International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Cell migration is a fundamental process in cell biology and refers to the movement of cells from one location to another. During cell migration, a series of elongated tubular structures are produced at the trailing edge of the cell. Notably, cell migration serves a key role in various biological processes, including embryonic development, wound healing, immune response, tissue regeneration and tumor metastasis (1). Extracellular vesicles (EVs) are important mediators of intercellular communication, having notable roles in various physiological and pathological processes; for example, cancer cell-derived EVs can modulate the tumor microenvironment, and EVs from endothelial progenitor cells can induce a proangiogenic phenotype in terminally differentiated endothelial cells and promote angiogenesis. Therefore, EVs exhibit potential as novel biomarkers for diseases, therapeutic agents and drug delivery vehicles (2,3). Previous studies have revealed that EVs, including exosomes, microvesicles and apoptotic bodies, serve important roles in a number of biological processes, including intercellular communication, tissue homeostasis, cell differentiation, organ development and remodeling (2–5). In 2015, a study by Ma et al (6) discovered a new type EV-like structure and proposed the concept of migrasomes as novel cellular organelles. Compared with other EVs, migrasomes exhibit distinct structural, compositional and functional characteristics. With diameters typically >1 µm, migrasomes are substantially larger than exosomes, which have diameters of 30–150 nm. Unlike the biogenesis of conventional EVs, such as exosomes derived from the endosomal pathway or microvesicles generated via plasma membrane budding, migrasome biogenesis is closely associated with cellular migration (6). This unique biogenesis mechanism suggests that migrasomes may serve as migration trail markers, whereas exosomes predominantly facilitate long-range intercellular communication (Table I). The discovery of migrasomes provides a new perspective on how cells transport materials and transmit information through extracellular structures (Fig. 1) (6–14).
The concept of migrasomes was first introduced through observations of elongated tubular structures located at the rear of migrating cells. In early studies of migrating cells, Taylor and Robbins (7) discovered and documented that elongated tubular structures formed when migrating cells retracted from a substrate. They designated these structures ‘retraction fibrils’, which were later named ‘retraction fibers’ (RFs). Despite the initial lack of interest in these structures from researchers, in 2012, a study at Tsinghua University (Beijing, China) led by Yu (15) used transmission electron microscopy to reveal the migration process of rat kidney cells. The study revealed that cells leave behind RFs, which, upon further study, are associated with vesicular structures ranging in diameter from 0.5 to 3 µm (6,15). These structures, situated behind migrating cells that attach to the RFs left behind during cell migration (6), were termed pomegranate-like structures (PLSs) due to their resemblance to pomegranate seeds. By purifying these structures, tetraspanin (TSPAN)4, a distinct marker protein for PLS, was identified via mass spectrometry, which served as a robust reference for future migrasome research. Through knockdown of the negative regulator Shank-associated RH domain interacting protein and treatment with cell migration inhibitors, it was demonstrated that the formation of PLSs is dependent on cell migration (6). Consequently, PLSs were renamed ‘migrasomes’, being defined as annular organelles formed at the tips or intersections of RFs, or at the rear edge of migrating cells (6).
The process of migrasome production can be visualized dynamically via time-lapse imaging techniques (6,16). Subsequent studies have confirmed the widespread presence of migrasomes across a variety of species, tissues, organs and cell types, including rat eyes, lungs, intestines, zebrafish embryos, chick chorioallantoic membranes and human coronary artery endothelial cells (6,11,17–20) (Fig. 2). As a novel type of organelle, migrasomes are characterized as membrane-bound vesicles with an ellipsoidal shape that harbor numerous smaller vesicles. Their composition primarily comprises proteins, lipids and nucleic acids (6,21). Notably, migrasomes exhibit a distinctive protein profile, which includes membrane proteins, contractile proteins, cytoskeletal proteins, chaperones, vesicular trafficking proteins and cell adhesion proteins, >50% of which are membrane-related (6,21,22). These proteins are engaged predominantly in biological processes, including cell migration, cell matrix adhesion, lipid degradation, protein glycosylation and glycoprotein metabolism (21).
In comparison with the cell membrane and overall lipid composition of the cell, migrasomes are notably enriched in sphingolipids, such as sphingomyelin (SM), ceramide, monosialodihexosylgangliosides and glycosphingolipids, including monoglycosylceramide, diglycosylceramide and triglycosylceramide. In a study by Liang et al (23), it was demonstrated that ceramide and SM are essential for the formation and maintenance of migrasomes. Furthermore, filipin III staining and quantitative analysis revealed that migrasomes are enriched in cholesterol, an important component for the physical properties and structural integrity of the migrasome membrane; notably, TSPAN4, TSPAN7 and cholesterol assemble into TSPAN-enriched microdomain (TEMs), the enrichment of which stiffens the plasma membrane, facilitating migrasome initiation (9). Ongoing lipidomics analysis of migrasomes anticipates the discovery of additional lipid components, thereby verifying the growing profile of known biological characteristics and functions of migrasomes.
Migrasomes are rich in nucleic acids. A study by Zhu et al (24) employed SYTO™ 14 fluorescence staining to demonstrate the presence of RNA within migrasomes. Sequencing analyses revealed that migrasomes predominantly contain long-chain mRNAs associated with cell metabolism, intracellular membrane transport, cell adhesion, vesicle fusion and the assembly of subcellular membrane structures. These mRNAs can be translated into proteins within recipient cells, participating in the biological responses of recipient cells and regulating their life processes. For example, the phosphatase and tensin homolog (PTEN) mRNA delivered from migrasomes to recipient cells is translated into the PTEN protein, which inhibits the proliferation of the recipient cells (24). Nonetheless, the mechanisms of migrasome RNA sorting and transport have yet to be elucidated, as does the presence of DNA within migrasomes, necessitating further investigation.
TSPAN4/7, and integrins α1, α3, α5 and β1, which are expressed on the migrasome membrane, serve as important structural markers of migrasomes, with TSPAN4 being the most distinguishing marker, which exhibits the clearest expression when examined by confocal microscopy (6). Nonetheless, the detection of migrasomes using fluorescently labeled marker proteins presents limitations, including complex procedures, extended experimental duration and difficulty (25). Consequently, Chen et al (25) investigated fluorescently-labelled wheat germ agglutinin (WGA), which is a more rapid, straightforward and less invasive marker than TSPAN4. However, WGA may non-specifically bind to structures containing sialic acid and N-acetyl-D-glucosamine, and its fluorescence intensity can be influenced by various factors; for example, it may also enter the cell and bind to intracellular components, leading to enhanced nonspecific signals. In addition, different cell lines or tissue types naturally exhibit variations in the glycosylation levels of their cell membrane surfaces, which can also interfere with the results, potentially limiting its application in quantitative migrasome analysis (25). Additional specific protein markers include N-deacetylase/N-sulfotransferase 1, phosphatidylinositol glycan anchor biosynthesis class K, carboxypeptidase Q and epidermal growth factor domain-specific O-linked N-acetylglucosamine transferase; however, due to marked variations in protein content across different cell types, these markers are not uniformly present in migrasomes derived from the same cell source (21,22). Furthermore, migrasome-associated mRNAs can be labelled with the nucleic acid stain SYTO 14, making them secondary markers for migrasomes (24,26).
