Skip to main content

Advances in regulating endothelial-mesenchymal transformation through exosomes

Abstract

Endothelial-mesenchymal transformation (EndoMT) is the process through which endothelial cells transform into mesenchymal cells, affecting their morphology, gene expression, and function. EndoMT is a potential risk factor for cardiovascular and cerebrovascular diseases, tumor metastasis, and fibrosis. Recent research has highlighted the role of exosomes, a mode of cellular communication, in the regulation of EndoMT. Exosomes from diseased tissues and microenvironments can promote EndoMT, increase endothelial permeability, and compromise the vascular barrier. Conversely, exosomes derived from stem cells or progenitor cells can inhibit the EndoMT process and preserve endothelial function. By modifying exosome membranes or contents, we can harness the advantages of exosomes as carriers, enhancing their targeting and ability to inhibit EndoMT. This review aims to systematically summarize the regulation of EndoMT by exosomes in different disease contexts and provide effective strategies for exosome-based EndoMT intervention.

Introduction

Endothelial-mesenchymal transformation (EndoMT) involves a series of cellular and molecular changes that cause endothelial cells(ECs) to lose their characteristics and acquire mesenchymal traits [1]. Initially, endothelial cells undergo morphological changes, shifting from an oval to a slender spindle shape. This process is accompanied by altered gene expression, characterized by the loss of endothelial markers (e.g., VE-cadherin, CD31) and the gain of mesenchymal markers (e.g., SM22α, α-SMA). Functional changes include enhanced cell migration and contraction, as well as increased invasive capabilities [1, 2]. EndoMT is increasingly recognized as a risk factor in cardiovascular diseases (e.g., pulmonary hypertension, atherosclerosis) [3, 4], and fibrotic diseases (e.g., heart and kidney fibrosis) [5, 6]. It also contributes to the breakdown of the endothelial barrier and increased vascular permeability, facilitating tumor cell invasion and metastasis [7, 8].

Exosomes, extracellular vesicles ranging from 30 to 150 nm in diameter, originate from endosomes, specifically multivesicular bodies (MVBs). When MVBs fuse with the plasma membrane, they release exosomes into the extracellular space [9,10,11]. Exosomes can deliver proteins, lipids, and nucleic acids (e.g., DNA, mRNA, microRNA, non-coding RNA) to target cells, regulating their functions [12, 13]. They can either promote or inhibit disease progression through the molecules they carry [14, 15]. Recent studies indicate that exosomal proteins and nucleic acids play critical roles in EndoMT regulation (Fig. 1). Therefore, this review systematically summarizes the regulatory effects and mechanisms of exosomes on EndoMT across different disease contexts, identifies the limitations of current researches, and outlines future directions to promote the translational application of exosome-mediated EndoMT regulation.

Fig. 1
figure 1

Schematic diagram of exosomes regulating EndoMT. Exosomes derived from tumor cells and immune cells in the tumor microenvironment may promote cancer metastasis by inducing EndoMT in microvascular endothelial cells. Additionally, in non-neoplastic diseases, exosomes from endothelial cells and immune cells can promote EndoMT, thereby damaging the barrier function of blood vessels

TAM, Tumor-related macrophages

Exosomes can regulate EndoMT in pathological environment

Exosomes regulate EndoMT in tumor tissue

EndoMT induced by exosomes plays a crucial role in the malignant progression of tumor invasion and metastasis. Recent studies have concentrated on malignant tumors of the digestive system, including hepatocellular carcinoma, colorectal cancer, pancreatic cancer and adenoid cystic carcinoma [8, 16,17,18,19,20]. Additionally, exosomes have been shown to promote EndoMT in nasopharyngeal carcinoma [7]. On one hand, exosomes secreted by cancer cells can directly act on endothelial cells, compromising the vascular endothelial barrier [7]. On the other hand, exosomes secreted by tumor tissues can circulate in fluid environments, such as bile and ascites, inducing EndoMT in certain blood vessels, thereby accelerating tumor progression or distant metastasis [8, 21]. Moreover, even within the same type of tumor, the ability of exosomes to regulate EndoMT can be heterogeneous. For example, HMGA2 protein in exosomes secreted by nasopharyngeal carcinoma (NPC) cells can upregulate the expression of the EndoMT-related transcription factor Snail, disrupt tight junctions between endothelial cells, and increase vascular permeability. But compared to EBV-negative NPC cells, exosomes enriched in HMGA2 protein predominantly originate from EBV-positive NPC cells [7]. This may be due to the impact of the virus itself on cancer cell functions, as viral infection significantly alters the proteome and RNA profiles encapsulated in exosomes, facilitating the spread of cytokines and RNAs from infected to uninfected cells (Table 1) [22,23,24,25,26,27,28,29,30,31]. Therefore, elucidating the impact of different viral infections on exosome production and secretion can further clarify their regulatory role in EndoMT. Additionally, elevated levels of TGF-β in the tumor microenvironment(TME) not only promote EndoMT but also induce EMT in tumor cells [32, 33]. Exosomes derived from EMT-induced tumor cells can likewise trigger EndoMT, and the degree of cellular transformation may be more pronounced compared to tumor cells that have not undergone EMT induction [34].

Table 1 Viral infections alter Exosome Communication in the tumor microenvironment

Additionally, previous studies have clearly demonstrated that the polarization state of macrophages affects cancer progression. M1 macrophages exhibit antitumor activity; in contrast, M2 macrophages promote tumor progression [35]. The tumor microenvironment (TME) skews macrophages toward a pro-tumor (M2) phenotype, regulating various stages of metastasis through the release of cytokines such as TGF-β, TNF-α, and IL-10 [36, 37]. The involvement of exosomes in tumor-associated macrophage (TAM)-mediated regulation of EndoMT, influencing cancer progression, has been increasingly recognized. M2 macrophage infiltration is closely associated with metastasis and poor prognosis in hepatocellular carcinoma (HCC). Exosomes from M2 macrophages are absorbed by HCC cells, promoting metastasis and epithelial-mesenchymal transition (EMT) [16]. These exosomes can also be internalized by endothelial cells, leading to the downregulation of tight junction proteins such as TJP-1, occludin, and claudin-5, thereby increasing vascular permeability and facilitating cancer cell metastasis [16]. Furthermore, miR-155 and miR-196a-5p are abundant in M2 TAMs and exosomes secreted by M2 TAMs. These microRNAs directly bind to the 3’-UTR of Ras association domain family member 4 (RASSF4), negatively regulating its expression and promoting the migration and invasion of non-small cell lung cancer (NSCLC) cells [38]. Although M1 macrophages are currently recognized for their antitumor activity, the role of exosome-mediated regulation in this process remains unclear. Further elucidation of how exosomes derived from different polarized macrophage types regulate EndoMT in various tumors is of critical importance for developing exosome-based interventions targeting distant metastasis in cancer.

