Next Article in Journal
Antibodies against Angiotensin II Type 1 and Endothelin 1 Type A Receptors in Cardiovascular Pathologies
Next Article in Special Issue
MicroRNA-mRNA Regulatory Network Mediates Activation of mTOR and VEGF Signaling in African American Prostate Cancer
Previous Article in Journal
Role of GALNT2 on Insulin Sensitivity, Lipid Metabolism and Fat Homeostasis
Previous Article in Special Issue
CircRNAs as Potential Blood Biomarkers and Key Elements in Regulatory Networks in Gastric Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Oncogenic Role of Exosomal Circular and Long Noncoding RNAs in Gastrointestinal Cancers

1
Department of Biomedical Science and Research, Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea
2
Postgraduate Program in Genetics, Department of Genetics, Federal University of Paraná, Curitiba 81531-990, Brazil
3
Department of Pharmaceutical Engineering and Biotechnology, SunMoon University, 70 Sunmoon-ro 221, Tangjeong-myeon, Asan-si 31460, Korea
4
Genome-Based BioIT Convergence Institute, 70 Sunmoon-ro 221, Tangjeong-myeon, Asan-si 31460, Korea
5
Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(2), 930; https://doi.org/10.3390/ijms23020930
Submission received: 9 November 2021 / Revised: 12 January 2022 / Accepted: 13 January 2022 / Published: 15 January 2022
(This article belongs to the Special Issue RNA Regulatory Networks at the Crossroad of Human Diseases 2.0)

Abstract

:
Circular RNAs (circRNAs) and long noncoding RNAs (lncRNAs) are differentially expressed in gastrointestinal cancers. These noncoding RNAs (ncRNAs) regulate a variety of cellular activities by physically interacting with microRNAs and proteins and altering their activity. It has also been suggested that exosomes encapsulate circRNAs and lncRNAs in cancer cells. Exosomes are then discharged into the extracellular environment, where they are taken up by other cells. As a result, exosomal ncRNA cargo is critical for cell–cell communication within the cancer microenvironment. Exosomal ncRNAs can regulate a range of events, such as angiogenesis, metastasis, immune evasion, drug resistance, and epithelial-to-mesenchymal transition. To set the groundwork for developing novel therapeutic strategies against gastrointestinal malignancies, a thorough understanding of circRNAs and lncRNAs is required. In this review, we discuss the function and intrinsic features of oncogenic circRNAs and lncRNAs that are enriched within exosomes.

1. Introduction

Solid cancers of the gastrointestinal system, such as colorectal cancer, esophageal cancer, gastric cancer, hepatocellular carcinoma, and pancreatic cancer, are among the most frequent types of malignancies. According to the GLOBOCAN database, gastrointestinal cancers are responsible for 26% of cancer incidence and 35% of cancer-related mortality worldwide [1]. Due to the lack of curative options, little progress has been made in the treatment of patients with gastrointestinal malignancies. Several classes of drugs, such as sorafenib, paclitaxel, and cetuximab, have been approved for use. However, the overall survival of patients and their quality of life have not improved significantly [2,3]. In order to develop more effective and appropriate treatment strategies, it is critical to identify novel target molecules associated with gastrointestinal cancers.
In solid cancers, the microenvironment is heterogeneous and composed of cellular and noncellular components, including cancer-associated fibroblasts (CAFs), immune cells, and the extracellular matrix [4,5]. Hypoxia, one of the characteristics of the cancer microenvironment, is caused by factors such as uncontrolled cancer growth and leads to metabolic reprogramming [6]. Reciprocal communication among cells enables them to share diverse cellular factors within the microenvironment. Exosomes are a class of extracellular vesicles that act as key players in this event, affecting cancer aggressiveness, anti-cancer immune responses, and treatment efficacy [7,8]. For instance, annexin A1 (ANXA1) proteins are cargo molecules in cancer cell-derived exosomes and activate epithelial–mesenchymal transition (EMT) in exosome-receiving cancer cells [9]. Another example is the potential of exosomal protein cargo, macrophage migration inhibitory factor (MIF), to reshape the phenotype of drug-sensitive cancer cells into temozolomide-resistant cells by positively modulating phosphoinositide 3-kinase (PI3K)/AKT signaling. The pharmacological inhibition of MIF prominently enhances temozolomide efficacy in vivo [10]. In addition, gemcitabine resistance is exacerbated by CAF-derived exosomes that harbor snail family transcriptional repressor 1 (SNAI1) messenger RNA (mRNA). Anti-pancreatic cancer effects of gemcitabine can be improved by pharmacological interruption of exosome secretion using GW4869 [11].
Circular RNAs (circRNAs) and long noncoding RNAs (lncRNAs) are aberrantly expressed in cancer and impinge on the activity of target molecules, including microRNA (miRNA) and proteins, therefore controlling cancer progression, therapeutic resistance, etc. [12,13,14,15,16]. One of the known functions of both circRNAs and lncRNAs is to act as competitive endogenous RNAs by sequestering miRNAs. For example, recent studies demonstrated that circ_ZFPM2 and lncRNA-ADPGK-AS1 can promote EMT by molecularly sponging miR-205-5p [17,18]. Furthermore, circRNAs interact with RNA-binding proteins to control gene transcription [19,20]. LncRNAs have been found to regulate a variety of events, including RNA splicing and protein degradation [21].
These noncoding RNAs (ncRNAs) can be incorporated and differentially loaded into exosomes. For instance, circ_EPB41L2 quantity is low in exosomes secreted by colorectal cancer cell lines. Treatment of cancer cells with exosomes acquired from circ_EPB41L2-overexpressing cells retards cell proliferation, survival, migration, and invasion by negatively modulating the effect of miR-21-5p and miR-942-5p on phosphatase and tensin homolog (PTEN) mRNA [22]. This review focuses on exosome-enriched circRNAs and lncRNAs to meticulously understand their oncogenic role in gastrointestinal cancers. We also present an overview of the relevant features of circRNAs and lncRNAs.

2. Exosomal CircRNAs

2.1. Colorectal Cancer

2.1.1. Circ_0000338

It was demonstrated that drug-sensitive parental cells cocultured with FOLFOX-resistant cells become desensitized to 5-fluorouracil (5-FU), implying that exosome-transferred molecules contribute to chemoresistance. Profiling of circRNAs revealed that circ_0000338 is one of the upregulated circRNAs in exosomes secreted from FOLFOX-resistant cells [23]. Another study further showed that circ_0000338 levels are upregulated in cancer tissues and cell lines and that the silencing of circ_0000338 sensitizes resistant cells to 5-FU via enhancing apoptotic cell death [24]. Treatment of cells with exosomes harboring circ_0000338 reduces the efficacy of 5-FU in vitro and in vivo. Mechanistically, it was identified that circ_0000338 directly interacts with and inactivates miR-217 and miR-485-3p [24] (Figure 1 and Table 1). It is known that miR-217 and miR-485-3p target mitogen-activated protein kinase 1 (MAPK1) and maternal embryonic leucine zipper kinase (MELK), respectively [25,26]. Since MAPK1 and MELK can promote 5-FU resistance [27,28], circ_0000338 may regulate drug sensitivity, partly via modulating the level of both MAPK1 and MELK.

2.1.2. Circ_0005963

Glycolysis has been found to be associated with therapeutic resistance toward several classes of anti-cancer drugs. Lactate, a glycolytic metabolite, can induce drug resistance by creating an acidic tumor microenvironment [29]. In addition, drug resistance can also be due to a glycolysis-induced upregulation of drug efflux pumps. For example, pyruvate kinase M2 type (PKM2), a glycolytic enzyme, facilitates oxaliplatin resistance via upregulating ATP-binding cassette subfamily B member 1 (ABCB1, also called P-glycoprotein 1) [30]. Recently, it was reported that circ_0005963 is enriched in exosomes from oxaliplatin-resistant cells and the serum of an oxaliplatin-resistant patient [31]. In this study, circ_0005963 was observed to be delivered from oxaliplatin-resistant cells to sensitive cells via exosomes. In recipient cells, circ_0005963 increases PKM2 expression via inactivating miR-122, ultimately diminishing the anti-colorectal cancer effect of oxaliplatin. Knockdown of circ_0005963 augments oxaliplatin-induced apoptotic cell death in vitro and inhibits cancer growth in vivo [31] (Figure 1 and Table 1).
Table 1. Exosomal circRNAs in gastrointestinal cancers (in alphanumerical order).
Table 1. Exosomal circRNAs in gastrointestinal cancers (in alphanumerical order).
Cargo MoleculeExpressionSource of ExosomeType of CancerTarget
Molecule
Clinical
Relevance
Ref.
Circ_0000337UpCisplatin-resistant EC9706 and KYSE30 cellsEsophageal cancermiR-377-3p-[32]
Circ_0000338UpFOLFOX-resistant HCT116 cellsColorectal cancer--[23]
Up5-FU-resistant SW480 and HCT116 cellsColorectal cancermiR-217 and miR-485-3p5-FU resistance is associated with high levels of circ_0000338[24]
Circ_0005963UpSerum from oxaliplatin-resistant patients. Oxaliplatin-resistant SW480 cellsColorectal cancermiR-122Highly expressed in oxaliplatin-resistant patients[31]
Circ_0010522UpSW480 and HCT116 cells exposed to a hypoxic condition (1% O2)Colorectal cancermiR-133a-3pPositively associated with the stages of cancer[33]
Circ_0032821UpOxaliplatin-resistant HGC-27 and AGS cellsGastric cancermiR-515-5p-[34]
Circ_0044366UpPatient plasma. SGC-7901 and MGC-803 cellsGastric cancermiR-29a-[35]
Circ_0061395UpPatient serumHepatocellular carcinomamiR-877-5p-[36]
Circ_0067835UpSerum from radiotherapy-treated patients. SW620 and HCT116 cellsColorectal cancermiR-296-5pA diagnostic biomarker candidate[37]
Circ_0088300UpPatient plasma. CAFsGastric cancermiR-1305Negative correlation between circ_0088300 expression and survival probability[38]
Circ_EIF3KUpHypoxic CAFsColorectal cancermiR-214Poor overall survival of patients with high circ_EIF3K levels[39]
Circ_FBLIM1UpPatient serumHepatocellular carcinomamiR-338-3p-[40]
Circ_IARSUpPatient plasma. Hs766T and Hs766T-L2 cellsPancreatic cancermiR-122Associated with vascular invasion; liver metastasis; and advanced tumor, node, and metastasis (TNM) stage[41]
Circ_IFT80UpPatient serum. SW480 and SW620 cellsColorectal cancermiR-296-5p-[42]
Circ_MMP2UpLM3 and 97H cellsHepatocellular carcinomamiR-136-5pLow survival rate of patients with high circ_MMP2 expression[43]
Circ_NEK9UpPatient plasmaGastric cancermiR-409-3pAssociated with TNM stage[44]
Circ_NHSL1UpPatient serum. HGC-27 and AGS cellsGastric cancermiR-149-5pAssociated with lymphatic metastasis and TNM stage[45]
Circ_PACRGLUpHCT116 and SW480 cellsColorectal cancermiR-142-3p and miR-506-3p-[46]
Circ_PDE8AUpPatient plasma. Hs766T-L2 cellsPancreatic cancermiR-338-3pAssociated with lymphatic invasion and TNM stage[47]
Circ_PRRX1UpDoxorubicin-resistant HGC-27 and AGS cellsGastric cancermiR-3064-5pAssociated with doxorubicin resistance[48]
Circ_PVT1UpSerum from cisplatin-resistant patients. Cisplatin-resistant HGC-27 and AGS cellsGastric cancermiR-30a-5pAssociated with lymph node metastasis and tumor size[49]
Circ_SHKBP1UpPatient serum. BGC823 and HGC27 cellsGastric cancermiR-582-3pPositively associated with poor prognosis, vascular invasion, and TNM stage[50]
Circ_TMEM45AUpPatient serumHepatocellular carcinomamiR-665Correlated with poor prognosis and clinicopathological parameters such as TNM stage[51]
Circ_UHRF1UpHCCLM3 and SMMC-7721 cellsHepatocellular carcinomamiR-449c-5pPoor clinical prognosis of patients with high circ_UHRF1 expression[52]
Circ_ZNF652UpPatient serum. SNU-387 and Huh7 cellsHepatocellular carcinomamiR-29a-3p-[53]
Circ_ZNF91UpHypoxic BxPC-3 cellsPancreatic cancermiR-23b-3pWorse overall survival of patients with high circ_ZNF91 expression[54]

2.1.3. Circ_0010522 and Circ_EIF3K

Guanine nucleotide exchange factor H1 (GEF-H1) serves as an activator of Ras homolog family member A (RhoA), promoting cell motility, invasion, and metastasis [55]. A recent study showed that circ_0010522 is present in exosomes secreted from hypoxic cancer cells [33]. Migratory capacities of normoxic cells are increased by treatment with hypoxic exosomes, suggesting a pro-migratory activity of circ_0010522. It was further shown that circ_0010522 absorbs miR-133a-3p, thereby elevating GEF-H1 and RhoA levels [33] (Figure 1 and Table 1).
Secretion of exosomes from CAFs is stimulated by hypoxic conditions [39]. Further investigation demonstrated that these exosomes carry circ_EIF3K, which sponges miR-214, which targets programmed cell death 1 ligand 1 (PD-L1). Colony formation of cancer cells is attenuated by culturing with conditioned media derived from circ_EIF3K-silenced CAFs. The growth of cancer is also impaired in xenografts treated with exosomes from circ_EIF3K-silenced CAFs. These results denote the role of exosomal circ_EIF3K in cancer progression [39] (Figure 1 and Table 1).

2.1.4. Circ_0067835 and Circ_IFT80

Circ_0067835 is abundant in blood-derived exosomes from patients who received radiotherapy, suggesting an involvement of this circRNA in radiosensitivity modulation. Indeed, it was noticed that radiotherapy-induced apoptosis is increased in cancer cells treated with exosomes derived from circ_0067835-silenced cells [37]. Furthermore, exosomal circ_IFT80 desensitizes recipient cells to radiation [42]. In their studies, circ_0067835 and circ_IFT80 were found to positively regulate the level of insulin-like growth factor 1 receptor (IGF1R) and musashi-1 (MSI1), respectively, by sponging miR-296-5p [37,42] (Figure 2 and Table 1). IGF1R and MSI1 have been investigated to reduce radiosensitivity by activating DNA damage repair [56,57].

2.1.5. Circ_PACRGL

Microarray analysis showed that circ_PACRGL is copiously detected in exosomes released from cancer cells [46]. Functional investigation exhibited that exosomal circ_PACRGL stimulates cell proliferation, migration, and invasion by sponging miR-142-3p and miR-506-3p, both of which directly regulate transforming growth factor beta 1 (TGFB1). In addition, exosomal circ_PACRGL stimulates neutrophil polarization towards a pro-tumorigenic N2 phenotype [46], suggesting that circ_PACRGL is a promising target for cancer treatment (Figure 2 and Table 1).

2.2. Esophageal Cancer

Circ_0000337
Circ_0000337 has been discovered to be an oncogenic factor. In osteosarcoma, circ_0000337 augments proliferation and migration by elevating BTB domain and CNC homolog 1 (BACH1) levels [58]. Besides this, circ_0000337 upmodulates methionine adenosyltransferase 2A (MAT2A) expression, thus accelerating migration and invasion in glioblastoma cells [59]. Circ_0000337 is highly expressed in esophageal cancer tissues and enhances the metastatic potential of cancer cells [60]. Moreover, it was found that circ_0000337 is highly expressed in cisplatin-resistant esophageal cancer cells and abundant in their secreted exosomes [32]. Non-resistant parental cells become less sensitive to cisplatin following exposure to exosomes from resistant cells in vitro and in vivo. Circ_0000337 can aggravate cisplatin resistance due to its interaction with miR-377-3p, which targets Janus kinase 2 (JAK2) [32] (Figure 1 and Table 1). However, miR-377-3p was shown to increase glycogen synthase kinase 3 beta (GSK3B) expression and activate nuclear factor kappa B (NF-κB) signaling [61]. Therefore, circ_0000337 knockdown may unexpectedly activate GSK3B/NF-κB signaling.

2.3. Gastric Cancer

2.3.1. Circ_0032821, Circ_PRRX1, and Circ_PVT1

It has been evaluated that miR-515-5p attenuates chemoresistance. The efficacy of vincristine and carboplatin is improved by miR-515-5p in retinoblastoma [62]. Further, miR-515-5p can negatively control interleukin 25 (IL25) levels and sensitize resistant cells to cisplatin as well as to 5-FU in nasopharyngeal cancer [63]. In gastric cancer, circ_0032821 competitively interacts with miR-515-5p to control SRY-box transcription factor 9 (SOX9) expression [34]. Owing to such an ability of circ_0032821, oxaliplatin resistance can be provoked in drug-sensitive cells after exposure to exosomal circ_0032821 derived from oxaliplatin-resistant cells [34] (Figure 1 and Table 1).
Protein tyrosine phosphatase non-receptor type 14 (PTPN14) functions as an oncogenic factor by advancing the proliferation and migration of gastric cancer cells via triggering Hippo signaling [64]. Additionally, PTPN14 overexpression can upregulate and downregulate the level of vimentin and caspase-3, respectively, in gastric cancer [65]. Analyses of the relationship between circ_PRRX1 and PTPN14 showed that circ_PRRX1 is able to positively control PTPN14 levels via sequestering miR-3064-5p, consequently intensifying doxorubicin resistance in gastric cancer [48]. Circ_PRRX1 is bountiful in exosomes released from doxorubicin-resistant cells. The effect of doxorubicin on proliferation, migration, and invasion is attenuated in exosome-receiving cancer cells [48] (Figure 1 and Table 1).
Circ_PVT1 is involved in the regulation of a variety of cellular events. Circ_PVT1 overexpression increases the invasive capacity of esophageal and gallbladder cancer cells [66,67]. In addition, circ_PVT1 makes lung cancer cells less responsive to Pemetrexed and cisplatin by elevating the expression of ABCC1 (also known as multidrug resistance-associated protein 1, MRP1) [68]. In gastric cancer, circ_PVT1 is profusely present in exosomes from cisplatin-resistant cancer cells [49]. In their study, circ_PVT1 was observed to negatively control miR-30a-5p, which targets Yes-associated protein 1 (YAP1). The silencing of circ_PVT1 re-sensitizes resistant cancer cells to cisplatin by augmenting apoptotic cell death and suppressing autophagy [49]. These findings suggest that exosomal circ_PVT1 is responsible for cisplatin resistance in recipient cells, at least partly via ABCC1 and YAP1 (Figure 1 and Table 1).

2.3.2. Circ_0044366 and Circ_SHKBP1

Exosomal circRNAs play a part in angiogenesis regulation. Treatment with cancer cell-derived exosomal circ_0044366 significantly enhances the proliferation, invasion, and tube formation of human umbilical vein endothelial cells (HUVECs) in vitro. Besides this, exosomal circ_0044366 promotes cancer growth and angiogenesis in vivo. Mechanistically, exosomal circ_0044366 leads to an increment of vascular endothelial growth factor (VEGF) levels via inactivating miR-29a in endothelial cells [35] (Figure 2 and Table 1).
Circ_SHKBP1 levels are higher in cancer tissues and exosomes derived from patient serum and cancer cell lines [50]. Exosomal circ_SHKBP1 facilitates cell proliferation, migration, and invasion. Furthermore, exosomal circ_SHKBP1 increases Hu-antigen R (HUR) levels via inactivating miR-582-3p, resulting in an increment of VEGF expression in cancer cells in vitro. Cancer growth and angiogenesis are activated by circ_SHKBP1 in vivo [50] (Figure 2 and Table 1). Other evidence showed that circ_SHKBP1 supports survival and tube formation of endothelial cells [69]. Therefore, these results suggest that circ_SHKBP1 can also be transferred to endothelial cells via exosomes, which, in turn, stimulate angiogenic events.

2.3.3. Circ_0088300

Once activated, JAK/signal transducer and activator of transcription (STAT) signaling promotes the expression of target genes involved in diverse cellular events, such as cell survival, migration, invasion, and therapeutic resistance [70]. Because JAK/STAT signaling is abnormally activated in gastric cancer, inhibiting this pathway has been proposed as a therapeutic choice [71]. Lately, circ_0088300 was identified as an exosomal circRNA from CAFs. Gastric cancer cells treated with conditioned media from CAFs express high circ_0088300 levels [38]. Circ_0088300 inactivates miR-1305, a negative regulator of JAK1 and STAT1. The overexpression of circ_0088300 reduces the level of pro-apoptotic factors such as BCL2-associated X protein (BAX) in cancer cells and enhances migration and invasion [38]. These observations reveal circ_0088300 as a mediator of CAF-induced cancer progression (Figure 1 and Table 1).

2.3.4. Circ_NEK9 and Circ_NHSL1

Both miR-149-5p and miR-409-3p perform a tumor-suppressive function. By targeting BCL2 and cell division cycle 42 (CDC42), miR-149-5p promotes apoptotic cell death, represses cell proliferation, and alleviates doxorubicin resistance in neuroblastomas [72]. In gastric cancer, miR-149-5p has an anti-metastatic property by suppressing forkhead box M1 (FOXM1) expression [73]. In addition, miR-409-3p is downregulated and capable of suppressing proliferation, invasion, survival, and metastasis by regulating the level of radixin and PHD finger protein 10 (PHF10) in gastric cancer [74,75].
Further functional studies showed that circ_NEK9 and circ_NHSL1 inactivate miR-409-3p and miR-149-5p, respectively. By suppressing such miRNAs, circ_NEK9 and circ_NHSL1 elevate the expression of microtubule-associated protein 7 (MAP7) and tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta (YWHAZ), respectively, in cancer cells [44,45]. Circ_NEK9 is upregulated in gastric cancer cells and abundant in exosomes from patient plasma. These results suggest that exosomal circ_NEK9 is originated from cancer cells rather than other cellular components within the tumor microenvironment. The metastatic ability of cancer cells is intensified by plasma exosomes, connoting the contribution of exosomal circ_NEK9 to gastric cancer progression by raising MAP7 levels [44]. Circ_NHSL1 is enriched in exosomes from cancer cells; therefore, it is feasible that exosomal circ_NHSL1 facilitates migration, invasion, and glutaminolysis in recipient cancer cells by upregulating YWHAZ expression [45] (Figure 2 and Table 1).

2.4. Hepatocellular Carcinoma

2.4.1. Circ_0061395

Circ_0061395 (also called circ_BACH1) is notably overexpressed in hepatocellular carcinomas. It was also observed that circ_0061395 can be incorporated into exosomes [36]. Functional analyses indicated that circ_0061395 inactivates miR-877-5p and contributes to cancer progression by positively modulating the expression of phosphoinositide-3-kinase regulatory subunit 3 (PIK3R3) [36] (Figure 2 and Table 1). Other studies demonstrated that circ_0061395 can promote cell proliferation and EMT via upregulating SERBP1 and translocating HUR to the cytoplasm [76,77]. Since EMT is one of the causes of therapeutic resistance [78,79], both EMT and drug resistance may be aggravated by the exosomal transport of circ_0061395 between hepatocellular carcinoma cells. Additionally, circ_0061395 is overexpressed in colorectal cancer; supports cell proliferation, migration, and invasion; and restricts apoptotic cell death [80]. Thus, it is worth investigating the existence and amount of circ_0061395 in exosomes released from colorectal cancer cells.

2.4.2. Circ_FBLIM1

It was denoted that circ_FBLIM1 is overexpressed in hepatocellular carcinoma tissues and cell lines [81]. Additionally, circ_FBLIM1 is plenteous in serum exosomes from patients [40], suggesting that exosomal circ_FBLIM1 can be originated from cancer cells. Treatment of hepatocellular carcinoma cells with serum exosomes leads to an upregulation of circ_FBLIM1 levels [40]. In their study, this circRNA was validated to deactivate miR-338-3p, thus triggering cancer progression and glycolysis by increasing the level of low-density lipoprotein receptor-related protein 6 (LRP6), which acts as a coreceptor of WNT ligands and activates WNT/β-catenin signaling [40]. Hence, circ_FBLIM1 may be transported between cancer cells to regulate the miR-338-3p/LRP6/WNT/β-catenin axis (Figure 2 and Table 1).
Another study showed that knockdown of circ_FBLIM1 hampers cell proliferation and invasion while promoting apoptotic cell death by augmenting miR-346 levels [81]. Even though circ_FBLIM1 can interact with miR-346, this miRNA has been reported to act as an oncogenic miRNA in various cancer types, including hepatocellular carcinoma [82,83,84]. Such findings point to the possibility that suppression of oncogenic circRNAs compensatorily activates a set of oncogenic miRNAs and their relevant signaling, eventually contributing to the recovery of damaged cancer cells.

2.4.3. Circ_MMP2

In lung cancer, circ_MMP2 (also known as circ_0039411) can bind to insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3), thus expediting cell proliferation and EMT by stabilizing FOXM1 transcripts [85]. Besides this, circ_MMP2 was determined to advance the progression of papillary thyroid cancer by post-transcriptionally upregulating ABCA9 and metastasis-associated 1 (MTA1) [86]. Such an oncogenic role of circ_MMP2 was also reported in hepatocellular carcinomas [43]. Circ_MMP2 is plentiful in cancer cell-derived exosomes and can be transported into other cancer cells. Circ_MMP2 is involved in an increase of the metastatic potential of exosome-receiving cancer cells via modulating the miR-136-5p/matrix metallopeptidase 2 (MMP2) axis [43] (Figure 2 and Table 1).

2.4.4. Circ_TMEM45A

Functional investigations indicated that this circRNA blocks apoptotic cell death and promotes in vivo cancer growth via regulating the miR-665/insulin growth factor 2 (IGF2) axis [51]. Circ_TMEM45A is one of the highly expressed circRNAs in hepatocellular carcinoma and is bountifully present in serum exosomes as well [51]. Therefore, sharing of circ_TMEM45A between cancer cells may contribute to IGF2 upregulation (Figure 2 and Table 1).
Inconsistent with the IGF2-targeting property of miR-665, other studies showed that miR-665 is overexpressed in hepatocellular carcinomas and prompts cell proliferation as well as metastasis [87,88]. Thus, circ_TMEM45A may interact with other potential target proteins and unidentified miRNAs, rather than miR-665, to fulfill its oncogenic role in hepatocellular carcinomas.

2.4.5. Circ_UHRF1

A recent study showed that circ_UHRF1 accelerates cell proliferation and EMT by elevating MYC proto-oncogene (MYC) expression in oral squamous cell carcinomas [89]. Further, circ_UHRF1 is upregulated in hepatocellular carcinoma tissues, and exosomal circ_UHRF1 is largely released from cancer cells [52]. Exosomal circ_UHRF1 was revealed to impair the function of natural killer (NK) cells by sequestering miR-449c-5p that directly targets hepatitis A virus cellular receptor 2 (HAVCR2, also named TIM3), ultimately diminishing the anti-cancer effects of nivolumab, anti-programmed cell death 1 (PD1) antibodies [52] (Figure 2 and Table 1).
It has been suggested that HAVCR2 is a molecular marker of activated NK cells and that NK cell-mediated cytotoxicity can be inhibited by anti-HAVCR2 antibodies [90]. By contrast, another study showed that NK cell-mediated cytotoxicity is improved by an HAVCR2 blockade [91]. Such a double-edged function of HAVCR2 is thought to be dependent on the type of HAVCR2 ligands [90,91]. Therefore, it is necessary to reveal the precise function of HAVCR2 and its contribution to the oncogenic effect of circ_UHRF1.

2.4.6. Circ_ZNF652

It has been proven that miR-203 and miR-502-5p are tumor-suppressive miRNAs in gastrointestinal cancers. For instance, miR-203 negatively modulates the level of eukaryotic translation initiation factor 5A2 (EIF5A2) in colorectal cancer, restraining cancer progression [92]. In the case of miR-502-5p, this miRNA can suppress cytoprotective autophagy in colorectal cancer [93]. In addition to this, miR-502-5p was recently explored to target Sp1 transcription factor (SP1), thus impeding migration and invasion of gastric cancer cells [94].
In hepatocellular carcinoma, miR-203 and miR-502-5p commonly target SNAI1, an EMT-promoting factor, and they are sponged by circ_ZNF652; therefore, lung metastasis is provoked by circ_ZNF652 [95]. Furthermore, circ_ZNF652 is an exosomal circRNA in hepatocellular carcinomas [53]. Circ_ZNF652 is significantly detected in exosomes derived from patient serum and cancer cells. The silencing of circ_ZNF652 suppresses migration, invasion, and glycolysis in exosome-receiving cells. The mechanism underlying circ_ZNF652-mediated cellular events involves the regulation of the miR-29a-3p/guanylyl cyclase domain containing 1 (GUCD1) axis [53] (Figure 2 and Table 1). Interestingly, circ_ZNF652 increases PTEN expression, thus positively regulating lipopolysaccharide-induced apoptosis in chondrocytes [96], indicating that the function of circ_ZNF652 can be distinct, depending on the cellular context.

2.5. Pancreatic Cancer

2.5.1. Circ_IARS

Endothelial tight junctions act as permeability barriers and restrict cancer metastasis. Transendothelial permeability of cancer cells is induced by the downregulation of Zona occludens-1 (ZO-1), a master regulator of tight junctions [97]. It has been noticed that endothelial tight junctions are disrupted by cancer-secreted factors, such as MMP2, MMP9, and miR-105, inducing vascular permeability and metastasis [98]. Besides, it was demonstrated that circ_IARS is overexpressed in pancreatic cancer tissues and especially abundant in plasma exosomes from patients with metastatic cancer [41]. Cancer cell-derived exosomal circ_IARS downregulates ZO-1 levels via inactivating miR-122 in endothelial cells and increases endothelial permeability, eventually reinforcing invasion and metastasis in vivo [41] (Figure 2 and Table 1).

2.5.2. Circ_PDE8A

In various cancer types, miR-338-3p functions as a tumor-suppressive miRNA. By inactivating MAPK signaling, miR-338-3p promotes apoptosis in osteosarcoma cells [99]. In ovarian cancer, the efficacy of cisplatin is augmented by miR-338-3p, which exerts suppressive effects on EMT and cell survival via targeting WNT2B [100]. Similarly, the migration and invasiveness of colorectal cancer cells are repressed by miR-338-3p that targets smoothened (SMO) [101]. It was further revealed that miR-338-3p directly controls the expression of metastasis-associated in colon cancer protein 1 (MACC1) [47], a transcriptional regulator of MET proto-oncogene (MET) [102]. Additional evidence indicated that miR-338-3p activity is limited by circ_PDE8A (also named circ_0036627), which is intensely expressed in pancreatic cancer tissues, plasma exosomes, and cell-derived exosomes [47]. Exosomal circ_PDE8A is capable of being transferred between cancer cells. In addition, this circRNA is able to augment the level of MET via the miR-338-3p/MACC1 axis, stimulating cancer growth and metastasis in vivo [47] (Figure 2 and Table 1).

2.5.3. Circ_ZNF91

Hypoxia-inducible factor-1α (HIF-1α) is stabilized by sirtuin 1 (SIRT1)-mediated deacetylation. SIRT1 knockdown reduces the invasion ability of cancer cells, together with a decrease in HIF-1α target genes, such as VEGF [103]. SIRT1 can be directly targeted by several miRNAs, including miR-34a-5p and miR-199a, resulting in the attenuation of HIF-1α levels [104,105]. Furthermore, circ_CIDN and circ_0076248 were uncovered to sponge miR-34a-5p and miR-181a, respectively, hence positively regulating SIRT1 expression [106,107].
Likewise, circ_ZNF91 competitively interacts with miR-23b-3p, enhancing HIF-1α stability by upregulating SIRT1 in pancreatic cancer [54]. Circ_ZNF91 is transcriptionally induced by HIF-1α, indicating a positive feedback loop between circ_ZNF91 and HIF-1α. Circ_ZNF91, which is carried in exosomes from hypoxic cancer cells, can be taken up by normoxic cancer cells, leading to gemcitabine resistance and glycolysis. In xenografts, gemcitabine resistance promoted by hypoxic exosomes is reversed by circ_ZNF91 silencing and miR-23b-3p upregulation [54] (Figure 1 and Table 1).

3. Exosomal LncRNAs

3.1. Colorectal Cancer

3.1.1. CCAL and LINC00659

Accumulating evidence has shown that the expression of CCAL is substantially higher in cancer than in non-cancerous tissues and that CCAL promotes angiogenesis and metastasis through escalating, for example, FOXM1 and angiopoietin-like 4 (ANGPTL4) expression [73,108]. In addition, CCAL regulates cell proliferation and apoptosis in colorectal cancer and plays a part in 5-FU resistance, owing to its ability to increase ABCB1 levels via activating WNT/β-catenin signaling [109]. Furthermore, CCAL is overexpressed in CAFs and the stromal region in colorectal cancer tissues [110]. Exosomal CCAL released from CAFs is delivered to cancer cells and activates β-catenin via directly interacting with HUR proteins, thus conferring resistance to oxaliplatin [110] (Figure 3 and Table 2).
LncRNA profiling identified that 201 lncRNAs have aberrant levels, including 165 upregulated and 36 downregulated lncRNAs in colorectal cancer. LINC00659 is one of the highly expressed lncRNAs, and the knockdown of this lncRNA suppresses cell cycle progression and cell survival through inactivating PI3K/AKT signaling [111]. Moreover, LINC00659 is abundantly expressed in CAF-derived exosomes, and LINC00659 uptake by cancer cells leads to enhanced proliferation and EMT [112]. A mechanistic investigation proved that LINC00659 acts as a sponge of miR-342-3p, significantly upregulating the expression of annexin A2 (ANXA2) [112], which is responsible for the activation of STAT3, an EMT-promoting factor [113] (Figure 3 and Table 2).
Table 2. Exosomal lncRNAs in gastrointestinal cancers (in alphanumerical order).
Table 2. Exosomal lncRNAs in gastrointestinal cancers (in alphanumerical order).
Cargo MoleculeExpressionSource of ExosomeType of CancerTarget
Molecule
Clinical
Relevance
Ref.
AFAP1-AS1UpM2 macrophageEsophageal cancermiR-26a-[114]
ASMTL-AS1UpResidual Huh7 cells following heat treatment using a 47 °C water bathHepatocellular carcinomamiR-342-3pCorrelated with distant metastasis and TNM stage[115]
CCALUpCAFsColorectal cancerHUR-[110]
CCAT1UpPatient plasma. PANC-1 cellsPancreatic cancermiR-138-5p-[116]
CEBPA-AS1UpBGC-823 and SGC-7901 cellsGastric cancer-Closely associated with Bormann type and TNM stage[117]
CRNDE-hUpPatient serum. SW480, HT29, and LOVO cellsColorectal cancerRORγtPositively associated with the proportion of Th17 cells[118]
DLX6-AS1UpSMMC-7721 and HepG2 cellsHepatocellular carcinomamiR-15a-5p-[119]
FAM72D-3UpPatient serumHepatocellular carcinomamiR-5787Also upregulated in patients with hepatitis and cirrhosis[120]
FAM225AUpECA109 and TE-1 cellsEsophageal cancermiR-206Associated with advanced stages and poor prognosis[121]
FMR1-AS1UpSerum of female patients. ECA-109 and KYSE-150 cellsEsophageal cancerTLR7Associated with a poor clinical outcome of female patients[122]
FRLnc1UpPatient serum. HGC-27 cellsGastric cancer-Associated with TNM stage and lymph node metastasis[123]
HOTTIPUpMitomycin-resistant SW620 and HCT116 cellsColorectal cancermiR-214Negatively associated with mitomycin response[124]
UpCisplatin-resistant MGC-803 and MKN-45 cellsGastric cancermiR-218Poor response to cisplatin in patients with high HOTTIP levels[125]
HULCUpPatient serum. PANC-1 cellsPancreatic cancermiR-133bA diagnostic biomarker candidate[126]
KCNQ1OT1UpSW1463 cellsColorectal cancermiR-30a-5pAssociated with vascular invasion, lymph node metastasis, and distant metastasis[127]
LINC00161UpPatient serum. Huh7, HCCLM3, MHCC-97L, and MHCC-97H cellsHepatocellular carcinomamiR-590-3pAssociated with poor prognosis[128]
LINC00659UpCAFsColorectal cancermiR-342-3pHighly expressed in patients with poor prognosis[112]
LINC01133UpCFPAC-1 and SW1990 cellsPancreatic cancerAXIN2Associated with poor overall survival and TNM stage[129]
LINC01559Upmesenchymal stem cellsGastric cancermiR-1343-3pPoor prognosis of patients with high LINC01559 levels[130]
LINC01711UpTE-1 cellsEsophageal cancermiR-326Associated with poor prognosis[131]
LINC02418UpPatient serumColorectal cancermiR-1273g-3pConsidered as a possible diagnostic marker[132]
MALAT1UpSW620 and LoVo cellsColorectal cancermiR-26a and 26bWorse survival probability of patients with high MALAT1 levels[133]
PCAT1UpPatient serum. Eight cell lines, including KYSE30Esophageal cancermiR-326-[134]
PCGEM1UpHypoxic AGS and MKN cellsGastric cancerSNAI1-[135]
POU3F3UpKYSE450 and TE12 cellsEsophageal cancer-Associated with poor survival rates of patients[136]
RPPH1UpPatient plasma. SW620 and HCT8 cellsColorectal cancerTUBB3Negatively correlated with overall survival[137]
SPRY4-IT1UpPatient serumGastric cancermiR-101-3pCorrelated with tumor size and TNM stage[138]
TUC339UpPLC/PRF/5 cellsHepatocellular carcinoma--[139]
UCA1UpPatient serumColorectal cancermiR-143Associated with advanced stages and distant metastasis[140]
UpPatient serum. Hypoxic MIA PaCa-2 cellsPancreatic cancermiR-96-5pAssociated with microvascular density, tumor size, lymphatic invasion, TNM stage, and overall survival[141]
ZFAS1UpEca109 cellsEsophageal cancermiR-124Associated with lymph node metastasis, TNM stage, and tumor size[142]

3.1.2. CRNDE-h, KCNQ1OT1, and RPPH1

Cancer-associated lncRNAs affect the tumor immune microenvironment in colorectal cancer. CRNDE-h levels are upregulated in naive CD4+ T cells exposed to cancer cell-secreted exosomes, indicating the transfer of CRNDE-h into CD4+ T cells via exosomes [118]. In addition, it was noted that exosomal CRNDE-h induces the differentiation of peripheral blood mononuclear cells (PBMC) into pro-inflammatory T helper 17 (Th17) cells by binding to and blocking ubiquitin-mediated degradation of RAR-related orphan receptor gammat (RORγt). The knockdown of CRNDE-h reduces the Th17 cell population in company with RORγt downregulation and retards cancer growth in an allograft mouse model [118] (Figure 4 and Table 2).
Numerous studies have suggested that oncogenic KCNQ1OT1 is implicated in migration, invasion, apoptosis, and therapeutic resistance. For example, KCNQ1OT1 significantly increases migration and invasion in hepatocellular carcinomas and osteosarcomas [143,144]. In small cell lung cancer, the inhibition of KCNQ1OT1 induces apoptosis and sensitizes cells to etoposide by interrupting JAK/STAT signaling [145]. Additionally, KCNQ1OT1 expression is upmodulated in methotrexate-resistant colorectal cancer cells, and KCNQ1OT1 silencing re-sensitizes resistant cells to methotrexate through downregulating cAMP-response element binding protein (CREB) and CREB-binding protein (CBP) levels [146]. Of late, KCNQ1OT1 was observed to support ubiquitin-specific peptidase 22 (USP22)-mediated stabilization of PD-L1 by deactivating miR-30a-5p and thereby inhibiting the anti-cancer immunity of CD8+ T cells [127]. Since KCNQ1OT1 is secreted from cancer cells via exosomes, the reciprocal transfer of KCNQ1OT1 between cancer cells can substantially increase PD-L1 levels, contributing to immune evasion [127] (Figure 4 and Table 2). KCNQ1OT1 knockdown may activate other oncogenic factors, since miR-30a-5p is intriguingly capable of negatively regulating tumor-suppressive genes [147,148].
Tubulin beta 3 class III (TUBB3) is induced by SNAI1 and facilitates cell migration and invasion [149]. The activity of focal adhesion kinase (FAK), SRC proto-oncogene (SRC), and STAT3 is downregulated by TUBB3 silencing, thus restraining metastasis [150]. Furthermore, it was proven that RPPH1 physically binds to and stabilizes TUBB3 proteins in colorectal cancer cells [137]. Since cancer cell-derived exosomes contain RPPH1, this lncRNA is able to affect TUBB3 expression in other surrounding cancer cells, encouraging colorectal cancer progression. Moreover, exosomal RPPH1 can mediate M2 polarization of macrophages, ultimately promoting migration, invasion, EMT, and metastasis [137] (Figure 4 and Table 2).

3.1.3. HOTTIP

Several studies have indicated that HOTTIP is interconnected with drug resistance. HOTTIP desensitizes lung cancer cells to doxorubicin, etoposide, and cisplatin, through increasing BCL2 levels [151]. Downregulation of HOTTIP turns WNT/β-catenin signaling inactive, improving anti-prostate cancer effects of cisplatin [152]. Similarly, HOTTIP enhances stemness by attenuating miR-205 effects and confers resistance to cisplatin in ovarian cancer [153].
In colorectal cancer, HOTTIP is overexpressed in mitomycin-resistant cancer cells and their exosomes. Exosomes from resistant cells weaken mitomycin-induced DNA damage in non-resistant cells via conveying HOTTIP, which impairs miR-214-mediated inhibition of karyopherin subunit alpha 3 (KPNA3) expression [124] (Figure 3 and Table 2). As mentioned above, HOTTIP inactivates miR-205. In endothelial cells, miR-205 can activate AKT signaling, thus promoting angiogenesis [154]. Therefore, careful monitoring of the effect of exosomal HOTTIP on angiogenesis is demanded.

3.1.4. LINC02418

A few studies provided evidence that LINC02418 is an oncogenic lncRNA. LINC02418 positively regulates lung cancer cell proliferation while limiting apoptotic cell death by sponging miR-4677-3p [155,156]. In a similar fashion, LINC02418 constrains apoptosis via controlling the miR-34b-5p/BCL2 axis, promoting colorectal cancer progression [157]. In addition, LINC02418 is overexpressed in colorectal cancer tissues, cell lines, and serum exosomes, suggesting the likelihood of the origin of exosomal LINC02418 from cancer cells [132]. Furthermore, it was discerned that LINC02418 upregulates the level of MELK by disabling miR-1273g-3p to support proliferation and cell survival in cancer cells [132]. These findings demonstrate that exosomal LINC02418 from cancer cells can act as one of the bioactive molecules in recipient cancer cells (Figure 4 and Table 2). However, it should be noted that miR-1273g-3p has a stimulatory effect on cell proliferation, migration, and invasion in LoVo cells, a colorectal cancer cell line [158]. Such findings indicate the interaction of miR-1273g-3p with both oncogenes and tumor-suppressive genes, eliciting different functions depending on the cellular context.

3.1.5. MALAT1 and UCA1

Exosomal MALAT1 and UCA1 serve as pro-metastatic factors in colorectal cancer [133,140]. Exosomal MALAT1 from metastatic cells can be internalized into other cancer cells and raise their migratory and invasive capacity in vitro. Furthermore, liver and lung metastases of colorectal cancer are advanced by exosomal MALAT1 in vivo. Such pro-metastatic effect of MALAT1 is partly due to the activation of PI3K/AKT signaling via sponging miR-26a and miR-26b [133] (Figure 4 and Table 2).
UCA1 is highly expressed in colorectal cancer tissues, cell lines, and patients’ serum exosomes [140]. This suggests the possibility that exosomal UCA1 is derived from cancer cells. Further, it was found that cancer cells can gain metastatic potential following treatment with serum exosomes harboring UCA1. In this study, it was also proposed that UCA1 is capable of impelling metastasis by downmodulating and upmodulating miR-143 and myosin VI (MYO6) levels, respectively [140] (Figure 4 and Table 2). However, UCA1 exerts anti-migratory and -invasive roles in esophageal cancer [159], showing that the function of UCA1 is dissimilar, depending on cancer types.

3.2. Esophageal Cancer

3.2.1. AFAP1-AS1

AFAP1-AS1, a highly expressed lncRNA in multiple cancers, was discovered to promote cancer progression, stemness, as well as therapeutic resistance. AFAP1-AS1 possesses stimulatory effects on cell proliferation, migration, and invasion in thyroid cancer as a consequence of sequestering miR-204-3p [160]. In laryngeal cancer, AFAP1-AS1 potentiates stemness and cisplatin resistance by raising the level of recombination signal-binding protein for immunoglobulin Kappa J region (RBPJ) [161].
Besides this, a profuse amount of AFAP1-AS1 was detected in exosomes unleashed by M2 macrophages. Exosomal AFAP1-AS1 can enter esophageal cancer cells and interact with miR-26a to promote the level of cyclic AMP-responsive element-binding protein 2 (CREB2, also known as activating transcription factor 2 (ATF2)), thereby boosting migration/invasion in vitro and lung metastasis in vivo [114] (Figure 3 and Table 2).

3.2.2. FAM225A and FMR1-AS1

Neuropilin and tolloid-like 2 (NETO2) activates the PI3K/AKT signaling-mediated NF-κB/SNAI1 axis, triggering the EMT process [162]. The silencing of NETO2 can induce apoptosis, along with an increase in cleaved caspase-3 and a decrease in BCL2 [163]. In addition, forkhead box P1 (FOXP1) supports cell survival via transcriptionally repressing the expression of proapoptotic genes, such as BCL2 interacting killer (BIK) [164]. Furthermore, the level of EMT-related genes, including SNAI1, is positively affected by FOXP1 overexpression [165]. Both NETO2 and FOXP1 are targets of miR-206 and are upregulated by FAM225A in esophageal cancer. FOXP1 can increase FAM225A levels, indicating a positive feedback loop between these molecules [121]. FAM225A is located in exosomes from cancer cells. Therefore, exosomal FAM225A can be transferred to other cancer cells, inciting cell survival and EMT [121] (Figure 4 and Table 2).
FMR1-AS1 is a highly expressed lncRNA and is sorted into exosomes in esophageal cancer [122]. This exosomal lncRNA is transcriptionally induced by NF-κB, which, in turn, activates NF-κB-MYC signaling by binding to toll-like receptor 7 (TLR7). Thus, FMR1-AS1 exerts anti-apoptotic and pro-invasive effects and strengthens stemness in recipient cancer cells. In vivo observation also indicated that exosomal FMR1-AS1 fuels cancer growth [122] (Figure 4 and Table 2).

3.2.3. LINC01711 and ZFAS1

LINC01711 is upregulated in esophageal cancer tissues and several cell lines. In addition, this lncRNA can be contained in exosomes produced by cancer cells, suggesting that exosomal LINC01711 can play an oncogenic role [131]. It has been shown that exosomal LINC01711 dampens the activity of miR-326, which targets fascin actin-bundling protein 1 (FSCN1), in recipient cancer cells, hence expediting proliferation, migration, and invasion [131] (Figure 4 and Table 2). Another study showed the positive correlation between LINC01711 and TGFB1 expression [166]. Thus, miR-326 activity may be modulated by the TGFB1/LINC01711 axis. FSCN1 is markedly increased in diverse cancers and affects various cellular processes, such as invasion, EMT, metastasis, as well as drug resistance [167,168,169]. Therefore, it is worth considering LINC01711 as a prospective target for esophageal cancer treatment.
In esophageal cancer, exosomal ZFAS1 is taken up by other surrounding cancer cells and exerts a positive influence on proliferation, migration, and invasion via the miR-124/STAT3 axis [142] (Figure 4 and Table 2). ZFAS1 has been known to abrogate tumor-suppressive functions of numerous miRNAs in gastrointestinal cancers. By targeting miR-150, ZFAS1 upregulates MMP14, MMP16, and zinc finger E-box-binding homeobox 1 (ZEB1) expression, consequently increasing the occurrence of lung metastasis in hepatocellular carcinomas [170]. ZFAS1 also invalidates miR-150 activity and facilitates angiogenesis in colorectal cancer. Of note, ZFAS1 enhances tube formation of HUVECs [171]. Exosomal ZFAS1 may therefore contribute to angiogenesis by affecting the biological traits of nearby endothelial cells in esophageal cancer.

3.2.4. PCAT1 and POU3F3

PCAT1 and POU3F3 have been regarded as oncogenic lncRNAs. PCAT1 physically binds to enhancer of Zeste 2 polycomb repressive complex 2 subunit (EZH2) and, thus, epigenetically silences PTEN expression, leading to cisplatin resistance in gastric cancer [172]. In hepatocellular carcinomas, PCAT1 silencing downregulates the level of high mobility group box 1 (HMGB1) and incapacitates the invasive and migratory potential [173]. In a similar manner, the degree of apoptosis and EMT process is attenuated in POU3F3-silenced colorectal cancer cells [174].
In esophageal cancer, both PCAT1 and POU3F3 regulate drug sensitivity [134,136]. PCAT1 was validated to sponge miR-326, which has an inhibitory action on AKT. Downregulation of PCAT1 induces G2/M cell cycle arrest and turns cancer cells more sensitive to paclitaxel [134]. This lncRNA is present in exosomes from cancer cells [134], demonstrating its potential to cause paclitaxel resistance in recipient cancer cells (Figure 4 and Table 2).
POU3F3 is also encompassed by exosomes generated from cancer cells and delivered to fibroblasts. POU3F3 ultimately accelerates the conversion of normal fibroblasts (NF) into CAFs, although the mechanisms of POU3F3-mediated fibroblast activation remain unclear. CAFs are then responsible for cisplatin resistance by secreting IL6 [136] (Figure 4 and Table 2). POU3F3 has been shown to upregulate the expression of rho-associated coiled-coil containing protein kinase (ROCK) [175]. ROCK is known to activate YAP1 [176], which stimulates NF-CAF transition [177]. For this reason, POU3F3 may regulate fibroblast activation via the ROCK/YAP1 axis.

3.3. Gastric Cancer

3.3.1. CEBPA-AS1

CEBPA-AS1 deactivates Notch signaling via sponging of oncogenic miR-10b-5p, and it increases apoptotic cell death in osteosarcomas [178]. By contrast, CEBPA-AS1 is highly expressed in exosomes from gastric cancer cells and suppresses apoptotic cell death in recipient cancer cells [117] (Figure 4 and Table 2). CEBPA-AS1 partakes in BCL2 upregulation in oral squamous cell carcinomas [179]. Although it is required to unravel the mechanisms underlying the oncogenic role of CEBPA-AS1 in gastric cancer, apoptosis may nonetheless be regulated by the CEBPA-AS1/BCL2 axis in gastric cancer as well.

3.3.2. FRLnc1 and PCGEM1

Exosomal FRLnc1 from gastric cancer cells can effectively augment cell proliferation, viability, and migration in receiver cancer cells [123] (Figure 4 and Table 2). Although direct targets of FRLnc1 remain to be identified, FRLnc1 is known to upregulate the level of TGFB1 and TWIST, favoring migration in vitro and lung metastasis in vivo in gastric cancer [180]. In addition, FRLnc1 can be induced by FOXM1 [180], and CCAL is capable of upmodulating FOXM1 levels (Section 3.1.1). Thus, CCAL may increase FRLnc1 expression in gastric cancer, resulting in an abundance of FRLnc1 in exosomes.
PCGEM1 stabilizes SNAI1 proteins via physically interacting with them, serving as a pro-migratory and -invasive lncRNA in gastric cancer [135]. Hypoxic cancer cells are sources of plentiful amounts of exosomal PCGEM1, which can be internalized into normoxic cancel cells. Exosomal PCGEM1 leads to an increase in SNAI1 levels and expedites migration and invasion in normoxic cancer cells [135] (Figure 3 and Table 2). Evidence from another study suggested that PCGEM1 also improves the invasive ability of gastric cancer cells by increasing the level of prolyl 4-hydroxylase subunit alpha 2 (P4HA2) [181]. Accordingly, PCGEM1 may transcriptionally and post-transcriptionally regulate SNAI1 expression, since P4HA2 transcriptionally induces SNAI1 [182].

3.3.3. HOTTIP

As mentioned in Section 3.1.3, HOTTIP regulates cellular factors and signaling linked to therapeutic resistance. In gastric cancer, cisplatin resistance in recipient cells is instigated by exosomal HOTTIP secreted from cisplatin-resistant cells. Mechanistically, exosomal HOTTIP augments the expression of high mobility group AT-hook 1 (HMGA1) via nullifying miR-218 activity [125] (Figure 3 and Table 2). In addition to cisplatin, HMGA1 contributes to cellular resistance to paclitaxel, 5-FU, and gefitinib [183,184,185]. Therefore, targeting HOTTIP may beneficially improve the effectiveness of various kinds of anti-cancer drugs.

3.3.4. LINC01559

Research-based evidence demonstrated the role of LINC01559 as an oncogenic lncRNA in gastrointestinal cancers. In pancreatic cancer, LINC01559 interacts with and enhances the transcriptional activity of YAP, boosting cell proliferation and migration [186]. Through sponging miR-6783-3p, LINC01559 renders hepatocellular carcinoma insensitive to oxaliplatin [187]. In addition, LINC01559 stabilizes ZEB1 mRNA via interacting with IGF2BP2 and acts as an EMT-promoting lncRNA in gastric cancer. ZEB1, in turn, transcriptionally activates LINC01559 expression, indicating a LINC01559–ZEB1 feedback loop [188].
Moreover, it was noticed that exosomes from mesenchymal stem cells convey LINC01559 to gastric cancer cells [130]. Exosomal LINC01559 counteracts the negative effect of miR-1343-3p on phosphoglycerate kinase 1 (PGK1) and epigenetically inhibits PTEN expression. By regulating PGK1 and PTEN levels, exosomal LINC01559 can activate PI3K/AKT signaling and promote cell proliferation, migration, and stemness in gastric cancer cells [130] (Figure 3 and Table 2).

3.3.5. SPRY4-IT1

SPRY4-IT1 has been proven to serve as either a tumor-suppressive or an oncogenic lncRNA. SPRY4-IT1 is downregulated in cisplatin-resistant lung cancer cells and takes part in the repression of EMT. Overexpression of SPRY4-IT1 reinforces cisplatin-induced apoptosis in resistant cells [189]. Conversely, EMT can be triggered by SPRY4-IT1 in cervical cancer, colorectal cancer, and hepatocellular carcinomas [190,191,192]. What is more, SPRY4-IT1 is transcriptionally activated by NF-κB and promotes staufen-mediated degradation of transcription elongation factor B subunit 1 (TCEB1) mRNA to upregulate HIF-1α levels [193]. Thus, it is probable that SPRY4-IT1 can control the expression of other ncRNAs, such as circ_ZNF91, which is regulated by HIF-1α (see Section 2.5.3 about circ_ZNF91).
Furthermore, SPRY4-IT1 is enriched in gastric cancer tissues, cancer cell lines, and serum exosomes [138]. Hence, exosomal SPRY4-IT1 can be released from cancer cells and regulate the biological properties of other cancer cells. SPRY4-IT1 functionally provokes cell proliferation, migration, and stemness via regulating the miR-101-3p/AMP-activated protein kinase (AMPK) axis [138] (Figure 4 and Table 2).

3.4. Hepatocellular Carcinoma

3.4.1. ASMTL-AS1

ASMTL-AS1 has a dual role in cancer. Overexpression of ASMTL-AS1 stabilizes spermidine/spermine N1-acetyltransferase 1 (SAT1) mRNA, retarding the growth of lung cancer along with ferroptosis [194]. ASMTL-AS1 impairs WNT/β-catenin signaling, restraining breast cancer growth in vivo [195]. By contrast, ASMTL-AS1 exacerbates osteosarcoma progression through the upmodulation of ADAM metallopeptidase domain 9 (ADAM9) expression [196].
In hepatocellular carcinoma, ASMTL-AS1 is upregulated in cancer tissues and further increased in residual tissues following insufficient radiofrequency ablation (RFA) [115]. Such an expression pattern of ASMTL-AS1 implies that this lncRNA may contribute to cancer recurrence. Indeed, ASMTL-AS1 is transcriptionally elevated by MYC and increases Nemo-like kinase (NLK) expression via rendering miR-342-3p ineffective, fundamentally reinforcing malignant phenotypes, such as EMT and metastasis [115]. Compared to its levels in non-treated cells, ASMTL-AS1 levels are increased in residual cancer cells following heat treatment mimicking insufficient RFA. In addition, ASMTL-AS1 can be enclosed by exosomes and conveyed from residual cells into other cancer cells [115]. These observations demonstrate the contribution of exosomal ASMTL-AS1 to cancer recurrence by increasing NLK signaling (Figure 3 and Table 2).

3.4.2. DLX6-AS1 and TUC339

Exosomal lncRNAs from hepatocellular carcinoma cells are accountable for M2 polarization, following an uptake by macrophages. Coculturing cancer cells with M2 macrophages stimulated by exosomal DLX6-AS1 promotes migration, invasion, and EMT in vitro. Furthermore, by activating M2 polarization, exosomal DLX6-AS1 triggers lung metastasis in vivo. Exosomal DLX6-AS1 exerts its function by upregulating C-X-C motif chemokine ligand 17 (CXCL17), which is targeted by miR-15a-5p [119] (Figure 4 and Table 2). Despite DLX6-AS1’s oncogenic ability, the knockdown of this lncRNA activates autophagy [197]. Since autophagy can be considered as the pro-survival pathway [198], double inhibition of DLX6-AS1 and autophagy may efficiently suppress cancer and synergistically improve the anti-cancer effect of conventional treatment.
In the case of TUC339, more studies are needed to unveil how TUC339 controls M2 polarization and what miRNAs and proteins this lncRNA interacts with once transferred to macrophages. Nonetheless, it was observed that exosomal TUC339 from cancer cells is required for M2 polarization and an increase in pro-inflammatory cytokines [139]. These results suggest that the downregulation of exosomal TUC339 can conceivably control cancer progression (Figure 4 and Table 2).

3.4.3. FAM72D-3

FAM72D-3 is one of the upregulated lncRNAs in serum exosomes from patients with hepatocellular carcinomas. FAM72D-3 silencing increases apoptotic cell death [120]. Thus, it is conceivable that the transfer of cancer cell-derived exosomal FAM72D-3 to other cancer cells enables them to escape apoptosis. In addition, miR-5787 levels are elevated in FAM72D-3-silencing cells, suggesting miR-5787 as a possible downstream effector of FAM72D-3 [120]. It is important to define whether FAM72D-3 directly regulates miR-5787 or not and if miR-5787 is an oncogenic or a tumor-suppressive factor (Figure 4 and Table 2).

3.4.4. LINC00161

LINC00161 upregulates interferon-induced protein with tetratricopeptide repeats 2 (IFIT2), a cell death-promoting factor, augmenting cisplatin-induced apoptosis in osteosarcoma cells [199]. On the other hand, LINC00161 is one of the highly expressed lncRNAs in cisplatin-resistant ovarian cancer cells and causes cisplatin resistance by upregulating ERK [200]. Besides, intercellular transport of LINC00161 from cancer cells to endothelial cells was noticed in hepatocellular carcinomas [128]. Exosomal LINC00161 can increase the level of ROCK by binding to miR-590-3p in endothelial cells, facilitating proliferation, migration, and tube formation. The silencing of LINC00161 markedly inhibits angiogenesis and metastasis in vivo [128] (Figure 4 and Table 2).

3.5. Pancreatic Cancer

3.5.1. CCAT1

In addition to the resistance-associated role of HMGA1 (Section 3.3.3), HMGA1 is involved in angiogenesis. For instance, HMGA1 can increase the transcriptional activity of FOXM1 on angiogenesis-related genes, such as VEGF [201]. In addition, HMGA1 is able to interact with HIF-1, thus mediating a hypoxia-induced increase in VEGF [202]. A recent report demonstrated that CCAT1 is released from pancreatic cancer cells through exosomes and positively modulates the level of HMGA1, a miR-138-5p target, in endothelial cells [116]. The effect of CCAT1 on migration and tube formation of endothelial cells is antagonized by miR-138-5p overexpression [116]. These results suggest that exosomal CCAT1 may regulate autocrine VEGF signaling via the miR-138-5p/HMGA1/VEGF axis in endothelial cells (Figure 4 and Table 2).

3.5.2. HULC

Identification of lncRNAs regulated by TGFB1, an EMT-promoting factor, showed that HULC is among the TGFB1-induced lncRNAs in pancreatic cancer cells. Both intracellular and intra-exosomal levels of HULC are upregulated by TGFB1 [126]. Treatment with exosomes derived from donor cancer cells increases the expression of HULC and EMT markers, such as vimentin, in recipient cancer cells and promotes invasion and migration. These tendencies are further reinforced by the treatment with exosomes derived from TGFB1-treated cancer cells. In addition, it was found that the effect of HULC is due to its suppressive action on miR-133b, an EMT-inhibiting miRNA [126] (Figure 4 and Table 2).

3.5.3. LINC01133

Disparate functions of LINC01133 depending on cancer types have been reported. LINC01133 hampers gastric and bladder cancer progression via somatostatin upregulation and WNT signaling inactivation, respectively [203,204]. Consistently, by suppressing SRY-box transcription factor 4 (SOX4) expression, LINC01133 is capable of inhibiting metastasis in breast cancer [205]. On the contrary, LINC01133 facilitates EMT via enhancing SNAI1 levels and activates STAT3 signaling in hepatocellular carcinoma [206]. In ovarian cancer, LINC01133 upregulates tumor protein D52 (TPD52) levels through sharing the same binding sites with miR-495-3p, acting as a pro-metastatic factor [207]. It was also identified that LINC01133 is transactivated by CCAAT/enhancer-binding protein β (CEBPB) and positively modulates cell proliferation via activating cyclin G1 expression in pancreatic cancer [208].
Moreover, LINC01133 is highly expressed in pancreatic cancer cells as well as in their secreted exosomes. LINC01133 interacts with EZH2 to silence the expression of axis inhibition protein 2 (AXIN2), activating β-catenin and promoting cell survival, proliferation, and EMT [129]. In this study, periostin (POSTN) was discovered to increase LINC01133 via the epidermal growth factor receptor (EGFR)/MYC axis and promote exosome secretion in pancreatic cancer cells [129]. These findings suggest that exosomal LINC01133 augmented by POSTN in donor cells can affect the progression of recipient cells via the AXIN2/β-catenin axis (Figure 4 and Table 2).

3.5.4. UCA1

Angiomotin-like 2 (AMOTL2) activates the SRC/ERK signaling pathway, playing a critical role in migration and proliferation of endothelial cells during angiogenesis [209]. More recently, exosomal UCA1 was proven to be released from hypoxic pancreatic cancer cells and transported to endothelial cells, in which UCA1 relieves the activity of miR-96-5p on AMOTL2. Exosomal UCA1 secreted from hypoxic cancer cells reinforces cancer growth and angiogenesis in vivo [141] (Figure 3 and Table 2).

4. Conclusions

Growing evidence introduced here shows that exosomal circRNAs and lncRNAs are involved in the intercellular communication between a wide range of cell types. Knockdown of a single exosomal ncRNA sufficiently blocks cancer progression and aggressiveness; therefore, targeting ncRNAs can be a reasonable option for gastrointestinal cancer treatment.
Nonetheless, it is critical to consider the characteristic features of these ncRNAs. These include the differences in function according to the type of cancer and cell. Inhibition of appropriately selected ncRNAs that are specifically relevant to the cancer type is required. Furthermore, to avoid side effects on normal cells, cancer-specific suppression of oncogenic ncRNA is essential. Additionally, even targeting ncRNAs themselves may hardly be free from therapeutic resistance. Therefore, it is attractive to explore innovative combination therapy with ncRNA inhibition and other anti-cancer treatments for pronouncedly controlling resistance problems.
In addition to the inhibition of exosome cargo, restraint of exosome secretion has therapeutic benefits. However, pharmacological agents, such as GW4869, are non-specific inhibitors of exosome-mediated intercellular dialogues and have undesirable effects [210,211]. Besides this, it has been found that exosome uptake can be controlled by cellular factors such as advanced glycosylation end-product specific receptor (AGER) [212]. Therefore, identifying genes involved in the secretion and uptake of exosomes will open the way for more specific inhibition of the cancer-supporting activity of exosomes through gene knockdown approaches.
Overall, an advanced understanding of the nature of ncRNAs and the mechanism underpinning the exosome itinerary will pave the way for overcoming gastrointestinal cancer.

Author Contributions

Conceptualization, B.D.Y. and J.K.P.; literature review and visualization, B.D.Y., Y.J.C., S.W.S., G.A.C., F.C.B.B., D.M., T.-J.O., H.J.K., S.Y.C. and J.K.P.; writing—original draft preparation, B.D.Y., Y.J.C. and J.K.P.; writing—review and editing, J.K.P. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant from the National Research Foundation of Korea (NRF), grant funded by the Korean government (MSIT) (2019R1A2C1089710) (J.K.P.) (2019R1A2B5B02070524) (H.J.K.), and the Basic Science Research Program through the National Research Foundation of Korea (NRF), funded by the Ministry of Education (2019R1A6A1A11036849) (S.Y.C.). Furthermore, this research was supported by the BioGreen21 Agri-Tech Innovation Program (Project No. PJ015710) (T.-J.O.), Rural Development Administration, Republic of Korea.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Arnold, M.; Abnet, C.C.; Neale, R.E.; Vignat, J.; Giovannucci, E.L.; McGlynn, K.A.; Bray, F. Global burden of 5 major types of gastrointestinal cancer. Gastroenterology 2020, 159, 335–349.e315. [Google Scholar] [CrossRef] [PubMed]
  2. Jiang, D.M.; Chan, K.K.W.; Jang, R.W.; Booth, C.; Liu, G.; Amir, E.; Mason, R.; Everest, L.; Elimova, E. Anticancer drugs approved by the food and drug administration for gastrointestinal malignancies: Clinical benefit and price considerations. Cancer Med. 2019, 8, 1584–1593. [Google Scholar] [CrossRef]
  3. Singh, D.; Dheer, D.; Samykutty, A.; Shankar, R. Antibody drug conjugates in gastrointestinal cancer: From lab to clinical development. J. Control. Release 2021, 340, 1–34. [Google Scholar] [CrossRef] [PubMed]
  4. Senthebane, D.A.; Rowe, A.; Thomford, N.E.; Shipanga, H.; Munro, D.; Mazeedi, M.; Almazyadi, H.A.M.; Kallmeyer, K.; Dandara, C.; Pepper, M.S.; et al. The role of tumor microenvironment in chemoresistance: To survive, keep your enemies closer. Int. J. Mol. Sci. 2017, 18, 1586. [Google Scholar] [CrossRef] [PubMed]
  5. Roma-Rodrigues, C.; Mendes, R.; Baptista, P.V.; Fernandes, A.R. Targeting tumor microenvironment for cancer therapy. Int. J. Mol. Sci. 2019, 20, 840. [Google Scholar] [CrossRef] [Green Version]
  6. Tirpe, A.A.; Gulei, D.; Ciortea, S.M.; Crivii, C.; Berindan-Neagoe, I. Hypoxia: Overview on hypoxia-mediated mechanisms with a focus on the role of hif genes. Int. J. Mol. Sci. 2019, 20, 6140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Yokoi, A.; Ochiya, T. Exosomes and extracellular vesicles: Rethinking the essential values in cancer biology. Semin. Cancer Biol. 2021, 74, 79–91. [Google Scholar] [CrossRef] [PubMed]
  8. Moeng, S.; Son, S.W.; Lee, J.S.; Lee, H.Y.; Kim, T.H.; Choi, S.Y.; Kuh, H.J.; Park, J.K. Extracellular vesicles (evs) and pancreatic cancer: From the role of evs to the interference with ev-mediated reciprocal communication. Biomedicines 2020, 8, 267. [Google Scholar] [CrossRef]
  9. Pessolano, E.; Belvedere, R.; Bizzarro, V.; Franco, P.; Marco, I.; Porta, A.; Tosco, A.; Parente, L.; Perretti, M.; Petrella, A. Annexin a1 may induce pancreatic cancer progression as a key player of extracellular vesicles effects as evidenced in the in vitro mia paca-2 model system. Int. J. Mol. Sci. 2018, 19, 3878. [Google Scholar] [CrossRef] [Green Version]
  10. Wei, Q.T.; Liu, B.Y.; Ji, H.Y.; Lan, Y.F.; Tang, W.H.; Zhou, J.; Zhong, X.Y.; Lian, C.L.; Huang, Q.Z.; Wang, C.Y.; et al. Exosome-mediated transfer of mif confers temozolomide resistance by regulating timp3/pi3k/akt axis in gliomas. Mol. Ther. Oncolytics 2021, 22, 114–128. [Google Scholar] [CrossRef]
  11. Richards, K.E.; Zeleniak, A.E.; Fishel, M.L.; Wu, J.; Littlepage, L.E.; Hill, R. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene 2017, 36, 1770–1778. [Google Scholar] [CrossRef] [Green Version]
  12. Lee, H.Y.; Son, S.W.; Moeng, S.; Choi, S.Y.; Park, J.K. The role of noncoding rnas in the regulation of anoikis and anchorage-independent growth in cancer. Int. J. Mol. Sci. 2021, 22, 627. [Google Scholar] [CrossRef]
  13. Son, S.W.; Song, M.G.; Yun, B.D.; Park, J.K. Noncoding rnas associated with therapeutic resistance in pancreatic cancer. Biomedicines 2021, 9, 263. [Google Scholar] [CrossRef] [PubMed]
  14. Son, S.W.; Yun, B.D.; Song, M.G.; Lee, J.K.; Choi, S.Y.; Kuh, H.J.; Park, J.K. The hypoxia-long noncoding rna interaction in solid cancers. Int. J. Mol. Sci. 2021, 22, 7261. [Google Scholar] [CrossRef] [PubMed]
  15. Fontemaggi, G.; Turco, C.; Esposito, G.; Di Agostino, S. New molecular mechanisms and clinical impact of circrnas in human cancer. Cancers 2021, 13, 3154. [Google Scholar] [CrossRef]
  16. Nisar, S.; Bhat, A.A.; Singh, M.; Karedath, T.; Rizwan, A.; Hashem, S.; Bagga, P.; Reddy, R.; Jamal, F.; Uddin, S.; et al. Insights into the role of circrnas: Biogenesis, characterization, functional, and clinical impact in human malignancies. Front. Cell Dev. Biol. 2021, 9, 617281. [Google Scholar] [CrossRef]
  17. Song, S.; Yu, W.; Lin, S.; Zhang, M.; Wang, T.; Guo, S.; Wang, H. Lncrna adpgk-as1 promotes pancreatic cancer progression through activating zeb1-mediated epithelial-mesenchymal transition. Cancer Biol. Ther. 2018, 19, 573–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Wang, D.; Cui, L.; Yang, Q.; Wang, J. Circular rna circzfpm2 promotes epithelial-mesenchymal transition in endometriosis by regulating mir-205-5p/zeb1 signalling pathway. Cell Signal. 2021, 87, 110145. [Google Scholar] [CrossRef] [PubMed]
  19. Limb, C.; Liu, D.S.K.; Veno, M.T.; Rees, E.; Krell, J.; Bagwan, I.N.; Giovannetti, E.; Pandha, H.; Strobel, O.; Rockall, T.A.; et al. The role of circular rnas in pancreatic ductal adenocarcinoma and biliary-tract cancers. Cancers 2020, 12, 3250. [Google Scholar] [CrossRef] [PubMed]
  20. Holdt, L.M.; Stahringer, A.; Sass, K.; Pichler, G.; Kulak, N.A.; Wilfert, W.; Kohlmaier, A.; Herbst, A.; Northoff, B.H.; Nicolaou, A.; et al. Circular non-coding rna anril modulates ribosomal rna maturation and atherosclerosis in humans. Nat. Commun. 2016, 7, 12429. [Google Scholar] [CrossRef] [Green Version]
  21. Zhang, X.; Wang, W.; Zhu, W.; Dong, J.; Cheng, Y.; Yin, Z.; Shen, F. Mechanisms and functions of long non-coding rnas at multiple regulatory levels. Int. J. Mol. Sci. 2019, 20, 5573. [Google Scholar] [CrossRef] [Green Version]
  22. Jiang, Z.; Hou, Z.; Li, L.; Liu, W.; Yu, Z.; Chen, S. Exosomal circepb41l2 serves as a sponge for mir-21-5p and mir-942-5p to suppress colorectal cancer progression by regulating the pten/akt signalling pathway. Eur. J. Clin. Investig. 2021, 51, e13581. [Google Scholar] [CrossRef] [PubMed]
  23. Hon, K.W.; Ab-Mutalib, N.S.; Abdullah, N.M.A.; Jamal, R.; Abu, N. Extracellular vesicle-derived circular rnas confers chemoresistance in colorectal cancer. Sci. Rep. 2019, 9, 16497. [Google Scholar] [CrossRef] [Green Version]
  24. Zhao, K.; Cheng, X.; Ye, Z.; Li, Y.; Peng, W.; Wu, Y.; Xing, C. Exosome-mediated transfer of circ_0000338 enhances 5-fluorouracil resistance in colorectal cancer through regulating microrna 217 (mir-217) and mir-485-3p. Mol. Cell Biol. 2021, 41, e00517-20. [Google Scholar] [CrossRef]
  25. Zhang, N.; Lu, C.; Chen, L. Mir-217 regulates tumor growth and apoptosis by targeting the mapk signaling pathway in colorectal cancer. Oncol. Lett. 2016, 12, 4589–4597. [Google Scholar] [CrossRef] [Green Version]
  26. Su, S.; Lu, W.; Liu, J.; Li, L.; Liu, L.; Li, X.; Ye, D.; Yu, Z. Circ_0007031 silencing inhibits cell proliferation and induces cell apoptosis via downregulating melk at a mir-485-3p-dependent way in colorectal cancer. Biochem. Genet. 2021, 1–22. [Google Scholar] [CrossRef]
  27. Blondy, S.; David, V.; Verdier, M.; Mathonnet, M.; Perraud, A.; Christou, N. 5-fluorouracil resistance mechanisms in colorectal cancer: From classical pathways to promising processes. Cancer Sci. 2020, 111, 3142–3154. [Google Scholar] [CrossRef] [PubMed]
  28. Choi, S.; Ku, J.L. Resistance of colorectal cancer cells to radiation and 5-fu is associated with melk expression. Biochem. Biophys. Res. Commun. 2011, 412, 207–213. [Google Scholar] [CrossRef] [PubMed]
  29. Apicella, M.; Giannoni, E.; Fiore, S.; Ferrari, K.J.; Fernandez-Perez, D.; Isella, C.; Granchi, C.; Minutolo, F.; Sottile, A.; Comoglio, P.M.; et al. Increased lactate secretion by cancer cells sustains non-cell-autonomous adaptive resistance to met and egfr targeted therapies. Cell Metab. 2018, 28, 848–865 e846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Lu, W.Q.; Hu, Y.Y.; Lin, X.P.; Fan, W. Knockdown of pkm2 and gls1 expression can significantly reverse oxaliplatin-resistance in colorectal cancer cells. Oncotarget 2017, 8, 44171–44185. [Google Scholar] [CrossRef] [Green Version]
  31. Wang, X.; Zhang, H.; Yang, H.; Bai, M.; Ning, T.; Deng, T.; Liu, R.; Fan, Q.; Zhu, K.; Li, J.; et al. Exosome-delivered circrna promotes glycolysis to induce chemoresistance through the mir-122-pkm2 axis in colorectal cancer. Mol. Oncol. 2020, 14, 539–555. [Google Scholar] [CrossRef]
  32. Zang, R.; Qiu, X.; Song, Y.; Wang, Y. Exosomes mediated transfer of circ_0000337 contributes to cisplatin (cddp) resistance of esophageal cancer by regulating jak2 via mir-377-3p. Front. Cell Dev. Biol. 2021, 9, 673237. [Google Scholar] [CrossRef] [PubMed]
  33. Yang, H.; Zhang, H.; Yang, Y.; Wang, X.; Deng, T.; Liu, R.; Ning, T.; Bai, M.; Li, H.; Zhu, K.; et al. Hypoxia induced exosomal circrna promotes metastasis of colorectal cancer via targeting gef-h1/rhoa axis. Theranostics 2020, 10, 8211–8226. [Google Scholar] [CrossRef]
  34. Zhong, Y.; Wang, D.; Ding, Y.; Tian, G.; Jiang, B. Circular rna circ_0032821 contributes to oxaliplatin (oxa) resistance of gastric cancer cells by regulating sox9 via mir-515-5p. Biotechnol. Lett. 2021, 43, 339–351. [Google Scholar] [CrossRef] [PubMed]
  35. 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]
  36. Yu, Y.; Bian, L.; Liu, R.; Wang, Y.; Xiao, X. Circular rna hsa_circ_0061395 accelerates hepatocellular carcinoma progression via regulation of the mir-877-5p/pik3r3 axis. Cancer Cell Int. 2021, 21, 10. [Google Scholar] [CrossRef] [PubMed]
  37. Wang, P.; Sun, Y.; Yang, Y.; Chen, Y.; Liu, H. Circ_0067835 knockdown enhances the radiosensitivity of colorectal cancer by mir-296-5p/igf1r axis. Onco Targets Ther. 2021, 14, 491–502. [Google Scholar] [CrossRef] [PubMed]
  38. Shi, H.; Huang, S.; Qin, M.; Xue, X.; Guo, X.; Jiang, L.; Hong, H.; Fang, J.; Gao, L. Exosomal circ_0088300 derived from cancer-associated fibroblasts acts as a mir-1305 sponge and promotes gastric carcinoma cell tumorigenesis. Front. Cell Dev. Biol. 2021, 9, 676319. [Google Scholar] [CrossRef]
  39. Yang, K.; Zhang, J.; Bao, C. Exosomal circeif3k from cancer-associated fibroblast promotes colorectal cancer (crc) progression via mir-214/pd-l1 axis. BMC Cancer 2021, 21, 933. [Google Scholar] [CrossRef]
  40. Lai, Z.; Wei, T.; Li, Q.; Wang, X.; Zhang, Y.; Zhang, S. Exosomal circfblim1 promotes hepatocellular carcinoma progression and glycolysis by regulating the mir-338/lrp6 axis. Cancer Biother. Radiopharm. 2020. [Google Scholar] [CrossRef] [PubMed]
  41. Li, J.; Li, Z.; Jiang, P.; Peng, M.; Zhang, X.; Chen, K.; Liu, H.; Bi, H.; Liu, X.; Li, X. Circular rna iars (circ-iars) secreted by pancreatic cancer cells and located within exosomes regulates endothelial monolayer permeability to promote tumor metastasis. J. Exp. Clin. Cancer Res. 2018, 37, 177. [Google Scholar] [CrossRef] [Green Version]
  42. Li, L.; Jiang, Z.; Zou, X.; Hao, T. Exosomal circ_ift80 enhances tumorigenesis and suppresses radiosensitivity in colorectal cancer by regulating mir-296-5p/msi1 axis. Cancer Manag. Res. 2021, 13, 1929–1941. [Google Scholar] [CrossRef]
  43. Liu, D.; Kang, H.; Gao, M.; Jin, L.; Zhang, F.; Chen, D.; Li, M.; Xiao, L. Exosome-transmitted circ_mmp2 promotes hepatocellular carcinoma metastasis by upregulating mmp2. Mol. Oncol. 2020, 14, 1365–1380. [Google Scholar] [CrossRef]
  44. Yu, L.; Xie, J.; Liu, X.; Yu, Y.; Wang, S. Plasma exosomal circnek9 accelerates the progression of gastric cancer via mir-409-3p/map7 axis. Dig. Dis. Sci. 2021, 66, 4274–4289. [Google Scholar] [CrossRef]
  45. Hui, C.; Tian, L.; He, X. Circular rna circnhsl1 contributes to gastric cancer progression through the mir-149-5p/ywhaz axis. Cancer Manag. Res. 2020, 12, 7117–7130. [Google Scholar] [CrossRef] [PubMed]
  46. Shang, A.; Gu, C.; Wang, W.; Wang, X.; Sun, J.; Zeng, B.; Chen, C.; Chang, W.; Ping, Y.; Ji, P.; et al. Exosomal circpacrgl promotes progression of colorectal cancer via the mir-142-3p/mir-506-3p- tgf-beta1 axis. Mol. Cancer 2020, 19, 117. [Google Scholar] [CrossRef] [PubMed]
  47. Li, Z.; Yanfang, W.; Li, J.; Jiang, P.; Peng, T.; Chen, K.; Zhao, X.; Zhang, Y.; Zhen, P.; Zhu, J.; et al. Tumor-released exosomal circular rna pde8a promotes invasive growth via the mir-338/macc1/met pathway in pancreatic cancer. Cancer Lett. 2018, 432, 237–250. [Google Scholar] [CrossRef]
  48. Wang, S.; Ping, M.; Song, B.; Guo, Y.; Li, Y.; Jia, J. Exosomal circprrx1 enhances doxorubicin resistance in gastric cancer by regulating mir-3064-5p/ptpn14 signaling. Yonsei Med. J. 2020, 61, 750–761. [Google Scholar] [CrossRef] [PubMed]
  49. Yao, W.; Guo, P.; Mu, Q.; Wang, Y. Exosome-derived circ-pvt1 contributes to cisplatin resistance by regulating autophagy, invasion, and apoptosis via mir-30a-5p/yap1 axis in gastric cancer cells. Cancer Biother. Radiopharm. 2021, 36, 347–359. [Google Scholar] [CrossRef]
  50. 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] [PubMed]
  51. Zhang, T.; Jing, B.; Bai, Y.; Zhang, Y.; Yu, H. Circular rna circtmem45a acts as the sponge of microrna-665 to promote hepatocellular carcinoma progression. Mol. Ther. Nucleic Acids 2020, 22, 285–297. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, P.F.; Gao, C.; Huang, X.Y.; Lu, J.C.; Guo, X.J.; Shi, G.M.; Cai, J.B.; Ke, A.W. Cancer cell-derived exosomal circuhrf1 induces natural killer cell exhaustion and may cause resistance to anti-pd1 therapy in hepatocellular carcinoma. Mol. Cancer 2020, 19, 110. [Google Scholar] [CrossRef] [PubMed]
  53. Li, Y.; Zang, H.; Zhang, X.; Huang, G. Exosomal circ-znf652 promotes cell proliferation, migration, invasion and glycolysis in hepatocellular carcinoma via mir-29a-3p/gucd1 axis. Cancer Manag. Res. 2020, 12, 7739–7751. [Google Scholar] [CrossRef] [PubMed]
  54. Zeng, Z.; Zhao, Y.; Chen, Q.; Zhu, S.; Niu, Y.; Ye, Z.; Hu, P.; Chen, D.; Xu, P.; Chen, J.; et al. Hypoxic exosomal hif-1alpha-stabilizing circznf91 promotes chemoresistance of normoxic pancreatic cancer cells via enhancing glycolysis. Oncogene 2021, 40, 5505–5517. [Google Scholar] [CrossRef]
  55. Cao, J.; Yang, T.; Tang, D.; Zhou, F.; Qian, Y.; Zou, X. Increased expression of gef-h1 promotes colon cancer progression by rhoa signaling. Pathol. Res. Pract. 2019, 215, 1012–1019. [Google Scholar] [CrossRef] [PubMed]
  56. Liu, X.; Chen, H.; Xu, X.; Ye, M.; Cao, H.; Xu, L.; Hou, Y.; Tang, J.; Zhou, D.; Bai, Y.; et al. Insulin-like growth factor-1 receptor knockdown enhances radiosensitivity via the hif-1alpha pathway and attenuates atm/h2ax/53bp1 DNA repair activation in human lung squamous carcinoma cells. Oncol. Lett. 2018, 16, 1332–1340. [Google Scholar] [PubMed] [Green Version]
  57. de Araujo, P.R.; Gorthi, A.; da Silva, A.E.; Tonapi, S.S.; Vo, D.T.; Burns, S.C.; Qiao, M.; Uren, P.J.; Yuan, Z.M.; Bishop, A.J.; et al. Musashi1 impacts radio-resistance in glioblastoma by controlling DNA-protein kinase catalytic subunit. Am. J. Pathol. 2016, 186, 2271–2278. [Google Scholar] [CrossRef] [Green Version]
  58. Fang, Y.; Long, F. Circular rna circ_0000337 contributes to osteosarcoma via the mir-4458/bach1 pathway. Cancer Biomark. 2020, 28, 411–419. [Google Scholar] [CrossRef] [PubMed]
  59. Liu, N.Z.; Li, T.; Liu, C.M.; Liu, F.R.; Wang, Y.X. Hsa_circ_0000337 promotes proliferation, migration and invasion in glioma by competitively binding mirna-942-5p and thus upregulates mat2a. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 12251–12257. [Google Scholar]
  60. Song, H.; Xu, D.; Shi, P.; He, B.; Li, Z.; Ji, Y.; Agbeko, C.K.; Wang, J. Upregulated circ rna hsa_circ_0000337 promotes cell proliferation, migration, and invasion of esophageal squamous cell carcinoma. Cancer Manag. Res. 2019, 11, 1997–2006. [Google Scholar] [CrossRef] [Green Version]
  61. Liu, W.Y.; Yang, Z.; Sun, Q.; Yang, X.; Hu, Y.; Xie, H.; Gao, H.J.; Guo, L.M.; Yi, J.Y.; Liu, M.; et al. Mir-377-3p drives malignancy characteristics via upregulating gsk-3beta expression and activating nf-kappab pathway in hcrc cells. J. Cell Biochem. 2018, 119, 2124–2134. [Google Scholar] [CrossRef]
  62. Yuan, X.W.; Yan, T.Q.; Tong, H. Effect of mir-515-5p on proliferation and drug sensitivity of retinoblastoma cells. Cancer Manag. Res. 2020, 12, 12087–12098. [Google Scholar] [CrossRef]
  63. Lin, J.; Qin, H.; Han, Y.; Li, X.; Zhao, Y.; Zhai, G. Circnrip1 modulates the mir-515-5p/il-25 axis to control 5-fu and cisplatin resistance in nasopharyngeal carcinoma. Drug Des. Devel. Ther. 2021, 15, 323–330. [Google Scholar] [CrossRef] [PubMed]
  64. Han, X.; Sun, T.; Hong, J.; Wei, R.; Dong, Y.; Huang, D.; Chen, J.; Ren, X.; Zhou, H.; Tian, W.; et al. Nonreceptor tyrosine phosphatase 14 promotes proliferation and migration through regulating phosphorylation of yap of hippo signaling pathway in gastric cancer cells. J. Cell Biochem. 2019, 120, 17723–17730. [Google Scholar] [CrossRef]
  65. Liu, Y.P.; Sun, X.H.; Cao, X.L.; Jiang, W.W.; Wang, X.X.; Zhang, Y.F.; Wang, J.L. Microrna-217 suppressed epithelial-to-mesenchymal transition in gastric cancer metastasis through targeting ptpn14. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 1759–1767. [Google Scholar]
  66. Zhong, R.; Chen, Z.; Mo, T.; Li, Z.; Zhang, P. Potential role of circpvt1 as a proliferative factor and treatment target in esophageal carcinoma. Cancer Cell Int. 2019, 19, 267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Wang, S.; Su, T.T.; Tong, H.; Shi, W.; Ma, F.; Quan, Z. Circpvt1 promotes gallbladder cancer growth by sponging mir-339-3p and regulates mcl-1 expression. Cell Death Discov. 2021, 7, 191. [Google Scholar] [CrossRef]
  68. Zheng, F.; Xu, R. Circpvt1 contributes to chemotherapy resistance of lung adenocarcinoma through mir-145-5p/abcc1 axis. Biomed. Pharmacother. 2020, 124, 109828. [Google Scholar] [CrossRef] [PubMed]
  69. He, Q.; Zhao, L.; Liu, Y.; Liu, X.; Zheng, J.; Yu, H.; Cai, H.; Ma, J.; Liu, L.; Wang, P.; et al. Circ-shkbp1 regulates the angiogenesis of u87 glioma-exposed endothelial cells through mir-544a/foxp1 and mir-379/foxp2 pathways. Mol. Ther. Nucleic Acids 2018, 10, 331–348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Brooks, A.J.; Putoczki, T. Jak-stat signalling pathway in cancer. Cancers 2020, 12, 1971. [Google Scholar] [CrossRef]
  71. Khanna, P.; Chua, P.J.; Bay, B.H.; Baeg, G.H. The jak/stat signaling cascade in gastric carcinoma (review). Int. J. Oncol. 2015, 47, 1617–1626. [Google Scholar] [CrossRef] [Green Version]
  72. Mao, F.; Zhang, J.; Cheng, X.; Xu, Q. Mir-149 inhibits cell proliferation and enhances chemosensitivity by targeting cdc42 and bcl2 in neuroblastoma. Cancer Cell Int. 2019, 19, 357. [Google Scholar] [CrossRef] [PubMed]
  73. Luo, X.; Wang, G.H.; Bian, Z.L.; Li, X.W.; Zhu, B.Y.; Jin, C.J.; Ju, S.Q. Long non-coding rna ccal/mir-149/foxm1 axis promotes metastasis in gastric cancer. Cell Death Dis. 2018, 9, 993. [Google Scholar] [CrossRef]
  74. Zheng, B.; Liang, L.; Huang, S.; Zha, R.; Liu, L.; Jia, D.; Tian, Q.; Wang, Q.; Wang, C.; Long, Z.; et al. Microrna-409 suppresses tumour cell invasion and metastasis by directly targeting radixin in gastric cancers. Oncogene 2012, 31, 4509–4516. [Google Scholar] [CrossRef] [Green Version]
  75. Li, C.; Nie, H.; Wang, M.; Su, L.; Li, J.; Yu, B.; Wei, M.; Ju, J.; Yu, Y.; Yan, M.; et al. Microrna-409-3p regulates cell proliferation and apoptosis by targeting phf10 in gastric cancer. Cancer Lett. 2012, 320, 189–197. [Google Scholar] [CrossRef]
  76. Li, G.; Du, P.; He, J.; Li, Y. Circrna circbach1 (hsa_circ_0061395) serves as a mir-656-3p sponge to facilitate hepatocellular carcinoma progression through increasing serbp1 expression. Biochem. Biophys. Res. Commun. 2021, 556, 1–8. [Google Scholar] [CrossRef]
  77. Liu, B.; Yang, G.; Wang, X.; Liu, J.; Lu, Z.; Wang, Q.; Xu, B.; Liu, Z.; Li, J. Circbach1 (hsa_circ_0061395) promotes hepatocellular carcinoma growth by regulating p27 repression via hur. J. Cell Physiol. 2020, 235, 6929–6941. [Google Scholar] [CrossRef] [PubMed]
  78. Katsuno, Y.; Meyer, D.S.; Zhang, Z.; Shokat, K.M.; Akhurst, R.J.; Miyazono, K.; Derynck, R. Chronic tgf-beta exposure drives stabilized emt, tumor stemness, and cancer drug resistance with vulnerability to bitopic mtor inhibition. Sci. Signal. 2019, 12, eaau8544. [Google Scholar] [CrossRef] [PubMed]
  79. Seo, H.A.; Moeng, S.; Sim, S.; Kuh, H.J.; Choi, S.Y.; Park, J.K. Microrna-based combinatorial cancer therapy: Effects of micrornas on the efficacy of anti-cancer therapies. Cells 2019, 9, 29. [Google Scholar] [CrossRef] [Green Version]
  80. Li, J.; Tang, Q.; Dong, W.; Wang, Y. Circbach1/let-7a-5p axis enhances the proliferation and metastasis of colorectal cancer by upregulating creb5 expression. J. Gastrointest. Oncol. 2020, 11, 1186–1199. [Google Scholar] [CrossRef]
  81. Bai, N.; Peng, E.; Qiu, X.; Lyu, N.; Zhang, Z.; Tao, Y.; Li, X.; Wang, Z. Circfblim1 act as a cerna to promote hepatocellular cancer progression by sponging mir-346. J. Exp. Clin. Cancer Res. 2018, 37, 172. [Google Scholar] [CrossRef] [Green Version]
  82. Yu, Q.; Yang, X.; Duan, W.; Li, C.; Luo, Y.; Lu, S. Mirna-346 promotes proliferation, migration and invasion in liver cancer. Oncol. Lett. 2017, 14, 3255–3260. [Google Scholar] [CrossRef] [Green Version]
  83. Pan, S.; Wu, W.; Ren, F.; Li, L.; Li, Y.; Li, W.; Wang, A.; Liu, D.; Dong, Y. Mir-346-5p promotes colorectal cancer cell proliferation in vitro and in vivo by targeting fbxl2 and activating the beta-catenin signaling pathway. Life Sci. 2020, 244, 117300. [Google Scholar] [CrossRef] [PubMed]
  84. Sun, C.C.; Li, S.J.; Yuan, Z.P.; Li, D.J. Microrna-346 facilitates cell growth and metastasis, and suppresses cell apoptosis in human non-small cell lung cancer by regulation of xpc/erk/snail/e-cadherin pathway. Aging 2016, 8, 2509–2524. [Google Scholar] [CrossRef] [Green Version]
  85. Lv, X.; Huang, H.; Feng, H.; Wei, Z. Circ-mmp2 (circ-0039411) induced by foxm1 promotes the proliferation and migration of lung adenocarcinoma cells in vitro and in vivo. Cell Death Dis. 2020, 11, 426. [Google Scholar] [CrossRef] [PubMed]
  86. Yang, Y.; Ding, L.; Li, Y.; Xuan, C. Hsa_circ_0039411 promotes tumorigenesis and progression of papillary thyroid cancer by mir-1179/abca9 and mir-1205/mta1 signaling pathways. J. Cell Physiol. 2020, 235, 1321–1329. [Google Scholar] [CrossRef] [PubMed]
  87. Hu, Y.; Yang, C.; Yang, S.; Cheng, F.; Rao, J.; Wang, X. Mir-665 promotes hepatocellular carcinoma cell migration, invasion, and proliferation by decreasing hippo signaling through targeting ptprb. Cell Death Dis. 2018, 9, 954. [Google Scholar] [CrossRef] [Green Version]
  88. Qu, Z.; Wu, J.; Wu, J.; Ji, A.; Qiang, G.; Jiang, Y.; Jiang, C.; Ding, Y. Exosomal mir-665 as a novel minimally invasive biomarker for hepatocellular carcinoma diagnosis and prognosis. Oncotarget 2017, 8, 80666–80678. [Google Scholar] [CrossRef] [Green Version]
  89. Zhao, W.; Cui, Y.; Liu, L.; Qi, X.; Liu, J.; Ma, S.; Hu, X.; Zhang, Z.; Wang, Y.; Li, H.; et al. Splicing factor derived circular rna circuhrf1 accelerates oral squamous cell carcinoma tumorigenesis via feedback loop. Cell Death Differ. 2020, 27, 919–933. [Google Scholar] [CrossRef]
  90. Ndhlovu, L.C.; Lopez-Verges, S.; Barbour, J.D.; Jones, R.B.; Jha, A.R.; Long, B.R.; Schoeffler, E.C.; Fujita, T.; Nixon, D.F.; Lanier, L.L. Tim-3 marks human natural killer cell maturation and suppresses cell-mediated cytotoxicity. Blood 2012, 119, 3734–3743. [Google Scholar] [CrossRef] [Green Version]
  91. Xu, L.; Huang, Y.; Tan, L.; Yu, W.; Chen, D.; Lu, C.; He, J.; Wu, G.; Liu, X.; Zhang, Y. Increased tim-3 expression in peripheral nk cells predicts a poorer prognosis and tim-3 blockade improves nk cell-mediated cytotoxicity in human lung adenocarcinoma. Int. Immunopharmacol. 2015, 29, 635–641. [Google Scholar] [CrossRef]
  92. Deng, B.; Wang, B.; Fang, J.; Zhu, X.; Cao, Z.; Lin, Q.; Zhou, L.; Sun, X. Mirna-203 suppresses cell proliferation, migration and invasion in colorectal cancer via targeting of eif5a2. Sci. Rep. 2016, 6, 28301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Zhai, H.; Song, B.; Xu, X.; Zhu, W.; Ju, J. Inhibition of autophagy and tumor growth in colon cancer by mir-502. Oncogene 2013, 32, 1570–1579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Peng, X.; Wu, M.; Liu, W.; Guo, C.; Zhan, L.; Zhan, X. Mir-502-5p inhibits the proliferation, migration and invasion of gastric cancer cells by targeting sp1. Oncol. Lett. 2020, 20, 2757–2762. [Google Scholar] [CrossRef] [PubMed]
  95. Guo, J.; Duan, H.; Li, Y.; Yang, L.; Yuan, L. A novel circular rna circ-znf652 promotes hepatocellular carcinoma metastasis through inducing snail-mediated epithelial-mesenchymal transition by sponging mir-203/mir-502-5p. Biochem. Biophys. Res. Commun. 2019, 513, 812–819. [Google Scholar] [CrossRef]
  96. Yuan, X.; Zhang, Y.; Cai, C.; Liu, C.; Xie, J.; Yi, C. Circular rna circznf652 is overexpressed in osteoarthritis and positively regulates lps-induced apoptosis of chondrocytes by upregulating pten. Autoimmunity 2021, 54, 415–421. [Google Scholar] [CrossRef]
  97. Kang, L.; Shen, L.; Lu, L.; Wang, D.; Zhao, Y.; Chen, C.; Du, L.; Gong, J.; Zhang, Y.; Mi, X.; et al. Asparaginyl endopeptidase induces endothelial permeability and tumor metastasis via downregulating zonula occludens protein zo-1. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 2267–2275. [Google Scholar] [CrossRef]
  98. Martin, T.A. The role of tight junctions in cancer metastasis. Semin. Cell Dev. Biol. 2014, 36, 224–231. [Google Scholar] [CrossRef]
  99. Jia, F.; Zhang, Z.; Zhang, X. Microrna-338-3p inhibits tumor growth and metastasis in osteosarcoma cells by targeting runx2/cdk4 and inhibition of mapk pathway. J. Cell Biochem. 2019, 120, 6420–6430. [Google Scholar] [CrossRef]
  100. Niu, Q.; Liu, Z.; Gao, J.; Wang, Q. Mir-338-3p enhances ovarian cancer cell sensitivity to cisplatin by downregulating wnt2b. Yonsei Med. J. 2019, 60, 1146–1156. [Google Scholar] [CrossRef]
  101. Xue, Q.; Sun, K.; Deng, H.J.; Lei, S.T.; Dong, J.Q.; Li, G.X. Microrna-338-3p inhibits colorectal carcinoma cell invasion and migration by targeting smoothened. Jpn. J. Clin. Oncol. 2014, 44, 13–21. [Google Scholar] [CrossRef] [Green Version]
  102. Stein, U.; Walther, W.; Arlt, F.; Schwabe, H.; Smith, J.; Fichtner, I.; Birchmeier, W.; Schlag, P.M. Macc1, a newly identified key regulator of hgf-met signaling, predicts colon cancer metastasis. Nat. Med. 2009, 15, 59–67. [Google Scholar] [CrossRef] [PubMed]
  103. Joo, H.Y.; Yun, M.; Jeong, J.; Park, E.R.; Shin, H.J.; Woo, S.R.; Jung, J.K.; Kim, Y.M.; Park, J.J.; Kim, J.; et al. Sirt1 deacetylates and stabilizes hypoxia-inducible factor-1alpha (hif-1alpha) via direct interactions during hypoxia. Biochem. Biophys. Res. Commun. 2015, 462, 294–300. [Google Scholar] [CrossRef] [PubMed]
  104. Li, A.; Peng, R.; Sun, Y.; Liu, H.; Peng, H.; Zhang, Z. Lincrna 1700020i14rik alleviates cell proliferation and fibrosis in diabetic nephropathy via mir-34a-5p/sirt1/hif-1alpha signaling. Cell Death Dis. 2018, 9, 461. [Google Scholar] [CrossRef]
  105. Rane, S.; He, M.; Sayed, D.; Vashistha, H.; Malhotra, A.; Sadoshima, J.; Vatner, D.E.; Vatner, S.F.; Abdellatif, M. Downregulation of mir-199a derepresses hypoxia-inducible factor-1alpha and sirtuin 1 and recapitulates hypoxia preconditioning in cardiac myocytes. Circ. Res. 2009, 104, 879–886. [Google Scholar] [CrossRef] [PubMed]
  106. Xiang, Q.; Kang, L.; Wang, J.; Liao, Z.; Song, Y.; Zhao, K.; Wang, K.; Yang, C.; Zhang, Y. Circrna-cidn mitigated compression loading-induced damage in human nucleus pulposus cells via mir-34a-5p/sirt1 axis. EBioMedicine 2020, 53, 102679. [Google Scholar] [CrossRef]
  107. Lei, B.; Huang, Y.; Zhou, Z.; Zhao, Y.; Thapa, A.J.; Li, W.; Cai, W.; Deng, Y. Circular rna hsa_circ_0076248 promotes oncogenesis of glioma by sponging mir-181a to modulate sirt1 expression. J. Cell Biochem. 2019, 120, 6698–6708. [Google Scholar] [CrossRef] [Green Version]
  108. Chen, S.; Yang, M.; Chang, S. Lncrna ccal promotes angiogenesis through regulating the mir-29b/angptl4 axis in osteosarcoma. Cancer Manag. Res. 2020, 12, 10521–10530. [Google Scholar] [CrossRef] [PubMed]
  109. Ma, Y.; Yang, Y.; Wang, F.; Moyer, M.P.; Wei, Q.; Zhang, P.; Yang, Z.; Liu, W.; Zhang, H.; Chen, N.; et al. Long non-coding rna ccal regulates colorectal cancer progression by activating wnt/beta-catenin signalling pathway via suppression of activator protein 2alpha. Gut 2016, 65, 1494–1504. [Google Scholar] [CrossRef]
  110. Deng, X.; Ruan, H.; Zhang, X.; Xu, X.; Zhu, Y.; Peng, H.; Zhang, X.; Kong, F.; Guan, M. Long noncoding rna ccal transferred from fibroblasts by exosomes promotes chemoresistance of colorectal cancer cells. Int. J. Cancer 2020, 146, 1700–1716. [Google Scholar] [CrossRef]
  111. Tsai, K.W.; Lo, Y.H.; Liu, H.; Yeh, C.Y.; Chen, Y.Z.; Hsu, C.W.; Chen, W.S.; Wang, J.H. Linc00659, a long noncoding rna, acts as novel oncogene in regulating cancer cell growth in colorectal cancer. Mol. Cancer 2018, 17, 72. [Google Scholar] [CrossRef] [PubMed]
  112. Zhou, L.; Li, J.; Tang, Y.; Yang, M. Exosomal lncrna linc00659 transferred from cancer-associated fibroblasts promotes colorectal cancer cell progression via mir-342-3p/anxa2 axis. J. Transl Med. 2021, 19, 8. [Google Scholar] [CrossRef]
  113. Rocha, M.R.; Barcellos-de-Souza, P.; Sousa-Squiavinato, A.C.M.; Fernandes, P.V.; de Oliveira, I.M.; Boroni, M.; Morgado-Diaz, J.A. Annexin a2 overexpression associates with colorectal cancer invasiveness and tgf-ss induced epithelial mesenchymal transition via src/anxa2/stat3. Sci. Rep. 2018, 8, 11285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Mi, X.; Xu, R.; Hong, S.; Xu, T.; Zhang, W.; Liu, M. M2 macrophage-derived exosomal lncrna afap1-as1 and microrna-26a affect cell migration and metastasis in esophageal cancer. Mol. Ther. Nucleic Acids 2020, 22, 779–790. [Google Scholar] [CrossRef]
  115. Ma, D.; Gao, X.; Liu, Z.; Lu, X.; Ju, H.; Zhang, N. Exosome-transferred long non-coding rna asmtl-as1 contributes to malignant phenotypes in residual hepatocellular carcinoma after insufficient radiofrequency ablation. Cell Prolif. 2020, 53, e12795. [Google Scholar] [CrossRef]
  116. Han, W.; Sulidankazha, Q.; Nie, X.; Yilidan, R.; Len, K. Pancreatic cancer cells-derived exosomal long non-coding rna ccat1/microrna-138-5p/hmga1 axis promotes tumor angiogenesis. Life Sci. 2021, 278, 119495. [Google Scholar] [CrossRef]
  117. Piao, H.Y.; Guo, S.; Wang, Y.; Zhang, J. Exosomal long non-coding rna cebpa-as1 inhibits tumor apoptosis and functions as a non-invasive biomarker for diagnosis of gastric cancer. Onco Targets Ther. 2020, 13, 1365–1374. [Google Scholar] [CrossRef] [Green Version]
  118. Sun, J.; Jia, H.; Bao, X.; Wu, Y.; Zhu, T.; Li, R.; Zhao, H. Tumor exosome promotes th17 cell differentiation by transmitting the lncrna crnde-h in colorectal cancer. Cell Death Dis. 2021, 12, 123. [Google Scholar] [CrossRef] [PubMed]
  119. Wang, L.P.; Lin, J.; Ma, X.Q.; Xu, D.Y.; Shi, C.F.; Wang, W.; Jiang, X.J. Exosomal dlx6-as1 from hepatocellular carcinoma cells induces m2 macrophage polarization to promote migration and invasion in hepatocellular carcinoma through microrna-15a-5p/cxcl17 axis. J. Exp. Clin. Cancer Res. 2021, 40, 177. [Google Scholar] [CrossRef] [PubMed]
  120. Yao, Z.; Jia, C.; Tai, Y.; Liang, H.; Zhong, Z.; Xiong, Z.; Deng, M.; Zhang, Q. Serum exosomal long noncoding rnas lnc-fam72d-3 and lnc-epc1-4 as diagnostic biomarkers for hepatocellular carcinoma. Aging 2020, 12, 11843–11863. [Google Scholar] [CrossRef]
  121. 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]
  122. Li, W.; Zhang, L.; Guo, B.; Deng, J.; Wu, S.; Li, F.; Wang, Y.; Lu, J.; Zhou, Y. Exosomal fmr1-as1 facilitates maintaining cancer stem-like cell dynamic equilibrium via tlr7/nfkappab/c-myc signaling in female esophageal carcinoma. Mol. Cancer 2019, 18, 22. [Google Scholar] [CrossRef] [PubMed]
  123. Zhang, Y.; Chen, L.; Ye, X.; Wu, Z.; Zhang, Z.; Sun, B.; Fu, H.; Fu, C.; Liang, X.; Jiang, H. Expression and mechanism of exosome-mediated a foxm1 related long noncoding rna in gastric cancer. J. Nanobiotechnol. 2021, 19, 133. [Google Scholar] [CrossRef]
  124. Chen, X.; Liu, Y.; Zhang, Q.; Liu, B.; Cheng, Y.; Zhang, Y.; Sun, Y.; Liu, J.; Gen, H. Exosomal long non-coding rna hottip increases resistance of colorectal cancer cells to mitomycin via impairing mir-214-mediated degradation of kpna3. Front. Cell Dev. Biol. 2020, 8, 582723. [Google Scholar] [CrossRef] [PubMed]
  125. Wang, J.; Lv, B.; Su, Y.; Wang, X.; Bu, J.; Yao, L. Exosome-mediated transfer of lncrna hottip promotes cisplatin resistance in gastric cancer cells by regulating hmga1/mir-218 axis. Onco Targets Ther. 2019, 12, 11325–11338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Takahashi, K.; Ota, Y.; Kogure, T.; Suzuki, Y.; Iwamoto, H.; Yamakita, K.; Kitano, Y.; Fujii, S.; Haneda, M.; Patel, T.; et al. Circulating extracellular vesicle-encapsulated hulc is a potential biomarker for human pancreatic cancer. Cancer Sci. 2020, 111, 98–111. [Google Scholar] [CrossRef] [Green Version]
  127. Xian, D.; Niu, L.; Zeng, J.; Wang, L. Lncrna kcnq1ot1 secreted by tumor cell-derived exosomes mediates immune escape in colorectal cancer by regulating pd-l1 ubiquitination via mir-30a-5p/usp22. Front. Cell Dev. Biol. 2021, 9, 653808. [Google Scholar] [CrossRef]
  128. 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] [PubMed]
  129. Liu, Y.; Tang, T.; Yang, X.; Qin, P.; Wang, P.; Zhang, H.; Bai, M.; Wu, R.; Li, F. Tumor-derived exosomal long noncoding rna linc01133, regulated by periostin, contributes to pancreatic ductal adenocarcinoma epithelial-mesenchymal transition through the wnt/beta-catenin pathway by silencing axin2. Oncogene 2021, 40, 3164–3179. [Google Scholar] [CrossRef]
  130. Wang, L.; Bo, X.; Yi, X.; Xiao, X.; Zheng, Q.; Ma, L.; Li, B. Exosome-transferred linc01559 promotes the progression of gastric cancer via pi3k/akt signaling pathway. Cell Death Dis. 2020, 11, 723. [Google Scholar] [CrossRef]
  131. Xu, M.L.; Liu, T.C.; Dong, F.X.; Meng, L.X.; Ling, A.X.; Liu, S. Exosomal lncrna linc01711 facilitates metastasis of esophageal squamous cell carcinoma via the mir-326/fscn1 axis. Aging 2021, 13, 19776–19788. [Google Scholar] [CrossRef] [PubMed]
  132. Zhao, Y.; Du, T.; Du, L.; Li, P.; Li, J.; Duan, W.; Wang, Y.; Wang, C. Long noncoding rna linc02418 regulates melk expression by acting as a cerna and may serve as a diagnostic marker for colorectal cancer. Cell Death Dis. 2019, 10, 568. [Google Scholar] [CrossRef] [PubMed]
  133. Xu, J.; Xiao, Y.; Liu, B.; Pan, S.; Liu, Q.; Shan, Y.; Li, S.; Qi, Y.; Huang, Y.; Jia, L. Exosomal malat1 sponges mir-26a/26b to promote the invasion and metastasis of colorectal cancer via fut4 enhanced fucosylation and pi3k/akt pathway. J. Exp. Clin. Cancer Res. 2020, 39, 54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Huang, L.; Wang, Y.; Chen, J.; Wang, Y.; Zhao, Y.; Wang, Y.; Ma, Y.; Chen, X.; Liu, W.; Li, Z.; et al. Long noncoding rna pcat1, a novel serum-based biomarker, enhances cell growth by sponging mir-326 in oesophageal squamous cell carcinoma. Cell Death Dis. 2019, 10, 513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Piao, H.Y.; Guo, S.; Wang, Y.; Zhang, J. Exosome-transmitted lncrna pcgem1 promotes invasive and metastasis in gastric cancer by maintaining the stability of snai1. Clin. Transl. Oncol. 2021, 23, 246–256. [Google Scholar] [CrossRef] [PubMed]
  136. Tong, Y.; Yang, L.; Yu, C.; Zhu, W.; Zhou, X.; Xiong, Y.; Wang, W.; Ji, F.; He, D.; Cao, X. Tumor-secreted exosomal lncrna pou3f3 promotes cisplatin resistance in escc by inducing fibroblast differentiation into cafs. Mol. Ther. Oncolytics 2020, 18, 1–13. [Google Scholar] [CrossRef] [PubMed]
  137. Liang, Z.X.; Liu, H.S.; Wang, F.W.; Xiong, L.; Zhou, C.; Hu, T.; He, X.W.; Wu, X.J.; Xie, D.; Wu, X.R.; et al. Lncrna rpph1 promotes colorectal cancer metastasis by interacting with tubb3 and by promoting exosomes-mediated macrophage m2 polarization. Cell Death Dis. 2019, 10, 829. [Google Scholar] [CrossRef] [Green Version]
  138. Cao, S.; Lin, L.; Xia, X.; Wu, H. Lncrna spry4-it1 regulates cell proliferation and migration by sponging mir-101-3p and regulating ampk expression in gastric cancer. Mol. Ther. Nucleic Acids 2019, 17, 455–464. [Google Scholar] [CrossRef] [Green Version]
  139. Li, X.; Lei, Y.; Wu, M.; Li, N. Regulation of macrophage activation and polarization by hcc-derived exosomal lncrna tuc339. Int. J. Mol. Sci. 2018, 19, 2958. [Google Scholar] [CrossRef] [Green Version]
  140. Luan, Y.; Li, X.; Luan, Y.; Zhao, R.; Li, Y.; Liu, L.; Hao, Y.; Oleg Vladimir, B.; Jia, L. Circulating lncrna uca1 promotes malignancy of colorectal cancer via the mir-143/myo6 axis. Mol. Ther. Nucleic Acids 2020, 19, 790–803. [Google Scholar] [CrossRef]
  141. 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]
  142. Li, Z.; Qin, X.; Bian, W.; Li, Y.; Shan, B.; Yao, Z.; Li, S. Exosomal lncrna zfas1 regulates esophageal squamous cell carcinoma cell proliferation, invasion, migration and apoptosis via microrna-124/stat3 axis. J. Exp. Clin. Cancer Res. 2019, 38, 477. [Google Scholar] [CrossRef] [PubMed]
  143. Cheng, J.L.; Li, D.J.; Lv, M.Y.; Pei, Y.J.; Zhang, X.J.; Li, L.; Liu, X.Y.; Fan, A.H. Lncrna kcnq1ot1 regulates the invasion and migration of hepatocellular carcinoma by acting on s1pr1 through mir-149. Cancer Gene Ther. 2021, 28, 850–863. [Google Scholar] [CrossRef]
  144. Huang, A.; Jin, S.; Han, W.; Wang, Y.; Ma, S.; Wang, Z.; Lin, K.; Zou, Q.; Zhou, J.; Li, Z.; et al. Long noncoding rna kcnq1ot1 contributes to tumor growth and activates wnt/betacatenin signaling in osteosarcoma by targeting the mir3666/klf7 axis. Int. J. Mol. Med. 2021, 47, 387–396. [Google Scholar] [CrossRef]
  145. Zhu, Y.; Shen, Y.; Chen, R.; Li, H.; Wu, Y.; Zhang, F.; Huang, W.; Guo, L.; Chen, Q.; Liu, H. Kcnq1ot1 lncrna affects the proliferation, apoptosis, and chemoresistance of small cell lung cancer cells via the jak2/stat3 axis. Ann. Transl. Med. 2021, 9, 891. [Google Scholar] [CrossRef]
  146. Xian, D.; Zhao, Y. Lncrna kcnq1ot1 enhanced the methotrexate resistance of colorectal cancer cells by regulating mir-760/ppp1r1b via the camp signalling pathway. J. Cell Mol. Med. 2019, 23, 3808–3823. [Google Scholar] [CrossRef]
  147. Zhang, J.W.; Wang, X.; Li, G.C.; Wang, D.; Han, S.; Zhang, Y.D.; Luo, C.H.; Wang, H.W.; Jiang, W.J.; Li, C.X.; et al. Mir-30a-5p promotes cholangiocarcinoma cell proliferation through targeting socs3. J. Cancer 2020, 11, 3604–3614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Wang, Z.; Dai, X.; Chen, Y.; Sun, C.; Zhu, Q.; Zhao, H.; Liu, G.; Huang, Q.; Lan, Q. Mir-30a-5p is induced by wnt/beta-catenin pathway and promotes glioma cell invasion by repressing ncam. Biochem. Biophys. Res. Commun. 2015, 465, 374–380. [Google Scholar] [CrossRef] [PubMed]
  149. Sobierajska, K.; Wieczorek, K.; Ciszewski, W.M.; Sacewicz-Hofman, I.; Wawro, M.E.; Wiktorska, M.; Boncela, J.; Papiewska-Pajak, I.; Kwasniak, P.; Wyroba, E.; et al. Beta-iii tubulin modulates the behavior of snail overexpressed during the epithelial-to-mesenchymal transition in colon cancer cells. Biochim. Biophys. Acta 2016, 1863, 2221–2233. [Google Scholar] [CrossRef]
  150. Kanojia, D.; Morshed, R.A.; Zhang, L.; Miska, J.M.; Qiao, J.; Kim, J.W.; Pytel, P.; Balyasnikova, I.V.; Lesniak, M.S.; Ahmed, A.U. Betaiii-tubulin regulates breast cancer metastases to the brain. Mol. Cancer Ther. 2015, 14, 1152–1161. [Google Scholar] [CrossRef] [Green Version]
  151. Sun, Y.; Hu, B.; Wang, Q.; Ye, M.; Qiu, Q.; Zhou, Y.; Zeng, F.; Zhang, X.; Guo, Y.; Guo, L. Long non-coding rna hottip promotes bcl-2 expression and induces chemoresistance in small cell lung cancer by sponging mir-216a. Cell Death Dis. 2018, 9, 85. [Google Scholar] [CrossRef] [Green Version]
  152. Jiang, H.; Xiong, W.; Chen, L.; Lv, Z.; Yang, C.; Li, Y. Knockdown of the long noncoding rna hottip inhibits cell proliferation and enhances cell sensitivity to cisplatin by suppressing the wnt/beta-catenin pathway in prostate cancer. J. Cell Biochem. 2019, 120, 8965–8974. [Google Scholar] [CrossRef] [PubMed]
  153. Dong, Y.J.; Feng, W.; Li, Y. Hottip-mir-205-zeb2 axis confers cisplatin resistance to ovarian cancer cells. Front. Cell Dev. Biol. 2021, 9, 707424. [Google Scholar] [CrossRef] [PubMed]
  154. He, L.; Zhu, W.; Chen, Q.; Yuan, Y.; Wang, Y.; Wang, J.; Wu, X. Ovarian cancer cell-secreted exosomal mir-205 promotes metastasis by inducing angiogenesis. Theranostics 2019, 9, 8206–8220. [Google Scholar] [CrossRef] [PubMed]
  155. Han, B. Lncrna linc02418 regulates proliferation and apoptosis of non-small cell lung cancer cells by regulating mir-4677-3p/sec61g. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 10354–10362. [Google Scholar]
  156. Wang, T.; Zhai, R.; Lv, X.; Wang, K.; Xu, J. Linc02418 promotes malignant behaviors in lung adenocarcinoma cells by sponging mir-4677-3p to upregulate knl1 expression. BMC Pulm. Med. 2020, 20, 217. [Google Scholar] [CrossRef]
  157. Tian, J.; Cui, P.; Li, Y.; Yao, X.; Wu, X.; Wang, Z.; Li, C. Linc02418 promotes colon cancer progression by suppressing apoptosis via interaction with mir-34b-5p/bcl2 axis. Cancer Cell Int. 2020, 20, 460. [Google Scholar] [CrossRef] [PubMed]
  158. Li, M.; Qian, X.; Zhu, M.; Li, A.; Fang, M.; Zhu, Y.; Zhang, J. Mir1273g3p promotes proliferation, migration and invasion of lovo cells via cannabinoid receptor 1 through activation of erbb4/pik3r3/mtor/s6k2 signaling pathway. Mol. Med. Rep. 2018, 17, 4619–4626. [Google Scholar]
  159. Zhu, Z.; Wang, H.; Pang, Y.; Hu, H.; Zhang, H.; Wang, W. Exosomal long non-coding rna uca1 functions as growth inhibitor in esophageal cancer. Aging 2020, 12, 20523–20539. [Google Scholar] [CrossRef]
  160. Tang, J.; Zhong, G.; Wu, J.; Chen, H.; Jia, Y. Long noncoding rna afap1-as1 facilitates tumor growth through enhancer of zeste homolog 2 in colorectal cancer. Am. J. Cancer Res. 2018, 8, 892–902. [Google Scholar] [PubMed]
  161. Yuan, Z.; Xiu, C.; Song, K.; Pei, R.; Miao, S.; Mao, X.; Sun, J.; Jia, S. Long non-coding rna afap1-as1/mir-320a/rbpj axis regulates laryngeal carcinoma cell stemness and chemoresistance. J. Cell Mol. Med. 2018, 22, 4253–4262. [Google Scholar] [CrossRef] [PubMed]
  162. Liu, J.Y.; Jiang, L.; He, T.; Liu, J.J.; Fan, J.Y.; Xu, X.H.; Tang, B.; Shi, Y.; Zhao, Y.L.; Qian, F.; et al. Neto2 promotes invasion and metastasis of gastric cancer cells via activation of pi3k/akt/nf-kappab/snail axis and predicts outcome of the patients. Cell Death Dis. 2019, 10, 162. [Google Scholar] [CrossRef] [PubMed]
  163. Xu, J.C.; Chen, T.Y.; Liao, L.T.; Chen, T.; Li, Q.L.; Xu, J.X.; Hu, J.W.; Zhou, P.H.; Zhang, Y.Q. Neto2 promotes esophageal cancer progression by inducing proliferation and metastasis via pi3k/akt and erk pathway. Int. J. Biol. Sci. 2021, 17, 259–270. [Google Scholar] [CrossRef]
  164. van Keimpema, M.; Gruneberg, L.J.; Mokry, M.; van Boxtel, R.; Koster, J.; Coffer, P.J.; Pals, S.T.; Spaargaren, M. Foxp1 directly represses transcription of proapoptotic genes and cooperates with nf-kappab to promote survival of human b cells. Blood 2014, 124, 3431–3440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Choi, E.J.; Seo, E.J.; Kim, D.K.; Lee, S.I.; Kwon, Y.W.; Jang, I.H.; Kim, K.H.; Suh, D.S.; Kim, J.H. Foxp1 functions as an oncogene in promoting cancer stem cell-like characteristics in ovarian cancer cells. Oncotarget 2016, 7, 3506–3519. [Google Scholar] [CrossRef] [PubMed]
  166. Vishnubalaji, R.; Alajez, N.M. Epigenetic regulation of triple negative breast cancer (tnbc) by tgf-beta signaling. Sci. Rep. 2021, 11, 15410. [Google Scholar] [CrossRef]
  167. Liu, H.; Zhang, Y.; Li, L.; Cao, J.; Guo, Y.; Wu, Y.; Gao, W. Fascin actin-bundling protein 1 in human cancer: Promising biomarker or therapeutic target? Mol. Ther. Oncolytics 2021, 20, 240–264. [Google Scholar] [CrossRef]
  168. Zhang, Y.; Lu, Y.; Zhang, C.; Huang, D.; Wu, W.; Zhang, Y.; Shen, J.; Cai, Y.; Chen, W.; Yao, W. Fscn1 increases doxorubicin resistance in hepatocellular carcinoma through promotion of epithelial-mesenchymal transition. Int. J. Oncol. 2018, 52, 1455–1464. [Google Scholar]
  169. Hayashi, Y.; Osanai, M.; Lee, G.H. Fascin-1 expression correlates with repression of e-cadherin expression in hepatocellular carcinoma cells and augments their invasiveness in combination with matrix metalloproteinases. Cancer Sci. 2011, 102, 1228–1235. [Google Scholar] [CrossRef] [PubMed]
  170. Li, T.; Xie, J.; Shen, C.; Cheng, D.; Shi, Y.; Wu, Z.; Deng, X.; Chen, H.; Shen, B.; Peng, C.; et al. Amplification of long noncoding rna zfas1 promotes metastasis in hepatocellular carcinoma. Cancer Res. 2015, 75, 3181–3191. [Google Scholar] [CrossRef] [Green Version]
  171. Chen, X.; Zeng, K.; Xu, M.; Hu, X.; Liu, X.; Xu, T.; He, B.; Pan, Y.; Sun, H.; Wang, S. Sp1-induced lncrna-zfas1 contributes to colorectal cancer progression via the mir-150-5p/vegfa axis. Cell Death Dis. 2018, 9, 982. [Google Scholar] [CrossRef] [PubMed]
  172. Li, H.; Ma, X.; Yang, D.; Suo, Z.; Dai, R.; Liu, C. Pcat-1 contributes to cisplatin resistance in gastric cancer through epigenetically silencing pten via recruiting ezh2. J. Cell Biochem. 2020, 121, 1353–1361. [Google Scholar] [CrossRef]
  173. Zhang, D.; Cao, J.; Zhong, Q.; Zeng, L.; Cai, C.; Lei, L.; Zhang, W.; Liu, F. Long noncoding rna pcat-1 promotes invasion and metastasis via the mir-129-5p-hmgb1 signaling pathway in hepatocellular carcinoma. Biomed. Pharmacother. 2017, 95, 1187–1193. [Google Scholar] [CrossRef] [PubMed]
  174. Shan, T.D.; Xu, J.H.; Yu, T.; Li, J.Y.; Zhao, L.N.; Ouyang, H.; Luo, S.; Lu, X.J.; Huang, C.Z.; Lan, Q.S.; et al. Knockdown of linc-pou3f3 suppresses the proliferation, apoptosis, and migration resistance of colorectal cancer. Oncotarget 2016, 7, 961–975. [Google Scholar] [CrossRef] [Green Version]
  175. Wan, X.; Xiang, J.; Zhang, Q.; Bian, C. Long noncoding rna pou3f3 promotes cancer cell proliferation in prostate carcinoma by upregulating rho-associated protein kinase 1. J. Cell Biochem. 2018, 120, 8195–8200. [Google Scholar] [CrossRef]
  176. Lee, H.J.; Diaz, M.F.; Price, K.M.; Ozuna, J.A.; Zhang, S.; Sevick-Muraca, E.M.; Hagan, J.P.; Wenzel, P.L. Fluid shear stress activates yap1 to promote cancer cell motility. Nat. Commun. 2017, 8, 14122. [Google Scholar] [CrossRef] [Green Version]
  177. Shen, T.; Li, Y.; Zhu, S.; Yu, J.; Zhang, B.; Chen, X.; Zhang, Z.; Ma, Y.; Niu, Y.; Shang, Z. Yap1 plays a key role of the conversion of normal fibroblasts into cancer-associated fibroblasts that contribute to prostate cancer progression. J. Exp. Clin. Cancer Res. 2020, 39, 36. [Google Scholar] [CrossRef] [PubMed]
  178. Xia, P.; Gu, R.; Zhang, W.; Sun, Y.F. Lncrna cebpa-as1 overexpression inhibits proliferation and migration and stimulates apoptosis of os cells via notch signaling. Mol. Ther. Nucleic Acids 2020, 19, 1470–1481. [Google Scholar] [CrossRef] [PubMed]
  179. Guo, Y.; Ma, Y.; Hu, X.; Song, R.; Zhu, L.; Zhong, M. Long non-coding rna cebpa-as1 correlates with poor prognosis and promotes tumorigenesis via cebpa/bcl2 in oral squamous cell carcinoma. Cancer Biol. Ther. 2018, 19, 205–213. [Google Scholar] [CrossRef] [Green Version]
  180. Cai, H.; Chen, J.; He, B.; Li, Q.; Li, Y.; Gao, Y. A foxm1 related long non-coding rna contributes to gastric cancer cell migration. Mol. Cell Biochem. 2015, 406, 31–41. [Google Scholar] [CrossRef]
  181. Zhang, T.; Piao, H.Y.; Guo, S.; Zhao, Y.; Wang, Y.; Zheng, Z.C.; Zhang, J. Lncrna pcgem1 enhances metastasis and gastric cancer invasion through targeting of mir-129-5p to regulate p4ha2 expression. Exp. Mol. Pathol. 2020, 116, 104487. [Google Scholar] [CrossRef]
  182. Lin, J.; Jiang, L.; Wang, X.; Wei, W.; Song, C.; Cui, Y.; Wu, X.; Qiu, G. P4ha2 promotes epithelial-to-mesenchymal transition and glioma malignancy through the collagen-dependent pi3k/akt pathway. J. Oncol. 2021, 2021, 1406853. [Google Scholar] [CrossRef]
  183. Wang, C.Q. Mir-195 reverses 5-fu resistance through targeting hmga1 in gastric cancer cells. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 3771–3778. [Google Scholar] [PubMed]
  184. Kim, D.K.; Seo, E.J.; Choi, E.J.; Lee, S.I.; Kwon, Y.W.; Jang, I.H.; Kim, S.C.; Kim, K.H.; Suh, D.S.; Seong-Jang, K.; et al. Crucial role of hmga1 in the self-renewal and drug resistance of ovarian cancer stem cells. Exp. Mol. Med. 2016, 48, e255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Wang, Y.T.; Pan, S.H.; Tsai, C.F.; Kuo, T.C.; Hsu, Y.L.; Yen, H.Y.; Choong, W.K.; Wu, H.Y.; Liao, Y.C.; Hong, T.M.; et al. Phosphoproteomics reveals hmga1, a ck2 substrate, as a drug-resistant target in non-small cell lung cancer. Sci. Rep. 2017, 7, 44021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Lou, C.; Zhao, J.; Gu, Y.; Li, Q.; Tang, S.; Wu, Y.; Tang, J.; Zhang, C.; Li, Z.; Zhang, Y. Linc01559 accelerates pancreatic cancer cell proliferation and migration through yap-mediated pathway. J. Cell Physiol. 2020, 235, 3928–3938. [Google Scholar] [CrossRef]
  187. Dong, S.; Fu, Y.; Yang, K.; Zhang, X.; Miao, R.; Long, Y.; Liu, C. Linc01559 served as a potential oncogene and promoted resistance of hepatocellular carcinoma to oxaliplatin by directly sponging mir-6783-3p. Anticancer Agents Med. Chem 2021, 21, 278–286. [Google Scholar] [CrossRef] [PubMed]
  188. Shen, H.; Zhu, H.; Chen, Y.; Shen, Z.; Qiu, W.; Qian, C.; Zhang, J. Zeb1-induced linc01559 expedites cell proliferation, migration and emt process in gastric cancer through recruiting igf2bp2 to stabilize zeb1 expression. Cell Death Dis. 2021, 12, 349. [Google Scholar] [CrossRef] [PubMed]
  189. Ye, Y.; Gu, J.; Liu, P.; Wang, H.; Jiang, L.; Lei, T.; Yu, S.; Han, G.; Wang, Z. Long non-coding rna spry4-it1 reverses cisplatin resistance by downregulating mpzl-1 via suppressing emt in nsclc. Onco Targets Ther. 2020, 13, 2783–2793. [Google Scholar] [CrossRef] [Green Version]
  190. Zhou, M.; Zhang, X.Y.; Yu, X. Overexpression of the long non-coding rna spry4-it1 promotes tumor cell proliferation and invasion by activating ezh2 in hepatocellular carcinoma. Biomed. Pharmacother. 2017, 85, 348–354. [Google Scholar] [CrossRef]
  191. Shen, F.; Cai, W.S.; Feng, Z.; Chen, J.W.; Feng, J.H.; Liu, Q.C.; Fang, Y.P.; Li, K.P.; Xiao, H.Q.; Cao, J.; et al. Long non-coding rna spry4-it1 pormotes colorectal cancer metastasis by regulate epithelial-mesenchymal transition. Oncotarget 2017, 8, 14479–14486. [Google Scholar] [CrossRef]
  192. Fan, M.J.; Zou, Y.H.; He, P.J.; Zhang, S.; Sun, X.M.; Li, C.Z. Long non-coding rna spry4-it1 promotes epithelial-mesenchymal transition of cervical cancer by regulating the mir-101-3p/zeb1 axis. BioSci. Rep. 2019, 39, BSR20181339. [Google Scholar] [CrossRef] [Green Version]
  193. Zhao, L.; Jiang, L.; Zhang, M.; Zhang, Q.; Guan, Q.; Li, Y.; He, M.; Zhang, J.; Wei, M. Nf-kappab-activated spry4-it1 promotes cancer cell metastasis by downregulating tceb1 mrna via staufen1-mediated mrna decay. Oncogene 2021, 40, 4919–4929. [Google Scholar] [CrossRef] [PubMed]
  194. Sui, X.; Hu, N.; Zhang, Z.; Wang, Y.; Wang, P.; Xiu, G. Asmtl-as1 impedes the malignant progression of lung adenocarcinoma by regulating sat1 to promote ferroptosis. Pathol. Int. 2021, 71, 741–751. [Google Scholar] [CrossRef]
  195. Sun, J.; Li, X.; Yu, E.; Liu, J.; Sun, L.; He, Q.; Lu, Q. A novel tumor suppressor asmtl-as1 regulates the mir-1228-3p/sox17/beta-catenin axis in triple-negative breast cancer. Diagn. Pathol. 2021, 16, 45. [Google Scholar] [CrossRef]
  196. Hou, C.; Sun, F.; Sun, M. Long non-coding rna asmtl-as1 deteriorates the oncogenicity of osteosarcoma by decoying microrna-342-3p and consequently raising adam9 expression. Biochem. Biophys. Res. Commun. 2021, 579, 89–96. [Google Scholar] [CrossRef]
  197. Feng, L.; Wang, R.; Wang, Y.; Shen, X.; Shi, Q.; Lian, M.; Ma, H.; Fang, J. Silencing long non-coding rna dlx6-as1 or restoring microrna-193b-3p enhances thyroid carcinoma cell autophagy and apoptosis via depressing hoxa1. J. Cell Mol. Med. 2021, 25, 9319–9330. [Google Scholar] [CrossRef] [PubMed]
  198. Lee, Y.J.; Jang, B.K. The role of autophagy in hepatocellular carcinoma. Int. J. Mol. Sci. 2015, 16, 26629–26643. [Google Scholar] [CrossRef] [Green Version]
  199. Wang, Y.; Zhang, L.; Zheng, X.; Zhong, W.; Tian, X.; Yin, B.; Tian, K.; Zhang, W. Long non-coding rna linc00161 sensitises osteosarcoma cells to cisplatin-induced apoptosis by regulating the mir-645-ifit2 axis. Cancer Lett. 2016, 382, 137–146. [Google Scholar] [CrossRef]
  200. Xu, M.; Zhou, K.; Wu, Y.; Wang, L.; Lu, S. Linc00161 regulated the drug resistance of ovarian cancer by sponging microrna-128 and modulating mapk1. Mol. Carcinog. 2019, 58, 577–587. [Google Scholar] [CrossRef]
  201. Zanin, R.; Pegoraro, S.; Ros, G.; Ciani, Y.; Piazza, S.; Bossi, F.; Bulla, R.; Zennaro, C.; Tonon, F.; Lazarevic, D.; et al. Hmga1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of foxm1. J. Exp. Clin. Cancer Res. 2019, 38, 313. [Google Scholar] [CrossRef] [Green Version]
  202. Messineo, S.; Laria, A.E.; Arcidiacono, B.; Chiefari, E.; Luque Huertas, R.M.; Foti, D.P.; Brunetti, A. Cooperation between hmga1 and hif-1 contributes to hypoxia-induced vegf and visfatin gene expression in 3t3-l1 adipocytes. Front. Endocrinol. 2016, 7, 73. [Google Scholar] [CrossRef]
  203. Zhang, L.; Pan, K.; Zuo, Z.; Ye, F.; Cao, D.; Peng, Y.; Tang, T.; Li, X.; Zhou, S.; Duan, L. Linc01133 hampers the development of gastric cancer through increasing somatostatin via binding to microrna-576-5p. Epigenomics 2021, 13, 1205–1219. [Google Scholar] [CrossRef] [PubMed]
  204. Yang, H.; Qu, H.; Huang, H.; Mu, Z.; Mao, M.; Xie, Q.; Wang, K.; Hu, B. Exosomes-mediated transfer of long noncoding rna linc01133 represses bladder cancer progression via regulating the wnt signaling pathway. Cell Biol. Int. 2021, 45, 1510–1522. [Google Scholar] [CrossRef]
  205. Song, Z.; Zhang, X.; Lin, Y.; Wei, Y.; Liang, S.; Dong, C. Linc01133 inhibits breast cancer invasion and metastasis by negatively regulating sox4 expression through ezh2. J. Cell Mol. Med. 2019, 23, 7554–7565. [Google Scholar] [CrossRef] [Green Version]
  206. Yin, D.; Hu, Z.Q.; Luo, C.B.; Wang, X.Y.; Xin, H.Y.; Sun, R.Q.; Wang, P.C.; Li, J.; Fan, J.; Zhou, Z.J.; et al. Linc01133 promotes hepatocellular carcinoma progression by sponging mir-199a-5p and activating annexin a2. Clin. Transl Med. 2021, 11, e409. [Google Scholar] [CrossRef]
  207. Liu, S.; Xi, X. Linc01133 contribute to epithelial ovarian cancer metastasis by regulating mir-495-3p/tpd52 axis. Biochem. Biophys. Res. Commun. 2020, 533, 1088–1094. [Google Scholar] [CrossRef]
  208. Huang, C.S.; Chu, J.; Zhu, X.X.; Li, J.H.; Huang, X.T.; Cai, J.P.; Zhao, W.; Yin, X.Y. The c/ebpbeta-linc01133 axis promotes cell proliferation in pancreatic ductal adenocarcinoma through upregulation of ccng1. Cancer Lett. 2018, 421, 63–72. [Google Scholar] [CrossRef]
  209. Wang, Y.; Li, Z.; Xu, P.; Huang, L.; Tong, J.; Huang, H.; Meng, A. Angiomotin-like2 gene (amotl2) is required for migration and proliferation of endothelial cells during angiogenesis. J. Biol. Chem 2011, 286, 41095–41104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Sahoo, S.; Emanueli, C. Exosomes in diabetic cardiomyopathy: The next-generation therapeutic targets? Diabetes 2016, 65, 2829–2831. [Google Scholar] [CrossRef] [Green Version]
  211. Catalano, M.; O’Driscoll, L. Inhibiting extracellular vesicles formation and release: A review of ev inhibitors. J. Extracell. Vesicles 2020, 9, 1703244. [Google Scholar] [CrossRef] [Green Version]
  212. Dai, E.; Han, L.; Liu, J.; Xie, Y.; Kroemer, G.; Klionsky, D.J.; Zeh, H.J.; Kang, R.; Wang, J.; Tang, D. Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic kras protein. Autophagy 2020, 16, 2069–2083. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Exosomal circRNAs secreted from drug-resistant cancer cells, hypoxic cancer cells, CAFs, and hypoxic CAFs. CircRNAs (black), and their biological functions (blue) are indicated in rounded rectangles. Arrows indicate the upregulation (↑) and downregulation (↓).
Figure 1. Exosomal circRNAs secreted from drug-resistant cancer cells, hypoxic cancer cells, CAFs, and hypoxic CAFs. CircRNAs (black), and their biological functions (blue) are indicated in rounded rectangles. Arrows indicate the upregulation (↑) and downregulation (↓).
Ijms 23 00930 g001
Figure 2. Exosomal circRNAs released from cancer cells and their effects in recipient cells. CircRNAs (black), and their biological functions (blue) are denoted in rounded rectangles. Arrows indicate the upregulation (↑) and downregulation (↓).
Figure 2. Exosomal circRNAs released from cancer cells and their effects in recipient cells. CircRNAs (black), and their biological functions (blue) are denoted in rounded rectangles. Arrows indicate the upregulation (↑) and downregulation (↓).
Ijms 23 00930 g002
Figure 3. Exosomal lncRNAs derived from various cell types. LncRNAs (black), and their biological functions (blue) are indicated in rounded rectangles. An arrow (↑) indicates the upregulation.
Figure 3. Exosomal lncRNAs derived from various cell types. LncRNAs (black), and their biological functions (blue) are indicated in rounded rectangles. An arrow (↑) indicates the upregulation.
Ijms 23 00930 g003
Figure 4. Exosomal lncRNAs derived from cancer cells and their biological functions in endothelial cells, macrophages, T cells, and fibroblasts. Rounded rectangles denote lncRNAs (black) and their functions (blue). Arrows indicate the upregulation (↑) and downregulation (↓).
Figure 4. Exosomal lncRNAs derived from cancer cells and their biological functions in endothelial cells, macrophages, T cells, and fibroblasts. Rounded rectangles denote lncRNAs (black) and their functions (blue). Arrows indicate the upregulation (↑) and downregulation (↓).
Ijms 23 00930 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yun, B.D.; Choi, Y.J.; Son, S.W.; Cipolla, G.A.; Berti, F.C.B.; Malheiros, D.; Oh, T.-J.; Kuh, H.J.; Choi, S.Y.; Park, J.K. Oncogenic Role of Exosomal Circular and Long Noncoding RNAs in Gastrointestinal Cancers. Int. J. Mol. Sci. 2022, 23, 930. https://doi.org/10.3390/ijms23020930

AMA Style

Yun BD, Choi YJ, Son SW, Cipolla GA, Berti FCB, Malheiros D, Oh T-J, Kuh HJ, Choi SY, Park JK. Oncogenic Role of Exosomal Circular and Long Noncoding RNAs in Gastrointestinal Cancers. International Journal of Molecular Sciences. 2022; 23(2):930. https://doi.org/10.3390/ijms23020930

Chicago/Turabian Style

Yun, Ba Da, Ye Ji Choi, Seung Wan Son, Gabriel Adelman Cipolla, Fernanda Costa Brandão Berti, Danielle Malheiros, Tae-Jin Oh, Hyo Jeong Kuh, Soo Young Choi, and Jong Kook Park. 2022. "Oncogenic Role of Exosomal Circular and Long Noncoding RNAs in Gastrointestinal Cancers" International Journal of Molecular Sciences 23, no. 2: 930. https://doi.org/10.3390/ijms23020930

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