During cell migration, RFs are extended from the posterior end of the cell, with migrasomes situated at the termini or bifurcations of these fibers (6). When RFs break, migrasomes may leak or rupture, thereby releasing their contents into the extracellular space (6). The formation of migrasomes is a complex process that involves the regulation of multiple molecules and signaling pathways.
The formation of migrasomes is contingent upon cellular migration, a finding initially presented by Ma et al (6). A study revealed that, among 563 compounds capable of reducing migrasome numbers in cultured cells, 507 also decreased RF formation, reinforcing the association of migrasome production with cell migration (27). Subsequent research conducted by Fan et al (28) revealed a lower migrasome count in turning cells compared with those migrating in a straight line, highlighting the importance of migration continuity and speed in migrasome formation. Directional changes during migration lead to fewer RFs and migrasomes, with the removal of vimentin from cells having been shown to impair migration and reduce migrasome numbers (29,30). In conclusion, these findings suggested that the formation of migrasomes is markedly regulated by cell migration behavior.
SM, synthesized from ceramide by SMS, is a key component of the plasma membrane, and is involved in signaling and membrane transport (23). A previous study has shown that SM and ceramide are enriched on migrasomes and are present at the sites of migrasome formation; furthermore, ceramide is unevenly distributed on different migrasomes, and as migrasome biogenesis proceeds, SM levels continuously increase, indicating that ceramide can be converted into SM on migrasomes (23). Hydrolyzing SM on migrasomes or knocking out ceramide synthase 5 to reduce SM synthesis severely impairs the formation of migrasomes, demonstrating the importance of SM for migrasome formation (23).
Mammalian cells contain two principal types of SMS: i) SMS1, which is localized in the Golgi apparatus; and ii) SMS2, which is located in both the Golgi apparatus and the plasma membrane (31). SMS2 synthesizes SM from ceramide in the plasma membrane (23,31). Consequently, it is plausible that SMS2 modulates migrasome formation by facilitating SM production. Experimental evidence, including the knockout of SMS2 and treatment with SMS2 inhibitors that hinder SM synthesis, has demonstrated that migrasome formation and growth are impeded in SM-depleted conditions, whereas the reintroduction of SM restores migrasome production (23). Furthermore, impaired SM synthesis reduces cholesterol recruitment, thereby affecting TEM assembly, since TSPAN4, TSPAN7 and cholesterol assemble into TEMs, the enrichment of which stiffens the plasma membrane, which is an important condition for migrasome formation (9).
Research has revealed that SMS2 localizes to foci on the basal membrane at the leading edge of a cell, which predestines migrasome formation sites (23). These foci mature into migrasomes during cell migration (23). Intracellular SMS2 foci initially adhere to the section of the membrane of the migrating cell that is interacting with a substrate to generate motility, resembling focal adhesions (FAs), which are the sites where cells are linked to the ECM in which integrins are highly enriched. However, previous studies have failed to detect FA markers within migrasomes, and active forms or markers of FA components such as integrin α5, integrin β1 or the FA kinase paxillin do not colocalize with intracellular SMS2 foci, underscoring that SMS2 focus formation is independent of FAs (23,32). To elucidate the role of SMS2 foci in migrasome formation, researchers identified an SMS2 mutant, S217A, that cannot form foci. The formation of migrasomes in cells expressing this mutation has been shown to be markedly diminished (23). Notably, the inability to form SMS2 foci prevents migrasome formation, even when exogenous SM is added (23). These findings suggest that SMS2 foci not only regulate migrasome formation by synthesizing SM, but may also be involved in other important intracellular signaling processes that are required for migrasome biosynthesis and function. Future investigations should explore the precise mechanisms of SMS2 focus assembly, the selection of assembly sites and the adhesive mechanisms of SMS2 foci to fully elucidate the contribution of SMS2 foci to migrasome formation and function.
TSPANs constitute a family of small hydrophobic proteins characterized by four transmembrane domains, comprising a total of 33 distinct members in mammalian cells. These proteins facilitate the organization of functional higher-order protein complexes on the cell membrane through interactions with adhesion molecules, enzymes and signaling proteins, thereby forming the structures known as TEMs (33,34). Initial research identified TSPAN4 as a marker of migrasomes; however, subsequent investigations have suggested that the roles of TSPAN family members may extend beyond this initial characterization. By establishing a stable normal rat kidney (NRK) cell line that expresses various levels of TSPAN4 and green fluorescent protein (GFP), a study by Huang et al (9) demonstrated that the overexpression of 14 types of TSPAN family members enhances migrasome formation in a dose-dependent manner, with TSPAN4 exhibiting the most pronounced effect. Conversely, knockout of the TSPAN4 gene was shown to markedly reduce the number of migrasomes, underscoring the importance of TSPAN4 for migrasome formation. During the migrasome formation process, the signal from TSPAN4-GFP during the growth phase of migrasomes rapidly re-emerges on their surface, whereas no such recovery occurs when migrasomes tend to mature and stabilize (9). Furthermore, high-speed imaging revealed that TSPAN4-GFP forms rapidly-moving-discrete spots on RFs and migrasomes that assemble on the surface of migrasomes during their growth phase. Taken together, these findings suggest that TSPAN4 is recruited to migrasomes specifically during the growth phase (9).
By developing an in vitro system to simulate migrasome formation, researchers have demonstrated that TSPAN4 and cholesterol are sufficient to reconstitute migrasome-like structures, further validating their role in this process (9). To elucidate how the assembly of TEMs promotes migrasome formation, a theoretical model was proposed, identifying three key energetic drivers: i) The bending energy of the migrasome and RF membranes; ii) the membrane tension energy; and iii) the boundary energy at the migrasome-RF interface (9). A subsequent study using a biomimetic system divided migrasome growth into two phases: i) A local swelling phase, driven by membrane tension and potentially other cellular factors (such as the cytoskeleton, specific lipids, ion fluxes, mechanosensitive signaling and adhesion complexes); and ii) a TSPAN4 migration-enrichment phase mediated by TSPAN family proteins (35). Notably, the biomimetic system lacks cytoskeletal components, which may regulate migrasome formation in vivo; therefore, discrepancies between artificial and cellular systems may exist (6,17,21,22). TSPAN4, TSPAN7 and cholesterol assemble into TEMs, whose enrichment stiffens the plasma membrane, facilitating migrasome initiation. Changes in TSPAN4 curvature will affect the swelling of the plasma membrane, the reduced intrinsic curvature of TSPAN4 directs TEMs to low-curvature membrane swellings, stabilizing these protrusions and promoting migrasome maturation (9,10,18,35,36). In summary, TEMs composed of TSPAN4 and cholesterol are important for migrasome biogenesis.
