Next Article in Journal
Stabilization of Keratinocyte Monolayer Integrity in the Presence of Anti-Desmoglein-3 Antibodies through FcRn Blockade with Efgartigimod: Novel Treatment Paradigm for Pemphigus?
Next Article in Special Issue
MicroRNA as a Potential Therapeutic Molecule in Cancer
Previous Article in Journal
Batch Effects during Human Bone Marrow Stromal Cell Propagation Prevail Donor Variation and Culture Duration: Impact on Genotype, Phenotype and Function
Previous Article in Special Issue
miRNA-Dependent Regulation of AKT1 Phosphorylation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Noncoding RNAs of Extracellular Vesicles in Tumor Angiogenesis: From Biological Functions to Clinical Significance

1
School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
2
Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
3
Watford General Hospital, Watford Herts WD18 0HB, UK
4
Institute of Life Science, Yinfeng Biological Group, Jinan 250000, China
5
Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
6
Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
7
NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this paper.
Cells 2022, 11(6), 947; https://doi.org/10.3390/cells11060947
Submission received: 14 February 2022 / Revised: 6 March 2022 / Accepted: 8 March 2022 / Published: 10 March 2022
(This article belongs to the Special Issue Regulatory Roles of Non-coding RNAs in Cancer)

Abstract

:
Extracellular vesicles (EVs) act as multifunctional regulators of intercellular communication and are involved in diverse tumor phenotypes, including tumor angiogenesis, which is a highly regulated multi-step process for the formation of new blood vessels that contribute to tumor proliferation. EVs induce malignant transformation of distinct cells by transferring DNAs, proteins, lipids, and RNAs, including noncoding RNAs (ncRNAs). However, the functional relevance of EV-derived ncRNAs in tumor angiogenesis remains to be elucidated. In this review, we summarized current research progress on the biological functions and underlying mechanisms of EV-derived ncRNAs in tumor angiogenesis in various cancers. In addition, we comprehensively discussed the potential applications of EV-derived ncRNAs as cancer biomarkers and novel therapeutic targets to tailor anti-angiogenic therapy.

1. Introduction

Under physiological and pathological conditions, almost all types of cells can secrete membrane vesicles collectively referred to as small extracellular vesicles (sEVs) and large EVs (lEVs) [1,2]. Exosomes, small EVs, play a crucial role in intercellular communication by transporting proteins, mRNAs, and ncRNAs [2]. In the past decade, ncRNAs, including microRNAs (miRNAs), PIWI-interacting RNAs (piRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), have been intensively studied in many human diseases, particularly cancers [3,4,5,6]. Studies have indicated that exosomal ncRNAs acting as versatile regulators at the transcriptional and post-transcriptional levels may contribute to cancer progression and metastasis [7,8]. Recently, it was found that EV-derived ncRNAs exhibit a diverse range of biological functions in cancer hallmarks, including tumor angiogenesis.
Angiogenesis, a multi-step process involving the formation of new blood vessels from existing vasculature, occurs during the natural growth, development, and progression of diseases, such as solid cancer [9]. Tumor angiogenesis participates in the whole process of tumorigenesis, proliferation, and metastasis, from the initial stages of pre-tumor to carcinoma in situ and, subsequently, to malignant tumor [10]. Proliferation and metastasis of tumor cells both depend on the adequate supply of oxygen and nutrients and the elimination of waste products [11]. Newly formed blood vessels can fulfil this requirement. Angiogenesis is caused by the imbalance between pro- and anti-angiogenic factors resulting from the excessive production of vascular endothelial growth factor (VEGF) induced by hypoxia [12]. Studies have reported that EV-derived ncRNAs regulate gene expression via a diverse range of regulatory mechanisms, contributing to diverse cancer phenotypes and tumor angiogenesis.
Recently, certain reviews have focused on the general role of EVs or miRNAs in angiogenesis [13,14,15]; however, the functions and mechanisms of EV-derived ncRNAs have not been comprehensively explored in tumor angiogenesis. In this review, we summarized the functions of EV-derived ncRNAs in tumor angiogenesis and highlighted their potential clinical applications in cancer diagnosis and therapy.

2. Biology of EVs, Angiogenesis, and ncRNAs

2.1. Characteristics of EVs and ncRNAs

EVs are extracellular structures enclosed in a lipid bilayer that can be secreted by almost all known cell types [16]. Several studies have indicated that EVs play an essential role in intercellular communication between tumor cells and the tumor microenvironment (TME). EVs are highly heterogeneous; therefore, their characterization and classification are crucial for further research to avoid generating inconclusive results. Based on their size, biogenesis, and release pathways, EVs can be broadly divided into three main subtypes: exosomes, microvesicles (MVs), and apoptotic bodies. Exosomes, also called small EVs, possess a diameter ranging from 30 to 150 nm and are derived from multivesicular endosomal pathways, which are formed by inward budding of the endosomal membrane in a process that sequesters particular proteins and lipids [17]. On the contrary, MVs are generated by regulated outward budding of the plasma membrane [18]. The mechanisms of exosomal biogenesis involve multiple factors, and the most well-known regulator is endosomal sorting complex required for transport (ESCRT) [18]. Exosomal biogenesis involves inward budding of the plasma membrane to form endosomes, leading to production of multivesicular bodies (MVBs), fusion of MVBs with the plasma membrane, and release of exosomes into the extracellular space. A core component of this mechanism is the ESCRT machinery, which consists of four protein complexes and auxiliary proteins that bind to future exosome cargoes and form intraluminal vesicles that incorporate those cargoes [19,20]. Several studies have found that exosomes can be formed despite the depletion of the ESCRT complex, which reveals an ESCRT-independent approach [21]. ESCRT-independent exosomal biogenesis is regulated by sphingolipid ceramide, which is produced from the hydrolysis of sphingomyelin by neutral sphingomyelinase 2 [17]. The contents of EVs include various nucleic acids, lipids, and proteins [22]. ncRNAs carried by EVs can regulate various physiological and pathological processes through multiple mechanisms (Figure 1).
In the past few decades, the development of high-throughput sequencing technology has indicated that the transcription rate of the human genome generally exceeds 70%. However, <2% of the transcript is translated into proteins; most human transcriptomes are ncRNAs. Emerging studies have shown that despite being ‘transcriptional junk’, ncRNAs, such as miRNAs, piRNAs, circRNAs, snoRNAs, and the attractive lncRNAs, play a versatile role in manipulating gene expression [23]. miRNAs, a type of endogenous small RNA with a length of 20–24 nucleotides (nt), have many essential adjustable functions in cells. By complementarily binding to the 3’-untranslated region (UTR) of targeted mRNAs, miRNAs act as regulators of gene expression, thereby inhibiting post-transcriptional gene expression [24,25,26]. Recent studies have reported that miRNAs are selectively sorted into EVs and participate in intercellular communication in the TME [27]. In addition, EV-derived miRNAs in biofluids can be used as ideal biomarkers for various types of tumors due to their easy accessibility, high abundance, and good stability [28]. piRNAs, a type of small RNA with a length of 21–35 nt, specifically interact with PIWI protein to perform multifaceted functions in germline development and somatic tissues [29,30,31].
In addition to small RNAs, large ncRNAs also participate in gene regulation in various biological processes. lncRNAs, collectively referred to as transcripts with more than 200 nt, have limited potential to encode proteins [32]. They perform their functions through multiple molecular and cellular mechanisms, such as interacting with epigenetic factors or TFs to modulate gene transcription, sequestering miRNAs, interacting with proteins, and encoding functional small peptides [33,34,35,36]. In addition, lncRNAs can also be selectively sorted into EVs and participate in cell-to-cell communication in the TME [37]. circRNAs, generated by a particular form of alternative splicing called back-splicing, regulate gene transcription and translation by interacting with DNAs, RNAs, and proteins [38]. Emerging studies have indicated that circRNAs participate in multiple physiological and pathological processes, including tumor angiogenesis [39,40,41].

2.2. EV-Derived ncRNAs: New Players in Tumor Angiogenesis

Angiogenesis is a multi-step process and has two types: sprouting and intussusceptive angiogenesis [42]. Various types of cells, including endothelial cells (ECs), tumor cells, stromal cells, and immune cells, regulate angiogenesis in the blood vessels. In addition, some regulatory and signalling molecules govern angiogenesis, including growth factors (e.g., VEGF, PDGF, FGF, and EGF) and transcription factors, such as HIFs [43,44,45]. Because angiogenesis is crucial for tumor growth and metastasis, targeting tumor-associated angiogenesis is a promising strategy for cancer treatment [46,47,48]. Currently, anti-angiogenic therapies targeting VEGF and VEGFR are used for the treatment of various tumors [49].
Several studies have indicated that EVs can be used as ncRNA carriers to play diverse roles in regulating tumor hallmarks, including angiogenesis. For example, in non-small cell lung cancer (NSCLC), RCAN1.4 has been identified as a target of miR-619-5p, and its suppression promotes angiogenesis [50]. The exosomal lncRNA FAM225A upregulates NETO2 and FOXP1 expression by sponging miR-206 to accelerate oesophageal squamous cell carcinoma (ESCC) progression and angiogenesis [51]. In addition, circSHKBP1 sponges miR-582-3p to enhance HUR expression and VEGF mRNA stability, which promotes angiogenesis and gastric tumor progression [52]. Moreover, emerging studies have indicated that EV-derived ncRNAs can regulate tumor angiogenesis by influencing a wide variety of tumor-associated molecules. The functions and mechanisms of EV-derived ncRNAs in tumor angiogenesis are summarized in Table 1. These studies suggest that EV-derived ncRNAs play an essential role in tumor angiogenesis. However, new technologies and animal models are required to further investigate the precise mechanisms of EV-derived ncRNAs in the regulation of tumor angiogenesis.

3. Emerging Functions of EV-Derived ncRNAs in Tumor Angiogenesis

The TME is composed of the extracellular matrix, blood vessels, immune cells, stromal cells, and bioactive substances, all of which enrich the local environment of tumor cells. It plays an integral role in tumor migration, tube formation, cell adhesion, invasion, proliferation, angiogenesis, and treatment resistance [104]. EVs contain various signalling molecules that are important for the reprogramming function of intercellular communication in the TME [19,105]. Tumor-derived EV-derived ncRNAs can regulate the behaviour of surrounding immune and stromal cells, ultimately creating a suitable microenvironment for tumor growth [106]. In addition, immune and stromal cells secrete EV-derived ncRNAs in the TME, which, in turn, affects tumor progression [107]. In the subsequent sections, we have summarized the emerging roles of EV-derived ncRNAs in tumor angiogenesis and highlighted the crosstalk between tumor cells and the surrounding cells within the TME (Figure 2).

