miR‑338‑3p regulates the proliferation, apoptosis and migration of SW480 cells by targeting MACC1

  • Authors:
    • Mingliang Lu
    • Hua Huang
    • Jinhui Yang
    • Jun Li
    • Gongfang Zhao
    • Weihua Li
    • Xinhua Li
    • Guobin Liu
    • Li Wei
    • Baoping Shi
    • Chunping Zhao
    • Yan Fu
  • View Affiliations

  • Published online on: February 12, 2019     https://doi.org/10.3892/etm.2019.7260
  • Pages: 2807-2814
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The mortality and incidence rates of colorectal cancer (CRC) vary widely worldwide. miR‑338‑3p inhibits tumor cell proliferation in several types of cancer, however, the role of miR‑338‑3p on CRC remains unknown. The aim of the current study was to investigate the cellular function of miRNA‑338‑3p (miR‑338‑3p) in CRC, the malignant behavior of CRC cells and the interaction between miR‑338‑3p and metastasis‑associated in colon cancer‑1 (MACC1). miR‑338‑3p expression was significantly decreased in CRC tissue compared with adjacent normal tissue. In the CRC cell line SW480, miR‑338‑3p overexpression suppressed cell proliferation and migration and induced G1/S cell cycle arrest and apoptosis. By contrast, miR‑338‑3p knockdown significantly enhanced cell proliferation and migration, and suppressed G1/S cell cycle arrest and apoptosis. Furthermore, the dual‑luciferase reporter assay confirmed MACC1 as a direct target of miR‑338‑3p. In addition, miR‑338‑3p overexpression reduced the level of MACC1 protein expression and MACC1 expression was significantly upregulated in CRC tissue samples. MACC1 siRNA significantly reduced CRC cell proliferation and migration, whilst cell apoptosis was significantly increased. In conclusion, miR‑338‑3p expression was decreased in CRC. miR‑338‑3p regulated the proliferation, apoptosis and migration of CRC cells by targeting MACC1.

Introduction

The incidence rate of colorectal cancer (CRC) varies widely between different regions of the world (1). In Western countries, the incidence rate of CRC is the second highest among all malignant tumors (2). In China, the incidence and mortality rates of CRC ranks fourth among all malignant tumors, with 310,244 diagnoses made in 2011 alone (3). The incidence and mortality rates of CRC vary significantly among countries and different regions of China, as well as between urban and rural areas (3).

Although improvements in diagnosis and treatment have contributed to the early detection and good prognosis of patients with CRC, those with more advanced lesions, particularly those with distant metastasis exhibit a poor prognosis and efficient therapeutic methods are lacking (4). Metastasis occurs when tumor cells detach from the primary tumor site, invade the circulatory and lymphatic vessels and travel to distant organs or tissues where they invade and proliferate forming new tumors that affect multiple organs, which may result in mortality (5). Therefore, an effective therapeutic strategy is needed and targeting the molecular and cellular mechanism underlying cancer cell invasion and metastasis may be an appropriate therapeutic strategy.

MicroRNAs (miRNAs) are small, non-coding RNAs 18–25 nucleotides in length and are widely expressed in mammals (6). miRNAs are regulators of gene expression, which affect cell proliferation, differentiation and death (7,8). miR-338-3p is located on chromosome 17q25.3 within the 8th intron of the apoptosis-associated tyrosine kinase gene, which produces two forms: miR-338-3p and miR-338-5p (914). It has been reported that chromosome 17q23-25 is a region associated with mutations in several malignant tumors (15,16). In addition, this locus is associated with many malignant biological behaviors which include tumor vascular invasion and distant metastasis (13). Previous studies have demonstrated that miR-338-3p inhibits glioblastoma proliferation (9,12), renal carcinoma invasion (10), gastric cancer progression (11,17), ovarian epithelial carcinoma growth (18) and hepatocellular carcinoma proliferation (14). However, studies investigating the cellular function of miR-338-3p in CRC are lacking.

Metastasis-associated in colon cancer-1 (MACC1) was identified by Stein et al (19) in 2009 and is located on chromosome 7 (7p21.1). A previous study demonstrated that MACC1 exhibits 49.3 and 43.7% homology with SH3 domain-binding protein 4 between nucleotide and amino acid sequences, respectively (20) The protein structure of MACC1 includes a SH3 domain and a P-X-X-P motif, which is required for binding to SH3 domains (20). In addition, there are multiple phosphorylation sites, which indicate that MACC1 may be required for important signal transduction pathways (20). MACC1 expression in CRC is an independent predictor of metastasis (19). A previous study demonstrated that miR-338-3p regulates MACC1 expression in cervical cancer as well as cancer progression by targeting MACC1 via the mitogen-activated protein kinase (MAPK) signaling pathway (21). miR-338-3p also inhibits epithelial-mesenchymal transition (EMT) by targeting zinc finger E-box-binding homeobox 2 (ZEB2) and MACC1 (11). The aggressive features associated with glioma cells are suppressed by miR-338-3p by directly silencing MACC1 expression (12). However, whether there is an association between miR-338-3p and MACC1 in CRC remains unknown.

Therefore, it was hypothesized that the miR-338-3p/MACC1 axis may participate in the occurrence and development of CRC. The aim of the current study was to investigate miR-338-3p expression in CRC tissues and its underlying mechanism. The results of the current study may facilitate the identification of a potential therapeutic target for the treatment of CRC.

Materials and methods

Patient samples

The present study analyzed tissue samples from 15 patients (9 males, 6 females; mean age, 52.5±9.3 years) with CRC who underwent surgery at the Second Affiliated Hospital of Kunming Medical University (Kunming, China) between January 2015 and May 2015. CRC was confirmed in all patients following pathological examination and patients did not receive neoadjuvant chemotherapy or radiotherapy. CRC tissue and adjacent normal colon tissue (≥2 cm) were collected and stored in liquid nitrogen until further use. The study was approved by the Ethics Committee of the Second Affiliated Hospital of Kunming Medical University and all patients provided their written informed consent.

Cell culture

The human CRC cell line SW480 and the 293T cell line were provided by the Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (Shanghai, China). Cells were cultured in high glucose Dulbecco's modified Eagle's medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) and maintained at 37°C in a 5% CO2-humidified incubator. Cells were passaged every 2–3 days, according to cell density. Experiments were performed when cells had reached a confluence of 80% confluence.

MACC1 siRNA lentivirus construction, packaging and transfection

The siRNA fragments targeting MACC1 were identified and amplified as previously described (21). MACCI siRNA was cloned into the lentiviral transfer plasmid LV-3 (pGLVH1/GFP/Furo; cat. no. c06003, Shanghai GenePharma Co. Ltd., Shanghai, China). 293T cells were subsequently transfected with the transfer plasmid (10 µg) encoding MACC1 siRNA, lentiviral packaging plasmid pRSV-Rev (2 µg; cat. no. #12253) and the envelope plasmid pMD2.G (2 µg; #12259; both Addgene, Inc., Cambridge, MA, USA) using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Following 48-h incubation at 37°C, the lentiviral supernatant was harvested and pelleted by centrifugation at 4,000 × g for 10 min at 4°C. The lentiviral supernatant was subsequently filtered using 0.4 µm cellulose acetate filters and used to transfect SW480 cells. SW480 cells were seeded at a density of 1×104 cells/well prior to transfection with lentivirus. Following 48-h transfection, SW480 cells were in subsequent experimentation.

Cell proliferation assay

miR-338-3p mimic (5′-UUUGAGCAGCACUCAUUUUUGC-3′), miR-338-3p inhibitor (5′-CAACAAAAUCACUGAUGCUGGA-3′), and miR control (5′-CAGUACUUUUAGUGUGUACAA-3′) were purchased from Shanghai GenePharma Co., Ltd. (Shanghai, China). Briefly, SW480 cells were seeded into 96-well plates at a density of 2×104 cells/well. Once at 70–80% confluency, cells were transfected with 50 nM miR control (transfection control), miR-338-3p mimic or miR-338-3p inhibitor using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Following 24-h transfection, 10 µg of MTT was added to each well and incubated for a further 4 h at 37°C. Following incubation, 50 µl DMSO was added to each well and plates were shaken for 10 min. DMSO was used as control. Cell proliferation was determined by measuring absorbance at a wavelength of 570 nm using a microplate reader.

Cell apoptosis assay

Cell apoptosis was analyzed using an Annexin V-fluorescein isothiocyanate (FITC) Apoptosis Detection kit (cat. no. 401003; BestBio, Shanghai, China), according to the manufacturer's protocol. Following 48 h transfection with controls, miR-338-3p mimics and miR-338-3p inhibitors, SW480 cells were washed twice with PBS, dissociated with trypsin and pelleted using centrifugation at 1,500 × g for 5 min at 4°C. Cells were resuspended in 500 µl 1X binding buffer and subsequently stained with 1 µl Annexin V-FITC for 15 min at 4°C in the dark followed by 5 µl propidium iodine (PI) for 15 min at 4°C in the dark. Apoptotic cells were detected using a BD FACS Calibur flow cytometer (BD Biosciences, Franklin Lakes, NJ, USA). Data were analyzed using CellQuest Pro software (version 5.1; BD Biosciences).

Cell cycle analysis

Following 48 h transfection with controls, miR-338-3p mimics and miR-338-3p inhibitors, SW480 cells were washed twice with PBS, dissociated with trypsin, and pelleted using centrifugation at 1,500 × g for 5 min at 4°C. Cells were then resuspended in 100 µl of RNase A and incubated for 30 min at 37°C. Cells were subsequently stained with 400 µl PI (BestBio) and incubated for 30 min at 4°C for 30 min in the dark. Flow cytometry was used to detect the cell cycle distribution.

Transwell migration assay

Migration assays were performed using a 24-well Transwell chambers with 8.0 µm pore size inserts (Corning Incorporated, Corning, NY, USA). Following 24-h transfection with controls, miR-338-3p mimics and miR-338-3p inhibitors, SW480 cells were seeded in the upper chambers in serum-free medium at a density of 3×104 cells/well, whilst medium (containing 10% FBS) was added to the lower chambers. Following 24-h incubation at 37°C, the chamber was removed and cells that migrated through the membrane were fixed with 4% formaldehyde (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) and acetic acid for 15 min at 37°C, washed with PBS and stained with crystal violet solution for 10 min at 37°C. The number of cells were observed and counted from five randomly selected visual fields under a light microscope (magnification, ×200).

Dual-luciferase reporter assay

TargetScan Bioinformatics software (www.targetscan.org/vert_72) was used to predict the putative target genes of miR-338-3p. To confirm direct target binding, the wild-type (wt) or mutant (mut) 3′-untranslated region (UTR) MACC1 were cloned into the pMIR-REPORT™ Luciferase miRNA Expression reporter vector (cat. no. AM5795; Ambion; Thermo Fisher Scientific, Inc.) to generate pmir-MACC1-wtUTR and pmir-MACC1-mutUTR. 293T cells were co-transfected with miR-338-3p mimics or miR-negative controls (NCs; Wuhan Biofavor Biotech Services Co., Ltd., Wuhan, China) and pmir-MACC1-wtUTR or pmir-MACC1-mutUTR using Lipofectamine® 2000 (cat. no. 15596026; Invitrogen; Thermo Fisher Scientific, Inc.) for 24 h at 37°C. Following 24-h incubation, cells were collected and luciferase activity was detected using the Dual-Luciferase Assay system (Promega Corporation, Madison, WI, USA). Firefly luciferase activity was normalized to Renilla luciferase activity.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

RT-qPCR was used to detect the expression of miR-338-3p in CRC and adjacent normal tissue samples. Total RNA was extracted from tissue samples using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Total RNA was reverse transcribed into cDNA using a PrimeScript™ II. 1st strand DNA Synthesis kit (cat. no. 6210A; Takara Bio, Inc., Otsu, Japan). qPCR was subsequently performed using SYBR® Green PCR Master mix (Takara Bio, Inc.) with the FQ-PCR detection system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The following primer pairs were used for the qPCR: miR-338-3p forward, 5′-GGTCCAGCATCAGTGA-3′ and reverse, 5′-GAGCAGGCTGGAGAA-3′; and U6 forward, 5′-CTCGCTTCGGCAGCACATA-3′ and reverse, 5′-CGCTTCACGAATTTGCGTG-3′. The following thermocycling conditions were used for the qPCR: Initial denaturation at 94°C for 4 min; 40 cycles of 95°C for 20 sec, 57°C for 30 sec and 72°C for 30 sec. miR-338-3p levels were quantified using the 2−ΔΔCq method (22) and normalized to the internal reference gene U6.

Western blot analysis

Total protein was extracted from SW480 cells following 48 h transfection with controls, miR-338-3p inhibitors and miR-338-3p mimics using radioimmunoprecipitation assay buffer. Total protein was quantified using a bicinchoninic acid assay and 50 µg protein/lane was separated via SDS-PAGE on a 15% gel. The separated proteins were subsequently transferred onto polyvinylidene difluoride membranes and blocked for 1 h at 37°C with the 5% skimmed milk (Sangon Biotech Co., Ltd., Shanghai, China). Membranes were then incubated with primary antibodies against MACC1 (1:1,000; cat. no. ab106579) and GAPDH (1:2,000; cat. no. ab9485; both Abcam, Cambridge, UK) overnight at 4°C. Following primary incubation, membranes were incubated with horse radish peroxidase (HRP)-labeled goat anti-rabbit IgG secondary antibody (1:5,000; cat. no. AQ132P; Sigma-Aldrich; Merck KGaA) for 1 h at 37°C. Protein bands were visualized using the Pierce™ ECL Western Blotting Substrate (cat. no. 32109; Pierce; Thermo Fisher Scientific Inc.). Protein expression was analyzed using Labworks™ Analaysis software (version 4.0; UVP, Upland, CA, USA) with GAPDH as the loading control.

Immunohistochemistry

Tissue samples were fixed in 4% paraformaldehyde (Sigma-Aldrich; Merck KGaA) at 37°C for 30 min and embedded in paraffin. Paraffin-embedded tissue samples were cut into 3–5-µm thick sections. The tissue sections were deparaffinized in xylene at 55°C and rehydrated in a descending alcohol series. Deparaffinized tissue sections were blocked for 1 h at room temperature with 10% FBS and incubated with 3% H2O2 (Sangon Biotech Co., Ltd.) for 10 min at 37°C. Tissue sections were washed in triplicate with distilled water for 2 min. Tissue sections were incubated with primary antibody against MACC1 (1:1,000; cat. no. ab106579; Abcam) overnight at 4°C. Subsequently, the tissues were incubated with HRP-labeled goat anti-rabbit IgG secondary antibody (1:2,000; cat. no. AQ132P; Sigma-Aldrich; Merck KGaA) for 1 h at 37°C. Tissue sections were subsequently stained with 3, 3′-diaminobenzidine (DAB) solution for color development at 37°C for 5 min and observed under a light microscope (magnification, ×200; BX51; Olympus, Tokyo, Japan). DAB precipitates as a dark brown pigment allowing for the visualization of positively stained cells. PBS alone was used as a negative control. Two independent pathologists blindly analyzed MACC1 staining. The percentage of positively stained cells was quantified as follows: 0 (no positive cells), 1 (<10% positive cells), 2 (10–50% positive cells) and 3 (>50% positive cells). Staining intensity was graded as follows: 0 (no color), 1 (light yellow), 2 (claybank) and 3 (sepia). The two scores were multiplied and MACC1 expression was determined according to the overall score whereby: ≤4 indicates negative expression and ≥6 indicates positive expression.

Statistical analysis

Data presented as the mean ± standard deviation. All statistical analyses were performed using SPSS software (version 13.0; SPSS, Inc., Chicago, IL, USA). A Student's t-test was used to analyze differences between two groups. One-way analysis of variance followed by followed by a Bonferroni post-hoc test was used to analyze differences among multiple groups. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-338-3p expression is decreased in CRC tissue samples

To assess the function of miR-338-3p in CRC progression, miR-338-3p expression in human CRC and adjacent normal tissue samples was determined via RT-qPCR. The results demonstrated that miR-338-3p expression was significantly decreased in the CRC tissue compared with adjacent normal tissue samples (P<0.001; Fig. 1).

miR-338-3p regulates SW480 cell proliferation, apoptosis and migration

SW480 cell proliferation was examined following transfection with controls, miR-338-3p mimics or miR-338-3p inhibitors. The miR-338-3p inhibitor significantly enhanced cell proliferation, whilst the miR-338-3p mimic significantly inhibited cell proliferation compared with the control group (P<0.01; Fig. 2). Flow cytometry revealed that the miR-338-3p inhibitor had no significant effect on cell apoptosis, whilst the miR-338-3p mimic significantly enhanced cell apoptosis (P<0.01; Fig. 3). Furthermore, miR-338-3p had a significant effect on SW480 cell cycle. The miR-338-3p inhibitor significantly increased the proportion of cells in S phase and significantly decreased the proportion of cells in G1/G2 phase compared with the control group (P<0.0001; Fig. 4). However, the miR-338-3p mimic significantly decreased the proportion of cells in S phase and significantly increased the proportion of cells in G1/G2 phase compared with the control group (P<0.0001; Fig. 4). Additionally, the miR-338-3p inhibitor significantly enhanced cell migration, whilst the miR-338-3p mimic significantly suppressed cell migration compared with the control group (P<0.01; Fig. 5), suggesting that miR-338-3p may regulate the migration of the CRC cell line SW480. Taken together, these results indicate that miR-338-3p may suppress the aggressive clinicopathological features associated with CRC.

MACC1 is a direct target of miR-338-3p

To further assess the role of miR-338-3p in human CRC, potential targets of miR-338-3p were determined. TargetScanHuman bioinformatics software was used to identify MACC1 as a putative target gene of miR-338-3p (Fig. 6A). The luciferase assay revealed that miR-338-3p significantly reduced wt MACC1 expression compared with the NC group (P<0.01; Fig. 6B). In addition, miR-338-3p had no effect on mut MACC1 expression compared with the NC group (Fig. 6B). However, the positive expression rate of MACC1 was increased in CRC tissue compared with the normal adjacent tissue samples (Fig. 6C). Among the 15 CRC tissue samples, 10 (66.7%) were positive for MACC1 expression, whilst in the 15 adjacent normal tissue samples, 3 (20.0%) were positive for MACC1 expression (data not shown). The current study demonstrated that the miR-338-3p inhibitor upregulated the level of MACC1 protein expression compared with the control group (Fig. 6D). These results indicate that MACC1 is a direct target of miR-338-3p.

MACC1 silencing inhibits SW480 cell proliferation, migration and induces cell apoptosis

To further investigate the role of MACC1 in CRC, SW480 cell growth was examined following transfection with MACC1 siRNA. The current study demonstrated that MACC1 siRNA significantly decreased the level of MACC1 protein expression compared with the control SW480 group (P<0.01; Fig. 7A). Cell proliferation rate was significantly reduced (P<0.05; Fig. 7B), whilst cell apoptosis was significantly enhanced (P<0.05; Fig. 7C) in SW480 cells following transfection with MACC1 siRNA compared with the control SW480 group. Furthermore, cell migration was significantly reduced in SW480 cells following transfection with MACC1 siRNA compared with the control SW480 group (P<0.05; Fig. 7D).

Discussion

miR-338-3p has been reported to be involved in the progression and development of various types of cancer (914,17,18,21). However the cellular function of miR-338-3and its molecular mechanism in CRC remains unknown. The aim of the current study was to assess the cellular function of miR-338-3p in CRC, the malignant behavior of CRC cells and the interaction between miR-338-3p and MACC1. The current study demonstrated that miR-338-3p expression was significantly downregulated in CRC tissues. In CRC, miR-338-3p may act as a tumor suppressor, inhibiting CRC progression by targeting MACC1. miR-338-3p may therefore be a potential novel target in the treatment of CRC.

