Skip to main content
Journal of Lipid Research logoLink to Journal of Lipid Research
. 2018 May 31;59(8):1325–1340. doi: 10.1194/jlr.R083915

Role of sphingolipids in the biogenesis and biological activity of extracellular vesicles

Claudia Verderio *,†,1, Martina Gabrielli *, Paola Giussani §
PMCID: PMC6071771  PMID: 29853528

Abstract

Extracellular vesicles (EVs) are membrane vesicles released by both eukaryotic and prokaryotic cells; they not only serve physiological functions, such as disposal of cellular components, but also play pathophysiologic roles in inflammatory and degenerative diseases. Common molecular mechanisms for EV biogenesis are evident in different cell biological contexts across eukaryotic phyla, and inhibition of this biogenesis may provide an avenue for therapeutic research. The involvement of sphingolipids (SLs) and their enzymes on EV biogenesis and release has not received much attention in current research. Here, we review how SLs participate in EV biogenesis by shaping membrane curvature and how they contribute to EV action in target cells. First, we describe how acid and neutral SMases, by generating the constitutive SL, ceramide, facilitate biogenesis of EVs at the plasma membrane and inside the endocytic compartment. We then discuss the involvement of other SLs, such as sphingosine-1-phosphate and galactosyl-sphingosine, in EV formation and cargo sorting. Last, we look ahead at some biological effects of EVs mediated by changes in SL levels in recipient cells.

Keywords: exosomes, ectosomes, microvesicles, ceramide, sphingosine-1-phosphate, psychosine, sphingomyelinase, endocannabinoids


Extracellular vesicles (EVs) are membrane vesicles released by both eukaryotic and prokaryotic cells, such as Gram-positive and -negative bacteria, mycobacteria, and Archaea (1). EVs are highly heterogeneous in size, structure, and molecular content (2). While the release of bacterial EVs is predominantly by direct outward “blebbing” and pinching of the external cellular membranes (2) in eukaryotic cells, EVs are generated both at the plasma membrane [ectosomes, also called shed microparticles or microvesicles (MVs)] or inside multivesicular bodies (MVBs) (exosomes) through two membrane inversion processes: the inward budding of the limiting membrane of the multivesicular endosome to form intraluminal vesicles and the fusion of MVBs with the plasma membrane. As a result, both ectosomes and exosomes have the same membrane topology of donor cells. However, EVs, especially ectosomes, often lose transmembrane lipid asymmetry and have phosphatidylserine (PS) residues externalized in the outer leaflet of the vesicle (35).

Plasma membrane-derived EVs are on average larger than exosomes in size, ranging from 100 to 300 nm up to 1 μm, while exosomes are generally 30–100 nm in diameter and are isolated by centrifugation at higher speeds compared with ectosomes (100,000 g vs. 10,000 g). Both types of EVs are recognized as mediators of cell-to-cell communication due to their ability to transfer biomolecules among cells (69) and to influence the extracellular microenvironment via regulation of critical nutrients (10, 11). EVs not only serve several physiological functions, such as neurotrophic support (12, 13) and disposal of unwanted cellular components (14), but also play pathophysiological roles in inflammatory and degenerative diseases (1517). Therefore, inhibiting the biogenesis and release of EVs may be an important therapeutic goal, as recently outlined by Tricarico, Clancy, and D’Souza-Schorey (18).

The mechanism of EV biogenesis has been a matter of intense research in the recent years. For discussion of the role of proteins, including members of the endosomal sorting complex required for transport (ESCRT), small GTPases, and glutaminase, in both exo- and ectosome biogenesis and fission, we refer readers to these other excellent reviews (18, 19). Here, we focus on the involvement of sphingolipids (SLs) and their metabolic enzymes in the biogenesis and release of ectosomes and exosomes, which has received less attention.

SLs are a class of lipids located in the plasma membrane and at intracellular organelle membranes, which have not only structural roles but also signaling function. Ceramide (Cer) and sphingosine-1-phosphate (S1P) are the main SLs that act as signaling molecules, controlling a vast number of cellular processes, such as cell growth, adhesion, migration, senescence, cell death, and inflammatory response (2022). In these cellular activities, dynamic balance between S1P and Cer levels is crucial. This process is known as “SL rheostat” (23).

SLs are constituted by a sphingoid base. In bilayer-forming SLs, a fatty acid is tethered/covalently linked to the sphingoid base. All SLs may be modified at the first hydroxyl group with a variable polar head group, and also by an esterified fatty acid tethered to the amino group of the sphingosine (Sph).

Cer, composed of a long-chain amino alcohol, known as Sph, linked to an amide bond to a long-chain fatty acid, is the structural unit of membrane-forming SLs (not present in Sph and S1P), which represent the most abundant SLs in cells. Cer, can originate from “de novo synthesis,” degradation of complex SLs (24), or recycling of long-chain bases through a salvage pathway (21, 25). The “de novo” biosynthesis of SLs begins with the condensation of palmitoyl-CoA with l-serine to form sphinganine and dihydroceramide, respectively, after reduction and acylation. In the de novo biosynthesis, dihydroceramide is then desaturated with the consequent formation of Cer. All of the enzymes involved in the de novo biosynthesis of Cer are localized in the endoplasmic reticulum (ER) membrane and act on the cytosolic surface of this subcellular organelle. The products of the reactions that are catalyzed by these enzymes remain anchored to the ER. In the salvage pathway, Cer is synthesized from Sph (26). During the degradation of complex SLs, glycosphingolipids (GSLs) are constitutively degraded to monosaccharides and Cer (27, 28), while SM is degraded to Cer and phosphocholine (29).

Once generated, Cer can be cleaved by a ceramidase (CDase) to generate Sph that, in turn, can be phosphorylated for the synthesis of the crucial bioactive SL, S1P. On the other hand, Cer is a substrate for various enzymes, mainly by the addition of phosphocholine to produce SM or by the addition of monosaccharides to produce GSLs. Among GSLs, gangliosides are a particular class characterized by the presence of one or more sialic acid residues (26). The GSLs, together with simple SLs, are located in the plasma membrane, where they interact with growth factor receptors, integrins, and other key molecules that participate in cell-cell and cell-matrix interactions (30). Importantly, several SL metabolites have been associated with development of several pathologies, including diabetes, cancer, microbial infections, neurological syndromes, and cardiovascular disease (21, 25, 26, 3135).

In this review, we will critically summarize current knowledge on the action of SL-metabolizing enzymes in the formation and release of the two main types of EVs (ectosomes and exosomes) as well as past and current efforts in targeting these enzymes pharmacologically or genetically for therapeutic purposes. We then focus on the SL content of EVs and discuss the molecular mechanisms underlying the capability of EVs to promote SL metabolism in target cells.

ROLE OF SLs IN EXOSOME AND ECTOSOME RELEASE

Cer-dependent exosome biogenesis: role for neutral SMase

Plasma and endosomal membranes display an asymmetric lipid distribution, with SM and phosphatidylcholine (PC) enriched on the noncytosolic (luminal) side, and PS and other lipid classes enriched in the leaflet facing cytosol (36). Consolidated evidence indicates that changing the local lipid composition is a way to alter the membrane fluidity and curvature. For example, breakdown of SM, which has a high affinity for membrane cholesterol (37), results in increased efflux of cholesterol from the membrane and increased membrane fluidity (38, 39), while generation of Cer, which has a cone-shaped structure, can give a spontaneous curvature to the membrane (40) (Fig. 1A).

Fig. 1.

Fig. 1.

MV and exosome biogenesis: the role of SMase and its product Cer. A: On the left, SM (cylinder shaped) stabilizes membrane structure; on the right, when a-SMase translocates from the lysosomal compartment to the outer leaflet of the plasma membrane, SM hydrolysis and Cer accumulation into the inner leaflet of the plasma membrane favor blebbing and MV evagination, thanks to the inverted cone shape of Cer. B: n-SMase resides on the cytosolic leaflet of endosomes/MVBs. As in A, before n-SMase action SM stabilizes membrane structure, while Cer production by n-SMase induces spontaneous negative curvature to the membrane of endosomes, thus promoting the formation of internal vesicles inside MVBs.

SM is hydrolyzed into phosphorylcholine and Cer by the action of SMases, a family of enzymes that possesses acid, neutral, or alkaline optimal pH (41). Neutral SMase (n-SMase) 2, also known as SM phosphodiesterase (SMPD)3, mostly localizes to the Golgi and ER (42), while n-SMase 1, also called SMPD2, is localized to the Golgi and nucleus (43, 44). However, plasma membrane-associated n-SMases have also been characterized (45, 46), and n-SMase 2 has been reported to recycle via the endosomal/recycling compartment (42). Activity of n-SMases is present at the cytosolic leaflet of the membrane (45). By contrast, acid SMase (a-SMase) activity is present in lysosomes and on the outer plasma membrane leaflet following lysosome fusion to the cell membrane (47).

Membrane destabilization driven by SM hydrolysis was initially reported to play a key role in membrane blebbing and release of apoptotic bodies during the late phase of cell damage (48). In 2002, Nurminen et al. (49) visualized the macroscopic consequence of SM breakdown by SMases on lipid membrane using fluorescence and differential interference contrast microscopy. SMase was immobilized onto synthetic microspheres and brought into contact with a giant liposome membrane by a micromanipulator: the contact caused formation of membrane microdomains and subsequent shedding of small vesicles from the membrane into the interior of the giant liposome (49). This elegant study was followed in 2008 by the seminal work by Trajokovic et al. (50), who first described that SM hydrolysis and Cer formation participate in ESCRT-independent biogenesis of intraluminal vesicles inside MVBs, the vesicles that are released as exosomes upon MVB fusion with the plasma membrane. While investigating sorting mechanisms of protein cargo into exosomes in an oligodendrocyte cell line (oli-Neu), Trajkovic et al. (50) observed that total Cer was enriched more than 3-fold in a subpopulation of exosomes containing proteolipid protein (PLP). The role of Cer in the biogenesis and release of PLP-positive exosomes was analyzed by treating Oli-neu cells with GW4869, an inhibitor of n-SMase 1 and n-SMase 2, and two structurally unrelated n-SMase blockers, spiroepoxide and glutathione. Exosome release was markedly reduced after treatment of the cells with all the n-SMase inhibitors. siRNA depletion of n-SMase 2 confirmed the involvement of the enzyme in the formation and secretion of PLP-positive exosomes. On this basis, Trajkovic et al. (50) proposed a lipid-based mechanism for membrane bending and exosome formation. They proposed that biogenesis of PLP-positive exosomes occurs at membrane domains enriched in SLs through formation of Cer microdomains and their coalescence into larger domains, which promote inward vesicle budding (50) (Fig. 1A). The cone-shaped structure of Cer induces spontaneous negative curvature to the membrane of endosomes and favors formation of internal vesicles inside MVBs.

Subsequent in vitro works reported that, while exogenous cell permeable C6 Cer dose-dependently increases the number of exosomes released from multiple myeloma cells (51), pharmacological or genetic block of n-SMase inhibited packaging of the prion protein into exosomes (52) and reduced exosome release among others in HEK cells (53), T cells (54), N2a cells (55), astrocytes (56), microglia (57), macrophages (58), hepatocytes (59), and multiple myeloma cells (51). Also, in vivo, in the brain and serum of 5XFAD mice, a transgenic model of Alzheimer’s disease (AD), the n-SMase inhibitor, GW4869, decreased exosome concentration (60). Importantly, decreased exosome production was reported by the same group in 5XFAD mice crossed with fro/fro mice, which lack n-SMase 2 (61), confirming the enzyme involvement in exosome release. Conversely, elevation of Cer caused by block of Cer conversion to SM by the SM synthase inhibitor, D609, promoted exosome secretion (55, 60).

In agreement with these studies, it has recently been shown that basal exosome production can be influenced by cellular components that control n-SMase activity. For example, in hepatocytes, the chemokine receptor, CXCR2, negatively regulates n-SMase activity and cells lacking CXCR2 expression produce significantly more exosomes than wild-type hepatocytes (62). Interestingly, inhibition of exosome release by CXCR2 is unrelated to ligand binding to the receptor and may occur in endosomal membranes where n-SMase localizes and exosomes are generated (62).

Collectively, these studies led to the idea that Cer is important for exosome release in general. However, exosomes are a heterogeneous population of vesicles and it is important to remember that blocking of n-SMases does not block release of all exosomes or impair exosome biogenesis in all the cells tested (6365), as recently highlighted by Skotland, Sandvig, and Llorente (66).

S1P-dependent cargo sorting in exosomes

Trajkovic et al. (50) revealed a role for Cer in exosome formation by inward budding inside MVBs. However, the mechanism underlying cargo sorting remained unanswered. A recent discovery by Nakamura’s group provided new insights into mechanisms of both Cer-dependent exosome formation and cargo sorting (67). Kajimoto et al. (67) initially showed that exosomal formation inside MVBs depends on S1P, the Cer metabolite generated by sequential activity of CDase and Sph kinase (SK). Specifically, they demonstrated that the S1P type 1 receptor (S1P1), an inhibitory G protein-coupled receptor, is present on MVBs and its sustained activation controls exosomal maturation and cargo sorting in HEK cells. In fact, downregulation of either S1P1 or the S1P synthesizing enzyme, SK-2, decreased CD63, CD81, and flotilin-2 content in exosomes (67). In a subsequent study, the same group further dissected the molecular mechanism necessary for cargo sorting by showing that downstream signaling of S1P1, mediated by the β and γ subunits of the inhibitory G protein (Giβγ), constantly activates the Rho family GTPases, Cdc42 and rac1, on MVBs. Through this mechanism, S1P promotes formation of F-actin networks that are essential for cargo sorting in exosomes. This is indicated by decreased content of CD63 in exosomes released under pharmacological inhibition of both F-actin and S1P formation (68).

Does n-SMase control biogenesis of MVs at the plasma membrane?

Until very recently, the action of n-SMase activity had been explored in the biogenesis of exosomes, but not in a larger EV population, such as MVs (69). The only study that analyzed the impact of n-SMase inhibitors on MV secretion from the plasma membrane was conducted on primary microglia under stimulation with ATP (70), a stimulus particularly effective in promoting release of both ectosomes and exosomes from brain immune cells (57, 71). This work showed no changes in ATP-induced MV shedding under pharmacological block of n-SMase by GW4869 or manumicyn, another n-SMase inhibitor, revealing that n-SMase activity is not required for ATP-induced MV shedding at the cell surface (70).

Recently, Menck et al. (69) investigated the effects of n-SMase on basal MV production from epithelial cells. Using pharmacological and genetic inhibitors of n-SMases, they observed an increase rather than a decrease in ectosome release from the cell surface. Consistently, overexpression of n-SMases (SMPD2 or SMPD3) decreased MV shedding (69). Although further work is necessary to define the mechanisms underlying enhanced MV production under block of n-SMase, this study clearly showed that n-SMase differentially controls the release of exosomes and MVs in epithelial cells. Enhanced basal MV production in cells treated with GW4869, a common tool to block exosome release, may have important consequences: specific proteins may be redirected from exosomes to MVs for secretion when exosome production is blocked. Consistent with this possibility, Menck et al. (69) showed that exosome-enriched proteins, such as CD81 and Wnt, are released in association with MVs under n-SMase inhibition. Importantly, MVs and exosomes derived from many cells share a number of common proteins. For example, both MVs and exosomes released from brain cells in the course of neurodegenerative diseases carry misfolded proteins (Aβ, tau, PrP, and α-synuclein), which form pathological aggregates (72). Although prolonged treatment of AD mice with GW4869 has been shown to reduce exosome levels in brain and serum and to decrease Aβ and tau aggregates (57, 60), how the n-SMase inhibitor impacts release of tau/Aβ in association with MVs has yet to be defined.

Cer-dependent MV biogenesis: role for a-SMase

A specialized type of EV release exists for cells that express the ATP receptor, P2X7, which sheds MVs formed by evaginations of the surface membrane when the receptor is activated. P2X7 receptor-positive cells include monocytes, macrophages, mast cells, dendritic cells, T cells, microglia, and astrocytes. In myeloid or lymphoid cells, the P2X7 receptor functions as an ATP-gated nonselective cation channel and its activation shapes the intensity or duration of innate immune and inflammatory responses (73). The most relevant consequence of P2X7 channel opening is net K+ efflux and assembly of the NLR family pyrin domain containing 3 (NLRP3) inflammasome complexes. These complexes enable activation of caspase-1 protease, which in turn leads to proteolytic cleavage and release of the pro-inflammatory cytokines, interleukin (IL)-1β, and IL-18 (74).

Before the mechanism of NLRP3 inflammasome assembly was discovered (75), the pioneering work by MacKenzie et al. (5) provided evidence that P2X7-induced MV shedding acts as a secretory pathway for IL-1β release. They reported that, in THP-1 monocytes, the P2X7 receptor induces formation of blebs, which are subsequently shed as MVs into the extracellular space within the first few minutes of receptor activation. Importantly, membrane blebs accumulate IL-1β, which is then packaged into MVs and released extracellularly. P2X7-induced membrane blebbing is preceded by loss of plasma membrane asymmetry and PS externalization, a commonly accepted marker for cell apoptosis. However, alterations of membrane asymmetry are reversible after brief ATP stimulation, dissociating bleb formation and MV shedding from apoptotic cell death. Subsequent studies reported that a similar mechanism for IL-1β and IL-18 release occurs in microglial cells (71), dendritic cells (76), and macrophages (77), suggesting a general role for P2X7-dependent MV shedding in rapid secretion of cytokines lacking a secretory sequence. In macrophages, MVs shed in response to P2X7 receptor activation also bear active tissue factor and have high pro-coagulant activity (78).

However, an unsolved question was how signaling by the P2X7 receptor led to alterations of the biophysical properties of the plasma membrane, which, together with actin-cytoskeleton reorganization, is a prerequisite for membrane blebbing and vesiculation. In 2009, Bianco et al. (70) identified a-SMase as the key enzyme responsible for P2X7-dependent MV biogenesis at the surface of glial cells (microglia and astrocytes). They showed that following P2X7 receptor activation, a src-protein tyrosine kinase interacts with the C terminus of the receptor (79) and promptly phosphorylates P38 MAP kinase. P38 phosphorylation, in turn, induces translocation of a-SMase from lysosomes to the plasma membrane outer leaflet, where it catalyzes Cer formation from SM (70). As mentioned above, SM to Cer conversion perturbs membrane curvature and fluidity, favoring budding of MVs. Specifically, redistribution of inverted cone-shaped Cer molecules, synthesized extracellularly, into the inner membrane leaflet probably drives membrane evagination (80) (Fig. 1B).

ATP-induced translocation of a-SMase to the plasma membrane and SM hydrolysis were recently confirmed by an independent group in macrophages (81), the peripheral counterpart of brain resident microglia. Immunostaining with an anti-a-SMase antibody revealed the presence of the enzyme on the surface of ATP-stimulated macrophages, while labeling was limited primarily to the cytoplasm in untreated cells. ATP-dependent SM hydrolysis was indicated by the release of the [14C]-labeled phosphorylcholine head group into the supernatant medium. Importantly, inhibition of a-SMase expression or activity not only attenuated ATP-induced SM hydrolysis but also attenuated release of tissue factor-positive MVs, which exhibit pro-coagulant activity (81).

Of note, a-SMase-dependent MV formation likely occurs at SM-enriched domains of the plasma membrane, where the P2X7 receptor localizes (82). As a consequence, cytoskeleton/membrane proteins interacting with the long cytoplasmic C terminus of the P2X7 receptor can be recruited and sorted into MVs. Consistent with this sorting mechanism, MVs released from ATP-stimulated microglia contain several proteins previously shown to bind to the P2X7 receptor C terminus, which are not present in MVs constitutively released by microglia (83).

Recent works suggest that a-SMase can be recruited to the plasma membrane to rapidly enhance EV release upon stimulation of other surface receptors besides P2X7. Initially, Truman et al. (84) found that a-SMase is activated in response to engagement of surface Fcγ receptors by oxidized LDL-containing immune complexes and plays a role in the release of IL-1β in association with exosomes in the human macrophage cell line, U937. This evidence was documented by reduced exosome and IL-1β secretion from cells in which a-SMase was pharmacologically (desipramine) or genetically inhibited (a-SMase siRNA) (84). More recently, Marrone et al. (85) reported that a brief stimulation of the transient potential receptor vanilloid type 1 (TRPV1), also known as vanilloid receptor 1, promotes shedding of MVs to the same extent as P2X7 receptor activation in primary murine microglia. TRPV1 receptor is a nonselective cationic channel highly expressed in brain immune cells, which is exogenously activated by capsaicin (86) and endogenously activated by inflammatory molecules, such as the signaling lipid, lysophosphatidic acid (87). Importantly, p38 MAPK, which is essential for P2X7-dependent a-SMase translocation onto the plasma membrane, is activated downstream of TRPV1 receptors (88) and its inhibition blocks capsaicin-induced MV shedding from microglia in vitro and in cortical brain slices (85), consistent with a role of a-SMase in TRPV1-dependent MV production.

Besides the consolidated role of a-SMase in MV shedding from myeloid cells, the enzyme has also been implicated in the release of EVs from red blood cells (RBCs), at least under pathological conditions. In 2014, Awojoodu et al. (89) reported that plasma membrane stresses and alterations in sickle cell disease, an inherited RBC disorder, enhance the activation of a-SMase and result in increased EV production from erythrocytes. Although erythrocytes do not possess SMase activity of their own (90) and SMases have been reported at low levels in erythrocytes (91, 92), RBCs can be exposed to a-SMase secreted by myeloid cells or by vascular endothelium (93). RBC-derived EVs display pro-inflammatory activities and promote monocyte adhesion to endothelial cells (89). Importantly, treatment with amitriptyline, an indirect inhibitor of a-SMase, reduces EV generation in in vitro and in vivo models of sickle cell disease, thereby representing a possible treatment to mitigate inflammatory processes (93). A very recent study corroborated the role of a-SMase in the formation of EVs from RBCs under stressful conditions, i.e., during packed storage (94). Standard blood banking practice allows storage of RBCs up to 42 days of age. However, as stored human packed RBCs age, they release a heterogeneous population of EVs, which cause lung inflammation and worse clinical outcomes in patients with anemia or hemorrhage. Hoehn et al. (94) showed that a-SMase activity is increased in aged compared with fresh RBC-derived EVs, whereas a-SMase activity is decreased on stored erythrocytes as compared with fresh. This suggests that a-SMase changes location and is concentrated in EVs over the course of RBC storage. Interestingly, the authors demonstrated that inhibition of a-SMase by amitriptyline reduces EV production during storage, supporting a primary role of the enzyme in EV biogenesis, and leads to a change in the composition of EVs that ultimately prevents lung inflammation. In fact, less lung inflammation was observed in the recipient mice transfused with equal numbers of EVs produced from RBCs treated with amitriptyline, revealing that a change in a-SMase content and/or Cer concentration in EVs likely preserves the integrity of endothelial cells in the lung and prevents endothelial cell apoptosis (94).

Another recent study implicated a-SMase in the release of EVs evoked by cigarette smoke (CS) from endothelial cells (95). Following exposure to CS, endothelial cells contract their bodies and release EVs from the tips of retracting filopodia-like structures without appreciable cell death. Given that Cer production is activated by CS in lung endothelial cells and a-SMase is rapidly activated by stress in the cells, Serban et al. (95) interrogated the role of a-SMase in the mechanism of CS-induced EV release. They found that treatment with imipramine, a functional inhibitor of a-SMase, markedly decreased CS-induced EV production and that a-SMase-null endothelial cells produced fewer EVs compared with wild-type cells. The role of a-SMase in EV production was corroborated by the observation that mice lacking a-SMase (Smpd1−/−) are characterized by reduced levels of EVs in plasma following CS exposure, while mice overexpressing a-SMase in endothelial cells display increased levels of circulating EVs. Besides pointing to a key role of a-SMase activation in CS-induced EV release, this work showed that EVs derived from endothelial cells are Cer rich, suggesting that EVs may act as a vehicle for the transport of Cer, a highly lipophilic molecule, in aqueous environments, such as plasma or bronchoalveolar lavage fluid. Also, in human macrophages, CS may promote MV shedding through an a-SMase-dependent pathway. This is suggested by the work of Li et al. (96) who found that production of MVs from human macrophages relies on a series of regulated steps that include activation of p38 MAPK, the enzyme that drives a-SMase translocation onto the plasma membrane (70).

While the role of a-SMase in enhancing MV shedding in response to danger signals (ATP, TRPV1 agonists) or stressful membrane conditions (CS, sickle cell disease, packed storage) is established (Table 1), the enzyme may not play a role in the constitutive shedding that occurs without any stimulation. Indeed, a very recent study showed that long exposure of microglial cells or monocytes to pro-inflammatory or pro-regenerative cytokines (24 h treatment) enhances basal production of MVs, but pharmacological inhibition of a-SMase by imipramine or by the structurally unrelated indirect inhibitor, siramesine, fails to reduce cytokine-induced MV release. Intriguingly, both constitutive release of MVs and cytokine-induced MV release were efficiently blocked by a sub-toxic concentration of actinomycin D, a strong inhibitor of transcription. While ruling out a role for a-SMase in constitutive release of MVs, this study suggests that cytokines may strengthen constitutive MV production through modulation of the transcriptional activity in myeloid cells (97).

TABLE 1.

a-SMase involvement in EV release

Reference
(70) (81) (84) (85) (93, 94) (95) (96)
Cell type Glial cells Macrophages Macrophage cell line U937 Primary microglia RBCs Endothelial cells Macrophages
Species Rat, mouse Human Human Mouse Human, mouse Mouse Human
EV subpopulation MVs MVs Exosomes MVs EVs EVs MVs
Stimulus ATP on P2X7 receptor ATP on P2X7 receptor oxLDL-IC on Fcγ receptor Capsaicin on TRPV1 receptor Stressful membrane conditions (sickle cell disease, packed storage) CS CS
Evidence Pharmacological and genetic Pharmacological and genetic Pharmacological and genetic Pharmacological Pharmacological Pharmacological and genetic Pharmacological

oxLDL-IC, oxidized LDL-containing immune complex.

Galactosyl-Sph-dependent EV biogenesis

A recent study revealed a role for the GSL, galactosyl-Sph, and its metabolic enzyme, galactosyl-CDase, in the control of plasma membrane structure and EV shedding. D’Auria et al. (98) highlighted the involvement of galactosyl-Sph in alteration of membrane organization and microvesiculation from oligodendrocytes, the glial cell type that deposits myelin along neuronal axons in the central nervous system.

Galactosyl-Sph, also known as psychosine, is an inverted cone GSL composed of a hydrophobic sphingoid base and a hydrophilic polar head group that accumulates in brain membranes of Krabbe’s disease, a genetic sphingolipidosis caused by mutations in the psychosine-degrading enzyme, galactosyl-CDase. Different from SM, psychosine is not ubiquitously present, but generated quite specifically in oligodendrocytes, where it is considered the main SL responsible for demyelination and oligodendrocyte loss in Krabbe’s disease. Indeed, psychosine accumulates in lipid rafts and disrupts their structures, altering signaling pathways important for oligodendrocyte differentiation and survival (99). However, until recently, no clear insights have been provided into how psychosine causes demyelination. D’Auria et al. (98) studied psychosine-mediated membrane disorganization in RBCs and showed that psychosine, due to the quite large size of its polar head group, affects the organization of the SM-enriched domain and elicits outward membrane blebbing and EV shedding. It should be noted, however, that, in this study, specificity of psychosine action has not been assessed by the use of an equal concentration of similarly structured lipids, e.g., octyl glucoside or hexadecylglycerol. Findings obtained in RBCs were confirmed in oligodendrocytes established from Twitcher mice, a model of Krabbe’s disease, which were more prone to produce MVs than wild-type cells upon exposure to the psychosine. Vesiculation was accompanied by lateral mobility of SM and cholesterol toward areas in the plasma membrane with high focal rigidity, which may introduce focal weak points in the myelin sheath contributing to myelin disruption. Importantly, this model of MV biogenesis implies that MVs do not originate from membrane blebs, which progressively bud from the plasma membrane and are then pinched off from the cell surface, a process that requires membrane bending and increased membrane fluidity. However, no mechanistic explanation of MV formation is provided by the author.

Independent of the underlying mechanisms, psychosine-dependent MV production may spread disease from sick to healthy cells. Indeed, while clearing some of the accumulated SL material via EV secretion, brain cells may shuttle pathogenic SLs [e.g., psychosine or proapoptotic Cer (56)] to cells that are not intrinsically affected, contributing to establishing non-cell-autonomous defects (100).

SL CONTENT OF EVs

Comparative analysis of SL composition in exosomes/MVs and in parental cells

To date, high resolution lipidomic analyses of EVs have been reported for a limited number of cell lines (101), thus limiting current knowledge of EV lipid composition and complexity. However, it is well-established that EVs contain lipids, such as aminophospholipids and Cer, which, along with GSLs and SM, are the most enriched lipids in EVs of different cellular origin (Table 2) (66). The SL content of less represented SL species is still very uncertain.

TABLE 2.

SL content in exosomes and enrichment from the originating cells

Lipids PC-3 Cells (102) Oli-neu Cells (50) B-Cells (104) Mast Cells (4) Dendritic Cells (4) Reticulocytes (105) Mesenchymal Stem Cells (101) U87 Cells (101) Huh7 Cells (101) FEMX1 Cells (153) DU145 Cells (154) VCaP (154) LNCaP (154)
% F % F % F % F % F % F % F % F % F % F % F % F % F
SM 16.3 2.4 8.2 1.5 23.0 2.3 12 2.8 20 2.2 8.4 1.31 5 1 20 2.35 4 0.8 4
GSLs 0.78 3.8 2.0 1.8 3.1 3
Cer 0.32 1.3 3.3 0.04 1 0.2 1 0.04 0.25

Data are based on the EV network database, Vesiclepedia. Only the reports that allowed us to calculate the changes of SLs in EVs compared with the parental cells are reported. %, percent of total lipid quantified; F, factor of enrichment from cells to exosomes or MVs.

There are major differences in the lipid composition of exosomes/MVs compared with whole cell lipid extract of parental cells, demonstrating the existence of specific mechanisms of lipid sorting into EVs. Interestingly, SM, GSLs, and Cer are among EV-enriched lipids compared with most donor cells (Table 2) (4, 50, 66, 102105).

However, Llorente et al. (102) demonstrated that exosomes released from reticulocytes and rat basophil leukemia cells exhibit a lower phospholipid-to-protein ratio than parental cells. Specifically, they found that most lipid classes were more abundant in exosomes, and that the relative amounts of specific lipids, such as GSLs, cholesterol, SM, and PS, were highly enriched in exosomes (102). Other studies confirmed enrichment from 1.3- to 2.8-fold for SM (4, 50, 66, 102105) and from 2.0- to 3.8-fold for GSLs (50, 102, 103) in exosomes depending on the types of donor cells. Exosomes derived from mouse Oli-neu cells show less enrichment in SM and GSLs and a much higher enrichment in Cer, which is crucial for exosome biogenesis inside MVBs (50). Specifically, SM is 1.5 times enriched in exosomes isolated from Oli-neu cells, compared with parental cells, while it is 2.4 times enriched in PC-3 cell-derived exosomes (102), 2.3 times enriched in human B-cell-derived exosomes (104), 2.8 times enriched in mast cell RBL-2H3-derived exosomes (4), and 2.2 times enriched in exosomes released from dendritic cells with respect to donor cells (105). GSLs are 2.0 times enriched in exosomes of Oli-neu cells, compared with their parental cells (50), while the enrichment in GSLs is of about 3.8 times in exosomes from PC-3 cells (102) and even 18 times in those produced from murine neuroblastoma Neuro2a cells (106).

Different methods have been used to analyze the SL content of EVs, including TLC, gas LC, and MS. Wubbolts et al. (104) were the first group to use MS (in addition to TLC) to study the SL content of exosomes released from human B-cells. By this approach, they described a large SL enrichment in exosomes and also highlighted similarities between the lipid classes abundant in exosomes and in detergent-resistant membranes, the membrane regions from which EVs are generated. The use of different methodologies could account for the slightly different SL composition of EVs produced by different cell types. However, the lipid content may actually vary depending on source cell type and even among exosomes and MVs released by the same donor cells. A few studies support this possibility: i) The comprehensive analysis of lipid composition of exosomes, MVs, and source cells in three different cell types by Haraszti et al. (101) showing specific enrichment in SMs in MVs derived from Huh7 hepatocellular carcinoma cells or mesenchymal stem cells with respect to MVs released by U87 glioblastoma cells (Table 3). Importantly this study also revealed that the MVs and exosomes differ in their types of lipid contents. Enrichment in glycolipids and free fatty acids characterized exosomes, whereas enrichment in Cers and SMs characterized MVs derived from Huh7 hepatocellular carcinoma cells as well as mesenchymal stem cells (101). ii) The work by Dang et al. (107) reporting significant differences in the lipid composition of exosomes secreted across the apical compared with the basolateral plasma membrane of epithelial cells. In particular, this study showed that SM is more represented in exosomes secreted across apical compared with the basolateral plasma membrane (107). iii) SL profiling of human oligodendroglioma (HOG) cell-derived exosomes, revealing differences in the SL composition, in particular of C16-, C24-, and C24:1-Cer species; C16-, C24-, and C24:1-dihydroCer species; and C16-, C24-, and C24:1-SM species, between exosomes released constitutively or under stimulation with inflammatory cytokines (108). iv) The work by Carayon et al. (109) showing that the exosomal content of Cer versus SM is significantly increased during reticulocyte maturation. v) The study by Baig et al. (110) demonstrating that GM3 levels are significantly downregulated in MVs derived from syncytiotrophoblasts of preeclampsia patients.

TABLE 3.

SL content in MVs and enrichment from the originating cells

Lipids Bone Marrow-Derived Mesenchymal Stem Cells (101) U87 Cells (101) Huh7 Cells (101) Platelets (155)
% F % F % F % F
SM 10 2 15,2 0.97 17 2 17 3.4
GSLs
Cer 0.48 2 0.68 2.4 0.24 1.5

Data are based on the EV network database, Vesiclepedia. Only the reports that allowed us to calculate the changes of SLs in EVs compared with the parental cells are reported. %, percent of total lipid quantified; F, factor of enrichment from cells to exosomes or MVs.

SL species in EVs as potential biomarkers

EVs are attracting increasing interest as a source of noninvasive biomarkers for several diseases, including cancer and multiple sclerosis, because these vesicles contain pathogenic molecules of donor cells that can be analyzed in biofluids (111113). Interestingly, SLs have recently been identified among EV components that are relevant for disease diagnosis.

In depth lipidomic analysis of exosomes secreted by a colorectal cancer cell line first revealed selective changes in SL abundance in EVs compared with parent colon cancer cells (114) and suggested that specific exosome SL(s), i.e., Cer and SM, could serve as effective diagnostic or prognostic biomarkers for colon cancer. The diagnostic potential of exosome SL(s) was then demonstrated by Skotland et al. (115), who found that levels of lactosylceramide (d18:1/16:0) in urinary exosomes are different between prostate cancer patients and healthy controls. Importantly, measurements of lactosylceramide in combination with other exosomal lipids were shown to have the capacity to distinguish cancer patients from healthy subjects with high sensitivity (93%) and specificity (100%) (115).

Another recent work revealed that a distinct SL (C16:0 sulfatide), a sulfated galactosylceramide with long-chain fatty acid, in blood EVs represents a unique indicator of multiple sclerosis, a chronic neuroinflammatory disease (116). Specifically, using ultra-high-performance LC tandem MS, Moyano et al. (116) detected a significant increase in C16:0 sulfatide in EVs isolated from multiple sclerosis patients when compared with healthy subjects.

SL-MEDIATED EV ACTION ON RECIPIENT CELLS

Several lines of evidence point to the involvement of exosome GSLs in the pathogenic aggregation of amyloidogenic peptide Aβ (106) that typically forms extracellular aggregates in the brain of subjects affected by AD, the most common neurodegenerative disease. GSLs present on the exosome surface of Neuro2a cells were shown to form clusters and bind to Aβ peptide. Aβ-GSL complexes, in turn, were shown to act as templates for further Aβ aggregation and fibril formation (106). In agreement with this study, Yanagisawa et al. (117) found that the monosialoganglioside, GM1, associates with Aβ in the brain of AD patients. On these bases, Yuyama et al. (106) proposed that there may be multiple GSLs on exosome membranes that organize themselves into specific areas that bind to Aβ and induce its assembly. Moreover, cholesterol and SM, which were highly enriched in exosomes compared with their parent cells, were also shown to promote Aβ assembly via the lateral packing of gangliosides on membranes (106, 119, 120).

Emerging evidence indicates that the SL cargo of EVs may be responsible for detrimental EV action. In 2012, an interesting study by Bieberich’s group revealed the involvement of Cer-enriched exosomes in Aβ-dependent apoptosis, which critically contributes to AD (56). The authors of this study showed that Cer levels are regulated by n-SMase 2 in astrocytes and are critical for astrocyte apoptosis caused by Aβ, which is not observed in n-SMase 2-deficient cells. Astrocyte apoptosis was accompanied by release of Cer-enriched exosomes, which propagated a death signal into recipient astrocytes not exposed to Aβ protein (56).

A subsequent study confirmed that Cer-enriched exosomes mediate cytotoxic effects in recipient cells. Specifically, exosomes released from HOG cells treated with a combination of inflammatory cytokines were found to be Cer-enriched and to induce cell death in fresh (naïve) HOG cells (108). Interestingly, Cer levels did not increase much in donor HOG cells under cytokine treatment, suggesting that Cer is immediately exported out of the cells by exosomes after generation.

Another recent work highlighted the involvement of the Cer metabolite, S1P, in exosome-mediated neoplastic transformation. Exosome-like particles produced by the intestinal mucosa in response to enterobacteria were shown to contain S1P, which favors the recruitment and proliferation of inflammatory Th17 T cells in the intestine and promotes tumor growth in animal models of colorectal cancer (121).

Finally, Wang et al. (122) identified S1P as the primary mediator of endothelial cell exosome actions on hepatic stellate cells, responsible for the pathological migration and aberrant phenotype of hepatic stellate cells during liver fibrosis. Mechanistically, this study revealed that exosome-induced hepatic stellate cell migration is dependent on exosome adhesion to target cells, which facilitates exosome uptake through dynamin-dependent endocytosis (122).

Involvement of SL enzymatic machinery or SL receptors in the biological activity of EVs

Accumulating evidence indicates that EVs can influence SL metabolism in receiving cells by providing them with key SL metabolic enzymes, among other enzymes of lipid metabolism borne by EVs (123).

The presence of the S1P-generating enzymes, neutral CDase (n-CDase) and SK-1, was first described in EVs shed by hepatocarcinoma and carcinoma cultured cells and was suggested to contribute to the proangiogenic activity of EVs (124). Later on, SK-1 was detected within exosomes derived from endothelial cells and, together with its product, S1P, identified as the active agent responsible for pathological migration on hepatic stellate cells, as mentioned above (122). In addition, Zhu et al. (125) found that active n-CDase is also released in association with exosomes by β-pancreatic INS-1 cells exposed to a low concentration of inflammatory cytokines. Exosome-packaged n-CDase inhibited INS-1β-cell destruction induced by a high concentration of inflammatory cytokines and the protective action was mediated by S1P generation and activation of S1P type 2 receptor (S1P2). These findings revealed a novel n-CDase-S1P2-dependent mechanism by which exosomes produced in response to a low level of inflammatory cytokines protect pancreatic β-cells from apoptosis (125).

In agreement with this work, hepatocyte-derived exosomes were recently shown to be incorporated into target hepatocytes and deliver both n-CDase and SK-2 to the cells, increasing S1P levels. By this mechanism, exosomes carrying S1P metabolic enzymes induce dose-dependent proliferation of hepatocytes in vitro and contribute to liver repair and regeneration after an ischemia/reperfusion injury, a major clinical problem that affects liver transplantation and other hepatic surgeries (59). Accordingly, exosomes produced by mesenchymal stem cells differentiated from human pluripotent stem cells were recently shown to exert protective and proliferative effects on primary hepatocytes and their beneficial action was also ascribed to activation of the synthetic machinery for S1P production in hepatocytes upon exosome internalization (126). Indeed, either the SK inhibitor, SKI-II, or the S1P1 antagonist, VPC23019, completely abolished the proliferative effect of exosomes on primary hepatocytes.

Not only the transfer of SL(s) and/or their enzymatic machinery but also the transfer of SL receptors may participate in the biological action of EVs on target cells. This hypothesis has recently been proposed by Pyne et al. (127), who found that S1P2 is released from breast cancer cells through exosomes, and it is cleaved into a smaller constitutively active form lacking the N terminus (red) into recipient fibroblasts, where the receptor stimulates the ERK-1/2 pathway and promotes proliferation. However, to the best of our knowledge, experimental data supporting the role of exosomal S1P2 cargo in cross-talk between cancer cells and fibroblasts are still unpublished. A previous work revealed a specific enrichment in S1P types 3 and 4 receptors (S1P3 and S1P4) in MVs and exosomes derived from a mammary luminal epithelial cell line overexpressing HER2/ERBB2 oncogene. Their presence may indeed be relevant for the biology of HER2+ tumors, by controlling tumor invasion and cell migration. However, their functional significance has yet to be evaluated (128).

EV-dependent activation of SL metabolism in target cells

In the last years, evidence has been provided that EVs can stimulate SL metabolism in receiving cells not only via the transfer of SL enzymatic machinery but also through activation of SL metabolic enzymes in recipient cells. Data from our laboratory showed that lipid species housed on the membrane of microglia-derived MVs, through activation of contact-mediated signaling pathways, act on neuronal a-SMase promoting both Cer and Sph production. The latter SL increased spontaneous release from excitatory terminals in cultured hippocampal neurons by enhancing glutamate release probability (129, 130) (Fig. 2B). The stimulation of SL metabolism in neurons required the action of neuronal a-SMase and was not due to the enzyme transfer from microglia in association with MVs. In fact, hippocampal cultures established from a-SMase KO mice exposed to MVs did not show any significant alteration in miniature excitatory postsynaptic current (mEPSC) frequency relative to untreated transgenic cultures, whereas wild-type cultures treated with MVs displayed an increase in mEPSC frequency compared with control neurons (Fig. 2B). Neuronal Sph was accountable for MV-mediated potentiation of excitatory transmission, as the blockade of Sph production through the CDase inhibitor, N-oleoylethanolamine (NOE), strongly prevented MV effects on mEPSCs, whereas the blockade of S1P synthesis from Sph by the Sph kinase inhibitor, SKI-1, was ineffective (Fig. 2A, B). However, in a subsequent study, we found that the SL cascade induced by microglial MVs in neurons does not stop at Sph, but proceeds to S1P, which contributes to enhance glutamate transmission by targeting synapsin I, the phosphoprotein controlling synaptic vesicle availability for exocytosis (131). The key role of S1P and its receptor, S1P3, in controlling synapsin I activity was defined by the use of pharmacological inhibitors of the SL cascade (i.e, the a-SMase inhibitor, imipramine, the CDase inhibitor, NOE, and the SK inhibitor, SKI-1) as well as S1P receptor antagonists (the pan receptor antagonist, S-FTY720-vinylphosphonate, the S1P3 antagonist, CAY1044, and the S1P1 antagonist, W146) (Fig. 2A, B). These findings were corroborated by a recent study reporting that the inhibition of a-CDase by the potent inhibitor, ARN14988, completely prevents the increase of mEPSC frequency induced by microglial-derived MVs in cortical slices (85). In this case, MV release from microglia was triggered by capsaicin acting on TRPV1 receptors, a receptor coupled to P38-MAPK, the enzyme that drives a-SMase (see also above). Given that TRPV1 receptors are antagonized by the broadly used a-CDase inhibitor, NOE, the authors employed the CDase inhibitor, ARN14988, instead of NOE in order to block a-CDase activity. ARN14988 prevented the enhancement of mEPSC frequency caused by MVs without interfering with capsaicin-dependent MV release or MV function on mEPSCs (85).

Fig. 2.

Fig. 2.

MV-dependent activation of SL metabolism in neurons. A: The SL cascade on the plasma membrane and the targets of genetic/pharmacological inhibitors. B: The cartoons elucidate the mode of action of MV released by microglial cells on excitatory glutamatergic synapses and the effects of genetic/pharmacological inhibitors. Microglial MVs interact with the plasma membrane of neurons through surface contact facilitated by receptor-ligand interaction between PS residues (typically exposed on the MV membrane) and PS neuronal receptor (in blue). Subsequently, unidentified lipid(s) on the surface of MVs activate a-SMase (translocated to the outer leaflet of plasma membrane from lysosomes), thus stimulating SL metabolism in neurons. Sph and S1P have been identified as the SLs responsible for the MV effects on the excitatory presynaptic terminal (see also the “a-SMase KO animals,” “CDase inhibitor,” and “SK inhibitor/S1P3 inhibitor/S1P3 siRNA” panels). Sph acts on the synaptic vesicle-associated SNARE protein, synaptobrevin, activating it and thereby promoting the assembly of the SNARE complex (which is necessary for synaptic vesicle fusion at the presynaptic terminal and, therefore, neurotransmitter release). This event increases the probability of neurotransmitter release, empowering excitatory synaptic transmission in neurons targeted by MVs. On the other hand, S1P, a SL able to move outside the membranes, travels extracellularly to engage its receptor on the presynaptic terminal (likely the S1P3 receptor). S1P3 receptor activation translates into downstream signaling in the target neuron, inducing ERK activation and synapsin I phosphorylation. Synapsin I is a presynaptic protein responsible for anchoring synaptic vesicles of the reserve pool to the actin cytoskeleton, therefore preventing their exocytosis. Dephosphorylated synapsin I binds synaptic vesicles and the actin cytoskeleton, tying the two of them together. When phosphorylated, synapsin I breaks the link between vesicles and actin, leaving synaptic vesicles free to migrate to the presynaptic terminal, ready for exocytosis upon stimulation. This increases the number of synaptic vesicles that undergo exocytosis when neurotransmitter release is triggered. The actions of genetic/pharmacological inhibitors are described in the a-SMase KO animals, CDase inhibitor, and SK inhibitor/S1P3 inhibitor/S1P3 siRNA panels.

Importantly, a recent study showed that EVs have the epigenetic machinery to target SL metabolism in recipient cells. Schatz et al. (132) observed that EVs released by a cosmopolitan alga (Emiliania huxleyi) during infection with its specific virus (EhV, Phycodnaviridae) have a unique lipid composition that differs from that of viruses and infected host cells, highly enriched in triacylglycerols. In addition, RNA isolation and analysis revealed that EV cargo is composed of specific microRNAs that are predicted to target SL metabolism (132134). Together with our findings, this evidence suggests that SL metabolism may be a highly conserved pathway activated in recipient cells by EVs either through their lipid or miRNA cargoes.

ENDOCANNABINOID-MEDIATED EV ACTION ON RECIPIENT CELLS

Another intriguing aspect of lipid involvement in EV biological activity concerns the role of endocannabinoids (eCBs).

eCBs are lipid mediators that regulate a large number of physiological functions in the central and peripheral nervous system (135137). The two major eCBs, 2-arachidonoylglycerol and N-arachidonoylethanolamine (AEA), also called anandamide, primarily act as retrograde messengers: typically produced by neurons, eCBs are synthesized on demand in response to high synaptic activity and rapidly released extracellularly. In the extracellular space, eCBs move toward the presynaptic membrane and activate eCB type 1 (CB1) receptors to inhibit neurotransmitter release (135, 138140). However, eCBs are also produced by astrocytes and microglial cells (141143), and control glia-neuron interaction (137, 144) with microglia being the major provider of eCBs under inflammatory conditions (145).

A long-standing question in the field of eCB research has been how these highly hydrophobic lipids are transported through the aqueous extracellular environment to engage their targets (146, 147). Studies conducted in our laboratory uncovered the role of MVs in the transport of eCB species outside brain cells (148). We found that EVs released from primary microglial cells are enriched in the eCB, AEA (while 2-arachidonoylglycerol was under the limit of detection), which likely localizes on the vesicle surface. This was indicated by the affinity of a biotinylated analog of AEA (biotinyl-AEA) (149) to the MV external membrane. We also found that vesicular eCBs are biologically active: they bind presynaptic CB1 receptors, activate CB1 receptor downstream signaling, and induce an impairment of inhibitory synaptic transmission (Fig. 3). In fact, primary hippocampal neurons exposed to MVs and recorded through patch-clamp electrophysiological technique displayed reduced miniature inhibitory postsynaptic current frequency compared with control untreated cells, revealing that MVs reproduce the action of the CB1 receptor agonist, WIN 55,212-2 (150, 151), on inhibitory hippocampal synapses (152). Furthermore, the involvement of vesicular AEA in this phenomenon was inferred from the ability of the CB1 receptor antagonist, SR141716A (150152), to block the depression of miniature inhibitory postsynaptic current frequency evoked by MVs. Finally, MV treatment also determined an increase in phosphorylated ERK levels in neurons, an event downstream CB1 receptor activation, which was again abolished in the presence of SR141716A.

Fig. 3.

Fig. 3.

Effects on inhibitory/excitatory synapses of MV-associated bioactive lipids. Center: Microglial cells release EVs (MVs and exosomes). MV interaction with the neuronal surface is mediated by PS residues, typically exposed on the MV membrane, which bind their specific neuronal receptor (in blue). Left: On inhibitory synapses, vesicular eCB(s) (likely AEA exposed on the surface of MVs) interact with presynaptic CB1 receptors. CB1 receptor activation induced by vesicular eCBs causes an increase in phosphorylated ERK levels and a downregulation of inhibitory synaptic transmission. Right: As already described in Fig. 2, on excitatory synapses, microglial MVs activate SL metabolism. Sph and S1P mediate MV action, inducing an increase in excitatory transmission.

CONCLUSIONS AND FUTURE DIRECTIONS

Progress has been made in the last 10 years in elucidating how EVs are formed and a fundamental role for Cer during formation of membrane negative curvature (away from the cytoplasm) has been established, which is necessary for both exosome and MV biogenesis. In the examples of EV biogenesis discussed above n- and a-SMase have been extensively studied and shown to mediate release of exosomes and MVs, respectively. In particular, a clear role for a-SMase has been demonstrated in MV release evoked by stimulation of various surface receptors or occurring under stressful conditions, while a more general role for n-SMase in exosome biogenesis has been shown, despite the fact that the enzyme does not mediate exosome release in all the systems tested. However, a more complex scenario is emerging with the n-SMase inhibitor, GW4869 (a common tool to block exosome release), enhancing rather than blocking production of MVs from the cell surface of epithelial cells.

Consolidated evidence indicates that SLs and their enzymatic machinery are enriched in EVs, despite the SL content of less represented SL species is still uncertain.

Besides attracting interest as potential disease biomarkers in cancer and inflammatory diseases, vesicular SLs and their metabolic enzymes have been shown to contribute to EV action by influencing SL levels in recipient cells. Interestingly EVs may also influence SL activity in receiving cells via transfer of microRNAs targeting SL metabolism, as indicated by transcriptomic analysis of EVs produced by prokaryotic cells.

We expect that future characterization of EVs in diverse cell physiology will have a wide impact in understanding biogenesis and signaling functions of these highly conserved mediators of intercellular communications in mammals.

Footnotes

Abbreviations:

AD
Alzheimer’s disease
AEA
N-arachidonoylethanolamine (anandamide)
a-SMase
acid SMase
CB1
endocannabinoid type 1 receptor
CDase
ceramidase
Cer
ceramide
CS
cigarette smoke
eCB
endocannabinoid
ER
endoplasmic reticulum
ESCRT
endosomal sorting complex required for transport
EV
extracellular vesicle
GSL
glycosphingolipid
HOG
human oligodendroglioma
IL
interleukin
mEPSC
miniature excitatory postsynaptic current
MV
microvesicle
MVB
multivesicular body
n-CDase
neutral ceramidase
n-SMase
neutral SMase
NOE
N-oleoylethanolamine
PC
phosphatidylcholine
PLP
proteolipid protein
PS
phosphatidylserine
RBC
red blood cell
SK
sphingosine kinase
SL
sphingolipid
SMPD
SM phosphodiesterase
S1P
sphingosine-1-phosphate
S1P1–4
sphingosine-1-phosphate types 1–4 receptor
Sph
sphingosine
TRPV1
transient potential receptor vanilloid type 1

This work was supported by Fondazione Italiana Sclerosi Multipla Grants 2016/R/30 (C.V.) and 2016/B/2 (M.G.), and Grant Multiple Sclerosis Innovation (GMSI) 2015 (C.V.). M.G. was also financed or co-financed with the “5 per Mille” public funding.

REFERENCES

  • 1.Deatherage B. L., and Cookson B. T.. 2012. Membrane vesicle release in bacteria, eukaryotes, and archaea: a conserved yet underappreciated aspect of microbial life. Infect. Immun. 80: 1948–1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Dauros Singorenko P., Chang V., Whitcombe A., Simonov D., Hong J., Phillips A., Swift S., and Blenkiron C.. 2017. Isolation of membrane vesicles from prokaryotes: a technical and biological comparison reveals heterogeneity. J. Extracell. Vesicles. 6: 1324731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Fitzner D., Schnaars M., van Rossum D., Krishnamoorthy G., Dibaj P., Bakhti M., Regen T., Hanisch U. K., and Simons M.. 2011. Selective transfer of exosomes from oligodendrocytes to microglia by macropinocytosis. J. Cell Sci. 124: 447–458. [DOI] [PubMed] [Google Scholar]
  • 4.Laulagnier K., Motta C., Hamdi S., Roy S., Fauvelle F., Pageaux J. F., Kobayashi T., Salles J. P., Perret B., Bonnerot C., et al. 2004. Mast cell- and dendritic cell-derived exosomes display a specific lipid composition and an unusual membrane organization. Biochem. J. 380: 161–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.MacKenzie A., Wilson H. L., Kiss-Toth E., Dower S. K., North R. A., and Surprenant A.. 2001. Rapid secretion of interleukin-1beta by microvesicle shedding. Immunity. 15: 825–835. [DOI] [PubMed] [Google Scholar]
  • 6.Janowska-Wieczorek A., Majka M., Kijowski J., Baj-Krzyworzeka M., Reca R., Turner A. R., Ratajczak J., Emerson S. G., Kowalska M. A., and Ratajczak M. Z.. 2001. Platelet-derived microparticles bind to hematopoietic stem/progenitor cells and enhance their engraftment. Blood. 98: 3143–3149. [DOI] [PubMed] [Google Scholar]
  • 7.Ratajczak J., Wysoczynski M., Hayek F., Janowska-Wieczorek A., and Ratajczak M. Z.. 2006. Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication. Leukemia. 20: 1487–1495. [DOI] [PubMed] [Google Scholar]
  • 8.Valadi H., Ekstrom K., Bossios A., Sjostrand M., Lee J. J., and Lotvall J. O.. 2007. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9: 654–659. [DOI] [PubMed] [Google Scholar]
  • 9.Skog J., Wurdinger T., van Rijn S., Meijer D. H., Gainche L., Sena-Esteves M., Curry W. T. Jr., Carter B. S., Krichevsky A. M., and Breakefield X. O.. 2008. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell Biol. 10: 1470–1476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Iraci N., Gaude E., Leonardi T., Costa A. S. H., Cossetti C., Peruzzotti-Jametti L., Bernstock J. D., Saini H. K., Gelati M., Vescovi A. L., et al. 2017. Extracellular vesicles are independent metabolic units with asparaginase activity. Nat. Chem. Biol. 13: 951–955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Zhao H., Yang L., Baddour J., Achreja A., Bernard V., Moss T., Marini J. C., Tudawe T., Seviour E. G., San Lucas F. A., et al. 2016. Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism. eLife. 5: e10250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Wang S., Cesca F., Loers G., Schweizer M., Buck F., Benfenati F., Schachner M., and Kleene R.. 2011. Synapsin I is an oligomannose-carrying glycoprotein, acts as an oligomannose-binding lectin, and promotes neurite outgrowth and neuronal survival when released via glia-derived exosomes. J. Neurosci. 31: 7275–7290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Frühbeis C., Fröhlich D., Kuo W. P., Amphornrat J., Thilemann S., Saab A. S., Kirchhoff F., Möbius W., Goebbels S., Nave K. A., et al. 2013. Neurotransmitter-triggered transfer of exosomes mediates oligodendrocyte-neuron communication. PLoS Biol. 11: e1001604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Yáñez-Mo M., Siljander P. R., Andreu Z., Zavec A. B., Borràs F. E., Buzas E. I., Buzas K., Casal E., Cappello F., Carvalho J., et al. 2015. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles. 4: 27066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Quek C., and Hill A. F.. 2017. The role of extracellular vesicles in neurodegenerative diseases. Biochem. Biophys. Res. Commun. 483: 1178–1186. [DOI] [PubMed] [Google Scholar]
  • 16.Joshi P., Benussi L., Furlan R., Ghidoni R., and Verderio C.. 2015. Extracellular vesicles in Alzheimer’s disease: friends or foes? Focus on abeta-vesicle interaction. Int. J. Mol. Sci. 16: 4800–4813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Croese T., and Furlan R.. 2018. Extracellular vesicles in neurodegenerative diseases. Mol. Aspects Med. 60: 52–61. [DOI] [PubMed] [Google Scholar]
  • 18.Tricarico C., Clancy J., and D’Souza-Schorey C.. 2017. Biology and biogenesis of shed microvesicles. Small GTPases. 8: 220–232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Juan T., and Furthauer M.. 2018. Biogenesis and function of ESCRT-dependent extracellular vesicles. Semin. Cell Dev. Biol. 74: 66–77. [DOI] [PubMed] [Google Scholar]
  • 20.Giussani P., Tringali C., Riboni L., Viani P., and Venerando B.. 2014. Sphingolipids: key regulators of apoptosis and pivotal players in cancer drug resistance. Int. J. Mol. Sci. 15: 4356–4392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hannun Y. A., and Obeid L. M.. 2008. Principles of bioactive lipid signalling: lessons from sphingolipids. Nat. Rev. Mol. Cell Biol. 9: 139–150. [DOI] [PubMed] [Google Scholar]
  • 22.Chiricozzi E., Loberto N., Schiumarini D., Samarani M., Mancini G., Tamanini A., Lippi G., Dechecchi M. C., Bassi R., Giussani P., et al. 2018. Sphingolipids role in the regulation of inflammatory response: From leukocyte biology to bacterial infection. J. Leukoc. Biol. 103: 445–456. [DOI] [PubMed] [Google Scholar]
  • 23.Cuvillier O., Pirianov G., Kleuser B., Vanek P. G., Coso O. A., Gutkind S., and Spiegel S.. 1996. Suppression of ceramide-mediated programmed cell death by sphingosine-1-phosphate. Nature. 381: 800–803. [DOI] [PubMed] [Google Scholar]
  • 24.Gault C. R., Obeid L. M., and Hannun Y. A.. 2010. An overview of sphingolipid metabolism: from synthesis to breakdown. Adv. Exp. Med. Biol. 688: 1–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Tettamanti G., Bassi R., Viani P., and Riboni L.. 2003. Salvage pathways in glycosphingolipid metabolism. Biochimie. 85: 423–437. [DOI] [PubMed] [Google Scholar]
  • 26.Kitatani K., Idkowiak-Baldys J., and Hannun Y. A.. 2008. The sphingolipid salvage pathway in ceramide metabolism and signaling. Cell Signal. 20: 1010–1018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hakomori S. 2000. Traveling for the glycosphingolipid path. Glycoconj. J. 17: 627–647. [DOI] [PubMed] [Google Scholar]
  • 28.Ichikawa S., and Hirabayashi Y.. 1998. Glucosylceramide synthase and glycosphingolipid synthesis. Trends Cell Biol. 8: 198–202. [DOI] [PubMed] [Google Scholar]
  • 29.Marchesini N., and Hannun Y. A.. 2004. Acid and neutral sphingomyelinases: roles and mechanisms of regulation. Biochem. Cell Biol. 82: 27–44. [DOI] [PubMed] [Google Scholar]
  • 30.Hakomori S. I. 2010. Glycosynaptic microdomains controlling tumor cell phenotype through alteration of cell growth, adhesion, and motility. FEBS Lett. 584: 1901–1906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Véret J., Bellini L., Giussani P., Ng C., Magnan C., and Le Stunff H.. 2014. Roles of sphingolipid metabolism in pancreatic beta cell dysfunction induced by lipotoxicity. J. Clin. Med. 3: 646–662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Airola M. V., and Hannun Y. A.. 2013. Sphingolipid metabolism and neutral sphingomyelinases. Handb. Exp. Pharmacol. 215: 57–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Bienias K., Fiedorowicz A., Sadowska A., Prokopiuk S., and Car H.. 2016. Regulation of sphingomyelin metabolism. Pharmacol. Rep. 68: 570–581. [DOI] [PubMed] [Google Scholar]
  • 34.Ogretmen B. 2018. Sphingolipid metabolism in cancer signalling and therapy. Nat. Rev. Cancer. 18: 33–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Zhou Y., Salker M. S., Walker B., Munzer P., Borst O., Gawaz M., Gulbins E., Singh Y., and Lang F.. 2016. Acid sphingomyelinase (ASM) is a negative regulator of regulatory T cell (Treg) development. Cell. Physiol. Biochem. 39: 985–995. [DOI] [PubMed] [Google Scholar]
  • 36.Devaux P. F., and Morris R.. 2004. Transmembrane asymmetry and lateral domains in biological membranes. Traffic. 5: 241–246. [DOI] [PubMed] [Google Scholar]
  • 37.Simons K., and Ikonen E.. 1997. Functional rafts in cell membranes. Nature. 387: 569–572. [DOI] [PubMed] [Google Scholar]
  • 38.Slotte J. P., Hedstrom G., Rannstrom S., and Ekman S.. 1989. Effects of sphingomyelin degradation on cell cholesterol oxidizability and steady-state distribution between the cell surface and the cell interior. Biochim. Biophys. Acta. 985: 90–96. [DOI] [PubMed] [Google Scholar]
  • 39.Neufeld E. B., Cooney A. M., Pitha J., Dawidowicz E. A., Dwyer N. K., Pentchev P. G., and Blanchette-Mackie E. J.. 1996. Intracellular trafficking of cholesterol monitored with a cyclodextrin. J. Biol. Chem. 271: 21604–21613. [DOI] [PubMed] [Google Scholar]
  • 40.Jarsch I. K., Daste F., and Gallop J. L.. 2016. Membrane curvature in cell biology: an integration of molecular mechanisms. J. Cell Biol. 214: 375–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Gulbins E., and Kolesnick R.. 2000. Measurement of sphingomyelinase activity. Methods Enzymol. 322: 382–388. [DOI] [PubMed] [Google Scholar]
  • 42.Milhas D., Clarke C. J., Idkowiak-Baldys J., Canals D., and Hannun Y. A.. 2010. Anterograde and retrograde transport of neutral sphingomyelinase-2 between the Golgi and the plasma membrane. Biochim. Biophys. Acta. 1801: 1361–1374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Tomiuk S., Zumbansen M., and Stoffel W.. 2000. Characterization and subcellular localization of murine and human magnesium-dependent neutral sphingomyelinase. J. Biol. Chem. 275: 5710–5717. [DOI] [PubMed] [Google Scholar]
  • 44.Mizutani Y., Tamiya-Koizumi K., Nakamura N., Kobayashi M., Hirabayashi Y., and Yoshida S.. 2001. Nuclear localization of neutral sphingomyelinase 1: biochemical and immunocytochemical analyses. J. Cell Sci. 114: 3727–3736. [DOI] [PubMed] [Google Scholar]
  • 45.Veldman R. J., Maestre N., Aduib O. M., Medin J. A., Salvayre R., and Levade T.. 2001. A neutral sphingomyelinase resides in sphingolipid-enriched microdomains and is inhibited by the caveolin-scaffolding domain: potential implications in tumour necrosis factor signalling. Biochem. J. 355: 859–868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Goñi F. M., and Alonso A.. 2002. Sphingomyelinases: enzymology and membrane activity. FEBS Lett. 531: 38–46. [DOI] [PubMed] [Google Scholar]
  • 47.Truman J. P., Al Gadban M. M., Smith K. J., and Hammad S. M.. 2011. Acid sphingomyelinase in macrophage biology. Cell. Mol. Life Sci. 68: 3293–3305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Tepper A. D., Ruurs P., Wiedmer T., Sims P. J., Borst J., and van Blitterswijk W. J.. 2000. Sphingomyelin hydrolysis to ceramide during the execution phase of apoptosis results from phospholipid scrambling and alters cell-surface morphology. J. Cell Biol. 150: 155–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Nurminen T. A., Holopainen J. M., Zhao H., and Kinnunen P. K.. 2002. Observation of topical catalysis by sphingomyelinase coupled to microspheres. J. Am. Chem. Soc. 124: 12129–12134. [DOI] [PubMed] [Google Scholar]
  • 50.Trajkovic K., Hsu C., Chiantia S., Rajendran L., Wenzel D., Wieland F., Schwille P., Brugger B., and Simons M.. 2008. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 319: 1244–1247. [DOI] [PubMed] [Google Scholar]
  • 51.Cheng Q., Li X., Wang Y., Dong M., Zhan F. H., and Liu J.. 2018. The ceramide pathway is involved in the survival, apoptosis and exosome functions of human multiple myeloma cells in vitro. Acta Pharmacol. Sin. 39: 561–568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Guo B. B., Bellingham S. A., and Hill A. F.. 2015. The neutral sphingomyelinase pathway regulates packaging of the prion protein into exosomes. J. Biol. Chem. 290: 3455–3467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kosaka N., Iguchi H., Yoshioka Y., Takeshita F., Matsuki Y., and Ochiya T.. 2010. Secretory mechanisms and intercellular transfer of microRNAs in living cells. J. Biol. Chem. 285: 17442–17452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mittelbrunn M., Gutierrez-Vazquez C., Villarroya-Beltri C., Gonzalez S., Sanchez-Cabo F., Gonzalez M. A., Bernad A., and Sanchez-Madrid F.. 2011. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat. Commun. 2: 282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Yuyama K., Sun H., Mitsutake S., and Igarashi Y.. 2012. Sphingolipid-modulated exosome secretion promotes clearance of amyloid-beta by microglia. J. Biol. Chem. 287: 10977–10989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Wang G., Dinkins M., He Q., Zhu G., Poirier C., Campbell A., Mayer-Proschel M., and Bieberich E.. 2012. Astrocytes secrete exosomes enriched with proapoptotic ceramide and prostate apoptosis response 4 (PAR-4): potential mechanism of apoptosis induction in Alzheimer disease (AD). J. Biol. Chem. 287: 21384–21395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Asai H., Ikezu S., Tsunoda S., Medalla M., Luebke J., Haydar T., Wolozin B., Butovsky O., Kugler S., and Ikezu T.. 2015. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat. Neurosci. 18: 1584–1593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Xu Y., Liu Y., Yang C., Kang L., Wang M., Hu J., He H., Song W., and Tang H.. 2016. Macrophages transfer antigens to dendritic cells by releasing exosomes containing dead-cell-associated antigens partially through a ceramide-dependent pathway to enhance CD4(+) T-cell responses. Immunology. 149: 157–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Nojima H., Freeman C. M., Schuster R. M., Japtok L., Kleuser B., Edwards M. J., Gulbins E., and Lentsch A. B.. 2016. Hepatocyte exosomes mediate liver repair and regeneration via sphingosine-1-phosphate. J. Hepatol. 64: 60–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Dinkins M. B., Dasgupta S., Wang G., Zhu G., and Bieberich E.. 2014. Exosome reduction in vivo is associated with lower amyloid plaque load in the 5XFAD mouse model of Alzheimer’s disease. Neurobiol. Aging. 35: 1792–1800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Dinkins M. B., Enasko J., Hernandez C., Wang G., Kong J., Helwa I., Liu Y., Terry A. V. Jr., and Bieberich E.. 2016. Neutral sphingomyelinase-2 deficiency ameliorates alzheimer’s disease pathology and improves cognition in the 5XFAD mouse. J. Neurosci. 36: 8653–8667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Nojima H., Konishi T., Freeman C. M., Schuster R. M., Japtok L., Kleuser B., Edwards M. J., Gulbins E., and Lentsch A. B.. 2016. Chemokine receptors, CXCR1 and CXCR2, differentially regulate exosome release in hepatocytes. PLoS One. 11: e0161443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.van Niel G., Charrin S., Simoes S., Romao M., Rochin L., Saftig P., Marks M. S., Rubinstein E., and Raposo G.. 2011. The tetraspanin CD63 regulates ESCRT-independent and -dependent endosomal sorting during melanogenesis. Dev. Cell. 21: 708–721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Colombo M., Moita C., van Niel G., Kowal J., Vigneron J., Benaroch P., Manel N., Moita L. F., Thery C., and Raposo G.. 2013. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J. Cell Sci. 126: 5553–5565. [DOI] [PubMed] [Google Scholar]
  • 65.Phuyal S., Hessvik N. P., Skotland T., Sandvig K., and Llorente A.. 2014. Regulation of exosome release by glycosphingolipids and flotillins. FEBS J. 281: 2214–2227. [DOI] [PubMed] [Google Scholar]
  • 66.Skotland T., Sandvig K., and Llorente A.. 2017. Lipids in exosomes: current knowledge and the way forward. Prog. Lipid Res. 66: 30–41. [DOI] [PubMed] [Google Scholar]
  • 67.Kajimoto T., Okada T., Miya S., Zhang L., and Nakamura S.. 2013. Ongoing activation of sphingosine 1-phosphate receptors mediates maturation of exosomal multivesicular endosomes. Nat. Commun. 4: 2712. [DOI] [PubMed] [Google Scholar]
  • 68.Kajimoto T., Mohamed N. N. I., Badawy S. M. M., Matovelo S. A., Hirase M., Nakamura S., Yoshida D., Okada T., Ijuin T., and Nakamura S. I.. 2018. Involvement of Gβγ subunits of Gi protein coupled with S1P receptor on multivesicular endosomes in F-actin formation and cargo sorting into exosomes. J. Biol. Chem. 293: 245–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Menck K., Sonmezer C., Worst T. S., Schulz M., Dihazi G. H., Streit F., Erdmann G., Kling S., Boutros M., Binder C., et al. 2017. Neutral sphingomyelinases control extracellular vesicles budding from the plasma membrane. J. Extracell. Vesicles. 6: 1378056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Bianco F., Perrotta C., Novellino L., Francolini M., Riganti L., Menna E., Saglietti L., Schuchman E. H., Furlan R., Clementi E., et al. 2009. Acid sphingomyelinase activity triggers microparticle release from glial cells. EMBO J. 28: 1043–1054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Bianco F., Pravettoni E., Colombo A., Schenk U., Moller T., Matteoli M., and Verderio C.. 2005. Astrocyte-derived ATP induces vesicle shedding and IL-1 beta release from microglia. J. Immunol. 174: 7268–7277. [DOI] [PubMed] [Google Scholar]
  • 72.Ciregia F., Urbani A., and Palmisano G.. 2017. Extracellular vesicles in brain tumors and neurodegenerative diseases. Front. Mol. Neurosci. 10: 276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ferrari D., Pizzirani C., Adinolfi E., Lemoli R. M., Curti A., Idzko M., Panther E., and Di Virgilio F.. 2006. The P2X7 receptor: a key player in IL-1 processing and release. J. Immunol. 176: 3877–3883. [DOI] [PubMed] [Google Scholar]
  • 74.Próchnicki T., Mangan M. S., and Latz E.. 2016. Recent insights into the molecular mechanisms of the NLRP3 inflammasome activation. F1000Res. 5: 1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Martinon F., Burns K., and Tschopp J.. 2002. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol. Cell. 10: 417–426. [DOI] [PubMed] [Google Scholar]
  • 76.Pizzirani C., Ferrari D., Chiozzi P., Adinolfi E., Sandona D., Savaglio E., and Di Virgilio F.. 2007. Stimulation of P2 receptors causes release of IL-1beta-loaded microvesicles from human dendritic cells. Blood. 109: 3856–3864. [DOI] [PubMed] [Google Scholar]
  • 77.Gulinelli S., Salaro E., Vuerich M., Bozzato D., Pizzirani C., Bolognesi G., Idzko M., Di Virgilio F., and Ferrari D.. 2012. IL-18 associates to microvesicles shed from human macrophages by a LPS/TLR-4 independent mechanism in response to P2X receptor stimulation. Eur. J. Immunol. 42: 3334–3345. [DOI] [PubMed] [Google Scholar]
  • 78.Furlan-Freguia C., Marchese P., Gruber A., Ruggeri Z. M., and Ruf W.. 2011. P2X7 receptor signaling contributes to tissue factor-dependent thrombosis in mice. J. Clin. Invest. 121: 2932–2944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Denlinger L. C., Fisette P. L., Sommer J. A., Watters J. J., Prabhu U., Dubyak G. R., Proctor R. A., and Bertics P. J.. 2001. Cutting edge: the nucleotide receptor P2X7 contains multiple protein- and lipid-interaction motifs including a potential binding site for bacterial lipopolysaccharide. J. Immunol. 167: 1871–1876. [DOI] [PubMed] [Google Scholar]
  • 80.Subra C., Laulagnier K., Perret B., and Record M.. 2007. Exosome lipidomics unravels lipid sorting at the level of multivesicular bodies. Biochimie. 89: 205–212. [DOI] [PubMed] [Google Scholar]
  • 81.Wang J., Pendurthi U. R., and Rao L. V. M.. 2017. Sphingomyelin encrypts tissue factor: ATP-induced activation of A-SMase leads to tissue factor decryption and microvesicle shedding. Blood Adv. 1: 849–862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Barth K., Weinhold K., Guenther A., Young M. T., Schnittler H., and Kasper M.. 2007. Caveolin-1 influences P2X7 receptor expression and localization in mouse lung alveolar epithelial cells. FEBS J. 274: 3021–3033. [DOI] [PubMed] [Google Scholar]
  • 83.Drago F., Lombardi M., Prada I., Gabrielli M., Joshi P., Cojoc D., Franck J., Fournier I., Vizioli J., and Verderio C.. 2017. ATP modifies the proteome of extracellular vesicles released by microglia and influences their action on astrocytes. Front. Pharmacol. 8: 910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Truman J. P., Al Gadban M. M., Smith K. J., Jenkins R. W., Mayroo N., Virella G., Lopes-Virella M. F., Bielawska A., Hannun Y. A., and Hammad S. M.. 2012. Differential regulation of acid sphingomyelinase in macrophages stimulated with oxidized low-density lipoprotein (LDL) and oxidized LDL immune complexes: role in phagocytosis and cytokine release. Immunology. 136: 30–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Marrone M. C., Morabito A., Giustizieri M., Chiurchiu V., Leuti A., Mattioli M., Marinelli S., Riganti L., Lombardi M., Murana E., et al. 2017. TRPV1 channels are critical brain inflammation detectors and neuropathic pain biomarkers in mice. Nat. Commun. 8: 15292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Caterina M. J., Schumacher M. A., Tominaga M., Rosen T. A., Levine J. D., and Julius D.. 1997. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature. 389: 816–824. [DOI] [PubMed] [Google Scholar]
  • 87.Nieto-Posadas A., Picazo-Juarez G., Llorente I., Jara-Oseguera A., Morales-Lazaro S., Escalante-Alcalde D., Islas L. D., and Rosenbaum T.. 2011. Lysophosphatidic acid directly activates TRPV1 through a C-terminal binding site. Nat. Chem. Biol. 8: 78–85. [Erratum. 2012. Nat. Chem. Biol. 8: 737.] [DOI] [PubMed] [Google Scholar]
  • 88.Amantini C., Mosca M., Nabissi M., Lucciarini R., Caprodossi S., Arcella A., Giangaspero F., and Santoni G.. 2007. Capsaicin-induced apoptosis of glioma cells is mediated by TRPV1 vanilloid receptor and requires p38 MAPK activation. J. Neurochem. 102: 977–990. [DOI] [PubMed] [Google Scholar]
  • 89.Awojoodu A. O., Keegan P. M., Lane A. R., Zhang Y., Lynch K. R., Platt M. O., and Botchwey E. A.. 2014. Acid sphingomyelinase is activated in sickle cell erythrocytes and contributes to inflammatory microparticle generation in SCD. Blood. 124: 1941–1950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Hanada K., Mitamura T., Fukasawa M., Magistrado P. A., Horii T., and Nishijima M.. 2000. Neutral sphingomyelinase activity dependent on Mg2+ and anionic phospholipids in the intraerythrocytic malaria parasite Plasmodium falciparum. Biochem. J. 346: 671–677. [PMC free article] [PubMed] [Google Scholar]
  • 91.D’Alessandro A., Righetti P. G., and Zolla L.. 2010. The red blood cell proteome and interactome: an update. J. Proteome Res. 9: 144–163. [DOI] [PubMed] [Google Scholar]
  • 92.Record M., Loyter A., and Gatt S.. 1980. Utilization of membranous lipid substrates by membranous enzymes. Hydrolysis of sphingomyelin in erythrocyte ‘ghosts’ and liposomes by the membranous sphingomyelinase of chicken erythrocyte ‘ghosts’. Biochem. J. 187: 115–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Jenkins R. W., Canals D., and Hannun Y. A.. 2009. Roles and regulation of secretory and lysosomal acid sphingomyelinase. Cell. Signal. 21: 836–846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Hoehn R. S., Jernigan P. L., Japtok L., Chang A. L., Midura E. F., Caldwell C. C., Kleuser B., Lentsch A. B., Edwards M. J., Gulbins E., et al. 2017. Acid sphingomyelinase inhibition in stored erythrocytes reduces transfusion-associated lung inflammation. Ann. Surg. 265: 218–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Serban K. A., Rezania S., Petrusca D. N., Poirier C., Cao D., Justice M. J., Patel M., Tsvetkova I., Kamocki K., Mikosz A., et al. 2016. Structural and functional characterization of endothelial microparticles released by cigarette smoke. Sci. Rep. 6: 31596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Li C. J., Liu Y., Chen Y., Yu D., Williams K. J., and Liu M. L.. 2013. Novel proteolytic microvesicles released from human macrophages after exposure to tobacco smoke. Am. J. Pathol. 182: 1552–1562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Colombo F., Bastoni M., Nigro A., Podini P., Finardi A., Casella G., Ramesh M., Farina C., Verderio C., and Furlan R.. 2018. Cytokines stimulate the release of microvesicles from myeloid cells independently from the P2X7 receptor/acid sphingomyelinase pathway. Front. Immunol. 9: 204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.D’Auria L., Reiter C., Ward E., Moyano A. L., Marshall M. S., Nguyen D., Scesa G., Hauck Z., van Breemen R., Givogri M. I., et al. 2017. Psychosine enhances the shedding of membrane microvesicles: Implications in demyelination in Krabbe’s disease. PLoS One. 12: e0178103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.White A. B., Givogri M. I., Lopez-Rosas A., Cao H., van Breemen R., Thinakaran G., and Bongarzone E. R.. 2009. Psychosine accumulates in membrane microdomains in the brain of Krabbe patients, disrupting the raft architecture. J. Neurosci. 29: 6068–6077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Scesa G., Moyano A. L., Bongarzone E. R., and Givogri M. I.. 2016. Port-to-port delivery: Mobilization of toxic sphingolipids via extracellular vesicles. J. Neurosci. Res. 94: 1333–1340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Haraszti R. A., Didiot M. C., Sapp E., Leszyk J., Shaffer S. A., Rockwell H. E., Gao F., Narain N. R., DiFiglia M., Kiebish M. A., et al. 2016. High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources. J. Extracell. Vesicles. 5: 32570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Llorente A., Skotland T., Sylvanne T., Kauhanen D., Rog T., Orlowski A., Vattulainen I., Ekroos K., and Sandvig K.. 2013. Molecular lipidomics of exosomes released by PC-3 prostate cancer cells. Biochim. Biophys. Acta. 1831: 1302–1309. [DOI] [PubMed] [Google Scholar]
  • 103.Phuyal S., Skotland T., Hessvik N. P., Simolin H., Overbye A., Brech A., Parton R. G., Ekroos K., Sandvig K., and Llorente A.. 2015. The ether lipid precursor hexadecylglycerol stimulates the release and changes the composition of exosomes derived from PC-3 cells. J. Biol. Chem. 290: 4225–4237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Wubbolts R., Leckie R. S., Veenhuizen P. T., Schwarzmann G., Mobius W., Hoernschemeyer J., Slot J. W., Geuze H. J., and Stoorvogel W.. 2003. Proteomic and biochemical analyses of human B cell-derived exosomes. Potential implications for their function and multivesicular body formation. J. Biol. Chem. 278: 10963–10972. [DOI] [PubMed] [Google Scholar]
  • 105.Vidal M., Sainte-Marie J., Philippot J. R., and Bienvenue A.. 1989. Asymmetric distribution of phospholipids in the membrane of vesicles released during in vitro maturation of guinea pig reticulocytes: evidence precluding a role for “aminophospholipid translocase”. J. Cell. Physiol. 140: 455–462. [DOI] [PubMed] [Google Scholar]
  • 106.Yuyama K., Sun H., Sakai S., Mitsutake S., Okada M., Tahara H., Furukawa J., Fujitani N., Shinohara Y., and Igarashi Y.. 2014. Decreased amyloid-beta pathologies by intracerebral loading of glycosphingolipid-enriched exosomes in Alzheimer model mice. J. Biol. Chem. 289: 24488–24498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Dang V. D., Jella K. K., Ragheb R. R. T., Denslow N. D., and Alli A. A.. 2017. Lipidomic and proteomic analysis of exosomes from mouse cortical collecting duct cells. FASEB J. 31: 5399–5408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Podbielska M., Szulc Z. M., Kurowska E., Hogan E. L., Bielawski J., Bielawska A., and Bhat N. R.. 2016. Cytokine-induced release of ceramide-enriched exosomes as a mediator of cell death signaling in an oligodendroglioma cell line. J. Lipid Res. 57: 2028–2039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Carayon K., Chaoui K., Ronzier E., Lazar I., Bertrand-Michel J., Roques V., Balor S., Terce F., Lopez A., Salome L., et al. 2011. Proteolipidic composition of exosomes changes during reticulocyte maturation. J. Biol. Chem. 286: 34426–34439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Baig S., Lim J. Y., Fernandis A. Z., Wenk M. R., Kale A., Su L. L., Biswas A., Vasoo S., Shui G., and Choolani M.. 2013. Lipidomic analysis of human placental syncytiotrophoblast microvesicles in adverse pregnancy outcomes. Placenta. 34: 436–442. [DOI] [PubMed] [Google Scholar]
  • 111.Nilsson J., Skog J., Nordstrand A., Baranov V., Mincheva-Nilsson L., Breakefield X. O., and Widmark A.. 2009. Prostate cancer-derived urine exosomes: a novel approach to biomarkers for prostate cancer. Br. J. Cancer. 100: 1603–1607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Colombo E., Borgiani B., Verderio C., and Furlan R.. 2012. Microvesicles: novel biomarkers for neurological disorders. Front. Physiol. 3: 63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Martins V. R., Dias M. S., and Hainaut P.. 2013. Tumor-cell-derived microvesicles as carriers of molecular information in cancer. Curr. Opin. Oncol. 25: 66–75. [DOI] [PubMed] [Google Scholar]
  • 114.Lydic T. A., Townsend S., Adda C. G., Collins C., Mathivanan S., and Reid G. E.. 2015. Rapid and comprehensive ‘shotgun’ lipidome profiling of colorectal cancer cell derived exosomes. Methods. 87: 83–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Skotland T., Ekroos K., Kauhanen D., Simolin H., Seierstad T., Berge V., Sandvig K., and Llorente A.. 2017. Molecular lipid species in urinary exosomes as potential prostate cancer biomarkers. Eur. J. Cancer. 70: 122–132. [DOI] [PubMed] [Google Scholar]
  • 116.Moyano A. L., Li G., Boullerne A. I., Feinstein D. L., Hartman E., Skias D., Balavanov R., van Breemen R. B., Bongarzone E. R., Mansson J. E., et al. 2016. Sulfatides in extracellular vesicles isolated from plasma of multiple sclerosis patients. J. Neurosci. Res. 94: 1579–1587. [DOI] [PubMed] [Google Scholar]
  • 117.Yanagisawa K., Odaka A., Suzuki N., and Ihara Y.. 1995. GM1 ganglioside-bound amyloid beta-protein (A beta): a possible form of preamyloid in Alzheimer’s disease. Nat. Med. 1: 1062–1066. [DOI] [PubMed] [Google Scholar]
  • 118. Deleted in proof. [Google Scholar]
  • 119.Yanagisawa K., and Matsuzaki K.. 2002. Cholesterol-dependent aggregation of amyloid beta-protein. Ann. N. Y. Acad. Sci. 977: 384–386. [DOI] [PubMed] [Google Scholar]
  • 120.Yuyama K., and Yanagisawa K.. 2010. Sphingomyelin accumulation provides a favorable milieu for GM1 ganglioside-induced assembly of amyloid beta-protein. Neurosci. Lett. 481: 168–172. [DOI] [PubMed] [Google Scholar]
  • 121.Deng Z., Mu J., Tseng M., Wattenberg B., Zhuang X., Egilmez N. K., Wang Q., Zhang L., Norris J., Guo H., et al. 2015. Enterobacteria-secreted particles induce production of exosome-like S1P-containing particles by intestinal epithelium to drive Th17-mediated tumorigenesis. Nat. Commun. 6: 6956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Wang R., Ding Q., Yaqoob U., de Assuncao T. M., Verma V. K., Hirsova P., Cao S., Mukhopadhyay D., Huebert R. C., and Shah V. H.. 2015. Exosome adherence and internalization by hepatic stellate cells triggers sphingosine 1-phosphate-dependent migration. J. Biol. Chem. 290: 30684–30696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Record M., Carayon K., Poirot M., and Silvente-Poirot S.. 2014. Exosomes as new vesicular lipid transporters involved in cell-cell communication and various pathophysiologies. Biochim. Biophys. Acta. 1841: 108–120. [DOI] [PubMed] [Google Scholar]
  • 124.Rigogliuso S., Donati C., Cassara D., Taverna S., Salamone M., Bruni P., and Vittorelli M. L.. 2010. An active form of sphingosine kinase-1 is released in the extracellular medium as component of membrane vesicles shed by two human tumor cell lines. J. Oncol. 2010: 509329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Zhu Q., Kang J., Miao H., Feng Y., Xiao L., Hu Z., Liao D. F., Huang Y., Jin J., and He S.. 2014. Low-dose cytokine-induced neutral ceramidase secretion from INS-1 cells via exosomes and its anti-apoptotic effect. FEBS J. 281: 2861–2870. [DOI] [PubMed] [Google Scholar]
  • 126.Du Y., Li D., Han C., Wu H., Xu L., Zhang M., Zhang J., and Chen X.. 2017. Exosomes from human-induced pluripotent stem cell-derived mesenchymal stromal cells (hiPSC-MSCs) protect liver against hepatic ischemia/reperfusion injury via activating sphingosine kinase and sphingosine-1-phosphate Signaling Pathway. Cell. Physiol. Biochem. 43: 611–625. [DOI] [PubMed] [Google Scholar]
  • 127.Pyne N. J., El Buri A., Adams D. R., and Pyne S.. 2018. Sphingosine 1-phosphate and cancer. Adv. Biol. Regul. 68: 97–106. [DOI] [PubMed] [Google Scholar]
  • 128.Amorim M., Fernandes G., Oliveira P., Martins-de-Souza D., Dias-Neto E., and Nunes D.. 2014. The overexpression of a single oncogene (ERBB2/HER2) alters the proteomic landscape of extracellular vesicles. Proteomics. 14: 1472–1479. [DOI] [PubMed] [Google Scholar]
  • 129.Antonucci F., Turola E., Riganti L., Caleo M., Gabrielli M., Perrotta C., Novellino L., Clementi E., Giussani P., Viani P., et al. 2012. Microvesicles released from microglia stimulate synaptic activity via enhanced sphingolipid metabolism. EMBO J. 31: 1231–1240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Darios F., Wasser C., Shakirzyanova A., Giniatullin A., Goodman K., Munoz-Bravo J. L., Raingo J., Jorgacevski J., Kreft M., Zorec R., et al. 2009. Sphingosine facilitates SNARE complex assembly and activates synaptic vesicle exocytosis. Neuron. 62: 683–694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Riganti L., Antonucci F., Gabrielli M., Prada I., Giussani P., Viani P., Valtorta F., Menna E., Matteoli M., and Verderio C.. 2016. Sphingosine-1-phosphate (S1P) impacts presynaptic functions by regulating synapsin I localization in the presynaptic compartment. J. Neurosci. 36: 4624–4634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Schatz D., Rosenwasser S., Malitsky S., Wolf S. G., Feldmesser E., and Vardi A.. 2017. Communication via extracellular vesicles enhances viral infection of a cosmopolitan alga. Nat. Microbiol. 2: 1485–1492. [DOI] [PubMed] [Google Scholar]
  • 133.Ziv C., Malitsky S., Othman A., Ben-Dor S., Wei Y., Zheng S., Aharoni A., Hornemann T., and Vardi A.. 2016. Viral serine palmitoyltransferase induces metabolic switch in sphingolipid biosynthesis and is required for infection of a marine alga. Proc. Natl. Acad. Sci. USA. 113: E1907–E1916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Rosenwasser S., Mausz M. A., Schatz D., Sheyn U., Malitsky S., Aharoni A., Weinstock E., Tzfadia O., Ben-Dor S., Feldmesser E., et al. 2014. Rewiring host lipid metabolism by large viruses determines the fate of Emiliania huxleyi, a bloom-forming alga in the ocean. Plant Cell. 26: 2689–2707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Augustin S. M., and Lovinger D. M.. Functional relevance of endocannabinoid-dependent synaptic plasticity in the central nervous system. ACS Chem. Neurosci. Epub ahead of print. February 19, 2018; doi:10.1021/acschemneuro.7b00508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Zou S., and Kumar U.. 2018. Cannabinoid receptors and the endocannabinoid system: signaling and function in the central nervous system. Int. J. Mol. Sci. 19: E833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Benarroch E. E. 2014. Synaptic effects of cannabinoids: complexity, behavioral effects, and potential clinical implications. Neurology. 83: 1958–1967. [DOI] [PubMed] [Google Scholar]
  • 138.Ohno-Shosaku T., Maejima T., and Kano M.. 2001. Endogenous cannabinoids mediate retrograde signals from depolarized postsynaptic neurons to presynaptic terminals. Neuron. 29: 729–738. [DOI] [PubMed] [Google Scholar]
  • 139.Wilson R. I., and Nicoll R. A.. 2001. Endogenous cannabinoids mediate retrograde signalling at hippocampal synapses. Nature. 410: 588–592. [DOI] [PubMed] [Google Scholar]
  • 140.Katona I., and Freund T. F.. 2012. Multiple functions of endocannabinoid signaling in the brain. Annu. Rev. Neurosci. 35: 529–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Carrier E. J., Kearn C. S., Barkmeier A. J., Breese N. M., Yang W., Nithipatikom K., Pfister S. L., Campbell W. B., and Hillard C. J.. 2004. Cultured rat microglial cells synthesize the endocannabinoid 2-arachidonylglycerol, which increases proliferation via a CB2 receptor-dependent mechanism. Mol. Pharmacol. 65: 999–1007. [DOI] [PubMed] [Google Scholar]
  • 142.Stella N. 2010. Cannabinoid and cannabinoid-like receptors in microglia, astrocytes, and astrocytomas. Glia. 58: 1017–1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Walter L., Franklin A., Witting A., Moller T., and Stella N.. 2002. Astrocytes in culture produce anandamide and other acylethanolamides. J. Biol. Chem. 277: 20869–20876. [DOI] [PubMed] [Google Scholar]
  • 144.Navarrete M., and Araque A.. 2008. Endocannabinoids mediate neuron-astrocyte communication. Neuron. 57: 883–893. [DOI] [PubMed] [Google Scholar]
  • 145.Stella N. 2009. Endocannabinoid signaling in microglial cells. Neuropharmacology. 56 (Suppl. 1): 244–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Nicolussi S., and Gertsch J.. 2015. Endocannabinoid transport revisited. Vitam. Horm. 98: 441–485. [DOI] [PubMed] [Google Scholar]
  • 147.Haj-Dahmane S., Shen R. Y., Elmes M. W., Studholme K., Kanjiya M. P., Bogdan D., Thanos P. K., Miyauchi J. T., Tsirka S. E., Deutsch D. G., et al. 2018. Fatty-acid-binding protein 5 controls retrograde endocannabinoid signaling at central glutamate synapses. Proc. Natl. Acad. Sci. USA. 115: 3482–3487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Gabrielli M., Battista N., Riganti L., Prada I., Antonucci F., Cantone L., Matteoli M., Maccarrone M., and Verderio C.. 2015. Active endocannabinoids are secreted on extracellular membrane vesicles. EMBO Rep. 16: 213–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Fezza F., Oddi S., Di Tommaso M., De Simone C., Rapino C., Pasquariello N., Dainese E., Finazzi-Agro A., and Maccarrone M.. 2008. Characterization of biotin-anandamide, a novel tool for the visualization of anandamide accumulation. J. Lipid Res. 49: 1216–1223. [DOI] [PubMed] [Google Scholar]
  • 150.Katona I., Sperlagh B., Sik A., Kafalvi A., Vizi E. S., Mackie K., and Freund T. F.. 1999. Presynaptically located CB1 cannabinoid receptors regulate GABA release from axon terminals of specific hippocampal interneurons. J. Neurosci. 19: 4544–4558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Hoffman A. F., and Lupica C. R.. 2000. Mechanisms of cannabinoid inhibition of GABA(A) synaptic transmission in the hippocampus. J. Neurosci. 20: 2470–2479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Irving A. J., Coutts A. A., Harvey J., Rae M. G., Mackie K., Bewick G. S., and Pertwee R. G.. 2000. Functional expression of cell surface cannabinoid CB(1) receptors on presynaptic inhibitory terminals in cultured rat hippocampal neurons. Neuroscience. 98: 253–262. [DOI] [PubMed] [Google Scholar]
  • 153.Rappa G., Mercapide J., Anzanello F., Pope R. M., and Lorico A.. 2013. Biochemical and biological characterization of exosomes containing prominin-1/CD133. Mol. Cancer. 12: 62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Hosseini-Beheshti E., Pham S., Adomat H., Li N., and Tomlinson Guns E. S.. 2012. Exosomes as biomarker enriched microvesicles: characterization of exosomal proteins derived from a panel of prostate cell lines with distinct AR phenotypes. Mol. Cell. Proteomics. 11: 863–885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Losito I., Patruno R., Conte E., Cataldi T. R., Megli F. M., and Palmisano F.. 2013. Phospholipidomics of human blood microparticles. Anal. Chem. 85: 6405–6413. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Lipid Research are provided here courtesy of American Society for Biochemistry and Molecular Biology

RESOURCES