MicroRNA‑337 inhibits colorectal cancer progression by directly targeting KRAS and suppressing the AKT and ERK pathways

Retraction in: /10.3892/or.2022.8394

  • Authors:
    • Xuerong Liu
    • Yan Wang
    • Jinglong Zhao
  • View Affiliations

  • Published online on: September 25, 2017     https://doi.org/10.3892/or.2017.5997
  • Pages: 3187-3196
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Colorectal cancer (CRC) is the third most common cancer and the fourth most common cause of cancer-related death worldwide. Tumour progression and development in CRC is a multi-step process involving a large number of genetic and epigenetic alterations. Previous studies indicated that abnormally expressed microRNAs play critical roles in CRC through regulation of oncogenic and tumour-suppressor genes. Hence, determination of the function of microRNAs may provide novel therapeutic targets for CRC diagnosis and treatments. MicroRNA‑337 (miR‑337) has been reported to be downregulated in several cancer types. However, the expression, function and underlying mechanisms of miR‑337 in CRC have not been clearly elucidated. In this study, miR‑337 was significantly decreased in CRC tissues and cell lines. Low miR‑337 expression level was correlated with lymph node metastasis, distant metastasis and TNM stage of CRC patients. In addition, upregulation of miR‑337 suppressed cell proliferation and invasion and promoted apoptosis in CRC. Based on bioinformatics analysis, we assumed that Kirsten rat sarcoma viral oncogene homolog (KRAS) was directly modulated by miR‑337 in CRC. Luciferase reporter assay demonstrated the direct interaction between miR‑337 and 3'‑UTR of KRAS mRNA. Furthermore, reverse transcription-quantitative polymerase chain reaction and western blot analysis indicated that miR‑337 could negatively regulate endogenous KRAS expression in CRC cells at both mRNA and protein levels. Moreover, KRAS was highly expressed in CRC tissues and inversely correlated with miR‑337 expression in CRC tissues. KRAS knockdown recapitulates effects similar to those induced by miR‑337 overexpression in CRC cells, whereas KRAS overexpression partially restored the tumour suppressive effects of miR‑337. Besides, ectopic expression of miR‑337 inactivates the AKT and ERK signalling pathways in CRC. These results suggested that miR‑337 may act as a tumour suppressor during the process of CRC malignant transformation by interacting with KRAS.

Introduction

Colorectal cancer (CRC) is the third most common cancer and the fourth most common cause of cancer-related deaths worldwide, with almost 1.23 million new cases and 0.6 million deaths per year (1,2). Until recently, several risk factors for CRC have been identified; these factors include age, obesity, smoking, dietary patterns, physical inactivity, metabolic syndrome and genetic and epigenetic factors (3,4). Despite advances in the diagnosis and therapy of CRC, which have improved the life expectancy of patients, the prognosis of patients with CRC remains poor (5). Approximately half of CRC patients remarkably develop distant metastases and high rates of recurrence after surgery, which are the main causes of death in the patients (6). Therefore, a better understanding of the molecular mechanisms underlying CRC formation and progression is vital to identify novel diagnostic markers and develop therapeutic strategies for patients with CRC.

Recently, microRNAs (miRNAs) have emerged as a major group of regulatory genes in plants and animals (7). miRNAs are a family of endogenous, noncoding and short RNA containing ~19–23 nucleotides (8). They negatively regulate the expression of their target mRNAs by interacting with complementary sites in the 3′ untranslated regions (UTRs), leading to translational repression or degradation of target mRNAs (9). miRNAs are often located in genomic breakpoint regions and play important roles in numerous physiological and pathological processes, including differentiation, proliferation, apoptosis, migration, invasion and survival of cells (10,11). Abnormal expression of miRNAs is observed in various human disorders, including cancer (12). Moreover, dysregulation of miRNAs is implicated in the initiation and progression of human cancers, such as CRC (13), gastric cancer(14), lung cancer (15), prostate cancer (16) and breast cancer (17). Functionally, miRNAs may act in either a tumour suppressing or promoting role in various kinds of human cancer, depending on whether they specifically target oncogenes or tumour suppressor genes (18). Therefore, miRNAs may be promising prognostic markers and therapeutic targets for patients with CRC.

miR-337, which is located at chromosome 14q32.2, aberrantly expressed several types of human cancers (1921). However, the expression, function and underlying mechanisms of miR-337 in CRC have not been clearly elucidated. In the present study, we investigated the expression level, functions of miR-337 in CRC and its underlying molecular mechanisms.

Materials and methods

Tissue specimens

This study was approved by the Ethics Committee of First Affiliated Hospital of Harbin Medical University, and written informed consent was also obtained from each patient enrolled in this research. A total of 54 paired CRC tissues and normal adjacent tissues were obtained from CRC patients who received surgical resection at First Affiliated Hospital of Harbin Medical University between July 2014 and March 2016. These 54 CRC patients have not been treated with radiotherapy or chemotherapy prior to surgical resection. All tissue samples were immediately frozen in liquid nitrogen at the time of surgery and stored at −80°C until further use.