3.1. Tumor-Derived EVs Affect Tumor Angiogenesis by Transferring ncRNAs

The TME is a ‘complex society’ of many cell types and their extracellular matrices. Different immune molecules exist in the TME, which include macrophages, neutrophils, mast cells, dendritic cells (DCs), natural killer (NK) cells, and T and B lymphocytes [108]. In addition, the TME also consists of tumor stromal cells, including mesenchymal stromal cells (MSCs), fibroblasts, ECs, and pericytes [109,110]. Primary tumors recruit immune and stromal cells to their microenvironments to induce angiogenesis mediated by EV-derived ncRNAs, leading to increased tumor proliferation and metastatic potential (Figure 2).
EV-derived ncRNAs derived from tumor cells promote the M2 polarization of macrophages in the TME, thereby affecting angiogenesis. A study found that EVs derived from hypoxic lung cancer (LC) cells increased the polarization of M2 macrophages through the miR-103a/PTEN axis. The M2-type cytokines (IL-10) and cancer-promoting cytokines (VEGF-A) produced by M2-polarized macrophages further promoted cancer progression and angiogenesis [65] (Figure 2A). Similarly, the EMT transcription factor Snail directly stimulates the production of miR-21-rich tumor-derived exosomes (TEX). Furthermore, CD14+ human monocytes phagocytize exosomes containing miR-21, thereby inhibiting the expression of M1 markers, increasing the expression of M2 markers, and eventually promoting angiogenesis [111].
Cancer-associated fibroblasts (CAFs) differentiated from fibroblasts and MSCs were shown to promote tumor growth [112]. Exosome-mediated transfer of ncRNAs can also enhance the activation of CAF-promoting angiogenic transformation. The transmission of EV-derived ncRNAs from hepatocellular carcinoma (HCC) melanoma cells induced the transformation of fibroblasts to CAFs, leading to cancer-supporting angiogenesis. Zhou et al. demonstrated that cancer-derived exosomal miR-21 could convert hepatic stellate cells (HSCs) to CAFs by downregulating PTEN, thereby activating CAFs and promoting cancer angiogenesis by upregulating the expression of VEGF-α, MMP2, MMP9, bFGF, and transforming growth factor-β (TGF-β). [80]. In addition, exosomal miR-155 secreted by melanoma cells could activate the JAK2/STAT3 signalling pathway by reducing SOCS1 expression, thereby triggering the reprogramming of normal fibroblasts into pro-angiogenic CAFs. CAFs further increase the expression and secretion of VEGF-α, FGF2, and MMP9, thus promoting the proliferation, migration, and tube formation of ECs and angiogenesis in melanoma [113] (Figure 2A). Therefore, current evidence indicates that EV-derived ncRNAs influence tumor angiogenesis through diverse cell types, including macrophages and CAFs.
ECs receive various information from their environment, eventually leading to their rapid growth to form new blood vessels [114]. Accumulating evidence indicates that tumor-cell-derived ncRNAs may be packaged into EVs and transferred to recipient cells, resulting in the regulation of angiogenesis by influencing the proliferation and migration of ECs. In gastric cancer (GC), exosomes can function as vehicles to deliver different miRNAs (e.g., miR-155, miR-130a, and miR-135b), eventually enhancing the growth of blood vessels and promoting the metastasis of GC [53,54,55,56]. In colorectal cancer (CRC), exosomal miR-25-3p targets KLF2 and KLF4, thereby modulating the expression of VEGFR2, ZO-1 and Claudin-5 in ECs and promoting vascular permeability and angiogenesis [61]. In ESCC, miR-181b-5p is transferred from ESCC cells to vascular ECs through exosomes, as evidenced by angiogenesis induced by targeting PTEN and PHLPP2. Moreover, higher expression of miR-181b-5p in patients with ESCC predicts poorer overall survival [81]. In leukemia, exosomal miR-210 downregulates EFNA3, thereby enhancing tube formation in human umbilical vein endothelial cells (HUVECs) [72]. In multiple myeloma, the number of exosomes carrying miR-135b is significantly increased, and these exosomes directly inhibit factor-inhibiting hypoxia-inducible factor 1 (FIH-1) in ECs, resulting in the formation of endothelial tubes through the HIF–FIH signalling pathway [57]. In glioma, exosomes carrying miR-9 absorbed by vascular ECs can increase angiogenesis by directly targeting COL18A1, THBS2, PTCH1, and PHD3. This process can be activated by MYC and OCT4 [70]. In addition, another type of small ncRNA, piRNA, was also reported to regulate angiogenesis. piRNA-823 delivered by MM-derived EVs promoted angiogenesis by increasing the secretion of VEGF and IL-6 from ECs [103].
In addition to the small ncRNAs mentioned above, EV-lncRNAs and circRNAs derived from tumor cells also mediate tumor angiogenesis. In NSCLC, exosomal lncRNA-p21 modulates miRNA-containing exosomal cargoes and influences EC behaviour, thereby regulating angiogenesis and EC permeability [115] (Figure 2A). Exosomal lncRNAs (such as MALAT1 and RAMP2-AS1) are transferred to recipient HUVECs and eventually promote angiogenesis by increasing VEGF and VEGFR2 expression in epithelial ovarian cancer (OC) and chondrosarcoma, respectively [88,94]. In breast cancer (BC), exosomal SNHG1 promotes the proliferation, migration, and angiogenesis of HUVECs via the miR-216b-5p/JAK2 axis [92]. In pancreatic cancer (PC), exosomal UCA1 is delivered to HUVECs and promotes angiogenesis via the miR-96-5p/AMOTL2 axis [90]. In GC, exosomal X26nt decreases vascular endothelial cadherin (VE-cadherin) expression by directly binding to the 3’UTR of VE-cadherin mRNA, thereby increasing vascular permeability and angiogenesis in HUVECs [116]. Exosomal AC073352.1 secreted by BC cells is internalised by HUVECs and promotes angiogenesis by interacting with YBX1 and stabilizing its protein expression [93]. Exosomal H19 released by cancer stem cells promotes endothelial tube production by upregulating VEGF [97]. In addition, circRNA-100338 expression is high in both highly metastatic HCC cells and HCC cell-secreted exosomes. Exosomal circRNA-100338 enhances the pro-angiogenic ability and permeability of HCC cells by interacting with NOVA2, thereby affecting cancer proliferation (Figure 2A) [98].
Tumor-derived EV-derived ncRNAs can be useful indicators of tumor burden, prognosis, and treatment because they regulate many aspects of the angiogenic microenvironment. Therefore, specific EVs and their genetic cargoes in the TME may be used as biomarkers for the prevention and diagnosis of tumors or as novel targets for cancer treatment.

3.2. Immune and Stromal Cell-Derived EVs Affect Tumor Angiogenesis by Transferring ncRNAs

Tumors can release extracellular signals to create an angiogenic TME suitable for their growth and development. In turn, the diverse immune and stromal cells in the microenvironment can also manipulate tumor development and progression. EV-derived ncRNAs secreted by immune and stromal cells in the TME regulate tumor angiogenesis. For example, Zhang et al. found that miR-130b-3p enclosed in M2 macrophage-derived EVs was delivered to GC cells, thereby promoting angiogenesis via both the downregulation of MLL3 and upregulation of GRHL2 (Figure 2B) [84]. In another study, M2 macrophage-derived exosomes increased the blood vessel density of mouse pancreatic ducts in vitro and promoted tumor growth by targeting E2F2 [76]. In addition, miR-10a-5p derived from CAFs promotes angiogenesis and tumorigenesis in cervical squamous cell carcinoma by activating Hedgehog signalling by inhibiting TBX5 (Figure 2B) [83]. Pakravan et al. showed that the transfer of exosomal miR-100 from human bone marrow MSCs to BC cells downregulated the expression of VEGF in BC-derived cells by regulating the mTOR/HIF-1α signalling axis, thereby inhibiting angiogenesis (Figure 2B) [78]. miR-126 in MSC-derived EVs modulates the pro-angiogenic capacity of ECs by upregulating VEGF [66]. Therefore, fibroblasts, macrophages, and MSCs have been shown to exert synergistic effects to promote tumor angiogenesis in the TME.
EV-derived ncRNAs, as signalling molecules for communication between tumor cells and other components of the TME, play an important role in remodelling the TME to regulate tumor angiogenesis, thereby leading to tumor progression. However, the current research progress on EVs secreted by immune and stromal cells is far behind that of EVs secreted by tumor cells. Therefore, more studies are warranted to further understand the potential impact of EV-derived ncRNAs on the TME, particularly on tumor angiogenesis. To date, a majority of biological studies on EV-derived ncRNAs have been conducted using cell culture systems in vitro. However, it remains unclear whether EV-derived ncRNAs derived from artificial 2-dimensional cell culture systems can accurately reflect the biological conditions of the body. To address this concern, tumor organoids should be considered suitable models to investigate the potential impacts of EV-derived ncRNAs on the TME.

4. Signalling Pathways Regulated by EV-Derived ncRNAs in Tumor Angiogenesis

Angiogenesis requires the participation of various molecules and can be adjusted by multiple factors. Studies have revealed that a series of signalling molecules and pathways contribute to the angiogenic response [47]. To achieve a better understanding of regulatory pathways involved in angiogenesis, in this section, we have discussed the features and functional roles of EV-derived ncRNAs in angiogenesis-associated pathways, including the JAK/STAT, TGF-β, PI3K/AKT, and nuclear factor-κB (NF-κB) pathways.
The JAK/STAT and TGF-β pathways have been implicated in distinct essential cellular functions related to proliferation, survival, differentiation, and angiogenesis. STAT and SMAD are excessively activated in different tumor types and act as crucial signalling nodes in the TME [117,118]. A study revealed that the transfer of exosomal miR-210 derived from HCCs to ECs promoted tumor angiogenesis by targeting STAT6 and SMAD4 [73]. Abnormal TGF-β signal transduction may lead to various diseases, such as abnormal embryonic development, immune diseases, tissue fibrosis, and cancerous diseases. TGF-β regulates not only physiological angiogenesis but also angiogenesis in the later stages of tumor development [119]. A similar phenomenon also exists in hypoxic papillary carcinoma: hypoxic papillary thyroid carcinoma cells secrete exosomes rich in miR-21-5p, thereby promoting endothelial tube formation by inhibiting the expression of TGF-βI and COL4A1 [77]. In addition, Lawson J et al. found that miR-142-3p, a known cancer inhibitor in lung adenocarcinoma, was enriched in lung adenocarcinoma cell (LAC)-derived exosomes. The transfer of miR-142-3p from LACs to ECs accelerated angiogenesis by repressing TGFβR1 expression [64]. Another study revealed that exosomes promoted angiogenesis by transferring lnc-CCAT2 and lnc-POU3F3 to ECs, leading to the activation of VEGFA, TGF-β, bFGF, and bFGFR in glioma [87,120]. PTEN, a cancer suppressor gene, is a key mediator of the PI3K/AKT pathway and plays a vital role in the formation of normal blood vessels during tumor progression [121]. A recent study found that exosomal miR-205 secreted by OCs induced angiogenesis by silencing PTEN and subsequently activating its downstream AKT pathway. Moreover, PI3K or AKT inhibitors significantly delay cell migration and tube formation induced by exosomal miR-205 [68]. In glioma stem cells (GSCs), the expression of miR-26a increases, whereas that of PTEN decreases. Exosomal miR-26a facilitates the angiogenesis of human brain microvascular endothelial cells (HBMECs) by targeting PTEN to activate the PI3K/AKT signalling pathway [74]. Another study showed that nasopharyngeal carcinoma (NPC) cells secreted exosomes that enclose miR-9, which suppressed endothelial tube formation and migration by targeting MDK and modulating the PDK/AKT signalling pathway [69]. Emerging studies have indicated that EV-derived ncRNAs also orchestrate the crossroads of angiogenic signalling pathways. A recent study showed that exosomal miR-141-3p secreted by OCs promoted angiogenesis by upregulating the expression of VEGFR-2 in ECs, thereby activating the JAK/STAT3 and NF-κB signalling pathways [67].
To date, several studies have reported the underlying mechanisms of EV-derived ncRNAs in various signalling pathways and their regulatory functions in tumor angiogenesis to a certain extent. However, more studies are required to elucidate the precise molecular mechanisms of ncRNAs in regulating signalling pathways relevant to tumor angiogenesis. Furthermore, intervention in these signalling pathways may offer promising strategies for cancer treatment. More importantly, it is likely that other signalling pathways that induce tumor angiogenesis will be identified in the future. Understanding the mechanisms that mediate tumor angiogenesis will help scholars to further develop novel strategies and therapeutic approaches for inhibiting angiogenesis [122]. However, the crosstalk between angiogenesis-related signalling pathways remains unclear and warrants further investigation. Understanding these pathways may have significant implications in the development of novel anti-angiogenic cancer therapy.

