Skip to main content
Oncotarget logoLink to Oncotarget
. 2016 Apr 9;7(19):28460–28487. doi: 10.18632/oncotarget.8668

Regulation of UHRF1 by dual-strand tumor-suppressor microRNA-145 (miR-145-5p and miR-145-3p): inhibition of bladder cancer cell aggressiveness

Ryosuke Matsushita 1, Hirofumi Yoshino 1, Hideki Enokida 1, Yusuke Goto 2, Kazutaka Miyamoto 1, Masaya Yonemori 1, Satoru Inoguchi 1, Masayuki Nakagawa 1, Naohiko Seki 2
PMCID: PMC5053739  PMID: 27072587

Abstract

In microRNA (miRNA) biogenesis, the guide-strand of miRNA integrates into the RNA induced silencing complex (RISC), whereas the passenger-strand is inactivated through degradation. Analysis of our miRNA expression signature of bladder cancer (BC) by deep-sequencing revealed that microRNA (miR)-145-5p (guide-strand) and miR-145-3p (passenger-strand) were significantly downregulated in BC tissues. It is well known that miR-145-5p functions as a tumor suppressor in several types of cancer. However, the impact of miR-145-3p on cancer cells is still ambiguous. The aim of the present study was to investigate the functional significance of miR-145-3p and BC oncogenic pathways and targets regulated by miR-145-5p/miR-145-3p. Ectopic expression of either miR-145-5p or miR-145-3p in BC cells significantly suppressed cancer cell growth, migration and invasion and it also induced apoptosis. The gene encoding ubiquitin-like with PHD and ring finger domains 1 (UHRF1) was a direct target of these miRNAs. Silencing of UHRF1 induced apoptosis and inhibited cancer cell proliferation, migration, and invasion in BC cells. In addition, overexpressed UHRF1 was confirmed in BC clinical specimens, and the high UHRF1 expression group showed a significantly poorer cause specific survival rate in comparison with the low expression group. Taken together, our present data demonstrated that both strands of miR-145 (miR-145-5p: guide-strand and miR-145-3p: passenger-strand) play pivotal roles in BC cells by regulating UHRF1. The identification of the molecular target of a tumor suppressive miRNAs provides novel insights into the potential mechanisms of BC oncogenesis and suggests novel therapeutic strategies.

Keywords: miR-145-5p, miR-145-3p, tumor-suppressor, UHRF1, bladder cancer

INTRODUCTION

In 2012, more than 400,000 new cases of bladder cancer (BC) were diagnosed and 165,000 patients died worldwide [1]. As for the prevalence of BC, men are three times more frequently diagnosed with BC than women [2]. The reasons for this disparity between sexes are not fully understood. BC is pathologically classified into two groups: non-muscle-invasive BC (NMIBC) and muscle-invasive BC (MIBC). Most BC patients (approximately 50%–80%) are diagnosed with NMIBC and this disease can be treated by removing the tumor by transurethral approaches [3]. In NMIBC, disease may recur, and some patients (approximately 25%) progress to MIBC [3]. Patients with advanced BC are generally treated with combination chemotherapy (gemcitabine and cisplatin), but progression-free survival is of limited duration [4]. Therefore, it is important to elucidate the molecular mechanisms of recurrence and invasiveness of BC cells to develop new treatment strategies.

The discovery of non-coding RNA in the human genome changed approaches in cancer research [5, 6]. Molecular mechanisms of post transcriptional gene regulation by protein-coding RNA/non-coding RNA networks are being studied on a genome-wide scale. MicroRNA (miRNA) is a class of small non-coding RNAs, and they are known to be involved in the repression or degradation of target RNA transcripts in a sequence-dependent manner [7]. A single miRNA can regulate thousands of target transcripts, and more than 60% of protein-coding genes may be influenced by miRNAs [8, 9]. Accumulating evidence indicates that aberrantly expressed miRNAs disturb normally regulated RNA networks, leading to pathologic responses in cancer cells [6]. Strategies to identify aberrant expression of miRNA-mediated cancer pathways are being developed as a new direction in cancer research in the post genome sequencing era.

To seek out differentially expressed miRNAs in BC cells, we used BC clinical specimens to establish deep sequencing-based miRNA expression signatures [10]. In general, the guide-strand RNA from duplex miRNA is retained to direct recruitment of the RNA induced silencing complex (RISC) to target messenger RNAs, whereas the passenger-strand RNA is degraded [1113]. Recently, we revealed that both strands of microRNA (miR)-144-5p and miR-144-3p derived from pre-miR-144 acted as tumor suppressors in BC cells [14]. Moreover, miR-144-5p (passenger-strand) directly targeted cyclin E1 and E2 in BC cells, suggesting that the passenger-strand of miRNA has a physiological role in cells [14].

In this study, we focused on miR-145-5p and miR-145-3p because these miRNAs were significantly downregulated in BC cells as determined in our deep sequencing signature [10]. It is well known that miR- 145- 5p functions as a tumor suppressor in several types of cancer, including BC [15]. However, the role of miR-145-3p on cancer cells is still ambiguous. The aims of the present study were to investigate the anti-tumor effects of miR-145-3p as well as miR-145-5p, and to determine the BC oncogenic pathways and target genes regulated by these miRNAs. The discovery that miR- 145- 5p and miR-145-3p coordinately regulate pathways and targets provides new insight into the mechanisms of BC progression and metastasis.

RESULTS

The expression levels of miR-145-5p and miR-145-3p in BC specimens and cell lines

We evaluated the expression levels of miR-145-5p and miR-145-3p in BC tissues (n = 69), normal bladder epithelia (NBE) (n = 12), and two BC cell lines (T24 and BOY). The expression levels of miR-145-5p and miR- 145- 3p were significantly lower in tumor tissues and BC cell lines compared with NBE (Figure 1A). Spearman's rank test showed a positive correlation between the expression of these miRNAs (r = 0.986 and P < 0.0001) (Figure 1B). On the other hand, there were no significant relationships between any of the clinicopathological parameters (i.e., tumor grade, stage, metastasis, or survival rate) and the expression levels of miR-145-5p and miR-145-3p (data not shown).

Figure 1. The expression levels of miR-145-5p and miR-145-3p, and their effects in BC cells.

Figure 1

(A) Expression levels of miR- 145- 5p and miR-145-3p in clinical specimens and BC cell lines were determined by qRT-PCR. Data were normalized to RNU48 expression. (B) Correlation of miR-145-5p and miR-145-3p expression. (C) Cell growth was determined by XTT assays 72 hours after transfection with 10 nM miR-145-5p or miR-145-3p. *P < 0.0001. (D) Cell migration activity was determined by the wound-healing assays. *P < 0.0001. (E) Cell invasion activity was determined using Matrigel invasion assays. *P < 0.0001.

Effect of restoring miR-145-5p or miR-145- 3p expression on cell growth, migration, and invasion in BC cell lines

We performed gain-of-function studies using transfection of these miRNAs to investigate their functional roles. XTT, cell migration, and invasion assays demonstrated that cell proliferation, cell migration, and cell invasion were significantly inhibited in miR-145-5p and miR-145-3p transfectants in comparison with mock or miR-control transfectants (each P < 0.0001, Figure 1C, 1D, and 1E). These results suggested that miR-145-3p as well as miR-145-5p could have a tumor suppressive function in BC cells.

To investigate the synergistic effects of miR- 145- 5p and miR-145-3p, we performed proliferation, migration, and invasion assays with co-transfection of miR- 145-5p and miR-145-3p in BC cells (T24 and BOY), but they did not show synergistic effects of these miRNAs transfection (Supplementary Figure 1).

Effects of miR-145-5p and miR-145-3p transfection on apoptosis and cell cycle in BC cell lines

Because miR-145-5p and miR-145-3p transfection strongly inhibited cell proliferation in BC cell lines, we hypothesized that these miRNAs may induce apoptosis. Hence, we performed flow cytometric analyses to determine the number of apoptotic cells following restoration of miR- 145-5p or miR-145-3p expression.

The apoptotic cell numbers (apoptotic and early apoptotic cells) were significantly larger in miR-145-5p or miR-145-3p transfectants than in mock or miR-control transfectants (Figure 2A and 2C). Western blot analyses showed that cleaved PARP expression was significantly increased in miR-145-5p or miR-145-3p transfectants compared with mock or miR-control transfectants (Figure 2B and 2D).

Figure 2. Effects of miR-145-5p and miR-145-3p on apoptosis.

Figure 2

(A, C) Apoptosis assays were carried out using flow cytometry. Early apoptotic cells are in area R4 and apoptotic cells are in area R2. The normalized ratios of apoptotic cells are shown in the histograms. Cycloheximide (2 μg/mL) was used as positive control. *P = 0.0266 and **P < 0.0001. (B, D) Western blot analyses for apoptotic markers (cleaved PARP) in BC cell lines. GAPDH was used as a loading control.

We also investigated the cell cycle assays using miR-145-5p and miR-145-3p transfectants. The fraction of cells in the G2/M phase was significantly larger in miR-145-5p and miR-145-3p transfectants in T24 cells in comparison with mock or miR-control transfectants (Supplementary Figure 2). In contrast, miR-145-5p and miR-145-3p transfection induced cell cycle arrest at the G1 phase in BOY cells (Supplementary Figure 2). The reason why the cell cycle arrest (G2 arrest in T24 and G1 arrest in BOY) varies according to a cell types is a future problem.

Identification of common target genes regulated by miR-145-5p and miR-145-3p in BC cells

To gain further insight into the molecular mechanisms and pathways regulated by tumor suppressive miR-145-5p and miR-145-3p in BC cells, we used a combination of in silico analyses and gene expression analyses. Figure 3 shows our strategy to narrow down the common target genes of miR-145-5p and miR-145-3p.

Figure 3. Flow chart illustrates the strategy for analysis of miR-145-5p and miR-145-3p target genes.

Figure 3

A total of 4,555 and 6,295 downregulated genes in expression analysis of miR-145-5p and miR-145-3p transfected BC cell lines, respectively, (T24 and BOY) were selected as putative target genes. Next we merged the data of those selected genes and the microRNA.org database. The analyses showed 398 common putative target genes between miR-145-5p and miR-145-3p. We then analyzed gene expression with available GEO data sets (GSE11783 + GSE31684). The analyses showed that 79 genes were significantly upregulated in BC specimens compared with NBE.

In gene expression analyses, a total of 4,555 and 6,295 genes were downregulated in miR-145-5p and miR- 145-3p transfectants, respectively, in comparison with control transfectants (Gene Expression Omnibus (GEO), accession number: GSE66498). Of those downregulated genes, 1,735 and 1,680 genes, respectively, had putative binding sites for miR-145-5p and miR-145- 3p in their 3′ untranslated regions (UTRs) according to the microRNA.org database. We found that there were 398 common genes targeted by both miRNAs, and among them, we ultimately identified 79 genes that were upregulated in the clinical BC samples from the GEO (accession numbers: GSE11783, GSE31684) (Table 1). We subsequently focused on the ubiquitin-like with PHD and ring finger domains 1 (UHRF1) gene because it was the top ranked gene in the list.

Table 1. Highly expressed genes putatively regulated by miR-145-5p and miR-145-3p.