Exosomes regulate EndoMT in non-tumor tissue

In non-tumor diseases, EndoMT is currently considered to be closely associated with disease progression, particularly in fibrosis and cardiovascular diseases such as atherosclerosis and vascular calcification [39,40,41,42]. Atherosclerosis is a widespread chronic inflammatory disease, and studies have shown that EndoMT is involved in the onset and development of atherosclerosis [39]. Various factors, including inflammation, shear stress, and oxidative stress, play roles in atherosclerosis and can also promote EndoMT [39, 43, 44]. In particular, inflammation involving cytokines such as TNF-α and IL-1β is a key factor in atherosclerosis, promoting EndoMT through both TGF-β-dependent and independent pathways [43]. Chronic inflammation caused by factors such as hypercholesterolemia or persistent stimuli can disrupt the protective cell signaling pathways of FGF signaling, thereby activating the EndoMT process. Once EndoMT is initiated, it exacerbates inflammation, creating a vicious cycle [45]. Vascular calcification (VC) is a pathological change in the atherosclerotic process that leads to arterial stiffening and reduced compliance [46]. Evidence suggests that BMP activation triggers EndoMT, leading endothelial cells transform into osteoblast-like cells, contributing to vascular calcification [47]. BMP is inhibited by matrix Gla protein (MGP), and the loss of MGP leads to rapid progressive calcification of vessels and abnormal endothelial cells [40]. The internal elastic lamina (IEL) is gradually degraded due to excessive proteolysis, causing ECs to lose their anchorage and become more susceptible to EndoMT [40, 48]. In MGP-deficient, diabetic, and atherosclerotic mice, enhanced BMP signaling triggers EndoMT, causing ECs to become more plastic and transition into osteoblast-like cells, thereby leading to vascular calcification [40,41,42]. Recent observations of EndoMT-related genes and molecular changes suggest that EndoMT also plays a critical role in the abnormal vascular development of cerebrovascular malformations. Increased EndoMT markers have been observed in cavernous malformations [49, 50]. Similarly, enhanced EndoMT-associated transcription factors (e.g., Snail, Twist) and mesenchymal markers have been shown in human brain arteriovenous malformations (bAVM) [51, 52]. Mutations may be involved in EndoMT and the development of bAVM. For example, a series of molecular analyses confirmed EndoMT characteristics in KRAS-overexpressing human umbilical vein endothelial cells (HUVECs), including increased mesenchymal markers and decreased EC markers [53, 54].

Significant progress has been made in understanding the impact of exosomes on EndoMT in non-tumor diseases, particularly in the context of diabetic complications such as diabetic retinopathy and diabetic nephropathy [55, 56]. By analyzing exosomes derived from the vitreous fluid of patients with proliferative diabetic retinopathy, it was found that these exosomes are enriched with long non-coding RNA LOC100132249. LOC100132249 functions as a competitive endogenous sponge for miR-199a-5p, thereby regulating the EndoMT process through the transcription factor SNAI1, leading to endothelial dysfunction [55]. Studies have confirmed that hyperglycemia itself can induce the EndoMT process by directly increasing both TGF-β-dependent and non-TGF-β signaling pathways. TGF-β signaling, as part of the EC metabolic memory, is activated by hyperglycemia and can lead to EndoMT in ECs even after the glucose levels are normalized in culture conditions [57]. Hyperglycemia can activate various pathways and pro-inflammatory factors that induce endothelial dysfunction. It triggers the non-canonical nuclear factor kappa-B (NF-κB) signaling pathway, resulting in the production of cytokines and chemokines that promote inflammation, which is a key factor in the occurrence of EndoMT [58]. However, research on the impact of hyperglycemia on exosome production and secretion is still lacking, and the precise mechanisms by which hyperglycemia regulates EndoMT progression via exosomes require further investigation.

In addition to the regulation by blood glucose, both direct physical trauma and secondary inflammatory responses can influence the EndoMT process through changes in exosomes [59, 60]. Oscillatory shear stress (OSS) can induce EndoMT in endothelial progenitor cells by promoting the release of exosomes enriched with circ-1199 [59]. These exosomes downregulate let-7 g-5p, which subsequently upregulates HMGA2, leading to the elevated expression of EndoMT-related transcription factors [59]. Research on spinal cord injury (SCI) has revealed the role of secondary injury in the development of EndoMT. After blood-spinal cord barrier disruption, M1-polarized macrophages accumulate at the injury site, playing a crucial role in the SCI process. In vitro studies have shown that exosomes from M1-polarized bone marrow-derived macrophages carry miR-155, which induces EndoMT in bEnd.3 cells through exosomal transport, exacerbating blood-spinal cord barrier damage [60]. In addition to acute injuries, chronic progressive cerebrovascular diseases of unknown etiology, such as Moyamoya disease, and congenital cerebrovascular anomalies, such as brain arteriovenous malformations (bAVMs), also involve EndoMT. Similarly, in these diseases, exosome-induced EndoMT is attributed to exosome-enriched RNA molecules [61, 62]. For example, a recent study reported that exosomal miR-3131 promotes EndoMT in KRAS-mutant bAVMs, suggesting that miR-3131 could be a potential biomarker and therapeutic target for KRAS-mutant bAVMs [62].

As mentioned above, exosomes derived from polarized macrophages carrying miR-155 exacerbate EndoMT in spinal cord injury, highlighting the critical role of macrophage-derived exosomes in the pathological process of vascular injury [60]. This mechanism differs from the regulation of EndoMT by polarized macrophages in tumor tissues, as discussed in Sect. 1.1. In the tumor microenvironment, M1 macrophages exhibit anti-tumor activity, negatively regulating the invasion and migration of tumor cells [36, 37]. However, in non-tumor tissues, M1 macrophages significantly promote the EndoMT process [63, 64]. Keith Q et al. demonstrated in their study on hemangioma regression mechanisms that M1-polarized, rather than M2-polarized, macrophages induce EndoMT in endothelial cells, promoting hemangioma regression, which could potentially serve as a novel therapeutic approach for this vascular anomaly [63]. Similarly, molecular studies on atherosclerosis have found that supernatants from M1 macrophage-derived foam cells (M1-FCs), but not from M2 macrophage-derived foam cells (M2-FCs), induce EndoMT. Specifically, M1-FCs upregulate CCL-4, which induces EndoMT, increases endothelial permeability, and promotes monocyte adhesion. This may further elucidate the pivotal role of EndoMT in the progression of atherosclerosis [65]. Therefore, a clearer understanding of the regulatory role of exosomes from polarized macrophages in vascular processes may contribute to the development of novel therapies for cardiovascular and fibrotic diseases.

Signaling pathways of exosomes regulating EndoMT

Signaling pathways of EndoMT

The signaling pathways involved in EndoMT are similar to those in epithelial-mesenchymal transition (EMT), with transforming growth factor-β (TGF-β) signaling being the most prominent [66,67,68,69,70]. TGF-β is a cytokine found in various cell types that regulates cell proliferation and differentiation, and its levels are elevated in immune diseases and inflammatory conditions [71]. Members of the TGF-β family, including TGF-β1, TGF-β2, and TGF-β3, bind to TGF-β receptors, initiating the EndoMT process in endothelial cells by activating downstream Smad-dependent and Smad-independent signaling pathways [72, 73] (Fig. 2). In the Smad-dependent pathway, Smad proteins, which are intracellular effectors of TGF-β signaling, are activated by the receptor and translocate to the nucleus to regulate transcription [73]. Besides TGF-β signaling, the Wnt/β-catenin signaling pathway also plays a role in the EndoMT process. Glycogen synthase kinase 3 beta (GSK3β) is a serine-threonine kinase that phosphorylates β-catenin. Wnt signal transduction inhibits GSK3β activity, thereby promoting the stability and function of the EndoMT transcription factor Snail1 [74]. Other pathways, such as the Notch pathway and the Hedgehog pathway, may also be involved in regulating the EndoMT process [75,76,77,78]. The signaling pathways involved in EndoMT are briefly summarized here, but they have been discussed in greater detail in previous reviews. For further information, refer to references [79,80,81].

Fig. 2
figure 2

Smad-dependent and Smad-independent TGFβ signaling in EndoMT. In the Smad-dependent signaling pathway, TGFβ binds to TGFβ receptor 1 or TGFβ receptor 2 on the cell membrane, promoting the phosphorylation of Smad2 and Smad3. Smad4 then forms a complex with phosphorylated Smad2 and Smad3, which is translocated into the nucleus, leading to the expression of EndoMT-related transcription factors and promoting the occurrence of EndoMT. In the Smad-independent signaling pathway, after TGFβ binds to its receptors, downstream signaling pathways such as PI3K, RAS, and MAPK are activated, resulting in the downregulation of endothelial-specific gene transcription and the upregulation of mesenchymal-specific gene expression, thereby promoting the occurrence of EndoMT

Mechanisms of exosome-mediated regulation of EndoMT

Non-coding RNAs include microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). These non-coding RNAs influence EndoMT pathways during both developmental and pathological processes [82]. As previously mentioned, in both tumor and non-tumor diseases, exosome-mediated regulation of EndoMT is often driven by the non-coding RNAs contained within exosomes [19, 55, 60, 61]. Therefore, this section will analyze the mechanisms by which exosomes regulate EndoMT through miRNAs, lncRNAs, and circRNAs.