Integrins are transmembrane receptors that bind ECM proteins and link to the cytoskeleton via intracellular adapters. These heterodimeric proteins, which are composed of α and β chains, are important for cell adhesion, spreading, migration and matrix remodeling (37). Migrasomes, which adhere to ECM sites during cell migration, are enriched with integrin α5β1, as revealed by mass spectrometry (32). Fluorescence staining confirms that integrin β1 in migrasomes is in an activated, ligand-bound state, demonstrating direct ECM binding. Three-dimensional imaging and total internal reflection fluorescence microscopy have further revealed the localization of integrins α5 and β1 at the migrasome base, supporting their role in migrasome anchorage (32).
Although FAs also contain high integrin concentrations, FA markers are absent in migrasomes, indicating a distinct adhesion mechanism (32,38). A functional study has revealed that NRK cells produce markedly more migrasomes on fibronectin-coated surfaces than on laminin 511- and collagen I-coated surfaces, with minimal migrasome formation on uncoated coverslips. This increase in migrasome formation on fibronectin-coated surfaces compared with other ECM components is associated with an elevated integrin α5 expression in NRK cells, as α5 knockout impairs migrasome production on fibronectin but not on other ECM components (32). Conversely, integrin α3 overexpression increases the number of migrasomes on laminin 511. Similarly, integrin α1 overexpression enhances migrasome formation on collagen IV in Chinese hamster ovarian cells without affecting formation on other ECM components (32). These findings demonstrate that migrasome formation depends on specific integrin-ECM pairings.
PIP2, a plasma membrane lipid synthesized by phosphatidylinositol 4-phosphate 5-kinase type-1α kinases, regulates key subcellular processes, including the regulation of ion channels and transporters, clathrin-dependent and -independent endocytosis, exocytosis regulation, actin polymerization, efficient FA turnover and the regulation of cell-cell contacts) (39,40). PIP2 localizes to migrasomes, as confirmed by phospholipase Cγ-PH domain-GFP fusion protein probes and antibody staining (41). Kinetic experiments have revealed that PIP2 recruitment precedes TSPAN4 and integrin α5 recruitment, both of which are important for migrasome biogenesis. Phosphatidylinositol-4-phosphate 5-kinase type-1α (PIP5K1A) inhibition disrupts migrasome formation, implicating PIP2 synthesis in this process (41).
PIP2 likely functions by recruiting binding partners, such as Rab35, a GTPase involved in organelle biogenesis, endosomal trafficking and actin regulation (42). Rab35 is recruited to migrasome formation sites in a PIP2-dependent manner, and its depletion disrupts RF elongation and migrasome assembly (41). Rab35 also interacts with integrin α5 via the GFFKR motif, recruiting integrins α5 to migrasome sites, although the direct binding mechanism remains unconfirmed (41,43). Thus, the PIP2-Rab35 axis orchestrates migrasome formation by coupling lipid signaling with integrin trafficking. Future studies should address PIP5K1A recruitment dynamics, Rab35-integrin α5 interactions and the clinical relevance of the PIP2-Rab35 signaling axis in cancer and migration-associated diseases.
Through a chemical genetic screen designed to identify compounds and protein targets that disrupt migrasome formation, a study by Lu et al (27) identified SAR407899, an inhibitor of ROCK1 and ROCK2, which stably suppresses migrasome biogenesis. ROCK1 and ROCK2 are serine/threonine kinases that act downstream of the small GTPase Ras homolog family member A (RhoA), regulating diverse cellular processes, including actin cytoskeleton organization, cell adhesion and migration, via the ROCK-Rho signaling pathway (44). Although ROCK2 knockdown does not affect migrasome formation, ROCK1 depletion markedly reduces the migrasome abundance per cell (27). Notably, ROCK1 knockdown also impairs cell migration. To distinguish the effects of RFs and migration from migrasome formation, researchers have quantified the number of migrasomes per 100 µm RFs (27). The findings of this experiment revealed that ROCK1-depleted cells generate notably weaker traction forces than healthy cells, supporting the premise that migrasome formation depends on ROCK1-mediated traction and other ECM components (such as fibronectin) adhesion (27).
PD-L1 is best known as an immune checkpoint molecule that facilitates cancer cell migration (45). Emerging evidence has indicated that PD-L1 has an intrinsic capability to facilitate sustained cellular migration, a process important for migrasome biogenesis. High-resolution time-lapse microscopy has demonstrated that PD-L1 accumulates at the trailing edge of migrating cancer cells, where it forms distinct structures that move directionally during retraction (46). Given that RFs form at the rear of the cell during migration, a subsequent study demonstrated that PD-L1 localizes not only in RFs, but also in migrasomes. Notably, PD-L1 enhances migrasome formation independently of cell migration (46). PD-L1 closely associates with integrin β4, co-localizing at the rear of the cell, and later in RFs and migrasomes. A further investigation revealed that PD-L1 recruits integrin β4 to the trailing edge; this recruitment stimulates contractility via the dynamics of the cell, a mechanism by which PD-L1 maintains rear polarity and reduces membrane tension (46). Additionally, PD-L1 activates RhoA by coupling integrin β4 to the cytoskeleton, further promoting rear contraction (actin cytoskeleton-mediated contractility) (46). Taken together, these findings underscore the dual role of PD-L1 in facilitating cell migration and migrasome formation. However, the precise mechanisms of PD-L1 in migrasome biogenesis, and its broader functional implications, warrant further exploration.
Regarding the formation mechanisms of migrasomes in different cell types, the universal core mechanism involves the following sequence of events: i) Cell migration initiation; ii) the formation of TEMs; and subsequently iii) the recruitment, fusion and maturation of microvesicles (the precursors of migrasomes) (6,9). However, migrasome formation is a complex biological process influenced by multiple factors, and the underlying mechanisms may vary across cell types. Some examples are as follows: i) Triggering signals vary, for example monocytes and macrophages may initiate migrasome generation through tissue damage and clearance signals, with these migrasomes potentially participating in angiogenesis regulation or damaged mitochondrial clearance, whereas cancer cells may trigger migrasome formation via oncogenic or metastasis-driving signals to promote cancer progression; and ii) regulatory factors may be cell type-specific. For example, normal human dermal fibroblasts exhibit modulated migrasome formation through peptide-modified matrices, which affect contractile fiber quantity and length (different peptide-modified substrates influence the strength of cell migration, such as migration distance and duration, thereby affecting the number and length of RFs) (47). Mouse embryonic stem cells exhibit calcium ion and synaptotagmin-1 (Syt1)-regulated migrasome production, where calcium promotes migrasome formation via Syt1 (48). Furthermore, in H4 human glioma cells, osmotic regulation may control migrasome formation, as hypotonic conditions induce the formation of TSPAN4-enriched migrasome-like vesicles on RFs. These cholesterol-dependent vesicles exhibit migrasome characteristics but originate from osmotic stress rather than from cell migration (49). While conclusive identification of these cholesterol-dependent vesicles as migrasomes requires further evidence, these observations provide valuable perspectives on cellular osmoregulation and migrasome biophysical responses in tissues. A previous report indicated that high-salt diets exacerbate ischemic brain injury by promoting excessive migrasome formation in microglia and macrophages, reducing their post-ischemic populations alongside astrocytes (17).
