Open Access

miR‑151a‑5p promotes the proliferation and metastasis of colorectal carcinoma cells by targeting AGMAT

  • Authors:
    • Yaya Xie
    • Yue Zhang
    • Xianju Liu
    • Lijun Cao
    • Mengting Han
    • Chunmei Wang
    • Jinlian Chen
    • Xingxing Zhang
  • View Affiliations

  • Published online on: January 20, 2023     https://doi.org/10.3892/or.2023.8487
  • Article Number: 50
  • Copyright: © Xie et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Colorectal carcinoma (CRC) is one of the most common types of digestive cancer. It has been reported that the ectopic expression of microRNAs (miRs) plays a critical role in the occurrence and progression of CRC. In addition, it has also been suggested that miR‑151a‑5p may serve as a useful biomarker for the early detection and treatment of different types of cancer and particularly CRC. However, the specific effects and underlying mechanisms of miR‑151a‑5p in CRC remain elusive. The results of the current study demonstrated that miR‑151a‑5p was upregulated in CRC cell lines and clinical tissues derived from patients with CRC. Functionally, the results showed that miR‑151a‑5p significantly promoted CRC cell proliferation, migration and invasion. Additionally, dual luciferase reporter assays verified that agmatinase (AGMAT) was a direct target of miR‑151a‑5p and it was positively associated with miR‑151a‑5p expression. Mechanistically, miR‑151a‑5p could enhance the epithelial‑mesenchymal transition of CRC cells. Taken together, the results of the current study revealed a novel molecular mechanism indicating that the miR‑151a‑5p/AGMAT axis could serve a crucial role in the regulation of CRC and could therefore be considered as a potential therapeutic strategy for CRC.

Introduction

Colorectal carcinoma (CRC) is one of the most common types of malignant tumors, ranking third in newly diagnosed cancer cases and cancer-related deaths, preceded by lung cancer and breast cancer in humans, thus seriously endangering the safety and lives of humans (1). In recent years, due to the rapid development of science and technology, and changes in people's lifestyles, there is a substantial increasing trend of CRC incidence in younger population. Although progress has been made in revealing the molecular mechanism and treatment of CRC, its prognosis remains very poor, possibly due to the rapid progression and development of CRC, chemoresistance and lack of effective screening and treatment approaches (24). Therefore, identifying specific diagnostic biomarkers and effective therapeutic targets for CRC is of great importance.

Agmatinase (AGMAT), located in mitochondria, is a key enzyme involved in polyamine metabolism pathway and is mainly distributed in the kidneys and liver (5). The polyamine metabolic disorder is commonly caused by the abnormal expression of enzymes involved in the metabolic pathway. Therefore, the abnormal expression of AGMAT could promote the occurrence and development of cancer via affecting polyamine metabolism. A previous study showed that AGMAT was highly expressed in lung adenocarcinoma tissues and it was closely associated with tumor stage and prognosis in patient with lung adenocarcinoma; furthermore, the study also demonstrated that AGMAT promoted inducible nitric oxide (NO) synthase expression via activating the MAPK and PI3K/Akt signaling pathways and inducing NO release to enhance lung cancer cell invasion and metastasis in vitro (6). Additionally, AGMAT could promote pancreatic cancer cell proliferation, invasion and metastasis via the TGF-β/Smad pathway (7). Although AGMAT plays an essential role in the metabolism of amino acids, its effects on CRC have not been previously investigated.

With the discovery of microRNAs (miRNAs or miRs), their role in the onset and progression of CRC has become a significant area of interest (8). Increasing evidence has suggested that the dysregulation of miRNAs is involved in the development of several types of cancer, including CRC (9). miRNAs also play a significant role in different biological and cellular processes, such as in cell proliferation, differentiation, apoptosis, death and metastasis (1013). miR-151a-5p is a well-characterized oncogenic miRNA, which is dysregulated in CRC and is therefore recognized as a promising biomarker for the early detection and treatment of CRC (14). As a member of the miR-151 family, miR-151a-5p is a small non-coding RNA molecule that acts as a proto-oncogene and is expressed in several types of cancer, such as lung cancer (15), prostate cancer (16) and lymphoblastic leukemia (17). It has been reported that miR-151a-5p is involved in numerous biological processes. For instance, a previous study suggested that miR-151a-5p could be involved in the regulation of cell respiration and ATP production in mitochondria via targeting cytochrome B (18). As a member of housekeeping miRNAs, miR-151a-5p was identified to be stably expressed in endothelial cells and macrophages in an inflammatory setting (19). Furthermore, Guo et al (20) showed that miR-151a-5p was upregulated in lung cancer, while miR-151a-5p silencing could inhibit the proliferation, invasion and migration, and induce the apoptosis of A549 lung cancer cells. These findings suggested that miR-151a-5p could be a potential therapeutic target for several types of cancer. Although miR-151a-5p could be used as a potential non-invasive serological diagnostic marker for the detection of CRC, the particular effects and mechanisms of miR-151a-5p in CRC remain unclear (14).

Therefore, the present study aimed to explore the effect and molecular mechanisms of miR-151a-5p in CRC in vitro.

Materials and methods

Datasets analysis

Co-expression analyses of the interaction between miR-151a-5p and AGMAT were downloaded from the starbase database (http://starbase.sysu.edu.cn/), including COAD samples (n=450). The potential binding sites between miR-151a-5p and AGMAT were downloaded from the miRmap database (http://www.mirmap.ezlab.org). The expression levels of miR-151a-5p in tumor tissues (n=254) were directly compared with those in adjacent normal tissues (n=8) using the expression data from ~32 common types of cancer downloaded from the Gene Expression Display Server (http://bioinfo.life.hust.edu.cn/web/GEDS/). Additionally, the expression levels of miR-151a-5p in tumor tissues (n=445) were also directly compared with those in adjacent normal tissues (n=8) downloaded from the UCSC Xena database (http://xena.ucsc.edu/).

