Skip to main content
American Journal of Cancer Research logoLink to American Journal of Cancer Research
. 2019 Jun 1;9(6):1118–1126.

Pivotal role of microRNA-138 in human cancers

Margaret Yeh 1, Christina S Oh 2, Ji Young Yoo 1, Balveen Kaur 1, Tae Jin Lee 1
PMCID: PMC6610051  PMID: 31285946

Abstract

Aberrant expression of certain microRNAs (miRNAs) has been implicated in cancers as a promising druggable target due to the fact that a modulation of the deregulated single miRNA seems to revert the therapeutically unfavorable gene expressions in cancer cell by targeting multiple genes. Global miRNA profiling from a number of patient cohorts in various type of human cancers has identified miR-138 as a signature of tumor suppressor that are down-regulated in most types of human cancer. As a tumor suppressor, miR-138 can inhibit oncogenic proteins by directly bind to their mRNAs. However, in rare cases of cancer stem cell population from glioblastoma, miR-138 seems to be down-regulated and plays an oncogenic function. This review will summarize accumulating evidence that has shown the expression and functional role of miR-138 in various human cancers with its target genes and pathways in a hope to find a better therapeutic option to treat human cancers.

Keywords: MicroRNA, miR-138, cancer, tumor suppressor, oncogene, glioblastoma

Introduction

Cancer is a complex disease caused by genetic abnormalities. Although cancer has been extensively studied over decades through the lens of tumor-suppressor genes and oncogenes, current understanding on its molecular level of tumorigenesis, progression and metastasis to identify genetic markers are still not sufficient to win the battle. Those genetic markers that are clearly related to cancers can be used not only as diagnostic or prognostic markers but also druggable targets for the development of new therapeutics.

MicroRNAs, approximately 23 nucleotides in length, are the smallest member of noncoding RNA molecules that play key roles in regulating the expression of coding genes [1]. Since the discovery of miRNAs and its involvement in all stages of cancer: tumorigenesis, progression and metastasis, a push for miRNA research in medical science has drawn a great attention from cancer researchers in a hope to find a new therapeutic target. Currently, it has been claimed that approximately 60% of human gene expressions are regulated by miRNAs [1,2]. In cancer cells, a number of different pathways have been shown to be involved with miRNAs, including cell proliferation and apoptosis [3], differentiation [4], cancer cell metabolism [5] and cell cycle [6]. Due to their crucial roles in controlling diverse metabolic and cellular biological processes, any dysregulation of genetic information could potentially develop into cancers. First miRNA was discovered in Caenorhabditis elegans by Victor Ambros laboratory as small RNAs transcribed from the gene lin-4 [7]. They found that both short and long (22 and 61 nucleotides in length) RNAs negatively regulates the lin-14 gene via attachment to the 3’ untranslated region (3’ UTR) region [7,8]. Shortly after, a similar finding of a small RNA molecule transcribed from the let-7 gene in C. elegans [9]. These findings led to the classification of these short RNA molecules as small temporal RNAs (stRNAs) [10], which were later termed miRNAs through the discovery of additional regulatory RNAs [2]. Since then, many miRNAs have been identified in variety of organisms including human and catalogued. Seminal finding in cancer biology was reported in 2002 by Carlo Croce laboratory, in which they found that miR-15/16 cluster is frequently deleted in chronic lymphocytic leukemia (CLL) patients as tumor suppressive players since the miR-15/16 can negatively regulate oncogene BCL2 expression [11]. This first pathological evidence that miRNAs are directly linked to human cancer development and progression has stimulated extensive studies on miRNAs in human cancers, and led to countless findings showing specific targeting of coding genes by specific miRNAs [12].

Biogenesis of miRNA

In order to effectively use those miRNAs as potential biomarkers and/or therapeutics, understanding the mechanism of miRNA biogenesis is critical. Mature form of miRNA is generated through a two-step cleavage of a long primary miRNA (pri-miRNA) after transcribed by RNA polymerase II [13]. The pri-miRNA is then processed by Drosha, a nuclear RNase III molecule, into a shorter ~70 nt hairpin structure, called a precursor miRNA (pre-miR) [14]. A nuclear membrane channel protein Exportin-5 (EXPO5) then transports the pre-miR into the cytoplasm [15], which is further cleaved again by a Dicer enzyme, creating a mature miRNA [16]. From there, the miRNA forms an RNA-induced silencing complex (RISC) and is guided to the 3’ UTR of target mRNA. The resulting miRNA/mRNA complex inhibits the ribosomal complex responsible for protein synthesis, effectively inhibiting the translation of gene expression [17]. Due to the nature of partial complementary binding between the 3’ UTR and the seed sequences of miRNA, the mature miRNA can bind to hundreds of distinct mRNA target strands [1]. It makes it extremely versatile and effective during the function of miRNAs in gene regulation processes. Furthermore, miRNAs have also been identified to irreversibly initiate mRNA degradation via deadenylation, which trumps the inhibition of translation by miRNA [18,19].

