Next Article in Journal
Mass Spectrometry-Based Disulfide Mapping of Lysyl Oxidase-like 2
Previous Article in Journal
Ion Channels of Spermatozoa: Structure, Function, and Regulation Mechanisms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential of the miR-200 Family as a Target for Developing Anti-Cancer Therapeutics

Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 24341, Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(11), 5881; https://doi.org/10.3390/ijms23115881
Submission received: 2 May 2022 / Revised: 20 May 2022 / Accepted: 21 May 2022 / Published: 24 May 2022

Abstract

:
MicroRNAs (miRNAs) are small non-coding RNAs (18–24 nucleotides) that play significant roles in cell proliferation, development, invasion, cancer development, cancer progression, and anti-cancer drug resistance. miRNAs target multiple genes and play diverse roles. miRNAs can bind to the 3′UTR of target genes and inhibit translation or promote the degradation of target genes. miR-200 family miRNAs mostly act as tumor suppressors and are commonly decreased in cancer. The miR-200 family has been reported as a valuable diagnostic and prognostic marker. This review discusses the clinical value of the miR-200 family, focusing on the role of the miR-200 family in the development of cancer and anti-cancer drug resistance. This review also provides an overview of the factors that regulate the expression of the miR-200 family, targets of miR-200 family miRNAs, and the mechanism of anti-cancer drug resistance regulated by the miR-200 family.

1. Biogenesis of MicroRNAs

MicroRNAs (miRNAs/miRs) are noncoding single-stranded RNAs of 18–24 nucleotides in length. They can modulate gene expression through post-transcriptional control and are involved in cancer cell proliferation [1,2], apoptosis [3,4], invasion [5,6], metastasis [7], and anti-cancer drug resistance [8].
miRNAs are transcribed as primary miRNAs, which are subsequently cleaved to precursor miRNAs (pre-miRNAs) and further processed into mature single-stranded ~22-nt miRNAs (Figure 1). The biogenesis of miRNAs requires RNase III enzymes DROSHA and DICER1, members of the Argonaute family (AGO1–4), and RNA polymerase II [9,10,11]. Mature miRNAs regulate gene expression by the cleavage of mRNA, translational repression, and the recruitment of epigenetic modifiers such as histone deacetylases (HDACs) and histone methyltransferases (HMT) [9,10,11] (Figure 1). The deletion of miRNA biogenesis proteins can result in embryonic lethality [12]. This suggests the role of miRNA biogenesis in developmental processes. The downregulation of DICER1, a regulator of miRNA biogenesis, decreased the expression of E-cadherin and enhanced the invasion of endometrial cancer cells [13]. This implies the role of DICER1 in epithelial-mesenchymal transition (EMT). DICER1 was highly expressed in cisplatin-resistant non-small cell lung cancer cells and induced cisplatin resistance by promoting autophagy [14] (Figure 1). DICER1 promoted colon cancer cell invasion by increasing the small non-coding RNA tRF-20-MEJB5Y13 [15] (Figure 1). DICER1 targeted the miR-200 family to promote the development of colon cancer and metastasis [16] (Figure 1). The overexpression of DICER1 enhanced the invasion and migration of lung cancer cells [17] (Figure 1). These findings suggest that the biogenesis of miRNAs plays a critical role in cancer development, cancer progression, and anti-cancer drug resistance.

2. miR-200 Family as a Diagnostic and Prognostic Marker

The decreased expression of the miR-200 family has been reported in various cancers. miR-200b was reported to be frequently downregulated in renal cell carcinoma [18]. Low levels of miR-200 were associated with high-grade glioblastomas [19]. High expression levels of miR-200b and miR-200c were associated with the high overall survival (OS) and progression-free survival (PFS) of patients with clear cell renal cell carcinoma (ccRCC) [20]. Low levels of miR-200b predicted the poor survival of patients with gastric cancer [21]. The low expression of miR-200b was strongly associated with the poor survival of patients with hepatocellular carcinomas [22]. High levels of the miR-200 family were strongly associated with better survival of bladder cancer patients [23]. The high expression of miR-200b was correlated with better responses of lung cancer patients to immunotherapy involving anti-PD-l antibodies such as pembrolizumab [24].
Circulating miRNAs (miRs) have been increasingly recognized as potential biomarkers in cancer [25,26]. Serum levels of miR-141 and miR-200a were decreased in hepatocellular carcinoma and could predict cancer metastasis [27]. Serum levels of miR-200b were lower in patients with non-small cell lung cancers than in healthy controls [28]. Circulating miR-200b is higher in metastatic breast cancer than in early breast cancer [29]. High levels of circulating miR-200c and miR-141 were associated with the poor OS of colon cancer patients [30]. Circulating miR-200 family members could predict OS and PFS in patients undergoing systemic therapy for metastatic breast cancer [31]. High levels of miR200 family members were strongly associated with reduced OS and PFS [31]. These reports indicate that the miR-200 family could serve as both diagnostic and prognostic markers. The above reports also suggest that the miR-200 family could be a target for developing anti-cancer drugs. Table 1 summarizes the clinical value of the miR-200 family as diagnostic and prognostic markers.

3. Regulation of miR-200 Family Expression

miR-200 family miRNAs are among the most extensively studied miRNAs. The miR-200 family consists of miR-200a, miR-200b, miR-200c, miR-141, and miR-429. These five miRNAs are clustered in two chromosomal locations. miR-200a, miR-200b, and miR-429 are located on chromosome 1, and miR-141 and miR-200c are located on chromosome 12 (Figure 2A). Figure 2B shows the seed sequences of miR-200 family miRNAs. The seed sequences of miR-200b/-c/-429 show one nucleotide difference from the seed sequence of miR-141/-200a (Figure 2B).
The promoter sequences of miR-200a/-b/-c contain a potential binding site for p53 (Figure 3A). p53 is known to increase the expression of the miR-200 family [32]. The downregulation of miR-200c by p53 mutation induced the resistance of breast cancer cells to doxorubicin [33]. p53-binding protein 1 (53BP1) inhibited epithelial-mesenchymal transition (EMT) in breast cancer cells by directly regulating the expression levels of miR-200b and miR-429 [34]. P53 might directly bind to the promoter sequences of the miR-200 family.
The promoter sequences of miR-200a/-b/-c contain a potential binding site for paired box-5 (Pax-5) (Figure 3A). Pax-5 could inhibit the invasion and proliferation of breast cancer cells by suppressing EMT [35]. Pax-5 inhibited the proliferation of breast cancer cells by increasing the expression of miR-215 [36]. Since miR-200 family miRNAs mostly function as tumor suppressors, Pax-5 might increase the expression of the miR-200 family.
The promoter sequences of miR-200a/-b/-c contain a potential binding site for nuclear receptors such as glucocorticoid receptor alpha (GR-α) (Figure 3A). GR activation suppressed pancreatic tumor growth [37]. It might increase the expression of the miR-200 family. Estrogen receptor (ER)-positive breast cancer cells were shown to express higher levels of miR-200c than triple-negative breast cancer cells [38]. The destabilization of the progesterone receptor by insulin-like growth factor-II mRNA-binding proteins 2 and 3 (IMP2 and IMP3) decreased the expression of miR-200a [39]. The decreased expression of miR-200a was seen in triple-negative breast cancer cells [39]. The overexpression of peroxisome proliferator-activated receptor alpha (PPARα) increased the expression of miR-200c in hepatocellular carcinoma cells [40]. Therefore, it is necessary to examine whether PPARα can suppress cancer growth.
The promoter sequences of miR-200a/-b/-c contain a potential binding site for Yin Yang 1 (YY1) (Figure 3A). The expression of YY1 was inversely correlated with miR-200a in Burkitt′s lymphoma (BL) tissue [41]. The promoter sequences of miR-200a/-b/-c contain a potential binding site for CAAT/enhancer-binding protein (C/EBP)-β (Figure 3A). The downregulation of C/EBP-β induced cisplatin resistance in malignant pleural mesothelioma cells [42]. Thus, it is necessary to examine the binding of YY1 and/or C/EBP-β to the promoter sequences of miR-200a/-b/-c. YY1 and C/EBP-β might regulate the expression of the miR-200 family.
Long non-coding RNAs can regulate the development of cancer, cancer cell proliferation, and invasion/migration of cancer cells by binding to mRNA, miRNA, and protein [43]. Long non-coding RNA LncARSR bound to the miR-200 family and increased the expression of zinc finger E-box binding homeobox (ZEB1)/ZEB2, a master regulator of EMT to induce EMT in ovarian cancer cells [43]. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) induced docetaxel resistance in breast cancer cells by decreasing the expression of miR-200b [44]. The expression of long non-coding RNA ZFAS1 was higher in colon cancer tissue than in normal tissue [45]. ZFAS1 targeted miR-200b/-c to promote EMT in colon cancer cells [45]. CCAT2, a long non-coding RNA, promoted the invasion and tumorigenic potential of esophageal squamous cell carcinoma (ESCC) by decreasing the expression of miR-200b [46]. Lnc-ATB, a long non-coding RNA, decreased the expression of miR-200c to promote cholangiocarcinoma (CCA) growth [47]. Lnc RNA H19 targeted miR-200a and promoted the invasion and migration of glioma cells [48].
Epigenetic modifications play critical roles in EMT and cancer cell proliferation [49]. The promoter regions of miR-200 clusters contain CpG islands that undergo DNA methylation [50,51,52,53]. Promoter methylation of miR-200b promoted the proliferation and invasion of endometrial cancer cells [54]. DNMT1 and enhancer of zeste homolog 2 (EZH2), a histone methyltransferase, could bind to the promoter sequences of miR-200b/a/429, leading to the downregulation of the miR-200 family [55]. Promoter methylation by MYC and DNMT3A decreased the expression of miR-200b in triple-negative breast cancer cells [56]. Therefore, it would be interesting to examine the effects of DNMT1 and DNMT3 A on EMT, cancer growth, and anti-cancer drug resistance. TargetScan analysis predicted histone deacetylase 4 (HDAC4) as a target of the miR-200 family. HDAC4 induced EMT and cancer stem cell-like properties in cancer cells [57]. The overexpression of HDAC4 decreased the expression of miR-200b, which led to the resistance of lung cancer cells to anti-cancer drugs [58].
MicroRNA-200b (miR-200b) is a downstream target of p38γ mitogen-activated protein kinase p38γ MAPK and is inhibited by p38γ MAPK [59]. The activation of Kindlin-2-integrin β1-AKT signaling was associated with the decreased expression of miR-200b in esophageal squamous cell carcinoma cells [60]. Hepatocyte nuclear factors (HNFs) were reported to bind to promoter sequences of miR-200b, increase the expression of miR-200b, and suppress the stemness of colorectal cancer cells [61]. HNF-1β promoted EMT and the tumorigenic potential of hepatocellular carcinoma cells by activating Notch signaling [62]. Figure 3B shows factors that regulate the expression of the miR-200 family.