miR-338-3p has been associated with tumor development in various types of cancer. Huang et al (11) demonstrated that miR-338-3p inhibits EMT in gastric cancer by targeting ZEB2 and the MACC1/EMT/serine/threonine kinase 1 (AKT) signaling pathway. Another study demonstrated that miR-338-3p suppressed gastric cancer progression by targeting phosphatidylinositol 3,4,5-trisphosphate Rac exchanger 2a and inhibiting the phosphatase and tensin homolog (PTEN)-AKT pathway (17). In normal glioblastoma, miR-338-3p regulates the malignant biological behavior of glioma cells via MACC1 (9,12). Wang et al (14) demonstrated that miR-338-3p regulates the survival of hepatocellular carcinoma cells by downregulating forkhead box P4. Hua et al (21) revealed that miR338-3p regulates cervical cancer progression by targeting MACC1 via the MAPK signaling pathway. In ovarian cancer, miR-338-3p suppresses tumor growth by targeting runt related transcription factor 2 (Runx2) (18). In renal cell carcinoma, transforming growth factor beta receptor 1 is downregulated by miR-338-3p (10). The current study indicated that miR-338-3p overexpression inhibited CRC cell proliferation and induced cell cycle arrest and apoptosis. These results suggest that miR-338-3p may be involved in CRC progression and development by acting as a tumor suppressor in CRC. Tang et al (23) revealed that cell proliferation and clone formation, as well as cell invasion and migration capabilities of colon cancer cells were significantly increased following miR-338-3p overexpression. Furthermore, decreased cell proliferation, smaller and fewer clones, as well as weakened cell migratory and invasive capabilities were observed in SW1116 and HCT116 cells treated with MACC1 siRNA. In addition, Sun et al (24) demonstrated that the downregulation of miR338-3p in CRC is associated with poor prognosis. Previous studies have also demonstrated that the effects of miR-338-3p in tumor progression and development are mediated through several pathways, including MET/AKT, PTEN/AKT, Runx2 and MAPK (25,26). However, whether these pathways are tissue-specific remains unknown.

Previous in vitro and in vivo studies have demonstrated that MACC1 is a transduction molecule of the hepatocyte growth factor (HGF)/MET signaling pathway and that MACC1 stimulates the proliferation and motility of CRC cells, accelerating metastatic spread (19,23,2729). HGF is a ligand that, following binding with MET on the cell membrane, activates downstream signals that positively regulate the proliferation, invasion, vascularization and EMT of tumor cells (23,28,29). The HGF/MET signaling pathway is important in CRC (30). In addition, MACC1 contributes to the basolateral polarity of epithelial cells, which indicates a potential role of MACC1 activation in tumor neovascularization (13). These results indicate that miR-338-3p inhibits the development of aggressive CRC features by directly targeting the MACC1 3′-UTR. In the current study, the level of MACC1 protein expression was significantly higher in CRC tissue compared with adjacent normal tissue samples. In addition, Wang et al (27) demonstrated that MACC1 overexpression was closely associated with survival in solid tumors. Taken together, these results indicate that MACC1 serves a role in the progression and development of CRC. A previous study has reported that miR338-3p regulates cervical cancer cell growth by targeting MACC1 gene expression (21). The current study demonstrated that miR-338-3p serves a role in CRC cell proliferation and further verified the effects of MACC1 silencing in CRC cells.

The present study has several limitations. A relatively small number of CRC tissue samples were utilized and only one CRC cell line was assessed. Therefore, further studies using several CRC cell lines that exhibit a different level of cellular tumoral aggressiveness is required to determine the potential pathways involved in CRC. In addition, the underlying mechanism of miR-338-3p in CRC was not assessed in the current study. Therefore, a comprehensive analysis of the main factors associated with the MET/AKT, PTEN/AKT, Runx2 and MAPK signaling pathways should be undertaken.

In conclusion, miR-338-3p expression is significantly decreased whilst MACC1 expression is significantly increased in CRC tissues compared with adjacent normal tissue samples. MACC1 is a direct target gene of miR-338-3p. miR-338-3p overexpression suppresses CRC cell proliferation and migration, and induces apoptosis. These results indicate that miR-338-3p may regulate CRC progression via the direct targeting of MACC1. miR-338-3p/MACC1 may therefore be a potential therapeutic target for patients with CRC.

Acknowledgements

Not applicable.

Funding

The present study was supported by grants obtained from the Application and Basic Research of Yunnan Province (grant no. 2015FB2015), the Hundred Young and Middle-aged Academic and Technical Backbone Project of Kunming Medical University (grant no. 60117190431) and Yunsheng Yang Work station (grant no. 2017IC028).

Availability of data and materials

All datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

ML, HH, YF, JY and JL made substantial contributions to the conception and design of the current study. ML designed the experiments. GZ, WL, XL, GL and LW performed the experiments. BS and CZ analyzed the data; ML prepared the manuscript. ML and YF made manuscript revisions. All authors reviewed the results and approved the final manuscript.

Ethics approval and consent to participate

The study was approved by the Ethics Committee of the Second Affiliated Hospital of Kunming Medical University (Kunming, China) and all patients provided written informed consent.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CRC

colorectal cancer

EMT

epithelial- mesenchymal transition

MACC1

metastasis-associated in colon cancer-1

miRNA

microRNA

References

1 

Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global patterns and trends in colorectal cancer incidence and mortality. Gut. 66:683–691. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Liu S, Zheng R, Zhang M, Zhang S, Sun X and Chen W: Incidence and mortality of colorectal cancer in China, 2011. Chin J Cancer Res. 27:22–28. 2015.PubMed/NCBI

4 

Marley AR and Nan H: Epidemiology of colorectal cancer. Int J Mol Epidemiol Genet. 7:105–114. 2016.PubMed/NCBI

5 

Steeg PS: Tumor metastasis: Mechanistic insights and clinical challenges. Nat Med. 12:895–904. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Slaby O, Svoboda M, Michalek J and Vyzula R: MicroRNAs in colorectal cancer: Translation of molecular biology into clinical application. Mol Cancer. 8:1022009. View Article : Google Scholar : PubMed/NCBI

7 

Sun K, Wang W, Zeng JJ, Wu CT, Lei ST and Li GX: MicroRNA-221 inhibits CDKN1C/p57 expression in human colorectal carcinoma. Acta Pharmacol Sin. 32:375–384. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Sassen S, Miska EA and Caldas C: MicroRNA: Implications for cancer. Virchows Arch. 452:1–10. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Howe JR VIth, Li ES, Streeter SE, Rahme GJ, Chipumuro E, Russo GB, Litzky JF, Hills LB, Rodgers KR, Skelton PD and Luikart BW: MiR-338-3p regulates neuronal maturation and suppresses glioblastoma proliferation. PLoS One. 12:e01776612017. View Article : Google Scholar : PubMed/NCBI

10 

Zhang X, Wang C, Li H, Niu X, Liu X, Pei D, Guo X, Xu X and Li Y: miR-338-3p inhibits the invasion of renal cell carcinoma by downregulation of ALK5. Oncotarget. 8:64106–64113. 2017.PubMed/NCBI

11 

Huang N, Wu Z, Lin L, Zhou M, Wang L, Ma H, Xia J, Bin J, Liao Y and Liao W: MiR-338-3p inhibits epithelial-mesenchymal transition in gastric cancer cells by targeting ZEB2 and MACC1/Met/Akt signaling. Oncotarget. 6:15222–15234. 2015.PubMed/NCBI

12 

Shang C, Hong Y, Guo Y and Xue YX: Mir-338-3p inhibits malignant biological behaviors of glioma cells by targeting MACC1 gene. Med Sci Monit. 22:710–716. 2016.PubMed/NCBI

13 

Tsuchiya S, Oku M, Imanaka Y, Kunimoto R, Okuno Y, Terasawa K, Sato F, Tsujimoto G and Shimizu K: MicroRNA-338-3p and microRNA-451 contribute to the formation of basolateral polarity in epithelial cells. Nucleic Acids Res. 37:3821–3827. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Wang G and Sun Y, He Y, Ji C, Hu B and Sun Y: MicroRNA-338-3p inhibits cell proliferation in hepatocellular carcinoma by target forkhead box P4 (FOXP4). Int J Clin Exp Pathol. 8:337–344. 2015.PubMed/NCBI

15 

Kelemen LE, Wang X, Fredericksen ZS, Pankratz VS, Pharoah PD, Ahmed S, Dunning AM, Easton DF, Vierkant RA, Cerhan JR, et al: Genetic variation in the chromosome 17q23 amplicon and breast cancer risk. Cancer Epidemiol Biomarkers Prev. 18:1864–1868. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Hirata Y, Murai N, Yanaihara N, Saito M, Saito M, Urashima M, Murakami Y, Matsufuji S and Okamoto A: MicroRNA-21 is a candidate driver gene for 17q23-25 amplification in ovarian clear cell carcinoma. BMC Cancer. 14:7992014. View Article : Google Scholar : PubMed/NCBI

