microRNA-18b is upregulated in breast cancer and modulates genes involved in cell migration

  • Authors:
    • Miguel A. Fonseca-Sanchéz
    • Carlos Pérez-Plasencia
    • Jorge Fernández-Retana
    • Elena Arechaga-Ocampo
    • Laurence A. Marchat
    • Sergio Rodríguez-Cuevas
    • Veronica Bautista-Piña
    • Zaira E. Arellano-Anaya
    • Ali Flores-Pérez
    • José Diaz-Chávez
    • César López-Camarillo
  • View Affiliations

  • Published online on: August 22, 2013     https://doi.org/10.3892/or.2013.2691
  • Pages: 2399-2410
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

microRNAs are small non-coding RNAs of ~22 nucleotides that function at post-transcriptional level as negative regulators of gene expression. Aberrant expression of microRNAs could promote uncontrolled proliferation, migration and invasion of human cancer cells. In this study, we analyzed the expression of microRNA-18b (miR-18b) in breast cancer cell lines and in a set of clinical specimens. Our results showed that miR-18b was upregulated in four out of five breast cancer cell lines and also in breast tumors. In order to identify potential gene targets, we carried out transcriptional profiling of MDA-MB-231 breast cancer cells that ectopically expressed miR-18b. Our results showed that 263 genes were significantly modulated in miR-18b-deficient cells (fold change >1.5; P≤0.05). We found that knock-down of miR-18b induced the upregulation of 55 olfactory receptor (OR) genes and nine genes (NLRP7, KLK3, OLFM3, POSTN, MAGED4B, KIR3DL3, CRX, SEMG1 and CEACAM5) with key roles in cell migration and metastasis. Consistently, we found that ectopic inhibition of miR-18b suppressed the migration of two breast cancer cell models in vitro. In conclusion, we have uncovered genes directly or indirectly modulated by miR-18b which may represent potential therapeutic targets in breast cancer. Our data also pointed out a role of miR-18b in migration of breast cancer cells.

Introduction

microRNAs are small non-coding RNAs of ~22 nucleotides that function at post-transcriptional level as negative regulators of gene expression. In normal cells, microRNAs maintain a balance of key cellular processes including cell growth, proliferation, differentiation and cell death. Abnormal expression of microRNAs occurs frequently in breast cancer where they act as oncogenes and tumor-suppressors. microRNAs may contribute to tumor initiation and progression by inhibiting apoptosis and promoting uncontrolled proliferation, invasion and metastasis. In consequence, they represent potential prognostic markers and novel therapeutic targets in human cancer (1). In animal cells, microRNAs are frequently transcribed together as polycistronic primary transcripts that are processed into multiple individual mature microRNAs. Particularly, microRNA-18b (miR-18b) is located at chromosome X within the miR-106a-363 cluster that is constituted by six different microRNAs (2,3). It has been proposed that expression of this microRNAs cluster is undetectable or very low in mouse (4). Diverse reports associated the aberrant expression of miR-18b to diverse human pathologies including cardiac hypertrophy (5), multiple sclerosis (6) and hepatitis B virus chronic infection (7). In human cancer, miR-18b expression is also deregulated. It has been reported that miR-18b is highly expressed in gastric cancer (8), in malignant serous tumors from borderline and benign ovarian tumors (9), in basal cell carcinoma of the skin (10), and in colonic cancer (11). Some miR-18b targets have been experimentally identified. In stem-like cells isolated from glioblastoma, it was reported that miR-18b targets the NOTCH2, NEDD9 and MEKK1 genes (12). In breast cancer miR-18a, miR-18b, and other 21 microRNAs negatively regulate the estrogen receptor (ER)-α signaling (13). In addition, miR-18a and miR-18b showed higher expression levels in ER-α negative clinical specimens (13). In another study, a low expression of miR-18b was associated with improved survival in human epidermal receptor (HER) 2-negative breast cancer, although it was not correlated with ER-α protein levels (14). Recently, higher levels of circulating miR-18b were proposed as a potential marker of breast cancer (15). Although knowledge on the roles of miR-18b in cancer is increasing, the physiological functions still remain unclear and the number of detected miR-18b targets in cancer is scarce. Here, we studied the miR-18b expression in breast cancer cell lines and clinical tumors. In addition, we performed a genomic-wide study using DNA microarrays in order to identify genes regulated by miR-18b with putative roles in breast cancer.

Materials and methods

Tissue samples

Human primary breast tumors and adjacent non-tumoral tissues were obtained from the Institute of Breast Diseases-FUCAM, Mexico. Biopsies were obtained from breast cancer patient after selection following the regulations approved by the FUCAM Ethics Committee, including patient informed consent for research use. None of the patients recruited in this study received anti-neoplastic therapy prior to surgery. After tumor resection, specimens were embedded in Tissue-Tek and snap-frozen in liquid nitrogen at −80°C until analysis. A pathologist analyzed samples for the amount of tumoral cells and to validate non-tumoral tissues. Tumor tissues containing at least 80% of tumor cells and non-tumoral tissues confirmed by the pathologist were used for subsequent analysis.

Cell line cultures

Human MDA-MB-231, MDA-MB-453, ZR-75, T47-D and MCF-7 breast cancer cell lines were cultured in Dulbecco's modified Eagle's medium (DMEM; Invitrogen, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 mg/ml streptomycin. Cultures were maintained in a 5% CO2 humidified atmosphere at 37°C.

RNA isolation and qRT-PCR analysis of miR-18b expression

Total RNA of tissues and cell lines was extracted using the TRIzol reagent (Invitrogen) according to the manufacturer's protocol. RNA integrity was assessed using capillary electrophoresis system Agilent 2100 Bioanalyzer with the eukaryotic nano-chip. Only samples with a RIN value of 6 or higher were processed. The expression of miR-18b was performed using microRNA assays (Applied Biosystems, Foster City, CA, USA). Total RNA (100 ng) obtained from clinical specimens and cell lines was reverse transcribed using a looped RT primer specific for miR-18b, 0.15 μl of dNTPs (100 mM), 1.0 μl of reverse transcriptase MultiScribe™ (50 U/μl), 1.5 μl of 10X buffer, 0.19 μl of RNase inhibitor (20 U/μl) and 4.16 μl of RNase-free water. Then, diluted retrotranscription reaction (1:15) was mixed with 10 μl of master mix TaqMan (Universal PCR Master mix, No AmpErase® UNG, 2X), 7.67 μl of RNase-free water, and 1.0 μl of probe PCR. PCR reaction was done in a GeneAmp System 9700 (Applied Biosystems) as follows: 95°C for 10 min, and 40 cycles at 95°C for 15 sec and 60°C for 1 min. The expression of miR-18b levels were measured by qRT-PCR using the comparative Ct (2−ΔΔCt) method. RNU44 and RNU48 were used as a control for normalization of data.

Transfection of the miR-18b inhibitor

MDA-MB-231 and MCF-7 cells were transfected with miR-18b inhibitor (#MH10466; Life Technologies) and scrambled at indicated final concentrations using siPORT™ amine transfection agent (Ambion, Inc., Austin, TX, USA). The miR-18b inhibitor was diluted in 25 μl of Opti-MEM® to 50–200 nM concentrations, and individually added to wells containing cultured cells in 450 μl of DMEM. Knock-down of miR-18b expression was evaluated by qRT-PCR at 48 h post-transfection.

DNA microarray analysis

To analyze the effects of miR-18b inhibition on gene expression we compared the gene expression profiles of MDA-MB-231 cells transfected with miR-18b inhibitor and with siPORT amine transfection agent (control) using DNA microarrays. Total RNA (1 μg) was employed for cDNA synthesis and labeling using Amino Allyl MessageAmp™ II aRNA Amplification kit (Ambion) following manufacturer's instructions. Total RNA obtained 48 h after transfection with empty transfection agent was labeled with Cy3, whereas cDNA from cells transfected with miR-18b inhibitor was Cy5 labeled. Dye incorporation efficiency was analyzed measuring specific absorbance. Samples (500 pmol) were hybridized onto Human Exonic Evidence Based Oligonucleotide (HEEBO) arrays obtained from the Stanford Functional Genomics Facility (Stanford, CA, USA). Each microarray contained 43,000 spots, including 85.8% I.M.A.G.E. consortium clones from the Research Genetics sequence verified clone set and 4.4% control spots, corresponding to 18,141 mapped human genes. Hybridization was performed at 42°C for 18 h. Following hybridization, arrays were washed and scanned using a GenePix 4100A Axon scanner (Axon Instruments, Inc., Union City, CA, USA). Fluorescence ratios were extracted using GenePix 5.0 software (Axon Instruments, Inc.). We defined well-measured spots by: i) having a ratio of normalized signal intensity to background noise of >2 for either the Cy5 signal derived from miR-18b inhibitor transfected cells, or the Cy3 signal derived from empty transfected cells and, ii) with no flag assignation by GenePix software which corresponds to poor spot quality caused by non-reliable hybridization and corruption by noise. We performed an algorithm on Microsoft Excel to accomplish both conditions. Background-subtracted fluorescence log2 ratios were globally normalized for each array, and then mean-centered for each gene. Each microarray experiment was repeated as technical replicates for statistical robustness. Finally, to avoid bias caused by different properties of the two cyanine dyes we balanced the intensity level of both channels using Lowess normalization method included on ArrayNorm software (16).

MTT cell viability assay

To evaluate the cell viability after inhibition of miR-18b in MDA-MB-231 and MCF-7 cells a standard 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) colorimetric assay was performed. Cells (2×104) transfected with miR-18b inhibitor (50 nM), scramble sequence, and siPORT transfection reagent as controls were seeded in 48-well culture dishes and incubated in MTT (1 mg/ml) at 37°C for 4 h. The medium was removed and formazan dye crystals were solubilized with 500 μl isopropanol, 4 mM HCl, NP-40 0.1% for 5 min. Absorbance was measured in a spectrophotometer at 540 nm wavelength. Assays were performed in triplicate.

Cell migration assays

To evaluate the effect of miR-18b inhibition in MDA-MB-231 and MCF-7 cell migration a scratch/wound-healing assay was performed. Briefly, cells transfected with the miR-18b inhibitor (50 nM), scramble sequence, and siPORT transfection reagent as controls were seeded in triplicate in a 6-well plate and grown to 80% confluence. Twenty-four hours post-transfection a vertical wound was traced in the cell monolayer with a sterile plastic tip. After 4 and 24 h cells were fixed with 4% paraformaldehyde in PBS pH 7.0 and monolayer restoration was imaged through an inverted phase contrast microscope (magnification, ×10). The scratched area was quantified at each time point and the resulting values were graphed. To quantify migratory capacity of cells, transwell chambers (Corning) with 6.5-mm diameter and 8-μm pore size polycarbonate membrane were used. MDA-MB-231 and MCF-7 cells (1×105) transfected with miR-18b inhibitor, scramble or siPORT transfection reagent (mock) were transferred to 0.5 ml serum-free medium and placed in the upper chamber, whereas the lower chamber was loaded with 0.8 ml medium containing 10% FBS. The total number of cells that migrated into the lower chamber was counted after 24 h incubation at 37°C.

Statistical analysis

Each experiment was performed at least three times, and the results are presented as the mean ± SD. One-way analysis of variance (ANOVA) followed by Tukey's test were used to compare the differences between means. P<0.05 was considered as statistically significant.

Results

miRNA-18b expression is upregulated in breast cancer cell lines and clinical tumors

In our previous studies, we analyzed a limited set of clinical breast tumors and identified 54 microRNAs that exhibit a significant differential expression between tumor and non-tumor tissues (our unpublished data). Of these, miR-18b was overexpressed in breast cancer and it was selected for further analysis. Here, we studied the miR-18b levels in an independent set of clinical specimens and in five human breast cancer cell lines. We extended our analysis by assaying the miR-18b expression using qRT-PCR TaqMan microRNA assays in clinical specimens obtained from 14 human breast cancer patients who did not receive anti-neoplastic therapy prior to surgery. Tumors were stratified based on clinical subtypes as luminal, HER2-positive and triple negative. Clinical features of breast tumors including hormonal receptors status, tumor size, histology, clinical stage, and tumor grade are summarized in Table I. Our results showed that miR-18b expression was increased in breast tumors in comparison to non-tumoral tissues (Fig. 1A). Particularly, the miR-18b expression average was augmented in luminal (2.5-fold), HER2-positive (4.6-fold), and triple negative (17-fold) tumor subtypes relative to non-tumoral specimens (Fig. 1B). We found no relationship between the expression of miR-18b and clinical characteristics of the patients such as tumor grade, clinical stage, tumor size or histological subtype. Then, we analyzed the miR-18b expression in breast cancer cell lines by qRT-PCR using TaqMan microRNA assays. Results showed that miR-18b levels were significantly increased (2- to 7-fold) in MDA-MB-231, MDA-MB-453, ZR-75 and T47-D cell lines in comparison to non-tumoral breast tissues used as control samples (Fig. 1C). In contrast, miR-18b expression was similar in MCF-7 cell line and normal breast tissues. Interestingly, the triple negative MDA-MB-231 cell line and triple negative breast tumors exhibit the highest miR-18b expression levels. We decided to use the MDA-MB-231 cell line as model for searching potential miR-18b targets.

Table I

Clinicopathological features of breast cancer patients and tumors analyzed for miR-18b expression.

Table I

Clinicopathological features of breast cancer patients and tumors analyzed for miR-18b expression.

Patient no.Age (years)Tumor size (mm)Clinical stageTumor gradeHER2ERPRClassificationHistological subtype
154535ND2++HER2-positiveInfiltrating ductal carcinoma
30412002+++Luminal BIn situ ductal carcinoma
755025IIB2+HER2-positiveInfiltrating ductal carcinoma
764920IIB3+HER2-positiveInfiltrating ductal carcinoma
788147IIIB3+HER2-positiveInfiltrating ductal carcinoma
795615IIA2+Luminal AInfiltrating ductal carcinoma
825925IIBND+Luminal AInfiltrating ductal carcinoma
854727IIA3++HER2-positiveInfiltrating ductal carcinoma
986530IIAND+Luminal AInfiltrating ductal carcinoma
1053923IIIC2++HER2-positiveInfiltrating ductal carcinoma
1394430IIB3Triple negativeInfiltrating ductal carcinoma
1696540IIBNDTriple negativeInfiltrating medular carcinoma
1915539IIB2+HER2-positiveInfiltrating ductal carcinoma

[i] ER, estrogen receptor; PR, progesterone receptor; HER2, human epidermal receptor 2; ND, not determined.

Transcriptional profiles of miR-18b-deficient MDA-MB-231 cells

In order to obtain a comprehensive analysis of cellular transcripts that may represent potential targets of miR-18b in triple negative breast cancer, we studied the effects of its inhibition on the transcriptome of MDA-MB-231 cells. We confirmed the effective knock-down of miR-18b expression by qRT-PCR assays using antagomiRs (data not shown). Total RNA was extracted from cells transfected with miR-18b inhibitor (50 nM) and siPORT amine transfection agent (control) cells, and hybridized to Human Gene Expression 12×135 K DNA microarrays (NimbleGen, Roche). We performed data extraction, normalization of replicates and controls, and selection of spots with a significant modulation (cut-off of ≥1.5-fold) as described in Materials and methods. Our microarray analysis identified 263 genes with a significant modulation (adjusted P-value = 0.05). Of these, 43 genes were downregulated and 220 were upregulated. These results indicated that repression of miR-18b had positive effects in global gene expression of MDA-MB-231 cells, as expected for a negative regulator of gene expression. In Tables II and III we show an overview of the top 20 modulated genes in miR-18b-deficient MDA-MB-231 cells.

Table II

Top 20 of upregulated genes in miR-18b-deficient human MDA-MB-231 breast cancer cells.

Table II

Top 20 of upregulated genes in miR-18b-deficient human MDA-MB-231 breast cancer cells.

Gene symbolaProtein namebFold changeP-value
OR8D2Olfactory receptor, family 8, subfamily D, member 27.100.039
OR8G2Olfactory receptor, family 8, subfamily G, member 26.440.011
OR8D1Olfactory receptor, family 8, subfamily D, member 16.360.017
REXO1L1REX1, RNA exonuclease 1 homolog-like 15.940.013
KRTAP13-1Keratin associated protein 13-15.870.013
ERC2 ELKS/RAB6-interacting/CAST family member 25.770.044
OR11H1Olfactory receptor, family 11, subfamily H, member 15.650.032
OR9G1Olfactory receptor, family 9, subfamily G, member 15.630.021
KIR3DL3Killer cell immunoglobulin-like receptor5.460.047
OR52E2Olfactory receptor, family 5, subfamily E, member 25.460.046
NLRP7NLR family, pyrin domain containing 75.330.018
OR52J3Olfactory receptor, family 52, subfamily J, member 35.330.013
KCND2Potassium voltage-gated channel5.330.040
PRAMEF5PRAME family member 55.320.031
REG1BRegenerating islet-derived 1 β5.260.014
OR4A47Olfactory receptor, family 4, subfamily A, member 475.190.006
OR8H3Olfactory receptor, family 8, subfamily H, member 35.180.041
LOC391747Similar to hCG1807616; TBP-associated factor 115.040.017
OR4A15Olfactory receptor, family 4, subfamily A, member 155.000.006
OR10G8Olfactory receptor, family 10, subfamily G, member 84.980.001

a GenBank databases;

b UniProt database (recommended name).

Table III

Top 20 of downregulated genes in miR-18b-deficient human MDA-MB-231 breast cancer cells.

Table III

Top 20 of downregulated genes in miR-18b-deficient human MDA-MB-231 breast cancer cells.

Gene symbolaProtein namebFold changeP-value
PELP1Proline, glutamate and leucine rich protein 1−8.6640.007
FBXW4F-box and WD repeat domain containing 4−5.5600.041
MR1Major histocompatibility complex, class I-related−4.160.044
MARCH1Membrane-associated ring finger (C3HC4) 1, E3 ubiquitin protein ligase−4.0650.042
MAKMale germ cell-associated kinase−4.0090.047
MAGED4BMelanoma-associated antigen D 4B−3.4510.043
EXOC6Exocyst complex component 6−3.0890.042
PLEKHH2Pleckstrin homology domain containing, family H member 2−2.9190.012
FAM113BFamily with sequence similarity 113, member B−2.7830.031
BAIAP2L2BAI1-associated protein 2-like 2−2.630.043
THAP9THAP domain containing 9−2.5470.009
KCNK7Potassium channel, subfamily K, member 7−2.4290.044
C17orf57Chromosome 17 open reading frame 57−2.3210.035
BEND2BEN domain containing 2−2.3000.016
TMEM27Transmembrane protein 27−2.2840.038
SOX4SRY (sex determining region Y)-box 4−2.2810.040
BCAT1Branched chain aminotransferase 1, cytosolic−2.200.031
TMEM19Transmembrane protein 19−2.1480.023
ADPGKADP-dependent glucokinase−2.120.032
SPNSialophorin−2.070.043

a GenBank databases;

b UniProt database (recommended name).

Olfactory receptor (OR) genes were upregulated in miR-18b- deficient breast cancer cells

Surprisingly, we evidenced the upregulation of 55 OR genes in MDA-MB-231 cells deficient for miR-18b (Table IV). The OR proteins are members of a large family of G-protein-coupled receptors which are expressed not only in the sensory neurons of the olfactory epithelium, but also in various other non-chemosensory tissues (17) and in tumor tissues including prostate cancer where their activation inhibits cell proliferation (18,19). However, the relevance and function of OR genes is still unknown in breast cancer. Here, we found that ten upregulated ORs genes were clustered in 11q12.1 chromosome region and six were grouped in 11q24.2 locus. To understand the coordinated upregulation of these genes, we searched for miR-18b binding sites in the 3′UTR of OR genes using miRanda software (20). Intriguingly, results showed that none of modulated OR genes presented potential miR-18b binding sites. This raises the possibility that OR gene expression could be regulated by a miR-18b-mediated indirect mechanism. To test this hypothesis, we searched for miR-18b binding sites in the 3′UTRs of transcription factors leading to activation of OR genes (21). Interestingly, we found that pancreatic and duodenal homeobox 1 (PDX1), a transcription factor regulating ORs, has potential binding sites for miR-18b. In agreement, we found that PDX1 gene was also up-regulated in our microarray assays (fold change 1.98; P=0.039). These data suggested that OR gene expression could be regulated through a miR-18b-indirect mechanism by targeting transcription factors leading to OR activation. However, further experiments are needed to support these assumptions.

Table IV

Upregulated olfactory receptor genes in miR-18b- deficient MDA-MB-231 breast cancer cells.

Table IV

Upregulated olfactory receptor genes in miR-18b- deficient MDA-MB-231 breast cancer cells.

Gene symbolFold changeP-valueLocus
OR8D27.100.03911q24.2
OR8G26.440.01111q24.2
OR8D16.360.01711q24.2
OR11H15.650.03222q11.1
OR9G15.630.02111q12.1
OR52E25.460.04611p15.4
OR52J35.330.01311p15.4
OR4A475.190.00611p11.2
OR8H35.180.04111q12.1
OR4A155.000.00611q11
OR10G84.980.00111q24.2
OR2T84.970.0351q44
OR4K24.920.04914q11.2
OR1C14.710.0361q44
OR5J24.560.03611q12.1
OR1S14.490.04011q12.1
OR4Q34.440.00114q11.2
OR10H44.360.05019p13.12
OR10H34.240.01419p13.12
OR5D164.230.02011q11
OR8H24.200.00711q12.1
OR6K24.170.0151q23.1
OR6N14.150.0251q23.1
OR7C24.120.01019p13.12
OR10K23.950.0381q23.1
OR4C133.950.00411p11.12
OR56A43.930.02611p15.4
OR2T273.890.0361q44
OR52K23.840.02211p15.4
OR1E13.770.01617p13.2
OR2J33.690.0196p22.1
OR6N23.660.0431q23.1
OR8B33.450.04611q24.2
OR4F173.360.02919p13.3
OR8H13.490.01111q12.1
OR10Q13.350.02311q12.1
OR5M83.310.01711q12.1
OR5M112.690.01011q12.1
OR5AR12.740.02811q12.1
OR14I13.310.0401q44
OR1J43.230.0269q33.2
OR5AU13.110.03414q11.2
OR2T293.000.0041q44
OR8B82.400.00911q24.2
OR4C462.200.00311p11.12
OR6K32.160.0011q23.1
OR2T52.100.0151q44
OR10S12.090.04211q24.1
OR3A22.020.02017p13.3
OR2J21.980.0366p22.1
OR2AK21.880.0351q44
OR5D141.820.04411q11
OR1L61.790.0259q33.2
OR13C51.600.0289q31.1
OR6Y11.520.0351q23.1

[i] Genes clustered in 11q24.2 chromosomal region are underlined. Genes clustered in 11q12.1 chromosomal region are shown in bold.

miR-18b modulates genes involved in tumorigenesis

Further analysis of microarray data leads us to the identification of diverse genes involved in cancer whose expression was modulated when miR-18b expression was inhibited in breast cancer cells. In Table V we show an overview of upregulated genes which have been reported previously in different malignancies. These genes have been implicated in tumor growth (CHRM2 and POSTN), cell proliferation (NLRP7 and CHMR2), anoikis (OLFM3), apoptosis (REG1B, SCN3B and POSTN), and angiogenesis (KLK3, CHRM2 and POSTN). Interestingly, eight genes (KIR3DL3, NLRP7, KLK3, OLFM3, SEMG1, CRX, POSTN and CEACAM5) have been shown to be involved in invasion and metastasis of cancer cells. To determine whether the overexpression of these genes was related to direct binding of miR-18b, we searched for potential miR-18b binding sites in their 3′UTR region. Eleven genes were predicted to have binding sites for miR-18b, nine of them with one site and two with more than one binding site. Among these, four genes (KIR3DL3, KLK3, CRX and POSTN) were implicated in invasion and metastasis of cancer cells. For instance, the KLK3 gene encodes prostate-specific antigen (PSA), which is a serin protease that has been established as a tumor marker of prostatic adenocarcinoma. This protein was found elevated in women with renal cell carcinoma (22), it has been shown to exert anti-angiogenic properties, and to present mutations in breast tumors (23). Another upregulated gene, POSTN encodes periostin, a secretory protein which has been suggested to function as a cell adhesion molecule participating in osteoblast recruitment, attachment and spreading (24,25). POSTN was found overexpressed in several cancers with enhanced invasiveness, and correlated with metastasis in colorectal and liver cancers, as well in oral cancer cell lines (26).