MiRNAs are small RNA fragments typically 20–25 nucleotides long, which bind to complementary sequences within mRNA targets, inhibiting the translation of mRNA into proteins [83]. MiRNAs that have been confirmed in exosomes to regulate EndoMT include miR-155, miR-30a-5p, miR-3131, miR-218, miR-122-5p, miR-92a-3p, miRNA151a-3p, and miRNA125b-5p [60,61,62, 84,85,86,87] (Table 2). Among these, miR-155 is upregulated in bone marrow-derived macrophages (BMDMs) during traumatic spinal cord injury, where it activates the NF-κB signaling pathway by targeting suppressor of cytokine signaling 6 (SOCS6), inhibiting SOCS6-mediated ubiquitination and degradation of p65, and thus regulating the EndoMT process in vascular endothelial cells [60]. However, the regulation of SOCS6 by miR-155 is not unique. In non-exosome studies, miR-155-5p has been shown to influence EndoMT progression via multiple targets or pathways, including SIRT1, SHIP-1, and c-Ski [88,89,90]. miR-155-5p modulates the expression of SIRT1 and downstream targets Nrf2 and HO-1, affecting the production of reactive oxygen species (ROS) in endothelial cells, thereby regulating endothelial-mesenchymal transition in a type 2 diabetes mouse model and impacting wound healing [88]. In pulmonary fibrosis models, endothelial miR-155 plays a key role in the fibrotic response through EndoMT, with SHIP-1 as its target. The regulation of EndoMT by endothelial miR-155 and SHIP-1 involves multiple signaling pathways, including PI3K/AKT, STAT3, and SMAD/STAT pathways [89]. In studies of cardiac fibrosis, miR-155 has been found to regulate TGF-β-induced EndoMT by targeting c-Ski, suggesting that the miR-155/c-Ski axis could be a novel biomarker and therapeutic target for cardiac fibrosis [90]. Similar to miR-155, other miRNAs in exosomes that regulate EndoMT are also associated with multiple targets or pathways. Therefore, the regulation of EndoMT by the same miRNA within exosomes is likely mediated through multiple targets and mechanisms, which increases the complexity of elucidating the mechanisms by which exosomes regulate endothelial injury and the challenges of developing therapeutic interventions.

Table 2 Exosomal molecules with proven regulatory effects on EndoMT functions

When non-coding RNAs consist of more than 200 nucleotides, they are classified as lncRNAs. LncRNAs are involved in post-transcriptional regulation, controlling processes such as mRNA processing, stability, or translation. They can act as sponges to block the effects of miRNAs or serve as sources of other small RNAs [91]. Compared to miRNAs, there are fewer well-established lncRNAs that have been confirmed to regulate EndoMT via exosomes. Among them are SNHG7 and LOC100132249 [55, 92] (Table 2). LncRNA SNHG7 primarily functions in diabetic retinopathy. Specifically, Megakaryocytic leukemia 1 (MKL1), a transcription modulator, is highly expressed in high glucose-induced retinal epithelial cells and promotes EndoMT by activating TWIST1 [93, 94]. In 2022, Cao et al. discovered that SNHG7 can weaken MKL1 mRNA stability by interacting with the RNA-binding protein KHSRP. This SNHG7/KHSRP/MKL1 axis regulates endothelial-to-mesenchymal transition in diabetic retinopathy [95]. Consistently, MSC-derived exosomes highly express SNHG7, which can inhibit EndoMT and tube formation in diabetic retinopathy. However, unlike the mechanism targeting MKL1, exosomal SNHG7 from MSCs exerts its effects through the miR-34a-5p/XBP1 axis [92]. LncRNA LOC100132249, primarily derived from endothelial cells, is enriched in the vitreous of patients with proliferative diabetic retinopathy and has been shown to participate in angiogenesis. Mechanistically, LOC100132249 acts as a competitive endogenous sponge for miRNA-199a-5p, regulating the EndoMT initiator SNAI1 via activation of the Wnt/β-catenin pathway, ultimately leading to endothelial dysfunction [55].

CircRNAs represent another subclass of non-coding RNAs, typically formed by back-splicing. Similar to other lncRNAs, they function as miRNA sponges, RNA-binding protein sequestration factors, and can regulate gene expression by controlling mRNA transcription [96, 97]. As described in Sect. 1.2, oscillatory shear stress induces the release of circ-1199-enriched exosomes from endothelial progenitor cells, which downregulate let-7 g-5p, eventually activating p-Smad3/Smad3 and Snail to promote EndoMT in endothelial progenitor cells (Table 2) [59]. Beyond this, there are few circRNAs that have been directly confirmed to regulate EndoMT via exosomes. Notably, exosomal circDLGAP4 is involved in the pathological changes of vascular injury in diseases such as diabetic nephropathy and cerebral ischemia [98, 99]. Research on ischemic stroke has also revealed that circDLGAP4 regulates EndoMT related to blood-brain barrier integrity by targeting miR-143, thereby improving ischemic stroke outcomes [100]. Further research is needed to determine whether the effects of exosomal circDLGAP4 on vascular injury are mediated through the regulation of EndoMT in endothelial cells and to elucidate the specific mechanisms involved.

Exosome therapy for EndoMT

While EndoMT is an adverse factor in cardiovascular diseases, its reverse process plays an active role in repairing damaged myocardium. Exosomal miR-218-5p/miR-363-3p from endothelial progenitor cells improve myocardial infarction outcomes by targeting the p53/JMY signaling pathway. Further studies have shown that these microRNAs promote mesenchymal-to-endothelial transformation and inhibit myocardial fibrosis [101]. In the diseased state, exosomes can promote the progression of EndoMT and disrupt the barrier function of endothelial cells. However, exosomes from certain stem or progenitor cells can exert therapeutic effects by inhibiting the EndoMT process. Exosomes derived from human umbilical cord mesenchymal stem cells (hucMSCs) are rich in miR-218, which possesses anti-fibrotic properties and inhibits EndoMT through the MeCP2/BMP2 pathway, offering a novel approach to preventing pulmonary fibrosis [85]. In experimental models of pulmonary hypertension, hucMSC-Exos have been shown to effectively reduce right ventricular systolic blood pressure and right ventricular hypertrophy index, as well as inhibit pulmonary vascular remodeling and EndoMT processes [102]. Additionally, bone marrow-derived mesenchymal stem cells have demonstrated significant effects in inhibiting EndoMT. Exosomes derived from these cells, containing lncRNA SNHG7, negatively regulate miR-34a-5p and inhibit EndoMT in human retinal microvascular endothelial cells through the miR-34a-5p/XBP1 axis, providing new insights for the treatment of diabetic retinopathy [92].

Further modification of therapeutic exosomes or the construction of exosome-based drug delivery platforms as biocompatible carriers can achieve more precise regulation of EndoMT. Nintedanib, a triple tyrosine kinase inhibitor with anti-fibrotic and antioxidant properties, is used clinically to treat pulmonary fibrosis. Researchers have developed a system for delivering Nintedanib using adipose-derived stem cell exosomes, finding it effective in alleviating bleomycin-induced EndoMT and reducing oxidative stress levels in pulmonary fibrosis models, significantly enhancing therapeutic effects [103]. Moreover, modifying the surface of exosomes to improve their targeting in vivo is a promising research direction. For instance, exosomes from mouse aortic endothelial cells were cultured and extracted, then combined with DSPE-PEG and CD34 antibodies to form exosomal vectors (Exosome-DSPE-PEG-AbCD34) targeting endothelial cells. High expression of SMAD7 in endothelial cells can effectively reverse TGF-β1-mediated EndoMT. Loading a SMAD7 plasmid into Exosome-DSPE-PEG-AbCD34 achieved high targeting of exosomes to endothelial cells, significantly increased SMAD7 levels in these cells, decreased TGF-β1 expression, and effectively reversed TGF-β1-mediated EndoMT [104]. Therefore, the modifiability of exosome membranes enhances their advantages as delivery vectors and further boosts their ability to inhibit EndoMT.