5. Potential Clinical Applications of EV-Derived ncRNAs in Cancers

5.1. EV-Derived ncRNAs as Promising Tumor Biomarkers

EVs that possess great potential as disease biomarkers and therapeutic carriers have attracted increasing attention [123,124]. Biomarkers are molecules that can be used for diagnosis or prognosis. An ideal biomarker should have the following four most important characteristics: specificity, sensitivity, stability, and easy accessibility in a relatively non-invasive way. Most studies have focused on extracellular ncRNAs as potential biomarkers because they are stable and can be easily extracted from liquid biopsy, such as blood, urine, or other body fluids, using simple, sensitive, and relatively inexpensive assays. It has been reported that the quality of EV-derived ncRNAs is almost unaffected even after the samples are stored for many years because of the uniqueness of the source cell components and protection via encapsulation in the membrane [125,126]. Therefore, EVs are stable in circulation and under various storage conditions. We conducted a comprehensive literature search of EV-derived ncRNAs from different sources and found valuable biomarkers for the diagnosis and prognosis of multiple cancers (Figure 3A).
Researchers are interested in investigating candidate miRNAs, lncRNAs, and circRNAs carried by EVs that may serve as biomarkers for the diagnosis, prognosis, and treatment of tumors [7]. For example, in a study, ROC curve analysis demonstrated that plasma miR-601 and miR-760 could both be used as promising diagnostic biomarkers for advanced CRC [127]. In a study involving patients with CRC, Ogata et al. reported that the area under the curve (AUC) was 0.953, 0.948, and 0.798. In another study, exosomal miR-23a, miR-1246, and miR-21 could distinguish CRC (all stages) from the control area [128]. Furthermore, higher levels of plasma miR-320-EV and miR-126-EV in patients with high-risk LC can promote angiogenesis and can be significantly associated with poor overall survival [66]. A similar study showed that serum exosomes of patients with GC were rich in lncRNA ZFAS1. In addition, the upregulation of ZFAS1 was significantly associated with tumor lymphatic metastasis and TNM staging. These studies indicate that exosomal ZFAS1 may serve as a potential prognostic biomarker for GC [129]. Another study reported that exosomal ENSG00000258332.1 and LINC00635 could be used to differentiate patients with HCC from those with chronic hepatitis B with high specificity. Therefore, serum exosomal ENSG00000258332.1 and LINC00635, which are highly sensitive and can be obtained non-invasively, may be used as biomarkers for HCC [130]. Similarly, recent studies have reported serum exosomal circRNAs as novel and useful tools for the non-invasive diagnosis of cancer [131]. Exosomal circPRMT5 is highly expressed in the serum and urine of patients with bladder cancer and is closely related to tumor metastasis [132]. In addition, certain diagnostic clinical trials are currently underway. In one such trial, exosomal lncRNAs are isolated from serum samples for the diagnosis of lung cancer (NCT03830619).
A large number of studies have focused on the diagnostic, prognostic, and therapeutic significance of EV-derived ncRNAs in different tumor types. However, the specific role of EV-derived ncRNAs in angiogenesis-related diseases remains unclear. Furthermore, almost all studies have focused on cellular experiments and EV-ncRNA-associated applications in vitro; therefore, further studies are required to validate the findings for in vivo models. In addition, the potential of EV-derived ncRNAs as biomarkers remains to be further verified in multi-center, large-scale clinical trials.

5.2. EV-Derived ncRNAs as Potential Anti-Angiogenic Therapeutic Targets

Because of their negligible antigenicity, minimal cytotoxicity, and ability to bypass endocytic pathways and phagocytosis, EVs are considered ideal natural carriers for the delivery of ncRNAs [133]. In a study, engineered exosomes modified with DSPE–PEG2K–RGD loaded with miR-92b-3p produced synergistic anti-tumor and anti-angiogenesis effects with apatinib in nude mice models of abdominal tumors [3]. Another study showed that the delivery of miR-29a/c using cell-derived MVs inhibited angiogenesis in GC [134]. Furthermore, EV-derived ncRNAs have been demonstrated to be functional towards tumor hallmarks in different cell lines. Huang et al. showed that exosomes derived from HCC cells silenced with circRNA-100338 could significantly decrease the invasive ability of HCC cells. In addition, these exosomes could reduce cell proliferation, angiogenesis, permeability, vasculogenic mimicry (VM) formation ability of HUVECs, and tumor metastasis [98]. Bai et al. demonstrated that exosomal miR-135b secreted by GC cells inhibited the expression of FOXO1 protein and enhanced the growth of blood vessels in mouse models of tumor transplantation [56]. Using an NPC model, Wang et al. found that overexpressed EBV-miR-BART10-5p and hsa-miR-18a upregulated VEGF and HIF-1α in a Spry3-dependent manner and strongly promoted angiogenesis. Moreover, in both in vitro and in vivo NPC models, treatment with iRGD-tagged exosomes enclosing antagomiR-BART10-5p and antagomiR-18a inhibited angiogenesis [135]. Therefore, exosome engineering is a promising tool in RNA-based therapeutics for cancer treatment (Figure 3B). Recently, RNA interference (RNAi)-based strategies, CRISPR/Cas9-mediated circRNA knockout, CRISPR/Cas13-mediated circRNA knockdown and circRNA-induced overexpressed plasmids were developed to target ncRNAs for therapeutic purposes both in vitro and in vivo [136]. In a study, the expression of pro-angiogenic factors in HUVECs was significantly reduced after miR-92a-3p was knocked down in exosomes using an miR-92a-3p inhibitor (miR-92a-3p-i) [137]. Furthermore, because EV-derived ncRNAs perform significant biological functions, specifically targeting EV-derived ncRNAs may be a promising strategy for treating many types of tumors. Currently, many studies aimed at regulating the production of EVs or blocking the uptake of EVs to achieve the goal of treating patients with cancer are underway. Using EVs as a delivery platform is a promising strategy; however, due to high costs and strict ethical regulations, the mass production of EVs is not easy to achieve to develop commercial viability.
Acquired resistance to chemotherapy remains a major obstacle in treating tumors. Previous studies have reported that EV-derived ncRNAs not only impact chemosensitivity but also regulate mechanisms underlying the development and progression of anticancer drug resistance [138,139]. Recent studies have indicated that EV-derived ncRNAs play a pivotal role in anticancer drug resistance. Deng et al. showed that doxorubicin treatment increased MDSC-produced exosomal miR-126a, which rescued doxorubicin-induced MDSC death and promoted tumor angiogenesis in an S100A8/A9-dependent manner [140]. In addition, Fu et al. found that multidrug-resistant HCC Bel/5-FU cells transported exosomal miR-32-5p to Bel7402-sensitive cells and activated the PI3K/AKT pathway, thereby regulating angiogenesis and EMT to further induce multidrug resistance [141]. Additionally, exosomal miR-549a promotes angiogenesis and enhances vascular permeability by regulating the expression of HIF-1α in vascular ECs, thereby promoting tumorigenesis and metastasis in TKI-resistant clear cell renal cell carcinoma [142]. Therefore, overcoming drug resistance during cancer treatment is key to achieving good results. Because some EV-derived ncRNAs can induce drug resistance, EVs may be engineered to be used as ncRNA carriers to overcome chemotherapeutic drug resistance [143]. Studies have shown that EVs can deliver anti-angiogenic ncRNAs, thus providing favorable evidence for their potential use in therapeutic applications. Various ncRNAs, other types of exogenous therapeutic drugs (such as anti-angiogenic drugs), and their combination can be loaded into EVs for cancer treatment. Moreover, EV-derived ncRNAs are considered promising tools for the diagnosis, prognosis, and treatment of various tumors. However, further investigation is required before these therapeutic approaches can be used in clinical settings for cancer treatment.

6. Concluding Remarks and Future Perspectives

This review highlights the mechanisms of EV-derived ncRNAs in tumor angiogenesis and anti-angiogenic resistance, their potential as diagnostic biomarkers, and their therapeutic applications. To date, studies have indicated that EV-derived ncRNAs play a versatile role in tumor angiogenesis by remodelling the TME involved in the crosstalk of tumor cells with various immune and stromal cells. Furthermore, these EV-derived ncRNAs can activate several classical signalling pathways by regulating various angiogenesis-related molecules. However, the following questions remain to be addressed to accelerate translational research on EV-derived ncRNAs in tumor diagnosis and treatment: How do EV-derived ncRNAs regulate angiogenesis at the transcriptional and post-transcriptional levels? How do immune cells in the TME, such as NK and B cells, release EVs containing ncRNAs to affect angiogenesis? How can new technologies be used to develop novel therapeutic agents for cancer therapy? More studies should be conducted to investigate EVs derived from different cancer types and to examine their respective roles in tumor angiogenesis. A better understanding of the interactions between tumor and other cells in the TME, such as immune and stromal cells, may provide new insights into EV biology and EV-based therapeutics to improve cancer patient outcomes.
Studies have indicated that EV-derived ncRNAs act as a promising source for diagnostic biomarkers because they are more tissue-specific, and they have more stable characteristics than traditional protein biomarkers. Although significant progress has been made in identifying candidate biomarkers, more large-scale, multi-center clinical trials should be well-designed to verify their utility.
Owing to the unique biological characteristics of EVs, the use of EVs to deliver therapeutic agents is recognized as a new approach with great potential in cancer therapy. Although EV engineering is a prime focus area for research, the findings of most studies are basic. For instance, current study findings on EV engineering are mainly derived from traditional 2-dimensional cell culture systems, which cannot be used to stimulate the TME. Therefore, more reliable models should be developed and validated for investigating anti-angiogenesis. Fortunately, scholars are now creating 3-dimensional organoids made from co-cultures of cancer cells with immune or stromal cells to investigate tumor angiogenesis, which will accelerate the development of new therapeutic approaches for cancer treatment using EVs as natural carriers of drugs and ncRNAs. Therefore, we can expect that more novel RNA-based therapeutic agents will be translated from the bench to the bedside in the future.

Author Contributions

Z.M. conceived the review article. Z.M. and L.W. designed the review and provided supervision. M.H. drafted the manuscript and prepared the figures. J.L., C.-G.L., R.M.W.J.G. and F.Y. helped to modify the figures and revise the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Natural Science Foundation of Hubei Province (2019CFB591 to Z.M.), the Joint Development Fund for Medical Research of Yangtze University (WJ2019-13 to Z.M.), and the Bureau of Science and Technology of Jingzhou Municipality (2020CB21-35 to Z.M.) This research was also supported by the National Research Foundation Singapore and the Singapore Ministry of Education under its Research Centers of Excellence initiative. Joint NCIS-N2CR Seed Grant (R-713-002-272-733, PI: LW).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef] [PubMed]
  2. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Teo, A.; Xiang, X.; Le, M.; Wong, A.; Zeng, Q.; Wang, L.; Goh, B.-C. Tiny miRNAs Play a Big Role in the Treatment of Breast Cancer Metastasis. Cancers 2021, 13, 337. [Google Scholar] [CrossRef] [PubMed]
  4. Cheng, J.-T.; Wang, L.; Wang, H.; Tang, F.-R.; Cai, W.-Q.; Sethi, G.; Xin, H.-W.; Ma, Z. Insights into Biological Role of LncRNAs in Epithelial-Mesenchymal Transition. Cells 2019, 8, 1178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Xiao, W.; Li, J.; Hu, J.; Wang, L.; Huang, J.; Sethi, G.; Ma, Z. Circular RNAs in cell cycle regulation: Mechanisms to clinical significance. Cell Prolif. 2021, 54, e13143. [Google Scholar] [CrossRef]
  6. Slack, F.J.; Chinnaiyan, A.M. The Role of Non-coding RNAs in Oncology. Cell 2019, 179, 1033–1055. [Google Scholar] [CrossRef]
  7. Li, J.; Zhang, G.; Liu, C.-G.; Xiang, X.; Le, M.T.; Sethi, G.; Wang, L.; Goh, B.-C.; Ma, Z. The potential role of exosomal circRNAs in the tumor microenvironment: Insights into cancer diagnosis and therapy. Theranostics 2022, 12, 87–104. [Google Scholar] [CrossRef]
  8. Xie, Y.; Dang, W.; Zhang, S.; Yue, W.; Yang, L.; Zhai, X.; Yan, Q.; Lu, J. The role of exosomal noncoding RNAs in cancer. Mol. Cancer 2019, 18, 37. [Google Scholar] [CrossRef] [Green Version]
  9. Herbert, S.P.; Stainier, D.Y.R. Molecular control of endothelial cell behaviour during blood vessel morphogenesis. Nat. Rev. Mol. Cell Biol. 2011, 12, 551–564. [Google Scholar] [CrossRef] [Green Version]
  10. Aguilar-Cazares, D.; Chavez-Dominguez, R.; Carlos-Reyes, A.; Lopez-Camarillo, C.; de la Cruz, O.N.H.; Lopez-Gonzalez, J.S. Contribution of Angiogenesis to Inflammation and Cancer. Front. Oncol. 2019, 9, 1399. [Google Scholar] [CrossRef] [Green Version]
  11. Nishida, N.; Yano, H.; Nishida, T.; Kamura, T.; Kojiro, M. Angiogenesis in cancer. Vasc. Health Risk Manag. 2006, 2, 213–219. [Google Scholar] [CrossRef] [PubMed]
  12. Jászai, J.; Schmidt, M.H. Trends and Challenges in Tumor Anti-Angiogenic Therapies. Cells 2019, 8, 1102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Song, W.; Yan, D.; Wei, T.; Liu, Q.; Zhou, X.; Liu, J. Tumor-derived extracellular vesicles in angiogenesis. Biomed. Pharmacother. 2018, 102, 1203–1208. [Google Scholar] [CrossRef] [PubMed]
  14. Wang, Y.; Wang, L.; Chen, C.; Chu, X. New insights into the regulatory role of microRNA in tumor angiogenesis and clinical implications. Mol. Cancer 2018, 17, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Olejarz, W.; Kubiak-Tomaszewska, G.; Chrzanowska, A.; Lorenc, T. Exosomes in Angiogenesis and Anti-angiogenic Therapy in Cancers. Int. J. Mol. Sci. 2020, 21, 5840. [Google Scholar] [CrossRef] [PubMed]
  16. György, B.; Szabó, T.G.; Pásztói, M.; Pál, Z.; Misják, P.; Aradi, B.; László, V.; Pállinger, É.; Pap, E.; Kittel, Á.; et al. Membrane vesicles, current state-of-the-art: Emerging role of extracellular vesicles. Cell. Mol. Life Sci. 2011, 68, 2667–2688. [Google Scholar] [CrossRef] [Green Version]
  17. Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brügger, B.; Simons, M. Ceramide Triggers Budding of Exosome Vesicles into Multivesicular Endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef]
  18. Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  19. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  20. Christ, L.; Raiborg, C.; Wenzel, E.M.; Campsteijn, C.; Stenmark, H. Cellular Functions and Molecular Mechanisms of the ESCRT Membrane-Scission Machinery. Trends Biochem. Sci. 2016, 42, 42–56. [Google Scholar] [CrossRef]
  21. Stuffers, S.; Wegner, C.S.; Stenmark, H.; Brech, A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic 2009, 10, 925–937. [Google Scholar] [CrossRef] [PubMed]
  22. Kikuchi, S.; Yoshioka, Y.; Prieto-Vila, M.; Ochiya, T. Involvement of Extracellular Vesicles in Vascular-Related Functions in Cancer Progression and Metastasis. Int. J. Mol. Sci. 2019, 20, 2584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Nagano, T.; Fraser, P. No-Nonsense Functions for Long Noncoding RNAs. Cell 2011, 145, 178–181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ingenito, F.; Roscigno, G.; Affinito, A.; Nuzzo, S.; Scognamiglio, I.; Quintavalle, C.; Condorelli, G. The Role of Exo-miRNAs in Cancer: A Focus on Therapeutic and Diagnostic Applications. Int. J. Mol. Sci. 2019, 20, 4687. [Google Scholar] [CrossRef] [Green Version]
  25. Qin, Q.; Wei, F.; Zhang, J.; Li, B. miR-134 suppresses the migration and invasion of non-small cell lung cancer by targeting ITGB1. Oncol. Rep. 2017, 37, 823–830. [Google Scholar] [CrossRef] [Green Version]
  26. Chen, L.; Qian, H.; Xue, J.; Zhang, J.; Chen, H. MicroRNA-152 regulates insulin secretion and pancreatic β cell proliferation by targeting PI3Kα. Mol. Med. Rep. 2018, 18, 4113–4121. [Google Scholar] [CrossRef] [Green Version]
  27. Godlewski, J.; Krichevsky, A.M.; Johnson, M.D.; Chiocca, E.A.; Bronisz, A. Belonging to a network—microRNAs, extracellular vesicles, and the glioblastoma microenvironment. Neuro Oncol. 2014, 17, 652–662. [Google Scholar] [CrossRef] [Green Version]
  28. Saadatpour, L.; Fadaee, E.; Fadaei, S.; Mansour, R.N.; Mohammadi, M.; Mousavi, S.M.; Goodarzi, M.; Verdi, J.; Mirzaei, H. Glioblastoma: Exosome and microRNA as novel diagnosis biomarkers. Cancer Gene Ther. 2016, 23, 415–418. [Google Scholar] [CrossRef]
  29. Grimson, A.; Srivastava, M.; Fahey, B.; Woodcroft, B.; Chiang, H.R.; King, N.; Degnan, B.; Rokhsar, D.; Bartel, D.P. Early origins and evolution of microRNAs and Piwi-interacting RNAs in animals. Nature 2008, 455, 1193–1197. [Google Scholar] [CrossRef]
  30. Ozata, D.M.; Gainetdinov, I.; Zoch, A.; O'Carroll, D.; Zamore, P.D. PIWI-interacting RNAs: Small RNAs with big functions. Nat. Rev. Genet. 2018, 20, 89–108. [Google Scholar] [CrossRef] [Green Version]
  31. Peng, Q.; Chiu, P.K.-F.; Wong, C.Y.-P.; Cheng, C.K.-L.; Teoh, J.Y.-C.; Ng, C.-F. Identification of piRNA Targets in Urinary Extracellular Vesicles for the Diagnosis of Prostate Cancer. Diagnostics 2021, 11, 1828. [Google Scholar] [CrossRef] [PubMed]
  32. Gutschner, T.; Diederichs, S. The hallmarks of cancer. RNA Biol. 2012, 9, 703–719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kopp, F.; Mendell, J.T. Functional Classification and Experimental Dissection of Long Noncoding RNAs. Cell 2018, 172, 393–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Li, J.; Long, J.; Zhang, Q.; Shen, H.; Guo, A.-Y.; Ma, Z.; Zhang, G. Hypothalamic long noncoding RNA AK044061 is involved in the development of dietary obesity in mice. Int. J. Obes. 2021, 45, 2638–2647. [Google Scholar] [CrossRef] [PubMed]
  35. Qian, H.; Chen, L.; Huang, J.; Wang, X.; Ma, S.; Cui, F.; Luo, L.; Ling, L.; Luo, K.; Zheng, G. The lncRNA MIR4435-2HG promotes lung cancer progression by activating β-catenin signalling. Klin. Wochenschr. 2018, 96, 753–764. [Google Scholar] [CrossRef] [PubMed]
  36. Gu, C.; Zou, S.; He, C.; Zhou, J.; Qu, R.; Wang, Q.; Qi, J.; Zhou, M.; Yan, S.; Ye, Z. Long non-coding RNA CCAT1 promotes colorectal cancer cell migration, invasiveness and viability by upregulating VEGF via negative modulation of microRNA-218. Exp. Ther. Med. 2020, 19, 2543–2550. [Google Scholar] [CrossRef]
  37. Wang, M.; Zhou, L.; Yu, F.; Zhang, Y.; Li, P.; Wang, K. The functional roles of exosomal long non-coding RNAs in cancer. Exp. 2019, 76, 2059–2076. [Google Scholar] [CrossRef]
  38. Li, X.; Yang, L.; Chen, L.-L. The Biogenesis, Functions, and Challenges of Circular RNAs. Mol. Cell 2018, 71, 428–442. [Google Scholar] [CrossRef] [Green Version]
  39. Yang, X.; Li, S.; Wu, Y.; Ge, F.; Chen, Y.; Xiong, Q. The circular RNA CDR1as regulate cell proliferation via TMED2 and TMED10. BMC Cancer 2020, 20, 1–12. [Google Scholar] [CrossRef] [Green Version]
  40. Yang, G.; Zhang, Y.; Yang, J. Identification of Potentially Functional CircRNA-miRNA-mRNA Regulatory Network in Gastric Carcinoma using Bioinformatics Analysis. Med. Sci. Monit. 2019, 25, 8777–8796. [Google Scholar] [CrossRef]
  41. Liu, C.-G.; Li, J.; Xu, Y.; Li, W.; Fang, S.-X.; Zhang, Q.; Xin, H.-W.; Ma, Z. Long non-coding RNAs and circular RNAs in tumor angiogenesis: From mechanisms to clinical significance. Mol. Ther. Oncolytics 2021, 22, 336–354. [Google Scholar] [CrossRef] [PubMed]
  42. Adams, R.H.; Alitalo, K. Molecular regulation of angiogenesis and lymphangiogenesis. Nat. Rev. Mol. Cell Biol. 2007, 8, 464–478. [Google Scholar] [CrossRef] [PubMed]
  43. Cook, K.M.; Figg, W.D. Angiogenesis Inhibitors: Current Strategies and Future Prospects. CA Cancer J. Clin. 2010, 60, 222–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Ma, Z.; Wang, L.Z.; Cheng, J.-T.; Lam, W.S.T.; Ma, X.; Xiang, X.; Wong, A.L.-A.; Goh, B.C.; Gong, Q.; Sethi, G.; et al. Targeting Hypoxia-Inducible Factor-1-Mediated Metastasis for Cancer Therapy. Antioxidants Redox Signal. 2021, 34, 1484–1497. [Google Scholar] [CrossRef] [PubMed]
  45. Peng, T.; Deng, X.; Tian, F.; Li, Z.; Jiang, P.; Zhao, X.; Chen, G.; Chen, Y.; Zheng, P.; Li, D.; et al. The interaction of LOXL2 with GATA6 induces VEGFA expression and angiogenesis in cholangiocarcinoma. Int. J. Oncol. 2019, 55, 657–670. [Google Scholar] [CrossRef] [Green Version]
  46. De Bock, K.; Mazzone, M.; Carmeliet, P. Antiangiogenic therapy, hypoxia, and metastasis: Risky liaisons, or not? Nat. Rev. Clin. Oncol. 2011, 8, 393–404. [Google Scholar] [CrossRef] [Green Version]
  47. Weis, S.M.; Cheresh, D.A. Tumor angiogenesis: Molecular pathways and therapeutic targets. Nat. Med. 2011, 17, 1359–1370. [Google Scholar] [CrossRef]
  48. Ma, Z.; Xiang, X.; Li, S.; Xie, P.; Gong, Q.; Goh, B.-C.; Wang, L. Targeting hypoxia-inducible factor-1, for cancer treatment: Recent advances in developing small-molecule inhibitors from natural compounds. Semin. Cancer Biol. 2020, 80, 379–390. [Google Scholar] [CrossRef]
  49. Jayson, G.C.; Hicklin, D.J.; Ellis, L.M. Antiangiogenic therapy—evolving view based on clinical trial results. Nat. Rev. Clin. Oncol. 2012, 9, 297–303. [Google Scholar] [CrossRef]
  50. Kim, D.H.; Park, S.; Kim, H.; Choi, Y.J.; Kim, S.Y.; Sung, K.J.; Sung, Y.H.; Choi, C.-M.; Yun, M.; Yi, Y.-S.; et al. Tumor-derived exosomal miR-619-5p promotes tumor angiogenesis and metastasis through the inhibition of RCAN1.4. Cancer Lett. 2020, 475, 2–13. [Google Scholar] [CrossRef]
  51. Zhang, C.; Luo, Y.; Cao, J.; Wang, X.; Miao, Z.; Shao, G. Exosomal lncRNA FAM225A accelerates esophageal squamous cell carcinoma progression and angiogenesis via sponging miR-206 to upregulate NETO2 and FOXP1 expression. Cancer Med. 2020, 9, 8600–8611. [Google Scholar] [CrossRef] [PubMed]
  52. Xie, M.; Yu, T.; Jing, X.; Ma, L.; Fan, Y.; Yang, F.; Ma, P.; Jiang, H.; Wu, X.; Shu, Y.; et al. Exosomal circSHKBP1 promotes gastric cancer progression via regulating the miR-582-3p/HUR/VEGF axis and suppressing HSP90 degradation. Mol. Cancer 2020, 19, 1–22. [Google Scholar] [CrossRef] [PubMed]
  53. Deng, T.; Zhang, H.; Yang, H.; Wang, H.; Bai, M.; Sun, W.; Wang, X.; Si, Y.; Ning, T.; Zhang, L.; et al. Exosome miR-155 Derived from Gastric Carcinoma Promotes Angiogenesis by Targeting the c-MYB/VEGF Axis of Endothelial Cells. Mol. Ther. Nucleic Acids 2020, 19, 1449–1459. [Google Scholar] [CrossRef]
  54. Zhou, Z.; Zhang, H.; Deng, T.; Ning, T.; Liu, R.; Liu, D.; Bai, M.; Ying, G.; Ba, Y. Exosomes Carrying MicroRNA-155 Target Forkhead Box O3 of Endothelial Cells and Promote Angiogenesis in Gastric Cancer. Mol. Ther. Oncolytics 2019, 15, 223–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Yang, H.; Zhang, H.; Ge, S.; Ning, T.; Bai, M.; Li, J.; Li, S.; Sun, W.; Deng, T.; Zhang, L.; et al. Exosome-Derived miR-130a Activates Angiogenesis in Gastric Cancer by Targeting C-MYB in Vascular Endothelial Cells. Mol. Ther. 2018, 26, 2466–2475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Bai, M.; Li, J.; Yang, H.; Zhang, H.; Zhou, Z.; Deng, T.; Zhu, K.; Ning, T.; Fan, Q.; Ying, G.; et al. miR-135b Delivered by Gastric Tumor Exosomes Inhibits FOXO1 Expression in Endothelial Cells and Promotes Angiogenesis. Mol. Ther. 2019, 27, 1772–1783. [Google Scholar] [CrossRef] [PubMed]
  57. Umezu, T.; Tadokoro, H.; Azuma, K.; Yoshizawa, S.; Ohyashiki, K.; Ohyashiki, J.H. Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood 2014, 124, 3748–3757. [Google Scholar] [CrossRef]
  58. Du, J.; Liang, Y.; Li, J.; Zhao, J.-M.; Lin, X.-Y. Gastric Cancer Cell-Derived Exosomal microRNA-23a Promotes Angiogenesis by Targeting PTEN. Front. Oncol. 2020, 10, 326. [Google Scholar] [CrossRef] [Green Version]
  59. Hsu, Y.-L.; Hung, J.-Y.; Chang, W.-A.; Lin, Y.-S.; Pan, Y.-C.; Tsai, P.-H.; Wu, C.-Y.; Kuo, P.-L. Hypoxic lung cancer-secreted exosomal miR-23a increased angiogenesis and vascular permeability by targeting prolyl hydroxylase and tight junction protein ZO-1. Oncogene 2017, 36, 4929–4942. [Google Scholar] [CrossRef]
  60. Moh-Moh-Aung, A.; Fujisawa, M.; Ito, S.; Katayama, H.; Ohara, T.; Ota, Y.; Yoshimura, T.; Matsukawa, A. Decreased miR-200b-3p in cancer cells leads to angiogenesis in HCC by enhancing endothelial ERG expression. Sci. Rep. 2020, 10, 10418. [Google Scholar] [CrossRef]
  61. Zeng, Z.; Li, Y.; Pan, Y.; Lan, X.; Song, F.; Sun, J.; Zhou, K.; Liu, X.; Ren, X.; Wang, F.; et al. Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nat. Commun. 2018, 9, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Hu, H.-Y.; Yu, C.-H.; Zhang, H.-H.; Zhang, S.-Z.; Yu, W.-Y.; Yang, Y.; Chen, Q. Exosomal miR-1229 derived from colorectal cancer cells promotes angiogenesis by targeting HIPK2. Int. J. Biol. Macromol. 2019, 132, 470–477. [Google Scholar] [CrossRef] [PubMed]
  63. Shang, A.; Wang, X.; Gu, C.; Liu, W.; Sun, J.; Zeng, B.; Chen, C.; Ji, P.; Wu, J.; Quan, W.; et al. Exosomal miR-183-5p promotes angiogenesis in colorectal cancer by regulation of FOXO1. Aging 2020, 12, 8352–8371. [Google Scholar] [CrossRef] [PubMed]
  64. Lawson, J.; Dickman, C.; Towle, R.; Jabalee, J.; Javer, A.; Garnis, C. Extracellular vesicle secretion of miR-142-3p from lung adenocarcinoma cells induces tumor promoting changes in the stroma through cell-cell communication. Mol. Carcinog. 2018, 58, 376–387. [Google Scholar] [CrossRef]
  65. Hsu, Y.-L.; Hung, J.-Y.; Chang, W.A.; Jian, S.-F.; Lin, Y.-S.; Pan, Y.-C.; Wu, C.-Y.; Kuo, P.-L. Hypoxic Lung-Cancer-Derived Extracellular Vesicle MicroRNA-103a Increases the Oncogenic Effects of Macrophages by Targeting PTEN. Mol. Ther. 2018, 26, 568–581. [Google Scholar] [CrossRef] [Green Version]
  66. Pontis, F.; Roz, L.; Mensah, M.; Segale, M.; Moro, M.; Bertolini, G.; Petraroia, I.; Centonze, G.; Ferretti, A.M.; Suatoni, P.; et al. Circulating extracellular vesicles from individuals at high-risk of lung cancer induce pro-tumorigenic conversion of stromal cells through transfer of miR-126 and miR-320. J. Exp. Clin. Cancer Res. 2021, 40, 1–16. [Google Scholar] [CrossRef]
  67. Masoumi-Dehghi, S.; Babashah, S.; Sadeghizadeh, M. microRNA-141-3p-containing small extracellular vesicles derived from epithelial ovarian cancer cells promote endothelial cell angiogenesis through activating the JAK/STAT3 and NF-κB signaling pathways. J. Cell Commun. Signal. 2020, 14, 233–244. [Google Scholar] [CrossRef]
  68. He, L.; Zhu, W.; Chen, Q.; Yuan, Y.; Wang, Y.; Wang, J.; Wu, X. Ovarian cancer cell-secreted exosomal miR-205 promotes metastasis by inducing angiogenesis. Theranostics 2019, 9, 8206–8220. [Google Scholar] [CrossRef]
  69. Lu, J.; Liu, Q.-H.; Wang, F.; Tan, J.-J.; Deng, Y.-Q.; Peng, X.-H.; Liu, X.; Zhang, B.; Xu, X.; Li, X.-P. Exosomal miR-9 inhibits angiogenesis by targeting MDK and regulating PDK/AKT pathway in nasopharyngeal carcinoma. J. Exp. Clin. Cancer Res. 2018, 37, 1–12. [Google Scholar] [CrossRef] [Green Version]
  70. Chen, X.; Yang, F.; Zhang, T.; Wang, W.; Xi, W.; Li, Y.; Zhang, D.; Huo, Y.; Zhang, J.; Yang, A.; et al. MiR-9 promotes tumorigenesis and angiogenesis and is activated by MYC and OCT4 in human glioma. J. Exp. Clin. Cancer Res. 2019, 38, 1–16. [Google Scholar] [CrossRef] [Green Version]
  71. Bao, L.; You, B.; Shi, S.; Shan, Y.; Zhang, Q.; Yue, H.; Zhang, J.; Zhang, W.; Shi, Y.; Liu, Y.; et al. Metastasis-associated miR-23a from nasopharyngeal carcinoma-derived exosomes mediates angiogenesis by repressing a novel target gene TSGA10. Oncogene 2018, 37, 2873–2889. [Google Scholar] [CrossRef] [PubMed]
  72. Tadokoro, H.; Umezu, T.; Ohyashiki, K.; Hirano, T.; Ohyashiki, J.H. Exosomes derived from hypoxic leukemia cells enhance tube formation in endothelial cells. J. Biol. Chem. 2013, 288, 34343–34351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Lin, X.-J.; Fang, J.-H.; Yang, X.-J.; Zhang, C.; Yuan, Y.; Zheng, L.; Zhuang, S.M. Hepatocellular carcinoma cell-secreted exosomal microRNA-210 promotes angiogenesis in vitro and in vivo. Mol. Ther. Nucleic Acids 2018, 11, 243–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Wang, Z.-F.; Liao, F.; Wu, H.; Dai, J. Glioma stem cells-derived exosomal miR-26a promotes angiogenesis of microvessel endothelial cells in glioma. J. Exp. Clin. Cancer Res. 2019, 38, 1–15. [Google Scholar] [CrossRef] [Green Version]