Table V

The genes modulated by miR-18b inhibition with a role in human cancers.

Table V

The genes modulated by miR-18b inhibition with a role in human cancers.

Gene symbolaFold changeP-valueDescriptionGene function related to cancerRefs.miR-18b binding sitesb
ERC25.770.044 ELKS/RAB6-interacting/CAST family member 2Promotes oncogenesis in papillary thyroid carcinoma by fusion of ELKS to RET.(40)1
KIR3DL35.460.047Killer cell immunoglobulin-like receptorInteractions with MHC-I on melanoma inhibit tumor cells lysis by NK and T cells. Interactions between KIR and HLA in NK cell-controls uveal melanoma metastasis.(41,42)1
NLRP75.330.018NLR family, pyrin domain containing 7Cell proliferation and tumorigenesis in testicular germ cell tumor. Promote tumor invasion and poor prognosis in endometrial cancer.(43,44)None
KCND25.330.040Potassium voltage-gated channel subfamily D2Participates in brain cancer-induced cachexia.(45)1
REG1B5.260.014Regenerating islet-derived 1 βRegulates IL-6 and enhance the chemo- and radio-sensitivity on squamous esophageal cancer.(46,47)1
KIR2DS24.850.020Killer cell immunoglobulin-like receptorAssociated with better response to treatment and prolonged survival in non-small cell lung carcinoma. Activated KIR genes are associated with reduced risk for developing acute lymphoblastic leukemia.(48,49)None
KLK34.520.013Prostate-specific antigen (PSA). Kallikrein-related peptidase 3Contribute to bone metastasis in prostate cancer. Modulates cell invasion of prostate cancer cells. High levels of PSA may slow down prostate cancer progression by inhibiting angiogenesis. Regulates androgen receptor in prostate cancer. Decreased expression in ovarian tumors. Favorable indicator of prognosis in breast cancer.(5054)1
PRAMEF44.490.012PRAME family member 4Correlates with poor prognosis in head and neck cancer.(55)None
CHRM24.470.005Muscarinic acetylcholine receptor M2Involved in proliferation of murine adenocarcinoma cell lines. Involved in growth, proliferation and angiogenesis in breast cancer cells. Involved in inhibition of glioma cell proliferation. Involved in cell proliferation of chronic myelogenous leukemia.(5660)1
OLFM34.440.037Olfactomedin 3/noelin-3Contributes to anoikis-resistance in squamous lung carcinoma. Olfml3 targeting inhibits endothelioma cell migration, tumor growth and sprouting. OLFM3 is pro-angiogenic.(61,62)None
PCDH154.120.014Protocadherin 15Potential marker for NK/T cell lymphomas.(63)1
SCN3B3.960.029Sodium channel subunit β-3Mediates a p53-dependent apoptotic pathway (pro-apoptotic) in colorectal carcinoma cell line HCT116.(64)1
SEMG13.860.005Semenogelin IOverexpressed in OH-1 SCLC lung cancer cell lines and associated with cell surface adhesion complexes. Mutated in colorectal cancer. Inhibits motility of sperm cells. SEMG1 inhibits prostate-tumor growth.(6569)None
CRX3.820.012Cone-rod homeobox proteinOverexpressed in medulloblastomas. Upregulated in retinoblastoma tumors. Metastatic retinoblastoma marker.(7072)2
KLK153.780.003Kallikrein-related peptidase 15Upregulated by steroid hormones in the LNCaP prostate cancer cell line; associated with more aggressive forms of prostate cancer. Marker of unfavorable prognosis in ovarian cancer. Prognostic marker for progression-free survival in patients with radical prostatectomy. Favorable prognostic marker for breast cancer.(7376)None
POSTN3.770.043PeriostinEnhances invasion, angiogenesis and metastasis in oral cancer and head and neck squamous cell carcinoma. Promotes the metastatic growth of colon cancer. It is required for cancer stem cell maintenance, and blocking its function prevents metastasis. Reduces cell migration and suppressed metastasis in vivo. Inhibits breast cancer progression and metastasis by anti-periostin in a murine model.(7780)1
PCDHB22.390.019Protocadherin beta 2Mutation of PCDHB2 (cell adhesion protein) in pancreatic cancers.(81)None
C1QTNF42.160.010C1q and tumor necrosis factor related protein 4Stimulates the STAT3 and NF-κB pathways and promotes cell survival in human cancer cells.(82)None
FOXD4L41.950.010Forkhead box D4-like 4FOX4DL4 gene amplification in pilocytic astrocytoma. Hypermethylated in ovarian tumors.(8385)3
CEACAM51.780.034Carcinoembryonic antigen-related cell adhesion molecule 5Methylated in non-small cell lung cancer tumors. Implied in bad prognosis in patients with adenocarcinoma. Overexpression enhances metastasis in coloncarcinoma. Brain metastasis development and poor survival associated with CEA level in NSCLC.(8688)None

a GenBank databases.

b As prediceted by TargetScan. Cell migration and invasion-related genes are denoted in bold.

Knockdown of miR-18b suppresses cell migration in vitro

Our transcriptional profiling of MDA-MB-231 cells with reduced miR-18b expression revealed that a set of modulated genes have key roles in migration and invasion of cancer cells. In order to evaluate whether miR-18b could be implicated in the regulation of migration properties of breast cancer cells, we performed scratch/wound-healing and Transwell assays in MDA-MB-231 cells with reduced expression of miR-18b. We first evaluated the effect of increasing concentrations of antagomiR-18b in cell viability by MTT assay. No significant changes in cell viability were observed using a range of concentrations (25–200 mM) of miR-18b inhibitor in metastatic MDA-MB-231 cells (Fig. 2A). Similar results were obtained in the MCF-7 breast cancer cell line (Fig. 2E). Then, we performed a scratch/wound-healing assay to evaluate whether the suppression of miR-18b expression could have effects on cancer cell migration. Monolayers of MDA-MB-231 and MCF-7 cells non-transfected and transfected with miR-18b inhibitor were grated, and the wounded areas of the cell monolayer were analyzed after 24 h. Results showed that the formation of monolayers was significantly delayed about 70 and 30% in MDA-MB-231 and MCF-7 cells transfected with miR-18b inhibitor, respectively, in comparison with control cells (Fig. 2B and F). By using Transwell chamber assays, we found that the number of migratory cells that crossed the membrane after 4 and 24 h was reduced in MDA-MB-231 (75%) and MCF-7 (40%) cells in comparison with controls confirming that miR-18b abolishment has negative effects in cell migration (Fig. 2D and H). These findings suggested that miR-18b is able to modulate the expression of genes that are important for migration of cancer cells in vitro.

Discussion

microRNA expression is frequently deregulated in breast cancer where they play key roles in tumorigenesis acting as oncogenes and tumor suppressors. In this study, we showed that miR-18b is commonly overexpressed in breast cancer cell lines and in clinical tumors, particularly in triple negative clinical specimens (17-fold). We found an inverse correlation between miR-18b expression and hormonal receptors and HER2/neu status in breast cell lines and tumors (Fig. 1). These data are in agreement with a previous study which reports that ER-α is a target of miR-18a and miR-18b suggesting that inhibition of this receptor in triple negative breast cancer may be due, at least in part, by miR-18b (13). Moreover, in a study of 204 lymph node-negative breast cancers, authors found an association among higher expression of miR-18b with negative expression of ER and strongly associated with basal-like breast cancer features (27), in agreement with our results. We showed that miR-18b is overexpressed in triple negative breast tumors. Triple negative breast cancer refers to a heterogeneous group of tumors that do not express estrogen, progesterone or HER2/neu receptors, accounting for approximately 12–15% of breast cancer diagnoses (28). It represents the most aggressive subtype of breast cancer, and is associated with high rates of mortality and poor survival. Notably, no specific therapeutics targets have been identified in triple negative tumors, thus treatment is restricted to conventional chemotherapy. Therefore, the search for novel potential therapeutic targets represents today a challenge for oncology community.