Conclusions

Whether in the distant metastasis of tumor cells or in non-neoplastic diseases such as cardiovascular damage, the role of exosomes in stimulating endothelial cells to undergo EndoMT by transferring proteins and nucleic acids has become increasingly recognized by researchers. However, research on exosomal regulation of EndoMT remains relatively limited in recent years. Firstly, the techniques for isolating and purifying exosomes have not yet been standardized. Different methods can affect the purity and activity of exosomal components, thereby influencing research outcomes. The lack of a unified standard for exosome preparation presents obstacles for subsequent research and clinical applications. Secondly, in terms of the mechanisms by which exosomes regulate EndoMT, most studies focus on miRNA, with a lack of research on lncRNA and circRNA. Furthermore, how to achieve precise targeted delivery of exosomes, particularly across the blood-brain barrier (for brain diseases) or other organ barriers, remains a technical challenge. Current delivery technologies struggle to ensure the efficacy and stability of exosomes in vivo.

Thus, future researches need to promote the standardization of techniques for exosome isolation, purification, and qualitative analysis, particularly focusing on the stability and activity of exosomes to ensure reproducibility across different laboratories. Additionally, more attention should be paid to investigating changes in lncRNA and circRNA in exosomes and their mechanisms in regulating EndoMT, which could help identify new therapeutic targets and refine the functional models of exosomes. Furthermore, clarifying the safety, appropriate dosage, and delivery pathways of exosomes is essential to reduce risks in the drug development process and lay the foundation for future clinical translation. On the other hand, advances in exosome research rely on interdisciplinary collaboration, involving fields such as biology, nanotechnology, and drug delivery. Future studies should emphasize cross-disciplinary cooperation to explore new exosome modification technologies that enhance their targeted delivery efficiency and therapeutic effects in vivo. Through further investigation of the relationship between exosomes and EndoMT, new therapeutic strategies may emerge for cancer, cardiovascular diseases, fibrosis, and wound healing.

Data availability

Not applicable.

Abbreviations

EndoMT:

Endothelial-mesenchymal transformation

MVB:

Multivesicular bodies

EBV:

Epstein-Barr virus

KLF4:

Kruppel-like factor 4

MRPL23-AS1:

MRPL23 antisense RNA 1

TAM:

Tumor-related macrophages

SCI:

Spinal cord injury

TGF-β:

Transforming growth factor-β

GSK3β:

Glycogen synthase kinase 3 beta

hucMSC:

human umbilical cord mesenchymal stem cells

NPC:

Nasopharyngeal carcinoma

HCC:

Hepatocellular carcinoma

HBV:

Hepatitis B virus

HPV:

Human papillomavirus

NADCs:

Non-AIDS-defining cancers

BC:

Breast cancer

EGFR:

Epidermal growth factor receptor

TAR:

Transactivation response

ARID2:

AT-rich interactive domain

VAMP2:

Vesicle-associated membrane protein 2

SOCS:

Suppressor of cytokine signaling

TLR3:

Toll-like receptor 3

M1-BMDMs:

M1-polarized bone marrow-derived macrophages

BSCB:

Blood-spinal-cord-barrier

HG:

High glucose

GECs:

Glomerular endothelial cells

HPMEC:

Human pulmonary microvascular endothelial cells

ARDS:

Acute respiratory distress syndrome

MSCs:

Mesenchymal stem cells

HRMECs:

Human retinal microvascular endothelial cells

References

  1. Kovacic JC, Dimmeler S, Harvey RP, Finkel T, Aikawa E, Krenning G, Baker AH. Endothelial to mesenchymal transition in Cardiovascular Disease: JACC State-of-the-art review. J Am Coll Cardiol. 2019;73:190–209. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jacc.2018.09.089.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Piera-Velazquez S, Jimenez SA. Endothelial to mesenchymal transition: role in physiology and in the Pathogenesis of Human diseases. Physiol Rev. 2019;99:1281–324. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/physrev.00021.2018.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ranchoux B, Antigny F, Rucker-Martin C, Hautefort A, Péchoux C, Bogaard HJ, Dorfmüller P, Remy S, Lecerf F, Planté S, Chat S, Fadel E, Houssaini A, Anegon I, Adnot S, Simonneau G, Humbert M, Cohen-Kaminsky S, Perros F. Endothelial-to-mesenchymal transition in pulmonary hypertension. Circulation. 2015;131:1006–18. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/circulationaha.114.008750.

    Article  CAS  PubMed  Google Scholar 

  4. Chen PY, Qin L, Baeyens N, Li G, Afolabi T, Budatha M, Tellides G, Schwartz MA, Simons M. Endothelial-to-mesenchymal transition drives atherosclerosis progression. J Clin Invest. 2015;125:4514–28. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci82719.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Piera-Velazquez S, Mendoza FA, Jimenez SA. Endothelial to mesenchymal transition (EndoMT) in the Pathogenesis of Human Fibrotic diseases. J Clin Med. 2016;5. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/jcm5040045.

  6. Medici D, Shore EM, Lounev VY, Kaplan FS, Kalluri R, Olsen BR. Conversion of vascular endothelial cells into multipotent stem-like cells. Nat Med. 2010;16:1400–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nm.2252.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Li DK, Chen XR, Wang LN, Wang JH, Li JK, Zhou ZY, Li X, Cai LB, Zhong SS, Zhang JJ, Zeng YM, Zhang QB, Fu XY, Lyu XM, Li MY, Huang ZX, Yao KT. Exosomal HMGA2 protein from EBV-positive NPC cells destroys vascular endothelial barriers and induces endothelial-to-mesenchymal transition to promote metastasis. Cancer Gene Ther. 2022;29:1439–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41417-022-00453-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kimoto A, Kadoi Y, Tsuruda T, Kim YS, Miyoshi M, Nomoto Y, Nakata Y, Miyake M, Miyashita K, Shimizu K, Ajiki T, Hori Y. Exosomes in ascites from patients with human pancreatic cancer enhance remote metastasis partially through endothelial-mesenchymal transition. Pancreatology. 2023;23:377–88. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.pan.2023.04.002.

    Article  CAS  PubMed  Google Scholar 

  9. Gurung S, Perocheau D, Touramanidou L, Baruteau J. The exosome journey: from biogenesis to uptake and intracellular signalling. Cell Commun Signal. 2021;19:47. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12964-021-00730-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Maas SLN, Breakefield XO, Weaver AM. Extracellular vesicles: Unique Intercellular Delivery vehicles. Trends Cell Biol. 2017;27:172–88. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tcb.2016.11.003.

    Article  CAS  PubMed  Google Scholar 

  11. van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19:213–28. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrm.2017.125.

    Article  CAS  PubMed  Google Scholar 

  12. Lu X, Yao C, Sun L, Li Z. Plasmon-enhanced biosensors for microRNA analysis and cancer diagnosis. Biosens Bioelectron. 2022;203:114041. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bios.2022.114041.

    Article  CAS  PubMed  Google Scholar 

  13. Li X, Wu L, Tian X, Zheng W, Yuan M, Tian X, Zuo H, Song H, Shen Z. miR-29a-3p in Exosomes from Heme Oxygenase-1 modified bone marrow mesenchymal stem cells alleviates Steatotic Liver Ischemia-Reperfusion Injury in rats by suppressing ferroptosis via Iron responsive element binding protein 2. Oxid Med Cell Longev. 2022;2022(6520789). https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2022/6520789.