MicroRNA-138 (miR-138) in human cancers

MiR-138 originates from two primary transcripts, pri-miR-138-1 and pri-miR-138-2 as encoded on chromosomes 3 (3p21) and 16 (16q13) which form the mature miR-138 [20,21]. The expression level of miR-138 was found to be regulated by many different epigenetic mechanisms. For examples, DNA hypermethylation of miR-138 promoter is negatively correlated with miR-138 expression in breast cancer [22]. Some of transcription factors, such as p53, HOXA4 and GATA1, were also shown to regulate the expression of miR-138 in non-small cell lung carcinoma (NSCLC) [23,24] and chronic myeloid leukemia (CLL) [25]. The expression level of a certain miRNA has been used to determine whether the specific miRNA plays a tumor suppressive or oncogenic role in cancers [12]. If a miRNA was found to be highly expressed in cancer cells, the miRNA can be expected to play an oncogenic functions by targeting tumor suppressor genes. On the other hand, low expression level of a miRNA has been regarded as a tumor suppressor and those miRNAs are often found to target oncogenes. It has been reported that miR-138 is dysregulated across many cancer types [26]. According to the global transcriptome analysis on human cancer patient samples by The Cancer Genome Atlas (TCGA) Consortium, miR-138 has been found to express at low level in many types of human cancer [20]. Cumulating data obtained from cell line studies including HeLa, melanoma, breast, neuroblastoma and pancreatic tumor suggest that miR-138 may play a tumor suppressive role by targeting various oncogenes. An ectopic overexpression of miR-138 directly suppresses focal adhesion kinase (FAK), resulting in lowered invasiveness and increased susceptibility to chemotherapy [27]. Changes in miR-138 expression can affect cell proliferation, metastatic ability and drug resistance. Although this review will mainly focus on the tumor suppressive aspect of miR-138 in various human cancers, rare case of reports, however, will also be covered regarding an oncogenic role of miR-138 in malignant glioma patients [28,29]. From these studies, putative targets of miR-138 is summarized in Table 1.

Table 1.

Putative targets of miR-138 in human cancers

Cancer type Function Target
Glioblastoma (GBM) Tumor suppressor EZH2 [32]
CDK6 [32]
E2F2 [32]
E2F3 [32]
PD-1 [33]
CTLA-4 [33]
Oncogene C/EBPβ [28]
BIM [34]
Non-small cell lung cancer (NSCLC) Tumor suppressor EGFR [24]
SIRT1 [36]
GIT1 [77]
SEMA4C [77]
EZH2 [38]
PDK1 [78]
HOXA4 [24]
SNHG12 [40]
Oral squamous cell carcinoma (OSCC) Tumor suppressor ISG15 [51]
ΔNp63 [52]
YAP1 [49]
EZH2 [53]
PD-1 [54]
CTLA-4 [54]
Colorectal cancer (CRC) Tumor suppressor TERT [59]
PD-L1 [58]
PODXL [60]
HMGA1 [61]
TWIST2 [55]
Lcn2 [57]
Renal cell cancer (RCC) Tumor suppressor EZH2 [69]
FOXP4 [70]
Hepatocellular carcinoma (HCC) Tumor suppressor SOX9 [63]
Cyclin D3 [62]
Vimentin [65]
CCND3 [65]
Prostate cancer Tumor suppressor EZH2 [44]
Cervical cancer Tumor suppressor SIRT1 [71,72]
Gastric cancer Tumor suppressor SOX4 [66]
HIF-1α [67]

MiR-138 in brain tumor

In normal brain tissues, miR-138 is highly enriched within dendrites and regulates a depalmitoylation enzyme acyl protein thioesterase 1 (APT1) to control dendritic spine morphogenesis [30]. In mice study, the high level of miR-138 expression was correlated with better memory performance of the mice since miR-138 negatively inhibits APT1 expression [31]. The study speculated that increased APT1 expression occurs with aging due to the reduction of miR-138 level. In a similar vein, miR-138 expression has been found to be reduced in brain tumors. Low expression levels of miR-138 were associated with progression-related poor survival of GBM patients according to the TCGA database [32]. The study also found EZH2, CDK6, E2F2 and E2F3 as the direct targets of miR-138, through which EZH2-CDK4/6-pRb-E2F1 signaling pathway was impaired by ectopic miR-138 expression. When human CD4+ T cells were transfected with miR-138, it was shown that the miR-138 can directly down-regulate two immune checkpoints, cytotoxic T-lymphocyte-associated molecule 4 (CTLA-4) and programmed cell death 1 (PD-1), and result in 43% increase in median survival time in mice models [33]. This study demonstrated a potential of miR-138 that can be translated into an immunotherapeutic agent.

In contrast, a few compelling reports have surfaced with a clue that miR-138 may act as an oncogenic miRNA. For examples, it was found that miR-138 was overexpressed in tumor-initiating glioma stem cell (GSC) [29]. Therefore, ectopic expression of miR-138 promoted self-renewal potential of GSCs resulting their growth and survival. They also found that the transcriptional activation of miR-138 in glioma is mediated by C/EBPβ [28]. In this regard, miR-138 was also found to promote acquired resistance to temozolomide (TMZ) by directly targeting an apoptosis regulator BIM [34].

MiR-138 in lung cancer

In primary lung cancer, miR-138 has been shown to be downregulated by pertinent exposure to TGFβ1 and result in increased stemness through epithelial mesenchymal transition (EMT) [35]. As the result, the population of CD44+CD90+ cancer stem cell (CSC) was increased. MiR-138 can also lower EMT properties through decreased activity of the AMPK signaling pathway by directly targeting SIRT1 [36]. ZEB2 was also found to be a direct target of miR-138 in lung adenocarcinoma, since the ectopic expression of miR-138 suppresses EMT, cell proliferation and metastasis [37]. Overexpression of miR-138 suppressed cell proliferation by reducing expression of EGFR [24] or EZH2 [38]. In NSCLC, the transcription of miR-138 was regulated by direct binding of certain transcription factors, such as p53 [23] HOXA4 [24]. Interestingly, certain long non-coding RNAs (lncRNAs) also regulate the expression of miR-138 in lung cancer. For examples, PFAR plays as a competing endogenous RNA (ceRNA) of miR-138 in targeting regulation of yes-associated protein 1 (YAP1) leading to lung fibrosis [39]. In addition, small nucleolar RNA host gene 12 (SNHG12) was identified to be upregulated in NSCLC and negatively regulated miR-138 expression [40]. Overexpression of miR-138 also can sensitize lung cancer cell to conventional chemotherapeutic drugs. For an example, NSCLC patients used to develop acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR TKIs) such as gefitinib. Ectopic expression of miR-138 in NSCLC cells restored gefitinib activity of cell killing by directly targeting G protein-coupled receptor 124 (GPR124) [41].

MiR-138 in prostate cancer

Comprehensive miRNA profiling study from 37 patients has identified miR-138 as one of the down-regulated miRNAs in prostate cancer [42]. Comparison of miRNA profile between normal stem cell and cancer stem cell in prostate cancer cells revealed the inverse correlation between miR-138 expression and KRAS [43]. Exogenous miR-138 expression in prostate cancer cells directly targeted EZH2 resulting in cell cycle arrest in the G1/G0 phase [44]. MiR-138 overexpression can also reduce tumor migratory ability through down-regulation of the Wnt/B-catenin pathway [45,46].

MiR-138 in head and neck cancer

MiR-138 expression has been found to be down-regulated in many types of head and neck cancer patients. Two-step reverse transcriptase-quantitative PCR (qRT-PCR) using TaqMan miRNA Assays on 42 oral squamous cell carcinoma (OSCC) tumors and 8 adjacent normal specimens revealed that miR-138 is one of the most significantly down-regulated miRNAs together with miR-184 [47]. In contrast, another miRNA profiling study on 35 oral cavity and oropharynx squamous cell carcinoma (SCC) samples and 10 non-neoplastic oral mucosa controls also identified miR-138 as one of up-regulated miRNAs in 50% of samples [48]. This discrepancy needs to be resolved by examining more patient groups by their grade levels.

As observed in lung cancer, miR-138 suppresses cell proliferation through targeting YAP1 in OSCC [49]. In another study with OSCC cells, forced expression of miR-138 in OSCC cells significantly decreased promoted cell adhesion and inhibited migration, invasion and EMT by directly targeting fascin to reduce filopodia formation and paxillin expression [50]. The low expression level of miR-138 was also correspond with high expression of interferon-simulated gene 15 (ISG15) in OSCC [51]. In the study, over-expression of miR-138 in OSCC cells resulted in reduced ISG15 expression through direct binding to its 3’ UTR causing inhibition of invasion, proliferation and migratory ability [51]. DeltaNp63 (ΔNp63), the predominant isoform of TP63, was found to be significantly up-regulated in OSCC tissues in the negative correlation with miR-138 expression. Direct targeting of ΔNp63 by miR-138 mitigated its overexpression in cancerous OSCC cells to reduce cell growth, metastasis and stemness [52]. LncRNA, H19, was found to be highly expressed in OSCC in an inverse correlation with miR-138 and play an oncogenic role by competing with miR-138 to bind to the 3’ UTR of EZH2 mRNA [53].

Recently, there was an attempt to restore miR-138 in OSCC as a therapeutic purpose. Li and colleagues used gamma-delta T cell-derived extracellular vesicles (γδTDEs) as drug delivery system (DDS) for miR-138 [54]. Interestingly, miR-138 immunization via γδTDEs into OSCC bearing immunocompetent mice did not inhibited the tumor growth. However, injection of the miR-138 in γδTDE into immunocompetent mice increased immune response to OSCC tumor by increasing the proliferation, interferon-gamma (IFN-gamma) production and cytotoxicity of CD8+ T cells possibly through direct targeting of programmed cell death 1 (PD-1) and CTLA-4 of CD8+ T cells by the cargo miR-138 [54]. This finding is interesting since miR-138 showed its potential for immunotherapy.

MiR-138 in colorectal cancer

In colorectal cancer (CRC) tumors and cell lines, miR-138 is mostly found to be downregulated [55,56]. Its downregulation is linked to increased metastasis and poor prognosis through up-regulation of Twist basic helix-loop-helix transcription factor 2 gene (TWIST2) [55], which is a target for miR-138. In a metastasis to livers, miR-138 was also shown to target lipocalin 2 (Lcn-2) in a relationship to increased lipid metabolism [57]. As in brain tumor, miR-138 was shown in CRC to target an immunocheck point PD-L1 [58,59]. Xu and colleagues reported that Podocalyxin-like (PODXL) plays an oncogenic role in the development and progression of CRC and miR-138 directly targets PODXL as a tumor suppressor [60]. LncRNA was also found to correlate with miR-138 in CRC. For an example, lncRNA H19, which is highly overexpressed in CRC, was inversely correlated with miR-138 in CRC and its downstream target high-mobility group A (HMGA1) was shown to be directly targeted by miR-138 [61]. For these observations, it was proposed that inhibition of oncogenic miR-21 and induction of tumor suppressive miR-138 will be beneficial in CRC treatment [56].

MiR-138 in liver cancer

In hepatocellular carcinoma (HCC), more than 70% of the HCC patients showed the down-regulated level of miR-138 expression [62]. It was shown that miR-138 inhibits cell proliferation and invasion by directly targeting SOX9 or cyclin D3, therefore ectopic expression of miR-138 in HCC cells was expected to function as a useful therapeutics [62,63]. In another study, down-regulated expression of miR-138 was found to be inversely correlated with a putative oncogenic LncRNA DBH-AS1 [64]. Interestingly, they found that DBH-AS1 functions as a molecular sponge against miR-138 resulting in attenuated miR-138-mediated tumor suppressive roles through the rescue of FAK/Src/ERK pathway from miR-138. Similarly, circular RNA (circRNA) circRBM23 was found to overexpress in HCC patients in an inverse correlation with miR-138 expression [65]. In the study, miR-138 was down-regulated by circRBM23 resulting overexpression of oncogenic proteins, such as vimentin and CCND3.

MiR-138 in other cancers

In other cancers, miR-138 was also mostly found to be down-regulated to function as a tumor suppressor. In gastric cancer, miR-138 is consistently down-regulated in tumor tissue and cell lines [66]. The study found that the low level of miR-138 expression corresponds with increased EMT from both tumor node and lymph nodes through its direct target SRY-related high mobility group box 4 (SOX4), a master mediator of EMT. LncRNA LINC00152 was found to overexpress in gastric cancer and functions as a molecular sponge for miR-138 to rescue hypoxia inducible factor-1α (HIF-1α) suppression [67]. CREPT regulates oncogenic beta-catenin/TCF4/cyclin D1 pathway in breast cancer, and is directly targeted miR-138 that is found to be down-regulated in most breast cancer patients [68]. MiR-138 is also down-regulated in renal cell carcinoma (RCC) and found to induce cell senescence by targeting EZH2 [69]. Interestingly, estrogen receptor β (ER-β) induces an oncogenic lncRNA, HOTAIR, in RCC, which antagonizes tumor suppressive miRNAs including miR-138 [26]. Recently, circRNA ZNF609, which was found to overexpress in RCC, was also shown to compete with miR-138 to target forkhead box P4 (FOXP4) [70]. In cervical cancer, SIRT1 was shown to be targeted by miR-138 which, in turn, can be sponged by up-regulated lncRNA TUG1 or H19 [71,72].

Perspectives

Discovery of miRNAs has highlighted their profound functions in almost every cellular pathways human cancers during cancer initiation, progression, migration and metastasis. Global miRNA profiling from a number of patient cohorts in various type of human cancers has identified miR-138 as a signature of tumor suppressor that are mostly down-regulated in cancer cells. Only in rare cases, miR-138 was found to be up-regulated, such as in cancer stem cells. As a tumor suppressor, miR-138 can inhibit oncogenic proteins by directly binding to their mRNAs. More importantly, accumulating evidence has shown that miR-138, as other miRNAs, can regulate multiple targets in cancer cells. These findings make miR-138 as an attractive therapeutic option to treat human cancers. Interestingly, it was observed only in brain tumors that miR-138 can function as an oncogenic miR in certain type of brain tumor cells. Although these controversial results regarding the functional role of miR-138 in brain tumors need to be further resolved, it may imply the possibility of dual functional role of miRNAs depending on the context. The most studies about down regulation and corresponding tumor suppressive function of miR-138 were obtained from primary brain tumors. However, the oncogenic role of miR-138 was observed in either GSCs or chemotherapy-resistant cells. It will be interesting to test whether the recurring brain tumor cells are related to the overexpression of miR-138 and how miR-138 changes its functions and targets depending on cell context not only in brain tumor also in other types of cancer. In order to use miR-138 for translational purpose of clinical therapeutics, it will be a critical issue to decide whose patients need to be treated with miR-138 overexpression or miR-138 inhibitors. Therefore, future studies will need to focus on how to deliver such therapeutically potent miR-138 into a specific cancer cell type to inhibit cancer progression and metastasis. Although many of such attempts to delivery miRNAs into cancer cells have not been successful, emerging new strategy for cell and tissue specific delivery are promising to accomplish this task. Our recent progress on RNA nanotechnology may solve these hard issues. We have shown that RNA-based nanoparticle or exosomes can successfully provide efficient and safe targeted delivery method for such small noncoding RNAs including siRNAs and miRNAs [73-76]. Nevertheless, miRNA-based cancer therapy needs further understanding in detail for their functional mechanism through target down-regulations in order to surpass or complement conventional treatment options to improve patient outcome.

Acknowledgements

This work was supported in part by Pilot/Feasibility Program from Texas Medical Center Digestive Diseases Center through National Institutes of Health National Institute of Diabetes And Digestive And Kidney Diseases (NIDDK) [Grant P30 DK056338] to T.J.L.; and the National Institutes of Health National Cancer Institute (NCI) [Grant R01 CA150153 and P01 CA163205] and National Institutes of Health National Institute of Neurological Disorders and Stroke (NINDS) [Grant R01 NS064607] to B.K.

Disclosure of conflict of interest

None.

References

  • 1.Bartel DP. Metazoan MicroRNAs. Cell. 2018;173:20–51. doi: 10.1016/j.cell.2018.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136:215–33. doi: 10.1016/j.cell.2009.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lynam-Lennon N, Maher SG, Reynolds JV. The roles of microRNA in cancer and apoptosis. Biol Rev Camb Philos Soc. 2009;84:55–71. doi: 10.1111/j.1469-185X.2008.00061.x. [DOI] [PubMed] [Google Scholar]
  • 4.Chen CZ, Li L, Lodish HF, Bartel DP. MicroRNAs modulate hematopoietic lineage differentiation. Science. 2004;303:83–6. doi: 10.1126/science.1091903. [DOI] [PubMed] [Google Scholar]
  • 5.Chen B, Li H, Zeng X, Yang P, Liu X, Zhao X, Liang S. Roles of microRNA on cancer cell metabolism. J Transl Med. 2012;10:228. doi: 10.1186/1479-5876-10-228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Singh DK, Bose S, Kumar S. Role of microRNA in regulating cell signaling pathways, cell cycle, and apoptosis in non-small cell lung cancer. Curr Mol Med. 2016 [Epub ahead of print] [PubMed] [Google Scholar]
  • 7.Lee RC, Feinbaum RL, Ambros V. The C. Elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75:843–54. doi: 10.1016/0092-8674(93)90529-y. [DOI] [PubMed] [Google Scholar]
  • 8.Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell. 1993;75:855–62. doi: 10.1016/0092-8674(93)90530-4. [DOI] [PubMed] [Google Scholar]
  • 9.Reinhart BJ, Slack FJ, Basson M, Pasquinelli AE, Bettinger JC, Rougvie AE, Horvitz HR, Ruvkun G. The 21-nucleotide let-7 RNA regulates developmental timing in caenorhabditis elegans. Nature. 2000;403:901–6. doi: 10.1038/35002607. [DOI] [PubMed] [Google Scholar]
  • 10.Pasquinelli AE, Reinhart BJ, Slack F, Martindale MQ, Kuroda MI, Maller B, Hayward DC, Ball EE, Degnan B, Muller P, Spring J, Srinivasan A, Fishman M, Finnerty J, Corbo J, Levine M, Leahy P, Davidson E, Ruvkun G. Conservation of the sequence and temporal expression of let-7 heterochronic regulatory RNA. Nature. 2000;408:86–9. doi: 10.1038/35040556. [DOI] [PubMed] [Google Scholar]
  • 11.Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, Aldler H, Rattan S, Keating M, Rai K, Rassenti L, Kipps T, Negrini M, Bullrich F, Croce CM. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A. 2002;99:15524–9. doi: 10.1073/pnas.242606799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006;6:857–66. doi: 10.1038/nrc1997. [DOI] [PubMed] [Google Scholar]
  • 13.Lee Y, Kim M, Han J, Yeom KH, Lee S, Baek SH, Kim VN. MicroRNA genes are transcribed by RNA polymerase II. EMBO J. 2004;23:4051–60. doi: 10.1038/sj.emboj.7600385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Radmark O, Kim S, Kim VN. The nuclear RNase III Drosha initiates microRNA processing. Nature. 2003;425:415–9. doi: 10.1038/nature01957. [DOI] [PubMed] [Google Scholar]
  • 15.Yi R, Qin Y, Macara IG, Cullen BR. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev. 2003;17:3011–6. doi: 10.1101/gad.1158803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Hutvagner G, McLachlan J, Pasquinelli AE, Balint E, Tuschl T, Zamore PD. A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA. Science. 2001;293:834–8. doi: 10.1126/science.1062961. [DOI] [PubMed] [Google Scholar]
  • 17.Olsen PH, Ambros V. The lin-4 regulatory RNA controls developmental timing in caenorhabditis elegans by blocking LIN-14 protein synthesis after the initiation of translation. Dev Biol. 1999;216:671–80. doi: 10.1006/dbio.1999.9523. [DOI] [PubMed] [Google Scholar]
  • 18.Bagga S, Bracht J, Hunter S, Massirer K, Holtz J, Eachus R, Pasquinelli AE. Regulation by let-7 and lin-4 miRNAs results in target mRNA degradation. Cell. 2005;122:553–63. doi: 10.1016/j.cell.2005.07.031. [DOI] [PubMed] [Google Scholar]
  • 19.Eichhorn SW, Guo H, McGeary SE, Rodriguez-Mias RA, Shin C, Baek D, Hsu SH, Ghoshal K, Villen J, Bartel DP. mRNA destabilization is the dominant effect of mammalian microRNAs by the time substantial repression ensues. Mol Cell. 2014;56:104–15. doi: 10.1016/j.molcel.2014.08.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sha HH, Wang DD, Chen D, Liu SW, Wang Z, Yan DL, Dong SC, Feng JF. MiR-138: a promising therapeutic target for cancer. Tumour Biol. 2017;39:1010428317697575. doi: 10.1177/1010428317697575. [DOI] [PubMed] [Google Scholar]
  • 21.Zhao X, Yang L, Hu J, Ruan J. miR-138 might reverse multidrug resistance of leukemia cells. Leuk Res. 2010;34:1078–82. doi: 10.1016/j.leukres.2009.10.002. [DOI] [PubMed] [Google Scholar]
  • 22.Munoz-Rodriguez JL, Vrba L, Futscher BW, Hu C, Komenaka IK, Meza-Montenegro MM, Gutierrez-Millan LE, Daneri-Navarro A, Thompson PA, Martinez ME. Differentially expressed microRNAs in postpartum breast cancer in Hispanic women. PLoS One. 2015;10:e0124340. doi: 10.1371/journal.pone.0124340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Li J, Xia W, Su X, Qin X, Chen Y, Li S, Dong J, Ding H, Li H, Huang A, Ge X, Hou L, Wang C, Sun L, Bai C, Shen X, Fang T, Liu Y, Zhang Y, Zhang H, Zhang H, Shao N. Species-specific mutual regulation of p53 and miR-138 between human, rat and mouse. Sci Rep. 2016;6:26187. doi: 10.1038/srep26187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Tang X, Jiang J, Zhu J, He N, Tan J. HOXA4-regulated miR-138 suppresses proliferation and gefitinib resistance in non-small cell lung cancer. Mol Genet Genomics. 2019;294:85–93. doi: 10.1007/s00438-018-1489-3. [DOI] [PubMed] [Google Scholar]