  75. Shang, D.; Xie, C.; Hu, J.; Tan, J.; Yuan, Y.; Liu, Z.; Yang, Z. Pancreatic cancer cell–derived exosomal microRNA-27a promotes angiogenesis of human microvascular endothelial cells in pancreatic cancer via BTG2. J. Cell. Mol. Med. 2019, 24, 588–604. [Google Scholar] [CrossRef] [Green Version]
  76. Yang, Y.; Guo, Z.; Chen, W.; Wang, X.; Cao, M.; Han, X.; Zhang, K.; Teng, B.; Cao, J.; Wu, W.; et al. M2 Macrophage-Derived Exosomes Promote Angiogenesis and Growth of Pancreatic Ductal Adenocarcinoma by Targeting E2F2. Mol. Ther. 2020, 29, 1226–1238. [Google Scholar] [CrossRef]
  77. Wu, F.; Li, F.; Lin, X.; Xu, F.; Cui, R.-R.; Zhong, J.-Y.; Zhu, T.; Shan, S.-K.; Liao, X.-B.; Yuan, L.-Q.; et al. Exosomes increased angiogenesis in papillary thyroid cancer microenvironment. Endocr. Relat. Cancer 2019, 26, 525–538. [Google Scholar] [CrossRef] [Green Version]
  78. Pakravan, K.; Babashah, S.; Sadeghizadeh, M.; Mowla, S.J.; Mossahebi-Mohammadi, M.; Ataei, F.; Dana, N.; Javan, M. MicroRNA-100 shuttled by mesenchymal stem cell-derived exosomes suppresses in vitro angiogenesis through modulating the mTOR/HIF-1α/VEGF signaling axis in breast cancer cells. Cell. Oncol. 2017, 40, 457–470. [Google Scholar] [CrossRef]
  79. Zhuang, H.; Wang, H.; Yang, H.; Li, H. Exosome-Encapsulated MicroRNA-21 from Esophageal Squamous Cell Carcinoma Cells Enhances Angiogenesis of Human Umbilical Venous Endothelial Cells by Targeting SPRY1. Cancer Manag. Res. 2020, 12, 10651–10667. [Google Scholar] [CrossRef]
  80. Zhou, Y.; Ren, H.; Dai, B.; Li, J.; Shang, L.; Huang, J.; Shi, X. Hepatocellular carcinoma-derived exosomal miRNA-21 contributes to tumor progression by converting hepatocyte stellate cells to cancer-associated fibroblasts. J. Exp. Clin. Cancer Res. 2018, 37, 324. [Google Scholar] [CrossRef] [Green Version]
  81. Wang, Y.; Lu, J.; Chen, L.; Bian, H.; Hu, J.; Li, D.; Xia, C.; Xu, H. Tumor-Derived EV-Encapsulated miR-181b-5p Induces Angiogenesis to Foster Tumorigenesis and Metastasis of ESCC. Mol. Ther. Nucleic Acids 2020, 20, 421–437. [Google Scholar] [CrossRef] [PubMed]
  82. Yao, X.; Xie, L.; Zeng, Y. MiR-9 Promotes Angiogenesis via Targeting on Sphingosine-1- Phosphate Receptor 1. Front. Cell Dev. Biol. 2020, 8, 755. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, X.; Wang, Y.; Wang, X.; Zou, B.; Mei, J.; Peng, X.; Wu, Z. Extracellular vesicles-encapsulated microRNA-10a-5p shed from cancer-associated fibroblast facilitates cervical squamous cell carcinoma cell angiogenesis and tumorigenicity via Hedgehog signaling pathway. Cancer Gene Ther. 2020, 28, 529–542. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, Y.; Meng, W.; Yue, P.; Li, X. M2 macrophage-derived extracellular vesicles promote gastric cancer progression via a microRNA-130b-3p/MLL3/GRHL2 signaling cascade. J. Exp. Clin. Cancer Res. 2020, 39, 1–20. [Google Scholar] [CrossRef] [PubMed]
  85. Cheng, Y.; Dai, X.; Yang, T.; Zhang, N.; Liu, Z.; Jiang, Y. Low Long Noncoding RNA Growth Arrest-Specific Transcript 5 Expression in the Exosomes of Lung Cancer Cells Promotes Tumor Angiogenesis. J. Oncol. 2019, 2019, 1–13. [Google Scholar] [CrossRef] [PubMed]
  86. Lang, H.-L.; Hu, G.-W.; Zhang, B.; Kuang, W.; Chen, Y.; Wu, L.; Xu, G.-H. Glioma cells enhance angiogenesis and inhibit endothelial cell apoptosis through the release of exosomes that contain long non-coding RNA CCAT2. Oncol. Rep. 2017, 38, 785–798. [Google Scholar] [CrossRef] [Green Version]
  87. Lang, H.-L.; Hu, G.-W.; Chen, Y.; Liu, Y.; Tu, W.; Lu, Y.-M.; Wu, L.; Xu, G.-H. Glioma cells promote angiogenesis through the release of exosomes containing long non-coding RNA POU3F3. Eur. Rev. Med Pharmacol. Sci. 2017, 21, 959–972. [Google Scholar]
  88. Cheng, C.; Zhang, Z.; Cheng, F.; Shao, Z. Exosomal lncRNA RAMP2-AS1 Derived from Chondrosarcoma Cells Promotes Angiogenesis Through miR-2355-5p/VEGFR2 Axis. Onco. Targets Ther. 2020, 13, 3291–3301. [Google Scholar] [CrossRef] [Green Version]
  89. Li, Y.; Lin, S.; Xie, X.; Zhu, H.; Fan, T.; Wang, S. Highly enriched exosomal lncRNA OIP5-AS1 regulates osteosarcoma tumor angiogenesis and autophagy through miR-153 and ATG5. Am. J. Transl. Res. 2021, 13, 4211–4223. [Google Scholar]
  90. Guo, Z.; Wang, X.; Yang, Y.; Chen, W.; Zhang, K.; Teng, B.; Huang, C.; Zhao, Q.; Qiu, Z. Hypoxic Tumor-Derived Exosomal Long Noncoding RNA UCA1 Promotes Angiogenesis via miR-96-5p/AMOTL2 in Pancreatic Cancer. Mol. Ther. Nucleic Acids 2020, 22, 179–195. [Google Scholar] [CrossRef]
  91. Fang, X.; Cai, Y.; Xu, Y.; Zhang, H. Exosome-mediated lncRNA SNHG11 regulates angiogenesis in pancreatic carcinoma through miR-324-3p/VEGFA axis. Cell Biol. Int. 2022, 46, 106–117. [Google Scholar] [CrossRef] [PubMed]
  92. Dai, G.; Yang, Y.; Liu, S.; Liu, H. Hypoxic Breast Cancer Cell-Derived Exosomal SNHG1 Promotes Breast Cancer Growth and Angiogenesis via Regulating miR-216b-5p/JAK2 Axis. Cancer Manag. Res. 2022, 14, 123–133. [Google Scholar] [CrossRef] [PubMed]
  93. Kong, X.; Li, J.; Li, Y.; Duan, W.; Qi, Q.; Wang, T.; Yang, Q.; Du, L.; Mao, H.; Wang, C. A novel long non-coding RNA AC073352.1 promotes metastasis and angiogenesis via interacting with YBX1 in breast cancer. Cell Death Dis. 2021, 12, 1–13. [Google Scholar] [CrossRef] [PubMed]
  94. Qiu, J.-J.; Lin, X.-J.; Tang, X.-Y.; Zheng, T.-T.; Lin, Y.-Y.; Hua, K.-Q. Exosomal Metastasis-Associated Lung Adenocarcinoma Transcript 1 Promotes Angiogenesis and Predicts Poor Prognosis in Epithelial Ovarian Cancer. Int. J. Biol. Sci. 2018, 14, 1960–1973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Lei, L.; Mou, Q. Exosomal taurine up-regulated 1 promotes angiogenesis and endothelial cell proliferation in cervical cancer. Cancer Biol. Ther. 2020, 21, 717–725. [Google Scholar] [CrossRef] [PubMed]
  96. You, L.-N.; Tai, Q.-W.; Xu, L.; Hao, Y.; Guo, W.-J.; Zhang, Q.; Tong, Q.; Zhang, H.; Huang, W.-K. Exosomal LINC00161 promotes angiogenesis and metastasis via regulating miR-590-3p/ROCK axis in hepatocellular carcinoma. Cancer Gene Ther. 2021, 28, 719–736. [Google Scholar] [CrossRef]
  97. Conigliaro, A.; Costa, V.; Dico, A.L.; Saieva, L.; Buccheri, S.; Dieli, F.; Manno, M.; Raccosta, S.; Mancone, C.; Tripodi, M.; et al. CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA. Mol. Cancer 2015, 14, 155. [Google Scholar] [CrossRef]
  98. Huang, X.-Y.; Huang, Z.-L.; Huang, J.; Xu, B.; Huang, X.-Y.; Xu, Y.-H.; Zhou, J.; Tang, Z.-Y. Exosomal circRNA-100338 promotes hepatocellular carcinoma metastasis via enhancing invasiveness and angiogenesis. J. Exp. Clin. Cancer Res. 2020, 39, 20. [Google Scholar] [CrossRef] [Green Version]
  99. Hu, K.; Li, N.; Li, J.; Chen, Z.; Wang, J.; Sheng, L. Exosome circCMTM3 promotes angiogenesis and tumorigenesis of hepatocellular carcinoma through miR-3619-5p/SOX9. Hepatol. Res. 2021, 51, 1139–1152. [Google Scholar] [CrossRef]
  100. Bai, L.; Gao, Z.; Jiang, A.; Ren, S.; Wang, B. Circular noncoding RNA circ_0007334 sequestrates miR-577 to derepress KLF12 and accelerate colorectal cancer progression. AntiCancer Drugs 2022, 33, e409–e422. [Google Scholar] [CrossRef]
  101. Zeng, W.; Liu, Y.; Li, W.; Zhu, J. CircFNDC3B sequestrates miR-937-5p to derepress TIMP3 and inhibit colorectal cancer progression. Mol. Oncol. 2020, 14, 2960–2984. [Google Scholar] [CrossRef] [PubMed]
  102. Li, Y.; Chen, J.; Chen, Z.; Xu, X.; Weng, J.; Zhang, Y.; Mo, Y.; Liu, Y.; Wang, J.; Ke, Y. CircGLIS3 Promotes High-Grade Glioma Invasion via Modulating Ezrin Phosphorylation. Front. Cell Dev. Biol. 2021, 9. [Google Scholar] [CrossRef] [PubMed]
  103. Li, B.; Hong, J.; Hong, M.; Wang, Y.; Yu, T.; Zang, S.; Wu, Q. piRNA-823 delivered by multiple myeloma-derived extracellular vesicles promoted tumorigenesis through re-educating endothelial cells in the tumor environment. Oncogene 2019, 38, 5227–5238. [Google Scholar] [CrossRef] [PubMed]
  104. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  105. Chistiakov, D.A.; Chekhonin, V.P. Extracellular vesicles shed by glioma cells: Pathogenic role and clinical value. Tumor Biol. 2014, 35, 8425–8438. [Google Scholar] [CrossRef]
  106. van der Vos, K.E.; Abels, E.R.; Zhang, X.; Lai, C.; Carrizosa, E.; Oakley, D.; Prabhakar, S.; Mardini, O.; Crommentuijn, M.H.W.; Skog, J.; et al. Directly visualized glioblastoma-derived extracellular vesicles transfer RNA to microglia/macrophages in the brain. Neuro Oncol. 2016, 18, 58–69. [Google Scholar] [CrossRef] [Green Version]
  107. Xie, F.; Zhou, X.; Fang, M.; Li, H.; Su, P.; Tu, Y.; Zhang, L.; Zhou, F. Extracellular Vesicles in Cancer Immune Microenvironment and Cancer Immunotherapy. Adv. Sci. 2019, 6, 1901779. [Google Scholar] [CrossRef]