  14. Zhu D, Liu S, Huang K, Wang Z, Hu S, Li J, Li Z, Cheng K. Intrapericardial Exosome Therapy dampens Cardiac Injury via activating Foxo3. Circ Res. 2022;131:e135–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/circresaha.122.321384.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Shi G, Long Z, De la Vega RE, Behfar A, Moran SL, Evans C, Zhao C. Purified exosome product enhances chondrocyte survival and regeneration by modulating inflammation and promoting chondrogenesis. Regen Med. 2023;18:55–71. https://doiorg.publicaciones.saludcastillayleon.es/10.2217/rme-2022-0132.

    Article  CAS  PubMed  Google Scholar 

  16. Lu Y, Han G, Zhang Y, Zhang L, Li Z, Wang Q, Chen Z, Wang X, Wu J. M2 macrophage-secreted exosomes promote metastasis and increase vascular permeability in hepatocellular carcinoma. Cell Commun Signal. 2023;21:299. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12964-022-00872-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wang D, Wang X, Si M, Yang J, Sun S, Wu H, Cui S, Qu X, Yu X. Exosome-encapsulated miRNAs contribute to CXCL12/CXCR4-induced liver metastasis of colorectal cancer by enhancing M2 polarization of macrophages. Cancer Lett. 2020;474:36–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.canlet.2020.01.005.

    Article  CAS  PubMed  Google Scholar 

  18. Ma J, Wang P, Liu Y, Zhao L, Li Z, Xue Y. Krüppel-like factor 4 regulates blood-tumor barrier permeability via ZO-1, occludin and claudin-5. J Cell Physiol. 2014;229:916–26. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jcp.24523.

    Article  CAS  PubMed  Google Scholar 

  19. Liu K, Dou R, Yang C, Di Z, Shi D, Zhang C, Song J, Fang Y, Huang S, Xiang Z, Zhang W, Wang S, Xiong B. Exosome-transmitted miR-29a induces colorectal cancer metastasis by destroying the vascular endothelial barrier. Carcinogenesis. 2023;44:356–67. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/carcin/bgad013.

    Article  CAS  PubMed  Google Scholar 

  20. Chen CW, Fu M, Du ZH, Zhao F, Yang WW, Xu LH, Li SL, Ge XY. Long noncoding RNA MRPL23-AS1 promotes adenoid cystic carcinoma lung metastasis. Cancer Res. 2020;80:2273–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.Can-19-0819.

    Article  CAS  PubMed  Google Scholar 

  21. Shu L, Li X, Liu Z, Li K, Shi A, Tang Y, Zhao L, Huang L, Zhang Z, Zhang D, Huang S, Lian S, Sheng G, Yan Z, Zhang Z, Xu Y. Bile exosomal miR-182/183-5p increases cholangiocarcinoma stemness and progression by targeting HPGD and increasing PGE2 generation. Hepatology. 2024;79:307–22. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/hep.0000000000000437.

    Article  PubMed  Google Scholar 

  22. Xu G, Shi X, Liu H, Shen C, Yang B, Zhang T, Chen X, Zhao D, Yang J, Hao Y, Cui H, Yuan X, Liu X, Zhang K, Zheng H. Functional analysis and Proteomics Profiling of Extracellular vesicles from swine plasma infected by African swine fever virus. Front Cell Infect Microbiol. 2022;12:809135. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fcimb.2022.809135.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Schwab A, Meyering SS, Lepene B, Iordanskiy S, van Hoek ML, Hakami RM, Kashanchi F. Extracellular vesicles from infected cells: potential for direct pathogenesis. Front Microbiol. 2015;6:1132. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmicb.2015.01132.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Aga M, Bentz GL, Raffa S, Torrisi MR, Kondo S, Wakisaka N, Yoshizaki T, Pagano JS, Shackelford J. Exosomal HIF1α supports invasive potential of nasopharyngeal carcinoma-associated LMP1-positive exosomes. Oncogene. 2014;33:4613–22. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/onc.2014.66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Li H, Chi X, Li R, Ouyang J, Chen Y. HIV-1-infected cell-derived exosomes promote the growth and progression of cervical cancer. Int J Biol Sci. 2019;15:2438–47. https://doiorg.publicaciones.saludcastillayleon.es/10.7150/ijbs.38146.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wei XC, Xia YR, Zhou P, Xue X, Ding S, Liu LJ, Zhu F. Hepatitis B core antigen modulates exosomal miR-135a to target vesicle-associated membrane protein 2 promoting chemoresistance in hepatocellular carcinoma. World J Gastroenterol. 2021;27:8302–22. https://doiorg.publicaciones.saludcastillayleon.es/10.3748/wjg.v27.i48.8302.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhao X, Sun L, Mu T, Yi J, Ma C, Xie H, Liu M, Tang H. An HBV-encoded miRNA activates innate immunity to restrict HBV replication. J Mol Cell Biol. 2020;12:263–76. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/jmcb/mjz104.

    Article  CAS  PubMed  Google Scholar 

  28. Dochi H, Kondo S, Komura S, Moriyama-Kita M, Komori T, Nanbo A, Sakaguchi M, Fukuyo M, Hamabe-Horiike T, Tanaka M, Mizokami H, Kano M, Kitagawa Y, Kobayashi E, Hirai N, Ueno T, Nakanishi Y, Endo K, Sugimoto H, Hanayama R, Kaneda A, Yoshizaki T. Peritumoral SPARC expression induced by exosomes from nasopharyngeal carcinoma infected Epstein-Barr virus: a poor prognostic marker. Int J Cancer. 2024;154:895–911. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ijc.34777.

    Article  CAS  PubMed  Google Scholar 

  29. Meckes DG Jr., Shair KH, Marquitz AR, Kung CP, Edwards RH, Raab-Traub N. Human tumor virus utilizes exosomes for intercellular communication. Proc Natl Acad Sci U S A. 2010;107:20370–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.1014194107.

    Article  PubMed  PubMed Central  Google Scholar 

  30. De Carolis S, Storci G, Ceccarelli C, Savini C, Gallucci L, Sansone P, Santini D, Seracchioli R, Taffurelli M, Fabbri F, Romani F, Compagnone G, Giuliani C, Garagnani P, Bonafè M, Cricca M. HPV DNA associates with breast Cancer malignancy and it is transferred to breast Cancer stromal cells by Extracellular vesicles. Front Oncol. 2019;9:860. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fonc.2019.00860.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Chen L, Feng Z, Yue H, Bazdar D, Mbonye U, Zender C, Harding CV, Bruggeman L, Karn J, Sieg SF, Wang B, Jin G. Exosomes derived from HIV-1-infected cells promote growth and progression of cancer via HIV TAR RNA. Nat Commun. 2018;9:4585. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-018-07006-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Takahashi K, Akatsu Y, Podyma-Inoue KA, Matsumoto T, Takahashi H, Yoshimatsu Y, Koinuma D, Shirouzu M, Miyazono K, Watabe T. Targeting all transforming growth factor-β isoforms with an fc chimeric receptor impairs tumor growth and angiogenesis of oral squamous cell cancer. J Biol Chem. 2020;295:12559–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.RA120.012492.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Miyazono K, Katsuno Y, Koinuma D, Ehata S, Morikawa M. Intracellular and extracellular TGF-β signaling in cancer: some recent topics. Front Med. 2018;12:387–411. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11684-018-0646-8.