Cell lines and culture

The human CRC cell lines HCT8, HCT116, HT29, SW480, SW620, LS174T and DLD1 were purchased from the American Type Culture Collection. In addition, the normal human colorectal cells NCM460 and 293T cells were conserved in the central laboratory of Shanghai Fengxian District Central Hospital. All cell lines were cultured in DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (both from Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin/streptomycin solution in a humidified incubator at 37°C with 5% CO2.

Plasmids and retroviral infection

The full-length sequence of the human AGMAT gene was subcloned into the pCD513B vector (Geneppl Technology Co., Ltd.) and transfected to generate cells stably overexpressing AGMAT. To stably knock down AGMAT in CRC cells, small hairpin RNAs (shRNAs) targeting AGMAT were synthesized and subcloned into the lentiviral pPLK-GFP + Puro vector (Geneppl Technology Co., Ltd.). The sequences of the shRNA clones used were listed in Table I. The concentrations of shRNA clones used were as follows: NC, 964 ng/µl; shAGMAT#1, 1,142 ng/µl; and shAGMAT#3, 795 ng/µl. The mixture of 1 ml DMEM, 10 µg DNA and 10 µl (µg) GM easy™ Lentivirus mix (Genomeditech) was co-transfected into 293T cells using 60 µl Hg transgene™ reagent (Genomeditech), according to the manufacturer's instructions (cat. no. GMeasy 40; Genomeditech). The lentiviral particles were collected at 48 h following cell transduction.

Table I.

The sequences of specific primers used for shRNA and miR-151a-5p.

Table I.

The sequences of specific primers used for shRNA and miR-151a-5p.

PrimerSequence (5′→3′)
shAGMAT#1 CGATGTGAATGTCAATCTTTA
shAGMAT#3 CGGGAAGAATCAGTGATGCTT
NC GTTCTCCGAACGTGTCACGTT
miR-151a-5p UUGUACUACACAAAAGUACUG
mimic NC
miR-151a-5p mimics UCGAGGAGCUCACAGUCUAGU
miR-151a-5p CAGUACUUUUGUGUAGUACAA
inhibitor NC
miR-151a-5p inhibitor ACUAGACUGUGAGCUCCUCGA

[i] sh-, short hairpin; NC, negative control; miR, microRNA.

Generation of stable cell lines and miR-151a-5p transfection

When the cell density of ~80–90%, CRC cells seeded into 6-cm culture-plates were infected with filtered lentiviral particles (the 2nd generation system) plus 8 µg/ml Polybrene (Sigma-Aldrich; Merck KGaA) for 24 h. Following transduction, cells were treated with 0.5 µg/ml puromycin (Invitrogen; Thermo Fisher Scientific, Inc.) for >7 days to obtain stably transfected cells; afterwards, the stably transfected cells were maintained with 0.3 µg/ml puromycin. Subsequently, when reached ~60-70% confluency in the cell culture plate, HCT116 cells, SW480 cells and stably AGMAT expressing CRC cells were treated with a mixture containing 5 µl Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), 100 pmol miR-151a-5p and control oligonucleotides, according to the manufacturer's instructions. Following transfection for 48 h, the relative expression levels of miR-151a-5p were determined by reverse transcription-quantitative PCR (RT-qPCR). The sequences of miR-151a-5p mimics or inhibitor used were listed in Table I (Genomeditech).

Dual luciferase reporter assay

The luciferase reporter constructs were generated by inserting the wild-type (WT) or mutant (MUT) 3′ untranslated region (3′ UTR) of AGMAT into the pGL3 luciferase vector (Geneppl Technology Co., Ltd.). Subsequently, the stable AGMAT overexpressing CRC cells were co-transfected with 2 µg WT or MUT AGMAT-3′ UTR, Renilla Luc, miR-151a-5p mimics or miR-NC using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions (Shanghai Yeasen Biotechnology Co., Ltd.). Following transfection for 48 h, the firefly and Renilla luciferase activities were determined by spectrophotometry using the Dual Luciferase Reporter Assay System (Glomax 96; Promega Corporation). The luciferase activity was then calculated. The results are expressed as the ratio of firefly luciferase activity to Renilla luciferase activity.

Cell counting kit-8 (CCK-8) assay

The cell proliferation ability was assessed using CCK-8 assay (Dojindo Molecular Technologies, Inc.), according to the manufacturer's protocol. Briefly, treated CRC cell lines were seeded into a 96-well plate at a density of 103 cells/well in 100 µl DMEM supplemented with 10% FBS and were then cultured for 10, 24, 48 and 72 h at 37°C with 5% CO2. Subsequently, cells were supplemented with 10 µl CCK-8 solution, followed by incubation for an additional 1 h in the dark. Finally, the optical density (OD) of each well was measured at a wavelength of 450 nm using a microplate reader (BioTek Instruments, Inc.).

Colony formation assay

For colony formation assays, treated CRC cells were seeded into a 12-well plate at a density of 3×103 cells/well and cultured in DMEM supplemented with 10% FBS for 7 or 10 days. The formed colonies were fixed with 4% paraformaldehyde at room temperature for 30 min and were then stained with 0.1% crystal violet at room temperature for 1 h followed by washing with ddH2O. Images of the formed colonies were captured under an inverted light microscope (Olympus Corporation). Finally, the number of colony cells (>50 cells per cell colony) were counted using ImageJ software (version 1.52a; National Institutes of Health).

Cell migration and invasion assay

The migration and invasion abilities of HCT116 and SW480 cells transfected with the aforementioned plasmids/miR mimics/inhibitors for 48 h were evaluated using Transwell chambers (0.8 µm; cat. no. 353097; Corning, Inc.). Briefly, treated HCT116 and SW480 cells at a density of 105 cells/well were added to the upper chamber of the Transwell chamber coated or not with Matrigel (0.8 µm; cat. no. 354480; Corning, Inc.). The upper chambers of the Transwell chamber with Matrigel were precoated with 500 µl medium without serum for 1 h at 37°C with 5% CO2. For migration assays, the lower chamber was supplemented with 600 µl medium with 10% FBS, cells were cultured for 48 h, while for the invasion assays, the lower chamber was supplemented with 600 µl medium with 20% FBS, cells were cultured for 24 h, the upper chamber was supplemented without serum. The migratory or invasive cells were fixed with 4% paraformaldehyde at room temperature for 30 min and were then stained with 0.1% crystal violet at room temperature for 1 h followed by ddH2O washing. Finally, migratory or invasive cells were observed and images were captured under a fluorescence microscope (magnification, ×100).

