Open Access

Regulation of the collagen cross-linking enzymes LOXL2 and PLOD2 by tumor-suppressive microRNA-26a/b in renal cell carcinoma

  • Authors:
    • Akira Kurozumi
    • Mayuko Kato
    • Yusuke Goto
    • Ryosuke Matsushita
    • Rika Nishikawa
    • Atsushi Okato
    • Ichiro Fukumoto
    • Tomohiko Ichikawa
    • Naohiko Seki
  • View Affiliations

  • Published online on: March 15, 2016     https://doi.org/10.3892/ijo.2016.3440
  • Pages: 1837-1846
  • Copyright: © Kurozumi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Our recent studies of microRNA (miRNA) expression signatures in human cancers revealed that microRNA-26a (miRNA-26a) and microRNA-26b (miRNA-26b) were significantly reduced in cancer tissues. To date, few reports have provided functional analyses of miR-26a or miR-26b in renal cell carcinoma (RCC). The aim of the present study was to investigate the functional significance of miR-26a and miR-26b in RCC and to identify novel miR-26a/b-mediated cancer pathways and target genes involved in RCC oncogenesis and metastasis. Downregulation of miR-26a or miR-26b was confirmed in RCC clinical specimens. Restoration of miR-26a or miR-26b in RCC cell lines (786-O and A498) revealed that these miRNAs significantly inhibited cancer cell migration and invasion. Our in silico analysis and luciferase reporter assays showed that lysyl oxidase-like 2 (LOXL2) and procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2) were directly regulated by these miRNAs. Moreover, downregulating the PLOD2 gene significantly inhibited cell migration and invasion in RCC cells. Thus, our data showed that two genes promoting metastasis, LOXL2 and PLOD2, were epigenetically regulated by tumor-suppressive microRNAs, miR-26a and miR-26b, providing important insights into the molecular mechanisms of RCC metastasis.

Introduction

Renal cell carcinoma (RCC) is a disease in which cells in the kidney tubules undergo oncogenic transformation. RCC has multiple subtypes and may occur in hereditary (2–3% of RCC) or sporadic forms (1,2). RCC is the third most common urological cancer and accounts for 3% of all adult neoplasias. Clear cell RCC (ccRCC) is the most common subtype of sporadic RCC (~80%) (1). The standard curative treatment for localized diseases remains surgical excision with total nephrectomy. In contrast, at diagnosis, ~30% of RCCs have already metastasized. The 5-year survival rate in patients with advanced stage RCC is poor (5–10%) due to recurrence or distant metastasis (3,4). Recent molecularly targeted therapy has improved the survival rate of patients with advanced RCC (5,6). However, almost all patients eventually relapse or show distant metastasis due to acquired resistance to molecularly targeted therapy. Identifying molecular pathways responsible for RCC metastasis could provide novel approaches for the development of therapies that block the RCC metastatic pathways.

The discovery of microRNA (miRNA) in the human genome provided new directions in cancer research. The miRNAs are endogenous small RNA molecules (19–22 bases long) that regulate protein coding gene expression by repressing translation or cleaving RNA transcripts in a sequence-specific manner (7,8). Numerous studies have shown that miRNAs are aberrantly expressed in many human cancers, and they have significant roles in the initiation, development and metastasis of those cancers (911). Moreover, normal regulatory mechanisms can be disrupted by the aberrant expression of tumor-suppressive or oncogenic miRNAs in cancer cells. Therefore, identifying aberrantly expressed miRNAs is an important first step toward elucidating miRNA-mediated oncogenic pathways.

Using miRNA expression signatures, we have identified molecular pathways in RCC that are mediated by aberrantly expressed miRNAs (1215). For example, downregulation of tumor-suppressive miR-218 promoted cancer cell migration and invasion through dysregulation of the focal adhesion pathway. In this regard, caveolin-2 has an oncogenic function in RCC cells (13). The epithelial-mesenchymal transition (EMT)-related miR-200 family (miR-200a/b/c, miR-141 and miR-429) is significantly downregulated in RCC where they act as tumor suppressors that target the focal adhesion and ErbB signaling pathways (14). The miR-143/145 cluster was frequently reduced in RCC tissues; restoration of these miRNAs significantly inhibited RCC cell proliferation and invasion through targeting of hexokinase-2 (16). More recently, expression of the miR-23b/27b cluster was significantly decreased in ccRCC tissues and associated with pathological grade and stage of the disease (17).

Our miRNA expression signatures of human cancers revealed that miR-26a and miR-26b were frequently downregulated in various types of cancer tissues (10,18,19), suggesting that these miRNAs act as tumor suppressors targeting several oncogenic pathways. Database searches revealed that there were few reports of functional analyses of miR-26a or miR-26b in RCC. The aim of the present study was to investigate the functional significance of miR-26a and miR-26b and to identify molecular targets and pathways contributing to metastasis in RCC cells by miR-26a or miR-26b regulation. We expect that this analysis will provide important insights into the potential molecular mechanisms of RCC oncogenesis and metastasis and will facilitate the development of therapeutic strategies for the treatment of the disease.

Materials and methods

RCC clinical specimens and cell culture

A total of 15 pairs of ccRCC specimens and corresponding non-cancerous specimens were collected from patients who had undergone radical nephrectomy at Chiba University Hospital (Chiba, Japan) from 2012 to 2015. These specimens were staged according to the General Rule for Clinical and Pathological Studies on Renal Cell Carcinoma based on the American Joint Committee on Cancer (AJCC)-UICC TNM classification. The clinicopathological characteristics of the patients are summarized in Table I. Before tissue collection, written informed consent of tissue donation for research purposes was obtained from all the patients.

Table I

Characteristics of ccRCC clinical specimens.

Table I

Characteristics of ccRCC clinical specimens.

No.PathologyGradepTINFvlyeg or igfcimrcrps
1Clear cellG2T1aa00eg10000
2Clear cellG1>G2T1aa00eg10000
3Clear cellG3>G2T1ba00eg10000
4Clear cellG2>G3>G1T1aa00eg10000
5Clear cellG2>G3T1ba00eg11000
6Clear cellG2>G3T3aa10eg10000
7Clear cellG2>G3>G1T3ab10ig01100
8Clear cellG2>G3>G1T3ab10ig10000
9Clear cellG3T3ab10ig00000
10Clear cellG1>G2T1ba00eg10000
11Clear cellG2>G1>G3T3ab10ig00000
12Clear cellG2T1aa00eg00000
13Clear cell G2>G1>>G3T1bb00eg10000
14Clear cellG2>G1T1ab00eg10000
15Clear cellG2T1ba00eg00000

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

We used two human RCC cell lines (786-O and A498) obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA) as previously described (1214).

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

The procedure for PCR quantification was previously described. TaqMan probes and primers for LOXL2 (P/N: Hs00158757_ml; Applied Biosystems, Foster City, CA, USA), PLOD2 (P/N: Hs01118190_ml; Applied Biosystems) and GUSB (the internal control; P/N: Hs00939627_ml; Applied Biosystems) were assay-on-demand gene expression products. The expression levels of miR-26a (assay ID: 000405; Applied Biosystems) and miR-26b (assay ID: 000407; Applied Biosystems) were analyzed by TaqMan quantitative real-time RT-PCR (TaqMan MicroRNA assay; Applied Biosystems) and normalized to the expression of RNU48 as previously described (12,20,21).

Transfection with mature miRNAs and siRNAs

The following mature miRNAs were used: Ambion Pre-miR miRNA precursor for hsa-miR-26a-5p (product ID: PM10249; Applied Biosystems) and for hsa-miR-26b-5p (product ID: PM12899; Applied Biosystems). The following siRNAs were used: Stealth Select RNAi si-RNA, si-PLOD2 (cat nos. HSS108124 and HSS182371; Invitrogen) and negative control miRNA/siRNA (P/N: AM17111; Applied Biosystems). RNAs were incubated with Opti-MEM (Invitrogen) and Lipofectamine RNAiMax transfection reagent (Invitrogen) as previously described (12,20,21).

Cell proliferation, migration and invasion assays

786-O and A498 cells were transfected with 10 nM miRNAs or si-RNAs by reverse transfection. Cell proliferation, migration and invasion assays were performed as previously described (12,20,21).

Western blotting

Cells were harvested 72 h after transfection, and lysates were prepared. Protein lysates (20 μg) were separated on Mini-PROTEAN TGX gels (Bio-Rad Laboratories, Hercules, CA, USA) and transferred to PVDF membranes. Immunoblotting was performed with rabbit anti-LOXL2 antibodies (1:1000; ab96233; Abcam, Cambridge, UK) and rabbit anti-PLOD2 antibodies (1:300; 21214-1-AP; Proteintech Group, Inc., Chicago, IL, USA). Anti-GAPDH antibodies (1:1,000; ab8245; Abcam) were used as an internal loading control. The membranes were washed and incubated with anti-rabbit IgG horseradish peroxidase (HRP)-linked antibodies (#7074; Cell Signaling Technology). Complexes were visualized with Clarity Western Substrate (Bio-Rad Laboratories).

Screening of miR-26a and miR-26b target genes using in silico analysis and gene expression data

To identify miR-26a/b target genes, we used in silico analysis and genome-wide gene expression analysis. First, we screened genes using TargetScan release 6.2 (http://www.targetscan.org/). Next, to identify upregulated genes in ccRCC clinical specimens, we analyzed publicly available gene expression profiles in the GEO database (accession nos. GSE22541 and GSE36895). Our strategies for miRNA target screening were previously described (12,20,21).

Plasmid construction and dual-luciferase reporter assay

Partial wild-type sequences of the LOXL2 and PLOD2 3′-untranslated region (UTR) or those with deleted miR-26a/b binding sites were inserted between the XhoI-PmeI restriction sites in the 3′-UTR of the hRluc gene in the psiCHECK-2 vector (C8021; Promega, Madison, WI, USA). The procedure for the dual-luciferase reporter assay was previously described (12,20,21).

Statistical analysis

The relationships between the two groups and the numerical values obtained by real-time RT-PCR were analyzed using the Mann-Whitney U-test. The relationships among the three variables and numerical values were analyzed using the Bonferroni-adjusted Mann-Whitney U test. Spearman's rank test was used to evaluate the correlations between the expression of (miR-26a and LOXL2), (miR-26a and PLOD2), (miR-26b and LOXL2) and (miR-26b and PLOD2). All analyses were performed using Expert StatView (version 5; SAS Institute, Inc., Cary, NC, USA).

Results

Expression levels of miR-26a and miR-26b in ccRCC clinical specimens and cell lines

The expression levels of miR-26a and miR-26b were significantly lower in ccRCC specimens than in corresponding non-cancerous specimens (P=0.0171 and P=0.0136, respectively; Fig. 1A and B). In 786-O and A498 cells, the expression levels of miR-26a or miR-26b were lower than in non-cancerous specimens.

Effects of miR-26a and miR-26b restoration on cell proliferation, migration and invasion activities in ccRCC cells

To investigate the functional effects of miR-26a or miR-26b, we performed gain-of-function studies using mature miRNA transfection of 786-O and A498 cells.

The XTT assays demonstrated that cell proliferation was not inhibited in miR-26a or miR-26b transfectants in comparison with the mock or miR-control transfectants (Fig. 1C).

In contrast, the migration assays demonstrated that cell migration activity was significantly inhibited in miR-26a or miR-26b transfectant cells in comparison with the mock or miR-control transfectants (Fig. 1D). The Matrigel invasion assays demonstrated that cell invasion activity was significantly inhibited in miR-26a or miR-26b transfectant cells in comparison with the mock or miR-control transfectants (Fig. 1E).

Identification of candidate target genes of miR-26a and miR-26b in ccRCC cells

To identify target genes of miR-26a and miR-26b (the seed sequences of the two miRNAs are identical), we used in silico analysis and genome-wide gene expression data. First, we searched the TargetScan database (release 6.2: http://www.targetscan.org/) and identified 3,419 genes that had putative target sites for miR-26a and miR-26b in their 3′-UTRs. Next, we pared down the list of putative candidate genes based on upregulated genes determined by the gene expression data set of RCC clinical specimens in the GEO (Gene Expression Omnibus) database (accession numbers: GSE36895, GSE22541). The flow chart outlining our strategy for identification of candidate target genes of miR-26a and miR-26b is shown in Fig. 2.

From this selection, 39 candidate genes were identified as targets of miR-26a and miR-26b (Table II). Among these candidate genes, we focused on LOXL2 and PLOD2 genes because these genes have two conserved target sites for miR-26a and miR-26b in their 3′-UTRs, and function as collagen cross-linking enzymes associated with extracellular matrix (ECM) stiffness. Recent studies showed that aberrantly expressed ECM contributes to cancer cell metastasis (22,23). Therefore, these two genes were chosen for further analysis.

Table II

Putative candidate target genes regulated by miR-26a and miR-26b in RCC cells.

Table II

Putative candidate target genes regulated by miR-26a and miR-26b in RCC cells.

Entrez gene IDSymbolGene nameLocationNo. of conserved target sitesNo. of poorly conserved target sitesGEO (GSE36895, GSE22541 average fold-change
5352PLOD2Procollagen-lysine, 2-oxoglutarate 5-dioxygenase 23q24202.2220507
4017LOXL2Lysyl oxidase-like 28p21.3202.7719142
2146EZH2Enhancer of zeste homolog 2 (Drosophila)7q35-q36102.0032272
3625INHBBInhibin, β B2cen-q13103.7558112
3678ITGA5Integrin, α 5 (fibronectin receptor, α polypeptide)12q11-q13102.8391342
23023TMCC1Transmembrane and coiled-coil domain family 13q22.1112.226072
1404HAPLN1Hyaluronan and proteoglycan link protein 15q14.3112.7813237
7903ST8SIA4ST8 α-N-acetyl-neuraminide
α-2,8-sialyltransferase 4
5q21103.1741676
1846DUSP4Dual specificity phosphatase 48p12-p11102.1518986
6890TAP1Transporter 1, ATP-binding cassette, sub-family B (MDR/TAP)6p21.3012.0403051
7272TTKTTK protein kinase6q14.1012.3837836
170384FUT11Fucosyltransferase 11 (α (1,3) fucosyltransferase)10q22.2012.0443428
22974TPX2TPX2, microtubule-associated, homolog (Xenopus laevis)20q11.2012.662108
2210FCGR1BFc fragment of IgG, high affinity Ib, receptor (CD64)1p11.2012.294377
4747NEFLNeurofilament, light polypeptide8p21012.1319628
5836PYGLPhosphorylase, glycogen, liver14q21-q22012.0643747
1234CCR5Chemokine (C-C motif) receptor 53p21.31013.3846455
55165CEP55Centrosomal protein 55 kDa10q23.33012.0711598
10288LILRB2Leukocyte immunoglobulin-like receptor, subfamily B (with TM and ITIM domains), member 219q13.4012.454539
1356CPCeruloplasmin (ferroxidase)3q23-q25013.9467278
3910LAMA4Laminin, α 46q21012.2182174
163404LPPR5Lipid phosphate phosphatase-related protein type 51p21.3012.450066
5027P2RX7Purinergic receptor P2X, ligand-gated ion channel, 712q24033.0084689
330BIRC3Baculoviral IAP repeat containing 311q22012.2927191
6507SLC1A3Solute carrier family 1 (glial high affinity glutamate transporter), member 35p13012.1052346
2335FN1Fibronectin 12q34012.4469628
8701DNAH11Dynein, axonemal, heavy chain 117p21012.2785249
79850FAM57AFamily with sequence similarity 57, member A17p13.3012.2900116
1462VCANVersican5q14.3012.524361
128346 C1orf162Chromosome 1 open reading frame 1621p13.2012.2255776
4015LOXLysyl oxidase5q23.2013.3194032
115761ARL11ADP-ribosylation factor-like 1113q14.2012.4013827
286336FAM78AFamily with sequence similarity 78, member A9q34012.1942985
6664SOX11SRY (sex determining region Y)-box 112p25012.577679
9770RASSF2Ras association (RalGDS/AF-6) domain family member 220p13012.619857
57823SLAMF7SLAM family member 71q23.1-q24.1012.063896
58475MS4A7Membrane-spanning 4-domains, subfamily A, member 711q12012.0315962
79742CXorf36Chromosome X open reading frame 36Xp11.3012.3148956
146857SLFN13Schlafen family member 1317q12012.6972997
Direct regulation of LOXL2 and PLOD2 by miR-26a and miR-26b in ccRCC cells

We first performed qRT-PCR and Western blotting to investigate whether expression of the LOXL2 gene and protein were reduced by restoration of miR-26a or miR-26b in 786-O and A498 cells. We found that the mRNA and protein expression levels of LOXL2/LOXL2 were significantly repressed in miR-26a or miR-26b transfectant cells in comparison with mock or miR-control transfectants (Fig. 3A and B).

Next, to investigate whether LOXL2 mRNA had target sites for miR-26a or miR-26b, we performed luciferase reporter assays in 786-O cells. We used vectors encoding either the partial wild-type sequence of the 3′-UTR of LOXL2, including the predicted miR-26a/b target sites, or deletion vectors lacking the miR-26a/b target sites. We found that the luminescence intensities were significantly reduced by transfection with miR-26a or miR-26b and vectors carrying the wild-type 3′-UTR of LOXL2, whereas transfection with deletion vectors blocked the decrease in luminescence. These data suggested that miR-26a or miR-26b bound directly to specific sites in the 3′-UTR of LOXL2 (Fig. 3C and D).

We also found that the mRNA and protein expression levels of PLOD2/PLOD2 were significantly repressed in miR-26a or miR-26b transfectant cells in comparison with mock or miR-control transfectants (Fig. 4A and B). We also observed that the luminescence intensities were significantly reduced by transfection with miR-26a or miR-26b and vectors carrying the wild-type 3′-UTR of PLOD2, whereas transfection with deletion vectors blocked the decrease in luminescence. These data suggested that miR-26a or miR-26b bound directly to specific sites in the 3′-UTR of PLOD2 (Fig. 4C and D).

Silencing PLOD2 affected cell proliferation, migration and invasion activities in ccRCC cells

We recently presented a loss-of-function study of LOXL2 in RCC cells (786-O and A498) by using two siRNAs (786-O and A498) (12). Those data showed that the silencing of LOXL2 significantly suppressed cancer cell migration and invasion activities in RCC cells.

To investigate the functional role of PLOD2 in ccRCC cells, we performed a loss-of-function study using si-PLOD2 transfected cells. First, we evaluated the knockdown efficiency of si-PLOD2 transfection in 786-O and A498 cells. qRT-PCR and western blotting indicated that si-PLOD2 transfection effectively downregulated PLOD2 expression in both cell lines (786-O, P<0.0001; A498, P<0.0001; Fig. 5A and B).

The XTT assay demonstrated that cell proliferation was not inhibited significantly in si-PLOD2 transfectant cells in comparison with the mock or negative control transfectants (Fig. 5C).

In contrast, the migration assay demonstrated that cell migration activity was significantly inhibited in si-PLOD2 transfectants in comparison with the mock or negative control transfectants (Fig. 5D). The Matrigel invasion assay demonstrated that invasive activity was significantly inhibited in si-PLOD2 transfectants in comparison with the mock or negative control transfectants (Fig. 5E).

Expression of LOXL2 and PLOD2 in ccRCC clinical specimens

A total of 15 pairs of ccRCC specimens and corresponding non-cancerous specimens were used for expression studies of LOXL2 and PLOD2 using RT-PCR. We showed that LOXL2 and PLOD2 were significantly upregulated in cancer tissues compared with normal tissues (P=0.0001 and P<0.0001, respectively; Fig. 6A and B). Furthermore, Spearman's rank test showed a negative correlation between the expression of miR-26a/PLOD2 and miR-26b/PLOD2 (Fig. 6E and F).

Discussion

A growing body of evidence has shown that aberrantly expressed miRNAs can disrupt tightly regulated RNA networks in cancer cells and promote human oncogenesis and metastasis (7,9,2426). Recently, our studies identified a variety of novel RCC molecular pathways regulated by tumor-suppressive miRNAs (1215). In the present study, we focused on miR-26a and miR-26b because the expression levels of these miRNAs were reduced in the miRNA signatures of various types of cancers (10,18,19,27). Moreover, the functional roles of these miRNAs in RCC cells are not clear. Our present data showed that miR-26a and miR-26b act as tumor suppressors that modulate cancer cell migration and invasion in RCC cells. Our previous studies of oral cancer and prostate cancer demonstrated the tumor-suppressive roles of these miRNAs (19,20), and those findings support the present results obtained with RCC cells. Downregulation and tumor-suppressive roles of miR-26a or miR-26b have been reported in several types of cancer, such as bladder, breast, hepatocellular carcinoma and oral cancer (19,2830).

In the human genome, the miR-26 family consists of three subtypes of miRNAs: miR-26a-1, miR-26a-2 and miR-26b. The mature sequences of miR-26a-1 and miR-26a-2 are identical, whereas the two nucleotides differ from that of miR-26b (miRBase release 21; http://www.mirbase.org/). The molecular mechanisms responsible for silencing the expression of the miR-26 family are still unclear. A recent study indicated that MYC oncogene directly bound to the promoter regions of miR-26a-1, miR-26a-2 and miR-26b and negatively regulated expression of these miRNAs in prostate cancer cells (31). Overexpression of MYC was observed in RCC clinical specimens (15,32), suggesting MYC might be a mediator for expression control of tumor-suppressive miRNAs in cancer cells.

A single miRNA may regulate multiple protein-coding genes; indeed, bioinformatics studies have shown that miRNAs regulate >30–60% of the protein-coding genes in the human genome (7,33). Reduced expression of tumor-suppressive miRNAs may cause overexpression of oncogenic genes in cancer cells. To better understand RCC oncogenesis and metastasis, we identified miR-26a and miR-26b target genes using in silico analysis. Recent miRNA studies in our laboratory have utilized this strategy to identify novel molecular targets and pathways regulated by tumor-suppressive miRNAs in several cancers, including RCC (12,20).

A total of 39 putative target genes of miR-26a and miR-26b were identified in the present study. Among these genes, we focused on LOXL2 and PLOD2 because they function as collagen cross-linking enzymes. Numerous studies have shown that aberrant expression of collagen cross-linking enzymes promotes extracellular matrix (ECM) stiffening, resulting in enhanced cancer cell migration and invasion (22,3439). Overexpression of ECM components has been observed in several cancers (21,23,40). Recently, a number of studies indicated that several miRNAs regulated ECM component genes, and aberrantly expressed miRNAs have contributed to cancer cell progression by dysregulation of cell adhesion, polarity and ECM remodeling (21,23). Our past studies found that the tumor-suppressive miR-29-family (miR-29a, miR-29b and miR-29c) and miR-218 directly regulated laminins (LAMC2 and LAMB3) and integrins (ITGA6 and ITGB3), such that restoration of these miRNAs inhibited cancer cell migration and invasion (21,41,42).

Once collagen is secreted, collagen cross-linking occurs on lysine and hydroxylysine residues by the lysyl oxidase (LOX) family of enzymes (22,43). More recently, we showed that the miR-29s-family directly targeted LOXL2 in RCC and lung cancers (12). Overexpression of LOXL2 was observed in RCC clinical specimens and silencing of LOXL2 inhibited cancer cell migration and invasion in ccRCC cell lines (12). Other research groups found that increased expression of LOXL2 is correlated with disease progression, including RCC (34,44). The function of the LOX-family is covalent crosslinking of collagen and/or elastin in the ECM (35,36). Aberrant expression of LOX-family proteins has been reported in several diseases, including cancers (3439). Interestingly, LOXL2 is a direct transcriptional target of HIF-1. Moreover, nuclear LOXL2 interacts with transcription factor SNAIL1 and represses E-cadherin as well as induces EMT (45,46). In this study, we demonstrated direct regulation of LOXL2 by miR-26a and miR-26b in RCC cells as observed with the miR-29s-family. These findings showed that tumor-suppressive miR-26a/b-LOXL2 is the pivotal pathway contributing to cancer cell migration and invasion in RCC.

In this study, we also focused on the PLOD2 (procollagen-lysine 2-oxyglutarate-dioxygenase) gene as a target of miR-26a and miR-26b and demonstrated the direct regulation of these miRNAs by luciferase reporter assays. PLOD2 encodes an enzyme that mediates collagen lysine hydroxylation. Collagen cross-linking that are derived from hydroxylated lysine residues have increased stability compared with non-hydroxylated lysine residues (22,47). Overexpression of PLOD2 in ccRCC clinical specimens and promoting migration and invasion in cancer cells were observed in the present study. In breast cancer, Kaplan-Meier curves of disease-specific survival stratified by PLOD2 expression revealed that high PLOD2 expression was significantly associated with decreased disease-specific survival (48). Moreover, PLOD2 expression promoted tumor stiffness and was required for metastasis to lymph nodes and lungs (22,48).

In conclusion, miR-26a and miR-26b were significantly downregulated in ccRCC clinical specimens and appeared to function as tumor suppressors through regulation of collagen cross-linking enzymes, LOXL2 and PLOD2, both of which function as oncogenes in this disease. The identification of novel molecular targets and pathways regulated by the tumor-suppressive miR-26a and miR-26b may lead to a better understanding of ccRCC and the development of new therapeutic strategies to treat this disease.

Acknowledgements

The present study was supported by the Japanese Society for the Promotion of Science (KAKENHI), grant numbers (C) 15K10801, (C) 15K20070, (C) 15K20071, and (B) 25293333.

References

1 

Rini BI, Campbell SC and Escudier B: Renal cell carcinoma. Lancet. 373:1119–1132. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Randall JM, Millard F and Kurzrock R: Molecular aberrations, targeted therapy, and renal cell carcinoma: Current state-of-the-art. Cancer Metastasis Rev. 33:1109–1124. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Gupta K, Miller JD, Li JZ, Russell MW and Charbonneau C: Epidemiologic and socioeconomic burden of metastatic renal cell carcinoma (mRCC): A literature review. Cancer Treat Rev. 34:193–205. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Motzer RJ and Russo P: Systemic therapy for renal cell carcinoma. J Urol. 163:408–417. 2000. View Article : Google Scholar : PubMed/NCBI

5 

Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Oudard S, Negrier S, Szczylik C, Pili R, Bjarnason GA, et al: Overall survival and updated results for sunitinib compared with interferon alfa in patients with meta-static renal cell carcinoma. J Clin Oncol. 27:3584–3590. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, et al: Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 356:115–124. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Filipowicz W, Bhattacharyya SN and Sonenberg N: Mechanisms of post-transcriptional regulation by microRNAs: Are the answers in sight? Nat Rev Genet. 9:102–114. 2008. View Article : Google Scholar : PubMed/NCBI

8 

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

9 

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

10 

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

11 

Esquela-Kerscher A and Slack FJ: Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 6:259–269. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Nishikawa R, Chiyomaru T, Enokida H, Inoguchi S, Ishihara T, Matsushita R, Goto Y, Fukumoto I, Nakagawa M and Seki N: Tumour-suppressive microRNA-29s directly regulate LOXL2 expression and inhibit cancer cell migration and invasion in renal cell carcinoma. FEBS Lett. 589:2136–2145. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Yamasaki T, Seki N, Yoshino H, Itesako T, Hidaka H, Yamada Y, Tatarano S, Yonezawa T, Kinoshita T, Nakagawa M, et al: MicroRNA-218 inhibits cell migration and invasion in renal cell carcinoma through targeting caveolin-2 involved in focal adhesion pathway. J Urol. 190:1059–1068. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Yoshino H, Enokida H, Itesako T, Tatarano S, Kinoshita T, Fuse M, Kojima S, Nakagawa M and Seki N: Epithelial-mesenchymal transition-related microRNA-200s regulate molecular targets and pathways in renal cell carcinoma. J Hum Genet. 58:508–516. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Yamada Y, Hidaka H, Seki N, Yoshino H, Yamasaki T, Itesako T, Nakagawa M and Enokida H: Tumor-suppressive microRNA-135a inhibits cancer cell proliferation by targeting the c-MYC oncogene in renal cell carcinoma. Cancer Sci. 104:304–312. 2013. View Article : Google Scholar

16 

Yoshino H, Enokida H, Itesako T, Kojima S, Kinoshita T, Tatarano S, Chiyomaru T, Nakagawa M and Seki N: Tumor-suppressive microRNA-143/145 cluster targets hexokinase-2 in renal cell carcinoma. Cancer Sci. 104:1567–1574. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Ishihara T, Seki N, Inoguchi S, Yoshino H, Tatarano S, Yamada Y, Itesako T, Goto Y, Nishikawa R, Nakagawa M, et al: Expression of the tumor suppressive miRNA-23b/27b cluster is a good prognostic marker in clear cell renal cell carcinoma. J Urol. 192:1822–1830. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Kikkawa N, Hanazawa T, Fujimura L, Nohata N, Suzuki H, Chazono H, Sakurai D, Horiguchi S, Okamoto Y and Seki N: miR-489 is a tumour-suppressive miRNA target PTPN11 in hypopharyngeal squamous cell carcinoma (HSCC). Br J Cancer. 103:877–884. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Fukumoto I, Hanazawa T, Kinoshita T, Kikkawa N, Koshizuka K, Goto Y, Nishikawa R, Chiyomaru T, Enokida H, Nakagawa M, et al: MicroRNA expression signature of oral squamous cell carcinoma: Functional role of microRNA-26a/b in the modulation of novel cancer pathways. Br J Cancer. 112:891–900. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Kato M, Goto Y, Matsushita R, Kurozumi A, Fukumoto I, Nishikawa R, Sakamoto S, Enokida H, Nakagawa M, Ichikawa T, et al: MicroRNA-26a/b directly regulate La-related protein 1 and inhibit cancer cell invasion in prostate cancer. Int J Oncol. 47:710–718. 2015.PubMed/NCBI

21 

Kinoshita T, Hanazawa T, Nohata N, Kikkawa N, Enokida H, Yoshino H, Yamasaki T, Hidaka H, Nakagawa M, Okamoto Y, et al: Tumor suppressive microRNA-218 inhibits cancer cell migration and invasion through targeting laminin-332 in head and neck squamous cell carcinoma. Oncotarget. 3:1386–1400. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Gilkes DM, Semenza GL and Wirtz D: Hypoxia and the extracellular matrix: Drivers of tumour metastasis. Nat Rev Cancer. 14:430–439. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Kurozumi A, Goto Y, Matsushita R, Fukumoto I, Kato M, Nishikawa R, Sakamoto S, Enokida H, Nakagawa M, Ichikawa T, et al: Tumor-suppressive microRNA-223 inhibits cancer cell migration and invasion by targeting ITGA3/ITGB1 signaling in prostate cancer. Cancer Sci. 107:84–94. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Wiemer EA: The role of microRNAs in cancer: No small matter. Eur J Cancer. 43:1529–1544. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Singh SK, Pal Bhadra M, Girschick HJ and Bhadra U: MicroRNAs - micro in size but macro in function. FEBS J. 275:4929–4944. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Iorio MV and Croce CM: MicroRNAs in cancer: Small molecules with a huge impact. J Clin Oncol. 27:5848–5856. 2009. View Article : Google Scholar : PubMed/NCBI

27 

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

28 

Lin Y, Chen H, Hu Z, Mao Y, Xu X, Zhu Y, Xu X, Wu J, Li S, Mao Q, et al: miR-26a inhibits proliferation and motility in bladder cancer by targeting HMGA1. FEBS Lett. 587:2467–2473. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Zhang B, Liu XX, He JR, Zhou CX, Guo M, He M, Li MF, Chen GQ and Zhao Q: Pathologically decreased miR-26a antagonizes apoptosis and facilitates carcinogenesis by targeting MTDH and EZH2 in breast cancer. Carcinogenesis. 32:2–9. 2011. View Article : Google Scholar

30 

Zhu Y, Lu Y, Zhang Q, Liu JJ, Li TJ, Yang JR, Zeng C and Zhuang SM: MicroRNA-26a/b and their host genes cooperate to inhibit the G1/S transition by activating the pRb protein. Nucleic Acids Res. 40:4615–4625. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Koh CM, Iwata T, Zheng Q, Bethel C, Yegnasubramanian S and De Marzo AM: Myc enforces overexpression of EZH2 in early prostatic neoplasia via transcriptional and post-transcriptional mechanisms. Oncotarget. 2:669–683. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Tang SW, Chang WH, Su YC, Chen YC, Lai YH, Wu PT, Hsu CI, Lin WC, Lai MK and Lin JY: MYC pathway is activated in clear cell renal cell carcinoma and essential for proliferation of clear cell renal cell carcinoma cells. Cancer Lett. 273:35–43. 2009. View Article : Google Scholar

33 

Friedman RC, Farh KK, Burge CB and Bartel DP: Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 19:92–105. 2009. View Article : Google Scholar :

34 

Barker HE, Cox TR and Erler JT: The rationale for targeting the LOX family in cancer. Nat Rev Cancer. 12:540–552. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Kagan HM and Li W: Lysyl oxidase: Properties, specificity, and biological roles inside and outside of the cell. J Cell Biochem. 88:660–672. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Vadasz Z, Kessler O, Akiri G, Gengrinovitch S, Kagan HM, Baruch Y, Izhak OB and Neufeld G: Abnormal deposition of collagen around hepatocytes in Wilson's disease is associated with hepatocyte specific expression of lysyl oxidase and lysyl oxidase like protein-2. J Hepatol. 43:499–507. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Kim YM, Kim EC and Kim Y: The human lysyl oxidase-like 2 protein functions as an amine oxidase toward collagen and elastin. Mol Biol Rep. 38:145–149. 2011. View Article : Google Scholar