Migrasomes from different cell types participate in distinct biological activities. Nevertheless, research on most cell-type-specific migrasomes remains preliminary, and the generation mechanisms of these migrasomes have not been fully elucidated. Currently, evidence confirms only their existence and functional roles in various biological responses. For example, migrasomes produced by zebrafish embryonic cells can regulate the formation of zebrafish embryonic organs; migrasomes derived from adipose stem cells serve a key role in adipose tissue regeneration; and migrasomes from neutrophils affect the coagulation function of the body (18,50). Specialized investigations into cell-type-specific formation mechanisms remain lacking, preventing targeted discussion of these mechanisms at present. Exploring these heterogeneous-origin migrasomes and their impacts on cellular functions and disease pathogenesis constitute an important future research direction.
Migrasomes, notable organelles in cell migration, are increasingly recognized for their roles in cellular physiology and pathology. Their formation and function are closely associated not only with cellular-migratory capacity, but also with intercellular communication, tissue remodeling and immune regulation. These microvesicles serve as carriers for the intercellular transfer of biomolecules, such as proteins, mRNAs and microRNAs, thereby modulating the gene expression and functional states of recipient cells. The present section elucidates the biological functions of migrasomes in organ morphogenesis, angiogenesis, mitochondrial quality control and immune regulation, highlighting their notable roles in maintaining tissue homeostasis and contributing to disease pathogenesis (Fig. 3). A deeper understanding of these functions underscores the complexity and diversity of migrasomes in biological systems and their potential as therapeutic targets in medical research.
Migrasomes constitute a notable secretory pathway in migrating cells, encapsulating diverse cytoplasmic contents (6,51). Migrasomes form and mature on RFs of migrating cells before they detach, rupture and release their contents (6) (Fig. 3A). Migrasomes can also be internalized by neighboring cells, or adhere to cell surfaces or the ECM without being engulfed. For example, interactions with the ECM enable migrasomes to attach to specific cell surfaces or tissues, facilitating intercellular communication (32). These vesicles transport intracellular biomolecules, including proteins, mRNAs and microRNAs. Recent studies have indicated that secretory proteins, such as signaling molecules, are actively transported from migrating cells into migrasomes via kinesin-mediated carriers, akin to targeted neurotransmitter release in neuronal systems (51). These molecules can be transferred to adjacent or distant cells, altering recipient cell-gene expression and function. For example, Zhu et al (24) added purified migrasomes derived from L929 cells to U87-MG, MDA-MB-468 and PC3 cells that do not express the PTEN protein, and demonstrated that migrasomes mediate the transfer of PTEN mRNA and protein, inhibiting proliferation in recipient cells.
Migrasomes are enriched with cytokines, including chemokines and morphogens, which enables them to act as signaling molecule-carriers, and thereby influence cell behavior. In zebrafish embryos, gastrula-derived migrasomes contain high levels of C-X-C motif chemokine ligand (CXCL)12a (18). These migrasomes, produced during mesoderm and endoderm cell gastrulation, regulate organ development. Mutations that reduce migrasome numbers lead to severe developmental defects, which can be rescued by migrasome supplementation, underscoring their role in organogenesis (18). Further studies have revealed that CXCL12 in migrasomes interacts with C-X-C chemokine receptor type 4 (CXCR4) on dorsal precursor cells (DFCs), inducing chemotaxis and ensuring typical organ morphogenesis (35). Migrasomes thus spatially and temporally distribute signaling molecules, releasing CXCL12 upon rupture to modulate DFC behavior. Similarly, adipose-derived stem cells produce CXCL12-enriched migrasomes that promote adipose tissue regeneration via CXCR4/RhoA signaling (50).
A study by Zhang et al (11) identified migrasome-producing monocytes in the chorioallantoic membrane of chicken embryos, where these vesicles are rich in proangiogenic factors, including TGF-β3, VEGFA and CXCL12. In this context, migrasomes induce endothelial tube formation; their depletion via monocyte clearance or TSPAN4 knockout inhibits angiogenesis, whereas supplementation restores monocyte recruitment and capillary formation (11). Monocytes leverage migrasomes to deliver angiogenic factors, thereby creating a microenvironment conducive to blood vessel growth. Notably, migrasome-derived CXCL12 recruits additional monocytes, forming a positive feedback loop (11,52).
Multipotent mesenchymal stem cells (MSCs) are precursors to various cell types, with the ability to support tissue homeostasis, promote hematopoiesis and interact with cancer cells. Previous research on MSCs has focused on MSC-derived EVs (MSC-EVs), indicating that MSC-EVs have functions similar to those of MSC (53–58). A recent investigation demonstrated that MSCs generate migrasomes that attract leukemia KG-1a cells and CD34+ hematopoietic stem cells via the CXCR4-CXCL12 axis (59). These migrasomes, enriched in TSPANs, including CD166 and TSPAN4, and endosomal markers, such as Rab7 and CD63, are influenced by ECM components such as fibronectin and laminins. In addition to influencing cell migration and thereby affecting the formation of retraction fibers, ECM components can also impact the process of TEM formation (59). Migrasomes produced by MSCs attract leukemia KG-1a cells and CD34+ hematopoietic stem cells through the CXCR4-CXCL12 axis, thereby facilitating communication between the MSCs and these cells, thus highlighting the role of migrasomes in MSC-hematopoietic cell interactions, offering insights into their functions in health and disease.
Neutrophil-derived migrasomes adsorb coagulation factors from the plasma; migrasomes actively bind and enrich coagulation factors on their surface by virtue of their unique membrane composition, particularly cholesterol esters, and then localize to injury sites and modulate coagulation by activating platelets (60). These migrasomes also regulate immune responses by delivering cytokines and chemokines to injury sites, enhancing immune cell recruitment during inflammation (60). Conversely, migrasomes have been shown to suppress immunity by transporting immunosuppressive molecules, thereby maintaining immune tolerance (46).