Cell lines and cell transfection

Five human CRC cell lines (LoVo, HCT116, HT29, SW480, SW620), normal human colon epithelium cell line FHC and human embryonic kidney 293T cells (HEK293T) were acquired from American Type Culture Collection (Manassas, VA, USA). All cells were routinely grown in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 10% foetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), 100 U/ml penicillin (Sigma, St. Louis, MO, USA) and 100 µg/ml streptomycin (Sigma). Cell cultures were kept in a humidified environment with 5% CO2 at 37°C.

miR-337 mimics and corresponding negative control miRNA (miR-NC) were purchased from GenePharma, Co., Ltd. (Shanghai, China). Small interfering RNA targeting Kirsten rat sarcoma viral oncogene homolog (si-KRAS) and the corresponding control siRNA (si-NC) were purchased from Ribobio (Guangzhou, China). pCMV-KRAS and blank plasmid pCMV were obtained from GeneCopoeia (Guangzhou, China). Cells were plated in 6-well clusters at a density of 60–70% confluence. Cell transfection was performed using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc. Waltham, MA, USA) according to the manufacturer's instructions. Transfected cells were then incubated at 37°C with 5% CO2, and the culture medium was replaced with fresh DMEM supplemented with 10% FBS at 8 h post-transfection.

RNA isolation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA from tissue samples or cells was isolated using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's instructions. The concentration of total RNA was examined using a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, Madrid, Spain). To detect miR-337 expression, complementary DNA (cDNA) was prepared using TaqMan MicroRNA Reverse Transcription kit (Applied Biosystems, Carlsbad, CA, USA). Real-time PCR was then conducted using a TaqMan MicroRNA PCR kit (Applied Biosystems) on an ABI PRISM 7900 Sequence Detection system (Applied Biosystems). To quantify KRAS mRNA expression, total RNA was converted into cDNA using PrimeScript RT Reagent kit (Takara Biotechnology Co., Ltd., Dalian, China), followed by quantitative PCR with SYBR Premix Ex Taq™ (Takara Biotechnology Co., Ltd.). U6 and GAPDH served as internal control for miR-337 and KRAS, respectively. Primers used in this study were as follows: miR-337 forward, 5′-ACACTCCAGCTGGGCTCCTATATGATGC-3′ and reverse 5′-ACTCCACGACACCAGTTGAG-3′; U6 forward, 5′-CTCGCTTCGGCAGCACA-3′ and reverse 5′-AACGCTTCACGAATTTGCGT-3′; KRAS forward, 5′-ATTCCTTTTATTGAAACATCAGCA-3′ and reverse 5′-TCGGATCTCCCTCACCAAT-3′; GAPDH forward, 5′-GACTCATGACCACAGTCCATGC-3′ and reverse 5′-AGAGGCAGGGATGATGTTCTG-3′. Relative gene expression was calculated using comparative 2−∆∆Ct method (22).

Cell Counting Kit-8 (CCK-8) assay

Cells were seeded at a density of 3×103 cells/well in 96-well plates. After transfection, cells were grown at 37°C with 5% CO2 for 0, 24, 48 and 72 h. At indicated time-point, CCK8 assay was performed to determine cell proliferation. In brief, 10 µl CCK-8 reagent (Dojindo Laboratories, Kumamoto, Japan) was added into each well and incubated for another 2 h at 37°C. The optical density (OD) value was detected using a plate reader (ELx808 Bio-Tek Instruments, USA) at a wavelength of 450 nm. Three independent experiments were performed in triplicate.

Cell invasion assay

Matrigel (BD Biosciences, San Jose, CA, USA)-coated Transwell inserts containing a membrane with 8-µm pores (Costar; Corning Inc., Corning, NY, USA) were used to perform cell invasion assays. In brief, 1×105 cells suspended in 200 µl FBS-free DMEM medium were plated on top of Transwell insert, whereas DMEM medium containing 20% FBS was placed in the lower chamber as a chemoattract. After 20-h incubation, cells remaining on the upper membrane of the filter were removed by wiping with a cotton swab. The invasive cells on the lower membrane surface were fixed in 20% methanol and stained with 0.1% crystal violet. Photographs were taken, and the number of invasive cells was counted in five randomly selected fields under an inverted microscope (×200; Olympus, Tokyo, Japan). The assay was independently repeated thrice.

Flow cytometry analysis

After 48 h of transfection, transfected cells were harvested, washed with phosphate-buffered saline (PBS) and fixed with 80% ice-cold ethanol in PBS. Cell apoptosis was detected using Annexin V-FITC apoptosis detection kit (Invitrogen Corp., CA, USA). In brief, cells were re-suspended in 300 µl 1X binding buffer, followed by staining with 5 µl FITC-Annexin V and 5 µl propidium iodide for 20 min in the dark at room temperature. Cell apoptosis was recognised by a FACScan flow cytometer (Becton-Dickinson, San Jose, CA, USA), and data were analysed using CellQuest® software (Becton-Dickinson, Heidelberg, Germany).