RT-qPCR analysis

Total RNA was extracted from treated cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). The nuclear and cytoplasmic extracts were isolated using the cytoplasmic and nuclear RNA purification kit (cat. no. 21000; Norgen Biotek Corp.). Subsequently, 1 µg RNA was reverse transcribed into cDNA using the Evo M-MLV RT Mix kit with gDNA Clean for qPCR (cat. no. AG11728; Accurate Bio-Medical Technology Co., Ltd.) and the miRNA 1st strand cDNA synthesis kit (by stem-loop) (cat. no. MR101-02; Vazyme Biotech Co., Ltd.). qPCR was performed on the quantum Studio 6 real-time PCR system (Thermo Fisher Scientific, Inc.) using the SYBR Green premix Pro Taq HS qPCR kit (Rox plus) (cat. no. AG1718; Accurate Bio-Medical Technology Co., Ltd.). The thermocycling conditions used for reverse transcription PCR were as follows: For gDNA clean, 42°C 2 min, 4°C; for synthesis of first strand cDNA of miR-151a-5p, 25°C for 5 min, 50°C for 15 min, 85°C for 5 min; for synthesis of common primer cDNA, 37°C for 15 min, 85°C for 5 min, 4°C. The thermocycling conditions used for qPCR were as follows: For miR-151a-5p, 95°C for 5 min, 95°C 10 sec, 60°C for 5 min; for common primers, 95°C for 30 sec, 95°C for 5 sec, 60°C for 30 sec. The relative mRNA expression levels of the target genes were calculated using the 2−ΔΔCq method (21). GAPDH served as an internal reference gene for AGMAT. The primers for U1 were purchased from Guangzhou RiboBio Co., Ltd. The primer sequences used are listed in Table II (Sangon Biotech Co., Ltd.).

Table II.

The sequences of specific primers used for reverse transcription-quantitative PCR.

Table II.

The sequences of specific primers used for reverse transcription-quantitative PCR.

Gene nameSequence (5′→3′)
GAPDHF: TTGGTATCGTGGAAGGACTCA
R: TGTCATCATATTTGGCAGGTT
microRNA-151a-5pF: CGCGTCGAGGAGCTCACAG
R: AGTGCAGGGTCCGAGGTATT
AGMATF: CTTGTCGAAGTTTCACCACCGTA
R: CTTTGGGGAGAGCACATAGCATC
AZIN2F: CAACTCAGCCTTGGACCTGTACTTC
R: CTGCTCCGTGGATGGTTTCTTCTG
AMD1F: CCCGACGCAAACCAAGGATCTG
R: TTCAACAGGGGAACCAGTGCTTTC
ODC1F: CACTGTTGCTGCTGCCTCTACG
R: GGTTCTGGAATTGCTGCATGAGTTG
OAZ1F: TCTCCCTCCACTGCTGTAGTAACC
R: TGACTATTCCCTCGCCCACCTG
SRMF: GATGATCGCCAACCTGCCTCTC
R: ATCTCACACTGGACCACGGACTC
SMSF: TTGGCAGAGAGTGATTTGGCATATACC
R: CCACCTCCCAGAATGAGTACATCTTTG
SAT1F: TGGTTGCAGAAGTGCCGAAAGAG
R: ATAACTTGCCAATCCACGGGTCATAG
SMOXF: CACACCCTCACCTACCCACCTG
R: CACTGCCTCGTCATCACACTTCTC
PMF1F: GCGATGACACAGCAAATCTATGACAAG
R: AGGCATTCAAGACAGCTTCTAGGTTC
TP53F: GCCCATCCTCACCATCATCACAC
R: GCACAAACACGCACCTCAAAGC
PAOXF: TTCCAGTGTCGGTAGAGTGTGAGG
R: ATCTTCCTGATTGCTTCTGCCTTCTC

[i] AGMAT, agmatinase; F, forward; R, reverse.

Immunofluorescence assay

The transfected cells were cultured in 48-well plates at a density of 6×104 cells/well for 24 h. Following fixation with 4% paraformaldehyde at room temperature for 30 min, the cells were permeabilized with 1% Triton X-100 at room temperature for 15 min followed by blocking with 3% BSA (cat. no. 4240GR100; Biofroxx; neoFroxx) at 37°C for 1 h. After cell incubation with primary antibodies at 4°C overnight, the cells were treated with the corresponding secondary Goat anti-Rabbit IgG (H + L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor™ 568 (Thermo Fisher Scientific, Inc.) at room temperature for 30 min in the dark. Nuclei were counterstained with DAPI (5 mg/ml; cat. no. C1006; Beyotime Institute of Biotechnology) at room temperature for 20 min in the dark. The stained cells were observed under a fluorescence microscope (magnification, ×200). The antibodies used for immunofluorescence are listed in Table III.

Table III.

Antibodies used for immunofluorescent staining.

Table III.

Antibodies used for immunofluorescent staining.

TargetSupplierCat. no.DilutionIsotypeHost species
E-cadherinProteintech Group, Inc.20874-1-AP1:200IgGRabbit
VimentinProteintech Group, Inc.10366-1-AP1:200IgGRabbit
Western blot analysis

Total proteins were extracted from treated CRC cells using RIPA lysis buffer (New cell & Molecular Biotech Co., Ltd.) supplemented with phosphatase inhibitors (Beyotime Institute of Biotechnology) and protein concentration was measured with the BCA Protein Assay Kit (cat. no. P0012; Beyotime Institute of Biotechnology). Subsequently, the proteins (10–20 µl/lane, 25 mg/ml) were separated by 10% SDS-PAGE (New cell & Molecular Biotech Co., Ltd.) and were then transferred onto a nitrocellulose membrane (Pall Corporation). Following blocking with 5% skimmed milk (Beyotime Institute of Biotechnology) at room temperature for 1 h, the membrane was incubated with primary antibodies at 4°C overnight. After incubation with the corresponding secondary antibodies at room temperature for 1 h, the immunoreactive bands were visualized using an ECL Kit (Vazyme Biotech Co., Ltd.) and images were captured under the Tanon 4600 system (Tanon Science and Technology Co., Ltd.). The density of each band was measured using a computer-assisted imaging analysis system (Adobe Systems Inc.). The expression levels of the target proteins were normalized to those of β-tubulin. The following primary and secondary antibodies were used: Anti-AGMAT, anti-MMP2, anti-MMP9, anti-N-cadherin, anti-E-cadherin, anti-Vimentin, anti-β-catenin, HRP-conjugated Affinipure Goat anti-Mouse IgG (H+L) and HRP-conjugated Affinipure Goat anti-Ribbit IgG (H+L) (Table IV). The protein expression levels were assessed using ImageJ software (National Institutes of Health).

Table IV.

Antibodies used for western blot analyses.

Table IV.

Antibodies used for western blot analyses.

Primary antibodies

TargetSupplierCat. no.DilutionIsotypeHost speciesMW (KDa)
AGMATAbcamAb2318941:2,000IgGRabbit38
MMP2Proteintech Group, Inc.10373-2-AP1:1,000IgGRabbit60
MMP9Proteintech Group, Inc.10375-2-AP1:1,000IgGRabbit92
N-cadherinProteintech Group, Inc.22018-1-AP1:2,000IgGRabbit130
E-cadherinProteintech Group, Inc.20874-1-AP1:5,000IgGRabbit135
VimentinProteintech Group, Inc.10366-1-AP1:2,000IgGRabbit54
β-CateninProteintech Group, Inc.51067-2-AP1:5,000IgGRabbit92
β-TubulinProteintech Group, Inc.66240-1-Ig1:10,000IgG2aMouse55

Secondary antibodies

Target Suppliercat. no. Dilution

HRP-conjugated Affinipure Goat anti-mouse IgG (H+L)Proteintech Group, Inc.SA00001-11:5,000
HRP-conjugated Affinipure Goat anti-rabbit IgG (H+L)Proteintech Group, Inc.SA00001-21:5,000
Statistical analysis

All experiments were repeated at least in triplicate. Data are expressed as the mean ± SD. All statistical analyses were carried out using GraphPad Prism 8 (GraphPad Software, Inc.). The differences between two groups were compared using unpaired Student's t-test. Comparisons among multiple groups were conducted using with one- or two-way ANOVA followed by Dunnett's multiple comparisons test used as post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-151a-5p is upregulated in CRC cell lines and tissues

To explore the pathophysiological relevance of miR-151a-5p in human CRC, the expression levels of miR-151a-5p were determined in CRC and adjacent normal tissues obtained from UCSC Xena database and the Gene Expression Display Server. Therefore, compared with normal colorectal tissues, miR-151a-5p was significantly upregulated in CRC tissues (Fig. 1A and B). These data suggested that miR-151a-5p was upregulated in CRC tissues and could therefore regulate the occurrence and development of CRC via acting as a tumor-promoting gene. In addition, the expression levels of miR-151a-5p were further detected in several CRC cell lines by RT-qPCR. The analysis showed that the expression levels of miR-151a-5p were higher in CRC cell lines compared with normal colorectal cells. Among all CRC cell lines examined, SW480 cells exhibited the highest miR-151a-5p expression levels, while HCT116 cells the lowest one (Fig. 1C). Therefore, HCT116 and SW480 cells were selected for the subsequent experiments. Furthermore, the transfection efficiency of miR-151a-5p in CRC cells was also verified (Fig. 1D and E).

miR-151a-5p promotes CRC cell proliferation, migration and invasion

To further uncover the potential role of miR-151a-5p in CRC, HCT116 or SW480 cells were transfected with miR-151a-5p mimics or inhibitor, respectively. Following cell transfection for 48 h, the cell proliferation ability was assessed by CCK-8 and colony formation assays. The results showed that compared with the NC group, the change in the overexpression levels of miR-151a-5p significantly promoted the proliferation of HCT116 cells in time-dependent manner (Fig. 2A), while as predicted, the proliferation capacity of SW480 cells transfected with miR-151a-5p inhibitor was significantly inhibited (Fig. 2B). In addition, the effect of miR-151a-5p on the proliferation of HCT116 and SW480 cells was evaluated by colony formation assay. The results demonstrated that the overexpression levels of miR-151a-5p promoted the proliferation of HCT116 cells (Fig. 2C). By contrast, the proliferation of SW480 cells was significantly inhibited after transfection with miR-151a-5p inhibitor (Fig. 2D). The enhanced migration and invasion abilities of CRC cells could also serve a key role in the growth of CRC. Therefore, to further evaluate the effect of miR-151a-5p on CRC cell invasion and migration, Transwell assays coated with Matrigel or not, respectively, were used. The data revealed that compared with the corresponding NC group, the migration and invasion abilities of HCT116 cells were significantly increased following cell transfection with miR-151a-5p mimics (Fig. 2E-G). However, the opposite effect was observed in miR-151a-5p-depleted SW480 cells (Fig. 2H-J). The aforementioned findings indicated that miR-151a-5p could significantly enhance the proliferation, migration and invasion abilities of CRC cells.

miR-151a-5p enhances the epithelial-mesenchymal transition (EMT) of CRC cells

Epithelial cells are characterized by mesenchymal phenotype and changes in the expression of three significant biomarkers, including E-cadherin, vimentin and N-cadherin, eventually leading to reduced cell adhesion, loss of polarity and tight junction (22). Matrix metalloproteinases (MMPs) are proteins involved in the degradation of base membranes, thus affecting cell metastasis during the development of malignant tumors (23). Therefore, western blot analysis was performed and the result revealed that miR-151a-5p overexpression in HCT116 cells downregulated E-cadherin and upregulated N-cadherin, vimentin and β-catenin. Correspondingly, western blot analysis was also performed to evaluate the effect of miR-151a-5p on the expression levels of metastasis-related MMPs, such as MMP2 and MMP9. The results demonstrated that compared with the NC group, miR-151a-5p enhanced the protein expression levels of both MMP2 and MMP9 in HCT116 cells. By contrast, miR-151a-5p silencing in SW480 cells exhibited the opposite effect (Fig. 3A-C). To further determine the expression of EMT-related proteins in CRC cell lines, the expression of E-cadherin and vimentin was assessed by immunofluorescence analysis. The immunofluorescence analysis results demonstrated that miR-151a-5p overexpression in HCT116 cells downregulated E-cadherin and upregulated vimentin. However, miR-151a-5p silencing upregulated E-cadherin and downregulated vimentin (Fig. 3D). The aforementioned results verified that miR-151a-5p could promote EMT in CRC cells.

