Skip to main content
Molecular Therapy. Nucleic Acids logoLink to Molecular Therapy. Nucleic Acids
. 2021 Oct 8;26:892–926. doi: 10.1016/j.omtn.2021.10.004

The role of non-coding RNAs in chemotherapy for gastrointestinal cancers

Fatemeh Dashti 1,2,10, Seyed Mohammad Ali Mirazimi 1,2,10, Nikta Rabiei 3, Reza Fathazam 3, Negin Rabiei 3, Haleh Piroozmand 4, Massoud Vosough 5, Neda Rahimian 6,, Michael R Hamblin 7,8,∗∗, Hamed Mirzaei 9,∗∗∗
PMCID: PMC8551789  PMID: 34760336

Abstract

Gastrointestinal (GI) cancers, including colorectal, gastric, hepatic, esophageal, and pancreatic tumors, are responsible for large numbers of deaths around the world. Chemotherapy is the most common approach used to treat advanced GI cancer. However, chemoresistance has emerged as a critical challenge that prevents successful tumor elimination, leading to metastasis and recurrence. Chemoresistance mechanisms are complex, and many factors and pathways are involved. Among these factors, non-coding RNAs (ncRNAs) are critical regulators of GI tumor development and subsequently can induce resistance to chemotherapy. This occurs because ncRNAs can target multiple signaling pathways, affect downstream genes, and modulate proliferation, apoptosis, tumor cell migration, and autophagy. ncRNAs can also induce cancer stem cell features and affect the epithelial-mesenchymal transition. Thus, ncRNAs could possibly act as new targets in chemotherapy combinations to treat GI cancer and to predict treatment response.

Keywords: gastrointestinal cancers, non-coding RNAs, response to chemotherapy

Graphical abstract

graphic file with name fx1.jpg


ncRNAs can also induce cancer stem cell features and affect the epithelial-mesenchymal transition. Thus, ncRNAs could be possible targets in new chemotherapy combinations to treat GI cancer and to predict treatment response.

Introduction

Gastrointestinal (GI) cancers are a major cause of death worldwide, and they impose a tremendous financial burden on many countries. Some therapeutic approaches for inoperable GI cancer, such as radiotherapy and chemotherapy, have been slowly developed along with advances in technology, and they have been shown to be partly effective in patients. Today, chemotherapy is the first-line standard procedure for most patients with cancer, particularly where surgery is impractical. There are already hundreds of chemotherapy drugs that have been approved to treat cancer, along with more new drugs being developed. The mechanisms of these anti-cancer drugs are different; while many aim to inhibit elements of the basic cellular function, others aim to kill the proliferating cells. They include compounds such as DNA-modifying agents (cisplatin [DDP]), molecular drugs targeting hormones such as estrogen receptor (ER) blockers (tamoxifen [TAM]), drugs that interfere with microtubules (taxol), and drugs that interfere with metabolic activity (methotrexate [MTX]).1 Radiotherapy is often used as an adjuvant therapy and is used in the treatment of nearly 50% of patients with cancer around the world. The treatment principles can be divided into two broad groups: (1) tumor cells have a restricted capability to repair damaged DNA; and (2) cancer cells are usually rapidly dividing and show higher sensitivity to chemotherapy and radiation compared to slower dividing normal cells.2 Radiotherapy and chemotherapy have both been shown to improve the overall survival (OS) and/or progression-free survival of patients. However, a problem is very likely to gradually appear; that is, cancer cells usually develop resistance to cytotoxic drugs or radiation. Both acquired and intrinsic resistance can dramatically reduce the therapeutic efficacy, leading to poor prognosis, recurrence, and eventual tumor metastasis.3 Nowadays, the discovery of approaches to overcome these challenges is an urgent need.

It has been reported that non-coding DNAs account for 95% of the DNA sequences within the human genome. A majority of the non-coding DNAs have been shown to be transcribed into more than 10,000 different functional non-coding RNAs (ncRNAs), including microRNAs (miRNAs), small interfering RNAs (siRNAs), antisense RNAs (asRNAs), and long non-coding RNAs (lncRNAs).4, 5, 6, 7 Recent studies have identified a new type of ncRNA, termed circular RNA (circRNA).8, 9, 10 Many circRNAs have been shown to be produced from the exons of coding genes, and several of them do not translate to proteins.8,10,11 Figure 1 summarizes the biogenesis of various ncRNAs, including miRNAs,5,12,13 siRNAs, lncRNAs,4 and circRNAs.8,10,11 Another class of ncRNAs that are produced to act as active enhancers are called enhancer RNAs (eRNAs).14 eRNAs are transcribed from genomic enhancer regions, which are generally considered as transcription factor binding sites distal to gene transcription start sites.15 The role of most eRNAs has remained enigmatic. Some studies have suggested that eRNAs carry out crucial tasks in regulating the chromatin conformation or transcription activation.16

Figure 1.

Figure 1

The biogenesis process of different types of ncRNAs

(A) RNA polymerase II (RNA Pol II) transcribes most miRNA genes and generates pre-miRNAs longer than 200 nt, composed of a hairpin structure containing the miRNA sequence. The pre-miRNAs are cleaved in the nucleus by RNase III enzyme Drosha into nearly 70-nt-long pre-miRNAs with a stem-loop motif. The pre-miRNAs are exported to the cytoplasm and are then cleaved by another RNase III enzyme called Dicer. Eventually, the ∼22 miRNA duplex is loaded into the RNA-induced silencing complex (RISC) where the mature single-stranded miRNA binds to complementary mRNA targets. (B) lncRNAs have been shown to be transcribed from genomic sequences. lncRNAs are classified into various categories, including promoter-related lncRNAs, enhancer-related lncRNAs, intronic and exonic lncRNAs, antisense lncRNAs, and long intergenic lncRNAs. (C) Numerous circRNAs are obtained from the pre-mRNAs, depending on the spliceosomes. circRNAs are classified into different types, including intron circRNAs, exon circRNAs, and extron-intron circRNAs. Read-through circRNA (rt-circRNA) is a new type of circRNA (dotted line).

Many studies have shown that several ncRNAs are deregulated (higher or lower) in cancer, where they are involved in cancer-associated processes, such as promotion of cancer stem cells (CSCs), drug resistance, and metastasis, underlining the role of ncRNAs as possible new targets in cancer.5,6,8,10,11,17, 18, 19, 20, 21 Some miRNAs have entered clinical trials.21, 22, 23 Moreover, circRNAs and lncRNAs have both been shown to be involved in cancer growth and treatment response by several different mechanisms.6,8 Some successful preclinical studies of nucleic acid-based therapeutics have suggested that ncRNAs could be used to treat cancer.5,21, 22, 23

miRNAs and response to chemotherapy in GI cancer

miRNAs and response to chemotherapy in colorectal cancer (CRC)

miR-135b is involved in various cancer cell processes, such as proliferation, apoptosis, and invasion in CRC.24 Moreover, miR-135b has been shown to inhibit phosphatase and tensin homolog (PTEN) signaling in CRC.25 miR-182 can promote drug resistance, cell proliferation, and metastasis in several malignancies.26,27 Knockdown of miR-182 could inhibit cell proliferation in triple-negative breast cancer.28 Some miRNAs were overexpressed in 5-fluorouracil (5-FU)-resistant CRC cell-line and tumor tissues.24

The levels of α-N-acetylgalactosaminide-α-2,6-sialyltransferase (ST6GALNAC2) were reduced by forced expression of miR-135b and miR-182. Moreover, ST6GALNAC2 was found to be a direct target of miR-135b and miR-182, and its expression showed an opposite pattern compared to miR-135b and miR-182 in CRC cell lines and tissues. Proliferation was increased by upregulation of miR-182 and miR-135b, and their upregulation resulted in reduced apoptosis in 5-FU-resistant CRC cell lines. Repression of these miRs by miR-135b/miR-182 inhibitors showed an opposite effect by increasing ST6GALNAC2 expression and promoting CRC progression. miR-135b and miR-182 were shown to modulate the activity of the AKT/ phosphatidylinositol 3-kinase (PI3K) pathway. Inhibition of the AKT/PI3K pathway increased the effects of 5-FU in LoVo and HCT-8 cells. Hence, miR-135b and miR-182 could be targets for CRC treatment.24

miR-138-5p has been found to act as an inhibitor in several different cancers.29, 30, 31 In pancreatic cancer (PC), for example, miR-138-5p played a tumor suppressor role.32 A study by Gao et al.33 showed that miR-138-5p improved the effect of radiotherapy on nasopharyngeal cancer through targeting EIF4EBP1. Roberto et al.34 showed that the expression of miR-138-5p changed the prognosis in osteosarcoma. Zhao et al.35 showed that RHBDD1 (rhomboid domain containing 1) was a direct target of miR-138-5p, and the reduction of RHBDD1 could inhibit breast cancer progression. Zhu et al.36 showed that the reduction of RHBDD1 by miR-138-5p suppressed lung cancer development.

Xu et al.37 showed that the expression of miR-138-5p was downregulated in CRC cells. miR-138-5p inhibited the migration of CRC cells and may also overcome chemoresistance. miR-138-5p controlled the expression of Snail1 by targeting NFIB, which is involved in the chemoresistance and migration of CRC cells. Their research showed that miR-138-5p could inhibit migration and reduce chemoresistance by affecting the NFIB/Snail axis, and could be a target in CRC treatment.37

miR-133b functions as a tumor repressor in CRC. For example, miR-133b reduced metastasis and proliferation of CRC cells in vitro and in vivo.38,39 Additionally, the miR-133b expression level was correlated with the risk of metastasis and survival in CRC patients40 and could be used as a prognostic indicator.41 Lv et al.42 reported that miR-133b was underexpressed in CRC spheroids enriched with CSCs, which also showed increased chemoresistance. Also, overexpression of miR-133b resulted in overcoming chemoresistance to oxaliplatin (OXP) and 5-5-FU and reducing CRC stemness. It was shown that miR-133b inhibited CRC chemoresistance and stemness by downregulating telomeric silencing disruptor 1-like (DOT1L) and a specific H3K79 methyl transferase.43

DOT1L replacement abrogated the inhibitory effects of miR-133b on CRC chemoresistance and stemness. These findings suggest that miR-133b may be a new target for overcoming CRC stemness and chemoresistance.42

miR-375 is a tumor suppressor that targets crucial oncogenes in several cancer types, such as hepatocellular carcinoma (HCC), CRC, gastric cancer (GC), and cervical cancer. In addition, miR-375 can be used as a therapeutic target because it can suppress tumor cell growth in vivo and in vitro.44 miR-375 is correlated with the susceptibility to chemotherapy in prostate cancer, breast cancer, and HCC.45, 46, 47 miR-375 also plays a key role in determining which patients should receive preoperative chemoradiation therapy for CRC.48,49 To date, more studies are required to prove a connection between drug resistance and miR-375 in CRC.50

Xu et al.51 reported that miR-375 was remarkably underexpressed in CRC cell lines and tissues, and that low expression of miR-375 was correlated with poor prognosis in CRC patients. In vivo and in vitro overexpression of miR-375 rendered the CRC cells more sensitive to a wide range of chemotherapy drugs. Further mechanistic analysis showed that, by directly targeting SP1 and YAP1, miR-375 improved the chemosensitivity of CRC to 5-FU. miR-375 negatively regulated YAP1, leading to a decrease in the expression of the downstream genes of Hippo-YAP1 (also known as survivin), such as CTGF, cyclin D1, and the BIRC55 pathway. Overall, miR-375 could be a potential molecular biomarker for chemoresistance in CRC patients, and also could have a role in the treatment of CRC patients, particularly those with chemoresistant tumors.51

Most studies about the role of miR-552 in tumor biology have concentrated on CRC.29, 30, 31, 32 In addition, measuring the miR-552 expression profile improved the prediction of lung metastasis in CRC patients.52 The involvement of aberrant miR-552 expression in the undifferentiated and highly proliferating tumors characteristic of DNA mismatch repair-deficient (dMMR) CRC was recently reported.53 Also, miR-552 can enhance the progression of HCC by stimulating the epithelial-mesenchymal transition (EMT).54

Zhao et al.55 found significantly downregulated miR-552 expression in 5-FU-resistant CRC cells and tissues, as well in dMMR tumors with poor prognosis after chemotherapy. Overexpression of miR-552 increased apoptosis and reduced 5-FU resistance, while inhibiting miR-552 expression increased 5-FU resistance in CRC cells. From a mechanistic point of view, miR-552 directly targeted the 3′ UTR of SMAD2. Overall, they demonstrated that the miR-552/SMAD2 pathway governed the response to 5-FU chemotherapy in dMMR CRC tumors.55

Previous studies have shown that miR-106a was overexpressed in CRC tumor tissues, fecal samples, and in plasma.56, 57, 58 A lower level of miR-106a was correlated with decreased overall survival as well as disease-free survival (DFS) in CRC patients.59 It was shown that miR-106a is involved in the regulation of CRC oxaliplatin sensitivity.60 miR-106a transfection mildly suppressed the growth of CRC cells and rendered them more sensitive to oxaliplatin. Furthermore, miR-106a overexpression decreased the FOXQ1 level in CRC cells at both mRNA and protein levels. Increased miR-106a expression also increased the expression of Wnt target genes, including matrix metallopeptidase 2 and vascular endothelial growth factor-A, which are known to be modulated by FOXQ1. It was predicted that miR-106a could directly bind to the 3′ UTR of FOXQ1, and this was confirmed by the finding of higher miR-106a and lower FOXQ1 levels in tumor tissues from oxaliplatin-sensitive CRC patients, compared to oxaliplatin-resistant CRC patients. They concluded that miR-106a increased CRC sensitivity to oxaliplatin through direct suppression of FOXQ1 expression.60

miR-744 was upregulated in head and neck tumors compared to normal tissues.61 High plasma levels of miR-744 correlated with chemoresistance and poor prognosis in patients with PC undergoing gemcitabine-based chemotherapy.62,63 Moreover, there were higher levels of serum miR-744 in GC patients compared to controls.64 Zhou et al.65 reported that miR-744 levels were substantially higher in CRC tissues from patients receiving oxaliplatin prior to surgery, and in oxaliplatin-resistant HCT116 cells. Oxaliplatin chemoresistance was increased by miR-744 overexpression, while miR-744 inhibition in HCT116 and T84 cells increased oxaliplatin sensitivity. In addition, the level of BIN1 protein was reduced by miR-744, and the oxaliplatin chemoresistance caused by miR-744 was reversed by BIN1 overexpression. In addition, a luciferase reporter assay demonstrated that BIN1 was a direct target of miR-744. Taken together, these results showed that by suppressing BIN1 expression in CRC cells, miR-744 could increase oxaliplatin drug resistance and could be a strategy to reverse oxaliplatin resistance in CRC therapy.65

miR-214 plays a role in several cancer types and has been implicated in many cellular pathways.66,67 miR-214 has been found to act as a tumor suppressor in CRC68,69 and can bind to the 3′ UTR of ARL2 (ADP ribosylation factor like GTPase 2). Also, miR-214 can control the signaling of the ErbB2/ErbB3 pathway and target the cellular adhesion molecule Necl-2.70 A study by Yang et al.71 showed that miR-214 was downregulated in 5-FU-resistant CRC cells in comparison with normal cells. 5-FU-sensitive and 5-FU-resistant CRC cells could both be sensitized to 5-FU by increasing the expression of miR-214. In terms of function, miR-214 suppressed cell growth and colony formation and increased 5-FU-mediated caspase-3 activity and apoptosis levels. Western blotting and dual-luciferase reporter assays showed that miR-214 could target heat shock protein 27 (Hsp27). Hsp27 makes LoVo and HT-29 cells sensitive to 5-FU by increasing apoptosis. Hsp27 overexpression blocked the action of miR-214, and the 5-FU sensitivity was affected. In summary, miR-214 targeted Hsp27 and sensitized CRC cells to 5-FU, suggesting a possible role in chemotherapy.71

miR-34a has received some interest in CRC because p53, which is a major transcriptional regulator of apoptosis, is regulated upstream.72 In CRC biopsies, miR-34a expression was significantly lower as compared to healthy colon tissue.73 It was proposed that miR-34a could control the asymmetric division of stem cells,74 inhibit the EMT, and reduce proliferation, tumorigenicity, and metastasis in CRC.75 The mechanism of miR-34a as a tumor suppressor miRNA works by controlling Notch,76 affecting multiple oncogenes, as well as PDGFRA signaling.77

Li et al.78 also found that miR-34a expression was significantly downregulated in clinical CRC specimens from oxaliplatin-resistant patients and could be experimentally used to develop multidrug-resistant CRC cells. Multidrug-resistant HCT-8/OR cells could be resensitized to oxaliplatin by exogenous expression of miR-34a, while miR-34a inhibition increased oxaliplatin resistance in chemosensitive HCT-8 cells. Mechanistically, miR-34a positively increased the stability of ornithine decarboxylase antizyme 2 (OAZ2) mRNA by directly targeting its 3′ UTR. Inhibition of miR-34a/OAZ2 signaling increased chemoresistance by activating anti-apoptotic pathways and multidrug resistance (MDR) ATP-binding cassette (ABC) drug efflux pumps, contributing to MDR development in CRC cells.78

Table 1 lists some miRNAs that affect the response to chemotherapy agents in CRC.

Table 1.

Various microRNAs that affect the chemotherapy response in colorectal cancer

MicroRNA Expression in CRC Target Drug Model Type of cell line Ref.
miR-224 up KRAS 5-FU in vitro HCT116 79
miR-31 up oxaliplatin human 80
miR-135b, miR-182 up ST6GALNAC2 5-FU in vitro HCT-8, 5-FU-resistant HCT-8 cells (HCT-8/5-FU), LoVo 24
miR-1914, miR-1915 down NFIX 5-FU, oxaliplatin in vitro HCT116 81
miR-107, miR-99a-3p up CCND1, DICER1, DROSHA, NFKB1 fluoropyrimidine human 82
miR-5323p down ETS1, TGM2 cisplatin, 5-FU in vitro RKO, SW480, SW620, HCT116, CaCo2, HT29, LoVo, colorectal mucosal cell line FHC, kidney cell 293FT 83
miR-587 up PPP2R1B 5-FU in vitro RKO, HCT116, FET, GEO 84
miRNA-17-5p up PTEN oxaliplatin, irinotecan, 5-FU human human CRC cell lines COLO205 (CCL-222), SW480 (CCL-228) 85
miR-143 down IGF-IR oxaliplatin in vitro SW1116 86
miR-138-5p down NFIB, Snail1 5-FU, doxorubicin, cisplatin in vitro LOVO, HCT116, HT29, SW620, SW480 37
miR-195-5p down GDPD5 5-FU in vitro Caco-2, HCT8, HCT116, SW480 87
miR-93-5p up cyclin-dependent kinase inhibitor 1A (CDKN1A) multiple drugs in vitro HCT-8 88
miR-143 down FXYD3 fluoropyrimidine human 89
miR-199b down SET oxaliplatin in vitro W480 (ATCC CCL-228), WiDr (ATCC CCL-218), DLD-1 (ATCC CCL-221), HT-29 (ATCC HTB-38), SW620 (ATCC CCL-227) 90
miR-133b down DOT1L oxaliplatin, 5-FU in vitro HT29, HCT116, SW620, HEK293 42
miR-375-3p down YAP1, SP1 5-FU in vitro HCT116 51
miR-204-5p down RAB22A oxaliplatin in vitro Caco2, DLD1, HCT8, HCT116, HT29, LoVo, SW480, SW620 91
miR-503-5p up PUMA oxaliplatin in vitro, in vivo, human HCT116, HT29 92
miR-214 up AEBP1 oxaliplatin in vitro, human HT29 93
miR-761 Down FOXM1 5-FU in vitro HT29, SW480, SW620, DLD-1, FHC 94
miR-940 down MACC1 anlotinib in vitro, in vivo SW620, Lovo 95
miR-330 down thymidylate synthase (TYMS) 5-FU in vitro HCT116, HT29, SW480, SW620, FHC, HEK293T 96
miR-218 down BIRC5, TS 5-FU in vitro HT29, HCT116 97
miR-197 up TYMS 5-FU in vitro HCT8, HCT116, SW480 98
miR-520 g up p21 5-FU in vitro RKO, HCT116, FET, GEO 99
miR-153 up FOXO3a cisplatin in vitro SW480 100
miR-552 down SMAD2 5-FU in vitro SW-480, SW-620, HT-116, CCD-18Co 55
miR200c down JNK2 multiple drugs human HCT8, HCT116, SGC7901, Bel7402 101
miR-20a up ASK1 cisplatin in vitro FHC,HT29, SW480, LoVo 102
miR-106a up FOXQ1 oxaliplatin in vitro 293T, HCT116, HT-29 60
miR-149 down FOXM1 5-FU in vitro HCT-8, LoVo 103
miR-1271 down mTOR cisplatin in vitro SW480 104
miR-143 up ERK5, NF-κB, Bcl-2 5-FU in vitro HCT116 105
miR-744 up BIN1 oxaliplatin in vitro HCT116 65
miR-199a/b up β-catenin, Gsk3β, ABCG2 cisplatin in vitro ALDHA1+ cells, ALDHA1 cells 106
miR-214 down Hsp27 5-FU in vitro HT-29, LoVo 71
miR-129 down BCL2 5-FU human HCT116, RKO, SW480 107
miR-21 up RECK 5-FU, oxaliplatin human ? 108
miR-106a up DUSP2 5-FU human HCT116/SW620 109
miR-361 up FOXM1 5-FU in vitro HCT116, HT29 110
miR-139-5p down NOTCH-1 5-FU in vitro HCT-116, LoVo, HCT-8 111
miR-195 down G2 checkpoint kinase WEE1, CHK1 5-FU in vitro HCT-116 112
miRNA-497 down Smurf1 5-FU in vitro SW480 113
miR-519c down ABCG2, HuR 5-FU in vitro S1, S1M1 80 114
miR-543 up PTEN 5-FU in vitro HCT8, HCT8/FU 115
miR-34a down TGF-β, Smad4 oxaliplatin in vitro HT29 116
miRNA-126 up capecitabine, oxaliplatin human N/A 117
miR-1290 up hMSH2 5-FU in vitro RKO, SW480, HCT116, LoVo 118
miR-770-5p HIPK1 methotrexate in vitro HT-29 119
miR-200c down PTEN,E-cadherin 5-FU in vitro HCT-116 120
miR-20a down, up BNIP2 5-FU, L-OHP, VM-26 in vitro SW620, SW480 121
miR-101 down 5-FU, cisplatin human HT-29, RKO 122
miR-203 down SIK2 paclitaxel (Taxol) in vitro CCD-18Co, CCD-33Co, LoVo, CaCo2, T-84, SW480, DLD-1 NCIN87, SKBR3, LNCaP 123
miR-196b-5p up SOCS1, SOCS3 5-FU in vitro Caco-2, COLO 205, COLO 320DM, CW-2, DLD-1, HCT15, HCT116, HT-29, NCI-H716, LS 174T, LoVo, RKO, SW480, SW620, SW948, SW1116 124
miR-409-3p down Beclin-1 oxaliplatin in vitro LoVo, HCT 116, DLD-1, SW480, HT-29, RKO, FHC, CCD-18Co 125
miR-338-3p down mTOR 5-FU in vitro HCT116, HT29 126
miR-107 CAB39 DCA, oxaliplatin (l-OHP) in vitro HCT-8, LoVo, HEK293T 127
miR-210-3p up succinate dehydrogenase subunits D 5-FU in vitro HT29 128
miR-145 down RAD18 5-FU in vitro SW620 129
miR-874-3p down YAP, TAZ 5-FU in vitro HCT116, SW480 130
miR-34a down LDHA 5-FU in vitro DLD-1 131
miR-203 up ATM kinase oxaliplatin in vitro HT29, RKO, HCT116 132
miR-34a down OAZ2 multiple drugs in vitro HCT-8, HCT-116, SW-480 78
miR-492 down CD147 oxaliplatin in vitro LS174T 133
miR-21 up hMSH2 5-FU in vitro HT-29, HT-29/5-FU 134
miR-222 down ADAM-17 multiple drugs in vitro HCT-8,
HCT-116
135

miRNAs and response to chemotherapy in PC

miR-1291-5p (or miR-1291) is underexpressed in samples from patients with PC, compared to adjacent normal tissues.136 Moreover, miR-1291 has been shown to act as a tumor inhibitor by affecting the cell metabolome, inducing apoptosis and cell cycle arrest.136,137 Arginine succinate synthase 1 (ASS1) is extensively downregulated in PC samples, and it might be a target of miR-1291.136 miR-1291 has been reported to suppress renal carcinoma cell viability by direct targeting of GLUT11.138

A new approach to develop recombinant miR-1291 agents (different from standard synthetic miRNA mimics) was reported by Tu et al.139 First, they showed that when the bioengineered miR-1291 agent was expressed in human PC cells it was processed into functional miR-1291-5p at high levels. miR-1291 disturbed arginine homeostasis by repressing ASS1 protein levels and rendered ASS1-abundant L3.3 cells sensitive to arginine deprivation treatment. In contrast, treatment with miR-1291 decreased the level of GLUT1 protein in both PANC-1 and AsPC-1 cells, thereby lowering glucose uptake and reducing glycolysis. As a result, miR-1291 greatly increased the ability of cisplatin to inhibit PC cell viability. The findings showed that miR-1291 could sensitize PC cells to arginine deprivation therapy and chemotherapy by targeting GLUT1-mediated glycolysis and ASS1 arginolysis, and it may help in a treatment for lethal PC.139

miR-301-3p was downregulated in PANC-1 cells in vitro by treatment with luteolin, and the knockdown of miR-301-3p showed antiproliferative effects, suggesting that miR-301-3p was an oncogenic lncRNA. miR-301-3p specifically targeted caspase-8, which is required for apoptosis, and miR-301-3p knockdown sensitized PANC-1 cells to the effects of TRAIL. TRAIL is considered to be a potent anticancer agent due to its ability to selectively kill malignant cells. However, resistance to TRAIL is the main factor that limits its application.140 Inhibition of various cell survival pathways, such as PI3K/AKT, inhibitory nuclear factor κB (NF-κB) (IκB) kinase (IKK), and protein kinase C (PKC) have been suggested to combat TRAIL resistance.141 Upregulation of caspase-8 makes cancer cells more susceptible to TRAIL, while downregulation of capsase-8 makes them more resistant.142,143 Epigenetic modifications can potentiate TRAIL cytotoxicity by controlling death receptor expression, inducing cell cycle arrest and increasing caspase-8 levels.140,142 Multiple genes have been identified as targets for miR-301-3p. In PC cells, miR-301-3p directly targeted PTEN and activated PI3K/AKT signaling.144 Inhibition of miR-301-3p could potentiate the cytotoxicity of gemcitabine. The NF-κB-repressing factor (NKRF) is another confirmed target of miR-301-3p. Thus, miR-301-3p overexpression can stimulate NF-κB signaling by repressing NKRF in PC cells.145

Moeng et al.146 studied the role of miR-301-3p in controlling cell proliferation, target gene expression, and TRAIL sensitivity. PANC-1 cells exposed to luteolin, a small molecule that suppresses PANC1 cell growth and sensitizes the cells to TRAIL, showed lower expression of miR-301-3p. The knockdown of miR-301-3p suppressed the proliferation and increased the TRAIL cytotoxicity. Additionally, caspase-8 was specifically targeted by miR-301-3p. The interaction between caspase-8 and miR-301-3p could also explain the function of miR-301-3p in different types of cancer.146

miR-137 has been shown to possess tumor-suppressor properties in several different types of human cancer.147 Moreover, it has been shown that miR-137 reduces tumor development and increases the chemosensitvity in lung cancer148 and in neuroblastoma cells.149 More specifically, miR-137 has been shown to inhibit mitophagy by controlling two mitophagy receptors, NIX and FUNDC1.150 In glioma cells, miR-137 upregulation inhibited autophagy, while treatment with a miR-137 antagomir promoted autophagy.151 Previous studies have shown that miR-137 expression inhibits tumor formation and invasion and increases the effects of chemotherapy in human PC cells.152,153

Wang et al.154 hypothesized that miR-137 could control autophagy and thereby sensitize PC cells to chemotherapy. The findings revealed that doxorubicin (DOX) caused autophagy in PC cells, but it decreased the expression level of miR-137. In contrast, overexpression of miR-137 increased the effects of DOX to reduce viability, trigger apoptosis, and inhibit autophagy by affecting the PC cell autophagic flux. ATG5 was a direct target of miR-137. In contrast, ATG5 overexpression significantly inhibited apoptosis and prevented autophagy induced by overexpression of miR-137. miR-137 made PANC-1 tumor xenografts more sensitive to DOX by inhibiting autophagy and ATG5 in vivo.154

The effects of glucocorticoids (GCs) on the differential control of miRNAs has been examined in only a small number of studies. Smith et al.155 reported that the inhibition of Dicer in response to treatment with glucocorticoids led to the decreased expression of several specific miRNAs. There was a high-density CpG island in the promoter region of miR-132, which showed a 2-fold increase in DNA methylation after treatment with dexamethasone (DEX), leading to the extensive silencing of many genes.156 Hypermethylation after DEX treatment was identified by a detailed examination of the promoter region of miR-132. Treatment with a demethylating agent could restore the expression of miR-132. Another investigation in prostate cancer found that hypermethylation regulated the miR-132 promoter region, resulting in its decreased expression.157

Abukiwan et al.158 analyzed 35 PC tissue samples from patients who received or did not receive glucocorticoids before surgery, as well as one primary and two established PC cell lines. They detected 268 miRNAs differentially expressed between DEX-treated and untreated cells by miRNA microarray analysis, qRT-PCR, and in silico analysis. They selected miR-132 and its target gene, transforming growth factor β2 (TGF-β2), as the lead candidates that could be involved in cancer progression. As shown by the enhanced luciferase activity, miR-132 mimics bound directly to the 3′ UTR of TGF-β2 luciferase and enhanced its expression. In comparison, DEX inhibited miR-132 expression via promoter methylation. The DEX-induced migration, expression, and clonogenicity PC cells, and the expression of E-cadherin and vimentin were decreased by miR-132 mimics. The growth of PC xenografts was also reduced. Glucocorticoid admininstration to patients post-surgery increased the overall hypermethylation of the miR-132 promoter and TGF-β2 expression in tissues; miR-132 expression was observed but could not be quantitatively measured. DEX-mediated suppression of miR-132 as a component of PC treatment could provide a basis for miRNA-based treatment.158

Several assorted diseases, including calcified aortic stenosis, obesity, infertility, ischemic stroke, and immunoglobulin (Ig)A nephropathy, are all characterized by the dysregulation of miR-374b-5p.159, 160, 161, 162, 163 Some studies have shown the overexpression of miR-374b-5p in prostate, breast, head and neck, and GC, and also in melanoma.164, 165, 166, 167, 168, 169 Alternatively, the downregulation of miR-374b-5p was observed in CRC and T cell lymphoblastic lymphoma.170,171 The findings suggest that miR-374b-5p could have different functions in different tumor types. A report by Schreiber et al.172 revealed a correlation between miR-374b-5p downregulation and the development of the cisplatin-resistant phenotype after stepwise treatment with an increasing dose of cisplatin across >20 passages. This suggested that in PC, the reduced expression of miR-374b-5p was correlated to chemoresistance. Nonetheless, the clinical relevance and the main mechanism behind the effect of miR-374b-5p in PC has yet to be elucidated. In PC tissues, miR-374b-5p was downregulated and was correlated with chemoresistance and low progression-free and overall survival rates. miR-374b-5p upregulation leads to the downregulation of anti-apoptotic proteins, such as baculoviral IAP repeat-containing 3 (BIRC3), X-linked apoptosis inhibitor (XIAP), and B cell lymphoma 2 (BCL2), which reduces the response of PC cells to gemcitabine. This confirms that miR-374b-5p has a tumor-suppressor function and governs PC resistance to chemotherapy.172

miR-374b-5p has been proposed to be a diagnostic biomarker in multiple cancer types. Hanniford et al.166 reported that miR-374-b-5p could provide a diagnostic signature in brain metastasis of melanoma. The Cancer Genome Atlas (TCGA) data analysis has also shown that miR-374b-5p could predict the progression of breast cancer. In PC tissues, miR-374b-5p expression was significantly lower, and the downregulation of miR-374b-5p predicted poor progression-free and overall survival in PC patients.165 Moreover, Summerer et al.167 reported that higher plasma miR-374b-5p levels predicted poor outcomes in head and neck cancer patients. They suggested that miR-374b-5p could be used as a minimally invasive diagnostic marker in cancer patients.

Sun et al.173 quantified the expression of miR-374b-5p in PC patient samples using qRT-PCR. They then assessed whether the levels of miR-374b-5p were correlated with clinicopathological features and progression-free or overall survival in PC patients. They used loss- or gain-of-function experiments in vivo and in vitro to test the effects of miR-374b-5p expression on chemoresistance and characterized the possible targets of miR-374b-5p using western blotting, bioinformatics analysis, qRT-PCR, RNA immunoprecipitation (RIP) assays, and luciferase reporter RIP assays. They showed that reduced expression of miR-374b-5p increased the tolerance of PC cells to gemcitabine chemotherapy, by increasing several anti-apoptotic proteins, such as XIAP, BCL2, and BIRC3. The findings indicated that miR-374b-5p may be considered a new therapeutic strategy for treating patients with chemoresistant PC.173 Table 2 lists some miRNAs that may affect the response of PC to chemotherapy.

Table 2.