Mitochondria, the cellular ‘powerhouses’, maintain homeostasis through energy production and quality control mechanisms, such as mitophagy (61,62). A previous study demonstrated that migrasomes mediate mitocytosis, a process in which damaged mitochondria are selectively expelled to sustain cellular health (12) (Fig. 3C). Under stress conditions, such as carbonyl cyanide m-chlorophenyl hydrazone treatment, mitochondria undergo fragmentation via kinesin-5B (KIF5B)-mediated transport and myosin head domain-containing protein 1 (MYO19)/density-regulated protein 1-dependent fission, accumulating at the cell periphery for subsequent migrasome encapsulation. Knocking out TSPAN9 or taking other measures to block the formation of migrasomes has been shown to cause migrating cells to lose their mitocytosis capability, leading to the accumulation of damaged mitochondria within the cells and affecting cell viability (12,63,64).
Although both mitocytosis and mitophagy contribute to cellular homeostasis, their mechanisms differ markedly. Mitocytosis, which is mediated by migrasomes, selectively removes mildly damaged mitochondria from migrating cells. In this process, damaged mitochondria selectively bind to intracellular dynein to facilitate their transport out of the cell, they are then transported towards the cell periphery by outwards motor proteins, such as KIF5B. MYO19 further facilitates this process by anchoring mitochondria to cortical actin, thereby promoting their incorporation into migrasomes (12,63,64). By contrast, mitophagy primarily degrades damaged mitochondria via the autophagy pathway. Upon mitochondrial damage, PTEN-induced kinase 1 accumulates on the outer mitochondrial membrane, where it recruits parkin to ubiquitinate outer membrane proteins. This ubiquitination marks the mitochondria for autophagosomal engulfment, and subsequent lysosomal degradation (61,62).
Mitocytosis complements mitophagy by incrementally clearing mildly damaged mitochondria. Migrasomes may also transfer mitochondrial components, including mitochondrial DNA and mitochondrial microRNAs, to recipient cells, influencing their function.
Migrasomes facilitate viral dissemination, facilitating the evasion of antiviral therapies (Fig. 3B). Vaccinia virus induces migrasomes containing viral particles, enabling their spread despite tecovirimat treatment (13,65). Similarly, herpes simplex virus-2 exploits migrasomes as ‘Trojan horses’ for intercellular transmission (66). Taken together, these findings illuminate novel viral spread mechanisms and suggest potential targets for antiviral strategies.
As research advances, the understating of migrasomes is improving, with their structure and physiological functions becoming increasingly elucidated. Studies have demonstrated that migrasomes perform notable roles in intercellular communication and signal transduction, as well as in the pathogenesis of various diseases (Fig. 4). These findings highlight the potential of migrasomes for clinical applications in disease diagnosis and treatment.
The association between migrasomes and kidney disease is particularly notable. Studies have identified podocyte-derived migrasomes in urine as early biomarkers of podocyte injury (14,67). Podocytes, which regulate glomerular permeability, are terminally differentiated cells incapable of regeneration; therefore, early detection of their injury is important for managing glomerular diseases (14). Research indicates that podocytes generate migrasomes during migration, with their numbers markedly increasing during kidney injury. Furthermore, Rac-1 inhibitors that target a small Rho family GTPase that is overactivated in podocyte injury dose-dependently suppress lipopolysaccharide-induced migrasome release, underscoring the diagnostic potential of the urinary presence of podocyte migrasomes (14).
Migrasomes also exhibit therapeutic relevance for post-stroke pneumonia. Bone marrow (BM)-MSC-derived migrasomes, which are packed with dermcidin, display dual effects that reduce pulmonary bacteria load and enhance LC3-associated phagocytosis (LAP) of macrophages (68). These migrasomes not only exert antibacterial effects, but also augment LAP in macrophages, facilitating bacterial clearance. Consequently, BM-MSC-derived migrasomes represent a promising alternative to conventional antibiotics for preventing and treating post-stroke pneumonia.
In neurodegenerative disorders, migrasomes have been implicated in cerebral amyloid angiopathy (CAA). β-amyloid protein 40-induced macrophage-derived migrasomes adhere to the vasculature in biopsies from patients with CAA and mouse models, delivering the protein CD5L to vascular walls and triggering complement-dependent cytotoxicity, thereby compromising the blood-brain barrier (69). These observations suggest that macrophage-derived migrasomes and complement activation are potential biomarkers and therapeutic targets for CAA.
Emerging evidence further links migrasomes to atherosclerosis, myocardial infarction and malignancies. TSPAN4, a migrasome-forming protein, is strongly associated with atherosclerosis regression-associated macrophages according to single-cell sequencing, Gene Expression Omnibus datasets and The Cancer Genome Atlas analyses, and is also associated with plaque hemorrhage and rupture (70). In another study, low-intensity pulsed ultrasound has been shown to mitigate myocardial ischemia-reperfusion injury via migrasome-mediated mitochondrial quality control; the possible mechanism involves promoting the formation of migrasomes, potentially by enhancing cell motility, which enables migrasomes to exert mitocytosis activity, facilitating the removal of damaged mitochondria from cells (71). TSPAN4 is also upregulated in hepatocellular carcinoma, gastric cancer and glioblastoma (GBM), where it influences tumor-associated macrophages (72). In hepatocellular carcinoma, migrasomes guide cancer cell invasion (73); in pancreatic cancer, pancreatic cancer cell-derived migrasomes modulate immunosuppressive factors in the tumor microenvironment, thereby accelerating progression (74). In GBM, TSPAN4 promotes the progression of GBM by regulating epidermal growth factor receptor stability, whereas migrasome-autophagosome crosstalk alleviates endoplasmic reticulum stress and migrasome-mediated GBM-microenvironment communication may drive recurrence (75–77). Collectively, these findings underscore the therapeutic potential of targeting TSPAN4 and migrasomes in severe diseases.
Migrasomes, a novel class of EVs, have attracted considerable attention in the field of cell biology, with their important roles in cellular processes gradually being elucidated. Research on migrasomes has advanced markedly, from structural characterization to functional exploration. These vesicles not only serve important roles in normal physiological processes, but also modulate cell behavior and disease progression under pathological conditions, highlighting their potential as biomarkers and therapeutic targets.
The field of migrasome research is rapidly evolving, but still faces substantial challenges. Firstly, a deeper mechanistic understanding of migrasome biogenesis and its regulatory networks, particularly the contributions of lipid and protein components, is important. Secondly, further experimental validation is needed to clarify how migrasome-derived biomolecules, such as RNAs and proteins, influence recipient cell functions and mediate intercellular communication. Additionally, investigating migrasome heterogeneity across cell types and tissues, as well as their functional alterations in disease states, remains a key research direction. Advances in single-cell sequencing and high-resolution imaging technologies are expected to provide deeper insights into the biology and disease-related functions of migrasomes.
