Dual strands of pre-miR‑150 (miR‑150‑5p and miR‑150‑3p) act as antitumor miRNAs targeting SPOCK1 in naïve and castration-resistant prostate cancer

  • Authors:
    • Atsushi Okato
    • Takayuki Arai
    • Satoko Kojima
    • Keiichi Koshizuka
    • Yusaku Osako
    • Tetsuya Idichi
    • Akira Kurozumi
    • Yusuke Goto
    • Mayuko Kato
    • Yukio Naya
    • Tomohiko Ichikawa
    • Naohiko Seki
  • View Affiliations

  • Published online on: May 17, 2017     https://doi.org/10.3892/ijo.2017.4008
  • Pages: 245-256
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Analysis of our microRNA (miRNA) expression signature in human cancers has shown that guide and passenger strands of pre-miR‑150, i.e., miR‑150‑5p and miR‑150‑3p, are significantly downregulated in cancer tissues. In miRNA biogenesis, the passenger strand of miRNA is degraded and is thought to have no functions. Thus, the aim of this study was to investigate the functional significance of miR‑150‑5p and miR‑150‑3p in naïve prostate cancer (PCa) and castration-resistant prostate cancer (CRPC). Ectopic expression assays showed that both strands of miRNAs significantly suppressed cancer cell migration and invasion. Our strategies of miRNA target searching demonstrated that SPOCK1 (SPARC/osteonectin, cwcv and kazal like domains proteoglycan 1) was directly regulated by miR‑150‑5p and miR‑150‑3p. Knockdown of SPOCK1 by siRNA inhibited cancer cell aggressiveness. Moreover, overexpression of SPOCK1 was observed in naïve PCa and CRPC tissues. Taken together, dual strands of pre-miR‑150 (miR‑150‑5p and miR‑150‑3p) acted as antitumor miRNAs in naïve PCa and CRPC cells. Expression of oncogenic SPOCK1 was involved in naïve PCa and CRPC pathogenesis. Novel approaches to analysis of antitumor miRNA-regulated RNA networks in cancer cells may provide new insights into the pathogenic mechanisms of naïve PCa and CRPC.

Introduction

Prostate cancer (PCa) is the most frequently diagnosed cancer among men in developed countries (1). Although PCa is initially responsive to androgen-deprivation therapy (ADT), most patients experience disease relapse and develop castration-resistant prostate cancer (CRPC) (2). The survival rate of patients with CRPC is poor owing to the occurrence of metastasis (3) and patients with metastatic CRPC cannot be effectively treated using recently developed molecular-targeted therapies (2,3). Therefore, new treatment strategies are needed for these patients. The molecular mechanisms underlying the aggressiveness of CRPC cells and the acquisition of treatment resistance are still unclear.

MicroRNAs (miRNAs) are noncoding RNAs that act as sequence-specific fine tuners for regulating the expression levels of proteins and RNAs (4,5). Notably, a single miRNA can regulate a large number of RNA transcripts in human cells (6). Accordingly, dysregulation of miRNAs can disrupt the tightly regulated RNA networks in cancer cells, leading to cancer cell initiation, development, metastasis, and drug resistance (7,8). The discovery of miRNAs has complicated the analysis of intracellular RNA networks in cancer.

We recently constructed an RNA-sequence based miRNA expression signature using autopsy specimens from patients with CRPC (9). To elucidate the miRNA-mediated RNA networks in metastatic CRPC, we have sequentially identified antitumor miRNAs and oncogenic genes regulated by these miRNAs based on our miRNA expression signatures (9,10). Analysis of our miRNA signatures, including that in CRPC, has shown that both strands of pre-miR-150, i.e., miR-150-5p (the guide strand) and miR-150-3p (the passenger strand), are significantly reduced in cancer tissues (9).

In miRNA biogenesis, pre-miRNA is cleaved by Dicer into the miRNA duplex, containing the guide and passenger strands. The mature guide strand miRNA is incorporated into the RNA-induced silencing complex (RISC) and represses mRNA translation or cleaves mRNA (11). In contrast, the passenger strand of miRNA was previously thought to be degraded and to have no function (1214). However, our expression data have contradicted this established miRNA theory. We hypothesized that the passenger strand of miR-150-3p may have functions in naïve PCa and CRPC cells by targeting novel oncogenic genes. Accordingly, in this study, we aimed to investigate the functional significance of miR-150-5p and miR-150-3p in naïve PCa and CRPC cells and to identify oncogenic genes involved in the pathogenesis of the disease.

Materials and methods

Clinical prostate specimens

Clinical prostate specimens were obtained from patients admitted to Teikyo University Chiba Medical Center from 2014 to 2015. All patients had elevated prostate-specific antigen (PSA) and had undergone trans-rectal biopsies for definitive cancer diagnoses. Particularly, in patients with CRPC, neuroendocrine differentiation was excluded. The patient backgrounds are summarized in Table I. Samples were staged according to the UICC TNM classification (15).

Table I

Characteristics of patients with non-PCa and naïve PCa and CRPC.

Table I

Characteristics of patients with non-PCa and naïve PCa and CRPC.

No.DiagnosisAge (years)PSA (ng/ml)Gleason scoreTNMStage
1Non-PCa575.71
2Non-PCa749.45
3Non-PCa708.58
4Non-PCa734.8
5Non-PCa676.91
6Non-PCa507.05
7Non-PCa749.91
8Non-PCa7620.9
9Non-PCa594.5
10Non-PCa751.1
11Non-PCa607.29
12Non-PCa7338.7
13Non-PCa6911.9
14Non-PCa7723.3
15Non-PCa614.57
16Non-PCa597.37
17Non-PCa655.06
18PCa7075.74+5411D2
19PCa781,8004+5411D2
20PCa7568.45+4410D1
21PCa6238.74+52b10D1
22PCa7025.54+53b00C
23PCa751,2604+53b11D2
24PCa888884+53b11D2
25PCa6933.94+5401D2
26PCa6262.34+53b10D1
27PCa7854+52c01bD2
28PCa644494+53b11D2
29PCa813654+5411D2
30PCa767155+4411D2
31PCa795554+5311D2
32PCa631,1204+52c01bD2
33PCa674.954+5411bD2
34PCa7019.55+5411cD2
35CRPC6915.85+43b11D2
36CRPC722125+4411D2
37CRPC714.44+5411D2
38CRPC662954+5411D2
39CRPC687.544+5411bD2

All patients in this study provided written informed consent for tissue donation for research purposes. The protocol was approved by the Institutional Review Boards of Chiba University and Teikyo University.

Tissue collection and cell culture

Prostate tissues were immersed in RNAlater (Thermo Fisher Scientific, Waltham, MA, USA) and stored at 4°C until RNA extraction. Human prostate cancer cells (PC3 and PC3M cells) were obtained from the American Type Culture Collection (Manassas, VA, USA).

Quantitative real-time reverse transcription polymerase chain reaction (RT-q-PCR)

Stem-loop RT-PCR (TaqMan MicroRNA assays; product ID: 000473 for miR-150-5p and 002637 for miR-150-3p; Applied Biosystems, Foster City, CA, USA) was used in this assays. TaqMan probes and primers for SPOCK1 (product ID: Hs00270274_m1; Applied Biosystems) were assay-on-demand gene expression products. We used GUSB (product ID: Hs00939627_m1; Applied Biosystems), GAPDH (product ID: Hs02758991_g1; Applied Biosystems), and RNU48 (product ID: 001006; Applied Biosystems) as internal controls.

Cell proliferation, migration, and invasion assays

Cell proliferation, migration, and invasion assays were carried out as previously described (9,10).

Argonaute 2 (AGO2)-bound miRNA isolation by immunoprecipitation

PC3 cells were transfected with 10 nM miRNA by reverse transfection and plated in 10-cm plates at 1×105 cells/ml. After 48 h, immunoprecipitation was performed using a microRNA Isolation kit, Human Ago2 (Wako, Osaka, Japan) according to the manufacturer's protocol. Expression levels of miRNAs bound to Ago2 were measured by TaqMan RT-qPCR. miRNA expression data were normalized to the expression of miR-26a (product ID: 000404; Applied Biosystems), which was not affected by miR-150-5p and miR-150-3p.

Western blot analysis

Immunoblotting was conducted with diluted monoclonal anti-SPOCK1 antibodies (1:100 dilution; sc-398782; Santa Cruz Biotechnology, Santa Cruz, CA, USA) and with diluted anti-glyceraldehyde 3-phosphate dehydrogenase (GAPDH) antibodies (1:1,000 dilution; ab8245; Abcam, Cambridge, UK) as a loading control. The procedures were performed as previously described (16).

Selection of putative target genes regulated by miR-150-5p and miR-150-3p in PCa cells

To identify miR-150-5p and miR-150-3p target genes, we analyzed gene expression profile (Gene Expression Omnibus database; accession no. GSE29079). Gene expression data were obtained from miR-150-5p and miR-150-3p transfectant PC3 cells (A SurePrint G3 Human 60K, Agilent Technologies, Santa Clara, CA, USA). We merged these datasets and selected putative miR-150-5p and miR-150-3p target genes using the TargetScan database (Release 7.1; http://www.targetscan.org/vert_71/).

Plasmid construction and dual-luciferase assays

The partial wide-type sequences of the 3′-untranslated region (UTR) of SPOCK1 or those with a deleted miR-150-5p or miR-150-3p target site were inserted in the psiCHECK-2 vector (C8021; Promega, Madison, WI, USA). Alternatively, we used sequences that were missing the miR-150-5p target site (position 182–188) or miR-150-3p target sites (position 1477–1483, position 1749–1756, or position 2593–2599). The procedure for dual-luciferase reporter assays was described previously (17).

Immunohistochemistry using tissue microarrays

We used a tissue microarray of prostate cancer samples obtained from Provitro (Berlin, Germany; cat. no. 401–2209, Lot no. 146.1P), which contained a total of 78 prostate samples (PCa specimens: n=58, prostatic intraepithelial neoplasia: n=10, and normal prostate samples: n=10). Detailed information on these samples is summarized in Table II. The tissue microarray was immunostained following the manufacturer's protocol, as described previously (10,18).

Table II

Characteristics of patients, as evaluated using immunohistochemistry.

Table II

Characteristics of patients, as evaluated using immunohistochemistry.

No.DiagnosisAge (years)Gleason scoreTNIHC score of SPOCK1
1PCa644+33b05
2PCa673+42b03
3PCa583+42b05
4PCa6373b06
5PCa653+32b05
6PCa614+43b×4
7PCa623+42b×2
8PCa664+42b×4
9PCa613+43a×4
10PCa744+32b×5
11PCa592+32b×4
12PCa693+43a04
13PCa543+42c×6
14PCa683+43a05
15PCa583+43a05
16PCa673+43a04
17PCa653+32a04
18PCa773+4405
19PCa644+32b04
20PCa583+43a06
21PCa504+32b06
22PCa533+32b03
23PCa594+53a05
24PCa702+32b05
25PCa655+43a04
26PCa573+52b05
27PCa684+42b06
28PCa583+32b05
29PCa663+32b03
30PCa633+42b06
31PCa563+42b04
32PCa635+33a03
33PCa643+53a05
34PCa603+42b05
35PCa603+33a05
36PCa573+22b04
37PCa503+32a06
38PCa683+33a05
39PCa653+43b14
40PCa695+53a15
41PCa633+42b05
42PCa512+32b04
43PCa623+33a05
44PCa613+43a05
45PCa534+43b14
46PCa564+32b05
47PCa592+32b05
48PCa613+42b05
49PCa513+43a06
50PCa623+43b15
51PCa663+33a05
52PCa623+32b04
53PCa563+32b05
54PCa583+33a05
55PCa665+43a06
56PCa553+43a05
57PCa672+32b05
58PCa613+52b06
59PIN593
60PIN585
61PIN624
62PIN516
63PIN583
64PIN684
65PIN645
66PIN565
67PIN613
68PIN515
69Normal704
70Normal633
71Normal624
72Normal810
73Normal673
74Normal764
75Normal664
76Normal693
77Normal635
78Normal714

[i] PCa, prostate cancer; PIN, prostatic intra-epithelial neoplasia.

Statistical analysis

The relationships between 2 groups and expression values obtained by RT-PCR were analyzed using Mann-Whitney U tests. The correlations between miR-150-5p and miR-150-3p expression were evaluated using Spearman's rank test. The relationships among more than 3 variables and numerical values were analyzed using Bonferroni-adjusted Mann-Whitney U tests. We used Expert StatView software (version 5.0 SAS Institute Inc., Cary, NC, USA) for these analyses.

Results

The expression levels of miR-150-5p and miR-150-3p in naïve PCa and CRPC specimens and cell lines

We evaluated the expression levels of miR-150-5p and miR-150-3p in PCa tissues (PCa: n=17, CRPC: n=5), normal tissues (non-PCa: n=17), and PCa cell lines (PC3 and PC3M cells). The patient backgrounds are summarized in Table I. The expression levels of miR-150-5p and miR-150-3p were significantly lower in PCa and CRPC tissues than normal tissues (miR-150-5p: P=0.0266 and P=0.018, respectively; miR-150-3p: P=0.0025 and P=0.0002, respectively; Fig. 1A). There was a positive correlation between expression levels of miR-150-5p and miR-150-3p (r=0.854, P<0.0001; Fig. 1B).

Effects of ectopic expression of miR-150-5p and miR-150-3p on cell proliferation, migration, and invasion assays in PCa cell lines

To examine the functional roles of miR-150-5p and miR-150-3p, we performed gain-of-function studies by using PC3 and PC3M cells transfected with mature miRNAs.

XTT assays revealed that proliferation was significantly inhibited in PC3 and PC3M cells transfected with miR-150-5p and miR-150-3p in comparison with that of mock or miR-control-transfected cells (P<0.0001; Fig. 1C). Wound-healing and Matrigel invasion assays demonstrated significant inhibition of cell migration and invasion in both miR-150-5p and miR-150-3p transfectants (P<0.0001; Fig. 1D and E).

Both miR-150-5p and miR-150-3p bound to Ago2

We hypothesized that both miR-150-5p and miR-150-3p may be incorporated into and function as part of the RISC. To test this hypothesis, we performed immunoprecipitation with antibodies targeting Ago2, which plays a central role in the RISC. After transfection with miR-150-5p or miR-150-3p, Ago2-bound miRNAs were isolated, and RT-qPCR was carried out to determine whether miR-150-5p and miR-150-3p bound to Ago2 (Fig. 2A).

After transfection with miR-150-5p and immunoprecipitation by anti-Ago2 antibodies, miR-150-5p levels were significantly higher than those of mock- or miR control-transfected cells and those of miR-150-3p-transfected PC3 cells (P<0.0001; Fig. 2B). Similarly, after transfection with miR-150-3p and immunoprecipitation by anti-Ago2 antibodies, miR-150-3p levels were significantly higher than those of mock- or miR control-transfected cells and those of miR-150-5p-transfected PC3 cells (P<0.0001; Fig. 2B).

Screening of target genes regulated by miR-150-3p in PCa cells

To obtain further insights into the molecular mechanisms regulated by antitumor miR-150-3p in PCa cells, we screened these miRNA-regulated genes by using in silico and genome-wide gene expression analyses. First, we undertook genome-wide gene expression analysis using PC3 cells.

Analysis of the TargetScan database showed that 2,558 genes had putative target sites for miR-150-3p in their 3′-UTRs. Next, we screened 328 of these genes using genome-wide gene expression analysis. Finally, we found 14 genes that were upregulated (fold-change log2 >0.5) in cancer tissues by GEO database analyses (GEO accession no. GSE29079). Our strategy for analysis is shown in Fig. 3. Putative target genes of miR-150-3p are summarized in Table III. Among these candidate genes, SPOCK1 had a putative binding site for miR-150-5p according to the TargetScan database. Therefore, we focused on SPOCK1 as a candidate target gene of the dual strands of pre-miR-150 and performed further investigations of this target in PCa.

Table III

Putative target genes regulated by miR-150-3p in PCa cells.

Table III

Putative target genes regulated by miR-150-3p in PCa cells.

Entrez gene IDGene symbolGene namePC3 miR-150-3p transfectant (log2 ratio)Site countsGEO Fold change
55771SPOCK1Sparc/osteonectin, cwcv and kazal-like domains proteoglycan (testican) 1−2.71846531.112207
11113LIG3Ligase III, DNA, ATP-dependent−1.01395820.833472
9448MLECMalectin−1.0593910.817763
1017NETO2Neuropilin (NRP) and tolloid (TLL)-like 2−1.63935520.808991
51400 HIST1H3BHistone cluster 1, H3b−1.07023710.803539
22877PHF12PHD finger protein 12−1.53198310.779377
4615NCALDNeurocalcin δ−1.34562320.772963
6695SIM2Single-minded family bHLH transcription factor 2−2.06061610.707018
79071TAB3TGF-β activated kinase 1/MAP3K7 binding protein 3−1.34169810.642641
8358MAP4K4Mitogen-activated protein kinase kinase kinase kinase 4−1.04332810.580977
10186HNRNPABHeterogeneous nuclear ribonucleoprotein A/B−1.50129210.571322
11113FARP1FERM, RhoGEF (ARHGEF) and pleckstrin domain protein 1 (chondrocyte-derived)−1.37955810.536826
54475SPECC1LSperm antigen with calponin homology and coiled-coil domains 1-like−1.16863310.506925
6548NFIBNuclear factor I/B−1.18980410.505127
Regulation of SPOCK1 expression by miR-150-5p and miR-150-3p in PCa cells

Our studies revealed that SPOCK1 mRNA was significantly reduced in both miR-150-5p and miR-150-3p transfectants in comparison with those in mock or miR-control transfectants (P<0.0001 and P<0.0001; Fig. 4A). Expression of SPOCK1 protein was also repressed in these miRNAs transfectants (Fig. 4B).

Target prediction databases indicated that miR-150-5p had one putative target site in the 3′-UTR of SPOCK1 (Fig. 4C). Likewise, miR-150-3p had three putative target sites (Fig. 4C). To determine whether SPOCK1 mRNA contained functional target sites, we performed a dual-luciferase reporter assay.

The TargetScan database identified one putative target site in the 3′-UTR of SPOCK1 for miR-150-5p (position 182-188) and three target sites of SPOCK1 for miR-150-3p (positions 1477-1483, 1749-1756, and 2593-2599). We used vectors encoding a partial wild-type sequence of the 3′-UTR of SPOCK1 mRNA, including the predicted miR-150-5p and miR-150-3p target site, or a vector lacking the miR-150-5p and miR-150-3p target sites. We found that the luminescence intensity was significantly reduced by co-transfection with miR-150-5p or miR-150-3p and the vector carrying the wild-type 3′-UTR of SPOCK1 (P<0.05; Fig. 4D).

Effects of silencing SPOCK1 on cell proliferation, migration, and invasion in PCa cells

We evaluated the knockdown efficiency of si-SPOCK1 transfection in PC3 cells. Our present data showed that si-SPOCK1 transfection effectively downregulated SPOCK1 expression in PC3 and PC3M cells (Fig. 5A and B).

Functional assays demonstrated that cell proliferation, migration, and invasion were inhibited in si-SPOCK1 transfectants compared with those in mock- or miR control-transfected cells (P<0.0001, Fig. 5C–E).

Analysis of SPOCK1 expression in naïve PCa and CRPC clinical specimens by immunohistochemistry

Next, we examined the expression levels of SPOCK1 in naïve PCa specimens by immunohistochemical staining. SPOCK1 was strongly expressed in several cancer tissues, while low expression was observed in normal tissues (Fig. 6A). Moreover, the expression score for SPOCK1 protein was significantly higher in PCa tissues than in normal tissues (P<0.001, Fig. 6B). The patient backgrounds and clinicopathological characteristics are summarized in Table II.

To analyze SPOCK1 protein expression, immunohistochemistry was performed with CRPC specimens. Immunohistochemical staining demonstrated high expression of SPOCK1 in CRPC tissues. SPOCK1 was strongly expressed in the cytoplasm of the PCa cells almost in the same area where PSA was expressed (Fig. 6C).

Discussion

Androgen-dependent PCa initially responds to ADT, which can result in disease control. However, most PCa cells eventually acquire ADT-resistance mechanisms. Moreover, there are no curative treatments for patients with metastatic CRPC (19). One of the main challenges in treating CRPC is controlling aggressive, lethal metastatic PCa cells. We believe identification of the genes and pathways responsible for metastasis may lead to the development of new therapeutic strategies. Accordingly, we have focused on identifying antitumor miRNAs and oncogenic RNA networks mediated by these miRNAs in naïve PCa and CRPC cells (9,20,21). For example, antitumor miR-223 inhibits cancer cell migration and invasion by targeting ITGA3 and ITGB1 (18). Antitumor miR-218 suppresses migration and invasion by regulating LASP1 (22). Moreover, antitumor miRNAs (miR-26a/b, miR-29a/b/c and miR-218) function cooperatively to suppress metastasis-promoting LOXL (23).

In this study, we demonstrated that the dual strands of pre-miR-150, i.e., miR-150-5p and miR-150-3p, acted as antitumor miRNAs in naïve PCa and CRPC cells. According to the miRNA database (miRBase: http://www.mirbase.org/), miR-150-5p is a guide strand of pre-miR-150, and miR-150-3p is the corresponding passenger strand. Previous studies have shown that miR-150-5p (the guide strand) is frequently downregulated in cancer tissues and functions as an antitumor miRNA in several types of cancer (2426). In this study, we focused on miR-150-3p (the passenger strand) and investigated the antitumor roles of this miRNA in naïve PCa and CRPC cells because no prior studies had evaluated the functions of miR-150-3p in cancer cells.

Passenger strands of miRNA are thought to be degraded and are not expected to be incorporated into the RISC (4). However, our data showed that the passenger strand of miR-150 was incorporated into the RISC in PCa cells, and this is the first report of the antitumor function of miR-150-3p in cancer cells. Our recent studies demonstrated that miR-145-3p (the passenger strand of pre-miR-145) acted as an antitumor miRNA targeting oncogenic UHRF1 and MTDH in bladder and lung cancer, respectively (16,27). Similarly, we confirmed the antitumor function of miR-139-3p (a passenger strand of pre-miR-139) in bladder cancer (17). These findings suggested that the passenger strands of miRNAs may have some biological functions in human cells, similar to the guide strands of miRNAs. The involvement of passenger strand miRNAs in the regulation of cellular processes is a novel concept in RNA research.

One of the main challenges in miRNA studies is to seek out miRNA targeting genes and RNA networks mediated by these miRNAs in cancer cells. We revealed that SPOCK1 was a direct target of dual strands of miR-150-5p and miR-150-3p in PCa cells. Moreover, we demonstrated the overexpression of SPOCK1 in naïve PCa and CRPC clinical specimens. SPOCK1/testican-1 belongs to the Ca2+-binding proteoglycan family, which includes SPARC, testican-2, and testican-3 (28). Overexpression of SPOCK1 was observed in several cancers and has been shown to play pivotal roles in cancer cell progression, metastasis, and drug resistance (2931). SPOCK1 is upregulated in lung cancer and is associated with metastasis and survival (32). Interestingly, ectopic expression of SPOCK1 induces the epithelial-mesenchymal transition (EMT) in lung cancer cells (33). Another study demonstrated that SPOCK1 induces MET-dependent EMT signaling in lapatinib-resistant gastric cancer (34). Several reports have indicated that tyrosine kinase receptor inhibitors (TKIs) can frequently cause the acquisition of TKI resistance in cells and that the EMT is deeply involved in these events (35,36). These findings suggest that SPOCK1 mediates the EMT signaling to regulate cancer cell aggressiveness and drug resistance.

Most patients with PCa exhibit ADT failure and progress to CRPC with metastasis. Moreover, no curative treatments are available for advanced CRPC with metastasis (37). In CRPC cells, the EMT is associated with metastatic processes and is involved in drug resistance (38). Interestingly, androgens and androgen receptor-mediated signaling enhance the EMT and cancer cell aggressiveness (39). Transforming growth factor β (TGFβ) is a pivotal player that induces EMT in cancer cells (40). Increased expression of TGFβ and EMT-related proteins has been observed in CRPC bone metastasis (41). Interestingly, expression of SPOCK1 is elevated by TGFβ treatment in lung cancer cells, suggesting that SPOCK1 mediates downstream TGFβ signaling (33). We suggest that SPOCK1 expression may be related to induction of TGFβ signaling and epigenetic regulation of miR-150-5p and miR-150-3p in naïve PCa and CRPC cells. A recent study showed that overexpression of SPOCK1 in RWPE-1 cells (non-neoplastic adult human prostatic epithelial cells) promotes cell viability and cell migration and invasion abilities (42). These findings were supported by our present data. Thus, the expression of SPOCK1 may be involved in the pathogenesis of naïve PCa and CRPC, and SPOCK1-mediated signaling may be a promising therapeutic target in this disease.

In conclusiom, the dual strands of pre-miR-150, i.e., miR-150-5p and miR-150-3p, were significantly reduced in naïve PCa and CRPC tissues and acted as antitumor mRNAs. The passenger strand of miR-150-3p was found to have a specific function in cancer cells. SPOCK1 was directly regulated by dual strands of miR-150-5p and miR-150-3p in PCa cells. Overexpression of SPOCK1 was confirmed in naïve PCa and CRPC tissues and acted as an oncogene in this disease. Elucidation of miR-150/SPOCK1-mediated molecular networks may lead to a better understanding of the pathogenesis of naïve PCa and CRPC and facilitate the development of new treatment strategies.

Acknowledgments

This study was supported by KAKENHI grants 15K10801(C), 15K20071(C), 16K20125, and 16H05462(B).

References

1 

Siegel RL, Miller KD and Jemal A: Cancer Statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Sridhar SS, Freedland SJ, Gleave ME, Higano C, Mulders P, Parker C, Sartor O and Saad F: Castration-resistant prostate cancer: From new pathophysiology to new treatment. Eur Urol. 65:289–299. 2014. View Article : Google Scholar

3 

Sturge J, Caley MP and Waxman J: Bone metastasis in prostate cancer: Emerging therapeutic strategies. Nat Rev Clin Oncol. 8:357–368. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Carthew RW and Sontheimer EJ: Origins and mechanisms of miRNAs and siRNAs. Cell. 136:642–655. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Zhang Y, Wang Z and Gemeinhart RA: Progress in microRNA delivery. J Control Release. 172:962–974. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Bartel DP: MicroRNAs: Target recognition and regulatory functions. Cell. 136:215–233. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Nelson KM and Weiss GJ: MicroRNAs and cancer: Past, present, and potential future. Mol Cancer Ther. 7:3655–3660. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Goto Y, Kojima S, Nishikawa R, Kurozumi A, Kato M, Enokida H, Matsushita R, Yamazaki K, Ishida Y, Nakagawa M, et al: MicroRNA expression signature of castration-resistant prostate cancer: The microRNA-221/222 cluster functions as a tumour suppressor and disease progression marker. Br J Cancer. 113:1055–1065. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Okato A, Goto Y, Kurozumi A, Kato M, Kojima S, Matsushita R, Yonemori M, Miyamoto K, Ichikawa T and Seki N: Direct regulation of LAMP1 by tumor-suppressive microRNA-320a in prostate cancer. Int J Oncol. 49:111–122. 2016.PubMed/NCBI

11 

Gregory RI, Chendrimada TP, Cooch N and Shiekhattar R: Human RISC couples microRNA biogenesis and posttranscriptional gene silencing. Cell. 123:631–640. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Chendrimada TP, Gregory RI, Kumaraswamy E, Norman J, Cooch N, Nishikura K and Shiekhattar R: TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing. Nature. 436:740–744. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Hutvágner G and Zamore PD: A microRNA in a multiple-turnover RNAi enzyme complex. Science. 297:2056–2060. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Matranga C, Tomari Y, Shin C, Bartel DP and Zamore PD: Passenger-strand cleavage facilitates assembly of siRNA into Ago2-containing RNAi enzyme complexes. Cell. 123:607–620. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Sobin LH, Gospodarowicz MK and Wittekind Ch: TNM Classification of Malignant Tumours. 7th edition. Wiley-Blackwell; Chichester: 2009