4. miR-200 Family and Targets of the miR-200 Family

miR-200 family miRNAs mainly function as tumor suppressors [63,64]. Transgenic overexpression of the miR-200 family suppressed the development of mammary tumors [65].
miR-200b targeted CXCL12 and suppressed the invasion and metastatic potential of gastric cancer cells [66]. The overexpression of miR-200c repressed genes encoding immune suppressive factors, including CD274, HMOX-1, and GDF15 [38]. Blockade of CXCL12-CXCR4 signaling enhanced anti-tumor effects by inhibiting immune suppression in ovarian cancer [67].
miR-200b targets Neuregulin 1 (NRG1) to inhibit the invasion of gastric cancer cells [21]. miR-200b targeted Notch1 and inhibited the proliferation and tube-forming potential of human umbilical vein endothelial cells [68]. miR-200 deficiency activated Notch signaling and promoted the proliferation of cancer-associated fibroblasts and the metastatic potential of lung cancer cells [69]. TargetScan analysis predicted bone morphogenetic protein 4 (BMP4) as a target of the miR-200 family. BMP4 promoted the resistance of MDA-MB-231 cells to anti-cancer drugs by upregulating Notch signaling [70]. miR-200b-mimics inhibited p38γ MAPK-induced EMT [59]. miR-200b/-c targeted rho family GTPase 3 (RhoE) and inhibited the proliferation of non-small cell lung cancer cells [71].
miR-200b directly regulated the expression of high mobility group 3B (HMG3B) and inhibited the proliferation of hepatocellular carcinoma cells [22]. miR-200b and miR-200c suppressed the progression of glioblastoma by directly decreasing the expression of HMG3B [72]. E2F transcription factor 3 (E2F3) and ZEB1 are targets of miR-200b and regulated docetaxel resistance in lung adenocarcinoma cells [44].
EMT plays a critical role in the crosstalk of tumor cells within the microenvironment. EMT is a highly plastic program. In other words, the mesenchymal/EMT phenotype can revert to mesenchymal-epithelial transition. The miR-200 family can regulate EMT [73,74]. miR-200b plays a critical role in EMT by interacting with genes involved in EMT, including receptors, signaling, and the cell cycle [75]. The miR-200 family plays a major role in specifying the epithelial phenotype by preventing the expression of transcription repressors ZEB1 and ZEB2 (Smad-interacting protein 1) [76,77,78]. TargetScan analysis predicted ZEB2 as a target of the miR-200 family. miR-200b/200a/429 suppressed the metastatic potential of breast cancer cells by decreasing the expression of ZEB1 [76]. miR-200b/miR-429 decreased the expression of ZEB1/2 and inhibited the migration potential of oral squamous cell carcinoma (OSCC) cells [79]. miR-200b-3p and miR-429-5p suppressed the proliferation and invasion of triple-negative breast cancer cells by decreasing the expression of cyclinD1/CDK4/CDK6 [80]. The downregulation of cyclin D1 led to the inhibition of EMT and cell cycle arrest [81]. EMT-regulating transcription factors are known to regulate anti-cancer drug resistance [82]. The downregulation of SNAIL enhanced the sensitivity of prostate cancer cells to anti-cancer drugs [83]. EMT activated cancer stem cells (CSCs) resistant to chemotherapy and target therapy [84]. Figure 3C shows the targets of the miR-200 family and the roles of these targets in anti-cancer drug resistance.

5. Role of the miR-200 Family in Anti-Cancer Drug Resistance

miRNAs can regulate the responses to anti-cancer drugs [85]. miR-200a-3p was shown to target dual-specificity phosphatase 6 (DUSP6) and enhance the sensitivity of hepatocellular carcinoma cells to 5-fluorouracil (5-FU) [86]. miR-200c targeted tyrosine-protein kinase B (TrkB) to enhance the sensitivity of breast cancer cells to doxorubicin [87]. TrkB promoted the EMT of prostate cancer cells in an Akt-dependent manner [88]. miR-200b inhibited the tumorigenic potential and enhanced the sensitivity of lung cancer cells to cisplatin by inhibiting ERK/Akt signaling and targeting ribosomal protein S6 kinase 1 (p70S6K1) [89]. miR-429 enhanced the sensitivity of pancreatic cancer cells to gemcitabine by upregulating Akt-inhibited programmed cell death 4 (PDCD4) [90].
High levels of EMT markers (ZEB1/ZEB2) and low levels of the miR-200 family (miR-200a/-b/-c) resulted in the resistance of estrogen receptor (ER)-positive breast cancer cells to tamoxifen [91]. The downregulation of miR-200c restored EMT and conferred the resistance of prostate cancer cells to docetaxel [92]. miR-200b was the most significantly downregulated miRNA in doxorubicin-resistant breast cancer cells [93]. miR-200b targeted fibronectin 1 (FN1) and suppressed EMT phenotypes to overcome doxorubicin resistance in breast cancer cells [93].
DNMT1 directly decreased the expression levels of miR-200a/-b/-429 and inhibited the progression of gastric cancer and glioblastoma [55]. This indicates the role of epigenetic modifications and the miR-200 family in anti-cancer drug resistance. miR-200b and miR-200c synergistically enhanced the sensitivity of ovarian cancer cells to cisplatin by targeting DNA methyltransferase I (DNMT1) [94]. DNMT1 promoted the resistance of breast cancer cells to cisplatin by decreasing the expression of miR-200b [95]. Flap endonuclease (FEN1)/DNMT3a complex decreased the expression of miR-200a and promoted breast cancer cell proliferation [96]. This implies that miR-200a may enhance chemosensitivity in breast cancer cells.
The allelic loss of Beclin1, a marker of autophagy, has been reported in various cancers [97]. The high expression of autophagy markers was strongly associated with poor survival of hepatocellular cancer patients [98]. Autophagy induction promoted EMT in glioma cells [99]. These reports indicate the roles of autophagy in the development of cancers and anti-cancer drug resistance. Cancer cells may induce autophagy for survival in response to anti-cancer drugs [100,101]. Autophagy is positively associated with anti-cancer drug resistance [102,103]. The activation of phosphoinositide 3 (PI3K)/Akt signaling promoted autophagy and chemotherapy resistance in breast cancer cells [104]. Targeting autophagy is known to enhance the cytotoxic effects of trastuzumab on human epidermal growth factor receptor 2 (HER2)-positive gastric cancer cells [105]. Autophagy inhibition enhanced the sensitivity of non-small cell lung cancer cells to osimertinib by decreasing the expression of SRY-box transcription factor 2 (SOX2), a marker of stemness [106]. This implies that cancer stemness is closely associated with anti-cancer drug resistance.
ZEB1, a regulator of EMT, induced autophagy to cause anti-cancer drug resistance in breast cancer cells [101]. ZEB1 may induce chemotherapy resistance by promoting EMT. This also implies a negative regulatory role of miR-200s in autophagy and chemotherapy resistance. The expression of miR-200b was inversely correlated with autophagy-associated gene 12 (ATG12) in docetaxel-resistant lung adenocarcinoma cells [107]. The downregulation of ATG12 by miR-200b enhanced the chemosensitivity of lung adenocarcinoma cells to docetaxel [107]. miR-200b decreased the expression of ATG-5 and enhanced the sensitivity of breast cancer cells to cisplatin and docetaxel [108]. It is probable that the combination of a miRNA-mimic (miRNA-mimic) and an inhibitor of autophagy can overcome the resistance of cancer cells to anti-cancer drugs.
miR-200b inhibited the proliferation of skin cancer stem cells [109]. SOX2 is known to be a target of the miR-200 family in colorectal cancer [110]. miR-429 enhanced the sensitivity of gastric cancer cells to cisplatin by decreasing the expression of SOX2 and inhibiting PI3K/Akt/mammalian target of rapamycin (mTOR) signaling [111]. These reports further indicate the role of cancer stem cells in anti-cancer drug resistance.
Cancer-associated gene (CAGE), a cancer/testis antigen, could bind to SOX2 and regulate the cancer stem cell-like properties of breast cancer cells [108]. CAGE bound to HDAC2 and SNAIL and repressed the expression of p53 in anti-cancer drug-resistant melanoma cells [112] (Figure 4A). CAGE conferred resistance to epidermal growth factor receptor (EGFR)-TKIs (EGFR tyrosine kinase inhibitors) by promoting autophagy via binding to Beclin1 in non-small cell lung cancer cells [113]. Since p53 increases the expression of the miR-200 family [32], miR-200 family members might decrease the expression of CAGE. TargetScan analysis predicted miR-200b as a negative regulator of CAGE. Figure 4A shows the potential binding of the miR-200 family to the 3′ untranslated region (UTR) of CAGE. miR-200b and CAGE could form a negative feedback loop and regulate the response of melanoma cells to anti-cancer drugs [114] (Figure 4B). miR-200b exerted negative effects on cancer stemness by decreasing the expression levels of CAGE and autophagic flux in breast cancer cells [108]. miR-200b decreased the expression of EGFR and suppressed the migration of gastrointestinal stromal tumors [115]. CAGE bound to the EGFR and HER2 and regulated the responses of melanoma cells to trastuzumab and microtubule-targeting anti-cancer drugs [116] (Figure 4B). Therefore, the effects of other miR-200 family members on the responses of cancer cells to anti-cancer drugs targeting EGFR should be examined.
HDAC3 could bind to the promoter sequences of CAGE and inhibit the tumorigenic potential of anti-cancer drug-resistant melanoma cells [117] (Figure 4B). HDAC3 enhanced the sensitivity of anti-cancer drug-resistant melanoma cells to anti-cancer drugs by negatively regulating EGFR signaling and CAGE expression [117] (Figure 4B). miR-326 acted as a negative regulator of HDAC3 and enhanced the invasion and migration of melanoma cells [118] (Figure 4B). miR-326 and miR-200b formed a negative feedback loop to regulate the response of melanoma cells to anti-cancer drugs [118]. HDAC3 might regulate EMT and autophagic flux. It is probable that the miR-200 family and HDAC3 might form a positive feedback loop.
The downregulation of tubulin β3 by HDAC3 enhanced the sensitivity of melanoma cells to microtubule-targeting agents [119]. Tubulin β3 may confer resistance to anti-cancer drugs by promoting EMT and autophagic flux. The overexpression of miR-200c targets class III tubulin (TUBB3) restored the expression of E-cadherin and enhanced the sensitivity to microtubule targeting agents in ovarian cancer cells [120]. miR-200c also targeted ZEB1/ZEB2 and various mesenchymal genes (FN1 and QK1) [120]. Other members of the miR-200 family might regulate the expression of tubulin β3.
Immune checkpoint molecules such as programmed death ligand-1 (PD-L1) and PD-1 are known as targets for anti-cancer drug development [121]. Figure 4C shows the regulation of PD-L1 expression by the miR-200 family. miR-200a-3p and miR-200c-3p decreased the expression of PD-L1, suppressed the development of colorectal cancer, and promoted anti-cancer immune responses [121]. miR-429 directly targeted PD-L1 and suppressed gastric cancer cell proliferation [122]. PD-L1 served as a target of miR-200a in non-small cell lung cancer cells [123]. PD-L1 promoted EMT by preventing glycogen synthase kinase 3β (GSK3β) from degrading SNAIL in triple-negative breast cancer cells [124] (Figure 4C). PD-L1 bound to the EGFR and promoted TNF-related apoptosis-inducing ligand (TRAIL) resistance in gastric cancer cells [125] (Figure 4C). miR-429 targeted PD-L1 and enhanced the sensitivity of gastric cancer cells to TRAIL [125] (Figure 4C). CAGE inactivated GSK3β by binding to GSK3β. The inactivation of GSK3β led to the increased expression of cyclinD1, causing the resistance of melanoma cells to microtubule-targeting drugs [126] (Figure 4C). A CAGE-derived peptide (269GTGKT273) could bind to CAGE and prevent CAGE from inactivating GSK3β [126] (Figure 4C). The CAGE-derived peptide enhanced the sensitivity of melanoma cells to microtubule-targeting drugs [126] (Figure 4C). Additionally, the CAGE–derived peptide inhibited the binding of CAGE to the EGFR and enhanced the sensitivity of anti-cancer drug-resistant melanoma cells to gefitinib and trastuzumab [116]. These reports indicate the potential role of the miR-200 family-CAGE-EGFR-PD-L1 molecular network in the resistance to immune checkpoint inhibitors, EGFR-tyrosine kinase inhibitors (EGFR-TKIs), and other anti-cancer drugs.
TargetScan analysis predicted proline, glutamate, and leucine-rich protein 1 (PELP1) as a target of the miR-200 family. PELP1 is overexpressed in various cancers. Its high expression contributes to the pathogenesis of triple-negative breast cancer [127]. The high expression of PELP1 was strongly associated with the poor survival of lung adenocarcinoma patients [128]. PELP1 downregulation by miR-200-mimic or miR-141-mimic decreased the metastatic potential of cancer cells [129]. PELP1 directly bound to the promoter sequences of miR-200a and recruited HDAC2, which decreased the expression of miR-200a in breast cancer cells [129]. The downregulation of PELP1 enhanced the efficacy of chemotherapy by suppressing signal transducer and activator of transcription 3 (STAT3)/vascular endothelial growth factor (VEGF) signaling in colorectal cancer cells [130]. The downregulation of PELP1 enhanced the sensitivity of breast cancer cells to genotoxic agents by suppressing the cell cycle and enhancing apoptosis [131]. Since CAGE could bind to HDAC2 in anti-cancer drug-resistant melanoma cells, it will be interesting to examine whether CAGE could bind to PELP1. PELP1 might regulate the expression of the miR-200 family and bind to CAGE to exert its effect on the responses of cancer cells to anti-cancer drugs.
The epigenetic silencing of miRNAs has been reported in various cancers [132,133] and played critical roles in anti-cancer drug resistance [134]. The role of DNMT1 in chemotherapy resistance has been reported [135,136]. The epigenetic silencing of the miR-200 family was strongly associated with acquired EGFR-TKI resistance in non-small cell lung cancer (NSCLC) cells [137].
EGFR signaling mediated autophagy and anti-cancer drug resistance in non-small cell lung cancer cells [138]. Osimertinib-resistant non-small cell lung cancer cells displayed activated ERBB2 [139]. The overexpression of miR-200c inhibited the proliferation of acquired EGFR-TKI-resistant non-small cell lung cancer cells [137] and overcame resistance to gefitinib by inhibiting PI3K/Akt signaling in non-small cell lung cancer cells [140]. The activation of the hedgehog (Hh) signaling mediated the resistance of non-small cell lung cancer cells to EGFR-TKIs (gefitinib, afatinib, and osimertinib) [141]. miR-200b enhanced the sensitivity of non-small cell lung cancer cells to erlotinib and cisplatin by inhibiting Hh signaling [142]. The combination of a miR-200-mimic and inhibitor of EGFR signaling might enhance the sensitivity of cancer cells to EGFR-TKIs. Table 2 summarizes the mechanism of anti-cancer drug resistance regulated by the miR-200 family.