In order to identify potential targets of miR-18b that could represent novel therapeutic targets in triple negative breast cancer, we suppressed its expression in MDA-MB-231 cells using antagomiRs and performed a genome-wide analysis using DNA microarrays. Our analysis showed that several genes with key roles in cancer were significantly modulated (Table V). In addition, a set of 55 OR genes were also found as regulated in miR-18b-deficient cells. The OR gene family is the largest family in the genome with more than 800 genes located in clusters that are dispersed across almost all chromosomes. Although the majority of OR members are expressed on the olfactory sensory neurons in the olfactory epithelium, they are also expressed in a large number of different tissues, but their functions remain unclear. It has been suggested that they may control cell positioning during embryogenesis (29). Of interest, the expression of ORs has been also reported in cancer. Previous studies detected the expression of ORs in human primordial germ cells and prostate cancer (30,31). In a recent study it was reported that the activation of an OR (PSGR/OR51E2) inhibits proliferation of prostate cancer cells (18). In addition, four microRNA:mRNA modules containing the olfactory transduction pathway, are over-represented in metastatic prostate cancer (19). Most of the 55 upregulated ORs detected in this study were clustered in 11q12.1 and 11q24.2 chromosome regions, suggesting that a gain or loss of these genomic regions could explain the changes in their expression. The 11q12.1 locus is defined as an overlapping region of imbalance involved in testicular germ cell tumors (32), whereas homozygous loss of 11q24.2 region was observed in uterine leiomyosarcoma (33). OR genes located on 11q12.1 have been already reported as overexpressed in breast tumors. Of a set of 862 differentially expressed genes, 34 ORs were found to have elevated expression in breast tumors with CHEK2 1100delC mutation, where 16 ORs resided on chromosome 11, and 5 in a region flanking the centromere region 11.p11.2–11.q12.1 (34). Our results on the identification of OR genes modulated in miR-18b-deficient cells indicate that they could be involved in tumorigenesis, however additional experiments are needed to clarify their role in breast cancer. The absence for potential miR-18b binding sites in the OR genes suggested that they could be modulated by an miR-18b-driven indirect mechanism. Plessy et al(21) found binding sites for several transcription factors on OR promoters that may control their transcription. In particular, a binding site motif for PDX1 homeobox transcription factor was found in the promoter of mouse and human olfactory epithelium (MOE). PDX1 participates during branching morphogenesis in the developing mouse embryonic pancreas (35). In cancer, PDX1 is activated at various stages of gastric carcinogenesis (36), and it was suggested as a novel tumor suppressor in gastric cancer (37). It was also involved in pancreas development, in pediatric solid pseudopapillary tumors (38), and in precursor lesions of ductal adenocarcinomas (39). In our study, we found that PDX1 was upregulated after inhibition of miR-18b. From these findings, we suggested that miR-18b could indirectly regulate OR genes by direct modulation of PDX1, however, additional experimentation is needed to corroborate this hypothesis.

We have shown that the inhibition of miR-18b induces the modulation of genes with key roles in cancer. These genes have been implicated in tumor growth (CHRM2 and POSTN), cell proliferation (NLRP7 and CHMR2), anoikis (OLFM3), apoptosis (REG1B, SCN3B and POSTN), and angiogenesis (KLK3, CHRM2 and POSTN). Notably, eight genes (KIR3DL3, NLRP7, KLK3, OLFM3, SEMG1, CRX, POSTN and CEACAM5) are involved in invasion and metastasis of cancer cells (Table V). Moreover, these genes contain potential binding sites for miR-18b which strongly suggested that they could be targets of miR-18b. Our findings using wound-healing assays in miR-18b depleted cells showed that cell migration was significantly affected. By Transwell chamber assay, we corroborated the impact of miR-18b inhibition on the capacity of migration in MDA-MB-231 and MCF-7 cells. In both assays, results showed that the formation of monolayer was delayed about 40–75% in miR-18b-deficient cells in comparison to control migrating cells. Taken together, our findings strengthened the view that miR-18b is able to modulate the expression of genes that are important for migration of triple negative breast cancer cells in vitro.

Acknowledgements

We thank the Autonomous University of Mexico City and the Instituto de Ciencia y Tecnología del Distrito Federal, Mexico (grants ICyTDF/328/2011 and PIUTE10-147) for support. This study was supported by CONACyT FONSEC SALUD 115306 and 115591 grants. Miguel A. Fonseca-Sánchez and Ali Flores-Pérez were supported by ICYT-DF (179/2011). Zaira E. Arellano-Anaya was supported by a postdoctoral fellowship from CONACyT.

References

1 

Hammond SM: RNAi, microRNAs, and human disease. Cancer Chemother Pharmacol. 58:S63–S68. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Tanzer A and Stadler PF: Molecular evolution of a microRNA cluster. J Mol Biol. 339:327–335. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Mendell JT: miRiad roles for the miR-17-92 cluster in development and disease. Cell. 133:217–222. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Ventura A, Young AG, Winslow MM, Lintault L, Meissner A, Erkeland SJ, Newman J, Bronson RT, Crowley D, Stone JR, Jaenisch R, Sharp PA and Jacks T: Targeted deletion reveals essential and overlapping functions of the miR-17 through 92 family of miRNA clusters. Cell. 132:875–886. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Tatsuguchi M, Seok HY, Callis TE, Thomson JM, Chen JF, Newman M, Rojas M, Hammond SM and Wang DZ: Expression of microRNAs is dynamically regulated during cardiomyocyte hypertrophy. J Mol Cell Cardiol. 42:1137–1141. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Otaegui D, Baranzini SE, Armañanzas R, Calvo B, Muñoz-Culla M, Khankhanian P, Inza I, Lozano JA, Castillo-Triviño T, Asensio A, Olaskoaga J and López de Munain A: Differential micro RNA expression in PBMC from múltiple sclerosis patients. PloS One. 4:e63092009.PubMed/NCBI

7 

Zhang ZZ, Liu X, Wang DQ, Teng MK, Niu LW, Huang AL and Liang Z: Hepatitis B virus and hepatocellular carcinoma at the miRNA level. World J Gastroenterol. 17:3353–3358. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Guo J, Miao Y, Xiao B, Huan R, Jiang Z, Meng D and Wang Y: Differential expression of microRNA species in human gastric cancer versus non-tumorous tissues. J Gastroenterol Hepatol. 24:652–657. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Kim TH, Kim YK, Kwon Y, Heo JH, Kang H, Kim G and An HJ: Deregulation of miR-519a, 153, and 485-5p and its clinicopathological relevance in ovarian epithelial tumours. Histopathology. 57:734–743. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Sand M, Skrygan M, Sand D, Georgas D, Hahn SA, Gambichler T, Altmeyer P and Bechara FG: Expression of microRNAs in basal cell carcinoma. Br J Dermatol. 167:847–855. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Wang YX, Zhang XY, Zhang BF, Yang CQ, Chen XM and Gao HJ: Initial study of microRNA expression profiles of colonic cancer without lymph node metastasis. J Dig Dis. 11:50–54. 2010. View Article : Google Scholar : PubMed/NCBI

12 

He X, DiMeco F, Vescovi A, Heth J, Muraszko K and Fan X: MIR-18b regulates glioblastoma neurosphere growth through NOTCH2, NEDD9, and MEKK1. Neuro Oncol. 14(Suppl 6): vi142–vi152. 2012.

13 

Leivonen SK, Mäkelä R, Ostling P, Kohonen P, Haapa-Paananen S, Kleivi K, Enerly E, Aakula A, Hellström K, Sahlberg N, Kristensen VN, Børresen-Dale AL, Saviranta P, Perälä M and Kallioniemi O: Protein lysate microarray analysis to identify microRNAs regulating estrogen receptor signaling in breast cancer cell lines. Oncogene. 28:3926–3936. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Yoshimoto N, Toyama T, Takahashi S, Sugiura H, Endo Y, Iwasa M, Fujii Y and Yamashita H: Distinct expressions of microRNAs that directly target estrogen receptor α in human breast cancer. Breast Cancer Res Treat. 130:331–339. 2011.PubMed/NCBI

15 

Cookson VJ, Bentley MA, Hogan BV, Horgan K, Hayward BE, Hazelwood LD and Hughes TA: Circulating microRNA profiles reflect the presence of breast tumours but not the profiles of microRNA within the tumours. Cell Oncol. 35:301–308. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Simon A and Biot E: ANAIS: Analysis of NimbleGen arrays interface. Bioinformatics. 26:2468–2469. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Kang N and Koo J: Olfactory receptors in non-chemosensory tissues. BMB Rep. 5:612–622. 2012. View Article : Google Scholar

18 

Neuhaus EM, Zhang W, Gelis L, Deng Y, Noldus J and Hatt H: Activation of an olfactory receptor inhibits proliferation of prostate cancer cells. J Biol Chem. 284:16218–16225. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Zhang W, Edwards A, Fan W, Flemington EK and Zhang K: miRNA-mRNA correlation-network modules in human prostate cancer and the differences between primary and metastatic tumor subtypes. PLoS One. 7:e401302012. View Article : Google Scholar : PubMed/NCBI

20 

Betel D, Wilson M, Gabow A, Marks DS and Sander C: The microRNA.org resource: targets and expression. Nucleic Acids Res. 36:149–153. 2008.PubMed/NCBI

21 

Plessy C, Pascarella G, Bertin N, Akalin A, Carrieri C, Vassalli A, Lazarevic D, Severin J, Vlachouli C, Simone R, Faulkner GJ, Kawai J, Daub CO, Zucchelli S, Hayashizaki Y, Mombaerts P, Lenhard B, Gustincich S and Carninci P: Promoter architecture of mouse olfactory receptor genes. Genome Res. 22:486–497. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Clements J, Ward G, Kaushal A, Hi SI, Kennett C and Nicol D: A prostate-specific antigen-like protein associated with renal cell carcinoma in women. Clin Cancer Res. 3:1427–1431. 1997.PubMed/NCBI

23 

Majumdar S and Diamandis EP: The promoter and the enhancer region of the KLK3 (prostate specific antigen) gene is frequently mutated in breast tumours and in breast carcinoma cell lines. Br J Cancer. 79:1594–1602. 1999. View Article : Google Scholar : PubMed/NCBI

24 

Takeshita S, Kikuno R, Tezuka K and Amann E: Osteoblast-specific factor 2: cloning of a putative bone adhesion protein with homology with the insect protein fasciclin I. Biochem J. 15:271–278. 1993.PubMed/NCBI

25 

Horiuchi K, Amizuka N, Takeshita S, Takamatsu H, Katsuura M, Ozawa H, Toyama Y, Bonewald LF and Kudo A: Identification and characterization of a novel protein, periostin, with restricted expression to periosteum and periodontal ligament and increased expression by transforming growth factor beta. J Bone Miner Res. 14:1239–1249. 1999. View Article : Google Scholar : PubMed/NCBI

26 

Siriwardena BS, Kudo Y, Ogawa I, Kitagawa M, Kitajima S, Hatano H, Tilakaratne WM, Miyauchi M and Takata T: Periostin is frequently overexpressed and enhances invasion and angiogenesis in oral cancer. Br J Cancer. 95:1396–1403. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Jonsdottir K, Janssen SR, Da Rosa FC, Gudlaugsson E, Skaland I, Baak JP and Janssen EA: Validation of expression patterns for nine miRNAs in 204 lymph-node negative breast cancers. PLoS One. 7:e486922012. View Article : Google Scholar : PubMed/NCBI

28 

Fornier M and Fumoleau P: The paradox of triple negative breast cancer: novel approaches to treatment. Breast J. 1:41–51. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Dreyer WJ: The area code hypothesis revisited: olfactory receptors and other related transmembrane receptors may function as the last digits in a cell surface code for assembling embryos. Proc Natl Acad Sci USA. 95:9072–9077. 1998. View Article : Google Scholar

30 

Goto T, Salpekar A and Monk M: Expression of a testis-specific member of the olfactory receptor gene family in human primordial germ cells. Mol Hum Reprod. 7:553–558. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Weng J, Wang J, Hu X, Wang F, Ittmann M and Liu M: PSGR2, a novel G-protein coupled receptor, is overexpressed in human prostate cancer. Int J Cancer. 118:1471–1480. 2006. View Article : Google Scholar : PubMed/NCBI

32 

McIntyre A, Summersgill B, Jafer O, Rodriguez S, Zafarana G, Oosterhuis JW, Gillis AJ, Looijenga L, Cooper C, Huddart R, Clark J and Shipley J: Defining minimum genomic regions of imbalance involved in testicular germ cell tumors of adolescents and adults through genome wide microarray analysis of cDNA clones. Oncogene. 23:9142–9147. 2004. View Article : Google Scholar : PubMed/NCBI

33 

Raish M, Khurshid M, Ansari MA, Chaturvedi PK, Bae SM, Kim JH, Park EK, Park DC and Ahn WS: Analysis of molecular cytogenetic alterations in uterine leiomyosarcoma by array-based comparative genomic hybridization. J Cancer Res Clin Oncol. 138:1173–1186. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Muranen TA, Greco D, Fagerholm R, et al: Breast tumors from CHEK2 110delC-mutation carriers: genomic landscape and clinical implications. Breast Cancer Res. 13:R902011. View Article : Google Scholar : PubMed/NCBI

35 

Wescott MP, Rovira M, Reichert M, von Burstin J, Means A, Leach SD and Rustgi AK: Pancreatic ductal morphogenesis and the Pdx1 homeodomain transcription factor. Mol Biol Cell. 20:4838–4844. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Sakai H, Eishi Y, Li XL, Akiyama Y, Miyake S, Takizawa T, Konishi N, Tatematsu M, Koike M and Yuasa Y: PDX1 homeobox protein expression in pseudopyloric glands and gastric carcinomas. Gut. 53:323–330. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Ma J, Chen M, Wang J, Xia HH, Zhu S, Liang Y, Gu Q, Qiao L, Dai Y, Zou B, Li Z, Zhang Y, Lan H and Wong BC: Pancreatic duodenal homeobox-1 (PDX1) functions as a tumor suppressor in gastric cancer. Carcinogenesis. 29:1327–1333. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Galmiche L, Sarnacki S, Verkarre V, Boizet B, Duvillie B, Fabre M and Jaubert F: Transcription factors involved in pancreas development are expressed in paediatric solid pseudopapillary tumours. Histopathology. 53:318–324. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Park JY, Hong SM, Klimstra DS, Goggins MG, Maitra A and Hruban RH: Pdx1 expression in pancreatic precursor lesions and neoplasms. Appl Immunohistochem Mol Morphol. 19:444–449. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Nakata T, Kitamura Y, Shimizu K, Tanaka S, Fujimori M, Yokoyama S, Ito K and Emi M: Fusion of a novel gene, ELKS, to RET due to translocation t(10;12)(q11;p13) in a papillary thyroid carcinoma. Genes Chromosomes Cancer. 25:97–103. 1999. View Article : Google Scholar : PubMed/NCBI

41 

Bakker AB, Phillips JH, Figdor CG and Lanier LL: Killer cell inhibitory receptors for MHC class I molecules regulate lysis of melanoma cells mediated by NK cells, gamma delta T cells, and antigen-specific CTL. J Immunol. 160:5239–5245. 1998.PubMed/NCBI

42 

Maat W, van der Slik AR, Verhoeven DH, Alizadeh BZ, Ly LV, Verduijn W, Luyten GP, Mulder A, van Hall T, Koning F, Jager MJ and van Bergen J: Evidence for natural killer cell-mediated protection from metastasis formation in uveal melanoma patients. Invest Ophthalmol Vis Sci. 50:2888–2895. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Okada K, Hirota E, Mizutani Y, Fujioka T, Shuin T, Miki T, Nakamura Y and Katagiri T: Oncogenic role of NALP7 in testicular seminomas. Cancer Sci. 95:949–954. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Ohno S, Kinoshita T, Ohno Y, Minamoto T, Suzuki N, Inoue M and Suda T: Expression of NLRP7 (PYPAF3, NALP7) protein in endometrial cancer tissues. Anticancer Res. 28:2493–2498. 2008.PubMed/NCBI

45 

Coma M, Vicente R, Busquets S, Carbó N, Tamkun MM, López-Soriano FJ, Argilés JM and Felipe A: Impaired voltage-gated K+ channel expression in brain during experimental cancer cachexia. FEBS Lett. 536:45–50. 2003.

46 

Usami S, Motoyama S, Koyota S, Wang J, Hayashi-Shibuya K, Maruyama K, Takahashi N, Saito H, Minamiya Y, Takasawa S, Ogawa J and Sugiyama T: Regenerating gene I regulates interleukin-6 production in squamous esophageal cancer cells. Biochem Biophys Res Commun. 29:4–8. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Hayashi K, Motoyama S, Koyota S, Koizumi Y, Wang J, Takasawa S, Itaya-Hironaka A, Sakuramoto-Tsuchida S, Maruyama K, Saito H, Minamiya Y, Ogawa J and Sugiyama T: REG I enhances chemo- and radiosensitivity in squamous cell esophageal cancer cells. Cancer Sci. 99:2491–2495. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Wiśniewski A, Jankowska R, Passowicz-Muszyńska E, Wiśniewska E, Majorczyk E, Nowak I, Frydecka I and Kuśnierczyk P: KIR2DL2/S2 and HLA-C C1C1 genotype is associated with better response to treatment and prolonged survival of patients with non-small cell lung cancer in a Polish Caucasian population. Hum Immunol. 73:927–931. 2012.PubMed/NCBI

49 

Almalte Z, Samarani S, Iannello A, Debbeche O, Duval M, Infante-Rivard C, Amre DK, Sinnett D and Ahmad A: Novel associations between activating killer-cell immunoglobulin-like receptor genes and childhood leukemia. Blood. 118:1323–1328. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Cumming AP, Hopmans SN, Vukmirović-Popović S and Duivenvoorden WC: PSA affects prostate cancer cell invasion in vitro and induces an osteoblastic phenotype in bone in vivo. Prostate Cancer Prostatic Dis. 14:286–294. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Mattsson JM, Närvänen A, Stenman UH and Koistinen H: Peptides binding to prostate-specific antigen enhances its antiangiogenic activity. Prostate. 72:1588–1594. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Saxena P, Trerotola M, Wang T, Li J, Sayeed A, Vanoudenhove J, Adams DS, Fitzgerald TJ, Altieri DC and Languino LR: PSA regulates androgen receptor expression in prostate cancer cells. Prostate. 72:769–776. 2012. View Article : Google Scholar : PubMed/NCBI

53 

O'Mara TA, Nagle CM, Batra J, Kedda MA, Clements JA and Spurdle AB: Kallikrein-related peptidase 3 (KLK3/PSA) single nucleotide polymorphisms and ovarían cancer survival. Twin Res Hum Genet. 14:323–327. 2011.PubMed/NCBI

54 

Black MH and Diamandis EP: The diagnostic and prognostic utility of prostate-specific antigen for diseases of the breast. Breast Cancer Res Treat. 59:1–14. 2000. View Article : Google Scholar : PubMed/NCBI

55 

Szczepanski MJ, DeLeo AB, Łuczak M, Molinska-Glura M, Misiak J, Szarzynska B, Dworacki G, Zagor M, Rozwadowska N, Kurpisz M, Krzeski A, Kruk-Zagajewska A, Kopec T, Banaszewski J and Whiteside TL: PRAME expression in head and neck cancer correlates with markers of poor prognosis and might help in selecting candidates for retinoid chemoprevention in pre-malignant lesions. Oral Oncol. 49:144–151. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Español AJ and Sales MN: Different muscarinic receptors are involved in the proliferation of murine mammary adenocarcinoma cell lines. Int J Mol Med. 13:311–317. 2004.PubMed/NCBI

57 

Fiszman GL, Middonno MC, de la Torre E, Farina M, Español AJ and Sales ME: Activation of muscarinic cholinergic receptors induces MCF-7 cells proliferation and angiogenesis by stimulating nitric oxide synthase activity. Cancer Biol Ther. 6:1106–1113. 2007. View Article : Google Scholar

58 

Español AJ, de la Torre E, Fiszman GL and Sales ME: Role of non-neuronal cholinergic system in breast cancer progression. Life Sci. 80:2281–2285. 2007.PubMed/NCBI

59 

Ferretti M, Fabbiano C, Di Bari M, Ponti D, Calogero A and Tata AM: M2 muscarinic receptors inhibit cell proliferation in human glioblastoma cell lines. Life Sci. 91:1134–1137. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Cabadak H, Aydin B and Kan B: Regulation of M2, M3, and M4 muscarinic receptor expression in K562 chronic myelogenous leukemic cells by carbachol. J Recept Signal Transduct Res. 31:26–32. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Keenan J, Joyce H, Aherne S, O'Dea S, Doolan P, Lynch V and Clynes M: Olfactomedin III expression contributes to anoikis-resistance in clonal variants of a human lung squamous carcinoma cell line. Exp Cell Res. 318:593–602. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Miljkovic-Licina M, Hammel P, Garrido-Urbani S, Lee BP, Meguenani M, Chaabane C, Bochaton-Piallat ML and Imhof BA: Targeting olfactomedin-like 3 inhibits tumor growth by impairing angiogenesis and pericyte coverage. Mol Cancer Ther. 11:2588–2599. 2012. View Article : Google Scholar : PubMed/NCBI

63 

Rouget-Quermalet V, Giustiniani J, Marie-Cardine A, Beaud G, Besnard F, Loyaux D, Ferrara P, Leroy K, Shimizu N, Gaulard P, Bensussan A and Schmitt C: Protocadherin 15 (PCDH15): a new secreted isoform and a potential marker for NK/T cell lymphomas. Oncogene. 25:2807–2811. 2006. View Article : Google Scholar : PubMed/NCBI

64 

Adachi K, Toyota M, Sasaki Y, Yamashita T, Ishida S, Ohe-Toyota M, Maruyama R, Hinoda Y, Saito T, Imai K, Kudo R and Tokino T: Identification of SCN3B as a novel p53-inducible proapoptotic gene. Oncogene. 23:7791–7798. 2004. View Article : Google Scholar : PubMed/NCBI

65 

Rodrigues RG, Panizo-Santos A, Cashel JA, Krutzsch HC, Merino MJ and Roberts DD: Semenogelins are ectopically expressed in small cell lung carcinoma. Clin Cancer Res. 7:854–860. 2001.PubMed/NCBI

66 

Zhang Y, Wang Z, Zhang J, Farmer B and Lim SH: Semenogelin I expression in myeloma cells can be upregulated pharmacologically. Leuk Res. 32:1889–1894. 2008. View Article : Google Scholar : PubMed/NCBI

67 

Hienonen T, Sammalkorpi H, Enholm S, Alhopuro P, Barber TD, Lehtonen R, Nupponen NN, Lehtonen H, Salovaara R, Mecklin JP, Järvinen H, Koistinen R, Arango D, Launonen V, Vogelstein B, Karhu A and Aaltonen LA: Mutations in two short noncoding mononucleotide repeats in most microsatellite-unstable colorectal cancers. Cancer Res. 65:4607–4613. 2005. View Article : Google Scholar : PubMed/NCBI

68 

Mitra A, Richardson RT and O'Rand MG: Analysis of recombinant human semenogelin as an inhibitor of human sperm motility. Biol Reprod. 82:489–496. 2010. View Article : Google Scholar : PubMed/NCBI

69 

Canacci AM, Izumi K, Zheng Y, Gordetsky J, Yao JL and Miyamoto H: Expression of semenogelins I and II and its prognostic significance in human prostate cancer. Prostate. 71:1108–1114. 2011. View Article : Google Scholar : PubMed/NCBI

70 

Santagata S, Maire CL, Idbaih A, Geffers L, Correll M, Holton K, Quackenbush J and Ligon KL: CRX is a diagnostic marker of retinal and pineal lineage tumors. PLoS One. 4:e79322009. View Article : Google Scholar : PubMed/NCBI

