Tetraspanins are a family of 33 proteins in mammals believed to play a crucial role in the compartmentalization of various associated proteins within cells and membranes. Recent studies have elucidated the structure of several tetraspanin members, revealing that while the four transmembrane domains typically adopt a cone-shaped configuration in crystals, other conformations are also possible. This cone-shaped structure may explain why tetraspanins are often enriched in curved and tubular cellular structures, such as microvilli, tunneling nanotubes, retraction fibers, or at the site of virus budding, and may contribute to the formation or maintenance of these structures. Tetraspanins have also been detected on midbody remnants and migrasomes, as well as on extracellular vesicles (EVs), for which CD9, CD81, and CD63 are widely used as markers. Although their impact on certain membrane structures and their ability to regulate the function and trafficking of associated proteins would suggest a potential role of tetraspanins either in EV formation or in regulating their protein composition, or both, efforts to characterize these roles have been complicated by conflicting results. In line with the interaction of certain tetraspanins with cholesterol, two recent studies have suggested that the presence or organization of oxysterols and cholesterol in EVs may be regulated by Tspan6 and CD63, respectively, paving the way for further research on the influence of tetraspanins on the lipid composition of EVs.

Tetraspanins are a family of proteins characterized by four transmembrane domains, expressed across all metazoans, and consist of 33 members in mammals [1,2]. Genetic studies in mice and the identification of rare mutations in humans have underscored the critical roles of specific tetraspanins in various biological processes, including immunity, vision, kidney function, fertilization, muscle regeneration, and the regulation of distinct cell populations within various organs (for review [3-5]). Additionally, numerous studies have highlighted their involvement in cellular infection by intracellular pathogens and the regulation of cancer progression (for review [6,7]). Here, we discuss the role of tetraspanins on membrane organelles and trafficking, with a focus on extracellular vesicles (EVs).

Tetraspanins are relatively small proteins, with a backbone of 200–300 amino acids but are variably glycosylated. In addition to their four transmembrane domains, tetraspanins share several structural features, including conserved residues, particularly cysteines. Intracellular cysteines serve as sites for palmitoylation, while extracellular cysteines contribute to the specific folding of the larger one of the two extracellular domains [4,5,8]. Recent studies have provided new insights into the structure of tetraspanins ([9-15], reviewed in [8]). In the CD9, CD81, and CD53 crystal structures, as well as in the Tspan15 cryo-EM structure (determined in association with ADAM10), the four transmembrane domains are arranged in two pairs of antiparallel helices (TM1/TM2 and TM3/TM4) that form a cone-shaped structure that converges near the cytoplasmic side of the membrane, creating a central intramembrane cavity (Figure 1). The large extracellular loop (LEL) bends toward the membrane and sits over the cavity formed by the transmembrane in a configuration referred to as the ‘closed’ structure [9-11,15]. Other studies have shown that tetraspanins can adopt an ‘open’ structure, in which the transmembrane domains do not form an intramembrane cavity and the LEL extends away from the membrane. This is the case of peripherin-2 (PRPH2) and rod outer segment membrane protein 1 (ROM1), two distant tetraspanins that form a heterodimer involved in photoreceptor morphogenesis [14]. CD81 also adopts an open conformation in the complex with CD19 (Figure 1 bottom, cryo-EM), showcasing the structural plasticity of this tetraspanin [13]. Additionally, the LEL of CD81 exhibits significant flexibility, as different crystal structures have revealed slightly varied arrangements, indicating some adaptability [16-18]. This structural versatility may enable CD81 to interact with a range of structural partners, including the E2 envelope protein of the hepatitis C virus for which CD81 serves as an obligatory receptor [19].

Structure of four tetraspanins.

Figure 1:
Structure of four tetraspanins.

The four transmembrane domains are in blue, the three structurally conserved helices in the large extracellular domain in red, and the variable region in green. The small extracellular domain is in gold. All structures were downloaded from PDB (www.rcsb.org). Top: the crystal structures of CD9 (PDB # 6K4J) and CD53 (PDB # 6WVG) and the cryo-EM structure of Tspan15 in association with ADAM10 (not shown). Bottom: the structure of CD81 (PDB # 5TCX) in crystals and that determined by cryo-EM (PDB # 7JIC) in which CD81 is in complex with CD19. Three different views of this structure are shown, including one showing CD19. Note that in this structure, CD81 does not show an intramembrane cavity and that the LEL extends from the plane of the membrane.

Figure 1:
Structure of four tetraspanins.

The four transmembrane domains are in blue, the three structurally conserved helices in the large extracellular domain in red, and the variable region in green. The small extracellular domain is in gold. All structures were downloaded from PDB (www.rcsb.org). Top: the crystal structures of CD9 (PDB # 6K4J) and CD53 (PDB # 6WVG) and the cryo-EM structure of Tspan15 in association with ADAM10 (not shown). Bottom: the structure of CD81 (PDB # 5TCX) in crystals and that determined by cryo-EM (PDB # 7JIC) in which CD81 is in complex with CD19. Three different views of this structure are shown, including one showing CD19. Note that in this structure, CD81 does not show an intramembrane cavity and that the LEL extends from the plane of the membrane.

Close modal

While many tetraspanins, such as CD9, CD81, and CD151, are predominantly expressed at the cell membrane (see, e.g., among many others, [20-22]), others, like CD63 and Tspan6, show a prominent intracellular localization at steady state [23-25]. Many tetraspanins including CD9, CD81, and CD82 have been shown to be enriched in various membrane protrusions [26,27]. Among these protrusions, Tspan4 has gained recent attention because it is enriched (together with other tetraspanins) on retraction fibers, long tubular structures the cells leave behind them during migration, as well as migrasomes, micrometer-sized structures that form by local swelling of these fibers (Figure 2, [34]). Some tetraspanins are present on EVs released by cells into the extracellular space (Figure 2) [35], as well as on at least two enveloped viruses, influenza and HIV [36-41]. Among tetraspanins, CD9 and CD81 have been identified on both viruses, which is consistent with these two molecules being recruited to the site of HIV or influenza virus budding (Figure 2, [30,42]). A common feature between many of these cell structures is their high curvature, which aligns well with recent research using a biomimetic system of membrane tubules, which demonstrated that Tspan4 and CD9 sense membrane curvature and preferentially localize to regions with high positive curvature [43]. This behavior may be due to the cone-shaped structure of the tetraspanin intramembrane region [9-11].

Examples of tetraspanin localization on remarkable membrane structures.

Figure 2:
Examples of tetraspanin localization on remarkable membrane structures.

(A): Localization of GFP-tagged CD9 on tunneling nanotubes in SH-SY5Y cells (image kindly provided by Dr. C Brou (see: [28]). (B): Localization of CD9 (red) and CD81 (green) on midbody remnants in HeLa cells, similarly to what has been described in [29]. (C): CD9 (red), detected by super-resolution microscopy concentrates in areas in which the HIV protein GAG (white) accumulates and induces membrane bending (image kindly provided by Dr. PE Milhiet; see: [30]). (D): CD9 expression in oocytes. The punctuated pattern reflects its preferential localization on microvilli as shown by [31]. (E): GFP-tagged Tspan4 localizes on retraction fibers and migrasomes. (image reproduced from [32]; https://doi.org/10.1016/j.cellin.2021.100003). (F): Small extracellular vesicles are released by either direct budding from the plasma membrane (ectosomes, enriched in CD9 and CD81) or following the fusion of multivesicular endosome resulting in the release of their intraluminal vesicles (exosomes, enriched in CD63). A labeling of CD63 on extracellular vesicle analyzed by electronic microscopy is also shown (image reproduced from [33], https://doi.org/10.3390/antib9030029).

Figure 2:
Examples of tetraspanin localization on remarkable membrane structures.

(A): Localization of GFP-tagged CD9 on tunneling nanotubes in SH-SY5Y cells (image kindly provided by Dr. C Brou (see: [28]). (B): Localization of CD9 (red) and CD81 (green) on midbody remnants in HeLa cells, similarly to what has been described in [29]. (C): CD9 (red), detected by super-resolution microscopy concentrates in areas in which the HIV protein GAG (white) accumulates and induces membrane bending (image kindly provided by Dr. PE Milhiet; see: [30]). (D): CD9 expression in oocytes. The punctuated pattern reflects its preferential localization on microvilli as shown by [31]. (E): GFP-tagged Tspan4 localizes on retraction fibers and migrasomes. (image reproduced from [32]; https://doi.org/10.1016/j.cellin.2021.100003). (F): Small extracellular vesicles are released by either direct budding from the plasma membrane (ectosomes, enriched in CD9 and CD81) or following the fusion of multivesicular endosome resulting in the release of their intraluminal vesicles (exosomes, enriched in CD63). A labeling of CD63 on extracellular vesicle analyzed by electronic microscopy is also shown (image reproduced from [33], https://doi.org/10.3390/antib9030029).