    Article  PubMed  Google Scholar 

  34. Kobayashi M, Fujiwara K, Takahashi K, Yoshioka Y, Ochiya T, Podyma-Inoue KA, Watabe T. Transforming growth factor-β-induced secretion of extracellular vesicles from oral cancer cells evokes endothelial barrier instability via endothelial-mesenchymal transition. Inflamm Regen. 2022;42:38. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s41232-022-00225-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Liu J, Geng X, Hou J, Wu G. New insights into M1/M2 macrophages: key modulators in cancer progression. Cancer Cell Int. 2021;21:389. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12935-021-02089-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ricketts TD, Prieto-Dominguez N, Gowda PS, Ubil E. Mechanisms of macrophage plasticity in the Tumor Environment: manipulating activation state to improve outcomes. Front Immunol. 2021;12:642285. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2021.642285.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lin Y, Xu J, Lan H. Tumor-associated macrophages in tumor metastasis: biological roles and clinical therapeutic applications. J Hematol Oncol. 2019;12:76. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-019-0760-3.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Li X, Chen Z, Ni Y, Bian C, Huang J, Chen L, Xie X, Wang J. Tumor-associated macrophages secret exosomal miR-155 and miR-196a-5p to promote metastasis of non-small-cell lung cancer. Transl Lung Cancer Res. 2021;10:1338–54. https://doiorg.publicaciones.saludcastillayleon.es/10.21037/tlcr-20-1255.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Huang Q, Gan Y, Yu Z, Wu H, Zhong Z. Endothelial to mesenchymal transition: an insight in atherosclerosis. Front Cardiovasc Med. 2021;8:734550. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fcvm.2021.734550.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Yao J, Guihard PJ, Blazquez-Medela AM, Guo Y, Moon JH, Jumabay M, Boström KI, Yao Y. Serine protease activation essential for endothelial-mesenchymal transition in vascular calcification. Circ Res. 2015;117:758–69. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/circresaha.115.306751.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Yao Y, Jumabay M, Ly A, Radparvar M, Cubberly MR, Boström KI. A role for the endothelium in vascular calcification. Circ Res. 2013;113:495–504. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/circresaha.113.301792.

    Article  CAS  PubMed  Google Scholar 

  42. Boström KI, Jumabay M, Matveyenko A, Nicholas SB, Yao Y. Activation of vascular bone morphogenetic protein signaling in diabetes mellitus. Circ Res. 2011;108:446–57. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/circresaha.110.236596.

    Article  PubMed  Google Scholar 

  43. Chen PY, Schwartz MA, Simons M. Endothelial-to-mesenchymal transition, vascular inflammation, and atherosclerosis. Front Cardiovasc Med. 2020;7:53. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fcvm.2020.00053.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhou M, Yu Y, Chen R, Liu X, Hu Y, Ma Z, Gao L, Jian W, Wang L. Wall shear stress and its role in atherosclerosis. Front Cardiovasc Med. 2023;10:1083547. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fcvm.2023.1083547.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Schwartz MA, Vestweber D, Simons M. A unifying concept in vascular health and disease. Science. 2018;360:270–1. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.aat3470.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Jiang H, Li L, Zhang L, Zang G, Sun Z, Wang Z. Role of endothelial cells in vascular calcification. Front Cardiovasc Med. 2022;9:895005. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fcvm.2022.895005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Yao J, Wu X, Qiao X, Zhang D, Zhang L, Ma JA, Cai X, Boström KI, Yao Y. Shifting osteogenesis in vascular calcification. JCI Insight. 2021;6. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci.insight.143023.

  48. Zhang L, Yao J, Yao Y, Boström KI. Contributions of the endothelium to vascular calcification. Front Cell Dev Biol. 2021;9:620882. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fcell.2021.620882.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Malinverno M, Maderna C, Abu Taha A, Corada M, Orsenigo F, Valentino M, Pisati F, Fusco C, Graziano P, Giannotta M, Yu QC, Zeng YA, Lampugnani MG, Magnusson PU, Dejana E. Endothelial cell clonal expansion in the development of cerebral cavernous malformations. Nat Commun. 2019;10:2761. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-019-10707-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Bravi L, Malinverno M, Pisati F, Rudini N, Cuttano R, Pallini R, Martini M, Larocca LM, Locatelli M, Levi V, Bertani GA, Dejana E, Lampugnani MG. Endothelial cells lining sporadic cerebral cavernous malformation Cavernomas Undergo endothelial-to-mesenchymal transition. Stroke. 2016;47:886–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/strokeaha.115.011867.

    Article  PubMed  Google Scholar 

  51. Shoemaker LD, McCormick AK, Allen BM, Chang SD. Evidence for endothelial-to-mesenchymal transition in human brain arteriovenous malformations. Clin Transl Med. 2020;10:e99. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ctm2.99.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Li H, Nam Y, Huo R, Fu W, Jiang B, Zhou Q, Song D, Yang Y, Jiao Y, Weng J, Yan Z, Di L, Li J, Wang J, Xu H, Wang S, Zhao J, Wen Z, Wang J, Cao Y. De Novo Germline and somatic variants Convergently promote endothelial-to-mesenchymal transition in Simplex Brain Arteriovenous Malformation. Circ Res. 2021;129:825–39. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/circresaha.121.319004.

    Article  CAS  PubMed  Google Scholar 

  53. Nikolaev SI, Vetiska S, Bonilla X, Boudreau E, Jauhiainen S, Rezai Jahromi B, Khyzha N, DiStefano PV, Suutarinen S, Kiehl TR, Mendes Pereira V, Herman AM, Krings T, Andrade-Barazarte H, Tung T, Valiante T, Zadeh G, Tymianski M, Rauramaa T, Ylä-Herttuala S, Wythe JD, Antonarakis SE, Frösen J, Fish JE, Radovanovic I. Somatic activating KRAS mutations in arteriovenous malformations of the brain. N Engl J Med. 2018;378:250–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa1709449.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Xu H, Huo R, Li H, Jiao Y, Weng J, Wang J, Yan Z, Zhang J, Zhao S, He Q, Sun Y, Wang S, Cao Y. KRAS mutation-induced EndMT of brain arteriovenous malformation is mediated through the TGF-β/BMP-SMAD4 pathway. Stroke Vasc Neurol. 2023;8:197–206. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/svn-2022-001700.

    Article  PubMed  Google Scholar 

  55. Hu Z, Wang J, Pan T, Li X, Tao C, Wu Y, Wang X, Zhang Z, Liu Y, Zhang W, Xu C, Wu X, Gu Q, Fan Y, Qian H, Mugisha A, Yuan S, Liu Q, Xie P. The exosome-transmitted lncRNA LOC100132249 induces endothelial dysfunction in Diabetic Retinopathy. Diabetes. 2023;72:1307–19. https://doiorg.publicaciones.saludcastillayleon.es/10.2337/db22-0435.

    Article  CAS  PubMed  Google Scholar 

  56. Li B, Sun G, Yu H, Meng J, Wei F. Exosomal circTAOK1 contributes to diabetic kidney disease progression through regulating SMAD3 expression by sponging miR-520 h. Int Urol Nephrol. 2022;54:2343–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11255-022-03139-y.

    Article  CAS  PubMed  Google Scholar 

  57. Yao Y, Song Q, Hu C, Da X, Yu Y, He Z, Xu C, Chen Q, Wang QK. Endothelial cell metabolic memory causes cardiovascular dysfunction in diabetes. Cardiovasc Res. 2022;118:196–211. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/cvr/cvab013.

    Article  CAS  PubMed  Google Scholar 

  58. Meyerovich K, Ortis F, Cardozo AK. The non-canonical NF-κB pathway and its contribution to β-cell failure in diabetes. J Mol Endocrinol. 2018;61:F1–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1530/jme-16-0183.

    Article  CAS  PubMed  Google Scholar 

  59. Li L, Wen J, Li H, He Y, Cui X, Zhang X, Guan X, Li Z, Cheng M. Exosomal circ-1199 derived from EPCs exposed to oscillating shear stress acts as a sponge of let-7 g-5p to promote endothelial-mesenchymal transition of EPCs by increasing HMGA2 expression. Life Sci. 2023;312:121223. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lfs.2022.121223.

