosylation of Cancer Extracellular Vesicles: Capture Strategies, Functional Roles and Potential Clinical Applications" Cells 10, no. 1: 109. https://doi.org/10.3390/cells10010109

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop