Next Article in Journal
The Emerging Role of Non-Coding RNAs (ncRNAs) in Plant Growth, Development, and Stress Response Signaling
Next Article in Special Issue
Topographic Distribution of miRNAs (miR-30a, miR-223, miR-let-7a, miR-let-7f, miR-451, and miR-486) in the Plasma Extracellular Vesicles
Previous Article in Journal
Circular RNAs, Noncoding RNAs, and N6-methyladenosine Involved in the Development of MAFLD
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Functional Relevance of Extracellular Vesicle-Derived Long Non-Coding and Circular RNAs in Cancer Angiogenesis

by
José A. Peña-Flores
*,
Daniela Muela-Campos
,
Rebeca Guzmán-Medrano
,
Diego Enríquez-Espinoza
and
Karla González-Alvarado
Doctoral Program in Biomedical and Stomatological Sciences, Faculty of Dentistry, Autonomous University of Chihuahua, Chihuahua 31000, Mexico
*
Author to whom correspondence should be addressed.
Non-Coding RNA 2024, 10(1), 12; https://doi.org/10.3390/ncrna10010012
Submission received: 19 December 2023 / Revised: 2 February 2024 / Accepted: 4 February 2024 / Published: 6 February 2024
(This article belongs to the Special Issue Extracellular Vesicles and ncRNA)

Abstract

:
Extracellular vesicles (EVs) are defined as subcellular structures limited by a bilayer lipid membrane that function as important intercellular communication by transporting active biomolecules, such as proteins, amino acids, metabolites, and nucleic acids, including long non-coding RNAs (lncRNAs). These cargos can effectively be delivered to target cells and induce a highly variable response. LncRNAs are functional RNAs composed of at least 200 nucleotides that do not code for proteins. Nowadays, lncRNAs and circRNAs are known to play crucial roles in many biological processes, including a plethora of diseases including cancer. Growing evidence shows an active presence of lnc- and circRNAs in EVs, generating downstream responses that ultimately affect cancer progression by many mechanisms, including angiogenesis. Moreover, many studies have revealed that some tumor cells promote angiogenesis by secreting EVs, which endothelial cells can take up to induce new vessel formation. In this review, we aim to summarize the bioactive roles of EVs with lnc- and circRNAs as cargo and their effect on cancer angiogenesis. Also, we discuss future clinical strategies for cancer treatment based on current knowledge of circ- and lncRNA-EVs.

1. Introduction

1.1. Cancer Generalities

Cancer is defined as a group of diseases that are multifactorial in nature and represent a challenge in their diagnosis and treatment due to their etiological diversity [1]. More than 200 types of human cancer have been identified based on the cell or tissue from where they originate, the somatic mutations acquired at any time of the progression of the disease, and the microenvironment influences in which they develop [2]. One of the hallmark features of cancer is its rapid and uncontrolled progression due to mutations that alter the cell cycle and overpass checkpoint regulation between the cell cycle phases, promoting the accumulation of mutations passed down to the progeny [3]. For this rapid progression to occur, the growing tumor has a high demand for nutrients and other components; thus, these cells generate molecular signaling to promote the formation of new blood vessels from preexisting ones, a process denominated angiogenesis [4]. The angiogenesis process is fundamental for cancer to advance locally and facilitate metastasis, and therefore, it has been extensively studied in most cancer types [5,6,7]. Multiple efforts have been made to develop antiangiogenic therapies to halt tumor growth and prevent metastasis [8,9]. Recently, the role of extracellular vesicles (EVs) between vessels in tumor communication has triggered the interest of many researchers.

1.2. Extracellular Vesicles

Extracellular vesicles are subcellular structures that are heterogeneous in nature and surrounded by a lipid bilayer membrane that exerts multiple functions in intercellular communication [10]. Based on how they are delivered from the original cell to the extracellular medium, EVs can be released by inward budding of the endosomal membrane or outward budding of the cellular membrane [11]. The recipient cell can then internalize EVs through endocytosis or membrane fusion to unload their contents into the cell cytoplasm (Figure 1) [12]. Since their discovery in the early 1980s, many biomolecules have been identified as cargo in EVs, including proteins, amino acids, signaling lipids, and different genetic molecules like DNA, RNA, and non-coding RNAs, promoting both physiological and pathological processes [13]. Based on their biogenesis, EVs are generally classified into microvesicles and exosomes [10], although some authors suggest a further division into apoptotic bodies and proteasomes [14]. Microvesicles are generated by outward budding of the plasma membrane and range from 50 nm to 1000 nm. In contrast, exosomes are membrane vesicles smaller in size (30–100 nm) and are formed by inward budding of the endosomal membrane to be later secreted by fusion with the cell membrane [14,15]. The role of EVs as cell-to-cell mediators in respiratory disease [16], neurodegenerative disease [17], kidney disease [18], cardiovascular disease [19], and cancer progression and metastasis [20,21,22] has been documented to ameliorate the understanding of the behavior or these subcellular structures.

1.3. Long Non-Coding and circRNAs

Long non-coding RNAs (lncRNAs) are a diverse group of RNAs that are not translated into proteins, and they are at least 200 nucleotides in length [24]. Recent advances in genomic sciences through RNA sequencing have offered the identification of lncRNAs performing functions to control chromatin complexes, recruit transcription factors, regulate alternative splicing, affect mRNA translation, sponge micro-RNAs by binding, degrade other RNAs, and serve as scaffolds for protein interactions (Figure 2) [25,26,27]. Evidence suggests an active role of lncRNAs in most physiological processes, and their involvement in disease has been the focus of active research in recent years [28,29]. The involvement of lncRNAs as oncogenes or tumor suppressors in many cancer types has also been documented. However, as new lncRNAs are discovered, the general landscape becomes complicated as the roles they can perform become more complex [30]. For instance, lncRNAs can influence the progression of cancer by promoting metastasis [31], drug resistance [32], epithelial-to-mesenchymal transition (EMT) [33], and angiogenesis [34].
It has recently been shown that some lncRNAs can take a circular shape and join covalently at the ends, and these are called circRNAs [35]. This type of lncRNA can perform similar functions to linear lncRNAs, as sponges to recruit specific miRNAs or as effectors to regulate the expression of certain genes [36]. circRNAs have recently been widely studied, arousing interest due to their stability, since, unlike linear non-coding RNAs, they are difficult to degrade [37].
Figure 2. Molecular functions of lncRNAs. (A) lncRNAs can guide chromatin complexes, controlling between transcriptionally active euchromatin and silent heterochromatin. (B) The recruitment of polymerase II and transcription factors can be inhibited or facilitated by lncRNAs. (C) lncRNAs contribute to transcriptome complexity by regulating alternative splicing of pre-mRNAs. (D) lncRNAs affect the stability and translation of mRNA by base-pairing with mRNA molecules. (E) They influence the expression of miRNAs by binding to them and preventing their function. (F) lncRNAs can act as siRNAs and target other RNAs, which subsequently could result in target degradation. (G) lncRNAs can join multiple protein factors as flexible scaffolds to interact with or cooperate in protein–protein interactions. (H,I) The scaffold function is also important for protein activity and localization as well as subcellular structures. Modified from Peña-Flores et al. [38].
Figure 2. Molecular functions of lncRNAs. (A) lncRNAs can guide chromatin complexes, controlling between transcriptionally active euchromatin and silent heterochromatin. (B) The recruitment of polymerase II and transcription factors can be inhibited or facilitated by lncRNAs. (C) lncRNAs contribute to transcriptome complexity by regulating alternative splicing of pre-mRNAs. (D) lncRNAs affect the stability and translation of mRNA by base-pairing with mRNA molecules. (E) They influence the expression of miRNAs by binding to them and preventing their function. (F) lncRNAs can act as siRNAs and target other RNAs, which subsequently could result in target degradation. (G) lncRNAs can join multiple protein factors as flexible scaffolds to interact with or cooperate in protein–protein interactions. (H,I) The scaffold function is also important for protein activity and localization as well as subcellular structures. Modified from Peña-Flores et al. [38].
Ncrna 10 00012 g002
Recently, the presence of both coding and non-coding RNA in EVs has motivated research to elucidate RNA’s role in various biological mechanisms in cancer and other diseases [39,40]. This review aims to provide recent evidence on the influence of exosomal lncRNAs on cancer angiogenesis. A systematic screening of papers was performed on PubMed, Google Scholar, Cochrane Library, Web of Science, and EMBASE up to July 2023 for articles matching the following criteria: (long non-coding RNA or lncRNA or ceRNA or circRNA or circular RNA) and (angiogenesis or lymphangiogenesis or vasculogenic mimicry) and (extracellular vesicles or EV or exosome or exosomal). The titles and abstracts were carefully read, and full-text manuscripts relevant to the subject of study were acquired for further analysis.