    Article  CAS  PubMed  Google Scholar 

  60. Ge X, Tang P, Rong Y, Jiang D, Lu X, Ji C, Wang J, Huang C, Duan A, Liu Y, Chen X, Chen X, Xu Z, Wang F, Wang Z, Li X, Zhao W, Fan J, Liu W, Yin G, Cai W. Exosomal miR-155 from M1-polarized macrophages promotes EndoMT and impairs mitochondrial function via activating NF-κB signaling pathway in vascular endothelial cells after traumatic spinal cord injury. Redox Biol. 2021;41:101932. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.redox.2021.101932.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Liu J, Chen C, Qin X, Wang J, Zhang B, Jin F. Plasma-derived exosomes contributes to endothelial-to-mesenchymal transition in Moyamoya disease. Heliyon. 2024;10:e26748. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.heliyon.2024.e26748.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. He Q, Huo R, Wang J, Xu H, Zhao S, Zhang J, Sun Y, Jiao Y, Weng J, Zhao J, Cao Y. Exosomal miR-3131 derived from endothelial cells with KRAS mutation promotes EndMT by targeting PICK1 in brain arteriovenous malformations. CNS Neurosci Ther. 2023;29:1312–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cns.14103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Wu KQ, Muratore CS, So EY, Sun C, Dubielecka PM, Reginato AM, Liang OD. M1 Macrophage-Induced endothelial-to-mesenchymal transition promotes infantile Hemangioma regression. Am J Pathol. 2017;187:2102–11. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ajpath.2017.05.014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Gui Z, Zhang X, Han Q, Hang Z, Tan R, Gu M, Wang Z. Macrophage polarization induces endothelium-to-myofibroblast transition in chronic allograft dysfunction. Ren Fail. 2023;45:2220418. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/0886022x.2023.2220418.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Yang Y, Luo NS, Ying R, Xie Y, Chen JY, Wang XQ, Gu ZJ, Mai JT, Liu WH, Wu MX, Chen ZT, Fang YB, Zhang HF, Zuo ZY, Wang JF, Chen YX. Macrophage-derived foam cells impair endothelial barrier function by inducing endothelial-mesenchymal transition via CCL-4. Int J Mol Med. 2017;40:558–68. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/ijmm.2017.3034.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Zhao S, Song C, Chen F, Li M. LncRNA XIST/miR-455-3p/HOXC4 axis promotes breast cancer development by activating TGF-β/SMAD signaling pathway. Funct Integr Genomics. 2024;24:159. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10142-024-01442-8.

    Article  CAS  PubMed  Google Scholar 

  67. Lee JH, Sánchez-Rivera FJ, He L, Basnet H, Chen FX, Spina E, Li L, Torner C, Chan JE, Yarlagadda DVK, Park JS, Sussman C, Rudin CM, Lowe SW, Tammela T, Macias MJ, Koche RP, Massagué J. TGF-β and RAS jointly unmask primed enhancers to drive metastasis. Cell. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2024.08.014.

    Article  PubMed  PubMed Central  Google Scholar 

  68. You Z, Hu Z, Hou C, Ma C, Xu X, Zheng Y, Sun X, Ke Y, Liang J, Xie Z, Shu L, Liu Y. FABP4 facilitates epithelial-mesenchymal transition via elevating CD36 expression in glioma cells. Neoplasia. 2024;57:101050. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neo.2024.101050.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Zhang W, Gonzalez L, Li X, Bai H, Li Z, Taniguchi R, Langford J, Ohashi Y, Thaxton C, Yukihiko A, Yatsula B, Martin K, Goodwin J, Tellides G, Long X, Shu C, Dardik A. Endothelial TGF-β signaling regulates endothelial-mesenchymal transition during Arteriovenous Fistula Remodeling in mice with chronic kidney disease. Arterioscler Thromb Vasc Biol. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/atvbaha.124.320933.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Vuong CK, Fukushige M, Ngo NH, Yamashita T, Obata-Yasuoka M, Hamada H, Osaka M, Tsukada T, Hiramatsu Y, Ohneda O. Extracellular vesicles derived from type 2 Diabetic Mesenchymal stem cells induce endothelial mesenchymal transition under high glucose conditions through the TGFβ/Smad3 signaling pathway. Stem Cells Dev. 2024;33:262–75. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/scd.2023.0262.

    Article  CAS  PubMed  Google Scholar 

  71. Makinde T, Murphy RF, Agrawal DK. The regulatory role of TGF-beta in airway remodeling in asthma. Immunol Cell Biol. 2007;85:348–56. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sj.icb.7100044.

    Article  CAS  PubMed  Google Scholar 

  72. Pardali E, Sanchez-Duffhues G, Gomez-Puerto MC, Ten Dijke P. TGF-β-Induced endothelial-mesenchymal transition in Fibrotic diseases. Int J Mol Sci. 2017;18. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms18102157.

  73. Derynck R, Zhang YE. Smad-dependent and smad-independent pathways in TGF-beta family signalling. Nature. 2003;425:577–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature02006.

    Article  CAS  PubMed  Google Scholar 

  74. Zhou BP, Deng J, Xia W, Xu J, Li YM, Gunduz M, Hung MC. Dual regulation of snail by GSK-3beta-mediated phosphorylation in control of epithelial-mesenchymal transition. Nat Cell Biol. 2004;6:931–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ncb1173.

    Article  CAS  PubMed  Google Scholar 

  75. Chang AC, Garside VC, Fournier M, Smrz J, Vrljicak P, Umlandt P, Fuller M, Robertson G, Zhao Y, Tam A, Jones SJ, Marra MA, Hoodless PA, Karsan A. A notch-dependent transcriptional hierarchy promotes mesenchymal transdifferentiation in the cardiac cushion. Dev Dyn. 2014;243:894–905. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/dvdy.24127.

    Article  CAS  PubMed  Google Scholar 

  76. Timmerman LA, Grego-Bessa J, Raya A, Bertrán E, Pérez-Pomares JM, Díez J, Aranda S, Palomo S, McCormick F, Izpisúa-Belmonte JC, de la Pompa JL. Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev. 2004;18:99–115. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/gad.276304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2014;15:178–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrm3758.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Li X, Deng W, Nail CD, Bailey SK, Kraus MH, Ruppert JM, Lobo-Ruppert SM. Snail induction is an early response to Gli1 that determines the efficiency of epithelial transformation. Oncogene. 2006;25:609–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sj.onc.1209077.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Ciszewski WM, Wawro ME, Sacewicz-Hofman I, Sobierajska K. Cytoskeleton reorganization in EndMT-The role in Cancer and Fibrotic diseases. Int J Mol Sci. 2021;22. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms222111607.

  80. Derada Troletti C, de Goede P, Kamermans A, de Vries HE. Molecular alterations of the blood–brain barrier under inflammatory conditions: the role of endothelial to mesenchymal transition. Biochimica et Biophysica Acta (BBA) -. Mol Basis Disease. 2016;1862:452–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbadis.2015.10.010.

    Article  CAS  Google Scholar 

  81. Derynck R, Zhang YE. Smad-dependent and smad-independent pathways in TGF-β family signalling. Nature. 2003;425:577–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature02006.

    Article  CAS  PubMed  Google Scholar 

  82. Xu S, Kamato D, Little PJ, Nakagawa S, Pelisek J, Jin ZG. Targeting epigenetics and non-coding RNAs in atherosclerosis: from mechanisms to therapeutics. Pharmacol Ther. 2019;196:15–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.pharmthera.2018.11.003.

    Article  CAS  PubMed  Google Scholar 

  83. Gu S, Kay MA. (2010) How do miRNAs mediate translational repression? Silence 1:11. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1758-907x-1-11

  84. Ning Y, Zhou X, Wang G, Zhang L, Wang J. Exosome miR-30a-5p regulates glomerular endothelial cells’ EndMT and angiogenesis by modulating Notch1/VEGF signaling pathway. Curr Gene Ther. 2024;24:159–77. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/0115665232258527230919071328.