  • 25.Xu C, Fu H, Gao L, Wang L, Wang W, Li J, Li Y, Dou L, Gao X, Luo X, Jing Y, Chim CS, Zheng X, Yu L. BCR-ABL/GATA1/miR-138 mini circuitry contributes to the leukemogenesis of chronic myeloid leukemia. Oncogene. 2014;33:44–54. doi: 10.1038/onc.2012.557. [DOI] [PubMed] [Google Scholar]
  • 26.Ding J, Yeh CR, Sun Y, Lin C, Chou J, Ou Z, Chang C, Qi J, Yeh S. Estrogen receptor beta promotes renal cell carcinoma progression via regulating LncRNA HOTAIR-miR-138/200c/204/217 associated CeRNA network. Oncogene. 2018;37:5037–53. doi: 10.1038/s41388-018-0175-6. [DOI] [PubMed] [Google Scholar]
  • 27.Golubovskaya VM, Sumbler B, Ho B, Yemma M, Cance WG. MiR-138 and MiR-135 directly target focal adhesion kinase, inhibit cell invasion, and increase sensitivity to chemotherapy in cancer cells. Anticancer Agents Med Chem. 2014;14:18–28. doi: 10.2174/187152061401140108113435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Di Pascale F, Nama S, Muhuri M, Quah S, Ismail HM, Chan XHD, Sundaram GM, Ramalingam R, Burke B, Sampath P. C/EBPbeta mediates RNA polymerase III-driven transcription of oncomiR-138 in malignant gliomas. Nucleic Acids Res. 2018;46:336–49. doi: 10.1093/nar/gkx1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chan XH, Nama S, Gopal F, Rizk P, Ramasamy S, Sundaram G, Ow GS, Ivshina AV, Tanavde V, Haybaeck J, Kuznetsov V, Sampath P. Targeting glioma stem cells by functional inhibition of a prosurvival oncomiR-138 in malignant gliomas. Cell Rep. 2012;2:591–602. doi: 10.1016/j.celrep.2012.07.012. [DOI] [PubMed] [Google Scholar]
  • 30.Siegel G, Obernosterer G, Fiore R, Oehmen M, Bicker S, Christensen M, Khudayberdiev S, Leuschner PF, Busch CJ, Kane C, Hubel K, Dekker F, Hedberg C, Rengarajan B, Drepper C, Waldmann H, Kauppinen S, Greenberg ME, Draguhn A, Rehmsmeier M, Martinez J, Schratt GM. A functional screen implicates microRNA-138-dependent regulation of the depalmitoylation enzyme APT1 in dendritic spine morphogenesis. Nat Cell Biol. 2009;11:705–16. doi: 10.1038/ncb1876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Tatro ET, Risbrough V, Soontornniyomkij B, Young J, Shumaker-Armstrong S, Jeste DV, Achim CL. Short-term recognition memory correlates with regional CNS expression of microRNA-138 in mice. Am J Geriatr Psychiatry. 2013;21:461–73. doi: 10.1016/j.jagp.2012.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Qiu S, Huang D, Yin D, Li F, Li X, Kung HF, Peng Y. Suppression of tumorigenicity by microRNA-138 through inhibition of EZH2-CDK4/6-pRb-E2F1 signal loop in glioblastoma multiforme. Biochim Biophys Acta. 2013;1832:1697–707. doi: 10.1016/j.bbadis.2013.05.015. [DOI] [PubMed] [Google Scholar]
  • 33.Wei J, Nduom EK, Kong LY, Hashimoto Y, Xu S, Gabrusiewicz K, Ling X, Huang N, Qiao W, Zhou S, Ivan C, Fuller GN, Gilbert MR, Overwijk W, Calin GA, Heimberger AB. MiR-138 exerts anti-glioma efficacy by targeting immune checkpoints. Neuro Oncol. 2016;18:639–48. doi: 10.1093/neuonc/nov292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Stojcheva N, Schechtmann G, Sass S, Roth P, Florea AM, Stefanski A, Stuhler K, Wolter M, Muller NS, Theis FJ, Weller M, Reifenberger G, Happold C. MicroRNA-138 promotes acquired alkylator resistance in glioblastoma by targeting the Bcl-2-interacting mediator BIM. Oncotarget. 2016;7:12937–50. doi: 10.18632/oncotarget.7346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Zhang F, Li T, Han L, Qin P, Wu Z, Xu B, Gao Q, Song Y. TGFbeta1-induced down-regulation of microRNA-138 contributes to epithelial-mesenchymal transition in primary lung cancer cells. Biochem Biophys Res Commun. 2018;496:1169–75. doi: 10.1016/j.bbrc.2018.01.164. [DOI] [PubMed] [Google Scholar]
  • 36.Ye Z, Fang B, Pan J, Zhang N, Huang J, Xie C, Lou T, Cao Z. miR-138 suppresses the proliferation, metastasis and autophagy of non-small cell lung cancer by targeting Sirt1. Oncol Rep. 2017;37:3244–52. doi: 10.3892/or.2017.5619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.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]
  • 38.Zhang H, Zhang H, Zhao M, Lv Z, Zhang X, Qin X, Wang H, Wang S, Su J, Lv X, Liu H, Du W, Zhou W, Chen X, Fei K. MiR-138 inhibits tumor growth through repression of EZH2 in non-small cell lung cancer. Cell Physiol Biochem. 2013;31:56–65. doi: 10.1159/000343349. [DOI] [PubMed] [Google Scholar]
  • 39.Zhao X, Sun J, Chen Y, Su W, Shan H, Li Y, Wang Y, Zheng N, Shan H, Liang H. lncRNA PFAR Promotes Lung Fibroblast Activation and Fibrosis by Targeting miR-138 to Regulate the YAP1-Twist Axis. Mol Ther. 2018;26:2206–17. doi: 10.1016/j.ymthe.2018.06.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Wang X, Qi G, Zhang J, Wu J, Zhou N, Li L, Ma J. Knockdown of long noncoding RNA small nucleolar RNA host gene 12 inhibits cell growth and induces apoptosis by upregulating mir-138 in nonsmall cell lung cancer. DNA Cell Biol. 2017;36:892–900. doi: 10.1089/dna.2017.3830. [DOI] [PubMed] [Google Scholar]
