Synthetic miR-145 mimic inhibits multiple myeloma cell growth in vitro and in vivo

Retraction in: /10.3892/or.2021.8125

  • Authors:
    • Qi Zhang
    • Weiqun Yan
    • Yang Bai
    • Hao Xu
    • Changhao Fu
    • Wenwen Zheng
    • Yingqiao Zhu
    • Jie Ma
  • View Affiliations

  • Published online on: November 5, 2014     https://doi.org/10.3892/or.2014.3591
  • Pages: 448-456
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Multiple myeloma (MM) is a disease with an adverse outcome and new therapeutic strategies are required to combat this disease. It is well known that tumor‑suppressor microRNA (miRNA) acts as a new potential anticancer agent. Accumulating evidence showed that microRNA-145 (miR-145) is a candidate tumor suppressor miRNA. However, whether miR-145 is involved in the development and progression of MM reamins to be determined. In the present study, we investigated the therapeutic potential of synthetic miR-145 against human MM cells in vitro and in vivo. The results showed that miR-145 was reduced in MM tissues and cell lines. Enforced expression of miR-145 by transfection with miR-145 mimics inhibited cell proliferation, migration, and the invasion abilities of H929 cells. Furthermore, our results demonstrated that the enforced expression of miR-145 in H929 cells profoundly decreased the levels of p-AKT and p-PI3K, which may contribute to some extent to the inhibition of MM cell proliferation and survival. The enforced expression of miR-145 in a xenograft mouse model suppressed tumor growth. In conclusion, our findings suggested that miR-145 may act as a tumor suppressor and contributes to the progression of MM. Additionally, miR-145 mimics is a potential therapeutic agent for the treatment of MM.

Introduction

Multiple myeloma (MM) is a B-cell malignancy that develops as a consequence of a multistep transformation process of normal plasma cells (PCs) within the human bone marrow milieu (huBMM), which results in paraprotein release in the serum, osteolytic bone disease, anemia and renal failure (1). Although in recent years important achievements in the understanding of MM pathogenesis have been obtained and novel research platforms and new therapeutics have been adopted (25), MM remains an incurable disease since available treatments do not have a curative effect. Therefore, the development of novel and more effective therapies is required (6,7).

MicroRNAs (miRNAs) are a class of short (~19 to 25 nucleotides) and highly conserved non-coding RNA sequences able to silence target genes by inhibiting the protein translation or by inducing mRNA degradation (8). miRNAs are important modulators of key regulatory cell pathways through the influence on target genes, and may play a crucial role in tumorigenesis (9), as deregulated miRNAs can act as oncogenes (onco-miRNAs) or tumor suppressors (TS-miRNAs) (10,11) with the former being upregulated and the latter downregulated in cancer cells (12). Underexpressed miRNAs in various types of cancer may function as tumor-suppressor genes mainly by downregulating oncogenic targets, onco-miRNAs are widely considered as oncogenes that promote tumor development by inhibiting tumor-suppressor genes and/or genes promoting cell differentiation or apoptosis (13). Therefore, miRNAs have elicited a growing interest in the cancer research community as new potential tumor cell targets (onco-miRNAs) or as new potential anticancer agents (TS-miRNAs) (14). Specifically, several miRNAs have been found to be abnormally upregulated or downregulated in primary MM cells or cell lines and involved in the pathogenesis of human MM (1417). Of note, miRNAs function as negative regulators of gene expression (18) and play a role in the pathogenesis of MM (19).

Of the miRNAs that are frequently deregulated in human cancer, microRNA-145 (miR-145) is of special interest in the field of miRNA therapeutics (2022). miR-145 is a 22-nt miRNA, whose genomic site is located in a fragile region of chromosome 5q. miR-145 has been noted for its underexpression in a various types of cancer such as, hepatocellular carcinoma (20), thyroid cancer (21), glioma (22), lung adenocarcinoma (23), bladder cancer (24), colon cancer (25) and breast cancer (26). Accumulating evidence has demonstrated that miR-145 acts as a tumor suppressor and exerts an inhibitory effect on cancer cell proliferation, migration and invasion by targeting different genes (2027). However, the biological role of miR-145 in MM has yet to be studied. Thus, the aim of this study was to characterize the expression of miR-145 in MM, identify its function in MM cells in vitro and in vivo, as well as to determine its utility in MM diagnosis and therapy.

