microRNA‑145 inhibits cell proliferation, migration and invasion by targeting matrix metallopeptidase-11 in renal cell carcinoma

  • Authors:
    • Deyao Wu
    • Min Li
    • Linmao Wang
    • Yunfeng Zhou
    • Jian Zhou
    • Huixing Pan
    • Ping Qu
  • View Affiliations

  • Published online on: April 15, 2014     https://doi.org/10.3892/mmr.2014.2149
  • Pages: 393-398
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

microRNA‑145 (miR‑145) has been reported to be frequently downregulated in various types of cancer, including renal, prostate, bladder, lung and colon cancer, as well as B‑cell malignancies. The present study examined the effects of miR‑145 on the cell proliferation, migration and invasion of renal cell carcinoma (RCC). Following transfection of miR‑145, an MTT, cell migration, cell invasion and luciferase assays, and western blot analysis were conducted in RCC cell lines. The present study demonstrated that miR‑145 inhibited cell proliferation, migration and invasion in 786‑O and A498 cells. The present study also demonstrated for the first time, to the best of our knowledge, that miR‑145 may directly target matrix metallopeptidase‑11 (MMP‑11) in RCC. miR‑145 was demonstrated to suppress cell proliferation, migration and invasion by targeting MMP‑11 in RCC cell lines. These results suggested that it may be investigated as a predictive marker for the early detection of tumor metastasis and for targeting therapeutic drugs to inhibit the invasion of RCC.

Introduction

Renal cell carcinoma (RCC) is a kidney cancer that originates from the proximal convoluted tubule. It represents the leading cause of mortality among urological malignancies (1,2). It is the 10th most common type of cancer in Europe (3). In the United States, there are ~65,000 novel cases and almost 14,000 mortalities from RCC annually (4). RCC includes several different histological subtypes that possess distinct biological behaviors and prognoses. The most common type of RCC is clear cell RCC (ccRCC), which originates in the lining of the proximal renal tubule, representing >75–80% of all cases of RCC (5). Approximately 30% of patients have metastatic disease when diagnosed with RCC and radical nephrectomy remains the main treatment for RCC patients due to resistance to radiation and chemotherapy (6). Although the overall survival rate is >60% over 5 years, ~30% of patients with a diagnosis of localized RCC develop metastatic recurrence (7,8). Patients with metastatic RCC face a poor prognosis and have limited therapeutic options. The median survival rate in a recent cohort was only 1.5 years with <10% of patients surviving to 5 years (9). The histological grade combined with clinical stage, which is considered to be the gold standard method for the prediction of patient prognosis, is not accurate when used alone (10). Thus, molecular markers and novel treatments are required in order to improve the prognosis for patients with RCC.

MicroRNAs (miRNAs) are a class of naturally occurring, endogenous small non-coding RNA, in the size range of 19–25 nt. miRNAs regulate gene expression at the post-transcriptional level by binding through partial sequence homology, to the 3′ untranslated region (3′UTR) of mammalian target mRNAs and causing translational inhibition and/or mRNA degradation (11). Since they were initially described almost 20 years ago, there has been a steady increase in their identification and the latest Release 20 of miRBase has 24,521 entries of miRNAs from various species, including 734 mature miRNAs from Gallus gallus (12). It has been predicted that as many as 30% of protein-encoding genes may be regulated by miRNAs and they may function as oncogenes and tumor suppressors (13). Upregulated miRNAs in cancer may function as oncogenes by negatively regulating tumor suppressors. By contrast, downregulated miRNAs may normally function as tumor suppressor genes and inhibit cancer by regulating oncogenes (14,15). It has demonstrated the important roles of miRNAs in regulating various cellular functions, including cell apoptosis, cell proliferation, neural development and stem cell differentiation (16,17). Recently, miRNAs have also been revealed to be important in tumorigenesis, cancer invasion and metastasis (18).

miR-145 has been reported to be frequently downregulated in various types of cancer, including renal, prostate, bladder, lung and colon cancer, as well as B-cell malignancies. However, the functions of miR-145 are yet to be investigated in RCC. The present study focused on the functions and direct target of miR-145 in RCC.

Materials and methods

Cells and culture conditions

The human ccRCC-derived cell lines 786-O and A498 were obtained from the Shanghai Institute of Cell Biology, Chinese Academy of Sciences (Shanghai, China). The cells were incubated in RPMI-1640 (HyClone Laboratories, Inc., Logan, UT, USA) or Dulbecco’s modified Eagle’s medium (DMEM; Gibco-BRL, Carlsbad, CA, USA) supplemented with 10% heat-inactivated fetal calf serum, 100 U/ml penicillin and 100 mg/l streptomycin at 37°C in a humidified atmosphere containing 5% CO2.

Transfection of miR-145 mimics, scrambled control (NC) and luciferase reporter plasmid

Mature miR-145 mimics, the NC and the luciferase reporter plasmid were designed and synthesized by GenePharma (Shanghai, China). The sequence of miR-145 mimics was 5′-GUCCAGUUUUCC CAGGAAUCCCU-3′. The sequence of NC mimics was 5′-UUCUCCGAACGUGUCACGUTT-3′. The insertion fragment was confirmed by DNA sequencing. Cell transfection and cotransfection were performed using Lipofectamine 2000 (Invitrogen Life Technologies) according to the manufacturer’s instructions.

Cell viability assay

The cell proliferation was determined by the MTT assay. The cells transfected with miR-145 mimics or the NC were seeded in 96-well plates at a density of 3,000 cells per well. Cell proliferation was documented every 24 h for five days according to the manufacturer’s instructions. Briefly, MTT solution was added into each well and incubated at 37°C for 4 h. The plates were spun (200 × g for 10 min), and the purple colored precipitates of formazan were dissolved in 200 μl dimethylsulfoxide. Absorbance (optical density, OD) was measured at 490 nm using an automatic multi-well spectrophotometer (Bio-Rad, Richmond, CA, USA). There were six replicate wells for every time point in each group. The suppression rate was calculated using the formula: Suppression rate = (1 - ODmiR-145 / ODmiR-NC) × 100%. All the experiments were performed in triplicate.

Cell migration and invasion assay

Cell motility was measured using 8 μm-pore polycarbonate membrane Boyden chambers inserted in a transwell apparatus (Costar, Cambridge, MA, USA). The transfected cells (miR-145 mimics and NC) growing in the log phase were treated with trypsin/EDTA solution, washed once with serum-containing RPMI-1640 medium, centrifuged (200 × g for 10 mins), and re-suspended as single-cell solutions. A total of 1×105 cells in 0.2 ml serum-free RPMI-1640 medium were seeded on a transwell apparatus (Costar). RPMI-1640 (600 μl) containing 20% fetal bovine serum was added to the lower chamber. An invasion assay was performed by the same procedure except that the filters of the transwell chambers were coated with 30 μg Matrigel (BD Biosciences, San Jose, CA, USA). After the cells were incubated for 12–24 h at 37°C in a 5% CO2 incubator, the cells on the top surface of the insert were removed by wiping with a cotton swab. The cells that migrated to the bottom surface of the insert were fixed in 100% methanol for 2 min, stained in 0.5% crystal violet for 2 min, rinsed in phosphate-buffered saline and then subjected to microscopic inspection (magnification, ×200; BX51WI-DPMC; Olympus, Tokyo, Japan). The values for invasion and migration were obtained by counting five fields per membrane and represent the average of three independent experiments.

Western blot analysis

The primary antibodies used in the present study, including anti-MMP-11 and anti-β-actin were products of Bioworld Technology (St. Louis Park, MN, USA). The total protein of cells was prepared using RIPA lysis buffer. The protein concentration in the resulting lysate was determined using the bicinchoninic acid protein assay (Beyotime Institute of Biotechnology, Shanghai, China). Equal quantities of protein were loaded onto SDS-PAGE and transferred onto polyvinylidene difluoride membranes. Following inhibition with 5% degreased milk in Tris-buffered saline with Tween-20 (TBST; containing 0.1% Tween-20), the membranes were incubated overnight with the appropriate primary antibody. The membranes were then washed and incubated with the corresponding horseradish peroxidase-conjugated secondary antibody (goat anti-rabbit) at 1:1,000 dilution in TBST. The blot was developed with enhanced chemiluminescence solution (Pierce Biotechnology, Inc., Rockford, IL, USA) and images were captured by a FluorChem imaging system (Alpha Innotech, San Leandro, CA, USA). The intensity of each spot was read and analyzed using AlphaEaseFC software (Alpha Innotech, San Leandro, CA, USA). β-actin was used as a loading control.

Luciferase assay

The cells were plated in a 12-well plate at ~90% confluence and transfected with 0.5 μg reporter plasmid, 40 nmol miR-145 mimics or their negative control by Lipofectamine 2000. Each sample was also cotransfected with 0.05 μg pRL-CMV plasmid expressing Renilla luciferase (Promega Corporation, Madison, WI, USA) as an internal control for transfection efficiency. The cells were harvested with passive lysis buffer, a component of the Dual-Luciferase Reporter Assay system (Tecan, Theale, UK), 48 h after transfection according to the manufacturer’s instructions. An appropriate volume of cell lysate was added to a well of the F96 MicroWell Plates, followed by 25 μl Luciferase Assay Reagent II (Tecan). Firefly luciferase activities and Renilla luciferase activities were measured using a luminometer (Tecan). Firefly luciferase activity was normalized to Renilla luciferase activity for each transfected well. Each assay was replicated three times.

Statistical analysis

Data are presented as the mean ± standard deviation and compared using Student’s t-test in Stata 10.0 (College Station, TX, USA). Double-tailed P<0.05 was considered to indicate a statistically significant difference.

Results

miR-145 suppresses cell proliferation in RCC cell lines

To measure the effect of miR-145 on cell proliferation, an MTT assay was used. As shown in Fig. 1, the upregulation of miR-145 significantly inhibited cell proliferation. MTT assays revealed that following 144 h of treatment, the suppression rate of miR-145 reached 35.81%±4.1% in 786-O cells and 41.15±3.5% in A498 cells. These results indicated that miR-145 may be important in 786-O and A498 cells.

miR-145 inhibits cell migration and invasion in RCC cell lines

To measure the effect of miR-145 on tumor cell migration and invasion, the transwell apparatus assay was used (Fig. 2). The transfected cells (miR-145 mimics and NC mimics) growing in the log phase were collected and cultured on transwell apparatus. Following 12 h incubation, cell migration was significantly decreased in the miR-145 groups compared with the control group (P<0.05). Using transwell apparatus pre-coated with Matrigel, the effects of miR-145 on cell invasiveness were examined. Following 24 h incubation, miR-145 transfected cells demonstrated significantly decreased invasiveness compared with the control cells (P<0.05). These results indicated that miR-145 inhibited cell migration and invasion in RCC cell lines.

miR-145 suppresses the expression of MMP-11 in RCC cell lines

Sachdeva et al revealed that miR-145 was able to target multiple metastasis related genes, including MMP-11 and ADAM-17 (19). Western blot analysis was performed to examine whether the protein level of MMP-11 was decreased following the transfection of miR-145. As shown in Fig. 3, MMP-11 was significantly decreased in 786-O and A498 cell lines 72 h after transfection of miR-145. Thus, miR-145 reduces the protein level of MMP-11 in RCC cells.

MMP-11 is a direct target of miR-145

Luciferase reporter assays were performed to evaluate whether the site was able to directly mediate the inhibition of expression. As shown in Fig. 4, the overexpression of miR-145 was able to suppress MMP-11 3′UTR-luciferase activity by 57% in 786-O cells and 45% in A498 cells (P<0.05). Therefore, MMP-11 may be a direct target of miR-145 in vitro.

Discussion

miR-145 was initially identified in mice from heart tissues using small RNA cloning techniques and then later reported in humans, revealing a unique seed sequence that is conserved in Xenopus and mammals (20). Human miR-145 (hsa-miR-145) is enriched in germline and mesoderm-derived tissues, including in the uterus, ovary, testis, prostate, spleen and heart (21). It is located on chromosome 5 (5q32–33) within a 4.09 kb region. The first study regarding the downregulation of miR-145 in various types of tumor was conducted by Michael et al (22). The authors demonstrated that the total number of clones sequenced for miR-145 was two from patients with colon adenocarcinomas compared with eight from normal tissue using the small RNA cloning approach. The results were confirmed by northern blot analysis. Notably, the authors also found a decreased level of miR-145 in precancerous adenomatous polyps, suggesting a possible role in tumor initiation.

miR-145 has been reported to be frequently downregulated in various types of cancer, including renal, prostate, bladder, lung and colon cancer, as well as B-cell malignancies (2327). The identification of miR-145 target genes is critical for understanding its role in tumorigenesis and is important for defining novel therapeutic targets. miR-145 suppresses tumor cell growth by targeting insulin receptor substrate-1 (28), c-Myc (29) and several other genes associated with carcinogenesis (19). miR-145 impacts migration, invasion and metastasis by targeting Fli 1 (30) and mucin 1 (31), respectively, and also affects p53-mediated cell cycle arrest by targeting p21 (32). In addition, the downregulation of miR-145 is associated with an aggressive phenotype and poor prognosis in prostate cancer (33,34). Therefore, upregulating miR-145 or providing analogous pharmaceutical compounds exogenously, may be effective cancer therapies for several types of tumor resulting from the activation or overexpression of these oncogenes. The present study revealed that miR-145 reduced cell migration and invasion by downregulating the expression of MMP-11. The results suggested that miR-145 may be used for the development of novel molecular markers and therapeutic approaches to inhibit the metastasis of RCC.

Metastasis is a multi-step process that requires cancer cells to detach from the main tumor, migrate and invade through the stroma, intravasate, survive in the circulatory system, arrive at a secondary site and extravasate, invade and grow at the secondary site (35). MMPs have been previously shown to contribute to migration and invasion in various types of cancer. They are a family of zinc-dependent extracellular endoproteinases that are collectively capable of degrading essentially all components of the extracellular matrix and basement membrane (36). They regulate and shape the tumor microenvironment, and are synthesized and secreted by multiple cell types, including corneal epithelial cells and fibroblasts (37). To date, at least 24 different human MMPs have been identified and they can be classified into five groups on the basis of substrate specificity, including interstitial collagenases, gelatinases, stromelysins, matrilysins and membrane-type MMPs (38,39).

MMPs are found in normal and pathological tissues in which matrix remodeling is involved, including embryonic development, wound healing, arthritis and angiogenesis, as well as tumor invasion and metastasis (40). Therefore, elevated levels of MMPs have been detected in the serum and urine of patients with several different types of cancer, including cancer of the bladder, breast, lung, colon, head and neck as well as melanoma (41). These MMPs are secreted as inactive zymogens (pro-MMPs) requiring extracellular activation and their activity is tightly regulated by specific tissue inhibitors (42). In view of the important role in tumor invasion and metastasis, inhibitors of MMP activity have been investigated as a method of preventing/decreasing tumor spread.

MMP-11, also termed stromelysin-3, is encoded by the MMP11 gene located on chromosome 22 q11.23 (43). It was originally identified by screening a breast cancer cDNA library for genes that were expressed at higher levels in invasive carcinomas compared with breast fibroadenomas (44). Additional investigation has demonstrated that MMP-11 is usually overexpressed in numerous types of human carcinoma, including breast, non-small cell lung and colorectal carcinomas, however, is rarely expressed in normal tissue, including the normal tissue surrounding the tumor. Notably, only adults have been shown to express MMP-11 in tumors and regenerating or healing tissues (4547). The functions of MMP-11 in cancer progression have been demonstrated by several preclinical observations. The upregulated expression of MMP-11 enhances tumor incidence in mice (48), homing of malignant epithelial cells (49), cancer progression by remodeling extracellular matrix (50) and has antiapoptotic and antinecrotic effects on tumor cells (51,52). MMP-11 deficiency increases tumor-free survival rate and modulates local or distant invasion (53). The knockdown of MMP-11 mRNA in gastric cancer cells suppresses tumor growth in vitro and in vivo and inhibits the spread of murine hepatocarcinoma cells to lymph nodes (53). The levels of MMP-11 expression may serve as a marker for transformation and invasion in several types of cancer, otherwise, it may be a target for cancer therapy in order to inhibit metastasis. The results from the present study suggested that miR-145 suppressed the migration and invasion of RCC cells through the downregulation of MMP-11. It may be investigated as a predictive marker for the early detection of tumor metastasis and for targeting therapeutic drugs to inhibit the invasion of RCC.

In conclusion, to the best of our knowledge this is the first study to demonstrate that miR-145 inhibited RCC cell migration and invasion by downregulating the expression of MMP-11. These findings have therapeutic implications and may be exploited for further treatment of RCC.

Future investigation is required to address whether the potential of miR-145 may be fully realized in cancer treatment. If so, it may be beneficial for the treatment of RCC.

References

1 

Siegel R, Ward E, Brawley O and Jemal A: Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin. 61:212–236. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Siegel R, DeSantis C, Virgo K, Stein K, Mariotto A, Smith T, Cooper D, Gansler T, Lerro C, Fedewa S, et al: Cancer treatment and survivorship statistics, 2012. CA Cancer J Clin. 62:220–241. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar

5 

Yan BC, Mackinnon AC and Al-Ahmadie HA: Recent developments in the pathology of renal tumors: morphology and molecular characteristics of select entities. Arch Pathol Lab Med. 133:1026–1032. 2009.PubMed/NCBI

6 

Yu ZH, Zhang Q, Wang YD, Chen J, Jiang ZM, Shi M, Guo X, Qin J, Cui GH, Cai ZM, et al: Overexpression of cyclooxygenase-1 correlates with poor prognosis in renal cell carcinoma. Asian Pac J Cancer Prev. 14:3729–3734. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Jemal A, Siegel R, Ward E, Hao Y, Xu J and Thun MJ: Cancer statistics, 2009. CA Cancer J Clin. 59:225–249. 2009. View Article : Google Scholar

8 

Zisman A, Pantuck AJ, Wieder J, Chao DH, Dorey F, Said JW, deKernion JB, Figlin RA and Belldegrun AS: Risk group assessment and clinical outcome algorithm to predict the natural history of patients with surgically resected renal cell carcinoma. J Clin Oncol. 20:4559–4566. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Xue YJ, Xiao RH, Long DZ, Zou XF, Wang XN, Zhang GX, Yuan YH, Wu GQ, Yang J, Wu YT, et al: Overexpression of FoxM1 is associated with tumor progression in patients with clear cell renal cell carcinoma. J Transl Med. 10:2002012. View Article : Google Scholar : PubMed/NCBI

10 

Choi YD, Kim KS, Ryu S, Park Y, Cho NH, Rha SH, Jang JJ, Ro JY, Juhng SW and Choi C: Claudin-7 is highly expressed in chromophobe renal cell carcinoma and renal oncocytoma. J Korean Med Sci. 22:305–310. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Garzon R, Pichiorri F, Palumbo T, Visentini M, Aqeilan R, Cimmino A, Wang H, Sun H, Volinia S, Alder H, et al: MicroRNA gene expression during retinoic acid-induced differentiation of human acute promyelocytic leukemia. Oncogene. 26:4148–4157. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Yao Y, Charlesworth J, Nair V and Watson M: MicroRNA expression profiles in avian haemopoietic cells. Front Genet. 4:1532013.PubMed/NCBI

13 

Lewis BP, Burge CB and Bartel DP: Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 120:15–20. 2005. View Article : Google Scholar : PubMed/NCBI

14 

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

15 

Calin GA and Croce CM: MicroRNA signatures in human cancers. Nat Rev Cancer. 6:857–866. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Yoo AS, Staahl BT, Chen L and Crabtree GR: MicroRNA-mediated switching of chromatin-remodelling complexes in neural development. Nature. 460:642–646. 2009.PubMed/NCBI

17 

le Sage C, Nagel R, Egan DA, Schrier M, Mesman E, Mangiola A, Anile C, Maira G, Mercatelli N, Ciafrè SA, et al: Regulation of the p27(Kip1) tumor suppressor by miR-221 and miR-222 promotes cancer cell proliferation. EMBO J. 26:3699–3708. 2007.PubMed/NCBI

18 

Zhang H, Li Y and Lai M: The microRNA network and tumor metastasis. Oncogene. 29:937–948. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Sachdeva M and Mo YY: miR-145-mediated suppression of cell growth, invasion and metastasis. Am J Transl Res. 2:170–180. 2010.PubMed/NCBI

20 

Lagos-Quintana M, Rauhut R, Lendeckel W and Tuschl T: Identification of novel genes coding for small expressed RNAs. Science. 294:853–858. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Lakshmipathy U, Love B, Adams C, Thyagarajan B and Chesnut JD: Micro RNA profiling: an easy and rapid method to screen and characterize stem cell populations. Methods Mol Biol. 407:97–114. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Michael MZ, O’ Connor SM, van Holst Pellekaan NG, Young GP and James RJ: Reduced accumulation of specific microRNAs in colorectal neoplasia. Mol Cancer Res. 1:882–891. 2003.PubMed/NCBI

23 

Doberstein K, Steinmeyer N, Hartmetz AK, Eberhardt W, Mittelbronn M, Harter PN, Juengel E, Blaheta R, Pfeilschifter J and Gutwein P: MicroRNA-145 targets the metalloprotease ADAM17 and is suppressed in renal cell carcinoma patients. Neoplasia. 15:218–230. 2013.PubMed/NCBI

24 

Ozen M, Creighton CJ, Ozdemir M and Ittmann M: Widespread deregulation of microRNA expression in human prostate cancer. Oncogene. 27:1788–1793. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Ichimi T, Enokida H, Okuno Y, Kunimoto R, Chiyomaru T, Kawamoto K, Kawahara K, Toki K, Kawakami K, Nishiyama K, et al: Identification of novel microRNA targets based on microRNA signatures in bladder cancer. Int J Cancer. 125:345–352. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Akao Y, Nakagawa Y and Naoe T: MicroRNA-143 and -145 in colon cancer. DNA Cell Biol. 26:311–320. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Akao Y, Nakagawa Y, Kitade Y, Kinoshita T and Naoe T: Downregulation of microRNAs-143 and -145 in B-cell malignancies. Cancer Sci. 98:1914–1920. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Shi B, Sepp-Lorenzino L, Prisco M, Linsley P, deAngelis T and Baserga R: Micro RNA 145 targets the insulin receptor substrate-1 and inhibits the growth of colon cancer cells. J Biol Chem. 282:32582–32590. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Sachdeva M, Zhu S, Wu F, Wu H, Walia V, Kumar S, Elble R, Watabe K and Mo YY: p53 represses c-Myc through induction of the tumor suppressor miR-145. Proc Natl Acad Sci USA. 106:3207–3212. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Larsson E, Fredlund Fuchs P, Heldin J, Barkefors I, Bondjers C, Genové G, Arrondel C, Gerwins P, Kurschat C, Schermer B, et al: Discovery of microvascular miRNAs using public gene expression data: miR-145 is expressed in pericytes and is a regulator of Fli1. Genome Med. 1:1082009. View Article : Google Scholar : PubMed/NCBI

31 

Sachdeva M and Mo YY: MicroRNA-145 suppresses cell invasion and metastasis by directly targeting mucin 1. Cancer Res. 70:378–387. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Seoane J, Le HV and Massagué J: Myc suppression of the p21(Cip1) Cdk inhibitor influences the outcome of the p53 response to DNA damage. Nature. 419:729–734. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Wang L, Tang H, Thayanithy V, Subramanian S, Oberg AL, Cunningham JM, Cerhan JR, Steer CJ and Thibodeau SN: Gene networks and microRNAs implicated in aggressive prostate cancer. Cancer Res. 69:9490–9497. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Chen X, Gong J, Zeng H, Chen N, Huang R, Huang Y, Nie L, Xu M, Xia J, Zhao F, et al: MicroRNA145 targets BNIP3 and suppresses prostate cancer progression. Cancer Res. 70:2728–2738. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Kwon YJ, Hurst DR, Steg AD, Yuan K, Vaidya KS, Welch DR and Frost AR: Gli1 enhances migration and invasion via up-regulation of MMP-11 and promotes metastasis in ERalpha negative breast cancer cell lines. Clin Exp Metastasis. 28:437–449. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Yan D, Dai H and Liu JW: Serum levels of MMP-11 correlate with clinical outcome in Chinese patients with advanced gastric adenocarcinoma. BMC Cancer. 11:1512011. View Article : Google Scholar : PubMed/NCBI

37 

Li DQ, Shang TY, Kim HS, Solomon A, Lokeshwar BL and Pflugfelder SC: Regulated expression of collagenases MMP-1, -8, and -13 and stromelysins MMP-3, -10, and -11 by human corneal epithelial cells. Invest Ophthalmol Vis Sci. 44:2928–2936. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Kähäri VM and Saarialho-Kere U: Matrix metalloproteinases and their inhibitors in tumour growth and invasion. Ann Med. 31:34–45. 1999.PubMed/NCBI

39 

Clark AF: New discoveries on the roles of matrix metalloproteinases in ocular cell biology and pathology. Invest Ophthalmol Vis Sci. 39:2514–2516. 1998.PubMed/NCBI

40 

Wu D, Ding J, Wang L, Pan H, Zhou Z, Zhou J and Qu P: microRNA-125b inhibits cell migration and invasion by targeting matrix metallopeptidase 13 in bladder cancer. Oncol Lett. 5:829–834. 2013.PubMed/NCBI

41 

Moses MA, Wiederschain D, Loughlin KR, Zurakowski D, Lamb CC and Freeman MR: Increased incidence of matrix metalloproteinases in urine of cancer patients. Cancer Res. 58:1395–1399. 1998.PubMed/NCBI

42 

Egeblad M and Werb Z: New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2:161–174. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Valdivia A, Peralta R, Matute-González M, Garcia Cebada JM, Casasola I, Jiménez-Medrano C, Aguado-Pérez R, Villegas V, Gonzalez-Bonilla C, Manuel-Apolinar L, et al: Co-expression of metalloproteinases 11 and 12 in cervical scrapes cells from cervical precursor lesions. Int J Clin Exp Pathol. 4:674–682. 2011.PubMed/NCBI

44 

Pan W, Arnone M, Kendall M, Grafstrom RH, Seitz SP, Wasserman ZR and Albright CF: Identification of peptide substrates for human MMP-11 (stromelysin-3) using phage display. J Biol Chem. 278:27820–27827. 2003. View Article : Google Scholar

45 

Rouyer N, Wolf C, Chenard MP, Rio MC, Chambon P, Bellocq JP and Basset P: Stromelysin-3 gene expression in human cancer: an overview. Invasion Metastasis. 14:269–275. 1994.PubMed/NCBI