  • 41.Gao Y, Fan X, Li W, Ping W, Deng Y, Fu X. miR-138-5p reverses gefitinib resistance in non-small cell lung cancer cells via negatively regulating G protein-coupled receptor 124. Biochem Biophys Res Commun. 2014;446:179–86. doi: 10.1016/j.bbrc.2014.02.073. [DOI] [PubMed] [Google Scholar]
  • 42.Walter BA, Valera VA, Pinto PA, Merino MJ. Comprehensive microRNA profiling of prostate cancer. J Cancer. 2013;4:350–7. doi: 10.7150/jca.6394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ngalame NN, Tokar EJ, Person RJ, Xu Y, Waalkes MP. Aberrant microRNA expression likely controls RAS oncogene activation during malignant transformation of human prostate epithelial and stem cells by arsenic. Toxicol Sci. 2014;138:268–77. doi: 10.1093/toxsci/kfu002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Erdmann K, Kaulke K, Rieger C, Salomo K, Wirth MP, Fuessel S. MiR-26a and miR-138 block the G1/S transition by targeting the cell cycle regulating network in prostate cancer cells. J Cancer Res Clin Oncol. 2016;142:2249–61. doi: 10.1007/s00432-016-2222-4. [DOI] [PubMed] [Google Scholar]
  • 45.Erdmann K, Kaulke K, Rieger C, Wirth MP, Fuessel S. Induction of alpha-methylacyl-CoA racemase by miR-138 via up-regulation of beta-catenin in prostate cancer cells. J Cancer Res Clin Oncol. 2017;143:2201–10. doi: 10.1007/s00432-017-2484-5. [DOI] [PubMed] [Google Scholar]
  • 46.Yu Z, Wang Z, Li F, Yang J, Tang L. miR138 modulates prostate cancer cell invasion and migration via Wnt/betacatenin pathway. Mol Med Rep. 2018;17:3140–5. doi: 10.3892/mmr.2017.8273. [DOI] [PubMed] [Google Scholar]
  • 47.Manikandan M, Deva Magendhra Rao AK, Rajkumar KS, Rajaraman R, Munirajan AK. Altered levels of miR-21, miR-125b-2*, miR-138, miR-155, miR-184, and miR-205 in oral squamous cell carcinoma and association with clinicopathological characteristics. J Oral Pathol Med. 2015;44:792–800. doi: 10.1111/jop.12300. [DOI] [PubMed] [Google Scholar]
  • 48.Brito BL, Lourenco SV, Damascena AS, Kowalski LP, Soares FA, Coutinho-Camillo CM. Expression of stem cell-regulating miRNAs in oral cavity and oropharynx squamous cell carcinoma. J Oral Pathol Med. 2016;45:647–54. doi: 10.1111/jop.12424. [DOI] [PubMed] [Google Scholar]
  • 49.Xu R, Zeng G, Gao J, Ren Y, Zhang Z, Zhang Q, Zhao J, Tao H, Li D. miR-138 suppresses the proliferation of oral squamous cell carcinoma cells by targeting Yes-associated protein 1. Oncol Rep. 2015;34:2171–8. doi: 10.3892/or.2015.4144. [DOI] [PubMed] [Google Scholar]
  • 50.Rodrigues PC, Sawazaki-Calone I, Ervolino de Oliveira C, Soares Macedo CC, Dourado MR, Cervigne NK, Miguel MC, Ferreira do Carmo A, Lambert DW, Graner E, Daniela da Silva S, Alaoui-Jamali MA, Paes Leme AF, Salo TA, Coletta RD. Fascin promotes migration and invasion and is a prognostic marker for oral squamous cell carcinoma. Oncotarget. 2017;8:74736–54. doi: 10.18632/oncotarget.20360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Zhang Q, He Y, Nie M, Cai W. Roles of miR-138 and ISG15 in oral squamous cell carcinoma. Exp Ther Med. 2017;14:2329–34. doi: 10.3892/etm.2017.4720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zhuang Z, Xie N, Hu J, Yu P, Wang C, Hu X, Han X, Hou J, Huang H, Liu X. Interplay between DeltaNp63 and miR-138-5p regulates growth, metastasis and stemness of oral squamous cell carcinoma. Oncotarget. 2017;8:21954–73. doi: 10.18632/oncotarget.15752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hong Y, He H, Sui W, Zhang J, Zhang S, Yang D. Long non-coding RNA H1 promotes cell proliferation and invasion by acting as a ceRNA of miR138 and releasing EZH2 in oral squamous cell carcinoma. Int J Oncol. 2018;52:901–12. doi: 10.3892/ijo.2018.4247. [DOI] [PubMed] [Google Scholar]
  • 54.Li L, Lu S, Liang X, Cao B, Wang S, Jiang J, Luo H, He S, Lang J, Zhu G. gammadeltaTDEs: an efficient delivery system for miR-138 with anti-tumoral and immunostimulatory roles on oral squamous cell carcinoma. Mol Ther Nucleic Acids. 2019;14:101–13. doi: 10.1016/j.omtn.2018.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Long L, Huang G, Zhu H, Guo Y, Liu Y, Huo J. Down-regulation of miR-138 promotes colorectal cancer metastasis via directly targeting TWIST2. J Transl Med. 2013;11:275. doi: 10.1186/1479-5876-11-275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.You C, Jin L, Xu Q, Shen B, Jiao X, Huang X. Expression of miR-21 and miR-138 in colon cancer and its effect on cell proliferation and prognosis. Oncol Lett. 2019;17:2271–7. doi: 10.3892/ol.2018.9864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Cristobal I, Torrejon B, Gonzalez-Alonso P, Manso R, Rojo F, Garcia-Foncillas J. Downregulation of miR-138 as a contributing mechanism to Lcn-2 overexpression in colorectal cancer with liver metastasis. World J Surg. 2016;40:1021–2. doi: 10.1007/s00268-015-3241-z. [DOI] [PubMed] [Google Scholar]
  • 58.Zhao L, Yu H, Yi S, Peng X, Su P, Xiao Z, Liu R, Tang A, Li X, Liu F, Shen S. The tumor suppressor miR-138-5p targets PD-L1 in colorectal cancer. Oncotarget. 2016;7:45370–84. doi: 10.18632/oncotarget.9659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Zhang XL, Xu LL, Wang F. Hsa_circ_0020397 regulates colorectal cancer cell viability, apoptosis and invasion by promoting the expression of the miR-138 targets TERT and PD-L1. Cell Biol Int. 2017;41:1056–64. doi: 10.1002/cbin.10826. [DOI] [PubMed] [Google Scholar]