Close modal

Tetraspanins not only sense curvature but can also influence the shape of membrane structures. It has long been recognized that alterations in tetraspanin expression levels or the use of tetraspanin-targeting antibodies can lead to membrane remodeling (reviewed in [26,27]). A recent example is the demonstration that CD9 and CD81 are expressed on tubular nanotubes, long and thin membranous conduits between cells, and regulate their number or their ability to transfer material (Figure 2, [28]). A striking example is that of CD9, a tetraspanin essential for sperm–egg fusion [44], which localizes on egg microvilli (Figure 2) that become shorter and thicker in the absence of CD9 [31]. It is not known whether these modifications are the consequence of a direct effect of CD9 or secondary to cell signaling or cytoskeleton rearrangements. A potential direct effect of CD9 on membrane shape is suggested by the finding that in the CD9 crystals, the arrayed CD9 molecules in the lipidic environment triggered wavy layers in the crystalline lattice caused by the induction of curvature in the lipid membranes [10]. In addition, several tetraspanins have been shown to regulate migrasome formation and biomimetic in vitro models demonstrated that Tspan4 could generate or stabilize migrasome-like structures, particularly in the presence of cholesterol, suggesting a direct role for this tetraspanin in migrasome formation [34,45].

Many studies have investigated the role of tetraspanins, particularly in cancer, by identifying the proteins with which they associate, such as adhesion molecules, growth factor receptors, and ectoenzymes, and analyzing their influence on these proteins’ functions. Through these investigations, the most extensively studied tetraspanins, including CD9, CD81, CD82, CD53, CD151, and Tspan8, have been found to co-immunoprecipitate the same repertoire of integral membrane proteins and to associate with one another (reviewed in [3-5]). This observation led to the hypothesis that tetraspanins organize a dynamic network of interactions, referred to as the ‘tetraspanin web’ [46]. Tetraspanins have also been shown to partition into the light–density fractions of sucrose gradients, similarly to lipid-enriched microdomain (lipid raft) resident proteins, although under distinct experimental conditions. This finding led to the proposal that tetraspanins form tetraspanin-enriched microdomains, which could serve to cluster specific proteins and lipids, potentially playing a role in cell signaling [47,48]. This concept evolved into the broader idea that tetraspanins contribute to membrane compartmentalization [4,5]. It later became apparent that many of these interactions are likely indirect, partly secondary to tetraspanin–tetraspanin interactions [48-52]. Concurrently, the discovery of a limited number of well-characterized direct (primary) complexes [48-52] underscored the crucial and functionally relevant role of tetraspanins in regulating the trafficking and function of their direct interaction partners. This is particularly well illustrated by extensive studies on integrins, CD19, and ADAM10, which have been shown to associate with multiple tetraspanins but are distinctly regulated by the specific tetraspanins with which they directly interact.

Laminin-binding integrins such as α3β1, α6β1, and α6β4 interact directly with the tetraspanin CD151 [49,51]. CD151 silencing has been shown to reduce α3β1 integrin endocytosis without affecting the total or surface levels of the integrin, suggesting that CD151 influences integrin recycling in addition to internalization [53]. Moreover, CD151 regulates adhesion strengthening following the interaction of α6β1 integrin with laminin, a process essential for reinforcing the interaction between integrins and their ligands, allowing cells to withstand mechanical forces and avoid detachment [54]. This functional importance is highlighted by the phenotypes observed in patients with CD151 mutations or in CD151 knockout (KO) mice, which exhibit conditions similar to integrin deficiencies, such as skin blistering and kidney failure [55-57]. In the case of kidney failure, this defect is linked to the inability of podocytes, the filtering cells in the kidney’s filtration system, to resist the expansive forces generated by the pressure gradient within the kidney [57].

In contrast with CD151, CD81 is critical for the biosynthetic trafficking of its major partner protein in B lymphoid cells, the co-stimulatory molecule CD19, which is essential for optimal B-cell activation [58]. A patient with a CD81 mutation exhibited a severely impaired humoral immune response due to the absence of CD19 on the cell surface [59]. In mice, in which CD19 surface reduction is less pronounced, the humoral response was also affected and CD81 was shown to influence CD19 dynamics and organization [60,61]. As noted above, in a cryo-EM structure of CD81 associated with CD19, CD81 adopts an ‘open’ conformation, losing its intramembrane cavity as the LEL extends from the plasma membrane [13].

Another well-characterized example is the tetraspanin-dependent regulation of ADAM10. This metalloprotease is responsible for ectodomain shedding of various transmembrane proteins, which constitutes the normal secretion pathway for certain cytokine and growth factors, and is especially important for Notch signaling as it is the first step allowing the release of the Notch intracellular domain which acts as a transcriptional cofactor [62]. The efficient exit of ADAM10 from the ER requires the presence of one of a subset of six tetraspanins known as TspanC8, which are characterized by eight cysteines in their LEL [63-65]. TspanC8 tetraspanins not only facilitate ADAM10 egress from the ER but also regulate subsequent steps in its trafficking, membrane compartmentalization, and substrate selectivity [66-68]. Interestingly, TspanC8 tetraspanins are among the few tetraspanins with orthologs in invertebrates, which also regulate ADAM10 trafficking and Notch signaling in Caenorhabditis elegans and Drosophila melanogaster [64,69,70].

CD63, the best-characterized intracellular tetraspanin, traffics to the cell surface but is rapidly internalized due to a C-terminal GYxxΦ (GYEVM) motif that interacts with AP2 and AP3 adaptor proteins, targeting CD63 to late endosomes and lysosome-related organelles (LROs), including lysosomes and multivesicular endosomes (MVEs) [23]. Consistent with this, CD63 has been shown to address several interacting molecules to CD63-positive intracellular compartments, such as lysosomes. In its absence or when the GYxxΦ motif is mutated, these molecules localize at the plasma membrane. Examples of molecules the trafficking of which is regulated by CD63 include the H,K-ATPase β-subunit, colonic H+-K+ ATPase, Ca2+ sensor synaptotagmin VII, Pmel17, CXCR4, and MT1-MMP [71-76]. It remains unclear whether CD63 directly interacts with these proteins. Nevertheless, tetraspanins can also influence the trafficking of proteins with which they may not interact directly. For instance, Tspan7 has been shown to regulate AMPA receptor trafficking by interacting with the intracellular PDZ protein PICK1 through its PDZ-binding domain, competing with the AMPAR subunit GluA2 for PICK1 binding [77].

Cells secrete vesicles of various size delimited by a lipid bilayer that have been implicated in various physiological and pathological processes, including immunity, inflammation, neurodegeneration, and cancer. These EVs support intercellular communication through the exchange of proteins, lipids, and nucleic acids. EV signaling also occurs by contact with the plasma membrane of recipient cells [78,79]. The mechanisms of EV biogenesis vary between cell types and signaling status. Besides apoptotic bodies, two main subtypes of EVs—ectosomes and exosomes—have been categorized based on their subcellular origin. Ectosomes bud from the plasma membrane, and their diameter ranges from nanometer to micrometer scale. The formation of small ectosomes depends on endosomal sorting complexes required for transport (ESCRT) components governing membrane-budding and abscission and on the actin cytoskeleton. Exosomes are strictly nanosized vesicles that form upon budding of endosomal membranes, creating so-called intraluminal vesicles (ILVs) located inside MVEs. The fusion of MVEs with the plasma membrane releases the ILVs into the extracellular space as ‘exosomes’. Exosome biogenesis also depends on ESCRT and additional non-ESCRT components including ceramide, the PDZ domain-containing protein syntenin and syndecans. Yet one cannot exclude these components also contribute to the formation of small ectosomes [80].

It has long been known that EVs carry many tetraspanins. Following the initial demonstrations that CD9, CD37, CD53, CD63, and CD82 are present on EV released by immune cells [81,82], numerous proteomic analyses uncovered the presence of various tetraspanins on EVs (compiled in Vesiclepedia, http://microvesicles.org) [83]. Among them, CD9, CD81, and CD63, are widely used as markers of EVs. Consistent with its presence on ILVs [74,81], CD63 may preferentially decorate exosomes although it may also be present on ectosomes. In contrast, CD9 and CD81, which are mainly expressed at the plasma membrane, decorate ectosomes [21,84]. These two tetraspanins are also present on midbody remnants and migrasomes [29,34], which are released in the extracellular medium [85-87].

Several non-exclusive hypotheses have been explored regarding the roles of tetraspanins in EVs: (1) regulation of EV formation; (2) regulation of EV functional properties, including uptake and fusion; and (3) regulation of EV composition. However, current research has produced contradictory results, and a unified understanding of the function of tetraspanins in EVs remains elusive.

Some studies suggest that altering the expression of tetraspanins like CD9 or CD63 can affect the extent of EV release [88-91]. However, other studies found no such effects [22,84,92-95]. Moreover, the results are sometimes contradictory; for instance, CD63 was reported to positively influence EV release in some studies [88] but negatively in others [91]. These discrepancies suggest that tetraspanins are not essential components of the machinery responsible for EV formation. It is possible that the impact of these tetraspanins on EV release is dependent on the cellular context. In this regard, it was shown that the depletion of CD63 in melanocytes resulted in the formation of MVEs with lower number of ILVs [74]. This may be specific for this cell type and related to the process of melanogenesis, which occurs in LRO and depends on ILVs, since there was no modification of ILVs number in the absence of CD63 in another cell line [95]. The observed discrepancies might also stem from variations in isolation protocols or other technical differences, such as methods of quantification, purification, or characterization of EVs.