Materials and methods

Tissue preparation and cell lines

The plasma specimens were obtained from patients with MM and normal donors from the First Hospital of Jilin University (Changchun, China). Plasma cells from MM patients and normal controls were purified from bone marrow aspirates using CD138 magnetic beads (Miltenyi Biotec) as described (19). All the patients provided written informed consent to participate in the study. This study was approved by the Ethics Committee of Jilin University (Changchun, China). CD138-negative mononuclear cells were used to establish long-term bone marrow stem cells (BMSC). The H929, MM1S, and RPMI-8226 MM cell lines were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). The cells were grown in RPMI-1640 medium supplemented with 10% fetal calf serum (FCS; Sigma-Aldrich, St. Louis, MO, USA), in 5% CO2 in humidified air at 37°C. The cells were routinely used and tested using Human Cell Line Genotyping System (Promega, Madison, WI, USA) when the cells were frozen and thawed.

siRNA and miRNA transfection

miR-145 mimic and negative control miRNA (mirVanaR miRNA mimic/inhibitor, Life Technologies, Carlsbad, CA, USA) were transiently transfected into MM cell lines in 6-well plates using Oligofectamine™ Transfection Reagent (Invitrogen Life Technologies, Carlsbad, CA, USA) according to the manufacturer’s instructions.

Quantitative reverse transcriptase polymerase chain reaction (RT-qPCR)

Isolation of total RNA from cells and clinical samples was performed using QIAzol reagent and the miRNeasy mini kit (Qiagen, Valencia, CA, USA) according to the manufacturer’s instructions. microRNA was reverse transcribed using the One Step PrimeScript miRNA cDNA Synthesis kit (Qiagen) and then quantified by real-time RT-PCR using SYBR Premix Ex Taq (Takara, Dalian, China). The PCR reactions were detected by ABI 7300 Fast system (Applied Biosystems, Foster City, CA, USA). The primers used were: U6 (HmiRQP9001); miR-145 (HmiRQP0192) (both from GeneCopoeia, Inc.). The integrity of the miRNA and the efficiency of qPCR in each sample were confirmed by the endogenous control U6 small RNA. The relative quantification of each miRNA was presented as the fold-change after normalization to the U6 RNA for the equation 2−ΔΔCt in the Rotor-Gene 6000 Series Software 1.7 (Qiagen, Hilden, Germany).

Cell proliferation and colony formation assays

Cells (5×103) were seeded in 96-well plates in triplicate, 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) working solution was added, and the cells were incubated for 4 h at 37°C. The medium was then removed and 200 μl of dimethyl sulfoxide was added to dissolve the formazan crystals. Cell viability was assessed for five consecutive days by absorbance at 570 nm using a microplate reader (Microplate Reader; Bio-Rad, Gaithersburg, MD, USA). All the experiments were repeated three times and the averages were calculated. The proliferation rate of cells was determined by measuring the incorporation of bromodeoxyuridine (BrdU) into the genomic DNA. Briefly, 2×103 cells/well were seeded in 96-well plates. A 5-bromodeoxyuridine (BrdU) incorporation assay was performed using the BrdU Cell Proliferation Assay kit (Chemicon International, Temecula, CA, USA) according to the manufacturer’s instructions. The growth rate of cells was calculated as described previously (27).

For the colony formation assay, the cells were seeded in 6-well plates at a low density (1×103 cells/well) and cultured for 7 days. The cells were fixed with 4% paraformaldehyde for 20 min and counted after staining with 1% crystal violet. The experiments were carried out in triplicate wells at least three times.

Cell cycle analysis

The cells were transfected with miR-145 or negative control miRNA. After culturing for 48 h, the cells were collected by trypsinisation, washed with ice-cold phosphate-buffered saline (PBS 7.2), and the fixed cells were incubated with DNA binding dye propidium iodide (PI, 20 μg/ml; Sigma-Aldrich) and RNase (1.0 mg/ml) for 30 min at 37°C in the dark, and then analysed by flow cytometry (BD Biosciences, Mansfield, MA, USA). Experiments were conducted in triplicate.

Apoptosis analysis

To determine the number of apoptotic cells, terminal deoxynucleotidyl transferase-mediated nick end-labeling (TUNEL) assay was performed. Briefly, cellular DNA fragmentation was measured with the ApopTag Red In Situ Apoptosis detection kit (Chemicon International) according to the manufacturer’s instructions when cells were transfected with miR-145 mimics and the corresponding negative control for 48 h. To quantify the apoptotic cells, the TUNEL-positive cells were counted using confocal microscopy (Olympus, Tokyo, Japan).

Caspase-3, -8 and -9 activity by ELISA as an additional indicator of apoptosis was also detected.