Various microRNAs that affect chemotherapy response in pancreatic cancer

MicroRNA Expression in pancreatic cancer Target Drug Model Type of cell line Ref.
miR-1291-5p up GLUT1, ASS1 cisplatin in vitro AsPC-1, PANC-1, L3.3 174
miR-138-5p up vimentin 5-FU in vivo, human AsPC-1, BxPc-3, Capan-1, Capan-2, CFPAC-1, PANC-1, MIA PaCa-2, SW1990 32
miR-100 up FGFR3 cisplatin in vitro, in vivo AsPC1, BxPc-3, Capan-1, Capan-2, CFPAC-1, PANC-1, MIA PaCa-2, SW1990 175
miR-137 up pleiotropic growth factor (PTN) 5-FU in vivo, in vitro AsPC-1, BxPc-3, Capan-1, Capan-2, CFPAC-1, PANC-1, MIA PaCa-2, SW1990 153
miR-137 down ATG5 doxorubicin in vivo PANC-1-Dox, PANC-1 176
miR-20a-5p down ribonucleotide reductase subunit M2 (RRM2) gemcitabine in vivo MIA-PaCa2, HEK293 177
miR-21 up p85α gemcitabine in vivo MIA-PaCa2, PANC-1, Hs766T 178
miR-21 up PDCD4 5-FU in vivo PATU8988, PANC-1 179
miR-429 down PDCD4 gemcitabine in vivo SW1990 180
miR-374b-5p down gemcitabine in vivo, in vitro BxPC-3, PANC-1, AsPC-1, SW1990, Capan-1, Capan-2, CFPAC-1, MIA PaCa-2 173
miR-132 down TGF-β2 dexamethasone in vitro AsPC-1, PANC-1 158
miR-30a down SNAI1-IRS1-AKT (snal1) gemcitabine in vitro, in vivo SW1990, SW1990-R 181
miR-221-3p up RB1 5-FU in vivo PANC-1, PATU8988 182
miR-320a up PDCD4 5-FU in vivo PANC-1, PATU8988 183
miR-21 down FasL gemcitabine human, in vivo PANC-1 BxPC3 184
miR-1285 down YAP1 gemcitabine in vivo AsPC-1, BxPC-3, MIA PaCa-2, PANC-1, SU86.86 T3M4 185
miR-3656 Down RHOF gemcitabine in vitro, in vivo GR PANC-1 (PANC-1-GR) 186
miR-663a up (gem), down (OSI-027) gemcitabine in vivo Panc-1, BxPC-3, T3-M4, MIA PaCa-2 187
miR-320c up SMARCC1 gemcitabine in vitro, human MIA PaCa2, PSN1 188
miR-21 down RRM2 gemcitabine in vivo SUIT2-028, SUIT2, SUIT2-007 189
miR-101-3p down RRM1 gemcitabine in vitro, human PANC-1, AsPC-1, MIA-PaCa2, AsanPaCa, BxPC-3 190
miR-410-3p down 3′ UTR of HMGB1 gemcitabine in vivo, in vitro MIA PaCa-2, PANC-1 191
miR-21 down PDCD4 gemcitabine in vivo PANC-1 192
miR-301-3p down caspase-8 luteolin in vitro PANC-1 146

miRNAs and response to chemotherapy in GC

Zheng et al.193 investigated the effects of miR-34c (and its upstream transcription factor E2F1) on the susceptibility of GC cells to paclitaxel in combination with cisplatin. They compared paired samples of GC tissue and adjacent normal tissue from 74 GC patients. Western blotting was used for E2F1 and real-time qPCR for miR-34c. Steadily increasing drug concentrations induced cisplatin and paclitaxel resistance in GC cells. They found that E2F1 inhibited miR-34c to promote proliferation of the GC cells and increase the resistance to cisplatin plus paclitaxel. Alternatively, silencing of E2F1 led to an increase in the effectiveness of paclitaxel plus cisplatin in GC cells.193

The roles of miR-567 in targeting FGF5 and inhibiting cell proliferation, invasion, and migration were previously reported in osteosarcoma.194 Other studies showed that miR-567 served as a tumor suppressor and an inhibitor of tumorigenesis in breast cancer.195, 196, 197, 198 Zhang et al.199 investigated the biological effects of miR-567 on gastric tumorigenesis and chemoresistance and the potential mechanism thereof. In vitro and loss- and gain-of-function assays demonstrated that miR-567 decreased proliferation and sensitized the GC cells to oxaliplatin and 5-FU. The tumorigenesis of GC cells in vivo was significantly inhibited by overexpression of miR-567. The mechanistic investigation revealed that PIK3AP1 triggered AKT phosphorylation in GC cells. miR-567 directly targeted PIK3AP1 to inhibit the PI3K/AKT/c-Myc pathway, and c-Myc negatively modulated miR-567 expression in a miR-567-PIK3AP1-PI3K/AKT-c-Myc feedback loop. They concluded that miR-567 was a tumor suppressor and inhibited GC carcinogenesis and chemoresistance via a miR-567-PI3K/AKT-PIK3AP1-c-Myc feedback loop. miR-567 may serve as a possible biomarker for GC prognosis and response to therapy.199

miR-4766-5p was recently identified as a possible tumor suppressor in breast cancer.200 Liang et al.200 discovered that knockdown of miR-4766-5p could increase proliferation, metastasis, and chemoresistance.

NKAP is a notch signaling transcriptional repressor that is critically required for T cell development.201,202 In 2011, Hsu et al.202 observed that NKAP was engaged in T cell maturation and was required for T cell functional competence. Only a few researchers have studied the role of NKAP in GC. Juan et al.203 suggested in 2010 that NKAP could be causally involved in GC because it was overexpressed during GC development. NKAP was found to be a nuclear speckle protein with roles in RNA splicing and processing. Knockdown of NKAP resulted in an increased proportion of chromosome misalignment, more unprocessed precursor (pre-)mRNAs, and cell cycle arrest.204,205

A study conducted by Wei et al.206 examined the expression of miR-4766-5pin GC cells and tissues using qRT-PCR. RNA interference (RNAi) was used to alter miR-4766-5p expression in the GC cell lines MKN45 and AGS. Cell proliferation was assessed by colony formation and Cell Counting Kit-8 (CCK-8) assays. Cell cycle and apoptosis were examined by flow cytometry. A dual-luciferase reporter assay was used to demonstrate the association between miR-4766-5p and NKAP. Western blotting was used to measure the protein expression in various signaling pathways. Their results showed that miR-4766-5p was downregulated in GC cell and tissue samples. miR-4766-5p decreased the proliferation of GC cell lines and also inhibited invasion and migration. miR-4766-5p triggered apoptosis in cells. The NKAP gene was directly targeted by miR-4766-5p, and the Akt/mTOR pathway was inhibited. They concluded that miR-4766-5p inhibited cell metastasis and proliferation by attacking NKAP and could have applications as a diagnostic and prognostic biomarker in GC.206

miR-31 has been shown to be involved in many pathways associated with cancer cells. For example, low miR-31 expression was correlated with poor prognosis and accelerated tumor progression in patients with bladder cancer.207 miR-31 was found to be responsible for higher chemosensitivity and lower infiltration of colon cancer cells.208 In GC samples, miR-31 expression was found to be lower, which was related to poor prognosis, and therefore was clinically important.209,210 miR-31 can target the enhancer of zeste homolog 2 (EZH2), a polycomb protein that has been implicated in the tumorigenesis, progression, and metastasis of various types of cancer. Higher expression of EZH2 in liver cancer increased metastasis via downregulation of tumor suppressor miRNAs.211 Likewise, EZH2 was overexpressed in prostate cancer and was correlated with metastasis.212 EZH2 was also upregulated in CRC and could serve as a biomarker.213

Sun et al.214 studied the tumor-repressor function of miR-31 in GC by regulation of EZH2. They observed significant downregulation of miR-31 in GC cell lines. Upregulation of miR-31 decreased the viability and colony formation by induction of G2/M cell cycle arrest in AGS GC cells. In addition, overexpression of miR-31 increased the response to 5-FU. EZH2 was suggested to be the target of miR-31 in AGS cells by using in silico analysis and a dual-luciferase reporter assay.

The expression of EZH2 was upregulated in GC cell lines, while miR-31 overexpression in AGS cells led to EZH2 suppression. In addition, silencing of EZH2 in AGS cells reduced colony formation and proliferation and caused G2/M cell cycle arrest. In addition, overexpression of EZH2 could at least partly abrogate the tumor-suppressor effects of miR-31. It was suggested that EZH2 was directly involved in the miR-31-mediated inhibition of proliferation in AGS cells. Overexpression of miR-31 inhibited AGS invasion and migration and downregulated the mesenchymal markers (vimentin and N-cadherin) as well as upregulation of the epithelial marker (E-cadherin) in AGS cells. It was concluded that miR-31 is a tumor suppressor gene and may have a role in GC treatment.214

miR-200c is a member of the miR-200 family, which is underexpressed in GC.215 Studies have also suggested that miR-200c could reduce chemoresistance.216, 217, 218 Many studies have shown the key contribution of miR-200c to cancer cell apoptosis and metastasis affecting various genes such as E-cadherin, FN1, FAP1, ZEB1, and ZEB2, among others. Furthermore, miR-200c downregulation was strongly associated with cancer cell metastasis and proliferation.120,215,219 Other studies demonstrated that miR-200c plays a significant role in chemoresistance, and that the response to chemotherapy could be enhanced by the upregulation of miR-200c in cancer cells.120,218,220

The possible mechanism and role of miR-200c in improving the activity of cisplatin to inhibit migration and induce apoptosis in GC cells were investigated by Ghasabi et al.221 First, miR-200c and locked nucleic acid (LNA)-anti-miR-200c mimics were transfected into KATOIII cells. In addition, the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay showed that cisplatin and increased miR-200c expression could further inhibit KATOIII cell proliferation. The migration of KATOIII cells in a wound-healing assay was inhibited by miR-200c overexpression. The combination of miR-200 and cisplatin could suppress colony formation in KATOIII cells. In order to determine the possible targets of miR-200c, they measured its effects on RhoE, VEGFR, MMP9, and PTEN expression levels. A decrease in the mRNA and protein levels of MMP9 and VEGFR was produced by increased miR-200c expression, suggesting that MMP9 and VEGFR were miR-200c targets. Furthermore, RT-PCR revealed that the target gene of miR-200c was RhoE, and PTEN expression was reduced by LNA-anti-miR-200c. DAPI staining and flow cytometry showed increased cisplatin-mediated apoptosis by miR-200c expression that could be related to the inhibition of RhoE in KATOIII cells. Moreover, cell cycle analysis showed arrest at the G2 stage. They concluded that miR-200c acts in KATOIII cells as a tumor suppressor gene and could be a therapeutic strategy to overcome cisplatin resistance in GC treatment.221

miR-362-5p also plays a role in different cancer types. Ying et al.177 used miRNA expression profiling datasets from seven widely accessible renal cell carcinomas (RCCs) to show that hsa-miR-362-5p was downregulated in RCC. Ni et al.222 showed that miR-362-5p could target the tumor suppressor gene for cylindromatosis (CYLD), thus promoting proliferation and metastasis of HCC. It has also been shown that upregulation of miR-362-5p increases proliferation, migration, and invasion of MCF7 human breast cancer cells.177

Wei et al.223 examined the functional miRNAs that can modulate the cisplatin sensitivity of human GC cells. miRNA microarray analysis revealed differentially expressed miRNAs between SGC7901 (a human cisplatin-sensitive GC cell line) and SGC7901/DDP (the cisplatin-resistant counterpart). miR-362-5p was found to be downregulated in SGC7901/DDP cells compared to SGC7901 cells using qRT-PCR. Overexpression of miR-362-5p enhanced cisplatin susceptibility and apoptosis, whereas downregulation of miR-362-5p showed the opposite results. Database prediction suggested that the suppressor of zeste 12 protein (SUZ12) could be a miR-362-5p target. Moreover, relative to SGC7901 cells, the mRNA and protein levels of SUZ12 were substantially higher in SGC7901/DDP cells and were negatively correlated with the expression of miR-362-5p. Furthermore, in SGC7901/DDP cells, western blotting and MTT assays showed that knockdown of SUZ12 increased cisplatin sensitivity and also reduced the protein levels of NF-κB/p65. Furthermore, overexpression of miR-362-5p in SGC7901/DDP cells lowered the expression level of SUZ12 protein, while downregulation of miR-362-5p increased its levels. Deregulation of miR-362-5p could target SUZ12 to increase cisplatin resistance and reduce cisplatin-induced apoptosis. miR-362-5p could be a potential clinical approach for cisplatin-resistant GC patients.223

Table 3 lists some miRNAs that could affect the response to chemotherapy drugs in GC.

Table 3.

Various microRNAs that can affect chemotherapy response in gastric cancer

MicroRNA Expression in gastric cancers Target Drug Model Type of cell line Ref.
miR-214 down cisplatin in vitro, in vivo SGC7901, SGC7901/DDP 224
miR-31 up RhoA 5-FU in vivo MKN-45, HEK293T 225
miR-125b down HER2 cisplatin in vivo HGC-27, MGC-803 226
miR-204 down Bcl-2 5-FU in vitro, in vivo GTL-16, N87 GC 227
mir-204 down TGFBR2 5-FU in vitro, in vivo GES-1, AGS, SGC-7901, MKN-45, MGC-803, BGC-823 228
miR-128 down MAPK signaling cisplatin in vivo GES-1, BGC823, SGC7901 229
miR-567 down PIK3AP1 5-FU, oxaliplatin in vivo GES-1, MKN45, BGC823, AGS, MGC803, BGC803, MKN28 199
miR-647 down NK2, FAK, MMP2, MMP12, CD44, SNAIL1 vincristine in vitro, in vivo SGC7901/VCR 230
miRNA-29 down catenin-δ (CTNND1) cisplatin, docetaxel in vitro, in vivo GES-1 HGC27, MGC803, BGC823, MKN45 231
miR-200c down ZEB1/ZEB2 trastuzumab in vivo SGC7901, BGC803, MKN28, NCI-N87 232
miR-let-7 down AURKB cisplatin in vivo SGC7901/DDP 233
miR-135a up DAPK2, E2F oxaliplatin in vitro, in vivo SNU-5 NCI-N87 SGC7901/OXA, MGC-803/OXA, SGC7901, MGC-803 234
miR-181a up ATG5 cisplatin in vitro, in vivo SGC7901/CDDP 235
miR-200c up VEGFR, MMP9, RhoE cisplatin in vitro, in vivo KATOIII 221
miR-31 down EZH2 5-FU in vivo GES-1 (AGS, SNU-1, SNU-5, SNU16 214
miR-21-5p down PTEN, TIMP3 doxorubicin in vivo SGC7901/DOX 236
miR-26a down NRAS, E2F2 cisplatin in vivo SGC-7901, SGC7901/DDP 237
miRNA-200c down RhoE cisplatin in vivo SGC7901, SGC7901/DDP 217
miR-15a-5p down PHLPP2 cisplatin, oxaliplatin in vivo 238
miR-524-5p up SOX9 cisplatin in vivo SC-M1, AZ521 239
miR-34a up met cisplatin in vivo SGC7901/DDP 240
miR-218 up survivin cisplatin in vivo SGC7901, SGC7901/DDP 241
miR-375 down ERBB2 cisplatin in vivo SGC7901/DDP 242
miR-362-5p down SUZ12 cisplatin in vivo SGC7901, SGC7901/DDP 223
miR-126 down EZH2 vincristine, doxorubicin in vivo SGC7901/VCR, SGC7901/ADR 243
miR-495 up mTOR, ERBB2 cisplatin, 5-FU human 244
miR-20a up NFKBIB cisplatin in vivo SGC7901/DDP, SGC7901 245
miR-185 down ARC cisplatin, doxorubicin in vivo, human SGC-7901, NCI-N87, MGC-803, BGC-823, AGS 246
miR-429 down Bcl-2 5-FU in vivo AGS 247
miR-20a down CYLD cisplatin in vivo, human SGC7901/DDP, SGC7901 248
miR-16-1 up FUBP1 doxorubicin in vitro, in vivo, human SGC790 249
miR-21 up PTEN/PI3K/Akt pathway cisplatin in vivo SGC7901/DDP, SGC7901 250
miR-223 up FBXW7 cisplatin in vivo SGC-7901, BGC-823 251
miR-129 down P-gp cisplatin in vivo BGC823, MKN45 252
miR-21 up PTEN pathway trastuzumab in vitro, in vivo MKN45, NUGC4, NCI-N87 253
miR-4290 down PDK1 (pyruvate dehydrogenase kinase 1) cisplatin in vitro, in vivo GES-1 SGC7901, MKN45, HGC27, HEK293 254

miRNAs and response to chemotherapy in hepatocellular cancer

It has been observed that the expression of miR-144-3p is altered in thyroid cancer, where it can target ZEB1 and ZEB2 to reduce the invasion and migration capability of thyroid cancer cells.255 Additionally, since miR-144-3p plays a direct regulatory role in the expression of ZFX (zinc finger protein X-linked), it could not only prevent the growth of NSCLC tumor cells, but also induce apoptosis.256 miR-144-3p was underexpressed in HCC samples.257 Although numerous investigations have suggested that miR-144-3p could inhibit HCC proliferation via targeting E2F3 and AKT3,258,259 the role of miR-144-3p in tumor angiogenesis is still unclear.260,261

Wu et al.262 showed that miR-144-3p was downregulated in HCC. Restoring the expression of miR-144-3p in HCC cells could reduce proliferation, migration, and angiogenic potential both in vitro and in vivo. In addition, a clinical review demonstrated an association between low expression of miR-144-3p and shorter disease-free survival in HCC patients. From a mechanistic point of view, the TargetScan database263 predicted the target of miR-144-3p to be glucocorticoid kinase 3 (SGK3). When SGK3 was inhibited by miR-144-3p, a PI3K-independent pathway was activated to suppress the activation of mTOR-VEGF downstream signaling. Therefore, miR-144-3p, which is often downregulated in HCC, can suppress migration, proliferation, and angiogenesis by modulating SGK3 activation by a PI3K-independent signaling pathway, and could act as a prognostic marker for HCC patients.262

Researchers have also shown the underexpression of the liver-specific miR-122 in primary HCC samples.264,265 Downregulation of miR-122 may be a poor prognostic factor for liver cancer and its risk of metastasis.266, 267, 268 Restoration of miR-122 expression in HCC using an adenoviral vector (Ad-miR122) led to cell cycle arrest and increased apoptosis.269 Restoration of miR-122 in HCC cells could increase the response to DOX and vincristine (VCR). Moreover, overexpression of miR-122 in HCC cells using the adenovirus could increase the sensitivity to VCR or DOX. Cell cycle analysis showed that the anti-proliferative role of miR-122 was related to a higher number of cells in the G2/M phase. Ad-miR122 led to increased HCC cell sensitivity to DOX or VCR by downregulation of MDR efflux pumps, as well as other genes, such as GST-π, cell cycle-associated gene cyclin B1, and the anti-apoptotic gene BCL-w. Ad-miR122 combined with chemotherapy drugs inhibited HCC cells via G2/M arrest and downregulation of cyclin B1 and MDR-associated genes (at least in part).265

miR-122 has a crucial function in modulating normal hepatocyte growth, differentiation, and cholesterol metabolism.270 As mentioned above, miR-122 downregulation was associated with HCC growth and progression,267,271 as well as HCC cell chemoresistance. miR-122 downregulation is also associated with the EMT and HCC metastasis. Ectopic expression of miR-122 could increase the cell sensitivity to sorafenib (SOR) and DOX in HepG2 and Hep3B cells.266,272 Earlier work suggested that miR-122 may directly target the Wnt/β-catenin pathway,273 which has a critical role in tumor growth and the modulation of MDR1 expression.274,275

Cao and Yin276 confirmed the underexpression of miR-122 in HCC cells. Overexpression of miR-122 or inhibition of Wnt/β-catenin signaling increased HCC cell apoptosis and enhanced the sensitivity of HCC cells to oxaliplatin. miR-122 inhibited the Wnt/β-catenin pathway, as well as reduced the expression of MDR1 and increased the sensitivity of HCC cells to oxaliplatin. miR-122 may be a new therapeutic target for HCC treatment.276

miR-182 acts as an oncogenic miRNA in many cancers, including HCC, ovarian carcinoma, and breast cancer.27,277,278 miR-182 is overexpressed in HCC and could stimulate HCC metastasis by suppressing MTSS11 (metastasis suppressor 11) protein.27 Moreover, miR-182 may be involved in chemoresistance. Husted et al.279 used a microarray-based approach to survey drug resistance-related miRNAs and discovered that miR-182 was overexpressed in MDR Ehrlich ascites tumor cells. Suppression of miR-182 is a possible therapeutic approach because it is involved in glucocorticoid resistance via targeting FOXO3A in lymphoblastic malignancies.280

Qin et al.281 found that the level of miR-182 was higher in cisplatin-resistant HepG2cells compared to parental HepG2 cells. miR-182 overexpression increased cell viability while miR-182 suppression decreased cell viability during cisplatin therapy. This was more pronounced in HepG2-R cells. Upregulation of miR-182 decreased the expression of tumor protein 53-induced nuclear protein 1 (TP53INP1; tumor suppressor gene) in vitro. miR-182 overexpression decreased the tumor response to cisplatin in vivo, partly by targeting TP53INP1.

Human miR-204 has been shown to be underexpressed in multiple types of cancer, such as CRC282 and acute myeloid leukemia (AML).283 miR-204 expression was remarkably reduced in HCC samples compared to adjacent normal liver tissue.284 miR-204 targeted S1RT1 to inhibit the development of HCC.285 miR-204 has also been shown to be associated with venous metastasis of HCC.286

Yu et al.287 reported that miR-204 could enhance drug sensitivity in HCC by reducing the expression of NUAK1 (novel nua kinase family 1). miR-204 was identified as a tumor suppressor by reducing NUAK1 in HCC. This suggested that both NUAK1 and miR-204 could be promising targets for liver cancer treatment.287

miR-223 often shows lower expression in HCC cells, and it has been shown to be involved in many critical pathological and physiological processes, such as HCC proliferation, maintenance of stemness, and metastasis. miR-223 is also involved in MDR in HCC cells.288,289 Targeted therapy with miR-223 may be clinically relevant.290, 291, 292, 293 Moreover, miR-223 inhibited uncontrolled autophagy in cardiomyocytes.294

One study by Zhou et al.295 found lower levels of miR-223 expression in DOX-treated HCC cells. They suggested that overexpression of miR-223 suppressed DOX-mediated autophagy that participates in chemoresistance. Inhibition of autophagic flux by chloroquine abrogated the ability of miR-223 overexpression to reverse the resistance of HCC cells to DOX. FOXO3a has been characterized as a direct downstream target of miR-223, and it could be a central intermediate of the activity of miR-223 on DOX-mediated autophagy and chemoresistance in HCC cells.295

miR-125b is downregulated in many types of cancer, such as HCC. Ectopic expression of miR-125b suppressed the proliferation, invasion, and tumorigenic properties of HCC cells, suggesting that it can act as a liver cancer suppressor.296, 297, 298, 299 Although the function of miR-125b in drug resistance remains unknown, it has been suggested as a prognostic biomarker in HCC patients.300

Comparison of oxaliplatin-resistant and oxaliplatin-sensitive HCC cell lines showed that miR-125b was underexpressed in the resistant cells. Therefore, miR-125b was proposed to increase the ability of oxaliplatin to suppress proliferation, cell migration, and EMT.301 In addition, higher expression of EVA1A was demonstrated in tissue specimens from oxaliplatin-resistant HCC patients. Ectopic expression of EVA1A increased autophagy and abrogated the effects of miR-125b on inhibiting oxaliplatin-resistant cell lines and xenograft tumors. A pathway involving the downregulation of EVA1A-mediated autophagy and miR-125b may play a role in HCC cell chemoresistance.301

Researchers have also revealed a critical contribution of miR-222 to tumor cell proliferation, oncogenesis, migration, invasion, and drug resistance.302,303 They also detected the overexpression of miR-222 in many different kinds of cancer, such as CRC, HCC, glioblastoma, and breast cancer.135,304, 305, 306, 307 Several studies have shown that miR-222 is involved in chemoresistance. Miller et al.305 reported that miR-221/222 increased tamoxifen resistance in breast cancer. Zhong et al.308 showed that miR-222 was implicated in PTEN-mediated resistance to DOX and docetaxel. Garofalo et al.309 found that miR-221/222 induced TRAIL resistance via targeting the tumor inhibitors TIMP3 and PTEN and increased cell migration.

It has been shown that miRNA inhibitors designed to specifically target miR-222 could suppress cell migration, proliferation, and invasion, cause G1/S cell cycle arrest, and induce apoptosis in HepG2 cells.310 Furthermore, miR-222 could increase resistance to SOR in HepG2 cells. They found that miR-222 inhibitors could increase susceptibility to the antitumor activity of SOR in HepG2 cells. The phosphorylation of PI3K and AKT was regulated by miR-222, explaining the induction of SOR resistance. Therefore, miR-222 can enhance proliferation, migration, and invasion. They concluded that miR-222 could trigger the PI3K/AKT signaling pathway and increase HCC cell resistance to SOR.310

Table 4 lists some miRNAs that can affect the response to chemotherapy agents in HCC .

Table 4.

Various microRNAs that can affect chemotherapy response in hepatocellular cancer

MicroRNA Expression in GI cancers Target Drug Model Type of cell line Ref.
miR-21 up AP-1 5-fluorouracil, pirarubicin in vitro Hep3b, SMMC7721 311
miR133a, miR326 down Bcl-xl 5-FU, cisplatin in vitro HepG2 HCC 312
miR-133b up PP2A-B55δ cisplatin in vitro L02, HCC cell lines HepG2, MHCC97H, MHCC97L, Hep3B, amd Huh7, human embryonic kidney 293T cells (HEK293T 313
miR-215 up P53, P21 adriamycin human HepG2 (cat. no. HB-8065), Hep3B (cat. no. HB-8064), 786-O (cat. no. CRL1932), and ACHN (cat. no. CRL-1611), RCC cell lines, and A549 (cat. no. CRM-CCL-185), H1299 (cat. no. CRL-5803) 314
miR-223 down FOXO3a doxorubicin in vitro HepG2, Huh7, SNU387, and SNU449, and human embryonic kidney cell line (HEK293T) 315
miR-122 down cyclin B1 adriamycin (ADM), vincristine (VCR) in vitro Hep3B, HepG2, Huh7, and PLC/PRF/5 265
miR-33a-5p down HSPA8? cisplatin in vitro Hep3B and 97L 316
miR-590-5p up YAP1 adriamycin human HepG2 and Huh7 317
miR-125b down EVA1A oxaliplatin human HepG2, Huh6, Mahlavu, and SK-Hep1 301
miR-122 down MDR1 oxaliplatin in vitro HepG2, Bel-7402, SMMC-7721, Huh7, and normal liver cell line (WRL-68) 276
miR-26 down ULK1? doxorubicin in vitro HepG2, Huh7, 293T 318
miR-34a down MDR1/P-gp, AXL doxorubicin in vitro HepG2 319
miR-16 down IKBKB paclitaxel in vitro SMMC-7721, PLC, BEL-7402, BEL-7404, HepG2, HCCLM3, and the normal liver cell line LO2 320
miR-1258 down CKS1B doxorubicin In vitro HuH7, HCCLM3 320
miR122 down PCDH20 5-FU In vitro SNU-449, MHCC97, HepG2, and SMMC-7721 321
miR-367-3p down MDM2 sorafenib human SKhep1, HepG2, Huh7, SNU398, HA22T, SNU423 322
miR-589-5p up SOCS members SOCS2 and SOCS5, and tyrosine phosphatases members PTPN1, PTPN5, PTPN7, PTPN11, PTPN13, PTPN18, and PTPN20 doxorubicin in vitro 323
miR-125b down HK II 5-FU in vitro SMMC-7221, Huh7, MHCC-97L, HepG2, HepG3, and BEL-7402 human HCC cell lines, THLE-2 and THLE-3 normal human liver cell lines 324
miR-124 down SIRT1/ROS/JNK pathway cisplatin in vitro HepG2, Huh7 325
miR-222 up PI3K/AKT signaling pathway sorafenib in vitro HepG2, normal human hepatocyte cell line HL-7702 310
miR-520c-3p down Mcl-1 doxorubicin in vitro HepG2? 326
miR-101 down Mcl-1 doxorubicin in vitro HepG2, Hep3B, Huh7, PLC human HCC cell lines, and the L-O2 normal liver cell line 327
miR-182 up TP53INP1 cisplatin in vitro HEK293, HepG2 281
miR-340 down Nrf2 cisplatin in vitro HepG2 328
miR-193b down Mcl-1 cisplatin in vitro normal hepatic cell line L02, and HCC cell lines Huh7, HepG2, and PLC 329
miR-363 down Mcl-1 cisplatin human HepG2 330
miR-130a up RUNX3? cisplatin human Huh7, HEK293 331
miR-3163 ADAM-17 in vitro L-02 (a non-tumor hepatic cell line), MHCC97-H, or LM-3 (two highly metastatic cell lines of HCC), HepG2, Hu7, BEL-7402, or SMMC-7721, and MHCC97-L (a lowly metastatic cell line of HCC) 332
microRNA-122 down ADAM10, SRF, and Igf1R sorafenib in vitro HepG2, Hep3B, and SK-Hep-1 272
miR-122 down IGF-1R sorafenib in vitro Huh7, Huh7-DR3 333
miR-221 up caspase-3 sorafenib human Huh-7 334
miR-137 down ANT2 sorafenib human Huh7 335
miR-23a up TOP1 etoposide in vitro HepG2 and embryonic kidney cell line HEK293T 336
miR-27b down p53, CYP1B1 in vitro HepG2 337
miR-372-3p down Mcl-1? doxorubicin in vitro HepG2 338
miR-223 down ABCB1 doxorubicin and paclitaxel in vitro HCC3, LM-6, SMMC7721, Huh-7, SK-Hep-1, HepG2, BEL-7402, Hep3B 289

miRNAs and response to chemotherapy in esophageal cancer

miR-133b has been identified as an important miRNA in muscle that modulates myoblast differentiation and is involved in some myogenic disorders.339 miR-133b functions as a tumor suppressor and is downregulated in many cancers, including bladder cancer,340 prostate cancer,339 lung cancer,341 and GC.342 miR-133b was also downregulated in cells and tissues from esophageal squamous cell carcinoma (ESCC).343 However, the functional molecular mechanism of miR-133b in ESCC is still uncertain. The epidermal growth factor receptor (EGFR) is a 170 kDa transmembrane protein that binds to its cognate ligands, to trigger its tyrosine kinase activity.344 EGFR has a critical contribution to tumor growth by binding to EGF. EGFR is known to positively affect proliferation, migration, invasion, and apoptosis. EGFR expression is significantly higher in ESCC tissues where it is associated with poor prognosis, clinical stage, and invasion.345,346 ESCC patients who had a low copy number of EGFR genes were shown to have a higher survival rate than cases with a higher copy number.347,348 However, the association between miR-133b and EGFR in ESCC and the respective mechanism should be clarified. It was reported that in prostate cancer, miR-133b inhibited migration, growth, and invasion of PC3 and DU145 cell lines via EGFR targeting.339,349

Lower levels of miR-133b expression and higher expression of EGFR were found to occur together in ESCC specimens.343 Cell proliferation, invasion, and migration in ESCC cells were inhibited either by miR-133b overexpression or by EGFR knockdown, and the percentage of cells in the G1 stage was increased. The overexpression of miR-133b and knockdown of EGFR both increased apoptosis in ESCC cells. Overexpression of miR-133b decreased the phosphorylation of AKT, extracellular signal-regulated kinase (ERK), and PI3K via direct reduction of EGFR. Higher levels of CK-18 and E-cadherin, and lower levels of N-cadherin and vimentin were observed following the transfection with miR-133b mimics or short hairpin RNA (shRNA) EGFR (shEGFR). They concluded that overexpression of miR-133b could block the PI3K/AKT and mitogen-activated protein kinase (MAPK)/ERK signaling pathways via EGFR targeting and inhibit ESCC cell proliferation, invasion, and migration, suggesting a role of miR-133b in ESCC treatment.343

miR-145 is a well-characterized tumor suppressor in different cancer types.350, 351, 352 miR-145 was first reported to be downregulated in ESCC.353 miR-145 inhibited proliferation, invasion, and the EMT and increased differentiation in ESCC.354,355 Moreover, miR-145 increased the sensitivity of human colon cancer cells toward 5-FU.356

The downregulation of miR-145 in ESCC cells and tissues was correlated with the upregulation of REV3L (protein reversionless 3-like) in ESCC tumor specimens. In the KYSE150 ESCC cell line, miR-145 overexpression reduced the level of REV3L mRNA and protein. On the contrary, miR-145 decreased REV3L mRNA and protein in the normal esophageal epithelium cell line (HEEC).357 Additionally, ESCC cell viability was decreased following transfection with a miR-145 mimic. Overexpression of miR-145 strongly suppressed viability and increased the apoptosis rate after application of 5-FU. Furthermore, transfection with miR-145 mimics altered the expression of apoptosis-related genes (Bax, caspase-3, Bcl-2) in 5-FU-treated ESCC cells. Finally, Chen et al.357 suggested that miR-145 could be a therapeutic agent to treat ESCC.

miR-193a-3p has previously been observed to reduce the invasion of cancer cells and prevent metastasis, while promoting apoptosis via ERBB44 downregulation in lung cancer cells.358 Additionally, miR-193a-3p could increase apoptosis by affecting Mcl-1 in both U-251 and HeLa cells.359 miR-193a suppressed the expression of c-kit and served as a methylation-silencing tumor suppressor in AML.360

Meng et al.361 suggested a role of miR-193a-3p in the regulation of radioresistance and chemoresistance in ESCC cells (KYSE410 cells are moderately radiation resistant while KYSE150 cells are radiation sensitive). Upregulation of miR-193a-3p increased chemoresistance and radioresistance of the ESCC cells. Furthermore, miR-193a-3p downregulation decreased the radioresistance and chemoresistance of the ESCC cells. Additionally, miR-193a-3p was involved in DNA damage as shown by quantification of the γ-H2AX level in relationship to miR-193a-3p. Also, the siRNA-mediated suppression of the PSEN1 gene had similar effects as miR-193a-3p overexpression. They concluded that miR-193a-3p participated in radioresistance and chemoresistance in esophageal cancer via PSEN1 downregulation. Therefore, PSEN1 and miR-193a-3p might be biomarkers for resistance to chemotherapy and radiotherapy.361

miR-181a-5p is involved in cell apoptosis, proliferation, and migration, and it could be a new potential biomarker.362,363 Although the precise molecular mechanism and function of miR-181a-5p in cisplatin resistance in ESCC are unclear, Hummel et al.364 showed that miR-181a-5p was deregulated in samples from ESCC patients who were resistant to cisplatin.

