Next Article in Journal
A Novel C1-Esterase Inhibitor Oxygenator Coating Prevents FXII Activation in Human Blood
Next Article in Special Issue
Role of microRNA/Epithelial-to-Mesenchymal Transition Axis in the Metastasis of Bladder Cancer
Previous Article in Journal
Synthesis and Anticancer Activity of Dimeric Polyether Ionophores
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

MicroRNAs and Their Influence on the ZEB Family: Mechanistic Aspects and Therapeutic Applications in Cancer Therapy

1
Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran
2
Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
3
Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
4
Department of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
5
General Practitioner, Kerman University of Medical Sciences, Kerman 7616913555, Iran
6
Laboratory for Stem Cell Research, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
7
Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417414418, Iran
8
Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur 9318614139, Iran
9
Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey
10
Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzia, Istanbul 34956, Turkey
11
Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
12
Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7619813159, Iran
*
Authors to whom correspondence should be addressed.
Biomolecules 2020, 10(7), 1040; https://doi.org/10.3390/biom10071040
Submission received: 13 May 2020 / Revised: 6 July 2020 / Accepted: 10 July 2020 / Published: 12 July 2020
(This article belongs to the Special Issue EMT and Cancer)

Abstract

:
Molecular signaling pathways involved in cancer have been intensively studied due to their crucial role in cancer cell growth and dissemination. Among them, zinc finger E-box binding homeobox-1 (ZEB1) and -2 (ZEB2) are molecules that play vital roles in signaling pathways to ensure the survival of tumor cells, particularly through enhancing cell proliferation, promoting cell migration and invasion, and triggering drug resistance. Importantly, ZEB proteins are regulated by microRNAs (miRs). In this review, we demonstrate the impact that miRs have on cancer therapy, through their targeting of ZEB proteins. MiRs are able to act as onco-suppressor factors and inhibit the malignancy of tumor cells through ZEB1/2 down-regulation. This can lead to an inhibition of epithelial-mesenchymal transition (EMT) mechanism, therefore reducing metastasis. Additionally, miRs are able to inhibit ZEB1/2-mediated drug resistance and immunosuppression. Additionally, we explore the upstream modulators of miRs such as long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), as these regulators can influence the inhibitory effect of miRs on ZEB proteins and cancer progression.

1. Introduction

Epithelial-mesenchymal transition (EMT) process was first introduced by Greenburg and his colleagues in 1982 [1]. To date, three major types of EMT have been identified: type I EMT, which occurs during embryogenesis, type II EMT, which is activated during wound healing, tissue regeneration and organ fibrosis, and type III EMT, which occurs during metastasis of cancer cells [2]. EMT is the process of cellular transition wherein epithelial cells are bio-transformed into mesenchymal cells with fibroblast-like properties [3,4,5,6]. In the EMT mechanism, cadherins play a significant role. Cadherins promote cell-cell adhesion and are located at the adherens’ junctions. There are different kinds of cadherins including E, N, P, VE, proto, desmosomal, and FAT cadherins, but N-cadherin and E-cadherin are the most important ones in EMT mechanism. A decrease in E-cadherin levels, and an increase in N-cadherin levels lead to stimulation of EMT, and enhanced migratory ability of cancer cells [7,8]. Additionally, upon EMT stimulation, morphology changes and alterations in cytoskeleton occur in cells and affect their migratory ability and adhesion to neighboring cells. These molecular and structural changes promote the dissemination of cells into other sites [9]. Essentially, this increased cell migration is beneficial in normal cells to accelerate physiological processes such as wound healing and embryogenesis. It has been reported that EMT occurs to provide the required flexibility for mesoderm and neural crest formations [10,11]. However, cancer cells can exploit the EMT mechanism for metastasis to distant sites [12,13,14]. There is increased attention towards the EMT mechanism in cancer therapy not only because of its contribution toward metastasis, but also due to the fact that the EMT mechanism can trigger chemoresistance of cancer cells, and decrease sensitivity to apoptosis [15,16]. Therefore, understanding the molecular pathways regulating EMT is a crucial in the field of cancer studies.
EMT is regulated by a variety EMT-promoting transcription factors (EMT-TFs) such as Snail, Slug, Twist, TBX-2, SIX, transforming growth factor--β (TGF-β), and Zinc finger E-box-binding homeobox protein (ZEB) [17]. These upstream EMT-TFs can induce EMT and promote the biotransformation of cells from epithelial phenotype into mesenchymal phenotype by affecting levels of cadherins. Different studies have shown the involvement of ZEB proteins in modulating EMT during normal development and in pathological conditions [18,19,20,21]. Our aim in the present review is to 1) show that ZEB proteins are able to regulate metastasis of cancer cells via affecting EMT, 2) understand how different microRNAs (miRs) can regulate the ZEB/EMT axis, and 3) demonstrate how other upstream mediators can regulate the miR/ZEB/EMT axis.

2. ZEB Family

The ZEB family, which was first discovered in Drosophila melanogaster, consists of two key members ZEB1 and ZEB2 [22]. Both ZEB1 and ZEB2 possess the amino-terminal (NZF) and carboxy-terminal zinc finger cluster (CZF), thereby allowing them to bind to regulatory DNA sequences in their target promoters [23,24,25]. This has led to their involvement in different biological events, such as embryogenesis, hematopoiesis, and more importantly, EMT. In fact, ZEB proteins are well-known due to their ability in stimulation of EMT [20]. In this section, we provide an overview of ZEB1 and ZEB2 proteins to shed some light on their role in cancer cells.

2.1. ZEB1

ZEB1 gene is located on chromosome 10p11.2, and its protein is made up of two zinc-finger clusters at N- and C-terminal ends, while the middle portion of the ZEB1 protein contains three distinct parts including a homeodomain, a Smad interaction domain and a C-terminal binding protein (CtBP). The CtBP is involved in the regulation of ZEB1 function [26,27]. Primarily, the zinc-finger clusters allow ZEB1 to bind to E-boxes. ZEB1 regulates its downstream effectors through binding to E-promoter DNA sequence (5′-CANNTG-3′) [28]. Various publications have also highlighted ZEB1′s association with enhanced viability and invasiveness of cancer cells. In colorectal cancer (CRC) cells, it was found that tumor suppressor death domain-associated protein (DAXX) is able to prevent ZEB1 modulation on E-cadherin to inhibit the invasion and proliferation of tumor cells. Down-regulation of DAXX enhanced ZEB-1 suppression of E-cadherin, leading to the enhanced proliferation and malignancy of cancer cells [29]. It has also been highlighted that EMT may contribute to chemoresistance of cancer cells [30,31]. In pancreatic cancer, Rho associated coiled coil containing protein kinase 2 (ROCK2) enhances the expression of ZEB1. This in turn leads to ZEB1-mediated EMT induction, which contributes to gemcitabine resistance in pancreatic cancer cells [32]. In CRC cells, TCF4 enhances expression of ZEB1 to promote stemness and migration of cancer cells, thereby promoting chemotherapy resistance [33]. In prostate cancer cells, ZEB1 stimulates up-regulation of ATP-binding cassette subfamily C member 10 (MRP4) to export docetaxel out of cancer cells, resulting in their decreased sensitivity to chemotherapy [34]. These studies support the modulation of ZEB1, and highlights that it may be beneficial in enhancing the efficacy of chemotherapy and in reducing the migratory ability of cancer cells. Overall, ZEB1 is an important mediator to enhance the invasion and proliferation of tumor cells. More importantly, ZEB1 may significantly reduce the efficiency of chemotherapy.

2.2. ZEB2

ZEB2 is another member of ZEB family and is located on chromosome 2q22.3 [25]. Structurally, the N-terminal end of ZEB2 consists of four zinc fingers, while the C-terminal end has three zinc fingers [25]. Similar to ZEB1, ZEB2 appears to play a crucial role in migration and invasion. In non-small cell lung cancer (NSCLC), MDM2 binding protein (MTBP) behaves as an oncogene to increase EMT through ZEB2 up-regulation [35]. This in turn enhanced the migration and metastasis of NSCLC tumor cells. In bladder cancer, it was found that indoleamine-2,3-dioxygenase-1 (IDO1) induces ZEB2 overexpression, which in turns increases the viability and proliferation of cancer cells [36]. ZEB2 has also been found to increase the expression of ETS proto-oncogene 1 (ETS1) to up-regulate other EMT proteins such as matrix metalloproteinase 9 (MMP-9) and Twist [37]. Importantly, ZEB2 is also capable of inducing chemoresistance via EMT activation. Phosphatidylinositol 3-kinase (PI3K)/protein kinase-B (Akt) pathway is a down-stream pathway of ZEB2 that induces EMT by reducing the level of E-cadherin protein, leading to the generation of cisplatin resistance in NSCLC cells [38] In all, ZEB2 appears to mediate EMT, and may be a potential therapeutic target in cancer treatment.

3. MicroRNAs

Non-coding RNAs (ncRNAs) comprise a huge part of human genome and are involved in various molecular pathways and processes [39,40,41,42,43,44,45,46,47,48]. They are divided into two characteristic groups: house-keeping and regulatory molecules [49]. They play a remarkable role in vital biological processes such as apoptosis, autophagy, differentiation, cell cycle, proliferation, and migration by targeting various down-stream molecular pathways [50,51]. Additionally, they contribute to the transcription, post-transcriptional modifications, and signal transduction networking. MiRs are house-keeping molecules belonging to the small nucleolar RNAs (SnoRNAs) family [52,53,54]. In this section, we first provide an introduction about miRs and their biosynthesis, followed by a highlight of their potential roles in cancer.
MiRs are single-stranded RNA molecules with a length of 19–24 nucleotides and may possess regulatory functions [55,56]. In total, 60% of all human genome has a binding site for miRs. This highlights the influence of miRs as they control many cellular processes and their dysregulation is related to the development of diseases [57,58,59,60,61]. MiRs are capable of post-transcriptional regulation of their target through RNA interference. These small RNA molecules bind to their target via 3′-untranslated region (3′-UTR). It has been demonstrated that the level of miRs has a negative relationship with the expression of their down-stream targets [62,63,64]. Moreover, one miR is able to target more than one messenger RNA (mRNA), again highlighting their widespread influence in many cellular processes [65]. In the synthesis of miR, a primary miR (pri-miR) is first produced by the action of RNA polymerase. The pri-miR is long with more than 500 nucleotides. It is then processed by Drosha/Pasha and DICER1 proteins, which cleave the pri-miR to generate a mature miR. Next, the mature miR is incorporated in a complex to form miR-RNA-induced silencing complex assembly [66,67,68,69,70].

MicroRNAs in Cancer Metastasis

When focusing on the cancer context, miRs can have oncogenic, or tumor suppressing properties. Onco-suppressor miRs that inhibit invasion of cancer cells undergo down-regulation during cancer development. Enhancing the expression of such miRs can aid in the down-regulation of factors involved in migration of cancer cells such as PRMT5 [71]. Additionally, MiR-506-3p up-regulation considerably reduces the viability and proliferation of ovarian cancer cells and stimulates apoptotic cell death. Investigation of underlying molecular pathways shows that miR-506-3p inhibits Akt/Forkhead box O3 (FOXO3a) by inhibition of sirtuin 1 (SIRT1) [72]. Elevating the expression of miR-506-3p is a potential strategy in ovarian cancer treatment. In the gastric cancer model, it was also observed that a reverse relationship between miR-612 and nin one binding protein (NOB1) helped reduce the migration and invasion of cervical cancer cells [73]. Similarly, in pancreatic cancer, it was also found that overexpression miRs could lead to better prognosis. Enhancing the expression of miR-519 appears to sensitize pancreatic cancer cells to apoptosis and inhibits their proliferation and migration. This miR prevents the activation of programmed death ligand 1 (PD-L1), under hypoxic conditions to suppress tumorigenesis [74]. In all, various studies have shown that miRs are efficient upstream mediators that target various molecular pathways. Enhancing the expression of tumor suppressor miRs may prove to be an advantageous strategy and extensive research is currently being performed to exploit this strategy [74,75,76,77,78]. Conversely, oncogenic miRs are able to elevate the malignancy and proliferation of cancer cells and are associated with poor prognosis. Their downregulation is of interest in cancer therapy [79,80]. For instance, miR-424-5p is able to induce anoikis resistance to promote migratory ability of cancer cells [81]. The targeting of miRs may therefore be considered a promising candidate in cancer therapy. Interestingly, EMT-TFs are considered as potential down-stream targets of miRs in cancer metastasis. MiR-582-3p and miR-582-5p suppress migration of cancer cells via down-regulation of TGF-β in cancer cells [82]. This concurs that miRs can play a significant role in the regulation of metastasis via targeting different pathways and mechanisms. In the following sections, we focus on the regulation of ZEB proteins by miRs and their association with cancer metastasis and chemoresistance.

4. MicroRNA, ncRNA, and ZEB: Role in EMT and Cancer Metastasis

This section specifically demonstrates the impact that miRs have on cell migration and invasion, through their targeting of ZEB proteins. Upstream modulators of miRs such as lncRNAs and circRNAs are also extensively discussed. As mentioned, miRs are able to act as both onco-suppressor as well as promoter of cancer dissemination. Particularly, they are able to exert these effects through their modulation of ZEB proteins, to result in changes in the EMT mechanism. For instance, it appears that miR-200c plays a dual role in cancer therapy. Some studies have demonstrated that miR-200c elevates the viability and proliferation of tumor cells, while another study showed that miR-200c sensitizes cancer cells into chemotherapy by targeting neurophilin 1 and reducing cancer malignancy [83,84,85,86]. It is believed that miR-200c exerts an inhibitory impact on TGF-β-mediated EMT through down-regulation of both ZEB1 and ZEB2 proteins [87].

4.1. ZEB1

4.1.1. MiRs as Modulators of ZEB1

Breast cancer is one of the leading causes of death among females [88]. Importantly, metastasis is a prevalent concern in breast cancer development [89]. Notably, ZEB1 has been identified as a key player in migration of breast cancer cells. MiR-200a was found to down-regulate ZEB1 in suppressing cancer cell migration. It appears that by down-regulating ZEB1, miR-200a can enhance E-cadherin levels and inhibit EMT [90]. This study demonstrates that the relationship between miRs and ZEB proteins is crucial in the regulation of metastasis. Additionally, other onco-suppressor miRs have also been identified to regulate the migration of cancer cells. MiR-1271 have been found to significantly decrease the viability and proliferation of tumor cells [91,92,93]. In ovarian cancer cells, miR-1271 inhibits EMT via ZEB1 down-regulation (binding into 3′-UTR), leading to the decreased viability, proliferation, invasion, and migration of tumor cells. As a consequence of ZEB1 down-regulation by miR-1271, levels of E-cadherin undergo up-regulation, accompanied by a decrease in the levels of N-cadherin [94]. In gastric cancer cells, expression of miR-203 undergoes down-regulation, resulting in an up-regulation of ZEB1 and resistance of cancer cells to radiotherapy. It has been suggested that enhancing the expression of miR-203 is a potential strategy in sensitizing cancer cells to radiotherapy, since miR-203 binds to the 3′-UTR of ZEB1 to repress its expression. This then results in a decrease in the malignancy of cancer cells and an increased sensitivity to radiotherapy [95]. Additionally, inhibition of ZEB1 by miRs such as miR-101-3p, miR-525-5p and miR-186-5p is also corelated with a diminution in metastasis of cancer cells due to EMT inhibition by E-cadherin up-regulation [96,97,98]. Taken together, the miR/ZEB1 axis is an important factor in cancer dissemination and may be an important and relevant target in cancer therapeutics. A newly published study has investigated efficacy of ursolic acid in affecting miR-220c/ZEB1 axis. Ursolic acid enhances expression of miR-200c, as an onco-suppressor factor that, in turn, reduces expression of TGF-β1, providing the condition for down-regulation of ZEB1 and inhibiting metastasis of CRC cells [97].
As aforementioned, EMT-TFs such as TGF-β can stimulate EMT. ZEB1 engages in a feedback loop with TGF-β and miR, thereby promoting metastasis of cancer cells. Normally, miR-33a-5p suppresses TGF-β to inhibit ZEB1 activation, leading to suppression of metastasis. However, in cancer conditions, TGF-β and ZEB1 cooperate with each other to promote migration of cancer cells. TGF-β can enhance copy numbers of ZEB1, while ZEB1 suppresses miR-33a-5p, an inhibitor of TGF-β signaling. This cooperation between ZEB1 and TGF-β leads to inhibition of miR-33a-5p, and stimulation of EMT [99]. This once again highlights that onco-suppressor miRs may suppress ZEB1 via affecting other EMT-TFs such as TGF-β, and that ZEB1 can form a negative feedback loop with onco-suppressor miRs in promoting metastasis of cancer cells.
The Akt/mammalian target of rapamycin (mTOR) signaling pathway is another pathway that is commonly deregulated in cancer [100,101,102]. Phosphorylated Akt can induce mTOR to promote the motility and invasion of tumor cells [103,104,105,106,107,108]. It appears that miR-205 is able to target the Akt/mTOR signaling pathway to regulate malignancy and progression of cancer cells [109]. By suppressing Akt/mTOR signaling pathway, miR-708 acts as an anti-tumor agent to inhibit ZEB1, leading to the suppressing EMT mechanism [110]. Finally, miR-126 was also found to inhibit ZEB1 to suppress MMP-2, MMP-9, and oncogenic JAK2/STAT3 signaling pathway, leading to the reduced migration and metastasis of cervical cancer cells [111].
Additionally, Wnt signaling pathway contributes to cancer cell growth and dissemination. Abnormal expression of Wnt signaling pathway can be observed in cancers [112,113,114,115,116,117]. Wnt/β-catenin signaling pathway can promote EMT through ZEB1 up-regulation to elevate the invasion and malignancy of tumor cells. Enhancing the expression of miR-33b effectively inhibits Wnt/β-catenin/ZEB1 axis to suppress cancer malignancy through EMT inhibition [118]. Similarly, miR-200a is capable of decreasing gastric adenocarcinoma invasion via down-regulation of Wnt/β-catenin and subsequent suppressing of ZEB1 and ZEB2 [119]. In malignant meningioma, miR-4652-3p down-regulates the expression of ZEB1 by suppressing Wnt and nuclear translocation of β-catenin. Conversely, lncRNA LINC00702 can activate Wnt/β-catenin signaling pathway by sponging miR-4652-3p to induce ZEB1 and promote the metastasis and invasion of malignant meningioma [120]. These studies highlight the fact that firstly, Wnt can promote metastasis of cancer cells via ZEB1 up-regulation; secondly, the Wnt/ZEB1 axis can be inhibited by onco-suppressor miRs; thirdly, miRs affect both expression of Wnt and nuclear translocation of β-catenin; and finally, lncRNAs can regulate miR/Wnt/ZEB1 axis. The mediation of the miR/ZEB1 axis by lncRNAs will be more extensively discussed in the next section.

4.1.2. LncRNAs as Modulators of miR/ZEB1 Axis

Long non-coding RNAs (lncRNAs) belong to a category of ncRNAs with regulatory effect on biological events [121,122]. They consist of at least 200 nucleotides and they are able to function as upstream mediators of miRs [111]. LncRNAs suppress the expression of miRs via acting as competitive endogenous RNA (ceRNA) [123]. The effect of lncRNAs on miR/ZEB1 axis has been investigated in cancer cells. For instance, miR-429 was found to inhibit EMT through ZEB1 inhibition and its expression is typically down regulated in pancreatic cancer cells. MiR-429 can be regulated by lncRNA XIST, which is up-regulated in pancreatic cancer cells to reduce miR-429 levels. This in turn increases ZEB1 expression and promotes EMT. Additionally, through targeting the miR-429/ZEB1 axis, XIST also affects morphology of cancer cells, such that silencing XIST results in a change in cell morphology, from the original spindle shape to a rounded one [124]. In another instance, LncRNA IUR is a onco-suppressor factor that has shown a great capability in suppressing tumorigenesis [125]. LncRNA IUR can inhibit the migration and metastasis of prostate cancer cells via enhancing the expression of miR-200, which in turn inhibits ZEB1 [126].
Conversely, lncRNA TDRG1 is an oncogenic factor that is able to regulate miRs in cancer cells [127,128]. In lung cancer cells, TDRG1 enhances the migration, metastasis, and malignancy of cancer cells by promoting ZEB1 expression through miR-873-5p down-regulation [129]. LncRNA TTN-AS1 is also considered an oncogenic factor that induces ZEB1 through miR-4677-3p down-regulation, leading to the enhanced migration and metastasis of NSCLC cells [130]. LncRNA (Nuclear Enriched Abundant Transcript 1) NEAT1 contributes to enhancing the malignancy of cancer cells [131]. It has been demonstrated that NEAT1 can target miRs to regulate cancer proliferation and migration [132]. In breast cancer cells, NEAT1 reduces the expression of miR-448 to elevate the metastasis and invasion of cancer cells through ZEB1 up-regulation [133]. LncRNA TP73-AS1 reduces the expression of miR-200a to up-regulate ZEB1, leading to the enhanced progression and malignancy of tumor cells. There appears to be a feedback loop, wherein TP73-AS1-activated ZEB1 has a stimulatory effect on the expression of TP73-AS1 to enhance its inhibitory activity on miR-200a, leading to increased induction of ZEB1 [90].
In renal cell carcinoma (RCC), miR-429 typically reduces the expression of ZEB1 to suppress RCC progression. However, miR-429 can be inhibited by SCAMP1, a lncRNA that is activated by oxidative stress [134]. This highlights that stimulation of oxidative stress negatively impacts cancer therapy. Generally, it is believed that enhancing level of oxidative stress can lead to a reduction in the viability of cancer cells by predisposing them into apoptosis [135,136]. However, as mentioned, increasing levels of oxidative stress may also activate lncRNAs involved in cancer metastasis. Therefore, careful considerations are warranted before using oxidative stress in cancer therapy, keeping in mind the possible adverse effects of this treatment method.
MiR-139-5 was also found to suppress ZEB1 levels. However, lncRNA human leukocyte antigen (HLA) complex 5 (HCP5) is able to induce ZEB1 and EMT by suppressing miR-139-5 [137]. LncRNA MAGI2-AS3 has also been explored in cancer and it appears that MAGI2-AS3 is able to modulate molecular pathways such as Fas/FasL to suppress breast cancer, bladder cancer, and hepatocellular carcinoma [138,139]. Particularly, in gastric cancer cells, miR-141/200a diminishes the invasion and migration of tumor cells via suppressing ZEB1. LncRNA MAGI2-AS3 down-regulates the expression of miR-141/200a to induce ZEB1, leading to the stimulation of EMT and enhanced invasion of tumor cells [125].
LncRNA LINC00511 is located on chromosome 17q24.3 and has been associated with increased malignancy in cancer [140]. In glioblastoma (GBM) cells, miR-524-5p inhibits ZEB1 to suppress GBM invasion and migration. LINC00511 has been found to decrease the expression of miR-524-5p to up-regulate YB1 [141]. YB1 is a transcription factor that can enhance the expression of ZEB1 in cancer [128]. The inhibition of miR-524-5p by LINC00511 promotes ZEB1 expression through YB1 up-regulation, leading to enhanced EMT and malignancy of GBM cells [141]. These studies again demonstrate that lncRNAs can disrupt inhibitory effects of miRs on ZEB1 to promote metastasis of cancer cells. In glioma cells, miR-205-3p inhibits TGF-β, while lncRNA linc00645 functions as an upstream mediator and activates TGF-β via suppressing miR-205-3p, leading to an increase in ZEB1 levels and subsequent EMT activation [137].
LncRNA MALAT1 located on the chromosome 11q13, is also suggested to be involved in elevating the malignancy of cancer cells. A variety of factors act as down-stream mediators for lncRNA MALAT1 and it appears that MALAT1 is capable of targeting miRs in cancer cells [105,142,143]. MALAT1 was found to enhance the expression of ZEB1 through miR-143-3p down-regulation, resulting in elevated migration and metastasis of tumor cells [144]. Another downstream target of MALAT1 is miR-429, which is considered as a potential biomarker for diagnosis of different cancers [145]. MALAT1 was found to inhibit miR-429 to accelerate the malignancy and invasion of cervical cancer cells [146]. Notably, miR-429 can inhibit the metastasis of cancer cells and stimulate apoptotic cell death through ZEB1 down-regulation [110]. Interestingly, it has been demonstrated that fine particulate matter (PM2.5, aerodynamic diameter, 2.5 μm) is able to induce oxidative stress, inflammation, genetic mutations, and DNA damage [147,148]. It has been found that miR-204 can reduce the expression of ZEB1 to suppress EMT. PM2.5 activates MALAT1 via stimulation of NF-κB, as an inflammatory pathway. MALAT1 in turn induces ZEB1 through miR-204 down-regulation to enhance the malignancy and invasion of tumor cells via EMT induction [149]. These two studies demonstrate that lncRNAs can affect more than one downstream miR to mediate ZEB1 levels, and that other molecular pathways such as NF-κB can act as upstream mediator of lncRNA/miR/ZEB1 axis.
HOXA distal transcript antisense RNA (HOTTIP) is located at the distal end of HOXA gene cluster [150]. This lncRNA undergoes abnormal expressions in different cancers and it has been shown that HOTTIP is related to the increased proliferation and progression of cancer cells [120]. It is held that lncRNA HOTTIP down-regulates the expression of miR-101 to elevate ZEB1 levels, leading to an increase in EMT [151]. A study has also shown that miR-205 down-regulates the expression of ZEB proteins and HOXD9 to suppress the malignancy and invasion of cancer cells through EMT inhibition [152]. Finally, it has been demonstrated that lncRNA HOXC-AS2 induces ZEB1 by sponging miR-876-5p, leading to the stimulation of EMT and enhanced migration and invasion of tumor cells [153]. Taken together, the relationship between lncRNAs and miRs in the regulation of ZEB1 in cancer cells are dynamic and complicated, and understanding these pathways is an essential part of effective cancer therapy.

4.1.3. CircRNAs as Modulators of miR/ZEB1 Axis

Circular RNAs (circRNAs) are endogenous, conserved ncRNAs that are sometimes employed as biomarkers for cancer diagnosis [154,155]. Similar to lncRNA, circRNAs are able to modulate the expression of their targets [156]. In lung cancer cells, hsa-circ-0023404 decreases the expression of miR-217 to enhance the expression of its target, ZEB1, leading to the increased migration and invasion of cancer cells [157]. The laryngeal carcinoma is considered as one of the common cancers among head and neck tumors and is mainly diagnosed in elder people [158]. In spite of the low incidence rate, this cancer results in high mortality worldwide [159]. It was discovered that miR-200c is capable of inhibiting ZEB1 to prevent the metastasis and invasion of laryngeal cancer cells. Hsa-circ-005748 up-regulates ZEB1 by sponging miR-200c, leading to the metastasis of these cancer cells [160]. Therefore, inhibition of hsa-circ-005748 may in turn increase miR-200c expression to suppress ZEB1 and cancer metastasis. Similarly, in lung adenocarcinoma (LUAD) cells, miR-665 is able to inhibit cancer metastasis via ZEB1 down-regulation. The circ-TSPAN4 enhances the expression of ZEB1 by miR-665 down-regulation to promote the metastasis of LUAD cells [161]. These studies concur that down-regulation of onco-suppressor miRs in cancer cells may also be mediated by upstream circRNAs. This in turn promotes up-regulation of ZEB1 and enhanced metastasis of cancer cells.

4.2. ZEB2

4.2.1. MiRs as Modulators of ZEB2

MiR-124 is suggested to be an onco-suppressor miR. Recently, an effort has been made to suppress the prostate cancer invasion. It is held that cationic polymer nanoparticles are able to deliver miR-124 in prostate cancer cells to inhibit their proliferation, motility, and colony formation [162]. In TNBC cells, miR-124 effectively decreases the malignancy and invasion of tumor cells by EMT inhibition through ZEB2 down-regulation [163]. Similarly, miR-145 has been widely established as a tumor suppressor. It negatively affects the invasion and migration of thyroid carcinoma cells by down-regulation of NF-κB signaling pathway [164]. Furthermore, lncRNA-ROR down-regulates the expression of miR-145 to remove its inhibitory impact and induce EMT in tumor cells [165]. Importantly, it was found that miR-145 decreases the expression of ZEB2 to inhibit EMT, and consequently, suppress the proliferation, progression, and migration of NSCLC cells [166]. These studies demonstrate that the downregulation of ZEB2 by onco-suppressor miRs can lead to a decrease in the metastasis of cancer cells.
Another onco-suppressor miR is miR-30a. In breast cancer cells, miR-30a suppresses the nuclear translocation of β-catenin to attenuate cancer proliferation and progression, and is associated with favorable prognosis of patients with breast cancer [167]. Furthermore, miR-30a appears to be beneficial in sensitizing cancer cells to chemotherapy via affecting Akt signaling pathway [168]. MiR-30a was found to inhibit ZEB2 to result in a reduction of triple negative breast cancer (TNBC) cells malignancy [169]. Finally, miR-3653 is an onco-suppressor that is down-regulated in hepatocellular carcinoma (HCC) cells [170]. MiR-3653 was found to bind to the 3′-UTR of ZEB2 to diminish its expression, leading to the reduced invasion and malignancy of colon cancer cells [171]. MiR-138-5p uses a same strategy in inhibition of lung adenocarcinoma cell malignancy, by suppressing EMT through ZEB2 inhibition to attenuate metastasis of tumor cells [172].
Osteosarcoma typically has a high recurrence rate and low survival rate [173,174]. Therefore, understanding the pathways involved in malignancy and cancer progression may pave the road for improved treatment of this type of cancer. Investigation of molecular pathways has shown that miR-101 up-regulation inhibits ZEB2 and affects proliferation and metastasis of osteosarcoma cells [175]. Unfortunately, miR-101 is down-regulated in osteosarcoma cells compared to the normal cells. Enhancing the expression of miR-101 may reduce malignancy and progression of osteosarcoma cells.

4.2.2. LncRNAs as Modulators of miR/ZEB2 Axis

In the previous section, we demonstrated that lncRNAs are able to function as ceRNA in affecting miR expression. Notably, increasing evidence has also demonstrated that lncRNAs can effectively target ZEB2 via affecting miRs. For instance, LncRNA HOTAIRM1, which has dual properties as it interacts with both onco-suppressor and oncogenic miRs. HOTAIRM1 is located on human HOXA gene cluster and suggested to be involved in myeloid cell development [176]. A newly published article has shown the anti-tumor activity of lncRNA HOTAIRM1 by up-regulation of ARHGAP24 through miR-106a-5p inhibition [177]. However, it has been found that HOTAIRM1 is related to the elevated migration and invasion of tumor cells [178]. It is held that lncRNA HOTAIRM1 diminishes the expression of miR-873-5p to induce ZEB2, resulting in an increase in cancer cell proliferation and suppressing apoptotic cell death [179].
MiR-505 is also considered an onco-suppressor miR that interacts with IGF-1 and HMGB1 to suppress the growth and malignancy of tumor cells [180,181,182]. Various studies have demonstrated that lncRNAs such as lncRNA CRAL, LEF-AS1, and DLX6-AS1 are able to target miR-505 in different cancers such as gastric cancer, CRC, and breast cancer [183,184,185]. In cervical cancer, lncRNA CTS was found to target miR-505. MiR-505 down-regulates ZEB2 levels to inhibit EMT and invasion of cervical cancer cells. LncRNA CTS, therefore, stimulates ZEB2-mediated EMT through miR-505 sponging, leading to the enhanced viability, proliferation, and malignancy of cervical cancer cells [186]. Taken together, stimulation of ZEB2 by lncRNAs not only enhances metastasis of cancer cells via EMT induction, but also promotes cell proliferation. This decrease in apoptosis by ZEB2 induction is of importance in chemotherapy, since cancer cells can attain chemoresistance via reducing their sensitivity into chemotherapy-mediated apoptosis. As such, targeting miR/ZEB2 axis may be a promising strategy in cancer therapy, as it increases the sensitivity of cancer cells toward chemotherapy.
In gastric cancer cells, miR-203 diminishes cancer metastasis through ZEB2 down-regulation. LncRNA UCA1 enhances the progression and metastasis of tumor cells through disrupting the miR-203/ZEB2 axis [187]. Particularly, lncRNAs can affect upstream transcription factors of ZEB2 in cancer metastasis. In NSCLC cells, slug was found to behave as an upstream mediator to induce EMT through increasing ZEB2 levels. MiR-218 was able to disrupt the Slug/ZEB2 axis to suppress NSCLC migration. Conversely, miR-218 undergoes down-regulation by lncRNA SNHG12 to stimulate Slug/ZEB2 and promote metastasis of NSCLC cells [188].
Glioma is an intracranial tumor that emanates from neuroglial stem or progenitor cells [189]. Again, this is an alarming cancer with high mortality and morbidity rate [190,191]. The migration and invasion of cancer cells into neighboring cells and tissues reduces the survival time of patients [192,193]. It has been demonstrated that lncRNA SNHG5 can inhibit miR-205-5p expression. Reduced miR-205-5p expression triggers the induction of ZEB2, which in turn enhances the migration ability of tumor cells [194]. Up-regulation of miR-205-5p may therefore be beneficial in reducing glioma malignancy.

4.2.3. CircRNAs as Modulators of miR/ZEB2 Axis

Increasing evidence highlights the role of miR-377 as an onco-suppressor in cancer cells. MiR-377 can target Akt signaling to suppress the proliferation and invasion of tumor cells, and induce cell cycle arrest [195]. Normally, miR-377 reduces the expression of ZEB2. In bladder cancer cells, the expression of miR-377 undergoes down-regulation by circZFR to promote cancer metastasis through ZEB2 stimulation [196]. MiR-653 also appears to be an onco-suppressor miR in bladder cancer cells. CircRNA ciRs-6 reduces miR-653 expression to induce March1, leading to the increased proliferation of tumor cells [110]. MiR-653 is similarly suppressed in breast cancer cells, by another circRNA hsa-circ-0004771. Knockdown of hsa-circ-0004771 sensitizes cancer cells to apoptosis and inhibits their progression through miR-653 up-regulation and subsequent inhibition of ZEB2 [197]. Evidently, ZEB2 induction dually enhances proliferation and metastasis of cancer cells. Hence, targeting the circRNA/miR/ZEB2 axis can pave the way into effective inhibition of proliferation and migration of cancer cells.
In renal cancer, patients typically have poorer survival rates and treatment strategies can be improved [198,199,200]. MiR-153 was found to exert inhibitory impact on ZEB2 expression to suppress renal cancer, while circPCNXL2 stimulates ZEB2 expression via miR-153 sponging to elevate the invasion and proliferation of renal cancer cells [201]. Therefore, decreasing the expression of circPCNXL2 may yield an up-regulation of miR-153 and suppresses ZEB2 expression to eliminate renal cancer.
In all, these studies highlight the extensive influence that miRs have on ZEB proteins (Figure 1 and Figure 2). Across a wide range of cancers, different miRs work to inhibit ZEB and halt cancer progression. Additionally, lncRNAs and circRNAs are able to act as upstream mediators of miRs to affect ZEB2 expression. Through the revealing of these molecular pathways, we may better understand these promising candidates in cancer therapy.

5. MicroRNAs, ZEB, and Their Role in Tumor Resistance

Multidrug resistance (MDR) is a complicated and challenging phenomenon accounting for cross-resistance towards structurally unrelated drugs [202,203]. It is estimated that approximately 70% of solid and hematological tumors demonstrate MDR. This percentage elevates after chemotherapy, since cancer cells are able to switch among molecular pathways to obtain chemoresistance, and frequent application of chemotherapeutic agents speeds up MDR [204,205]. Therefore, when trying to understand the role of miR and ZEB proteins in cancer, it also important to explore how miR’s modulation on ZEB can affect tumor resistance. Particularly, miR is implicated in tumor resistance. For instance, cisplatin is a potential chemotherapeutic agent with the ability of inhibiting the proliferation and viability of various cancers [206]. In ovarian cancer, it has been reported that miR-137 reduces the expression of MCL1 to sensitize tumor cells into cisplatin-induced apoptosis [207]. In this section, we seek to understand how miR modulation on ZEB can contribute to tumor resistance.

5.1. ZEB1

5.1.1. Paclitaxel Resistance

Paclitaxel (PTX) is a chemotherapeutic agent that is frequently employed in cancer therapy to prevent cell proliferation due to its anti-mitotic capabilities [208]. Unfortunately, PTX resistance is an important obstacle, which has reduced the feasibility of this agent [209,210,211,212]. Notably, ZEB1 can promote cancer cells resistance towards PTX, and down-regulation of ZEB1 may be a key toward re-sensitizing cancer cells to PTX chemotherapy [213]. MiR-124-3p suppresses ZEB1 to sensitize gastric cancer cells into PTX therapy. Circular RNA Circ-PVT1 reverses this axis by sponging miR-124-3p and elevating the expression of ZEB1 to induce PTX resistance in gastric cancer cells [214]. LncRNA NEAT1 was also found to mediate PTX resistance in ovarian cancer cells. Normally, miR-194 undergoes up-regulation to inhibit ZEB1 and subsequently, reduce the malignancy and invasion of cancer cells. LncRNA NEAT1 suppresses the inhibitory effect of miR-194 on ZEB1 to induce the resistance of ovarian cancer cells into PTX chemotherapy [215].

5.1.2. Gemcitabine Resistance

Gemcitabine is a chemotherapeutic agent isolated from deoxycytidine, which is frequently applied in the treatment of breast cancer [216]. Gemcitabine triggers cell cycle arrest by binding into DNA or suppressing ribonucleotide reductase [217,218]. It appears that ZEB1 contributes to the gemcitabine resistance in TNBC cells. This study found that ZEB1 associates with Yes associated protein (YAP) to enhance cancer progression and proliferation and induces chemoresistance. Importantly, ZEB1 was found to be a target of miR-873, and that increasing miR-873 expression down-regulates the expression of YAP and ZEB1, and sensitizes tumor cells into gemcitabine therapy [219].

5.1.3. Cisplatin Resistance

Another important factor to consider when exploring acquired tumor resistance is lncRNA, which can regulate miR, to in turn affect ZEB levels. Prostate cancer-associated transcription 1 (PCAT1) undergoes up-regulation in cancer cells to suppress cell death [220]. In gastric cancer cells, PCAT-1 induces the resistance of cancer cells into cisplatin therapy by stimulation of ZEB1 through miR-128 inhibition, leading to the enhanced progression and malignancy of gastric cancer cells [221]. Therefore, targeting the miR/ZEB1 axis may alleviate cisplatin resistance.

5.1.4. 5-Fluorouracil

The most common chemotherapeutic agent in treatment of cancer is 5-FU [222]. Different molecular pathways are involved in resistance into 5-FU, and miRs are key players [223,224]. LncRNA NEAT1 have been found to possess oncogenic activity and enhance the progression and malignancy of cancer cells via targeting miRs such as miR-144-3p and miR-410 [225,226]. In CRC cells, NEAT1 is involved in 5-FU resistance through miR-34a regulation [227] (Figure 3).

5.2. ZEB2

In osteosarcoma, miR-200b diminishes the progression and motility of tumor cells by inhibition of PI3K/Akt and AMPK signaling pathways, leading to the downregulation of vascular endothelial growth factor (VEGF). LncRNA CCAT2 reverses this axis by induction of VEGF through miR-200b inhibition [228]. It is worth mentioning that enhancing the expression of miR-200b is beneficial in sensitizing cancer cells into chemotherapy, so that arrestin domain containing 3 (ARRDC3) elevates the efficacy of chemotherapy in TNBC cells via miR-200b up-regulation [229]. It appears that up-regulation of miR-200b enhances apoptosis in lung cancer cells and remarkably increases the efficacy of chemotherapy [230]. Another member of miR-200 family, known as miR-200c, sensitizes gastric cancer cells to cisplatin and enhances chemotherapeutic efficacy by suppressing ZEB2 expression [215].

6. MicroRNAs Target ZEB Family in Immune Cells

Other than ZEB’s prevalent role in EMT, ZEB’s involvement with the tumor microenvironment and immune system is also crucial in its mediation of cancer dissemination and development. Tumor cells use immunosuppressive cells such as CD4+ T cells to escape from the anti-cancer activity of CD8+ T cells [231,232,233]. Notably, it has been demonstrated that cytotoxic CD8+ tumor infiltrating lymphocytes (CD8+ TILs) are able to eliminate cancer cells [234], while sustained exposure of tumor cells into CD8+ TILs reduces their anti-tumor activity [235]. It is worth mentioning that PD-1/PD-L1 axis may be involved in driving CD8+ T cell exhaustion and therapies targeting PD-L1 have been explored [236,237,238,239,240]. PD-L1 binds to PD-1 to induce apoptotic cell death in CD8+ T cells and ensure the survival of cancer cells [241,242,243,244].
In diffuse large B cell lymphoma (DLBCL) cells, miR-8890-3p is capable of suppressing ZEB1, while lncRNA SNHG14 conversely reduces the expression of miR-8890-3p to activate ZEB1. Consequently, ZEB1 stimulates PD-L1 to protect cancer cells against the cytotoxic effects of immune cells, resulting in promoting the survival and migration of DLBCL cells [245].
In another instance, it has been reported that ZEB1 is an efficient factor in elevating the malignancy of tumor cells, through the induction of PD-L1 expression to enhance the levels of CD8+ T-cell immunosuppression and cancer metastasis. Enhancing the expression of miR-200 disrupts ZEB1 expression to suppress PD-L1 and immunosuppression, resulting in decreased metastasis and invasion of cancer cells [246]. ZEB1 can induce EMT in breast cancer cells via activation of PD-L1. It has been reported that miR-200 overexpression reduces the levels of ZEB1 to inhibit EMT through interfering with PD-L1 activation, as an immunosuppressive factor [247]. Unfortunately, there are currently no reports about the relationship between miRs and ZEB2 in cancer immunotherapy, and further studies can focus on revealing relationship between miR/ZEB2 axis and cancer immunotherapy. Table 1, Table 2, Table 3, Table 4, Table 5 and Table 6 demonstrate the regulation of ZEB1 and ZEB2 by various miRs proteins in mediating cancer metastasis. Upstream mediators of miR such as lncRNAs and circRNAs are also highlighted in Table 1 through Table 6. Figure 4 further summarizes the effect of miR/ZEB axis on immune system.

7. Conclusions

In this article, we provided a comprehensive review about the relationship between miRs and ZEB family in cancer cells and how this relationship affects the progression and metastasis of tumor cells. After miRs discovery, an exponential amount of research has been performed to understand their role in different biological processes such as cell differentiation, apoptosis, and migration. We typically observe aberrant miR expression in cancer cells and restoring the normal expression of miRs may be crucial in cancer therapy. It is also vital to explore the relevance of ZEB1 and ZEB2 proteins in cancer therapy. It has been reported that ZEB proteins are able to enhance the proliferation and malignancy of tumor cells. One of the most important pathways affected by ZEB proteins is the EMT mechanism. It appears that induction of EMT by ZEB proteins not only enhances the progression and metastasis of cancer cells, but also stimulates drug resistance. Therefore, revealing the underlying molecular pathways involved in ZEB regulation can be beneficial for further studies in the field of cancer therapy and elevating the efficacy of chemotherapy. In this review, we also detailed how and which miRs affect ZEB proteins in various cancers. We consolidated the factors that may function as upstream modulators to negatively affect miRs, leading to the induction of ZEB expression. As it is shown in Table 1, Table 2, Table 3, Table 4, Table 5 and Table 6, lncRNAs and circRNAs can act as oncogenic factors. These upstream mediators induce and enhance the expression of ZEB1 and -2 through sponging their target miRs, resulting in an increase in malignancy and invasion of tumor cells. Identification of these factors and further targeting of them can significantly diminish the malignancy of tumor cells and pave the road for the effective cancer therapy. Finally, we highlighted ZEB1’s role in immunosuppression. Through it, we identified a crucial knowledge gap wherein the relationship between miRs, ZEB2, and immune cells in the cancer context is still a mystery. In all, we dissected the different effects of miR on ZEB proteins, which may in turn help us develop better treatment strategies in attenuating metastasis of cancer cells.

Author Contributions

M.A., H.L.A., E.R.M., S.M., V.Z., K.H., S.S., A.Z., and M.N. did the literature review presented in this study and wrote the manuscript. A.Z. and M.N. did the figures. E.R.M., S.M., V.Z., K.H., and S.S. did the tables. R.M. and A.P.K. conceived the idea for this review and directed the entire study. R.M., H.L.A., and A.P.K. finalized this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

A.P.K. is supported by the National Medical Research Council of Singapore. A.P.K. is also supported by the National Medical Research Council of Singapore and the Singapore Ministry of Education under its Research Centres of Excellence initiative to Cancer Science Institute of Singapore, National University of Singapore.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

EMT, epithelial-to-mesenchymal transition; PCD, programmed cell death; MET, mesenchymal-to-epithelial transition; TGF-β, transforming growth factor-beta; EMT-TFs, EMT-promoting transcription factors; ZEB, zinc finger E-box-binding homeobox; CZF, carboxy-terminal zinc finger cluster; CtBP, C-terminal binding protein; CRC, colorectal cancer; DAXX, death domain-associated protein; ROCK2, Rho associated coiled-coil containing protein kinase 2; NSCLC, non-small cell lung cancer; MTBP, MDM2 binding protein; IDO1, indoleamine-2,3-dioxygenase-1; MMP, matrix metalloproteinase; PI3K, phosphatidylinositol 3-kinase; Akt, protein kinase B; ncRNAs, non-coding RNAs; SnoRNAs, small nucleolar RNAs; 3′-UTR, 3′-untranslated region; mRNA, messenger RNA; pri-miR, primary miR; SIRT1, sirtuin 1; FOXO3a, Forkhead box O3; NOB1, nin one binding protein; PD-L1, programmed death ligand 1; OSCC, oral squamous cell carcinoma; mTOR, mammalian target of rapamycin; lncRNAs, long non-coding RNAs; ceRNA, competitive endogenous RNA; NEAT1, Nuclear Enriched Abundant Transcript 1; RCC, renal cell carcinoma; HLA, human leukocyte antigen; HCP5, HLA complex 5; HOTTIP, HOXA distal transcript antisense RNA; α-SMA, α-smooth muscle actin; circRNAs, circular RNAs; LUAD, lung adenocarcinoma; TNBC, triple negative breast cancer; HCC, hepatocellular carcinoma; MDR, multidrug resistance; P-gp, P-glycoprotein; PTX, paclitaxel; YAP, Yes-associated protein; PCAT1, prostate cancer-associated transcription 1; VEGF, vascular endothelial growth factor; ARRDC3, arrestin domain containing 3; CD8+ TILs, CD8+ tumor infiltrating lymphocytes; DLBCL, diffuse large B cell lymphoma.

References

  1. Wu, X.; Xin, Z.; Zou, Z.; Lu, C.; Yu, Z.; Feng, S.; Pan, P.; Hao, G.; Dong, Y.; Yang, Y. SRY-related high-mobility-group box 4: Crucial regulators of the EMT in cancer. In Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2019. [Google Scholar]
  2. Wang, Y.; Zhou, B.P. Epithelial-mesenchymal transition—a hallmark of breast cancer metastasis. Cancer Hallm. 2013, 1, 38–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Diaz, V.M.; de Herreros, A.G. F-box proteins: Keeping the epithelial-to-mesenchymal transition (EMT) in check. In Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2016; pp. 71–79. [Google Scholar]
  4. Kar, R.; Jha, N.K.; Jha, S.K.; Sharma, A.; Dholpuria, S.; Asthana, N.; Chaurasiya, K.; Singh, V.K.; Burgee, S.; Nand, P. A “NOTCH” Deeper into the Epithelial-To-Mesenchymal Transition (EMT) Program in Breast Cancer. Genes 2019, 10, 961. [Google Scholar] [CrossRef] [Green Version]
  5. Seccia, T.; Caroccia, B.; Piazza, M.; Rossi, G.P. The Key Role of Epithelial to Mesenchymal Transition (EMT) in Hypertensive Kidney Disease. Int. J. Mol. Sci. 2019, 20, 3567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Chakraborty, S.; Mir, K.B.; Seligson, N.D.; Nayak, D.; Kumar, R.; Goswami, A. Integration of EMT and cellular survival instincts in reprogramming of programmed cell death to anastasis. Cancer Metastasis Rev. 2020, 39, 553–566. [Google Scholar] [CrossRef] [PubMed]
  7. Wang, P.; Liu, X.; Han, G.; Dai, S.; Ni, Q.; Xiao, S.; Huang, J. Downregulated lncRNA UCA1 acts as ceRNA to adsorb microRNA-498 to repress proliferation, invasion and epithelial mesenchymal transition of esophageal cancer cells by decreasing ZEB2 expression. Cell Cycle 2019, 18, 2359–2376. [Google Scholar] [CrossRef]
  8. Seton-Rogers, S. Epithelial–mesenchymal transition: Untangling EMT’s functions. Nat. Rev. Cancer 2015, 16, 1. [Google Scholar] [CrossRef]
  9. Yilmaz, M.; Christofori, G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 2009, 28, 15–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Hernandez-Martinez, R.; Ramkumar, N.; Anderson, K.V. p120-catenin regulates WNT signaling and EMT in the mouse embryo. Proc. Natl. Acad. Sci. USA 2019, 116, 16872–16881. [Google Scholar] [CrossRef] [Green Version]
  11. Moly, P.K.; Cooley, J.R.; Zeltzer, S.L.; Yatskievych, T.A.; Antin, P.B. Gastrulation EMT is independent of P-cadherin downregulation. PLoS ONE 2016, 11, e0153591. [Google Scholar] [CrossRef] [Green Version]
  12. Zhu, J.; Zheng, Y.; Zhang, H.; Liu, Y.; Sun, H.; Zhang, P. Galectin-1 induces metastasis and epithelial-mesenchymal transition (EMT) in human ovarian cancer cells via activation of the MAPK JNK/p38 signalling pathway. Am. J. Transl. Res. 2019, 11, 3862–3878. [Google Scholar]
  13. Chi, Y.; Wang, F.; Zhang, T.; Xu, H.; Zhang, Y.; Shan, Z.; Wu, S.; Fan, Q.; Sun, Y. miR-516a-3p inhibits breast cancer cell growth and EMT by blocking the Pygo2/Wnt signalling pathway. J. Cell. Mol. Med. 2019, 23, 6295–6307. [Google Scholar] [CrossRef] [PubMed]
  14. Zhou, P.; Li, Y.; Li, B.; Zhang, M.; Liu, Y.; Yao, Y.; Li, D. NMIIA promotes tumor growth and metastasis by activating the Wnt/β-catenin signaling pathway and EMT in pancreatic cancer. Oncogene 2019, 38, 5500–5515. [Google Scholar] [CrossRef] [PubMed]
  15. Liu, W.; Yang, Y.J.; An, Q. LINC00963 Promotes Ovarian Cancer Proliferation, Migration and EMT via the miR-378g/CHI3L1 Axis. Cancer Manag. Res. 2020, 12, 463–473. [Google Scholar] [CrossRef] [Green Version]
  16. Schulz, A.; Gorodetska, I.; Behrendt, R.; Fuessel, S.; Erdmann, K.; Foerster, S.; Datta, K.; Mayr, T.; Dubrovska, A.; Muders, M.H. Linking NRP2 With EMT and Chemoradioresistance in Bladder Cancer. Front. Oncol. 2019, 9, 1461. [Google Scholar] [CrossRef] [PubMed]
  17. Afzal Ashaie, M.; Hoque Chowdhury, E. Cadherins: The superfamily critically involved in breast cancer. Curr. Pharm. Des. 2016, 22, 616–638. [Google Scholar] [CrossRef] [PubMed]
  18. Carpinelli, M.R.; de Vries, M.E.; Auden, A.; Butt, T.; Deng, Z.; Partridge, D.D.; Miles, L.B.; Georgy, S.R.; Haigh, J.J.; Darido, C.; et al. Inactivation of Zeb1 in GRHL2-deficient mouse embryos rescues mid-gestation viability and secondary palate closure. Dis. Models Mech. 2020. [Google Scholar] [CrossRef] [Green Version]
  19. Cho, H.J.; Oh, N.; Park, J.H.; Kim, K.S.; Kim, H.K.; Lee, E.; Hwang, S.; Kim, S.J.; Park, K.S. ZEB1 Collaborates with ELK3 to Repress E-Cadherin Expression in Triple-Negative Breast Cancer Cells. Mol. Cancer Res. MCR 2019, 17, 2257–2266. [Google Scholar] [CrossRef] [Green Version]
  20. Yoshimoto, S.; Tanaka, F.; Morita, H.; Hiraki, A.; Hashimoto, S. Hypoxia-induced HIF-1alpha and ZEB1 are critical for the malignant transformation of ameloblastoma via TGF-beta-dependent EMT. Cancer Med. 2019, 8, 7822–7832. [Google Scholar] [CrossRef] [Green Version]
  21. Zhuang, W.; Li, Z.; Dong, X.; Zhao, N.; Liu, Y.; Wang, C.; Chen, J. Schisandrin B inhibits TGF-beta1-induced epithelial-mesenchymal transition in human A549 cells through epigenetic silencing of ZEB1. Exp. Lung Res. 2019, 45, 157–166. [Google Scholar] [CrossRef] [PubMed]
  22. Soen, B.; Vandamme, N.; Berx, G.; Schwaller, J.; Van Vlierberghe, P.; Goossens, S. ZEB Proteins in Leukemia: Friends, Foes, or Friendly Foes? HemaSphere 2018, 2, e43. [Google Scholar] [CrossRef]
  23. Comijn, J.; Berx, G.; Vermassen, P.; Verschueren, K.; van Grunsven, L.; Bruyneel, E.; Mareel, M.; Huylebroeck, D.; van Roy, F. The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol. Cell 2001, 7, 1267–1278. [Google Scholar] [CrossRef] [Green Version]
  24. Verschueren, K.; Remacle, J.E.; Collart, C.; Kraft, H.; Baker, B.S.; Tylzanowski, P.; Nelles, L.; Wuytens, G.; Su, M.T.; Bodmer, R.; et al. SIP1, a novel zinc finger/homeodomain repressor, interacts with Smad proteins and binds to 5’-CACCT sequences in candidate target genes. J. Biol. Chem. 1999, 274, 20489–20498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Remacle, J.E.; Kraft, H.; Lerchner, W.; Wuytens, G.; Collart, C.; Verschueren, K.; Smith, J.C.; Huylebroeck, D. New mode of DNA binding of multi-zinc finger transcription factors: deltaEF1 family members bind with two hands to two target sites. EMBO J. 1999, 18, 5073–5084. [Google Scholar] [CrossRef] [Green Version]
  26. Shi, Y.; Sawada, J.-i.; Sui, G.; Affar, E.B.; Whetstine, J.R.; Lan, F.; Ogawa, H.; Luke, M.P.-S.; Nakatani, Y.; Shi, Y. Coordinated histone modifications mediated by a CtBP co-repressor complex. Nature 2003, 422, 735–738. [Google Scholar] [CrossRef] [PubMed]
  27. Zhang, P.; Sun, Y.; Ma, L. ZEB1: At the crossroads of epithelial-mesenchymal transition, metastasis and therapy resistance. Cell Cycle 2015, 14, 481–487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Shargh, S.A.; Sakizli, M.; Khalaj, V.; Movafagh, A.; Yazdi, H.; Hagigatjou, E.; Sayad, A.; Mansouri, N.; Mortazavi-Tabatabaei, S.A.; Khorshid, H.R.K. Downregulation of E-cadherin expression in breast cancer by promoter hypermethylation and its relation with progression and prognosis of tumor. Med. Oncol. 2014, 31, 250. [Google Scholar] [CrossRef] [PubMed]
  29. Peiffer, D.S.; Wyatt, D.; Zlobin, A.; Piracha, A.; Ng, J.; Dingwall, A.K.; Albain, K.S.; Osipo, C. DAXX Suppresses Tumor-Initiating Cells in Estrogen Receptor–Positive Breast Cancer Following Endocrine Therapy. Cancer Res. 2019, 79, 4965–4977. [Google Scholar] [CrossRef] [Green Version]
  30. Liang, F.; Ren, C.; Wang, J.; Wang, S.; Yang, L.; Han, X.; Chen, Y.; Tong, G.; Yang, G. The crosstalk between STAT3 and p53/RAS signaling controls cancer cell metastasis and cisplatin resistance via the Slug/MAPK/PI3K/AKT-mediated regulation of EMT and autophagy. Oncogenesis 2019, 8, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Wu, Y.; Zhou, Y.; He, J.; Sun, H.; Jin, Z. Long non-coding RNA H19 mediates ovarian cancer cell cisplatin-resistance and migration during EMT. Int. J. Clin. Exp. Pathol. 2019, 12, 2506. [Google Scholar] [PubMed]
  32. Zhou, Y.; Zhou, Y.; Wang, K.; Li, T.; Zhang, M.; Yang, Y.; Wang, R.; Hu, R. ROCK2 Confers Acquired Gemcitabine Resistance in Pancreatic Cancer Cells by Upregulating Transcription Factor ZEB1. Cancers 2019, 11, 1881. [Google Scholar] [CrossRef] [Green Version]
  33. Sun, S.; Yang, X.; Qin, X.; Zhao, Y. TCF4 promotes colorectal cancer drug resistance and stemness via regulating ZEB1/ZEB2 expression. Protoplasma 2020. [Google Scholar] [CrossRef] [PubMed]
  34. Orellana-Serradell, O.; Herrera, D.; Castellon, E.A.; Contreras, H.R. The transcription factor ZEB1 promotes chemoresistance in prostate cancer cell lines. Asian J. Androl. 2019, 21, 460–467. [Google Scholar] [CrossRef] [PubMed]
  35. Pan, B.; Han, H.; Wu, L.; Xiong, Y.; Zhang, J.; Dong, B.; Yang, Y.; Chen, J. MTBP promotes migration and invasion by regulation of ZeB2-mediated epithelial–mesenchymal transition in lung cancer cells. OncoTargets Ther. 2018, 11, 6741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Tsai, Y.-S.; Jou, Y.-C.; Tsai, H.-T.; Cheong, I.-S.; Tzai, T.-S. Indoleamine-2, 3-dioxygenase-1 expression predicts poorer survival and up-regulates ZEB2 expression in human early stage bladder cancer. In Urologic Oncology: Seminars and Original Investigations; Elsevier: Amsterdam, The Netherlands, 2019. [Google Scholar]
  37. Yalim-Camci, I.; Balcik-Ercin, P.; Cetin, M.; Odabas, G.; Tokay, N.; Sayan, A.E.; Yagci, T. ETS1 is coexpressed with ZEB2 and mediates ZEB2-induced epithelial-mesenchymal transition in human tumors. Mol. Carcinog. 2019, 58, 1068–1081. [Google Scholar] [CrossRef] [PubMed]
  38. Wu, D.M.; Zhang, T.; Liu, Y.B.; Deng, S.H.; Han, R.; Liu, T.; Li, J.; Xu, Y. The PAX6-ZEB2 axis promotes metastasis and cisplatin resistance in non-small cell lung cancer through PI3K/AKT signaling. Cell Death Dis. 2019, 10, 349. [Google Scholar] [CrossRef]
  39. ElKhouly, A.M.; Youness, R.; Gad, M. MicroRNA-486-5p and MicroRNA-486-3p: Multifaceted Pleiotropic Mediators in Oncological and Non-Oncological Conditions. Non-coding RNA Res. 2020, 5, 11–21. [Google Scholar] [CrossRef]
  40. Delihas, N. Discovery and characterization of the first non-coding RNA that regulates gene expression, micF RNA: A historical perspective. World J. Biol. Chem. 2015, 6, 272. [Google Scholar] [CrossRef]
  41. Anastasiadou, E.; Jacob, L.S.; Slack, F.J. Non-coding RNA networks in cancer. Nat. Rev. Cancer 2018, 18, 5. [Google Scholar] [CrossRef]
  42. Chen, X.; Tang, F.R.; Arfuso, F.; Cai, W.Q.; Ma, Z.; Yang, J.; Sethi, G. The Emerging Role of Long Non-Coding RNAs in the Metastasis of Hepatocellular Carcinoma. Biomolecules 2019, 10, 66. [Google Scholar] [CrossRef] [Green Version]
  43. Ma, Z.; Wang, Y.Y.; Xin, H.W.; Wang, L.; Arfuso, F.; Dharmarajan, A.; Kumar, A.P.; Wang, H.; Tang, F.R.; Warrier, S.; et al. The expanding roles of long non-coding RNAs in the regulation of cancer stem cells. Int. J. Biochem. Cell Biol. 2019, 108, 17–20. [Google Scholar] [CrossRef]
  44. Cheng, J.T.; Wang, L.; Wang, H.; Tang, F.R.; Cai, W.Q.; Sethi, G.; Xin, H.W.; Ma, Z. Insights into Biological Role of LncRNAs in Epithelial-Mesenchymal Transition. Cells 2019, 8, 1178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Pourhanifeh, M.H.; Mahjoubin-Tehran, M.; Karimzadeh, M.R.; Mirzaei, H.R.; Razavi, Z.S.; Sahebkar, A.; Hosseini, N.; Mirzaei, H.; Hamblin, M.R. Autophagy in cancers including brain tumors: Role of MicroRNAs. Cell Commun. Signal. 2020, 18, 88. [Google Scholar] [CrossRef] [PubMed]
  46. Yousefi, F.; Shabaninejad, Z.; Vakili, S.; Derakhshan, M.; Movahedpour, A.; Dabiri, H.; Ghasemi, Y.; Mahjoubin-Tehran, M.; Nikoozadeh, A.; Savardashtaki, A.; et al. TGF-β and WNT signaling pathways in cardiac fibrosis: Non-coding RNAs come into focus. Cell Commun. Signal. 2020, 18, 87. [Google Scholar] [CrossRef]
  47. Jamali, Z.; Taheri-Anganeh, M.; Shabaninejad, Z.; Keshavarzi, A.; Taghizadeh, H.; Razavi, Z.S.; Mottaghi, R.; Abolhassan, M.; Movahedpour, A.; Mirzaei, H. Autophagy regulation by microRNAs: Novel insights into osteosarcoma therapy. IUBMB Life 2020, 72, 1306–1321. [Google Scholar] [CrossRef] [PubMed]
  48. Naeli, P.; Yousefi, F.; Ghasemi, Y.; Savardashtaki, A.; Mirzaei, H. The Role of MicroRNAs in Lung Cancer: Implications for Diagnosis and Therapy. Curr. Mol. Med. 2020, 20, 90–101. [Google Scholar] [CrossRef] [PubMed]
  49. Losko, M.; Kotlinowski, J.; Jura, J. Long noncoding RNAs in metabolic syndrome related disorders. Mediat. Inflamm. 2016, 2016, 5365209. [Google Scholar] [CrossRef] [Green Version]
  50. Goh, J.N.; Loo, S.Y.; Datta, A.; Siveen, K.S.; Yap, W.N.; Cai, W.; Shin, E.M.; Wang, C.; Kim, J.E.; Chan, M.; et al. microRNAs in breast cancer: Regulatory roles governing the hallmarks of cancer. Biol. Rev. Camb. Philos. Soc. 2016, 91, 409–428. [Google Scholar] [CrossRef]
  51. Varghese, E.; Liskova, A.; Kubatka, P.; Samuel, S.M.; Büsselberg, D. Anti-Angiogenic Effects of Phytochemicals on miRNA Regulating Breast Cancer Progression. Biomolecules 2020, 10, 191. [Google Scholar] [CrossRef] [Green Version]
  52. Magalhaes, M.; Jorge, J.; Goncalves, A.C.; Sarmento-Ribeiro, A.B.; Carvalho, R.; Figueiras, A.; Santos, A.C.; Veiga, F. miR-29b and retinoic acid co-delivery: A promising tool to induce a synergistic antitumoral effect in non-small cell lung cancer cells. Drug Deliv. Transl. Res. 2020. [Google Scholar] [CrossRef]
  53. Ragheb, M.A.; Soliman, M.H.; Elzayat, E.M.; Mohamed, M.S.; El-Ekiaby, N.; Abdelaziz, A.I.; Abdelwahab, A.A. MiR-520c-3p Modulates Doxorubicin-chemosensitivity in HepG2 cells. Anti-Cancer Agents Med. Chem. 2020. [Google Scholar] [CrossRef]
  54. Taherdangkoo, K.; Kazemi Nezhad, S.R.; Hajjari, M.R.; Tahmasebi Birgani, M. miR-485-3p suppresses colorectal cancer via targeting TPX2. Bratisl. Lek. Listy 2020, 121, 302–307. [Google Scholar] [CrossRef]
  55. Park, J.H.; Jeong, G.H.; Lee, K.S.; Lee, K.H.; Suh, J.S.; Eisenhut, M.; van der Vliet, H.J.; Kronbichler, A.; Stubbs, B.; Solmi, M.; et al. Genetic variations in MicroRNA genes and cancer risk: A field synopsis and meta-analysis. Eur. J. Clin. Investig. 2020. [Google Scholar] [CrossRef] [PubMed]
  56. Salomão, K.B.; Pezuk, J.A.; de Souza, G.R.; Chagas, P.; Pereira, T.C.; Valera, E.T.; Brassesco, M.S. MicroRNA dysregulation interplay with childhood abdominal tumors. Cancer Metastasis Rev. 2019, 38, 783–811. [Google Scholar] [CrossRef] [PubMed]
  57. 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 2020, 9, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Jiao, X.; Qian, X.; Wu, L.; Li, B.; Wang, Y.; Kong, X.; Xiong, L. microRNA: The Impact on Cancer Stemness and Therapeutic Resistance. Cells 2020, 9, 8. [Google Scholar] [CrossRef] [Green Version]
  59. Wu, J.; Bao, J.; Kim, M.; Yuan, S.; Tang, C.; Zheng, H.; Mastick, G.S.; Xu, C.; Yan, W. Two miRNA clusters, miR-34b/c and miR-449, are essential for normal brain development, motile ciliogenesis, and spermatogenesis. Proc. Natl. Acad. Sci. USA 2014, 111, E2851–E2857. [Google Scholar] [CrossRef] [Green Version]
  60. Chen, C.Z.; Schaffert, S.; Fragoso, R.; Loh, C. Regulation of immune responses and tolerance: The micro RNA perspective. Immunol. Rev. 2013, 253, 112–128. [Google Scholar] [CrossRef] [Green Version]
  61. Follert, P.; Cremer, H.; Béclin, C. MicroRNAs in brain development and function: A matter of flexibility and stability. Front. Mol. Neurosci. 2014, 7, 5. [Google Scholar] [CrossRef] [Green Version]
  62. Takahashi, R.-u.; Miyazaki, H.; Ochiya, T. The role of microRNAs in the regulation of cancer stem cells. Front. Genet. 2014, 4, 295. [Google Scholar] [CrossRef] [Green Version]
  63. Raza, U.; Zhang, J.D.; Şahin, Ö. MicroRNAs: Master regulators of drug resistance, stemness, and metastasis. J. Mol. Med. 2014, 92, 321–336. [Google Scholar] [CrossRef] [Green Version]
  64. Filipowicz, W.; Bhattacharyya, S.N.; Sonenberg, N. Mechanisms of post-transcriptional regulation by microRNAs: Are the answers in sight? Nat. Rev. Genet. 2008, 9, 102–114. [Google Scholar] [CrossRef]
  65. Eulalio, A.; Huntzinger, E.; Izaurralde, E. Getting to the root of miRNA-mediated gene silencing. Cell 2008, 132, 9–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Sartorius, K.; Makarova, J.; Sartorius, B.; An, P.; Winkler, C.; Chuturgoon, A.; Kramvis, A. The Regulatory Role of MicroRNA in Hepatitis-B Virus-Associated Hepatocellular Carcinoma (HBV-HCC) Pathogenesis. Cells 2019, 8, 1504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Syed, S.N.; Frank, A.C.; Raue, R.; Brune, B. MicroRNA-A Tumor Trojan Horse for Tumor-Associated Macrophages. Cells 2019, 8, 1482. [Google Scholar] [CrossRef] [Green Version]
  68. Handa, H.; Murakami, Y.; Ishihara, R.; Kimura-Masuda, K.; Masuda, Y. The Role and Function of microRNA in the Pathogenesis of Multiple Myeloma. Cancers 2019, 11, 1738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Cosentino, G.; Plantamura, I.; Cataldo, A.; Iorio, M.V. MicroRNA and Oxidative Stress Interplay in the Context of Breast Cancer Pathogenesis. Int. J. Mol. Sci. 2019, 20, 5143. [Google Scholar] [CrossRef] [Green Version]
  70. Guo, Y.; Jia, Y.; Wang, S.; Liu, N.; Gao, D.; Zhang, L.; Lin, Z.; Wang, S.; Kong, F.; Peng, C. Downregulation of MUTYH contributes to cisplatin-resistance of esophageal squamous cell carcinoma cells by promoting Twist-mediated EMT. Oncol. Rep. 2019, 42, 2716–2727. [Google Scholar] [CrossRef] [PubMed]
  71. Sun, C.M.; Zhang, G.M.; Qian, H.N.; Cheng, S.J.; Wang, M.; Liu, M.; Li, D. MiR-1266 suppresses the growth and metastasis of prostate cancer via targeting PRMT5. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 6436–6444. [Google Scholar] [CrossRef] [PubMed]
  72. Xia, X.Y.; Yu, Y.J.; Ye, F.; Peng, G.Y.; Li, Y.J.; Zhou, X.M. MicroRNA-506-3p inhibits proliferation and promotes apoptosis in ovarian cancer cell via targeting SIRT1/AKT/FOXO3a signaling pathway. Neoplasma 2020, 67, 344–353. [Google Scholar] [CrossRef] [Green Version]
  73. Jin, Y.; Zhou, X.; Yao, X.; Zhang, Z.; Cui, M.; Lin, Y. MicroRNA-612 inhibits cervical cancer progression by targeting NOB1. J. Cell. Mol. Med. 2020, 24, 3149–3156. [Google Scholar] [CrossRef] [PubMed]
  74. Nong, K.; Zhang, D.; Chen, C.; Yang, Y.; Yang, Y.; Liu, S.; Cai, H. MicroRNA-519 inhibits hypoxia-induced tumorigenesis of pancreatic cancer by regulating immune checkpoint PD-L1. Oncol. Lett. 2020, 19, 1427–1433. [Google Scholar] [CrossRef] [PubMed]
  75. ZHAO, L.; YU, A.; ZHANG, Y.; WANG, X.; HAN, B.; WANG, X. MicroRNA-149 suppresses the malignant phenotypes of ovarian cancer via downregulation of MSI2 and inhibition of PI3K/AKT pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 55–64. [Google Scholar] [PubMed]
  76. Allahverdi, A.; Arefian, E.; Soleimani, M.; Ai, J.; Nahanmoghaddam, N.; Yousefi-Ahmadipour, A.; Ebrahimi-Barough, S. MicroRNA-4731-5p delivered by AD-mesenchymal stem cells induces cell cycle arrest and apoptosis in glioblastoma. J. Cell. Physiol. 2020. [Google Scholar] [CrossRef] [PubMed]
  77. Guo, X.; Piao, H.; Zhang, Y.; Sun, P.; Yao, B. Overexpression of microRNA-129-5p in glioblastoma inhibits cell proliferation, migration, and colony-forming ability by targeting ZFP36L1. Bosn. J. Basic Med. Sci. 2019. [Google Scholar] [CrossRef] [PubMed]
  78. Wang, W.; He, B. MiR-760 inhibits the progression of non-small cell lung cancer through blocking ROS1/Ras/Raf/MEK/ERK pathway. Biosci. Rep. 2020, BSR20182483. [Google Scholar] [CrossRef]
  79. Huang, C.; Liu, J.; Pan, X.; Peng, C.; Xiong, B.; Feng, M.; Yang, X. miR-454 promotes survival and induces oxaliplatin resistance in gastric carcinoma cells by targeting CYLD. Exp. Ther. Med. 2020, 19, 3604–3610. [Google Scholar] [CrossRef]
  80. Qin, X.; Wang, X.Y.; Fei, J.W.; Li, F.H.; Han, J.; Wang, H.X. MiR-20a promotes lung tumorigenesis by targeting RUNX3 via TGF-beta signaling pathway. J. Biol. Regul. Homeost. Agents 2020, 34. [Google Scholar] [CrossRef]
  81. Liu, X.; Fu, Y.; Zhang, G.; Zhang, D.; Liang, N.; Li, F.; Li, C.; Sui, C.; Jiang, J.; Lu, H.; et al. miR-424-5p Promotes Anoikis Resistance and Lung Metastasis by Inactivating Hippo Signaling in Thyroid Cancer. Mol. Ther. Oncolytics 2019, 15, 248–260. [Google Scholar] [CrossRef] [Green Version]
  82. Huang, S.; Zou, C.; Tang, Y.; Wa, Q.; Peng, X.; Chen, X.; Yang, C.; Ren, D.; Huang, Y.; Liao, Z.; et al. miR-582-3p and miR-582-5p Suppress Prostate Cancer Metastasis to Bone by Repressing TGF-β Signaling. Mol. Ther. Nucleic Acids 2019, 16, 91–104. [Google Scholar] [CrossRef] [Green Version]
  83. Vescarelli, E.; Gerini, G.; Megiorni, F.; Anastasiadou, E.; Pontecorvi, P.; Solito, L.; De Vitis, C.; Camero, S.; Marchetti, C.; Mancini, R.; et al. MiR-200c sensitizes Olaparib-resistant ovarian cancer cells by targeting Neuropilin 1. J. Exp. Clin. Cancer Res. CR 2020, 39, 3. [Google Scholar] [CrossRef]
  84. Meng, Z.; Zhang, R.; Wang, Y.; Zhu, G.; Jin, T.; Li, C.; Zhang, S. miR-200c/PAI-2 promotes the progression of triple negative breast cancer via M1/M2 polarization induction of macrophage. Int. Immunopharmacol. 2019, 106028. [Google Scholar] [CrossRef] [PubMed]
  85. Basu, S.; Chaudhary, A.; Chowdhury, P.; Karmakar, D.; Basu, K.; Karmakar, D.; Chatterjee, J.; Sengupta, S. Evaluating the role of hsa-miR-200c in reversing the epithelial to mesenchymal transition in prostate cancer. Gene 2020, 730, 144264. [Google Scholar] [CrossRef]
  86. Tang, H.; Song, C.; Ye, F.; Gao, G.; Ou, X.; Zhang, L.; Xie, X.; Xie, X. miR-200c suppresses stemness and increases cellular sensitivity to trastuzumab in HER2+ breast cancer. J. Cell. Mol. Med. 2019, 23, 8114–8127. [Google Scholar] [CrossRef]
  87. Lin, H.; Yang, L.; Tian, F.; Nie, S.; Zhou, H.; Liu, J.; Chen, W. Up-regulated LncRNA-ATB regulates the growth and metastasis of cholangiocarcinoma via miR-200c signals. OncoTargets Ther. 2019, 12, 7561–7571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. DeSantis, C.E.; Ma, J.; Gaudet, M.M.; Newman, L.A.; Miller, K.D.; Goding Sauer, A.; Jemal, A.; Siegel, R.L. Breast cancer statistics, 2019. CA A Cancer J. Clin. 2019, 69, 438–451. [Google Scholar] [CrossRef]
  89. Abolghasemi, M.; Tehrani, S.S.; Yousefi, T.; Karimian, A.; Mahmoodpoor, A.; Ghamari, A.; Jadidi-Niaragh, F.; Yousefi, M.; Kafil, H.S.; Bastami, M.; et al. MicroRNAs in breast cancer: Roles, functions, and mechanism of actions. J. Cell. Physiol. 2020, 235, 5008–5029. [Google Scholar] [CrossRef] [PubMed]
  90. Zou, Q.; Zhou, E.; Xu, F.; Zhang, D.; Yi, W.; Yao, J. A TP73-AS1/miR-200a/ZEB1 regulating loop promotes breast cancer cell invasion and migration. J. Cell. Biochem. 2018, 119, 2189–2199. [Google Scholar] [CrossRef] [PubMed]
  91. Zhu, Y.; Yang, Z.; Luo, X.H.; Xu, P. Long noncoding RNA TTN-AS1 promotes the proliferation and migration of prostate cancer by inhibiting miR-1271 level. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 10678–10684. [Google Scholar] [CrossRef] [PubMed]
  92. Tian, Y.; Chen, Y.Y.; Han, A.L. MiR-1271 inhibits cell proliferation and metastasis by targeting LDHA in endometrial cancer. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 5648–5656. [Google Scholar] [CrossRef]
  93. Yao, H.; Sun, Q.; Zhu, J. miR-1271 enhances the sensitivity of colorectal cancer cells to cisplatin. Exp. Ther. Med. 2019, 17, 4363–4370. [Google Scholar] [CrossRef]
  94. Jiao, Y.; Zhu, G.; Yu, J.; Li, Y.; Wu, M.; Zhao, J.; Tian, X. miR-1271 inhibits growth, invasion and epithelial–mesenchymal transition by targeting ZEB1 in ovarian cancer cells. OncoTargets Ther. 2019, 12, 6973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Jiang, Y.; Jin, S.; Tan, S.; Shen, Q.; Xue, Y. MiR-203 acts as a radiosensitizer of gastric cancer cells by directly targeting ZEB1. OncoTargets Ther. 2019, 12, 6093–6104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Li, J.; Xia, L.; Zhou, Z.; Zuo, Z.; Xu, C.; Song, H.; Cai, J. MiR-186-5p upregulation inhibits proliferation, metastasis and epithelial-to-mesenchymal transition of colorectal cancer cell by targeting ZEB1. Arch. Biochem. Biophys. 2018, 640, 53–60. [Google Scholar] [CrossRef] [PubMed]
  97. Zhang, L.; Cai, Q.Y.; Liu, J.; Peng, J.; Chen, Y.Q.; Sferra, T.J.; Lin, J.M. Ursolic acid suppresses the invasive potential of colorectal cancer cells by regulating the TGF-beta1/ZEB1/miR-200c signaling pathway. Oncol. Lett. 2019, 18, 3274–3282. [Google Scholar] [CrossRef] [Green Version]
  98. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Dai, Y.; Wu, Z.; Lang, C.; Zhang, X.; He, S.; Yang, Q.; Guo, W.; Lai, Y.; Du, H.; Peng, X.; et al. Copy number gain of ZEB1 mediates a double-negative feedback loop with miR-33a-5p that regulates EMT and bone metastasis of prostate cancer dependent on TGF-β signaling. Theranostics 2019, 9, 6063–6079. [Google Scholar] [CrossRef]
  100. Sharma, N.; Nanta, R.; Sharma, J.; Gunewardena, S.; Singh, K.P.; Shankar, S.; Srivastava, R.K. PI3K/AKT/mTOR and sonic hedgehog pathways cooperate together to inhibit human pancreatic cancer stem cell characteristics and tumor growth. Oncotarget 2015, 6, 32039. [Google Scholar] [CrossRef]
  101. Ong, P.S.; Wang, L.Z.; Dai, X.; Tseng, S.H.; Loo, S.J.; Sethi, G. Judicious Toggling of mTOR Activity to Combat Insulin Resistance and Cancer: Current Evidence and Perspectives. Front. Pharmacol. 2016, 7, 395. [Google Scholar] [CrossRef]
  102. Singh, S.S.; Yap, W.N.; Arfuso, F.; Kar, S.; Wang, C.; Cai, W.; Dharmarajan, A.M.; Sethi, G.; Kumar, A.P. Targeting the PI3K/Akt signaling pathway in gastric carcinoma: A reality for personalized medicine? World J. Gastroenterol. 2015, 21, 12261–12273. [Google Scholar] [CrossRef]
  103. Huang, J.; Wang, X.; Wen, G.; Ren, Y. miRNA2055p functions as a tumor suppressor by negatively regulating VEGFA and PI3K/Akt/mTOR signaling in renal carcinoma cells. Oncol. Rep. 2019, 42, 1677–1688. [Google Scholar] [CrossRef]