Caspase activity

The activity of caspase-3, -8 and -9 was determined by using caspase colorimetric protease assay kits (Millipore Corporation, Billerica, MA, USA) as per the manufacturer’s instructions. Briefly, the cells were washed twice with ice-cold PBS and harvested by centrifugation at 1,000 × g for 5 min. The cell pellets were then lysed in 150 μl buffer provided in the kit. Protein concentrations of lysates were measured by the Lowry method. An aliquot of lysates (80 μl) was incubated with 10 μl substrate of each caspase at 37°C for 1 h. The samples were analyzed at 405 nm in a microplate reader (Thermo Fisher Scientific Inc., Waltham, MA, USA). The relative caspase activity of the negative control group was referred as 100.

Cell migration and invasion assays

For the Transwell migration assays (BD Biosciences, Bedford, MA, USA), 5×104 H929 cells transfected with miR-145 mimics and the corresponding negative control were plated into the upper chamber, respectively. For the invasion assays, the upper chamber was precoated with Matrigel (BD Biosciences). In the two assays, the cells were plated in RPMI-1640 without serum, and RPMI-1640 supplemented with 10% FBS was added to the lower chamber and used as a chemoattractant. After 48-h incubation, the cells that migrated to the lower surface of the membrane were fixed with 100% methanol and stained with 0.2% crystal violet, imaged, and counted under an inverted microscope (Olympus). Experiments were performed in triplicate.

ELISA test

Culture media were collected and the VEGF ELISA was performed using Quantikine Human VEGF Immunoassay according to the manufacturer’s instructions (R&D Systems, Minneapolis, MN, USA). The amount of VEGF protein was compared to total protein as determined with the BCA reagent (Thermo Scientific, Rockford, IL, USA).

Western blot analysis

For western blot analysis, after 48-h transfection, the cells were harvested and rinsed once with ice-cold PBS and lysed in ice-cold cell lysis buffer (Walterson, London, UK) containing complete protease inhibitors cocktail (Sigma-Aldrich, Munich, Germany). The protein concentration was determined using the BCA protein assay kit (KeyGEN Biotech, Nanjing, China) using a c-globulin standard curve. Equal amounts of protein (20 μg/lane) from the cell lysates were separated on an 8–15% SDS-polyacrylamide gel (SDS-PAGE) and transferred onto nitrocellulose membranes (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA). The membranes were incubated for 2 h in PBS plus 0.1% Tween-20 and 5% non-fat skim milk to block non-specific binding. The membranes were incubated overnight at 4°C using the following antibodies: anti-survivin (1:1,000, Abcam); anti-AKT (1:1,000), anti-p-AKTSer473 162 (1:1,000), anti-Bcl-2 (1:1,000), anti-cyclin D1 (1:1,000) (all from Cell Signaling Technology, Beverly, MA, USA); anti-MMP-2 (1:1,000; EMD Millipore, Billerica, MA, USA); anti-MMP-9 (1:5,000), and anti-p21 (1:250), anti-PI3K (1:5,000) and anti-p-PI3K (Tyr458, 1:5,000), (all from Santa Cruz Biotechnology, Inc.). Anti-β-actin (1:10,000; Santa Cruz Biotechnology, Inc.) was used as a loading control. The membranes were incubated with the appropriate horseradish peroxidase-conjugated IgG (anti-rabbit 1:3,000; Cell Signaling Technology, or anti-mouse 1:10,000; Santa Cruz Biotechnology, Inc.) and the proteins were detected by enhanced chemiluminescence (ECL; Thermo Scientific).

Tumor growth in vivo

To determine whether miR-145 can inhibit tumor development, we manipulated miR-145 levels in MM H929 cells and then implanted the cells into the NOD-SCID mice. Thirty male severe combined immunodeficient (SCID) mice, 5–6 weeks old, were purchased from HFK Bio-Technology, Co., Ltd. (Beijing, China) and were maintained under specific pathogen-free (SPF) conditions and provided with food and water ad libitum. Cells (1×106) of H929, miR-145 expressing and negative control expression cells suspended in 50 μl of PBS were injected into the flanks of mice (n=10), respectively. The mice were monitored weekly for tumor growth. Tumor size was measured on a weekly basis, and tumor volume was calculated as 0.5236 × width2 × length. The mice were sacrificed humanely on week 4, and tumors were resected and weighed, with aliquots of tumors being fixed in 10% PBS to TUNEL. We also measured the miR-145 and VEGF level in tumor tissue by RT-qPCR and ELISA, respectively. TUNEL staining was performed on 5 μm sections of the excised tumors using ApopTag Red In Situ Apoptosis detection kit (Chemicon International) according to the manufacturer’s instructions. The number of apoptotic cells in five random high-power fields were counted and expressed as a percentage of total cells (apoptotic fraction).

The animal experiments were performed following the standards of animal care as outlined in the Guide for the Care and Use of Experimental Animals of Jilin University, following a protocol approved by the Ethics Committees of the Disease Model Research Center, Jilin University.

Statistical analysis

Data from at least three independent experiments are expressed as mean ± SD. The Student’s t-test or ANOVA was used when appropriate. The data were analyzed using the SPSS® statistical package, version 19.0 (SPSS Inc., Chicago, IL, USA) and the GraphPad Prism version 5.01 (GraphPad Software, San Diego, CA, USA) for Windows®. P<0.05 was considered to be statistically significant.

Results

miR-145 is decreased in MM plasma and cell lines

The expression of miR-145 levels in plasmas of 20 MM patients and 20 normal donors was examined by RT-qPCR. It was found that the plasma level of miR-145 was significantly downregulated in patients with MM compared with healthy control subjects (P<0.05, Fig. 1A). In addition, the expression of miR-145 in the H929, MM1S and RPMI-8226 MM cell lines, was significantly decreased compared with normal BM-derived CD138+ PCs from healthy donors (P<0.05, Fig. 1B).

miR-145 inhibits MM cell proliferation and colony formation

Having documented the significant downregulation of miR-145 in the MM cell lines and clinical MM, we examined how miR-145 affects MM cell biological behavior. To this end, miR-145 mimics and the corresponding negative control were synthesized and transfected into H929 cells, respectively. The results of RT-qPCR demonstrated that the expression of miR-145 in cells transfected with miR-145 mimics was increased compared with cells transfected with the negative control (P<0.05, Fig. 2A). Cell proliferation was determined by MTT and BrdU incorporation assays. As shown in Fig. 2B, the viability of H929 cells was markedly decreased by the transfection of miR-145 mimics (P<0.05 compared to the negative control), and the enhanced effect of miR-145 mimics on cell proliferation was observed beginning on day 2, although it became more obvious on days 4 and 5 (P<0.05, Fig. 2B). Consistent with MTT assays, BrdU incorporation assays also demonstrated that the proliferation rate of cells transfected with miR-145 mimics was significantly decreased compared to cells transfected with the negative control (P<0.05, Fig. 2C). The effect of miR-145 on cell colony formation was determined and it was found that cells transfected with miR-145 mimics significantly inhibited cell colony formation compared to cells transfected with the negative control (P<0.05, Fig. 2D). These findings suggested that the miR-145 overexpression markedly inhibited cell proliferation and colony formation in H929 cells.

Effects of miR-145 on the cell cycle in H929 cells

In order to determine the effects of miR-145 on the cell cycle, FACScan flow cytometry assays were performed. A flow cytometric analysis revealed that the G1-phase cell population was observed in the cells transfection miR-145 mimics as compared with the cells transfected with the negative control (P<0.05, Fig. 3A and B). In addition, transfection with miR-145 mimics resulted in a much lower percentage of cells in the S phase compared with those cells transfected with the negative control (P<0.05, Fig. 3A and B). There were no significant differences in cells in the G2/M phases among the groups.

We also analyzed the effects of miR-145 on the expression of cell-cycle relevant proteins, such as cyclin D1 and p21 by western blot analysis. As shown in Fig. 3C and D, compared to cells transfected with the negative control, p21 expression was significantly increased, whereas, cyclin D1 expression was significantly decreased in cells transfected with miR-145 mimics (P<0.05, Fig. 3D).

miR-145 induces cell apoptosis in H929 cells

The role of miR-145 in MM cell apoptosis was examined. Cell apoptotic analysis of MM cells (H929) was performed by TUNEL 48 h after transfection. The cell apoptotic rate of tumor cells transfected with miR-145 was higher than that of cells transfected with the negative control (P<0.05, Fig. 4A).

We analyzed the effects of miR-145 on caspase-3, -8 and -9 activity by ELISA. Caspase-3, -8 and -9 activity in cells transfected with miR-145 mimics was significantly increased compared with those cells transfected with the negative control (P<0.05) (Fig. 4B–D).

To determine the potential mechanism of the miR-145 effect on cell apoptosis, apoptosis-related protein, survivin and Bcl-2 expression was examined by western blot analysis. Western blot analysis revealed a significant decrease in survivin and Bcl-2 expression in cells transfected with miR-145 compared to cells transfected with the negative control (P<0.05, Fig. 4E and F). These results suggested that the overexpression of miR-145 induces cell apoptosis in H929 cells.

miR-145 inhibits cell migration and invasion in H929 cells

To detect whether miR-145 affects the motility of cancer cells, migration assay was performed. The results showed that the cell motility expressing miR-145 was significantly reduced in H929 cells (P<0.05; Fig. 5A). Subsequently, the ability of miR-145 mimics to reduce the invasiveness of H929 cells was investigated using the Transwell system assay. It was found that invasion was also significantly decreased in cells expressing miR-145 compared to cells expressing the negative control (P<0.05; Fig. 5B).

To determine the potential mechanism of the miR-145 effect on cell migration and invasion, VEGF secretion was examined by ELISA. The results showed that overexpression of miR-145 significantly inhibited VEGF secretion in H929 cells (P<0.05, Fig. 5C). In addition, MMP-2 and MMP-9 expression levels were determined by western blot analysis. Western blot analysis showed that MMP-2 and MMP-9 expression was significantly reduced in cells transfected with miR-145 compared to cells transfected with the negative control (Fig. 5D).

Effect of miR-145 on PI3K/AKT signaling pathway

The PI3K/AKT pathway is important in cell proliferation and survival for various types of cancer. Thus, in this study, we evaluated the effect of miR-145 on several key downstream molecules involved in the PI3K/AKT signaling pathway. Therefore, measurements of the phosphorylation/activation pattern of PI3K and AKT was performed by western blot analysis. The results of western blot analysis demonstrated that cells transfected with miR-145 mimics inhibited the phosphorylation of PI3K and p-AKT expression compared to the untreated cells and cells transfected with the negative control, without altering the total protein levels of PI3K or AKT in H299 cells (Fig. 6).

miR-145 suppresses tumor growth in the nude mouse model

We determined whether miR-145 was able to inhibit tumor growth in the xenograft tumor model. Tumor growth was monitored for four weeks. On day 28, the mice were sacrificed and final tumor weights and tumor volume were determined. The results showed that tumor weight and volume of mice treated with miR-145 mimics were significantly reduced when compared to the untreated cells and cells treated with the negative control (P<0.05; Fig. 7A–C). In addition, in this study, we examined the miR-145 level and VEGF secretion in grafted tumor tissues by RT-qPCR and ELISA, respectively. RT-qPCR revealed that miR-145 expression levels were obviously increased in the groups treated with miR-145 mimics compared to the untreated and negative control groups (P<0.05, Fig. 7D). The results of ELISA showed that VEGF secretion levels were obviously decreased in the groups treated with miR-145 mimics compared to the untreated and negative control groups (P<0.05, Fig. 7E). We also assessed the efficacy of miR-145 in inducing cell apoptosis in vivo using the TUNEL assay. As shown in Fig. 7F, the cells treated with miR-145 significantly induced apoptosis compared to the untreated and negative control groups (P<0.05). These results suggested that overexpression of miR-145 inhibited tumor growth in vivo.

Discussion

In this study, we have shown that miR-145 expression was downregulated in MM plasma and cell lines, and that the enforced expression of miR-145 by miR-145 mimics inhibited cell proliferation, migration, and invasion in H929 cells. We also found that the enforced expression of miR-145 suppressed tumor growth of MM in a nude mouse model. In addition, our results demonstrated that the enforced expression of miR-145 in H929 cells profoundly decreased levels of p-AKT and p-PI3K, which may contribute to the inhibition of MM cell proliferation and survival to some extent. Therefore, our study identifies that miR-145 may be a tumor suppressor in the progression of MM. To the best of our knowledge, this is the first experimental evidence of antitumor activity of miR-145 in the treatment for MM in vitro and in vivo.

It is well known that miRNAs are crucial in the regulation of cell proliferation, cycle, migration and invasion by regulating target gene, and that miRNA dysregulation is causally involved in the initiation and progression of cancer (28,29). miR-145, a major tumor-suppressor miRNA, is downregulated in many types of cancer, including hepatocellular carcinoma, thyroid cancer, glioma, lung adenocarcinoma, bladder cancer, colon cancer, as well as breast cancer (2026). miR-145 also plays an important role in regulating smooth muscle cell differentiation (30) and inducing apoptosis (31). Genes currently identified as targeted genes of miR-145 are AKT, Fascin1, Pai-1, Oct-4, Sox-2, Klf4, c-Myc, insulin receptor substrate-1 (IRS1), Muc1, YES and Stat1 (21,24,3237), which are involved in several different signaling pathways, suggesting that miR-145 is a tumor-suppressor miRNA and plays a crucial role in the initiation and progression of tumor. Accumulating evidence has proved that enforced miR-145 reduced the expression of differentiation markers, decreased cell proliferation, migration and invasion, and induced cell cycle arrest and apoptosis (2026,3237). For example, Lu et al (38) reported that miR-145 suppressed cell proliferation and motility and induced cell apoptosis via targeting two oncogenes, ANGPT2 and NEDD9 in renal cell carcinoma. Li et al (39) reported that miR-145 may act as a tumor suppressor and contribute to the progression of OS by targeting Rho-associated protein kinase 1 (ROCK1). Xing et al (40) demonstrated that miR-145 suppressed anchorage-independent growth and cell motility in the Hep-G2 liver cancer cell line and the MKN-45 gastric cancer cell line, and inhibited cell proliferation in a cell type-specific manner by targeting IRS1. Feng et al (41) reported that miR-145 was found to suppress the invasion and metastasis of colorectal cancer (CRC) by functioning as a tumor suppressor through the direct repression of Fascin1. Recently, Wang et al (42) showed that miR-145 inhibited HCC by targeting IRS1 and its downstream signaling, suggesting the loss of miR-145 regulation is a potential molecular mechanism causing aberrant oncogenic signaling in HCC. Shao et al (43) suggested that miR-145 exerts its tumor-suppressor function by targeting c-Myc and Cdk6, leading to the inhibition of OSCC cell growth. Similar to the aforementioned studies, in the present study, we found that miR-145 was significantly reduced in the plasma of patients with MM compared with the plasma of normal controls. miR-145 inhibited cell proliferation, migration, and invasion in MM cells. These findings suggest miR-145 acts as a tumor suppressor of MM, and that miR-145 expression inhibits tumor growth of MM.

It is well known that the PI3K/AKT pathway is an important oncogenic pathway and is frequently activated during tumorigenesis, playing a crucial role in cell proliferation and survival for various types of cancer (44,45). It has been shown that the activation of PI3K/AKT reduces the levels of p21Cip1 and p27Kip1, and increases the expression of CCND1 (46,47), which contributes to in the regulation of cell cycle progression through the G1 phase (48). In addition, AKT prevents apoptosis by increasing the level of anti-apoptotic proteins Bcl-2 and Bcl-xL, while inactivating proapoptotic proteins such as Bad, Bax and Bim [Maddika et al (49); Datta et al (50)]. Of note, results of a recently study showed that miR-145 has a tumor-suppressor function and directly targets AKT3 to regulate the PI3K/AKT signaling pathway in thyroid cancer (21). In the present study, we found that the upregulation of miR-145 inhibited cell proliferation, migration and invasion, induced cell cycle and apoptosis, and decrease AKT and PI3K phosphorylation. Thus, we suggest that the molecular mechanism by which miR-145 inhibited MM cell proliferation and tumorigenicity may be attributed to, at least in part, suppression of the PI3K/AKT signaling pathway.

Taken together, the findings reported in the present study suggest that miR-145 inhibits cell proliferation, migration and invasion, induces cell apoptosis, and suppresses tumor growth at least in part by inhibiting the PI3K/AKT signaling pathway. Our data suggest that miR-145 is a potential therapeutic target for MM treatment.

Acknowledgements

The authors gratefully acknowledge the financial support provided by Science and Technology Research and Innovation Team funded of Jilin province (JL2012058).

References

1 

Palumbo A and Anderson K: Multiple myeloma. N Engl J Med. 364:1046–1060. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Terpos E, Morgan G, Dimopoulos MA, et al: International Myeloma Working Group recommendations for the treatment of multiple myeloma-related bone disease. J Clin Oncol. 31:2347–2357. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Munshi NC and Anderson KC: New strategies in the treatment of multiple myeloma. Clin Cancer Res. 19:3337–3344. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Rossi M, Di Martino MT, Morelli E, et al: Molecular targets for the treatment of multiple myeloma. Curr Cancer Drug Targets. 12:757–767. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Gao Y, Gao F, Ma JL, Sun WZ and Song LP: The potential clinical applications and prospects of microRNAs in lung cancer. Onco Targets Ther. 7:901–906. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Nana-Sinkam SP and Croce CM: Clinical applications for microRNAs in cancer. Clin Pharmacol Ther. 93:98–104. 2013. View Article : Google Scholar

7 

Rajkumar SV: Multiple myeloma: 2011 update on diagnosis, risk-stratification, and management. Am J Hematol. 86:57–65. 2011. View Article : Google Scholar

8 

Lionetti M, Agnelli L, Mosca L, et al: Integrative high-resolution microarray analysis of human myeloma cell lines reveals deregulated miRNA expression associated with allelic imbalances and gene expression profiles. Genes Chromosomes Cancer. 48:521–531. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Calin GA, Sevignani C, Dumitru CD, et al: Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA. 101:2999–3004. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Iorio MV and Croce CM: Causes and consequences of microRNA dysregulation. Cancer J. 18:215–222. 2012. 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 

Garzon R, Marcucci G and Croce CM: Targeting microRNAs in cancer: rationale, strategies and challenges. Nat Rev Drug Discov. 9:775–789. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Tagliaferri P, Rossi M, Di Martino MT, et al: Promises and challenges of MicroRNA-based treatment of multiple myeloma. Curr Cancer Drug Targets. 12:838–846. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Lionetti M, Biasiolo M, Agnelli L, et al: Identification of microRNA expression patterns and definition of a microRNA/mRNA regulatory network in distinct molecular groups of multiple myeloma. Blood. 114:e20–e26. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Calin GA and Croce CM: MicroRNA-cancer connection: the beginning of a new tale. Cancer Res. 66:7390–7394. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Chesi M and Bergsagel PL: Molecular pathogenesis of multiple myeloma: basic and clinical updates. Int J Hematol. 97:313–323. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Klein B, Seckinger A, Moehler T and Hose D: Molecular pathogenesis of multiple myeloma: chromosomal aberrations, changes in gene expression, cytokine networks, and the bone marrow microenvironment. Recent Results Cancer Res. 183:39–86. 2011. View Article : Google Scholar : PubMed/NCBI

18 

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

19 

Pichiorri F, Suh SS, Rocci A, et al: Downregulation of p53-inducible microRNAs 192, 194, and 215 impairs the p53/MDM2 autoregulatory loop in multiple myeloma development. Cancer Cell. 18:367–381. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Noh JH, Chang YG, Kim MG, et al: MiR-145 functions as a tumor suppressor by directly targeting histone deacetylase 2 in liver cancer. Cancer Lett. 335:455–462. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Boufraqech M, Zhang L, Jain M, et al: miR-145 suppresses thyroid cancer growth and metastasis and targets AKT3. Endocr Relat Cancer. 21:517–531. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Lee SJ, Kim SJ, Seo HH, et al: Over-expression of miR-145 enhances the effectiveness of HSVtk gene therapy for malignant glioma. Cancer Lett. 320:72–80. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Cho WC, Chow AS and Au JS: Restoration of tumour suppressor hsa-miR-145 inhibits cancer cell growth in lung adenocarcinoma patients with epidermal growth factor receptor mutation. Eur J Cancer. 45:2197–2206. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Chiyomaru T, Enokida H, Tatarano S, et al: miR-145 and miR-133a function as tumour suppressors and directly regulate FSCN1 expression in bladder cancer. Br J Cancer. 102:883–891. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Zhang J, Guo H, Zhang H, et al: Putative tumor suppressor miR-145 inhibits colon cancer cell growth by targeting oncogene Friend leukemia virus integration 1 gene. Cancer. 117:86–95. 2011. View Article : Google Scholar

26 

Spizzo R, Nicoloso MS, Lupini L, et al: miR-145 participates with TP53 in a death-promoting regulatory loop and targets estrogen receptor-alpha in human breast cancer cells. Cell Death Differ. 17:246–254. 2010. View Article : Google Scholar

27 

Lin Y, Peng S and Yu H: RNAi-mediated downregulation of NOB1 suppresses the growth and colony-formation ability of human ovarian cancer cells. Med Oncol. 29:311–317. 2012. View Article : Google Scholar

28 

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

29 

Manikandan J, Aarthi JJ, Kumar SD and Pushparaj PN: Oncomirs: the potential role of non-coding microRNAs in understanding cancer. Bioinformation. 2:330–334. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Cordes KR, Sheehy NT, White MP, et al: miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature. 460:705–710. 2009.PubMed/NCBI

31 

Ostenfeld MS, Bramsen JB, Lamy P, et al: miR-145 induces caspase-dependent and -independent cell death in urothelial cancer cell lines with targeting of an expression signature present in Ta bladder tumors. Oncogene. 29:1073–1084. 2010. View Article : Google Scholar

32 

Villadsen SB, Bramsen JB, Ostenfeld MS, et al: The miR-143/-145 cluster regulates plasminogen activator inhibitor-1 in bladder cancer. Br J Cancer. 106:366–374. 2012. View Article : Google Scholar :

33 

Xu N, Papagiannakopoulos T, Pan G, Thomson JA and Kosik KS: MicroRNA-145 regulates OCT4, SOX2, and KLF4 and represses pluripotency in human embryonic stem cells. Cell. 137:647–658. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Sachdeva M, Zhu S, Wu F, et al: 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

35 

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

36 

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 :

37 

Gregersen LH, Jacobsen AB, Frankel LB, Wen J, Krogh A and Lund AH: MicroRNA-145 targets YES and STAT1 in colon cancer cells. PLoS One. 5:e88362010. View Article : Google Scholar : PubMed/NCBI

38 

Lu R, Ji Z, Li X, et al: miR-145 functions as tumor suppressor and targets two oncogenes, ANGPT2 and NEDD9, in renal cell carcinoma. J Cancer Res Clin Oncol. 140:387–397. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Li E, Zhang J, Yuan T and Ma B: miR-145 inhibits osteosarcoma cells proliferation and invasion by targeting ROCK1. Tumour Biol. 35:7645–7650. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Xing AY, Wang B, Shi DB, et al: Deregulated expression of miR-145 in manifold human cancer cells. Exp Mol Pathol. 95:91–97. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Feng Y, Zhu J, Ou C, et al: MicroRNA-145 inhibits tumour growth and metastasis in colorectal cancer by targeting fascin-1. Br J Cancer. 110:2300–2309. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Wang Y, Hu C, Cheng J, et al: MicroRNA-145 suppresses hepatocellular carcinoma by targeting IRS1 and its downstream Akt signaling. Biochem Biophys Res Commun. 446:1255–1260. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Shao Y, Qu Y, Dang S, Yao B and Ji M: MiR-145 inhibits oral squamous cell carcinoma (OSCC) cell growth by targeting c-Myc and Cdk6. Cancer Cell Int. 13:512013. View Article : Google Scholar : PubMed/NCBI

44 

Chandramohan V, Jeay S, Pianetti S and Sonenshein GE: Reciprocal control of Forkhead box O 3a and c-Myc via the phosphatidylinositol 3-kinase pathway coordinately regulates p27Kip1 levels. J Immunol. 172:5522–5527. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Cheng GZ, Park S, Shu S, et al: Advances of AKT pathway in human oncogenesis and as a target for anti-cancer drug discovery. Curr Cancer Drug Targets. 8:2–6. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Medema RH, Kops GJ, Bos JL and Burgering BM: AFX-like Forkhead transcription factors mediate cell cycle regulation by Ras and PKB through p27kip1. Nature. 404:782–787. 2000. View Article : Google Scholar : PubMed/NCBI

47 

Roy SK, Srivastava RK and Shankar S: Inhibition of PI3K/AKT and MAPK/ERK pathways causes activation of FOXO transcription factor, leading to cell cycle arrest and apoptosis in pancreatic cancer. J Mol Signal. 5:102010. View Article : Google Scholar : PubMed/NCBI

48 

Massague J: G1 cell-cycle control and cancer. Nature. 432:298–306. 2004. View Article : Google Scholar : PubMed/NCBI

49 

Maddika S, Ande SR, Panigrahi S, et al: Cell survival, cell death and cell cycle pathways are interconnected: implications for cancer therapy. Drug Resist Updat. 10:13–29. 2007. View Article : Google Scholar : PubMed/NCBI

50 

Datta SR, Dudek H, Tao X, et al: Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell. 91:231–241. 1997. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2015
Volume 33 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang Q, Yan W, Bai Y, Xu H, Fu C, Zheng W, Zhu Y and Ma J: Synthetic miR-145 mimic inhibits multiple myeloma cell growth in vitro and in vivo Retraction in /10.3892/or.2021.8125. Oncol Rep 33: 448-456, 2015
APA
Zhang, Q., Yan, W., Bai, Y., Xu, H., Fu, C., Zheng, W. ... Ma, J. (2015). Synthetic miR-145 mimic inhibits multiple myeloma cell growth in vitro and in vivo Retraction in /10.3892/or.2021.8125. Oncology Reports, 33, 448-456. https://doi.org/10.3892/or.2014.3591
MLA
Zhang, Q., Yan, W., Bai, Y., Xu, H., Fu, C., Zheng, W., Zhu, Y., Ma, J."Synthetic miR-145 mimic inhibits multiple myeloma cell growth in vitro and in vivo Retraction in /10.3892/or.2021.8125". Oncology Reports 33.1 (2015): 448-456.
Chicago
Zhang, Q., Yan, W., Bai, Y., Xu, H., Fu, C., Zheng, W., Zhu, Y., Ma, J."Synthetic miR-145 mimic inhibits multiple myeloma cell growth in vitro and in vivo Retraction in /10.3892/or.2021.8125". Oncology Reports 33, no. 1 (2015): 448-456. https://doi.org/10.3892/or.2014.3591