Yang et al.365 found strong expression of miR-181a-5p in OE19/CDDP-resistant cells, whereas CBLB (E3 ubiquitin-protein ligase) was underexpressed in the OE19 cell lines. The overexpression of miR-181a-5p or the knockdown CBLB inhibited cell viability and induced apoptosis in cisplatin-resistant CDDP/OE19 cells. In addition, miR-181a-5p suppression or CBLB upregulation increased cell viability and prevented apoptosis in the cisplatin-sensitive OE19 cells. CBLB was confirmed to be a miR-181a-5p target. Furthermore, a rescue assay demonstrated that CBLB upregulation abrogated the effects of miR-181a-5p on OE19/CDDP cell viability. Moreover, miR-181a-5p overexpression increased the effects of cisplatin on ESCC xenografts in vivo.365

miR-10b has been found to act as an oncogene in several cancer types, including breast cancer,366 GC,367 lung cancer,368 and HCC.369 Moreover, higher expression of miR-10b was associated with chemoresistance. For example, one study showed that miR-10b increased CRC cell resistance to 5-FU, likely by suppressing the pro-apoptotic BIM.370 Zhang et al.371 showed that miR-10b was overexpressed in cisplatin-resistant nasopharyngeal cancer cells. miR-10b deficiency reversed the EMT phenotype and reduced cisplatin resistance by regulating the Notch1/KLF4/E-cadherin pathway. Another study showed that miR-10b overexpression in ER-positive breast cancer resulted in enhanced tolerance to tamoxifen, partially by downregulating HDAC44.372 An earlier investigation showed that miR-10b could increase invasion and migration in ESCC cells by targeting the KLF44 transcription factor.373

Wu et al.374 found that overexpression of miR-10b and underexpression of the peroxisome proliferator-activated receptor-γ (PPARγ) were associated together in ESCC cells and tissues. PPARγ has been shown to be a target of miR-10b. Additionally, miR-10b inhibition increased the cisplatin chemosensitivity of ESCC cells in vitro and tumors in vivo. Furthermore, miR-10b overexpression abrogated the PPARγ-induced cisplatin sensitivity. Therefore, miR-10b targeted PPARγ and thereby activated the AKT/mTOR/p70S6K signaling pathway. AKT inhibitor (GSK690693) inactivated the AKT/mTOR/p70S6K signaling pathway and abrogated the miR-10b-mediated cisplatin resistance in the ESCC cells. Overall, miRNA-10b might be used to increase the response of esophageal cancer patients to DDP treatment.374

miR-96 expression has been found to be upregulated in non-small cell lung cancer,375 breast cancer,376 prostate cancer,377 and bladder cancer.378 Also, recent studies suggested that miR-96 could promote tumor growth by downregulating RECK expression in ESCC.375, 376, 377, 378, 379

Table 5 lists some miRNAs that could affect the response to chemotherapy agents in esophageal cancer.

Table 5.

Various microRNAs that affect chemotherapy response in esophageal cancer

MicroRNA Expression in EC Target Drug Model Cell line Ref.
miR-27b-3p, miR148a-3p up Sp1 and PPARy, DNMT-1, MSK-1, Bcl-2 and Bim cisplatin human OE-19,OE-33, Flo-1, SKGT4, OACM5.1, OACP4C 380
miR-21 up PDCD4 cisplatin in vitro Eca109/DDP 381
miR-143 up lncRNA CCAT1 cisplatin in vitro ECA-109, TE-1, KYSE140, KYSE70, KYSE150, KYSE450 382
miR-142-5p down SREBP1 fatostatin human OE21, OE33 383
miR-141-3p up PTEN 5-FU, oxaliplatin human EC109, EC9706, TE-1, KYSE150 384
miR-224 up DESC1 cisplatin, 5-FU + doxorubicin, doxorubicin + cisplatin, 5-FU + paclitaxel in vitro TE-13, KYSE140, EC9706, KYSE30 385
miR-130a, miR-148a-3p up, down Bcl-2 cisplatin, 5-FU In vitro KYSE70, KYSE140, KYSE270, KYSE410 380
miR-27 up TGF–β cisplatin human CAFs, NOFs 386
miR-196a up cyclin B1, ABCG2 cisplatin in vitro TE1, EC109 387
miR-145 down REV3L 5-FU in vitro HEEC, TE-8, KYSE150, TE-1 357
miR-148a down MSK1, DNMT3B cisplatin, 5-FU in vitro KYSE410 388
Let-7b down IL-6/STAT3 pathway cisplatin in vitro TE1, TE5, TE8, TE9, TE10, TE11, TE13 389
miR-432-3p up KEAP1 cisplatin, 5-FU, actinomycin D in vitro KYSE170, KYSE770, KYSE2270 390
miR-499 down polβ cisplatin in vitro EC9706, KYSE30 391
miR-200c up PPP2R1B cisplatin in vitro TE-1, TE-8, TE-10, TE-13, TE-15 392
miR-193a-3p down PSEN1 docetaxel, paclitaxel, vinorelbine, 5-FU in vitro KYSE150, KYSE410, KYSE450, KYSE510 361
miR-21 up cisplatin, 5-FU in vitro KYSE170 393
miR-483-3p up E124 doxorubicin, cisplatin in vitro EC109, EC9706, TE-1 394
miR-181a-5p up CBLB cisplatin in vitro OE19 365
miR-141 up Yap1 cisplatin in vitro KYSE 28
miR-338-5p down Id-1 5-FU in vitro KYSE410, KYSE150, KYSE270 395
miR-10b up PPARγ cisplatin in vitro EC109, TE10 374
miR-29c down FBXO31 5-FU in vitro KYSE150FR, KYSE410FR 396

lncRNAs and response to chemotherapy in GI cancer

lncRNAs and response to chemotherapy in GC

Some lncRNAs are critically involved in tumor development and are involved in various chemoresistance mechanisms, such as mutation of drug targets, increased drug efflux, inhibition of apoptosis, and DNA damage repair.397 Furthermore, lncRNAs most often increase chemoresistance, whereas they hardly ever have the opposite effect. Figure 2 summarizes the respective pathways by which lncRNAs are involved in HCC, breast cancer, and lung cancer.

Figure 2.

Figure 2

An overview of the role of lncRNAs in chemoresistance

(A) lncRNA-associated pathways in HCC. (B) lncRNA-associated pathways in breast cancer. (C) lncRNA-associated pathways in lung cancer.

The lncRNA UCA1 increased metastasis and tumor growth in GC.398,399 He et al.400 reported that UCA1 expression could predict a poor prognosis in patients, and it could also modulate GC cell migration and proliferation in vitro. It was also reported that UCA1 could increase cisplatin resistance in ovarian cancer,401 non-small cell lung cancer,402 oral squamous cell carcinoma,403 bladder cancer,404 and GC.405 UCA1 knockdown was reported to increase apoptosis by modulating the expression of Bax and cleaved caspase-3/9.406 The activated protein Bax causes release of pro-apoptotic factors from the mitochondria into the cytoplasm to increase apoptosis.407 EZH2 has a key role in modulating gene expression to increase cisplatin resistance in GC.408,409 Overexpression of EZH2 promoted tumor development by activating the PI3K/AKT pathway.398,410 The PI3K/AKT pathway also has a significant role in increasing chemoresistance.411 Therefore, inhibition of the PI3K/AKT pathway will decrease drug resistance, thus restoring tumor sensitivity to chemotherapy drugs.411 It has also been found that the PI3K/AKT pathway affects chemotherapy response by modulating anti-apoptotic proteins and drug efflux pumps.412,413 It has been shown that the PI3K/AKT signaling pathway inhibits apoptosis by reducing the activity of caspase-9 and caspase-3.412,413

Dai et al.414 investigated the role of UCA1 in the response of GC to cisplatin, as well as the underlying mechanism. Apoptosis assays and CCK-8 were employed to explore the effects of various doses of cisplatin on GC apoptosis and proliferation. They also investigated the association between EZH2 and UCA1 using western blotting and qRT-PCR. RNA pull-down and RIP assays were used to detect the relationship between EZH2 and UCA1. TCGA and the GEO database suggested that a higher expression of UCA1 in GC tissues was connected with worse patient prognosis. Moreover, UCA1 overexpression increased GC cell proliferation and prevented apoptosis caused by cisplatin. Knockdown of UCA1 produced the opposite results. UCA1 exerted its role by increasing EZH2 expression, while EZH2 knockdown lowered GC cell cisplatin resistance. Therefore, by increasing EZH2 and activating the PI3K/AKT pathway, UCA1 promoted GC cisplatin tolerance. Interventions directed at EZH2 or UCA1 could provide a therapeutic approach for cisplatin-resistant GC patients.414

The lncRNA FAM83H-AS1 is linked with poor prognosis in colon cancer, luminal subtype breast cancer, and cervical cancer.415, 416, 417 Da et al.418 found that FAM83H-AS1 was overexpressed in GC and may be a prognostic indicator in patients. However, they found that the expression level of FAM83H-AS1 was not linked to the clinical pathology, such as tumor-lymph node-metastasis (TNM) stage and tumor size.

Wang et al.419 reported that FAM83H-AS1 was overexpressed in GC cell lines and tissues. It was correlated with invasion depth, poor differentiation, and chemoresistance in GC patients. FAM83H-AS1 was overexpressed in chemoresistant GC cell lines (SGC7901/R) and tissues. Furthermore, silencing of FAM83H-AS1 could sensitize SGC7901/R cells to 5-FU and cisplatin. Silencing of FAM83H-AS1 in SGC7901/R cells resulted in the inactivation of the Wnt/β-catenin signaling pathway. Activation of the Wnt/β-catenin signaling pathway reversed the effects of FAM83H-AS1 silencing on increasing chemosensitivity. They concluded that FAM83H-AS1 was involved in chemoresistance in GC patients by activating the Wnt/β-catenin signaling pathway.419

The relationship between miRNAs and lncRNAs has been widely investigated.420 lncRNAs either stimulate or compete with certain specific miRNAs, which subsequently affect their target mRNAs, and some miRNAs may lower the longevity of certain lncRNAs.420

Cheng et al.421 performed a study to explore the role and potential underlying mechanism of action of HOTAIR in modulating resistance to cisplatin in GC. Their study demonstrated that expression of HOTAIR was increased in GC tissues and cell lines, while miR-34a was downregulated. Using RIP assays and luciferase reporter assays, HOTAIR was found to have the potential to directly bind to miR-34a. miR-34a was remarkably upregulated in cells transfected with siRNA (si-)HOTAIR. In GC cells resistant to cisplatin, anti-miR-34a reduced the effect of si-HOTAIR on resistance to cisplatin and inhibited the Wnt/β-catenin and PI3K/Akt signaling pathways, as well as apoptosis-related genes, which suggests that the HOTAIR effects mainly depend on miR-34a. Additionally, HOTAIR suppression promoted the inhibitory effect of cisplatin on proliferation of tumors in vivo. Consequently, knockdown of HOTAIR suppressed resistance to cisplatin in GC cells through increasing expression of miR-34a. The HOTAIR/miR-34a axis may exert its effects on GC cells via the Wnt/β-catenin and PI3K/Akt signaling pathways. Their study suggested that HOTAIR may be a promising therapeutic target in patients with GC.421

Table 6 lists some lncRNAs that can affect the response to chemotherapy agents in GC.

Table 6.

Various lncRNAs that affect chemotherapy response in gastric cancer

lncRNA Expression in GC Target Drug Model Type of cell line Ref.
HOX transcript antisense RNA (HOTAIR) down miR-34a cisplatin in vivo SGC7901, MGC803, GES1 421
lncR-D63785 down miR-422a doxorubicin in vitro, in vivo BGC823 422
LOC_006753 up PI3K/AKT/mTOR cisplatin, 5-FU in vivo SGC-7901/5-FU, SGC-7901/DDP 423
PVT-1 up cisplatin in vivo BGC823, SGC7901 424
SNGH3 up miR-3619-5p cisplatin in vitro SGC7901 and BGC823 425
lncRNA UCA1 up EZH2 and activation of PI3K/AKT pathway cisplatin human HGC27, MKN45, MGC803, MKN28, AGS, SGC7901 GES-1 414
FAM83H-AS1 up Wnt/β-catenin signaling cisplatin, 5-FU human GES-1 SNU216, BGC823, SGC7901 419

lncRNAs and response to chemotherapy in PC

The lncRNA LINC00346 was upregulated in non-small cell lung and bladder cancer, and it was proposed to be a tumor promoter in these cancer types.426,427 New research has shown overexpression of LINC00346 in PC, which could be a prognostic marker.428 Overexpression of LINC00346 was correlated with shorter overall survival in PC patients.428

The function of LINC00346 in PC was investigated by Shi et al.426 The effects of overexpression and knockdown of LINC00346 on proliferation, apoptosis, cell cycle, and resistance to gemcitabine were studied in PC cell lines. To explore the possible miRNAs that could be affected by LINC00346, a luciferase reporter assay, bioinformatics analysis, and RIP were employed. LINC00346 overexpression significantly increased PC cell proliferation, colony formation, and tumorigenesis. Conversely, LINC00346 knockdown suppressed the proliferation of PC cells and resulted in G2/M cell cycle arrest. LINC00346 depletion increased the sensitivity of PC cells and xenograft tumors to gemcitabine. Mechanistic analysis showed that LINC00346 acted as a miR-188-3p sponge and blocked the downregulation of BRD4 (bromodomain-containing protein 4) caused by miR-188-3p in PC cells. A clinical study in PC tissue samples revealed a negative association between miR-188-3p and LINC00346. Rescue studies confirmed that LINC00346 attenuated the chemosensitizing and growth-inhibiting properties of miR-188-3p in PC. Silencing of BRD4 abrogated the increased colony formation and cell proliferation caused by LINC00346. They concluded that LINC00346 promoted PC resistance to gemcitabine by antagonizing miR-188-3p and increasing BRD4. LINC00346 targeting could boost the therapeutic effectiveness of gemcitabine in PC therapy.426

Table 7 lists some lncRNAs that could affect the response to chemotherapy agents in pancraatic cancer.

Table 7.

Various lncRNAs that affect the chemotherapy response in pancreatic cancer

lncRNA Expression in pancreatic cancer Target Drug Model Type of cell line Ref.
LINC00346 up BRD4 gemcitabine in vitro, in vivo PANC-1, MIA PaCa-2, Capan-1, BxPC-3 426
SBF2-AS1 up miR-142-3p gemcitabine in vitro, in vivo AsPC-1/GEM, PANC-1/GEM 429

lncRNAs and response to chemotherapy in hepatic cancer

The lncRNA GAS5 (growth arrest-specific 5) was first identified to be involved in cell growth arrest.430 Recent studies have demonstrated abnormal expression of GAS5 in a variety of human cancers. Researchers have shown downregulation of GAS5 in several cancer types, such as HCC, breast cancer, bladder cancer, and prostate cancer.431,432 Decreased expression levels of GAS5 were detected in HCC cell lines and tissues compared to matched tissue samples. Low expression of GAS5 was associated with a shorter survival rate and lymph node metastasis in HCC patients. Cell cycle progression was inhibited by overexpression of GAS5, while proliferation and cell viability was enhanced by knockdown of GAS5 in several cancer cells, such as lung, gastric, and breast cancer.433,434

Wang et al.435 investigated the role of lncRNA GAS5 and its molecular mechanism in HCC. They measured miR-21, GAS5, and PTEN levels using qRT-PCR. They used MTT and cell counting assays to measure proliferation in vitro, together with an in vivo xenograft mouse model. RIP and luciferase reporter assays were used to evaluate the correlation between GAS5 and miR-21. They demonstrated that GAS5 was underexpressed in tumor tissues as well as HCC cell lines. GAS5 knockdown was correlated with increased proliferation of HCC cells in vitro and in tumors in vivo. GAS5 knockdown also increased DOX resistance in HCC cells via acting as a sponge to silence miR-21, thereby resulting in PTEN overexpression. Their data revealed that GAS5 was a tumor suppressor in HCC via regulating the miR-21/PTEN signaling axis and indicated a possible role of GAS5 in HCC treatment.435

The lncRNA called cancer susceptibility candidate 2 (CASC2) is situated on chromosome 10q26 and has been found to act as a tumor suppressor in several human cancer types.436 CASC2 has been found to be downregulated in HCC,437 endometrial cancer,438 and prostate cancer.439 However, the role of CASC2 in HCC needs to be better understood. As mentioned above, many lncRNAs act by sponging certain specific miRNAs, and they thereby can affect the expression of the target genes of these miRNAs.440,441 For instance, CASC2 was found to act as a competing endogenous RNA (ceRNA) to sponge miR-18a and therefore upregulate the target of miR-18a, which is PTEN. This could increase the chemosensitivity of cervical cancer cells to cisplatin.441,442 By inhibiting autophagy and enhancing cell death, CASC2 was shown to function as a sponge of miR193a-5p and to increase glioma cell sensitivity to temozolomide. CASC2 could be a new therapeutic target for chemoresistant GC, since its overexpression increased the cisplatin sensitivity of GC cells by sponging miR-19a.443

The function and mechanism of CASC2 were investigated in a study by Liu et al. who looked at cisplatin-resistant HCC cells. The results showed that CASC2 was downregulated in HCC cells and tissues, particularly in those that were cisplatin resistant. Lower expression of CASC2 was correlated with shorter survival in HCC patients. CASC2 overexpression sensitized cisplatin-resistant HCC cell lines (SMMC-7721/cisplatin and Huh7/cisplatin) to cisplatin. Mechanistically, CASC2 increased the cisplatin sensitivity of HCC cells by sponging miR-222. These results showed that upregulation of CASC2 reduced cisplatin resistance in HCC by sponging miR-222, and could provide a route to overcome chemoresistance in HCC patients.444

The lncRNA celled TPTEP1 (transmembrane phosphatase with tensin homology pseudogene 1) has been found to have lower expression in kidney, liver, lung, and stomach cancer, because it is silenced by DNA methylation and has three transcript variants.445 The expression of TPTEP1 could be restored by histone deacetylase inhibitors and by DNA demethylation.445 Many studies have shown the effects of epigenetic alterations, such as DNA methylation, on the susceptibility of various cancers to chemotherapy,446 including cisplatin sensitivity in cancer cells.447,448

A study by Ding et al.449 explored the mechanism and differential expression of various lncRNAs using RNA sequencing (RNA-seq) in HCC cells during cisplatin therapy. They identified TPTEP1 as an important lncRNA in HCC cell lines (QYG-7703 and MHCC97H). Colony formation, qRT-PCR, cell invasion, cell proliferation, and flow cytometry assays were employed. Gain- and loss-of-function analysis, RNA pull-down, subcellular fractionation, western blotting, RIP, and dual-luciferase reporter assays were used to investigate the mechanism by which TPTEP1 could sensitize HCC cells to cisplatin. A subcutaneous xenograft animal model of HCC was used in vivo. qRT-PCR was used to detect TPTEP1 expression levels in clinical tumor specimens. The lncRNA TPTEP1 was found to be overexpressed in HCC cells treated with cisplatin and correlated with cisplatin-mediated apoptosis. TPTEP1 upregulation suppressed, whereas TPTEP1 knockdown increased, the proliferation, invasion, and tumorigenicity of HCC cells. TPTEP1 was found to exert its anti-tumor activity by interaction with signal transducer and transcription activator 3 (STAT3), where it inhibited STAT3 phosphorylation, homodimerization, nuclear translocation, and transcription of downstream genes. Furthermore, in an in vivo subcutaneous xenograft model of HCC, TPTEP1 upregulation clearly suppressed tumor growth in vivo. This agreed with the observation that TPTEP1 is often underexpressed in HCC tissues in comparison to normal liver samples. lncRNA TPTEP1 inhibited HCC development via altering interleukin (IL)-6/STAT3 signaling, and it could improve the response to chemotherapy.449

Table 8 lists some lncRNAs that could affect the response to chemotherapy agents in HCC.

Table 8.

Various lncRNAs that can affect chemotherapy response in hepatic cancer

lncRNA Expression in HCC Target Drug Model Type of cell line Ref.
lncRNA PVT1 down IL-6/STAT3 signaling cisplatin in vitro, in vivo HepG2, SMMC-7721, QGY-7703, Huh-7, MHCC97h, SNU-449, Sk-hep1, L02 449
NR2F1-AS up ABCC1 oxaliplatin in vitro, in vivo Huh7, HepG2, Lo-2 450
p34710_v4 up JAK-STAT signaling pathway teriflunomide in vivo SMMC-7721 451
LINC000607 down NF-κB p65/p53 signaling 5-FU, doxorubicin in vitro, in vivo MHCC97H, HCCLM3, PLC, Hep3B, HepG2, 7721 452
KCNQ1OT1 up miR-7-5p/ABCC1 axis oxaliplatin in vivo SMMC-7721, Huh7, SK-Hep-1, HepG2, Lo-2 453
TUC338 up RASAL1 sorafenib in vivo HepG2, SMMC-7721, BEK-7402, Hep3B, Huh-7, L02 454
Growth arrest-specific 5 (GAS5) down miR-21-PTEN signaling pathway doxorubicin in vitro, in vivo HepG2 HepB3, LO2 435
FOXD2-AS1 up TMEM9 expression sorafenib human, in vivo HepG2, HUH7 455
CASC2 up miR-222 cispatin in vivo, human Huh7, SMMC-7721, HL-7702 456

lncRNAs and response to chemotherapy in CRC

BCYRN1 (brain cytoplasmic RNA 1) is a recently discovered lncRNA, which is activated by c-MYC.457 BCYRN1 has been reported to be overexpressed in several cancer types,458 such as HCC,459 GC,460 and lung cancer,457 compared to healthy control tissues. Moreover, BCYRN1 also has a role in smooth muscle cell differentiation and vascularization within the cardiovascular system.461,462

Yang et al.463 measured the expression levels of BCYRN1 in CRC tumor tissues and cell lines using RT-PCR. They used BCYRN1 knockdown in CRC cells to measure proliferation by 5-ethynyl-2′-deoxyuridine (EdU), CCK-8, and expression of proliferating cell nuclear antigen (PCNA) and Ki-67. Cell invasion and migration were assessed by a transwell assay and scratch wound healing. Flow cytometry analysis was used to determine whether BCYRN1 affected apoptosis. A dual-luciferase reporter assay was used to detect competitive binding of BCYRN1 to miR-204-3p, and in vivo experiments were conducted to assess the effects of BCYRN1 on tumor development. Rescue experiments confirmed that BCYRN1 could sponge miR-204-3p and thereby affect KRAS expression in CRC. Expression levels of BCYRN1 were higher in CRC cell lines and tumor tissues compared to normal intestinal epithelial cells and tissues. Knockdown of BCYRN1 inhibited proliferation, migration, and invasion and increased apoptosis. Furthermore, bioinformatics and a dual-luciferase reporter assay showed that BCYRN1 could competitively bind to miR-204-3p to promote CRC growth. Further experiments showed that miR-204-3p overexpression abrogated the effects of BCYRN1 on CRC. A dual-luciferase reporter assay and TargetScan suggested that KRAS was the miR-204-3p target gene. Tumorigenesis studies in a mouse model showed that tumor development was inhibited by BCYRN1 downregulation. Therefore, BCYRN1 modulates the KRAS/miR-204-3p axis and plays a tumor promoter function in CRC.463

Taurine upregulated gene 1 (TUG1) was initially characterized as a transcript that was overexpressed in the presence of taurine, and then found to be a lncRNA with a role in embryonic retinal growth.464 Moreover, TUG1 overexpression was detected in gastric, bladder, and cervical cancer.465, 466, 467 Alternatively, TUG1 was downregulated in non-small cell lung cancer,468 suggesting contrasting functions in different types of cancer. Researchers recently observed that TUG1 could induce methotrexate resistance in CRC via affecting the CPEB2/miR-186 axis.469

Wang et al.470 reported that the lncRNA TUG1 was associated with 5-FU resistance in CRC samples. TUG1 was substantially increased in samples from recurrent CRC patients. Kaplan-Meier survival analysis found that overexpression of TUG1 in CRC specimens was correlated with an increased risk of disease recurrence. In CRC cell lines, TUG1 knockdown could re-sensitize the cells to the effects of 5-FU. Additionally, bioinformatics research demonstrated that miR-197-3p could directly bind to TUG1, indicating that TUG1 might act as a miR-197-3p sponging ceRNA. TYMS (thymidylate synthase) was shown to be a direct target of miR-197-3p in CRC cells. The authors concluded that TUG1 could increase 5-FU resistance in CRC via the miR-197-3p/TYMS axis.470

Table 9 lists some lncRNAs that could affect the response to chemotherapy agents in CRC.

Table 9.

Various lncRNAs that affect chemotherapy response in colorectal cancer

lncRNA Expression in colorectal cancer Target Drug Model Type of cell line Ref.
PCAT6 up HMGA2/PI3K signaling 5-FU in vitro, in vivo HCT116, HT-29, SW620, SW480, DLD-1, RKO, LoVo, HEK293, CCD-112CoN 471
TUG1 up sponging miR-197-3p 5-FU in vivo HCT8Fu, HCT8 470
TUG1 up miR186/CPEB2 methotrexate in vivo HT-29-P, HT-29-R, HCT-8 469
LINC00473 up miR-15a Taxol in vitro, in vivo FHC, HCT116, HCT116/Taxol, SW620, LoVo 443

circRNAs and response to chemotherapy in GI cancer

The circ-ARHGAP26, known as circ_0074362, is located on chromosome 5 between location 142894237 and 142932125, with a length of 37,888 bp, and it has been detected in normal gastric cells and tissues.472,473 Microarray analysis demonstrated that circ-ARHGAP26 was overexpressed in GC tissues in comparison with paired normal tissues, while the expression of circ-ARHGAP26 in GC tissues was found to be downregulated in other studies.473,474

A study by Wangxia et al.475 reported that circ-ARHGAP26 was overexpressed in several GC cell lines (HGC-27, NCI-N87, AGS, SGC-7901, and BGC-823) compared to normal GSE-1 cells. Knockdown of circ-ARHGAP26 in HGC-27 cells decreased proliferation as measured by the CCK-8 assay at 48 and 72h, while flow cytometry showed that apoptosis was increased at 72 h. Western blotting showed that pro-apoptotic caspase-3 was increased while anti-apoptotic Bcl-2 was reduced at 72 h in the circ-ARHGAP26(−) group. Similar results were also found in AGS cells.475

It was shown that circRNAs may function as competitive inhibitors through binding to miRNAs, also known as “miRNA sponges,” or alternatively these ncRNAs may function as target mimics and suppress the activity of certain miRNAs.476, 477, 478, 479 The circRNAs have binding sites for the complementary miRNA, which is located in the 3′ UTR or non-coding transcript of a particular gene.480

circ-PVT1 has been found to act as a ceRNA in an oral squamous carcinoma cell (OSCC) line to affect the miR-125b/STAT3 axis and thereby increase proliferation.481 Upregulation of circ-PVT1 promoted invasion and proliferation in non-small cell lung cancer by activating E2F2 signaling.482 Recent results showed that upregulation of circ-PVT1 also increased proliferation in GC and could be a prognostic marker.483 A recent publication suggested that circ-PVT1 could increase resistance to DOX and cisplatin by regulating ABCB1 in osteosarcoma cells.484

Liu et al.115 found that circ-PVT1 was overexpressed in PTX-resistant GC cells and tissues, and it could reduce the expression of miR-124-3p. Reduction of circ-PVT1 expression promoted PTX sensitivity in PTX-resistant GC cells. ZEB was suggested to be a direct target of miR-124-3p, and therefore circ-PVT1 could increase ZEB1 expression by sponging miR-124-3p. circ-PVT1 knockdown led to an increase in PTX sensitivity of GC tumors in vivo. Taken together, circ-PVT1 increased resistance to PTX by upregulating ZEB by sponging miR-124-3p and could be a target for GC treatment.115

Table 10 lists some circRNAs that can affect the response to chemotherapy agents in GI cancer.

Table 10.

Various circular RNAs that affect chemotherapy response in GI cancer

Cancer Circular RNA Expression in GI cancers Target Drug Model Type of cell line Ref.
Esophageal circ-LARP4 down miR-1323 in vitro HEEPIC, ESCC (ECA109, TE-1, KYSE30, KYSE410) 485
Gastric rc-ARHGAP26 up not mentioned in vitro HGC-27, AGS, SGC-7901, BGC-823, NCI-N87, GSE-1 475
Gastric circ-PVT1 up ZEB1, miR-124-3p paclitaxel in vitro MKN-45, HGC-27, MGC-803, AGS 115
Gastric circ-MCTP2 down MTMR3, miR-99a-5p cisplatin in vitro BGC823, SGC7901, SGC7901CDDP 486
Gastric ciRS-7 up miR-7, PTEN, PI3K In vitro MGC-803, HGC-27, GES-1 487
Gastric circ-FN1 up miR-182-5p cisplatin in vitro SGC7901CDDP, BGC823C, DDP, SGC7901, BGC823 488
Hepatocellular circ-FBXO11 up miR-605, FOXO3 oxaliplatin human HepG2, Hep3B, SMMC-7721, Huh7, Lo-2 489
Pancreatic circ-HIPK3 up miR-330-5p, RASSF1 gemcitabine in vitro PANC-1, SW 1990 490
Colorectal circ_0032833 down miR-125-5p, MSI1 5-FU, oxaliplatin in vitro HCT116 491
Colorectal hsa_circ_001680 up miR-340, BMI1 irinotecan in vitro FHC, HCT116, SW480, HCT15, SW620, CACO2, DLD1, LOVO, HT29, HCT8, RKO 492
Colorectal ciRS-122 up miR-122, PKM2 oxaliplatin in vitro SW480, HCT116, HEK293T 493

Conclusions

Surgery is considered to be the main therapeutic approach for most GI cancers, but many cancers are diagnosed at an advanced stage, where they are then considered to be inoperable. Although chemotherapy and radiotherapy are then available, these approaches seldom result in a complete cure. Thus, there is an urgent for major improvements in chemotherapy regimens in GI treatment. The poor response of most GI cancers to chemotherapy can be attributed to multiple mechanisms. Chemoresistance can have many causes such as reduced drug uptake, increased drug efflux, less activation of pro-drugs, changes in the molecular targets, better DNA repair mechanisms, disruption of the pro-apoptotic machinery, or overexpression of anti-apoptotic genes. Many new techniques derived from biotechnology and molecular biology, such as bioinformatics analysis, genome modification, high-throughput sequencing, pharmaceutical chemistry, and mouse modeling, have been used to reveal the wide involvement of a range of ncRNAs in the initiation, biology, progression, and response to treatment of various cancers. Put differently, the expression patterns of ncRNAs are very different in tumor tissue compared to the corresponding normal tissue. This enables ncRNAs to be used as biomarkers for disease progression and tumor stage. In addition, many lncRNAs, miRNAs, and circRNAs are specifically related to prognosis and therapeutic response, because they control resistance to radiotherapy and chemotherapy. Researchers have identified several genes in animal models with the use of single-stranded antisense oligonucleotides (ASOs) and double-stranded RNAi. As an example, inhibiting MALAT1 with ASO may promote differentiation and inhibit metastasis in mouse models of cancer,494,495 including lung cancer.496

Researchers have attempted to employ ncRNA systems or carriers as therapeutic strategies, including oncolytic adenoviruses, nanoparticles (NPs), and direct delivery of modified ncRNAs. Some clinical trials have been based on ncRNA-mediated precision medicine (https://clinicaltrials.gov). Some clinical trials have reached phase 3, such as miR-31-5p and miR-31-3p for CRC,497 as well as miR-200 and miR-21 for oral cancer.498,499 Moreover, some circRNAs and lncRNAs have begun to be evaluated in trials, such as MALAT1 for breast cancer494,495 or lung cancer,500 and HOTAIR for thyroid cancer.501 Furthermore, siRNAs are usually incorporated into lipid nanoparticles for delivery, and these have been tested in clinical trials. Phase 2 clinical trials of siRNAs include the use of Atu027 to knock down the PKN3 gene to inhibit migration in metastatic pancreatic adenocarcinoma, and DCR-MYC to knock down the MYC gene to inhibit the cell cycle in HCC.3 In the future, ncRNAs and modifiers may be used to improve the response of various cancers to therapies. However, there are many challenges to overcome before these techniques can be widely applied in clinics. First, ncRNAs vary widely in their length and number of nucleotides, as well as their modes of action. Second, ncRNAs are very different in different tumors, and therefore the selection of the appropriate target from multiple candidates would be difficult. Additional information on the functional parameters and genomic strategies is needed from fundamental and translational research. Third, besides the need for a suitable target, an effective delivery strategy that allows specific binding affinity is not easy. The tumor microenvironment has a highly heterogeneous composition, which makes application and delivery of ncRNAs very difficult. These challenges include off-target effects, low transfection efficacy, and a short half-life due to RNA degradation and instability. The present delivery systems will need to be significantly improved in future studies to overcome these challenges.

The use of targeted carriers or smart carriers may be an alternative option. For instance, the conjugation of nanoparticles to tissue-specific receptors could improve the target specificity. Another challenge is the bioavailability of nucleic acid-based therapeutics inside the tumor tissue, and reduction of cytotoxicity to normal tissues. Moreover, assay techniques must be agreed upon to ensure high quality and calculate the relative efficiency. Finally, most of the studies on the use of ncRNAs and modifiers are still in the preclinical phase, and most of them have been limited to a single type or a few types of cancer. After deciding on a suitable gene candidate and an efficient delivery carrier, further efforts will be needed to evaluate the patient responses to the clinical therapies. This is essential to understand the long-term outcomes of new cancer treatments that have never been assessed before. If these challenges can be overcome the use of ncRNAs/modifiers, either as tumor suppressors or inhibitors of oncogenes, could be an addition to standard chemotherapy or radiotherapy regimens to deal with resistance and improve patient survival in the coming years.