  108. Turley, S.J.; Cremasco, V.; Astarita, J.L. Immunological hallmarks of stromal cells in the tumour microenvironment. Nat. Rev. Immunol. 2015, 15, 669–682. [Google Scholar] [CrossRef]
  109. De Visser, K.E.; Eichten, A.; Coussens, L.M. Paradoxical roles of the immune system during cancer development. Nat. Cancer 2006, 6, 24–37. [Google Scholar] [CrossRef]
  110. Joyce, J.A.; Pollard, J.W. Microenvironmental regulation of metastasis. Nat. Cancer 2008, 9, 239–252. [Google Scholar] [CrossRef]
  111. Hsieh, C.-H.; Tai, S.-K.; Yang, M.-H. Snail-overexpressing Cancer Cells Promote M2-Like Polarization of Tumor-Associated Macrophages by Delivering MiR-21-Abundant Exosomes. Neoplasia 2018, 20, 775–788. [Google Scholar] [CrossRef] [PubMed]
  112. Öhlund, D.; Elyada, E.; Tuveson, D. Fibroblast heterogeneity in the cancer wound. J. Exp. Med. 2014, 211, 1503–1523. [Google Scholar] [CrossRef] [PubMed]
  113. Zhou, X.; Yan, T.; Huang, C.; Xu, Z.; Wang, L.; Jiang, E.; Wang, H.; Chen, Y.; Liu, K.; Shao, Z.; et al. Melanoma cell-secreted exosomal miR-155-5p induce proangiogenic switch of cancer-associated fibroblasts via SOCS1/JAK2/STAT3 signaling pathway. J. Exp. Clin. Cancer Res. 2018, 37, 242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Quesada, A.M.R.; Muñoz-Chápuli, R.; Medina, M. Angiogenesis and signal transduction in endothelial cells. Exp. 2004, 61, 2224–2243. [Google Scholar] [CrossRef]
  115. Castellano, J.J.; Marrades, R.M.; Molins, L.; Viñolas, N.; Moises, J.; Canals, J.; Han, B.; Li, Y.; Martinez, D.; Monzó, M.; et al. Extracellular Vesicle lincRNA-p21 Expression in Tumor-Draining Pulmonary Vein Defines Prognosis in NSCLC and Modulates Endothelial Cell Behavior. Cancers 2020, 12, 734. [Google Scholar] [CrossRef] [Green Version]
  116. Chen, X.; Zhang, S.; Du, K.; Zheng, N.; Liu, Y.; Chen, H.; Xie, G.; Ma, Y.; Zhou, Y.; Zheng, Y.; et al. Gastric cancer–secreted exosomal X26nt increases angiogenesis and vascular permeability by targeting VE-cadherin. Cancer Sci. 2020, 112, 1839–1852. [Google Scholar] [CrossRef]
  117. Zou, S.; Tong, Q.; Liu, B.; Huang, W.; Tian, Y.; Fu, X. Targeting STAT3 in Cancer Immunotherapy. Mol. Cancer 2020, 19, 1–19. [Google Scholar] [CrossRef]
  118. Visan, I. Targeting TGF-β in cancer. Nat. Immunol. 2018, 19, 316. [Google Scholar] [CrossRef]
  119. Orlova, V.V.; Liu, Z.; Goumans, M.-J.; ten Dijke, P. Controlling angiogenesis by two unique TGF-β type I receptor signaling pathways. Histol. Histopathol. 2011, 26, 1219–1230. [Google Scholar] [CrossRef]
  120. The Blue Nile Project. Lancet 1985, 2, 1166–1167. [CrossRef]
  121. Bunney, T.; Katan, M. Phosphoinositide signalling in cancer: Beyond PI3K and PTEN. Nat. Cancer 2010, 10, 342–352. [Google Scholar] [CrossRef] [PubMed]
  122. Chew, C.L.; Conos, S.A.; Unal, B.; Tergaonkar, V. Noncoding RNAs: Master Regulators of Inflammatory Signaling. Trends Mol. Med. 2018, 24, 66–84. [Google Scholar] [CrossRef] [PubMed]
  123. Jiang, L.; Gu, Y.; Du, Y.; Liu, J. Exosomes: Diagnostic Biomarkers and Therapeutic Delivery Vehicles for Cancer. Mol. Pharm. 2019, 16, 3333–3349. [Google Scholar] [CrossRef] [PubMed]
  124. Zhang, W.; Su, X.; Li, S.; Liu, Z.; Wang, Q.; Zeng, H. Low serum exosomal miR-484 expression predicts unfavorable prognosis in ovarian cancer. Cancer Biomark. 2020, 27, 485–491. [Google Scholar] [CrossRef]
  125. Liu, C.-J.; Xie, G.-Y.; Miao, Y.-R.; Xia, M.; Wang, Y.; Lei, Q.; Zhang, Q.; Guo, A.-Y. EVAtlas: A comprehensive database for ncRNA expression in human extracellular vesicles. Nucleic Acids Res. 2021, 50, D111–D117. [Google Scholar] [CrossRef]
  126. Weber, J.A.; Baxter, D.H.; Zhang, S.; Huang, D.Y.; How Huang, K.; Jen Lee, M.; Galas, D.J.; Wang, K. The MicroRNA Spectrum in 12 Body Fluids. Clin. Chem. 2010, 56, 1733–1741. [Google Scholar] [CrossRef]
  127. Maishi, N.; Hida, K. Tumor endothelial cells accelerate tumor metastasis. Cancer Sci. 2017, 108, 1921–1926. [Google Scholar] [CrossRef] [Green Version]
  128. Ogata-Kawata, H.; Izumiya, M.; Kurioka, D.; Honma, Y.; Yamada, Y.; Furuta, K.; Gunji, T.; Ohta, H.; Okamoto, H.; Sonoda, H.; et al. Circulating Exosomal microRNAs as Biomarkers of Colon Cancer. PLoS ONE 2014, 9, e92921. [Google Scholar] [CrossRef]
  129. Pan, L.; Liang, W.; Fu, M.; Huang, Z.-H.; Li, X.; Zhang, W.; Zhang, P.; Qian, H.; Jiang, P.-C.; Xu, W.-R.; et al. Exosomes-mediated transfer of long noncoding RNA ZFAS1 promotes gastric cancer progression. J. Cancer Res. Clin. Oncol. 2017, 143, 991–1004. [Google Scholar] [CrossRef]
  130. Xu, H.; Chen, Y.; Dong, X.; Wang, X. Serum Exosomal Long Noncoding RNAs ENSG00000258332.1 and LINC00635 for the Diagnosis and Prognosis of Hepatocellular Carcinoma. Cancer Epidemiol. Biomark. Prev. 2018, 27, 710–716. [Google Scholar] [CrossRef] [Green Version]
  131. Xian, J.; Su, W.; Liu, L.; Rao, B.; Lin, M.; Feng, Y.; Qiu, F.; Chen, J.; Zhou, Q.; Zhao, Z.; et al. Identification of Three Circular RNA Cargoes in Serum Exosomes as Diagnostic Biomarkers of Non–Small-Cell Lung Cancer in the Chinese Population. J. Mol. Diagn. 2020, 22, 1096–1108. [Google Scholar] [CrossRef] [PubMed]
  132. Chen, X.; Chen, R.-X.; Wei, W.-S.; Li, Y.-H.; Feng, Z.-H.; Tan, L.; Chen, J.-W.; Yuan, G.-J.; Chen, S.-L.; Guo, S.-J.; et al. PRMT5 Circular RNA Promotes Metastasis of Urothelial Carcinoma of the Bladder through Sponging miR-30c to Induce Epithelial–Mesenchymal Transition. Clin. Cancer Res. 2018, 24, 6319–6330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Zhou, Y.; Zhou, G.; Tian, C.; Jiang, W.; Jin, L.; Zhang, C.; Chen, X. Exosome-mediated small RNA delivery for gene therapy. Wiley Interdiscip. Rev. RNA 2016, 7, 758–771. [Google Scholar] [CrossRef] [PubMed]
  134. Zhang, H.; Bai, M.; Deng, T.; Liu, R.; Wang, X.; Qu, Y.; Duan, J.; Zhang, L.; Ning, T.; Ge, S.; et al. Cell-derived microvesicles mediate the delivery of miR-29a/c to suppress angiogenesis in gastric carcinoma. Cancer Lett. 2016, 375, 331–339. [Google Scholar] [CrossRef]
  135. Wang, J.; Jiang, Q.; Faleti, O.D.; Tsang, C.-M.; Zhao, M.; Wu, G.; Tsao, S.-W.; Fu, M.; Chen, Y.; Ding, T.; et al. Exosomal Delivery of AntagomiRs Targeting Viral and Cellular MicroRNAs Synergistically Inhibits Cancer Angiogenesis. Mol. Ther. Nucleic Acids 2020, 22, 153–165. [Google Scholar] [CrossRef]
  136. He, A.T.; Liu, J.; Li, F.; Yang, B.B. Targeting circular RNAs as a therapeutic approach: Current strategies and challenges. Signal Transduct. Target. Ther. 2021, 6, 1–14. [Google Scholar] [CrossRef]
  137. Chen, S.; Chen, X.; Luo, Q.; Liu, X.; Wang, X.; Cui, Z.; He, A.; He, S.; Jiang, Z.; Wu, N.; et al. Retinoblastoma cell-derived exosomes promote angiogenesis of human vesicle endothelial cells through microRNA-92a-3p. Cell Death Dis. 2021, 12, 1–11. [Google Scholar] [CrossRef]
  138. Zhao, L.; Liu, W.; Xiao, J.; Cao, B. The role of exosomes and “exosomal shuttle microRNA” in tumorigenesis and drug resistance. Cancer Lett. 2014, 356, 339–346. [Google Scholar] [CrossRef]
  139. Li, G.; Gong, J.; Cao, S.; Wu, Z.; Cheng, D.; Zhu, J.; Huang, X.; Tang, J.; Yuan, Y.; Cai, W.; et al. The Non-Coding RNAs Inducing Drug Resistance in Ovarian Cancer: A New Perspective for Understanding Drug Resistance. Front. Oncol. 2021, 11. [Google Scholar] [CrossRef]
  140. Deng, Z.; Rong, Y.; Teng, Y.; Zhuang, X.; Samykutty, A.; Mu, J.; Zhang, L.; Cao, P.; Yan, J.; Miller, D.; et al. Exosomes miR-126a released from MDSC induced by DOX treatment promotes lung metastasis. Oncogene 2016, 36, 639–651. [Google Scholar] [CrossRef] [Green Version]
  141. Fu, X.; Liu, M.; Qu, S.; Ma, J.; Zhang, Y.; Shi, T.; Wen, H.; Yang, Y.; Wang, S.; Wang, J.; et al. Exosomal microRNA-32-5p induces multidrug resistance in hepatocellular carcinoma via the PI3K/Akt pathway. J. Exp. Clin. Cancer Res. 2018, 37, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Xuan, Z.; Chen, C.; Tang, W.; Ye, S.; Zheng, J.; Zhao, Y.; Shi, Z.; Zhang, L.; Sun, H.; Shao, C. TKI-Resistant Renal Cancer Secretes Low-Level Exosomal miR-549a to Induce Vascular Permeability and Angiogenesis to Promote Tumor Metastasis. Front. Cell Dev. Biol. 2021, 9. [Google Scholar] [CrossRef]
  143. Guo, C.; Liu, J.; Zhou, Q.; Song, J.; Zhang, Z.; Li, Z.; Wang, G.; Yuan, W.; Sun, Z. Exosomal Noncoding RNAs and Tumor Drug Resistance. Cancer Res. 2020, 80, 4307–4313. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Formation and release of EVs. The formation of EVs involves the following processes: Proteins are transported from the Golgi apparatus or internalised from the cell surface, and nucleic acids are endocytosed and transferred to early endosomes. Early endosomes gradually mature into late endosomes and MVBs, some of which are degraded, whereas others are secreted as exosomes. These exosomes carry multiple biological components, including proteins, lipids, and nucleic acids (e.g., ncRNAs), which are delivered to the recipient cell through different ways: a. phagocytosis, b. receptor–ligand interaction, and c. direct fusion.