    Article  CAS  PubMed  Google Scholar 

  85. Zhao Y, Du L, Sun J, Wang X, Cong Z, Chen S, Wang F, Li Z. Exosomal miR-218 derived from mesenchymal stem cells inhibits endothelial-to-mesenchymal transition by epigenetically modulating of BMP2 in pulmonary fibrosis. Cell Biol Toxicol. 2023;39:2919–36. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10565-023-09810-z.

    Article  CAS  PubMed  Google Scholar 

  86. Qi D, Deng W, Chen X, Fan S, Peng J, Tang X, Wang D, Yu Q. (2022) Adipose-Derived Circulating Exosomes Promote Protection of the Pulmonary Endothelial Barrier by Inhibiting EndMT and Oxidative Stress through Down-Regulation of the TGF-β Pathway: A Potential Explanation for the Obesity Paradox in ARDS. Oxid Med Cell Longev 2022:5475832. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2022/5475832

  87. Yamada NO, Heishima K, Akao Y, Senda T. Extracellular vesicles containing MicroRNA-92a-3p facilitate partial endothelial-mesenchymal transition and angiogenesis in endothelial cells. Int J Mol Sci. 2019;20. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms20184406.

  88. Zhang Y, Hei F, Xiao Y, Liu Y, Han J, Hu D, Wang H. Acidic fibroblast growth factor inhibits reactive oxygen species-induced epithelial-mesenchymal transdifferentiation in vascular endothelial cells via the miR-155-5p/SIRT1/Nrf2/HO-1 pathway to promote wound healing in diabetic mice. Burns Trauma. 2024;12:tkae010. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/burnst/tkae010.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Tang H, Mao J, Ye X, Zhang F, Kerr WG, Zheng T, Zhu Z. SHIP-1, a target of miR-155, regulates endothelial cell responses in lung fibrosis. Faseb j. 2020;34:2011–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1096/fj.201902063R.

    Article  CAS  PubMed  Google Scholar 

  90. Wang J, He W, Xu X, Guo L, Zhang Y, Han S, Shen D. The mechanism of TGF-β/miR-155/c-Ski regulates endothelial-mesenchymal transition in human coronary artery endothelial cells. Biosci Rep. 2017;37. https://doiorg.publicaciones.saludcastillayleon.es/10.1042/bsr20160603.

  91. Kung JT, Colognori D, Lee JT. Long noncoding RNAs: past, present, and future. Genetics. 2013;193:651–69. https://doiorg.publicaciones.saludcastillayleon.es/10.1534/genetics.112.146704.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Cao X, Xue LD, Di Y, Li T, Tian YJ, Song Y. MSC-derived exosomal lncRNA SNHG7 suppresses endothelial-mesenchymal transition and tube formation in diabetic retinopathy via miR-34a-5p/XBP1 axis. Life Sci. 2021;272:119232. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lfs.2021.119232.

    Article  CAS  PubMed  Google Scholar 

  93. Li Z, Chen B, Dong W, Kong M, Fan Z, Yu L, Wu D, Lu J, Xu Y. MKL1 promotes endothelial-to-mesenchymal transition and liver fibrosis by activating TWIST1 transcription. Cell Death Dis. 2019;10:899. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-019-2101-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Ding Y, Xu H, Li L, Yuan Y, Xu Y. Megakaryocytic leukemia 1 (MKL1) mediates high glucose induced epithelial-mesenchymal transition by activating LOX transcription. Biochem Biophys Res Commun. 2019;509:633–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbrc.2018.12.024.

    Article  CAS  PubMed  Google Scholar 

  95. Cao X, Song Y, Huang LL, Tian YJ, Wang XL, Hua LY. M(6)a transferase METTL3 regulates endothelial-mesenchymal transition in diabetic retinopathy via lncRNA SNHG7/KHSRP/MKL1 axis. Genomics. 2022;114:110498. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ygeno.2022.110498.

    Article  CAS  PubMed  Google Scholar 

  96. Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, Zhong G, Yu B, Hu W, Dai L, Zhu P, Chang Z, Wu Q, Zhao Y, Jia Y, Xu P, Liu H, Shan G. Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol. 2015;22:256–64. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nsmb.2959.

    Article  CAS  PubMed  Google Scholar 

  97. Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK, Kjems J. Natural RNA circles function as efficient microRNA sponges. Nature. 2013;495:384–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature11993.

    Article  CAS  PubMed  Google Scholar 

  98. Bai S, Xiong X, Tang B, Ji T, Li X, Qu X, Li W. Exosomal circ_DLGAP4 promotes diabetic kidney disease progression by sponging miR-143 and targeting ERBB3/NF-κB/MMP-2 axis. Cell Death Dis. 2020;11:1008. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-020-03169-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Feng J, He W, Xia J, Huang Q, Yang J, Gu WP, Zhang N, Liu YH. Human umbilical cord mesenchymal stem cells-derived exosomal circDLGAP4 promotes angiogenesis after cerebral ischemia-reperfusion injury by regulating miR-320/KLF5 axis. Faseb j. 2023;37:e22733. https://doiorg.publicaciones.saludcastillayleon.es/10.1096/fj.202201488R.

    Article  CAS  PubMed  Google Scholar 

  100. Bai Y, Zhang Y, Han B, Yang L, Chen X, Huang R, Wu F, Chao J, Liu P, Hu G, Zhang JH, Yao H. Circular RNA DLGAP4 ameliorates ischemic stroke outcomes by targeting miR-143 to regulate endothelial-mesenchymal transition Associated with Blood-Brain Barrier Integrity. J Neurosci. 2018;38:32–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1523/jneurosci.1348-17.2017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Ke X, Yang R, Wu F, Wang X, Liang J, Hu X, Hu C. (2021) Exosomal miR-218-5p/miR-363-3p from Endothelial Progenitor Cells Ameliorate Myocardial Infarction by Targeting the p53/JMY Signaling Pathway. Oxid Med Cell Longev 2021:5529430. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2021/5529430

  102. Ge L, Jiang W, Zhang S, Wang J, Xin Q, Sun C, Li K, Qi T, Luan Y. Mesenchymal stromal cell-derived Exosomes Attenuate Experimental Pulmonary arterial hypertension. Curr Pharm Biotechnol. 2021;22:1654–62. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/1389201022666201231113127.

    Article  CAS  PubMed  Google Scholar 

  103. Cai L, Wang J, Yi X, Yu S, Wang C, Zhang L, Zhang X, Cheng L, Ruan W, Dong F, Su P, Shi Y. Nintedanib-loaded exosomes from adipose-derived stem cells inhibit pulmonary fibrosis induced by bleomycin. Pediatr Res. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41390-024-03024-7.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Wei Z, Zhao Y, Hsu P, Guo S, Zhang C, Zhong B. Exosomes for gene therapy effectively inhibit the endothelial-mesenchymal transition in mouse aortic endothelial cells. BMC Musculoskelet Disord. 2021;22:1000. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12891-021-04896-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors declare that they have not used Artificial Intelligence in this study.

Funding

This work was supported by the National High Level Hospital Clinical Research Funding(2022-PUMCH-C-032), the National Key R&D Program of China (2018YFA0108600), the CAMS Initiative for Innovative Medicine (2021-1-I2M-019), and National High Level Hospital Clinical Research Funding (2022-PUMCH-C-042).

Author information

Authors and Affiliations

Authors

Contributions

Data acquisition, analysis, and writing, SS and GL; review and editing, LP; project administration and funding acquisition, WJ and BX.

Corresponding authors

Correspondence to Bao Xinjie or Wei Junji.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sishuai, S., Lingui, G., Pengtao, L. et al. Advances in regulating endothelial-mesenchymal transformation through exosomes. Stem Cell Res Ther 15, 391 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-024-04010-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-024-04010-w

Keywords