  • 60.Xu Y, Pan ZG, Shu L, Li QJ. Podocalyxin-like, targeted by miR-138, promotes colorectal cancer cell proliferation, migration, invasion and EMT. Eur Rev Med Pharmacol Sci. 2018;22:8664–74. doi: 10.26355/eurrev_201812_16631. [DOI] [PubMed] [Google Scholar]
  • 61.Yang Q, Wang X, Tang C, Chen X, He J. H19 promotes the migration and invasion of colon cancer by sponging miR-138 to upregulate the expression of HMGA1. Int J Oncol. 2017;50:1801–9. doi: 10.3892/ijo.2017.3941. [DOI] [PubMed] [Google Scholar]
  • 62.Wang W, Zhao LJ, Tan YX, Ren H, Qi ZT. MiR-138 induces cell cycle arrest by targeting cyclin D3 in hepatocellular carcinoma. Carcinogenesis. 2012;33:1113–20. doi: 10.1093/carcin/bgs113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Liu Y, Zhang W, Liu K, Liu S, Ji B, Wang Y. miR-138 suppresses cell proliferation and invasion by inhibiting SOX9 in hepatocellular carcinoma. Am J Transl Res. 2016;8:2159–68. [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 64.Bao J, Chen X, Hou Y, Kang G, Li Q, Xu Y. LncRNA DBH-AS1 facilitates the tumorigenesis of hepatocellular carcinoma by targeting miR-138 via FAK/Src/ERK pathway. Biomed Pharmacother. 2018;107:824–33. doi: 10.1016/j.biopha.2018.08.079. [DOI] [PubMed] [Google Scholar]
  • 65.Wang B, Chen H, Zhang C, Yang T, Zhao Q, Yan Y, Zhang Y, Xu F. Effects of hsa_circRBM23 on hepatocellular carcinoma cell viability and migration as produced by regulating miR-138 expression. Cancer Biother Radiopharm. 2018;33:194–202. doi: 10.1089/cbr.2017.2424. [DOI] [PubMed] [Google Scholar]
  • 66.Pang L, Li B, Zheng B, Niu L, Ge L. miR-138 inhibits gastric cancer growth by suppressing SOX4. Oncol Rep. 2017;38:1295–302. doi: 10.3892/or.2017.5745. [DOI] [PubMed] [Google Scholar]
  • 67.Cai Q, Wang Z, Wang S, Weng M, Zhou D, Li C, Wang J, Chen E, Quan Z. Long non-coding RNA LINC00152 promotes gallbladder cancer metastasis and epithelial-mesenchymal transition by regulating HIF-1alpha via miR-138. Open Biol. 2017;7 doi: 10.1098/rsob.160247. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 68.Liang Z, Feng Q, Xu L, Li S, Zhou L. CREPT regulated by miR-138 promotes breast cancer progression. Biochem Biophys Res Commun. 2017;493:263–9. doi: 10.1016/j.bbrc.2017.09.033. [DOI] [PubMed] [Google Scholar]
  • 69.Liang J, Zhang Y, Jiang G, Liu Z, Xiang W, Chen X, Chen Z, Zhao J. MiR-138 induces renal carcinoma cell senescence by targeting EZH2 and is downregulated in human clear cell renal cell carcinoma. Oncol Res. 2013;21:83–91. doi: 10.3727/096504013X13775486749218. [DOI] [PubMed] [Google Scholar]
  • 70.Xiong Y, Zhang J, Song C. CircRNA ZNF609 functions as a competitive endogenous RNA to regulate FOXP4 expression by sponging miR-138-5p in renal carcinoma. J Cell Physiol. 2019;234:10646–54. doi: 10.1002/jcp.27744. [DOI] [PubMed] [Google Scholar]
  • 71.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;8:65253–64. doi: 10.18632/oncotarget.18224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Ou L, Wang D, Zhang H, Yu Q, Hua F. Decreased expression of miR-138-5p by lncRNA H19 in cervical cancer promotes tumor proliferation. Oncol Res. 2018;26:401–10. doi: 10.3727/096504017X15017209042610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lee TJ, Haque F, Shu D, Yoo JY, Li H, Yokel RA, Horbinski C, Kim TH, Kim SH, Kwon CH, Nakano I, Kaur B, Guo P, Croce CM. RNA nanoparticle as a vector for targeted siRNA delivery into glioblastoma mouse model. Oncotarget. 2015;6:14766–76. doi: 10.18632/oncotarget.3632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Lee TJ, Haque F, Vieweger M, Yoo JY, Kaur B, Guo P, Croce CM. Functional assays for specific targeting and delivery of RNA nanoparticles to brain tumor. Methods Mol Biol. 2015;1297:137–52. doi: 10.1007/978-1-4939-2562-9_10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lee TJ, Yoo JY, Shu D, Li H, Zhang J, Yu JG, Jaime-Ramirez AC, Acunzo M, Romano G, Cui R, Sun HL, Luo Z, Old M, Kaur B, Guo P, Croce CM. RNA nanoparticle-based targeted therapy for glioblastoma through inhibition of oncogenic miR-21. Mol Ther. 2017;25:1544–1555. doi: 10.1016/j.ymthe.2016.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Pi F, Binzel DW, Lee TJ, Li Z, Sun M, Rychahou P, Li H, Haque F, Wang S, Croce CM, Guo B, Evers BM, Guo P. Nanoparticle orientation to control RNA loading and ligand display on extracellular vesicles for cancer regression. Nat Nanotechnol. 2018;13:82–9. doi: 10.1038/s41565-017-0012-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Li J, Wang Q, Wen R, Liang J, Zhong X, Yang W, Su D, Tang J. MiR-138 inhibits cell proliferation and reverses epithelial-mesenchymal transition in non-small cell lung cancer cells by targeting GIT1 and SEMA4C. J Cell Mol Med. 2015;19:2793–805. doi: 10.1111/jcmm.12666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Ye XW, Yu H, Jin YK, Jing XT, Xu M, Wan ZF, Zhang XY. miR-138 inhibits proliferation by targeting 3-phosphoinositide-dependent protein kinase-1 in non-small cell lung cancer cells. Clin Respir J. 2015;9:27–33. doi: 10.1111/crj.12100. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Cancer Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES