Your browser does not fully support modern features. Please upgrade for a smoother experience.
Submitted Successfully!
Thank you for your contribution! You can also upload a video entry or images related to this topic. For video creation, please contact our Academic Video Service.
Version Summary Created by Modification Content Size Created at Operation
1 Zhaowu Ma -- 2819 2022-04-10 16:36:29 |
2 Format Change Jessie Wu -43 word(s) 2776 2022-04-11 03:06:45 | |
3 Format Change Jessie Wu Meta information modification 2776 2022-04-11 03:07:52 | |
4 Format Change Jessie Wu Meta information modification 2776 2022-04-11 08:47:51 | |
5 Format Change Jessie Wu Meta information modification 2776 2022-04-11 08:49:37 |

Video Upload Options

We provide professional Academic Video Service to translate complex research into visually appealing presentations. Would you like to try it?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Ma, Z.; Wang, L.; , .; Liu, C. Noncoding RNAs of Extracellular Vesicles in Tumor Angiogenesis. Encyclopedia. Available online: https://encyclopedia.pub/entry/21547 (accessed on 14 January 2026).
Ma Z, Wang L,  , Liu C. Noncoding RNAs of Extracellular Vesicles in Tumor Angiogenesis. Encyclopedia. Available at: https://encyclopedia.pub/entry/21547. Accessed January 14, 2026.
Ma, Zhaowu, Lingzhi Wang,  , Chen-Guang Liu. "Noncoding RNAs of Extracellular Vesicles in Tumor Angiogenesis" Encyclopedia, https://encyclopedia.pub/entry/21547 (accessed January 14, 2026).
Ma, Z., Wang, L., , ., & Liu, C. (2022, April 10). Noncoding RNAs of Extracellular Vesicles in Tumor Angiogenesis. In Encyclopedia. https://encyclopedia.pub/entry/21547
Ma, Zhaowu, et al. "Noncoding RNAs of Extracellular Vesicles in Tumor Angiogenesis." Encyclopedia. Web. 10 April, 2022.
Noncoding RNAs of Extracellular Vesicles in Tumor Angiogenesis
Edit

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. 

noncoding RNAs extracellular vesicles tumor angiogenesis

1. Characteristics of Extracellular Vesicles and ncRNAs

Extracellular vesicles (EVs) are extracellular structures enclosed in a lipid bilayer that can be secreted by almost all known cell types [1]. 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 [2]. On the contrary, MVs are generated by regulated outward budding of the plasma membrane [3]. The mechanisms of exosomal biogenesis involve multiple factors, and the most well-known regulator is endosomal sorting complex required for transport (ESCRT) [3]. 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 [4][5]. Several studies have found that exosomes can be formed despite the depletion of the ESCRT complex, which reveals an ESCRT-independent approach [6]. ESCRT-independent exosomal biogenesis is regulated by sphingolipid ceramide, which is produced from the hydrolysis of sphingomyelin by neutral sphingomyelinase 2 [2]. The contents of EVs include various nucleic acids, lipids, and proteins [7]. ncRNAs carried by EVs can regulate various physiological and pathological processes through multiple mechanisms (Figure 1).
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.
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 [8]. 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 [9][10][11]. Recent studies have reported that miRNAs are selectively sorted into EVs and participate in intercellular communication in the TME [12]. 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 [13]. 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 [14][15][16].
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 [17]. 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 [18][19][20][21]. In addition, lncRNAs can also be selectively sorted into EVs and participate in cell-to-cell communication in the TME [22]. circRNAs, generated by a particular form of alternative splicing called back-splicing, regulate gene transcription and translation by interacting with DNAs, RNAs, and proteins [23]. Emerging studies have indicated that circRNAs participate in multiple physiological and pathological processes, including tumor angiogenesis [24][25][26].

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

Angiogenesis is a multi-step process and has two types: sprouting and intussusceptive angiogenesis [27]. 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 [28][29][30]. Because angiogenesis is crucial for tumor growth and metastasis, targeting tumor-associated angiogenesis is a promising strategy for cancer treatment [31][32][33]. Currently, anti-angiogenic therapies targeting VEGF and VEGFR are used for the treatment of various tumors [34].
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 [35]. The exosomal lncRNA FAM225A upregulates NETO2 and FOXP1 expression by sponging miR-206 to accelerate oesophageal squamous cell carcinoma (ESCC) progression and angiogenesis [36]. In addition, circSHKBP1 sponges miR-582-3p to enhance HUR expression and VEGF mRNA stability, which promotes angiogenesis and gastric tumor progression [37]. 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.
Table 1. The emerging roles of EV-derived ncRNAs in tumor angiogenesis.
EV-Derived ncRNAs Expression Source Cell Function and Mechanism Tumor Type Reference

miR-155

Upregulated

Tumor cell

Promotes angiogenesis via the c-MYB/VEGF axis

Gastric cancer

[38]

Upregulated

Tumor cell

Promotes angiogenesis by inhibiting FOXO3a

Gastric cancer

[39]

miR-130a

Upregulated

Tumor cell

Activates angiogenesis by inhibiting c-MYB

Gastric cancer

[40]

miR-135b

Upregulated

Tumor cell

Promotes angiogenesis by inhibiting FOXO1

Gastric cancer

[41]

Upregulated

Tumor cell

Regulates the HIF/FIH signalling pathway

Multiple myeloma

[42]

miR-23a

Upregulated

Tumor cell

Inhibits PTEN and activates the AKT pathway

Gastric cancer

[43]

Upregulated

Tumor cell

Increases angiogenesis by inhibiting ZO-1

Lung cancer

[44]

miR-200b-3p

Downregulated

Tumor cell

Enhances endothelial ERG expression

Hepatocellular carcinoma

[45]

miR-25-3p

Upregulated

Tumor cell

Inhibits KLF2 and KLF4, thereby elevating VEGFR2 expression

Colorectal cancer

[46]

miR-1229

Upregulated

Tumor cell

Inhibits HIPK2, thereby activating the VEGF pathway

Colorectal cancer

[47]

miR-183-5p

Upregulated

Tumor cell

Inhibits FOXO1, thereby promoting expression of VEGFA, VEGFAR2, ANG2, PIGF, MMP-2, and MMP-9

Colorectal cancer

[48]

miR-142-3p

Upregulated

Tumor cell

Inhibits TGFβR1

Lung adenocarcinoma

[49]

miR-103a

Upregulated

Tumor cell

Inhibits PTEN, thereby promoting the polarization of M2 macrophages

Lung cancer

[50]

miR-126

Upregulated

MSCs

Upregulates CD34 and CXCR4, thereby promoting expression of VEGF

Lung cancer

[51]

miR-141-3p

Upregulated

Tumor cell

Inhibits SOCS5, thereby activating JAK/STAT3 and NF-κB signalling pathways

Ovarian cancer

[52]

miR-205

Upregulated

Tumor cell

Regulates the PTEN/AKT pathway

Ovarian cancer

[53]

miR-9

Downregulated

Tumor cell

Inhibits MDK, thereby regulating the PDK/AKT signalling pathway

Nasopharyngeal carcinoma

[54]

Upregulated

Tumor cell

Promotes angiogenesis by targeting COL18A1, THBS2, PTCH1, and PHD3

Glioma

[55]

miR-23a

Upregulated

Tumor cell

Promotes angiogenesis by inhibiting TSGA10

Nasopharyngeal carcinoma

[56]

miR-210

Upregulated

Tumor cell

Enhances tube formation by inhibiting EFNA3

Leukemia

[57]

Upregulated

Tumor cell

Promotes angiogenesis by inhibiting SMAD4 and STAT6

Hepatocellular carcinoma

[58]

miR-26a

Upregulated

Tumor cell

Inhibits PTEN, thereby activating the PI3K/AKT signalling pathway

Glioma

[59]

miR-27a

Upregulated

Tumor cell

Inhibits BTG2, thereby promoting VEGF, VEGFR, MMP-2, and MMP-9 expression

Pancreatic cancer

[60]

miR-155-5p

/miR-221-5p

Upregulated

M2 macrophages

Promotes angiogenesis by targeting E2F2

Pancreatic cancer

[61]

miR-21-5p

Upregulated

Tumor cell

Promotes angiogenesis by targeting TGFBI and COL4A1

Papillary carcinoma

[62]

miR-100

- -

MSCs

Regulates the mTOR/HIF-1α signalling axis

Breast cancer

[63]

miR-21

Upregulated

Tumor cell

Inhibits SPRY1, thereby promoting VEGF expression

Oesophageal squamous cell carcinoma

[64]