2. Mechanism of Angiogenesis

The angiogenesis process embodies forming new blood vessels from existing vessels in response to physiological and pathological mechanisms [41]. During embryogenesis, the vascular network develops through a combination of vasculogenesis, referred to as the de novo formation of the heart and new blood vessels from stem endothelial cells, namely, angioblasts, and angiogenesis, which expands the initial primitive vascular plexus [42]. Although most blood vessels remain quiescent under physiological conditions, tissue repair and regeneration through wound healing, ovulation, and endometrial thickening throughout the menstrual cycle are based on angiogenesis for proper functioning [43,44,45]. While vascular growth varies depending on where angiogenesis is initiated and the tissue to which they will provide a new blood supply, several mechanisms are common in forming these vessels [46]. In a hypoxic state, the recruitment of cells that promote inflammation; angiogenic growth factor production; degradation of the basement membrane; and endothelial cells (ECs) sprouting, migrating, proliferating, differentiating, and modulating vascular support cells are some of the shared characteristics in angiogenesis [47].
The angiogenic process (Figure 3) comprises several stages involving the sprouting, migration, and proliferation of ECs guided by the vascular endothelial growth factor (VEGF) [48]. Following VEGF stimulation, pericytes from the vessel wall detach, and the basal membrane is weakened by proteolytic degradation. At the same time, ECs adopt an invasive and motile phenotype called tip cells that send out filamentous pseudopodia to guide vascular budding [49]. The cells behind the tip cells are denominated stalk cells, which proliferate to maintain the integrity of the structure and function of the nascent vessels, mainly expanding the vascular lumen [50]. ECs modify their shape by negatively charging glycoproteins on the apical surface to repel each other and open the lumen while redistributing cell-to-cell adhesion to the periphery [51]. For maturation to occur, pericytes must be recruited by the platelet-derived growth factor subunit B (PDGF-B) and angiopoietin 1 (Ang1) signaling along with the strengthening and consolidation of the adhesion between ECs with junctional molecules such as VE-cadherin, while a basement membrane is deposited by tissue inhibitors of metalloproteinases (TIMPs) [52,53].
Under physiological conditions, angiogenesis is strongly regulated by factors ranging from metabolites to hormones [54]. Various molecular pathways have been extensively studied that comprise the angiogenesis process, including the VEGF-VEGFR, Angiopoietin-Tie, Delta-Notch, and Ephrin-Eph [55]. Angiogenesis, vasculogenesis, and lymphangiogenesis are mostly regulated by six VEGF members encoded in the human genome, namely, VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, and the placenta growth factor (P1GF) [48,56]. According to their molecular configuration and affinity, the aforementioned factors may bind to different tyrosine kinase receptors VEGFR-1, VEGFR-2, and/or VEGFR-3; VEGF-A binds to VEGFR-2 to contribute to angiogenesis, whereas VEGF-C and VEGF-D enhance lymphatic vessel sprouting by binding to VEGFR-3 [57]. Another group of angiogenesis inducers are the platelet-derived growth factors (PDGF), which induce ECs proliferation and migration by binding to two tyrosine kinase receptors, PDGFR-α and PDGFR-β [58]. Another angiogenesis factor is the fibroblast growth factor (FGF) 2, responsible for inducing metalloproteinase (MMP) secretion to degrade the basement membrane and promote vessel sprouting along with VEGF [59]. Angiopoietin 1, interleukin 8 (IL-8), epidermal growth factor (EGF), and tumor necrosis factor α (TNF-α) also exert a pro-angiogenic effect in ECs through several signaling pathways [60,61,62,63]. Conversely, angiopoietin 2, angiostatin, endostatin, vasostatin, and TIMPs inhibit angiogenesis and play an important role in achieving vascular homeostasis [64,65].
Extracellular vesicles produced by many cellular lineages under specific circumstances can be taken up by ECs to promote and regulate angiogenesis [66]. For instance, a study in endometrial stromal cells (HESCs) found active secretion of EVs during decidualization in a controlled manner by the hypoxia-inducible factor 2 alpha (HIF2α)–Ras-related protein Rab-27B (RAB27B) cascade, revealing a cargo with a variety of growth regulators, signaling molecules, metabolic modulators, and factors that control the expansion and remodeling of ECs [67]. In a myocardial infarction animal model, stem cell-derived small extracellular vesicles (sEVs) loaded with miR-486-5p promoted cardiac angiogenesis via fibroblastic MMP19-VEGFA cleavage signaling [68]. Moreover, Gregorius et al. [69] evaluated the effects of mesenchymal stromal cell (MSC)-derived sEVs on the proliferation, migration, and tube formation of cerebral microvascular ECs. Interestingly, hypoxic preconditioning enhanced angiogenesis and increased post-ischemic endothelial survival by regulating several miRNAs through the uptake of protein-enriched sEVs cargo. Another study demonstrated that HS-1 protein X-1 (HAX1), a major regulator of myeloid homeostasis, was present in EVs secreted by nasopharyngeal carcinoma (NPC) tumors, promoting an angiogenesis phenotype by activating the focal adhesion kinase (FAK) pathway in ECs by increasing the expression level of integrin subunit beta 6 (ITGB6) [70]. Conversely, EVs produced by bone marrow MSCs were found to be enriched in the cluster of differentiation 39 (CD39), TIMP-1, and CD73, inhibiting tumor angiogenesis by targeting the extracellular matrix remodeling and the endothelial cell migration [71].

3. Exosomal Long Non-Coding and Circular RNAs in Cancer Angiogenesis

LncRNAs in EV cargo have been demonstrated lately, mainly in cancer [72,73,74]. A recent study launched an online repository of EV long RNAs (exLRs) in diverse human body fluids, comprising 19,643 mRNAs, 15,645 lncRNAs, and 79,084 circRNAs obtained from human blood, cerebrospinal fluid, bile, and urine samples. The database provides novel exLR signatures to help discover new biomarkers that could aid in diagnosing and treating many diseases [75]. Based on available recent research, Casado-Díaz et al. [76] concluded that lncRNAs and other RNAs included in MSC-derived EVs can be applied in chronic skin ulcers to improve accelerated healing and decrease scar formation due to immunosuppressive and immunomodulatory properties. Conversely, in a diabetic wound-healing animal model, upregulated lncRNAs packed in EVs from fibroblasts enhanced keratinocyte MMP-9 expression to induce collagen degradation, delaying wound healing [77]. Recently, the long non-coding repressor of NFAT (NRON) was detected in BMSC-derived EVs, inhibiting osteoclast differentiation and osteoporotic bone loss in vitro and in vivo [78].
In tumors, the high rate of cell proliferation forces the formation of new blood vessels [79]. However, in most cases, these blood vessels are dilated, tortuous, and immature, leading to excessive permeability and increased hypoxia [80]. In addition, vascular disorganization causes heterogeneity in the tumor blood vessel network, creating highly vascularized tumor areas and other hypoxic areas with low vascular density [47]. Thus, hypoxia becomes a major driver of tumor angiogenesis, along with other mechanisms promoted by activated oncogenes or loss of tumor suppressor genes, in which lncRNAs play an important role, mainly through acting as competing endogenous RNAs for miRNAs [81]. Similarly, circRNAs have been extensively studied in cancer, elucidating important roles in tumor development, growth, and angiogenesis [82]. For instance, VEGFR-related pathways have been linked to circRNAs by affecting tumor angiogenesis by sponging miRNAs [83,84]. The landscape of exosomal lnc- and circRNAs in angiogenesis in cancer is summarized in Table 1 and Figure 4.

3.1. Bone Malignancies

Some studies have been dedicated to studying exosomal lncRNAs in bone malignancies. LncRNA Opa-interacting protein 5-antisense 1 (OIP5-AS1) was found to be overexpressed in exosomes secreted by osteosarcoma cells, increasing angiogenesis in tubule formation assays by mechanistically sponging miR-153 and increasing the autophagy-related 5 protein (ATG5) [85]. Interestingly, serum samples from osteosarcoma patients could transfer via EVs the myocardial infarction-associated transcript (MIAT), promoting the proliferation of osteosarcoma cell lines and angiogenesis in HUVECs by sponging miR-613 and upregulating G protein-coupled receptor 158 (GPR158) [86]. In an in vitro and animal model, BMSC-EVs carried the non-coding RNA activated by DNA damage (NORAD) into osteosarcoma cells and upregulated CREB-binding protein (CREBBP) by sponging miR-877-3p to promote proliferation, invasion, migration, and angiogenesis [87]. Another lncRNA called Ewing sarcoma-associated transcript 1 (EWSAT1) was found to regulate osteosarcoma-induced angiogenesis via two mechanisms: (1) by increasing in sensitivity/reactivity of vascular endothelial cells triggered by exosomes carrying EWSAT1, and (2) by increasing angiogenic factors secretion [88]. Moreover, exosomes secreted by chondrosarcoma cells were loaded with the receptor activity-modifying protein 2 antisense 1 (RAMP2-AS1). They could enhance HUVECs proliferation, migration, and tube formation by acting as a ceRNA for miR2355-5p to regulate VEGFR2 expression. In addition, the overexpression of RAMP2-AS1 in the serum of chondrosarcoma patients was demonstrated to be closely related to local invasiveness, distant metastasis, and poor prognosis [89].

3.2. Esophageal, Gastric, and Colorectal Cancers

Some of the most prevalent tumors of the gastrointestinal (GI) tract have been explored regarding the role played in angiogenesis by exosomes loaded with different lncRNAs and circRNAs. For instance, the exosomal lncRNA family with sequence similarity 225 member A (FAM225A) was highly expressed in esophageal squamous cell carcinoma (ESCC), upregulating neuropilin and tolloid-like 2 (NETO2) and forkhead box P1 (FOXP1) expression by sponging miR-206 to accelerate tumor progression and angiogenesis [93].
In gastric cancer patients, exosomal circ-SHKBP1 was overexpressed in tumor and blood samples. When the exosomes were isolated and exposed to different cell lines, cells showed a promoted proliferation, invasion, migration, and angiogenesis rate by mechanistically regulating the miR-582-3p/HUR/VEGF axis and suppressing heat shock protein 90 (HSP90) degradation [116]. Similarly, 30 blood samples and tissues from gastric cancer patients were taken to analyze circ-FCH and mu domain-containing endocytic adaptor 2 (FCHO2). It was found that circ-FCHO2 up-modulation led to a poor outcome, while circ-FCHO2 silencing weakened the proliferation, invasion, angiogenesis, and stem cell characteristics, presumably by activating the Janus kinase 1 (JAK1)/signal transducer and activator of transcription 2 (STAT2) pathway via sponging miR-194-5p [117]. Conversely, by acting as a miR-587 sponge to adjust the expression of the sclerostin domain-containing 1 (SOSTDC1), circ-0001190 overexpression inhibited cell viability, proliferation, angiogenesis, migration, and invasion of gastric cancer cell lines [114]. Moreover, circ-0044366 was highly expressed in gastric cancer and impaired the proliferation, migration, and tube formation of HUVECs by exosomal communication by acting as miR-29a ceRNA and regulating the VEGF pathway [115].
In colorectal cancer (CRC), tumor growth, angiogenesis, and liver metastasis were suppressed by exosomal circ-fibronectin type III domain-containing 3B (FNDC3B) overexpression by acting via the miR-97-5p/TIMP3 pathway [98]. Similarly, exosomes derived from lncRNA adenomatous polyposis coli (APC1)-silenced CRC cells promoted angiogenesis by activating the mitogen-activated protein kinase 1 (MAPK) pathway in endothelial cells, while enforced APC1 was sufficient to inhibit CRC growth, metastasis, and tumor angiogenesis by suppressing exosome production [99]. Interestingly, exosomes loaded with circ-tubulin gamma complex component 4 (TUBGCP4) derived from CRC cells enhanced vascular endothelial cell migration and tube formation via inducing filopodia formation and endothelial cell tipping by upregulating the pyruvate dehydrogenase kinase 2 (PDK2) to activate the AKT serine/threonine kinase 1 (AKT) signaling pathway and by sponging miR-146b-3p [100]. A very interesting study by Zhi et al. [125] compared EVs derived from the b-Raf proto-oncogene (BRAF) wild-type CRC and the BRAFV600E mutant patients to find the overexpression of 13 lncRNAs and downregulation of 22 lncRNAs in exosomes from the BRAFV600E mutation type. This difference showed a higher microvascular and micro-lymphatic vessel density of the BRAFV600E mutant CRC tissues.

3.3. Liver and Pancreatic Cancers

LncRNA-loaded exosomes from tumors from other organs related to the GI tract have also shown some relationship with tumor angiogenesis. You et al. [101] reported high levels of Linc-00161 in serum-derived exosomes from hepatocellular cancer (HCC) patients and the supernatants of HCC cell lines, which are associated with poor survival. Mechanistically, Linc-00161 promoted angiogenesis in HUVECs by inhibiting miR-590-3p and activating the Rho-associated coiled-coil-containing protein kinase 2 (ROCK2) axis. In an in vitro study, exosomes with lncRNA H19 were released by CD90+ HCC cells and modulated endothelial cells, promoting an angiogenic phenotype and cell-to-cell adhesion [103]. Similarly, lncRNA ubiquitin-conjugating enzyme E2 C pseudogene 3 (UBE2CP3) was overexpressed in HCC EVs. It promoted HUVEC proliferation, migration, and tube formation via the activation of the ERK/HIF-1α/p70S6K/VEGFA signaling cascade, promoting HCC tumorigenicity [102]. In another study, exosomal circ-100388 affected the cell proliferation, angiogenesis, permeability, and vasculogenic mimicry formation ability of HUVECs and HCC tumor metastasis [104].
In cholangiocarcinoma (CCA), the cholangiocarcinoma-associated circular RNA 1 (circ-CCAC1) from CCA-derived EVs was transferred to endothelial monolayer cells, disrupting endothelial barrier integrity and inducing angiogenesis. Interestingly, circ-CCAC1 increased cell leakiness by sequestering the enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) gene, thus elevating the SH3 domain-containing GRB2 like 2, endophilin A1 (SH3GL2) expression to reduce levels of intercellular junction proteins [124].
In pancreatic cancer, the expression levels of the lncRNA urothelial cancer-associated 1 (UCA1) in exosomes derived from the serum of patients were associated with poor survival, promoting angiogenesis and tumor growth by acting as a ceRNA of miR-96-5p, relieving the repressive effects on the expression of its target gene angiomotin like 2 (AMOTL2) [90]. Moreover, the exosomal small nucleolar RNA host gene 11 (SNHG11) promoted cell proliferation, migration, and angiogenesis in pancreatic cancer cell lines but impeded cell apoptosis via sponging miR-324-3p to upregulate VEGFA expression [126].

3.4. Renal and Bladder Cancers

Some urinary system tumors have observed a relationship between lncRNA-loaded EVs and angiogenesis. In renal cell carcinoma (RCC), RCC-derived exosomes had an lncRNA Ars operon (ARSR) that promoted macrophage polarization, cytokine release, phagocytosis, angiogenesis, and tumor development by sponging miR34/miR-449 and upregulating the signal transducer and activator of transcription 3 (STAT3) pathway [108]. Similarly, RCC-derived exosomal circular scaffold attachment factor B2 (circ-SAFB2) facilitated the progression, invasion, angiogenesis, and metastasis of RCC by inducing the polarization of M2 macrophages through the miR-620/JAK1/STAT3 axis [109]. Conversely, exosomal circular spire type actin nucleation factor 1 (circ-SPIRE1) suppressed angiogenesis and vessel permeability through regulating ELAV-like RNA-binding protein 1-mRNA, binding and upregulating polypeptide N-Acetylgalactosaminyltransferase 3 (GALNT3) and KH domain RNA-binding protein (QKI) expression [110].
In bladder carcinoma (BCa), exosomal brain cytoplasmic RNA 1 (BCYRN1) promoted the tube formation and migration of human lymphatic endothelial cells (HLECs), upregulating the Wnt family member 5A (WNT5A) gene expression by inducing hnRNPA1-associated H2K4 trimethylation in WNAT5a promoter, which activated Wnt/β-catenin signaling to facilitate the secretion of VEGF-C in BCa [111]. Moreover, lymph node metastasis-associated transcript 2 (LNMAT2)-loaded exosomes from BCa tissues and blood samples stimulated the tube formation and migration of HLECs and enhanced tumor lymphangiogenesis and lymph node metastasis by upregulation of prospero homeobox 1 (PROX1) gene expression by recruitment of hnRNPA2B1 and increasing H3K4 trimethylation [112]. Comparably, BCa cell-secreted EVs mediated intercellular communication with HLECs through the transmission of the small nucleolar RNA host gene 16 (ELNAT1) and promoted lymphangiogenesis by inducing the ubiquitin-conjugating enzyme E2 (UBC9) gene overexpression to catalyze the small ubiquitin-like modifier (SUMO) binding of hnRNPA1 at the lysine 113 residue [113].

3.5. Nasopharyngeal and Lung Cancers

LncRNAs in exosomes derived from nasopharyngeal squamous cell carcinoma (NPSCC) and their relationship with tumor angiogenesis have been mildly explored. In serum samples from newly diagnosed NPSCC patients, the long intergenic non-protein-coding RNA, regulator of reprogramming (linc-ROR), was substantially expressed in exosomes that could be taken up by HUVECs, increasing proliferation, migration, and angiogenesis in vitro by mechanistically upregulating the p-AKT/p-VEGFR2 pathway [91]. Similarly, lncRNA colon cancer-associated transcript 2 (CCAT2) was found in EVs derived from NPSCC patients, promoting HUVEC proliferation and angiogenesis promotion [92].
In non-small cell lung carcinoma (NSCLC), NSCLC cells secreted exosomes with melanotransferrin antisense 1 (MFI2-AS1) to induce tube formation by HUVECs, promoting angiogenesis and metastasis by sponging miR-107, which in turn activated the PI3K/AKT pathway [105]. Similarly, high EV Linc-p21 was found in NSCLC blood samples from tumor-draining pulmonary veins before tumor surgical resection. EVs with Linc-p21 were taken up by HUVECs and promoted tube formation and enhanced tumor cell adhesion to endothelial cells by sponging miR-23a, miR-146bv, miR-330, and miR-494 [106]. In contrast, GAS5 was lowly expressed in human lung cancer tissues, lung cancer cells, and cell culture supernatant exosomes. The exosomes of lung cancer cells containing high GAS5 levels inhibited HUVECs proliferation and tube formation, increasing their apoptosis by sponging miR-29-3p and upregulating phosphatase and tensin homolog (PTEN) and inhibiting PI3K/AKT phosphorylation [107].

3.6. Glioma and Gliobastoma

A few studies have reported evidence of the role of exosomal lncRNAs in glioma and glioblastoma angiogenesis. An in vitro study with glioma cell lines demonstrated that HUVECs can take up exosomal CCAT2 to promote migration, proliferation, tubular-like structure formation, and arteriole formation [94]. Similarly, the POU class 3 homeobox 3 (POU3F3) was upregulated in glioma tissue. When human brain microvascular endothelial cells (HBMVECs) were treated with exosomes loaded with POU3F3, they exhibited better migration, proliferation, tubular-like structure formation, and arteriole formation. Mechanistically, POU3F3 was shown to upregulate bFGF, bFGFR, VEGFA, and Angio [95]. Moreover, cell line A172 was cultured to demonstrate that EVs loaded with the HOX transcript antisense RNA (HOTAIR) had a pro-angiogenic activity in HBMVECS via VEGFA [96]. In glioblastoma, lncRNA HIF1A antisense RNA 2 (AHIF) was found upregulated in tissue samples, and when cultured with glioblastoma cell lines, exosomal AHIF regulated factors associated with migration and angiogenesis [97].

3.7. Other Cancer Types

In ovarian cancer, an in vitro study revealed that lncRNA activated by TGF-β (ATB) promoted viability and angiogenesis of HUVECs by sponging miR-204-3p and thus upregulating TGFβ-R2 [127]. Similarly, elevated serum exosomal metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) promoted angiogenesis and was highly correlated with an advanced and metastatic phenotype of epithelial ovarian cancer [118]. Another study demonstrated taurine-upregulated 1 (TUG1) overexpression in human cervical cancer cell lines. When TUG1 was depleted, the exosome-mediated pro-angiogenic potential of HUVECs was impaired by modulating angiogenesis-related genes like VEGFA, MMP9, TGFβ, IL-8, and bFGF [119]. In breast cancer cell lines, the metadherin (MTDH) gene improved cell viability and angiogenesis in endothelial cells. The molecular cascade was promoted by exosomal circular homeodomain-interacting protein kinase 3 (circ-HIPK3), which sponged miR-124-3p and in turn upregulated MTDH [120]. Liu et al. Campo [121] demonstrated that exosomal overexpression of the FYVE, RhoGF, and PH domain-containing 5 antisense 1 (FGD5-AS1) enhanced the proliferation, migration, angiogenesis, and permeability of HUVECs by regulating the endothelial miR-6838-5p/Vav guanine nucleotide exchange factor 2 (VAV2) axis. A total of 25 peripheral blood samples from 20 multiple myeloma patients and 5 matched healthy controls showed overexpression of the circular ATPase phospholipid-transporting 10A (circ-ATP10A) in the multiple myeloma samples, mechanistically acting as a sponge of several miRNAs to consequently regulate the expression of downstream VEGFB, HIF1A, PDGFA, and FGF [122].