Acknowledgments

Author contributions

H.M. and M.R.H. contributed to conception, design, statistical analysis, and drafting of the manuscript. F.D., S.M.A.M., Nikta Rabiei, R.F., Negin Rabiei, H.P., and M.V. contributed to data collection and manuscript drafting. All authors approved the final version of the manuscript for submission.

Declaration of interests

M.R.H. declares the following potential conflicts of interest. Scientific Advisory Boards: Transdermal Cap, Inc, Cleveland, OH, USA; BeWell Global, Inc, Wan Chai, Hong Kong; Hologenix, Inc, Santa Monica, CA, USA; LumiThera, Inc, Poulsbo, WA, USA; Vielight, Toronto, ON, Canada; Bright Photomedicine, Sao Paulo, Brazil; Quantum Dynamics, LLC, Cambridge, MA, USA; Global Photon, Inc, Bee Cave, TX, USA; Medical Coherence, Boston, MA, USA; NeuroThera, Newark, DE, USA; JOOVV, Inc, Minneapolis-St. Paul, MN, USA; AIRx Medical, Pleasanton, CA, USA; FIR Industries, Inc, Ramsey, NJ, USA; UVLRx Therapeutics, Oldsmar, FL, USA; Ultralux UV, Inc, Lansing, MI, USA; Illumiheal and PetThera, Shoreline, WA, USA; MB Lasertherapy, Houston, TX, USA; ARRC LED, San Clemente, CA, USA; Varuna Biomedical Corp, Incline Village, NV, USA; Niraxx Light Therapeutics, Inc, Boston, MA, USA. Consulting: Lexington International, LLC, Boca Raton, FL, USA; USHIO Corp, Japan; Merck KGaA, Darmstadt, Germany; Philips Electronics Nederland BV, Eindhoven, the Netherlands; Johnson & Johnson, Inc, Philadelphia, PA, USA; Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany. Stockholdings: Global Photon, Inc, Bee Cave, TX; Mitonix, Newark, DE. The remaining authors declare no competing interests.

Contributor Information

Neda Rahimian, Email: rahimian.n@iums.ac.ir.

Michael R. Hamblin, Email: hamblin.lab@gmail.com.

Hamed Mirzaei, Email: mirzaei-h@kaums.ac.ir.