Upregulated

Tumor cell

Inhibits PTEN, thereby activating PDK1/AKT signalling

Hepatocellular carcinoma

[65]

miR-181b-5p

Upregulated

Tumor cell

Inhibits PTEN and PHLPP2, thereby activating AKT signalling

Oesophageal squamous cell carcinoma

[66]

miR-9

Upregulated

Tumor cell

Inhibits S1P, thereby promoting VEGF expression

Medulloblastoma and xenoglioblastoma

[67]

miR-10a-5p

Upregulated

CAFs

Inhibits TBX5, thereby activating Hedgehog signalling

Cervical squamous cell carcinoma

[68]

miR-135b

Upregulated

Tumor cell

Enhances angiogenesis by targeting FIH-1

Multiple myeloma

[42]

miR-130b-3p

Upregulated

M2 macrophages

Regulates the miR-130b-3p/MLL3/GRHL2 signalling cascade

Gastric cancer

[69]

lncGAS5

Downregulated

Tumor cell

Inhibits angiogenesis by regulating the miR-29-3p/PTEN axis

Lung cancer

[70]

lnc-CCAT2

Upregulated

Tumor cell

Promotes VEGFA and TGF-β expression

Glioma

[71]

lnc-POU3F3

Upregulated

Tumor cell

Promotes bFGF, bFGFR, and VEGFA expression

Glioma

[72]

lncRNA RAMP2-AS1

Upregulated

Tumor cell

Promotes angiogenesis through the miR-2355-5p/VEGFR2 axis

Chondrosarcoma

[73]

OIP5-AS1

Upregulated

Tumor cell

Regulates angiogenesis and autophagy through miR-153/ATG5 axis

Osteosarcoma

[74]

FAM225A

Upregulated

Tumor cell

Promotes angiogenesis through the miR-206/NETO2/FOXP1 axis

Oesophageal squamous cell carcinoma

[36]

UCA1

Upregulated

Tumor cell

Promotes angiogenesis through the miR-96-5p/AMOTL2 axis

Pancreatic cancer

[75]

SNHG11

Upregulated

Tumor cell

Promotes angiogenesis through the miR-324-3p/VEGFA axis

Pancreatic cancer

[76]

SNHG1

Upregulated

Tumor cell

Promotes angiogenesis by regulating the miR-216b-5p/JAK2 axis

Breast cancer

[77]

AC073352.1

Upregulated

Tumor cell

Binds and stabilizes the YBX1 protein

Breast cancer

[78]

MALAT1

Upregulated

Tumor cell

Facilitates angiogenesis and predicts poor prognosis

Ovarian cancer

[79]

TUG1

Upregulated

Tumor cell

Promotes angiogenesis by inhibiting caspase-3 activity

Cervical cancer

[80]

LINC00161

Upregulated

Tumor cell

Promotes angiogenesis and metastasis by regulating the miR-590-3p/ROCK axis

Hepatocellular carcinoma

[81]

H19

Upregulated

Cancer stem cell

Promotes VEGF production and release in ECs

Liver cancer

[82]

circSHKBP1

Upregulated

Tumor cell

Enhances VEGF mRNA stability by the miR-582-3p/HUR axis

Gastric cancer

[37]

circRNA-100,338

Upregulated

Tumor cell

Facilitates HCC metastasis by enhancing invasiveness and angiogenesis

Hepatocellular carcinoma

[83]

circCMTM3

Upregulated

Tumor cell

Promotes angiogenesis and HCC tumor growth by the miR-3619-5p/SOX9 axis

Hepatocellular carcinoma

[84]

circ_0007334

Upregulated

Tumor cell

Accelerates CRC tumor growth and angiogenesis by the miR-577/KLF12 axis

Colorectal cancer

[85]

CircFNDC3B

Downregulated

Tumor cell

Inhibits angiogenesis and CRC progression by the miR-937-5p/TIMP3 axis

Colorectal cancer

[86]

circGLIS3

Upregulated

Tumor cell

Induces endothelial cell angiogenesis by promoting Ezrin T567 phosphorylation

Glioma

[87]

piRNA-823

Upregulated

Tumor cell

Promotes VEGF and IL-6 expression

Multiple myeloma

[88]

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

3.1. EV-Derived ncRNAs as Promising Tumor Biomarkers

EVs that possess great potential as disease biomarkers and therapeutic carriers have attracted increasing attention [89][90]. 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 [91][92]. Therefore, EVs are stable in circulation and under various storage conditions. Researchers 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 2A).
Figure 2. 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.
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 [93]. 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 [94]. 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 [95]. 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 [51]. 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 [96]. 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 [97]. Similarly, recent studies have reported serum exosomal circRNAs as novel and useful tools for the non-invasive diagnosis of cancer [98]. Exosomal circPRMT5 is highly expressed in the serum and urine of patients with bladder cancer and is closely related to tumor metastasis [99]. 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.

3.2 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 [100]. 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 [101]. Another study showed that the delivery of miR-29a/c using cell-derived MVs inhibited angiogenesis in GC [102]. 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 [83]. 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 [41]. 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 [103]. Therefore, exosome engineering is a promising tool in RNA-based therapeutics for cancer treatment (Figure 2B). 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 [104]. 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) [105]. 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.

References

  1. 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.
  2. 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.
  3. 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.
  4. 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.
  5. 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.
  6. Stuffers, S.; Wegner, C.S.; Stenmark, H.; Brech, A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic 2009, 10, 925–937.
  7. 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.
  8. Nagano, T.; Fraser, P. No-Nonsense Functions for Long Noncoding RNAs. Cell 2011, 145, 178–181.
  9. 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.
  10. 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.
  11. 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.
  12. 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.
  13. 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.
  14. 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.
  15. 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.
  16. 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.
  17. Gutschner, T.; Diederichs, S. The hallmarks of cancer. RNA Biol. 2012, 9, 703–719.
  18. Kopp, F.; Mendell, J.T. Functional Classification and Experimental Dissection of Long Noncoding RNAs. Cell 2018, 172, 393–407.
  19. 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.
  20. 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.
  21. 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.
  22. 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.
  23. Li, X.; Yang, L.; Chen, L.-L. The Biogenesis, Functions, and Challenges of Circular RNAs. Mol. Cell 2018, 71, 428–442.
  24. 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.
  25. 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.
  26. 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.
  27. Adams, R.H.; Alitalo, K. Molecular regulation of angiogenesis and lymphangiogenesis. Nat. Rev. Mol. Cell Biol. 2007, 8, 464–478.
  28. Cook, K.M.; Figg, W.D. Angiogenesis Inhibitors: Current Strategies and Future Prospects. CA Cancer J. Clin. 2010, 60, 222–243.
  29. 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.
  30. 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.
  31. De Bock, K.; Mazzone, M.; Carmeliet, P. Antiangiogenic therapy, hypoxia, and metastasis: Risky liaisons, or not? Nat. Rev. Clin. Oncol. 2011, 8, 393–404.
  32. Weis, S.M.; Cheresh, D.A. Tumor angiogenesis: Molecular pathways and therapeutic targets. Nat. Med. 2011, 17, 1359–1370.
  33. 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.
  34. 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.
  35. 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.
  36. 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.
  37. 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.
  38. 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.
  39. 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.
  40. 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.
  41. 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.
  42. 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.
  43. 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.
  44. 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.
  45. 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.
  46. 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.
  47. 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.
  48. 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.
  49. 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.
  50. 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.
  51. 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.
  52. 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.
  53. 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.
  54. 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.
  55. 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.
  56. 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.
  57. 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.
  58. 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.
  59. 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.
  60. 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.
  61. 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.
  62. 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.
  63. 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.
  64. 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.
  65. 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.
  66. 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.
  67. 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.
  68. 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.
  69. 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.
  70. 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.
  71. 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.
  72. 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.
  73. 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.
  74. 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.
  75. 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.
  76. 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.
  77. 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.
  78. 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.
  79. 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.
  80. 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.
  81. 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.
  82. 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.
  83. 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.
  84. 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.
  85. 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.
  86. 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.
  87. 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.
  88. 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.
  89. Jiang, L.; Gu, Y.; Du, Y.; Liu, J. Exosomes: Diagnostic Biomarkers and Therapeutic Delivery Vehicles for Cancer. Mol. Pharm. 2019, 16, 3333–3349.
  90. 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.
  91. 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.
  92. 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.
  93. 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.
  94. Maishi, N.; Hida, K. Tumor endothelial cells accelerate tumor metastasis. Cancer Sci. 2017, 108, 1921–1926.
  95. 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.
  96. 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.
  97. 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.
  98. 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.
  99. 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.
  100. 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.
  101. 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.
  102. 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.
  103. 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.
  104. 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.
  105. 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.
More
Upload a video for this entry
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : Zhaowu Ma , Lingzhi Wang , , Chen-Guang Liu
View Times: 591
Revisions: 5 times (View History)
Update Date: 11 Apr 2022
Notice
You are not a member of the advisory board for this topic. If you want to update advisory board member profile, please contact office@encyclopedia.pub.
OK
Confirm
Only members of the Encyclopedia advisory board for this topic are allowed to note entries. Would you like to become an advisory board member of the Encyclopedia?
Yes
No
${ textCharacter }/${ maxCharacter }
Submit
Cancel
There is no comment~
${ textCharacter }/${ maxCharacter }
Submit
Cancel
${ selectedItem.replyTextCharacter }/${ selectedItem.replyMaxCharacter }
Submit
Cancel
Confirm
Are you sure to Delete?
Yes No
Academic Video Service