38 

Fong SF, Dietzsch E, Fong KS, Hollosi P, Asuncion L, He Q, Parker MI and Csiszar K: Lysyl oxidase-like 2 expression is increased in colon and esophageal tumors and associated with less differentiated colon tumors. Genes Chromosomes Cancer. 46:644–655. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Peinado H, Moreno-Bueno G, Hardisson D, Pérez-Gómez E, Santos V, Mendiola M, de Diego JI, Nistal M, Quintanilla M, Portillo F, et al: Lysyl oxidase-like 2 as a new poor prognosis marker of squamous cell carcinomas. Cancer Res. 68:4541–4550. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Liu LX, Jiang HC, Liu ZH, Zhou J, Zhang WH, Zhu AL, Wang XQ and Wu M: Integrin gene expression profiles of human hepatocellular carcinoma. World J Gastroenterol. 8:631–637. 2002. View Article : Google Scholar : PubMed/NCBI

41 

Kinoshita T, Nohata N, Hanazawa T, Kikkawa N, Yamamoto N, Yoshino H, Itesako T, Enokida H, Nakagawa M, Okamoto Y, et al: Tumour-suppressive microRNA-29s inhibit cancer cell migration and invasion by targeting laminin-integrin signalling in head and neck squamous cell carcinoma. Br J Cancer. 109:2636–2645. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Yamamoto N, Kinoshita T, Nohata N, Itesako T, Yoshino H, Enokida H, Nakagawa M, Shozu M and Seki N: Tumor suppressive microRNA-218 inhibits cancer cell migration and invasion by targeting focal adhesion pathways in cervical squamous cell carcinoma. Int J Oncol. 42:1523–1532. 2013.PubMed/NCBI

43 

Gordon MK and Hahn RA: Collagens. Cell Tissue Res. 339:247–257. 2010. View Article : Google Scholar :

44 

Hase H, Jingushi K, Ueda Y, Kitae K, Egawa H, Ohshio I, Kawakami R, Kashiwagi Y, Tsukada Y, Kobayashi T, et al: LOXL2 status correlates with tumor stage and regulates integrin levels to promote tumor progression in ccRCC. Mol Cancer Res. 12:1807–1817. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Schietke R, Warnecke C, Wacker I, Schödel J, Mole DR, Campean V, Amann K, Goppelt-Struebe M, Behrens J, Eckardt KU, et al: The lysyl oxidases LOX and LOXL2 are necessary and sufficient to repress E-cadherin in hypoxia: Insights into cellular transformation processes mediated by HIF-1. J Biol Chem. 285:6658–6669. 2010. View Article : Google Scholar :

46 

Moon HJ, Finney J, Xu L, Moore D, Welch DR and Mure M: MCF-7 cells expressing nuclear associated lysyl oxidase-like 2 (LOXL2) exhibit an epithelial-to-mesenchymal transition (EMT) phenotype and are highly invasive in vitro. J Biol Chem. 288:30000–30008. 2013. View Article : Google Scholar : PubMed/NCBI

47 

van der Slot AJ, Zuurmond AM, van den Bogaerdt AJ, Ulrich MM, Middelkoop E, Boers W, Karel Ronday H, DeGroot J, Huizinga TW and Bank RA: Increased formation of pyridinoline cross-links due to higher telopeptide lysyl hydroxylase levels is a general fibrotic phenomenon. Matrix Biol. 23:251–257. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Gilkes DM, Bajpai S, Wong CC, Chaturvedi P, Hubbi ME, Wirtz D and Semenza GL: Procollagen lysyl hydroxylase 2 is essential for hypoxia-induced breast cancer metastasis. Mol Cancer Res. 11:456–466. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2016
Volume 48 Issue 5

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kurozumi A, Kato M, Goto Y, Matsushita R, Nishikawa R, Okato A, Fukumoto I, Ichikawa T and Seki N: Regulation of the collagen cross-linking enzymes LOXL2 and PLOD2 by tumor-suppressive microRNA-26a/b in renal cell carcinoma. Int J Oncol 48: 1837-1846, 2016
APA
Kurozumi, A., Kato, M., Goto, Y., Matsushita, R., Nishikawa, R., Okato, A. ... Seki, N. (2016). Regulation of the collagen cross-linking enzymes LOXL2 and PLOD2 by tumor-suppressive microRNA-26a/b in renal cell carcinoma. International Journal of Oncology, 48, 1837-1846. https://doi.org/10.3892/ijo.2016.3440
MLA
Kurozumi, A., Kato, M., Goto, Y., Matsushita, R., Nishikawa, R., Okato, A., Fukumoto, I., Ichikawa, T., Seki, N."Regulation of the collagen cross-linking enzymes LOXL2 and PLOD2 by tumor-suppressive microRNA-26a/b in renal cell carcinoma". International Journal of Oncology 48.5 (2016): 1837-1846.
Chicago
Kurozumi, A., Kato, M., Goto, Y., Matsushita, R., Nishikawa, R., Okato, A., Fukumoto, I., Ichikawa, T., Seki, N."Regulation of the collagen cross-linking enzymes LOXL2 and PLOD2 by tumor-suppressive microRNA-26a/b in renal cell carcinoma". International Journal of Oncology 48, no. 5 (2016): 1837-1846. https://doi.org/10.3892/ijo.2016.3440