References

  • 1.Kamb A., Wee S., Lengauer C. Why is cancer drug discovery so difficult? Nat. Rev. Drug Discov. 2007;6:115–120. doi: 10.1038/nrd2155. [DOI] [PubMed] [Google Scholar]
  • 2.Lievens Y., Gospodarowicz M., Grover S., Jaffray D., Rodin D., Torode J., Yap M.L., Zubizarreta E., GIRO Steering and Advisory Committees Global impact of radiotherapy in oncology: Saving one million lives by 2035. Radiother. Oncol. 2017;125:175–177. doi: 10.1016/j.radonc.2017.10.027. [DOI] [PubMed] [Google Scholar]
  • 3.Wang W.T., Han C., Sun Y.M., Chen T.Q., Chen Y.Q. Noncoding RNAs in cancer therapy resistance and targeted drug development. J. Hematol. Oncol. 2019;12:55. doi: 10.1186/s13045-019-0748-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Guttman M., Rinn J.L. Modular regulatory principles of large non-coding RNAs. Nature. 2012;482:339–346. doi: 10.1038/nature10887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Esteller M. Non-coding RNAs in human disease. Nat. Rev. Genet. 2011;12:861–874. doi: 10.1038/nrg3074. [DOI] [PubMed] [Google Scholar]
  • 6.Matsui M., Corey D.R. Non-coding RNAs as drug targets. Nat. Rev. Drug Discov. 2017;16:167–179. doi: 10.1038/nrd.2016.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Yoon S., Rossi J.J. Therapeutic potential of small activating RNAs (saRNAs) in human cancers. Curr. Pharm. Biotechnol. 2018;19:604–610. doi: 10.2174/1389201019666180528084059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Dong Y., He D., Peng Z., Peng W., Shi W., Wang J., Li B., Zhang C., Duan C. Circular RNAs in cancer: An emerging key player. J. Hematol. Oncol. 2017;10:2. doi: 10.1186/s13045-016-0370-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Jafarpour S.M., Safaei M., Mohseni M., Salimian M., Aliasgharzadeh A., Farhood B. The radioprotective effects of curcumin and trehalose against genetic damage caused by I-131. Indian J. Nucl. Med. 2018;33:99–104. doi: 10.4103/ijnm.IJNM_158_17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Vo J.N., Cieslik M., Zhang Y., Shukla S., Xiao L., Zhang Y., Wu Y.M., Dhanasekaran S.M., Engelke C.G., Cao X. The landscape of circular RNA in cancer. Cell. 2019;176:869–881.e13. doi: 10.1016/j.cell.2018.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Li X., Yang L., Chen L.L. The biogenesis, functions, and challenges of circular RNAs. Mol. Cell. 2018;71:428–442. doi: 10.1016/j.molcel.2018.06.034. [DOI] [PubMed] [Google Scholar]
  • 12.Yu A.M., Jian C., Yu A.H., Tu M.J. RNA therapy: Are we using the right molecules? Pharmacol. Ther. 2019;196:91–104. doi: 10.1016/j.pharmthera.2018.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hayes J., Peruzzi P.P., Lawler S. MicroRNAs in cancer: biomarkers, functions and therapy. Trends Mol. Med. 2014;20:460–469. doi: 10.1016/j.molmed.2014.06.005. [DOI] [PubMed] [Google Scholar]
  • 14.Sartorelli V., Lauberth S.M. Enhancer RNAs are an important regulatory layer of the epigenome. Nat. Struct. Mol. Biol. 2020;27:521–528. doi: 10.1038/s41594-020-0446-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ørom U.A. Springer; 2017. Enhancer RNAs: Methods and Protocols. [Google Scholar]
  • 16.Ye R., Cao C., Xue Y. Enhancer RNA: Biogenesis, function, and regulation. Essays Biochem. 2020;64:883–894. doi: 10.1042/EBC20200014. [DOI] [PubMed] [Google Scholar]
  • 17.Timmons L. The long and short of siRNAs. Mol. Cell. 2002;10:435–437. doi: 10.1016/s1097-2765(02)00657-3. [DOI] [PubMed] [Google Scholar]
  • 18.Zhang M., Xin Y. Circular RNAs: A new frontier for cancer diagnosis and therapy. J. Hematol. Oncol. 2018;11:21. doi: 10.1186/s13045-018-0569-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Guo S., Fesler A., Wang H., Ju J. MicroRNA based prognostic biomarkers in pancreatic cancer. Biomark. Res. 2018;6:18. doi: 10.1186/s40364-018-0131-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Fort R.S., Mathó C., Oliveira-Rizzo C., Garat B., Sotelo-Silveira J.R., Duhagon M.A. An integrated view of the role of miR-130b/301b miRNA cluster in prostate cancer. Exp. Hematol. Oncol. 2018;7:10. doi: 10.1186/s40164-018-0102-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Smolle M.A., Calin H.N., Pichler M., Calin G.A. Noncoding RNAs and immune checkpoints-clinical implications as cancer therapeutics. FEBS J. 2017;284:1952–1966. doi: 10.1111/febs.14030. [DOI] [PubMed] [Google Scholar]
  • 22.Bader A.G. miR-34—A microRNA replacement therapy is headed to the clinic. Front. Genet. 2012;3:120. doi: 10.3389/fgene.2012.00120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Rupaimoole R., Slack F.J. MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat. Rev. Drug Discov. 2017;16:203–222. doi: 10.1038/nrd.2016.246. [DOI] [PubMed] [Google Scholar]
  • 24.Liu B., Liu Y., Zhao L., Pan Y., Shan Y., Li Y., Jia L. Upregulation of microRNA-135b and microRNA-182 promotes chemoresistance of colorectal cancer by targeting ST6GALNAC2 via PI3K/AKT pathway. Mol. Carcinog. 2017;56:2669–2680. doi: 10.1002/mc.22710. [DOI] [PubMed] [Google Scholar]
  • 25.Xiang S., Fang J., Wang S., Deng B., Zhu L. MicroRNA-135b regulates the stability of PTEN and promotes glycolysis by targeting USP13 in human colorectal cancers. Oncol. Rep. 2015;33:1342–1348. doi: 10.3892/or.2014.3694. [DOI] [PubMed] [Google Scholar]
  • 26.Yang W.B., Chen P.H., Hsu T., Fu T.F., Su W.C., Liaw H., Chang W.C., Hung J.J. Sp1-mediated microRNA-182 expression regulates lung cancer progression. Oncotarget. 2014;5:740–753. doi: 10.18632/oncotarget.1608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wang J., Li J., Shen J., Wang C., Yang L., Zhang X. MicroRNA-182 downregulates metastasis suppressor 1 and contributes to metastasis of hepatocellular carcinoma. BMC Cancer. 2012;12:227. doi: 10.1186/1471-2407-12-227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Gao J., Liu X., Yang F., Liu T., Yan Q., Yang X. By inhibiting Ras/Raf/ERK and MMP-9, knockdown of EpCAM inhibits breast cancer cell growth and metastasis. Oncotarget. 2015;6:27187–27198. doi: 10.18632/oncotarget.4551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Yang J., Yang X.S., Zhang Q., Zhuang X., Dong X.K., Jiang Y.H., Tao Y.N., Yang C.H. Downregulated LINC01614 ameliorates hypoxia/reoxygenation-stimulated myocardial injury by directly sponging microRNA-138-5p. Dose Response. 2020;18 doi: 10.1177/1559325820913786. 1559325820913786. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 30.Deng Y., Chen D., Gao F., Lv H., Zhang G., Sun X., Liu L., Mo D., Ma N., Song L. Exosomes derived from microRNA-138-5p-overexpressing bone marrow-derived mesenchymal stem cells confer neuroprotection to astrocytes following ischemic stroke via inhibition of LCN2. J. Biol. Eng. 2019;13:71. doi: 10.1186/s13036-019-0193-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Yu C., Wang M., Li Z., Xiao J., Peng F., Guo X., Deng Y., Jiang J., Sun C. MicroRNA-138-5p regulates pancreatic cancer cell growth through targeting FOXC1. Cell Oncol. (Dordr.) 2015;38:173–181. doi: 10.1007/s13402-014-0200-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yu C., Wang M., Chen M., Huang Y., Jiang J. Upregulation of microRNA-138-5p inhibits pancreatic cancer cell migration and increases chemotherapy sensitivity. Mol. Med. Rep. 2015;12:5135–5140. doi: 10.3892/mmr.2015.4031. [DOI] [PubMed] [Google Scholar]
  • 33.Gao W., Lam J.W., Li J.Z., Chen S.Q., Tsang R.K., Chan J.Y., Wong T.S. MicroRNA-138-5p controls sensitivity of nasopharyngeal carcinoma to radiation by targeting EIF4EBP1. Oncol. Rep. 2017;37:913–920. doi: 10.3892/or.2017.5354. [DOI] [PubMed] [Google Scholar]
  • 34.Roberto G.M., Lira R.C., Delsin L.E., Vieira G.M., Silva M.O., Hakime R.G., Yamashita M.E., Engel E.E., Scrideli C.A., Tone L.G., Brassesco M.S. MicroRNA-138-5p as a worse prognosis biomarker in pediatric, adolescent, and young adult osteosarcoma. Pathol. Oncol. Res. 2020;26:877–883. doi: 10.1007/s12253-019-00633-0. [DOI] [PubMed] [Google Scholar]
  • 35.Zhao C., Ling X., Li X., Hou X., Zhao D. MicroRNA-138-5p inhibits cell migration, invasion and EMT in breast cancer by directly targeting RHBDD1. Breast Cancer. 2019;26:817–825. doi: 10.1007/s12282-019-00989-w. [DOI] [PubMed] [Google Scholar]
  • 36.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. doi: 10.1016/j.prp.2019.01.029. [DOI] [PubMed] [Google Scholar]
  • 37.Xu W., Chen B., Ke D., Chen X. MicroRNA-138-5p targets the NFIB-Snail1 axis to inhibit colorectal cancer cell migration and chemoresistance. Cancer Cell Int. 2020;20:475. doi: 10.1186/s12935-020-01573-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hu G., Chen D., Li X., Yang K., Wang H., Wu W. miR-133b regulates the MET proto-oncogene and inhibits the growth of colorectal cancer cells in vitro and in vivo. Cancer Biol. Ther. 2010;10:190–197. doi: 10.4161/cbt.10.2.12186. [DOI] [PubMed] [Google Scholar]
  • 39.Wang X., Bu J., Liu X., Wang W., Mai W., Lv B., Zou J., Mo X., Li X., Wang J. miR-133b suppresses metastasis by targeting HOXA9 in human colorectal cancer. Oncotarget. 2017;8:63935–63948. doi: 10.18632/oncotarget.19212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Akçakaya P., Ekelund S., Kolosenko I., Caramuta S., Ozata D.M., Xie H., Lindforss U., Olivecrona H., Lui W.O. miR-185 and miR-133b deregulation is associated with overall survival and metastasis in colorectal cancer. Int. J. Oncol. 2011;39:311–318. doi: 10.3892/ijo.2011.1043. [DOI] [PubMed] [Google Scholar]
  • 41.Duan F.T., Qian F., Fang K., Lin K.Y., Wang W.T., Chen Y.Q. miR-133b, a muscle-specific microRNA, is a novel prognostic marker that participates in the progression of human colorectal cancer via regulation of CXCR4 expression. Mol. Cancer. 2013;12:164. doi: 10.1186/1476-4598-12-164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lv L., Li Q., Chen S., Zhang X., Tao X., Tang X., Wang S., Che G., Yu Y., He L. miR-133b suppresses colorectal cancer cell stemness and chemoresistance by targeting methyltransferase DOT1L. Exp. Cell Res. 2019;385:111597. doi: 10.1016/j.yexcr.2019.111597. [DOI] [PubMed] [Google Scholar]
  • 43.Nguyen A.T., Zhang Y. The diverse functions of Dot1 and H3K79 methylation. Genes Dev. 2011;25:1345–1358. doi: 10.1101/gad.2057811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Yan J.W., Lin J.S., He X.X. The emerging role of miR-375 in cancer. Int. J. Cancer. 2014;135:1011–1018. doi: 10.1002/ijc.28563. [DOI] [PubMed] [Google Scholar]
  • 45.Zhao Z., Yang S., Cheng Y., Zhao X. MicroRNA-655 inhibits cell proliferation and invasion in epithelial ovarian cancer by directly targeting vascular endothelial growth factor. Mol. Med. Rep. 2018;18:1878–1884. doi: 10.3892/mmr.2018.9090. [DOI] [PubMed] [Google Scholar]
  • 46.Fan Y.P., Liao J.Z., Lu Y.Q., Tian D.A., Ye F., Zhao P.X., Xiang G.Y., Tang W.X., He X.X. miR-375 and doxorubicin co-delivered by liposomes for combination therapy of hepatocellular carcinoma. Mol. Ther. Nucleic Acids. 2017;7:181–189. doi: 10.1016/j.omtn.2017.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Wang Y., Lieberman R., Pan J., Zhang Q., Du M., Zhang P., Nevalainen M., Kohli M., Shenoy N.K., Meng H. miR-375 induces docetaxel resistance in prostate cancer by targeting SEC23A and YAP1. Mol. Cancer. 2016;15:70. doi: 10.1186/s12943-016-0556-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Gaedcke J., Grade M., Camps J., Søkilde R., Kaczkowski B., Schetter A.J., Difilippantonio M.J., Harris C.C., Ghadimi B.M., Møller S. The rectal cancer microRNAome—MicroRNA expression in rectal cancer and matched normal mucosa. Clin. Cancer Res. 2012;18:4919–4930. doi: 10.1158/1078-0432.CCR-12-0016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Conde-Muiño R., Cano C., Sánchez-Martín V., Herrera A., Comino A., Medina P.P., Palma P., Cuadros M. Preoperative chemoradiotherapy for rectal cancer: The sensitizer role of the association between miR-375 and c-Myc. Oncotarget. 2017;8:82294–82302. doi: 10.18632/oncotarget.19393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Mussnich P., Rosa R., Bianco R., Fusco A., D’Angelo D. miR-199a-5p and miR-375 affect colon cancer cell sensitivity to cetuximab by targeting PHLPP1. Expert Opin. Ther. Targets. 2015;19:1017–1026. doi: 10.1517/14728222.2015.1057569. [DOI] [PubMed] [Google Scholar]
  • 51.Xu X., Chen X., Xu M., Liu X., Pan B., Qin J., Xu T., Zeng K., Pan Y., He B. miR-375-3p suppresses tumorigenesis and partially reverses chemoresistance by targeting YAP1 and SP1 in colorectal cancer cells. Aging (Albany NY) 2019;11:7357–7385. doi: 10.18632/aging.102214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kim H.K., Lim N.J., Jang S.G., Lee G.K. miR-592 and miR-552 can distinguish between primary lung adenocarcinoma and colorectal cancer metastases in the lung. Anticancer Res. 2014;34:2297–2302. [PubMed] [Google Scholar]
  • 53.Sarver A.L., French A.J., Borralho P.M., Thayanithy V., Oberg A.L., Silverstein K.A., Morlan B.W., Riska S.M., Boardman L.A., Cunningham J.M. Human colon cancer profiles show differential microRNA expression depending on mismatch repair status and are characteristic of undifferentiated proliferative states. BMC Cancer. 2009;9:401. doi: 10.1186/1471-2407-9-401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Qu W., Wen X., Su K., Gou W. miR-552 promotes the proliferation, migration and EMT of hepatocellular carcinoma cells by inhibiting AJAP1 expression. J. Cell. Mol. Med. 2019;23:1541–1552. doi: 10.1111/jcmm.14062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.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. doi: 10.1007/s00280-019-03866-7. [DOI] [PubMed] [Google Scholar]
  • 56.Feng B., Dong T.T., Wang L.L., Zhou H.M., Zhao H.C., Dong F., Zheng M.H. Colorectal cancer migration and invasion initiated by microRNA-106a. PLoS ONE. 2012;7:e43452. doi: 10.1371/journal.pone.0043452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Koga Y., Yamazaki N., Yamamoto Y., Yamamoto S., Saito N., Kakugawa Y., Otake Y., Matsumoto M., Matsumura Y. Fecal miR-106a is a useful marker for colorectal cancer patients with false-negative results in immunochemical fecal occult blood test. Cancer Epidemiol. Biomarkers Prev. 2013;22:1844–1852. doi: 10.1158/1055-9965.EPI-13-0512. [DOI] [PubMed] [Google Scholar]
  • 58.Liu G.H., Zhou Z.G., Chen R., Wang M.J., Zhou B., Li Y., Sun X.F. Serum miR-21 and miR-92a as biomarkers in the diagnosis and prognosis of colorectal cancer. Tumour Biol. 2013;34:2175–2181. doi: 10.1007/s13277-013-0753-8. [DOI] [PubMed] [Google Scholar]
  • 59.Díaz R., Silva J., García J.M., Lorenzo Y., García V., Peña C., Rodríguez R., Muñoz C., García F., Bonilla F., Domínguez G. Deregulated expression of miR-106a predicts survival in human colon cancer patients. Genes Chromosomes Cancer. 2008;47:794–802. doi: 10.1002/gcc.20580. [DOI] [PubMed] [Google Scholar]
  • 60.Liu Z., Qin Y., Dong S., Chen X., Huo Z., Zhen Z. Overexpression of miR-106a enhances oxaliplatin sensitivity of colorectal cancer through regulation of FOXQ1. Oncol. Lett. 2020;19:663–670. doi: 10.3892/ol.2019.11151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Nurul-Syakima A.M., Yoke-Kqueen C., Sabariah A.R., Shiran M.S., Singh A., Learn-Han L. Differential microRNA expression and identification of putative miRNA targets and pathways in head and neck cancers. Int. J. Mol. Med. 2011;28:327–336. doi: 10.3892/ijmm.2011.714. [DOI] [PubMed] [Google Scholar]
  • 62.Miyamae M., Komatsu S., Ichikawa D., Kawaguchi T., Hirajima S., Okajima W., Ohashi T., Imamura T., Konishi H., Shiozaki A. Plasma microRNA profiles: Identification of miR-744 as a novel diagnostic and prognostic biomarker in pancreatic cancer. Br. J. Cancer. 2015;113:1467–1476. doi: 10.1038/bjc.2015.366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Zhou W., Li Y., Gou S., Xiong J., Wu H., Wang C., Yan H., Liu T. miR-744 increases tumorigenicity of pancreatic cancer by activating Wnt/β-catenin pathway. Oncotarget. 2015;6:37557–37569. doi: 10.18632/oncotarget.5317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Song M.Y., Pan K.F., Su H.J., Zhang L., Ma J.L., Li J.Y., Yuasa Y., Kang D., Kim Y.S., You W.C. Identification of serum microRNAs as novel non-invasive biomarkers for early detection of gastric cancer. PLoS ONE. 2012;7:e33608. doi: 10.1371/journal.pone.0033608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Zhou Y., He A., Zhang L., Yi G. miR-744 mediates the oxaliplatin chemoresistance in colorectal cancer through inhibiting BIN1. Neoplasma. 2020;67:296–303. doi: 10.4149/neo_2019_190508N411. [DOI] [PubMed] [Google Scholar]
  • 66.Sharma T., Hamilton R., Mandal C.C. miR-214: A potential biomarker and therapeutic for different cancers. Future Oncol. 2015;11:349–363. doi: 10.2217/fon.14.193. [DOI] [PubMed] [Google Scholar]
  • 67.Penna E., Orso F., Taverna D. miR-214 as a key hub that controls cancer networks: Small player, multiple functions. J. Invest. Dermatol. 2015;135:960–969. doi: 10.1038/jid.2014.479. [DOI] [PubMed] [Google Scholar]
  • 68.Yang H., Kong W., He L., Zhao J.J., O’Donnell J.D., Wang J., Wenham R.M., Coppola D., Kruk P.A., Nicosia S.V., Cheng J.Q. MicroRNA expression profiling in human ovarian cancer: miR-214 induces cell survival and cisplatin resistance by targeting PTEN. Cancer Res. 2008;68:425–433. doi: 10.1158/0008-5472.CAN-07-2488. [DOI] [PubMed] [Google Scholar]
  • 69.Long L.M., He B.F., Huang G.Q., Guo Y.H., Liu Y.S., Huo J.R. MicroRNA-214 functions as a tumor suppressor in human colon cancer via the suppression of ADP-ribosylation factor-like protein 2. Oncol. Lett. 2015;9:645–650. doi: 10.3892/ol.2014.2746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Momose K., Minami A., Shimono Y., Mizutani K., Nobutani K., Azuma T., Takai Y. miR-214 and hypoxia down-regulate Necl-2/CADM1 and enhance ErbB2/ErbB3 signaling. Genes Cells. 2013;18:195–202. doi: 10.1111/gtc.12027. [DOI] [PubMed] [Google Scholar]
  • 71.Yang Y., Bao Y., Yang G.K., Wan J., Du L.J., Ma Z.H. miR-214 sensitizes human colon cancer cells to 5-FU by targeting Hsp27. Cell. Mol. Biol. Lett. 2019;24:22. doi: 10.1186/s11658-019-0143-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Song C., Lu P., Sun G., Yang L., Wang Z., Wang Z. miR-34a sensitizes lung cancer cells to cisplatin via p53/miR-34a/MYCN axis. Biochem. Biophys. Res. Commun. 2017;482:22–27. doi: 10.1016/j.bbrc.2016.11.037. [DOI] [PubMed] [Google Scholar]
  • 73.Gao J., Li N., Dong Y., Li S., Xu L., Li X., Li Y., Li Z., Ng S.S., Sung J.J. miR-34a-5p suppresses colorectal cancer metastasis and predicts recurrence in patients with stage II/III colorectal cancer. Oncogene. 2015;34:4142–4152. doi: 10.1038/onc.2014.348. [DOI] [PubMed] [Google Scholar]
  • 74.Wang L., Bu P., Ai Y., Srinivasan T., Chen H.J., Xiang K., Lipkin S.M., Shen X. A long non-coding RNA targets microRNA miR-34a to regulate colon cancer stem cell asymmetric division. eLife. 2016;5:e14620. doi: 10.7554/eLife.14620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lai M., Du G., Shi R., Yao J., Yang G., Wei Y., Zhang D., Xu Z., Zhang R., Li Y. miR-34a inhibits migration and invasion by regulating the SIRT1/p53 pathway in human SW480 cells. Mol. Med. Rep. 2015;11:3301–3307. doi: 10.3892/mmr.2015.3182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Bu P., Chen K.Y., Chen J.H., Wang L., Walters J., Shin Y.J., Goerger J.P., Sun J., Witherspoon M., Rakhilin N. A microRNA miR-34a-regulated bimodal switch targets Notch in colon cancer stem cells. Cell Stem Cell. 2013;12:602–615. doi: 10.1016/j.stem.2013.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Li C., Wang Y., Lu S., Zhang Z., Meng H., Liang L., Zhang Y., Song B. miR-34a inhibits colon cancer proliferation and metastasis by inhibiting platelet-derived growth factor receptor α. Mol. Med. Rep. 2015;12:7072–7078. doi: 10.3892/mmr.2015.4263. [DOI] [PubMed] [Google Scholar]
  • 78.Li Y., Gong P., Hou J.X., Huang W., Ma X.P., Wang Y.L., Li J., Cui X.B., Li N. miR-34a regulates multidrug resistance via positively modulating OAZ2 signaling in colon cancer cells. J. Immunol. Res. 2018;2018:7498514. doi: 10.1155/2018/7498514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Amankwatia E.B., Chakravarty P., Carey F.A., Weidlich S., Steele R.J., Munro A.J., Wolf C.R., Smith G. MicroRNA-224 is associated with colorectal cancer progression and response to 5-fluorouracil-based chemotherapy by KRAS-dependent and -independent mechanisms. Br. J. Cancer. 2015;112:1480–1490. doi: 10.1038/bjc.2015.125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Li Y., Xin S., Wu H., Xing C., Duan L., Sun W., Hu X., Lin R., Zhang F. High expression of microRNA-31 and its host gene LOC554202 predict favorable outcomes in patients with colorectal cancer treated with oxaliplatin. Oncol. Rep. 2018;40:1706–1724. doi: 10.3892/or.2018.6571. [DOI] [PubMed] [Google Scholar]
  • 81.Hu J., Cai G., Xu Y., Cai S. The plasma microRNA miR-1914∗ and -1915 suppresses chemoresistant in colorectal cancer patients by down-regulating NFIX. Curr. Mol. Med. 2016;16:70–82. doi: 10.2174/1566524016666151222144656. [DOI] [PubMed] [Google Scholar]
  • 82.Molina-Pinelo S., Carnero A., Rivera F., Estevez-Garcia P., Bozada J.M., Limon M.L., Benavent M., Gomez J., Pastor M.D., Chaves M. miR-107 and miR-99a-3p predict chemotherapy response in patients with advanced colorectal cancer. BMC Cancer. 2014;14:656. doi: 10.1186/1471-2407-14-656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Gu C., Cai J., Xu Z., Zhou S., Ye L., Yan Q., Zhang Y., Fang Y., Liu Y., Tu C. miR-532-3p suppresses colorectal cancer progression by disrupting the ETS1/TGM2 axis-mediated Wnt/β-catenin signaling. Cell Death Dis. 2019;10:739. doi: 10.1038/s41419-019-1962-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Zhang Y., Talmon G., Wang J. MicroRNA-587 antagonizes 5-FU-induced apoptosis and confers drug resistance by regulating PPP2R1B expression in colorectal cancer. Cell Death Dis. 2015;6:e1845. doi: 10.1038/cddis.2015.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Fang L., Li H., Wang L., Hu J., Jin T., Wang J., Yang B.B. MicroRNA-17-5p promotes chemotherapeutic drug resistance and tumour metastasis of colorectal cancer by repressing PTEN expression. Oncotarget. 2014;5:2974–2987. doi: 10.18632/oncotarget.1614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Qian X., Yu J., Yin Y., He J., Wang L., Li Q., Zhang L.Q., Li C.Y., Shi Z.M., Xu Q. MicroRNA-143 inhibits tumor growth and angiogenesis and sensitizes chemosensitivity to oxaliplatin in colorectal cancers. Cell Cycle. 2013;12:1385–1394. doi: 10.4161/cc.24477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Feng C., Zhang L., Sun Y., Li X., Zhan L., Lou Y., Wang Y., Liu L., Zhang Y. GDPD5, a target of miR-195-5p, is associated with metastasis and chemoresistance in colorectal cancer. Biomed. Pharmacoth. 2018;101:945–952. doi: 10.1016/j.biopha.2018.03.028. [DOI] [PubMed] [Google Scholar]
  • 88.Wang S.J., Cao Y.F., Yang Z.Q., Jiang Z.Y., Cai B., Guo J., Zhang S., Zhang X.L., Gao F. MicroRNA-93-5p increases multidrug resistance in human colorectal carcinoma cells by downregulating cyclin dependent kinase inhibitor 1A gene expression. Oncol. Lett. 2017;13:722–730. doi: 10.3892/ol.2016.5463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Simmer F., Venderbosch S., Dijkstra J.R., Vink-Börger E.M., Faber C., Mekenkamp L.J., Koopman M., De Haan A.F., Punt C.J., Nagtegaal I.D. MicroRNA-143 is a putative predictive factor for the response to fluoropyrimidine-based chemotherapy in patients with metastatic colorectal cancer. Oncotarget. 2015;6:22996–23007. doi: 10.18632/oncotarget.4035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Cristóbal I., Caramés C., Rincón R., Manso R., Madoz-Gúrpide J., Torrejón B., González-Alonso P., Rojo F., García-Foncillas J. Downregulation of microRNA-199b predicts unfavorable prognosis and emerges as a novel therapeutic target which contributes to PP2A inhibition in metastatic colorectal cancer. Oncotarget. 2017;8:40169–40180. doi: 10.18632/oncotarget.11174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Yin Y., Zhang B., Wang W., Fei B., Quan C., Zhang J., Song M., Bian Z., Wang Q., Ni S. miR-204-5p inhibits proliferation and invasion and enhances chemotherapeutic sensitivity of colorectal cancer cells by downregulating RAB22A. Clin. Cancer Res. 2014;20:6187–6199. doi: 10.1158/1078-0432.CCR-14-1030. [DOI] [PubMed] [Google Scholar]
  • 92.Xu K., Chen G., Qiu Y., Yuan Z., Li H., Yuan X., Sun J., Xu J., Liang X., Yin P. miR-503-5p confers drug resistance by targeting PUMA in colorectal carcinoma. Oncotarget. 2017;8:21719–21732. doi: 10.18632/oncotarget.15559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Li S., Li C., Fang Z. MicroRNA 214 inhibits adipocyte enhancer-binding protein 1 activity and increases the sensitivity of chemotherapy in colorectal cancer. Oncol. Lett. 2019;17:55–62. doi: 10.3892/ol.2018.9623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Cao S., Lin L., Xia X., Wu H. MicroRNA-761 promotes the sensitivity of colorectal cancer cells to 5-Fluorouracil through targeting FOXM1. Oncotarget. 2017;9:321–331. doi: 10.18632/oncotarget.20109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wang Y., Zhao M., Zhao H., Cheng S., Bai R., Song M. MicroRNA-940 restricts the expression of metastasis-associated gene MACC1 and enhances the antitumor effect of Anlotinib on colorectal cancer. OncoTargets Ther. 2019;12:2809–2822. doi: 10.2147/OTT.S195364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Xu W., Jiang H., Zhang F., Gao J., Hou J. MicroRNA-330 inhibited cell proliferation and enhanced chemosensitivity to 5-fluorouracil in colorectal cancer by directly targeting thymidylate synthase. Oncol. Lett. 2017;13:3387–3394. doi: 10.3892/ol.2017.5895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Puthanveetil P., Chen S., Feng B., Gautam A., Chakrabarti S. Long non-coding RNA MALAT1 regulates hyperglycaemia induced inflammatory process in the endothelial cells. J. Cell. Mol. Med. 2015;19:1418–1425. doi: 10.1111/jcmm.12576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Sun Z., Zhou N., Han Q., Zhao L., Bai C., Chen Y., Zhou J., Zhao R.C. MicroRNA-197 influences 5-fluorouracil resistance via thymidylate synthase in colorectal cancer. Clin. Trans. Oncol. 2015;17:876–883. doi: 10.1007/s12094-015-1318-7. [DOI] [PubMed] [Google Scholar]
  • 99.Zhang Y., Geng L., Talmon G., Wang J. MicroRNA-520g confers drug resistance by regulating p21 expression in colorectal cancer. J. Biol. Chem. 2015;290:6215–6225. doi: 10.1074/jbc.M114.620252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Wang G., Wang J., Luo J., Wang L., Chen X., Zhang L., Jiang S. PEG2000-DPSE-coated quercetin nanoparticles remarkably enhanced anticancer effects through induced programed cell death on C6 glioma cells. J. Biomed. Mater. Res. A. 2013;101:3076–3085. doi: 10.1002/jbm.a.34607. [DOI] [PubMed] [Google Scholar]
  • 101.Sui H., Cai G.X., Pan S.F., Deng W.L., Wang Y.W., Chen Z.S., Cai S.J., Zhu H.R., Li Q. miR200c attenuates P-gp-mediated MDR and metastasis by targeting JNK2/c-Jun signaling pathway in colorectal cancer. Mol. Cancer Ther. 2014;13:3137–3151. doi: 10.1158/1535-7163.MCT-14-0167. [DOI] [PubMed] [Google Scholar]
  • 102.Zhang L., He L., Zhang H., Chen Y. Knockdown of miR-20a enhances sensitivity of colorectal cancer cells to cisplatin by increasing ASK1 expression. Cell. Physiol. Biochem. 2018;47:1432–1441. doi: 10.1159/000490834. [DOI] [PubMed] [Google Scholar]
  • 103.Liu X., Xie T., Mao X., Xue L., Chu X., Chen L. MicroRNA-149 increases the sensitivity of colorectal cancer cells to 5-fluorouracil by targeting forkhead box transcription factor FOXM1. Cell. Physiol. Biochem. 2016;39:617–629. doi: 10.1159/000445653. [DOI] [PubMed] [Google Scholar]
  • 104.Yao H., Sun Q., Zhu J. miR-1271 enhances the sensitivity of colorectal cancer cells to cisplatin. Exp. Ther. Med. 2019;17:4363–4370. doi: 10.3892/etm.2019.7501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Borralho P.M., Kren B.T., Castro R.E., da Silva I.B., Steer C.J., Rodrigues C.M. MicroRNA-143 reduces viability and increases sensitivity to 5-fluorouracil in HCT116 human colorectal cancer cells. FEBS J. 2009;276:6689–6700. doi: 10.1111/j.1742-4658.2009.07383.x. [DOI] [PubMed] [Google Scholar]
  • 106.Chen B., Zhang D., Kuai J., Cheng M., Fang X., Li G. Upregulation of miR-199a/b contributes to cisplatin resistance via Wnt/β-catenin-ABCG2 signaling pathway in ALDHA1+ colorectal cancer stem cells. Tumour Biol. 2017;39 doi: 10.1177/1010428317715155. 1010428317715155. [DOI] [PubMed] [Google Scholar]
  • 107.Karaayvaz M., Zhai H., Ju J. miR-129 promotes apoptosis and enhances chemosensitivity to 5-fluorouracil in colorectal cancer. Cell Death Dis. 2013;4:e659. doi: 10.1038/cddis.2013.193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Liu K., Li G., Fan C., Zhou X., Wu B., Li J. Increased expression of microRNA-21and its association with chemotherapeutic response in human colorectal cancer. J. Int. Med. Res. 2011;39:2288–2295. doi: 10.1177/147323001103900626. [DOI] [PubMed] [Google Scholar]
  • 109.Qin Y., Chen X., Liu Z., Tian X., Huo Z. miR-106a reduces 5-fluorouracil (5-FU) sensitivity of colorectal cancer by targeting dual-specificity phosphatases 2 (DUSP2) Med. Sci. Monit. 2018;24:4944–4951. doi: 10.12659/MSM.910016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Zhang L., Li B., Zhang B., Zhang H., Suo J. miR-361 enhances sensitivity to 5-fluorouracil by targeting the FOXM1-ABCC5/10 signaling pathway in colorectal cancer. Oncol. Lett. 2019;18:4064–4073. doi: 10.3892/ol.2019.10741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Liu H., Yin Y., Hu Y., Feng Y., Bian Z., Yao S., Li M., You Q., Huang Z. miR-139-5p sensitizes colorectal cancer cells to 5-fluorouracil by targeting NOTCH-1. Pathol. Res. Pract. 2016;212:643–649. doi: 10.1016/j.prp.2016.04.011. [DOI] [PubMed] [Google Scholar]
  • 112.Kim C., Hong Y., Lee H., Kang H., Lee E.K. MicroRNA-195 desensitizes HCT116 human colon cancer cells to 5-fluorouracil. Cancer Lett. 2018;412:264–271. doi: 10.1016/j.canlet.2017.10.022. [DOI] [PubMed] [Google Scholar]
  • 113.Liu L., Zheng W., Song Y., Du X., Tang Y., Nie J., Han W. miRNA-497 enhances the sensitivity of colorectal cancer cells to neoadjuvant chemotherapeutic drug. Curr. Protein Pept. Sci. 2015;16:310–315. doi: 10.2174/138920371604150429154142. [DOI] [PubMed] [Google Scholar]
  • 114.To K.K., Leung W.W., Ng S.S. Exploiting a novel miR-519c-HuR-ABCG2 regulatory pathway to overcome chemoresistance in colorectal cancer. Exp. Cell Res. 2015;338:222–231. doi: 10.1016/j.yexcr.2015.09.011. [DOI] [PubMed] [Google Scholar]
  • 115.Liu G., Zhou J., Dong M. Down-regulation of miR-543 expression increases the sensitivity of colorectal cancer cells to 5-fluorouracil through the PTEN/PI3K/AKT pathway. Biosci. Rep. 2019;39 doi: 10.1042/BSR20190249. BSR20190249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Sun C., Wang F.J., Zhang H.G., Xu X.Z., Jia R.C., Yao L., Qiao P.F. miR-34a mediates oxaliplatin resistance of colorectal cancer cells by inhibiting macroautophagy via transforming growth factor-β/Smad4 pathway. World J. Gastroenterol. 2017;23:1816–1827. doi: 10.3748/wjg.v23.i10.1816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Hansen T.F., Sørensen F.B., Lindebjerg J., Jakobsen A. The predictive value of microRNA-126 in relation to first line treatment with capecitabine and oxaliplatin in patients with metastatic colorectal cancer. BMC Cancer. 2012;12:83. doi: 10.1186/1471-2407-12-83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Ye L., Jiang T., Shao H., Zhong L., Wang Z., Liu Y., Tang H., Qin B., Zhang X., Fan J. miR-1290 is a biomarker in DNA-mismatch-repair-deficient colon cancer and promotes resistance to 5-fluorouracil by directly targeting hMSH2. Mol. Ther. Nucleic Acids. 2017;7:453–464. doi: 10.1016/j.omtn.2017.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Zhang D., Li Y., Sun P. miR-770-5p modulates resistance to methotrexate in human colorectal adenocarcinoma cells by downregulating HIPK1. Exp. Ther. Med. 2020;19:339–346. doi: 10.3892/etm.2019.8221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Heydari K., Saidijam M., Sharifi M.R., Dermani F.K., Soleimani Asl S., Shabab N., Najafi R. The effect of mir-200c inhibition on chemosensitivity (5-fluorouracil) in colorectal cancer. Pathol. Oncol. Res. 2018;24:145–151. doi: 10.1007/s12253-017-0222-6. [DOI] [PubMed] [Google Scholar]
  • 121.Chai H., Liu M., Tian R., Li X., Tang H. miR-20a targets BNIP2 and contributes chemotherapeutic resistance in colorectal adenocarcinoma SW480 and SW620 cell lines. Acta Biochim. Biophys. Sin. (Shanghai) 2011;43:217–225. doi: 10.1093/abbs/gmq125. [DOI] [PubMed] [Google Scholar]
  • 122.Chen L.G., Xia Y.J., Cui Y. Upregulation of miR-101 enhances the cytotoxic effect of anticancer drugs through inhibition of colon cancer cell proliferation. Oncol. Rep. 2017;38:100–108. doi: 10.3892/or.2017.5666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Liu Y., Gao S., Chen X., Liu M., Mao C., Fang X. Overexpression of miR-203 sensitizes paclitaxel (Taxol)-resistant colorectal cancer cells through targeting the salt-inducible kinase 2 (SIK2) Tumour Biol. 2016;37:12231–12239. doi: 10.1007/s13277-016-5066-2. [DOI] [PubMed] [Google Scholar]
  • 124.Ren D., Lin B., Zhang X., Peng Y., Ye Z., Ma Y., Liang Y., Cao L., Li X., Li R. Maintenance of cancer stemness by miR-196b-5p contributes to chemoresistance of colorectal cancer cells via activating STAT3 signaling pathway. Oncotarget. 2017;8:49807–49823. doi: 10.18632/oncotarget.17971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Tan S., Shi H., Ba M., Lin S., Tang H., Zeng X., Zhang X. miR-409-3p sensitizes colon cancer cells to oxaliplatin by inhibiting Beclin-1-mediated autophagy. Int. J. Mol. Med. 2016;37:1030–1038. doi: 10.3892/ijmm.2016.2492. [DOI] [PubMed] [Google Scholar]
  • 126.Han J., Li J., Tang K., Zhang H., Guo B., Hou N., Huang C. miR-338-3p confers 5-fluorouracil resistance in p53 mutant colon cancer cells by targeting the mammalian target of rapamycin. Exp. Cell Res. 2017;360:328–336. doi: 10.1016/j.yexcr.2017.09.023. [DOI] [PubMed] [Google Scholar]
  • 127.Liang Y., Zhu D., Hou L., Wang Y., Huang X., Zhou C., Zhu L., Wang Y., Li L., Gu Y. miR-107 confers chemoresistance to colorectal cancer by targeting calcium-binding protein 39. Br. J. Cancer. 2020;122:705–714. doi: 10.1038/s41416-019-0703-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Pranzini E., Leo A., Rapizzi E., Ramazzotti M., Magherini F., Giovannelli L., Caselli A., Cirri P., Taddei M.L., Paoli P. miR-210-3p mediates metabolic adaptation and sustains DNA damage repair of resistant colon cancer cells to treatment with 5-fluorouracil. Mol. Carcinog. 2019;58:2181–2192. doi: 10.1002/mc.23107. [DOI] [PubMed] [Google Scholar]
  • 129.Liu R.L., Dong Y., Deng Y.Z., Wang W.J., Li W.D. Tumor suppressor miR-145 reverses drug resistance by directly targeting DNA damage-related gene RAD18 in colorectal cancer. Tumour Biol. 2015;36:5011–5019. doi: 10.1007/s13277-015-3152-5. [DOI] [PubMed] [Google Scholar]
  • 130.Que K., Tong Y., Que G., Li L., Lin H., Huang S., Wang R., Tang L. Downregulation of miR-874-3p promotes chemotherapeutic resistance in colorectal cancer via inactivation of the Hippo signaling pathway. Oncol. Rep. 2017;38:3376–3386. doi: 10.3892/or.2017.6041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Li X., Zhao H., Zhou X., Song L. Inhibition of lactate dehydrogenase A by microRNA-34a resensitizes colon cancer cells to 5-fluorouracil. Mol. Med. Rep. 2015;11:577–582. doi: 10.3892/mmr.2014.2726. [DOI] [PubMed] [Google Scholar]
  • 132.Zhou Y., Wan G., Spizzo R., Ivan C., Mathur R., Hu X., Ye X., Lu J., Fan F., Xia L. miR-203 induces oxaliplatin resistance in colorectal cancer cells by negatively regulating ATM kinase. Mol. Oncol. 2014;8:83–92. doi: 10.1016/j.molonc.2013.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Peng L., Zhu H., Wang J., Sui H., Zhang H., Jin C., Li L., Xu T., Miao R. miR-492 is functionally involved in oxaliplatin resistance in colon cancer cells LS174T via its regulating the expression of CD147. Mol. Cell. Biochem. 2015;405:73–79. doi: 10.1007/s11010-015-2397-z. [DOI] [PubMed] [Google Scholar]
  • 134.Deng J., Lei W., Fu J.C., Zhang L., Li J.H., Xiong J.P. Targeting miR-21 enhances the sensitivity of human colon cancer HT-29 cells to chemoradiotherapy in vitro. Biochem. Biophys. Res. Commun. 2014;443:789–795. doi: 10.1016/j.bbrc.2013.11.064. [DOI] [PubMed] [Google Scholar]
  • 135.Xu K., Liang X., Shen K., Sun L., Cui D., Zhao Y., Tian J., Ni L., Liu J. miR-222 modulates multidrug resistance in human colorectal carcinoma by down-regulating ADAM-17. Exp. Cell Res. 2012;318:2168–2177. doi: 10.1016/j.yexcr.2012.04.014. [DOI] [PubMed] [Google Scholar]
  • 136.Tu M.J., Pan Y.Z., Qiu J.X., Kim E.J., Yu A.M. MicroRNA-1291 targets the FOXA2-AGR2 pathway to suppress pancreatic cancer cell proliferation and tumorigenesis. Oncotarget. 2016;7:45547–45561. doi: 10.18632/oncotarget.9999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Bi H.C., Pan Y.Z., Qiu J.X., Krausz K.W., Li F., Johnson C.H., Jiang C.T., Gonzalez F.J., Yu A.M. N-methylnicotinamide and nicotinamide N-methyltransferase are associated with microRNA-1291-altered pancreatic carcinoma cell metabolome and suppressed tumorigenesis. Carcinogenesis. 2014;35:2264–2272. doi: 10.1093/carcin/bgu174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Yamasaki T., Seki N., Yoshino H., Itesako T., Yamada Y., Tatarano S., Hidaka H., Yonezawa T., Nakagawa M., Enokida H. Tumor-suppressive microRNA-1291 directly regulates glucose transporter 1 in renal cell carcinoma. Cancer Sci. 2013;104:1411–1419. doi: 10.1111/cas.12240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Tu M.J., Duan Z., Liu Z., Zhang C., Bold R.J., Gonzalez F.J., Kim E.J., Yu A.M. MicroRNA-1291-5p sensitizes pancreatic carcinoma cells to arginine deprivation and chemotherapy through the regulation of arginolysis and glycolysis. Mol. Pharmacol. 2020;98:686–694. doi: 10.1124/molpharm.120.000130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Zhang B., Liu B., Chen D., Setroikromo R., Haisma H.J., Quax W.J. Histone deacetylase inhibitors sensitize TRAIL-induced apoptosis in colon cancer cells. Cancers (Basel) 2019;11:645. doi: 10.3390/cancers11050645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Eberle J. Countering trail resistance in melanoma. Cancers (Basel) 2019;11:656. doi: 10.3390/cancers11050656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Hopkins-Donaldson S., Bodmer J.-L., Bourloud K.B., Brognara C.B., Tschopp J., Gross N. Loss of caspase-8 expression in highly malignant human neuroblastoma cells correlates with resistance to tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis. Cancer Res. 2000;60:4315–4319. [PubMed] [Google Scholar]
  • 143.Yang X., Merchant M.S., Romero M.E., Tsokos M., Wexler L.H., Kontny U., Mackall C.L., Thiele C.J. Induction of caspase 8 by interferon γ renders some neuroblastoma (NB) cells sensitive to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) but reveals that a lack of membrane TR1/TR2 also contributes to TRAIL resistance in NB. Cancer Res. 2003;63:1122–1129. [PubMed] [Google Scholar]
  • 144.Xia X., Zhang K., Luo G., Cen G., Cao J., Huang K., Qiu Z. Downregulation of miR-301a-3p sensitizes pancreatic cancer cells to gemcitabine treatment via PTEN. Am. J. Transl. Res. 2017;9:1886–1895. [PMC free article] [PubMed] [Google Scholar]
  • 145.Lu Z., Li Y., Takwi A., Li B., Zhang J., Conklin D.J., Young K.H., Martin R., Li Y. miR-301a as an NF-κB activator in pancreatic cancer cells. EMBO J. 2011;30:57–67. doi: 10.1038/emboj.2010.296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Moeng S., Son S.W., Seo H.A., Lee J.S., Kim C.K., Kuh H.J., Park J.K. Luteolin-regulated microRNA-301-3p targets caspase-8 and modulates TRAIL sensitivity in PANC-1 cells. Anticancer Res. 2020;40:723–731. doi: 10.21873/anticanres.14003. [DOI] [PubMed] [Google Scholar]
  • 147.Li D., Shan W., Fang Y., Wang P., Li J. miR-137 acts as a tumor suppressor via inhibiting CXCL12 in human glioblastoma. Oncotarget. 2017;8:101262–101270. doi: 10.18632/oncotarget.20589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Shen H., Wang L., Ge X., Jiang C.-F., Shi Z.-M., Li D.-M., Liu W.-T., Yu X., Shu Y.-Q. MicroRNA-137 inhibits tumor growth and sensitizes chemosensitivity to paclitaxel and cisplatin in lung cancer. Oncotarget. 2016;7:20728–20742. doi: 10.18632/oncotarget.8011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Takwi A.A., Wang Y.-M., Wu J., Michaelis M., Cinatl J., Chen T. miR-137 regulates the constitutive androstane receptor and modulates doxorubicin sensitivity in parental and doxorubicin-resistant neuroblastoma cells. Oncogene. 2014;33:3717–3729. doi: 10.1038/onc.2013.330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Li W., Zhang X., Zhuang H., Chen H.G., Chen Y., Tian W., Wu W., Li Y., Wang S., Zhang L. MicroRNA-137 is a novel hypoxia-responsive microRNA that inhibits mitophagy via regulation of two mitophagy receptors FUNDC1 and NIX. J. Biol. Chem. 2014;289:10691–10701. doi: 10.1074/jbc.M113.537050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Zeng Y., Huo G., Mo Y., Wang W., Chen H. MIR137 regulates starvation-induced autophagy by targeting ATG7. J. Mol. Neurosci. 2015;56:815–821. doi: 10.1007/s12031-015-0514-9. [DOI] [PubMed] [Google Scholar]
  • 152.Neault M., Mallette F.A., Richard S. miR-137 modulates a tumor suppressor network-inducing senescence in pancreatic cancer cells. Cell Rep. 2016;14:1966–1978. doi: 10.1016/j.celrep.2016.01.068. [DOI] [PubMed] [Google Scholar]
  • 153.Xiao J., Peng F., Yu C., Wang M., Li X., Li Z., Jiang J., Sun C. MicroRNA-137 modulates pancreatic cancer cells tumor growth, invasion and sensitivity to chemotherapy. Int. J. Clin. Exp. Pathol. 2014;7:7442–7450. [PMC free article] [PubMed] [Google Scholar]
  • 154.Wang Z.C., Huang F.Z., Xu H.B., Sun J.C., Wang C.F. MicroRNA-137 inhibits autophagy and chemosensitizes pancreatic cancer cells by targeting ATG5. Int. J. Biochem. Cell Biol. 2019;111:63–71. doi: 10.1016/j.biocel.2019.01.020. [DOI] [PubMed] [Google Scholar]
  • 155.Smith L.K., Shah R.R., Cidlowski J.A. Glucocorticoids modulate microRNA expression and processing during lymphocyte apoptosis. J. Biol. Chem. 2010;285:36698–36708. doi: 10.1074/jbc.M110.162123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Herr I., Ucur E., Herzer K., Okouoyo S., Ridder R., Krammer P.H., von Knebel Doeberitz M., Debatin K.-M. Glucocorticoid cotreatment induces apoptosis resistance toward cancer therapy in carcinomas. Cancer Res. 2003;63:3112–3120. [PubMed] [Google Scholar]
  • 157.Qin J., Ke J., Xu J., Wang F., Zhou Y., Jiang Y., Wang Z. Downregulation of microRNA-132 by DNA hypermethylation is associated with cell invasion in colorectal cancer. OncoTargets Ther. 2015;8:3639–3648. doi: 10.2147/OTT.S91560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Abukiwan A., Nwaeburu C.C., Bauer N., Zhao Z., Liu L., Gladkich J., Gross W., Benner A., Strobel O., Fellenberg J., Herr I. Dexamethasone-induced inhibition of miR-132 via methylation promotes TGF-β-driven progression of pancreatic cancer. Int. J. Oncol. 2019;54:53–64. doi: 10.3892/ijo.2018.4616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Jones A., Danielson K.M., Benton M.C., Ziegler O., Shah R., Stubbs R.S., Das S., Macartney-Coxson D. miRNA signatures of insulin resistance in obesity. Obesity (Silver Spring) 2017;25:1734–1744. doi: 10.1002/oby.21950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Xu H.-X., Wang Y., Zheng D.-D., Wang T., Pan M., Shi J.-H., Zhu J.-H., Li X.-F. Differential expression of microRNAs in calcific aortic stenosis. Clin. Lab. 2017;63:1163–1170. doi: 10.7754/Clin.Lab.2017.170108. [DOI] [PubMed] [Google Scholar]
  • 161.Tan J.R., Tan K.S., Yong F.L., Armugam A., Wang C.W., Jeyaseelan K., Wong P.T.-H. MicroRNAs regulating cluster of differentiation 46 (CD46) in cardioembolic and non-cardioembolic stroke. PLoS ONE. 2017;12:e0172131. doi: 10.1371/journal.pone.0172131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Hu S., Bao H., Xu X., Zhou X., Qin W., Zeng C., Liu Z. Increased miR-374b promotes cell proliferation and the production of aberrant glycosylated IgA1 in B cells of IgA nephropathy. FEBS Lett. 2015;589(24 Pt B):4019–4025. doi: 10.1016/j.febslet.2015.10.033. [DOI] [PubMed] [Google Scholar]
  • 163.Wang C., Yang C., Chen X., Yao B., Yang C., Zhu C., Li L., Wang J., Li X., Shao Y. Altered profile of seminal plasma microRNAs in the molecular diagnosis of male infertility. Clin. Chem. 2011;57:1722–1731. doi: 10.1373/clinchem.2011.169714. [DOI] [PubMed] [Google Scholar]
  • 164.Zhang K., Wang Y.-W., Wang Y.-Y., Song Y., Zhu J., Si P.-C., Ma R. Identification of microRNA biomarkers in the blood of breast cancer patients based on microRNA profiling. Gene. 2017;619:10–20. doi: 10.1016/j.gene.2017.03.038. [DOI] [PubMed] [Google Scholar]
  • 165.Chang J.T., Wang F., Chapin W., Huang R.S. Identification of MicroRNAs as breast cancer prognosis markers through the cancer genome atlas. PLoS ONE. 2016;11:e0168284. doi: 10.1371/journal.pone.0168284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Hanniford D., Zhong J., Koetz L., Gaziel-Sovran A., Lackaye D.J., Shang S., Pavlick A., Shapiro R., Berman R., Darvishian F. A miRNA-based signature detected in primary melanoma tissue predicts development of brain metastasis. Clin. Cancer Res. 2015;21:4903–4912. doi: 10.1158/1078-0432.CCR-14-2566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Summerer I., Unger K., Braselmann H., Schuettrumpf L., Maihoefer C., Baumeister P., Kirchner T., Niyazi M., Sage E., Specht H.M. Circulating microRNAs as prognostic therapy biomarkers in head and neck cancer patients. Br. J. Cancer. 2015;113:76–82. doi: 10.1038/bjc.2015.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Xie J., Tan Z.-H., Tang X., Mo M.-S., Liu Y.-P., Gan R.-L., Li Y., Zhang L., Li G.-Q. miR-374b-5p suppresses RECK expression and promotes gastric cancer cell invasion and metastasis. World J. Gastroenterol. 2014;20:17439–17447. doi: 10.3748/wjg.v20.i46.17439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.He H.C., Han Z.D., Dai Q.S., Ling X.H., Fu X., Lin Z.Y., Deng Y.H., Qin G.Q., Cai C., Chen J.H. Global analysis of the differentially expressed miRNAs of prostate cancer in Chinese patients. BMC Genomics. 2013;14:757. doi: 10.1186/1471-2164-14-757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Qian D., Chen K., Deng H., Rao H., Huang H., Liao Y., Sun X., Lu S., Yuan Z., Xie D., Cai Q. MicroRNA-374b suppresses proliferation and promotes apoptosis in T-cell lymphoblastic lymphoma by repressing AKT1 and Wnt-16. Clin. Cancer Res. 2015;21:4881–4891. doi: 10.1158/1078-0432.CCR-14-2947. [DOI] [PubMed] [Google Scholar]
  • 171.Wu X., Li S., Xu X., Wu S., Chen R., Jiang Q., Li Y., Xu Y. The potential value of miR-1 and miR-374b as biomarkers for colorectal cancer. Int. J. Clin. Exp. Pathol. 2015;8:2840–2851. [PMC free article] [PubMed] [Google Scholar]
  • 172.Schreiber R., Mezencev R., Matyunina L.V., McDonald J.F. Evidence for the role of microRNA 374b in acquired cisplatin resistance in pancreatic cancer cells. Cancer Gene Ther. 2016;23:241–245. doi: 10.1038/cgt.2016.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Sun D., Wang X., Sui G., Chen S., Yu M., Zhang P. Downregulation of miR-374b-5p promotes chemotherapeutic resistance in pancreatic cancer by upregulating multiple anti-apoptotic proteins. Int. J. Oncol. 2018;52:1491–1503. doi: 10.3892/ijo.2018.4315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Tu M.J., Duan Z., Liu Z., Zhang C., Bold R.J., Gonzalez F.J., Kim E.J., Yu A.M. MicroRNA-1291-5p sensitizes pancreatic carcinoma cells to arginine deprivation and chemotherapy through the regulation of arginolysis and glycolysis. Mol. Pharmacol. 2020;98:686–694. doi: 10.1124/molpharm.120.000130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Li Z., Li X., Yu C., Wang M., Peng F., Xiao J., Tian R., Jiang J., Sun C. MicroRNA-100 regulates pancreatic cancer cells growth and sensitivity to chemotherapy through targeting FGFR3. Tumour Biol. 2014;35:11751–11759. doi: 10.1007/s13277-014-2271-8. [DOI] [PubMed] [Google Scholar]
  • 176.Sheu-Gruttadauria J., Pawlica P., Klum S.M., Wang S., Yario T.A., Oakdale N.T.S., Steitz J.A., MacRae I.J. Structural basis for target-directed microrna degradation. Mol. Cell. 2019;75:1243–1255.e7. doi: 10.1016/j.molcel.2019.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Ying G., Wu R., Xia M., Fei X., He Q.E., Zha C. Identification of eight key miRNAs associated with renal cell carcinoma: a meta-analysis. Oncol Lett. 2018;16:5847–5855. doi: 10.3892/ol.2018.9384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Toste P.A., Li L., Kadera B.E., Nguyen A.H., Tran L.M., Wu N., Madnick D.L., Patel S.G., Dawson D.W., Donahue T.R. p85α is a microRNA target and affects chemosensitivity in pancreatic cancer. J. Surg. Res. 2015;196:285–293. doi: 10.1016/j.jss.2015.02.071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Wei X., Wang W., Wang L., Zhang Y., Zhang X., Chen M., Wang F., Yu J., Ma Y., Sun G. MicroRNA-21 induces 5-fluorouracil resistance in human pancreatic cancer cells by regulating PTEN and PDCD4. Cancer Med. 2016;5:693–702. doi: 10.1002/cam4.626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.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. [PMC free article] [PubMed] [Google Scholar]
  • 181.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. doi: 10.1038/s41419-019-1326-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Zhao L., Zou D., Wei X., Wang L., Zhang Y., Liu S., Si Y., Zhao H., Wang F., Yu J. miRNA-221-3p desensitizes pancreatic cancer cells to 5-fluorouracil by targeting RB1. Tumour Biol. 2016;37:16053–16063. doi: 10.1007/s13277-016-5445-8. [DOI] [PubMed] [Google Scholar]
  • 183.Wang W., Zhao L., Wei X., Wang L., Liu S., Yang Y., Wang F., Sun G., Zhang J., Ma Y. MicroRNA-320a promotes 5-FU resistance in human pancreatic cancer cells. Sci. Rep. 2016;6:27641. doi: 10.1038/srep27641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Wang P., Zhuang L., Zhang J., Fan J., Luo J., Chen H., Wang K., Liu L., Chen Z., Meng Z. The serum miR-21 level serves as a predictor for the chemosensitivity of advanced pancreatic cancer, and miR-21 expression confers chemoresistance by targeting FasL. Mol. Oncol. 2013;7:334–345. doi: 10.1016/j.molonc.2012.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Huang H., Xiong G., Shen P., Cao Z., Zheng L., Zhang T., Zhao Y. MicroRNA-1285 inhibits malignant biological behaviors of human pancreatic cancer cells by negative regulation of YAP1. Neoplasma. 2017;64:358–366. doi: 10.4149/neo_2017_306. [DOI] [PubMed] [Google Scholar]
  • 186.Yang R.M., Zhan M., Xu S.W., Long M.M., Yang L.H., Chen W., Huang S., Liu Q., Zhou J., Zhu J., Wang J. miR-3656 expression enhances the chemosensitivity of pancreatic cancer to gemcitabine through modulation of the RHOF/EMT axis. Cell Death Dis. 2017;8:e3129. doi: 10.1038/cddis.2017.530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Xu X., Wang Y., Mojumdar K., Zhou Z., Jeong K.J., Mangala L.S., Yu S., Tsang Y.H., Rodriguez-Aguayo C., Lu Y. A-to-I-edited miRNA-379-5p inhibits cancer cell proliferation through CD97-induced apoptosis. J. Clin. Invest. 2019;129:5343–5356. doi: 10.1172/JCI123396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Iwagami Y., Eguchi H., Nagano H., Akita H., Hama N., Wada H., Kawamoto K., Kobayashi S., Tomokuni A., Tomimaru Y. miR-320c regulates gemcitabine-resistance in pancreatic cancer via SMARCC1. Br. J. Cancer. 2013;109:502–511. doi: 10.1038/bjc.2013.320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Maftouh M., Avan A., Funel N., Frampton A.E., Fiuji H., Pelliccioni S., Castellano L., Galla V., Peters G.J., Giovannetti E. miR-211 modulates gemcitabine activity through downregulation of ribonucleotide reductase and inhibits the invasive behavior of pancreatic cancer cells. Nucleosides Nucleotides Nucleic Acids. 2014;33:384–393. doi: 10.1080/15257770.2014.891741. [DOI] [PubMed] [Google Scholar]
  • 190.Fan P., Liu L., Yin Y., Zhao Z., Zhang Y., Amponsah P.S., Xiao X., Bauer N., Abukiwan A., Nwaeburu C.C. MicroRNA-101-3p reverses gemcitabine resistance by inhibition of ribonucleotide reductase M1 in pancreatic cancer. Cancer Lett. 2016;373:130–137. doi: 10.1016/j.canlet.2016.01.038. [DOI] [PubMed] [Google Scholar]
  • 191.Xiong J., Wang D., Wei A., Ke N., Wang Y., Tang J., He S., Hu W., Liu X. MicroRNA-410-3p attenuates gemcitabine resistance in pancreatic ductal adenocarcinoma by inhibiting HMGB1-mediated autophagy. Oncotarget. 2017;8:107500–107512. doi: 10.18632/oncotarget.22494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Paik W.H., Kim H.R., Park J.K., Song B.J., Lee S.H., Hwang J.H. Chemosensitivity induced by down-regulation of microRNA-21 in gemcitabine-resistant pancreatic cancer cells by indole-3-carbinol. Anticancer Res. 2013;33:1473–1481. [PubMed] [Google Scholar]
  • 193.Zheng H., Wang J.J., Yang X.R., Yu Y.L. Upregulation of miR-34c after silencing E2F transcription factor 1 inhibits paclitaxel combined with cisplatin resistance in gastric cancer cells. World J. Gastroenterol. 2020;26:499–513. doi: 10.3748/wjg.v26.i5.499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Liu D., Zhang C., Li X., Zhang H., Pang Q., Wan A. MicroRNA-567 inhibits cell proliferation, migration and invasion by targeting FGF5 in osteosarcoma. EXCLI J. 2018;17:102–112. doi: 10.17179/excli2017-932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Shao Y., Liang B., Long F., Jiang S.-J. Diagnostic microRNA biomarker discovery for non-small-cell lung cancer adenocarcinoma by integrative bioinformatics analysis. BioMed Res. Int. 2017;2017:2563085. doi: 10.1155/2017/2563085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Wang F., Wong S.C., Chan L.W., Cho W.C., Yip S.P., Yung B.Y. Multiple regression analysis of mRNA-miRNA associations in colorectal cancer pathway. BioMed Res. Int. 2014;2014:676724. doi: 10.1155/2014/676724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Cava C., Bertoli G., Ripamonti M., Mauri G., Zoppis I., Della Rosa P.A., Gilardi M.C., Castiglioni I. Integration of mRNA expression profile, copy number alterations, and microRNA expression levels in breast cancer to improve grade definition. PLoS ONE. 2014;9:e97681. doi: 10.1371/journal.pone.0097681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Bertoli G., Cava C., Diceglie C., Martelli C., Rizzo G., Piccotti F., Ottobrini L., Castiglioni I. MicroRNA-567 dysregulation contributes to carcinogenesis of breast cancer, targeting tumor cell proliferation, and migration. Breast Cancer Res. Treat. 2017;161:605–616. doi: 10.1007/s10549-016-4079-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Zhang F., Li K., Yao X., Wang H., Li W., Wu J., Li M., Zhou R., Xu L., Zhao L. A miR-567-PIK3AP1-PI3K/AKT-c-Myc feedback loop regulates tumour growth and chemoresistance in gastric cancer. EBioMedicine. 2019;44:311–321. doi: 10.1016/j.ebiom.2019.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Liang Y., Song X., Li Y., Sang Y., Zhang N., Zhang H., Liu Y., Duan Y., Chen B., Guo R. A novel long non-coding RNA-PRLB acts as a tumor promoter through regulating miR-4766-5p/SIRT1 axis in breast cancer. Cell Death Dis. 2018;9:563. doi: 10.1038/s41419-018-0582-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Pajerowski A.G., Nguyen C., Aghajanian H., Shapiro M.J., Shapiro V.S. NKAP is a transcriptional repressor of notch signaling and is required for T cell development. Immunity. 2009;30:696–707. doi: 10.1016/j.immuni.2009.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Hsu F.-C., Pajerowski A.G., Nelson-Holte M., Sundsbak R., Shapiro V.S. NKAP is required for T cell maturation and acquisition of functional competency. J. Exp. Med. 2011;208:1291–1304. doi: 10.1084/jem.20101874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Juan C., Li F., Puett D., Hong C., Xu Y. 2010. Protein markers identification for gastric cancer diagnosis. Patent application publication WO2010104662A1, filed February 19, 2010, and published September 16, 2010. [Google Scholar]
  • 204.Burgute B.D., Peche V.S., Steckelberg A.-L., Glöckner G., Gaßen B., Gehring N.H., Noegel A.A. NKAP is a novel RS-related protein that interacts with RNA and RNA binding proteins. Nucleic Acids Res. 2014;42:3177–3193. doi: 10.1093/nar/gkt1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Li T., Chen L., Cheng J., Dai J., Huang Y., Zhang J., Liu Z., Li A., Li N., Wang H. SUMOylated NKAP is essential for chromosome alignment by anchoring CENP-E to kinetochores. Nat. Commun. 2016;7:12969. doi: 10.1038/ncomms12969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Wei Y., Wang Y., Zang A., Wang Z., Fang G., Hong D. miR-4766-5p inhibits the development and progression of gastric cancer by targeting NKAP. OncoTargets Ther. 2019;12:8525–8536. doi: 10.2147/OTT.S220234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Wang S., Li Q., Wang K., Dai Y., Yang J., Xue S., Han F., Zhang Q., Liu J., Wu W. Decreased expression of microRNA-31 associates with aggressive tumor progression and poor prognosis in patients with bladder cancer. Clin. Transl. Oncol. 2013;15:849–854. doi: 10.1007/s12094-013-1014-4. [DOI] [PubMed] [Google Scholar]
  • 208.Wang C.-J., Stratmann J., Zhou Z.-G., Sun X.-F. Suppression of microRNA-31 increases sensitivity to 5-FU at an early stage, and affects cell migration and invasion in HCT-116 colon cancer cells. BMC Cancer. 2010;10:616. doi: 10.1186/1471-2407-10-616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Wang H., Zhang X., Liu Y., Ni Z., Lin Y., Duan Z., Shi Y., Wang G., Li F. Downregulated miR-31 level associates with poor prognosis of gastric cancer and its restoration suppresses tumor cell malignant phenotypes by inhibiting E2F2. Oncotarget. 2016;7:36577–36589. doi: 10.18632/oncotarget.9288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Zhang Y., Guo J., Li D., Xiao B., Miao Y., Jiang Z., Zhuo H. Down-regulation of miR-31 expression in gastric cancer tissues and its clinical significance. Med. Oncol. 2010;27:685–689. doi: 10.1007/s12032-009-9269-x. [DOI] [PubMed] [Google Scholar]
  • 211.Au S.L.K., Wong C.C.L., Lee J.M.F., Fan D.N.Y., Tsang F.H., Ng I.O.L., Wong C.M. Enhancer of zeste homolog 2 epigenetically silences multiple tumor suppressor microRNAs to promote liver cancer metastasis. Hepatology. 2012;56:622–631. doi: 10.1002/hep.25679. [DOI] [PubMed] [Google Scholar]
  • 212.Saramäki O.R., Tammela T.L., Martikainen P.M., Vessella R.L., Visakorpi T. The gene for polycomb group protein enhancer of zeste homolog 2 (EZH2) is amplified in late-stage prostate cancer. Genes Chromosomes Cancer. 2006;45:639–645. doi: 10.1002/gcc.20327. [DOI] [PubMed] [Google Scholar]
  • 213.Mimori K., Ogawa K., Okamoto M., Sudo T., Inoue H., Mori M. Clinical significance of enhancer of zeste homolog 2 expression in colorectal cancer cases. Eur. J. Surg. Oncol. 2005;31:376–380. doi: 10.1016/j.ejso.2004.11.001. [DOI] [PubMed] [Google Scholar]
  • 214.Sun K.K., Shen X.J., Yang D., Gan M.Q., Liu G., Zhang Y.F., Hua P., Wang H.D., Wu X.Y. MicroRNA-31 triggers G2/M cell cycle arrest, enhances the chemosensitivity and inhibits migration and invasion of human gastric cancer cells by downregulating the expression of zeste homolog 2 (ZH2) Arch. Biochem. Biophys. 2019;663:269–275. doi: 10.1016/j.abb.2019.01.023. [DOI] [PubMed] [Google Scholar]
  • 215.Feng X., Wang Z., Fillmore R., Xi Y. miR-200, a new star miRNA in human cancer. Cancer Lett. 2014;344:166–173. doi: 10.1016/j.canlet.2013.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Liu X., Du P., Han L., Zhang A., Jiang K., Zhang Q. Effects of miR-200a and FH535 combined with taxol on proliferation and invasion of gastric cancer. Pathol. Res. Pract. 2018;214:442–449. doi: 10.1016/j.prp.2017.12.004. [DOI] [PubMed] [Google Scholar]
  • 217.Chang L., Guo F., Wang Y., Lv Y., Huo B., Wang L., Liu W. MicroRNA-200c regulates the sensitivity of chemotherapy of gastric cancer SGC7901/DDP cells by directly targeting RhoE. Pathol. Oncol. Res. 2014;20:93–98. doi: 10.1007/s12253-013-9664-7. [DOI] [PubMed] [Google Scholar]
  • 218.Jiang T., Dong P., Li L., Ma X., Xu P., Zhu H., Wang Y., Yang B., Liu K., Liu J. MicroRNA-200c regulates cisplatin resistance by targeting ZEB2 in human gastric cancer cells. Oncol. Rep. 2017;38:151–158. doi: 10.3892/or.2017.5659. [DOI] [PubMed] [Google Scholar]
  • 219.Zhang H., Sun Z., Li Y., Fan D., Jiang H. MicroRNA-200c binding to FN1 suppresses the proliferation, migration and invasion of gastric cancer cells. Biomed. Pharmacother. 2017;88:285–292. doi: 10.1016/j.biopha.2017.01.023. [DOI] [PubMed] [Google Scholar]
  • 220.Liu Y., Zhu S.-T., Wang X., Deng J., Li W.-H., Zhang P., Liu B.-S. miR-200c regulates tumor growth and chemosensitivity to cisplatin in osteosarcoma by targeting AKT2. Sci. Rep. 2017;7:13598. doi: 10.1038/s41598-017-14088-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Ghasabi M., Majidi J., Mansoori B., Mohammadi A., Shomali N., Shirafkan N., Baghbani E., Kazemi T., Baradaran B. The effect of combined miR-200c replacement and cisplatin on apoptosis induction and inhibition of gastric cancer cell line migration. J. Cell. Physiol. 2019;234:22581–22592. doi: 10.1002/jcp.28823. [DOI] [PubMed] [Google Scholar]
  • 222.Ni F., Zhao H., Cui H., Wu Z., Chen L., Hu Z., Guo C., Liu Y., Chen Z., Wang X. MicroRNA-362-5p promotes tumor growth and metastasis by targeting CYLD in hepatocellular carcinoma. Cancer Lett. 2015;356(2 Pt B):809–818. doi: 10.1016/j.canlet.2014.10.041. [DOI] [PubMed] [Google Scholar]
  • 223.Wei X., Gao M., Ahmed Y., Gao M., Liu W., Zhang Y., Xie X., Zhao Q., Wang H., Gu K. MicroRNA-362-5p enhances the cisplatin sensitivity of gastric cancer cells by targeting suppressor of zeste 12 protein. Oncol. Lett. 2019;18:1607–1616. doi: 10.3892/ol.2019.10496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Wang X., Zhang H., Bai M., Ning T., Ge S., Deng T., Liu R., Zhang L., Ying G., Ba Y. Exosomes serve as nanoparticles to deliver anti-miR-214 to reverse chemoresistance to cisplatin in gastric cancer. Mol. Ther. 2018;26:774–783. doi: 10.1016/j.ymthe.2018.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Korourian A., Roudi R., Shariftabrizi A., Madjd Z. MicroRNA-31 inhibits RhoA-mediated tumor invasion and chemotherapy resistance in MKN-45 gastric adenocarcinoma cells. Exp. Biol. Med. (Maywood) 2017;242:1842–1847. doi: 10.1177/1535370217728460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Zhang X., Yao J., Guo K., Huang H., Huai S., Ye R., Niu B., Ji T., Han W., Li J. The functional mechanism of miR-125b in gastric cancer and its effect on the chemosensitivity of cisplatin. Oncotarget. 2017;9:2105–2119. doi: 10.18632/oncotarget.23249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Sacconi A., Biagioni F., Canu V., Mori F., Di Benedetto A., Lorenzon L., Ercolani C., Di Agostino S., Cambria A.M., Germoni S. miR-204 targets Bcl-2 expression and enhances responsiveness of gastric cancer. Cell Death Dis. 2012;3:e423. doi: 10.1038/cddis.2012.160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Li L.Q., Pan D., Chen Q., Zhang S.W., Xie D.Y., Zheng X.L., Chen H. Sensitization of gastric cancer cells to 5-FU by microRNA-204 through targeting the TGFBR2-mediated epithelial to mesenchymal transition. Cell. Physiol. Biochem. 2018;47:1533–1545. doi: 10.1159/000490871. [DOI] [PubMed] [Google Scholar]
  • 229.Guo Y., Yue P., Wang Y., Chen G., Li Y. PCAT-1 contributes to cisplatin resistance in gastric cancer through miR-128/ZEB1 axis. Biomed. Pharmacother. 2019;118:109255. doi: 10.1016/j.biopha.2019.109255. [DOI] [PubMed] [Google Scholar]
  • 230.Cao W., Wei W., Zhan Z., Xie D., Xie Y., Xiao Q. Regulation of drug resistance and metastasis of gastric cancer cells via the microRNA647-ANK2 axis. Int. J. Mol. Med. 2018;41:1958–1966. doi: 10.3892/ijmm.2018.3381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Wang Y., Liu C., Luo M., Zhang Z., Gong J., Li J., You L., Dong L., Su R., Lin H. Chemotherapy-induced miRNA-29c/catenin-δ signaling suppresses metastasis in gastric cancer. Cancer Res. 2015;75:1332–1344. doi: 10.1158/0008-5472.CAN-14-0787. [DOI] [PubMed] [Google Scholar]
  • 232.Zhou X., Men X., Zhao R., Han J., Fan Z., Wang Y., Lv Y., Zuo J., Zhao L., Sang M. miR-200c inhibits TGF-β-induced-EMT to restore trastuzumab sensitivity by targeting ZEB1 and ZEB2 in gastric cancer. Cancer Gene Ther. 2018;25:68–76. doi: 10.1038/s41417-017-0005-y. [DOI] [PubMed] [Google Scholar]
  • 233.Han X., Zhang J.J., Han Z.Q., Zhang H.B., Wang Z.A. Let-7b attenuates cisplatin resistance and tumor growth in gastric cancer by targeting AURKB. Cancer Gene Ther. 2018;25:300–308. doi: 10.1038/s41417-018-0048-8. [DOI] [PubMed] [Google Scholar]
  • 234.Yan L.H., Chen Z.N., Li-Li, Chen J., Wei W.E., Mo X.W., Qin Y.Z., Lin Y., Chen J.S. miR-135a promotes gastric cancer progression and resistance to oxaliplatin. Oncotarget. 2016;7:70699–70714. doi: 10.18632/oncotarget.12208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Zhao J., Nie Y., Wang H., Lin Y. miR-181a suppresses autophagy and sensitizes gastric cancer cells to cisplatin. Gene. 2016;576:828–833. doi: 10.1016/j.gene.2015.11.013. [DOI] [PubMed] [Google Scholar]
  • 236.Chen J., Zhou C., Li J., Xiang X., Zhang L., Deng J., Xiong J. miR-21-5p confers doxorubicin resistance in gastric cancer cells by targeting PTEN and TIMP3. Int. J. Mol. Med. 2018;41:1855–1866. doi: 10.3892/ijmm.2018.3405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Wen L., Cheng F., Zhou Y., Yin C. miR-26a enhances the sensitivity of gastric cancer cells to cisplatin by targeting NRAS and E2F2. Saudi J. Gastroenterol. 2015;21:313–319. doi: 10.4103/1319-3767.166206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Pang K., Song J., Bai Z., Zhang Z. miR-15a-5p targets PHLPP2 in gastric cancer cells to modulate platinum resistance and is a suitable serum biomarker for oxaliplatin resistance. Neoplasma. 2020;67:1114–1121. doi: 10.4149/neo_2020_190904N861. [DOI] [PubMed] [Google Scholar]
  • 239.Wang J., Xue X., Hong H., Qin M., Zhou J., Sun Q., Liang H., Gao L. Upregulation of microRNA-524-5p enhances the cisplatin sensitivity of gastric cancer cells by modulating proliferation and metastasis via targeting SOX9. Oncotarget. 2017;8:574–582. doi: 10.18632/oncotarget.13479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Zhang Z., Kong Y., Yang W., Ma F., Zhang Y., Ji S., Ma E.M., Liu H., Chen Y., Hua Y. Upregulation of microRNA-34a enhances the DDP sensitivity of gastric cancer cells by modulating proliferation and apoptosis via targeting MET. Oncol. Rep. 2016;36:2391–2397. doi: 10.3892/or.2016.5016. [DOI] [PubMed] [Google Scholar]
  • 241.Zhang Z., Kong Y., Yang W., Zhang B., Ma F., Liu H., Hua Y. MicroRNA-218 enhances gastric cancer cell cisplatin sensitivity by targeting survivin. Exp. Ther. Med. 2018;16:4796–4802. doi: 10.3892/etm.2018.6802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Zhou N., Qu Y., Xu C., Tang Y. Upregulation of microRNA-375 increases the cisplatin-sensitivity of human gastric cancer cells by regulating ERBB2. Exp. Ther. Med. 2016;11:625–630. doi: 10.3892/etm.2015.2920. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 243.Wang P., Li Z., Liu H., Zhou D., Fu A., Zhang E. MicroRNA-126 increases chemosensitivity in drug-resistant gastric cancer cells by targeting EZH2. Biochem. Biophys. Res. Commun. 2016;479:91–96. doi: 10.1016/j.bbrc.2016.09.040. [DOI] [PubMed] [Google Scholar]
  • 244.Li N., Han M., Zhou N., Tang Y., Tang X.S. MicroRNA-495 confers increased sensitivity to chemotherapeutic agents in gastric cancer via the mammalian target of rapamycin (mTOR) signaling pathway by interacting with human epidermal growth factor receptor 2 (ERBB2) Med. Sci. Monit. 2018;24:5960–5972. doi: 10.12659/MSM.909458. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 245.Pires A.O., Mendes-Pinheiro B., Teixeira F.G., Anjo S.I., Ribeiro-Samy S., Gomes E.D., Serra S.C., Silva N.A., Manadas B., Sousa N., Salgado A.J. Unveiling the differences of secretome of human bone marrow mesenchymal stem cells, adipose tissue-derived stem cells, and human umbilical cord perivascular cells: a proteomic analysis. Stem Cells Dev. 2016;25:1073–1083. doi: 10.1089/scd.2016.0048. [DOI] [PubMed] [Google Scholar]
  • 246.Li Q., Wang J.X., He Y.Q., Feng C., Zhang X.J., Sheng J.Q., Li P.F. MicroRNA-185 regulates chemotherapeutic sensitivity in gastric cancer by targeting apoptosis repressor with caspase recruitment domain. Cell Death Dis. 2014;5:e1197. doi: 10.1038/cddis.2014.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Zhu P., Zhang J., Zhu J., Shi J., Zhu Q., Gao Y. miR-429 induces gastric carcinoma cell apoptosis through Bcl-2. Cell. Physiol. Biochem. 2015;37:1572–1580. doi: 10.1159/000438524. [DOI] [PubMed] [Google Scholar]
  • 248.Zhu M., Zhou X., Du Y., Huang Z., Zhu J., Xu J., Cheng G., Shu Y., Liu P., Zhu W., Wang T. miR-20a induces cisplatin resistance of a human gastric cancer cell line via targeting CYLD. Mol. Med. Rep. 2016;14:1742–1750. doi: 10.3892/mmr.2016.5413. [DOI] [PubMed] [Google Scholar]
  • 249.Zhao D., Zhang Y., Song L. miR-16-1 targeted silences far upstream element binding protein 1 to advance the chemosensitivity to adriamycin in gastric cancer. Pathol. Oncol. Res. 2018;24:483–488. doi: 10.1007/s12253-017-0263-x. [DOI] [PubMed] [Google Scholar]
  • 250.Yang S.M., Huang C., Li X.F., Yu M.Z., He Y., Li J. miR-21 confers cisplatin resistance in gastric cancer cells by regulating PTEN. Toxicology. 2013;306:162–168. doi: 10.1016/j.tox.2013.02.014. [DOI] [PubMed] [Google Scholar]
  • 251.Zhou X., Jin W., Jia H., Yan J., Zhang G. miR-223 promotes the cisplatin resistance of human gastric cancer cells via regulating cell cycle by targeting FBXW7. J. Exp. Clin. Cancer Res. 2015;34:28. doi: 10.1186/s13046-015-0145-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Lu C., Shan Z., Li C., Yang L. miR-129 regulates cisplatin-resistance in human gastric cancer cells by targeting P-gp. Biomed. Pharmacother. 2017;86:450–456. doi: 10.1016/j.biopha.2016.11.139. [DOI] [PubMed] [Google Scholar]
  • 253.Eto K., Iwatsuki M., Watanabe M., Ida S., Ishimoto T., Iwagami S., Baba Y., Sakamoto Y., Miyamoto Y., Yoshida N., Baba H. The microRNA-21/PTEN pathway regulates the sensitivity of HER2-positive gastric cancer cells to trastuzumab. Ann. Surg. Oncol. 2014;21:343–350. doi: 10.1245/s10434-013-3325-7. [DOI] [PubMed] [Google Scholar]
  • 254.Qian Y., Wu X., Wang H., Hou G., Han X., Song W. MicroRNA-4290 suppresses PDK1-mediated glycolysis to enhance the sensitivity of gastric cancer cell to cisplatin. Braz. J. Med. Biol. Res. 2020;53:e9330. doi: 10.1590/1414-431X20209330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.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. doi: 10.1007/s12020-014-0326-7. [DOI] [PubMed] [Google Scholar]
