Skip to main content
Cancer Science logoLink to Cancer Science
. 2018 Mar 9;109(4):1239–1253. doi: 10.1111/cas.13526

Impact of novel oncogenic pathways regulated by antitumor miR‐451a in renal cell carcinoma

Yasutaka Yamada 1,2, Takayuki Arai 1,2, Sho Sugawara 1,2, Atsushi Okato 1,2, Mayuko Kato 2, Satoko Kojima 3, Kazuto Yamazaki 4, Yukio Naya 3, Tomohiko Ichikawa 2, Naohiko Seki 1,
PMCID: PMC5891191  PMID: 29417701

Abstract

Recent analyses of our microRNA (miRNA) expression signatures obtained from several types of cancer have provided novel information on their molecular pathology. In renal cell carcinoma (RCC), expression of microRNA‐451a (miR‐451a) was significantly downregulated in patient specimens and low expression of miR‐451a was significantly associated with poor prognosis of RCC patients (P = .00305) based on data in The Cancer Genome Atlas. The aims of the present study were to investigate the antitumor roles of miR‐451a and to identify novel oncogenic networks it regulated in RCC cells. Ectopic expression of miR‐451a significantly inhibited cancer cell migration and invasion by RCC cell lines, suggesting that miR‐451a had antitumor roles. To identify oncogenes regulated by miR‐451a in RCC cells, we analyzed genome‐wide gene expression data and examined information in in silico databases. A total of 16 oncogenes and were found to be possible targets of miR‐451a regulation. Interestingly, high expression of 9 genes (PMM2,CRELD2,CLEC2D,SPC25,BST2,EVL,TBX15,DPYSL3, and NAMPT) was significantly associated with poor prognosis. In this study, we focused on phosphomannomutase 2 (PMM2), which was the most strongly associated with prognosis. Overexpression of PMM2 was detected in clinical specimens and Spearman's rank test indicated a negative correlation between the expression levels of miR‐451a and PMM2 (P = .0409). Knockdown of PMM2 in RCC cells inhibited cancer cell migration and invasion, indicating overexpression of PMM2 could promote malignancy. Analytic strategies based on antitumor miRNAs is an effective tool for identification of novel pathways of cancer.

Keywords: antitumor, microRNA, miR‐451a, PMM2, renal cell carcinoma

1. INTRODUCTION

Renal cell carcinoma (RCC) is the most common form of kidney cancer and is diagnosed in more than 350 000 patients worldwide, making it the seventh most common site for tumors.1 Although patients with stage I RCC had a 5‐years survival rate above 90%, those with advanced RCC had a 5‐years survival rate of only 23%.2 In fact, 25%‐30% of patients have metastasis at the time of diagnosis.3 Furthermore, distant metastasis and recurrence are found even when surgical resection is carried out for localized RCC, cases that are associated with poor prognosis. Recently, molecular targeted therapy and immunotherapy have been used for patients with metastatic or recurrent RCC. However, the therapeutic benefits are limited, and RCC is generally insensitive to radiation and chemotherapy. Thus, development of novel therapeutic strategies is needed.2 We believe that novel genomic approaches are required to elucidate the underlying molecular mechanism of metastatic RCC.

MicroRNA (miRNA) is a single‐stranded low molecular RNA of 19‐22 bases that possesses important functions. For example, miRNA regulates the expression of target genes by inhibiting translation and/or accelerates the degradation of functional RNAs (protein coding/non‐protein coding genes).4, 5 In human cells, a single miRNA can regulate many different protein‐coding or non‐coding mRNAs and a single mRNA can be regulated by several different miRNAs.6 Thus, aberrant expression of miRNA could disrupt regulated RNA networks in cancer cells. Therefore, it is important to elucidate the aberrant expression of miRNAs in each type of cancer to better understand the molecular mechanism of cancer pathogenesis.4, 7, 8

We previously identified antitumor miRNAs that regulated novel oncogenic pathways based on miRNA expression signatures.9, 10, 11, 12 Downregulation of miR‐451a was detected by our studies of miRNA signatures and several types of cancers.12, 13, 14, 15, 16, 17, 18 Moreover, a large cohort analysis using The Cancer Genome Atlas (TCGA) database (https://cancergenome.nih.gov/) showed that low expression of miR‐451a was associated with poor survival of patients with RCC. The antitumor role of miR‐451a in RCC has been reported;19 however, the molecular pathways regulated by miR‐451a have not been fully elucidated. MicroRNA biogenesis is unique, in that a single miRNA can target a vast number of RNAs in cells. Continuous analyses of antitumor miRNA‐regulated molecular pathways are essential for understanding RCC pathogenesis. Here, we aimed to investigate novel oncogenic pathways regulated by antitumor miR‐451a in RCC cells and involving in RCC pathogenesis.

2. MATERIALS AND METHODS

2.1. Patients, cancer tissue collection, cell lines, and cell culture

We postoperatively collected cancerous and normal tissues from 15 RCC patients at Chiba University Hospital (Chiba, Japan) between 2012 and 2015. Clinicopathological characteristics of the 15 patients are listed in Table 1. Tumor stages were determined by the General Rule for Clinical and Pathological Studies on Renal Cell Carcinoma based on the AJCC‐UICC TNM classification. All patients gave signed, informed consent for the use of the tissues for research purposes.

Table 1.

Clinical features of 15 patients with clear cell renal cell carcinoma

Patient no. Age, years Gender Grade pT INF v ly eg or ig fc im rc rp s
1 71 F G2 T1a a 0 0 eg 1 0 0 0 0
2 74 M G1 > G2 T1a a 0 0 eg 1 0 0 0 0
3 59 M G3 > G2 T1b a 0 0 eg 1 0 0 0 0
4 52 M G2 > G3 > G1 T1a a 0 0 eg 1 0 0 0 0
5 64 M G2 > G3 T1b a 0 0 eg 1 1 0 0 0
6 67 M G2 > G3 > G1 T3a b 1 0 ig 0 1 1 0 0
7 67 M G2 > G3 > G1 T3a b 1 0 ig 1 0 0 0 0
8 59 M G3 > G2 T3a b 1 0 ig 0 0 0 0 0
9 73 M G1 > G3 T2a a 0 1 eg 1 0 0 0 0
10 77 M G1 > G2 T1b a 0 0 eg 1 0 0 0 0
11 77 M G2 > G1 T3a a 1 0 eg 1 0 0 0 0
12 51 M G2 > G1 T1 a 0 0 eg 0 0 0 0 0
13 78 M G2 > G1 > G3 T1b b 0 0 eg 1 0 0 0 0
14 57 M G1 > G2 T1a a 0 0 eg 1 0 0 0 0
15 54 M G2 > G1 T3a a 0 0 eg 0 0 1 0 0

eg, expansive growth; F, female; fc, capsular formation; ig, infiltrative growth; im, intrarenal metastasis; INF, infiltration; ly, lymph node; M, male; rc, renal capsule invasion; rp, pelvis invasion; s, sinus invasion; v, vein.

We used 2 human RCC cell lines (786‐O and A498) obtained from ATCC (Manassas, VA, USA) as previously described.20, 21, 22, 23, 24, 25 These cell lines were maintained in RPMI‐1640 with 10% FBS (HyClone, Logan, UT, USA).

2.2. Transfections with mature miRNA, siRNA, or plasmid vectors

We used the following mature miRNA species in these experiments: mature miRNA and pre‐miR miRNA precursors (has‐miR‐451a; P/N: AM17100; Applied Biosystems, Foster City, CA, USA). The following siRNAs were used: Stealth Select RNAi siRNA, si‐PMM2 (HSS108164 and HSS108165; Invitrogen, Carlsbad, CA, USA), and negative control miRNA/siRNA (P/N: AM17111; Applied Biosystems). PMM2 plasmid vectors were designed and provided by OriGene (cat. no. RC203472; Rockville, MD, USA). MicroRNAs and siRNAs were incubated with Opti‐MEM (Invitrogen) and Lipofectamine RNAiMax transfection reagent (Invitrogen), as previously described.22, 26 Plasmid vectors were incubated with Opti‐MEM and Lipofectamine 3000 reagent (Invitrogen) by forward transfection following the manufacturer's protocol.

2.3. Quantitative real‐time RT‐PCR

Total RNA was extracted from human tissues and cell lines using TRIzol reagent (Invitrogen) according to the manufacturer's protocol, as we described previously.20, 21, 22, 23, 24, 25 The procedure for PCR quantification has been outlined. Expression levels of miR‐451a (assay ID:001141; Applied Biosystems) were analyzed by TaqMan quantitative real‐time RT‐PCR (qRT‐PCR) (TaqMan MicroRNA Assay; Applied Biosystems) and normalized to the expression of RNU48 (assay ID: 001006; Applied Biosystems). TaqMan probes and primers for PMM2 (P/N: Hs00756707_m1; Applied Biosystems), GAPDH (internal control; P/N: Hs02758991_m1; Applied Biosystems), and GUSB (internal control; P/N: Hs00939627_ml; Applied Biosystems) were assay‐on‐demand gene expression products. The relative expression levels were calculated using the 2−ΔΔCT method.

2.4. Cell proliferation, migration, and invasion assays

Cell proliferation, migration, and invasion assays have been described.20, 21, 22, 23, 24, 25

2.5. Identification of putative target genes regulated by miR‐451a in RCC cells

To identify miR‐451a target genes, we used in silico analyses and genome‐wide gene expression analyses, as outlined previously.20, 21, 22, 23, 24, 25 We used the TargetScanHuman 7.0 (August 2015 release) (http://www.targetscan.org/), TCGA, and OncoLnc datasets (http://www.oncolnc.org/) to select and narrow down putative miRNA target genes.27, 28, 29 An oligo microarray (Human Ge 60K; Agilent Technologies) was used for gene expression analysis. We deposited the microarray data into the Gene Expression Omnibus (GEO) database.

2.6. Western blot analysis

Cells were collected 48 hours after transfection, and lysates were prepared. Immunoblotting was undertaken with rabbit anti‐PMM2 antibodies (1:500 dilution, SAB2702078; Sigma‐Aldrich, St. Louis, MO, USA). Anti‐GAPDH antibodies (1:10 000, ab8245; Abcam) were used as an internal loading control. The procedures have been described.20, 21, 22, 23, 24, 25

2.7. Plasmid construction and dual luciferase reporter assays

The partial wild‐type sequence of the PMM2 3′‐UTR or that with deletion of the miR‐451a putative target site was inserted between the SgfI‐PmeI restriction sites in the 3′‐UTR of the hRluc gene in the psiCHECK‐2 vector (C8021; Promega, Madison, WI, USA). The procedures were covered in earlier reports.20, 21, 22, 23, 24, 25

2.8. Immunohistochemistry

We used a tissue microarray that was incubated overnight at 4°C with anti‐PMM2 antibodies (1:100 dilution, SAB2702078; Sigma‐Aldrich). The sliced slides were treated with biotinylated goat antibodies (Histofine SAB‐PO kit; Nichirei, Tokyo, Japan). The procedures were described earlier.20, 21, 22, 23, 24, 25

2.9. Regulation of targets downstream of PMM2 in RCC

We analyzed PMM2‐regulated pathways in RCC cells. We analyzed gene expression using si‐PMM2‐transfected 786‐O cells. Microarray data were used for expression profiling of si‐PMM2 transfectants. The microarray data were deposited into the GEO database (accession no. GSE107008).

2.10. The Cancer Genome Atlas database analysis of RCC

To assess the clinical significance of miRNAs and their targeted genes, we used the RNA sequencing database in TCGA. The definition of high and low expression divided half of the clinical data population in the order of expression. Gene expression and clinical data were obtained from cBioportal and OncoLnc (data downloaded November 1, 2017).27, 28, 29

2.11. Statistical analysis

Relationships between 2 groups and the numerical values obtained by qRT‐PCR were analyzed by Mann‐Whitney U‐tests and paired t tests. Spearman's rank test was used to analyze the correlation between the expression levels of miR‐451a and PMM2. Relationships among more than 3 variables and numerical values were analyzed with Bonferroni‐adjusted Mann‐Whitney U‐tests. Survival analysis was carried out using the Kaplan‐Meier method, log‐rank tests, and multivariable Cox hazard regression analyses with JMP software (version 13; SAS Institute, Cary, NC, USA). Other statistical analyses were carried out using Expert StatView (version 5; SAS Institute).

3. RESULTS

3.1. Expression levels of miR‐451a in RCC clinical specimens and cell lines

Expression levels of miR‐451a were significantly downregulated in RCC tissues compared with those in non‐cancerous tissues (P = .0166; Figure 1A). Furthermore, expression levels of miR‐451a in 786‐O and A498 cells were markedly downregulated (Figure 1A). In the human genome, miR‐451a has formed a miRNA cluster with other miRNAs (miR‐144‐5p, miR‐144‐3p, miR‐451b, and miR‐4732) on human chromosome 17q11.2 region (Figure S1). We also checked expression of these miRNAs in RCC clinical specimens. Expression levels of miR‐144‐5p and miR‐144‐3p were significantly downregulated in RCC tissues. On the contrary, it was revealed that the expression levels of miR‐451b and miR‐4732 were extremely low in normal kidney and RCC tissues (Figure S1).

Figure 1.

Figure 1

Antitumor functions of miR‐451a in renal cell carcinoma. A, Expression levels of miR‐451a in renal cell carcinoma clinical specimens and cell lines. RNU48 was used as an internal control. B, Kaplan‐Meier survival curves, as determined using data from The Cancer Genome Atlas database. C, Cell proliferation was determined by XTT assays 72 hours after transfection with miR‐451a. D, Cell migration activity. E, Cell invasion activity was determined using Matrigel assays. *P < .0001

To investigate the molecular mechanisms of silencing of miR‐451a in RCC cells, 786‐O cells were treated with the demethylating agent 5‐aza‐2′‐deoxycytidine. Expression of miR‐451a was not dramatically elevated by 5‐aza‐2′‐deoxycytidine treatment (Figure S1).

A large cohort analysis (n = 506) using data from TCGA database showed that low expression of miR‐451a was associated with poor survival of patients with RCC (P = .00305; Figure 1B).

3.2. Effects of ectopic expression of miR‐451a in RCC cell lines

To investigate the antitumor functions of miR‐451a in RCC cells, we applied to gain‐of‐function analyses in this study. Ectopic expression of miR‐451a in RCC cell lines (786‐O and A498) did not affect cell proliferation (Figure 1C). Cell migration and invasion activities were significantly inhibited in miR‐451a transfectant cells compared with those in mock or miRNA‐control transfectant cells (Figure 1D,E).

3.3. Screening of candidate targets by miR‐451a regulation in RCC cells

We undertook in silico and gene expression analyses to identify genes targeted by miR‐451a. The strategy for selection of miR‐451a target genes is shown in Figure 2. Using the TargetScanHuman 7.0 database, we identified 541 genes that had putative target sites for miR‐451a in their 3′‐UTRs. Among these genes, we identified 64 that showed increased expression levels in RCC tissues (fold‐change >1.5) using the database (GEO database accession number: GSE36895).

Figure 2.

Figure 2

Flow chart illustrating the analytic strategy for miR‐451a targets in renal cell carcinoma cells. A total of 541 genes were putative targets of miR‐451a in the TargetScan database analysis (release 7.0). We selected 16 genes as putative targets of miR‐451a in renal cell carcinoma cells. GEO, Gene Expression Omnibus; TCGA, The Cancer Genome Atlas

Next, we identified 16 genes that were downregulated after transfection of 786‐O and A498 cells with miR‐451a (average log2 ratio < −1.0; Table 2). Next, using the OncoLnc database, we investigated whether the expression levels of the 16 candidate genes affected the prognosis of RCC patients. Kaplan‐Meier survival curves revealed that high expression levels of 9 of the genes were associated with poor prognosis in patients with RCC (Figure 3). Finally, we focused on PMM2 because it showed the most evident difference in OncoLnc prognostic analysis (P = .000000218; Table 2, Figure 3) and there are few reports on cancer studies.

Table 2.

Candidate target genes regulated by miR‐451a in renal cell carcinoma cells

Gene symbol Gene name Conserved sites count Poorly conserved sites count GEO expression data fold‐change (tumor/normal) A498 miR‐451a transfection (Log2 ratio) 786‐O miR‐451a transfection (Log2 ratio) Average A498/786‐O miR‐451a transfection (Log2 ratio) Cytoband TCGA data OS (P‐value)
PMM2 Phosphomannomutase 2 1 0 1.580 −1.617 −1.020 −1.319 hs|16p13.2 .000000218
CRELD2 Cysteine‐rich with EGF‐like domains 2 0 1 1.656 −1.724 −2.256 −1.990 hs|22q13.33 .000000831
CLEC2D C‐type lectin domain family 2, member D 0 1 2.557 −1.014 −1.455 −1.235 hs|12p13.31 .00009140
SPC25 SPC25, NDC80 kinetochore complex component 0 1 3.213 −2.497 −2.321 −2.409 hs|2q31.1 .00009800
BST2 Bone marrow stromal cell antigen 2 0 1 2.061 −0.668 −1.723 −1.196 hs|19p13.11 .00015300
EVL Enah/Vasp‐like 0 1 1.896 −1.197 −1.510 −1.354 hs|14q32.2 .00125000
TBX15 T‐box 15 0 1 4.118 −1.224 −1.618 −1.421 hs|1p12 .00193000
DPYSL3 Dihydropyrimidinase‐like 3 0 1 2.326 −1.325 −1.344 −1.335 hs|5q32 .00389000
NAMPT Nicotinamide phosphoribosyltransferase 0 1 2.174 −2.369 −0.534 −1.452 hs|7q22.3 .01380000
MEGF6 Multiple EGF‐like‐domains 6 0 1 2.112 −0.917 −1.811 −1.364 hs|1p36.32 .15100000
CRIP2 Cysteine‐rich protein 2 0 1 1.590 −1.899 −1.593 −1.746 hs|14q32.33 .18800000
CAV1 Caveolin 1, caveolae protein, 22kda 0 1 6.729 −1.196 −2.144 −1.670 hs|7q31.2 .20100000
PSMB8 Proteasome (prosome, macropain) subunit, beta type, 8 1 0 2.682 −1.502 −1.906 −1.704 hs|6p21.32 .25500000
CDH11 Cadherin 11, type 2, OB‐cadherin (osteoblast) 0 1 1.847 −0.791 −1.788 −1.290 hs|16q21 .42600000
EGLN3 Egl‐9 family hypoxia‐inducible factor 3 0 1 13.668 −0.740 −1.940 −1.340 hs|14q13.1 .68800000
SLC39A14 Solute carrier family 39 (zinc transporter), member 14 0 1 2.057 −1.391 −1.386 −1.389 hs|8p21.3 .83600000

GEO, Gene Expression Omnibus; OS, overall survival; TCGA, The Cancer Genome Atlas.

Figure 3.

Figure 3

The Cancer Genome Atlas database analysis of putative targets of miR‐451a in renal cell carcinoma. Kaplan‐Meier plots of overall survival with log‐rank tests for 16 genes with high and low expression from The Cancer Genome Atlas database

3.4. PMM2 directly regulated by miR‐451a in RCC cells

Expression levels of PMM2/PMM2 were reduced by miR‐451a transfection at mRNA and protein levels (Figure 4A,B).

Figure 4.

Figure 4

Regulation of PMM2 expression by miR‐451a in renal cell carcinoma cells. A, Expression levels of PMM2 mRNA 48 hours after transfection of 10 nmol/L miR‐451a into cell lines. GUSB was used as an internal control. *P < .0001. B, Protein expression of phosphomannomutase 2 (PMM2) 72 hours after transfection with miR‐451a. GAPDH was used as a loading control. C, miR‐451a binding sites in the 3′‐UTR of PMM2 mRNA. D, Dual luciferase reporter assays using vectors encoding putative miR‐451a target sites (positions 1127‐1133) in the PMM2 3′‐UTR for both wild‐type and deleted regions. Normalized data were calculated as the ratio of Renilla/firefly luciferase activities. *< .005

Furthermore, we carried out luciferase reporter assays to elucidate whether PMM2 mRNA had a functional target site for miR‐451a. The TargetScan database predicted that miR‐451a was bound at position 1127‐1133 in the 3′‐UTR of PMM2 (Figure 4C). We used vectors encoding a partial wild‐type sequence of the 3′‐UTR of PMM2 mRNA, including the predicted miR‐451a target site, or a vector lacking the miR‐451a target site. Luminescence intensity was significantly reduced by co‐transfection with miR‐451a and the vector carrying the wild‐type 3′‐UTR of PMM2. In contrast, luminescence intensity was not reduced when the target site of miR‐451a was deleted from the vectors (Figure 4D).

3.5. Effects of PMM2 knockdown in RCC cell lines

To investigate the functional significance of PMM2, we carried out loss‐of‐function studies using transfection of si‐PMM2 into 786‐O and A498 cells. First, we evaluated the knockdown efficiency of si‐PMM2 transfection, using 2 types of si‐PMM2 (si‐PMM2_1 and si‐PMM2_2). Using qRT‐PCR and Western blot analyses, we confirmed that the expression levels of PMM2 mRNA and protein were significantly reduced (Figure 5A,B). Furthermore, functional assays showed that si‐PMM2 transfection significantly inhibited cell proliferation, migration, and invasion in comparison with mock or siRNA‐control transfected cells (Figure 5C‐E).

Figure 5.

Figure 5

Effects of PMM2 silencing in renal cell carcinoma cell lines. A, PMM2 mRNA expression 48 hours after transfection with 10 nM si‐PMM2 into renal cell carcinoma cell lines. GUSB was used as an internal control. B, Phosphomannomutase 2 (PMM2) protein expression 72 h after transfection with si‐PMM2. GAPDH was used as a loading control. C, Cell proliferation was determined with XTT assays 72 hours after transfection with 10 nM si‐PMM2_1 or si‐PMM2_2. D, Cell migration assessed by wound healing assays. E, Cell invasion activity was determined using a Matrigel system. *P < .0001

3.6. Expression of PMM2/PMM2 in RCC clinical specimens and cell lines

We used qRT‐PCR to investigate the mRNA expression levels of PMM2 in 15 pairs of RCC tissues, adjacent noncancerous tissues and RCC cell lines. Expression of PMM2 was significantly upregulated in RCC tissues compared with that in normal tissues (P = .0026; Figure 6A) and also markedly upregulated in RCC cell lines. Furthermore, Spearman's rank test revealed a negative correlation between the expression levels of miR‐451a and PMM2 (P = .0409, R = −.38; Figure 6B).

Figure 6.

Figure 6

Expression of PMM2 in clinical specimens of renal cell carcinoma. A, Expression levels of PMM2 in renal cell carcinoma clinical specimens. GUSB was used as an internal control. B, A negative correlation between PMM2 expression and miR‐451a (R = −.38 and P = .0409). Spearman's rank test was used to evaluate the correlation. C, Immunostaining showed that phosphomannomutase 2 (PMM2) was strongly expressed in several cancer lesions (magnification, ×100 [left panels] and ×400 [right panels])

Moreover, we carried out immunohistochemistry to analyze PMM2 protein expression in an RCC tissue microarray (cat. no. KD806; US Biomax, Rockville, MD, USA). Patient characteristics for samples used in the tissue microarray are as described in http://www.biomax.us/tissue-arrays/Kidney/KD806. The PMM2 protein was strongly expressed in several cancer lesions (Figure 6C).

3.7. Effects of co‐transfection of PMM2/miR‐451a in 786‐O cells

We undertook PMM2 rescue studies in 786‐O cells to elucidate whether the molecular pathway of PMM2/miR‐451a was significant for the progression of RCC. Figure 7A shows the results of Western blot analysis of PMM2 protein expression. Functional assays showed that the migration and invasive abilities of RCC cells were significantly recovered by PMM2 and miR‐451a transfection compared with cells transfected with miR‐451a alone (Figure 7C,D). These results suggested that PMM2 had an important role in the aggressiveness of RCC.

Figure 7.

Figure 7

Effects of co‐transfection of PMM2/miR‐451a in 786‐O renal cell carcinoma cells. A, Phosphomannomutase 2 (PMM2) protein expression was evaluated by Western blot analysis of 786‐O cells 72 hours after reverse transfection with miR‐451a and 48 hours after forward transfection with the PMM2 vector. GAPDH was used as a loading control. B, Cell proliferation was determined using XTT assays 72 hours after reverse transfection with miR‐451a and 48 hours after forward transfection with the PMM2 vector. C, Cell migration activity was assessed by wound healing assays 48 hours after reverse transfection with miR‐451a and 24 hours after forward transfection with the PMM2 vector. D, Cell invasive activity was characterized by invasion assays 48 hours after reverse transfection with miR‐451a and 24 h after forward transfection with PMM2 vector. *P < .0001. VC, Vector Control

3.8. Downstream genes affected by silencing of PMM2 in 786‐O cells

We carried out genome‐wide gene expression analyses by transfecting si‐PMM2 into 786‐O cells to elucidate which genes were modulated by PMM2 knockdown. We focused on genes that were significantly downregulated by transfection of both si‐PMM2_1 and si‐PMM2_2 (average log2 [si‐PMM2/mock] < −1.0). Genes significantly downregulated by silencing of PMM2 are listed in Table 3. Among these genes, high expression levels of CD7, CCNE2, ASB2, RCN3, HSF1, PAQR4, CD37, SOX11, XAF1, and DEPDC1 were associated with poor prognosis in RCC patients based on TCGA database (Figure 8).

Table 3.

Candidate downstream genes modulated by PMM2 in renal cell carcinoma cells

Gene symbol Gene name GEO expression data fold‐change (tumor/normal) Log2 (si‐PMM2_1/mock) Log2 (si‐PMM2_2/mock) Average Log2 (si‐PMM2/mock) Cytoband TCGA data OS (P‐value)
GNLY Granulysin 5.987 −2.465 −2.548 −2.506 hs|2p11.2 .09360000
TTF2 Transcription termination factor, RNA polymerase II 1.312 −3.407 −1.139 −2.273 hs|1p13.1 .27800000
CD7 CD7 molecule 1.981 −1.935 −2.522 −2.228 hs|17q25.3 .00001220
CCNE2 Cyclin E2 2.430 −3.101 −1.251 −2.176 hs|8q22.1 .00664000
NLGN1 Neuroligin 1 2.422 −1.377 −2.500 −1.938 hs|3q26.31 .03910000*
TMEM184B Transmembrane protein 184B 1.525 −1.128 −2.639 −1.883 hs|22q13.1 .96300000
ASB2 Ankyrin repeat and SOCS box containing 2 1.804 −2.534 −1.194 −1.864 hs|14q32.12 .01680000
ASPH Aspartate β‐hydroxylase 1.727 −1.414 −2.244 −1.829 hs|8q12.3 .22000000
MAML3 Mastermind‐like 3 (Drosophila) 1.364 −1.644 −1.923 −1.784 hs|4q31.1 .00010700*
RCN3 Reticulocalbin 3, EF‐hand calcium binding domain 1.314 −2.325 −1.103 −1.714 hs|19q13.33 .00000636
CAMK1 Calcium/calmodulin‐dependent protein kinase I 1.324 −1.566 −1.471 −1.519 hs|3p25.3 .54600000
P2RX7 Purinergic receptor P2X, ligand gated ion channel, 7 8.047 −1.746 −1.134 −1.440 hs|12q24.31 .95700000
KDM1B Lysine (K)‐specific demethylase 1B 1.342 −1.291 −1.544 −1.417 hs|6p22.3 .00939000*
OAS3 2′‐5′‐oligoadenylate synthetase 3, 100kda 2.919 −1.490 −1.287 −1.388 hs|12q24.13 .97300000
HSF1 Heat shock transcription factor 1 1.324 −1.685 −1.083 −1.384 hs|8q24.3 .00126000
PMM2 Phosphomannomutase 2 1.580 −1.304 −1.376 −1.340 hs|16p13.2 .000000218
CDKN2B Cyclin‐dependent kinase inhibitor 2B (p15, inhibits CDK4) 3.355 −1.237 −1.434 −1.335 hs|9p21.3 .00542000*
PAQR4 Progestin and adipoq receptor family member IV 5.134 −1.567 −1.087 −1.327 hs|16p13.3 .00152000
CD37 CD37 molecule 6.272 −1.154 −1.420 −1.287 hs|19q13.33 .01880000
SOX11 SRY (sex determining region Y)‐box 11 5.969 −1.334 −1.236 −1.285 hs|2p25.2 .04050000
XAF1 XIAP associated factor 1 2.349 −1.044 −1.489 −1.266 hs|17p13.1 .00001720
DEPDC1 DEP domain containing 1 2.606 −1.253 −1.197 −1.225 hs|1p31.2 .00011100
SAMHD1 SAM domain and HD domain 1 2.764 −1.225 −1.163 −1.194 hs|20q11.23 .16600000
P2RY1 Purinergic receptor p2y, g‐protein coupled, 1 1.427 −1.167 −1.142 −1.155 hs|3q25.2 .00051400*
LOC100422737 Uncharacterized loc100422737 1.322 −1.093 −1.199 −1.146 hs|6q21 No data
DOCK9 Dedicator of cytokinesis 9 1.614 −1.158 −1.087 −1.123 hs|13q32.3 .00001100*
SLC39A10 Solute carrier family 39 (zinc transporter), member 10 1.331 −1.074 −1.137 −1.105 hs|2q32.3 .01820000*
RNF125 Ring finger protein 125, e3 ubiquitin protein ligase 1.526 −1.060 −1.060 −1.060 hs|18q12.1 .03820000*

GEO, Gene Expression Omnibus; OS, overall survival; TCGA, The Cancer Genome Atlas.

*, poor prognosis with low expression.

Figure 8.

Figure 8

The Cancer Genome Atlas database analysis of PMM2‐mediated downstream genes in renal cell carcinoma. Kaplan‐Meier curves of 10 genes whose high expression levels were associated with poor prognosis in renal cell carcinoma

3.9. Clinical significance of PMM2 in RCC pathogenesis using TCGA database

To investigate the clinical significance of PMM2 in RCC pathogenesis, we asked whether the PMM2 expression level affected disease‐free survival (DFS) in RCC. We found that high expression levels of PMM2 were significantly associated with low DFS in RCC patients (P < .0001; Figure 9A). Furthermore, we analyzed the relationships among PMM2 expression levels and tumor T stage, lymph node metastasis, disease stage, and histologic grade in RCC. PMM2 expression levels were significantly higher in the more advanced tumor stages and histologic grades (Figure 9B‐E).

Figure 9.

Figure 9

Kaplan‐Meier curves for disease‐free survival based on PMM2 expression in patients with renal cell carcinoma, and expression levels of PMM2 according to T stage, N stage, tumor stage, and histologic grade. All patients’ data were obtained from The Cancer Genome Atlas database. A, Kaplan‐Meier survival curves for disease‐free survival based on PMM2 expression in patients with renal cell carcinoma. B‐E, Relationships between expression levels of PMM2 and T stage, N stage, disease stage, and histologic grade. *P < .05, **P < .005, ***P < .0001

Additionally, we undertook univariable and multivariable Cox hazard regression analyses to investigate the clinical significance of PMM2 expression along with other clinical factors in the overall survival of RCC patients. Multivariate analysis showed that high PMM2 expression and tumor stage were independent predictive factors for overall survival (hazard ratio = 1.26, P = .0487 and hazard ratio = 0.76, P = .0353, respectively; Table 4). These results suggested that PMM2 may be closely associated with tumor progression and malignancy in RCC.

Table 4.

Univariable and multivariable Cox hazard regression models for overall survival in renal cell carcinoma

Variable Group Univariable Multivariable
HR 95% CI P‐value HR 95% CI P‐value
PMM2 expression High/low 1.35 1.10‐1.68 .0047 1.26 1.002‐1.58 .0487
Age, years <60/≥60 0.78 0.64‐0.97 .0265 0.82 0.66‐1.01 .0703
Gender Male/female 1.13 0.90‐1.41 .279
Stage I+II/III+IV 0.67 0.52‐0.84 .0007 0.76 0.59‐0.98 .0353
Histologic grade G1+2/G3+4 0.68 0.55‐0.85 .0006 0.81 0.64‐1.03 .0896
Lymph node metastasis Positive/negative 1.25 0.44‐2.74 .648

‐, Not analyzed in multivariate analysis; CI, confidence interval; HR, hazard ratio.

4. DISCUSSION

The aberrant expression of miRNAs disrupts regulated RNA networks in various type of cancer cells and therefore contributes substantially to the pathogenesis of human cancer.30 Therefore, understanding miRNA signatures is essential to clarifying their roles in the pathology of human cancer cells. We previously characterized several miRNAs that had significant antitumor functions in RCC cells. For example, miR‐101 was significantly reduced in RCC tissues and the restoration of miR‐101 inhibited RCC aggressiveness through targeting the UHRF1 gene.9 More recently, we showed that miR‐10a‐5p was downregulated in primary and tyrosine‐kinase inhibitor‐treated RCC specimens and directly regulated the SKA1 gene. SKA1 was overexpressed and knockdown of SKA1 inhibited migration and invasion of RCC cells.22

Our present data showed that expression of miR‐451a was significantly reduced in primary RCC. Furthermore, ectopic expression of miR‐451a resulted in inhibition of cancer cell migratory and invasive abilities in RCC, indicating that miR‐451a functions as a tumor suppressor in RCC. Previous reports showed that expression of miR‐451 was downregulated in several types of human cancers. In bladder cancer, miR‐451 expression was significantly reduced in cancer tissues compared with adjacent tissues and that restoration of miR‐451 reduced cancer cell migration and invasive abilities.31, 32 Furthermore, low expression of miR‐451 was associated with advanced tumor stage and high pathological grade in bladder cancer.32 Our previous data showed that miR‐451a acted as an antitumor miRNA through targeting ESDN/DCBLD2 in head and neck squamous cell carcinoma.12

In this study, we identified 16 putative oncogenic targets by miR‐451a regulation in RCC cells. Interestingly, among these targets, expression of 9 genes (PMM2, CRELD2, CLEC2D, SPC25, BST2, EVL, TBX15, DPYSL3, and NAMPT) were significantly associated with poor prognosis of the patients with RCC by TCGA database analyses. These targets might be potential therapeutic targets for RCC and the search for RNA networks controlled by antitumor miR‐451a and its targets are important for elucidation of the pathogenesis of RCC. In the present study, we identified PMM2 as an oncogenic gene in RCC cells. Moreover, overexpressed PMM2 was involved RCC pathogenesis. PMM2 codes for an enzyme that converts mannose‐6‐phosphate to mannose‐1‐phosphate and participates in a metabolic pathway in glycan synthesis.33, 34, 35 Protein glycosylation is an important contributor to cancer progression, including cell growth, tumor‐induced immunomodulation, and eventual metastasis.36 Previous reports indicated that cancer cells, including RCC, are characterized by an aberrant increase in protein N‐glycosylation.37, 38 In clear cell RCC, upregulation of protein glycosylation in cancer cells may be useful in diagnosis and determining disease prognosis.39 Furthermore, N‐glycosylation is involved in cell adhesion and is associated with reduced expression of E‐cadherin, which modulates the metastatic potential of cancer cells.37, 40 We hypothesize that this pathway might contribute to cancer pathogenesis such that upregulation of PMM2 might enhance cancer cell aggressiveness by increased N‐glycosylation. In support of this hypothesis, TCGA database showed that high expression of PMM2 was associated with poor prognosis even in other types of cancer, for example, bladder cancer, breast cancer, head and neck squamous cell carcinoma, glioma, and melanoma (Figure S2).

Interestingly, a previous report showed that miR‐451 was controlled by glucose levels and regulated cancer aggressiveness through the AMP‐activated protein kinase pathway and mTOR activation in glioblastoma.15 Thus, miR‐451 regulated pathways may be involved in glucose‐related metabolic pathways and regulate cancer aggressiveness. Further research into miR‐451/PMM2‐modulated pathways in cancer will be necessary. To investigate PMM2‐mediated pathways in RCC, we undertook genome‐wide gene expression analyses using PMM2 knockdown cells. We identified 27 genes that were regulated by PMM2 in RCC. Among them, the expression of 10 genes was elevated (CD7, CCNE2, ASB2, RCN3, HSF1, PAQR4, CD37, SOX11, XAF1, and DEPDC1) and their expression levels were significantly associated with poor prognosis in RCC patients according to TCGA database (P < .05). Elucidation of novel PMM2‐mediated pathways may improve our understanding of RCC aggressiveness.

In conclusion, our data revealed that expression of miR‐451a was significantly downregulated in clinical RCC cells. Moreover, miR‐451a acted as a tumor suppressor through the targeting of PMM2. Phosphomannomutase 2 was strongly expressed in RCC cells and its silencing significantly inhibited cancer cell migration and invasive abilities. In clinical analysis, high expression of PMM2 was significantly associated with shorter DFS and lower survival rates. In short, PMM2‐regulated genes are deeply involved in RCC pathogenesis. Elucidation of the pathways mediated by the miR‐451a/PMM2 axis should improve our understanding of oncogenic mechanisms and lead to new treatment strategies in RCC.

CONFLICT OF INTEREST

The authors have no conflict of interest.

Supporting information

 

 

ACKNOWLEDGMENTS

The present study was supported by the Japan Society for the Promotion of Science (KAKENHI grant nos. 16K20125, 17K11160, 16H05462, and 15K10801).

Yamada Y, Arai T, Sugawara S, et al. Impact of novel oncogenic pathways regulated by antitumor miR‐451a in renal cell carcinoma. Cancer Sci. 2018;109:1239–1253. https://doi.org/10.1111/cas.13526

Funding information

Japan Society for the Promotion of Science (16K20125, 17K11160, 16H05462, and 15K10801).

REFERENCES

  • 1. Capitanio U, Montorsi F. Renal cancer. Lancet. 2016;387:894‐906. [DOI] [PubMed] [Google Scholar]
  • 2. Figlin R, Sternberg C, Wood CG. Novel agents and approaches for advanced renal cell carcinoma. J Urol. 2012;188:707‐715. [DOI] [PubMed] [Google Scholar]
  • 3. Gupta K, Miller JD, Li JZ, Russell MW, Charbonneau C. Epidemiologic and socioeconomic burden of metastatic renal cell carcinoma (mRCC): a literature review. Cancer Treat Rev. 2008;34:193‐205. [DOI] [PubMed] [Google Scholar]
  • 4. Goto Y, Kurozumi A, Enokida H, Ichikawa T, Seki N. Functional significance of aberrantly expressed microRNAs in prostate cancer. Int J Urol. 2015;22:242‐252. [DOI] [PubMed] [Google Scholar]
  • 5. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281‐297. [DOI] [PubMed] [Google Scholar]
  • 6. Friedman RC, Farh KK, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009;19:92‐105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Koshizuka K, Hanazawa T, Fukumoto I, Kikkawa N, Okamoto Y, Seki N. The microRNA signatures: aberrantly expressed microRNAs in head and neck squamous cell carcinoma. J Hum Genet. 2017;62:3‐13. [DOI] [PubMed] [Google Scholar]
  • 8. Kurozumi A, Goto Y, Okato A, Ichikawa T, Seki N. Aberrantly expressed microRNAs in bladder cancer and renal cell carcinoma. J Hum Genet. 2017;62:49‐56. [DOI] [PubMed] [Google Scholar]
  • 9. Goto Y, Kurozumi A, Nohata N, et al. The microRNA signature of patients with sunitinib failure: regulation of UHRF1 pathways by microRNA‐101 in renal cell carcinoma. Oncotarget. 2016;7:59070‐59086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Goto Y, Kojima S, Nishikawa R, 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. 2015;113:1055‐1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Koshizuka K, Nohata N, Hanazawa T, et al. Deep sequencing‐based microRNA expression signatures in head and neck squamous cell carcinoma: dual strands of pre‐miR‐150 as antitumor miRNAs. Oncotarget. 2017;8:30288‐30304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Fukumoto I, Kinoshita T, Hanazawa T, et al. Identification of tumour suppressive microRNA‐451a in hypopharyngeal squamous cell carcinoma based on microRNA expression signature. Br J Cancer. 2014;111:386‐394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Pan X, Wang R, Wang ZX. The potential role of miR‐451 in cancer diagnosis, prognosis, and therapy. Mol Cancer Ther. 2013;12:1153‐1162. [DOI] [PubMed] [Google Scholar]
  • 14. Minna E, Romeo P, Dugo M, et al. miR‐451a is underexpressed and targets AKT/mTOR pathway in papillary thyroid carcinoma. Oncotarget. 2016;7:12731‐12747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Godlewski J, Bronisz A, Nowicki MO, Chiocca EA, Lawler S. microRNA‐451: a conditional switch controlling glioma cell proliferation and migration. Cell Cycle. 2010;9:2742‐2748. [PubMed] [Google Scholar]
  • 16. Liu Z, Miao T, Feng T, et al. miR‐451a inhibited cell proliferation and enhanced tamoxifen sensitive in breast cancer via macrophage migration inhibitory factor. Biomed Res Int. 2015;2015:207684. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 17. Gao Z, Liu R, Liao J, et al. Possible tumor suppressive role of the miR‐144/451 cluster in esophageal carcinoma as determined by principal component regression analysis. Mol Med Rep. 2016;14:3805‐3813. [DOI] [PubMed] [Google Scholar]
  • 18. Zhang F, Huang W, Sheng M, Liu T. MiR‐451 inhibits cell growth and invasion by targeting CXCL16 and is associated with prognosis of osteosarcoma patients. Tumour Biol. 2015;36:2041‐2048. [DOI] [PubMed] [Google Scholar]
  • 19. Tang Y, Wan W, Wang L, Ji S, Zhang J. microRNA‐451 inhibited cell proliferation, migration and invasion through regulation of MIF in renal cell carcinoma. Int J Clin Exp Pathol. 2015;8:15611‐15621. [PMC free article] [PubMed] [Google Scholar]
  • 20. Okato A, Arai T, Kojima S, et al. Dual strands of pre‐miR150 (miR1505p and miR1503p) act as antitumor miRNAs targeting SPOCK1 in naive and castration‐resistant prostate cancer. Int J Oncol. 2017;51:245‐256. [DOI] [PubMed] [Google Scholar]
  • 21. Yamada Y, Koshizuka K, Hanazawa T, et al. Passenger strand of miR‐145‐3p acts as a tumor‐suppressor by targeting MYO1B in head and neck squamous cell carcinoma. Int J Oncol. 2018;52:166‐178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Arai T, Okato A, Kojima S, et al. Regulation of spindle and kinetochore‐associated protein 1 by antitumor miR‐10a‐5p in renal cell carcinoma. Cancer Sci. 2017;108:2088‐2101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Yamada Y, Nishikawa R, Kato M, et al. Regulation of HMGB3 by antitumor miR‐205‐5p inhibits cancer cell aggressiveness and is involved in prostate cancer pathogenesis. J Hum Genet. 2018;63:195‐205. [DOI] [PubMed] [Google Scholar]
  • 24. Koshizuka K, Hanazawa T, Kikkawa N, et al. Regulation of ITGA3 by the anti‐tumor miR‐199 family inhibits cancer cell migration and invasion in head and neck cancer. Cancer Sci. 2017;108:1681‐1692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Nishikawa R, Goto Y, Sakamoto S, et al. Tumor‐suppressive microRNA‐218 inhibits cancer cell migration and invasion via targeting of LASP1 in prostate cancer. Cancer Sci. 2014;105:802‐811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Kurozumi A, Goto Y, Matsushita R, et al. Tumor‐suppressive microRNA‐223 inhibits cancer cell migration and invasion by targeting ITGA3/ITGB1 signaling in prostate cancer. Cancer Sci. 2016;107:84‐94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. OncoLnc JA. Linking TCGA survival data to mRNAs, miRNAs, and lncRNAs. PeerJ Comput Sci. 2016;2:e67. [Google Scholar]
  • 28. Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6:pl1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401‐404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Esquela‐Kerscher A, Slack FJ. Oncomirs ‐ microRNAs with a role in cancer. Nat Rev Cancer. 2006;6:259‐269. [DOI] [PubMed] [Google Scholar]
  • 31. Wang J, Zhao X, Shi J, et al. miR‐451 suppresses bladder cancer cell migration and invasion via directly targeting c‐Myc. Oncol Rep. 2016;36:2049‐2058. [DOI] [PubMed] [Google Scholar]
  • 32. Zeng T, Peng L, Chao C, et al. miR‐451 inhibits invasion and proliferation of bladder cancer by regulating EMT. Int J Clin Exp Pathol. 2014;7:7653‐7662. [PMC free article] [PubMed] [Google Scholar]
  • 33. Freeze HH. Towards a therapy for phosphomannomutase 2 deficiency, the defect in CDG‐Ia patients. Biochem Biophys Acta. 2009;1792:835‐840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Schiff M, Roda C, Monin ML, et al. Clinical, laboratory and molecular findings and long‐term follow‐up data in 96 French patients with PMM2‐CDG (phosphomannomutase 2‐congenital disorder of glycosylation) and review of the literature. J Med Genet. 2017;54:843‐851. [DOI] [PubMed] [Google Scholar]
  • 35. Chan B, Clasquin M, Smolen GA, et al. A mouse model of a human congenital disorder of glycosylation caused by loss of PMM2. Hum Mol Genet. 2016;25:2182‐2193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Stowell SR, Ju T, Cummings RD. Protein glycosylation in cancer. Annu Rev Pathol. 2015;10:473‐510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Borzym‐Kluczyk M, Radziejewska I, Cechowska‐Pasko M, Darewicz B. Reduced expression of E‐cadherin and increased sialylation level in clear cell renal cell carcinoma. Acta Biochim Pol. 2017;64:465‐470. [DOI] [PubMed] [Google Scholar]
  • 38. Dennis JW, Granovsky M, Warren CE. Protein glycosylation in development and disease. BioEssays. 1999;21:412‐421. [DOI] [PubMed] [Google Scholar]
  • 39. Gbormittah FO, Bones J, Hincapie M, Tousi F, Hancock WS, Iliopoulos O. Clusterin glycopeptide variant characterization reveals significant site‐specific glycan changes in the plasma of clear cell renal cell carcinoma. J Proteome Res. 2015;14:2425‐2436. [DOI] [PubMed] [Google Scholar]
  • 40. Hsiao CT, Cheng HW, Huang CM, et al. Fibronectin in cell adhesion and migration via N‐glycosylation. Oncotarget. 2017;8:70653‐70668. [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 Cancer Science are provided here courtesy of Wiley

RESOURCES