Given the influence of tetraspanins on specific cellular functions and their known interactions with various proteins, researchers have examined whether tetraspanins can regulate the sorting of particular proteins into EVs and the functions of EVs. For example, CD9 has been shown to interact with the ectopeptidase CD10/neprilysin and to enhance its release via EVs [96]. Additionally, overexpression of CD9 or CD82 was found to reduce Wnt/β-catenin signaling by increasing the release of β-catenin through EVs, an effect partly dependent on E-cadherin expression [97]. Similarly, Tspan8 expression in breast cancer cells was associated with increased levels of E-cadherin in EVs, probably as a consequence of the regulation of E-cadherin expression by Tspan8 [98]. In pancreatic cancer cells, Tspan8 transfection resulted in the targeting of the integrin α4 to EVs, enabling these EVs to bind to endothelial cells via VCAM1 and promoting angiogenesis [99].

Other studies have found that tetraspanins affect the functionality of EVs, which could indirectly suggest that they regulate EV composition or influence the function of specific components. For instance, Tspan8 transfection in breast cancer cells was shown to enhance EV attachment to recipient cells and stimulate their migration [100]. In another study, pre-incubation of cancer-associated fibroblast-derived EVs with a CD9 monoclonal antibody reduced their uptake by pancreatic cancer cells and their ability to stimulate cell migration [101]. However, it remains unclear whether this effect directly reflects the role of CD9 in this model. Elsewhere, transfection of CD9 in colon cancer cells was found to decrease EV uptake by recipient cells, potentially by regulating the interaction between EV-associated ADAM17 and the integrin α5β1 [102]. Yet, recent quantitative approaches suggest that the absence of CD9 or CD63 does not significantly alter the delivery of EV contents to recipient cells, indicating no major change in the uptake or internalization mechanisms [94].

Several studies have used proteomic approaches to analyze the composition of EVs in the absence of specific tetraspanins [22,90,93,95,103,104]. One study compared the composition of EVs secreted by lymphoblasts from wild type (WT) and CD81 KO mice [103]. Given that CD81 plays a crucial role in the immune system, particularly in the humoral response [105], the observed alterations in EV composition might stem from changes in lymphocyte subpopulations. This could explain the observed increase in CD4 and the decrease in B-cell molecules, such as CD20, immunoglobulins, or MHC class II antigens. Other studies examined EV protein composition in cell lines deficient in CD9, CD81, or CD63 [22,90,93,95,104]. While these studies reported several changes—both increases and decreases in protein levels—there were few commonalities, and some results were even contradictory. For instance, the absence of CD9 affected the secretion of proteasome subunits differently across studies, with PSMB7 showing a decrease in one study, an increase in another, and no change in a third [22,90,93]. These changes, along with changes in metabolic enzymes or ribosomal/RNA-binding proteins, are unexpected since these proteins are not typically found in tetraspanin-decorated EVs [106]. This suggests that some observed changes might be indirect effects of tetraspanin absence, or perhaps directly related to the experimental or analytical methods used, reflecting variations in the methods for isolation, quantification or characterization of EVs used in the different studies. Overall, these findings suggest that CD9, CD81, and CD63 do not play a general role in sorting protein cargos into EVs. However, it is possible that certain effects of tetraspanin deletion went undetected due to potential functional redundancies among tetraspanins. In this regard, one study [22] found that two immunoglobulin domain proteins, CD9P-1/EWI-F (encoded by the PTGFRN gene) and EWI-2 (encoded by the IGSF8 gene), which directly associate with both CD9 and CD81 [3-5], were decreased in EVs lacking both tetraspanins, but not only one of them. While the reduction in EWI-2 was primarily due to a lower cellular level of the protein, the decrease in CD9P-1 was not, indicating that CD9 and CD81 contribute to its sorting into EVs. In contrast, β-catenin levels in EVs were unaffected by the absence of CD63, CD9, CD81, or both CD9 and CD81. This suggests that the CD9-dependent regulation of β-catenin targeting to EVs, previously reported [97], is not a general phenomenon.

Tspan6 has gained significant attention recently for its role in EV formation. Similar to CD63 and CD81 [107,108], Tspan6 interacts directly with the scaffolding PDZ protein syntenin [24,25] that plays an important role in the formation of EVs [92,109]. In HEK 293 cells, Tspan6 primarily resides after transfection in LAMP1-positive organelles, akin to CD63, making it a resident of LROs. Overexpression of Tspan6 in these cells was shown to alter endosomal structures and enhance EV release, an effect not observed in syntenin-deficient cells [24]. However, contrasting results were observed in breast cancer MCF7 cells, where Tspan6 limited EV release by promoting syndecan-4-dependent syntenin lysosomal degradation [25]. Further complicating the picture, transfection of Tspan6 in another breast cancer cell line did not alter EV release [110], and organoids derived from APC mutant Tspan6 KO mice produced similar amounts of EVs as those expressing Tspan6 [111]. Importantly, in this latter study, Tspan6 was found to interact with TGFα via syntenin, and its absence led to increased TGFα release in EVs, making organoid growth independent of exogenous EGF and enhancing tumor formation in APC mutant animals.

Tetraspanins have long been recognized for their physical and functional interactions with lipids, such as cholesterol and gangliosides like GM3 [112-117]. The crystal structure of CD81, generated in the presence of cholesterol, revealed the presence of this lipid in the central cavity formed by the transmembrane domains [9]. Several studies have explored how tetraspanin absence affects the lipid composition of EVs [90,95,110]. Following the discovery that Tspan6 enhances the chemoattractive potential of breast cancer cells for B lymphocytes in an EV-dependent manner, it was shown that this effect relied on the function of liver X receptors, which are nuclear receptors for oxysterols, in B cells. Tspan6 expression was associated with increased levels of oxysterol species (25-HC and 27-HC) in EVs as well as in crude cellular membranes, without any change in the total cell oxysterol levels suggesting that it participates to the transport of oxysterols [110]. Importantly, a mutation in Tspan6 predicted to disrupt oxysterol binding abolished its ability to elevate oxysterol levels in EVs and stimulate B-cell migration.

Molecular modeling also suggests that CD63 could accommodate cholesterol within an intramembrane cavity [95]. Although the absence of CD63 did not affect the uptake of exogenous cholesterol from LDL, it altered the intracellular trafficking of endogenous cholesterol. Specifically, CD63 deficiency resulted in reduced labeling of ILVs in MVEs by the D4 fragment of the cholesterol-binding toxin perfringolysin O, alongside increased Golgi apparatus labeling. EVs from CD63-KO cells also exhibited reduced D4 labeling, which was fully reversed by reintroducing CD63 but not by expressing a mutant CD63 predicted not to accommodate cholesterol in its intramembrane cavity. As not all approaches indicated a decreased cholesterol level in EVs released by CD63 KO cells, it is possible that D4 detects discrete cholesterol pools modified by the absence of CD63. Importantly, EVs from CD63 KO cells loaded with a fluorescent analogue of cholesterol delivered less of this fluorescent probe to recipient cells than EVs from WT cells.

Tetraspanins have long been associated with a wide range of cellular functions and molecular interactions. Their ability to directly interact with partner proteins has led to the emerging concept of tetraspanins as regulators of cell membrane compartmentalization. Recent structural studies of full-length tetraspanins provide insight into their localization in and their impact on curved and tubular cellular structures. Small EVs represent a typical example of curved structures enriched in several tetraspanins. Early hypotheses suggesting that discrete tetraspanins play a role in EV formation or protein composition have been challenged by conflicting experimental data. However, recent studies indicate a possible role for tetraspanins in the trafficking and/or organization of sterols within EVs. These findings open new avenues for research into how tetraspanins influence the lipid composition of EVs.

Perspectives

  • Many cells release extracellular vesicles (EVs) in their environment, which often bear tetraspanins such as CD9, CD81, and CD63. These three tetraspanins are commonly used as markers of EVs although some EVs do not carry any of them.

  • The ability of tetraspanins to regulate the trafficking of associated partner proteins and to have an impact on curved or tubular structures, possibly due to their conical structure, may suggest a role in the formation or protein composition of EVs. However, these hypotheses have been confounded by contradictory results.

  • Future studies could revisit these hypotheses by analyzing discrete subtypes of EVs and could seek to confirm the impact of tetraspanins on the composition or organization of lipids in EVs.

ER declares that there are no competing interests associated with the manuscript. PZ and CT are inventors on respectively 1 and 2 filed patents on the therapeutic use of EVs.

This work was funded by core fundings from the Institut National de la Santé et de la Recherche Médicale (INSERM) and the Institut Curie, by a grant from the Agence Nationale de la Recherche (ANR-18-CE13-0017-02) and by the internal funds of the KU Leuven (C14/20/105).

Writing ȓ original draft: ER; Writing – review & editing: CT and PZ

We thank Dr. Christel Brou and Pierre-Emmanuel Milhiet for providing images included in Figure 2. We also thank Dr. Brou for critical reading of the manuscript. A short review as this one is inevitably incomplete. We apologize to all those colleagues whose studies could not be covered due to space limitations. The original references can be found in the different reviews we refer to.

ESCRT

endosomal sorting complexes required for transport

EVs

extracellular vesicles

ILVs

intraluminal vesicles

LEL

large extracellular loop

LRO

lysosome-related organelle

MVEs

multivesicular endosomes/bodies

PRPH2

peripherin-2

ROM1

rod outer segment membrane protein 1

1
Huang
,
S.
,
Yuan
,
S.
,
Dong
,
M.
,
Su
,
J.
,
Yu
,
C.
,
Shen
,
Y.
et al.
(
2005
)
The phylogenetic analysis of tetraspanins projects the evolution of cell-cell interactions from unicellular to multicellular organisms
.
Genomics
86
,
674
684
https://doi.org/10.1016/j.ygeno.2005.08.004
2
Garcia-España
,
A.
,
Chung
,
P.J.
,
Sarkar
,
I.N.
,
Stiner
,
E.
,
Sun
,
T.T.
and
Desalle
,
R
. (
2008
)
Appearance of new tetraspanin genes during vertebrate evolution
.
Genomics
91
,
326
334
https://doi.org/10.1016/j.ygeno.2007.12.005
3
Hemler
,
M.E
. (
2005
)
Tetraspanin functions and associated microdomains
.
Nat. Rev. Mol. Cell Biol.
6
,
801
811
https://doi.org/10.1038/nrm1736
4
Charrin
,
S.
,
Le Naour
,
F.
,
Silvie
,
O.
,
Milhiet
,
P.E.
,
Boucheix
,
C.
and
Rubinstein
,
E
. (
2009
)
Lateral organization of membrane proteins: tetraspanins spin their web
.
Biochem. J.
420
,
133
154
https://doi.org/10.1042/BJ20082422
5
Charrin
,
S.
,
Jouannet
,
S.
,
Boucheix
,
C.
and
Rubinstein
,
E
. (
2014
)
Tetraspanins at a glance
.
J. Cell. Sci.
127
,
3641
3648
https://doi.org/10.1242/jcs.154906
6
Florin
,
L.
and
Lang
,
T
. (
2018
)
Tetraspanin assemblies in virus infection
.
Front. Immunol.
9
, 1140 https://doi.org/10.3389/fimmu.2018.01140
7
Hemler
,
M.E
. (
2014
)
Tetraspanin proteins promote multiple cancer stages
.
Nat. Rev. Cancer
14
,
49
60
https://doi.org/10.1038/nrc3640
8
Susa
,
K.J.
,
Kruse
,
A.C.
and
Blacklow
,
S.C
. (
2024
)
Tetraspanins: structure, dynamics, and principles of partner-protein recognition
.
Trends Cell Biol.
34
,
509
522
https://doi.org/10.1016/j.tcb.2023.09.003
9
Zimmerman
,
B.
,
Kelly
,
B.
,
McMillan
,
B.J.
,
Seegar
,
T.C.M.
,
Dror
,
R.O.
,
Kruse
,
A.C.
, et al.
(
2016
)
Crystal structure of a full-length human tetraspanin reveals a cholesterol-binding pocket
.
Cell
167
,
1041
1051
https://doi.org/10.1016/j.cell.2016.09.056
10
Umeda
,
R.
,
Satouh
,
Y.
,
Takemoto
,
M.
,
Nakada-Nakura
,
Y.
,
Liu
,
K.
,
Yokoyama
,
T.
et al.
(
2020
)
Structural insights into tetraspanin CD9 function
.
Nat. Commun.
11
,
1606
https://doi.org/10.1038/s41467-020-15459-7
11
Yang
,
Y.
,
Liu
,
X.R.
,
Greenberg
,
Z.J.
,
Zhou
,
F.
,
He
,
P.
,
Fan
,
L.
et al.
(
2020
)
Open conformation of tetraspanins shapes interaction partner networks on cell membranes
.
EMBO J.
39
, e105246 https://doi.org/10.15252/embj.2020105246
12
Oosterheert
,
W.
,
Xenaki
,
K.T.
,
Neviani
,
V.
,
Pos
,
W.
,
Doulkeridou
,
S.
,
Manshande
,
J.
et al.
(
2020
)
Implications for tetraspanin-enriched microdomain assembly based on structures of CD9 with EWI-F
.
Life Sci. Alliance
3
, e202000883 https://doi.org/10.26508/lsa.202000883
13
Susa
,
K.J.
,
Rawson
,
S.
,
Kruse
,
A.C.
and
Blacklow
,
S.C
. (
2021
)
Cryo-EM structure of the B cell co-receptor CD19 bound to the tetraspanin CD81
.
Science
371
,
300
305
https://doi.org/10.1126/science.abd9836
14
El Mazouni
,
D.
and
Gros
,
P
. (
2022
)
Cryo-EM structures of peripherin-2 and ROM1 suggest multiple roles in photoreceptor membrane morphogenesis
.
Sci. Adv.
8
, eadd3677 https://doi.org/10.1126/sciadv.add3677
15
Lipper
,
C.H.
,
Egan
,
E.D.
,
Gabriel
,
K.H.
and
Blacklow
,
S.C
. (
2023
)
Structural basis for membrane-proximal proteolysis of substrates by ADAM10
.
Cell
186
,
3632
3641
https://doi.org/10.1016/j.cell.2023.06.026
16
Kitadokoro
,
K.
,
Bordo
,
D.
,
Galli
,
G.
,
Petracca
,
R.
,
Falugi
,
F.
,
Abrignani
,
S.
et al.
(
2001
)
CD81 extracellular domain 3D structure: insight into the tetraspanin superfamily structural motifs
.
EMBO J.
20
,
12
18
https://doi.org/10.1093/emboj/20.1.12
17
Kitadokoro
,
K.
,
Ponassi
,
M.
,
Galli
,
G.
,
Petracca
,
R.
,
Falugi
,
F.
,
Grandi
,
G.
et al.
(
2002
)
Subunit association and conformational flexibility in the head subdomain of human CD81 large extracellular loop
.
Biol. Chem.
383
,
1447
1452
https://doi.org/10.1515/BC.2002.164
18
Cunha
,
E.S.
,
Sfriso
,
P.
,
Rojas
,
A.L.
,
Roversi
,
P.
,
Hospital
,
A.
,
Orozco
,
M.
, et al.
(
2017
)
Mechanism of structural tuning of the hepatitis C virus human cellular receptor CD81 large extracellular loop
.
Structure
25
,
53
65
https://doi.org/10.1016/j.str.2016.11.003
19
Fénéant
,
L.
,
Levy
,
S.
and
Cocquerel
,
L
. (
2014
)
CD81 and hepatitis C virus (HCV) infection
.
Viruses
6
,
535
572
https://doi.org/10.3390/v6020535
20
Yáñez-Mó
,
M.
,
Tejedor
,
R.
,
Rousselle
,
P.
and
Sánchez -Madrid
,
F
. (
2001
)
Tetraspanins in intercellular adhesion of polarized epithelial cells: spatial and functional relationship to integrins and cadherins
.
J. Cell. Sci.
114
,
577
587
https://doi.org/10.1242/jcs.114.3.577
21
Mathieu
,
M.
,
Névo
,
N.
,
Jouve
,
M.
,
Valenzuela
,
J.I.
,
Maurin
,
M.
,
Verweij
,
F.J.
et al.
(
2021
)
Specificities of exosome versus small ectosome secretion revealed by live intracellular tracking of CD63 and CD9
.
Nat. Commun.
12
,
4389
https://doi.org/10.1038/s41467-021-24384-2
22
Fan
,
Y.
,
Pionneau
,
C.
,
Cocozza
,
F.
,
Boëlle
,
P.Y.
,
Chardonnet
,
S.
,
Charrin
,
S.
et al.
(
2023
)
Differential proteomics argues against a general role for CD9, CD81 or CD63 in the sorting of proteins into extracellular vesicles
.
J. Extracell. Vesicles
12
, 12352 https://doi.org/10.1002/jev2.12352
23
Pols
,
M.S.
and
Klumperman
,
J
. (
2009
)
Trafficking and function of the tetraspanin CD63
.
Exp. Cell Res.
315
,
1584
1592
https://doi.org/10.1016/j.yexcr.2008.09.020
24
Guix
,
F.X.
,
Sannerud
,
R.
,
Berditchevski
,
F.
,
Arranz
,
A.M.
,
Horré
,
K.
,
Snellinx
,
A.
et al.
(
2017
)
Tetraspanin 6: a pivotal protein of the multiple vesicular body determining exosome release and lysosomal degradation of amyloid precursor protein fragments
.
Mol. Neurodegener.
12
,
25
,
25
https://doi.org/10.1186/s13024-017-0165-0
25
Ghossoub
,
R.
,
Chéry
,
M.
,
Audebert
,
S.
,
Leblanc
,
R.
,
Egea-Jimenez
,
A.L.
,
Lembo
,
F.
et al.
(
2020
)
Tetraspanin-6 negatively regulates exosome production
.
Proc. Natl. Acad. Sci. U.S.A.
117
,
5913
5922
https://doi.org/10.1073/pnas.1922447117
26
Zhang
,
X.A.
and
Huang
,
C
. (
2012
)
Tetraspanins and cell membrane tubular structures
.
Cell. Mol. Life Sci.
69
,
2843
2852
https://doi.org/10.1007/s00018-012-0954-0
27
Dharan
,
R.
and
Sorkin
,
R
. (
2024
)
Tetraspanin proteins in membrane remodeling processes
.
J. Cell. Sci.
137
, jcs261532 https://doi.org/10.1242/jcs.261532
28
Notario Manzano
,
R.
,
Chaze
,
T.
,
Rubinstein
,
E.
,
Penard
,
E.
,
Matondo
,
M.
,
Zurzolo
,
C.
et al.
(
2024
)
Proteomic landscape of tunneling nanotubes reveals CD9 and CD81 tetraspanins as key regulators
.
Elife
13
, RP99172 https://doi.org/10.7554/eLife.99172
29
Presle
,
A.
,
Frémont
,
S.
,
Salles
,
A.
,
Commere
,
P.H.
,
Sassoon
,
N.
,
Berlioz-Torrent
,
C.
, et al.
(
2021
)
The viral restriction factor tetherin/BST2 tethers cytokinetic midbody remnants to the cell surface
.
Curr. Biol.
31
,
2203
2213
https://doi.org/10.1016/j.cub.2021.02.039
30
Dahmane
,
S.
,
Doucet
,
C.
,
Le Gall
,
A.
,
Chamontin
,
C.
,
Dosset
,
P.
,
Murcy
,
F.
et al.
(
2019
)
Nanoscale organization of tetraspanins during HIV-1 budding by correlative dSTORM/AFM
.
Nanoscale
11
,
6036
6044
https://doi.org/10.1039/c8nr07269h
31
Runge
,
K.E.
,
Evans
,
J.E.
,
He
,
Z.Y.
,
Gupta
,
S.
,
McDonald
,
K.L.
,
Stahlberg
,
H.
et al.
(
2007
)
Oocyte CD9 is enriched on the microvillar membrane and required for normal microvillar shape and distribution
.
Dev. Biol.
304
,
317
325
https://doi.org/10.1016/j.ydbio.2006.12.041
32
Huang
,
Y.
and
Yu
,
L
. (
2022
)
Tetraspanin-enriched microdomains: the building blocks of migrasomes
.
Cell Insight
1
, 100003 https://doi.org/10.1016/j.cellin.2021.100003
33
Charrin
,
S.
,
Palmulli
,
R.
,
Billard
,
M.
,
Clay
,
D.
,
Boucheix
,
C.
,
Van Niel
,
G.
et al.
(
2020
)
Rapid isolation of rare isotype-switched hybridoma variants: application to the generation of IgG2a and IgG2b MAb to CD63, a late endosome and exosome marker
.
Antibodies
9
, 29 https://doi.org/10.3390/antib9030029
34
Huang
,
Y.
,
Zucker
,
B.
,
Zhang
,
S.
,
Elias
,
S.
,
Zhu
,
Y.
,
Chen
,
H.
et al.
(
2019
)
Migrasome formation is mediated by assembly of micron-scale tetraspanin macrodomains
.
Nat. Cell Biol.
21
,
991
1002
https://doi.org/10.1038/s41556-019-0367-5
35
Andreu
,
Z.
and
Yáñez-Mó
,
M
. (
2014
)
Tetraspanins in extracellular vesicle formation and function
.
Front. Immunol.
5
, 442 https://doi.org/10.3389/fimmu.2014.00442
36
Pelchen-Matthews
,
A.
,
Kramer
,
B.
and
Marsh
,
M
. (
2003
)
Infectious HIV-1 assembles in late endosomes in primary macrophages
.
J. Cell Biol.
162
,
443
455
https://doi.org/10.1083/jcb.200304008
37
Chertova
,
E.
,
Chertov
,
O.
,
Coren
,
L.V.
,
Roser
,
J.D.
,
Trubey
,
C.M.
,
Bess
,
J.W.
Jr
et al.
(
2006
)
Proteomic and biochemical analysis of purified human immunodeficiency virus type 1 produced from infected monocyte-derived macrophages
.
J. Virol.
80
,
9039
9052
https://doi.org/10.1128/JVI.01013-06
38
Booth
,
A.M.
,
Fang
,
Y.
,
Fallon
,
J.K.
,
Yang
,
J.M.
,
Hildreth
,
J.E.K.
and
Gould
,
S.J
. (
2006
)
Exosomes and HIV gag bud from endosome-like domains of the T cell plasma membrane
.
J. Cell Biol.
172
,
923
935
https://doi.org/10.1083/jcb.200508014
39
Jolly
,
C.
and
Sattentau
,
Q.J
. (
2007
)
Human immunodeficiency virus type 1 assembly, budding, and cell-cell spread in T cells take place in tetraspanin-enriched plasma membrane domains
.
J. Virol.
81
,
7873
7884
https://doi.org/10.1128/JVI.01845-06
40
Shaw
,
M.L.
,
Stone
,
K.L.
,
Colangelo
,
C.M.
,
Gulcicek
,
E.E.
and
Palese
,
P
. (
2008
)
Cellular proteins in influenza virus particles
.
PLOS Pathog.
4
, e1000085 https://doi.org/10.1371/journal.ppat.1000085
41
Martin-Jaular
,
L.
,
Nevo
,
N.
,
Schessner
,
J.P.
,
Tkach
,
M.
,
Jouve
,
M.
,
Dingli
,
F.
et al.
(
2021
)
Unbiased proteomic profiling of host cell extracellular vesicle composition and dynamics upon HIV-1 infection
.
EMBO J.
40
, e105492 https://doi.org/10.15252/embj.2020105492
42
He
,
J.
,
Sun
,
E.
,
Bujny
,
M.V.
,
Kim
,
D.
,
Davidson
,
M.W.
and
Zhuang
,
X
. (
2013
)
Dual function of CD81 in influenza virus uncoating and budding
.
PLOS Pathog.
9
, e1003701 https://doi.org/10.1371/journal.ppat.1003701
43
Dharan
,
R.
,
Goren
,
S.
,
Cheppali
,
S.K.
,
Shendrik
,
P.
,
Brand
,
G.
,
Vaknin
,
A.
et al.
(
2022
)
Transmembrane proteins tetraspanin 4 and CD9 sense membrane curvature
.
Proc. Natl. Acad. Sci. U.S.A.
119
, e2208993119 https://doi.org/10.1073/pnas.2208993119
44
Le Naour
,
F.
,
Rubinstein
,
E.
,
Jasmin
,
C.
,
Prenant
,
M.
and
Boucheix
,
C
. (
2000
)
Severely reduced female fertility in CD9-deficient mice
.
Science
287
,
319
321
https://doi.org/10.1126/science.287.5451.319
45
Dharan
,
R.
,
Huang
,
Y.
,
Cheppali
,
S.K.
,
Goren
,
S.
,
Shendrik
,
P.
,
Wang
,
W.
et al.
(
2023
)
Tetraspanin 4 stabilizes membrane swellings and facilitates their maturation into migrasomes
.
Nat. Commun.
14
,
1037
https://doi.org/10.1038/s41467-023-36596-9
46
Rubinstein
,
E.
,
Le Naour
,
F.
,
Lagaudrière-Gesbert
,
C.
,
Billard
,
M.
,
Conjeaud
,
H.
and
Boucheix
,
C
. (
1996
)
CD9, CD63, CD81, and CD82 are components of a surface tetraspan network connected to HLA-DR and VLA integrins
.
Eur. J. Immunol.
26
,
2657
2665
https://doi.org/10.1002/eji.1830261117
47
Claas
,
C.
,
Stipp
,
C.S.
and
Hemler
,
M.E
. (
2001
)
Evaluation of prototype transmembrane 4 superfamily protein complexes and their relation to lipid rafts
.
J. Biol. Chem.
276
,
7974
7984
https://doi.org/10.1074/jbc.M008650200
48
Berditchevski
,
F.
,
Odintsova
,
E.
,
Sawada
,
S.
and
Gilbert
,
E
. (
2002
)
Expression of the palmitoylation-deficient CD151 weakens the association of alpha 3 beta 1 integrin with the tetraspanin-enriched microdomains and affects integrin-dependent signaling
.
J. Biol. Chem.
277
,
36991
37000
https://doi.org/10.1074/jbc.M205265200
49
Yauch
,
R.L.
,
Berditchevski
,
F.
,
Harler
,
M.B.
,
Reichner
,
J.
and
Hemler
,
M.E
. (
1998
)
Highly stoichiometric, stable, and specific association of integrin α3β1 with CD151 provides a major link to phosphatidylinositol 4-kinase, and may regulate cell migration
.
Mol. Biol. Cell
9
,
2751
2765
https://doi.org/10.1091/mbc.9.10.2751
50
Horváth
,
G.
,
Serru
,
V.
,
Clay
,
D.
,
Billard
,
M.
,
Boucheix
,
C.
and
Rubinstein
,
E
. (
1998
)
CD19 is linked to the integrin-associated tetraspans CD9, CD81, and CD82
.
J. Biol. Chem.
273
,
30537
30543
https://doi.org/10.1074/jbc.273.46.30537
51
Serru
,
V.
,
Le Naour
,
F.
,
Billard
,
M.
,
Azorsa
,
D.O.
,
Lanza
,
F.
,
Boucheix
,
C.
et al.
(
1999
)
Selective tetraspan-integrin complexes (CD81/α4β1, CD151/α3β1, CD151/α6β1) under conditions disrupting tetraspan interactions
.
Biochem. J.
340
,
103
111
https://doi.org/10.1042/bj3400103
52
Charrin
,
S.
,
Manié
,
S.
,
Billard
,
M.
,
Ashman
,
L.
,
Gerlier
,
D.
,
Boucheix
,
C.
et al.
(
2003
)
Multiple levels of interactions within the tetraspanin web
.
Biochem. Biophys. Res. Commun.
304
,
107
112
https://doi.org/10.1016/s0006-291x(03)00545-x
53
Winterwood
,
N.E.
,
Varzavand
,
A.
,
Meland
,
M.N.
,
Ashman
,
L.K.
and
Stipp
,
C.S
. (
2006
)
A critical role for tetraspanin CD151 in α3β1 and α6β4 integrin-dependent tumor cell functions on laminin-5
.
Mol. Biol. Cell
17
,
2707
2721
https://doi.org/10.1091/mbc.e05-11-1042
54
Lammerding
,
J.
,
Kazarov
,
A.R.
,
Huang
,
H.
,
Lee
,
R.T.
and
Hemler
,
M.E
. (
2003
)
Tetraspanin CD151 regulates α6β1 integrin adhesion strengthening
.
Proc. Natl. Acad. Sci. U.S.A.
100
,
7616
7621
https://doi.org/10.1073/pnas.1337546100
55
Karamatic Crew
,
V.
,
Burton
,
N.
,
Kagan
,
A.
,
Green
,
C.A.
,
Levene
,
C.
,
Flinter
,
F.
et al.
(
2004
)
CD151, the first member of the tetraspanin (TM4) superfamily detected on erythrocytes, is essential for the correct assembly of human basement membranes in kidney and skin
.
Blood
104
,
2217
2223
https://doi.org/10.1182/blood-2004-04-1512
56
Rahmani
,
N.
,
Talebi
,
S.
,
Hoseini
,
R.
,
Asghari Kollahi
,
N.
and
Shojaei
,
A
. (
2022
)
New report of a different clinical presentation of CD151 splicing mutation (C.351+2T>C): could TSPAN11 be considered as a potential modifier gene for CD151
.
Mol. Syndromol.
13
,
212
220
https://doi.org/10.1159/000519633
57
Sachs
,
N.
,
Claessen
,
N.
,
Aten
,
J.
,
Kreft
,
M.
,
Teske
,
G.J.D.
,
Koeman
,
A.
, et al.
(
2012
)
Blood pressure influences end-stage renal disease of Cd151 knockout mice
.
J. Clin. Invest.
122
,
348
358
https://doi.org/10.1172/JCI58878
58
Shoham
,
T.
,
Rajapaksa
,
R.
,
Boucheix
,
C.
,
Rubinstein
,
E.
,
Poe
,
J.C.
,
Tedder
,
T.F.
et al.
(
2003
)
The tetraspanin CD81 regulates the expression of CD19 during B cell development in a postendoplasmic reticulum compartment
.
J. Immunol.
171
,
4062
4072
https://doi.org/10.4049/jimmunol.171.8.4062
59
Zelm
,
M.C.
,
Smet
,
J.
,
Adams
,
B.
,
Mascart
,
F.
,
Schandené
,
L.
,
Janssen
,
F.
, et al.
(
2010
)
CD81 gene defect in humans disrupts CD19 complex formation and leads to antibody deficiency
.
J. Clin. Invest.
120
,
1265
1274
https://doi.org/10.1172/JCI39748
60
Maecker
,
H.T.
and
Levy
,
S
. (
1997
)
Normal lymphocyte development but delayed humoral immune response in CD81-null mice
.
J. Exp. Med.
185
,
1505
1510
https://doi.org/10.1084/jem.185.8.1505
61
Mattila
,
P.K.
,
Feest
,
C.
,
Depoil
,
D.
,
Treanor
,
B.
,
Montaner
,
B.
,
Otipoby
,
K.L.
et al.
(
2013
)
The actin and tetraspanin networks organize receptor nanoclusters to regulate B cell receptor-mediated signaling
.
Immunity
38
,
461
474
https://doi.org/10.1016/j.immuni.2012.11.019
62
Saftig
,
P.
and
Lichtenthaler
,
S.F
. (
2015
)
The alpha secretase ADAM10: a metalloprotease with multiple functions in the brain
.
Prog. Neurobiol.
135
,
1
20
https://doi.org/10.1016/j.pneurobio.2015.10.003
63
Prox
,
J.
,
Willenbrock
,
M.
,
Weber
,
S.
,
Lehmann
,
T.
,
Schmidt-Arras
,
D.
,
Schwanbeck
,
R.
et al.
(
2012
)
Tetraspanin15 regulates cellular trafficking and activity of the ectodomain sheddase ADAM10
.
Cell. Mol. Life Sci.
69
,
2919
2932
https://doi.org/10.1007/s00018-012-0960-2
64
Dornier
,
E.
,
Coumailleau
,
F.
,
Ottavi
,
J.F.
,
Moretti
,
J.
,
Boucheix
,
C.
,
Mauduit
,
P.
et al.
(
2012
)
TspanC8 tetraspanins regulate ADAM10/Kuzbanian trafficking and promote notch activation in flies and mammals
.
J. Cell Biol.
199
,
481
496
https://doi.org/10.1083/jcb.201201133
65
Haining
,
E.J.
,
Yang
,
J.
,
Bailey
,
R.L.
,
Khan
,
K.
,
Collier
,
R.
,
Tsai
,
S.
et al.
(
2012
)
The TspanC8 subgroup of tetraspanins interacts with a disintegrin and metalloprotease 10 (ADAM10) and regulates its maturation and cell surface expression
.
J. Biol. Chem.
287
,
39753
39765
https://doi.org/10.1074/jbc.M112.416503
66
Jouannet
,
S.
,
Saint-Pol
,
J.
,
Fernandez
,
L.
,
Nguyen
,
V.
,
Charrin
,
S.
,
Boucheix
,
C.
et al.
(
2016
)
TspanC8 tetraspanins differentially regulate the cleavage of ADAM10 substrates, notch activation and ADAM10 membrane compartmentalization
.
Cell. Mol. Life Sci.
73
,
1895
1915
https://doi.org/10.1007/s00018-015-2111-z
67
Noy
,
P.J.
,
Yang
,
J.
,
Reyat
,
J.S.
,
Matthews
,
A.L.
,
Charlton
,
A.E.
,
Furmston
,
J.
et al.
(
2016
)
TspanC8 tetraspanins and a disintegrin and metalloprotease 10 (ADAM10) interact via their extracellular regions: evidence for distint binding mechanisms for different TspanC8 proteins
.
J. Biol. Chem.
291
,
3145
3157
https://doi.org/10.1074/jbc.M115.703058
68
Eschenbrenner
,
E.
,
Jouannet
,
S.
,
Clay
,
D.
,
Chaker
,
J.
,
Boucheix
,
C.
,
Brou
,
C.
et al.
(
2020
)
TspanC8 tetraspanins differentially regulate ADAM10 endocytosis and half-life
.
Life Sci. Alliance
3
, e201900444 https://doi.org/10.26508/lsa.201900444
69
Dunn
,
C.D.
,
Sulis
,
M.L.
,
Ferrando
,
A.A.
and
Greenwald
,
I
. (
2010
)
A conserved tetraspanin subfamily promotes notch signaling in Caenorhabditis elegans and in human cells
.
Proc. Natl. Acad. Sci. U.S.A.
107
,
5907
5912
https://doi.org/10.1073/pnas.1001647107
70
Wang
,
L.
,
Liu
,
Z.
,
Shi
,
H.
and
Liu
,
J
. (
2017
)
Two paralogous tetraspanins TSP-12 and TSP-14 function with the ADAM10 metalloprotease SUP-17 to promote BMP signaling in Caenorhabditis elegans
.
PLOS Genet.
13
, e1006568 https://doi.org/10.1371/journal.pgen.1006568
71
Duffield
,
A.
,
Kamsteeg
,
E.J.
,
Brown
,
A.N.
,
Pagel
,
P.
and
Caplan
,
M.J
. (
2003
)
The tetraspanin CD63 enhances the internalization of the H,K-ATPase β-subunit
.
Proc. Natl. Acad. Sci. U.S.A.
100
,
15560
15565
https://doi.org/10.1073/pnas.2536699100
72
Codina
,
J.
,
Li
,
J.
and
Dubose
,
T.D.
Jr
. (
2005
)
CD63 interacts with the carboxy terminus of the colonic H+-K+-ATPase to increase plasma membrane localization and 86Rb+ uptake
.
Am. J. Physiol., Cell Physiol.
288
,
C1279
C1286
https://doi.org/10.1152/ajpcell.00463.2004
73
Flannery
,
A.R.
,
Czibener
,
C.
and
Andrews
,
N.W
. (
2010
)
Palmitoylation-dependent association with CD63 targets the Ca2+ sensor synaptotagmin VII to lysosomes
.
J. Cell Biol.
191
,
599
613
https://doi.org/10.1083/jcb.201003021
74
Niel
,
G.
Charrin
,
S.
,
Simoes
,
S.
,
Romao
,
M.
,
Rochin
,
L.
,
Saftig
,
P.
et al.
(
2011
)
The tetraspanin CD63 regulates ESCRT-independent and -dependent endosomal sorting during melanogenesis
.
Dev. Cell
21
,
708
721
https://doi.org/10.1016/j.devcel.2011.08.019
75
Yoshida
,
T.
,
Kawano
,
Y.
,
Sato
,
K.
,
Ando
,
Y.
,
Aoki
,
J.
,
Miura
,
Y.
et al.
(
2008
)
A CD63 mutant inhibits T-cell tropic human immunodeficiency virus type 1 entry by disrupting CXCR4 trafficking to the plasma membrane
.
Traffic
9
,
540
558
https://doi.org/10.1111/j.1600-0854.2007.00700.x
76
Takino
,
T.
,
Miyamori
,
H.
,
Kawaguchi
,
N.
,
Uekita
,
T.
,
Seiki
,
M.
and
Sato
,
H
. (
2003
)
Tetraspanin CD63 promotes targeting and lysosomal proteolysis of membrane-type 1 matrix metalloproteinase
.
Biochem. Biophys. Res. Commun.
304
,
160
166
https://doi.org/10.1016/s0006-291x(03)00544-8
77
Bassani
,
S.
,
Cingolani
,
L.A.
,
Valnegri
,
P.
,
Folci
,
A.
,
Zapata
,
J.
,
Gianfelice
,
A.
et al.
(
2012
)
The X-linked intellectual disability protein TSPAN7 regulates excitatory synapse development and AMPAR trafficking
.
Neuron
73
,
1143
1158
https://doi.org/10.1016/j.neuron.2012.01.021
78
Yates
,
A.G.
,
Pink
,
R.C.
,
Erdbrügger
,
U.
,
Siljander
,
P.R.M.
,
Dellar
,
E.R.
,
Pantazi
,
P.
et al.
(
2022
)
In sickness and in health: the functional role of extracellular vesicles in physiology and pathology in vivo: part I: health and normal physiology
.
J. Extracell. Vesicles
11
, e12190 https://doi.org/10.1002/jev2.12190
79
Yates
,
A.G.
,
Pink
,
R.C.
,
Erdbrügger
,
U.
,
Siljander
,
P.R.M.
,
Dellar
,
E.R.
,
Pantazi
,
P.
et al.
(
2022
)
In sickness and in health: the functional role of extracellular vesicles in physiology and pathology in vivo: part II: pathology
.
J. Extracell. Vesicles
11
, e12190 https://doi.org/10.1002/jev2.12190
80
Dixson
,
A.C.
,
Dawson
,
T.R.
,
Di Vizio
,
D.
and
Weaver
,
A.M
. (
2023
)
Context-specific regulation of extracellular vesicle biogenesis and cargo selection
.
Nat. Rev. Mol. Cell Biol.
24
,
454
476
https://doi.org/10.1038/s41580-023-00576-0
81
Escola
,
J.M.
,
Kleijmeer
,
M.J.
,
Stoorvogel
,
W.
,
Griffith
,
J.M.
,
Yoshie
,
O.
and
Geuze
,
H.J
. (
1998
)
Selective enrichment of tetraspan proteins on the internal vesicles of multivesicular endosomes and on exosomes secreted by human B-lymphocytes
.
J. Biol. Chem.
273
,
20121
20127
https://doi.org/10.1074/jbc.273.32.20121
82
Théry
,
C.
,
Regnault
,
A.
,
Garin
,
J.
,
Wolfers
,
J.
,
Zitvogel
,
L.
,
Ricciardi-Castagnoli
,
P.
et al.
(
1999
)
Molecular characterization of dendritic cell-derived exosomes. Selective accumulation of the heat shock protein Hsc73
.
J. Cell Biol.
147
,
599
610
https://doi.org/10.1083/jcb.147.3.599
83
Pathan
,
M.
,
Fonseka
,
P.
,
Chitti
,
S.V.
,
Kang
,
T.
,
Sanwlani
,
R.
,
Van Deun
,
J.
et al.
(
2019
)
Vesiclepedia 2019: a compendium of RNA, proteins, lipids and metabolites in extracellular vesicles
.
Nucleic Acids Res.
47
,
D516
D519
https://doi.org/10.1093/nar/gky1029
84
Fordjour
,
F.K.
,
Guo
,
C.
,
Ai
,
Y.
,
Daaboul
,
G.G.
and
Gould
,
S.J
. (
2022
)
A shared, stochastic pathway mediates exosome protein budding along plasma and endosome membranes
.
J. Biol. Chem.
298
, 102394 https://doi.org/10.1016/j.jbc.2022.102394
85
Rai
,
A.
,
Greening
,
D.W.
,
Xu
,
R.
,
Chen
,
M.
,
Suwakulsiri
,
W.
and
Simpson
,
R.J
. (
2021
)
Secreted midbody remnants are a class of extracellular vesicles molecularly distinct from exosomes and microparticles
.
Commun. Biol.
4
,
400
https://doi.org/10.1038/s42003-021-01882-z
86
Ma
,
L.
,
Li
,
Y.
,
Peng
,
J.
,
Wu
,
D.
,
Zhao
,
X.
,
Cui
,
Y.
et al.
(
2015
)
Discovery of the migrasome, an organelle mediating release of cytoplasmic contents during cell migration
.
Cell Res.
25
,
24
38
https://doi.org/10.1038/cr.2014.135
87
Patel
,
S.A.
,
Park
,
S.
,
Zhu
,
D.
,
Torr
,
E.E.
,
Dureke
,
A.G.
,
McIntyre
,
A.
, et al.
(
2024
)
Extracellular vesicles, including large translating vesicles called midbody remnants, are released during the cell cycle
.
MBoC mbc
E23-10-0384 https://doi.org/10.1091/mbc.E23-10-0384
88
Hurwitz
,
S.N.
,
Conlon
,
M.M.
,
Rider
,
M.A.
,
Brownstein
,
N.C.
and
Meckes
,
D.G.
Jr
. (
2016
)
Nanoparticle analysis sheds budding insights into genetic drivers of extracellular vesicle biogenesis
.
J. Extracell. Vesicles
5
,
31295
https://doi.org/10.3402/jev.v5.31295
89
Böker
,
K.O.
,
Lemus-Diaz
,
N.
,
Rinaldi Ferreira
,
R.
,
Schiller
,
L.
,
Schneider
,
S.
and
Gruber
,
J
. (
2018
)
The impact of the CD9 tetraspanin on lentivirus infectivity and exosome secretion
.
Mol. Ther.
26
,
634
647
https://doi.org/10.1016/j.ymthe.2017.11.008
90
Suárez
,
H.
,
Andreu
,
Z.
,
Mazzeo
,
C.
,
Toribio
,
V.
,
Pérez-Rivera
,
A.E.
,
López-Martín
,
S.
et al.
(
2021
)
CD9 inhibition reveals a functional connection of extracellular vesicle secretion with mitophagy in melanoma cells
.
J. Extracell. Vesicles
10
, e12082 https://doi.org/10.1002/jev2.12082
91
Dogrammatzis
,
C.
,
Saud
,
R.
,
Waisner
,
H.
,
Lasnier
,
S.
,
Suma
,
S.M.
,
Grieshaber
,
B.
et al.
(
2024
)
Tracing the STING exocytosis pathway during herpes viruses infection
.
MBio
15
, e00373-24 https://doi.org/10.1128/mbio.00373-24
92
Baietti
,
M.F.
,
Zhang
,
Z.
,
Mortier
,
E.
,
Melchior
,
A.
,
Degeest
,
G.
,
Geeraerts
,
A.
et al.
(
2012
)
Syndecan-syntenin-ALIX regulates the biogenesis of exosomes
.
Nat. Cell Biol.
14
,
677
685
https://doi.org/10.1038/ncb2502
93
Brzozowski
,
J.S.
,
Bond
,
D.R.
,
Jankowski
,
H.
,
Goldie
,
B.J.
,
Burchell
,
R.
,
Naudin
,
C.
et al.
(
2018
)
Extracellular vesicles with altered tetraspanin CD9 and CD151 levels confer increased prostate cell motility and invasion
.
Sci. Rep.
8
,
8822
https://doi.org/10.1038/s41598-018-27180-z
94
Tognoli
,
M.L.
,
Dancourt
,
J.
,
Bonsergent
,
E.
,
Palmulli
,
R.
,
Jong
,
O.G.
,
Van Niel
,
G.
, et al.
(
2023
)
Lack of involvement of CD63 and CD9 tetraspanins in the extracellular vesicle content delivery process
.
Commun. Biol.
6
,
532
https://doi.org/10.1038/s42003-023-04911-1
95
Palmulli
,
R.
,
Couty
,
M.
,
Piontek
,
M.C.
,
Ponnaiah
,
M.
,
Dingli
,
F.
,
Verweij
,
F.J.
et al.
(
2024
)
CD63 sorts cholesterol into endosomes for storage and distribution via exosomes
.
Nat. Cell Biol.
26
,
1093
1109