46 

Kossakowska AE, Huchcroft SA, Urbanski SJ and Edwards DR: Comparative analysis of the expression patterns of metalloproteinases and their inhibitors in breast neoplasia, sporadic colorectal neoplasia, pulmonary carcinomas and malignant non-Hodgkin’s lymphomas in humans. Br J Cancer. 73:1401–1408. 1996.PubMed/NCBI

47 

Têtu B, Brisson J, Lapointe H and Bernard P: Prognostic significance of stromelysin 3, gelatinase A, and urokinase expression in breast cancer. Hum Pathol. 29:979–985. 1998.PubMed/NCBI

48 

Nöel AC, Lefebvre O, Maquoi E, VanHoorde L, Chenard MP, Mareel M, Foidart JM, Basset P and Rio MC: Stromelysin-3 expression promotes tumor take in nude mice. J Clin Invest. 97:1924–1930. 1996.PubMed/NCBI

49 

Masson R, Lefebvre O, Noël A, Fahime ME, Chenard MP, Wendling C, Kebers F, LeMeur M, Dierich A, Foidart JM, et al: In vivo evidence that the stromelysin-3 metalloproteinase contributes in a paracrine manner to epithelial cell malignancy. J Cell Biol. 140:1535–1541. 1998. View Article : Google Scholar : PubMed/NCBI

50 

Noël A, Boulay A, Kebers F, Kannan R, Hajitou A, Calberg-Bacq CM, Basset P, Rio MC and Foidart JM: Demonstration in vivo that stromelysin-3 functions through its proteolytic activity. Oncogene. 19:1605–1612. 2000.PubMed/NCBI

51 

Boulay A, Masson R, Chenard MP, El Fahime M, Cassard L, Bellocq JP, Sautès-Fridman C, Basset P and Rio MC: High cancer cell death in syngeneic tumors developed in host mice deficient for the stromelysin-3 matrix metalloproteinase. Cancer Res. 61:2189–2193. 2001.PubMed/NCBI

52 

Wu E, Mari BP, Wang F, Anderson IC, Sunday ME and Shipp MA: Stromelysin-3 suppresses tumor cell apoptosis in a murine model. J Cell Biochem. 82:549–555. 2001. View Article : Google Scholar : PubMed/NCBI

53 

Peruzzi D, Mori F, Conforti A, Lazzaro D, De Rinaldis E, Ciliberto G, La Monica N and Aurisicchio L: MMP11: a novel target antigen for cancer immunotherapy. Clin Cancer Res. 15:4104–4113. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2014
Volume 10 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu D, Li M, Wang L, Zhou Y, Zhou J, Pan H and Qu P: microRNA‑145 inhibits cell proliferation, migration and invasion by targeting matrix metallopeptidase-11 in renal cell carcinoma. Mol Med Rep 10: 393-398, 2014
APA
Wu, D., Li, M., Wang, L., Zhou, Y., Zhou, J., Pan, H., & Qu, P. (2014). microRNA‑145 inhibits cell proliferation, migration and invasion by targeting matrix metallopeptidase-11 in renal cell carcinoma. Molecular Medicine Reports, 10, 393-398. https://doi.org/10.3892/mmr.2014.2149
MLA
Wu, D., Li, M., Wang, L., Zhou, Y., Zhou, J., Pan, H., Qu, P."microRNA‑145 inhibits cell proliferation, migration and invasion by targeting matrix metallopeptidase-11 in renal cell carcinoma". Molecular Medicine Reports 10.1 (2014): 393-398.
Chicago
Wu, D., Li, M., Wang, L., Zhou, Y., Zhou, J., Pan, H., Qu, P."microRNA‑145 inhibits cell proliferation, migration and invasion by targeting matrix metallopeptidase-11 in renal cell carcinoma". Molecular Medicine Reports 10, no. 1 (2014): 393-398. https://doi.org/10.3892/mmr.2014.2149