  • 256.Zha W., Cao L., Shen Y., Huang M. Roles of Mir-144-ZFX pathway in growth regulation of non-small-cell lung cancer. PLoS ONE. 2013;8:e74175. doi: 10.1371/journal.pone.0074175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Huang X.P., Hou J., Shen X.Y., Huang C.Y., Zhang X.H., Xie Y.A., Luo X.L. MicroRNA-486-5p, which is downregulated in hepatocellular carcinoma, suppresses tumor growth by targeting PIK3R1. FEBS J. 2015;282:579–594. doi: 10.1111/febs.13167. [DOI] [PubMed] [Google Scholar]
  • 258.Bruhn M.A., Pearson R.B., Hannan R.D., Sheppard K.E. AKT-independent PI3-K signaling in cancer—Emerging role for SGK3. Cancer Manag. Res. 2013;5:281–292. doi: 10.2147/CMAR.S35178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Cao T., Li H., Hu Y., Ma D., Cai X. miR-144 suppresses the proliferation and metastasis of hepatocellular carcinoma by targeting E2F3. Tumour Biol. 2014;35:10759–10764. doi: 10.1007/s13277-014-2017-7. [DOI] [PubMed] [Google Scholar]
  • 260.Wang R., Zhao N., Li S., Fang J.H., Chen M.X., Yang J., Jia W.H., Yuan Y., Zhuang S.M. MicroRNA-195 suppresses angiogenesis and metastasis of hepatocellular carcinoma by inhibiting the expression of VEGF, VAV2, and CDC42. Hepatology. 2013;58:642–653. doi: 10.1002/hep.26373. [DOI] [PubMed] [Google Scholar]
  • 261.Yang X., Zhang X.F., Lu X., Jia H.L., Liang L., Dong Q.Z., Ye Q.H., Qin L.X. MicroRNA-26a suppresses angiogenesis in human hepatocellular carcinoma by targeting hepatocyte growth factor-cMet pathway. Hepatology. 2014;59:1874–1885. doi: 10.1002/hep.26941. [DOI] [PubMed] [Google Scholar]
  • 262.Wu M., Huang C., Huang X., Liang R., Feng Y., Luo X. MicroRNA-144-3p suppresses tumor growth and angiogenesis by targeting SGK3 in hepatocellular carcinoma. Oncol. Rep. 2017;38:2173–2181. doi: 10.3892/or.2017.5900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Agarwal V., Bell G.W., Nam J.-W., Bartel D.P. Predicting effective microRNA target sites in mammalian mRNAs. Elife. 2015;4:e05005. doi: 10.7554/eLife.05005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Kutay H., Bai S., Datta J., Motiwala T., Pogribny I., Frankel W., Jacob S.T., Ghoshal K. Downregulation of miR-122 in the rodent and human hepatocellular carcinomas. J. Cell. Biochem. 2006;99:671–678. doi: 10.1002/jcb.20982. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 265.Xu Y., Xia F., Ma L., Shan J., Shen J., Yang Z., Liu J., Cui Y., Bian X., Bie P., Qian C. MicroRNA-122 sensitizes HCC cancer cells to adriamycin and vincristine through modulating expression of MDR and inducing cell cycle arrest. Cancer Lett. 2011;310:160–169. doi: 10.1016/j.canlet.2011.06.027. [DOI] [PubMed] [Google Scholar]
  • 266.Fornari F., Gramantieri L., Giovannini C., Veronese A., Ferracin M., Sabbioni S., Calin G.A., Grazi G.L., Croce C.M., Tavolari S. miR-122/cyclin G1 interaction modulates p53 activity and affects doxorubicin sensitivity of human hepatocarcinoma cells. Cancer Res. 2009;69:5761–5767. doi: 10.1158/0008-5472.CAN-08-4797. [DOI] [PubMed] [Google Scholar]
  • 267.Coulouarn C., Factor V.M., Andersen J.B., Durkin M.E., Thorgeirsson S.S. Loss of miR-122 expression in liver cancer correlates with suppression of the hepatic phenotype and gain of metastatic properties. Oncogene. 2009;28:3526–3536. doi: 10.1038/onc.2009.211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Tsai W.C., Hsu P.W., Lai T.C., Chau G.Y., Lin C.W., Chen C.M., Lin C.D., Liao Y.L., Wang J.L., Chau Y.P. MicroRNA-122, a tumor suppressor microRNA that regulates intrahepatic metastasis of hepatocellular carcinoma. Hepatology. 2009;49:1571–1582. doi: 10.1002/hep.22806. [DOI] [PubMed] [Google Scholar]
  • 269.Ma L., Liu J., Shen J., Liu L., Wu J., Li W., Luo J., Chen Q., Qian C. Expression of miR-122 mediated by adenoviral vector induces apoptosis and cell cycle arrest of cancer cells. Cancer Biol. Ther. 2010;9:554–561. doi: 10.4161/cbt.9.7.11267. [DOI] [PubMed] [Google Scholar]
  • 270.Bandiera S., Pfeffer S., Baumert T.F., Zeisel M.B. miR-122—A key factor and therapeutic target in liver disease. J. Hepatol. 2015;62:448–457. doi: 10.1016/j.jhep.2014.10.004. [DOI] [PubMed] [Google Scholar]
  • 271.Morita K., Taketomi A., Shirabe K., Umeda K., Kayashima H., Ninomiya M., Uchiyama H., Soejima Y., Maehara Y. Clinical significance and potential of hepatic microRNA-122 expression in hepatitis C. Liver Int. 2011;31:474–484. doi: 10.1111/j.1478-3231.2010.02433.x. [DOI] [PubMed] [Google Scholar]
  • 272.Bai S., Nasser M.W., Wang B., Hsu S.H., Datta J., Kutay H., Yadav A., Nuovo G., Kumar P., Ghoshal K. MicroRNA-122 inhibits tumorigenic properties of hepatocellular carcinoma cells and sensitizes these cells to sorafenib. J. Biol. Chem. 2009;284:32015–32027. doi: 10.1074/jbc.M109.016774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Xu J., Zhu X., Wu L., Yang R., Yang Z., Wang Q., Wu F. MicroRNA-122 suppresses cell proliferation and induces cell apoptosis in hepatocellular carcinoma by directly targeting Wnt/β-catenin pathway. Liver Int. 2012;32:752–760. doi: 10.1111/j.1478-3231.2011.02750.x. [DOI] [PubMed] [Google Scholar]
  • 274.Grainger S., Willert K. Mechanisms of Wnt signaling and control. Wiley Interdiscip. Rev. Syst. Biol. Med. 2018;10:e1422. doi: 10.1002/wsbm.1422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Liu L.J., Xie S.X., Chen Y.T., Xue J.L., Zhang C.J., Zhu F. Aberrant regulation of Wnt signaling in hepatocellular carcinoma. World J. Gastroenterol. 2016;22:7486–7499. doi: 10.3748/wjg.v22.i33.7486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Cao F., Yin L.X. miR-122 enhances sensitivity of hepatocellular carcinoma to oxaliplatin via inhibiting MDR1 by targeting Wnt/β-catenin pathway. Exp. Mol. Pathol. 2019;106:34–43. doi: 10.1016/j.yexmp.2018.10.009. [DOI] [PubMed] [Google Scholar]
  • 277.Lei R., Tang J., Zhuang X., Deng R., Li G., Yu J., Liang Y., Xiao J., Wang H.Y., Yang Q., Hu G. Suppression of MIM by microRNA-182 activates RhoA and promotes breast cancer metastasis. Oncogene. 2014;33:1287–1296. doi: 10.1038/onc.2013.65. [DOI] [PubMed] [Google Scholar]
  • 278.Wang Y.Q., Guo R.D., Guo R.M., Sheng W., Yin L.R. MicroRNA-182 promotes cell growth, invasion, and chemoresistance by targeting programmed cell death 4 (PDCD4) in human ovarian carcinomas. J. Cell. Biochem. 2013;114:1464–1473. doi: 10.1002/jcb.24488. [DOI] [PubMed] [Google Scholar]
  • 279.Husted S., Søkilde R., Rask L., Cirera S., Busk P.K., Eriksen J., Litman T. MicroRNA expression profiles associated with development of drug resistance in Ehrlich ascites tumor cells. Mol. Pharm. 2011;8:2055–2062. doi: 10.1021/mp200255d. [DOI] [PubMed] [Google Scholar]
  • 280.Yang A., Ma J., Wu M., Qin W., Zhao B., Shi Y., Jin Y., Xie Y. Aberrant microRNA-182 expression is associated with glucocorticoid resistance in lymphoblastic malignancies. Leuk. Lymphoma. 2012;53:2465–2473. doi: 10.3109/10428194.2012.693178. [DOI] [PubMed] [Google Scholar]
  • 281.Qin J., Luo M., Qian H., Chen W. Upregulated miR-182 increases drug resistance in cisplatin-treated HCC cell by regulating TP53INP1. Gene. 2014;538:342–347. doi: 10.1016/j.gene.2013.12.043. [DOI] [PubMed] [Google Scholar]
  • 282.Sümbül A.T., Göğebakan B., Ergün S., Yengil E., Batmacı C.Y., Tonyalı Ö., Yaldız M. miR-204-5p expression in colorectal cancer: An autophagy-associated gene. Tumour Biol. 2014;35:12713–12719. doi: 10.1007/s13277-014-2596-3. [DOI] [PubMed] [Google Scholar]
  • 283.Butrym A., Rybka J., Baczyńska D., Tukiendorf A., Kuliczkowski K., Mazur G. Low expression of microRNA-204 (miR-204) is associated with poor clinical outcome of acute myeloid leukemia (AML) patients. J. Exp. Clin. Cancer Res. 2015;34:68. doi: 10.1186/s13046-015-0184-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Luo Y.H., Tang W., Zhang X., Tan Z., Guo W.L., Zhao N., Pang S.M., Dang Y.W., Rong M.H., Cao J. Promising significance of the association of miR-204-5p expression with clinicopathological features of hepatocellular carcinoma. Medicine (Baltimore) 2017;96:e7545. doi: 10.1097/MD.0000000000007545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Jiang G., Wen L., Zheng H., Jian Z., Deng W. miR-204-5p targeting SIRT1 regulates hepatocellular carcinoma progression. Cell Biochem. Funct. 2016;34:505–510. doi: 10.1002/cbf.3223. [DOI] [PubMed] [Google Scholar]
  • 286.Zeng L., Yu J., Huang T., Jia H., Dong Q., He F., Yuan W., Qin L., Li Y., Xie L. Differential combinatorial regulatory network analysis related to venous metastasis of hepatocellular carcinoma. BMC Genomics. 2012;13(Suppl 8):S14. doi: 10.1186/1471-2164-13-S8-S14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Yu Y., Wang Y., Xiao X., Cheng W., Hu L., Yao W., Qian Z., Wu W. miR-204 inhibits hepatocellular cancer drug resistance and metastasis through targeting NUAK1. Biochem. Cell Biol. 2019;97:563–570. doi: 10.1139/bcb-2018-0354. [DOI] [PubMed] [Google Scholar]
  • 288.Zheng H., Zou A.E., Saad M.A., Wang X.Q., Kwok J.G., Korrapati A., Li P., Kisseleva T., Wang-Rodriguez J., Ongkeko W.M. Alcohol-dysregulated microRNAs in hepatitis B virus-related hepatocellular carcinoma. PLoS ONE. 2017;12:e0178547. doi: 10.1371/journal.pone.0178547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Yang T., Zheng Z.M., Li X.N., Li Z.F., Wang Y., Geng Y.F., Bai L., Zhang X.B. miR-223 modulates multidrug resistance via downregulation of ABCB1 in hepatocellular carcinoma cells. Exp. Biol. Med. (Maywood) 2013;238:1024–1032. doi: 10.1177/1535370213497321. [DOI] [PubMed] [Google Scholar]
  • 290.Ji J., Zheng X., Forgues M., Yamashita T., Wauthier E.L., Reid L.M., Wen X., Song Y., Wei J.S., Khan J. Identification of microRNAs specific for epithelial cell adhesion molecule-positive tumor cells in hepatocellular carcinoma. Hepatology. 2015;62:829–840. doi: 10.1002/hep.27886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Wong Q.W., Lung R.W., Law P.T., Lai P.B., Chan K.Y., To K.F., Wong N. MicroRNA-223 is commonly repressed in hepatocellular carcinoma and potentiates expression of Stathmin1. Gastroenterology. 2008;135:257–269. doi: 10.1053/j.gastro.2008.04.003. [DOI] [PubMed] [Google Scholar]
  • 292.Dong Y.W., Wang R., Cai Q.Q., Qi B., Wu W., Zhang Y.H., Wu X.Z. Sulfatide epigenetically regulates miR-223 and promotes the migration of human hepatocellular carcinoma cells. J. Hepatol. 2014;60:792–801. doi: 10.1016/j.jhep.2013.12.004. [DOI] [PubMed] [Google Scholar]
  • 293.Zhou J., Yu L., Gao X., Hu J., Wang J., Dai Z., Wang J.F., Zhang Z., Lu S., Huang X. Plasma microRNA panel to diagnose hepatitis B virus-related hepatocellular carcinoma. J. Clin. Oncol. 2011;29:4781–4788. doi: 10.1200/JCO.2011.38.2697. [DOI] [PubMed] [Google Scholar]
  • 294.Liu X., Deng Y., Xu Y., Jin W., Li H. MicroRNA-223 protects neonatal rat cardiomyocytes and H9c2 cells from hypoxia-induced apoptosis and excessive autophagy via the Akt/mTOR pathway by targeting PARP-1. J. Mol. Cell. Cardiol. 2018;118:133–146. doi: 10.1016/j.yjmcc.2018.03.018. [DOI] [PubMed] [Google Scholar]
  • 295.Zhou Y., Chen E., Tang Y., Mao J., Shen J., Zheng X., Xie S., Zhang S., Wu Y., Liu H. miR-223 overexpression inhibits doxorubicin-induced autophagy by targeting FOXO3a and reverses chemoresistance in hepatocellular carcinoma cells. Cell Death Dis. 2019;10:843. doi: 10.1038/s41419-019-2053-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Budhu A., Jia H.L., Forgues M., Liu C.G., Goldstein D., Lam A., Zanetti K.A., Ye Q.H., Qin L.X., Croce C.M. Identification of metastasis-related microRNAs in hepatocellular carcinoma. Hepatology. 2008;47:897–907. doi: 10.1002/hep.22160. [DOI] [PubMed] [Google Scholar]
  • 297.Ura S., Honda M., Yamashita T., Ueda T., Takatori H., Nishino R., Sunakozaka H., Sakai Y., Horimoto K., Kaneko S. Differential microRNA expression between hepatitis B and hepatitis C leading disease progression to hepatocellular carcinoma. Hepatology. 2009;49:1098–1112. doi: 10.1002/hep.22749. [DOI] [PubMed] [Google Scholar]
  • 298.Kim J.K., Noh J.H., Jung K.H., Eun J.W., Bae H.J., Kim M.G., Chang Y.G., Shen Q., Park W.S., Lee J.Y. Sirtuin7 oncogenic potential in human hepatocellular carcinoma and its regulation by the tumor suppressors miR-125a-5p and miR-125b. Hepatology. 2013;57:1055–1067. doi: 10.1002/hep.26101. [DOI] [PubMed] [Google Scholar]
  • 299.Liang L., Wong C.M., Ying Q., Fan D.N., Huang S., Ding J., Yao J., Yan M., Li J., Yao M. MicroRNA-125b suppressesed human liver cancer cell proliferation and metastasis by directly targeting oncogene LIN28B2. Hepatology. 2010;52:1731–1740. doi: 10.1002/hep.23904. [DOI] [PubMed] [Google Scholar]
  • 300.Li W., Xie L., He X., Li J., Tu K., Wei L., Wu J., Guo Y., Ma X., Zhang P. Diagnostic and prognostic implications of microRNAs in human hepatocellular carcinoma. Int. J. Cancer. 2008;123:1616–1622. doi: 10.1002/ijc.23693. [DOI] [PubMed] [Google Scholar]
  • 301.Ren W.W., Li D.D., Chen X., Li X.L., He Y.P., Guo L.H., Liu L.N., Sun L.P., Zhang X.P. MicroRNA-125b reverses oxaliplatin resistance in hepatocellular carcinoma by negatively regulating EVA1A mediated autophagy. Cell Death Dis. 2018;9:547. doi: 10.1038/s41419-018-0592-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Stinson S., Lackner M.R., Adai A.T., Yu N., Kim H.J., O’Brien C., Spoerke J., Jhunjhunwala S., Boyd Z., Januario T. miR-221/222 targeting of trichorhinophalangeal 1 (TRPS1) promotes epithelial-to-mesenchymal transition in breast cancer. Sci. Signal. 2011;4:pt5. doi: 10.1126/scisignal.2002258. [DOI] [PubMed] [Google Scholar]
  • 303.Yang C.J., Shen W.G., Liu C.J., Chen Y.W., Lu H.H., Tsai M.M., Lin S.C. miR-221 and miR-222 expression increased the growth and tumorigenesis of oral carcinoma cells. J. Oral Pathol. Med. 2011;40:560–566. doi: 10.1111/j.1600-0714.2010.01005.x. [DOI] [PubMed] [Google Scholar]
  • 304.Lu Y., Roy S., Nuovo G., Ramaswamy B., Miller T., Shapiro C., Jacob S.T., Majumder S. Retraction. J. Biol. Chem. 2018;293:3588. doi: 10.1074/jbc.W118.002430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Miller T.E., Ghoshal K., Ramaswamy B., Roy S., Datta J., Shapiro C.L., Jacob S., Majumder S. MicroRNA-221/222 confers tamoxifen resistance in breast cancer by targeting p27Kip1. J. Biol. Chem. 2008;283:29897–29903. doi: 10.1074/jbc.M804612200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Zhang C.Z., Zhang J.X., Zhang A.L., Shi Z.D., Han L., Jia Z.F., Yang W.D., Wang G.X., Jiang T., You Y.P. miR-221 and miR-222 target PUMA to induce cell survival in glioblastoma. Mol. Cancer. 2010;9:229. doi: 10.1186/1476-4598-9-229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307.Wong Q.W., Ching A.K., Chan A.W., Choy K.W., To K.F., Lai P.B., Wong N. miR-222 overexpression confers cell migratory advantages in hepatocellular carcinoma through enhancing AKT signaling. Clin. Cancer Res. 2010;16:867–875. doi: 10.1158/1078-0432.CCR-09-1840. [DOI] [PubMed] [Google Scholar]
  • 308.Zhong S., Li W., Chen Z., Xu J., Zhao J. miR-222 and miR-29a contribute to the drug-resistance of breast cancer cells. Gene. 2013;531:8–14. doi: 10.1016/j.gene.2013.08.062. [DOI] [PubMed] [Google Scholar]
  • 309.Garofalo M., Di Leva G., Romano G., Nuovo G., Suh S.S., Ngankeu A., Taccioli C., Pichiorri F., Alder H., Secchiero P. miR-221&222 regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell. 2009;16:498–509. doi: 10.1016/j.ccr.2009.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 310.Liu K., Liu S., Zhang W., Ji B., Wang Y., Liu Y. miR-222 regulates sorafenib resistance and enhance tumorigenicity in hepatocellular carcinoma. Int. J. Oncol. 2014;45:1537–1546. doi: 10.3892/ijo.2014.2577. [DOI] [PubMed] [Google Scholar]
  • 311.He X., Li J., Guo W., Liu W., Yu J., Song W., Dong L., Wang F., Yu S., Zheng Y. Targeting the microRNA-21/AP1 axis by 5-fluorouracil and pirarubicin in human hepatocellular carcinoma. Oncotarget. 2015;6:2302–2314. doi: 10.18632/oncotarget.2955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Ma J., Wang T., Guo R., Yang X., Yin J., Yu J., Xiang Q., Pan X., Zu X., Peng C. MicroRNA-133a and microRNA-326 co-contribute to hepatocellular carcinoma 5-fluorouracil and cisplatin sensitivity by directly targeting B-cell lymphoma-extra large. Mol. Med. Rep. 2015;12:6235–6240. doi: 10.3892/mmr.2015.4134. [DOI] [PubMed] [Google Scholar]
  • 313.Zhuang Q., Zhou T., He C., Zhang S., Qiu Y., Luo B., Zhao R., Liu H., Lin Y., Lin Z. Protein phosphatase 2A-B55δ enhances chemotherapy sensitivity of human hepatocellular carcinoma under the regulation of microRNA-133b. J. Exp. Clin. Cancer Res. 2016;35:67. doi: 10.1186/s13046-016-0341-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314.Wang L., Wang Y.M., Xu S., Wang W.G., Chen Y., Mao J.Y., Tian B.L. MicroRNA-215 is upregulated by treatment with adriamycin and leads to the chemoresistance of hepatocellular carcinoma cells and tissues. Mol. Med. Rep. 2015;12:5274–5280. doi: 10.3892/mmr.2015.4012. [DOI] [PubMed] [Google Scholar]
  • 315.Zheng T., Feng H., Liu L., Peng J., Xiao H., Yu T., Zhou Z., Li Y., Zhang Y., Bai X. Enhanced antiproliferative effect of resveratrol in head and neck squamous cell carcinoma using GE11 peptide conjugated liposome. Int. J. Mol. Med. 2019;43:1635–1642. doi: 10.3892/ijmm.2019.4096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316.Meng W., Tai Y., Zhao H., Fu B., Zhang T., Liu W., Li H., Yang Y., Zhang Q., Feng Y., Chen G. Downregulation of miR-33a-5p in hepatocellular carcinoma: A possible mechanism for chemotherapy resistance. Med. Sci. Monit. 2017;23:1295–1304. doi: 10.12659/MSM.902692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317.Chen M., Wu L., Tu J., Zhao Z., Fan X., Mao J., Weng Q., Wu X., Huang L., Xu M., Ji J. miR-590-5p suppresses hepatocellular carcinoma chemoresistance by targeting YAP1 expression. EBioMedicine. 2018;35:142–154. doi: 10.1016/j.ebiom.2018.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Jin F., Wang Y., Li M., Zhu Y., Liang H., Wang C., Wang F., Zhang C.Y., Zen K., Li L. miR-26 enhances chemosensitivity and promotes apoptosis of hepatocellular carcinoma cells through inhibiting autophagy. Cell Death Dis. 2017;8:e2540. doi: 10.1038/cddis.2016.461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319.Zheng S.Z., Sun P., Wang J.P., Liu Y., Gong W., Liu J. miR-34a overexpression enhances the inhibitory effect of doxorubicin on HepG2 cells. World J. Gastroenterol. 2019;25:2752–2762. doi: 10.3748/wjg.v25.i22.2752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320.Huang Y., Chen G., Wang Y., He R., Du J., Jiao X., Tai Q. Inhibition of microRNA-16 facilitates the paclitaxel resistance by targeting IKBKB via NF-κB signaling pathway in hepatocellular carcinoma. Biochem. Biophys. Res. Commun. 2018;503:1035–1041. doi: 10.1016/j.bbrc.2018.06.113. [DOI] [PubMed] [Google Scholar]
  • 321.Wang W., Liu W.B., Huang B., Jia W., Ji C.S., Hu B. Targeting PCDH20 gene by microRNA-122 confers 5-FU resistance in hepatic carcinoma. Am. J. Cancer Res. 2016;6:1681–1694. [PMC free article] [PubMed] [Google Scholar]
  • 322.Yuan Z., Yu X., Ni B., Chen D., Yang Z., Huang J., Wang J., Chen D., Wang L. Overexpression of long non-coding RNA-CTD903 inhibits colorectal cancer invasion and migration by repressing Wnt/β-catenin signaling and predicts favorable prognosis. Int. J. Oncol. 2016;48:2675–2685. doi: 10.3892/ijo.2016.3447. [DOI] [PubMed] [Google Scholar]
  • 323.Long J., Jiang C., Liu B., Dai Q., Hua R., Chen C., Zhang B., Li H. Maintenance of stemness by miR-589-5p in hepatocellular carcinoma cells promotes chemoresistance via STAT3 signaling. Cancer Lett. 2018;423:113–126. doi: 10.1016/j.canlet.2017.11.031. [DOI] [PubMed] [Google Scholar]
  • 324.Jiang J.X., Gao S., Pan Y.Z., Yu C., Sun C.Y. Overexpression of microRNA-125b sensitizes human hepatocellular carcinoma cells to 5-fluorouracil through inhibition of glycolysis by targeting hexokinase II. Mol. Med. Rep. 2014;10:995–1002. doi: 10.3892/mmr.2014.2271. [DOI] [PubMed] [Google Scholar]
  • 325.Xu Y., Lai Y., Weng H., Tan L., Li Y., Chen G., Luo X., Ye Y. miR-124 sensitizes cisplatin-induced cytotoxicity against CD133+ hepatocellular carcinoma cells by targeting SIRT1/ROS/JNK pathway. Aging (Albany NY) 2019;11:2551–2564. doi: 10.18632/aging.101876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326.Ragheb M.A., Soliman M.H., Elzayat E.M., Mohamed M.S., El-Ekiaby N., Abdelaziz A.I., Abdel-Wahab A.A. MicroR-520c-3p modulates doxorubicin-chemosensitivity in HepG2 cells. Anticancer. Agents Med. Chem. 2021;21:237–245. doi: 10.2174/1871520620666200502004817. [DOI] [PubMed] [Google Scholar]
  • 327.He H., Tian W., Chen H., Deng Y. MicroRNA-101 sensitizes hepatocellular carcinoma cells to doxorubicin-induced apoptosis via targeting Mcl-1. Mol. Med. Rep. 2016;13:1923–1929. doi: 10.3892/mmr.2015.4727. [DOI] [PubMed] [Google Scholar]
  • 328.Shi L., Chen Z.G., Wu L.L., Zheng J.J., Yang J.R., Chen X.F., Chen Z.Q., Liu C.L., Chi S.Y., Zheng J.Y. miR-340 reverses cisplatin resistance of hepatocellular carcinoma cell lines by targeting Nrf2-dependent antioxidant pathway. Asian Pac. J. Cancer Prev. 2014;15:10439–10444. doi: 10.7314/apjcp.2014.15.23.10439. [DOI] [PubMed] [Google Scholar]
  • 329.Yin W., Nie Y., Zhang Z., Xie L., He X. miR-193b acts as a cisplatin sensitizer via the caspase-3-dependent pathway in HCC chemotherapy. Oncol. Rep. 2015;34:368–374. doi: 10.3892/or.2015.3996. [DOI] [PubMed] [Google Scholar]
  • 330.Ou Y., Zhai D., Wu N., Li X. Downregulation of miR-363 increases drug resistance in cisplatin-treated HepG2 by dysregulating Mcl-1. Gene. 2015;572:116–122. doi: 10.1016/j.gene.2015.07.002. [DOI] [PubMed] [Google Scholar]
  • 331.Xu N., Shen C., Luo Y., Xia L., Xue F., Xia Q., Zhang J. Upregulated miR-130a increases drug resistance by regulating RUNX3 and Wnt signaling in cisplatin-treated HCC cell. Biochem. Biophys. Res. Commun. 2012;425:468–472. doi: 10.1016/j.bbrc.2012.07.127. [DOI] [PubMed] [Google Scholar]
  • 332.Yang B., Wang C., Xie H., Wang Y., Huang J., Rong Y., Zhang H., Kong H., Yang Y., Lu Y. MicroRNA-3163 targets ADAM-17 and enhances the sensitivity of hepatocellular carcinoma cells to molecular targeted agents. Cell Death Dis. 2019;10:784. doi: 10.1038/s41419-019-2023-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 333.Xu Y., Huang J., Ma L., Shan J., Shen J., Yang Z., Liu L., Luo Y., Yao C., Qian C. MicroRNA-122 confers sorafenib resistance to hepatocellular carcinoma cells by targeting IGF-1R to regulate RAS/RAF/ERK signaling pathways. Cancer Lett. 2016;371:171–181. doi: 10.1016/j.canlet.2015.11.034. [DOI] [PubMed] [Google Scholar]
  • 334.Fornari F., Pollutri D., Patrizi C., La Bella T., Marinelli S., Casadei Gardini A., Marisi G., Baron Toaldo M., Baglioni M., Salvatore V. Hepatocellular carcinoma miR-221 modulates sorafenib resistance through inhibition of caspase-3-mediated apoptosis. Clin. Cancer Res. 2017;23:3953–3965. doi: 10.1158/1078-0432.CCR-16-1464. [DOI] [PubMed] [Google Scholar]
  • 335.Lu A.Q., Lv B., Qiu F., Wang X.Y., Cao X.H. Upregulation of miR-137 reverses sorafenib resistance and cancer-initiating cell phenotypes by degrading ANT2 in hepatocellular carcinoma. Oncol. Rep. 2017;37:2071–2078. doi: 10.3892/or.2017.5498. [DOI] [PubMed] [Google Scholar]
  • 336.Wang N., Zhu M., Tsao S.W., Man K., Zhang Z., Feng Y. miR-23a-mediated inhibition of topoisomerase 1 expression potentiates cell response to etoposide in human hepatocellular carcinoma. Mol. Cancer. 2013;12:119. doi: 10.1186/1476-4598-12-119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 337.Mu W., Hu C., Zhang H., Qu Z., Cen J., Qiu Z., Li C., Ren H., Li Y., He X. miR-27b synergizes with anticancer drugs via p53 activation and CYP1B1 suppression. Cell Res. 2015;25:477–495. doi: 10.1038/cr.2015.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 338.Soliman M.H., Ragheb M.A., Elzayat E.M., Mohamed M.S., El-Ekiaby N., Abdelaziz A.I., Abdelwahab A.A. miR-372-3p predicts response of TACE patients treated with doxorubicin and enhances chemosensitivity in hepatocellular carcinoma. Anticancer Agents Med. Chem. 2021;21:246–253. doi: 10.2174/1871520620666200516145830. [DOI] [PubMed] [Google Scholar]
  • 339.Tao J., Wu D., Xu B., Qian W., Li P., Lu Q., Yin C., Zhang W. MicroRNA-133 inhibits cell proliferation, migration and invasion in prostate cancer cells by targeting the epidermal growth factor receptor. Oncol. Rep. 2012;27:1967–1975. doi: 10.3892/or.2012.1711. [DOI] [PubMed] [Google Scholar]
  • 340.Zhou Y., Wu D., Tao J., Qu P., Zhou Z., Hou J. MicroRNA-133 inhibits cell proliferation, migration and invasion by targeting epidermal growth factor receptor and its downstream effector proteins in bladder cancer. Scand. J. Urol. 2013;47:423–432. doi: 10.3109/00365599.2012.748821. [DOI] [PubMed] [Google Scholar]
  • 341.Liu L., Shao X., Gao W., Zhang Z., Liu P., Wang R., Huang P., Yin Y., Shu Y. MicroRNA-133b inhibits the growth of non-small-cell lung cancer by targeting the epidermal growth factor receptor. FEBS J. 2012;279:3800–3812. doi: 10.1111/j.1742-4658.2012.08741.x. [DOI] [PubMed] [Google Scholar]
  • 342.Qiu T., Zhou X., Wang J., Du Y., Xu J., Huang Z., Zhu W., Shu Y., Liu P. miR-145, miR-133a and miR-133b inhibit proliferation, migration, invasion and cell cycle progression via targeting transcription factor Sp1 in gastric cancer. FEBS Lett. 2014;588:1168–1177. doi: 10.1016/j.febslet.2014.02.054. [DOI] [PubMed] [Google Scholar]
  • 343.Kano M., Seki N., Kikkawa N., Fujimura L., Hoshino I., Akutsu Y., Chiyomaru T., Enokida H., Nakagawa M., Matsubara H. miR-145, miR-133a and miR-133b: Tumor-suppressive miRNAs target FSCN1 in esophageal squamous cell carcinoma. Int. J. Cancer. 2010;127:2804–2814. doi: 10.1002/ijc.25284. [DOI] [PubMed] [Google Scholar]
  • 344.Ceresa B.P., Peterson J.L. Cell and molecular biology of epidermal growth factor receptor. Int. Rev. Cell Mol. Biol. 2014;313:145–178. doi: 10.1016/B978-0-12-800177-6.00005-0. [DOI] [PubMed] [Google Scholar]
  • 345.Centuori S.M., Martinez J.D. Differential regulation of EGFR-MAPK signaling by deoxycholic acid (DCA) and ursodeoxycholic acid (UDCA) in colon cancer. Dig. Dis. Sci. 2014;59:2367–2380. doi: 10.1007/s10620-014-3190-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Wang F., Xiao W., Sun J., Han D., Zhu Y. Retraction. Tumour Biol. 2014 doi: 10.1007/s13277-017-5487-6. Published online April 20, 2017. [DOI] [PubMed] [Google Scholar]
  • 347.Guo K., Wang W.P., Jiang T., Wang J.Z., Chen Z., Li Y., Zhou Y.A., Li X.F., Lu Q., Zhang L.J. Assessment of epidermal growth factor receptor mutation/copy number and K-ras mutation in esophageal cancer. J. Thorac. Dis. 2016;8:1753–1763. doi: 10.21037/jtd.2016.06.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348.Song J., Shi W., Zhang Y., Sun M., Liang X., Zheng S. Epidermal growth factor receptor and B7-H3 expression in esophageal squamous tissues correlate to patient prognosis. OncoTargets Ther. 2016;9:6257–6263. doi: 10.2147/OTT.S111691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349.Kashyap M.K., Abdel-Rahman O. Expression, regulation and targeting of receptor tyrosine kinases in esophageal squamous cell carcinoma. Mol. Cancer. 2018;17:54. doi: 10.1186/s12943-018-0790-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 350.Pan Y., Ye C., Tian Q., Yan S., Zeng X., Xiao C., Wang L., Wang H. miR-145 suppresses the proliferation, invasion and migration of NSCLC cells by regulating the BAX/BCL-2 ratio and the caspase-3 cascade. Oncol. Lett. 2018;15:4337–4343. doi: 10.3892/ol.2018.7863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351.Ye P., Shi Y., An N., Zhou Q., Guo J., Long X. miR-145 overexpression triggers alteration of the whole transcriptome and inhibits breast cancer development. Biomed. Pharmacother. 2018;100:72–82. doi: 10.1016/j.biopha.2018.01.167. [DOI] [PubMed] [Google Scholar]
  • 352.Azmi A.S., Li Y., Muqbil I., Aboukameel A., Senapedis W., Baloglu E., Landesman Y., Shacham S., Kauffman M.G., Philip P.A., Mohammad R.M. Exportin 1 (XPO1) inhibition leads to restoration of tumor suppressor miR-145 and consequent suppression of pancreatic cancer cell proliferation and migration. Oncotarget. 2017;8:82144–82155. doi: 10.18632/oncotarget.19285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Wu B.L., Xu L.Y., Du Z.P., Liao L.D., Zhang H.F., Huang Q., Fang G.Q., Li E.M. miRNA profile in esophageal squamous cell carcinoma: downregulation of miR-143 and miR-145. World J. Gastroenterol. 2011;17:79–88. doi: 10.3748/wjg.v17.i1.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 354.Mei L.L., Wang W.J., Qiu Y.T., Xie X.F., Bai J., Shi Z.Z. miR-145-5p suppresses tumor cell migration, invasion and epithelial to mesenchymal transition by regulating the Sp1/NF-κB signaling pathway in esophageal squamous cell carcinoma. Int. J. Mol. Sci. 2017;18:1833. doi: 10.3390/ijms18091833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 355.Cui X.B., Li S., Li T.T., Peng H., Jin T.T., Zhang S.M., Liu C.X., Yang L., Shen Y.Y., Li S.G. Targeting oncogenic PLCE1 by miR-145 impairs tumor proliferation and metastasis of esophageal squamous cell carcinoma. Oncotarget. 2016;7:1777–1795. doi: 10.18632/oncotarget.6499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 356.Akao Y., Khoo F., Kumazaki M., Shinohara H., Miki K., Yamada N. Extracellular disposal of tumor-suppressor miRs-145 and -34a via microvesicles and 5-FU resistance of human colon cancer cells. Int. J. Mol. Sci. 2014;15:1392–1401. doi: 10.3390/ijms15011392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 357.Chen Q., Hou J., Wu Z., Zhao J., Ma D. miR-145 regulates the sensitivity of esophageal squamous cell carcinoma cells to 5-FU via targeting REV3L. Pathol. Res. Pract. 2019;215:152427. doi: 10.1016/j.prp.2019.04.019. [DOI] [PubMed] [Google Scholar]
  • 358.Liang H., Liu M., Yan X., Zhou Y., Wang W., Wang X., Fu Z., Wang N., Zhang S., Wang Y. miR-193a-3p functions as a tumor suppressor in lung cancer by down-regulating ERBB4. J. Biol. Chem. 2015;290:926–940. doi: 10.1074/jbc.M114.621409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 359.Kwon J.E., Kim B.Y., Kwak S.Y., Bae I.H., Han Y.H. Ionizing radiation-inducible microRNA miR-193a-3p induces apoptosis by directly targeting Mcl-1. Apoptosis. 2013;18:896–909. doi: 10.1007/s10495-013-0841-7. [DOI] [PubMed] [Google Scholar]
  • 360.Gao X.N., Lin J., Li Y.H., Gao L., Wang X.R., Wang W., Kang H.Y., Yan G.T., Wang L.L., Yu L. MicroRNA-193a represses c-kit expression and functions as a methylation-silenced tumor suppressor in acute myeloid leukemia. Oncogene. 2011;30:3416–3428. doi: 10.1038/onc.2011.62. [DOI] [PubMed] [Google Scholar]
  • 361.Meng F., Qian L., Lv L., Ding B., Zhou G., Cheng X., Niu S., Liang Y. miR-193a-3p regulation of chemoradiation resistance in oesophageal cancer cells via the PSEN1 gene. Gene. 2016;579:139–145. doi: 10.1016/j.gene.2015.12.060. [DOI] [PubMed] [Google Scholar]
  • 362.Korhan P., Erdal E., Atabey N. miR-181a-5p is downregulated in hepatocellular carcinoma and suppresses motility, invasion and branching-morphogenesis by directly targeting c-Met. Biochem. Biophys. Res. Commun. 2014;450:1304–1312. doi: 10.1016/j.bbrc.2014.06.142. [DOI] [PubMed] [Google Scholar]
  • 363.Liu Z., Sun F., Hong Y., Liu Y., Fen M., Yin K., Ge X., Wang F., Chen X., Guan W. MEG2 is regulated by miR-181a-5p and functions as a tumour suppressor gene to suppress the proliferation and migration of gastric cancer cells. Mol. Cancer. 2017;16:133. doi: 10.1186/s12943-017-0695-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 364.Hummel R., Sie C., Watson D.I., Wang T., Ansar A., Michael M.Z., Van der Hoek M., Haier J., Hussey D.J. MicroRNA signatures in chemotherapy resistant esophageal cancer cell lines. World J. Gastroenterol. 2014;20:14904–14912. doi: 10.3748/wjg.v20.i40.14904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365.Yang S., Wang P., Wang S., Cong A., Zhang Q., Shen W., Li X., Zhang W., Han G. miRNA-181a-5p enhances the sensitivity of cells to cisplatin in esophageal adenocarcinoma by targeting CBLB. Cancer Manag. Res. 2020;12:4981–4990. doi: 10.2147/CMAR.S251264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Singh R., Pochampally R., Watabe K., Lu Z., Mo Y.Y. Exosome-mediated transfer of miR-10b promotes cell invasion in breast cancer. Mol. Cancer. 2014;13:256. doi: 10.1186/1476-4598-13-256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 367.Liu L.Z., Zhou X.D., Qian G., Shi X., Fang J., Jiang B.H. AKT1 amplification regulates cisplatin resistance in human lung cancer cells through the mammalian target of rapamycin/p70S6K1 pathway. Cancer Res. 2007;67:6325–6332. doi: 10.1158/0008-5472.CAN-06-4261. [DOI] [PubMed] [Google Scholar]
  • 368.Huang J., Sun C., Wang S., He Q., Li D. MicroRNA miR-10b inhibition reduces cell proliferation and promotes apoptosis in non-small cell lung cancer (NSCLC) cells. Mol. Biosyst. 2015;11:2051–2059. doi: 10.1039/c4mb00752b. [DOI] [PubMed] [Google Scholar]
  • 369.Guan L., Ji D., Liang N., Li S., Sun B. Up-regulation of miR-10b-3p promotes the progression of hepatocellular carcinoma cells via targeting CMTM5. J. Cell. Mol. Med. 2018;22:3434–3441. doi: 10.1111/jcmm.13620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 370.Nishida N., Yamashita S., Mimori K., Sudo T., Tanaka F., Shibata K., Yamamoto H., Ishii H., Doki Y., Mori M. MicroRNA-10b is a prognostic indicator in colorectal cancer and confers resistance to the chemotherapeutic agent 5-fluorouracil in colorectal cancer cells. Ann. Surg. Oncol. 2012;19:3065–3071. doi: 10.1245/s10434-012-2246-1. [DOI] [PubMed] [Google Scholar]
  • 371.Zhang P., Hong H., Sun X., Jiang H., Ma S., Zhao S., Zhang M., Wang Z., Jiang C., Liu H. MicroRNA-10b regulates epithelial-mesenchymal transition by modulating KLF4/Notch1/E-cadherin in cisplatin-resistant nasopharyngeal carcinoma cells. Am. J. Cancer Res. 2016;6:141–156. [PMC free article] [PubMed] [Google Scholar]
  • 372.Ahmad A., Ginnebaugh K.R., Yin S., Bollig-Fischer A., Reddy K.B., Sarkar F.H. Functional role of miR-10b in tamoxifen resistance of ER-positive breast cancer cells through down-regulation of HDAC4. BMC Cancer. 2015;15:540. doi: 10.1186/s12885-015-1561-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 373.Tian Y., Luo A., Cai Y., Su Q., Ding F., Chen H., Liu Z. MicroRNA-10b promotes migration and invasion through KLF4 in human esophageal cancer cell lines. J. Biol. Chem. 2010;285:7986–7994. doi: 10.1074/jbc.M109.062877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 374.Wu K., Hu Y., Yan K., Qi Y., Zhang C., Zhu D., Liu D., Zhao S. MicroRNA-10b confers cisplatin resistance by activating AKT/mTOR/P70S6K signaling via targeting PPARγ in esophageal cancer. J. Cell. Physiol. 2020;235:1247–1258. doi: 10.1002/jcp.29040. [DOI] [PubMed] [Google Scholar]
  • 375.Guo H., Li Q., Li W., Zheng T., Zhao S., Liu Z. miR-96 downregulates RECK to promote growth and motility of non-small cell lung cancer cells. Mol. Cell. Biochem. 2014;390:155–160. doi: 10.1007/s11010-014-1966-x. [DOI] [PubMed] [Google Scholar]
  • 376.Lin H., Dai T., Xiong H., Zhao X., Chen X., Yu C., Li J., Wang X., Song L. Unregulated miR-96 induces cell proliferation in human breast cancer by downregulating transcriptional factor FOXO3a. PLoS ONE. 2010;5:e15797. doi: 10.1371/journal.pone.0015797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 377.Fendler A., Jung M., Stephan C., Erbersdobler A., Jung K., Yousef G.M. The antiapoptotic function of miR-96 in prostate cancer by inhibition of FOXO1. PLoS ONE. 2013;8:e80807. doi: 10.1371/journal.pone.0080807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 378.Guo Y., Liu H., Zhang H., Shang C., Song Y. miR-96 regulates FOXO1-mediated cell apoptosis in bladder cancer. Oncol. Lett. 2012;4:561–565. doi: 10.3892/ol.2012.775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 379.Zhang J., Kong X., Li J., Luo Q., Li X., Shen L., Chen L., Fang L. miR-96 promotes tumor proliferation and invasion by targeting RECK in breast cancer. Oncol. Rep. 2014;31:1357–1363. doi: 10.3892/or.2013.2934. [DOI] [PubMed] [Google Scholar]