71 

Glubrecht DD, Kim JH, Russell L, Bamforth JS and Godbout R: Differential CRX and OTX2 expression in human retina and retinoblastoma. J Neurochem. 111:250–263. 2009. View Article : Google Scholar : PubMed/NCBI

72 

Terry J, Calicchio ML, Rodriguez-Galindo C and Perez-Atayde AR: Immunohistochemical expression of CRX in extracranial malignant small round cell tumors. Am J Surg Pathol. 36:1165–1169. 2012. View Article : Google Scholar : PubMed/NCBI

73 

Yousef GM, Scorilas A, Jung K, Ashworth LK and Diamandis EP: Molecular cloning of the human kallikrein 15 gene (KLK15). J Biol Chem. 276:53–61. 2001. View Article : Google Scholar : PubMed/NCBI

74 

Yousef GM, Scorilas A, Katsaros D, Fracchioli S, Iskander L, Borgono C, Rigault de la Longrais IA, Puopolo M, Massobrio M and Diamandis EP: Prognostic value of the human kallikrein gene 15 expression in ovarian cancer. J Clin Oncol. 21:3119–3126. 2003. View Article : Google Scholar : PubMed/NCBI

75 

Rabien A, Fritzsche FR, Jung M, Tölle A, Diamandis EP, Miller K, Jung K, Kristiansen G and Stephan C: KLK15 is a prognostic marker for progression-free survival in patients with radical prostatectomy. Int J Cancer. 127:2386–2394. 2010. View Article : Google Scholar : PubMed/NCBI

76 

Yousef GM, Scorilas A, Magklara A, Memari N, Ponzone R, Sismondi P, Biglia N, Abd Ellatif M and Diamandis EP: The androgen-regulated gene human kallikrein 15 (KLK15) is an indpendent and favourable prognostic marker for breast cancer. Br J Cancer. 87:1294–1300. 2002. View Article : Google Scholar : PubMed/NCBI

77 

Kudo Y, Ogawa I, Kitajima S, Kitagawa M, Kawai H, Gaffney PM, Miyauchi M and Takata T: Periotin promotes invasion and anchorage-independent growth in the metastatic process of head and neck cancer. Cancer Res. 66:6928–6935. 2006. View Article : Google Scholar : PubMed/NCBI

78 

Malanchi I, Santamaria-Martínez A, Susanto E, Peng H, Lehr HA, Delaloye JF and Huelsken J: Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 481:85–89. 2011. View Article : Google Scholar : PubMed/NCBI

79 

Kanno A, Satoh K, Masamune A, Hirota M, Kimura K, Umino J, Hamada S, Satoh A, Egawa S, Motoi F, Unno M and Shimosegawa T: Periostin, secreted from stromal cells, has biphasic effect on cell migration and correlates with the epitelial to mesenchymal transition of human pancreatic cancer cells. Int J Cancer. 22:2707–2718. 2008. View Article : Google Scholar : PubMed/NCBI

80 

Kyutoku M, Taniyama Y, Katsuragi N, Shimizu H, Kunugiza Y, Iekushi K, Koibuchi N, Sanada F, Oshita Y and Morishita R: Role of periostin in cancer progression and metastasis: Inhibition of breast progression and metastasis by anti-periostin antibody in a murine model. Int J Mol Med. 28:181–186. 2011.PubMed/NCBI

81 

Carter H, Samayoa J, Hruban RH and Karchin R: Prioritization of driver mutations in pancreatic cancer using cancer-specific high-throughput annotation of somatic mutations (CHASM). Cancer Biol Ther. 10:582–587. 2010. View Article : Google Scholar : PubMed/NCBI

82 

Li Q and Wang L, Tan W, Peng Z, Luo Y, Zhang Y, Zhang G, Na D, Jin P, Shi T, Ma D and Wang L: Identification of C1qTNF-related protein 4 as a potential cytokine that stimulates the STAT3 and NF-κB pathways and promotes cell survival in human cancer cells. Cancer Lett. 28:203–214. 2011.PubMed/NCBI

83 

Belirgen M, Berrak SG, Ozdag H, Bozkurt SU, Eksioglu-Demiralp E and Ozek MM: Biologic tumor behavior in pilocytic astrocytomas. Childs Nerv Syst. 28:375–389. 2012. View Article : Google Scholar : PubMed/NCBI

84 

Huang YW, Jansen RA, Fabbri E, Potter D, Liyanarachchi S, Chan MW, Liu JC, Crijns AP, Brown R, Nephew KP, van der Zee AG, Cohn DE, Yan PS, Huang TH and Lin HJ: Identification of candidate epigenetic biomarkers for ovarian cancer detection. Oncol Rep. 22:853–861. 2009.PubMed/NCBI

85 

Heller G, Babinsky VN, Ziegler B, Weinzierl M, Noll C, Altenberger C, Müllauer L, Dekan G, Grin Y, Lang G, End-Pfützenreuter A, Steiner I, Zehetmayer S, Döme B, Arns BM, Fong KM, Wright CM, Yang IA, Klepetko W, Posch M, Zielinski CC and Zöchbauer-Müller S: Genome-wide CpG island methylation analyses in non-small cell lung cancer patients. Carcinogenesis. 34:513–521. 2013. View Article : Google Scholar : PubMed/NCBI

86 

Wirth T, Soeth E, Czubayko F and Juhl H: Inhibition of endogenous carcinoembryonic antigen (CEA) increases the apoptotic rate of colon cancer cells and inhibits metastatic tumor growth. Clin Exp Metastasis. 19:155–160. 2002. View Article : Google Scholar : PubMed/NCBI

87 

Arrieta O, Saavedra-Perez D, Kuri R, Aviles-Salas A, Martinez L, Mendoza-Posada D, Castillo P, Astorga A, Guzman E and de la Garza J: Brain metastasis development and poor survival associated with carcinoembryonic antigen (CEA) level in advanced non-small cell lung cancer: a prospective analysis. BMC Cancer. 9:119–128. 2009. View Article : Google Scholar : PubMed/NCBI

88 

Liebhardt S, Ditsch N, Nieuwland R, Rank A, Jeschke U, Von Koch F, Friese K and Toth B: CEA-, Her2/neu-, BCRP- and Hsp27-positive microparticles in breast cancer patients. Anticancer Res. 30:1707–1712. 2010.PubMed/NCBI

Related Articles

Journal Cover

November 2013
Volume 30 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fonseca-Sanchéz MA, Pérez-Plasencia C, Fernández-Retana J, Arechaga-Ocampo E, Marchat LA, Rodríguez-Cuevas S, Bautista-Piña V, Arellano-Anaya ZE, Flores-Pérez A, Diaz-Chávez J, Diaz-Chávez J, et al: microRNA-18b is upregulated in breast cancer and modulates genes involved in cell migration. Oncol Rep 30: 2399-2410, 2013
APA
Fonseca-Sanchéz, M.A., Pérez-Plasencia, C., Fernández-Retana, J., Arechaga-Ocampo, E., Marchat, L.A., Rodríguez-Cuevas, S. ... López-Camarillo, C. (2013). microRNA-18b is upregulated in breast cancer and modulates genes involved in cell migration. Oncology Reports, 30, 2399-2410. https://doi.org/10.3892/or.2013.2691
MLA
Fonseca-Sanchéz, M. A., Pérez-Plasencia, C., Fernández-Retana, J., Arechaga-Ocampo, E., Marchat, L. A., Rodríguez-Cuevas, S., Bautista-Piña, V., Arellano-Anaya, Z. E., Flores-Pérez, A., Diaz-Chávez, J., López-Camarillo, C."microRNA-18b is upregulated in breast cancer and modulates genes involved in cell migration". Oncology Reports 30.5 (2013): 2399-2410.
Chicago
Fonseca-Sanchéz, M. A., Pérez-Plasencia, C., Fernández-Retana, J., Arechaga-Ocampo, E., Marchat, L. A., Rodríguez-Cuevas, S., Bautista-Piña, V., Arellano-Anaya, Z. E., Flores-Pérez, A., Diaz-Chávez, J., López-Camarillo, C."microRNA-18b is upregulated in breast cancer and modulates genes involved in cell migration". Oncology Reports 30, no. 5 (2013): 2399-2410. https://doi.org/10.3892/or.2013.2691