AGMAT is a target gene of miR-151a-5p and is positively associated with miR-151a-5p expression

Polyamines, such as putrescine, spermidine and spermine, produced during the metabolic process, are low molecular weight, aliphatic and polycationic nitrogen compounds containing two or more amino groups. Currently, the application of new technologies has increased our understanding of the genetics and potential molecular biology of polyamine function in normal and tumor cells. Recognition of the effect of the increased polyamine levels on tumor transformation and progression has deepen our understanding of the significant role of polyamines in cancer, thus providing reasonable targets for intervention (24). It has been reported that miRNAs can regulate tumor-associated proteins in CRC (25,26). However, the association between genes and miRNAs is not a one-to-one correspondence. The results of the current study showed that following miR-151a-5p overexpression or silencing in CRC cell lines, the levels of the polyamine metabolism-related metabolites and key enzymes significantly altered, while the most differences were observed in the expression levels of AGMAT at the molecular level (Fig. 4A and B). These findings indicated that miR-151a-5p could promote polyamine metabolism via regulating AGMAT. Therefore, subsequently, the present study aimed to explore whether miR-151a-5p could regulate CRC progression via targeting AGMAT. Bioinformatic analysis using the starbase database predicted that AGMAT encompassed a binding site for miR-151a-5p and revealed that AGMAT was positively associated with miR-151a-5p expression in CRC (Fig. 4C). In addition, it was further predicted using the miRmap database that the potential miR-151a-5p binding site was encompassed in AGMAT-3′ UTR (Fig. 4D). Previous studies suggested that the majority of miRNAs in the cytoplasm could act as tumor suppressors, thus resulting in gene silencing via inhibiting gene translation (27). By contrast, other studies showed that several miRNAs, located at the nucleus, could positively regulate gene transcription via binding to and activating their target enhancers (28,29). In the present study, RNA isolation from nuclear and cytoplasmic extracts revealed that miR-151a-5p was mainly localized in the nucleus, as evidenced by RT-qPCR analysis (Fig. 4E and F). To further verify whether miR-151a-5p could regulate the expression of AGMAT via binding to AGMAT-3′ UTR, a dual luciferase reporter assay was performed. As revealed in Fig. 4G, the luciferase activity of AGMAT-3′ UTR was significantly increased in cells transfected with miR-151a-5p mimics. However, miR-151a-5p overexpression showed a modest effect on luciferase activity in cells transfected with mutated putative binding site. For the convenience of follow-up tests, the stable AGMAT expression cell lines were designed, namely, the overexpression of AGMAT in HCT116 cells and the knockdown of AGMAT in SW480 cells. The transfection efficiency of AGMAT in stable transmutation cells was determined by RT-qPCR and western blot analysis. ShAGMAT#1 was selected from the AGMAT-stably knocked down SW480 cells due to the best knockdown effect (Fig. S1). Furthermore, RT-qPCR and western blot analysis were carried out to reveal the relationship between miR-151a-5p and AGMAT expression. The results identified that the mRNA expression levels of AGMAT were significantly reduced in AGMAT-stably overexpressing HCT116 cells transfected with miR-151a-5p inhibitor (Fig. 5A). However, the opposite effect was observed in the AGMAT-stably knocked down SW480 cells transfected with miR-151a-5p mimics (Fig. 5B). Western blot analysis further verified the positive regulatory association between miR-151a-5p and AGMAT (Fig. 5C and D). This indicated that miR-151a-5p could promote the expression of AGMAT in CRC cells. The aforementioned data suggested that AGMAT could be a potential target of miR-151a-5p and could be positively associated with miR-151a-5p expression in CRC.

miR-151a-5p promotes the effects of AGMAT on the proliferation, migration and invasion of CRC cells

To further verify whether miR-151a-5p could regulate the potential functions of AGMAT in CRC, AGMAT-stably expressing HCT116 or SW480 cells were transfected with miR-151a-5p inhibitor or mimics. Following transfection for 48 h, the proliferation ability of AGMAT-stably expressing CRC cells was evaluated by CCK-8 and colony formation assays. Therefore, compared with the NC group, the proliferation of the AGMAT-stably overexpressing HCT116 cells was significantly decreased after transfection with miR-151a-5p inhibitor in a time-dependent manner (Fig. 6A). However, the significant opposite result of the proliferation was observed in a time-dependent manner in the AGMAT-stably knocked down SW480 cells transfected with miR-151a-5p mimics (Fig. 6B). Additionally, the effect of miR-151a-5p targeting AGMAT on the proliferation of the AGMAT-stably overexpressing HCT116 or SW480 cells was also evaluated by colony formation assays. As expected, the proliferation ability of the AGMAT-stably overexpressing HCT116 cells transfected with miR-151a-5p inhibitor was significantly decreased (Fig. 6C), but the significant opposite result of the proliferation was observed in a time-dependent manner in the AGMAT-stably knocked down SW480 cells transfected with miR-151a-5p mimics (Fig. 6D). To further explore whether miR-151a-5p could regulate CRC cell invasion and migration via targeting AGMAT, Transwell invasion and migration assays were performed. The results showed that compared with the NC group, the migratory and invasive abilities of the AGMAT-stably overexpressing HCT116 cells transfected with miR-151a-5p inhibitor were significantly decreased (Fig. 6E-G). The completely opposite effect was obtained in the AGMAT-stably knocked down SW480 cells transfected with miR-151a-5p mimics (Fig. 6H-J). The aforementioned results indicated that miR-151a-5p could enhance the biofunction of CRC cells via targeting AGMAT.

Discussion

Emerging evidence has suggested that miRNAs can mediate the development of several diseases, such as CRC, through multiple genes and biological pathways (3032). It has been reported that miRNAs are abnormally expressed in CRC tissues, thus serving as powerful and promising biomarkers for the early screening and treatment of CRC (33,34). For example, Lopez-Camarillo et al (35) investigated the expression of several miRNAs, known as metaMIRs, which are involved in the initiation of cell invasion and metastasis via targeting numerous proteins associated with several cellular processes. Therefore, the aforementioned study highlighted the potential use of metaMIRs as novel specific markers for evaluating cancer progression. MicroRNAs play an important role in regulating gene expression under normal and pathological conditions, such as cancer (36). The majority of miRNAs act as tumor suppressors, thus promoting gene silencing through translational suppression, which mainly occurs in the cytoplasm (27). However, the aberrant overexpression of other miRNAs, known as super-enhancers, commonly localized in the nucleus, can promote tumor occurrence, growth and/or metastasis (28,29). It has been reported that several miRNAs can act as biomarkers in CRC via negatively regulating the expression of different genes. However, the application of miRNAs as super-enhancers in clinical practice remains limited. Previous studies demonstrated that miR-151a-5p could be used as a potential non-invasive serological diagnostic marker for the diagnosis of CRC (14) and endometrial cancer (37). However, the effect and underlying mechanism of miR-151a-5p on CRC remains unclear. Consistent with previous findings, the results of the current study revealed that miR-151a-5p was significantly upregulated in CRC. Additionally, miR-151a-5p overexpression enhanced the proliferation ability of CRC cells. Furthermore, the results demonstrated that the tumor-promoting effects of miR-151a-5p were mediated via improving the migration and invasion abilities of CRC cells. MMPs are significant calcium- and zinc-dependent proteolytic enzymes, which can degrade the components of the extracellular matrix and basement membrane. These proteins are involved in cell invasion and metastasis during cancer progression (38). The present study verified that miR-151a-5p overexpression notably reinforced the migration and invasion abilities of CRC cells and increased the protein expression levels of MMP2 and MMP9. In addition, miR-151a-5p promoted the EMT of CRC cells.

miRNAs are a class of endogenous small single-stranded non-encoding RNAs with a length of ~18–24 nucleotides, completely or incompletely complementary with the 3′ UTR of their target mRNAs. miRNAs can negatively regulate the expression of their target genes at the post-transcriptional level (39,40). In the present study, bioinformatic analysis revealed that the AGMAT 3′ UTR encompassed a complementary binding site for miR-151a-5p and AGMAT positively regulated miR-151a-5p expression. To further explore the combined effect of miR-151a-5p and AGMAT on the occurrence and development of CRC, a dual luciferase reporter assay was performed to verify that miR-151a-5p could directly target AGMAT. Similar to arginase, AGMAT, as a metal hydrolase, belongs to the urea hydrolase superfamily. The catalytic activity of AGMAT depends on Mn2+ ions (41). Agmatine, an endogenous polyamine located in mitochondria, is catalyzed by L-arginine decarboxylase and hydrolyzed by agmatine enzyme to form urea and putrescine. Previous studies suggested that AGMAT could not only regulate the biological effects of agmatine in mammalian cells (42), but it could also be considered as an alternative mechanism for polyamine biosynthesis (5). Several studies have reported the regulatory mechanisms of AGMAT and its analogs in several diseases (4345). However, the role of AGMAT in cancer remains elusive. Recent studies showed that AGMAT could play a significant role in lung adenocarcinoma (6) and pancreatic adenocarcinoma (7). In the present study, AGMAT was identified to be positively associated with miR-151a-5p expression. In addition, transfection of AGMAT-stably knocked down CRC cells with miR-151a-5p mimics promoted the proliferation, migration and invasion of CRC cells. The current study was the first to demonstrate that miR-151a-5p could play a significant role in regulating the proliferation and metastasis of CRC cells via targeting AGMAT.

In conclusion, the current study demonstrated that miR-151a-5p was significantly upregulated in CRC tissues and cell lines, while its ectopic expression promoted the proliferation, migration and invasion of CRC cells via targeting AGMAT. Furthermore, miR-151a-5p could enhance the EMT processes of CRC cells. Therefore, the miR-151a-5p/AGMAT axis could serve as a potential therapeutic target for the diagnosis and treatment of CRC.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by the Shanghai municipal health commission Foundation (grant no. 20204Y0181), the Health Commission research Program of Hunan (grant no. 202203034469), and the Fengxian Science and Technology Commission Project (grant no. 20211805).

Availability of data and materials

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

Authors' contributions

YX, YZ, LC and XL contributed to the experiments. CW and MH were responsible for the bioinformatic analysis and the experiment result analysis. YX and XZ confirm the authenticity of all the raw data. XL and YX contributed to writing the manuscript. JC and XZ designed project ideas, provided technical guidance and revised the manuscript. All authors have read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CRC

colorectal carcinoma

AGMAT

agmatinase

TGF

transforming growth factor

GEDS

Gene Expression Display Server

TCGA

The Cancer Genome Atlas

RT-qPCR

reverse transcription-quantitative PCR

GFP

green fluorescent protein

DAPI

4′,6-diamidino-2-phenylindole

MMP

matrix metalloproteinase

EMT

epithelial-mesenchymal transition

References

1 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2022. CA Cancer J Clin. 72:7–33. 2022. View Article : Google Scholar : PubMed/NCBI

2 

Kaplan MA, Isikdogan A, Gumus M, Arslan UY, Geredeli C, Ozdemir N, Koca D, Dane F, Suner A, Elkiran ET, et al: Childhood, adolescents, and young adults (≤25 y) colorectal cancer: Study of Anatolian society of medical oncology. J Pediatr Hematol Oncol. 35:83–89. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Heervä E, Lavonius M, Jaakkola P, Minn H and Ristamäki R: Overall survival and metastasis resections in patients with metastatic colorectal cancer using electronic medical records. J Gastrointest Cancer. 49:245–251. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Wang X, Ying W, Dunlap KA, Lin G, Satterfield MC, Burghardt RC, Wu G and Bazer FW: Arginine decarboxylase and agmatinase: An alternative pathway for de novo biosynthesis of polyamines for development of mammalian conceptuses. Biol Reprod. 90:842014. View Article : Google Scholar : PubMed/NCBI