Entrez Gene ID Gene Symbol Description Genomic location Gene Expression Omnibus (GSE11783 + GSE31684) Expression in miR-145-5ptransfectant (Log2 FC) Expression in miR-145-3ptransfectant (Log2 FC)
Expression Log2 FC P-value T24 BOY T24 BOY
29128 UHRF1 ubiquitin-like with PHD and ring finger domains 1 19p13.3 up 4.984 1.049E-03 −0.041 −0.274 −0.334 −0.901
54972 TMEM132A transmembrane protein 132A 11q12.2 up 3.458 1.049E-03 −0.006 −0.087 −0.178 −0.140
4288 MKI67 marker of proliferation Ki-67 10q26.2 up 3.182 1.049E-03 −0.070 −0.022 −0.609 −0.872
1111 CHEK1 checkpoint kinase 1 11q24.2 up 2.841 1.049E-03 −0.354 −0.204 −0.426 −0.583
25886 POC1A POC1 centriolar protein A 3p21.2 up 2.354 1.049E-03 −0.146 −0.194 −0.251 −0.161
400745 SH2D5 SH2 domain containing 5 1p36.12 up 2.299 1.049E-03 −0.512 −0.075 −0.136 −0.038
55215 FANCI Fanconi anemia, complementation group I 15q26.1 up 2.188 1.049E-03 −0.031 −0.079 −0.281 −0.320
51512 GTSE1 G-2 and S-phase expressed 1 22q13.31 up 2.147 1.049E-03 −0.028 −0.149 −0.713 −0.209
157570 ESCO2 establishment of sister chromatid cohesion N-acetyltransferase 2 8p21.1 up 2.028 1.049E-03 −0.441 −0.352 −0.585 −0.166
2175 FANCA Fanconi anemia, complementation group A 16q24.3 up 1.877 1.049E-03 −0.017 −0.166 −0.412 −0.532
6624 FSCN1 fascin homolog 1, actin-bundling protein (Strongylocentrotus purpuratus) 7p22.1 up 1.829 2.942E-03 −2.899 −0.732 −0.175 −1.133
22979 EFR3B EFR3 homolog B (S. cerevisiae) 2p23.3 up 1.803 1.247E-03 −0.312 −0.033 −1.189 −1.625
3918 LAMC2 laminin, gamma 2 1q25.3 up 1.797 1.791E-02 −0.839 −0.707 −0.125 −0.608
8349 HIST2H2BE histone cluster 2, H2be 1q21.2 up 1.764 1.524E-03 −0.266 −0.149 −0.524 −0.170
9455 HOMER2 homer homolog 2 (Drosophila) 15q25.2 up 1.706 2.526E-03 −0.360 −0.278 −0.132 −0.305
25902 MTHFD1L methylenetetrahydrofolate dehydrogenase (NADP+ dependent) 1-like 6q25.1 up 1.611 1.049E-03 −0.307 −0.024 −0.617 −0.505
55732 C1orf112 chromosome 1 open reading frame 112 1q24.2 up 1.461 1.685E-03 −0.099 −0.147 −0.030 −0.132
388389 CCDC103 coiled-coil domain containing 103 17q21.31 up 1.390 3.290E-02 −0.327 −0.266 −2.471 −1.838
6566 SLC16A1 solute carrier family 16 (monocarboxylate transporter), member 1 1p13.2 up 1.359 3.893E-02 −0.229 −0.137 −0.759 −1.259
23178 PASK PAS domain containing serine/threonine kinase 2q37.3 up 1.333 1.058E-03 −0.016 −0.001 −0.218 −0.443
5426 POLE polymerase (DNA directed), epsilon, catalytic subunit 12q24.33 up 1.241 1.247E-03 −0.094 −0.424 −0.295 −0.051
55379 LRRC59 leucine rich repeat containing 59 17q21.33 up 1.233 1.049E-03 −0.155 −0.198 −0.289 −0.283
6715 SRD5A1 steroid-5-alpha-reductase, alpha polypeptide 1 (3-oxo-5 alpha-steroid delta 4-dehydrogenase alpha 1) 5p15.31 up 1.170 5.069E-03 −0.329 −0.018 −0.823 −0.837
4602 MYB v-myb avian myeloblastosis viral oncogene homolog 6q23.3 up 1.160 4.501E-03 −0.105 −0.337 −0.111 −1.418
8940 TOP3B topoisomerase (DNA) III beta 22q11.22 up 1.157 9.078E-03 −0.108 −0.021 −0.840 −1.150
64768 IPPK inositol 1,3,4,5,6-pentakisphosphate 2-kinase 9q22.31 up 1.153 1.072E-03 −0.526 −0.102 −0.630 −0.296
9266 CYTH2 cytohesin 2 19q13.33 up 1.127 1.049E-03 −0.226 −0.104 −0.598 −0.377
221468 TMEM217 transmembrane protein 217 6p21.2 up 1.081 4.734E-02 −0.049 −0.008 −0.033 −0.337
25859 PART1 prostate androgen-regulated transcript 1 (non-protein coding) 5q12.1 up 1.025 4.873E-03 −0.144 −0.212 −0.097 −0.694
8566 PDXK pyridoxal (pyridoxine, vitamin B6) kinase 21q22.3 up 1.014 1.316E-03 −0.039 −0.842 −0.567 −0.558
11072 DUSP14 dual specificity phosphatase 14 17q12 up 1.008 2.440E-03 −0.126 −0.092 −0.924 −1.020
23516 SLC39A14 solute carrier family 39 (zinc transporter), member 14 8p21.3 up 0.999 3.435E-03 −0.540 −0.216 −2.083 −1.548
85414 SLC45A3 solute carrier family 45, member 3 1q32.1 up 0.977 3.435E-03 −0.578 −0.086 −0.782 −0.505
1163 CKS1B CDC28 protein kinase regulatory subunit 1B 1q21.3 up 0.941 1.857E-02 −0.370 −0.229 −0.678 −0.802
79929 MAP6D1 MAP6 domain containing 1 3q27.1 up 0.927 1.093E-03 −0.135 −0.210 −0.928 −0.529
65985 AACS acetoacetyl-CoA synthetase 12q24.31 up 0.919 1.058E-03 −0.555 −0.367 −0.816 −0.798
1263 PLK3 polo-like kinase 3 1p34.1 up 0.910 1.685E-03 −0.229 −0.092 −1.766 −2.103
64785 GINS3 GINS complex subunit 3 (Psf3 homolog) 16q21 up 0.891 1.740E-03 −0.185 −0.218 −0.853 −0.826
4957 ODF2 outer dense fiber of sperm tails 2 9q34.11 up 0.854 1.185E-03 −0.232 −0.409 −0.610 −0.963
57613 KIAA1467 KIAA1467 12p13.1 up 0.837 4.169E-03 −0.382 −0.282 −0.398 −0.456
7525 YES1 v-yes-1 Yamaguchi sarcoma viral oncogene homolog 1 18p11.32 up 0.794 2.526E-03 −0.382 −0.447 −0.256 −0.446
8751 ADAM15 ADAM metallopeptidase domain 15 1q22 up 0.787 6.433E-03 −0.233 −0.217 −0.383 −0.318
7172 TPMT thiopurine S-methyltransferase 6p22.3 up 0.786 1.524E-03 −0.167 −0.032 −0.482 −0.323
4615 MYD88 myeloid differentiation primary response 88 3p22.2 up 0.759 1.947E-03 −0.662 −0.118 −0.286 −0.113
1678 TIMM8A translocase of inner mitochondrial membrane 8 homolog A (yeast) Xq22.1 up 0.729 2.723E-03 −0.530 −0.187 −0.201 −0.267
3927 LASP1 LIM and SH3 protein 1 17q12 up 0.692 2.348E-03 −0.280 −0.014 −0.319 −0.069
10295 BCKDK branched chain ketoacid dehydrogenase kinase 16p11.2 up 0.685 6.186E-03 −0.281 −0.161 −0.439 −0.246
26088 GGA1 golgi-associated, gamma adaptin ear containing, ARF binding protein 1 22q13.1 up 0.668 1.049E-03 −0.010 −0.074 −0.180 −0.202
6240 RRM1 ribonucleotide reductase M1 11p15.4 up 0.667 4.582E-02 −0.206 −0.207 −1.158 −2.292
219902 TMEM136 transmembrane protein 136 11q23.3 up 0.667 3.574E-03 −0.449 −0.477 −0.386 −0.405
7019 TFAM transcription factor A, mitochondrial 10q21.1 up 0.644 1.274E-02 −0.163 −0.413 −0.543 −0.609
55775 TDP1 tyrosyl-DNA phosphodiesterase 1 14q32.11 up 0.624 1.316E-03 −0.151 −0.193 −0.651 −0.188
79858 NEK11 NIMA-related kinase 11 3q22.1 up 0.613 1.626E-03 −0.628 −0.563 −0.179 −0.189
1889 ECE1 endothelin converting enzyme 1 1p36.12 up 0.604 3.635E-02 −0.949 −0.274 −0.559 −0.639
65264 UBE2Z ubiquitin-conjugating enzyme E2Z 17q21.32 up 0.590 1.348E-03 −0.352 −0.187 −0.895 −1.241
9205 ZMYM5 zinc finger, MYM-type 5 13q12.11 up 0.582 7.805E-03 −0.413 −0.381 −0.699 −0.890
996 CDC27 cell division cycle 27 17q21.32 up 0.572 9.799E-03 −0.486 −0.018 −0.260 −0.099
22898 DENND3 DENN/MADD domain containing 3 8q24.3 up 0.570 1.016E-02 −0.235 −0.012 −0.597 −0.926
84314 TMEM107 transmembrane protein 107 17p13.1 up 0.570 2.965E-02 −0.471 −0.208 −0.199 −0.839
85464 SSH2 slingshot protein phosphatase 2 17q11.2 up 0.562 2.440E-03 −0.296 −0.173 −0.433 −0.220
56180 MOSPD1 motile sperm domain containing 1 Xq26.3 up 0.559 1.928E-02 −0.145 −0.237 −1.352 −1.270
6625 SNRNP70 small nuclear ribonucleoprotein 70kDa (U1) 19q13.33 up 0.554 1.725E-02 −0.373 −0.281 −0.663 −0.988
60490 PPCDC phosphopantothenoyl­-
cysteine decarboxylase
15q24.2 up 0.550 1.182E-02 −0.269 −0.338 −0.057 −0.130
147657 ZNF480 zinc finger protein 480 19q13.41 up 0.547 3.893E-02 −0.453 −0.035 −0.107 −0.047
159090 FAM122B family with sequence similarity 122B Xq26.3 up 0.543 2.865E-02 −0.356 −0.131 −1.379 −1.493
3150 HMGN1 high mobility group nucleosome binding
domain 1
21q22.2 up 0.522 7.521E-03 −0.884 −0.157 −0.162 −0.119
7421 VDR vitamin D (1,25-dihydroxyvitamin D3) receptor 12q13.11 up 0.494 3.290E-02 −0.001 −0.069 −0.428 −0.417
84705 GTPBP3 GTP binding protein 3 (mitochondrial) 19p13.11 up 0.485 1.999E-02 −0.156 −0.048 −0.488 −1.061
84818 IL17RC interleukin 17 receptor C 3p25.3 up 0.478 8.102E-03 −0.306 −0.009 −0.053 −0.194
10102 TSFM Ts translation elongation factor, mitochondrial 12q14.1 up 0.475 4.873E-03 −0.170 −0.026 −0.951 −0.608
27 ABL2 c-abl oncogene 2, non-receptor tyrosine kinase 1q25.2 up 0.455 9.799E-03 −0.211 −0.281 −0.230 −0.102
55285 RBM41 RNA binding motif
protein 41
Xq22.3 up 0.415 1.538E-02 −0.055 −0.215 −0.495 −0.559
57532 NUFIP2 nuclear fragile X mental retardation protein interacting protein 2 17q11.2 up 0.397 1.056E-02 −0.098 −0.256 −0.425 −0.904
84445 LZTS2 leucine zipper, putative tumor suppressor 2 10q24.31 up 0.394 4.155E-02 −0.174 −0.125 −0.288 −0.026
8243 SMC1A structural maintenance of chromosomes 1A Xp11.22 up 0.390 3.635E-02 −0.163 −0.061 −0.917 −0.297
54617 INO80 INO80 complex subunit 15q15.1 up 0.384 2.835E-03 −0.594 −0.006 −0.635 −0.350
7511 XPNPEP1 X-prolyl aminopeptidase (aminopeptidase P) 1, soluble 10q25.1 up 0.381 7.521E-03 −0.648 −0.272 −1.595 −1.701
23367 LARP1 La ribonucleoprotein domain family, member 1 5q33.2 up 0.377 4.155E-02 −0.049 −0.003 −0.091 −0.216
10146 G3BP1 GTPase activating protein (SH3 domain) binding protein 1 5q33.1 up 0.313 4.021E-02 −1.431 −0.040 −0.505 −0.475

UHRF1 was a direct target of miR-145-5p and miR-145-3p in BC cells

We performed quantitative real-time RT-PCR (qRT-PCR) to validate that miR-145-5p and miR-145-3p repressed UHRF1 mRNA expression in BC cell lines, and we did indeed observe that it was significantly reduced in transfectants of these miRNAs in comparison with mock or miR-control transfectants (P < 0.0001 and P = 0.0036, Figure 4A). The protein expression levels of UHRF1 were also repressed in the miRNAs transfectants (Figure 4B).

Figure 4. Direct regulation of UHRF1 by miR-145-5p and miR-145-3p.

Figure 4

(A) UHRF1 mRNA expression was evaluated by qRT-PCR in T24 and BOY 72 hours after transfection with miR-145-5p and miR-145-3p. GUSB was used as an internal control. *P = 0.0036 and **P < 0.0001. (B) UHRF1 protein expression was evaluated by Western blot analyses in T24 and BOY 72–96 hours after transfection with miR-145-5p or miR-145-3p. GAPDH was used as a loading control. (C) miR-145-5p and miR-145-3p binding sites in the 3′ UTR of UHRF1 mRNA. Dual Luciferase reporter assays using vectors encoding putative miR-145-5p and miR-145-3p target sites of the UHRF 3′ UTR (positions 1,179–1,198 and 287–292, respectively) for both wild-type and deleted regions. Normalized data were calculated as ratios of Renilla/firefly luciferase activities. *P < 0.0001.

We carried out dual luciferase reporter assays in T24 and BOY cells to determine whether the UHRF1 gene was directly regulated by miR-145-5p/3p. The microRNA.org database predicted that there was one binding site for miR- 145-5p in the 3′ UTR of UHRF1 (position 1,179– 1,198); for miR-145-3p, there was a binding site in the 3′ UTR at position 287–292. We used vectors encoding the partial wild-type sequence of the 3′ UTR of the mRNA, including the predicted miR-145-5p or miR-145- 3p target sites. We found that the luminescence intensity was significantly reduced by co-transfection with these miRNAs and the vector carrying the wild-type 3′ UTR, whereas no reduction of luminescence was observed by transfection with the deletion vector (binding site had been removed) (P < 0.0001, Figure 4C). These suggested that either of miR-145-5p and miR-145-3p were directly bounded to specific sites in the 3′ UTR of UHRF1 mRNA.

Effects of silencing UHRF1 in BC cell lines

To investigate the functional role of UHRF1 in BC cells, we carried out loss-of-function studies by using si-UHRF1 transfectants. First, we evaluated the knockdown efficiency of si-UHRF1 transfection in BC cell lines. In the present study, we used two types of si- UHRF1 (si- UHRF1-1 and si-UHRF1-2). The qRT- PCR and Western blot analyses showed that both siRNAs effectively downregulated UHRF1 expression in both cell lines (Figure 5A and 5B).

Figure 5. UHRF1 mRNA and protein expression after si-UHRF1 transfection and effects of UHRF1 silencing in BC cell lines.

Figure 5

(A) UHRF1 mRNA expression was evaluated by qRT-PCR in T24 and BOY 72 hours after transfection with si-UHRF1-1 and si-UHRF1-2. GUSB was used as an internal control. (B) UHRF1 protein expression was evaluated by Western blot analysis in T24 and BOY 72 - 96 hours after transfection with miR-145-5p or miR-145-3p. GAPDH was used as a loading control. (C) Cell proliferation was determined with the XTT assays 72 hours after transfection with 10 nM si-UHRF1-1 or si-UHRF1-2. *P < 0.0001. (D) Cell migration activity was determined by wound-healing assays. *P < 0.0001. (E) Cell invasion activity was determined using Matrigel invasion assays. *P < 0.0001.

XTT, cell migration, and invasion assays demonstrated that cell proliferation, cell migration, and cell invasion were inhibited in si-UHRF1 transfectants in comparison with the mock or siRNA-control transfectant cells (each P < 0.0001, Figure 5C, 5D, and 5E).

In the apoptosis assays, the apoptotic cell numbers were significantly greater in si-UHRF1 transfectants than in mock or siRNA-control transfectants (Figure 6A and 6C). Western blot analyses showed that cleaved PARP expression was significantly increased in si-UHRF1 transfectants compared with mock or siRNA-control transfectants (Figure 6B and 6D).

Figure 6. Effects of silencing UHRF1 on apoptosis in BC cell lines.

Figure 6

(A, C) Apoptosis assays were carried out using flow cytometry. Early apoptotic cells are in area R4 and apoptotic cells are in area R2. The normalized ratios of the apoptotic cells are shown in the histogram. Cycloheximide (2 μg/mL) was used as a positive control. *P < 0.0001 (B, D) Western blot analyses for apoptotic markers (cleaved PARP) in BC cell lines. GAPDH was used as a loading control.

Expression of UHRF1 in BC clinical specimens

The qRT-PCR analyses showed that the expression level of UHRF1 mRNA was significantly upregulated in 69 BC specimens and 2 BC cell lines compared with 12 NBE (P < 0.0001, Figure 7A). Spearman's rank test showed negative correlations between miR-145-5p/miR-145-3p expression and UHRF1 mRNA expression (r = −0.324 and −0.298, P = 0.0024 and 0.0051, Figure 7B). As shown in Figure 7C, the expression level of UHRF1 was significantly greater in high grade clinical BCs (P = 0.0135), MIBCs (T2 ≤) (P = 0.0379), BCs with positive lymph node invasion (N1) (P = 0.00182), and in BCs with positive distant metastasis (M1) (P = 0.0307) than in their counterparts. Kaplan-Meier analysis showed that the high UHRF1 expression group had significantly lower cause specific survival probabilities compared to the low UHRF1 expression group (P = 0.0259, Figure 8).

Figure 7. The expression level of UHRF1 mRNA in BC clinical specimens and cell lines, and association of UHRF1 expression with clinicopathological parameters.

Figure 7

(A) Expression levels of UHRF1 in clinical specimens and BC cell lines were determined by qRT-PCR. Data were normalized to GUSB expression. (B) The correlated expression among miR-145-5p, miR-145-3p, and UHRF1. (C) Association of UHRF1 expression with clinicopathological parameters. Relationships between two variables were analyzed using the Mann-Whitney U test.

Figure 8. The association between the expression level of UHRF1 and cause specific survival rate.

Figure 8

Kaplan-Meier survival curves for cause specific survival rates based on UHRF1 expression in 57 BC patients. P-values were calculated using the log-rank test.

We validated the expression status of UHRF1 in BC clinical specimens using immunohistochemical staining. UHRF1 was expressed moderately or strongly in several cancer lesions, and normal bladder tissues stained weakly (Figure 9).

Figure 9. Immunohistochemical staining of UHRF1 in BC clinical specimens.

Figure 9

UHRF1 was expressed more strongly in several cancer lesions than in noncancerous tissues. Left panel, original magnification ×40; Right panel, original magnification ×200. (A) Positively stained tumor lesion (High grade, T2bN0M0), (B) Positively stained tumor lesion (High grade, T1N0M0), (C) Positively stained tumor lesion (Low grade, T3N0M0), (D) Negative staining in normal bladder tissue.

Investigation of downstream genes regulated by UHRF1 in BC cells

To identify the downstream genes regulated by UHRF1, genome-wide gene expression analyses and in silico analyses were performed in two BC cell lines transfected with si-UHRF1. A total of 533 genes were downregulated (log2 FC < −1.5) by si-UHRF1 transfection, and a total of 704 genes were upregulated (log2 FC > 1.0) by si-UHRF1 transfection compared with negative control cells (GEO, accession number: GSE77790). Among the downregulated genes in the si-UHRF1 transfectants, 104 genes were upregulated in the BC clinical samples from GEO database (accession numbers: GSE11783, GSE31684), whereas among the upregulated genes, 62 genes were downregulated in the clinical BCs. These results imply that the 104 upregulated genes may act as oncogenes, and the 62 downregulated genes may act as tumor suppressors downstream from UHRF1 in BC (Tables 2 and 3).

Table 2. Significantly downregulated genes by si-UHRF1 in BC cell lines.

Entrez Gene ID Gene Symbol Description Genomic location Gene Expression Omnibus
(GSE11783 + GSE31684)
Expression
in si-UHRF1
transfectant
(Log2 FC)
Expression Log2FC P-value T24 BOY
7153 TOP2A topoisomerase (DNA) II alpha 170kDa 17q21.2 up 6.312 1.049E-03 −1.880 −1.681
29128 UHRF1 ubiquitin-like with PHD and ring finger domains 1 19p13.3 up 4.984 1.049E-03 −3.213 −2.907
259266 ASPM asp (abnormal spindle) homolog, microcephaly associated (Drosophila) 1q31.3 up 4.299 1.049E-03 −3.431 −3.444
332 BIRC5 baculoviral IAP repeat containing 5 17q25.3 up 4.110 1.049E-03 −2.258 −1.777
9928 KIF14 kinesin family member 14 1q32.1 up 3.866 1.049E-03 −3.294 −1.544
1063 CENPF centromere protein F, 350/400kDa 1q41 up 3.576 1.049E-03 −2.613 −3.307
1894 ECT2 epithelial cell transforming 2 3q26.31 up 3.469 1.049E-03 −1.928 −1.813
55247 NEIL3 nei endonuclease VIII-like 3 (E. coli) 4q34.3 up 3.428 1.049E-03 −1.728 −2.065
9401 RECQL4 RecQ protein-like 4 8q24.3 up 3.414 1.049E-03 −1.751 −2.102
3832 KIF11 kinesin family member 11 10q23.33 up 3.356 1.049E-03 −2.299 −1.657
57082 CASC5 cancer susceptibility candidate 5 15q15.1 up 3.230 1.049E-03 −2.470 −2.188
151176 FAM132B family with sequence similarity 132, member B 2q37.3 up 3.100 1.058E-03 −2.420 −2.184
151246 SGOL2 shugoshin-like 2 (S. pombe) 2q33.1 up 2.694 1.049E-03 −3.124 −2.407
1062 CENPE centromere protein E, 312kDa 4q24 up 2.689 1.058E-03 −3.676 −3.218
23529 CLCF1 cardiotrophin-like cytokine factor 1 11q13.2 up 2.646 1.049E-03 −1.905 −2.363
81930 KIF18A kinesin family member 18A 11p14.1 up 2.553 1.049E-03 −3.246 −2.128
7130 TNFAIP6 tumor necrosis factor, alpha-induced protein 6 2q23.3 up 2.531 2.835E-03 −1.795 −2.735
55502 HES6 hes family bHLH transcription factor 6 2q37.3 up 2.506 6.688E-03 −1.572 −1.508
5328 PLAU plasminogen activator, urokinase 10q22.2 up 2.244 1.740E-03 −2.417 −1.791
9824 ARHGAP11A Rho GTPase activating protein 11A 15q13.3 up 2.051 2.348E-03 −1.675 −1.613
23057 NMNAT2 nicotinamide nucleotide adenylyltransferase 2 1q25.3 up 2.050 1.247E-03 −1.707 −1.863
59285 CACNG6 calcium channel, voltage-dependent, gamma subunit 6 19q13.42 up 2.016 1.049E-03 −1.502 −1.763
675 BRCA2 breast cancer 2, early onset 13q13.1 up 2.015 1.049E-03 −1.764 −2.356
6524 SLC5A2 solute carrier family 5 (sodium/glucose cotransporter), member 2 16p11.2 up 1.900 1.214E-03 −1.855 −1.569
79412 KREMEN2 kringle containing transmembrane protein 2 16p13.3 up 1.893 1.348E-03 −2.309 −1.796
6274 S100A3 S100 calcium binding protein A3 1q21.3 up 1.825 8.102E-03 −2.215 −1.848
5331 PLCB3 phospholipase C, beta 3 (phosphatidylinositol-specific) 11q13.1 up 1.790 1.049E-03 −2.219 −1.735
55349 CHDH choline dehydrogenase 3p21.1 up 1.743 1.049E-03 −1.926 −2.008
811 CALR calreticulin 19p13.2 up 1.652 1.049E-03 −1.554 −1.500
4987 OPRL1 opiate receptor-like 1 20q13.33 up 1.627 2.626E-03 −1.927 −1.766
375248 ANKRD36 ankyrin repeat domain 36 2q11.2 up 1.530 8.102E-03 −3.873 −1.791
441054 C4orf47 chromosome 4 open reading frame 47 4q35.1 up 1.485 2.151E-02 −2.229 −2.522
201475 RAB12 RAB12, member RAS oncogene family 18p11.22 up 1.468 1.058E-03 −2.353 −2.947
286151 FBXO43 F-box protein 43 8q22.2 up 1.463 2.396E-02 −1.528 −2.082
9091 PIGQ phosphatidylinositol glycan anchor biosynthesis, class Q 16p13.3 up 1.434 3.574E-03 −1.594 −1.693
81575 APOLD1 apolipoprotein L domain containing 1 12p13.1 up 1.354 1.808E-03 −2.237 −2.383
132320 SCLT1 sodium channel and clathrin linker 1 4q28.2 up 1.340 1.049E-03 −3.140 −3.098
100131211 TMEM194B transmembrane protein 194B 2q32.2 up 1.325 1.049E-03 −1.573 −1.967
153642 ARSK arylsulfatase family, member K 5q15 up 1.252 1.049E-03 −2.052 −1.875
21 ABCA3 ATP-binding cassette, sub-family A (ABC1), member 3 16p13.3 up 1.170 4.892E-02 −1.879 −1.831
55036 CCDC40 coiled-coil domain containing 40 17q25.3 up 1.160 1.049E-03 −1.562 −1.531
84259 DCUN1D5 DCN1, defective in cullin neddylation 1, domain containing 5 11q22.3 up 1.151 1.247E-03 −1.591 −1.993
80381 CD276 CD276 molecule 15q24.1 up 1.146 1.072E-03 −2.656 −2.096
6487 ST3GAL3 ST3 beta-galactoside alpha-2,3-sialyltransferase 3 1p34.1 up 1.139 1.049E-03 −1.828 −2.380
5351 PLOD1 procollagen-lysine, 2-oxoglutarate 5-dioxygenase 1 1p36.22 up 1.104 2.942E-03 −1.650 −1.570
343099 CCDC18 coiled-coil domain containing 18 1p22.1 up 1.075 1.578E-03 −3.521 −2.428
30818 KCNIP3 Kv channel interacting protein 3, calsenilin 2q11.1 up 1.069 2.723E-03 −3.678 −2.733
10051 SMC4 structural maintenance of chromosomes 4 3q25.33 up 1.066 1.578E-03 −2.612 −1.745
51427 ZNF107 zinc finger protein 107 7q11.21 up 1.040 1.316E-03 −2.527 −2.104
10592 SMC2 structural maintenance of chromosomes 2 9q31.1 up 1.032 6.688E-03 −3.520 −2.180
20 ABCA2 ATP-binding cassette, sub-family A (ABC1), member 2 9q34.3 up 0.965 1.372E-02 −1.511 −2.291
55183 RIF1 replication timing regulatory factor 1 2q23.3 up 0.960 1.058E-03 −1.712 −1.605
9898 UBAP2L ubiquitin associated protein 2-like 1q21.3 up 0.952 1.049E-03 −1.587 −2.301
29780 PARVB parvin, beta 22q13.31 up 0.952 1.096E-02 −3.288 −1.888
9585 KIF20B kinesin family member 20B 10q23.31 up 0.933 5.720E-03 −2.282 −3.122
9534 ZNF254 zinc finger protein 254 19p12 up 0.920 3.863E-03 −2.072 −2.662
57520 HECW2 HECT, C2 and WW domain containing E3 ubiquitin protein ligase 2 2q32.3 up 0.884 3.179E-03 −1.838 −1.958
84083 ZRANB3 zinc finger, RAN-binding domain containing 3 2q21.3 up 0.873 1.578E-03 −1.987 −1.915
6498 SKIL SKI-like proto-oncogene 3q26.2 up 0.859 1.808E-03 −2.709 −1.845
64770 CCDC14 coiled-coil domain containing 14 3q21.1 up 0.842 6.943E-03 −2.453 −1.711
254065 BRWD3 bromodomain and WD repeat domain containing 3 Xq21.1 up 0.808 1.393E-03 −1.852 −2.546
22973 LAMB2P1 laminin, beta 2 pseudogene 1 3p21.31 up 0.804 7.521E-03 −2.336 −2.311
7525 YES1 YES proto-oncogene 1, Src family tyrosine kinase 18p11.32 up 0.794 2.526E-03 −3.127 −2.099
1984 EIF5A eukaryotic translation initiation factor 5A 17p13.1 up 0.793 5.486E-03 −2.297 −2.018
22852 ANKRD26 ankyrin repeat domain 26 10p12.1 up 0.787 3.303E-03 −2.798 −2.663
23322 RPGRIP1L RPGRIP1-like 16q12.2 up 0.778 1.182E-02 −1.517 −1.806
79677 SMC6 structural maintenance of chromosomes 6 2p24.2 up 0.764 8.401E-03 −1.909 −2.083
84920 ALG10 ALG10, alpha-1,2-glucosyltransferase 12p11.1 up 0.763 6.688E-03 −1.828 −2.360
8570 KHSRP KH-type splicing regulatory protein 19p13.3 up 0.762 3.303E-03 −1.767 −1.820
5819 PVRL2 poliovirus receptor-related 2 (herpesvirus entry mediator B) 19q13.32 up 0.757 9.078E-03 −3.014 −2.465
51575 ESF1 ESF1, nucleolar pre-rRNA processing protein, homolog (S. cerevisiae) 20p12.1 up 0.755 9.430E-03 −1.786 −1.732
51361 HOOK1 hook microtubule-tethering protein 1 1p32.1 up 0.689 3.067E-02 −2.156 −2.000
10198 MPHOSPH9 M-phase phosphoprotein 9 12q24.31 up 0.667 1.947E-03 −2.113 −1.502
4983 OPHN1 oligophrenin 1 Xq12 up 0.632 5.277E-03 −2.278 −1.747
4976 OPA1 optic atrophy 1 (autosomal dominant) 3q29 up 0.619 2.169E-03 −2.190 −1.526
168850 ZNF800 zinc finger protein 800 7q31.33 up 0.611 1.227E-02 −1.807 −1.867
26272 FBXO4 F-box protein 4 5p13.1 up 0.611 3.512E-02 −2.224 −2.445
7390 UROS uroporphyrinogen III synthase 10q26.13 up 0.605 6.433E-03 −3.120 −2.062
4683 NBN nibrin 8q21.3 up 0.590 5.720E-03 −2.986 −1.966
79670 ZCCHC6 zinc finger, CCHC domain containing 6 9q21.33 up 0.587 5.486E-03 −2.353 −1.839
79573 TTC13 tetratricopeptide repeat domain 13 1q42.2 up 0.587 6.943E-03 −1.740 −2.064
50840 TAS2R14 taste receptor, type 2, member 14 12p13.2 up 0.574 1.598E-02 −1.947 −1.509
79042 TSEN34 TSEN34 tRNA splicing endonuclease subunit 19q13.42 up 0.570 1.138E-02 −2.455 −1.761
6801 STRN striatin, calmodulin binding protein 2p22.2 up 0.563 2.723E-03 −1.964 −2.434
3597 IL13RA1 interleukin 13 receptor, alpha 1 Xq24 up 0.552 2.075E-02 −2.460 −2.403
147657 ZNF480 zinc finger protein 480 19q13.41 up 0.547 3.893E-02 −3.434 −3.276
8683 SRSF9 serine/arginine-rich splicing factor 9 12q24.31 up 0.534 1.227E-02 −1.523 −2.098
252983 STXBP4 syntaxin binding protein 4 17q22 up 0.516 2.151E-02 −1.776 −1.599
284325 C19orf54 chromosome 19 open reading frame 54 19q13.2 up 0.510 4.734E-02 −1.614 −2.171
91147 TMEM67 transmembrane protein 67 8q22.1 up 0.509 9.799E-03 −1.647 −2.069
114799 ESCO1 establishment of sister chromatid cohesion N-acetyltransferase 1 18q11.2 up 0.495 4.873E-03 −2.173 −2.401
57670 KIAA1549 KIAA1549 7q34 up 0.480 4.582E-02 −2.127 −1.789
6103 RPGR retinitis pigmentosa GTPase regulator Xp11.4 up 0.467 3.290E-02 −1.583 −2.025
5700 PSMC1 proteasome (prosome, macropain) 26S subunit, ATPase, 1 14q32.11 up 0.449 1.274E-02 −1.639 −1.711
253260 RICTOR RPTOR independent companion of MTOR, complex 2 5p13.1 up 0.442 2.666E-02 −2.458 −1.683
23241 PACS2 phosphofurin acidic cluster sorting protein 2 14q32.33 up 0.442 3.179E-03 −3.416 −2.028
27154 BRPF3 bromodomain and PHD finger containing, 3 6p21.31 up 0.440 5.720E-03 −1.772 −2.598
7703 PCGF2 polycomb group ring finger 2 17q12 up 0.439 2.865E-02 −1.828 −1.974
51105 PHF20L1 PHD finger protein 20-like 1 8q24.22 up 0.383 9.078E-03 −3.492 −2.007
57697 FANCM Fanconi anemia, complementation group M 14q21.2 up 0.364 3.067E-02 −1.648 −1.627
9730 VPRBP Vpr (HIV-1) binding protein 3p21.2 up 0.363 2.075E-02 −2.342 −1.568
5378 PMS1 PMS1 postmeiotic segregation increased 1 (S. cerevisiae) 2q32.2 up 0.350 4.734E-02 −2.701 −1.616
255520 ELMOD2 ELMO/CED-12 domain containing 2 4q31.1 up 0.334 4.582E-02 −2.360 −1.637
80124 VCPIP1 valosin containing protein (p97)/p47 complex interacting protein 1 8q13.1 up 0.304 3.893E-02 −3.107 −2.286

Table 3. Significantly upregulated genes by si-UHRF1 in BC cell lines.

Entrez Gene ID Gene Symbol Description Genomic location Gene Expression Omnibus
(GSE11783 + GSE31684)
Expression
in si-UHRF1
transfectant
(Log2 FC)
Expression Log2FC P-value T24 BOY
3043 HBB hemoglobin, beta 11p15.4 down −3.263 1.214E-03 1.204 2.109
137835 TMEM71 transmembrane protein 71 8q24.22 down −2.428 4.873E-03 2.813 3.920
8639 AOC3 amine oxidase, copper containing 3 17q21.31 down −2.188 1.434E-03 1.907 3.140
1408 CRY2 cryptochrome circadian clock 2 11p11.2 down −2.141 1.058E-03 2.134 2.108
7644 ZNF91 zinc finger protein 91 19p12 down −2.058 1.155E-03 1.435 2.063
197257 LDHD lactate dehydrogenase D 16q23.1 down −1.626 2.965E-02 1.844 1.362
316 AOX1 aldehyde oxidase 1 2q33.1 down −1.601 2.169E-03 1.841 1.049
26051 PPP1R16B protein phosphatase 1, regulatory subunit 16B 20q11.23 down −1.547 6.688E-03 1.076 1.198
63976 PRDM16 PR domain containing 16 1p36.32 down −1.439 2.075E-02 2.639 3.846
254827 NAALADL2 N-acetylated alpha-linked acidic dipeptidase-like 2 3q26.31 down −1.313 4.873E-03 1.621 3.168
154 ADRB2 adrenoceptor beta 2, surface 5q32 down −1.242 9.799E-03 2.384 2.302
10477 UBE2E3 ubiquitin-conjugating enzyme E2E 3 2q31.3 down −1.117 1.135E-03 1.053 2.755
7099 TLR4 toll-like receptor 4 9q33.1 down −1.053 6.943E-03 1.402 2.356
57478 USP31 ubiquitin specific peptidase 31 16p12.2 down −1.037 4.169E-03 1.570 1.234
57185 NIPAL3 NIPA-like domain containing 3 1p36.11 down −0.986 1.316E-03 1.329 1.189
30815 ST6GALNAC6 ST6 (alpha-N-acetyl-neuraminyl-2,3-beta-galactosyl-1,3)-N-acetylgalactosaminide alpha-2,6-sialyltransferase 6 9q34.11 down −0.936 1.660E-02 1.093 2.348
29915 HCFC2 host cell factor C2 12q23.3 down −0.928 1.393E-03 1.304 1.296
54741 LEPROT leptin receptor overlapping transcript 1p31.3 down −0.893 1.049E-03 1.280 2.248
7779 SLC30A1 solute carrier family 30 (zinc transporter), member 1 1q32.3 down −0.879 8.736E-03 1.267 1.262
79027 ZNF655 zinc finger protein 655 7q22.1 down −0.863 1.393E-03 1.570 1.589
64344 HIF3A hypoxia inducible factor 3, alpha subunit 19q13.32 down −0.845 1.016E-02 1.284 2.411
79844 ZDHHC11 zinc finger, DHHC-type containing 11 5p15.33 down −0.834 3.176E-02 1.505 1.890
79815 NIPAL2 NIPA-like domain containing 2 8q22.2 down −0.825 6.688E-03 1.929 1.259
7923 HSD17B8 hydroxysteroid (17-beta) dehydrogenase 8 6p21.32 down −0.821 3.512E-02 2.657 3.759
8629 JRK Jrk homolog (mouse) 8q24.3 down −0.820 1.740E-03 1.358 2.076
79591 C10orf76 chromosome 10 open reading frame 76 10q24.32 down −0.812 1.808E-03 1.099 1.917
599 BCL2L2 BCL2-like 2 14q11.2 down −0.775 2.835E-03 1.384 1.730
412 STS steroid sulfatase (microsomal), isozyme S Xp22.31 down −0.770 1.372E-02 1.440 1.471
56900 TMEM167B transmembrane protein 167B 1p13.3 down −0.755 2.626E-03 2.282 2.366
23509 POFUT1 protein O-fucosyltransferase 1 20q11.21 down −0.747 1.274E-02 1.400 2.132
25923 ATL3 atlastin GTPase 3 11q12.3 down −0.727 3.290E-02 1.179 1.907
79669 C3orf52 chromosome 3 open reading frame 52 3q13.2 down −0.708 4.021E-02 1.200 1.482
55844 PPP2R2D protein phosphatase 2, regulatory subunit B, delta 10q26.3 down −0.691 2.666E-02 1.422 1.303
5939 RBMS2 RNA binding motif, single stranded interacting protein 2 12q13.3 down −0.626 5.943E-03 1.193 1.438
6158 RPL28 ribosomal protein L28 19q13.42 down −0.618 1.808E-03 2.026 3.427
2145 EZH1 enhancer of zeste 1 polycomb repressive complex 2 subunit 17q21.2 down −0.618 1.393E-03 1.391 1.171
388969 C2orf68 chromosome 2 open reading frame 68 2p11.2 down −0.611 3.435E-03 1.309 1.192
55422 ZNF331 zinc finger protein 331 19q13.42 down −0.594 1.725E-02 2.855 2.230
92400 RBM18 RNA binding motif protein 18 9q33.2 down −0.594 8.401E-03 1.172 2.001
80017 C14orf159 chromosome 14 open reading frame 159 14q32.11 down −0.590 1.182E-02 1.072 1.748
7556 ZNF10 zinc finger protein 10 12q24.33 down −0.563 1.480E-02 1.592 1.127
55957 LIN37 lin-37 DREAM MuvB core complex component 19q13.12 down −0.543 1.857E-02 1.002 1.205
84267 C9orf64 chromosome 9 open reading frame 64 9q21.32 down −0.543 5.720E-03 1.215 1.299
8799 PEX11B peroxisomal biogenesis factor 11 beta 1q21.1 down −0.535 4.679E-03 1.083 1.163
8790 FPGT fucose-1-phosphate guanylyltransferase 1p31.1 down −0.524 2.075E-02 1.680 1.222
6992 PPP1R11 protein phosphatase 1, regulatory (inhibitor) subunit 11 6p22.1 down −0.517 6.433E-03 1.104 1.329
116224 FAM122A family with sequence similarity 122A 9q21.11 down −0.507 2.169E-03 1.231 1.549
51710 ZNF44 zinc finger protein 44 19p13.2 down −0.499 1.372E-02 2.385 1.001
7265 TTC1 tetratricopeptide repeat domain 1 5q33.3 down −0.487 1.182E-02 1.109 1.112
80213 TM2D3 TM2 domain containing 3 15q26.3 down −0.485 1.182E-02 1.342 1.742
81631 MAP1LC3B microtubule-associated protein 1 light chain 3 beta 16q24.2 down −0.480 1.725E-02 1.210 2.109
6016 RIT1 Ras-like without CAAX 1 1q22 down −0.473 2.666E-02 1.556 1.432
7247 TSN translin 2q14.3 down −0.467 4.582E-02 1.101 1.496
167227 DCP2 decapping mRNA 2 5q22.2 down −0.447 1.016E-02 1.284 1.104
11046 SLC35D2 solute carrier family 35 (UDP-GlcNAc/UDP-glucose transporter), member D2 9q22.32 down −0.431 1.227E-02 1.318 1.340
54946 SLC41A3 solute carrier family 41, member 3 3q21.2 down −0.402 4.294E-02 1.526 1.988
7799 PRDM2 PR domain containing 2, with ZNF domain 1p36.21 down −0.384 7.805E-03 1.438 1.294
6651 SON SON DNA binding protein 21q22.11 down −0.374 5.486E-03 1.126 1.155
80255 SLC35F5 solute carrier family 35, member F5 2q14.1 down −0.369 4.441E-02 1.143 1.619
55197 RPRD1A regulation of nuclear pre-mRNA domain containing 1A 18q12.2 down −0.364 3.893E-02 1.480 1.761
91603 ZNF830 zinc finger protein 830 17q12 down −0.358 2.075E-02 1.040 1.085
5094 PCBP2 poly(rC) binding protein 2 12q13.13 down −0.286 4.734E-02 1.454 1.158

To further investigate the UHRF1 downstream genes, we performed the classification of these candidate genes to known molecular pathways by using DAVID program (https://david.ncifcrf.gov/). Classification strategy of downstream genes by si-UHRF1 transfectants is shown in Figure 10A and 10B. Significantly upregulated and downregulated pathways and their involved genes are indicated in Tables 4 and 5. Several genes were classified into biological process categories and a variety of biological pathways, “M phase”, “cell cycle”, and “cell cycle phase” were significantly downregulated by si- UHRF1 transfectants (Table 4).

Figure 10. Flow chart demonstrating the strategy for analysis of genes regulated by UHRF1.

Figure 10

(A) A total of 2,222 and 1,512 downregulated genes in expression analyses of si-UHRF1 transfectants of BC cell lines (T24 and BOY, respectively) were selected. We then analyzed 533 common downregulated genes by using available GEO data sets (GSE11783 + GSE31684). The analyses showed that 104 genes were significantly upregulated in BC specimens compared with NBE. (B) A total of 2,665 and 2,434 upregulated genes in expression analysis of si-UHRF1 transfectants of BC cell lines (T24 and BOY, respectively) were selected. We then analyzed 704 common upregulated genes by using GEO data sets. The analyses showed that 62 genes were significantly downregulated in BC specimens compared with NBE.

Table 4. Downregulated genes by si-UHRF1 were classified by DAVID program.

Biological process Number of genes P-Value Genes
M phase 15 8.10E-09 ASPM, BIRC5, BRCA2, CENPE, CENPF, FBXO43, KIF11, KIF18A, KIF20B, MPHOSPH9, NBN, SGOL2, SMC2, SMC4, VCPIP1
cell cycle 20 1.10E-07 ASPM, BIRC5, BRCA2, CALR, CENPE, CENPF, ESCO1, FBXO43, KIF11, KIF18A, KIF20B, MPHOSPH9, NBN, PSMC1, RIF1, SGOL2, SMC2, SMC4, UHRF1, VCPIP1
cell cycle phase 15 1.40E-07 ASPM, BIRC5, BRCA2, CENPE, CENPF, FBXO43, KIF11, KIF18A, KIF20B, MPHOSPH9, NBN, SGOL2, SMC2, SMC4, VCPIP1
cell cycle process 17 1.90E-07 ASPM, BIRC5, BRCA2, CALR, CENPE, CENPF, FBXO43, KIF11, KIF18A, KIF20B, MPHOSPH9, NBN, PSMC1, SGOL2, SMC2, SMC4, VCPIP1
chromosome segregation 8 5.20E-07 BIRC5, CENPE, CENPF, KIF18A, SGOL2, SMC2, SMC4, TOP2A
M phase of mitotic cell cycle 11 8.50E-07 ASPM, BIRC5, CENPE, CENPF, KIF11, KIF18A, KIF20B, MPHOSPH9, SMC2, SMC4, VCPIP1
organelle fission 11 1.00E-06 ASPM, BIRC5, CENPE, CENPF, KIF11, KIF18A, KIF20B, OPA1, SMC2, SMC4, VCPIP1
mitosis 10 6.40E-06 ASPM, BIRC5, CENPE, CENPF, KIF11, KIF18A, KIF20B, SMC2, SMC4, VCPIP1
nuclear division 10 6.40E-06 ASPM, BIRC5, CENPE, CENPF, KIF11, KIF18A, KIF20B, SMC2, SMC4, VCPIP1
mitotic cell cycle 12 1.20E-05 ASPM, BIRC5, CENPE, CENPF, KIF11, KIF18A, KIF20B, MPHOSPH9, PSMC1, SMC2, SMC4, VCPIP1
DNA repair 10 4.90E-05 BRCA2, ESCO1, FANCM, NBN, NEIL3, PMS1, RECQL4, SMC6, TOP2A, UHRF1
cell division 10 6.50E-05 ASPM, BIRC5, BRCA2, CENPE, CENPF, KIF11, KIF20B, SGOL2, SMC2, SMC4
response to DNA damage stimulus 11 7.40E-05 BRCA2, ESCO1, FANCM, NBN, NEIL3, PMS1, RECQL4, RIF1, SMC6, TOP2A, UHRF1
establishment of chromosome localization 4 8.90E-05 BIRC5, CENPE, CENPF, KIF18A
chromosome localization 4 8.90E-05 BIRC5, CENPE, CENPF, KIF18A
chromosome organization 12 1.40E-04 BRCA2, BRPF3, CENPE, CENPF, FBXO4, KIF18A, NBN, PCGF2, SGOL2, SMC2, SMC4, TOP2A
DNA metabolic process 12 2.00E-04 BRCA2, CENPF, ESCO1, FANCM, FBXO4, NBN, NEIL3, PMS1, RECQL4, SMC6, TOP2A, UHRF1
microtubule-based movement 6 5.80E-04 CENPE, KIF11, KIF14, KIF18A, KIF20B, OPA1
regulation of cell cycle process 6 6.00E-04 BIRC5, BRCA2, CALR, CENPE, CENPF, KIF20B
microtubule-based process 8 7.90E-04 BRCA2, CENPE, HOOK1, KIF11, KIF14, KIF18A, KIF20B, OPA1
mitotic sister chromatid segregation 4 1.30E-03 CENPE, KIF18A, SMC2, SMC4
sister chromatid segregation 4 1.40E-03 CENPE, KIF18A, SMC2, SMC4
metaphase plate congression 3 1.90E-03 CENPE, CENPF, KIF18A
cellular response to stress 11 2.00E-03 BRCA2, ESCO1, FANCM, NBN, NEIL3, PMS1, RECQL4, RIF1, SMC6, TOP2A, UHRF1
regulation of mitotic cell cycle 6 2.20E-03 BIRC5, BRCA2, CENPE, CENPF, KIF20B, NBN
organelle localization 5 2.20E-03 ASPM, BIRC5, CENPE, CENPF, KIF18A
spindle checkpoint 3 2.20E-03 BIRC5, CENPE, CENPF
positive regulation of cell cycle 4 4.80E-03 BIRC5, BRCA2, CALR, CENPE
establishment of organelle localization 4 8.20E-03 BIRC5, CENPE, CENPF, KIF18A
chromosome condensation 3 9.70E-03 SMC2, SMC4, TOP2A
glucose transport 3 1.30E-02 SLC5A2, STXBP4, YES1
hexose transport 3 1.40E-02 SLC5A2, STXBP4, YES1
regulation of cell cycle 7 1.40E-02 BIRC5, BRCA2, CALR, CENPE, CENPF, KIF20B, NBN
monosaccharide transport 3 1.50E-02 SLC5A2, STXBP4, YES1
negative regulation of neuron differentiation 3 1.70E-02 ASPM, CALR, NBN
cell cycle checkpoint 4 1.70E-02 BIRC5, CENPE, CENPF, NBN
kinetochore assembly 2 1.80E-02 CENPE, CENPF
meiosis 4 2.10E-02 BRCA2, FBXO43, NBN, SGOL2
M phase of meiotic cell cycle 4 2.10E-02 BRCA2, FBXO43, NBN, SGOL2
meiotic cell cycle 4 2.20E-02 BRCA2, FBXO43, NBN, SGOL2
germ cell development 4 2.30E-02 BRCA2, CASC5, HOOK1, PVRL2
kinetochore organization 2 2.40E-02 CENPE, CENPF
DNA recombination 4 2.50E-02 BRCA2, NBN, RECQL4, SMC6
mitotic cell cycle checkpoint 3 2.70E-02 CENPE, CENPF, NBN
centromere complex assembly 2 3.50E-02 CENPE, CENPF
spermatid development 3 4.10E-02 CASC5, HOOK1, PVRL2
regulation of nuclear division 3 4.40E-02 CENPE, CENPF, KIF20B
regulation of mitosis 3 4.40E-02 CENPE, CENPF, KIF20B
negative regulation of macromolecule biosynthetic process 8 4.50E-02 BRCA2, CALR, CD276, CENPF, KCNIP3, PCGF2, SKIL, ZNF254
spermatid differentiation 3 4.60E-02 CASC5, HOOK1, PVRL2
cytoskeleton organization 7 4.60E-02 BRCA2, CALR, HOOK1, KIF11, KIF18A, OPHN1, RICTOR
negative regulation of cellular biosynthetic process 8 5.10E-02 BRCA2, CALR, CD276, CENPF, KCNIP3, PCGF2, SKIL, ZNF254
positive regulation of cellular protein metabolic process 5 5.10E-02 CLCF1, EIF5A, FBXO4, PSMC1, RICTOR
carbohydrate transport 3 5.20E-02 SLC5A2, STXBP4, YES1
mitotic metaphase plate congression 2 5.30E-02 CENPE, KIF18A
regulation of DNA replication 3 5.30E-02 BRCA2, CALR, NBN
double-strand break repair 3 5.30E-02 BRCA2, NBN, RECQL4
negative regulation of biosynthetic process 8 5.50E-02 BRCA2, CALR, CD276, CENPF, KCNIP3, PCGF2, SKIL, ZNF254
positive regulation of protein metabolic process 5 5.80E-02 CLCF1, EIF5A, FBXO4, PSMC1, RICTOR
microtubule cytoskeleton organization 4 5.80E-02 BRCA2, HOOK1, KIF11, KIF18A
negative regulation of mitotic metaphase/anaphase transition 2 6.40E-02 CENPE, CENPF
blastocyst growth 2 6.40E-02 BRCA2, NBN
mitotic cell cycle spindle assembly checkpoint 2 6.40E-02 CENPE, CENPF
positive regulation of mitotic cell cycle 2 7.00E-02 BIRC5, BRCA2
negative regulation of mitosis 2 7.00E-02 CENPE, CENPF
negative regulation of nuclear division 2 7.00E-02 CENPE, CENPF
negative regulation of macromolecule metabolic process 9 7.20E-02 BRCA2, CALR, CD276, CENPF, KCNIP3, PCGF2, PSMC1, SKIL, ZNF254
reproductive cellular process 4 7.30E-02 BRCA2, CASC5, HOOK1, PVRL2
mitotic chromosome condensation 2 7.50E-02 SMC2, SMC4
negative regulation of transcription from RNA polymerase II promoter 5 7.50E-02 CALR, KCNIP3, PCGF2, SKIL, ZNF254
protein localization 10 8.00E-02 CALR, CENPE, CENPF, EIF5A, HOOK1, KIF18A, RAB12, RPGR, SGOL2, STXBP4
negative regulation of nucleobase, nucleoside, nucleotide and nucleic acid metabolic process 7 8.60E-02 BRCA2, CALR, CENPF, KCNIP3, PCGF2, SKIL, ZNF254
establishment of protein localization 9 8.90E-02 CALR, CENPE, CENPF, EIF5A, HOOK1, KIF18A, RAB12, RPGR, STXBP4
in utero embryonic development 4 8.90E-02 BRCA2, NBN, PCGF2, RPGRIP1L
negative regulation of nitrogen compound metabolic process 7 9.10E-02 BRCA2, CALR, CENPF, KCNIP3, PCGF2, SKIL, ZNF254
positive regulation of cellular component organization 4 9.50E-02 CALR, CENPE, EIF5A, RICTOR
developmental growth 3 9.60E-02 BRCA2, NBN, PLAU

Table 5. Upregulated genes by si-UHRF1 were classified by DAVID program.

Biological process Number of genes P-Value Genes
regulation of transcription 15 1.40E-02 CRY2, ADRB2, EZH1, HCFC2, HIF3A, JRK, POFUT1, PRDM16, PRDM2, TLR4, ZNF10, ZNF331, ZNF44, ZNF655, ZNF91
regulation of transcription, DNA-dependent 10 7.00E-02 ADRB2, HCFC2, HIF3A, PRDM16, PRDM2, ZNF10, ZNF331, ZNF44, ZNF655, ZNF91
regulation of RNA metabolic process 10 7.90E-02 ADRB2, HCFC2, HIF3A, PRDM16, PRDM2, ZNF10, ZNF331, ZNF44, ZNF655, ZNF91
negative regulation of myeloid leukocyte differentiation 2 4.90E-02 PRDM16, TLR4
fucose metabolic process 2 5.20E-02 POFUT1, FPGT
brown fat cell differentiation 2 6.90E-02 ADRB2, PRDM16
negative regulation of myeloid cell differentiation 2 8.50E-02 PRDM16, TLR4

DISCUSSION

miRNAs are critical regulators of gene expression and they control many physiologic processes in mammalian cells [57]. There are abundant evidences that aberrantly expressed miRNAs can dysregulate otherwise well-controlled cellular RNA networks, thereby enhancing cancer cell development, progression, and metastasis [69]. The discovery of aberrantly expressed miRNAs and the resultant changes in RNA networks in cancer cells provide novel molecular explanations for cancer cell progression and metastasis. It is now apparent that dysregulated miRNAs play important roles in BC cell development [16]. Our past miRNA studies of BC cells showed that clustered miRNAs (including miR-1/133a (targeting TAGLN2), miR-23b/27b/24-1 (targeting EGFR, MET, and FOXM1), and miR-195/497 (targeting BIRC5 and WNT7A)) act as tumor-suppressive miRNAs through their regulation of several oncogenic genes and pathways [10, 1719].

Improved technological developments (next generation sequencing) have illuminated the role of miRNA networks in cancer cells. In this study, we examined the expression of miR-145-5p and miR-145 3p in BC cells because these miRNAs were significantly reduced in cancer cells as determined by deep sequencing. Our data demonstrated that miR-145-3p (the passenger-strand from pre-miR-145) had anti-tumor effects through targeting of UHRF1 in BC cells.

Downregulation of miR-145-5p (the guide-strand) is frequently observed in many types of cancer, and past studies have established the anti-tumor function of miR-145-5p through its regulation of several types of oncogenes in cancer cells [15]. Our group also identified the anti-tumor function of miR-145-5p in prostate cancer, renal cell carcinoma, bladder cancer, and esophageal squamous cell carcinoma [2023]. Importantly, p53 appears to transcriptionally regulate miR-145-5p by interaction with a potential p53 response element at the pre-miR-145 promoter region [24]. Moreover, c-MYC is directly repressed by miR-145-5p, indicating that it acts as a new member of the p53 regulatory network and contributes to the direct linkage between p53 and c-MYC in human cancer pathways [24]. In contrast to miR-145-5p, the functional significance of miR-145-3p in cancer cells has been obscure. This is the first report to evaluate the anti-tumor function of miR-145-3p in BC cells by gain-of-function assays.

miRNAs are often associated in clusters in the genome, and several studies have focused on the functional role of clustered miRNAs in human cancers [17, 18, 2023, 25]. In the human genome, 429 human miRNAs have been found to be clustered at 144 sites, with inter-miRNA distances of less than 5,000 base pair (miRBase, release 21). Both miR-143 and miR-145-5p are known to be located close together on human chromosome 5q32, where they form a cluster [26]. Based on our miRNA signatures, miR-143 and miR-145-5p are the most frequently downregulated miRNAs in various types of human cancers [26]. These two miRNAs have been reported as tumor suppressors and studied extensively for their role in oncogenic pathways in several cancers [15]. Our past studies demonstrated that hexokinase-2 (HK2) and Golgi membrane protein 1 (GOLM1) were directly regulated by miR-143 and miR-145-5p in renal carcinoma and prostate cancer, respectively [22, 23].

In this study, we speculated that miR-145-5p and miR-145-3p worked together to regulate pathways in BC cell progression and metastasis. Our present data showed that UHRF1 was directly regulated by both miR-145-5p and miR-145-3p in BC cells. In previous studies of miRNA regulation of UHRF1 in cancers, UHRF1 was regulated by miR-146a/146b in gastric cancer [27], miR-9 in colorectal cancer [28], and miR-124 in BC [29]. However, there have been no previous reports about the effects of miR-145-5p and miR-145-3p on UHRF1.

The UHRF1 gene was first cloned as a transcription factor that binds to the promoter region of the topoisomerase IIα (TOP2A) gene and controls its expression levels [30]. UHRF1 is involved in a wide range of physiological and pathological phenomena, including cancer development and metastasis [31]. UHRF1 plays a pivotal role in controlling gene expression through regulating epigenetic mechanisms, including DNA methylation, histone deacetylation, histone methylation, and histone ubiquitination [31]. Overexpression of UHRF1 occurs in many types of cancer, and aberrantly expressed UHRF1 causes cancer cell activation through hyper-methylation of tumor-suppressor genes such as BRCA1, CDKN2A, p73, and RASSF1 [32]. Expression of UHRF1 might be used as a progression marker in cancer [32]. For example, the expression of UHRF1 in MIBC was greater than in NMIBC, and upregulation was associated with an increased risk of progression after transurethral resection [33]. Our present data showed that knockdown of UHRF1 significantly induced apoptosis in BC cells and expression levels of the gene correlated with cause specific survival. Our data support the past studies of UHRF1 in cancer research, suggesting UHRF1 plays essential roles in BC cell progression and might be a molecular target for BC treatment.

In this study, we identified UHRF1-regulated BC pathways by using genome-wide gene expression analysis of si-UHRF1-transfected cells. Our expression data showed that UHRF1 and TOP2A were significantly reduced by si-UHRF1 transfection, indicating the usefulness of the present analytic approach. Our data showed that several anti-apoptosis genes and pro-proliferation genes were involved in pathways downstream of UHRF1, such as BIRC5 and CENPF. BIRC5 is a member of the inhibitor of apoptosis (IAP) family preferentially expressed by many cancers, including BC [10], and its mediated cellular networks are essential for cancer cell proliferation and viability [34]. CENPF is a master regulator of prostate cancer malignancy. Together, FOXM1 and CENPF regulate target gene expression and activation in cancer cells [35, 36]. The identification of these novel molecular pathways and targets mediated by the miR-145-5p/145-3p/UHRF1 axis may lead to a better understanding of BC cell progression and metastasis.

In conclusion, downregulation of dual-strand miR- 145-5p and miR-145-3p was validated in BC clinical specimens, and these miRNAs were shown to function as tumor suppressors in BC cells. To the best of our knowledge, this is the first report demonstrating that tumor suppressive miR-145-5p and miR-145-3p directly targeted UHRF1. Moreover, UHRF1 was upregulated in BC clinical specimens and contributed to anti-apoptotic effects through its regulation of several oncogenic genes. Expression of UHRF1 might be a useful prognostic marker for survival of BC patients. The identification of novel molecular pathways and targets regulated by the miR-145-5p/miR-145-3p/UHRF1 axis may lead to a better understanding of BC progression and aggressiveness.

MATERIALS AND METHODS

Clinical specimens and cell lines

Clinical tissue specimens were collected from BC patients (n = 69) who had undergone transurethral resection of their bladder tumors (TURBT, n = 59) or cystectomy (n = 10) at Kagoshima University Hospital between 2003 and 2013. NBE (n = 12) were derived from patients with noncancerous disease. The specimens were staged according to the American Joint Committee on Cancer-Union Internationale Contre le Cancer tumor-node-metastasis (TNM) classification and histologically graded [37]. Our study was approved by the Bioethics Committee of Kagoshima University; written prior informed consent and approval were obtained from all patients. Patient details and clinicopathological characteristics are listed in Table 6.

Table 6. Characteristic of patients.

Bladder cancer (BC)
Total number 69
Median age (range) 73 (40–94) years
Gender
 Male 53 76.8%
 Female 16 23.2%
Tumor grade
 Low grade 45 65.2%
 High grade 22 31.9%
 Unknown 2 2.9%
T stage
 Tis 2 2.9%
 Ta 7 10.1%
 T1 25 36.2%
 T2 27 39.1%
 T3 4 5.8%
 T4 4 5.8%
N stage
 N0 40 58.0%
 N1 8 11.6%
 Unknown 21 30.4%
M stage
 M0 58 84.1%
 M1 5 7.2%
 Unknown 6 8.7%
Operation method
 TURBT 59 85.5%
 Cystectomy 10 14.5%
Normal bladder epithelium
 Total number 12
 Median age (range) 61 (47–72)  years

Abbreviation: TURBT = transurethral resection of bladder tumor

We used two human BC cell lines: T24, which was invasive and obtained from the American Type Culture Collection; and BOY, which was established in our laboratory from an Asian male patient, 66 years old, who was diagnosed with stage III BC and lung metastasis [38, 39]. These cell lines were maintained in minimum essential medium supplemented with 10% fetal bovine serum in a humidified atmosphere of 5% CO2 and 95% air at 37°C.

Tissue collection and RNA extraction

Tissues were immersed in RNAlater (Thermo Fisher Scientific; Waltham, MA, USA) and stored at −20°C until RNA extraction was conducted. Total RNA, including miRNA, was extracted using the mirVana miRNA isolation kit (Thermo Fisher Scientific) following the manufacturer's protocol. The integrity of the RNA was checked with an RNA 6000 Nano Assay kit and a 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA) following the manufacturer's protocol.

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

The procedure for qRT-PCR quantification was described previously [40, 41]. Stem-loop RT-PCR (TaqMan MicroRNA Assays; product ID: 002278 for miR-145-5p and product ID: 002149 for miR-145-3p; Thermo Fisher Scientific) was used to quantify miRNAs according to previously published conditions [4042]. TaqMan probes and primers for UHRF1 (product ID: Hs 01086727_m1; Thermo Fisher Scientific) were assay-on-demand gene expression products. We used human GUSB (product ID: Hs99999908_m1; Thermo Fisher Scientific) and RNU48 (product ID: 001006; Thermo Fisher Scientific), respectively, as internal controls.

Transfections with miRNA mimic and small interfering RNA (siRNA) into BC cell lines

Mature miRNA molecules, Pre-miR miRNA precursors (hsa-miR-145-5p; product ID: PM11480, hsa-miR-145-3p; product ID: PM13036, and negative control miRNA; product ID: AM 17111; Thermo Fisher Scientific) were used in the gain-of-function experiments, whereas UHRF1 siRNA (product ID: HSS120939 and HSS179006; Thermo Fisher Scientific) and negative control siRNA (product ID: D-001810-10; Thermo Fisher Scientific) were used in the loss-of-function experiments. The transfection procedures and transfection efficiencies of miRNA in T24 and BOY cells were reported previously [4042].

Cell proliferation, migration, and invasion assays

To investigate the functional significance of the miR-145-5p, miR-145-3p, and UHRF1, we performed cell proliferation, migration, and invasion assays using T24 and BOY cells. The experimental procedures were performed as described in our previous studies [4042].

Apoptosis assays

BC cell lines were transiently transfected with reagent only (mock), miR-control, miR-145-5p, miR- 145- 3p, siRNA-control, or si-UHRF1 at 10 nM in 6 well tissue culture plates, as described previously [14, 1719]. Cells were harvested by trypsinization 72 hours after transfection and washed in cold phosphate-buffered saline. For apoptosis assays, double staining with FITC-Annexin V and propidium iodide was carried out using a FITC Annexin V Apoptosis Detection Kit (BD Biosciences, Bedford, MA, USA) according to the manufacturer's recommendations and analysed within 1 hour by flow cytometry (CyAn ADP analyzer; Beckman Coulter, Brea, CA, USA). Cells were identified as viable cells, dead cells, early apoptotic cells, and apoptotic cells using Summit 4.3 software (Beckman Coulter), and the percentages of early apoptotic and apoptotic cells from each experiment were then compared. As a positive control, we used 2 μg/mL cycloheximide.

Cell cycle assays

For the cell cycle analyses, cells were stained with PI using the Cycletest PLUS DNA Reagent Kit (BD Biosciences) following the protocol and analyzed by CyAn ADP analyzer (Beckman Coulter). The percentages of the cells in the G0/G1, S, and G2/M phases were determined and compared. Experiments were performed in triplicate.

Western blot analyses

Immunoblotting was performed with rabbit anti-UHRF1 antibodies (1:500, PA5-29884; Thermo Fisher Scientific), anti-PARP antibodies (1:500 #9542; Cell Signaling Technology; Danvers, MA, USA), anti-cleaved PARP antibodies (1:500 #5625; Cell Signaling Technology), and anti-GAPDH antibodies (1:10000 MAB374; Chemicon, Temecula, CA, USA). Specific complexes were visualized with an echochemiluminescence detection system (GE Healthcare, Little Chalfont, UK).

Immunohistochemistry

A tissue microarray of 68 urothelial cancers and 20 normal bladder tissues was obtained from US Biomax, Inc. (Rockville, MD, USA) (product ID: BL1002). Detailed information on all tumor specimens can be found at http://www.biomax.us/index.php. The tissue microarray was immunostained following the manufacturer's protocol with an Ultra Vision Detection System (Thermo Scientific). The primary rabbit polyclonal antibodies against UHRF1 (PA5-29884; Thermo Fisher Scientific) were diluted 1:300. Immunostaining was evaluated according to a scoring method as described previously [17].

Genome-wide gene expression and in silico analyses for the identification of genes regulated by miR-145-5p and miR-145-3p

To further investigate the specific genes affected by miR-145-5p and miR-145-3p, we performed a combination of in silico and genome-wide gene expression analyses. We attempted to identify target genes using a BC cell line transfected with these miRNAs. A Sure Print G3 Human GE 8 × 60K Microarray (Agilent Technologies) was used for expression profiling of miR-145-5p and miR-145-3p transfectants. The microarray data were deposited into GEO (http://www.ncbi.nlm.nih.gov/geo/) and were assigned GEO accession number GSE66498. Next, we selected putative miRNA target genes using the microRNA.org database (August, 2010 release, http://www.microrna.org). Finally, to identify upregulated genes in BC, we analyzed publicly available gene expression data sets in GEO (accession numbers: GSE11783, GSE31684). The data were normalized and analyzed with Gene Spring software (Agilent Technologies) as described previously [22, 23, 4042]. The strategy for investigation of the target genes is shown in Figure 3.

Plasmid construction and dual luciferase reporter assays

Partial wild-type sequences of the 3′ UTR of UHRF1 or those with a deleted miR-145-5p and miR- 145- 3p target site (positions 1,179–1,198 of UHRF1 3′ UTR for miR- 145-5p, and positions 287–292 of UHRF1 3′ UTR for miR-145-3p) were inserted between the XhoI and PmeI restriction sites in the 3′ UTR of the hRluc gene in the psiCHECK-2 vector (C8021; Promega, Madison, WI, USA). T24 and BOY cell lines were transfected with 50 ng of the vector and 10 nM miR-145-5p or miR-145-3p using Lipofectamine 2000 (Thermo Fisher Scientific) and Opti-MEM (Thermo Fisher Scientific). The activities of firefly and Renilla luciferases in cell lysates were determined with a dual luciferase reporter assay system according to the manufacturer's protocol (E1960; Promega). Normalized data were calculated as the ratio of Renilla/firefly luciferase activities.

Identification of downstream targets regulated by UHRF1 in BC

To investigate molecular targets regulated by UHRF1 in BC cells, we carried out gene expression analyses using si-UHRF1-transfected BC cell lines. Microarray data were used for expression profiling of si-UHRF1 transfectants. The microarray data were deposited into GEO (accession number: GSE77790). We analyzed common down or upregulated genes using the GEO dataset. The flow chart outlining the investigation of UHRF1 downstream genes is shown in Figure 10A and 10B.

Statistical analysis

Relationships among two or three variables and numerical values were analysed using the Mann-Whitney U test or Bonferroni-adjusted Mann-Whitney U test. Spearman's rank test was used to evaluate the correlation among the expressions of miR-145-5p, miR-145-3p, and UHRF1. We estimated cause specific survival of 57 BC patients by using the Kaplan-Meier method. Among the 69 BC patients, 12 died of other causes. Therefore, we analyzed cause specific survival of 57 BC patients. Patients were divided into two groups according to the median value of UHRF1 expression, and the differences between the two groups were evaluated by the log-rank tests. We used Expert Stat View software, version 5.0 (SAS Institute Inc., Cary, NC, USA), for these analyses.

SUPPLEMENTARY MATERIALS FIGURES

ACKNOWLEDGMENTS AND FUNDING

This study was supported by JSPS KAKENHI Grant Numbers 26293354, 25462490, and 26462416.

Footnotes

CONFLICTS OF INTEREST

The authors indicated no potential conflicts of interest.

REFERENCES

  • 1.Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108. doi: 10.3322/caac.21262. [DOI] [PubMed] [Google Scholar]
  • 2.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65:5–29. doi: 10.3322/caac.21254. [DOI] [PubMed] [Google Scholar]
  • 3.Schenk-Braat EA, Bangma CH. Immunotherapy for superficial bladder cancer. Cancer Immunol Immunother. 2005;54:414–423. doi: 10.1007/s00262-004-0621-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Collaboration ABCAM-a. Neoadjuvant chemotherapy in invasive bladder cancer: a systematic review and meta-analysis. Lancet. 2003;361:1927–1934. doi: 10.1016/s0140-6736(03)13580-5. [DOI] [PubMed] [Google Scholar]
  • 5.Di Leva G, Croce CM. Roles of small RNAs in tumor formation. Trends in molecular medicine. 2010;16:257–267. doi: 10.1016/j.molmed.2010.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Garzon R, Marcucci G, Croce CM. Targeting microRNAs in cancer: rationale, strategies and challenges. Nat Rev Drug Discov. 2010;9:775–789. doi: 10.1038/nrd3179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Carthew RW, Sontheimer EJ. Origins and Mechanisms of miRNAs and siRNAs. Cell. 2009;136:642–655. doi: 10.1016/j.cell.2009.01.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Friedman RC, Farh KK, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome research. 2009;19:92–105. doi: 10.1101/gr.082701.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB. Prediction of mammalian microRNA targets. Cell. 2003;115:787–798. doi: 10.1016/s0092-8674(03)01018-3. [DOI] [PubMed] [Google Scholar]
  • 10.Itesako T, Seki N, Yoshino H, Chiyomaru T, Yamasaki T, Hidaka H, Yonezawa T, Nohata N, Kinoshita T, Nakagawa M, Enokida H. The microRNA expression signature of bladder cancer by deep sequencing: the functional significance of the miR-195/497 cluster. PloS one. 2014;9:e84311. doi: 10.1371/journal.pone.0084311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Chendrimada TP, Gregory RI, Kumaraswamy E, Norman J, Cooch N, Nishikura K, Shiekhattar R. TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing. Nature. 2005;436:740–744. doi: 10.1038/nature03868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136:215–233. doi: 10.1016/j.cell.2009.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hutvagner G, Zamore PD. A microRNA in a multiple-turnover RNAi enzyme complex. Science. 2002;297:2056–2060. doi: 10.1126/science.1073827. [DOI] [PubMed] [Google Scholar]
  • 14.Matsushita R, Seki N, Chiyomaru T, Inoguchi S, Ishihara T, Goto Y, Nishikawa R, Mataki H, Tatarano S, Itesako T, Nakagawa M, Enokida H. Tumour-suppressive microRNA-144-5p directly targets CCNE1/2 as potential prognostic markers in bladder cancer. Br J Cancer. 2015;113:282–289. doi: 10.1038/bjc.2015.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Cui SY, Wang R, Chen LB. MicroRNA-145: a potent tumour suppressor that regulates multiple cellular pathways. JJ Cell Mol Med. 2014;18:1913–1926. doi: 10.1111/jcmm.12358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Yoshino H, Seki N, Itesako T, Chiyomaru T, Nakagawa M, Enokida H. Aberrant expression of microRNAs in bladder cancer. Nature reviews Urology. 2013;10:396–404. doi: 10.1038/nrurol.2013.113. [DOI] [PubMed] [Google Scholar]
  • 17.Yoshino H, Chiyomaru T, Enokida H, Kawakami K, Tatarano S, Nishiyama K, Nohata N, Seki N, Nakagawa M. The tumour-suppressive function of miR-1 and miR-133a targeting TAGLN2 in bladder cancer. Br J Cancer. 2011;104:808–818. doi: 10.1038/bjc.2011.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Chiyomaru T, Seki N, Inoguchi S, Ishihara T, Mataki H, Matsushita R, Goto Y, Nishikawa R, Tatarano S, Itesako T, Nakagawa M, Enokida H. Dual regulation of receptor tyrosine kinase genes EGFR and c-Met by the tumor-suppressive microRNA-23b/27b cluster in bladder cancer. Int J Oncol. 2015;46:487–496. doi: 10.3892/ijo.2014.2752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Inoguchi S, Seki N, Chiyomaru T, Ishihara T, Matsushita R, Mataki H, Itesako T, Tatarano S, Yoshino H, Goto Y, Nishikawa R, Nakagawa M, Enokida H. Tumour-suppressive microRNA-24-1 inhibits cancer cell proliferation through targeting FOXM1 in bladder cancer. FEBS Lett. 2014;588:3170–3179. doi: 10.1016/j.febslet.2014.06.058. [DOI] [PubMed] [Google Scholar]
  • 20.Chiyomaru T, Enokida H, Tatarano S, Kawahara K, Uchida Y, Nishiyama K, Fujimura L, Kikkawa N, Seki N, Nakagawa M. miR-145 and miR-133a function as tumour suppressors and directly regulate FSCN1 expression in bladder cancer. Br J Cancer. 2010;102:883–891. doi: 10.1038/sj.bjc.6605570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kano M, Seki N, Kikkawa N, Fujimura L, Hoshino I, Akutsu Y, Chiyomaru T, Enokida H, Nakagawa M, Matsubara H. miR-145, miR-133a and miR-133b: Tumor-suppressive miRNAs target FSCN1 in esophageal squamous cell carcinoma. Int J Cancer. 2010;127:2804–2814. doi: 10.1002/ijc.25284. [DOI] [PubMed] [Google Scholar]
  • 22.Yoshino H, Enokida H, Itesako T, Kojima S, Kinoshita T, Tatarano S, Chiyomaru T, Nakagawa M, Seki N. Tumor-suppressive microRNA-143/145 cluster targets hexokinase-2 in renal cell carcinoma. Cancer Sci. 2013;104:1567–1574. doi: 10.1111/cas.12280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kojima S, Enokida H, Yoshino H, Itesako T, Chiyomaru T, Kinoshita T, Fuse M, Nishikawa R, Goto Y, Naya Y, Nakagawa M, Seki N. The tumor-suppressive microRNA-143/145 cluster inhibits cell migration and invasion by targeting GOLM1 in prostate cancer. J Hum Genet. 2014;59:78–87. doi: 10.1038/jhg.2013.121. [DOI] [PubMed] [Google Scholar]
  • 24.Sachdeva M, Zhu S, Wu F, Wu H, Walia V, Kumar S, Elble R, Watabe K, Mo YY. p53 represses c-Myc through induction of the tumor suppressor miR-145. Proc Natl Acad Sci U S A. 2009;106:3207–3212. doi: 10.1073/pnas.0808042106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kim YK, Yu J, Han TS, Park SY, Namkoong B, Kim DH, Hur K, Yoo MW, Lee HJ, Yang HK, Kim VN. Functional links between clustered microRNAs: suppression of cell-cycle inhibitors by microRNA clusters in gastric cancer. Nucleic Acids Res. 2009;37:1672–1681. doi: 10.1093/nar/gkp002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Das AV, Pillai RM. Implications of miR cluster 143/145 as universal anti-oncomiRs and their dysregulation during tumorigenesis. Cancer Cell Int. 2015;15:92. doi: 10.1186/s12935-015-0247-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Zhou L, Zhao X, Han Y, Lu Y, Shang Y, Liu C, Li T, Jin Z, Fan D, Wu K. Regulation of UHRF1 by miR-146a/b modulates gastric cancer invasion and metastasis. FASEB J. 2013;27:4929–4939. doi: 10.1096/fj.13-233387. [DOI] [PubMed] [Google Scholar]
  • 28.Zhu M, Xu Y, Ge M, Gui Z, Yan F. Regulation of UHRF1 by miR-9 modulates colorectal cancer cell proliferation and apoptosis. Cancer Sci. 2015;106:883–839. doi: 10.1111/cas.12689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Wang X, Wu Q, Xu B, Wang P, Fan W, Cai Y, Gu X, Meng F. miR-124 exerts tumor suppressive functions on the cell proliferation, motility and angiogenesis of bladder cancer by fine-tuning UHRF1. FEBS J. 2015;282:4376–4388. doi: 10.1111/febs.13502. [DOI] [PubMed] [Google Scholar]
  • 30.Hopfner R, Mousli M, Jeltsch JM, Voulgaris A, Lutz Y, Marin C, Bellocq JP, Oudet P, Bronner C. ICBP90, a novel human CCAAT binding protein, involved in the regulation of topoisomerase IIalpha expression. Can Res. 2000;60:121–128. [PubMed] [Google Scholar]
  • 31.Unoki M, Brunet J, Mousli M. Drug discovery targeting epigenetic codes: the great potential of UHRF1, which links DNA methylation and histone modifications, as a drug target in cancers and toxoplasmosis. Biochem Pharmacol. 2009;78:1279–1288. doi: 10.1016/j.bcp.2009.05.035. [DOI] [PubMed] [Google Scholar]
  • 32.Bronner C, Krifa M, Mousli M. Increasing role of UHRF1 in the reading and inheritance of the epigenetic code as well as in tumorogenesis. Biochem Pharmacol. 2013;86:1643–1649. doi: 10.1016/j.bcp.2013.10.002. [DOI] [PubMed] [Google Scholar]
  • 33.Unoki M, Kelly JD, Neal DE, Ponder BA, Nakamura Y, Hamamoto R. UHRF1 is a novel molecular marker for diagnosis and the prognosis of bladder cancer. Br J Cancer. 2009;101:98–105. doi: 10.1038/sj.bjc.6605123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Athanasoula K, Gogas H, Polonifi K, Vaiopoulos AG, Polyzos A, Mantzourani M. Survivin beyond physiology: orchestration of multistep carcinogenesis and therapeutic potentials. Cancer Lett. 2014;347:175–182. doi: 10.1016/j.canlet.2014.02.014. [DOI] [PubMed] [Google Scholar]
  • 35.Aytes A, Mitrofanova A, Lefebvre C, Alvarez MJ, Castillo-Martin M, Zheng T, Eastham JA, Gopalan A, Pienta KJ, Shen MM, Califano A, Abate-Shen C. Cross-species regulatory network analysis identifies a synergistic interaction between FOXM1 and CENPF that drives prostate cancer malignancy. Cancer Cell. 2014;25:638–651. doi: 10.1016/j.ccr.2014.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lokody I. Signalling: FOXM1 and CENPF: co-pilots driving prostate cancer. Nat Rev Cancer. 2014;14:450–451. doi: 10.1038/nrc3772. [DOI] [PubMed] [Google Scholar]
  • 37.Sobin LH, Compton CC. TNM seventh edition: what's new, what's changed: communication from the International Union Against Cancer and the American Joint Committee on Cancer. Cancer. 2010;116:5336–5339. doi: 10.1002/cncr.25537. [DOI] [PubMed] [Google Scholar]
  • 38.Tatarano S, Chiyomaru T, Kawakami K, Enokida H, Yoshino H, Hidaka H, Nohata N, Yamasaki T, Gotanda T, Tachiwada T, Seki N, Nakagawa M. Novel oncogenic function of mesoderm development candidate 1 and its regulation by MiR-574-3p in bladder cancer cell lines. Int J Oncol. 2012;40:951–959. doi: 10.3892/ijo.2011.1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Tatarano S, Chiyomaru T, Kawakami K, Enokida H, Yoshino H, Hidaka H, Yamasaki T, Kawahara K, Nishiyama K, Seki N, Nakagawa M. miR-218 on the genomic loss region of chromosome 4p15. 31 functions as a tumor suppressor in bladder cancer. Int J Oncol. 2011;39:13–21. doi: 10.3892/ijo.2011.1012. [DOI] [PubMed] [Google Scholar]
  • 40.Kinoshita T, Hanazawa T, Nohata N, Kikkawa N, Enokida H, Yoshino H, Yamasaki T, Hidaka H, Nakagawa M, Okamoto Y, Seki N. Tumor suppressive microRNA-218 inhibites cancer cell migration and invasion through targeting laminin-332 in head and neck squamous cell carcinoma. Oncotarget. 2012;3:1386–1400. doi: 10.18632/oncotarget.709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Goto Y, Kojima S, Nishikawa R, Enokida H, Chiyomaru T, Kinoshita T, Nakagawa M, Naya Y, Ichikawa T, Seki N. The microRNA-23b/27b/24-1 cluster is a disease progression marker and tumor suppressor in prostate cancer. Oncotarget. 2014;5:7748–7759. doi: 10.18632/oncotarget.2294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Goto Y, Kojima S, Nishikawa R, Kurozumi A, Kato M, Enokida H, Matsushita R, Yamazaki K, Ishida Y, Nakagawa M, Naya Y, Ichikawa T, Seki N. MicroRNA expression signature of castration-resistant prostate cancer: the microRNA-221/222 cluster function as a tumour suppressor and disease progression marker. Br J Cancer. 2015;113:1055–1065. doi: 10.1038/bjc.2015.300. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials


Articles from Oncotarget are provided here courtesy of Impact Journals, LLC

RESOURCES