17 

Guo B, Liu L, Yao J, Ma R, Chang D, Li Z, Song T and Huang C: miR-338-3p suppresses gastric cancer progression through a PTEN-AKT axis by targeting P-REX2a. Mol Cancer Res. 12:313–321. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Wen C, Liu X, Ma H, Zhang W and Li H: miR-338-3p suppresses tumor growth of ovarian epithelial carcinoma by targeting Runx2. Int J Oncol. 46:2277–2285. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Stein U, Walther W, Arlt F, Schwabe H, Smith J, Fichtner I, Birchmeier W and Schlag PM: MACC1, a newly identified key regulator of HGF-MET signaling, predicts colon cancer metastasis. Nat Med. 15:59–67. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Dunlevy JR, Berryhill BL, Vergnes JP, SundarRaj N and Hassell JR: Cloning, chromosomal localization, and characterization of cDNA from a novel gene, SH3BP4, expressed by human corneal fibroblasts. Genomics. 62:519–524. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Hua FF, Liu SS, Zhu LH, Wang YH, Liang X, Ma N and Shi HR: MiRNA-338-3p regulates cervical cancer cells proliferation by targeting MACC1 through MAPK signaling pathway. Eur Rev Med Pharmacol Sci. 21:5342–5352. 2017.PubMed/NCBI

22 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Tang J, Chen JX, Chen L, Tang JY, Cui Z, Liu CH and Wang Z: Metastasis associated in colon cancer 1 (MACC1) promotes growth and metastasis processes of colon cancer cells. Eur Rev Med Pharmacol Sci. 20:2825–2834. 2016.PubMed/NCBI

24 

Sun K, Su G, Deng H, Dong J, Lei S and Li G: Relationship between miRNA-338-3p expression and progression and prognosis of human colorectal carcinoma. Chin Med J (Engl). 127:1884–1890. 2014.PubMed/NCBI

25 

Cohen-Solal KA, Boregowda RK and Lasfar A: RUNX2 and the PI3K/AKT axis reciprocal activation as a driving force for tumor progression. Mol Cancer. 14:1372015. View Article : Google Scholar : PubMed/NCBI

26 

Walker CL, Liu NK and Xu XM: PTEN/PI3K and MAPK signaling in protection and pathology following CNS injuries. Front Biol (Beijing). 8:2013.doi: 10.1007/s11515-013-1255-1. PubMed/NCBI

27 

Wang G, Fu Z and Li D: MACC1 overexpression and survival in solid tumors: A meta-analysis. Tumour Biol. 36:1055–1065. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Li H, Chen YX, Wen JG and Zhou HH: Metastasis-associated in colon cancer 1: A promising biomarker for the metastasis and prognosis of colorectal cancer. Oncol Lett. 14:3899–3908. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Ashktorab H, Hermann P, Nouraie M, Shokrani B, Lee E, Haidary T, Brim H and Stein U: Increased MACC1 levels in tissues and blood identify colon adenoma patients at high risk. J Transl Med. 14:2152016. View Article : Google Scholar : PubMed/NCBI

30 

Birchmeier C, Birchmeier W, Gherardi E and Vande Woude GF: Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 4:915–925. 2003. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2019
Volume 17 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lu M, Huang H, Yang J, Li J, Zhao G, Li W, Li X, Liu G, Wei L, Shi B, Shi B, et al: miR‑338‑3p regulates the proliferation, apoptosis and migration of SW480 cells by targeting MACC1. Exp Ther Med 17: 2807-2814, 2019
APA
Lu, M., Huang, H., Yang, J., Li, J., Zhao, G., Li, W. ... Fu, Y. (2019). miR‑338‑3p regulates the proliferation, apoptosis and migration of SW480 cells by targeting MACC1. Experimental and Therapeutic Medicine, 17, 2807-2814. https://doi.org/10.3892/etm.2019.7260
MLA
Lu, M., Huang, H., Yang, J., Li, J., Zhao, G., Li, W., Li, X., Liu, G., Wei, L., Shi, B., Zhao, C., Fu, Y."miR‑338‑3p regulates the proliferation, apoptosis and migration of SW480 cells by targeting MACC1". Experimental and Therapeutic Medicine 17.4 (2019): 2807-2814.
Chicago
Lu, M., Huang, H., Yang, J., Li, J., Zhao, G., Li, W., Li, X., Liu, G., Wei, L., Shi, B., Zhao, C., Fu, Y."miR‑338‑3p regulates the proliferation, apoptosis and migration of SW480 cells by targeting MACC1". Experimental and Therapeutic Medicine 17, no. 4 (2019): 2807-2814. https://doi.org/10.3892/etm.2019.7260