6 

Zhu HE, Yin JY, Chen DX, He S and Chen H: Agmatinase promotes the lung adenocarcinoma tumorigenesis by activating the NO-MAPKs-PI3K/Akt pathway. Cell Death Dis. 10:8542019. View Article : Google Scholar : PubMed/NCBI

7 

Zhang Y, Cao L, Xie Y, Wang C, Liu X, Zhang X and Chen J: Agmatinase facilitates the tumorigenesis of pancreatic adenocarcinoma through the TGFβ/Smad pathway. Exp Ther Med. 24:4902022. View Article : Google Scholar : PubMed/NCBI

8 

Hosseini M, Khatamianfar S, Hassanian SM, Nedaeinia R, Shafiee M, Maftouh M, Ghayour-Mobarhan M, ShahidSales S and Avan A: Exosome-encapsulated microRNAs as potential circulating biomarkers in colon cancer. Curr Pharm Des. 23:1705–1709. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Pidíkova P, Reis R and Herichova I: miRNA clusters with down-regulated expression in human colorectal cancer and their regulation. Int J Mol Sci. 21:46332020. View Article : Google Scholar : PubMed/NCBI

10 

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

11 

Wang H: MicroRNAs and apoptosis in colorectal cancer. Int J Mol Sci. 21:53532020. View Article : Google Scholar : PubMed/NCBI

12 

Liu G and Li B: Role of miRNA in transformation from normal tissue to colorectal adenoma and cancer. J Cancer Res Ther. 15:278–285. 2019.PubMed/NCBI

13 

Huang S, Tan X, Huang Z, Chen Z, Lin P and Fu SW: microRNA biomarkers in colorectal cancer liver metastasis. J Cancer. 9:3867–3873. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Zhang H, Zhu M, Shan X, Zhou X, Wang T, Zhang J, Tao J, Cheng W, Chen G, Li J, et al: A panel of seven-miRNA signature in plasma as potential biomarker for colorectal cancer diagnosis. Gene. 687:246–254. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Daugaard I, Sanders KJ, Idica A, Vittayarukskul K, Hamdorf M, Krog JD, Chow R, Jury D, Hansen LL, Hager H, et al: miR-151a induces partial EMT by regulating E-cadherin in NSCLC cells. Oncogenesis. 6:e3662017. View Article : Google Scholar : PubMed/NCBI

16 

Fredsøe J, Rasmussen AKI, Mouritzen P, Borre M, Ørntoft T and Sørensen KD: A five-microRNA model (pCaP) for predicting prostate cancer aggressiveness using cell-free urine. Int J Cancer. 145:2558–2567. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Almeida RS, Costa E, Silva M, Coutinho LL, Garcia Gomes R, Pedrosa F, Massaro JD, Donadi EA and Lucena-Silva N: MicroRNA expression profiles discriminate childhood T-from B-acute lymphoblastic leukemia. Hematol Oncol. 37:103–112. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Zhou R, Wang R, Qin Y, Ji J, Xu M, Wu W, Chen M, Wu D, Song L, Shen H, et al: Mitochondria-related miR-151a-5p reduces cellular ATP production by targeting CYTB in asthenozoospermia. Sci Rep. 5:177432015. View Article : Google Scholar : PubMed/NCBI

19 

Link F, Krohn K and Schumann J: Author correction: Identification of stably expressed housekeeping miRNAs in endothelial cells and macrophages in an inflammatory setting. Sci Rep. 9:144662019. View Article : Google Scholar : PubMed/NCBI

20 

Guo S, Zhang J, Zhao YY, Zhou LY, Xie Y, Wu XY, Bian X and Yu XY: The expressions of miR-151a-5p and miR-23b in lung cancer tissues and their effects on the biological functions of lung cancer A549 cells. Eur Rev Med Pharmacol Sci. 24:6779–6785. 2020.PubMed/NCBI

21 

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

22 

Baj J, Korona-Glowniak I, Forma A, Maani A, Sitarz E, Rahnama-Hezavah M, Radzikowska E and Portincasa P: Mechanisms of the epithelial-mesenchymal transition and tumor microenvironment in Helicobacter pylori-induced gastric cancer. Cells. 9:10552020. View Article : Google Scholar : PubMed/NCBI

23 

Adnan M, Siddiqui AJ, Hamadou WS, Snoussi M, Badraoui R, Ashraf SA, Jamal A, Awadelkareem AM, Sachidanandan M, Hadi S, et al: Deciphering the molecular mechanism responsible for efficiently inhibiting metastasis of human non-small cell lung and colorectal cancer cells targeting the matrix metalloproteinases by selaginella repanda. Plants (Basel). 10:9792021. View Article : Google Scholar : PubMed/NCBI

24 

Arruabarrena-Aristorena A, Zabala-Letona A and Carracedo A: Oil for the cancer engine: The cross-talk between oncogenic signaling and polyamine metabolism. Sci Adv. 4:eaar26062018. View Article : Google Scholar : PubMed/NCBI

25 

Yan L, Yao J and Qiu J: miRNA-495 suppresses proliferation and migration of colorectal cancer cells by targeting FAM83D. Biomed Pharmacother. 96:974–981. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Kang X, Kong B, Chen Q and Zhao S: Low expression of miR-138 inhibit the proliferation, migration and invasion of colorectal cancer and affect patient survival by targeting SIRT1. Transl Cancer Res. 10:3548–3559. 2021. View Article : Google Scholar : PubMed/NCBI

27 

Wang D, Qiu C, Zhang H, Wang J, Cui Q and Yin Y: Human microRNA oncogenes and tumor suppressors show significantly different biological patterns: From functions to targets. PLoS One. 5:e130672010. View Article : Google Scholar : PubMed/NCBI