https://doi.org/10.1038/s41556-024-01432-9
96
Mazurov
,
D.
,
Barbashova
,
L.
and
Filatov
,
A
. (
2013
)
Tetraspanin protein CD9 interacts with metalloprotease CD10 and enhances its release via exosomes
.
FEBS J.
280
,
1200
1213
https://doi.org/10.1111/febs.12110
97
Chairoungdua
,
A.
,
Smith
,
D.L.
,
Pochard
,
P.
,
Hull
,
M.
and
Caplan
,
M.J
. (
2010
)
Exosome release of β-catenin: a novel mechanism that antagonizes Wnt signaling
.
J. Cell Biol.
190
,
1079
1091
https://doi.org/10.1083/jcb.201002049
98
Voglstaetter
,
M.
,
Thomsen
,
A.R.
,
Nouvel
,
J.
,
Koch
,
A.
,
Jank
,
P.
,
Navarro
,
E.G.
et al.
(
2019
)
Tspan8 is expressed in breast cancer and regulates E-cadherin/catenin signalling and metastasis accompanied by increased circulating extracellular vesicles
.
J. Pathol.
248
,
421
437
https://doi.org/10.1002/path.5281
99
Nazarenko
,
I.
,
Rana
,
S.
,
Baumann
,
A.
,
McAlear
,
J.
,
Hellwig
,
A.
,
Trendelenburg
,
M.
et al.
(
2010
)
Cell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation
.
Cancer Res.
70
,
1668
1678
https://doi.org/10.1158/0008-5472.CAN-09-2470
100
Wang
,
T.
,
Wang
,
X.
,
Wang
,
H.
,
Li
,
L.
,
Zhang
,
C.
,
Xiang
,
R.
et al.
(
2021
)
High TSPAN8 expression in epithelial cancer cell-derived small extracellular vesicles promote confined diffusion and pronounced uptake
.
J. Extracell. Vesicles
10
, e12167 https://doi.org/10.1002/jev2.12167
101
Nigri
,
J.
,
Leca
,
J.
,
Tubiana
,
S.S.
,
Finetti
,
P.
,
Guillaumond
,
F.
,
Martinez
,
S.
et al.
(
2022
)
CD9 mediates the uptake of extracellular vesicles from cancer-associated fibroblasts that promote pancreatic cancer cell aggressiveness
.
Sci. Signal.
15
, eabg8191 https://doi.org/10.1126/scisignal.abg8191
102
Cardeñes
,
B.
,
Clares
,
I.
,
Toribio
,
V.
,
Pascual
,
L.
,
López-Martín
,
S.
,
Torres-Gomez
,
A.
et al.
(
2021
)
Cellular integrin α5β1 and exosomal ADAM17 mediate the binding and uptake of exosomes produced by colorectal carcinoma cells
.
Int. J. Mol. Sci.
22
, 9938 https://doi.org/10.3390/ijms22189938
103
Perez-Hernandez
,
D.
,
Gutiérrez-Vázquez
,
C.
,
Jorge
,
I.
,
López-Martín
,
S.
,
Ursa
,
A.
,
Sánchez-Madrid
,
F.
et al.
(
2013
)
The intracellular interactome of tetraspanin-enriched microdomains reveals their function as sorting machineries toward exosomes
.
J. Biol. Chem.
288
,
11649
11661
https://doi.org/10.1074/jbc.M112.445304
104
Hurwitz
,
S.N.
,
Cheerathodi
,
M.R.
,
Nkosi
,
D.
,
York
,
S.B.
and
Meckes
,
D.G
. (
2018
)
Tetraspanin CD63 bridges autophagic and endosomal processes to regulate exosomal secretion and intracellular signaling of epstein-barr virus LMP1
.
J. Virol.
92
, e01969-17 https://doi.org/10.1128/JVI.01969-17
105
Levy
,
S
. (
2014
)
Function of the tetraspanin molecule CD81 in B and T cells
.
Immunol. Res.
58
,
179
185
https://doi.org/10.1007/s12026-014-8490-7
106
Jeppesen
,
D.K.
,
Fenix
,
A.M.
,
Franklin
,
J.L.
,
Higginbotham
,
J.N.
,
Zhang
,
Q.
,
Zimmerman
,
L.J.
, et al.
(
2019
)
Reassessment of exosome composition
.
Cell
177
,
428
445
https://doi.org/10.1016/j.cell.2019.02.029
107
Latysheva
,
N.
,
Muratov
,
G.
,
Rajesh
,
S.
,
Padgett
,
M.
,
Hotchin
,
N.A.
,
Overduin
,
M.
et al.
(
2006
)
Syntenin-1 is a new component of tetraspanin-enriched microdomains: mechanisms and consequences of the interaction of syntenin-1 with CD63
.
Mol. Cell. Biol.
26
,
7707
7718
https://doi.org/10.1128/MCB.00849-06
108
Castro-Cruz
,
M.
,
Hyka
,
L.
,
Daaboul
,
G.
,
Leblanc
,
R.
,
Meeussen
,
S.
,
Lembo
,
F.
et al.
(
2023
)
PDZ scaffolds regulate extracellular vesicle production, composition, and uptake
.
Proc. Natl. Acad. Sci. U.S.A.
120
, e2310914120 https://doi.org/10.1073/pnas.2310914120
109
Ghossoub
,
R.
,
David
,
G.
and
Zimmermann
,
P
. (
2023
) Intercellular communication: what syndecan-syntenin may teach us about the molecular mechanisms that support vesicular exchanges.
In
Encyclopedia of Cell Biology
(
Bradshaw
,
R.A.
,
Hart
,
G.W.
, and
Stahl
,
P.D.
, eds), Second edition
ed
,
pp
.
401
407
,
Academic Press
110
Molostvov
,
G.
,
Gachechiladze
,
M.
,
Shaaban
,
A.M.
,
Hayward
,
S.
,
Dean
,
I.
,
Dias
,
I.H.K.
, et al.
(
2023
)
Tspan6 stimulates the chemoattractive potential of breast cancer cells for B cells in an EV- and LXR-dependent manner
.
Cell Rep.
42
,
112207
https://doi.org/10.1016/j.celrep.2023.112207
111
Andrijes
,
R.
,
Hejmadi
,
R.K.
,
Pugh
,
M.
,
Rajesh
,
S.
,
Novitskaya
,
V.
,
Ibrahim
,
M.
et al.
(
2021
)
Tetraspanin 6 is a regulator of carcinogenesis in colorectal cancer
.
Proc. Natl. Acad. Sci. U.S.A.
118
, e2011411118 https://doi.org/10.1073/pnas.2011411118
112
Charrin
,
S.
,
Manié
,
S.
,
Thiele
,
C.
,
Billard
,
M.
,
Gerlier
,
D.
,
Boucheix
,
C.
et al.
(
2003
)
A physical and functional link between cholesterol and tetraspanins
.
Eur. J. Immunol.
33
,
2479
2489
https://doi.org/10.1002/eji.200323884
113
Silvie
,
O.
,
Charrin
,
S.
,
Billard
,
M.
,
Franetich
,
J.F.
,
Clark
,
K.L.
,
Gemert
,
G.J.
, et al.
(
2006
)
Cholesterol contributes to the organization of tetraspanin-enriched microdomains and to CD81-dependent infection by malaria sporozoites
.
J. Cell. Sci.
119
,
1992
2002
https://doi.org/10.1242/jcs.02911
114
Ono
,
M.
,
Handa
,
K.
,
Sonnino
,
S.
,
Withers
,
D.A.
,
Nagai
,
H.
and
Hakomori
,
S
. (
2001
)
GM3 ganglioside inhibits CD9-facilitated haptotactic cell motility: coexpression of GM3 and CD9 is essential in the downregulation of tumor cell motility and malignancy
.
Biochemistry
40
,
6414
6421
https://doi.org/10.1021/bi0101998
115
Odintsova
,
E.
,
Butters
,
T.D.
,
Monti
,
E.
,
Sprong
,
H.
,
Meer
,
G.
and
Berditchevski
,
F
. (
2006
)
Gangliosides play an important role in the organization of CD82-enriched microdomains
.
Biochem. J.
400
,
315
325
https://doi.org/10.1042/BJ20060259
116
Fernandez
,
L.
,
Malrieu
,
M.
,
Bénistant
,
C.
,
Dosset
,
P.
,
Rubinstein
,
E.
,
Odintsova
,
E.
et al.
(
2021
)
CD82 and gangliosides tune CD81 membrane behavior
.
Int. J. Mol. Sci.
22
, 8459 https://doi.org/10.3390/ijms22168459
117
Huang
,
C.
,
Hays
,
F.A.
,
Tomasek
,
J.J.
,
Benyajati
,
S.
and
Zhang
,
X.A
. (
2020
)
Tetraspanin CD82 interaction with cholesterol promotes extracellular vesicle-mediated release of ezrin to inhibit tumour cell movement
.
J. Extracell. Vesicles
9
, 1692417 https://doi.org/10.1080/20013078.2019.1692417
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).