6. Conclusions

The miR-200 family has been reported to play diverse roles in cell proliferation, cancer development, EMT regulation, autophagy, and anti-cancer drug resistance. Many studies have reported the decreased expression of the miR-200 family in cancer tissues [18,19], and anti-cancer drug-resistant cancer cells [93]. The expression levels of miR-200 family miRNAs were shown to predict responses to anti-cancer therapeutics [19,20,21,22,23,24]. Thus, miR-200 family miRNAs can be a target for anti-cancer drug development. The global identification of targets of miR-200 family miRNAs is necessary to better understand the role of miR-200 family miRNAs in the development of cancer and anti-cancer drug resistance. These miR-200 family miRNA targets can also be employed for developing anti-cancer drugs.
Since miR-200 family miRNAs are mostly decreased in anti-cancer drug-resistant cells compared to parental sensitive cells, it is necessary to develop therapeutic miR-200 family miRNA-mimics (miR-200-mimics) that can overcome the resistance to anti-cancer drugs. The overexpression of miR-200 family miRNAs has been reported to enhance chemosensitivity [107,108,111]. Drugs targeting miR-200 family-regulated genes can be combined with miR-200 family miRNA-mimics to overcome the resistance to anti-cancer drugs.
miR-200 family miRNAs have been shown to target PD-L1 [121,122,123]. The combination of miR-200 family miRNA-mimics and immune checkpoint inhibitors might enhance the therapeutic potential of immune checkpoint inhibitors. miR-200 family miRNAs could negatively regulate EGFR signaling [115]. Combination therapy involving miR-200 family miRNA-mimics and EGFR-TKIs could overcome the resistance to EGFR-TKI. miR-200b was shown to form a negative feedback loop with the cancer/testis antigen CAGE to regulate anti-cancer drug resistance [114]. A CAGE-derived peptide enhanced the sensitivity of melanoma cells to anti-cancer drugs by disrupting CAGE-GSK3β interaction [126]. As with CAGE, PD-L1 could bind to the EGFR and inactivate GSK3β [124]. Thus, combining a miR-200 family miRNA-mimic and a CAGE-derived peptide can overcome resistance to immune check inhibitors.
Since CAGE forms a negative feedback loop with miR-200b, it is necessary to identify small molecules that target CAGE. These molecules may enhance the therapeutic value of miR-200-mimics. Polyphenols increased the expression of miR-200b and inhibited the proliferation of skin cancer stem cells [109]. Small molecules that regulate the expression of miRNA have been reported in breast cancer cells by employing a graphene-based biosensor [143]. Chemicals that increase the expression of miR-200 family miRNAs might overcome the resistance to anti-cancer drugs.
miRNA-based therapeutics employing mimics or inhibitors can be a reasonable tool to treat cancer and overcome the resistance to anti-cancer drugs [144] and are under development. Most clinical trials involving miRNAs are in phase I or phase II trials. However, no clinical trials involving mimics of miR-200 family miRNAs are underway. Compared to small interfering RNAs (siRNAs), few miRNA-mimics are in clinical trials. Unlike siRNAs, miRNAs target multiple genes, making miRNAs attractive targets for developing anti-cancer therapeutics. However, miRNA-mimics may cause off-target effects. In addition, miRNA-mimics are unstable with difficulty penetrating cell membranes due to their negative charge. Thus, the modification of miRNA-mimics is necessary to improve their pharmacokinetics and pharmacodynamics. The efficient delivery of miRNA-mimics is critical for the successful development of miRNA-based anti-cancer drugs. Delivery systems include viral vectors, lipid nanoparticles, cationic lipids, cell-penetrating peptides, polymer-based vectors, and bacterial mini cell vehicles. In the case of miRNA-mimics, the mimic concentration is also critical for successful clinical trials.
As mentioned above, efforts have been made to improve the specificity, immunogenicity, and delivery of miRNA-mimics. Although miRNA-mimics have not been successful in clinical trials, various efforts to enhance the clinical value of miRNA-mimics will eventually overcome cancer and anti-cancer drug resistance.

Author Contributions

D.J. conceived the idea and wrote the manuscript. H.J. and K.S. critically reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by National Research Foundation Grants (2020R1A2C1006996 and 2017M3A9G7072417) and a grant from the BK21 Plus Program.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Liu, W.; Ma, C.; Xu, H.; Wang, L.; Xu, W.; Zhang, H.; Wang, Z.; Li, J.; Zhang, J.; Liu, X.; et al. miR-184-5p inhibits cell proliferation, invasion and predicts prognosis of clear cell renal cell carcinoma by targeting NUS1 dehydrodolichyl diphosphate synthase subunit: Results from large-scale comprehensive identification and validation. J. Cancer 2022, 13, 1398–1409. [Google Scholar] [CrossRef]
  2. Chen, S.; Tu, Y.; Yuan, H.; Shi, Z.; Guo, Y.; Gong, W.; Tu, S. Regulatory functions of miR200b3p in tumor development (Review). Oncol. Rep. 2022, 47, 96. [Google Scholar] [CrossRef]
  3. Luo, H.; Lv, W.; Zhang, H.; Lin, C.; Li, F.; Zheng, F.; Zhong, B. miR-204-5p inhibits cell proliferation and induces cell apoptosis in esophageal squamous cell carcinoma by regulating Nestin. Int. J. Med. Sci. 2022, 19, 472–483. [Google Scholar] [CrossRef]
  4. Zhang, F.; Cheng, N.; Du, J.; Zhang, H.; Zhang, C. MicroRNA-200b-3p promotes endothelial cell apoptosis by targeting HDAC4 in atherosclerosis. BMC Cardiovasc. Disord. 2021, 21, 172. [Google Scholar] [CrossRef]
  5. Simpson, K.; Conquer-van Heumen, G.; Watson, K.L.; Roth, M.; Martin, C.J.; Moorehead, R.A. Re-expression of miR-200s in claudin-low mammary tumor cells alters cell shape and reduces proliferation and invasion potentially through modulating other miRNAs and SUZ12 regulated genes. Cancer Cell Int. 2021, 21, 89. [Google Scholar] [CrossRef]
  6. Tang, H.; Wang, X.; Zhang, M.; Yan, Y.; Huang, S.; Ji, J.; Xu, J.; Zhang, Y.; Cai, Y.; Yang, B.; et al. MicroRNA-200b/c-3p regulate epithelial plasticity and inhibit cutaneous wound healing by modulating TGF-beta-mediated RAC1 signaling. Cell Death Dis. 2020, 11, 931. [Google Scholar] [CrossRef]
  7. Quirico, L.; Orso, F.; Cucinelli, S.; Paradzik, M.; Natalini, D.; Centonze, G.; Dalmasso, A.; La Vecchia, S.; Coco, M.; Audrito, V.; et al. miRNA-guided reprogramming of glucose and glutamine metabolism and its impact on cell adhesion/migration during solid tumor progression. Cell Mol. Life Sci. 2022, 79, 216. [Google Scholar] [CrossRef]
  8. Lv, Q.; Xia, Q.; Li, A.; Wang, Z. circRNA_101277 Influences Cisplatin Resistance of Colorectal Cancer Cells by Modulating the miR-370/IL-6 Axis. Genet. Res. 2022, 2022, 4237327. [Google Scholar] [CrossRef]
  9. Liu, J.; Zhou, F.; Guan, Y.; Meng, F.; Zhao, Z.; Su, Q.; Bao, W.; Wang, X.; Zhao, J.; Huo, Z.; et al. The Biogenesis of miRNAs and Their Role in the Development of Amyotrophic Lateral Sclerosis. Cells 2022, 11, 572. [Google Scholar] [CrossRef]
  10. Tang, L. Recapitulating miRNA biogenesis in cells. Nat. Methods 2022, 19, 35. [Google Scholar] [CrossRef]
  11. Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef]
  12. Krill, K.T.; Gurdziel, K.; Heaton, J.H.; Simon, D.P.; Hammer, G.D. Dicer deficiency reveals microRNAs predicted to control gene expression in the developing adrenal cortex. Mol. Endocrinol. 2013, 27, 754–768. [Google Scholar] [CrossRef]
  13. Li, B.; Lu, W.; Qu, J.; Zhang, Y.; Wan, X. DICER1 regulates endometrial carcinoma invasion via histone acetylation and methylation. J. Cancer 2017, 8, 933–939. [Google Scholar] [CrossRef] [Green Version]
  14. Li, C.; Chen, L.; Song, W.; Peng, B.; Zhu, J.; Fang, L. DICER activates autophagy and promotes cisplatin resistance in non-small cell lung cancer by binding with let-7i-5p. Acta Histochem. 2021, 123, 151788. [Google Scholar] [CrossRef]
  15. Luan, N.; Mu, Y.; Mu, J.; Chen, Y.; Ye, X.; Zhou, Q.; Xu, M.; Deng, Q.; Hu, Y.; Tang, Z.; et al. Dicer1 Promotes Colon Cancer Cell Invasion and Migration Through Modulation of tRF-20-MEJB5Y13 Expression Under Hypoxia. Front. Genet. 2021, 12, 638244. [Google Scholar] [CrossRef]
  16. Iliou, M.S.; da Silva-Diz, V.; Carmona, F.J.; Ramalho-Carvalho, J.; Heyn, H.; Villanueva, A.; Munoz, P.; Esteller, M. Impaired DICER1 function promotes stemness and metastasis in colon cancer. Oncogene 2014, 33, 4003–4015. [Google Scholar] [CrossRef] [Green Version]
  17. Wang, J.; Zhang, Y.; Ge, F. MiR-186 Suppressed Growth, Migration, and Invasion of Lung Adenocarcinoma Cells via Targeting Dicer1. J. Oncol. 2021, 2021, 6217469. [Google Scholar] [CrossRef]
  18. Li, Y.; Guan, B.; Liu, J.; Zhang, Z.; He, S.; Zhan, Y.; Su, B.; Han, H.; Zhang, X.; Wang, B.; et al. MicroRNA-200b is downregulated and suppresses metastasis by targeting LAMA4 in renal cell carcinoma. EBioMedicine 2019, 44, 439–451. [Google Scholar] [CrossRef] [Green Version]
  19. Kong, X.; Gong, S.; Yan, T.; Yang, Y. MicroRNA-200b expression level is negatively associated with pathological grading in human gliomas. Cancer Manag. Res. 2018, 10, 2825–2834. [Google Scholar] [CrossRef]
  20. Saleeb, R.; Kim, S.S.; Ding, Q.; Scorilas, A.; Lin, S.; Khella, H.W.; Boulos, C.; Ibrahim, G.; Yousef, G.M. The miR-200 family as prognostic markers in clear cell renal cell carcinoma. Urol. Oncol. 2019, 37, 955–963. [Google Scholar] [CrossRef]
  21. Xu, T.; Xie, F.; Xu, D.; Xu, W.; Ge, X.; Lv, S.; Li, S. MiR-200b Suppresses Gastric Cancer Cell Migration and Invasion by Inhibiting NRG1 through ERBB2/ERBB3 Signaling. J. Oncol. 2021, 2021, 4470778. [Google Scholar] [CrossRef]
  22. Wang, L.K.; Xie, X.N.; Song, X.H.; Su, T.; Chang, X.L.; Xu, M.; Liang, B.; Huang, D.Y. Upregulation of miR-200b Inhibits Hepatocellular Carcinoma Cell Proliferation and Migration by Targeting HMGB3 Protein. Technol. Cancer Res. Treat. 2018, 17, 1533033818806475. [Google Scholar] [CrossRef] [Green Version]
  23. Mei, Y.; Zheng, J.; Xiang, P.; Liu, C.; Fan, Y. Prognostic value of the miR-200 family in bladder cancer: A systematic review and meta-analysis. Medicine 2020, 99, e22891. [Google Scholar] [CrossRef]
  24. Grenda, A.; Krawczyk, P.; Blach, J.; Chmielewska, I.; Kubiatowski, T.; Kieszko, S.; Wojas-Krawczyk, K.; Kucharczyk, T.; Jarosz, B.; Pasnik, I.; et al. Tissue MicroRNA Expression as a Predictor of Response to Immunotherapy in NSCLC Patients. Front. Oncol. 2020, 10, 563613. [Google Scholar] [CrossRef]
  25. Gharib, A.F.; Eed, E.M.; Khalifa, A.S.; Raafat, N.; Shehab-Eldeen, S.; Alwakeel, H.R.; Darwiesh, E.; Essa, A. Value of Serum miRNA-96-5p and miRNA-99a-5p as Diagnostic Biomarkers for Hepatocellular Carcinoma. Int. J. Gen. Med. 2022, 15, 2427–2436. [Google Scholar] [CrossRef]
  26. Santos, A.; Cristobal, I.; Rubio, J.; Carames, C.; Luque, M.; Sanz-Alvarez, M.; Morales-Gallego, M.; Madoz-Gurpide, J.; Rojo, F.; Garcia-Foncillas, J. MicroRNA-199b Deregulation Shows Oncogenic Properties and Promising Clinical Value as Circulating Marker in Locally Advanced Rectal Cancer Patients. Int J. Mol. Sci. 2022, 23, 2203. [Google Scholar] [CrossRef]
  27. Chen, S.; Zhang, J.; Chen, Q.; Cheng, J.; Chen, X.; Mao, Y.; Chen, W.; Liu, C.; Wu, H.; Lv, Y.; et al. MicroRNA-200a and microRNA-141 have a synergetic effect on the suppression of epithelial-mesenchymal transition in liver cancer by targeting STAT4. Oncol. Lett. 2021, 21, 137. [Google Scholar] [CrossRef]
  28. Zou, J.G.; Ma, L.F.; Li, X.; Xu, F.L.; Fei, X.Z.; Liu, Q.; Bai, Q.L.; Dong, Y.L. Circulating microRNA array (miR-182, 200b and 205) for the early diagnosis and poor prognosis predictor of non-small cell lung cancer. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 1108–1115. [Google Scholar]
  29. Papadaki, C.; Stoupis, G.; Tsalikis, L.; Monastirioti, A.; Papadaki, M.; Maliotis, N.; Stratigos, M.; Mastrostamatis, G.; Mavroudis, D.; Agelaki, S. Circulating miRNAs as a marker of metastatic disease and prognostic factor in metastatic breast cancer. Oncotarget 2019, 10, 966–981. [Google Scholar] [CrossRef] [Green Version]
  30. Santasusagna, S.; Moreno, I.; Navarro, A.; Martinez Rodenas, F.; Hernandez, R.; Castellano, J.J.; Munoz, C.; Monzo, M. Prognostic Impact of miR-200 Family Members in Plasma and Exosomes from Tumor-Draining versus Peripheral Veins of Colon Cancer Patients. Oncology 2018, 95, 309–318. [Google Scholar] [CrossRef]
  31. Fischer, C.; Deutsch, T.M.; Feisst, M.; Rippinger, N.; Riedel, F.; Hartkopf, A.D.; Brucker, S.Y.; Domschke, C.; Fremd, C.; Michel, L.; et al. Circulating miR-200 family as predictive markers during systemic therapy of metastatic breast cancer. Arch. Gynecol. Obstet. 2022, 6, e1263412. [Google Scholar] [CrossRef]
  32. Jing, J.; Xiong, S.; Li, Z.; Wu, J.; Zhou, L.; Gui, J.F.; Mei, J. A feedback regulatory loop involving p53/miR-200 and growth hormone endocrine axis controls embryo size of zebrafish. Sci. Rep. 2015, 5, 15906. [Google Scholar] [CrossRef] [Green Version]
  33. Alam, F.; Mezhal, F.; El Hasasna, H.; Nair, V.A.; Aravind, S.R.; Saber Ayad, M.; El-Serafi, A.; Abdel-Rahman, W.M. The role of p53-microRNA 200-Moesin axis in invasion and drug resistance of breast cancer cells. Tumour Biol. 2017, 39, 1010428317714634. [Google Scholar] [CrossRef] [Green Version]
  34. 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] [Green Version]
  35. Benzina, S.; Beauregard, A.P.; Guerrette, R.; Jean, S.; Faye, M.D.; Laflamme, M.; Maicas, E.; Crapoulet, N.; Ouellette, R.J.; Robichaud, G.A. Pax-5 is a potent regulator of E-cadherin and breast cancer malignant processes. Oncotarget 2017, 8, 12052–12066. [Google Scholar] [CrossRef] [Green Version]
  36. Leblanc, N.; Harquail, J.; Crapoulet, N.; Ouellette, R.J.; Robichaud, G.A. Pax-5 Inhibits Breast Cancer Proliferation Through MiR-215 Up-regulation. Anticancer Res. 2018, 38, 5013–5026. [Google Scholar] [CrossRef] [Green Version]
  37. Yao, Y.; Yao, Q.Y.; Xue, J.S.; Tian, X.Y.; An, Q.M.; Cui, L.X.; Xu, C.; Su, H.; Yang, L.; Feng, Y.Y.; et al. Dexamethasone inhibits pancreatic tumor growth in preclinical models: Involvement of activating glucocorticoid receptor. Toxicol. Appl. Pharmacol. 2020, 401, 115118. [Google Scholar] [CrossRef]
  38. Rogers, T.J.; Christenson, J.L.; Greene, L.I.; O’Neill, K.I.; Williams, M.M.; Gordon, M.A.; Nemkov, T.; D’Alessandro, A.; Degala, G.D.; Shin, J.; et al. Reversal of Triple-Negative Breast Cancer EMT by miR-200c Decreases Tryptophan Catabolism and a Program of Immunosuppression. Mol. Cancer Res. 2019, 17, 30–41. [Google Scholar] [CrossRef] [Green Version]
  39. Kim, H.Y.; Ha Thi, H.T.; Hong, S. IMP2 and IMP3 cooperate to promote the metastasis of triple-negative breast cancer through destabilization of progesterone receptor. Cancer Lett. 2018, 415, 30–39. [Google Scholar] [CrossRef]
  40. Zhang, Y.; Yang, Z.; Whitby, R.; Wang, L. Regulation of miR-200c by nuclear receptors PPARalpha, LRH-1 and SHP. Biochem Biophys Res. Commun. 2011, 416, 135–139. [Google Scholar] [CrossRef] [Green Version]
  41. Hafsi, S.; Candido, S.; Maestro, R.; Falzone, L.; Soua, Z.; Bonavida, B.; Spandidos, D.A.; Libra, M. Correlation between the overexpression of Yin Yang 1 and the expression levels of miRNAs in Burkitt’s lymphoma: A computational study. Oncol. Lett. 2016, 11, 1021–1025. [Google Scholar] [CrossRef] [Green Version]
  42. Kopecka, J.; Salaroglio, I.C.; Righi, L.; Libener, R.; Orecchia, S.; Grosso, F.; Milosevic, V.; Ananthanarayanan, P.; Ricci, L.; Capelletto, E.; et al. Loss of C/EBP-beta LIP drives cisplatin resistance in malignant pleural mesothelioma. Lung Cancer 2018, 120, 34–45. [Google Scholar] [CrossRef]
  43. Shu, C.; Yan, D.; Mo, Y.; Gu, J.; Shah, N.; He, J. Long noncoding RNA lncARSR promotes epithelial ovarian cancer cell proliferation and invasion by association with HuR and miR-200 family. Am. J. Cancer Res. 2018, 8, 981–992. [Google Scholar]
  44. Chen, J.; Liu, X.; Xu, Y.; Zhang, K.; Huang, J.; Pan, B.; Chen, D.; Cui, S.; Song, H.; Wang, R.; et al. TFAP2C-Activated MALAT1 Modulates the Chemoresistance of Docetaxel-Resistant Lung Adenocarcinoma Cells. Mol. Ther. Nucleic Acids 2019, 14, 567–582. [Google Scholar] [CrossRef] [Green Version]
  45. O’Brien, S.J.; Fiechter, C.; Burton, J.; Hallion, J.; Paas, M.; Patel, A.; Patel, A.; Rochet, A.; Scheurlen, K.; Gardner, S.; et al. Long non-coding RNA ZFAS1 is a major regulator of epithelial-mesenchymal transition through miR-200/ZEB1/E-cadherin, vimentin signaling in colon adenocarcinoma. Cell Death Discov. 2021, 7, 61. [Google Scholar] [CrossRef]
  46. Wu, X.; Fan, Y.; Liu, Y.; Shen, B.; Lu, H.; Ma, H. Long Non-Coding RNA CCAT2 Promotes the Development of Esophageal Squamous Cell Carcinoma by Inhibiting miR-200b to Upregulate the IGF2BP2/TK1 Axis. Front. Oncol. 2021, 11, 680642. [Google Scholar] [CrossRef]
  47. 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. Onco. Targets Ther. 2019, 12, 7561–7571. [Google Scholar] [CrossRef] [Green Version]
  48. Chen, X.; Li, Y.; Zuo, C.; Zhang, K.; Lei, X.; Wang, J.; Yang, Y.; Zhang, J.; Ma, K.; Wang, S.; et al. Long Non-Coding RNA H19 Regulates Glioma Cell Growth and Metastasis via miR-200a-Mediated CDK6 and ZEB1 Expression. Front. Oncol. 2021, 11, 757650. [Google Scholar] [CrossRef]
  49. Kim, S.L.; La, M.T.; Shin, M.W.; Kim, S.W.; Kim, H.K. A novel HDAC1 inhibitor, CBUD1001, exerts anticancer effects by modulating the apoptosis and EMT of colorectal cancer cells. Int. J. Oncol. 2020, 57, 1027–1038. [Google Scholar]
  50. Neves, R.; Scheel, C.; Weinhold, S.; Honisch, E.; Iwaniuk, K.M.; Trompeter, H.I.; Niederacher, D.; Wernet, P.; Santourlidis, S.; Uhrberg, M. Role of DNA methylation in miR-200c/141 cluster silencing in invasive breast cancer cells. BMC Res. Notes 2010, 3, 219. [Google Scholar] [CrossRef] [Green Version]
  51. Vrba, L.; Jensen, T.J.; Garbe, J.C.; Heimark, R.L.; Cress, A.E.; Dickinson, S.; Stampfer, M.R.; Futscher, B.W. Role for DNA methylation in the regulation of miR-200c and miR-141 expression in normal and cancer cells. PLoS ONE 2010, 5, e8697. [Google Scholar] [CrossRef] [PubMed]
  52. Wiklund, E.D.; Bramsen, J.B.; Hulf, T.; Dyrskjot, L.; Ramanathan, R.; Hansen, T.B.; Villadsen, S.B.; Gao, S.; Ostenfeld, M.S.; Borre, M.; et al. Coordinated epigenetic repression of the miR-200 family and miR-205 in invasive bladder cancer. Int. J. Cancer 2011, 128, 1327–1334. [Google Scholar] [CrossRef] [PubMed]
  53. Wee, E.J.; Peters, K.; Nair, S.S.; Hulf, T.; Stein, S.; Wagner, S.; Bailey, P.; Lee, S.Y.; Qu, W.J.; Brewster, B.; et al. Mapping the regulatory sequences controlling 93 breast cancer-associated miRNA genes leads to the identification of two functional promoters of the Hsa-mir-200b cluster, methylation of which is associated with metastasis or hormone receptor status in advanced breast cancer. Oncogene 2012, 31, 4182–4195. [Google Scholar] [PubMed] [Green Version]
  54. Li, B.L.; Lu, W.; Lu, C.; Qu, J.J.; Yang, T.T.; Yan, Q.; Wan, X.P. CpG island hypermethylation-associated silencing of microRNAs promotes human endometrial cancer. Cancer Cell Int. 2013, 13, 44. [Google Scholar] [CrossRef] [Green Version]
  55. Ning, X.; Shi, Z.; Liu, X.; Zhang, A.; Han, L.; Jiang, K.; Kang, C.; Zhang, Q. DNMT1 and EZH2 mediated methylation silences the microRNA-200b/a/429 gene and promotes tumor progression. Cancer Lett. 2015, 359, 198–205. [Google Scholar] [CrossRef]
  56. Pang, Y.; Liu, J.; Li, X.; Xiao, G.; Wang, H.; Yang, G.; Li, Y.; Tang, S.C.; Qin, S.; Du, N.; et al. MYC and DNMT3A-mediated DNA methylation represses microRNA-200b in triple negative breast cancer. J. Cell Mol. Med. 2018, 22, 6262–6274. [Google Scholar] [CrossRef] [Green Version]
  57. Kaowinn, S.; Kaewpiboon, C.; Koh, S.S.; Kramer, O.H.; Chung, Y.H. STAT1HDAC4 signaling induces epithelialmesenchymal transition and sphere formation of cancer cells overexpressing the oncogene, CUG2. Oncol. Rep. 2018, 40, 2619–2627. [Google Scholar]
  58. Chen, D.Q.; Pan, B.Z.; Huang, J.Y.; Zhang, K.; Cui, S.Y.; De, W.; Wang, R.; Chen, L.B. HDAC 1/4-mediated silencing of microRNA-200b promotes chemoresistance in human lung adenocarcinoma cells. Oncotarget 2014, 5, 3333–3349. [Google Scholar] [CrossRef] [Green Version]
  59. Xu, M.; Wang, S.; Wang, Y.; Wu, H.; Frank, J.A.; Zhang, Z.; Luo, J. Role of p38gamma MAPK in regulation of EMT and cancer stem cells. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 3605–3617. [Google Scholar] [CrossRef]
  60. Zhang, H.F.; Alshareef, A.; Wu, C.; Li, S.; Jiao, J.W.; Cao, H.H.; Lai, R.; Xu, L.Y.; Li, E.M. Loss of miR-200b promotes invasion via activating the Kindlin-2/integrin β1/AKT pathway in esophageal squamous cell carcinoma: An E-cadherin-independent mechanism. Oncotarget 2015, 6, 28949–28960. [Google Scholar] [CrossRef] [Green Version]
  61. Wang, Y.; Wei, C.; Yang, Y.; Luo, A.; Zhang, X.; Zheng, D.; Lu, X.; Zhang, K.; Duan, X.; Xu, X. Hepatocyte nuclear factor-1beta suppresses the stemness and migration of colorectal cancer cells through promoting miR-200b activity. Mol. Carcinog. 2020, 59, 989–999. [Google Scholar] [CrossRef] [PubMed]
  62. Zhu, J.N.; Jiang, L.; Jiang, J.H.; Yang, X.; Li, X.Y.; Zeng, J.X.; Shi, R.Y.; Shi, Y.; Pan, X.R.; Han, Z.P.; et al. Hepatocyte nuclear factor-1beta enhances the stemness of hepatocellular carcinoma cells through activation of the Notch pathway. Sci. Rep. 2017, 7, 4793. [Google Scholar] [CrossRef] [Green Version]
  63. Chen, X.; Wang, W.; Li, Y.; Huo, Y.; Zhang, H.; Feng, F.; Xi, W.; Zhang, T.; Gao, J.; Yang, F.; et al. MYSM1 inhibits human colorectal cancer tumorigenesis by activating miR-200 family members/CDH1 and blocking PI3K/AKT signaling. J. Exp. Clin. Cancer Res. 2021, 40, 341. [Google Scholar] [CrossRef] [PubMed]
  64. Ramirez-Moya, J.; Baker, A.R.; Slack, F.J.; Santisteban, P. ADAR1-mediated RNA editing is a novel oncogenic process in thyroid cancer and regulates miR-200 activity. Oncogene 2020, 39, 3738–3753. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Watson, K.L.; Yi, R.; Moorehead, R.A. Transgenic overexpression of the miR-200b/200a/429 cluster inhibits mammary tumor initiation. Transl. Oncol. 2021, 14, 101228. [Google Scholar] [CrossRef]
  66. Li, D.; Li, Q. MicroRNA-200b-3p restrains gastric cancer cell proliferation, migration, and invasion via C-X-C motif chemokine ligand 12/CXC chemokine receptor 7 axis. Bioengineered 2022, 13, 6509–6520. [Google Scholar] [CrossRef]
  67. Zeng, Y.; Li, B.; Liang, Y.; Reeves, P.M.; Qu, X.; Ran, C.; Liu, Q.; Callahan, M.V.; Sluder, A.E.; Gelfand, J.A.; et al. Dual blockade of CXCL12-CXCR4 and PD-1-PD-L1 pathways prolongs survival of ovarian tumor-bearing mice by prevention of immunosuppression in the tumor microenvironment. FASEB J. 2019, 33, 6596–6608. [Google Scholar] [CrossRef]
  68. Qiu, T.Y.; Huang, J.; Wang, L.P.; Zhu, B.S. Inhibition of miR-200b Promotes Angiogenesis in Endothelial Cells by Activating The Notch Pathway. Cell J. 2021, 23, 51–60. [Google Scholar]
  69. Xue, B.; Chuang, C.H.; Prosser, H.M.; Fuziwara, C.S.; Chan, C.; Sahasrabudhe, N.; Kuhn, M.; Wu, Y.; Chen, J.; Biton, A.; et al. miR-200 deficiency promotes lung cancer metastasis by activating Notch signaling in cancer-associated fibroblasts. Genes Dev. 2021, 35, 1109–1122. [Google Scholar] [CrossRef]
  70. Sharma, R.; Gogoi, G.; Saikia, S.; Sharma, A.; Kalita, D.J.; Sarma, A.; Limaye, A.M.; Gaur, M.K.; Bhattacharyya, J.; Jaganathan, B.G. BMP4 enhances anoikis resistance and chemoresistance of breast cancer cells through canonical BMP signaling. J. Cell Commun. Signal. 2022, 16, 191–205. [Google Scholar] [CrossRef]
  71. Tang, Q.; Li, M.; Chen, L.; Bi, F.; Xia, H. miR-200b/c targets the expression of RhoE and inhibits the proliferation and invasion of non-small cell lung cancer cells. Int. J. Oncol. 2018, 53, 1732–1742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Liu, J.; Wang, L.; Li, X. HMGB3 promotes the proliferation and metastasis of glioblastoma and is negatively regulated by miR-200b-3p and miR-200c-3p. Cell Biochem. Funct. 2018, 36, 357–365. [Google Scholar] [CrossRef] [PubMed]
  73. Cavallari, I.; Ciccarese, F.; Sharova, E.; Urso, L.; Raimondi, V.; Silic-Benussi, M.; D’Agostino, D.M.; Ciminale, V. The miR-200 Family of microRNAs: Fine Tuners of Epithelial-Mesenchymal Transition and Circulating Cancer Biomarkers. Cancers 2021, 13, 5874. [Google Scholar] [CrossRef]
  74. Yu, L.; Cao, C.; Li, X.; Zhang, M.; Gu, Q.; Gao, H.; Balic, J.J.; Xu, D.; Zhang, L.; Ying, L.; et al. Complete loss of miR-200 family induces EMT associated cellular senescence in gastric cancer. Oncogene 2022, 41, 26–36. [Google Scholar] [CrossRef]
  75. Gorecki, I.; Rak, B. The role of microRNAs in epithelial to mesenchymal transition and cancers; focusing on mir-200 family. Cancer Treat. Res. Commun. 2021, 28, 100385. [Google Scholar] [CrossRef] [PubMed]
  76. Watson, K.L.; Jones, R.A.; Bruce, A.; Moorehead, R.A. The miR-200b/200a/429 cluster prevents metastasis and induces dormancy in a murine claudin-low mammary tumor cell line. Exp. Cell Res. 2018, 369, 17–26. [Google Scholar] [CrossRef]
  77. 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]
  78. Gregory, P.A.; Bert, A.G.; Paterson, E.L.; Barry, S.C.; Tsykin, A.; Farshid, G.; Vadas, M.A.; Khew-Goodall, Y.; Goodall, G.J. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat. Cell Biol. 2008, 10, 593–601. [Google Scholar] [CrossRef]
  79. Kim, E.J.; Kim, J.S.; Lee, S.; Lee, H.; Yoon, J.S.; Hong, J.H.; Chun, S.H.; Sun, S.; Won, H.S.; Hong, S.A.; et al. QKI, a miR-200 target gene, suppresses epithelial-to-mesenchymal transition and tumor growth. Int. J. Cancer 2019, 145, 1585–1595. [Google Scholar] [CrossRef]
  80. Li, D.; Wang, H.; Song, H.; Xu, H.; Zhao, B.; Wu, C.; Hu, J.; Wu, T.; Xie, D.; Zhao, J.; et al. The microRNAs miR-200b-3p and miR-429–5p target the LIMK1/CFL1 pathway to inhibit growth and motility of breast cancer cells. Oncotarget 2017, 8, 85276–85289. [Google Scholar] [CrossRef] [Green Version]
  81. Du, J.; Zhu, Z.; Xu, L.; Chen, X.; Li, X.; Lan, T.; Li, W.; Yuan, K.; Zeng, Y. ARHGEF11 promotes proliferation and epithelial-mesenchymal transition of hepatocellular carcinoma through activation of beta-catenin pathway. Aging 2020, 12, 20235–20253. [Google Scholar] [CrossRef] [PubMed]
  82. Seo, J.; Ha, J.; Kang, E.; Cho, S. The role of epithelial-mesenchymal transition-regulating transcription factors in anti-cancer drug resistance. Arch. Pharm. Res. 2021, 44, 281–292. [Google Scholar] [CrossRef] [PubMed]
  83. Kasikci, E.; Aydemir, E.; Bayrak, O.F.; Sahin, F. Inhibition of Migration, Invasion and Drug Resistance of Pancreatic Adenocarcinoma Cells - Role of Snail, Slug and Twist and Small Molecule Inhibitors. Onco. Targets Ther. 2020, 13, 5763–5777. [Google Scholar] [CrossRef] [PubMed]
  84. Boukouris, A.E.; Zhang, Y.; Saleme, B.; Kinnaird, A.; Zhao, Y.Y.; Liu, Y.; Zervopoulos, S.D.; Das, S.K.; Mittal, R.D.; Haromy, A.; et al. A reversible metabolic stress-sensitive regulation of CRMP2A orchestrates EMT/stemness and increases metastatic potential in cancer. Cell Rep. 2022, 38, 110511. [Google Scholar] [CrossRef] [PubMed]
  85. Geretto, M.; Pulliero, A.; Rosano, C.; Zhabayeva, D.; Bersimbaev, R.; Izzotti, A. Resistance to cancer chemotherapeutic drugs is determined by pivotal microRNA regulators. Am. J. Cancer Res. 2017, 7, 1350–1371. [Google Scholar]
  86. Lee, H.; Kim, C.; Kang, H.; Tak, H.; Ahn, S.; Yoon, S.K.; Kuh, H.J.; Kim, W.; Lee, E.K. microRNA-200a-3p increases 5-fluorouracil resistance by regulating dual specificity phosphatase 6 expression. Exp. Mol. Med. 2017, 49, e327. [Google Scholar] [CrossRef] [Green Version]
  87. Kopp, F.; Oak, P.S.; Wagner, E.; Roidl, A. miR-200c sensitizes breast cancer cells to doxorubicin treatment by decreasing TrkB and Bmi1 expression. PLoS ONE 2012, 7, e50469. [Google Scholar] [CrossRef] [Green Version]
  88. Li, T.; Yu, Y.; Song, Y.; Li, X.; Lan, D.; Zhang, P.; Xiao, Y.; Xing, Y. Activation of BDNF/TrkB pathway promotes prostate cancer progression via induction of epithelial-mesenchymal transition and anoikis resistance. FASEB J. 2020, 34, 9087–9101. [Google Scholar] [CrossRef]
  89. Jin, H.F.; Wang, J.F.; Song, T.T.; Zhang, J.; Wang, L. MiR-200b Inhibits Tumor Growth and Chemoresistance via Targeting p70S6K1 in Lung Cancer. Front. Oncol. 2020, 10, 643. [Google Scholar] [CrossRef]
  90. Yu, G.; Jia, B.; Cheng, Y.; Zhou, L.; Qian, B.; Liu, Z.; Wang, Y. MicroRNA-429 sensitizes pancreatic cancer cells to gemcitabine through regulation of PDCD4. Am. J. Transl. Res. 2017, 9, 5048–5055. [Google Scholar]
  91. Gao, Y.; Zhang, W.; Liu, C.; Li, G. miR-200 affects tamoxifen resistance in breast cancer cells through regulation of MYB. Sci. Rep. 2019, 9, 18844. [Google Scholar] [CrossRef] [PubMed]
  92. Puhr, M.; Hoefer, J.; Schafer, G.; Erb, H.H.; Oh, S.J.; Klocker, H.; Heidegger, I.; Neuwirt, H.; Culig, Z. Epithelial-to-mesenchymal transition leads to docetaxel resistance in prostate cancer and is mediated by reduced expression of miR-200c and miR-205. Am. J. Pathol. 2012, 181, 2188–2201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Yang, X.; Hu, Q.; Hu, L.X.; Lin, X.R.; Liu, J.Q.; Lin, X.; Dinglin, X.X.; Zeng, J.Y.; Hu, H.; Luo, M.L.; et al. miR-200b regulates epithelial-mesenchymal transition of chemo-resistant breast cancer cells by targeting FN1. Discov. Med. 2017, 24, 75–85. [Google Scholar] [PubMed]
  94. Liu, J.; Zhang, X.; Huang, Y.; Zhang, Q.; Zhou, J.; Zhang, X.; Wang, X. miR-200b and miR-200c co-contribute to the cisplatin sensitivity of ovarian cancer cells by targeting DNA methyltransferases. Oncol. Lett. 2019, 17, 1453–1460. [Google Scholar] [CrossRef] [Green Version]
  95. Shindo, T.; Niinuma, T.; Nishiyama, N.; Shinkai, N.; Kitajima, H.; Kai, M.; Maruyama, R.; Tokino, T.; Masumori, N.; Suzuki, H. Epigenetic silencing of miR-200b is associated with cisplatin resistance in bladder cancer. Oncotarget 2018, 9, 24457–24469. [Google Scholar] [CrossRef] [Green Version]
  96. Zeng, X.; Qu, X.; Zhao, C.; Xu, L.; Hou, K.; Liu, Y.; Zhang, N.; Feng, J.; Shi, S.; Zhang, L.; et al. FEN1 mediates miR-200a methylation and promotes breast cancer cell growth via MET and EGFR signaling. FASEB J. 2019, 33, 10717–10730. [Google Scholar] [CrossRef]
  97. Aita, V.M.; Liang, X.H.; Murty, V.V.; Pincus, D.L.; Yu, W.; Cayanis, E.; Kalachikov, S.; Gilliam, T.C.; Levine, B. Cloning and genomic organization of beclin 1, a candidate tumor suppressor gene on chromosome 17q21. Genomics 1999, 59, 59–65. [Google Scholar] [CrossRef]
  98. Chen, Z.; Liu, S.; Xie, P.; Zhang, B.; Yu, M.; Yan, J.; Jin, L.; Zhang, W.; Zhou, B.; Li, X.; et al. Tumor-derived PD1 and PD-L1 could promote hepatocellular carcinoma growth through autophagy induction in vitro. Biochem. Biophys. Res. Commun. 2022, 605, 82–89. [Google Scholar] [CrossRef]
  99. Lai, M.; Liu, L.; Zhu, L.; Feng, W.; Luo, J.; Liu, Y.; Deng, S. Triptolide reverses epithelial-mesenchymal transition in glioma cells via inducing autophagy. Ann. Transl. Med. 2021, 9, 1304. [Google Scholar] [CrossRef]
  100. Hu, Y.L.; Jahangiri, A.; Delay, M.; Aghi, M.K. Tumor cell autophagy as an adaptive response mediating resistance to treatments such as antiangiogenic therapy. Cancer Res. 2012, 72, 4294–4299. [Google Scholar] [CrossRef] [Green Version]
  101. Fedorova, O.; Daks, A.; Parfenyev, S.; Shuvalov, O.; Netsvetay, S.; Vasileva, J.; Gudovich, A.; Golotin, V.; Semenov, O.; Petukhov, A.; et al. Zeb1-mediated autophagy enhances resistance of breast cancer cells to genotoxic drugs. Biochem. Biophys. Res. Commun. 2022, 589, 29–34. [Google Scholar] [CrossRef] [PubMed]
  102. Yeon, M.; Kim, Y.; Pathak, D.; Kwon, E.; Kim, D.Y.; Jeong, M.S.; Jung, H.S.; Jeoung, D. The CAGE-MiR-181b-5p-S1PR1 Axis Regulates Anticancer Drug Resistance and Autophagy in Gastric Cancer Cells. Front. Cell Dev. Biol. 2021, 9, 666387. [Google Scholar] [CrossRef] [PubMed]
  103. Bi, J.; Zhang, Y.; Malmrose, P.K.; Losh, H.A.; Newtson, A.M.; Devor, E.J.; Thiel, K.W.; Leslie, K.K. Blocking autophagy overcomes resistance to dual histone deacetylase and proteasome inhibition in gynecologic cancer. Cell Death Dis. 2022, 13, 59. [Google Scholar] [CrossRef] [PubMed]
  104. Pei, L.; Kong, Y.; Shao, C.; Yue, X.; Wang, Z.; Zhang, N. Heme oxygenase-1 induction mediates chemoresistance of breast cancer cells to pharmorubicin by promoting autophagy via PI3K/Akt pathway. J. Cell Mol. Med. 2018, 22, 5311–5321. [Google Scholar] [CrossRef] [PubMed]
  105. Zhang, J.; Fan, J.; Zeng, X.; Nie, M.; Chen, W.; Wang, Y.; Luan, J.; Zhu, Z.; Chang, X.; Ju, D.; et al. Targeting the autophagy promoted antitumor effect of T-DM1 on HER2-positive gastric cancer. Cell Death Dis. 2021, 12, 288. [Google Scholar] [CrossRef] [PubMed]
  106. Li, L.; Wang, Y.; Jiao, L.; Lin, C.; Lu, C.; Zhang, K.; Hu, C.; Ye, J.; Zhang, D.; Wu, H.; et al. Protective autophagy decreases osimertinib cytotoxicity through regulation of stem cell-like properties in lung cancer. Cancer Lett. 2019, 452, 191–202. [Google Scholar] [CrossRef] [PubMed]
  107. Pan, B.; Feng, B.; Chen, Y.; Huang, G.; Wang, R.; Chen, L.; Song, H. MiR-200b regulates autophagy associated with chemoresistance in human lung adenocarcinoma. Oncotarget 2015, 6, 32805–32820. [Google Scholar] [CrossRef]
  108. Kim, H.; Kim, Y.; Jeoung, D. DDX53 Promotes Cancer Stem Cell-Like Properties and Autophagy. Mol. Cells 2017, 40, 54–65. [Google Scholar] [CrossRef] [Green Version]
  109. Alsadi, N.; Mallet, J.F.; Matar, C. miRNA-200b Signature in the Prevention of Skin Cancer Stem Cells by Polyphenol-enriched Blueberry Preparation. J. Cancer Prev. 2021, 26, 162–173. [Google Scholar] [CrossRef]
  110. Rankovic, B.; Zidar, N.; Zlajpah, M.; Bostjancic, E. Epithelial-Mesenchymal Transition-Related MicroRNAs and Their Target Genes in Colorectal Cancerogenesis. J. Clin. Med. 2019, 8, 1603. [Google Scholar] [CrossRef] [Green Version]
  111. Wang, X.; He, R.; Geng, L.; Yuan, J.; Fan, H. Ginsenoside Rg3 Alleviates Cisplatin Resistance of Gastric Cancer Cells Through Inhibiting SOX2 and the PI3K/Akt/mTOR Signaling Axis by Up-Regulating miR-429. Front. Genet. 2022, 13, 823182. [Google Scholar] [CrossRef] [PubMed]
  112. Kim, Y.; Park, H.; Park, D.; Lee, Y.S.; Choe, J.; Hahn, J.H.; Lee, H.; Kim, Y.M.; Jeoung, D. Cancer/testis antigen CAGE exerts negative regulation on p53 expression through HDAC2 and confers resistance to anti-cancer drugs. J. Biol. Chem. 2010, 285, 25957–25968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Yeon, M.; Byun, J.; Kim, H.; Kim, M.; Jung, H.S.; Jeon, D.; Kim, Y.; Jeoung, D. CAGE Binds to Beclin1, Regulates Autophagic Flux and CAGE-Derived Peptide Confers Sensitivity to Anti-cancer Drugs in Non-small Cell Lung Cancer Cells. Front. Oncol. 2018, 8, 599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Kim, Y.; Park, D.; Kim, H.; Choi, M.; Lee, H.; Lee, Y.S.; Choe, J.; Kim, Y.M.; Jeoung, D. miR-200b and cancer/testis antigen CAGE form a feedback loop to regulate the invasion and tumorigenic and angiogenic responses of a cancer cell line to microtubule-targeting drugs. J. Biol. Chem. 2013, 288, 36502–36518. [Google Scholar] [CrossRef] [Green Version]
  115. Gyvyte, U.; Lukosevicius, R.; Inciuraite, R.; Streleckiene, G.; Gudoityte, G.; Bekampyte, J.; Valentini, S.; Salteniene, V.; Ruzgys, P.; Satkauskas, S.; et al. The Role of miR-375-3p and miR-200b-3p in Gastrointestinal Stromal Tumors. Int J. Mol Sci 2020, 21, 5151. [Google Scholar] [CrossRef]
  116. Kim, Y.; Kim, H.; Park, D.; Han, M.; Lee, H.; Lee, Y.S.; Choe, J.; Kim, Y.M.; Jeoung, D. miR-217 and CAGE form feedback loop and regulates the response to anti-cancer drugs through EGFR and HER2. Oncotarget 2016, 7, 10297–10321. [Google Scholar] [CrossRef] [Green Version]
  117. Kim, H.; Kim, Y.; Goh, H.; Jeoung, D. Histone Deacetylase-3/CAGE Axis Targets EGFR Signaling and Regulates the Response to Anti-Cancer Drugs. Mol. Cells 2016, 39, 229–241. [Google Scholar]
  118. Kim, Y.; Kim, H.; Park, H.; Park, D.; Lee, H.; Lee, Y.S.; Choe, J.; Kim, Y.M.; Jeoung, D. miR-326-histone deacetylase-3 feedback loop regulates the invasion and tumorigenic and angiogenic response to anti-cancer drugs. J. Biol. Chem. 2014, 289, 28019–28039. [Google Scholar] [CrossRef] [Green Version]
  119. Kwon, Y.; Kim, Y.; Jung, H.S.; Jeoung, D. Role of HDAC3-miRNA-CAGE Network in Anti-Cancer Drug-Resistance. Int. J. Mol. Sci 2018, 20, 51. [Google Scholar] [CrossRef] [Green Version]
  120. Cochrane, D.R.; Spoelstra, N.S.; Howe, E.N.; Nordeen, S.K.; Richer, J.K. MicroRNA-200c mitigates invasiveness and restores sensitivity to microtubule-targeting chemotherapeutic agents. Mol. Cancer Ther. 2009, 8, 1055–1066. [Google Scholar] [CrossRef] [Green Version]
  121. Shadbad, M.A.; Asadzadeh, Z.; Derakhshani, A.; Hosseinkhani, N.; Mokhtarzadeh, A.; Baghbanzadeh, A.; Hajiasgharzadeh, K.; Brunetti, O.; Argentiero, A.; Racanelli, V.; et al. A scoping review on the potentiality of PD-L1-inhibiting microRNAs in treating colorectal cancer: Toward single-cell sequencing-guided biocompatible-based delivery. Biomed. Pharmacother. 2021, 143, 112213. [Google Scholar] [CrossRef] [PubMed]
  122. Mu, L.; Wang, Y.; Su, H.; Lin, Y.; Sui, W.; Yu, X.; Lv, Z. HIF1A-AS2 Promotes the Proliferation and Metastasis of Gastric Cancer Cells Through miR-429/PD-L1 Axis. Dig. Dis. Sci. 2021, 66, 4314–4325. [Google Scholar] [CrossRef] [PubMed]
  123. Wei, S.; Wang, K.; Huang, X.; Zhao, Z.; Zhao, Z. LncRNA MALAT1 contributes to non-small cell lung cancer progression via modulating miR-200a-3p/programmed death-ligand 1 axis. Int. J. Immunopathol. Pharmacol. 2019, 33, 2058738419859699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Chen, C.; Li, S.; Xue, J.; Qi, M.; Liu, X.; Huang, Y.; Hu, J.; Dong, H.; Ling, K. PD-L1 tumor-intrinsic signaling and its therapeutic implication in triple-negative breast cancer. JCI Insight 2021, 6, e131458. [Google Scholar] [CrossRef]
  125. Lv, J.; Guo, T.; Qu, X.; Che, X.; Li, C.; Wang, S.; Gong, J.; Wu, P.; Liu, Y.; Liu, Y.; et al. PD-L1 Under Regulation of miR-429 Influences the Sensitivity of Gastric Cancer Cells to TRAIL by Binding of EGFR. Front. Oncol. 2020, 10, 1067. [Google Scholar] [CrossRef]
  126. Kim, Y.; Kim, H.; Park, D.; Lee, H.; Lee, Y.S.; Choe, J.; Kim, Y.M.; Jeon, D.; Jeoung, D. The pentapeptide Gly-Thr-Gly-Lys-Thr confers sensitivity to anti-cancer drugs by inhibition of CAGE binding to GSK3beta and decreasing the expression of cyclinD1. Oncotarget 2017, 8, 13632–13651. [Google Scholar] [CrossRef] [Green Version]
  127. Liu, Z.; Altwegg, K.A.; Liu, J.; Weintraub, S.T.; Chen, Y.; Lai, Z.; Sareddy, G.R.; Viswanadhapalli, S.; Vadlamudi, R.K. Global Genomic and Proteomic Analysis Identified Critical Pathways Modulated by Proto-Oncogene PELP1 in TNBC. Cancers 2022, 14, 930. [Google Scholar] [CrossRef]
  128. Zhang, D.; Dai, J.; Pan, Y.; Wang, X.; Qiao, J.; Sasano, H.; Zhao, B.; McNamara, K.M.; Guan, X.; Liu, L.; et al. Overexpression of PELP1 in Lung Adenocarcinoma Promoted E2 Induced Proliferation, Migration and Invasion of the Tumor Cells and Predicted a Worse Outcome of the Patients. Pathol. Oncol. Res. 2021, 27, 582443. [Google Scholar] [CrossRef]
  129. Roy, S.S.; Gonugunta, V.K.; Bandyopadhyay, A.; Rao, M.K.; Goodall, G.J.; Sun, L.Z.; Tekmal, R.R.; Vadlamudi, R.K. Significance of PELP1/HDAC2/miR-200 regulatory network in EMT and metastasis of breast cancer. Oncogene 2014, 33, 3707–3716. [Google Scholar] [CrossRef] [Green Version]
  130. Zhu, J.; Wang, L.; Liu, F.; Pan, J.; Yao, Z.; Lin, Y.; Yang, Y.; Xiong, X.; Li, K.; Yang, Y.; et al. Targeting PELP1 Attenuates Angiogenesis and Enhances Chemotherapy Efficiency in Colorectal Cancer. Cancers 2022, 14, 383. [Google Scholar] [CrossRef]
  131. Krishnan, S.R.; Nair, B.C.; Sareddy, G.R.; Roy, S.S.; Natarajan, M.; Suzuki, T.; Peng, Y.; Raj, G.; Vadlamudi, R.K. Novel role of PELP1 in regulating chemotherapy response in mutant p53-expressing triple negative breast cancer cells. Breast Cancer Res. Treat. 2015, 150, 487–499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Wang, W.; Feng, C.; Zhang, W.; Long, Y.; Fa, X. The epigenetic silencing of microRNA-433 facilitates the malignant phenotypes of non-small cell lung cancer by targeting CREB1. Am. J. Transl. Res. 2021, 13, 12302–12317. [Google Scholar] [PubMed]
  133. Stavast, C.J.; van Zuijen, I.; Karkoulia, E.; Ozcelik, A.; van Hoven-Beijen, A.; Leon, L.G.; Voerman, J.S.A.; Janssen, G.M.C.; van Veelen, P.A.; Burocziova, M.; et al. The tumor suppressor MIR139 is silenced by POLR2M to promote AML oncogenesis. Leukemia 2022, 36, 687–700. [Google Scholar] [CrossRef] [PubMed]
  134. Zhang, M.; Yang, L.; Hou, L.; Tang, X. LncRNA SNHG1 promotes tumor progression and cisplatin resistance through epigenetically silencing miR-381 in breast cancer. Bioengineered 2021, 12, 9239–9250. [Google Scholar] [CrossRef]
  135. Farah, E.; Zhang, Z.; Utturkar, S.M.; Liu, J.; Ratliff, T.L.; Liu, X. Targeting DNMTs to Overcome Enzalutamide Resistance in Prostate Cancer. Mol. Cancer Ther. 2022, 21, 193–205. [Google Scholar] [CrossRef]
  136. Liu, Y.; Bai, Z.; Chai, D.; Gao, Y.; Li, T.; Ma, Y.; Li, J. DNA methyltransferase 1 inhibits microRNA-497 and elevates GPRC5A expression to promote chemotherapy resistance and metastasis in breast cancer. Cancer Cell Int. 2022, 22, 112. [Google Scholar] [CrossRef]
  137. Sato, H.; Shien, K.; Tomida, S.; Okayasu, K.; Suzawa, K.; Hashida, S.; Torigoe, H.; Watanabe, M.; Yamamoto, H.; Soh, J.; et al. Targeting the miR-200c/LIN28B axis in acquired EGFR-TKI resistance non-small cell lung cancer cells harboring EMT features. Sci. Rep. 2017, 7, 40847. [Google Scholar] [CrossRef]
  138. Cao, Q.; You, X.; Xu, L.; Wang, L.; Chen, Y. PAQR3 suppresses the growth of non-small cell lung cancer cells via modulation of EGFR-mediated autophagy. Autophagy 2020, 16, 1236–1247. [Google Scholar] [CrossRef]
  139. Dent, P.; Booth, L.; Poklepovic, A.; Von Hoff, D.; Martinez, J.; Zhou, Y.; Hancock, J.F. Osimertinib-resistant NSCLC cells activate ERBB2 and YAP/TAZ and are killed by neratinib. Biochem. Pharmacol. 2021, 190, 114642. [Google Scholar] [CrossRef]
  140. 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]
  141. Della Corte, C.M.; Malapelle, U.; Vigliar, E.; Pepe, F.; Troncone, G.; Ciaramella, V.; Troiani, T.; Martinelli, E.; Belli, V.; Ciardiello, F.; et al. Efficacy of continuous EGFR-inhibition and role of Hedgehog in EGFR acquired resistance in human lung cancer cells with activating mutation of EGFR. Oncotarget 2017, 8, 23020–23032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Ahmad, A.; Maitah, M.Y.; Ginnebaugh, K.R.; Li, Y.; Bao, B.; Gadgeel, S.M.; Sarkar, F.H. Inhibition of Hedgehog signaling sensitizes NSCLC cells to standard therapies through modulation of EMT-regulating miRNAs. J. Hematol. Oncol. 2013, 6, 77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Ryoo, S.R.; Yim, Y.; Kim, Y.K.; Park, I.S.; Na, H.K.; Lee, J.; Jang, H.; Won, C.; Hong, S.; Kim, S.Y.; et al. High-throughput chemical screening to discover new modulators of microRNA expression in living cells by using graphene-based biosensor. Sci. Rep. 2018, 8, 11413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Slaby, O.; Laga, R.; Sedlacek, O. Therapeutic targeting of non-coding RNAs in cancer. Biochem. J. 2017, 474, 4219–4251. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Biogenesis of miRNAs. Transcription to primary miRNA is catalyzed by RNA polymerase II or III. Primary miRNA is processed to precursor miRNA (~70 nucleotides) with a stem and loop structure by RNase Drosha and DiGeorge Critical Region 8 (DGCR8). Precursor miRNA is transported into the cytoplasm by the Exportin 5-Ran-GTP complex. Precursor miRNA is further processed to mature miRNA (~22 nucleotides) by RNase DICER. Double-stranded miRNA is unwound by helicase, and guide miRNA is incorporated into the RNA-induced silencing complex (RISC). Passenger miRNA is degraded. Perfect base-pairing between seed and target mRNA leads to the degradation of the target mRNA. The degradation of the target mRNA involves deadenylation, decapping, and 5′–3′ exonuclease activity. Imperfect base-pairing leads to translational inhibition. DICER1, a master regulator of miRNA biogenesis, promotes autophagy, anti-cancer drug resistance, autophagy, and cancer development. TRBP denotes TAR-RNA binding protein, HMT denotes histone methyltransferase, ↑ denotes positive regulation, ↓ denotes negative regulation. The arrows with solid lines indicate each step of miRNA biogenesis.
Figure 1. Biogenesis of miRNAs. Transcription to primary miRNA is catalyzed by RNA polymerase II or III. Primary miRNA is processed to precursor miRNA (~70 nucleotides) with a stem and loop structure by RNase Drosha and DiGeorge Critical Region 8 (DGCR8). Precursor miRNA is transported into the cytoplasm by the Exportin 5-Ran-GTP complex. Precursor miRNA is further processed to mature miRNA (~22 nucleotides) by RNase DICER. Double-stranded miRNA is unwound by helicase, and guide miRNA is incorporated into the RNA-induced silencing complex (RISC). Passenger miRNA is degraded. Perfect base-pairing between seed and target mRNA leads to the degradation of the target mRNA. The degradation of the target mRNA involves deadenylation, decapping, and 5′–3′ exonuclease activity. Imperfect base-pairing leads to translational inhibition. DICER1, a master regulator of miRNA biogenesis, promotes autophagy, anti-cancer drug resistance, autophagy, and cancer development. TRBP denotes TAR-RNA binding protein, HMT denotes histone methyltransferase, ↑ denotes positive regulation, ↓ denotes negative regulation. The arrows with solid lines indicate each step of miRNA biogenesis.
Ijms 23 05881 g001
Figure 2. The chromosomal location and sequence of the miR-200 family. (A) The miR-200 family is divided into two clusters. Cluster 1 (human chromosome 1) contains miR-200a, miR-200b, and miR-429, and cluster 2 (human chromosome 10) contains miR-141and miR-200c. (B) The miR-200 family is divided into two groups based on the seed sequence. These two groups show a difference in the third nucleotide of the seed sequence. The seed sequences are underlined.
Figure 2. The chromosomal location and sequence of the miR-200 family. (A) The miR-200 family is divided into two clusters. Cluster 1 (human chromosome 1) contains miR-200a, miR-200b, and miR-429, and cluster 2 (human chromosome 10) contains miR-141and miR-200c. (B) The miR-200 family is divided into two groups based on the seed sequence. These two groups show a difference in the third nucleotide of the seed sequence. The seed sequences are underlined.
Ijms 23 05881 g002
Figure 3. Regulation of miR-200 family expression and targets of the miR-200 family. (A) Promoter analysis revealed potential binding sites for various transcription factors in the promoter sequences of the miR-200 family. → denotes transcription activation. (B) Various factors regulating the expression of the miR-200 family. The T-bar arrows denote negative regulation, ↑ denotes positive regulation. (C) Targets of the miR-200 family and roles of these targets in anti-cancer drug resistance. The T-bar arrows denote negative regulation.
Figure 3. Regulation of miR-200 family expression and targets of the miR-200 family. (A) Promoter analysis revealed potential binding sites for various transcription factors in the promoter sequences of the miR-200 family. → denotes transcription activation. (B) Various factors regulating the expression of the miR-200 family. The T-bar arrows denote negative regulation, ↑ denotes positive regulation. (C) Targets of the miR-200 family and roles of these targets in anti-cancer drug resistance. The T-bar arrows denote negative regulation.
Ijms 23 05881 g003
Figure 4. The proposed role of the HDAC3-CAGE-miR-200b-PD-L1 network in anti-cancer drug resistance. (A) CAGE binds to HDAC2 in anti-cancer drug-resistant melanoma cells. CAGE-HDAC2 complex binds to the promoter sequences of p53 to repress p53 expression. The decreased expression of p53 leads to resistance to anti-cancer drugs. ↓ denotes decreased expression. ↑ denotes positive regulation. The T-bar arrows denote transcription inhibition. The potential binding of the miR-200 family to the 3′ UTR of CAGE is shown. (B) HDAC3 forms a positive feedback loop with miR-200b and a negative feedback loop with miR-326. HDAC3 binds to the promoter sequences of CAGE, which decreases the expression of CAGE and pEGFRY845. miR-200b forms a negative feedback loop with CAGE. The binding of CAGE to the EGFR leads to anti-cancer drug resistance and the enhanced metastatic potential of cancer cells. ↓ denotes decreased expression. ↑ denotes positive regulation. Both side T-bars denote negative feedback. (C) The miR-200 family decreases the expression of PD-L1. The binding of the miR-200 family to the 3′ UTR of PD-L1 is shown. PD-L1 induces EMT by preventing GSK3β from degrading SNAIL. miR-429 decreases the expression of PD-L1 to enhance sensitivity to TRAIL. CAGE binds to GSK3β to inactivate GSK3β. The inactivation of GSK3β leads to increased cyclin D1 expression and promotes anti-cancer drug resistance. CAGE-derived peptide (269GTGKT273) inhibits the binding of CAGE to GSK3β to enhance chemosensitivity. GTGKT also inhibits the binding of CAGE to EGFR. ↑ denotes positive regulation. The T-bar arrows denote negative regulation.
Figure 4. The proposed role of the HDAC3-CAGE-miR-200b-PD-L1 network in anti-cancer drug resistance. (A) CAGE binds to HDAC2 in anti-cancer drug-resistant melanoma cells. CAGE-HDAC2 complex binds to the promoter sequences of p53 to repress p53 expression. The decreased expression of p53 leads to resistance to anti-cancer drugs. ↓ denotes decreased expression. ↑ denotes positive regulation. The T-bar arrows denote transcription inhibition. The potential binding of the miR-200 family to the 3′ UTR of CAGE is shown. (B) HDAC3 forms a positive feedback loop with miR-200b and a negative feedback loop with miR-326. HDAC3 binds to the promoter sequences of CAGE, which decreases the expression of CAGE and pEGFRY845. miR-200b forms a negative feedback loop with CAGE. The binding of CAGE to the EGFR leads to anti-cancer drug resistance and the enhanced metastatic potential of cancer cells. ↓ denotes decreased expression. ↑ denotes positive regulation. Both side T-bars denote negative feedback. (C) The miR-200 family decreases the expression of PD-L1. The binding of the miR-200 family to the 3′ UTR of PD-L1 is shown. PD-L1 induces EMT by preventing GSK3β from degrading SNAIL. miR-429 decreases the expression of PD-L1 to enhance sensitivity to TRAIL. CAGE binds to GSK3β to inactivate GSK3β. The inactivation of GSK3β leads to increased cyclin D1 expression and promotes anti-cancer drug resistance. CAGE-derived peptide (269GTGKT273) inhibits the binding of CAGE to GSK3β to enhance chemosensitivity. GTGKT also inhibits the binding of CAGE to EGFR. ↑ denotes positive regulation. The T-bar arrows denote negative regulation.
Ijms 23 05881 g004
Table 1. Potential value of miR-200 family miRNAs as diagnostic and prognostic markers. ↑ denotes increased expression and ↓ denotes decreased expression.
Table 1. Potential value of miR-200 family miRNAs as diagnostic and prognostic markers. ↑ denotes increased expression and ↓ denotes decreased expression.
Tumor TypeSample Type/SizeExpression of miR-200sApplicationReference
Renal cell carcinomaEighty tumor tissues and adjacent tissues↓ miR-200b in tumor tissuesPrediction of metastasis, prognosis[18]
GliomaEighty-nine glioma tissues and 41 non-tumor tissues↓ miR-200b in tumor tissuesDiagnosis and prognosis[19]
Clear cell renal cell carcinomaNormal tissues (n = 23), primary tumor tissues (n = 194), metastatic tumor tissues (n= 10)↓ miR-141, ↓ miR-200b in metastatic tumor tissues
High levels of miR-200b and miR-200c → longer progression-free survival
Diagnosis and prognosis[20]
Gastric cancerSixty tumor tissues and normal control tissuesHigh level of mR-200b → high overall survivalDiagnosis and prognosis[21]
Hepatocellular carcinoma371 tumor tissues↓ miR-200b ↑ HMG3B in tumor tissues
—miR-200b targets HMG3B
Diagnosis[22]
Bladder cancerTumor tissues from 1150 patientsHigh level of the miR-200 family → better prognosisPrognosis[23]
Non-small cell lung cancer (NSCLC)Tumor tissues from 60 NSCLC patientsHigh level of miR-200b → high progression-free survivalPrognosis in response to systemic immune therapy[24]
Hepatocellular carcinomaWhole blood from thirty patients and normal controls↓ miR-141 ↓ miR-200a in serum of cancer patientsDiagnosis[27]
NSCLCFifty cancer patients and 30 normal controls↓ miR-200b in serum of cancer patientsDiagnosis[28]
Breast cancerWhole blood from early (137) and metastatic patients (110)High levels of miR-200b and miR-200c in metastatic patients compared to early patients
High level of miR-200b → shorter overall survival
Diagnosis and prognosis[29]
Colon cancerPlasma and exosomes from fifty resected patientsLow levels of miR-141 and miR-200c → longer overall survivalDiagnosis[30]
Breast cancerSerum (n = 47) from metastatic cancer patientsHigh levels of the miR-200 family (miR-141, miR-200a, miR-200b, miR-429) → reduction in overall survival and progression-free survivalPrognosis[31]
Table 2. Role of miR-200 family miRNAs in anti-cancer drug resistance and the mechanisms associated with it.
Table 2. Role of miR-200 family miRNAs in anti-cancer drug resistance and the mechanisms associated with it.
miR-200 FamilyMechanismTargetEnhances Sensitivity to CancerReference
miR-200aSignalingDUSP65-FUHepatocellular carcinoma[86]
miR-200bmTOR pathwayP70S6K1CisplatinLung cancer[89]
FAK/Src signalingFN1DoxorubicinBreast cancer[93]
AutophagyATG-12DocetaxelLung cancer[107]
AutophagyATG-5Cisplatin DocetaxelBreast cancer[108]
Autophagy/cancer stemnessCAGEMicrotubule-targeting agentsMelanoma[112]
SignalingHh signalingErlotinibLung cancer[142]
miR-200b/-200cEMTC-MYBTamoxifenBreast cancer[91]
Epigenetic modificationDNMT1CisplatinOvarian cancer[94]
miR-200cSignalingTrkBDoxorubicinBreast cancer[87]
EMTE-cadherin, SNAILDocetaxelProstate cancer[92]
EMTTUBB3, ZEB1/ZEB2Microtubule-targeting agentsOvarian, Breast cancer[120]
SignalingPI3K/AktGefitinibLung cancer[137]
miR-200c/-141EMTE-cadherin, VimentinOxaliplatinOvarian cancer[95]
miR-429Cell deathPDCD4GemcitabinePancreatic cancer[90]
Cancer stemnessSOX2CisplatinGastric cancer[111]
Immune suppressionPD-L1TRAILGastric cancer[125]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jo, H.; Shim, K.; Jeoung, D. Potential of the miR-200 Family as a Target for Developing Anti-Cancer Therapeutics. Int. J. Mol. Sci. 2022, 23, 5881. https://doi.org/10.3390/ijms23115881

AMA Style

Jo H, Shim K, Jeoung D. Potential of the miR-200 Family as a Target for Developing Anti-Cancer Therapeutics. International Journal of Molecular Sciences. 2022; 23(11):5881. https://doi.org/10.3390/ijms23115881

Chicago/Turabian Style

Jo, Hyein, Kyeonghee Shim, and Dooil Jeoung. 2022. "Potential of the miR-200 Family as a Target for Developing Anti-Cancer Therapeutics" International Journal of Molecular Sciences 23, no. 11: 5881. https://doi.org/10.3390/ijms23115881

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