16 

Mataki H, Seki N, Mizuno K, Nohata N, Kamikawaji K, Kumamoto T, Koshizuka K, Goto Y and Inoue H: Dual-strand tumor-suppressor microRNA-145 (miR-145-5p and miR-145-3p) coordinately targeted MTDH in lung squamous cell carcinoma. Oncotarget. 7:72084–72098. 2016.PubMed/NCBI

17 

Yonemori M, Seki N, Yoshino H, Matsushita R, Miyamoto K, Nakagawa M and Enokida H: Dual tumor-suppressors miR-139-5p and miR-139-3p targeting matrix metalloprotease 11 in bladder cancer. Cancer Sci. 107:1233–1242. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Kurozumi A, Goto Y, Matsushita R, Fukumoto I, Kato M, Nishikawa R, Sakamoto S, Enokida H, Nakagawa M, Ichikawa T, et al: Tumor-suppressive microRNA-223 inhibits cancer cell migration and invasion by targeting ITGA3/ITGB1 signaling in prostate cancer. Cancer Sci. 107:84–94. 2016. View Article : Google Scholar

19 

Semenas J, Allegrucci C, Boorjian SA, Mongan NP and Persson JL: Overcoming drug resistance and treating advanced prostate cancer. Curr Drug Targets. 13:1308–1323. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Fuse M, Kojima S, Enokida H, Chiyomaru T, Yoshino H, Nohata N, Kinoshita T, Sakamoto S, Naya Y, Nakagawa M, et al: Tumor suppressive microRNAs (miR-222 and miR-31) regulate molecular pathways based on microRNA expression signature in prostate cancer. J Hum Genet. 57:691–699. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Goto Y, Kurozumi A, Enokida H, Ichikawa T and Seki N: Functional significance of aberrantly expressed microRNAs in prostate cancer. Int J Urol. 22:242–252. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Nishikawa R, Goto Y, Sakamoto S, Chiyomaru T, Enokida H, Kojima S, Kinoshita T, Yamamoto N, Nakagawa M, Naya Y, et al: Tumor-suppressive microRNA-218 inhibits cancer cell migration and invasion via targeting of LASP1 in prostate cancer. Cancer Sci. 105:802–811. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Kato M, Kurozumi A, Goto Y, Matsushita R, Okato A, Nishikawa R, Fukumoto I, Koshizuka K, Ichikawa T and Seki N: Regulation of metastasis-promoting LOXL2 gene expression by antitumor microRNAs in prostate cancer. J Hum Genet. 62:123–132. 2017. View Article : Google Scholar

24 

Abe F, Kitadate A, Ikeda S, Yamashita J, Nakanishi H, Takahashi N, Asaka C, Teshima K, Miyagaki T, Sugaya M, et al: Histone deacetylase inhibitors inhibit metastasis by restoring a tumor suppressive microRNA-150 in advanced cutaneous T-cell lymphoma. Oncotarget. 8:7572–7585. 2017.

25 

Srivastava SK, Bhardwaj A, Singh S, Arora S, Wang B, Grizzle WE and Singh AP: MicroRNA-150 directly targets MUC4 and suppresses growth and malignant behavior of pancreatic cancer cells. Carcinogenesis. 32:1832–1839. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Qu Y, Pan S, Kang M, Dong R and Zhao J: MicroRNA-150 functions as a tumor suppressor in osteosarcoma by targeting IGF2BP1. Tumour Biol. 37:5275–5284. 2016. View Article : Google Scholar

27 

Matsushita R, Yoshino H, Enokida H, Goto Y, Miyamoto K, Yonemori M, Inoguchi S, Nakagawa M and Seki N: Regulation of UHRF1 by dual-strand tumor-suppressor microRNA-145 (miR-145-5p and miR-145-3p): Inhibition of bladder cancer cell aggressiveness. Oncotarget. 7:28460–28487. 2016.PubMed/NCBI

28 

Bradshaw AD: Diverse biological functions of the SPARC family of proteins. Int J Biochem Cell Biol. 44:480–488. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Li Y, Chen L, Chan TH, Liu M, Kong KL, Qiu JL, Li Y, Yuan YF and Guan XY: SPOCK1 is regulated by CHD1L and blocks apoptosis and promotes HCC cell invasiveness and metastasis in mice. Gastroenterology. 144:179–191.e4. 2013. View Article : Google Scholar

30 

Ma LJ, Wu WJ, Wang YH, Wu TF, Liang PI, Chang IW, He HL and Li CF: SPOCK1 Overexpression confers a poor prognosis in urothelial carcinoma. J Cancer. 7:467–476. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Shu YJ, Weng H, Ye YY, Hu YP, Bao RF, Cao Y, Wang XA, Zhang F, Xiang SS, Li HF, et al: SPOCK1 as a potential cancer prognostic marker promotes the proliferation and metastasis of gallbladder cancer cells by activating the PI3K/AKT pathway. Mol Cancer. 14:122015. View Article : Google Scholar : PubMed/NCBI

32 

Kusakabe M, Kutomi T, Watanabe K, Emoto N, Aki N, Kage H, Hamano E, Kitagawa H, Nagase T, Sano A, et al: Identification of G0S2 as a gene frequently methylated in squamous lung cancer by combination of in silico and experimental approaches. Int J Cancer. 126:1895–1902. 2010.

33 

Miao L, Wang Y, Xia H, Yao C, Cai H and Song Y: SPOCK1 is a novel transforming growth factor-β target gene that regulates lung cancer cell epithelial-mesenchymal transition. Biochem Biophys Res Commun. 440:792–797. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Kim HP, Han SW, Song SH, Jeong EG, Lee MY, Hwang D, Im SA, Bang YJ and Kim TY: Testican-1-mediated epithelial-mesenchymal transition signaling confers acquired resistance to lapatinib in HER2-positive gastric cancer. Oncogene. 33:3334–3341. 2014. View Article : Google Scholar

35 

Uramoto H, Iwata T, Onitsuka T, Shimokawa H, Hanagiri T and Oyama T: Epithelial-mesenchymal transition in EGFR-TKI acquired resistant lung adenocarcinoma. Anticancer Res. 30:2513–2517. 2010.PubMed/NCBI

36 

Choe C, Shin YS, Kim C, Choi SJ, Lee J, Kim SY, Cho YB and Kim J: Crosstalk with cancer-associated fibroblasts induces resistance of non-small cell lung cancer cells to epidermal growth factor receptor tyrosine kinase inhibition. Onco Targets Ther. 8:3665–3678. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Crawford ED, Higano CS, Shore ND, Hussain M and Petrylak DP: Treating patients with metastatic castration resistant prostate xancer: A comprehensive review of available therapies. J Urol. 194:1537–1547. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Martin SK, Pu H, Penticuff JC, Cao Z, Horbinski C and Kyprianou N: Multinucleation and mesenchymal-to-epithelial transition alleviate resistance to combined cabazitaxel and anti-androgen therapy in advanced prostate cancer. Cancer Res. 76:912–926. 2016. View Article : Google Scholar

39 

Nakazawa M and Kyprianou N: Epithelial-mesenchymal-transition regulators in prostate cancer: Androgens and beyond. J Steroid Biochem Mol Biol. 166:84–90. 2017. View Article : Google Scholar

40 

Xu J, Lamouille S and Derynck R: TGF-beta-induced epithelial to mesenchymal transition. Cell Res. 19:156–172. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Haider M, Zhang X, Coleman I, Ericson N, True LD, Lam HM, Brown LG, Ketchanji M, Nghiem B, Lakely B, et al: Epithelial mesenchymal-like transition occurs in a subset of cells in castration resistant prostate cancer bone metastases. Clin Exp Metastasis. 33:239–248. 2016. View Article : Google Scholar :

42 

Chen Q, Yao YT, Xu H, Chen YB, Gu M, Cai ZK and Wang Z: SPOCK1 promotes tumor growth and metastasis in human prostate cancer. Drug Des Devel Ther. 10:2311–2321. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2017
Volume 51 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Okato A, Arai T, Kojima S, Koshizuka K, Osako Y, Idichi T, Kurozumi A, Goto Y, Kato M, Naya Y, Naya Y, et al: Dual strands of pre-miR‑150 (miR‑150‑5p and miR‑150‑3p) act as antitumor miRNAs targeting SPOCK1 in naïve and castration-resistant prostate cancer. Int J Oncol 51: 245-256, 2017
APA
Okato, A., Arai, T., Kojima, S., Koshizuka, K., Osako, Y., Idichi, T. ... Seki, N. (2017). Dual strands of pre-miR‑150 (miR‑150‑5p and miR‑150‑3p) act as antitumor miRNAs targeting SPOCK1 in naïve and castration-resistant prostate cancer. International Journal of Oncology, 51, 245-256. https://doi.org/10.3892/ijo.2017.4008
MLA
Okato, A., Arai, T., Kojima, S., Koshizuka, K., Osako, Y., Idichi, T., Kurozumi, A., Goto, Y., Kato, M., Naya, Y., Ichikawa, T., Seki, N."Dual strands of pre-miR‑150 (miR‑150‑5p and miR‑150‑3p) act as antitumor miRNAs targeting SPOCK1 in naïve and castration-resistant prostate cancer". International Journal of Oncology 51.1 (2017): 245-256.
Chicago
Okato, A., Arai, T., Kojima, S., Koshizuka, K., Osako, Y., Idichi, T., Kurozumi, A., Goto, Y., Kato, M., Naya, Y., Ichikawa, T., Seki, N."Dual strands of pre-miR‑150 (miR‑150‑5p and miR‑150‑3p) act as antitumor miRNAs targeting SPOCK1 in naïve and castration-resistant prostate cancer". International Journal of Oncology 51, no. 1 (2017): 245-256. https://doi.org/10.3892/ijo.2017.4008