Clinically, migrasome research may offer novel strategies for early disease diagnosis and therapy (Table II). For example, migrasomes could serve as biomarkers for monitoring kidney or neurodegenerative diseases, or their formation and function could be therapeutically targeted. Furthermore, migrasome involvement in viral transmission suggests their potential applications in infectious disease research. As knowledge on the function of migrasomes expands, their impact on research and clinical therapeutics is expected to grow. In summary, migrasome research presents both challenges and opportunities, with future discoveries poised to further elucidate the complexity of cellular processes while advancing diagnostic and therapeutic innovations.
Not applicable.
The present study was supported by the Jiangsu Provincial Key Medical Discipline (grant no. ZDXK202227) and the Wuxi Taihu Talent Program (grant no. WX0302B010507200065).
Not applicable.
YC and QW wrote the manuscript. YC, XL, BZ and HZ revised the manuscript. XL supervised the present review and guided each author who participated in writing the article. Data authentication is not applicable. All authors read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
EV |
extracellular vesicle |
|
RF |
retraction fiber |
|
PLS |
pomegranate-like structure |
|
TSPAN |
tetraspanin |
|
SM |
sphingomyelin |
|
WGA |
wheat germ agglutinin |
|
SMS |
sphingomyelin synthase |
|
TEM |
tetraspanin-enriched microdomain |
|
FA |
focal adhesion |
|
NRK |
normal rat kidney |
|
ECM |
extracellular matrix |
|
PIP2 |
phosphatidylinositol 4,5-bisphosphate |
|
GFP |
green fluorescent protein |
|
PIP5K1A |
phosphatidylinositol-4-phosphate 5-kinase type-1α |
|
PD-L1 |
programmed death-ligand 1 |
|
ROCK |
Rho-associated protein kinase |
|
RhoA |
Ras homolog family member A |
|
PTEN |
phosphatase and tensin homolog |
|
CXCL |
C-X-C motif chemokine ligand |
|
CXCR4 |
C-X-C chemokine receptor type 4 |
|
DFC |
dorsal precursor cell |
|
MSC |
mesenchymal stem cell |
|
KIF5B |
kinesin-5B |
|
MYO19 |
myosin head domain-containing protein 1 |
|
BM-MSCs |
bone marrow mesenchymal stem cells |
|
Syt1 |
synaptotagmin-1 |
|
CAA |
cerebral amyloid angiopathy |
|
GBM |
glioblastoma |
|
Trepat X, Chen Z and Jacobson K: Cell migration. Compr Physiol. 2:2369–2392. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Ratajczak J, Wysoczynski M, Hayek F, Janowska-Wieczorek A and Ratajczak MZ: Membrane-derived microvesicles: Important and underappreciated mediators of cell-to-cell communication. Leukemia. 20:1487–1495. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Bruno S, Chiabotto G, Favaro E, Deregibus MC and Camussi G: Role of extracellular vesicles in stem cell biology. Am J Physiol Cell Physiol. 317:C303–C313. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Hopkin K: Core concept: Extracellular vesicles garner interest from academia and biotech. Proc Natl Acad Sci USA. 113:9126–9128. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Kang T, Atukorala I and Mathivanan S: Biogenesis of extracellular vesicles. Subcell Biochem. 97:19–43. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Ma L, Li Y, Peng J, Wu D, Zhao X, Cui Y, Chen L, Yan X, Du Y and Yu L: Discovery of the migrasome, an organelle mediating release of cytoplasmic contents during cell migration. Cell Res. 25:24–38. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Taylor AC and Robbins E: Observations on microextensions from the surface of isolated vertebrate cells. Dev Biol. 6:660–673. 1963. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Y, Li Y, Ma L and Yu L: Detection of migrasomes. Methods Mol Biol. 1749:43–49. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Huang Y, Zucker B, Zhang S, Elias S, Zhu Y, Chen H, Ding T, Li Y, Sun Y, Lou J, et al: Migrasome formation is mediated by assembly of micron-scale tetraspanin macrodomains. Nat Cell Biol. 21:991–1002. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Tavano S and Heisenberg CP: Migrasomes take center stage. Nat Cell Biol. 21:918–920. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang C, Li T, Yin S, Gao M, He H, Li Y, Jiang D, Shi M, Wang J and Yu L: Monocytes deposit migrasomes to promote embryonic angiogenesis. Nat Cell Biol. 24:1726–1738. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Jiao H, Jiang D, Hu X, Du W, Ji L, Yang Y, Li X, Sho T, Wang X, Li Y, et al: Mitocytosis, a migrasome-mediated mitochondrial quality-control process. Cell. 184:2896–2910.e13. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Lv L and Zhang L: Identification of poxvirus inside migrasomes suggests a novel mode of mpox virus spread. J Infect. 87:160–162. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Li S, Rong W, Zeng C, Zhu X, Chen Q, Li L, Liu ZH and Zen K: Podocyte-released migrasomes in urine serve as an indicator for early podocyte injury. Kidney Dis (Basel). 6:422–433. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yu L: Migrasomes: The knowns, the known unknowns and the unknown unknowns: A personal perspective. Sci China Life Sci. 64:162–166. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wu J, Lu Z, Jiang D, Guo Y, Qiao H, Zhang Y, Zhu T, Cai Y, Zhang X, Zhanghao K, et al: Iterative tomography with digital adaptive optics permits hour-long intravital observation of 3D subcellular dynamics at millisecond scale. Cell. 184:3318–3332.e17. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Schmidt-Pogoda A, Strecker JK, Liebmann M, Massoth C, Beuker C, Hansen U, König S, Albrecht S, Bock S, Breuer J, et al: Dietary salt promotes ischemic brain injury and is associated with parenchymal migrasome formation. PLoS One. 13:e02098712018. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang D, Jiang Z, Lu D, Wang X, Liang H, Zhang J, Meng Y, Li Y, Wu D, Huang Y, et al: Migrasomes provide regional cues for organ morphogenesis during zebrafish gastrulation. Nat Cell Biol. 21:966–977. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Gagat M, Zielińska W, Mikołajczyk K, Zabrzyński J, Krajewski A, Klimaszewska-Wiśniewska A, Grzanka D and Grzanka A: CRISPR-based activation of endogenous expression of tpm1 inhibits inflammatory response of primary human coronary artery endothelial and smooth muscle cells induced by recombinant human tumor necrosis factor α. Front Cell Dev Biol. 9:6680322021. View Article : Google Scholar : PubMed/NCBI | |
|