28 

Xiao M, Li J, Li W, Wang Y, Wu F, Xi Y, Zhang L, Ding C, Luo H, Li Y, et al: MicroRNAs activate gene transcription epigenetically as an enhancer trigger. RNA Biol. 14:1326–1334. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Suzuki HI, Young RA and Sharp PA: Super-enhancer-mediated RNA processing revealed by integrative MicroRNA network analysis. Cell. 168:1000–1014.e15. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Chen E, Li Q, Wang H, Yang F, Min L and Yang J: MiR-92a promotes tumorigenesis of colorectal cancer, a transcriptomic and functional based study. Biomed Pharmacother. 106:1370–1377. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Guo S, Zhu KX, Yu WH, Wang T, Li S, Wang YX, Zhang CC and Guo JQ: SH3PXD2A-AS1/miR-330-5p/UBA2 ceRNA network mediates the progression of colorectal cancer through regulating the activity of the Wnt/β-catenin signaling pathway. Environ Toxicol. 36:1969–1980. 2021. View Article : Google Scholar : PubMed/NCBI

32 

Jafarzadeh M and Soltani BM: MiRNA-Wnt signaling regulatory network in colorectal cancer. J Biochem Mol Toxicol. 35:e228832021. View Article : Google Scholar : PubMed/NCBI

33 

Zhang N, Hu X, Du Y and Du J: The role of miRNAs in colorectal cancer progression and chemoradiotherapy. Biomed Pharmacother. 134:1110992021. View Article : Google Scholar : PubMed/NCBI

34 

Zuo Z, Jiang Y, Zeng S, Li Y, Fan J, Guo Y and Tao H: The value of microRNAs as the novel biomarkers for colorectal cancer diagnosis: A meta-analysis. Pathol Res Pract. 216:1531302020. View Article : Google Scholar : PubMed/NCBI

35 

Lopez-Camarillo C, Marchat LA, Arechaga-Ocampo E, Perez-Plasencia C, Del Moral-Hernandez O, Castaneda-Ortiz EJ and Rodriguez-Cuevas S: MetastamiRs: Non-coding MicroRNAs driving cancer invasion and metastasis. Int J Mol Sci. 13:1347–1379. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Kunej T, Godnic I, Horvat S, Zorc M and Calin GA: Cross talk between microRNA and coding cancer genes. Cancer J. 18:223–231. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Fan X, Cao M, Liu C, Zhang C, Li C, Cheng W, Zhang S, Zhang H and Zhu W: Three plasma-based microRNAs as potent diagnostic biomarkers for endometrial cancer. Cancer Biomark. 31:127–138. 2021. View Article : Google Scholar : PubMed/NCBI

38 

Pellikainen JM, Ropponen KM, Kataja VV, Kellokoski JK, Eskelinen MJ and Kosma VM: Expression of matrix metalloproteinase (MMP)-2 and MMP-9 in breast cancer with a special reference to activator protein-2, HER2, and prognosis. Clin Cancer Res. 10:7621–7628. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Fabian MR, Sonenberg N and Filipowicz W: Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 79:351–379. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Bracken CP, Scott HS and Goodall GJ: A network-biology perspective of microRNA function and dysfunction in cancer. Nat Rev Genet. 17:719–732. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Uribe E, Reyes MB, Martinez I, Mella K, Salas M, Tarifeño-Saldivia E, López V, Garcia-Robles M, Martinez-Oyanedel J, Figueroa M, et al: Functional analysis of the Mn2+ requirement in the catalysis of ureohydrolases arginase and agmatinase-a historical perspective. J Inorg Biochem. 202:1108122020. View Article : Google Scholar : PubMed/NCBI

42 

Chai J, Luo L, Hou F, Fan X, Yu J, Ma W, Tang W, Yang X, Zhu J, Kang W, et al: Agmatine reduces lipopolysaccharide-mediated oxidant response via activating PI3K/Akt pathway and up-regulating Nrf2 and HO-1 expression in macrophages. PLoS One. 11:e01636342016. View Article : Google Scholar : PubMed/NCBI

43 

Yılmaz E, Şekeroğlu MR, Yılmaz E and Çokluk E: Evaluation of plasma agmatine level and its metabolic pathway in patients with bipolar disorder during manic episode and remission period. Int J Psychiatry Clin Pract. 23:128–133. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Dallmann K, Junker H, Balabanov S, Zimmermann U, Giebel J and Walther R: Human agmatinase is diminished in the clear cell type of renal cell carcinoma. Int J Cancer. 108:342–347. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Bernstein HG, Stich C, Jäger K, Dobrowolny H, Wick M, Steiner J, Veh R, Bogerts B and Laube G: Agmatinase, an inactivator of the putative endogenous antidepressant agmatine, is strongly upregulated in hippocampal interneurons of subjects with mood disorders. Neuropharmacology. 62:237–246. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2023
Volume 49 Issue 3

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
Xie Y, Zhang Y, Liu X, Cao L, Han M, Wang C, Chen J and Zhang X: miR‑151a‑5p promotes the proliferation and metastasis of colorectal carcinoma cells by targeting AGMAT. Oncol Rep 49: 50, 2023
APA
Xie, Y., Zhang, Y., Liu, X., Cao, L., Han, M., Wang, C. ... Zhang, X. (2023). miR‑151a‑5p promotes the proliferation and metastasis of colorectal carcinoma cells by targeting AGMAT. Oncology Reports, 49, 50. https://doi.org/10.3892/or.2023.8487
MLA
Xie, Y., Zhang, Y., Liu, X., Cao, L., Han, M., Wang, C., Chen, J., Zhang, X."miR‑151a‑5p promotes the proliferation and metastasis of colorectal carcinoma cells by targeting AGMAT". Oncology Reports 49.3 (2023): 50.
Chicago
Xie, Y., Zhang, Y., Liu, X., Cao, L., Han, M., Wang, C., Chen, J., Zhang, X."miR‑151a‑5p promotes the proliferation and metastasis of colorectal carcinoma cells by targeting AGMAT". Oncology Reports 49, no. 3 (2023): 50. https://doi.org/10.3892/or.2023.8487