4. Clinical Relevance and Future Perspectives

In recent years, a plethora of studies have demonstrated the role played by lncRNAs and circRNAs in many molecular and cellular processes, ranging from early development to complex diseases such as cancer [128,129,130]. Although the intracellular expression of these non-coding RNAs has been documented, the presence of DNA and RNA fragments in extracellular vesicles and exosomes has also been discovered, demonstrating the ability of cells to transmit information not only to their environment and surrounding cells but also to distant areas through fluids like plasma, bile, and urine [131,132,133]. For instance, exosomes derived from serum from pancreatic cancer patients were associated with poor survival when loaded with UCA1 [90]. Similarly, elevated serum exosomal MALAT1 was an independent predictive factor for ovarian cancer overall survival [118]. In the sera of chondrosarcoma patients, exosomal RAMP2-AS1 was closely related to local invasiveness, distant metastasis, and poor prognosis [89]. Serum-derived exosomes loaded with linc-00161 from hepatocellular cancer patients were significantly associated with poor survival [101], and high EV linc-p21 levels in blood were associated with shorter time to relapse and shorter overall survival in lung cancer [106]. The presence of cholangiocarcinoma-derived EVs loaded with circ-CCAT1 was detected in bile samples from perihilar and distal CCA patients, demonstrating a worse overall prognosis [124]. Interestingly, 63% of patients with bladder cancer evaluated as lymph node metastasis (LN)-negative by CT were correctly predicted as being LN-positive by the detection of urinary EV-mediated ELNAT1 [113].
EV-mediated lncRNAs are promising early diagnostic biomarkers and potential therapeutic targets in many diseases. As the relationship between exosomal lnc- and circRNAs and their role in angiogenesis is further studied, tools may be developed for early and accurate diagnosis of diseases such as diabetes and cancer, establishing therapeutic pathways that promote a better prognosis for patients with these and other diseases. The sensitivity and specificity for the detection of circulating RNAs must be improved to apply these techniques in regular clinical practice.

5. Conclusions

Exosomes have recently been the subject of study due to the molecular cargo they possess since they apparently function as intercellular communication mechanisms. Among the components that can generate changes in other cells are lnc- and circRNAs, which can influence fundamental processes for functional and pathological development, such as angiogenesis. As the roles of exosomal lnc- and circRNAs in angiogenesis are elucidated, diagnostic and prognostic tools can be developed to improve advanced systemic disease therapies.

Author Contributions

J.A.P.-F. and D.E.-E. contributed to conception and design of the review. J.A.P.-F. and D.M.-C. organized the database. J.A.P.-F. wrote the first draft of the manuscript. J.A.P.-F., D.M.-C., R.G.-M. and K.G.-A. wrote sections of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors acknowledge CONAHCyT for JAPF’s (1194005) fellowship. The authors also would like to thank Alexis Álvarez-Ramírez, Kaori E. Galaso-Trujillo, Andrés A. Barraza-Gómez, Ángel A. Sáenz-Chávez, Gustavo Barajas-Aguirre, and Milena Corral-Apodaca.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Lewandowska, A.M.; Rudzki, M.; Rudzki, S.; Lewandowski, T.; Laskowska, B. Environmental risk factors for cancer—Review paper. Ann. Agric. Environ. Med. 2019, 26, 1–7. [Google Scholar] [CrossRef] [PubMed]
  2. Álvarez-Garcia, V.; Tawil, Y.; Wise, H.M.; Leslie, N.R. Mechanisms of PTEN loss in cancer: It’s all about diversity. Semin. Cancer Biol. 2019, 59, 66–79. [Google Scholar] [CrossRef] [PubMed]
  3. Leal-Esteban, L.C.; Fajas, L. Cell cycle regulators in cancer cell metabolism. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165715. [Google Scholar] [CrossRef] [PubMed]
  4. Li, S.; Xu, H.X.; Wu, C.T.; Wang, W.Q.; Jin, W.; Gao, H.L.; Li, H.; Zhang, S.R.; Xu, J.Z.; Qi, Z.H.; et al. Angiogenesis in pancreatic cancer: Current research status and clinical implications. Angiogenesis 2019, 22, 15–36. [Google Scholar] [CrossRef]
  5. Badodekar, N.; Sharma, A.; Patil, V.; Telang, G.; Sharma, R.; Patil, S.; Vyas, N.; Somasundaram, I. Angiogenesis induction in breast cancer: A paracrine paradigm. Cell Biochem. Funct. 2021, 39, 860–873. [Google Scholar] [CrossRef]
  6. Tan, H.W.; Xu, Y.M.; Qin, S.H.; Chen, G.F.; Lau, A.T.Y. Epigenetic regulation of angiogenesis in lung cancer. J. Cell Physiol. 2021, 236, 3194–3206. [Google Scholar] [CrossRef]
  7. Unterleuthner, D.; Neuhold, P.; Schwarz, K.; Janker, L.; Neuditschko, B.; Nivarthi, H.; Crncec, I.; Kramer, N.; Unger, C.; Hengstschläger, M.; et al. Cancer-associated fibroblast-derived WNT2 increases tumor angiogenesis in colon cancer. Angiogenesis 2020, 23, 159–177. [Google Scholar] [CrossRef]
  8. Liang, P.; Ballou, B.; Lv, X.; Si, W.; Bruchez, M.P.; Huang, W.; Dong, X. Monotherapy and Combination Therapy Using Anti-Angiogenic Nanoagents to Fight Cancer. Adv. Mater. 2021, 33, e2005155. [Google Scholar] [CrossRef]
  9. Lopes-Coelho, F.; Martins, F.; Pereira, S.A.; Serpa, J. Anti-Angiogenic Therapy: Current Challenges and Future Perspectives. Int. J. Mol. Sci. 2021, 22, 3765. [Google Scholar] [CrossRef]
  10. 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]
  11. Abels, E.R.; Breakefield, X.O. Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake. Cell Mol. Neurobiol. 2016, 36, 301–312. [Google Scholar] [CrossRef] [PubMed]
  12. Joshi, B.S.; de Beer, M.A.; Giepmans, B.N.G.; Zuhorn, I.S. Endocytosis of Extracellular Vesicles and Release of Their Cargo from Endosomes. ACS Nano 2020, 14, 4444–4455. [Google Scholar] [CrossRef]
  13. Fonseka, P.; Marzan, A.L.; Mathivanan, S. Introduction to the Community of Extracellular Vesicles. Subcell. Biochem. 2021, 97, 3–18. [Google Scholar] [CrossRef] [PubMed]
  14. Urabe, F.; Kosaka, N.; Ito, K.; Kimura, T.; Egawa, S.; Ochiya, T. Extracellular vesicles as biomarkers and therapeutic targets for cancer. Am. J. Physiol. Cell Physiol. 2020, 318, C29–C39. [Google Scholar] [CrossRef]
  15. D’Souza-Schorey, C.; Schorey, J.S. Regulation and mechanisms of extracellular vesicle biogenesis and secretion. Essays Biochem. 2018, 62, 125–133. [Google Scholar] [CrossRef]
  16. Carnino, J.M.; Lee, H. Extracellular vesicles in respiratory disease. Adv. Clin. Chem. 2022, 108, 105–127. [Google Scholar] [CrossRef] [PubMed]
  17. Hill, A.F. Extracellular Vesicles and Neurodegenerative Diseases. J. Neurosci. 2019, 39, 9269–9273. [Google Scholar] [CrossRef] [PubMed]
  18. Grange, C.; Bussolati, B. Extracellular vesicles in kidney disease. Nat. Rev. Nephrol. 2022, 18, 499–513. [Google Scholar] [CrossRef]
  19. Han, C.; Yang, J.; Sun, J.; Qin, G. Extracellular vesicles in cardiovascular disease: Biological functions and therapeutic implications. Pharmacol. Ther. 2022, 233, 108025. [Google Scholar] [CrossRef]
  20. Becker, A.; Thakur, B.K.; Weiss, J.M.; Kim, H.S.; Peinado, H.; Lyden, D. Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis. Cancer Cell 2016, 30, 836–848. [Google Scholar] [CrossRef]
  21. Chang, C.H.; Pauklin, S. Extracellular vesicles in pancreatic cancer progression and therapies. Cell Death Dis. 2021, 12, 973. [Google Scholar] [CrossRef]
  22. Wu, H.; Fu, M.; Liu, J.; Chong, W.; Fang, Z.; Du, F.; Liu, Y.; Shang, L.; Li, L. The role and application of small extracellular vesicles in gastric cancer. Mol. Cancer 2021, 20, 71. [Google Scholar] [CrossRef]
  23. Wang, M.; Zhou, L.; Yu, F.; Zhang, Y.; Li, P.; Wang, K. The functional roles of exosomal long non-coding RNAs in cancer. Cell Mol. Life Sci. 2019, 76, 2059–2076. [Google Scholar] [CrossRef]
  24. Dahariya, S.; Paddibhatla, I.; Kumar, S.; Raghuwanshi, S.; Pallepati, A.; Gutti, R.K. Long non-coding RNA: Classification, biogenesis and functions in blood cells. Mol. Immunol. 2019, 112, 82–92. [Google Scholar] [CrossRef]
  25. Entezari, M.; Ghanbarirad, M.; Taheriazam, A.; Sadrkhanloo, M.; Zabolian, A.; Goharrizi, M.; Hushmandi, K.; Aref, A.R.; Ashrafizadeh, M.; Zarrabi, A.; et al. Long non-coding RNAs and exosomal lncRNAs: Potential functions in lung cancer progression, drug resistance and tumor microenvironment remodeling. Biomed. Pharmacother. 2022, 150, 112963. [Google Scholar] [CrossRef]
  26. Meng, X.; Wang, Z.F.; Lou, Q.Y.; Rankine, A.N.; Zheng, W.X.; Zhang, Z.H.; Zhang, L.; Gu, H. Long non-coding RNAs in head and neck squamous cell carcinoma: Diagnostic biomarkers, targeted therapies, and prognostic roles. Eur. J. Pharmacol. 2021, 902, 174114. [Google Scholar] [CrossRef]
  27. Nojima, T.; Proudfoot, N.J. Mechanisms of lncRNA biogenesis as revealed by nascent transcriptomics. Nat. Rev. Mol. Cell Biol. 2022, 23, 389–406. [Google Scholar] [CrossRef] [PubMed]
  28. Joshi, M.; Rajender, S. Long non-coding RNAs (lncRNAs) in spermatogenesis and male infertility. Reprod. Biol. Endocrinol. 2020, 18, 103. [Google Scholar] [CrossRef] [PubMed]
  29. Schmitz, S.U.; Grote, P.; Herrmann, B.G. Mechanisms of long noncoding RNA function in development and disease. Cell Mol. Life Sci. 2016, 73, 2491–2509. [Google Scholar] [CrossRef] [PubMed]
  30. Lin, W.; Zhou, Q.; Wang, C.Q.; Zhu, L.; Bi, C.; Zhang, S.; Wang, X.; Jin, H. LncRNAs regulate metabolism in cancer. Int. J. Biol. Sci. 2020, 16, 1194–1206. [Google Scholar] [CrossRef] [PubMed]
  31. Liu, S.J.; Dang, H.X.; Lim, D.A.; Feng, F.Y.; Maher, C.A. Long noncoding RNAs in cancer metastasis. Nat. Rev. Cancer 2021, 21, 446–460. [Google Scholar] [CrossRef]
  32. Taghvimi, S.; Abbaszadeh, S.; Banan, F.B.; Fard, E.S.; Jamali, Z.; Najafabadi, M.A.; Savardashtaki, A.; Movahedpour, A. LncRNAs Roles in Chemoresistance of Cancer Cells. Curr. Mol. Med. 2022, 22, 691–702. [Google Scholar] [CrossRef]
  33. Kichi, Z.A.; Soltani, M.; Rezaei, M.; Shirvani-Farsani, Z.; Rojhannezhad, M. The Emerging Role of EMT-related lncRNAs in Therapy Resistance and their Applications as Biomarkers. Curr. Med. Chem. 2022, 29, 4574–4601. [Google Scholar] [CrossRef]
  34. Jin, K.T.; Yao, J.Y.; Fang, X.L.; Di, H.; Ma, Y.Y. Roles of lncRNAs in cancer: Focusing on angiogenesis. Life Sci. 2020, 252, 117647. [Google Scholar] [CrossRef]
  35. Qin, T.; Li, J.; Zhang, K.Q. Structure, Regulation, and Function of Linear and Circular Long Non-Coding RNAs. Front. Genet. 2020, 11, 150. [Google Scholar] [CrossRef]
  36. Zhou, W.Y.; Cai, Z.R.; Liu, J.; Wang, D.S.; Ju, H.Q.; Xu, R.H. Circular RNA: Metabolism, functions and interactions with proteins. Mol. Cancer 2020, 19, 172. [Google Scholar] [CrossRef]
  37. Patop, I.L.; Wüst, S.; Kadener, S. Past, present, and future of circRNAs. EMBO J. 2019, 38, e100836. [Google Scholar] [CrossRef]
  38. Peña-Flores, J.A.; Bermúdez, M.; Ramos-Payán, R.; Villegas-Mercado, C.E.; Soto-Barreras, U.; Muela-Campos, D.; Álvarez-Ramírez, A.; Pérez-Aguirre, B.; Larrinua-Pacheco, A.D.; López-Camarillo, C.; et al. Emerging role of lncRNAs in drug resistance mechanisms in head and neck squamous cell carcinoma. Front. Oncol. 2022, 12, 965628. [Google Scholar] [CrossRef] [PubMed]
  39. Kim, K.M.; Abdelmohsen, K.; Mustapic, M.; Kapogiannis, D.; Gorospe, M. RNA in extracellular vesicles. WIREs RNA 2017, 8, e1413. [Google Scholar] [CrossRef] [PubMed]
  40. Momen-Heravi, F.; Getting, S.J.; Moschos, S.A. Extracellular vesicles and their nucleic acids for biomarker discovery. Pharmacol. Ther. 2018, 192, 170–187. [Google Scholar] [CrossRef] [PubMed]
  41. Nowak-Sliwinska, P.; Alitalo, K.; Allen, E.; Anisimov, A.; Aplin, A.C.; Auerbach, R.; Augustin, H.G.; Bates, D.O.; van Beijnum, J.R.; Bender, R.H.F.; et al. Consensus guidelines for the use and interpretation of angiogenesis assays. Angiogenesis 2018, 21, 425–532. [Google Scholar] [CrossRef]
  42. Patan, S. Vasculogenesis and angiogenesis. Cancer Treat. Res. 2004, 117, 3–32. [Google Scholar] [CrossRef] [PubMed]
  43. Ng, S.W.; Norwitz, G.A.; Pavlicev, M.; Tilburgs, T.; Simón, C.; Norwitz, E.R. Endometrial Decidualization: The Primary Driver of Pregnancy Health. Int. J. Mol. Sci. 2020, 21, 4092. [Google Scholar] [CrossRef] [PubMed]