Ardalan M, Hosseiniyan Khatibi SM, Rahbar Saadat Y, Bastami M, Nariman-Saleh-Fam Z, Abediazar S, Khalilov R and Zununi Vahed S: Migrasomes and exosomes; different types of messaging vesicles in podocytes. Cell Biol Int. 46:52–62. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao X, Lei Y, Zheng J, Peng J, Li Y, Yu L and Chen Y: Identification of markers for migrasome detection. Cell Discov. 5:272019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Wang J, Ding Y, Zhang J, Xu Y, Xu J, Zheng S and Yang H: Migrasome and tetraspanins in vascular homeostasis: Concept, present, and future. Front Cell Dev Biol. 8:4382020. View Article : Google Scholar : PubMed/NCBI | |
|
Liang H, Ma X, Zhang Y, Liu Y, Liu N, Zhang W, Chen J, Liu B, Du W, Liu X and Yu L: The formation of migrasomes is initiated by the assembly of sphingomyelin synthase 2 foci at the leading edge of migrating cells. Nat Cell Biol. 25:1173–1184. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu M, Zou Q, Huang R, Li Y, Xing X, Fang J, Ma L, Li L, Yang X and Yu L: Lateral transfer of mRNA and protein by migrasomes modifies the recipient cells. Cell Res. 31:237–240. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chen L, Ma L and Yu L: WGA is a probe for migrasomes. Cell Discov. 5:132019. View Article : Google Scholar : PubMed/NCBI | |
|
Gustafson CM, Roffers-Agarwal J and Gammill LS: Chick cranial neural crest cells release extracellular vesicles that are critical for their migration. J Cell Sci. 135:jcs2602722022. View Article : Google Scholar : PubMed/NCBI | |
|
Lu P, Liu R, Lu D, Xu Y, Yang X, Jiang Z, Yang C, Yu L, Lei X and Chen Y: Chemical screening identifies ROCK1 as a regulator of migrasome formation. Cell Discov. 6:512020. View Article : Google Scholar : PubMed/NCBI | |
|
Fan C, Shi X, Zhao K, Wang L, Shi K, Liu YJ, Li H, Ji B and Jiu Y: Cell migration orchestrates migrasome formation by shaping retraction fibers. J Cell Biol. 221:e2021091682022. View Article : Google Scholar : PubMed/NCBI | |
|
Ivaska J, Pallari HM, Nevo J and Eriksson JE: Novel functions of vimentin in cell adhesion, migration, and signaling. Exp Cell Res. 313:2050–2062. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Jiu Y, Peränen J, Schaible N, Cheng F, Eriksson JE, Krishnan R and Lappalainen P: Vimentin intermediate filaments control actin stress fiber assembly through GEF-H1 and RhoA. J Cell Sci. 130:892–902. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Huitema K, van den Dikkenberg J, Brouwers JF and Holthuis JC: Identification of a family of animal sphingomyelin synthases. EMBO J. 23:33–44. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Wu D, Xu Y, Ding T, Zu Y, Yang C and Yu L: Pairing of integrins with ECM proteins determines migrasome formation. Cell Res. 27:1397–1400. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zuidscherwoude M, Göttfert F, Dunlock VM, Figdor CG, van den Bogaart G and van Spriel AB: The tetraspanin web revisited by super-resolution microscopy. Sci Rep. 5:122012015. View Article : Google Scholar : PubMed/NCBI | |
|
Hemler ME: Tetraspanin proteins mediate cellular penetration, invasion, and fusion events and define a novel type of membrane microdomain. Annu Rev Cell Dev Biol. 19:397–422. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Dharan R, Huang Y, Cheppali SK, Goren S, Shendrik P, Wang W, Qiao J, Kozlov MM, Yu L and Sorkin R: Tetraspanin 4 stabilizes membrane swellings and facilitates their maturation into migrasomes. Nat Commun. 14:10372023. View Article : Google Scholar : PubMed/NCBI | |
|
Dharan R, Goren S, Cheppali SK, Shendrik P, Brand G, Vaknin A, Yu L, Kozlov MM and Sorkin R: Transmembrane proteins tetraspanin 4 and CD9 sense membrane curvature. Proc Natl Acad Sci USA. 119:e22089931192022. View Article : Google Scholar : PubMed/NCBI | |
|
Zaidel-Bar R: Job-splitting among integrins. Nat Cell Biol. 15:575–577. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Wehrle-Haller B: Structure and function of focal adhesions. Curr Opin Cell Biol. 24:116–124. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Kolay S, Basu U and Raghu P: Control of diverse subcellular processes by a single multi-functional lipid phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. Biochem J. 473:1681–1692. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Hammond GRV: Does PtdIns(4,5)P2 concentrate so it can multi-task? Biochem Soc Trans. 44:228–233. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Ding T, Ji J, Zhang W, Liu Y, Liu B, Han Y, Chen C and Yu L: The phosphatidylinositol (4,5)-bisphosphate-Rab35 axis regulates migrasome formation. Cell Res. 33:617–627. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Klinkert K and Echard A: Rab35 GTPase: A central regulator of phosphoinositides and F-actin in endocytic recycling and beyond. Traffic. 17:1063–1077. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Allaire PD, Seyed Sadr M, Chaineau M, Seyed Sadr E, Konefal S, Fotouhi M, Maret D, Ritter B, Del Maestro RF and McPherson PS: Interplay between Rab35 and Arf6 controls cargo recycling to coordinate cell adhesion and migration. J Cell Sci. 126:722–731. 2013.PubMed/NCBI | |
|
Lock FE, Ryan KR, Poulter NS, Parsons M and Hotchin NA: Differential regulation of adhesion complex turnover by ROCK1 and ROCK2. PLoS One. 7:e314232012. View Article : Google Scholar : PubMed/NCBI | |
|
Yu W, Hua Y, Qiu H, Hao J, Zou K, Li Z, Hu S, Guo P, Chen M, Sui S, et al: PD-L1 promotes tumor growth and progression by activating WIP and β-catenin signaling pathways and predicts poor prognosis in lung cancer. Cell Death Dis. 11:5062020. View Article : Google Scholar : PubMed/NCBI | |
|
Wang M, Xiong C and Mercurio AM: PD-LI promotes rear retraction during persistent cell migration by altering integrin β4 dynamics. J Cell Biol. 221:e2021080832022. View Article : Google Scholar : PubMed/NCBI | |
|
Saito S, Tanaka M, Tatematsu S and Okochi M: Peptide-modified substrate enhances cell migration and migrasome formation. Mater Sci Eng C Mater Biol Appl. 131:1124952021. View Article : Google Scholar : PubMed/NCBI | |
|
Han Y and Yu L: Calcium ions promote migrasome formation via Synaptotagmin-1. J Cell Biol. 223:e2024020602024. View Article : Google Scholar : PubMed/NCBI | |
|