Figure 1. Formation and release of EVs. The formation of EVs involves the following processes: Proteins are transported from the Golgi apparatus or internalised from the cell surface, and nucleic acids are endocytosed and transferred to early endosomes. Early endosomes gradually mature into late endosomes and MVBs, some of which are degraded, whereas others are secreted as exosomes. These exosomes carry multiple biological components, including proteins, lipids, and nucleic acids (e.g., ncRNAs), which are delivered to the recipient cell through different ways: a. phagocytosis, b. receptor–ligand interaction, and c. direct fusion.
Cells 11 00947 g001
Figure 2. EV-derived ncRNAs regulate cell–cell communication in the angiogenic microenvironment. Tumor cells (A), immune cells, and stromal cells (B) release EVs containing miRNAs, lncRNAs, circRNAs, and piRNAs, contributing to tumor angiogenesis within the tumor microenvironment. ECs: endothelial cells, CAFs: cancer-associated fibroblasts, and MSCs: mesenchymal stromal cells.
Figure 2. EV-derived ncRNAs regulate cell–cell communication in the angiogenic microenvironment. Tumor cells (A), immune cells, and stromal cells (B) release EVs containing miRNAs, lncRNAs, circRNAs, and piRNAs, contributing to tumor angiogenesis within the tumor microenvironment. ECs: endothelial cells, CAFs: cancer-associated fibroblasts, and MSCs: mesenchymal stromal cells.
Cells 11 00947 g002
Figure 3. The potential clinical application of EV-derived ncRNAs in tumor angiogenesis (A) EV-derived ncRNAs can be detected from patient samples and are potential diagnostic and prognostic biomarkers. (B) A combination of targeting EV-derived ncRNAs and using conventional anti-angiogenic agents can enhance therapeutic efficacy.
Figure 3. The potential clinical application of EV-derived ncRNAs in tumor angiogenesis (A) EV-derived ncRNAs can be detected from patient samples and are potential diagnostic and prognostic biomarkers. (B) A combination of targeting EV-derived ncRNAs and using conventional anti-angiogenic agents can enhance therapeutic efficacy.
Cells 11 00947 g003
Table 1. The emerging roles of EV-derived ncRNAs in tumor angiogenesis.
Table 1. The emerging roles of EV-derived ncRNAs in tumor angiogenesis.
EV-Derived ncRNAs ExpressionSource Cell Function and MechanismTumor TypeReference
miR-155UpregulatedTumor cellPromotes angiogenesis via the c-MYB/VEGF axis Gastric cancer[53]
UpregulatedTumor cellPromotes angiogenesis by inhibiting FOXO3aGastric cancer[54]
miR-130aUpregulatedTumor cellActivates angiogenesis by inhibiting c-MYB Gastric cancer[55]
miR-135bUpregulatedTumor cellPromotes angiogenesis by inhibiting FOXO1 Gastric cancer[56]
UpregulatedTumor cellRegulates the HIF/FIH signalling pathwayMultiple myeloma[57]
miR-23aUpregulatedTumor cellInhibits PTEN and activates the AKT pathwayGastric cancer[58]
UpregulatedTumor cellIncreases angiogenesis by inhibiting ZO-1 Lung cancer [59]
miR-200b-3pDownregulatedTumor cellEnhances endothelial ERG expressionHepatocellular carcinoma[60]
miR-25-3pUpregulatedTumor cellInhibits KLF2 and KLF4, thereby elevating VEGFR2 expressionColorectal cancer[61]
miR-1229UpregulatedTumor cellInhibits HIPK2, thereby activating the VEGF pathwayColorectal cancer [62]
miR-183-5pUpregulatedTumor cellInhibits FOXO1, thereby promoting expression of VEGFA, VEGFAR2, ANG2, PIGF, MMP-2, and MMP-9 Colorectal cancer[63]
miR-142-3pUpregulatedTumor cellInhibits TGFβR1Lung adenocarcinoma [64]
miR-103aUpregulatedTumor cellInhibits PTEN, thereby promoting the polarization of M2 macrophages Lung cancer[65]
miR-126UpregulatedMSCsUpregulates CD34 and CXCR4, thereby promoting expression of VEGFLung cancer[66]
miR-141-3pUpregulatedTumor cellInhibits SOCS5, thereby activating JAK/STAT3 and NF-κB signalling pathwaysOvarian cancer[67]
miR-205UpregulatedTumor cellRegulates the PTEN/AKT pathwayOvarian cancer[68]
miR-9DownregulatedTumor cellInhibits MDK, thereby regulating the PDK/AKT signalling pathwayNasopharyngeal carcinoma[69]
UpregulatedTumor cellPromotes angiogenesis by targeting COL18A1, THBS2, PTCH1, and PHD3Glioma [70]
miR-23aUpregulatedTumor cellPromotes angiogenesis by inhibiting TSGA10Nasopharyngeal carcinoma[71]
miR-210UpregulatedTumor cellEnhances tube formation by inhibiting EFNA3Leukemia[72]
UpregulatedTumor cellPromotes angiogenesis by inhibiting SMAD4 and STAT6Hepatocellular carcinoma[73]
miR-26aUpregulatedTumor cellInhibits PTEN, thereby activating the PI3K/AKT signalling pathwayGlioma[74]
miR-27aUpregulatedTumor cellInhibits BTG2, thereby promoting VEGF, VEGFR, MMP-2, and MMP-9 expressionPancreatic cancer[75]
miR-155-5p
/miR-221-5p
UpregulatedM2 macrophagesPromotes angiogenesis by targeting E2F2Pancreatic cancer[76]
miR-21-5pUpregulatedTumor cellPromotes angiogenesis by targeting TGFBI and COL4A1Papillary carcinoma[77]
miR-100- -MSCsRegulates the mTOR/HIF-1α signalling axisBreast cancer[78]
miR-21UpregulatedTumor cellInhibits SPRY1, thereby promoting VEGF expressionOesophageal squamous cell carcinoma[79]
UpregulatedTumor cellInhibits PTEN, thereby activating PDK1/AKT signalling Hepatocellular carcinoma[80]
miR-181b-5pUpregulatedTumor cellInhibits PTEN and PHLPP2, thereby activating AKT signallingOesophageal squamous cell carcinoma[81]
miR-9UpregulatedTumor cellInhibits S1P, thereby promoting VEGF expressionMedulloblastoma and xenoglioblastoma[82]
miR-10a-5pUpregulatedCAFsInhibits TBX5, thereby activating Hedgehog signalling Cervical squamous cell carcinoma[83]
miR-135bUpregulatedTumor cellEnhances angiogenesis by targeting FIH-1Multiple myeloma [57]
miR-130b-3pUpregulatedM2 macrophagesRegulates the miR-130b-3p/MLL3/GRHL2 signalling cascadeGastric cancer[84]
lncGAS5DownregulatedTumor cellInhibits angiogenesis by regulating the miR-29-3p/PTEN axisLung cancer[85]
lnc-CCAT2UpregulatedTumor cellPromotes VEGFA and TGF-β expressionGlioma[86]
lnc-POU3F3UpregulatedTumor cellPromotes bFGF, bFGFR, and VEGFA expressionGlioma[87]
lncRNA RAMP2-AS1UpregulatedTumor cellPromotes angiogenesis through the miR-2355-5p/VEGFR2 axisChondrosarcoma [88]
OIP5-AS1UpregulatedTumor cellRegulates angiogenesis and autophagy through miR-153/ATG5 axisOsteosarcoma[89]
FAM225AUpregulatedTumor cellPromotes angiogenesis through the miR-206/NETO2/FOXP1 axis Oesophageal squamous cell carcinoma[51]
UCA1UpregulatedTumor cellPromotes angiogenesis through the miR-96-5p/AMOTL2 axisPancreatic cancer[90]
SNHG11UpregulatedTumor cellPromotes angiogenesis through the miR-324-3p/VEGFA axisPancreatic cancer[91]
SNHG1UpregulatedTumor cellPromotes angiogenesis by regulating the miR-216b-5p/JAK2 axisBreast cancer[92]
AC073352.1UpregulatedTumor cellBinds and stabilizes the YBX1 proteinBreast cancer[93]
MALAT1UpregulatedTumor cellFacilitates angiogenesis and predicts poor prognosisOvarian cancer[94]
TUG1UpregulatedTumor cellPromotes angiogenesis by inhibiting caspase-3 activity Cervical cancer[95]
LINC00161UpregulatedTumor cellPromotes angiogenesis and metastasis by regulating the miR-590-3p/ROCK axisHepatocellular carcinoma[96]
H19 UpregulatedCancer stem cellPromotes VEGF production and release in ECsLiver cancer[97]
circSHKBP1UpregulatedTumor cellEnhances VEGF mRNA stability by the miR-582-3p/HUR axis Gastric cancer [52]
circRNA-100,338UpregulatedTumor cellFacilitates HCC metastasis by enhancing invasiveness and angiogenesisHepatocellular carcinoma[98]
circCMTM3UpregulatedTumor cellPromotes angiogenesis and HCC tumor growth by the miR-3619-5p/SOX9 axisHepatocellular carcinoma[99]
circ_0007334UpregulatedTumor cellAccelerates CRC tumor growth and angiogenesis by the miR-577/KLF12 axisColorectal cancer [100]
CircFNDC3BDownregulatedTumor cellInhibits angiogenesis and CRC progression by the miR-937-5p/TIMP3 axisColorectal cancer [101]
circGLIS3UpregulatedTumor cellInduces endothelial cell angiogenesis by promoting Ezrin T567 phosphorylationGlioma[102]
piRNA-823UpregulatedTumor cellPromotes VEGF and IL-6 expressionMultiple myeloma[103]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hu, M.; Li, J.; Liu, C.-G.; Goh, R.M.W.J.; Yu, F.; Ma, Z.; Wang, L. Noncoding RNAs of Extracellular Vesicles in Tumor Angiogenesis: From Biological Functions to Clinical Significance. Cells 2022, 11, 947. https://doi.org/10.3390/cells11060947

AMA Style

Hu M, Li J, Liu C-G, Goh RMWJ, Yu F, Ma Z, Wang L. Noncoding RNAs of Extracellular Vesicles in Tumor Angiogenesis: From Biological Functions to Clinical Significance. Cells. 2022; 11(6):947. https://doi.org/10.3390/cells11060947

Chicago/Turabian Style

Hu, Miao, Juan Li, Chen-Guang Liu, Robby Miguel W. J. Goh, Fenggang Yu, Zhaowu Ma, and Lingzhi Wang. 2022. "Noncoding RNAs of Extracellular Vesicles in Tumor Angiogenesis: From Biological Functions to Clinical Significance" Cells 11, no. 6: 947. https://doi.org/10.3390/cells11060947

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

Article Metrics

Back to TopTop