  44. Veith, A.P.; Henderson, K.; Spencer, A.; Sligar, A.D.; Baker, A.B. Therapeutic strategies for enhancing angiogenesis in wound healing. Adv. Drug Deliv. Rev. 2019, 146, 97–125. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, F.; Qian, H.; Kong, L.; Wang, W.; Wang, X.; Xu, Z.; Chai, Y.; Xu, J.; Kang, Q. Accelerated Bone Regeneration by Astragaloside IV through Stimulating the Coupling of Osteogenesis and Angiogenesis. Int. J. Biol. Sci. 2021, 17, 1821–1836. [Google Scholar] [CrossRef] [PubMed]
  46. Dudley, A.C.; Griffioen, A.W. Pathological angiogenesis: Mechanisms and therapeutic strategies. Angiogenesis 2023, 26, 313–347. [Google Scholar] [CrossRef] [PubMed]
  47. Viallard, C.; Larrivée, B. Tumor angiogenesis and vascular normalization: Alternative therapeutic targets. Angiogenesis 2017, 20, 409–426. [Google Scholar] [CrossRef] [PubMed]
  48. Apte, R.S.; Chen, D.S.; Ferrara, N. VEGF in Signaling and Disease: Beyond Discovery and Development. Cell 2019, 176, 1248–1264. [Google Scholar] [CrossRef] [PubMed]
  49. Lee, H.W.; Xu, Y.; He, L.; Choi, W.; Gonzalez, D.; Jin, S.W.; Simons, M. Role of Venous Endothelial Cells in Developmental and Pathologic Angiogenesis. Circulation 2021, 144, 1308–1322. [Google Scholar] [CrossRef]
  50. Zeng, A.; Wang, S.R.; He, Y.X.; Yan, Y.; Zhang, Y. Progress in understanding of the stalk and tip cells formation involvement in angiogenesis mechanisms. Tissue Cell 2021, 73, 101626. [Google Scholar] [CrossRef]
  51. Strilić, B.; Kucera, T.; Eglinger, J.; Hughes, M.R.; McNagny, K.M.; Tsukita, S.; Dejana, E.; Ferrara, N.; Lammert, E. The molecular basis of vascular lumen formation in the developing mouse aorta. Dev. Cell 2009, 17, 505–515. [Google Scholar] [CrossRef]
  52. Mandel, E.R.; Uchida, C.; Nwadozi, E.; Makki, A.; Haas, T.L. Tissue Inhibitor of Metalloproteinase 1 Influences Vascular Adaptations to Chronic Alterations in Blood Flow. J. Cell Physiol. 2017, 232, 831–841. [Google Scholar] [CrossRef] [PubMed]
  53. Teichert, M.; Milde, L.; Holm, A.; Stanicek, L.; Gengenbacher, N.; Savant, S.; Ruckdeschel, T.; Hasanov, Z.; Srivastava, K.; Hu, J.; et al. Pericyte-expressed Tie2 controls angiogenesis and vessel maturation. Nat. Commun. 2017, 8, 16106. [Google Scholar] [CrossRef] [PubMed]
  54. Ribatti, D.; Tamma, R. Hematopoietic growth factors and tumor angiogenesis. Cancer Lett. 2019, 440–441, 47–53. [Google Scholar] [CrossRef] [PubMed]
  55. Shibuya, M. Vascular endothelial growth factor and its receptor system: Physiological functions in angiogenesis and pathological roles in various diseases. J. Biochem. 2013, 153, 13–19. [Google Scholar] [CrossRef] [PubMed]
  56. Simons, M.; Gordon, E.; Claesson-Welsh, L. Mechanisms and regulation of endothelial VEGF receptor signalling. Nat. Rev. Mol. Cell Biol. 2016, 17, 611–625. [Google Scholar] [CrossRef] [PubMed]
  57. 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]
  58. Andrae, J.; Gallini, R.; Betsholtz, C. Role of platelet-derived growth factors in physiology and medicine. Genes Dev. 2008, 22, 1276–1312. [Google Scholar] [CrossRef] [PubMed]
  59. Presta, M.; Andrés, G.; Leali, D.; Dell’Era, P.; Ronca, R. Inflammatory cells and chemokines sustain FGF2-induced angiogenesis. Eur. Cytokine Netw. 2009, 20, 39–50. [Google Scholar] [CrossRef]
  60. Fousek, K.; Horn, L.A.; Palena, C. Interleukin-8: A chemokine at the intersection of cancer plasticity, angiogenesis, and immune suppression. Pharmacol. Ther. 2021, 219, 107692. [Google Scholar] [CrossRef]
  61. Parmar, D.; Apte, M. Angiopoietin inhibitors: A review on targeting tumor angiogenesis. Eur. J. Pharmacol. 2021, 899, 174021. [Google Scholar] [CrossRef]
  62. Rao, L.; Giannico, D.; Leone, P.; Solimando, A.G.; Maiorano, E.; Caporusso, C.; Duda, L.; Tamma, R.; Mallamaci, R.; Susca, N.; et al. HB-EGF-EGFR Signaling in Bone Marrow Endothelial Cells Mediates Angiogenesis Associated with Multiple Myeloma. Cancers 2020, 12, 173. [Google Scholar] [CrossRef]
  63. Zhang, C.; Zhu, M.; Wang, W.; Chen, D.; Chen, S.; Zheng, H. TNF-α promotes tumor lymph angiogenesis in head and neck squamous cell carcinoma through regulation of ERK3. Transl. Cancer Res. 2019, 8, 2439–2448. [Google Scholar] [CrossRef] [PubMed]
  64. Akwii, R.G.; Sajib, M.S.; Zahra, F.T.; Mikelis, C.M. Role of Angiopoietin-2 in Vascular Physiology and Pathophysiology. Cells 2019, 8, 471. [Google Scholar] [CrossRef] [PubMed]
  65. Morse, M.A.; Sun, W.; Kim, R.; He, A.R.; Abada, P.B.; Mynderse, M.; Finn, R.S. The Role of Angiogenesis in Hepatocellular Carcinoma. Clin. Cancer Res. 2019, 25, 912–920. [Google Scholar] [CrossRef] [PubMed]
  66. Todorova, D.; Simoncini, S.; Lacroix, R.; Sabatier, F.; Dignat-George, F. Extracellular Vesicles in Angiogenesis. Circ. Res. 2017, 120, 1658–1673. [Google Scholar] [CrossRef] [PubMed]
  67. Ma, Q.; Beal, J.R.; Bhurke, A.; Kannan, A.; Yu, J.; Taylor, R.N.; Bagchi, I.C.; Bagchi, M.K. Extracellular vesicles secreted by human uterine stromal cells regulate decidualization, angiogenesis, and trophoblast differentiation. Proc. Natl. Acad. Sci. USA 2022, 119, e2200252119. [Google Scholar] [CrossRef]
  68. Li, Q.; Xu, Y.; Lv, K.; Wang, Y.; Zhong, Z.; Xiao, C.; Zhu, K.; Ni, C.; Wang, K.; Kong, M.; et al. Small extracellular vesicles containing miR-486-5p promote angiogenesis after myocardial infarction in mice and nonhuman primates. Sci. Transl. Med. 2021, 13, 584. [Google Scholar] [CrossRef] [PubMed]
  69. Gregorius, J.; Wang, C.; Stambouli, O.; Hussner, T.; Qi, Y.; Tertel, T.; Börger, V.; Mohamud Yusuf, A.; Hagemann, N.; Yin, D.; et al. Small extracellular vesicles obtained from hypoxic mesenchymal stromal cells have unique characteristics that promote cerebral angiogenesis, brain remodeling and neurological recovery after focal cerebral ischemia in mice. Basic. Res. Cardiol. 2021, 116, 40. [Google Scholar] [CrossRef]
  70. You, B.; Pan, S.; Gu, M.; Zhang, K.; Xia, T.; Zhang, S.; Chen, W.; Xie, H.; Fan, Y.; Yao, H.; et al. Extracellular vesicles rich in HAX1 promote angiogenesis by modulating ITGB6 translation. J. Extracell. Vesicles 2022, 11, e12221. [Google Scholar] [CrossRef]
  71. Angioni, R.; Liboni, C.; Herkenne, S.; Sánchez-Rodríguez, R.; Borile, G.; Marcuzzi, E.; Calì, B.; Muraca, M.; Viola, A. CD73+ extracellular vesicles inhibit angiogenesis through adenosine A(2B) receptor signalling. J. Extracell. Vesicles 2020, 9, 1757900. [Google Scholar] [CrossRef] [PubMed]
  72. Chen, F.; Chen, J.; Yang, L.; Liu, J.; Zhang, X.; Zhang, Y.; Tu, Q.; Yin, D.; Lin, D.; Wong, P.P.; et al. Extracellular vesicle-packaged HIF-1α-stabilizing lncRNA from tumour-associated macrophages regulates aerobic glycolysis of breast cancer cells. Nat. Cell Biol. 2019, 21, 498–510. [Google Scholar] [CrossRef] [PubMed]
  73. Ma, W.; Zhang, W.; Cui, B.; Gao, J.; Liu, Q.; Yao, M.; Ning, H.; Xing, L. Functional delivery of lncRNA TUG1 by endothelial progenitor cells derived extracellular vesicles confers anti-inflammatory macrophage polarization in sepsis via impairing miR-9-5p-targeted SIRT1 inhibition. Cell Death Dis. 2021, 12, 1056. [Google Scholar] [CrossRef] [PubMed]
  74. Romano, R.; Picca, A.; Eusebi, L.H.U.; Marzetti, E.; Calvani, R.; Moro, L.; Bucci, C.; Guerra, F. Extracellular Vesicles and Pancreatic Cancer: Insights on the Roles of miRNA, lncRNA, and Protein Cargos in Cancer Progression. Cells 2021, 10, 1361. [Google Scholar] [CrossRef] [PubMed]
  75. Lai, H.; Li, Y.; Zhang, H.; Hu, J.; Liao, J.; Su, Y.; Li, Q.; Chen, B.; Li, C.; Wang, Z.; et al. exoRBase 2.0: An atlas of mRNA, lncRNA and circRNA in extracellular vesicles from human biofluids. Nucleic Acids Res. 2022, 50, D118–D128. [Google Scholar] [CrossRef] [PubMed]
  76. Casado-Díaz, A.; Quesada-Gómez, J.M.; Dorado, G. Extracellular Vesicles Derived From Mesenchymal Stem Cells (MSC) in Regenerative Medicine: Applications in Skin Wound Healing. Front. Bioeng. Biotechnol. 2020, 8, 146. [Google Scholar] [CrossRef]
  77. Wu, Y.; Wu, X.; Wang, J.; Chen, S.; Chen, H.; Liu, J.; Zeng, T.; Hu, M.; Liang, Y.; Sun, K.; et al. Fibroblast-Derived Extracellular Vesicle-Packaged Long Noncoding RNA Upregulated in Diabetic Skin Enhances Keratinocyte MMP-9 Expression and Delays Diabetic Wound Healing. Lab. Investig. 2023, 103, 100019. [Google Scholar] [CrossRef]
  78. Yang, Z.; Liu, X.; Zhao, F.; Yao, M.; Lin, Z.; Yang, Z.; Liu, C.; Liu, Y.; Chen, X.; Du, C. Bioactive glass nanoparticles inhibit osteoclast differentiation and osteoporotic bone loss by activating lncRNA NRON expression in the extracellular vesicles derived from bone marrow mesenchymal stem cells. Biomaterials 2022, 283, 121438. [Google Scholar] [CrossRef]
  79. Al-Ostoot, F.H.; Salah, S.; Khamees, H.A.; Khanum, S.A. Tumor angiogenesis: Current challenges and therapeutic opportunities. Cancer Treat. Res. Commun. 2021, 28, 100422. [Google Scholar] [CrossRef] [PubMed]
  80. Jiang, X.; Wang, J.; Deng, X.; Xiong, F.; Zhang, S.; Gong, Z.; Li, X.; Cao, K.; Deng, H.; He, Y.; et al. The role of microenvironment in tumor angiogenesis. J. Exp. Clin. Cancer Res. 2020, 39, 204. [Google Scholar] [CrossRef] [PubMed]
  81. Zhuang, Y.; Liu, K.; He, Q.; Gu, X.; Jiang, C.; Wu, J. Hypoxia signaling in cancer: Implications for therapeutic interventions. MedComm 2023, 4, e203. [Google Scholar] [CrossRef]
  82. Chen, L.; Shan, G. CircRNA in cancer: Fundamental mechanism and clinical potential. Cancer Lett. 2021, 505, 49–57. [Google Scholar] [CrossRef]
  83. Jiang, S.; Fu, R.; Shi, J.; Wu, H.; Mai, J.; Hua, X.; Chen, H.; Liu, J.; Lu, M.; Li, N. CircRNA-Mediated Regulation of Angiogenesis: A New Chapter in Cancer Biology. Front. Oncol. 2021, 11, 553706. [Google Scholar] [CrossRef] [PubMed]