Yoshikawa K, Saito S, Kadonosono T, Tanaka M and Okochi M: Osmotic stress induces the formation of migrasome-like vesicles. FEBS Lett. 598:437–445. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Y, Li Y, Li B, Hu D, Dong Z and Lu F: Migrasomes from adipose derived stem cells enrich CXCL12 to recruit stem cells via CXCR4/RhoA for a positive feedback loop mediating soft tissue regeneration. J Nanobiotechnology. 22:2192024. View Article : Google Scholar : PubMed/NCBI | |
|
Jiao H, Li X, Li Y, Guo Y, Hu X, Sho T, Luo Y, Wang J, Cao H, Du W, et al: Localized, highly efficient secretion of signaling proteins by migrasomes. Cell Res. 34:572–585. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Strzyz P: Migrasomes promote angiogenesis. Nat Rev Mol Cell Biol. 24:842023. View Article : Google Scholar : PubMed/NCBI | |
|
Nawaz M, Fatima F, Vallabhaneni KC, Penfornis P, Valadi H, Ekström K, Kholia S, Whitt JD, Fernandes JD, Pochampally R, et al: Extracellular vesicles: Evolving factors in stem cell biology. Stem Cells Int. 2016:10731402016. View Article : Google Scholar : PubMed/NCBI | |
|
Rani S, Ryan AE, Griffin MD and Ritter T: Mesenchymal stem cell-derived extracellular vesicles: Toward cell-free therapeutic applications. Mol Ther. 23:812–823. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Tan X, Gong YZ, Wu P, Liao DF and Zheng XL: Mesenchymal stem cell-derived microparticles: A promising therapeutic strategy. Int J Mol Sci. 15:14348–14363. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Bruno S and Camussi G: Role of mesenchymal stem cell-derived microvesicles in tissue repair. Pediatr Nephrol. 28:2249–2254. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Chopp M, Meng Y, Katakowski M, Xin H, Mahmood A and Xiong Y: Effect of exosomes derived from multipluripotent mesenchymal stromal cells on functional recovery and neurovascular plasticity in rats after traumatic brain injury. J Neurosurg. 122:856–867. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Xin H, Li Y, Buller B, Katakowski M, Zhang Y, Wang X, Shang X, Zhang ZG and Chopp M: Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells. 30:1556–1564. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Deniz IA, Karbanová J, Wobus M, Bornhäuser M, Wimberger P, Kuhlmann JD and Corbeil D: Mesenchymal stromal cell-associated migrasomes: A new source of chemoattractant for cells of hematopoietic origin. Cell Commun Signal. 21:362023. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang D, Jiao L, Li Q, Xie R, Jia H, Wang S, Chen Y, Liu S, Huang D, Zheng J, et al: Neutrophil-derived migrasomes are an essential part of the coagulation system. Nat Cell Biol. 26:1110–1123. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Sugiura A, McLelland GL, Fon EA and McBride HM: A new pathway for mitochondrial quality control: Mitochondrial-derived vesicles. EMBO J. 33:2142–2156. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Poole LP and Macleod KF: Mitophagy in tumorigenesis and metastasis. Cell Mol Life Sci. 78:3817–3851. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Baumann K: Damaged mitochondria are discarded via migrasomes. Nat Rev Mol Cell Biol. 22:4422021. View Article : Google Scholar : PubMed/NCBI | |
|
Mehra C and Pernas L: Move it to lose it: Mitocytosis expels damaged mitochondria. Dev Cell. 56:2014–2015. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao W, Tang X and Zhang L: Virus-containing migrasomes enable poxviruses to evade tecovirimat/ST-246 treatment. J Infect. 88:203–205. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Zhu Z, Li Y, Yang M and Hu Q: Migrasomes released by HSV-2-infected cells serve as a conveyance for virus spread. Virol Sin. 38:643–645. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Yang R, Zhang H, Chen S, Lou K, Zhou M, Zhang M, Lu R, Zheng C, Li L, Chen Q, et al: Quantification of urinary podocyte-derived migrasomes for the diagnosis of kidney disease. J Extracell Vesicles. 13:e124602024. View Article : Google Scholar : PubMed/NCBI | |
|
Li T, Su X, Lu P, Kang X, Hu M, Li C, Wang S, Lu D, Shen S, Huang H, et al: Bone marrow mesenchymal stem cell-derived dermcidin-containing migrasomes enhance LC3-associated phagocytosis of pulmonary macrophages and protect against post-stroke pneumonia. Adv Sci (Weinh). 10:e22064322023. View Article : Google Scholar : PubMed/NCBI | |
|
Hu M, Li T, Ma X, Liu S, Li C, Huang Z, Lin Y, Wu R, Wang S, Lu D, et al: Macrophage lineage cells-derived migrasomes activate complement-dependent blood-brain barrier damage in cerebral amyloid angiopathy mouse model. Nat Commun. 14:39452023. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng Y, Lang Y, Qi B and Li T: TSPAN4 and migrasomes in atherosclerosis regression correlated to myocardial infarction and pan-cancer progression. Cell Adh Migr. 17:14–19. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Sun P, Li Y, Yu W, Chen J, Wan P, Wang Z, Zhang M, Wang C, Fu S, Mang G, et al: Low-intensity pulsed ultrasound improves myocardial ischaemia-reperfusion injury via migrasome-mediated mitocytosis. Clin Transl Med. 14:e17492024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Zhang T, Zhang R, Zhang Z and Tan S: Migrasomes and tetraspanins in hepatocellular carcinoma: Current status and future prospects. Future Sci OA. 9:FSO8902023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang K, Zhu Z, Jia R, Wang NA, Shi M, Wang Y, Xiang S, Zhang Q and Xu L: CD151-enriched migrasomes mediate hepatocellular carcinoma invasion by conditioning cancer cells and promoting angiogenesis. J Exp Clin Cancer Res. 43:1602024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang R, Peng J, Zhang Y, Zheng K, Chen Y, Liu L, Li T, Liu J, Li Y, Yang S, et al: Pancreatic cancer cell-derived migrasomes promote cancer progression by fostering an immunosuppressive tumor microenvironment. Cancer Lett. 605:2172892024. View Article : Google Scholar : PubMed/NCBI | |
|
Dong Y, Tang X, Zhao W, Liu P, Yu W, Ren J, Chen Y, Cui Y, Chen J and Liu Y: TSPAN4 influences glioblastoma progression through regulating EGFR stability. iScience. 27:1104172024. View Article : Google Scholar : PubMed/NCBI | |
|
Lee SY, Choi SH, Kim Y, Ahn HS, Ko YG, Kim K, Chi SW and Kim H: Migrasomal autophagosomes relieve endoplasmic reticulum stress in glioblastoma cells. BMC Biol. 22:232024. View Article : Google Scholar : PubMed/NCBI | |
|
Köktürk S, Doğan S, Yılmaz CE, Cetinkol Y and Mutlu O: Expression of brain-derived neurotrophic factor and formation of migrasome increases in the glioma cells induced by the adipokinetic hormone. Rev Assoc Med Bras (1992). 70:e202313372024. View Article : Google Scholar : PubMed/NCBI |