  • 380.Eichelmann A.K., Matuszcak C., Lindner K., Haier J., Hussey D.J., Hummel R. Complex role of miR-130a-3p and miR-148a-3p balance on drug resistance and tumor biology in esophageal squamous cell carcinoma. Sci. Rep. 2018;8:17553. doi: 10.1038/s41598-018-35799-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 381.Yang Y.C., Liu G.J., Yuan D.F., Li C.Q., Xue M., Chen L.J. Influence of exosome-derived miR-21on chemotherapy resistance of esophageal cancer. Eur. Rev. Med. Pharmacol. Sci. 2019;23:1513–1519. doi: 10.26355/eurrev_201902_17109. [DOI] [PubMed] [Google Scholar]
  • 382.Hu M., Zhang Q., Tian X.H., Wang J.L., Niu Y.X., Li G. lncRNA CCAT1 is a biomarker for the proliferation and drug resistance of esophageal cancer via the miR-143/PLK1/BUBR1 axis. Mol. Carcinog. 2019;58:2207–2217. doi: 10.1002/mc.23109. [DOI] [PubMed] [Google Scholar]
  • 383.Huang C.M., Huang C.S., Hsu T.N., Huang M.S., Fong I.H., Lee W.H., Liu S.C. Disruption of cancer metabolic SREBP1/miR-142-5p suppresses epithelial-mesenchymal transition and stemness in esophageal carcinoma. Cells. 2019;9:7. doi: 10.3390/cells9010007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 384.Jin Y.Y., Chen Q.J., Xu K., Ren H.T., Bao X., Ma Y.N., Wei Y., Ma H.B. Involvement of microRNA-141-3p in 5-fluorouracil and oxaliplatin chemo-resistance in esophageal cancer cells via regulation of PTEN. Mol. Cell. Biochem. 2016;422:161–170. doi: 10.1007/s11010-016-2816-9. [DOI] [PubMed] [Google Scholar]
  • 385.Chang Z.W., Jia Y.X., Zhang W.J., Song L.J., Gao M., Li M.J., Zhao R.H., Li J., Zhong Y.L., Sun Q.Z., Qin Y.R. lncRNA-TUSC7/miR-224 affected chemotherapy resistance of esophageal squamous cell carcinoma by competitively regulating DESC1. J. Exp. Clin. Cancer Res. 2018;37:56. doi: 10.1186/s13046-018-0724-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 386.Tanaka K., Miyata H., Sugimura K., Fukuda S., Kanemura T., Yamashita K., Miyazaki Y., Takahashi T., Kurokawa Y., Yamasaki M. miR-27 is associated with chemoresistance in esophageal cancer through transformation of normal fibroblasts to cancer-associated fibroblasts. Carcinogenesis. 2015;36:894–903. doi: 10.1093/carcin/bgv067. [DOI] [PubMed] [Google Scholar]
  • 387.Ma Y., Wang B., Guo Y., Zhang Y., Huang S., Bao X., Bai M. Inhibition of miR-196a affects esophageal cancer cell growth in vitro. Biomed. Pharmacother. 2016;84:22–27. doi: 10.1016/j.biopha.2016.09.013. [DOI] [PubMed] [Google Scholar]
  • 388.Hummel R., Watson D.I., Smith C., Kist J., Michael M.Z., Haier J., Hussey D.J. Mir-148a improves response to chemotherapy in sensitive and resistant oesophageal adenocarcinoma and squamous cell carcinoma cells. J. Gastrointest. Surg. 2011;15:429–438. doi: 10.1007/s11605-011-1418-9. [DOI] [PubMed] [Google Scholar]
  • 389.Sugimura K., Miyata H., Tanaka K., Hamano R., Takahashi T., Kurokawa Y., Yamasaki M., Nakajima K., Takiguchi S., Mori M. Let-7 expression is a significant determinant of response to chemotherapy through the regulation of IL-6/STAT3 pathway in esophageal squamous cell carcinoma. Clin. Cancer Res. 2012;18:5144–5153. doi: 10.1158/1078-0432.CCR-12-0701. [DOI] [PubMed] [Google Scholar]
  • 390.Akdemir B., Nakajima Y., Inazawa J., Inoue J. miR-432 induces NRF2 stabilization by directly targeting KEAP1. Mol. Cancer Res. 2017;15:1570–1578. doi: 10.1158/1541-7786.MCR-17-0232. [DOI] [PubMed] [Google Scholar]
  • 391.Wang Y., Feng J., Zang W., Du Y., Chen X., Sun Q., Dong Z., Zhao G. miR-499 enhances the cisplatin sensitivity of esophageal carcinoma cell lines by targeting DNA polymerase β. Cell. Physiol. Biochem. 2015;36:1587–1596. doi: 10.1159/000430321. [DOI] [PubMed] [Google Scholar]
  • 392.Hamano R., Miyata H., Yamasaki M., Kurokawa Y., Hara J., Moon J.H., Nakajima K., Takiguchi S., Fujiwara Y., Mori M. Overexpression of miR-200c induces chemoresistance in esophageal cancers mediated through activation of the Akt signaling pathway. Clin. Cancer Res. 2011;17:3029–3038. doi: 10.1158/1078-0432.CCR-10-2532. [DOI] [PubMed] [Google Scholar]
  • 393.Komatsu S., Ichikawa D., Kawaguchi T., Miyamae M., Okajima W., Ohashi T., Imamura T., Kiuchi J., Konishi H., Shiozaki A. Circulating miR-21 as an independent predictive biomarker for chemoresistance in esophageal squamous cell carcinoma. Am. J. Cancer Res. 2016;6:1511–1523. [PMC free article] [PubMed] [Google Scholar]
  • 394.Ma J., Hong L., Xu G., Hao J., Wang R., Guo H., Liu J., Zhang Y., Nie Y., Fan D. miR-483-3p plays an oncogenic role in esophageal squamous cell carcinoma by targeting tumor suppressor EI24. Cell Biol. Int. 2016;40:448–455. doi: 10.1002/cbin.10585. [DOI] [PubMed] [Google Scholar]
  • 395.Imanaka Y., Tsuchiya S., Sato F., Shimada Y., Shimizu K., Tsujimoto G. MicroRNA-141 confers resistance to cisplatin-induced apoptosis by targeting YAP1 in human esophageal squamous cell carcinoma. J. Hum. Genet. 2011;56:270–276. doi: 10.1038/jhg.2011.1. [DOI] [PubMed] [Google Scholar]
  • 396.Li B., Hong P., Zheng C.C., Dai W., Chen W.Y., Yang Q.S., Han L., Tsao S.W., Chan K.T., Lee N.P.Y. Identification of miR-29c and its target FBXO31 as a Key regulatory mechanism in esophageal cancer chemoresistance: Functional validation and clinical significance. Theranostics. 2019;9:1599–1613. doi: 10.7150/thno.30372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 397.Chen Q.N., Wei C.C., Wang Z.X., Sun M. Long non-coding RNAs in anti-cancer drug resistance. Oncotarget. 2017;8:1925–1936. doi: 10.18632/oncotarget.12461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 398.Wang Z.Q., Cai Q., Hu L., He C.Y., Li J.F., Quan Z.W., Liu B.Y., Li C., Zhu Z.G. Long noncoding RNA UCA1 induced by SP1 promotes cell proliferation via recruiting EZH2 and activating AKT pathway in gastric cancer. Cell Death Dis. 2017;8:e2839. doi: 10.1038/cddis.2017.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 399.Wang Z.Q., He C.Y., Hu L., Shi H.P., Li J.F., Gu Q.L., Su L.P., Liu B.Y., Li C., Zhu Z. Long noncoding RNA UCA1 promotes tumour metastasis by inducing GRK2 degradation in gastric cancer. Cancer Lett. 2017;408:10–21. doi: 10.1016/j.canlet.2017.08.013. [DOI] [PubMed] [Google Scholar]
  • 400.He X., Wang J., Chen J., Han L., Lu X., Miao D., Yin D., Geng Q., Zhang E. lncRNA UCA1 predicts a poor prognosis and regulates cell proliferation and migration by repressing p21 and SPRY1 expression in GC. Mol. Ther. Nucleic Acids. 2019;18:605–616. doi: 10.1016/j.omtn.2019.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 401.Xie J., Wang S., Li G., Zhao X., Jiang F., Liu J., Tan W. circEPSTI1 regulates ovarian cancer progression via decoying miR-942. J. Cell. Mol. Med. 2019;23:3597–3602. doi: 10.1111/jcmm.14260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 402.Li C., Fan K., Qu Y., Zhai W., Huang A., Sun X., Xing S. Deregulation of UCA1 expression may be involved in the development of chemoresistance to cisplatin in the treatment of non-small-cell lung cancer via regulating the signaling pathway of microRNA-495/NRF2. J. Cell. Physiol. 2020;235:3721–3730. doi: 10.1002/jcp.29266. [DOI] [PubMed] [Google Scholar]
  • 403.Fang Z., Zhao J., Xie W., Sun Q., Wang H., Qiao B. lncRNA UCA1 promotes proliferation and cisplatin resistance of oral squamous cell carcinoma by sunppressing miR-184 expression. Cancer Med. 2017;6:2897–2908. doi: 10.1002/cam4.1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 404.Pan J., Li X., Wu W., Xue M., Hou H., Zhai W., Chen W. Long non-coding RNA UCA1 promotes cisplatin/gemcitabine resistance through CREB modulating miR-196a-5p in bladder cancer cells. Cancer Lett. 2016;382:64–76. doi: 10.1016/j.canlet.2016.08.015. [DOI] [PubMed] [Google Scholar]
  • 405.Fang Q., Chen X., Zhi X. Long non-coding RNA (lncRNA) urothelial carcinoma associated 1 (UCA1) increases multi-drug resistance of gastric cancer via downregulating miR-27b. Med. Sci. Monit. 2016;22:3506–3513. doi: 10.12659/MSM.900688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 406.Qin L., Jia Z., Xie D., Liu Z. Retraction. J. Cell. Biochem. 2021;122:722. [Google Scholar]
  • 407.Elmore S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007;35:495–516. doi: 10.1080/01926230701320337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 408.Ye Y., Yang S., Han Y., Sun J., Xv L., Wu L., Ming L. HOXD-AS1 confers cisplatin resistance in gastric cancer through epigenetically silencing PDCD4 via recruiting EZH2. Open Biol. 2019;9:190068. doi: 10.1098/rsob.190068. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 409.Sun S., Zhao S., Yang Q., Wang W., Cai E., Wen Y., Yu L., Wang Z., Cai J. Enhancer of zeste homolog 2 promotes cisplatin resistance by reducing cellular platinum accumulation. Cancer Sci. 2018;109:1853–1864. doi: 10.1111/cas.13599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 410.Liu X., Lu X., Zhen F., Jin S., Yu T., Zhu Q., Wang W., Xu K., Yao J., Guo R. LINC00665 induces acquired resistance to gefitinib through recruiting EZH2 and activating PI3K/AKT pathway in NSCLC. Mol. Ther. Nucleic Acids. 2019;16:155–161. doi: 10.1016/j.omtn.2019.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 411.Zhang C., Liu J., Zhang Y., Luo C., Zhu T., Zhang R., Yao R. LINC01210 accelerates proliferation, invasion and migration in ovarian cancer through epigenetically downregulating KLF4. Biomed. Pharmacother. 2019;119:109431. doi: 10.1016/j.biopha.2019.109431. [DOI] [PubMed] [Google Scholar]
  • 412.Xi G., Hayes E., Lewis R., Ichi S., Mania-Farnell B., Shim K., Takao T., Allender E., Mayanil C.S., Tomita T. Retraction. Oncogene. 2016;35:5576. doi: 10.1038/onc.2016.64. [DOI] [PubMed] [Google Scholar]
  • 413.Zhao X., Cheng Z., Wang J. Long noncoding RNA FEZF1-AS1 promotes proliferation and inhibits apoptosis in ovarian cancer by activation of JAK-STAT3 pathway. Med. Sci. Monit. 2018;24:8088–8095. doi: 10.12659/MSM.911194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 414.Dai Q., Zhang T., Pan J., Li C. lncRNA UCA1 promotes cisplatin resistance in gastric cancer via recruiting EZH2 and activating PI3K/AKT pathway. J. Cancer. 2020;11:3882–3892. doi: 10.7150/jca.43446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 415.Yang L., Cui J., Wang Y., Tan J. FAM83H-AS1 is upregulated and predicts poor prognosis in colon cancer. Biomed. Pharmacother. 2019;118:109342. doi: 10.1016/j.biopha.2019.109342. [DOI] [PubMed] [Google Scholar]
  • 416.Barr J.A., Hayes K.E., Brownmiller T., Harold A.D., Jagannathan R., Lockman P.R., Khan S., Martinez I. Long non-coding RNA FAM83H-AS1 is regulated by human papillomavirus 16 E6 independently of p53 in cervical cancer cells. Sci. Rep. 2019;9:3662. doi: 10.1038/s41598-019-40094-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 417.Yang F., Lv S.-X., Lv L., Liu Y.-H., Dong S.-Y., Yao Z.-H., Dai X.X., Zhang X.-H., Wang O.-C. Identification of lncRNA FAM83H-AS1 as a novel prognostic marker in luminal subtype breast cancer. OncoTargets Ther. 2016;9:7039–7045. doi: 10.2147/OTT.S110055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 418.Da J., Liu P., Wang R., Bu L. Upregulation of the long non-coding RNA FAM83H-AS1 in gastric cancer and its clinical significance. Pathol. Res. Pract. 2019;215:152616. doi: 10.1016/j.prp.2019.152616. [DOI] [PubMed] [Google Scholar]
  • 419.Wang B., Guan G., Zhao D. Silence of FAM83H-AS1 promotes chemosensitivity of gastric cancer through Wnt/β-catenin signaling pathway. Biomed. Pharmacother. 2020;125:109961. doi: 10.1016/j.biopha.2020.109961. [DOI] [PubMed] [Google Scholar]
  • 420.Yoon J.H., Abdelmohsen K., Gorospe M. Functional interactions among microRNAs and long noncoding RNAs. Semin. Cell Dev. Biol. 2014;34:9–14. doi: 10.1016/j.semcdb.2014.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 421.Cheng C., Qin Y., Zhi Q., Wang J., Qin C. Knockdown of long non-coding RNA HOTAIR inhibits cisplatin resistance of gastric cancer cells through inhibiting the PI3K/Akt and Wnt/β-catenin signaling pathways by up-regulating miR-34a. Int. J. Biol. Macromol. 2018;107(Pt B):2620–2629. doi: 10.1016/j.ijbiomac.2017.10.154. [DOI] [PubMed] [Google Scholar]
  • 422.Zhou Z., Lin Z., He Y., Pang X., Wang Y., Ponnusamy M., Ao X., Shan P., Tariq M.A., Li P., Wang J. The long noncoding RNA D63785 regulates chemotherapy sensitivity in human gastric cancer by targeting miR-422a. Mol. Ther. Nucleic Acids. 2018;12:405–419. doi: 10.1016/j.omtn.2018.05.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 423.Zeng L., Liao Q., Zou Z., Wen Y., Wang J., Liu C., He Q., Weng N., Zeng J., Tang H. Long non-coding RNA XLOC_006753 promotes the development of multidrug resistance in gastric cancer cells through the PI3K/AKT/mTOR signaling pathway. Cell. Physiol. Biochem. 2018;51:1221–1236. doi: 10.1159/000495499. [DOI] [PubMed] [Google Scholar]
  • 424.Zhang X.W., Bu P., Liu L., Zhang X.Z., Li J. Overexpression of long non-coding RNA PVT1 in gastric cancer cells promotes the development of multidrug resistance. Biochem. Biophys. Res. Commun. 2015;462:227–232. doi: 10.1016/j.bbrc.2015.04.121. [DOI] [PubMed] [Google Scholar]
  • 425.Sun B., Han Y., Cai H., Huang H., Xuan Y. Long non-coding RNA SNHG3, induced by IL-6/STAT3 transactivation, promotes stem cell-like properties of gastric cancer cells by regulating the miR-3619-5p/ARL2 axis. Cell. Oncol. (Dordr.) 2020;44:179–192. doi: 10.1007/s13402-020-00560-2. [DOI] [PubMed] [Google Scholar]
  • 426.Shi W., Zhang C., Ning Z., Hua Y., Li Y., Chen L., Liu L., Chen Z., Meng Z. Long non-coding RNA LINC00346 promotes pancreatic cancer growth and gemcitabine resistance by sponging miR-188-3p to derepress BRD4 expression. J. Exp. Clin. Cancer Res. 2019;38:60. doi: 10.1186/s13046-019-1055-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 427.Ye T., Ding W., Wang N., Huang H., Pan Y., Wei A. Long noncoding RNA linc00346 promotes the malignant phenotypes of bladder cancer. Biochem. Biophys. Res. Commun. 2017;491:79–84. doi: 10.1016/j.bbrc.2017.07.045. [DOI] [PubMed] [Google Scholar]
  • 428.Zhang B., Li C., Sun Z. Long non-coding RNA LINC00346, LINC00578, LINC00673, LINC00671, LINC00261, and SNHG9 are novel prognostic markers for pancreatic cancer. Am. J. Transl. Res. 2018;10:2648–2658. [PMC free article] [PubMed] [Google Scholar]
  • 429.Hua Y.Q., Zhu Y.D., Xie G.Q., Zhang K., Sheng J., Zhu Z.F., Ning Z.Y., Chen H., Chen Z., Meng Z.Q., Liu L.M. Long non-coding SBF2-AS1 acting as a competing endogenous RNA to sponge microRNA-142-3p to participate in gemcitabine resistance in pancreatic cancer via upregulating TWF1. Aging (Albany NY) 2019;11:8860–8878. doi: 10.18632/aging.102307. [DOI] [PubMed] [Google Scholar]
  • 430.Coccia E.M., Cicala C., Charlesworth A., Ciccarelli C., Rossi G.B., Philipson L., Sorrentino V. Regulation and expression of a growth arrest-specific gene (gas5) during growth, differentiation, and development. Mol. Cell. Biol. 1992;12:3514–3521. doi: 10.1128/mcb.12.8.3514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 431.DiStefano J.K. The emerging role of long noncoding RNAs in human disease. Methods Mol. Biol. 2018;1706:91–110. doi: 10.1007/978-1-4939-7471-9_6. [DOI] [PubMed] [Google Scholar]
  • 432.Prensner J.R., Chinnaiyan A.M. The emergence of lncRNAs in cancer biology. Cancer Discov. 2011;1:391–407. doi: 10.1158/2159-8290.CD-11-0209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 433.Gao Q., Xie H., Zhan H., Li J., Liu Y., Huang W. Prognostic values of long noncoding RNA GAS5 in various carcinomas: An updated systematic review and meta-analysis. Front. Physiol. 2017;8:814. doi: 10.3389/fphys.2017.00814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 434.Pickard M.R., Williams G.T. Molecular and cellular mechanisms of action of tumour suppressor GAS5 lncRNA. Genes (Basel) 2015;6:484–499. doi: 10.3390/genes6030484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 435.Wang C., Ke S., Li M., Lin C., Liu X., Pan Q. Downregulation of lncRNA GAS5 promotes liver cancer proliferation and drug resistance by decreasing PTEN expression. Mol. Genet. Genomics. 2020;295:251–260. doi: 10.1007/s00438-019-01620-5. [DOI] [PubMed] [Google Scholar]
  • 436.Wang Y., Liu Z., Yao B., Li Q., Wang L., Wang C., Dou C., Xu M., Liu Q., Tu K. Long non-coding RNA CASC2 suppresses epithelial-mesenchymal transition of hepatocellular carcinoma cells through CASC2/miR-367/FBXW7 axis. Mol. Cancer. 2017;16:123. doi: 10.1186/s12943-017-0702-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 437.Baldinu P., Cossu A., Manca A., Satta M.P., Sini M.C., Palomba G., Dessole S., Cherchi P., Mara L., Tanda F., Palmieri G. CASC2a gene is down-regulated in endometrial cancer. Anticancer Res. 2007;27(1A):235–243. [PubMed] [Google Scholar]
  • 438.Gao W., Lin S., Cheng C., Zhu A., Hu Y., Shi Z., Zhang X., Hong Z. Long non-coding RNA CASC2 regulates Sprouty2 via functioning as a competing endogenous RNA for miR-183 to modulate the sensitivity of prostate cancer cells to docetaxel. Arch. Biochem. Biophys. 2019;665:69–78. doi: 10.1016/j.abb.2018.01.013. [DOI] [PubMed] [Google Scholar]
  • 439.Ambros V. MicroRNAs: Tiny regulators with great potential. Cell. 2001;107:823–826. doi: 10.1016/s0092-8674(01)00616-x. [DOI] [PubMed] [Google Scholar]
  • 440.Wang K., Long B., Zhou L.-Y., Liu F., Zhou Q.-Y., Liu C.-Y., Fan Y.-Y., Li P.-F. CARL lncRNA inhibits anoxia-induced mitochondrial fission and apoptosis in cardiomyocytes by impairing miR-539-dependent PHB2 downregulation. Nat. Commun. 2014;5:3596. doi: 10.1038/ncomms4596. [DOI] [PubMed] [Google Scholar]
  • 441.Feng Y., Zou W., Hu C., Li G., Zhou S., He Y., Ma F., Deng C., Sun L. Modulation of CASC2/miR-21/PTEN pathway sensitizes cervical cancer to cisplatin. Arch. Biochem. Biophys. 2017;623–624:20–30. doi: 10.1016/j.abb.2017.05.001. [DOI] [PubMed] [Google Scholar]
  • 442.Jiang C., Shen F., Du J., Fang X., Li X., Su J., Wang X., Huang X., Liu Z. Upregulation of CASC2 sensitized glioma to temozolomide cytotoxicity through autophagy inhibition by sponging miR-193a-5p and regulating mTOR expression. Biomed. Pharmacother. 2018;97:844–850. doi: 10.1016/j.biopha.2017.10.146. [DOI] [PubMed] [Google Scholar]
  • 443.Wang L., Zhang X., Sheng L., Qiu C., Luo R. LINC00473 promotes the Taxol resistance via miR-15a in colorectal cancer. Biosci. Rep. 2018;38 doi: 10.1042/BSR20180790. BSR20180790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 444.Yu L., Gui S., Liu Y., Qiu X., Zhang G., Zhang X., Pan J., Fan J., Qi S., Qiu B. Exosomes derived from microRNA-199a-overexpressing mesenchymal stem cells inhibit glioma progression by down-regulating AGAP2. Aging (Albany NY) 2019;11:5300–5318. doi: 10.18632/aging.102092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 445.Liang Q., Ding J., Xu R., Xu Z., Zheng S. The novel human endogenous retrovirus-related gene, psiTPTE22-HERV, is silenced by DNA methylation in cancers. Int. J. Cancer. 2010;127:1833–1843. doi: 10.1002/ijc.25213. [DOI] [PubMed] [Google Scholar]
  • 446.Choi S.J., Jung S.W., Huh S., Chung Y.-S., Cho H., Kang H. Alteration of DNA methylation in gastric cancer with chemotherapy. J. Microbiol. Biotechnol. 2017;27:1367–1378. doi: 10.4014/jmb.1704.04035. [DOI] [PubMed] [Google Scholar]
  • 447.Vera O., Jimenez J., Pernia O., Rodriguez-Antolin C., Rodriguez C., Sanchez Cabo F., Soto J., Rosas R., Lopez-Magallon S., Esteban Rodriguez I. DNA methylation of miR-7 is a mechanism involved in platinum response through MAFG overexpression in cancer cells. Theranostics. 2017;7:4118–4134. doi: 10.7150/thno.20112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 448.van Jaarsveld M.T., Helleman J., Boersma A.W., van Kuijk P.F., van Ijcken W.F., Despierre E., Vergote I., Mathijssen R.H., Berns E.M., Verweij J. miR-141 regulates KEAP1 and modulates cisplatin sensitivity in ovarian cancer cells. Oncogene. 2013;32:4284–4293. doi: 10.1038/onc.2012.433. [DOI] [PubMed] [Google Scholar]
  • 449.Ding H., Liu J., Zou R., Cheng P., Su Y. Long non-coding RNA TPTEP1 inhibits hepatocellular carcinoma progression by suppressing STAT3 phosphorylation. J. Exp. Clin. Cancer Res. 2019;38:189. doi: 10.1186/s13046-019-1193-0. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 450.Huang H., Chen J., Ding C.M., Jin X., Jia Z.M., Peng J. lncRNA NR2F1-AS1 regulates hepatocellular carcinoma oxaliplatin resistance by targeting ABCC1 via miR-363. J. Cell. Mol. Med. 2018;22:3238–3245. doi: 10.1111/jcmm.13605. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 451.Xu Y., Shen D., Liu J., Xu X., Tu J., Qin L., Jiang L., Qian H., Guo F. Long non-coding RNAs as targets for immunosuppressive drug teriflunomide in anti-cancer potential for hepatocellular carcinoma. J. Mol. Histol. 2020;51:659–673. doi: 10.1007/s10735-020-09912-6. [DOI] [PubMed] [Google Scholar]
  • 452.Sun Q.M., Hu B., Fu P.Y., Tang W.G., Zhang X., Zhan H., Sun C., He Y.F., Song K., Xiao Y.S. Long non-coding RNA 00607 as a tumor suppressor by modulating NF-κB p65/p53 signaling axis in hepatocellular carcinoma. Carcinogenesis. 2018;39:1438–1446. doi: 10.1093/carcin/bgy113. [DOI] [PubMed] [Google Scholar]
  • 453.Hu H., Yang L., Li L., Zeng C. Long non-coding RNA KCNQ1OT1 modulates oxaliplatin resistance in hepatocellular carcinoma through miR-7-5p/ ABCC1 axis. Biochem. Biophys. Res. Commun. 2018;503:2400–2406. doi: 10.1016/j.bbrc.2018.06.168. [DOI] [PubMed] [Google Scholar]
  • 454.Jin W., Chen L., Cai X., Zhang Y., Zhang J., Ma D., Cai X., Fu T., Yu Z., Yu F., Chen G. Long non-coding RNA TUC338 is functionally involved in sorafenib-sensitized hepatocarcinoma cells by targeting RASAL1. Oncol. Rep. 2017;37:273–280. doi: 10.3892/or.2016.5248. [DOI] [PubMed] [Google Scholar]
  • 455.Sui C., Dong Z., Yang C., Zhang M., Dai B., Geng L., Lu J., Yang J., Xu M. lncRNA FOXD2-AS1 as a competitive endogenous RNA against miR-150-5p reverses resistance to sorafenib in hepatocellular carcinoma. J. Cell. Mol. Med. 2019;23:6024–6033. doi: 10.1111/jcmm.14465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 456.Liu B., Zhang J., Yang D. miR-96-5p promotes the proliferation and migration of ovarian cancer cells by suppressing Caveolae1. J. Ovarian Res. 2019;12:57. doi: 10.1186/s13048-019-0533-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 457.Hu T., Lu Y.-R. BCYRN1, a c-MYC-activated long non-coding RNA, regulates cell metastasis of non-small-cell lung cancer. Cancer Cell Int. 2015;15:36. doi: 10.1186/s12935-015-0183-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 458.Booy E.P., McRae E.K., Koul A., Lin F., McKenna S.A. The long non-coding RNA BC200 (BCYRN1) is critical for cancer cell survival and proliferation. Mol. Cancer. 2017;16:109. doi: 10.1186/s12943-017-0679-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 459.Ding S., Jin Y., Hao Q., Kang Y., Ma R. lncRNA BCYRN1/miR-490-3p/POU3F2, served as a ceRNA network, is connected with worse survival rate of hepatocellular carcinoma patients and promotes tumor cell growth and metastasis. Cancer Cell Int. 2020;20:6. doi: 10.1186/s12935-019-1081-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 460.Zhai H., Li Y. BCYRN1 is correlated with progression and prognosis in gastric cancer. Biosci. Rep. 2019;39 doi: 10.1042/BSR20190505. BSR20190505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 461.Wang Y.-C., Chuang Y.-H., Shao Q., Chen J.-F., Chen S.-Y. Brain cytoplasmic RNA 1 suppresses smooth muscle differentiation and vascular development in mice. J. Biol. Chem. 2018;293:5668–5678. doi: 10.1074/jbc.RA117.001578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 462.Zhang X.-Y., Tang X.-Y., Ma L.-J., Guo Y.-L., Li X.-S., Zhao L.-M., Tian C.-J., Cheng D.-J., Chen Z.-C., Zhang L.-X. Schisandrin B down-regulated lncRNA BCYRN1 expression of airway smooth muscle cells by improving miR-150 expression to inhibit the proliferation and migration of ASMC in asthmatic rats. Cell Prolif. 2017;50:e12382. doi: 10.1111/cpr.12382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 463.Yang L., Zhang Y., Bao J., Feng J.F. Long non-coding RNA BCYRN1 exerts an oncogenic role in colorectal cancer by regulating the miR-204-3p/KRAS axis. Cancer Cell Int. 2020;20:453. doi: 10.1186/s12935-020-01543-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 464.Young T.L., Matsuda T., Cepko C.L. The noncoding RNA taurine upregulated gene 1 is required for differentiation of the murine retina. Curr. Biol. 2005;15:501–512. doi: 10.1016/j.cub.2005.02.027. [DOI] [PubMed] [Google Scholar]
  • 465.Tan J., Qiu K., Li M., Liang Y. Double-negative feedback loop between long non-coding RNA TUG1 and miR-145 promotes epithelial to mesenchymal transition and radioresistance in human bladder cancer cells. FEBS Lett. 2015;589(20 Pt B):3175–3181. doi: 10.1016/j.febslet.2015.08.020. [DOI] [PubMed] [Google Scholar]
  • 466.Zhang E., He X., Yin D., Han L., Qiu M., Xu T., Xia R., Xu L., Yin R., De W. Increased expression of long noncoding RNA TUG1 predicts a poor prognosis of gastric cancer and regulates cell proliferation by epigenetically silencing of p57. Cell Death Dis. 2016;7:e2109. doi: 10.1038/cddis.2015.356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 467.Zhu J., Shi H., Liu H., Wang X., Li F. Long non-coding RNA TUG1 promotes cervical cancer progression by regulating the miR-138-5p-SIRT1 axis. Oncotarget. 2017;8:65253–65264. doi: 10.18632/oncotarget.18224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 468.Zhang E.B., Yin D.D., Sun M., Kong R., Liu X.H., You L.H., Han L., Xia R., Wang K.M., Yang J.S. p53-regulated long non-coding RNA TUG1 affects cell proliferation in human non-small cell lung cancer, partly through epigenetically regulating HOXB7 expression. Cell Death Dis. 2014;5:e1243. doi: 10.1038/cddis.2014.201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 469.Li C., Gao Y., Li Y., Ding D. TUG1 mediates methotrexate resistance in colorectal cancer via miR-186/CPEB2 axis. Biochem. Biophys. Res. Commun. 2017;491:552–557. doi: 10.1016/j.bbrc.2017.03.042. [DOI] [PubMed] [Google Scholar]
  • 470.Wang M., Hu H., Wang Y., Huang Q., Huang R., Chen Y., Ma T., Qiao T., Zhang Q., Wu H. Long non-coding RNA TUG1 mediates 5-fluorouracil resistance by acting as a ceRNA of miR-197-3p in colorectal cancer. J. Cancer. 2019;10:4603–4613. doi: 10.7150/jca.32065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 471.Wu H., Zou Q., He H., Liang Y., Lei M., Zhou Q., Fan D., Shen L. Long non-coding RNA PCAT6 targets miR-204 to modulate the chemoresistance of colorectal cancer cells to 5-fluorouracil-based treatment through HMGA2 signaling. Cancer Med. 2019;8:2484–2495. doi: 10.1002/cam4.1809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 472.Doherty G.J., Åhlund M.K., Howes M.T., Morén B., Parton R.G., McMahon H.T., Lundmark R. The endocytic protein GRAF1 is directed to cell-matrix adhesion sites and regulates cell spreading. Mol. Biol. Cell. 2011;22:4380–4389. doi: 10.1091/mbc.E10-12-0936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 473.Xie Y., Shao Y., Sun W., Ye G., Zhang X., Xiao B., Guo J. Downregulated expression of hsa_circ_0074362 in gastric cancer and its potential diagnostic values. Biomarkers Med. 2018;12:11–20. doi: 10.2217/bmm-2017-0114. [DOI] [PubMed] [Google Scholar]
  • 474.Shao Y., Li J., Lu R., Li T., Yang Y., Xiao B., Guo J. Global circular RNA expression profile of human gastric cancer and its clinical significance. Cancer Med. 2017;6:1173–1180. doi: 10.1002/cam4.1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 475.Wangxia L.V., Fang Y., Liu Y., Zhao Y., Shi Z., Zhong H. Circular RNA ARHGAP26 is over-expressed and its downregulation inhibits cell proliferation and promotes cell apoptosis in gastric cancer cells. Saudi J. Gastroenterol. 2019;25:119–125. doi: 10.4103/sjg.SJG_283_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 476.Hansen T.B., Jensen T.I., Clausen B.H., Bramsen J.B., Finsen B., Damgaard C.K., Kjems J. Natural RNA circles function as efficient microRNA sponges. Nature. 2013;495:384–388. doi: 10.1038/nature11993. [DOI] [PubMed] [Google Scholar]
  • 477.Tay Y., Rinn J., Pandolfi P.P. The multilayered complexity of ceRNA crosstalk and competition. Nature. 2014;505:344–352. doi: 10.1038/nature12986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 478.Barrett S.P., Salzman J. Circular RNAs: Analysis, expression and potential functions. Development. 2016;143:1838–1847. doi: 10.1242/dev.128074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 479.Memczak S., Jens M., Elefsinioti A., Torti F., Krueger J., Rybak A., Maier L., Mackowiak S.D., Gregersen L.H., Munschauer M. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495:333–338. doi: 10.1038/nature11928. [DOI] [PubMed] [Google Scholar]
  • 480.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. doi: 10.1016/j.jbiotec.2016.09.011. [DOI] [PubMed] [Google Scholar]
  • 481.He T., Li X., Xie D., Tian L. Overexpressed circPVT1 in oral squamous cell carcinoma promotes proliferation by serving as a miRNA sponge. Mol. Med. Rep. 2019;20:3509–3518. doi: 10.3892/mmr.2019.10615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 482.Li X., Zhang Z., Jiang H., Li Q., Wang R., Pan H., Niu Y., Liu F., Gu H., Fan X. Circular RNA circPVT1 promotes proliferation and invasion through sponging miR-125b and activating E2F2 signaling in non-small cell lung cancer. Cell. Physiol. Biochem. 2018;51:2324–2340. doi: 10.1159/000495876. [DOI] [PubMed] [Google Scholar]
  • 483.Chen J., Li Y., Zheng Q., Bao C., He J., Chen B., Lyu D., Zheng B., Xu Y., Long Z. Circular RNA profile identifies circPVT1 as a proliferative factor and prognostic marker in gastric cancer. Cancer Lett. 2017;388:208–219. doi: 10.1016/j.canlet.2016.12.006. [DOI] [PubMed] [Google Scholar]
  • 484.Kun-Peng Z., Xiao-Long M., Chun-Lin Z. Overexpressed circPVT1, a potential new circular RNA biomarker, contributes to doxorubicin and cisplatin resistance of osteosarcoma cells by regulating ABCB1. Int. J. Biol. Sci. 2018;14:321–330. doi: 10.7150/ijbs.24360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 485.Chen Z., Yao N., Gu H., Song Y., Ye Z., Li L., Lu P., Shao Q. Circular RNA_LARP4 sponges miR-1323 and hampers progression of esophageal squamous cell carcinoma through modulating PTEN/PI3K/AKT pathway. Dig. Dis. Sci. 2020;65:2272–2283. doi: 10.1007/s10620-019-05973-0. [DOI] [PubMed] [Google Scholar]
  • 486.Sun G., Li Z., He Z., Wang W., Wang S., Zhang X., Cao J., Xu P., Wang H., Huang X. Circular RNA MCTP2 inhibits cisplatin resistance in gastric cancer by miR-99a-5p-mediated induction of MTMR3 expression. J. Exp. Clin. Cancer Res. 2020;39:246. doi: 10.1186/s13046-020-01758-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 487.Pan H., Li T., Jiang Y., Pan C., Ding Y., Huang Z., Yu H., Kong D. Overexpression of circular RNA ciRS-7 abrogates the tumor suppressive effect of miR-7 on gastric cancer via PTEN/PI3K/AKT signaling pathway. J. Cell. Biochem. 2018;119:440–446. doi: 10.1002/jcb.26201. [DOI] [PubMed] [Google Scholar]
  • 488.Huang X.X., Zhang Q., Hu H., Jin Y., Zeng A.L., Xia Y.B., Xu L. A novel circular RNA circFN1 enhances cisplatin resistance in gastric cancer via sponging miR-182-5p. J. Cell. Biochem. 2020;122:1009–1020. doi: 10.1002/jcb.29641. [DOI] [PubMed] [Google Scholar]
  • 489.Li J., Qin X., Wu R., Wan L., Zhang L., Liu R. Circular RNA circFBXO11 modulates hepatocellular carcinoma progress and oxaliplatin resistance through miR-605/FOXO3/ABCB1 axis. J. Cell. Mol. Med. 2020;24:5152–5161. doi: 10.1111/jcmm.15162. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 490.Liu Y., Xia L., Dong L., Wang J., Xiao Q., Yu X., Zhu H. circHIPK3 promotes gemcitabine (GEM) resistance in pancreatic cancer cells by sponging miR-330-5p and targets RASSF1. Cancer Manag. Res. 2020;12:921–929. doi: 10.2147/CMAR.S239326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 491.Li S., Zheng S. Down-regulation of circ_0032833 sensitizes colorectal cancer to 5-fluorouracil and oxaliplatin partly depending on the regulation of miR-125-5p and MSI1. Cancer Manag. Res. 2020;12:11257–11269. doi: 10.2147/CMAR.S270123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 492.Jian X., He H., Zhu J., Zhang Q., Zheng Z., Liang X., Chen L., Yang M., Peng K., Zhang Z. hsa_circ_001680 affects the proliferation and migration of CRC and mediates its chemoresistance by regulating BMI1 through miR-340. Mol. Cancer. 2020;19:20. doi: 10.1186/s12943-020-1134-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 493.Wang X., Zhang H., Yang H., Bai M., Ning T., Deng T., Liu R., Fan Q., Zhu K., Li J. Exosome-delivered circRNA promotes glycolysis to induce chemoresistance through the miR-122-PKM2 axis in colorectal cancer. Mol. Oncol. 2020;14:539–555. doi: 10.1002/1878-0261.12629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 494.Amodio N., Raimondi L., Juli G., Stamato M.A., Caracciolo D., Tagliaferri P., Tassone P. MALAT1: A druggable long non-coding RNA for targeted anti-cancer approaches. J. Hematol. Oncol. 2018;11:63. doi: 10.1186/s13045-018-0606-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 495.Arun G., Diermeier S., Akerman M., Chang K.C., Wilkinson J.E., Hearn S., Kim Y., MacLeod A.R., Krainer A.R., Norton L. Differentiation of mammary tumors and reduction in metastasis upon Malat1 lncRNA loss. Genes Dev. 2016;30:34–51. doi: 10.1101/gad.270959.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 496.Liu Y., Zhou H., Yin T., Gong Y., Yuan G., Chen L., Liu J. Quercetin-modified gold-palladium nanoparticles as a potential autophagy inducer for the treatment of Alzheimer’s disease. J. Colloid Interface Sci. 2019;552:388–400. doi: 10.1016/j.jcis.2019.05.066. [DOI] [PubMed] [Google Scholar]
  • 497.Taieb J., Tabernero J., Mini E., Subtil F., Folprecht G., Van Laethem J.L., Thaler J., Bridgewater J., Petersen L.N., Blons H., PETACC-8 Study Investigators Oxaliplatin, fluorouracil, and leucovorin with or without cetuximab in patients with resected stage III colon cancer (PETACC-8): An open-label, randomised phase 3 trial. Lancet Oncol. 2014;15:862–873. doi: 10.1016/S1470-2045(14)70227-X. [DOI] [PubMed] [Google Scholar]
  • 498.Arunkumar G., Deva Magendhra Rao A.K., Manikandan M., Prasanna Srinivasa Rao H., Subbiah S., Ilangovan R., Murugan A.K., Munirajan A.K. Dysregulation of miR-200 family microRNAs and epithelial-mesenchymal transition markers in oral squamous cell carcinoma. Oncol. Lett. 2018;15:649–657. doi: 10.3892/ol.2017.7296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 499.Lin C.Y., Hsieh P.L., Liao Y.W., Peng C.Y., Lu M.Y., Yang C.H., Yu C.C., Liu C.M. Berberine-targeted miR-21 chemosensitizes oral carcinomas stem cells. Oncotarget. 2017;8:80900–80908. doi: 10.18632/oncotarget.20723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 500.Gong N., Teng X., Li J., Liang X.J. Antisense oligonucleotide-conjugated nanostructure-targeting lncRNA MALAT1 inhibits cancer metastasis. ACS Appl. Mater. Interfaces. 2019;11:37–42. doi: 10.1021/acsami.8b18288. [DOI] [PubMed] [Google Scholar]
  • 501.Di W., Li Q., Shen W., Guo H., Zhao S. The long non-coding RNA HOTAIR promotes thyroid cancer cell growth, invasion and migration through the miR-1-CCND2 axis. Am. J. Cancer Res. 2017;7:1298–1309. [PMC free article] [PubMed] [Google Scholar]

Articles from Molecular Therapy. Nucleic Acids are provided here courtesy of The American Society of Gene & Cell Therapy

RESOURCES