  84. Zhong, Z.; Huang, M.; Lv, M.; He, Y.; Duan, C.; Zhang, L.; Chen, J. Circular RNA MYLK as a competing endogenous RNA promotes bladder cancer progression through modulating VEGFA/VEGFR2 signaling pathway. Cancer Lett. 2017, 403, 305–317. [Google Scholar] [CrossRef] [PubMed]
  85. 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] [PubMed]
  86. Wang, B.D.; Yu, X.J.; Hou, J.C.; Fu, B.; Zheng, H.; Liu, Q.K.; Wang, S.X.; Bi, Z.G.; Cao, Y. Bevacizumab attenuates osteosarcoma angiogenesis by suppressing MIAT encapsulated by serum-derived extracellular vesicles and facilitating miR-613-mediated GPR158 inhibition. Cell Death Dis. 2022, 13, 272. [Google Scholar] [CrossRef] [PubMed]
  87. Feng, D.; Li, Z.; Yang, L.; Liang, H.; He, H.; Liu, L.; Zhang, W. BMSC-EV-derived lncRNA NORAD Facilitates Migration, Invasion, and Angiogenesis in Osteosarcoma Cells by Regulating CREBBP via Delivery of miR-877-3p. Oxid. Med. Cell Longev. 2022, 2022, 8825784. [Google Scholar] [CrossRef] [PubMed]
  88. Tao, S.C.; Huang, J.Y.; Wei, Z.Y.; Li, Z.X.; Guo, S.C. EWSAT1 Acts in Concert with Exosomes in Osteosarcoma Progression and Tumor-Induced Angiogenesis: The “Double Stacking Effect”. Adv. Biosyst. 2020, 4, e2000152. [Google Scholar] [CrossRef] [PubMed]
  89. 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]
  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. Zhang, S.; Cai, J.; Ji, Y.; Zhou, S.; Miao, M.; Zhu, R.; Li, K.; Xue, Z.; Hu, S. Tumor-derived exosomal lincRNA ROR promotes angiogenesis in nasopharyngeal carcinoma. Mol. Cell Probes 2022, 66, 101868. [Google Scholar] [CrossRef]
  92. Zhou, S.K.; Gao, F.; Zhong, Z.S.; Yao, H. Long non-coding RNA colon cancer associated transcript-2 from nasopharyngeal carcinoma-derived exosomes promotes angiogenesis. Zhonghua Er Bi Yan Hou Tou Jing Wai Ke Za Zhi 2020, 55, 944–951. [Google Scholar] [CrossRef] [PubMed]
  93. 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]
  94. 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] [PubMed]
  95. 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] [PubMed]
  96. Ma, X.; Li, Z.; Li, T.; Zhu, L.; Li, Z.; Tian, N. Long non-coding RNA HOTAIR enhances angiogenesis by induction of VEGFA expression in glioma cells and transmission to endothelial cells via glioma cell derived-extracellular vesicles. Am. J. Transl. Res. 2017, 9, 5012–5021. [Google Scholar] [PubMed]
  97. Dai, X.; Liao, K.; Zhuang, Z.; Chen, B.; Zhou, Z.; Zhou, S.; Lin, G.; Zhang, F.; Lin, Y.; Miao, Y.; et al. AHIF promotes glioblastoma progression and radioresistance via exosomes. Int. J. Oncol. 2019, 54, 261–270. [Google Scholar] [CrossRef] [PubMed]
  98. Zeng, W.; Liu, Y.; Li, W.T.; Li, Y.; Zhu, J.F. CircFNDC3B sequestrates miR-937-5p to derepress TIMP3 and inhibit colorectal cancer progression. Mol. Oncol. 2020, 14, 2960–2984. [Google Scholar] [CrossRef] [PubMed]
  99. Wang, F.W.; Cao, C.H.; Han, K.; Zhao, Y.X.; Cai, M.Y.; Xiang, Z.C.; Zhang, J.X.; Chen, J.W.; Zhong, L.P.; Huang, Y.; et al. APC-activated long noncoding RNA inhibits colorectal carcinoma pathogenesis through reduction of exosome production. J. Clin. Investig. 2019, 129, 727–743. [Google Scholar] [CrossRef]
  100. Chen, C.; Liu, Y.; Liu, L.; Si, C.; Xu, Y.; Wu, X.; Wang, C.; Sun, Z.; Kang, Q. Exosomal circTUBGCP4 promotes vascular endothelial cell tipping and colorectal cancer metastasis by activating Akt signaling pathway. J. Exp. Clin. Cancer Res. 2023, 42, 46. [Google Scholar] [CrossRef]
  101. 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]
  102. Lin, J.; Cao, S.; Wang, Y.; Hu, Y.; Liu, H.; Li, J.; Chen, J.; Li, P.; Liu, J.; Wang, Q.; et al. Long non-coding RNA UBE2CP3 enhances HCC cell secretion of VEGFA and promotes angiogenesis by activating ERK1/2/HIF-1α/VEGFA signalling in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2018, 37, 113. [Google Scholar] [CrossRef]
  103. Conigliaro, A.; Costa, V.; Lo Dico, A.; 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] [PubMed]
  104. 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] [PubMed]
  105. Xu, J.; Wang, H.; Shi, B.; Li, N.; Xu, G.; Yan, X.; Xu, L. Exosomal MFI2-AS1 sponge miR-107 promotes non-small cell lung cancer progression through NFAT5. Cancer Cell Int. 2023, 23, 51. [Google Scholar] [CrossRef] [PubMed]
  106. 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]
  107. 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, 2476175. [Google Scholar] [CrossRef] [PubMed]
  108. Zhang, W.; Zheng, X.; Yu, Y.; Zheng, L.; Lan, J.; Wu, Y.; Liu, H.; Zhao, A.; Huang, H.; Chen, W. Renal cell carcinoma-derived exosomes deliver lncARSR to induce macrophage polarization and promote tumor progression via STAT3 pathway. Int. J. Biol. Sci. 2022, 18, 3209–3222. [Google Scholar] [CrossRef]
  109. Huang, X.; Wang, J.; Guan, J.; Zheng, Z.; Hao, J.; Sheng, Z.; Wang, M.; Xu, T.; Guo, G.; Yao, L. Exosomal Circsafb2 Reshaping Tumor Environment to Promote Renal Cell Carcinoma Progression by Mediating M2 Macrophage Polarization. Front. Oncol. 2022, 12, 808888. [Google Scholar] [CrossRef]
  110. Shu, G.; Lu, X.; Pan, Y.; Cen, J.; Huang, K.; Zhou, M.; Lu, J.; Dong, J.; Han, H.; Chen, W.; et al. Exosomal circSPIRE1 mediates glycosylation of E-cadherin to suppress metastasis of renal cell carcinoma. Oncogene 2023, 42, 1802–1820. [Google Scholar] [CrossRef]
  111. Zheng, H.; Chen, C.; Luo, Y.; Yu, M.; He, W.; An, M.; Gao, B.; Kong, Y.; Ya, Y.; Lin, Y.; et al. Tumor-derived exosomal BCYRN1 activates WNT5A/VEGF-C/VEGFR3 feedforward loop to drive lymphatic metastasis of bladder cancer. Clin. Transl. Med. 2021, 11, e497. [Google Scholar] [CrossRef] [PubMed]
  112. Chen, C.; Luo, Y.; He, W.; Zhao, Y.; Kong, Y.; Liu, H.; Zhong, G.; Li, Y.; Li, J.; Huang, J.; et al. Exosomal long noncoding RNA LNMAT2 promotes lymphatic metastasis in bladder cancer. J. Clin. Investig. 2020, 130, 404–421. [Google Scholar] [CrossRef]
  113. Chen, C.; Zheng, H.; Luo, Y.; Kong, Y.; An, M.; Li, Y.; He, W.; Gao, B.; Zhao, Y.; Huang, H.; et al. SUMOylation promotes extracellular vesicle-mediated transmission of lncRNA ELNAT1 and lymph node metastasis in bladder cancer. J. Clin. Investig. 2021, 131, 8. [Google Scholar] [CrossRef] [PubMed]
  114. Liu, C.; Yang, J.; Zhu, F.; Zhao, Z.; Gao, L. Exosomal circ_0001190 Regulates the Progression of Gastric Cancer via miR-586/SOSTDC1 Axis. Biochem. Genet. 2022, 60, 1895–1913. [Google Scholar] [CrossRef] [PubMed]
  115. Li, S.; Li, J.; Zhang, H.; Zhang, Y.; Wang, X.; Yang, H.; Zhou, Z.; Hao, X.; Ying, G.; Ba, Y. Gastric cancer derived exosomes mediate the delivery of circRNA to promote angiogenesis by targeting miR-29a/VEGF axis in endothelial cells. Biochem. Biophys. Res. Commun. 2021, 560, 37–44. [Google Scholar] [CrossRef]
  116. 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, 112. [Google Scholar] [CrossRef]
  117. Zhang, Z.; Sun, C.; Zheng, Y.; Gong, Y. circFCHO2 promotes gastric cancer progression by activating the JAK1/STAT3 pathway via sponging miR-194-5p. Cell Cycle 2022, 21, 2145–2164. [Google Scholar] [CrossRef]
  118. 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]
  119. 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]
  120. Shi, P.; Liu, Y.; Yang, H.; Hu, B. Breast cancer derived exosomes promoted angiogenesis of endothelial cells in microenvironment via circHIPK3/miR-124-3p/MTDH axis. Cell Signal 2022, 95, 110338. [Google Scholar] [CrossRef]
  121. Liu, B.; Chen, J.; Shang, F.; Lian, M.; Shen, X.; Fang, J. Tumor-Derived Exosome FGD5-AS1 Promotes Angiogenesis, Vascular Permeability, and Metastasis in Thyroid Cancer by Targeting the miR-6838-5p/VAV2 Axis. J. Oncol. 2022, 2022, 4702855. [Google Scholar] [CrossRef] [PubMed]
  122. Yu, M.; Yu, J.; Zhang, Y.; Sun, X.; Sun, R.; Xia, M.; Li, S.; Cui, X. A novel circRNA-miRNA-mRNA network revealed exosomal circ-ATP10A as a biomarker for multiple myeloma angiogenesis. Bioengineered 2022, 13, 667–683. [Google Scholar] [CrossRef]
  123. Lamichhane, T.N.; Leung, C.A.; Douti, L.Y.; Jay, S.M. Ethanol Induces Enhanced Vascularization Bioactivity of Endothelial Cell-Derived Extracellular Vesicles via Regulation of MicroRNAs and Long Non-Coding RNAs. Sci. Rep. 2017, 7, 13794. [Google Scholar] [CrossRef]
  124. Xu, Y.; Leng, K.; Yao, Y.; Kang, P.; Liao, G.; Han, Y.; Shi, G.; Ji, D.; Huang, P.; Zheng, W.; et al. A Circular RNA, Cholangiocarcinoma-Associated Circular RNA 1, Contributes to Cholangiocarcinoma Progression, Induces Angiogenesis, and Disrupts Vascular Endothelial Barriers. Hepatology 2021, 73, 1419–1435. [Google Scholar] [CrossRef] [PubMed]
  125. Zhi, J.; Jia, X.J.; Yan, J.; Wang, H.C.; Feng, B.; Xing, H.Y.; Jia, Y.T. BRAF(V600E) mutant colorectal cancer cells mediate local immunosuppressive microenvironment through exosomal long noncoding RNAs. World J. Gastrointest. Oncol. 2021, 13, 2129–2148. [Google Scholar] [CrossRef] [PubMed]
  126. 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]
  127. Yuan, D.; Guo, T.; Zhu, D.; Ge, H.; Zhao, Y.; Huang, A.; Wang, X.; Cao, X.; He, C.; Qian, H.; et al. Exosomal lncRNA ATB Derived from Ovarian Cancer Cells Promotes Angiogenesis via Regulating miR-204-3p/TGFβR2 Axis. Cancer Manag. Res. 2022, 14, 327–337. [Google Scholar] [CrossRef]
  128. Ghafouri-Fard, S.; Shirvani-Farsani, Z.; Hussen, B.M.; Taheri, M. The critical roles of lncRNAs in the development of osteosarcoma. Biomed. Pharmacother. 2021, 135, 111217. [Google Scholar] [CrossRef]