  104. Liu, Z.; Luo, S.; Wu, M.; Huang, C.; Shi, H.; Song, X. LncRNA GHET1 promotes cervical cancer progression through regulating AKT/mTOR and Wnt/beta-catenin signaling pathways. Biosci. Rep. 2020, 40. [Google Scholar] [CrossRef]
  105. Yin, C.; Lin, X.; Wang, Y.; Liu, X.; Xiao, Y.; Liu, J.; Snijders, A.M.; Wei, G.; Mao, J.H.; Zhang, P. FAM83D promotes epithelial-mesenchymal transition, invasion and cisplatin resistance through regulating the AKT/mTOR pathway in non-small-cell lung cancer. Cell. Oncol. (Dordrecht) 2020. [Google Scholar] [CrossRef] [PubMed]
  106. Cai, C.; Dang, W.; Shilei, L.; Huang, L.; Li, Y.; Li, G.; Yan, S.; Jiang, C.; Song, X.; Hu, Y.; et al. ANTXR1/TEM8 promotes gastric cancer progression through activation of the PI3K/AKT/mTOR signaling pathway. Cancer Sci. 2020. [Google Scholar] [CrossRef] [PubMed]
  107. Cui, Z.; Han, B.; Wang, X.; Li, Z.; Wang, J.; Lv, Y. Long Non-Coding RNA TTN-AS1 Promotes the Proliferation and Invasion of Colorectal Cancer Cells by Activating miR-497-Mediated PI3K/Akt/mTOR Signaling. OncoTargets Ther. 2019, 12, 11531–11539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Ren, S.; Liu, J.; Feng, Y.; Li, Z.; He, L.; Li, L.; Cao, X.; Wang, Z.; Zhang, Y. Knockdown of circDENND4C inhibits glycolysis, migration and invasion by up-regulating miR-200b/c in breast cancer under hypoxia. J. Exp. Clin. Cancer Res. CR 2019, 38, 388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Chen, W.; Kong, K.-K.; Xu, X.-K.; Chen, C.; Li, H.; Wang, F.-Y.; Peng, X.-F.; Zhang, Z.; Li, P.; Li, J.-L. Downregulation of miR-205 is associated with glioblastoma cell migration, invasion, and the epithelial-mesenchymal transition, by targeting ZEB1 via the Akt/mTOR signaling pathway. Int. J. Oncol. 2018, 52, 485–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Sun, S.; Hang, T.; Zhang, B.; Zhu, L.; Wu, Y.; Lv, X.; Huang, Q.; Yao, H. miRNA-708 functions as a tumor suppressor in colorectal cancer by targeting ZEB1 through Akt/mTOR signaling pathway. Am. J. Transl. Res. 2019, 11, 5338–5356. [Google Scholar]
  111. Xie, R.; Wang, M.; Zhou, W.; Wang, D.; Yuan, Y.; Shi, H.; Wu, L. Long Non-Coding RNA (LncRNA) UFC1/miR-34a Contributes to Proliferation and Migration in Breast Cancer. Med. Sci. Monit. 2019, 25, 7149–7157. [Google Scholar] [CrossRef]
  112. Du, H.; Wang, X.; Dong, R.; Hu, D.; Xiong, Y. miR-601 inhibits proliferation, migration and invasion of prostate cancer stem cells by targeting KRT5 to inactivate the Wnt signaling pathway. Int. J. Clin. Exp. Pathol. 2019, 12, 4361–4379. [Google Scholar]
  113. Kong, B.; Lv, Z.D.; Xia, J.; Jin, L.Y.; Yang, Z.C. DLC-3 suppresses cellular proliferation, migration, and invasion in triple-negative breast cancer by the Wnt/beta-catenin pathway. Int. J. Clin. Exp. Pathol. 2019, 12, 1224–1232. [Google Scholar]
  114. Guo, Y.; Li, B.; Yan, X.; Shen, X.; Ma, J.; Liu, S.; Zhang, D. Bisphenol A and polychlorinated biphenyls enhance the cancer stem cell properties of human ovarian cancer cells by activating the WNT signaling pathway. Chemosphere 2019, 246, 125775. [Google Scholar] [CrossRef] [PubMed]
  115. Gao, X.H.; Zhang, Y.L.; Zhang, Z.Y.; Guo, S.S.; Chen, X.B.; Guo, Y.Z. MicroRNA-96-5p represses breast cancer proliferation and invasion through Wnt/beta-catenin signaling via targeting CTNND1. Sci. Rep. 2020, 10, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Pohl, S.G.; Brook, N.; Agostino, M.; Arfuso, F.; Kumar, A.P.; Dharmarajan, A. Wnt signaling in triple-negative breast cancer. Oncogenesis 2017, 6, e310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Surana, R.; Sikka, S.; Cai, W.; Shin, E.M.; Warrier, S.R.; Tan, H.J.; Arfuso, F.; Fox, S.A.; Dharmarajan, A.M.; Kumar, A.P. Secreted frizzled related proteins: Implications in cancers. Biochim. Biophys. Acta 2014, 1845, 53–65. [Google Scholar] [CrossRef]
  118. Qu, J.; Li, M.; An, J.; Zhao, B.; Zhong, W.; Gu, Q.; Cao, L.; Yang, H.; Hu, C. MicroRNA-33b inhibits lung adenocarcinoma cell growth, invasion, and epithelial-mesenchymal transition by suppressing Wnt/beta-catenin/ZEB1 signaling. Int. J. Oncol. 2015, 47, 2141–2152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Cong, N.; Du, P.; Zhang, A.; Shen, F.; Su, J.; Pu, P.; Wang, T.; Zjang, J.; Kang, C.; Zhang, Q. Downregulated microRNA-200a promotes EMT and tumor growth through the wnt/beta-catenin pathway by targeting the E-cadherin repressors ZEB1/ZEB2 in gastric adenocarcinoma. Oncol. Rep. 2013, 29, 1579–1587. [Google Scholar] [CrossRef] [Green Version]
  120. Shang, A.; Wang, W.; Gu, C.; Chen, C.; Zeng, B.; Yang, Y.; Ji, P.; Sun, J.; Wu, J.; Lu, W.; et al. Long non-coding RNA HOTTIP enhances IL-6 expression to potentiate immune escape of ovarian cancer cells by upregulating the expression of PD-L1 in neutrophils. J. Exp. Clin. Cancer Res. 2019, 38, 411. [Google Scholar] [CrossRef]
  121. Abolghasemi, M.; Tehrani, S.S.; Yousefi, T.; Karimian, A.; Mahmoodpoor, A.; Ghamari, A.; Jadidi-Niaragh, F.; Yousefi, M.; Kafil, H.S.; Bastami, M.; et al. Critical roles of long noncoding RNAs in breast cancer. J. Cell. Physiol. 2020, 235, 5059–5071. [Google Scholar] [CrossRef]
  122. Vafadar, A.; Shabaninejad, Z.; Movahedpour, A.; Mohammadi, S.; Fathullahzadeh, S.; Mirzaei, H.R.; Namdar, A.; Savardashtaki, A.; Mirzaei, H. Long Non-Coding RNAs As Epigenetic Regulators in Cancer. Curr. Pharm. Des. 2019, 25, 3563–3577. [Google Scholar] [CrossRef]
  123. Yang, C.; Di Wu, L.G.; Liu, X.; Jin, Y.; Wang, D.; Wang, T.; Li, X. Competing endogenous RNA networks in human cancer: Hypothesis, validation, and perspectives. Oncotarget 2016, 7, 13479. [Google Scholar] [CrossRef] [Green Version]
  124. Shen, J.; Hong, L.; Yu, D.; Cao, T.; Zhou, Z.; He, S. LncRNA XIST promotes pancreatic cancer migration, invasion and EMT by sponging miR-429 to modulate ZEB1 expression. Int. J. Biochem. Cell Biol. 2019, 113, 17–26. [Google Scholar] [CrossRef]
  125. Li, D.; Wang, J.; Zhang, M.; Hu, X.; She, J.; Qiu, X.; Zhang, X.; Xu, L.; Liu, Y.; Qin, S. LncRNA MAGI2-AS3 Is Regulated by BRD4 and Promotes Gastric Cancer Progression via Maintaining ZEB1 Overexpression by Sponging miR-141/200a. Mol. Ther. Nucleic Acids 2020, 19, 109–123. [Google Scholar] [CrossRef] [PubMed]
  126. Sun, L.; Chen, T.; Li, T.; Yu, J. LncRNA IUR downregulates ZEB1 by upregulating miR-200 to inhibit prostate carcinoma. Physiol. Genom. 2019, 51, 607–611. [Google Scholar] [CrossRef] [PubMed]
  127. Jiang, H.; Jiang, Y.; Zhang, T.; Mo, K.; Su, S.; Wang, A.; Zhu, Y.; Huang, G.; Zhou, R. The lncRNA TDRG1 promotes cell proliferation, migration and invasion by targeting miR-326 to regulate MAPK1 expression in cervical cancer. Cancer Cell Int. 2019, 19, 152. [Google Scholar] [CrossRef] [Green Version]
  128. Deng, S.-j.; Chen, H.-y.; Ye, Z.; Deng, S.-c.; Zhu, S.; Zeng, Z.; He, C.; Liu, M.-l.; Huang, K.; Zhong, J.-x. Hypoxia-induced LncRNA-BX111 promotes metastasis and progression of pancreatic cancer through regulating ZEB1 transcription. Oncogene 2018, 37, 5811–5828. [Google Scholar] [CrossRef]
  129. Hu, X.; Mu, Y.; Wang, J.; Zhao, Y. LncRNA TDRG1 promotes the metastasis of NSCLC cell through regulating miR-873-5p/ZEB1 axis. J. Cell. Biochem. 2019. [Google Scholar] [CrossRef] [PubMed]
  130. Zhong, Y.; Wang, J.; Lv, W.; Xu, J.; Mei, S.; Shan, A. LncRNA TTN-AS1 drives invasion and migration of lung adenocarcinoma cells via modulation of miR-4677-3p/ZEB1 axis. J. Cell. Biochem. 2019, 120, 17131–17141. [Google Scholar] [CrossRef] [PubMed]
  131. Zhang, M.; Wu, W.; Wang, Z.; Wang, X. lncRNA NEAT1 is closely related with progression of breast cancer via promoting proliferation and EMT. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 1020–1026. [Google Scholar]
  132. Tan, X.; Wang, P.; Lou, J.; Zhao, J. Knockdown of lncRNA NEAT1 suppresses hypoxia-induced migration, invasion and glycolysis in anaplastic thyroid carcinoma cells through regulation of miR-206 and miR-599. Cancer Cell Int. 2020, 20, 132. [Google Scholar] [CrossRef] [Green Version]
  133. Jiang, X.; Zhou, Y.; Sun, A.J.; Xue, J.L. NEAT1 contributes to breast cancer progression through modulating miR-448 and ZEB1. J. Cell. Physiol. 2018, 233, 8558–8566. [Google Scholar] [CrossRef]
  134. Shao, Q.; Wang, Q.; Wang, J. LncRNA SCAMP1 regulates ZEB1/JUN and autophagy to promote pediatric renal cell carcinoma under oxidative stress via miR-429. Biomed. Pharmacother. 2019, 120, 109460. [Google Scholar] [CrossRef]
  135. Kirtonia, A.; Sethi, G.; Garg, M. The multifaceted role of reactive oxygen species in tumorigenesis. Cell. Mol. Life Sci. 2020. [Google Scholar] [CrossRef] [PubMed]
  136. Aggarwal, V.; Tuli, H.S.; Varol, A.; Thakral, F.; Yerer, M.B.; Sak, K.; Varol, M.; Jain, A.; Khan, M.A.; Sethi, G. Role of Reactive Oxygen Species in Cancer Progression: Molecular Mechanisms and Recent Advancements. Biomolecules 2019, 9, 735. [Google Scholar] [CrossRef] [Green Version]
  137. Yang, C.; Sun, J.; Liu, W.; Yang, Y.; Chu, Z.; Yang, T.; Gui, Y.; Wang, D. Long noncoding RNA HCP5 contributes to epithelial-mesenchymal transition in colorectal cancer through ZEB1 activation and interacting with miR-139-5p. Am. J. Transl. Res. 2019, 11, 953–963. [Google Scholar] [PubMed]
  138. Pu, J.; Wang, J.; Wei, H.; Lu, T.; Wu, X.; Wu, Y.; Shao, Z.; Luo, C.; Lu, Y. lncRNA MAGI2-AS3 Prevents the Development of HCC via Recruiting KDM1A and Promoting H3K4me2 Demethylation of the RACGAP1 Promoter. Mol. Ther. Nucleic Acids 2019, 18, 351–362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Yang, Y.; Yang, H.; Xu, M.; Zhang, H.; Sun, M.; Mu, P.; Dong, T.; Du, S.; Liu, K. Long non-coding RNA (lncRNA) MAGI2-AS3 inhibits breast cancer cell growth by targeting the Fas/FasL signalling pathway. Human Cell 2018, 31, 232–241. [Google Scholar] [CrossRef]
  140. Zhao, X.; Liu, Y.; Li, Z.; Zheng, S.; Wang, Z.; Li, W.; Bi, Z.; Li, L.; Jiang, Y.; Luo, Y. Linc00511 acts as a competing endogenous RNA to regulate VEGFA expression through sponging hsa-miR-29b-3p in pancreatic ductal adenocarcinoma. J. Cell. Mol. Med. 2018, 22, 655–667. [Google Scholar] [CrossRef]
  141. Du, X.; Tu, Y.; Liu, S.; Zhao, P.; Bao, Z.; Li, C.; Li, J.; Pan, M.; Ji, J. LINC00511 contributes to glioblastoma tumorigenesis and epithelial-mesenchymal transition via LINC00511/miR-524-5p/YB1/ZEB1 positive feedback loop. J. Cell. Mol. Med. 2019, 24, 1474–1487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Hao, T.; Wang, Z.; Yang, J.; Zhang, Y.; Shang, Y.; Sun, J. MALAT1 knockdown inhibits prostate cancer progression by regulating miR-140/BIRC6 axis. Biomed. Pharmacother. 2020, 123, 109666. [Google Scholar] [CrossRef] [PubMed]
  143. Wei, S.; Liu, Q. Long noncoding RNA MALAT1 modulates sepsis-induced cardiac inflammation through the miR-150-5p/NF-kappaB axis. Int. J. Clin. Exp. Pathol. 2019, 12, 3311–3319. [Google Scholar]
  144. Chen, L.; Yao, H.; Wang, K.; Liu, X. Long non-coding RNA MALAT1 regulates ZEB1 expression by sponging miR-143-3p and promotes hepatocellular carcinoma progression. J. Cell. Biochem. 2017, 118, 4836–4843. [Google Scholar] [CrossRef] [PubMed]
  145. Wang, Y.; Yu, X.J.; Zhou, W.; Chu, Y.X. MicroRNA-429 inhibits the proliferation and migration of esophageal squamous cell carcinoma cells by targeting RAB23 through the NF-kappaB pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 1202–1210. [Google Scholar] [CrossRef] [PubMed]
  146. Shen, F.; Zheng, H.; Zhou, L.; Li, W.; Xu, X. Overexpression of MALAT1 contributes to cervical cancer progression by acting as a sponge of miR-429. J. Cell Physiol. 2019, 234, 11219–11226. [Google Scholar] [CrossRef] [PubMed]
  147. Szigeti, T.; Dunster, C.; Cattaneo, A.; Cavallo, D.; Spinazze, A.; Saraga, D.E.; Sakellaris, I.A.; de Kluizenaar, Y.; Cornelissen, E.J.; Hanninen, O.; et al. Oxidative potential and chemical composition of PM2.5 in office buildings across Europe—The OFFICAIR study. Environ. Int. 2016, 92–93, 324–333. [Google Scholar] [CrossRef] [Green Version]
  148. Valavanidis, A.; Vlachogianni, T.; Fiotakis, K.; Loridas, S. Pulmonary oxidative stress, inflammation and cancer: Respirable particulate matter, fibrous dusts and ozone as major causes of lung carcinogenesis through reactive oxygen species mechanisms. Int. J. Environ. Res. Public Health 2013, 10, 3886–3907. [Google Scholar] [CrossRef]
  149. Luo, F.; Wei, H.; Guo, H.; Li, Y.; Feng, Y.; Bian, Q.; Wang, Y. LncRNA MALAT1, an lncRNA acting via the miR-204/ZEB1 pathway, mediates the EMT induced by organic extract of PM2.5 in lung bronchial epithelial cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 317, L87–Ll98. [Google Scholar] [CrossRef]
  150. Wang, K.C.; Yang, Y.W.; Liu, B.; Sanyal, A.; Corces-Zimmerman, R.; Chen, Y.; Lajoie, B.R.; Protacio, A.; Flynn, R.A.; Gupta, R.A. A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature 2011, 472, 120–124. [Google Scholar] [CrossRef] [Green Version]
  151. Zhang, S.; Wang, W.; Liu, G.; Xie, S.; Li, Q.; Li, Y.; Lin, Z. Long non-coding RNA HOTTIP promotes hypoxia-induced epithelial-mesenchymal transition of malignant glioma by regulating the miR-101/ZEB1 axis. Biomed. Pharmacother. 2017, 95, 711–720. [Google Scholar] [CrossRef]
  152. Dai, B.; Zhou, G.; Hu, Z.; Zhu, G.; Mao, B.; Su, H.; Jia, Q. MiR-205 suppresses epithelial-mesenchymal transition and inhibits tumor growth of human glioma through down-regulation of HOXD9. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef] [Green Version]
  153. Dong, N.; Guo, J.; Han, S.; Bao, L.; Diao, Y.; Lin, Z. Positive feedback loop of lncRNA HOXC-AS2/miR-876-5p/ZEB1 to regulate EMT in glioma. OncoTargets Ther. 2019, 12, 7601–7609. [Google Scholar] [CrossRef] [Green Version]
  154. Kulcheski, F.R.; Christoff, A.P.; Margis, R. Circular RNAs are miRNA sponges and can be used as a new class of biomarker. J. Biotechnol. 2016, 238, 42–51. [Google Scholar] [CrossRef] [PubMed]
  155. Meng, S.; Zhou, H.; Feng, Z.; Xu, Z.; Tang, Y.; Li, P.; Wu, M. CircRNA: Functions and properties of a novel potential biomarker for cancer. Mol. Cancer 2017, 16, 94. [Google Scholar] [CrossRef]
  156. Conn, S.J.; Pillman, K.A.; Toubia, J.; Conn, V.M.; Salmanidis, M.; Phillips, C.A.; Roslan, S.; Schreiber, A.W.; Gregory, P.A.; Goodall, G.J. The RNA binding protein quaking regulates formation of circRNAs. Cell 2015, 160, 1125–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Liu, C.; Zhang, Z.; Qi, D. Circular RNA hsa_circ_0023404 promotes proliferation, migration and invasion in non-small cell lung cancer by regulating miR-217/ZEB1 axis. OncoTargets Ther. 2019, 12, 6181–6189. [Google Scholar] [CrossRef] [PubMed]
  158. Re, M.; Magliulo, G.; Gioacchini, F.M.; Bajraktari, A.; Bertini, A.; Çeka, A.; Rubini, C.; Ferrante, L.; Procopio, A.D.; Olivieri, F. Expression levels and clinical significance of miR-21-5p, miR-let-7a, and miR-34c-5p in laryngeal squamous cell carcinoma. BioMed. Res. Int. 2017, 2017, 3921258. [Google Scholar] [CrossRef] [Green Version]
  159. Wei, Q.; Yu, D.; Liu, M.; Wang, M.; Zhao, M.; Liu, M.; Jia, W.; Ma, H.; Fang, J.; Xu, W. Genome-wide association study identifies three susceptibility loci for laryngeal squamous cell carcinoma in the Chinese population. Nat. Genet. 2014, 46, 1110. [Google Scholar] [CrossRef]
  160. Fu, D.; Huang, Y.; Gao, M. Hsa_circ_0057481 promotes laryngeal cancer proliferation and migration by modulating the miR-200c/ZEB1 axis. Int. J. Clin. Exp. Pathol. 2019, 12, 4066. [Google Scholar]
  161. Ying, X.; Zhu, J.; Zhang, Y. Circular RNA circ-TSPAN4 promotes lung adenocarcinoma metastasis by upregulating ZEB1 via sponging miR-665. Mol. Genet. Genom. Med. 2019, 7, e991. [Google Scholar] [CrossRef] [Green Version]
  162. Conte, R.; Valentino, A.; Di Cristo, F.; Peluso, G.; Cerruti, P.; Di Salle, A.; Calarco, A. Cationic Polymer Nanoparticles-Mediated Delivery of miR-124 Impairs Tumorigenicity of Prostate Cancer Cells. Int. J. Mol. Sci. 2020, 21, 869. [Google Scholar] [CrossRef] [Green Version]
  163. Chen, J.; Zhong, Y.; Li, L. miR-124 and miR-203 synergistically inactivate EMT pathway via coregulation of ZEB2 in clear cell renal cell carcinoma (ccRCC). J. Transl. Med. 2020, 18, 69. [Google Scholar] [CrossRef]
  164. Chen, L.; Hu, N.; Wang, C.; Zhao, H. HOTAIRM1 knockdown enhances cytarabine-induced cytotoxicity by suppression of glycolysis through the Wnt/beta-catenin/PFKP pathway in acute myeloid leukemia cells. Arch. Biochem. Biophys. 2020, 680, 108244. [Google Scholar] [CrossRef] [PubMed]
  165. Li, J.; Zhang, S.; Wu, L.; Pei, M. Interaction between LncRNA-ROR and miR-145 contributes to epithelial-mesenchymal transition of ovarian cancer cells. Gen. Physiol. Biophys. 2019, 38, 461–471. [Google Scholar] [CrossRef] [PubMed]
  166. Liu, Q.; Chen, J.; Wang, B.; Zheng, Y.; Wan, Y.; Wang, Y.; Zhou, L.; Liu, S.; Li, G.; Yan, Y. miR-145 modulates epithelial-mesenchymal transition and invasion by targeting ZEB2 in non-small cell lung cancer cell lines. J. Cell. Biochem. 2018. [Google Scholar] [CrossRef] [PubMed]
  167. Miao, Y.; Wang, L.; Zhang, X.; Xing, R.-G.; Zhou, W.-W.; Liu, C.-R.; Zhang, X.-L.; Tian, L. miR-30a inhibits breast cancer progression through the Wnt/β-catenin pathway. Int. J. Clin. Exp. Pathol. 2019, 12, 241. [Google Scholar]
  168. Wang, T.; Chen, G.; Ma, X.; Yang, Y.; Chen, Y.; Peng, Y.; Bai, Z.; Zhang, Z.; Pei, H.; Guo, W. MiR-30a regulates cancer cell response to chemotherapy through SNAI1/IRS1/AKT pathway. Cell Death Dis. 2019, 10, 153. [Google Scholar] [CrossRef]
  169. Di Gennaro, A.; Damiano, V.; Brisotto, G.; Armellin, M.; Perin, T.; Zucchetto, A.; Guardascione, M.; Spaink, H.P.; Doglioni, C.; Snaar-Jagalska, B.E. A p53/miR-30a/ZEB2 axis controls triple negative breast cancer aggressiveness. Cell Death Differ. 2018, 25, 2165–2180. [Google Scholar] [CrossRef]
  170. Zhang, L.; Zhang, T.; Deng, Z.; Sun, L. MicroRNA3653 inhibits the growth and metastasis of hepatocellular carcinoma by inhibiting ITGB1. Oncol. Rep. 2019, 41, 1669–1677. [Google Scholar] [CrossRef] [Green Version]
  171. Zhu, W.; Luo, X.; Fu, H.; Liu, L.; Sun, P.; Wang, Z. miR-3653 inhibits the metastasis and epithelial-mesenchymal transition of colon cancer by targeting Zeb2. Pathol. Res. Pract. 2019, 215, 152577. [Google Scholar] [CrossRef]
  172. Zhu, D.; Gu, L.; Li, Z.; Jin, W.; Lu, Q.; Ren, T. MiR-138-5p suppresses lung adenocarcinoma cell epithelial-mesenchymal transition, proliferation and metastasis by targeting ZEB2. Pathol. Res. Pract. 2019, 215, 861–872. [Google Scholar] [CrossRef]
  173. Eleutério, S.J.P.; Senerchia, A.A.; Almeida, M.T.; Costa, C.M.D.; Lustosa, D.; Calheiros, L.M.; Barreto, J.H.S.; Brunetto, A.L.; Macedo, C.R.P.D.; Petrilli, A.S. Osteosarcoma in patients younger than 12 years old without metastases have similar prognosis as adolescent and young adults. Pediatric Blood Cancer 2015, 62, 1209–1213. [Google Scholar] [CrossRef]
  174. Wang, S.-N.; Luo, S.; Liu, C.; Piao, Z.; Gou, W.; Wang, Y.; Guan, W.; Li, Q.; Zou, H.; Yang, Z.-Z. miR-491 inhibits osteosarcoma lung metastasis and chemoresistance by targeting αB-crystallin. Mol. Ther. 2017, 25, 2140–2149. [Google Scholar] [CrossRef]
  175. Lin, H.; Zheng, X.; Lu, T.; Gu, Y.; Zheng, C.; Yan, H. The proliferation and invasion of osteosarcoma are inhibited by miR-101 via targetting ZEB2. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef] [Green Version]
  176. Liu, N.; Feng, S.; Li, H.; Chen, X.; Bai, S.; Liu, Y. Long non-coding RNA MALAT1 facilitates the tumorigenesis, invasion and glycolysis of multiple myeloma via miR-1271-5p/SOX13 axis. J. Cancer Res. Clin. Oncol. 2020, 146, 367–379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Chao, H.; Zhang, M.; Hou, H.; Zhang, Z.; Li, N. HOTAIRM1 suppresses cell proliferation and invasion in ovarian cancer through facilitating ARHGAP24 expression by sponging miR-106a-5p. Life Sci. 2020, 117296. [Google Scholar] [CrossRef] [PubMed]
  178. Shi, T.; Guo, D.; Xu, H.; Su, G.; Chen, J.; Zhao, Z.; Shi, J.; Wedemeyer, M.; Attenello, F.; Zhang, L.; et al. HOTAIRM1, an enhancer lncRNA, promotes glioma proliferation by regulating long-range chromatin interactions within HOXA cluster genes. Mol. Biol. Rep. 2020, 47, 2723–2733. [Google Scholar] [CrossRef] [PubMed]
  179. Lin, Y.H.; Guo, L.; Yan, F.; Dou, Z.Q.; Yu, Q.; Chen, G. Long non-coding RNA HOTAIRM1 promotes proliferation and inhibits apoptosis of glioma cells by regulating the miR-873-5p/ZEB2 axis. Chin. Med. J. (Engl.) 2020, 133, 174–182. [Google Scholar] [CrossRef]
  180. Ren, L.; Yao, Y.; Wang, Y.; Wang, S. MiR-505 suppressed the growth of hepatocellular carcinoma cells via targeting IGF-1R. Biosci. Rep. 2019, 39, BSR20182442. [Google Scholar] [CrossRef] [Green Version]
  181. Tian, L.; Wang, Z.Y.; Hao, J.; Zhang, X.Y. miR-505 acts as a tumor suppressor in gastric cancer progression through targeting HMGB1. J. Cell. Biochem. 2019, 120, 8044–8052. [Google Scholar] [CrossRef]
  182. Shi, H.; Yang, H.; Xu, S.; Zhao, Y.; Liu, J. miR-505 functions as a tumor suppressor in glioma by targeting insulin like growth factor 1 receptor expression. Int. J. Clin. Exp. Pathol. 2018, 11, 4405. [Google Scholar]
  183. Gong, X.; Huang, A. LEF-AS1 participates in occurrence of colorectal cancer through adsorbing miR-505 and promoting KIF3B expression. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 9362–9370. [Google Scholar]
  184. Wang, Z.; Wang, Q.; Xu, G.; Meng, N.; Huang, X.; Jiang, Z.; Chen, C.; Zhang, Y.; Chen, J.; Li, A.; et al. The long noncoding RNA CRAL reverses cisplatin resistance via the miR-505/CYLD/AKT axis in human gastric cancer cells. RNA Biol. 2020, 1–14. [Google Scholar] [CrossRef] [PubMed]
  185. Zhao, P.; Guan, H.; Dai, Z.; Ma, Y.; Zhao, Y.; Liu, D. Long noncoding RNA DLX6-AS1 promotes breast cancer progression via miR-505-3p/RUNX2 axis. Eur. J. Pharmacol. 2019, 865, 172778. [Google Scholar] [CrossRef] [PubMed]
  186. Feng, S.; Liu, W.; Bai, X.; Pan, W.; Jia, Z.; Zhang, S.; Zhu, Y.; Tan, W. LncRNA-CTS promotes metastasis and epithelial-to-mesenchymal transition through regulating miR-505/ZEB2 axis in cervical cancer. Cancer Lett. 2019, 465, 105–117. [Google Scholar] [CrossRef] [PubMed]
  187. Gong, P.; Qiao, F.; Wu, H.; Cui, H.; Li, Y.; Zheng, Y.; Zhou, M.; Fan, H. LncRNA UCA1 promotes tumor metastasis by inducing miR-203/ZEB2 axis in gastric cancer. Cell Death Dis. 2018, 9, 1–14. [Google Scholar] [CrossRef] [PubMed]
  188. Wang, Y.; Liang, S.; Yu, Y.; Shi, Y.; Zheng, H. Knockdown of SNHG12 suppresses tumor metastasis and epithelial-mesenchymal transition via the Slug/ZEB2 signaling pathway by targeting miR-218 in NSCLC. Oncol. Lett. 2019, 17, 2356–2364. [Google Scholar] [CrossRef] [Green Version]
  189. Weller, M.; Wick, W.; Aldape, K.; Brada, M.; Berger, M.; Pfister, S.M.; Nishikawa, R.; Rosenthal, M.; Wen, P.Y.; Stupp, R. Glioma. Nat. Rev. Dis. Primers 2015, 1, 1–18. [Google Scholar] [CrossRef] [PubMed]
  190. Ostrom, Q.T.; Bauchet, L.; Davis, F.G.; Deltour, I.; Fisher, J.L.; Langer, C.E.; Pekmezci, M.; Schwartzbaum, J.A.; Turner, M.C.; Walsh, K.M. Response to “the epidemiology of glioma in adults: A ‘state of the science’review”. Neuro-Oncology 2015, 17, 624–626. [Google Scholar] [CrossRef] [Green Version]
  191. Hanif, F.; Muzaffar, K.; Perveen, K.; Malhi, S.M.; Simjee, S.U. Glioblastoma multiforme: A review of its epidemiology and pathogenesis through clinical presentation and treatment. Asian Pac. J. Cancer Prev. APJCP 2017, 18, 3. [Google Scholar]
  192. Rodriguez, A.; Tatter, S.B. Laser ablation of recurrent malignant gliomas: Current status and future perspective. Neurosurgery 2016, 79, S35–S39. [Google Scholar] [CrossRef] [Green Version]
  193. Hu, B.; Wang, Q.; Wang, Y.A.; Hua, S.; Sauvé, C.-E.G.; Ong, D.; Lan, Z.D.; Chang, Q.; Ho, Y.W.; Monasterio, M.M. Epigenetic activation of WNT5A drives glioblastoma stem cell differentiation and invasive growth. Cell 2016, 167, 1281–1295.e18. [Google Scholar] [CrossRef] [Green Version]
  194. Meng, X.; Deng, Y.; Lv, Z.; Liu, C.; Guo, Z.; Li, Y.; Liu, H.; Xie, B.; Jin, Z.; Lin, F. LncRNA SNHG5 Promotes Proliferation of Glioma by Regulating miR-205-5p/ZEB2 Axis. OncoTargets Ther. 2019, 12, 11487. [Google Scholar] [CrossRef] [Green Version]
  195. Wu, H.; Liu, H.Y.; Liu, W.J.; Shi, Y.L.; Bao, D. miR-377-5p inhibits lung cancer cell proliferation, invasion, and cell cycle progression by targeting AKT1 signaling. J. Cell. Biochem. 2019, 120, 8120–8128. [Google Scholar] [CrossRef] [PubMed]
  196. Zhang, W.Y.; Liu, Q.H.; Wang, T.J.; Zhao, J.; Cheng, X.H.; Wang, J.S. CircZFR serves as a prognostic marker to promote bladder cancer progression by regulating miR-377/ZEB2 signaling. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef] [PubMed]
  197. Xie, R.; Tang, J.; Zhu, X.; Jiang, H. Silencing of hsa_circ_0004771 inhibits proliferation and induces apoptosis in breast cancer through activation of miR-653 by targeting ZEB2 signaling pathway. Biosci. Rep. 2019, 39, BSR20181919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Siegel, R.; Naishadham, D.; Jemal, A. Cancer statistics, 2012. CA Cancer J. Clin. 2012, 62, 10–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Ljungberg, B.; Campbell, S.C.; Cho, H.Y.; Jacqmin, D.; Lee, J.E.; Weikert, S.; Kiemeney, L.A. The epidemiology of renal cell carcinoma. Eur. Urol. 2011, 60, 615–621. [Google Scholar] [CrossRef] [PubMed]
  200. Znaor, A.; Lortet-Tieulent, J.; Laversanne, M.; Jemal, A.; Bray, F. International variations and trends in renal cell carcinoma incidence and mortality. Eur. Urol. 2015, 67, 519–530. [Google Scholar] [CrossRef] [PubMed]
  201. Zhou, B.; Zheng, P.; Li, Z.; Li, H.; Wang, X.; Shi, Z.; Han, Q. CircPCNXL2 sponges miR-153 to promote the proliferation and invasion of renal cancer cells through upregulating ZEB2. Cell Cycle 2018, 17, 2644–2654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Riganti, C.; Contino, M. New Strategies to Overcome Resistance to Chemotherapy and Immune System in Cancer. Int. J. Mol. Sci. 2019, 20, 4783. [Google Scholar] [CrossRef] [Green Version]
  203. Nedeljković, M.; Damjanović, A. Mechanisms of Chemotherapy Resistance in Triple-Negative Breast Cancer-How We Can Rise to the Challenge. Cells 2019, 8, 957. [Google Scholar] [CrossRef] [Green Version]
  204. Sajid, A.; Raju, N.; Lusvarghi, S.; Vahedi, S.; Swenson, R.E.; Ambudkar, S.V. Synthesis and Characterization of Bodipy-FL-Cyclosporine A as a Substrate for Multidrug Resistance-Linked P-Glycoprotein (ABCB1). Drug Metab. Dispos. Biol. Fate Chem. 2019, 47, 1013–1023. [Google Scholar] [CrossRef]
  205. van Hoppe, S.; Jamalpoor, A.; Rood, J.J.M.; Wagenaar, E.; Sparidans, R.W.; Beijnen, J.H.; Schinkel, A.H. Brain accumulation of osimertinib and its active metabolite AZ5104 is restricted by ABCB1 (P-glycoprotein) and ABCG2 (breast cancer resistance protein). Pharmacol. Res. 2019, 146, 104297. [Google Scholar] [CrossRef] [PubMed]
  206. Shen, X.; Wang, Y.; Xi, L.; Su, F.; Li, S. Biocompatibility and paclitaxel/cisplatin dual-loading of nanotubes prepared from poly(ethylene glycol)-polylactide-poly(ethylene glycol) triblock copolymers for combination cancer therapy. Saudi Pharm. J. SPJ Off. Publ. Saudi Pharm. Soc. 2019, 27, 1025–1035. [Google Scholar] [CrossRef] [PubMed]
  207. Chen, W.; Du, J.; Li, X.; Zhi, Z.; Jiang, S. microRNA-137 downregulates MCL1 in ovarian cancer cells and mediates cisplatin-induced apoptosis. Pharmacogenomics 2020, 21, 195–207. [Google Scholar] [CrossRef] [PubMed]
  208. Abu Samaan, T.M.; Samec, M.; Liskova, A.; Kubatka, P.; Büsselberg, D. Paclitaxel’s Mechanistic and Clinical Effects on Breast Cancer. Biomolecules 2019, 9, 789. [Google Scholar] [CrossRef] [Green Version]
  209. Khodaverdi, S.; Jafari, A.; Movahedzadeh, F.; Madani, F.; Yousefi Avarvand, A.; Falahatkar, S. Evaluating Inhibitory Effects of Paclitaxel and Vitamin D3 Loaded Poly Lactic Glycolic Acid Co-Delivery Nanoparticles on the Breast Cancer Cell Line. Adv. Pharm. Bull. 2020, 10, 30–38. [Google Scholar] [CrossRef]
  210. Miyagawa, Y.; Yanai, A.; Yanagawa, T.; Inatome, J.; Egawa, C.; Nishimukai, A.; Takamoto, K.; Morimoto, T.; Kikawa, Y.; Suwa, H.; et al. Baseline neutrophil-to-lymphocyte ratio and c-reactive protein predict efficacy of treatment with bevacizumab plus paclitaxel for locally advanced or metastatic breast cancer. Oncotarget 2020, 11, 86–98. [Google Scholar] [CrossRef]
  211. Gao, L.; Zhao, P.; Li, Y.; Yang, D.; Hu, P.; Li, L.; Cheng, Y.; Yao, H. Reversal of PGlycoprotein-Mediated Multidrug Resistance by Novel Curcumin Analogues in Paclitaxel-resistant Human Breast Cancer Cells. Biochem. Cell Biol. 2020. [Google Scholar] [CrossRef] [PubMed]
  212. Bashmail, H.A.; Alamoudi, A.A.; Noorwali, A.; Hegazy, G.A.; Ajabnoor, G.M.; Al-Abd, A.M. Thymoquinone Enhances Paclitaxel Anti-Breast Cancer Activity via Inhibiting Tumor-Associated Stem Cells Despite Apparent Mathematical Antagonism. Molecules 2020, 25, 426. [Google Scholar] [CrossRef] [Green Version]
  213. Sakata, J.; Utsumi, F.; Suzuki, S.; Niimi, K.; Yamamoto, E.; Shibata, K.; Senga, T.; Kikkawa, F.; Kajiyama, H. Inhibition of ZEB1 leads to inversion of metastatic characteristics and restoration of paclitaxel sensitivity of chronic chemoresistant ovarian carcinoma cells. Oncotarget 2017, 8, 99482–99494. [Google Scholar] [CrossRef]
  214. Liu, Y.Y.; Zhang, L.Y.; Du, W.Z. Circular RNA circ-PVT1 contributes to paclitaxel resistance of gastric cancer cells through the regulation of ZEB1 expression by sponging miR-124-3p. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef]
  215. Jiang, G.; Wen, L.; Deng, W.; Jian, Z.; Zheng, H. Regulatory role of miR-211-5p in hepatocellular carcinoma metastasis by targeting ZEB2. Biomed. Pharmacother. 2017, 90, 806–812. [Google Scholar] [CrossRef]
  216. Papa, A.-L.; Sidiqui, A.; Balasubramanian, S.U.A.; Sarangi, S.; Luchette, M.; Sengupta, S.; Harfouche, R. PEGylated liposomal Gemcitabine: Insights into a potential breast cancer therapeutic. Cell. Oncol. 2013, 36, 449–457. [Google Scholar] [CrossRef]
  217. Samanta, K.; Setua, S.; Kumari, S.; Jaggi, M.; Yallapu, M.M.; Chauhan, S.C. Gemcitabine Combination Nano Therapies for Pancreatic Cancer. Pharmaceutics 2019, 11, 574. [Google Scholar] [CrossRef] [Green Version]
  218. Goel, S.; Sinha, R.J.; Bhaskar, V.; Aeron, R.; Sharma, A.; Singh, V. Role of gemcitabine and cisplatin as neoadjuvant chemotherapy in muscle invasive bladder cancer: Experience over the last decade. Asian J. Urol. 2019, 6, 222–229. [Google Scholar] [CrossRef] [PubMed]
  219. Wang, H.; Liu, K.; Chi, Z.; Zhou, X.; Ren, G.; Zhou, R.; Li, Y.; Tang, X.; Wang, X.J. Interplay of MKP-1 and Nrf2 drives tumor growth and drug resistance in non-small cell lung cancer. Aging 2019, 11, 11329–11346. [Google Scholar] [CrossRef]
  220. Yuan, Q.; Chu, H.; Ge, Y.; Ma, G.; Du, M.; Wang, M.; Zhang, Z.; Zhang, W. LncRNA PCAT1 and its genetic variant rs1902432 are associated with prostate cancer risk. J. Cancer 2018, 9, 1414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Ji, X.; Guo, H.; Yin, S.; Du, H. miR-139-5p functions as a tumor suppressor in cervical cancer by targeting TCF4 and inhibiting Wnt/beta-catenin signaling. OncoTargets Ther. 2019, 12, 7739–7748. [Google Scholar] [CrossRef] [Green Version]
  222. Guo, S.S.; Wang, Y.; Fan, Q.X. Raddeanin A promotes apoptosis and ameliorates 5-fluorouracil resistance in cholangiocarcinoma cells. World J. Gastroenterol. 2019, 25, 3380–3391. [Google Scholar] [CrossRef]
  223. Nakagawa, Y.; Kuranaga, Y.; Tahara, T.; Yamashita, H.; Shibata, T.; Nagasaka, M.; Funasaka, K.; Ohmiya, N.; Akao, Y. Induced miR-31 by 5-fluorouracil exposure contributes to the resistance in colorectal tumors. Cancer Sci. 2019, 110, 2540–2548. [Google Scholar] [CrossRef]
  224. Zhao, P.; Ma, Y.G.; Zhao, Y.; Liu, D.; Dai, Z.J.; Yan, C.Y.; Guan, H.T. MicroRNA-552 deficiency mediates 5-fluorouracil resistance by targeting SMAD2 signaling in DNA-mismatch-repair-deficient colorectal cancer. Cancer Chemother. Pharmacol. 2019, 84, 427–439. [Google Scholar] [CrossRef] [PubMed]
  225. Shan, G.; Tang, T.; Xia, Y.; Qian, H.-J. Long non-coding RNA NEAT1 promotes bladder progression through regulating miR-410 mediated HMGB1. Biomed. Pharmacother. 2020, 121, 109248. [Google Scholar] [CrossRef] [PubMed]
  226. Wang, W.; Ge, L.; Xu, X.J.; Yang, T.; Yuan, Y.; Ma, X.L.; Zhang, X.H. LncRNA NEAT1 promotes endometrial cancer cell proliferation, migration and invasion by regulating the miR-144-3p/EZH2 axis. Radiol. Oncol. 2019, 53, 434–442. [Google Scholar] [CrossRef] [Green Version]
  227. Liu, F.; Ai, F.Y.; Zhang, D.C.; Tian, L.; Yang, Z.Y.; Liu, S.J. LncRNA NEAT1 knockdown attenuates autophagy to elevate 5-FU sensitivity in colorectal cancer via targeting miR-34a. Cancer Med. 2020, 9, 1079–1091. [Google Scholar] [CrossRef] [Green Version]
  228. Liu, J.; Kong, D.; Sun, D.; Li, J. Long non-coding RNA CCAT2 acts as an oncogene in osteosarcoma through regulation of miR-200b/VEGF. Artif. Cells Nanomed. Biotechnol. 2019, 47, 2994–3003. [Google Scholar] [CrossRef] [Green Version]
  229. Soung, Y.H.; Chung, H.; Yan, C.; Ju, J.; Chung, J. Arrestin Domain Containing 3 Reverses Epithelial to Mesenchymal Transition and Chemo-Resistance of TNBC Cells by Up-Regulating Expression of miR-200b. Cells 2019, 8, 692. [Google Scholar] [CrossRef] [Green Version]
  230. Fang, S.; Zeng, X.; Zhu, W.; Tang, R.; Chao, Y.; Guo, L. Zinc finger E-box-binding homeobox 2 (ZEB2) regulated by miR-200b contributes to multi-drug resistance of small cell lung cancer. Exp. Mol. Pathol. 2014, 96, 438–444. [Google Scholar] [CrossRef]
  231. Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.S. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. In Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2015; pp. S185–S198. [Google Scholar]
  232. Spranger, S. Mechanisms of tumor escape in the context of the T-cell-inflamed and the non-T-cell-inflamed tumor microenvironment. Int. Immunol. 2016, 28, 383–391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Davis, R.J.; Van Waes, C.; Allen, C.T. Overcoming barriers to effective immunotherapy: MDSCs, TAMs, and Tregs as mediators of the immunosuppressive microenvironment in head and neck cancer. Oral Oncol. 2016, 58, 59–70. [Google Scholar] [CrossRef] [Green Version]
  234. Restifo, N.P.; Dudley, M.E.; Rosenberg, S.A. Adoptive immunotherapy for cancer: Harnessing the T cell response. Nat. Rev. Immunol. 2012, 12, 269–281. [Google Scholar] [CrossRef]
  235. Wherry, E.J. T cell exhaustion. Nat. Immunol. 2011, 12, 492–499. [Google Scholar] [CrossRef]
  236. Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.; Hwu, W.-J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K. Safety and activity of anti–PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Chen, L.; Flies, D.B. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat. Rev. Immunol. 2013, 13, 227–242. [Google Scholar] [CrossRef] [PubMed]
  238. Hamid, O.; Robert, C.; Daud, A.; Hodi, F.S.; Hwu, W.-J.; Kefford, R.; Wolchok, J.D.; Hersey, P.; Joseph, R.W.; Weber, J.S. Safety and tumor responses with lambrolizumab (anti–PD-1) in melanoma. N. Engl. J. Med. 2013, 369, 134–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B. Safety, activity, and immune correlates of anti–PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef] [PubMed]
  240. Youngblood, B.; Wherry, E.J.; Ahmed, R. Acquired transcriptional programming in functional and exhausted virus-specific CD8 T cells. Curr. Opin. HIV AIDS 2012, 7, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Dong, H.; Strome, S.E.; Salomao, D.R.; Tamura, H.; Hirano, F.; Flies, D.B.; Roche, P.C.; Lu, J.; Zhu, G.; Tamada, K. Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nat. Med. 2002, 8, 793–800. [Google Scholar] [CrossRef] [PubMed]
  242. Freeman, G.J.; Sharpe, A.H.; Kuchroo, V.K. Protect the killer: CTLs need defenses against the tumor. Nat. Med. 2002, 8, 787–789. [Google Scholar] [CrossRef]
  243. Hobo, W.; Maas, F.; Adisty, N.; de Witte, T.; Schaap, N.; van der Voort, R.; Dolstra, H. siRNA silencing of PD-L1 and PD-L2 on dendritic cells augments expansion and function of minor histocompatibility antigen–specific CD8+ T cells. Blood J. Am. Soc. Hematol. 2010, 116, 4501–4511. [Google Scholar] [CrossRef] [Green Version]
  244. Zha, H.; Han, X.; Zhu, Y.; Yang, F.; Li, Y.; Li, Q.; Guo, B.; Zhu, B. Blocking C5aR signaling promotes the anti-tumor efficacy of PD-1/PD-L1 blockade. Oncoimmunology 2017, 6, e1349587. [Google Scholar] [CrossRef]
  245. Zhao, L.; Liu, Y.; Zhang, J.; Liu, Y.; Qi, Q. LncRNA SNHG14/miR-5590-3p/ZEB1 positive feedback loop promoted diffuse large B cell lymphoma progression and immune evasion through regulating PD-1/PD-L1 checkpoint. Cell Death Dis. 2019, 10, 1–15. [Google Scholar] [CrossRef]
  246. Chen, L.; Gibbons, D.L.; Goswami, S.; Cortez, M.A.; Ahn, Y.H.; Byers, L.A.; Zhang, X.; Yi, X.; Dwyer, D.; Lin, W.; et al. Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat. Commun. 2014, 5, 5241. [Google Scholar] [CrossRef]
  247. Noman, M.Z.; Janji, B.; Abdou, A.; Hasmim, M.; Terry, S.; Tan, T.Z.; Mami-Chouaib, F.; Thiery, J.P.; Chouaib, S. The immune checkpoint ligand PD-L1 is upregulated in EMT-activated human breast cancer cells by a mechanism involving ZEB-1 and miR-200. Oncoimmunology 2017, 6, e1263412. [Google Scholar] [CrossRef]
  248. Wang, Y.; Luo, Y.; Guan, W.; Zhao, H. Role of miR-23a/Zeb1 negative feedback loop in regulating epithelial-mesenchymal transition and tumorigenicity of intraocular tumors. Oncol. Lett. 2018, 16, 2462–2470. [Google Scholar] [CrossRef]
  249. Majid, S.; Dar, A.A.; Saini, S.; Deng, G.; Chang, I.; Greene, K.; Tanaka, Y.; Dahiya, R.; Yamamura, S. MicroRNA-23b functions as a tumor suppressor by regulating Zeb1 in bladder cancer. PLoS ONE 2013, 8, e67686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  250. Zhang, P.; Huang, C.; Fu, C.; Tian, Y.; Hu, Y.; Wang, B.; Strasner, A.; Song, Y.; Song, E. Cordycepin (3’-deoxyadenosine) suppressed HMGA2, Twist1 and ZEB1-dependent melanoma invasion and metastasis by targeting miR-33b. Oncotarget 2015, 6, 9834–9853. [Google Scholar] [CrossRef] [Green Version]
  251. Jiang, R.; Zhang, C.; Liu, G.; Gu, R.; Wu, H. MicroRNA-126 Inhibits Proliferation, Migration, Invasion, and EMT in Osteosarcoma by Targeting ZEB1. J. Cell. Biochem. 2017, 118, 3765–3774. [Google Scholar] [CrossRef]
  252. Sun, X.; Li, Y.; Yu, J.; Pei, H.; Luo, P.; Zhang, J. miR-128 modulates chemosensitivity and invasion of prostate cancer cells through targeting ZEB1. Jpn. J. Clin. Oncol. 2015, 45, 474–482. [Google Scholar] [CrossRef] [Green Version]
  253. Dong, P.; Karaayvaz, M.; Jia, N.; Kaneuchi, M.; Hamada, J.; Watari, H.; Sudo, S.; Ju, J.; Sakuragi, N. Mutant p53 gain-of-function induces epithelial-mesenchymal transition through modulation of the miR-130b-ZEB1 axis. Oncogene 2013, 32, 3286–3295. [Google Scholar] [CrossRef] [Green Version]
  254. Qiu, G.; Lin, Y.; Zhang, H.; Wu, D. miR-139-5p inhibits epithelial-mesenchymal transition, migration and invasion of hepatocellular carcinoma cells by targeting ZEB1 and ZEB2. Biochem. Biophys. Res. Commun. 2015, 463, 315–321. [Google Scholar] [CrossRef]
  255. Yue, S.; Wang, L.; Zhang, H.; Min, Y.; Lou, Y.; Sun, H.; Jiang, Y.; Zhang, W.; Liang, A.; Guo, Y.; et al. miR-139-5p suppresses cancer cell migration and invasion through targeting ZEB1 and ZEB2 in GBM. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2015, 36, 6741–6749. [Google Scholar] [CrossRef]
  256. Al-Khalaf, H.H.; Aboussekhra, A. MicroRNA-141 and microRNA-146b-5p inhibit the prometastatic mesenchymal characteristics through the RNA-binding protein AUF1 targeting the transcription factor ZEB1 and the protein kinase AKT. J. Biol. Chem. 2014, 289, 31433–31447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Pan, Y.; Zhang, J.; Fu, H.; Shen, L. miR-144 functions as a tumor suppressor in breast cancer through inhibiting ZEB1/2-mediated epithelial mesenchymal transition process. OncoTargets Ther. 2016, 9, 6247–6255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  258. Guan, H.; Liang, W.; Xie, Z.; Li, H.; Liu, J.; Liu, L.; Xiu, L.; Li, Y. Down-regulation of miR-144 promotes thyroid cancer cell invasion by targeting ZEB1 and ZEB2. Endocrine 2015, 48, 566–574. [Google Scholar] [CrossRef]
  259. Yokobori, T.; Suzuki, S.; Tanaka, N.; Inose, T.; Sohda, M.; Sano, A.; Sakai, M.; Nakajima, M.; Miyazaki, T.; Kato, H.; et al. MiR-150 is associated with poor prognosis in esophageal squamous cell carcinoma via targeting the EMT inducer ZEB1. Cancer Sci. 2013, 104, 48–54. [Google Scholar] [CrossRef]
  260. Jin, M.; Yang, Z.; Ye, W.; Xu, H.; Hua, X. MicroRNA-150 predicts a favorable prognosis in patients with epithelial ovarian cancer, and inhibits cell invasion and metastasis by suppressing transcriptional repressor ZEB1. PLoS ONE 2014, 9, e103965. [Google Scholar] [CrossRef] [Green Version]
  261. Liang, L.; Zhang, Z.; Qin, X.; Gao, Y.; Zhao, P.; Liu, J.; Zeng, W. Gambogic Acid Inhibits Melanoma through Regulation of miR-199a-3p/ZEB1 Signalling. Basic Clin. Pharmacol. Toxicol. 2018, 123, 692–703. [Google Scholar] [CrossRef] [Green Version]
  262. Wang, J.; Zhou, F.; Yin, L.; Zhao, L.; Zhang, Y.; Wang, J. MicroRNA-199b targets the regulation of ZEB1 expression to inhibit cell proliferation, migration and invasion in nonsmall cell lung cancer. Mol. Med. Rep. 2017, 16, 5007–5014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Ungewiss, C.; Rizvi, Z.H.; Roybal, J.D.; Peng, D.H.; Gold, K.A.; Shin, D.H.; Creighton, C.J.; Gibbons, D.L. The microRNA-200/Zeb1 axis regulates ECM-dependent beta1-integrin/FAK signaling, cancer cell invasion and metastasis through CRKL. Sci. Rep. 2016, 6, 18652. [Google Scholar] [CrossRef]
  264. Asakura, T.; Yamaguchi, N.; Ohkawa, K.; Yoshida, K. Proteasome inhibitor-resistant cells cause EMT-induction via suppression of E-cadherin by miR-200 and ZEB1. Int. J. Oncol. 2015, 46, 2251–2260. [Google Scholar] [CrossRef] [Green Version]
  265. Huang, W.T.; Kuo, S.H.; Cheng, A.L.; Lin, C.W. Inhibition of ZEB1 by miR-200 characterizes Helicobacter pylori-positive gastric diffuse large B-cell lymphoma with a less aggressive behavior. Mod. Pathol. Off. J. USA Can. Acad. Pathol. Inc. 2014, 27, 1116–1125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Title, A.C.; Hong, S.J.; Pires, N.D.; Hasenohrl, L.; Godbersen, S.; Stokar-Regenscheit, N.; Bartel, D.P.; Stoffel, M. Genetic dissection of the miR-200-Zeb1 axis reveals its importance in tumor differentiation and invasion. Nat. Commun. 2018, 9, 4671. [Google Scholar] [CrossRef] [PubMed]
  267. Li, Y.; Zeng, C.; Tu, M.; Jiang, W.; Dai, Z.; Hu, Y.; Deng, Z.; Xiao, W. MicroRNA-200b acts as a tumor suppressor in osteosarcoma via targeting ZEB1. OncoTargets Ther. 2016, 9, 3101–3111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  268. Tsai, S.C.; Lin, C.C.; Shih, T.C.; Tseng, R.J.; Yu, M.C.; Lin, Y.J.; Hsieh, S.Y. The miR-200b-ZEB1 circuit regulates diverse stemness of human hepatocellular carcinoma. Mol. Carcinog. 2017, 56, 2035–2047. [Google Scholar] [CrossRef] [PubMed]
  269. Nishijima, N.; Seike, M.; Soeno, C.; Chiba, M.; Miyanaga, A.; Noro, R.; Sugano, T.; Matsumoto, M.; Kubota, K.; Gemma, A. miR-200/ZEB axis regulates sensitivity to nintedanib in non-small cell lung cancer cells. Int. J. Oncol. 2016, 48, 937–944. [Google Scholar] [CrossRef] [Green Version]
  270. Chen, M.L.; Liang, L.S.; Wang, X.K. miR-200c inhibits invasion and migration in human colon cancer cells SW480/620 by targeting ZEB1. Clin. Exp. Metastasis 2012, 29, 457–469. [Google Scholar] [CrossRef]
  271. Kurata, A.; Yamada, M.; Ohno, S.I.; Inoue, S.; Hashimoto, H.; Fujita, K.; Takanashi, M.; Kuroda, M. Expression level of microRNA-200c is associated with cell morphology in vitro and histological differentiation through regulation of ZEB1/2 and E-cadherin in gastric carcinoma. Oncol. Rep. 2018, 39, 91–100. [Google Scholar] [CrossRef] [Green Version]
  272. Shan, Y.; Zhang, L.; Bao, Y.; Li, B.; He, C.; Gao, M.; Feng, X.; Xu, W.; Zhang, X.; Wang, S. Epithelial-mesenchymal transition, a novel target of sulforaphane via COX-2/MMP2, 9/Snail, ZEB1 and miR-200c/ZEB1 pathways in human bladder cancer cells. J. Nutr. Biochem. 2013, 24, 1062–1069. [Google Scholar] [CrossRef]
  273. Karagur, E.R.; Ozay, C.; Mammadov, R.; Akca, H. Anti-invasive effect of Cyclamen pseudibericum extract on A549 non-small cell lung carcinoma cells via inhibition of ZEB1 mediated by miR-200c. J. Nat. Med. 2018, 72, 686–693. [Google Scholar] [CrossRef]
  274. Zhou, G.; Zhang, F.; Guo, Y.; Huang, J.; Xie, Y.; Yue, S.; Chen, M.; Jiang, H.; Li, M. miR-200c enhances sensitivity of drug-resistant non-small cell lung cancer to gefitinib by suppression of PI3K/Akt signaling pathway and inhibites cell migration via targeting ZEB1. Biomed. Pharmacother. 2017, 85, 113–119. [Google Scholar] [CrossRef]
  275. Gao, H.X.; Yan, L.; Li, C.; Zhao, L.M.; Liu, W. miR-200c regulates crizotinib-resistant ALK-positive lung cancer cells by reversing epithelial-mesenchymal transition via targeting ZEB1. Mol. Med. Rep. 2016, 14, 4135–4143. [Google Scholar] [CrossRef] [Green Version]
  276. Guo, E.; Wang, Z.; Wang, S. MiR-200c and miR-141 inhibit ZEB1 synergistically and suppress glioma cell growth and migration. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 3385–3391. [Google Scholar] [PubMed]
  277. Zhou, X.; Wang, Y.; Shan, B.; Han, J.; Zhu, H.; Lv, Y.; Fan, X.; Sang, M.; Liu, X.D.; Liu, W. The downregulation of miR-200c/141 promotes ZEB1/2 expression and gastric cancer progression. Med. Oncol. (Northwood Lond. Engl.) 2015, 32, 428. [Google Scholar] [CrossRef] [PubMed]
  278. Singh, T.; Prasad, R.; Katiyar, S.K. Therapeutic intervention of silymarin on the migration of non-small cell lung cancer cells is associated with the axis of multiple molecular targets including class 1 HDACs, ZEB1 expression, and restoration of miR-203 and E-cadherin expression. Am. J. Cancer Res. 2016, 6, 1287–1301. [Google Scholar]
  279. Wu, G.; Wang, J.; Chen, G.; Zhao, X. microRNA-204 modulates chemosensitivity and apoptosis of prostate cancer cells by targeting zinc-finger E-box-binding homeobox 1 (ZEB1). Am. J. Transl. Res. 2017, 9, 3599–3610. [Google Scholar]
  280. Niu, K.; Shen, W.; Zhang, Y.; Zhao, Y.; Lu, Y. MiR-205 promotes motility of ovarian cancer cells via targeting ZEB1. Gene 2015, 574, 330–336. [Google Scholar] [CrossRef]
  281. El Bezawy, R.; Tinelli, S.; Tortoreto, M.; Doldi, V.; Zuco, V.; Folini, M.; Stucchi, C.; Rancati, T.; Valdagni, R.; Gandellini, P.; et al. miR-205 enhances radiation sensitivity of prostate cancer cells by impairing DNA damage repair through PKCepsilon and ZEB1 inhibition. J. Exp. Clin. Cancer Res. CR 2019, 38, 51. [Google Scholar] [CrossRef]
  282. Zhang, P.; Wang, L.; Rodriguez-Aguayo, C.; Yuan, Y.; Debeb, B.G.; Chen, D.; Sun, Y.; You, M.J.; Liu, Y.; Dean, D.C.; et al. miR-205 acts as a tumour radiosensitizer by targeting ZEB1 and Ubc13. Nat. Commun. 2014, 5, 5671. [Google Scholar] [CrossRef] [Green Version]
  283. Li, L.; Li, S. miR-205-5p inhibits cell migration and invasion in prostatic carcinoma by targeting ZEB1. Oncol. Lett. 2018, 16, 1715–1721. [Google Scholar] [CrossRef] [Green Version]
  284. Hou, L.K.; Yu, Y.; Xie, Y.G.; Wang, J.; Mao, J.F.; Zhang, B.; Wang, X.; Cao, X.C. miR-340 and ZEB1 negative feedback loop regulates TGF-beta- mediated breast cancer progression. Oncotarget 2016, 7, 26016–26026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Yan, H.; Zhang, B.; Fang, C.; Chen, L. miR-340 alleviates chemoresistance of osteosarcoma cells by targeting ZEB1. Anti-Cancer Drugs 2018, 29, 440–448. [Google Scholar] [CrossRef] [PubMed]
  286. Barbachano, A.; Fernandez-Barral, A.; Pereira, F.; Segura, M.F.; Ordonez-Moran, P.; Carrillo-de Santa Pau, E.; Gonzalez-Sancho, J.M.; Hanniford, D.; Martinez, N.; Costales-Carrera, A.; et al. SPROUTY-2 represses the epithelial phenotype of colon carcinoma cells via upregulation of ZEB1 mediated by ETS1 and miR-200/miR-150. Oncogene 2016, 35, 2991–3003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  287. Zou, J.; Liu, L.; Wang, Q.; Yin, F.; Yang, Z.; Zhang, W.; Li, L. Downregulation of miR-429 contributes to the development of drug resistance in epithelial ovarian cancer by targeting ZEB1. Am. J. Transl. Res. 2017, 9, 1357–1368. [Google Scholar] [PubMed]
  288. Lei, W.; Liu, Y.E.; Zheng, Y.; Qu, L. MiR-429 inhibits oral squamous cell carcinoma growth by targeting ZEB1. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2015, 21, 383–389. [Google Scholar] [CrossRef]
  289. Sun, K.; Zeng, T.; Huang, D.; Liu, Z.; Huang, S.; Liu, J.; Qu, Z. MicroRNA-431 inhibits migration and invasion of hepatocellular carcinoma cells by targeting the ZEB1-mediated epithelial-mensenchymal transition. FEBS Open Bio 2015, 5, 900–907. [Google Scholar] [CrossRef] [Green Version]
  290. Ma, P.; Ni, K.; Ke, J.; Zhang, W.; Feng, Y.; Mao, Q. miR-448 inhibits the epithelial-mesenchymal transition in breast cancer cells by directly targeting the E-cadherin repressor ZEB1/2. Exp. Biol. Med. (Maywood, NJ, USA) 2018, 243, 473–480. [Google Scholar] [CrossRef] [PubMed]
  291. Li, Y.J.; Ping, C.; Tang, J.; Zhang, W. MicroRNA-455 suppresses non-small cell lung cancer through targeting ZEB1. Cell Biol. Int. 2016, 40, 621–628. [Google Scholar] [CrossRef]
  292. Hu, Y.; Xie, H.; Liu, Y.; Liu, W.; Liu, M.; Tang, H. miR-484 suppresses proliferation and epithelial-mesenchymal transition by targeting ZEB1 and SMAD2 in cervical cancer cells. Cancer Cell Int. 2017, 17, 36. [Google Scholar] [CrossRef] [Green Version]
  293. Gao, H.; Li, X.; Zhan, G.; Zhu, Y.; Yu, J.; Wang, J.; Li, L.; Wu, W.; Liu, N.; Guo, X. Long noncoding RNA MAGI1-IT1 promoted invasion and metastasis of epithelial ovarian cancer via the miR-200a/ZEB axis. Cell Cycle 2019, 18, 1393–1406. [Google Scholar] [CrossRef]
  294. Guo, S.J.; Zeng, H.X.; Huang, P.; Wang, S.; Xie, C.H.; Li, S.J. MiR-508-3p inhibits cell invasion and epithelial-mesenchymal transition by targeting ZEB1 in triple-negative breast cancer. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 6379–6385. [Google Scholar] [CrossRef]
  295. Wang, M.; Zhang, R.; Zhang, S.; Xu, R.; Yang, Q. MicroRNA-574-3p regulates epithelial mesenchymal transition and cisplatin resistance via targeting ZEB1 in human gastric carcinoma cells. Gene 2019, 700, 110–119. [Google Scholar] [CrossRef]
  296. Pang, H.; Zheng, Y.; Zhao, Y.; Xiu, X.; Wang, J. miR-590-3p suppresses cancer cell migration, invasion and epithelial-mesenchymal transition in glioblastoma multiforme by targeting ZEB1 and ZEB2. Biochem. Biophys. Res. Commun. 2015, 468, 739–745. [Google Scholar] [CrossRef]
  297. Yao, R.; Zheng, H.; Wu, L.; Cai, P. miRNA-641 inhibits the proliferation, migration, and invasion and induces apoptosis of cervical cancer cells by directly targeting ZEB1. OncoTargets Ther. 2018, 11, 8965–8976. [Google Scholar] [CrossRef] [Green Version]
  298. Deng, S.; Li, X.; Niu, Y.; Zhu, S.; Jin, Y.; Deng, S.; Chen, J.; Liu, Y.; He, C.; Yin, T.; et al. MiR-652 inhibits acidic microenvironment-induced epithelial-mesenchymal transition of pancreatic cancer cells by targeting ZEB1. Oncotarget 2015, 6, 39661–39675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  299. Harazono, Y.; Muramatsu, T.; Endo, H.; Uzawa, N.; Kawano, T.; Harada, K.; Inazawa, J.; Kozaki, K. miR-655 Is an EMT-suppressive microRNA targeting ZEB1 and TGFBR2. PLoS ONE 2013, 8, e62757. [Google Scholar] [CrossRef] [Green Version]
  300. Wang, J.; Zhang, Y.; Wei, H.; Zhang, X.; Wu, Y.; Gong, A.; Xia, Y.; Wang, W.; Xu, M. The mir-675-5p regulates the progression and development of pancreatic cancer via the UBQLN1-ZEB1-mir200 axis. Oncotarget 2017, 8, 24978–24987. [Google Scholar] [CrossRef] [Green Version]
  301. Li, G.; Xu, Y.; Wang, S.; Yan, W.; Zhao, Q.; Guo, J. MiR-873-5p inhibits cell migration, invasion and epithelial-mesenchymal transition in colorectal cancer via targeting ZEB1. Pathol. Res. Pract. 2019, 215, 34–39. [Google Scholar] [CrossRef]
  302. El Bezawy, R.; Cominetti, D.; Fenderico, N.; Zuco, V.; Beretta, G.L.; Dugo, M.; Arrighetti, N.; Stucchi, C.; Rancati, T.; Valdagni, R.; et al. miR-875-5p counteracts epithelial-to-mesenchymal transition and enhances radiation response in prostate cancer through repression of the EGFR-ZEB1 axis. Cancer Lett. 2017, 395, 53–62. [Google Scholar] [CrossRef]
  303. Liu, H.; Wang, H.; Liu, X.; Yu, T. miR-1271 inhibits migration, invasion and epithelial-mesenchymal transition by targeting ZEB1 and TWIST1 in pancreatic cancer cells. Biochem. Biophys. Res. Commun. 2016, 472, 346–352. [Google Scholar] [CrossRef]
  304. Wang, Y.; Yan, S.; Liu, X.; Zhang, W.; Li, Y.; Dong, R.; Zhang, Q.; Yang, Q.; Yuan, C.; Shen, K.; et al. miR-1236-3p represses the cell migration and invasion abilities by targeting ZEB1 in high-grade serous ovarian carcinoma. Oncol. Rep. 2014, 31, 1905–1910. [Google Scholar] [CrossRef] [Green Version]
  305. Liang, T.C.; Fu, W.G.; Zhong, Y.S. MicroRNA-1236-3p inhibits proliferation and invasion of breast cancer cells by targeting ZEB1. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 9988–9995. [Google Scholar] [CrossRef] [PubMed]
  306. Zhu, L.; Liu, Z.; Dong, R.; Wang, X.; Zhang, M.; Guo, X.; Yu, N.; Zeng, A. MicroRNA-3662 targets ZEB1 and attenuates the invasion of the highly aggressive melanoma cell line A375. Cancer Manag. Res. 2019, 11, 5845–5856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  307. Zhang, C.; Wang, L.; Yang, J.; Fu, Y.; Li, H.; Xie, L.; Cui, Y. MicroRNA-33a-5p suppresses esophageal squamous cell carcinoma progression via regulation of lncRNA DANCR and ZEB1. Eur. J. Pharmacol. 2019, 861, 172590. [Google Scholar] [CrossRef] [PubMed]
  308. Chang, L.; Yuan, Y.; Li, C.; Guo, T.; Qi, H.; Xiao, Y.; Dong, X.; Liu, Z.; Liu, Q. Upregulation of SNHG6 regulates ZEB1 expression by competitively binding miR-101-3p and interacting with UPF1 in hepatocellular carcinoma. Cancer Lett. 2016, 383, 183–194. [Google Scholar] [CrossRef]
  309. Liang, H.; Yu, T.; Han, Y.; Jiang, H.; Wang, C.; You, T.; Zhao, X.; Shan, H.; Yang, R.; Yang, L.; et al. LncRNA PTAR promotes EMT and invasion-metastasis in serous ovarian cancer by competitively binding miR-101-3p to regulate ZEB1 expression. Mol. Cancer 2018, 17, 119. [Google Scholar] [CrossRef] [PubMed]
  310. Li, Y.; Lv, M.; Song, Z.; Lou, Z.; Wang, R.; Zhuang, M. Long non-coding RNA NNT-AS1 affects progression of breast cancer through miR-142-3p/ZEB1 axis. Biomed. Pharmacother. 2018, 103, 939–946. [Google Scholar] [CrossRef]
  311. He, C.; Liu, Z.; Jin, L.; Zhang, F.; Peng, X.; Xiao, Y.; Wang, X.; Lyu, Q.; Cai, X. lncRNA TUG1-Mediated Mir-142-3p Downregulation Contributes to Metastasis and the Epithelial-to-Mesenchymal Transition of Hepatocellular Carcinoma by Targeting ZEB1. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2018, 48, 1928–1941. [Google Scholar] [CrossRef]
  312. Zhang, K.; Chen, J.; Song, H.; Chen, L.B. SNHG16/miR-140-5p axis promotes esophagus cancer cell proliferation, migration and EMT formation through regulating ZEB1. Oncotarget 2018, 9, 1028–1040. [Google Scholar] [CrossRef] [Green Version]
  313. Zhu, C.; Cheng, D.; Qiu, X.; Zhuang, M.; Liu, Z. Long Noncoding RNA SNHG16 Promotes Cell Proliferation by Sponging MicroRNA-205 and Upregulating ZEB1 Expression in Osteosarcoma. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2018, 51, 429–440. [Google Scholar] [CrossRef]
  314. Wang, B.; Qu, X.L.; Liu, J.; Lu, J.; Zhou, Z.Y. HOTAIR promotes osteosarcoma development by sponging miR-217 and targeting ZEB1. J. Cell. Physiol. 2019, 234, 6173–6181. [Google Scholar] [CrossRef]
  315. Yang, T.; He, X.; Chen, A.; Tan, K.; Du, X. LncRNA HOTAIR contributes to the malignancy of hepatocellular carcinoma by enhancing epithelial-mesenchymal transition via sponging miR-23b-3p from ZEB1. Gene 2018, 670, 114–122. [Google Scholar] [CrossRef] [PubMed]
  316. Xue, M.; Pang, H.; Li, X.; Li, H.; Pan, J.; Chen, W. Long non-coding RNA urothelial cancer-associated 1 promotes bladder cancer cell migration and invasion by way of the hsa-miR-145-ZEB1/2-FSCN1 pathway. Cancer Sci. 2016, 107, 18–27. [Google Scholar] [CrossRef] [PubMed]
  317. Liang, C.; Yang, Y.; Guan, J.; Lv, T.; Qu, S.; Fu, Q.; Zhao, H. LncRNA UCA1 sponges miR-204-5p to promote migration, invasion and epithelial-mesenchymal transition of glioma cells via upregulation of ZEB1. Pathol. Res. Pract. 2018, 214, 1474–1481. [Google Scholar] [CrossRef]
  318. Meng, L.; Ma, P.; Cai, R.; Guan, Q.; Wang, M.; Jin, B. Long noncoding RNA ZEB1-AS1 promotes the tumorigenesis of glioma cancer cells by modulating the miR-200c/141-ZEB1 axis. Am. J. Transl. Res. 2018, 10, 3395–3412. [Google Scholar] [PubMed]
  319. Qu, R.; Chen, X.; Zhang, C. LncRNA ZEB1-AS1/miR-409-3p/ZEB1 feedback loop is involved in the progression of non-small cell lung cancer. Biochem. Biophys. Res. Commun. 2018, 507, 450–456. [Google Scholar] [CrossRef] [PubMed]
  320. Xiong, W.C.; Han, N.; Wu, N.; Zhao, K.L.; Han, C.; Wang, H.X.; Ping, G.F.; Zheng, P.F.; Feng, H.; Qin, L.; et al. Interplay between long noncoding RNA ZEB1-AS1 and miR-101/ZEB1 axis regulates proliferation and migration of colorectal cancer cells. Am. J. Transl. Res. 2018, 10, 605–617. [Google Scholar]
  321. Jin, H.; Jin, X.; Chai, W.; Yin, Z.; Li, Y.; Dong, F.; Wang, W. Long non-coding RNA MIAT competitively binds miR-150-5p to regulate ZEB1 expression in osteosarcoma. Oncol. Lett. 2019, 17, 1229–1236. [Google Scholar] [CrossRef]
  322. Gao, H.; Li, S.P.; Xu, H.X.; Yu, Y.; He, J.D.; Wang, Z.; Xu, Y.J.; Wang, C.Y.; Zhang, H.M.; Zhang, R.X.; et al. LncRNA HULC enhances epithelial-mesenchymal transition to promote tumorigenesis and metastasis of hepatocellular carcinoma via the miR-200a-3p/ZEB1 signaling pathway. Oncotarget 2016, 7, 42431–42446. [Google Scholar] [CrossRef] [Green Version]
  323. Lu, Y.; Li, T.; Wei, G.; Liu, L.; Chen, Q.; Xu, L.; Zhang, K.; Zeng, D.; Liao, R. The long non-coding RNA NEAT1 regulates epithelial to mesenchymal transition and radioresistance in through miR-204/ZEB1 axis in nasopharyngeal carcinoma. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2016, 37, 11733–11741. [Google Scholar] [CrossRef]
  324. Zhong, Q.; Chen, Y.; Chen, Z. LncRNA MINCR regulates irradiation resistance in nasopharyngeal carcinoma cells via the microRNA-223/ZEB1 axis. Cell Cycle (Georgetown, Tex.) 2020, 19, 53–66. [Google Scholar] [CrossRef]
  325. Yuan, G.; Quan, J.; Dong, D.; Wang, Q. Long Noncoding RNA CAT104 Promotes Cell Viability, Migration, and Invasion in Gastric Carcinoma Cells Through Activation of MicroRNA-381-Inhibiting Zinc Finger E-box-Binding Homeobox 1 (ZEB1) Expression. Oncol. Res. 2018, 26, 1037–1046. [Google Scholar] [CrossRef]
  326. Wang, P.S.; Chou, C.H.; Lin, C.H.; Yao, Y.C.; Cheng, H.C.; Li, H.Y.; Chuang, Y.C.; Yang, C.N.; Ger, L.P.; Chen, Y.C.; et al. A novel long non-coding RNA linc-ZNF469-3 promotes lung metastasis through miR-574-5p-ZEB1 axis in triple negative breast cancer. Oncogene 2018, 37, 4662–4678. [Google Scholar] [CrossRef]
  327. Zheng, L.; Xu, M.; Xu, J.; Wu, K.; Fang, Q.; Liang, Y.; Zhou, S.; Cen, D.; Ji, L.; Han, W.; et al. ELF3 promotes epithelial-mesenchymal transition by protecting ZEB1 from miR-141-3p-mediated silencing in hepatocellular carcinoma. Cell Death Dis. 2018, 9, 387. [Google Scholar] [CrossRef] [PubMed]
  328. Guo, L.; Chen, C.; Shi, M.; Wang, F.; Chen, X.; Diao, D.; Hu, M.; Yu, M.; Qian, L.; Guo, N. Stat3-coordinated Lin-28-let-7-HMGA2 and miR-200-ZEB1 circuits initiate and maintain oncostatin M-driven epithelial-mesenchymal transition. Oncogene 2013, 32, 5272–5282. [Google Scholar] [CrossRef] [PubMed]
  329. Zhang, N.; Liu, Y.; Wang, Y.; Zhao, M.; Tu, L.; Luo, F. Decitabine reverses TGF-beta1-induced epithelial-mesenchymal transition in non-small-cell lung cancer by regulating miR-200/ZEB axis. Drug Des. Dev. Ther. 2017, 11, 969–983. [Google Scholar] [CrossRef] [Green Version]
  330. Chung, V.Y.; Tan, T.Z.; Tan, M.; Wong, M.K.; Kuay, K.T.; Yang, Z.; Ye, J.; Muller, J.; Koh, C.M.; Guccione, E.; et al. GRHL2-miR-200-ZEB1 maintains the epithelial status of ovarian cancer through transcriptional regulation and histone modification. Sci. Rep. 2016, 6, 19943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  331. Kong, X.; Ding, X.; Li, X.; Gao, S.; Yang, Q. 53BP1 suppresses epithelial-mesenchymal transition by downregulating ZEB1 through microRNA-200b/429 in breast cancer. Cancer Sci. 2015, 106, 982–989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  332. Lee, J.Y.; Park, M.K.; Park, J.H.; Lee, H.J.; Shin, D.H.; Kang, Y.; Lee, C.H.; Kong, G. Loss of the polycomb protein Mel-18 enhances the epithelial-mesenchymal transition by ZEB1 and ZEB2 expression through the downregulation of miR-205 in breast cancer. Oncogene 2014, 33, 1325–1335. [Google Scholar] [CrossRef] [Green Version]
  333. Zhao, W.; Wang, H.; Han, X.; Ma, J.; Zhou, Y.; Chen, Z.; Zhou, H.; Xu, H.; Sun, Z.; Kong, B.; et al. DeltaNp63alpha attenuates tumor aggressiveness by suppressing miR-205/ZEB1-mediated epithelial-mesenchymal transition in cervical squamous cell carcinoma. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2016, 37, 10621–10632. [Google Scholar] [CrossRef]
  334. Bian, Y.; Gao, G.; Zhang, Q.; Qian, H.; Yu, L.; Yao, N.; Qian, J.; Liu, B.; Qian, X. KCNQ1OT1/miR-217/ZEB1 feedback loop facilitates cell migration and epithelial-mesenchymal transition in colorectal cancer. Cancer Biol. Ther. 2019, 20, 886–896. [Google Scholar] [CrossRef]
  335. Xiao, T.; Xue, J.; Shi, M.; Chen, C.; Luo, F.; Xu, H.; Chen, X.; Sun, B.; Sun, Q.; Yang, Q.; et al. Circ008913, via miR-889 regulation of DAB2IP/ZEB1, is involved in the arsenite-induced acquisition of CSC-like properties by human keratinocytes in carcinogenesis. Met. Integr. Biometal Sci. 2018, 10, 1328–1338. [Google Scholar] [CrossRef] [PubMed]
  336. Wang, H.; An, X.; Yu, H.; Zhang, S.; Tang, B.; Zhang, X.; Li, Z. MiR-29b/TET1/ZEB2 signaling axis regulates metastatic properties and epithelial-mesenchymal transition in breast cancer cells. Oncotarget 2017, 8, 102119–102133. [Google Scholar] [CrossRef] [PubMed]
  337. Chen, Z.; Zhang, J.; Zhang, Z.; Feng, Z.; Wei, J.; Lu, J.; Fang, Y.; Liang, Y.; Cen, J.; Pan, Y.; et al. The putative tumor suppressor microRNA-30a-5p modulates clear cell renal cell carcinoma aggressiveness through repression of ZEB2. Cell Death Dis. 2017, 8, e2859. [Google Scholar] [CrossRef]
  338. Ji, H.; Sang, M.; Liu, F.; Ai, N.; Geng, C. miR-124 regulates EMT based on ZEB2 target to inhibit invasion and metastasis in triple-negative breast cancer. Pathol. Res. Pract. 2019, 215, 697–704. [Google Scholar] [CrossRef]
  339. Li, X.; Li, C.; Bi, H.; Bai, S.; Zhao, L.; Zhang, J.; Qi, C. Targeting ZEB2 By microRNA-129 In Non-Small Cell Lung Cancer Suppresses Cell Proliferation, Invasion And Migration Via Regulating Wnt/beta-Catenin Signaling Pathway And Epithelial-Mesenchymal Transition. OncoTargets Ther. 2019, 12, 9165–9175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  340. Zheng, Y.B.; Luo, H.P.; Shi, Q.; Hao, Z.N.; Ding, Y.; Wang, Q.S.; Li, S.B.; Xiao, G.C.; Tong, S.L. miR-132 inhibits colorectal cancer invasion and metastasis via directly targeting ZEB2. World J. Gastroenterol. 2014, 20, 6515–6522. [Google Scholar] [CrossRef] [PubMed]
  341. You, J.; Li, Y.; Fang, N.; Liu, B.; Zu, L.; Chang, R.; Li, X.; Zhou, Q. MiR-132 suppresses the migration and invasion of lung cancer cells via targeting the EMT regulator ZEB2. PLoS ONE 2014, 9, e91827. [Google Scholar] [CrossRef]
  342. Wu, S.-M.; Ai, H.-W.; Zhang, D.-Y.; Han, X.-Q.; Pan, Q.; Luo, F.-L.; Zhang, X.-L. MiR-141 targets ZEB2 to suppress HCC progression. Tumor Biol. 2014, 35, 9993–9997. [Google Scholar] [CrossRef]
  343. Li, W.; Wang, Q.; Su, Q.; Ma, D.; An, C.; Ma, L.; Liang, H. Honokiol suppresses renal cancer cells’ metastasis via dual-blocking epithelial-mesenchymal transition and cancer stem cell properties through modulating miR-141/ZEB2 signaling. Mol. Cells 2014, 37, 383–388. [Google Scholar] [CrossRef] [Green Version]
  344. Ren, D.; Wang, M.; Guo, W.; Huang, S.; Wang, Z.; Zhao, X.; Du, H.; Song, L.; Peng, X. Double-negative feedback loop between ZEB2 and miR-145 regulates epithelial-mesenchymal transition and stem cell properties in prostate cancer cells. Cell Tissue Res. 2014, 358, 763–778. [Google Scholar] [CrossRef]
  345. Jiang, S.B.; He, X.J.; Xia, Y.J.; Hu, W.J.; Luo, J.G.; Zhang, J.; Tao, H.Q. MicroRNA-145-5p inhibits gastric cancer invasiveness through targeting N-cadherin and ZEB2 to suppress epithelial-mesenchymal transition. OncoTargets Ther. 2016, 9, 2305–2315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  346. Zhou, J.; Xie, M.; Shi, Y.; Luo, B.; Gong, G.; Li, J.; Wang, J.; Zhao, W.; Zi, Y.; Wu, X.; et al. MicroRNA-153 functions as a tumor suppressor by targeting SET7 and ZEB2 in ovarian cancer cells. Oncol. Rep. 2015, 34, 111–120. [Google Scholar] [CrossRef] [PubMed]
  347. Lin, X.; Yang, Z.; Zhang, P.; Liu, Y.; Shao, G. miR-154 inhibits migration and invasion of human non-small cell lung cancer by targeting ZEB2. Oncol. Lett. 2016, 12, 301–306. [Google Scholar] [CrossRef] [Green Version]
  348. Pang, X.; Huang, K.; Zhang, Q.; Zhang, Y.; Niu, J. miR-154 targeting ZEB2 in hepatocellular carcinoma functions as a potential tumor suppressor. Oncol. Rep. 2015, 34, 3272–3279. [Google Scholar] [CrossRef]
  349. Fei, D.; Zhao, K.; Yuan, H.; Xing, J.; Zhao, D. MicroRNA-187 exerts tumor-suppressing functions in osteosarcoma by targeting ZEB2. Am. J. Cancer Res. 2016, 6, 2859–2868. [Google Scholar] [PubMed]
  350. Bendoraite, A.; Knouf, E.C.; Garg, K.S.; Parkin, R.K.; Kroh, E.M.; O’Briant, K.C.; Ventura, A.P.; Godwin, A.K.; Karlan, B.Y.; Drescher, C.W.; et al. Regulation of miR-200 family microRNAs and ZEB transcription factors in ovarian cancer: Evidence supporting a mesothelial-to-epithelial transition. Gynecol. Oncol. 2010, 116, 117–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  351. Perdigao-Henriques, R.; Petrocca, F.; Altschuler, G.; Thomas, M.; Le, M.; Tan, S.; Hide, W.; Lieberman, J. miR-200 promotes the mesenchymal to epithelial transition by suppressing multiple members of the Zeb2 and Snail1 transcriptional repressor complexes. Oncogene 2016, 35, 158–172. [Google Scholar] [CrossRef]
  352. Xia, H.; Ng, S.S.; Jiang, S.; Cheung, W.K.; Sze, J.; Bian, X.-W.; Kung, H.-f.; Lin, M.C. miR-200a-mediated downregulation of ZEB2 and CTNNB1 differentially inhibits nasopharyngeal carcinoma cell growth, migration and invasion. Biochem. Biophys. Res. Commun. 2010, 391, 535–541. [Google Scholar] [CrossRef]
  353. Yang, X.; Wang, J.; Qu, S.; Zhang, H.; Ruan, B.; Gao, Y.; Ma, B.; Wang, X.; Wu, N.; Li, X.; et al. MicroRNA-200a suppresses metastatic potential of side population cells in human hepatocellular carcinoma by decreasing ZEB2. Oncotarget 2015, 6, 7918–7929. [Google Scholar] [CrossRef]
  354. Wu, Q.; Guo, R.; Lin, M.; Zhou, B.; Wang, Y. MicroRNA-200a inhibits CD133/1+ ovarian cancer stem cells migration and invasion by targeting E-cadherin repressor ZEB2. Gynecol. Oncol. 2011, 122, 149–154. [Google Scholar] [CrossRef]
  355. Kurashige, J.; Kamohara, H.; Watanabe, M.; Hiyoshi, Y.; Iwatsuki, M.; Tanaka, Y.; Kinoshita, K.; Saito, S.; Baba, Y.; Baba, H. MicroRNA-200b regulates cell proliferation, invasion, and migration by directly targeting ZEB2 in gastric carcinoma. Ann. Surg. Oncol. 2012, 19 (Suppl. 3), S656–S664. [Google Scholar] [CrossRef]
  356. Li, J.; Yuan, J.; Yuan, X.; Zhao, J.; Zhang, Z.; Weng, L.; Liu, J. MicroRNA-200b inhibits the growth and metastasis of glioma cells via targeting ZEB2. Int. J. Oncol. 2016, 48, 541–550. [Google Scholar] [CrossRef] [PubMed]
  357. Lu, Y.-m.; Shang, C.; Ou, Y.-l.; Yin, D.; Li, Y.-N.; Li, X.; Wang, N.; Zhang, S.-l. miR-200c modulates ovarian cancer cell metastasis potential by targeting zinc finger E-box-binding homeobox 2 (ZEB2) expression. Med. Oncol. 2014, 31, 134. [Google Scholar] [CrossRef]
  358. Jiao, A.; Sui, M.; Zhang, L.; Sun, P.; Geng, D.; Zhang, W.; Wang, X.; Li, J. MicroRNA-200c inhibits the metastasis of non-small cell lung cancer cells by targeting ZEB2, an epithelial-mesenchymal transition regulator. Mol. Med. Rep. 2016, 13, 3349–3355. [Google Scholar] [CrossRef]
  359. Zhang, J.; Zhang, H.; Qin, Y.; Chen, C.; Yang, J.; Song, N.; Gu, M. MicroRNA-200c-3p/ZEB2 loop plays a crucial role in the tumor progression of prostate carcinoma. Ann. Transl. Med. 2019, 7, 141. [Google Scholar] [CrossRef] [PubMed]
  360. Duan, X.; Fu, Z.; Gao, L.; Zhou, J.; Deng, X.; Luo, X.; Fang, W.; Luo, R. Direct interaction between miR-203 and ZEB2 suppresses epithelial–mesenchymal transition signaling and reduces lung adenocarcinoma chemoresistance. Acta Biochim. Biophys. Sin. 2016, 48, 1042–1049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  361. Jiang, Q.; Zhou, Y.; Yang, H.; Li, L.; Deng, X.; Cheng, C.; Xie, Y.; Luo, X.; Fang, W.; Liu, Z. A directly negative interaction of miR-203 and ZEB2 modulates tumor stemness and chemotherapy resistance in nasopharyngeal carcinoma. Oncotarget 2016, 7, 67288. [Google Scholar] [CrossRef] [PubMed]
  362. Chen, Z.; Tang, Z.Y.; He, Y.; Liu, L.F.; Li, D.J.; Chen, X. miRNA-205 is a candidate tumor suppressor that targets ZEB2 in renal cell carcinoma. Oncology Res. Treat. 2014, 37, 658–664. [Google Scholar] [CrossRef]
  363. Chen, X.F.; Guo, J.F.; Xu, J.F.; Yin, S.H.; Cao, W.L. MiRNA-206 inhibits proliferation of renal clear cell carcinoma by targeting ZEB2. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 7826–7834. [Google Scholar] [CrossRef]
  364. Hou, Y.; Zhen, J.; Xu, X.; Zhen, K.; Zhu, B.; Pan, R.; Zhao, C. miR-215 functions as a tumor suppressor and directly targets ZEB2 in human non-small cell lung cancer. Oncol. Lett. 2015, 10, 1985–1992. [Google Scholar] [CrossRef] [Green Version]
  365. Sun, Z.; Zhang, Z.; Liu, Z.; Qiu, B.; Liu, K.; Dong, G. MicroRNA-335 inhibits invasion and metastasis of colorectal cancer by targeting ZEB2. Med. Oncol. (Northwood Lond. Engl.) 2014, 31, 982. [Google Scholar] [CrossRef] [PubMed]
  366. Kan, Q.; Su, Y.; Yang, H. MicroRNA-335 is downregulated in papillary thyroid cancer and suppresses cancer cell growth, migration and invasion by directly targeting ZEB2. Oncol. Lett. 2017, 14, 7622–7628. [Google Scholar] [CrossRef] [PubMed]
  367. Huang, N.; Wu, Z.; Lin, L.; Zhou, M.; Wang, L.; Ma, H.; Xia, J.; Bin, J.; Liao, Y.; Liao, W. MiR-338-3p inhibits epithelial-mesenchymal transition in gastric cancer cells by targeting ZEB2 and MACC1/Met/Akt signaling. Oncotarget 2015, 6, 15222. [Google Scholar] [CrossRef] [Green Version]
  368. Cui, J.; Pan, G.; He, Q.; Yin, L.; Guo, R.; Bi, H. MicroRNA-545 targets ZEB2 to inhibit the development of non-small cell lung cancer by inactivating Wnt/beta-catenin pathway. Oncol. Lett. 2019, 18, 2931–2938. [Google Scholar] [CrossRef] [Green Version]
  369. Tong, X.; Su, P.; Yang, H.; Chi, F.; Shen, L.; Feng, X.; Jiang, H.; Zhang, X.; Wang, Z. MicroRNA-598 inhibits the proliferation and invasion of non-small cell lung cancer cells by directly targeting ZEB2. Exp. Ther. Med. 2018, 16, 5417–5423. [Google Scholar] [CrossRef]
  370. Song, Q.; Pang, H.; Qi, L.; Liang, C.; Wang, T.; Wang, W.; Li, R. Low microRNA-622 expression predicts poor prognosis and is associated with ZEB2 in glioma. OncoTargets Ther. 2019, 12, 7387–7397. [Google Scholar] [CrossRef] [Green Version]
  371. Wang, K.; Yang, S.; Gao, Y.; Zhang, C.; Sui, Q. MicroRNA-769-3p inhibits tumor progression in glioma by suppressing ZEB2 and inhibiting the Wnt/beta-catenin signaling pathway. Oncol. Lett. 2020, 19, 992–1000. [Google Scholar] [CrossRef] [Green Version]
  372. Xu, R.; Zhou, F.; Yu, T.; Xu, G.; Zhang, J.; Wang, Y.; Zhao, L.; Liu, N. MicroRNA-940 inhibits epithelial-mesenchymal transition of glioma cells via targeting ZEB2. Am. J. Transl. Res. 2019, 11, 7351–7363. [Google Scholar] [PubMed]
  373. Gao, H.B.; Gao, F.Z.; Chen, X.F. MiRNA-1179 suppresses the metastasis of hepatocellular carcinoma by interacting with ZEB2. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 5149–5157. [Google Scholar] [CrossRef]
  374. Liu, Q.; Liu, H.; Cheng, H.; Li, Y.; Li, X.; Zhu, C. Downregulation of long noncoding RNA TUG1 inhibits proliferation and induces apoptosis through the TUG1/miR-142/ZEB2 axis in bladder cancer cells. OncoTargets Ther. 2017, 10, 2461–2471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  375. Li, C.; Lu, L.; Feng, B.; Zhang, K.; Han, S.; Hou, D.; Chen, L.; Chu, X.; Wang, R. The lincRNA-ROR/miR-145 axis promotes invasion and metastasis in hepatocellular carcinoma via induction of epithelial-mesenchymal transition by targeting ZEB2. Sci. Rep. 2017, 7, 4637. [Google Scholar] [CrossRef]
  376. Xiao, H.; Tang, K.; Liu, P.; Chen, K.; Hu, J.; Zeng, J.; Xiao, W.; Yu, G.; Yao, W.; Zhou, H. LncRNA MALAT1 functions as a competing endogenous RNA to regulate ZEB2 expression by sponging miR-200s in clear cell kidney carcinoma. Oncotarget 2015, 6, 38005. [Google Scholar] [CrossRef] [PubMed]
  377. Wang, Y.; Zhou, Y.; Yang, Z.; Chen, B.; Huang, W.; Liu, Y.; Zhang, Y. MiR-204/ZEB2 axis functions as key mediator for MALAT1-induced epithelial-mesenchymal transition in breast cancer. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2017, 39, 1010428317690998. [Google Scholar] [CrossRef]
  378. Chen, D.L.; Lu, Y.X.; Zhang, J.X.; Wei, X.L.; Wang, F.; Zeng, Z.L.; Pan, Z.Z.; Yuan, Y.F.; Wang, F.H.; Pelicano, H.; et al. Long non-coding RNA UICLM promotes colorectal cancer liver metastasis by acting as a ceRNA for microRNA-215 to regulate ZEB2 expression. Theranostics 2017, 7, 4836–4849. [Google Scholar] [CrossRef] [PubMed]
  379. Li, C.; Wan, L.; Liu, Z.; Xu, G.; Wang, S.; Su, Z.; Zhang, Y.; Zhang, C.; Liu, X.; Lei, Z. Long non-coding RNA XIST promotes TGF-β-induced epithelial-mesenchymal transition by regulating miR-367/141-ZEB2 axis in non-small-cell lung cancer. Cancer Lett. 2018, 418, 185–195. [Google Scholar] [CrossRef] [PubMed]
  380. Li, Y.; He, Q.; Wen, X.; Hong, X.; Yang, X.; Tang, X.; Zhang, P.; Lei, Y.; Sun, Y.; Zhang, J.; et al. EZH2-DNMT1-mediated epigenetic silencing of miR-142-3p promotes metastasis through targeting ZEB2 in nasopharyngeal carcinoma. Cell Death Differ. 2019, 26, 1089–1106. [Google Scholar] [CrossRef]
  381. Li, X.; Tian, Y.; Hu, Y.; Yang, Z.; Zhang, L.; Luo, J. CircNUP214 sponges miR-145 to promote the expression of ZEB2 in thyroid cancer cells. Biochem. Biophys. Res. Commun. 2018, 507, 168–172. [Google Scholar] [CrossRef]
  382. Brown, C.Y.; Dayan, S.; Wong, S.W.; Kaczmarek, A.; Hope, C.M.; Pederson, S.M.; Arnet, V.; Goodall, G.J.; Russell, D.; Sadlon, T.J. FOXP3 and miR-155 cooperate to control the invasive potential of human breast cancer cells by down regulating ZEB2 independently of ZEB1. Oncotarget 2018, 9, 27708. [Google Scholar] [CrossRef]
  383. Li, H.; Xu, L.; Li, C.; Zhao, L.; Ma, Y.; Zheng, H.; Li, Z.; Zhang, Y.; Wang, R.; Liu, Y.; et al. Ubiquitin ligase Cbl-b represses IGF-I-induced epithelial mesenchymal transition via ZEB2 and microRNA-200c regulation in gastric cancer cells. Mol. Cancer 2014, 13, 136. [Google Scholar] [CrossRef] [Green Version]
  384. Truong, H.H.; Xiong, J.; Ghotra, V.P.; Nirmala, E.; Haazen, L.; Le Devedec, S.E.; Balcioglu, H.E.; He, S.; Snaar-Jagalska, B.E.; Vreugdenhil, E.; et al. beta1 integrin inhibition elicits a prometastatic switch through the TGFbeta-miR-200-ZEB network in E-cadherin-positive triple-negative breast cancer. Sci. Signal. 2014, 7, ra15. [Google Scholar] [CrossRef]
Figure 1. The oncogenic upstream mediators of miRs activating ZEB proteins, which enhances metastasis and migration of cancer cells.
Figure 1. The oncogenic upstream mediators of miRs activating ZEB proteins, which enhances metastasis and migration of cancer cells.
Biomolecules 10 01040 g001
Figure 2. Metformin, lncRNAs, circRNAs, and other molecular pathways are able to function as upstream mediators of miRs in targeting ZEB proteins, and promote cancer progression.
Figure 2. Metformin, lncRNAs, circRNAs, and other molecular pathways are able to function as upstream mediators of miRs in targeting ZEB proteins, and promote cancer progression.
Biomolecules 10 01040 g002
Figure 3. The miRs as key player in regulation of tumor malignancy via targeting ZEB proteins.
Figure 3. The miRs as key player in regulation of tumor malignancy via targeting ZEB proteins.
Biomolecules 10 01040 g003
Figure 4. How tumor microenvironment components are affected by the relationship between miRs and ZEB proteins, and their regulation by lncRNAs.
Figure 4. How tumor microenvironment components are affected by the relationship between miRs and ZEB proteins, and their regulation by lncRNAs.
Biomolecules 10 01040 g004
Table 1. ZEB1 regulation by miRs in different cancers.
Table 1. ZEB1 regulation by miRs in different cancers.
MiRDown-Stream TargetCancer TypeMajor OutcomesRefs
MiR-23aZEB1Intraocular tumorA negative feedback loop between miR-23a and ZEB1 regulates EMT and overexpression of miR-23a inhibits EMT by ZEB1 down-regulation[248]
MiR-23bZEB1Bladder cancerMiR-23b induces apoptosis and cell cycle arrest, and decreases the invasion and EMT through ZEB1 inhibition[249]
MiR-33bZEB1MelanomaCordycepin enhances the expression of miR-33b to inhibit ZEB1 and induces mesenchymal-epithelial transition in cancer cells, resulting in decreased invasion and migration of cancer cells[250]
MiR-126ZEB1OsteosarcomaInhibition of EMT, migration, and metastasis of cancer cells through ZEB1 down-regulation[251]
MiR-128ZEB1Prostate cancerMiR-128 sensitizes cancer cells into cisplatin chemotherapy by ZEB1 down-regulation and decreasing the malignancy and invasion of cancer cells[252]
MiR-130bZEB1Endometrial cancerThe miR-130b down-regulates the expression of ZEB1 to inhibit the malignancy and invasion of cancer cells[253]
MiR-139-5pZEB1/2Hepatocellular carcinomaReduced invasion, migration, metastasis, and EMT by ZEB1/2 down-regulation through miR-139-5p[254]
Glioblastoma multiformeSuppressing the invasion and migration of cancer cells through ZEB1/2 inhibition[255]
MiR-141 and miR-146b-5pAUF1/ZEB1OsteosarcomaThese miRs are able to down-regulate the expression of AUF1 to repress ZEB1, resulting in an increase in epithelial markers (E-cadherin and Epcam) and a decrease in mesenchymal markers (N-cadherin and Vimentin)[256]
MiR-144
MiR-144
ZEB1/2Breast cancerMiR-144 is an onco-suppressor that inhibits EMT and migration invasion through ZEB1/2 down-regulation[257]
Thyroid cancerMiR-144 down-regulates the expression of ZEB1/2 to prevent cancer progression and proliferation[258]
MiR-150ZEB1Esophageal squamous cell carcinomaMiR-150 degrades ZEB1 to induce mesenchymal-epithelial transition (MET), resulting in a decrease in tumor depth, lymph node metastasis, and lymphatic invasion [259]
Ovarian cancerSuppressing the malignancy and invasion of cancer cells through ZEB1 inhibition[260]
MiR-199a-3pZEB1MelanomaThe administration of gambogic acid is associated with up-regulation of miR-199a-3p and subsequent inhibition of ZEB1 to suppress cancer progression both in vitro and in vivo[261]
MiR-199bZEB1Non-small cell lung cancerSuppressing the proliferation, migration, and invasion of cancer cells through ZEB1 down-regulation[262]
MiR-200ZEB1-FAK/SrcHuman lung cancerThe miR-200 up-regulation decreases the invasion and malignancy of cancer cells through enhancing ZEB1 expression and subsequent activation of FAK/Src[263]
ZEB1Endometrial carcinomaThe expression of miR-200 undergoes down-regulation in endometrial carcinoma cells to induce ZEB1 and subsequently, EMT mechanism to elevate the invasion and malignancy of cancer cells[264]
LymphomaGeneration of a less aggressive behavior by ZEB1 inhibition through miR-200[265]
Insulinoma mouse modelOverexpression of miR-200 is associated with ZEB1 inhibition and decreased migration and proliferation of cancer cells[266]
MiR-200bZEB1OsteosarcomaOverexpression of miR-200b is associated with down-regulation of ZEB1 and decreased invasion and malignancy of cancer cells[267]
Human hepatocellular carcinomaBy down-regulation of ZEB1, miR-200b reduces the stemness of cancer cells[268]
MiR-200b and miR-141ZEB1Non-small cell lung cancerThe overexpression of miR-200b and miR-141 is related to the inhibition of ZEB1 and sensitizing cancer cells into nintedanib[269]
MiR-200cZEB1Human colon cancerSuppressing the invasion and migration of cancer cells through ZEB1 down-regulation[270]
Gastric carcinomaOverexpression of miR-200c is related to the ZEB1 down-regulation and enhanced levels of E-cadherin protein[271]
Human bladder cancerAdministration of sulforaphane is associated with miR-200c induction and subsequently, inhibition of ZEB1 and malignancy of cancer cells[272]
Non-small cell lung carcinomaThe cyclamen pseudibericum extract up-regulates miR-200c to induce ZEB1 down-regulation, resulting in suppressing cancer progression and proliferation[273]
Non-small cell lung cancerMiR-200c sensitizes cancer cells to the gefitinib-mediated apoptosis by down-regulation of ZEB1 [274]
Lung cancerMiR-200c sensitizes lung cancer cells into crizotinib chemotherapy by inhibition of ZEB1, and subsequently, EMT inhibition[275]
MiR-200c and miR-141ZEB1Glioma cellThe miR-200c and -141 synergistically inhibit ZEB1 to prevent the malignancy and invasion of cancer cells[276]
ZEB1/2Gastric cancerMiR-200c/141 significantly decreases ZEB1/2 expression to suppress cancer malignancy[277]
MiR-203ZEB1Non-small cell lung cancerThe administration of silymarin enhances the expression of miR-203 to inhibit ZEB1 and elevate the levels of E-cadherin, resulting in suppressing cancer[278]
MiR-204ZEB1Prostate cancerMiR-204 up-regulation sensitizes cancer cells into docetaxel-mediated apoptosis through ZEB1 down-regulation[279]
MiR-205ZEB1Ovarian cancerMiR-205 enhances the invasion and migration of cancer cells via ZEB1 up-regulation. Reducing the expression of miR-205 is of interest in suppressing the malignancy of cancer cells[280]
Prostate cancerBy inhibition of ZEB1, miR-205 sensitizes cancer cells into radiotherapy and induces DNA damage[281]
Breast cancerMiR-205 sensitizes cancer cells into radiotherapy and prevents DNA repair by ZEB1 down-regulation[282]
MiR-205-5pZEB1Prostatic carcinomaSuppressing the migration and invasion of cancer cells by ZEB1 down-regulation[283]
MiR-340ZEB1/TGF-βBreast cancerMiR-340 inhibits ZEB1 to suppress TGF-β-mediated cancer progression[284]
ZEB1OsteosarcomaMiR-340 down-regulates the expression of ZEB1 to sensitize cancer cells into cisplatin-mediated apoptotic cell death[285]
MiR-409-3pZEB1Breast cancerMiR-409-3p binds to the 3′-UTR of ZEB1 to inhibit the progression and metastasis of cancer cells[286]
MiR-429ZEB1Ovarian cancerDown-regulation of miR-429 is related to the resistance of cancer cells into cisplatin chemotherapy. Up-regulation of miR-429 suppresses ZEB1 to sensitize cancer cells into apoptosis[287]
Oral squamous cell carcinomaMiR-429 suppresses the viability and progression of cancer cells via ZEB1 down-regulation[288]
Human thyroid cancerMiR-429 binds to the 3′-UTR to inhibit ZEB1, resulting in suppressing invasion of cancer cells[110]
MiR-431ZEB1Hepatocellular carcinomaMiR-431 suppresses the migration and invasion capabilities of cancer cells through inhibition of ZEB1-mediated EMT[289]
MiR-448ZEB1/2Breast cancerThe miR-448 significantly reduces the expressions of ZEB1/2 to inhibit the malignancy and invasion of cancer cells via EMT down-regulation[290]
MiR-455ZEB1Non-small cell lung cancerThe miR-455 reduces the expression of ZEB1 to inhibit the malignancy of cancer cells[291]
MiR-484Smad2/ZEB1Cervical cancerOverexpression of miR-484 inhibits Smad2/ZEB1 to suppress cancer malignancy and miR-484 expression can be considered as a biomarker[292]
MiR-508ZEB1Renal cell carcinomaUp-regulation of miR-508 significantly reduces the expression of ZEB1 to inhibit EMT, leading to a decrease in cancer migration and metastasis[293]
MiR-508-3pZEB1Triple negative breast cancerSuppressing the invasion and EMT of cancer cells by down-regulation of ZEB1[294]
MiR-574-3pZEB1Human gastric carcinomaMiR-574-3p reduces the expression of ZEB1 by binding into 3′-UTR to decrease the malignancy of cancer cells, and simultaneously, sensitize cancer cells into cisplatin therapy [295]
MiR-590-3pZEB1/2Glioblastoma multiformeDecreased invasion and migration of cancer cells by ZEB1/2 down-regulation[296]
MiR-641ZEB1Cervical cancerNegatively affecting the proliferation, migration, and invasion of cancer cells through ZEB1 down-regulation[297]
MiR-652ZEB1Pancreatic cancerAcidic microenvironment of tumor cells induces EMT through ZEB1 up-regulation. Enhancing the expression of miR-652 inhibits acidic-mediated EMT and ZEB1 induction[298]
MiR-655TGF-β/ZEB1Pancreatic cancerMiR-655 inhibits TGF-β/ZEB1 axis to suppress EMT in cancer cells[299]
MiR-675-5pUBQLN1/ZEB1/miR200Pancreatic cancerThe miR-675-5p reduces the malignancy of cancer cells and ZEB1 protein by up-regulation of UBQLN1 and down-regulation of miR-200[300]
MiR-873-5pZEB1Colorectal cancerThe inhibitory effect of miR-873-5p on the migration, EMT formation, and invasion of cancer cells is mediated through ZEB1 down-regulation[301]
MiR-875-5pEGFR/ZEB1Prostate cancerBy suppressing EGFR/ZEB1 axis, miR-875-5p inhibits EMT mechanism and sensitizes cancer cells to radiotherapy[302]
MiR-1271ZEB1Pancreatic cancerSuppressing the invasion, progression, and EMT in cancer cells by ZEB1 down-regulation[303]
MiR-1236-3pZEB1High-grade serous ovarian carcinomaThere is a negative relationship between miR-1236-3p and ZEB1 to suppress the migration and invasion of cancer cells[304]
MiR-1236-3pZEB1Breast cancerZEB1 inhibition by miR-1236-3p contributes to the inhibitory effect of this miR on the migration and invasion of cancer cells[305]
MiR-3662ZEB1MelanomaAmelioration of invasiveness and malignancy of cancer cells by ZEB1 down-regulation[306]
Table 2. miR/ZEB1 regulation by lncRNAs in different cancers.
Table 2. miR/ZEB1 regulation by lncRNAs in different cancers.
LncRNAMiRDown-Stream TargetCancer TypeMajor OutcomesRefs
LncRNA DANCRMiR-33a-5pZEB1Esophageal squamous cell carcinomaMiR-33a-5p suppresses cancer malignancy via reducing ZEB1 expression. LncRNA DANCR sponges miR-33a-5p to enhances the invasion via ZEB1 induction[307]
LncRNA SNHG6MiR-101-3pZEB1Hepatocellular carcinomaLncRNA SNHG6 down-regulates the expression of miR-101-3p to induce ZEB1 and enhance the malignancy of cancer cells[308]
LncRNA PTARMiR-101-3pZEB1Serous ovarian cancerLncRNA PTAR decreases the expression of miR-101-3p to induce ZEB1 and EMT mechanism, leading to the invasion and metastasis of cancer cells[309]
LncRNA NNT-AS1MiR-142-3pZEB1Breast cancerEnhancing the progression of cancer cells by sponging miR-142-3p and induction of ZEB1[310]
LncRNA TUG1MiR-142-3pZEB1Hepatocellular carcinomaBy down-regulation of miR-142-3p, lncRNA TUG1 enhances the expression of ZEB1 to ensure the proliferation and malignancy of cancer cells[311]
LncRNA SNHG16MiR-140-5pZEB1Esophageal squamous cell carcinoma The lncRNA SNHG16 functions as an oncogenic factor and neutralizes the inhibitory effect of miR-140-5p on ZEB1 to induce EMT and enhance the migration and invasion of cancer cells[312]
MiR-205ZEB1OsteosarcomaSNHG16 reduces the expression of miR-205 to elevate the expression of ZEB1, resulting in an increase in the viability, proliferation, and migration of cancer cells[313]
LncRNA HOTAIRMiR-217ZEB1OsteosarcomaBy reducing the expression of miR-217, lncRNA HOTAIR enhances the expression of ZEB1 and improves their malignancy[314]
MiR-23b-3pZEB1Hepatocellular carcinomaThe miR-23b-3p inhibits ZEB1 and lncRNA HOTAIR prevents the inhibitory effect of miR-23b-3p on ZEB1 to induce EMT[315]
lncRNA UCA1Has-miR-145ZEB1/2-FSCN1Bladder cancerThere is a reverse relationship between lncRNA UCA1 and has-miR-145. Decreased expression of has-miR-145 enhances the expression of ZEB1/2 and FSCN1 to elevate the migration and invasion of cancer cells[316]
MiR-204-5pZEB1Glioma cellsBy sponging miR-204-5p, lncRNA UCA1 stimulates ZEB1 and activates EMT mechanism[317]
LncRNA ZEB1-AS1MiR-200c/141ZEB1Glioma cancerLncRNA ZEB1-AS1 down-regulates the expression of miR-200c/141 to induce ZEB1 and enhance the malignancy and invasion of cancer cells[318]
MiR-409-3pZEB1Non-small cell lung cancerA feedback loop is involved, so that lncRNA ZEB1-AS1 induces ZEB1 through miR-409-3p down-regulation, leading to the metastasis and survival of cancer cells[319]
MiR-101ZEB1Colorectal cancerElevating the proliferation and migration of cancer cells via down-regulation of MiR-101 and up-regulation of ZEB1 by lncRNA ZEB1-AS1[320]
LncRNA MIATMiR-150-5pZEB1OsteosarcomaThe miR-150-5p is down-regulated by MIAT to induce ZEB1 and enhance the malignancy of cancer cells[321]
LncRNA MAGI1-IT1MiR-200aZEB1/2Ovarian cancerVia competitively binding into miR-200a, lncRNA MAGI1-IT1 enhances the expression of ZEB1/2 to ensure the invasion and metastasis of cancer cells[293]
LncRNA HULCMiR-200a-3pZEB1Hepatocellular carcinomaBy sequestering miR-200a-3p, lncRNA HULC stimulates ZEB1 to enhance the malignancy and progression of tumor cells[322]
LncRNA NEAT1MiR-204ZEB1Nasopharyngeal carcinomaMiR-204 inhibits EMT through ZEB1 down-regulation, and lncRNA NEAT1 reverse this axis to enhance the proliferation and viability of cancer cells[323]
LncRNA MINCRMiR-223ZEB1-Akt/PI3KNasopharyngeal carcinomaMINCR induces ZEB1 by sponging miR-223, resulting in activation of Akt/PI3K and resistance of cancer cells into radiotherapy[324]
LncRNA CAT104MiR-381ZEB1Gastric carcinomaLncRNA CAT104 down-regulates the expression of miR-381 to enhances ZEB1 levels, resulting in enhanced invasion of cancer cells. Additionally, there is a negative feedback loop between ZEB1 and miR-381.[325]
LncRNA ZNF469-3MiR-574-5pZEB1Triple negative breast cancerThe reverse relationship between ZNF469-3 and miR-574-5p paves the road for up-regulation of ZEB1 and subsequent activation of EMT, leading to the cancer progression and malignancy[326]
Table 3. miR/ZEB1 regulation by various molecular pathways in different cancers.
Table 3. miR/ZEB1 regulation by various molecular pathways in different cancers.
Upstream MediatorMiRDown-Stream TargetCancer TypeMajor OutcomesRefs
ELF3MiR-141-3pZEB1Hepatocellular carcinomaOverexpression of miR-141-3p down-regulates ZEB1. The ELF3 reduces the expression of miR-141-3p to induce ZEB1 and EMT mechanism[327]
SPROUTY-2MiR-200/miR-150ZEB1Colon cancerBy reducing the expression of miR-200/miR-150, SPROUTY-2 induces ZEB1 to facilitate the mesenchymal phenotype acquisition of cancer cells[286]
STAT3MiR-200ZEB1Invasive breast carcinomaZEB1 stimulation by miR-200 down-regulation via STAT3-dependent manner enhances the EMT acquisition in cancer cells[328]
TGF-β1MiR-200ZEB1/2Non-small cell lung cancerThe administration of decitabine induces miR-200 expression through TGF-β1 inhibition to down-regulate ZEB1/2, leading to the suppressing EMT and migration of cancer cells[329]
GRHL2MiR-200b/aZEB1Ovarian cancerGRHL2 down-regulates the expression of ZEB1 by miR-200a/b overexpression to preserve the epithelial phenotype[330]
53BP1MiR-200b and miR-429ZEB1Breast cancerThe 53BP1 enhances the expression of miR-200b and miR-429 to elevate E-cadherin levels and suppress EMT mechanism through ZEB1 down-regulation[331]
Mel-18MiR-205ZEB1/2Breast cancerMel-18 enhances the expression of miR-205 to inhibit ZEB1/2, resulting in decreased progression and invasion of cancer cells[332]
ΔNp63αMiR-205ZEB1Cervical squamous cell carcinomaΔNp63α alleviates cancer progression and malignancy by enhancing the expression of miR-205, subsequently down-regulating of ZEB1, and consequently, inhibition of EMT, and enhancing E-cadherin levels[333]
KCNQ1OT1MiR-217ZEB1Colorectal cancerKCNQ1OT1 inhibits miR-217 to stimulate ZEB1 and EMT mechanism in cancer cells. There is a feedback loop, so that ZEB1 also enhances the expression of KCNQ1OT1 to elevate its inhibitory effect on miR-217[334]
Circ008913MiR-889DAB2IP/ZEB1Skin carcinogenesisArsenite down-regulates the expression of circ008913 to up-regulate miR-889. Then, a decrease occurs in DAB2IP to induce ZEB1 and carcinogenesis[335]
Pituitary tumor-transforming gene 1MiR-3666ZEB1Cervical cancerThe expression of miR-3666 reduces to neutralize its inhibitory impact of ZEB1, and consequently, elevate the metastasis and progression of cancer cells [289]
Table 4. ZEB2 regulation by miRs in different cancers.
Table 4. ZEB2 regulation by miRs in different cancers.
MiRDown-Stream TargetCancer TypeMajor OutcomesRefs
MiR-29bTET1/ZEB2Breast cancerThe miR-29b is an oncogene miR that inhibits TET1 to induce ZEB2 expression, leading to the EMT and colony formation of cancer cells[336]
MiR-30a-5pZEB2Renal cancerThe miR-30a-5p reduces the expression of ZEB2 to be related with desirable prognosis of cancer cells[337]
MiR-101ZEB2OsteosarcomaSuppressing the invasion and proliferation of cancer cells through ZEB2 down-regulation[175]
MiR-124ZEB2Triple negative breast cancerMiR-124 diminishes the expression of ZEB2 to inhibit the EMT and invasion of cancer cells[338]
MiR-129Wnt-β-catenin/ZEB2Non-small cell lung cancerThe miR-129 disrupts Wnt/ZEB2 axis to inhibit EMT[339]
MiR-132ZEB2Colorectal cancerReducing the invasion and metastasis of cancer cells through ZEB2 down-regulation[340]
Lung cancerDiminishing the migration and invasion of cancer cells through ZEB2 inhibition[341]
MiR-138ZEB2Bladder cancerThe miR-138 binds to the 3′-UTR of ZEB2 to inhibit the metastasis and invasion of cancer cells[289]
MiR-141ZEB2Hepatocellular carcinomaThe miR-141 decreases the expression of ZEB2 to induce apoptosis and diminish viability and proliferation of cancer cells[342]
Renal cancerThe administration of honokiol is associated with miR-141 induction and subsequent downregulation of ZEB2 to inhibit the malignancy of cancer cells[343]
MiR-145ZEB2Non-small cell lung cancerMiR-145 acts as an onco-suppressor miR that negatively affects the expression of ZEB2 to inhibit the progression and malignancy of cancer cells[166]
Prostate cancerThere is a negative feedback loop between miR-145 and ZEB2, so that overexpression of miR-145 down-regulates the expression of ZEB2 to ensure the reduced viability and proliferation of cancer cells[344]
MiR-145-5pZEB2Gastric cancerThe miR-145-5p decreases the levels of N-cadherin by ZEB2 down-regulation[345]
MiR-153ZEB2Ovarian cancerActing as an onco-suppressor miR and reduces ZEB2 expression to EMT inhibition[346]
MiR-154ZEB2Non-small cell lung cancerThe miR-154 exerts an anti-tumor impact by ZEB2 down-regulation[347]
Hepatocellular carcinomaThe miR-154 functions as an onco-suppressor miR by inhibition ZEB2 expression and reducing cancer malignancy and proliferation[348]
MiR-155 and FOXP3ZEB2Colorectal cancerThe miR-155 and FOXP3 inhibit ZEB2 expression to suppress EMT via E-cadherin level up-regulation and Vimentin level downregulation[307]
MiR-187ZEB2OsteosarcomaThe miR-187 decreases the expression of ZEB2 to inhibit the malignancy and migration of tumor cells[349]
MiR-200ZEB1/2Ovarian cancerThe cancer cells acquire an epithelial phenotype by enhancing the expression of miR-200 and subsequent inhibition of ZEB1 and ZEB2 proteins[350]
ZEB2Breast cancerAs an onco-suppressor miR, miR-200 decreases the expression of ZEB2 and its targets gene Snail1 to induce mesenchymal to epithelial transition[351]
MiR-200aZEB2Nasopharyngeal carcinomaSuppressing the growth and invasion of cancer cells through ZEB2 down-regulation[352]
Hepatocellular carcinomaThe miR-200a diminishes the expression of ZEB2 to suppress EMT and invasion of cancer cells[353]
Ovarian cancerThe miR-200a increases the levels of E-cadherin by EMT inhibition and ZEB2 down-regulation [354]
MiR-200bZEB2Gastric carcinomaInhibition of ZEB2 by miR-200b suppresses invasion, metastasis, and migration of cancer cells[355]
GliomaReducing the growth and metastasis of ZEB2 inhibition[356]
MiR-200cZEB2Ovarian cancerMiR-200c reduces the expression of ZEB2 to inhibit EMT by enhancing E-cadherin levels and reducing Vimentin levels[357]
Non-small cell lung cancerThe miR-200c inhibits EMT mechanism by ZEB2 down-regulation [358]
MiR-200c-3pZEB2Prostate carcinomaThe miR-200c-3p functions as an anti-tumor miR that inhibits the progression and invasion of cancer cells through ZEB2 down-regulation[359]
MiR-203ZEB2Lung adenocarcinoma and nasopharyngeal carcinomaMiR-203 enhances the efficacy of cisplatin in chemotherapy and eradication of cancer cells, and also inhibits their invasion by EMT down-regulation through ZEB2 inhibition[360,361]
MiR-205ZEB2Renal cell carcinomaThe miR-205 is related to the favorable prognosis and reduced invasion of cancer cells through ZEB2 down-regulation[362]
MiR-206ZEB2Renal cancerDecreasing the proliferation of tumor cells through ZEB2 down-regulation[363]
MiR-211-5pZEB2Hepatocellular carcinomaThe miR-211-5p suppresses the metastasis of cancer cells via ZEB2 down-regulation[215]
MiR-215ZEB2Non-small cell lung cancerThe in vitro and in vivo experiments demonstrate the potential of miR-215 in down-regulation of ZEB2 and suppressing the invasion, progression, and malignancy of cancer cells, and induction of apoptotic cell death[364]
MiR-335ZEB2Colorectal cancerThe inhibition of metastasis and invasion of cancer cells through ZEB2 down-regulation[365]
Papillary thyroid cancerThrough reducing the expression of ZEB2, miR-335 suppresses the growth and metastasis of cancer cells[366]
MiR-338-3pZEB2Gastric cancerMiR-338-3p diminishes the expression of ZEB2 to inhibit EMT in cancer cells[367]
MiR-454-3p and miR-374b-5pZEB2Bladder cancerReducing the expression of ZEB2 significantly decreases the migration and invasion of cancer cells[325]
MiR-506ZEB2Gastric carcinomaThe miR-506 suppresses metastasis through ZEB2 down-regulation[130]
MiR-545Wnt-β-catenin/ZEB2Non-small cell lung cancerThe miR-545 reduces the expression of Wnt/β−catenin to down-regulate the expression of ZEB2, leading to the decreased migration and invasion of cancer cells[368]
MiR-598ZEB2Non-small cell lung cancerThe in vitro experiment demonstrated that miR-598 decreases the expression of ZEB2 to inhibit the migration and metastasis of cancer cells[369]
MiR-622ZEB2GliomaThe increased expression of miR-622 is related to the desirable prognosis via ZEB2 down-regulation[370]
MiR-769-3pWnt-β-catenin/ZEB2GliomaThe miR-769-3p down-regulates the expression of Wnt and inhibits nuclear translocation of β−catenin to suppress ZEB2, leading to the decreased viability, proliferation and invasion of cancer cells[371]
MiR-940ZEB2GliomaInhibition of cancer progression and EMT through ZEB2 down-regulation[372]
MiR-1179ZEB2Hepatocellular carcinomaThe miR-1179 reduces the expression of ZEB2 to inhibit cancer progression and malignancy[373]
MiR-3653ZEB2Colon cancerSuppressing metastasis and EMT by inhibition of ZEB2[171]
Table 5. MiR/ZEB2 regulation by lncRNAs in different cancers.
Table 5. MiR/ZEB2 regulation by lncRNAs in different cancers.
LncRNAMiRDown-Stream TargetCancer TypeMajor OutcomesRefs
LncRNA TUG1MiR-142ZEB2Bladder cancerThe lncRNA TUG1 stimulates ZEB2 through miR-142 down-regulation to inhibit apoptosis and enhance the proliferation of cancer cells[374]
LncRNA RORMiR-145ZEB2Hepatocellular carcinomaThe lncRNA ROR elevates the expression of ZEB2 through miR-145 sponging to inhibit the EMT and malignancy of cancer cells[375]
LncRNA MALAT1MiR-200sZEB2Kidney carcinomaThe lncRNA MALAT1 induces ZEB2 via miR-200s sponging, predisposing cancer cells into growth and proliferation[376]
MiR-204ZEB2Breast cancerThe negative relationship between MALAT1 and miR-204 results in ZEB2 induction to enhance the migration and invasion of cancer cells[377]
LncRNA UCA1MiR-203ZEB2Gastric cancerThis lncRNA sponges miR-203 to induce ZEB2, leading to the enhanced malignancy, invasion, and proliferation of tumor cells[187]
LncRNA SNHG5MiR-205-5pZEB2GliomaLncRNA SNHG5 stimulates ZEB2 by sponging miR-205-5p to elevate the proliferation of cancer cells[194]
LncRNA UICLMMiR-215ZEB2Colorectal cancerThe in vivo and in vitro experiments demonstrated that lncRNA induces ZEB2 via miR-215 down-regulation to enhance the migration and malignancy of cancer cells[378]
LncRNA SNHG12MiR-218Slug/ZEB2Non-small cell lung cancerMiR-218 inhibits Slug/ZEB2 axis to suppress EMT in cancer cells. LncRNA SNHG16 activates Slug/ZEB2 axis through miR-218 sponging[188]
LncRNA XISTMiR-367 and miR-141ZEB2Non-small cell lung cancerThe lncRNA XIST up-regulates the expression of ZEB2 by inhibition of miR-367 and miR-141, leading to the TGF- β-induced EMT[379]
LncRNA UCA1MiR-498ZEB2Esophageal cancerThe lncRNA UCA1 inhibits ZEB2 via miR-498 down-regulation to suppress the migration, proliferation, invasion, and EMT[7]
LncRNA CTSMiR-505ZEB2Cervical cancerDown-regulation of miR-505 by CTS is associated with increased malignancy of cancer cells through ZEB2 induction[186]
LncRNA HOTAIRM1MiR-873-5pZEB2GliomaLncRNA HOTAIRM1 decreases the expression of miR-873-5p by sponging to up-regulate the expression of ZEB2, leading to an increase in progression of glioma cells and a decrease in apoptotic cell death[179]
Table 6. miR/ZEB2 regulation by various molecular pathways in different cancers.
Table 6. miR/ZEB2 regulation by various molecular pathways in different cancers.
Upstream MediatorMiRDown-Stream TargetCancer TypeMajor OutcomesRefs
P53MiR-30aZEB2Breast cancerP53 stimulates the expression of miR-30a to upregulate ZEB2, resulting in reduced viability, proliferation, and invasion of cancer cells[169]
EZH2-DNMT1MiR-142-3pZEB2Nasopharyngeal carcinomaThe EZH2-DNMT1 induces ZEB2 through miR-142-3p sponging, resulting in an increase in cancer progression[380]
CircNUP214MiR-145ZEB2Thyroid cancerCircNUP214 induces ZEB2 through miR-145 down-regulation to enhance the malignancy and progression of cancer cells[381]
CircPCNXL2MiR-153ZEB2Renal cancerThe circPCNXL2 stimulates the expression of ZEB2 through miR-153 down-regulation to suppress the malignancy and invasion of cancer cells[201]
FOXP3MiR-155ZEB2Human breast cancerFOXP3 and miR-155 synergistically down-regulate the expression of ZEB2 to diminish the invasiveness of cancer cells[382]
Akt/ERKMiR-200cZEB2Gastric cancerThe inhibition of Akt/ERK enhances the expression of miR-200c to suppress IGF-I-mediated ZEB2, leading to the reduced invasion and EMT of cancer cells[383]
β1 integrinTGF-β/miR-200ZEB2Triple negative breast cancerEnhancing the expression of β1 integrin reduces the metastasis of cancer cells into lung. This is followed by disrupting TFG−β/miR-200/ZEB2, elevating the E-cadherin levels, and restoring the cohesion of cells[384]
CircZFRMiR-377ZEB2Bladder cancerEnhanced progression and malignancy of cancer cells result from down-regulation of miR-377 by circZFR and subsequent induction of ZEB2 [196]
Hsa-circ-0004771MiR-653ZEB2Breast cancerMiR-653 reduces the expression of ZEB2 and is associated with desirable prognosis. Hsa-circ-0004771 diminishes miR-653 expression to induce ZEB2, leading to the inhibition of apoptosis and enhanced migration and invasion of cancer cells[197]

Share and Cite

MDPI and ACS Style

Ashrafizadeh, M.; Ang, H.L.; Moghadam, E.R.; Mohammadi, S.; Zarrin, V.; Hushmandi, K.; Samarghandian, S.; Zarrabi, A.; Najafi, M.; Mohammadinejad, R.; et al. MicroRNAs and Their Influence on the ZEB Family: Mechanistic Aspects and Therapeutic Applications in Cancer Therapy. Biomolecules 2020, 10, 1040. https://doi.org/10.3390/biom10071040

AMA Style

Ashrafizadeh M, Ang HL, Moghadam ER, Mohammadi S, Zarrin V, Hushmandi K, Samarghandian S, Zarrabi A, Najafi M, Mohammadinejad R, et al. MicroRNAs and Their Influence on the ZEB Family: Mechanistic Aspects and Therapeutic Applications in Cancer Therapy. Biomolecules. 2020; 10(7):1040. https://doi.org/10.3390/biom10071040

Chicago/Turabian Style

Ashrafizadeh, Milad, Hui Li Ang, Ebrahim Rahmani Moghadam, Shima Mohammadi, Vahideh Zarrin, Kiavash Hushmandi, Saeed Samarghandian, Ali Zarrabi, Masoud Najafi, Reza Mohammadinejad, and et al. 2020. "MicroRNAs and Their Influence on the ZEB Family: Mechanistic Aspects and Therapeutic Applications in Cancer Therapy" Biomolecules 10, no. 7: 1040. https://doi.org/10.3390/biom10071040

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