  129. Monteiro, L.J.; Peñailillo, R.; Sánchez, M.; Acuña-Gallardo, S.; Mönckeberg, M.; Ong, J.; Choolani, M.; Illanes, S.E.; Nardocci, G. The Role of Long Non-Coding RNAs in Trophoblast Regulation in Preeclampsia and Intrauterine Growth Restriction. Genes 2021, 12, 970. [Google Scholar] [CrossRef]
  130. Sun, W.; Shen, N.M.; Fu, S.L. Involvement of lncRNA-mediated signaling pathway in the development of cervical cancer. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 3672–3687. [Google Scholar] [CrossRef]
  131. Kok, V.C.; Yu, C.C. Cancer-Derived Exosomes: Their Role in Cancer Biology and Biomarker Development. Int. J. Nanomed. 2020, 15, 8019–8036. [Google Scholar] [CrossRef] [PubMed]
  132. Liu, R.; Li, X.; Zhu, W.; Wang, Y.; Zhao, D.; Wang, X.; Gurley, E.C.; Liang, G.; Chen, W.; Lai, G.; et al. Cholangiocyte-Derived Exosomal Long Noncoding RNA H19 Promotes Hepatic Stellate Cell Activation and Cholestatic Liver Fibrosis. Hepatology 2019, 70, 1317–1335. [Google Scholar] [CrossRef] [PubMed]
  133. Movahedpour, A.; Khatami, S.H.; Karami, N.; Vakili, O.; Naeli, P.; Jamali, Z.; Shabaninejad, Z.; Tazik, K.; Behrouj, H.; Ghasemi, H. Exosomal noncoding RNAs in prostate cancer. Clin. Chim. Acta 2022, 537, 127–132. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Exosome biogenesis, release to the extracellular environment, and uptake by the recipient cell. (A) Exosome biogenesis begins with early endosome formation during endocytosis. (B) Early endosomes are then matured into late endosomes, generating multiple intraluminal vesicles (ILVs) by the inward budding of endosomal membranes. (C) The accumulation of ILVs leads to the formation of multivesicular endosomes (MVEs), and proteins and nucleic acids produced by the donor cell can be sorted into exosomes during MVE formation. (D) Exosomes are released into the extracellular environment by fusing MVEs with the cellular membrane. (EG) Microvesicles arise from the outward budding and shedding of the plasma membrane. (H) Extracellular vesicles are taken up by the recipient cell by direct fusion, receptor-mediated fusion, or endocytosis. (I) Exosomal lncRNAs can be subsequently delivered to the recipient cell to exert regulatory effects as sponges for miRNAs, protein scaffolds, transcription and translation regulators, and chromatin activators. The detailed functions of lncRNAs are depicted in Figure 2. Figure 1 is modified from Wang et al. [23].
Figure 1. Exosome biogenesis, release to the extracellular environment, and uptake by the recipient cell. (A) Exosome biogenesis begins with early endosome formation during endocytosis. (B) Early endosomes are then matured into late endosomes, generating multiple intraluminal vesicles (ILVs) by the inward budding of endosomal membranes. (C) The accumulation of ILVs leads to the formation of multivesicular endosomes (MVEs), and proteins and nucleic acids produced by the donor cell can be sorted into exosomes during MVE formation. (D) Exosomes are released into the extracellular environment by fusing MVEs with the cellular membrane. (EG) Microvesicles arise from the outward budding and shedding of the plasma membrane. (H) Extracellular vesicles are taken up by the recipient cell by direct fusion, receptor-mediated fusion, or endocytosis. (I) Exosomal lncRNAs can be subsequently delivered to the recipient cell to exert regulatory effects as sponges for miRNAs, protein scaffolds, transcription and translation regulators, and chromatin activators. The detailed functions of lncRNAs are depicted in Figure 2. Figure 1 is modified from Wang et al. [23].
Ncrna 10 00012 g001
Figure 3. Stages of the angiogenic process. (A) Angiogenic signals, such as VEGF, promote pericyte detachment from the basement membrane and weaken the extracellular matrix. (B) Endothelial cells display characteristic phenotypes after VEGF stimulation: migratory tip cells or proliferating stalk cells. (C) Attractive and repulsive forces control endothelial cells, forming a vessel lumen to initiate blood flow. (D) PDGF-B and Ang1 signaling lead to pericyte recruitment, while junctional molecules consolidate EC–EC adhesion. Modified from Viallard et al. [47].
Figure 3. Stages of the angiogenic process. (A) Angiogenic signals, such as VEGF, promote pericyte detachment from the basement membrane and weaken the extracellular matrix. (B) Endothelial cells display characteristic phenotypes after VEGF stimulation: migratory tip cells or proliferating stalk cells. (C) Attractive and repulsive forces control endothelial cells, forming a vessel lumen to initiate blood flow. (D) PDGF-B and Ang1 signaling lead to pericyte recruitment, while junctional molecules consolidate EC–EC adhesion. Modified from Viallard et al. [47].
Ncrna 10 00012 g003
Figure 4. Molecular landscape of exosomal lnc- and circRNAs in angiogenesis in cancer.
Figure 4. Molecular landscape of exosomal lnc- and circRNAs in angiogenesis in cancer.
Ncrna 10 00012 g004
Table 1. The landscape of exosomal lnc- and circRNAs in angiogenesis in cancer.
Table 1. The landscape of exosomal lnc- and circRNAs in angiogenesis in cancer.
Lnc/circRNAMolecular TargetDonor CellsRecipient CellsEffectReference
OSTEOSARCOMA
OIP5-AS1 miR-153/ATG5HOSHUVECsPromotesLi 2021 [85]
MIAT miR-613/GPR158U2OS, MG63, and 293THUVECsPromotesWang 2022 [86]
NORAD miR-877-3p/CREBBP143B, MG-63, Saos2, HOS, and U20SOsteosarcoma cellsPromotesFeng 2022 [87]
EWSAT1 miR-326/KRas143B, MNNG/HOS, MG63, U20SBMSCs, HMEC-1PromotesTao 2020 [88]
CHONDROSARCOMA
RAMP2-AS1miR-2355-5p/VEGFR2SW1353HUVECsPromotesCheng 2020 [89]
PANCREATIC
UCA1 miR-96-5p/AMOTL2PANC-1, MIA PaCa-2, BxPC-3, Aspc-1, Sw1990HUVECs, HEK293TPromotesGuo 2020 [90]
NASOPHARYNGEAL
Linc-ROR p-AKT/p-VEGFR2 pathwayCNE2HUVECsPromotesZhang 2022 [91]
CCAT2NRCNE2, NP69HUVECsPromotesZhou 2020 [92]
ESOPHAGEAL
FAM225A miR-206/NETO2 and FOXP1 ECA109, TE-1, KYSE150, KYSE140HET-1A, HUVECsPromotesZhang 2020 [93]
GLIOMA/GLIOBLASTOMA
CCAT2 VEGF-A and TGF-BA172, U87-MG, U251, T98GHUVECsPromotesLang 2017 [94]
POU3F3 bFGF/bFGFR/VEGFAA172, U87-MG, U251, T98GHBMECsPromotesLang 2017 [95]
HOTAIR VEGFAA172HBMVECsPromotesMa 2017 [96]
aHIFNR *U87-MG, U251-MG, A172, T98GHUVECsPromotesDai 2019 [97]
COLORECTAL
CircFNDC3B miR-97-5p/TIMP3 LoVo, SW480, SW602, HCT116HUVECsSuppressesZeng 2020 [98]
APC1 MAPK pathwayHTC116, DLD-1, SW480, LoVo, SW116HEK293T, HUVECsPromotesWang 2019 [99]
CircTUBGCP4 miR-146b-3p/PDK2/Akt SW480HEK297TPromotesChen 2023 [100]
LIVER/HEPATOCELLULAR
LINC00161 miR-590-3p/ROCK2 axisHuh-7, HCCLM3, MHCC-97L, MHCC-97HWRL-68, HUVECsPromotesYou 2021 [101]
UBE2CP3 ERK1/2/HIF-1α/VEGFAHepG2, SMMC-7721HUVECsPromotesLin 2018 [102]
H19NR *Huh-7, Sk-HepHUVECsPromotesConigliaro 2015 [103]
Circ100338 VE-CadherinHep3B, HLE, Huh-7, BEL7402, SMCC7721, MHCC97L, HCCLM3, MHCC97H, HCCLM6HUVECsPromotesHuang 2020 [104]
LUNG
MFI2-AS1 miR-107/PI3K/AKT pathwayPC9, A549, H1299HUVECsPromotesXu 2023 [105]
LincRNA-p21 miR-23a, miR-146b, miR-330, and miR-494H23, HCC44HUVECsPromotesCastellano 2020 [106]
GAS5 miR-29-3p/PI3K/Akt16HBE, A549, H1299, 95DHUVECsPromotesCheng 2019 [107]
RENAL CELL
ARSR miR-34 and miR-449 to upregulate STAT3 pathwayCaki-1, ACHN, 786-ONR *PromotesZheng 2022 [108]
CircSAFB2 miR-620/JAK1/STAT3 axisA498, 786-O, Caki-1, Caki-2, 769-P, ACHNTHP-1PromotesHuang 2022 [109]
CircSPIRE1 ELAVL1 proteinNR *NR *SuppressesShu 2023 [110]
BLADDER
BCYRN1 WNT5a/VEGF-C/VEGFR3T24, 5637, SVHUC-1HLECs, HDLECs, HUVECsPromotesZheng 2021 [111]
LNMAT2 PROX1/RNPA2B1/H3K4UM-UC-3, 5637, T24HLEC, SV-HUC-1PromotesChen 2020 [112]
ELNAT1 SOX18 UM-UC-1, RT112, RT4, UM-UC-3, T24, 5637HLEC, SV-HUC-1PromotesChen 2021 [113]
GASTRIC
Circ0001190 miR-587/SOSTDC1NR *NR *SuppressesLiu 2022 [114]
Circ29 miR-29a/VEGF pathwaySGC-7901, MGC-803HUVECs, HEK297TSuppressesLi 2021 [115]
CircSHKBP1 miR-582/HUR/VEGFAGS, HGC27, BGC823 MGC803, GES1HUVECs, HEK293TPromotesXie 2020 [116]
CircFCHO2 miR-194-5p/JAK1/STAT3 pathwayNR *NR *PromotesZhang 2022 [117]
OVARIAN
MALAT1 NR *SKOV3, HO8910SKOV3.ip1, HO8910.PMPromotesQiu 2018 [118]
CERVICAL
TUG1VEGF-A, MMP-9, IL-8HeLa, CaSkiHUVECsSuppressesLei 2020 [119]
BREAST
CircHIPK3 miR-124-3p/MTDHNR *NR *PromotesShi 2022 [120]
THYROID
FGD5-AS1 miR-6838-5p/VAV2 axisSW1736, KAT18HUVECsPromotesLiu 2022 [121]
MULTIPLE MIELOMA
CircATP10AmiR-66758-3p, miR-3977, miR-6804-3p, miR-1266-3p, miR-3620-3pNR *NR *PromotesYu 2022 [122]
ALCOHOL-INDUCED TUMOR
HOTAIR and MALAT1 NR *NR *HUVECs, HDMECsPromotesLamichhane 2017 [123]
CHOLANGIOCARCINOMA
CircCCAC1EZH2/SH3GL2CCA cellsHUVECsPromotesXu 2021 [124]
* NR: not reported.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Peña-Flores, J.A.; Muela-Campos, D.; Guzmán-Medrano, R.; Enríquez-Espinoza, D.; González-Alvarado, K. Functional Relevance of Extracellular Vesicle-Derived Long Non-Coding and Circular RNAs in Cancer Angiogenesis. Non-Coding RNA 2024, 10, 12. https://doi.org/10.3390/ncrna10010012

AMA Style

Peña-Flores JA, Muela-Campos D, Guzmán-Medrano R, Enríquez-Espinoza D, González-Alvarado K. Functional Relevance of Extracellular Vesicle-Derived Long Non-Coding and Circular RNAs in Cancer Angiogenesis. Non-Coding RNA. 2024; 10(1):12. https://doi.org/10.3390/ncrna10010012

Chicago/Turabian Style

Peña-Flores, José A., Daniela Muela-Campos, Rebeca Guzmán-Medrano, Diego Enríquez-Espinoza, and Karla González-Alvarado. 2024. "Functional Relevance of Extracellular Vesicle-Derived Long Non-Coding and Circular RNAs in Cancer Angiogenesis" Non-Coding RNA 10, no. 1: 12. https://doi.org/10.3390/ncrna10010012

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