Bioinformatics analysis and luciferase reporter assay

To determine the potential targets of miR-337, bioinformatics analysis was performed with the publicly available algorithms: TargetScan (http://www.targetscan.org), miRanda (http://www.microrna.org/microrna/getExprForm.do) and PICTA (http://pictar.mdc-berlin.de/).

Luciferase reporter assay

Luciferase reporter vector containing the wild-type (pGL3-KRAS-3′-UTR-Wt) or mutant (pGL3-KRAS-3′-UTR-Mut) 3′-UTR of KRAS were synthesised and confirmed by GenePharma Co., Ltd. HEK293T cells were seeded into 24-well plates at a density of 60–70% confluence. After incubation overnight, cells were transfected with miR-337 mimics or miR-NC, together with pGL3-KRAS-3′-UTR-Wt or pGL3-KRAS-3′-UTR-Mut using Lipofectamine 2000. Cells were collected 48 h after transfection, and luciferase activities were examined using a dual-luciferase reporter system (Promega, Madison, WI, USA) according to the manufacturer's protocols. Firefly luciferase activities were normalised to Renilla luciferase activities. Each assay was performed in triplicate.

Western blot analysis

Total protein was extracted from tissue samples or cells using radioimmunoprecipitation assay cell lysis buffer (Beyotime, Shanghai, China). Protein concentrations were quantified using BCA assay kit (Pierce, Rockford, IL, USA). Equal amounts of protein were separated by 10% SDS PAGE, transferred onto PVDF membrane (Millipore, Billerica, MA, USA) and then blocked in Tris-buffered saline with Tween (TBST) buffer with 5% fat-free milk for 2 h at 37°C. Subsequently, the membranes were incubated with primary antibodies: mouse anti-human monoclonal KRAS (sc-30; 1:1,000 dilution; Santa Cruz Biotechnology, CA, USA), mouse anti-human monoclonal p-AKT (sc-271966; 1:1,000 dilution; Santa Cruz Biotechnology), mouse anti-human monoclonal AKT (sc-81434; 1:1,000 dilution; Santa Cruz Biotechnology), mouse anti-human monoclonal p-ERK (sc-81492; 1:1,000 dilution; Santa Cruz Biotechnology), mouse anti-human monoclonal ERK (sc-514302; 1:1,000 dilution; Santa Cruz Biotechnology), and mouse anti-human monoclonal GAPDH antibody (sc-47724; 1:1,000 dilution; Santa Cruz Biotechnology). Following washing in TBST, the membranes were further probed with goat anti-mouse horseradish peroxidase-conjugated secondary antibody (sc-2005; 1:5,000 dilution; Santa Cruz Biotechnology) at room temperature for 1 h. Protein bands were visualised by applying ECL Protein Detection kit (Pierce Biotechnology, Inc., Rockford, IL, USA) and analysed using Quantity One software (Bio-Rad Laboratories, Inc., Hercules, CA, USA). GAPDH was used as the loading control.

Statistical analysis

Data are expressed as the mean ± standard deviation. All statistical analyses were performed using Student's t-test or one way ANOVA test with SPSS software (version 18.0; SPSS, Inc., Chicago, IL, USA). Student-Newman-Keuls was used to compare between two groups in multiple groups. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-337 expression and its association with clinicopathological characteristics in CRC

To investigate the expression pattern of miR-337 in CRC, we measured its expression in 54 paired CRC tissues and normal adjacent tissues using RT-qPCR. As shown in Fig. 1A, miR-337 was significantly downregulated in CRC tissues compared with that in normal adjacent tissues (P<0.05). Subsequently, we explored the association between miR-337 expression and clinicopathological characteristics of CRC patients. The colorectal cancer tissues were divided into either the miR-337 low-expression group (n=28) or the miR-337 high-expression group (n=26) using the median expression level of miR-337 as a cut-off. As shown in Table I, low expression levels of miR-337 were significantly correlated with lymph node metastasis (P=0.002), distant metastasis (P=0.014) and TNM stage (P<0.0001, significant difference), whereas no correlation was found with patient gender (P=0.276), age at diagnosis (P=0.177), location (P=0.394) or tumour size (P=0.279).

Table I.

Correlation of microRNA-337 expression and clinicopathological factors in colorectal cancer.

Table I.

Correlation of microRNA-337 expression and clinicopathological factors in colorectal cancer.

MicroRNA-337 level

FactorsNo. of casesLowHighP-value
Sex   0.276
  Male271215
  Female271611
Age at diagnosis (years)   0.177
  <501510  5
  ≥50391821
Location   0.394
  Colon241410
  Rectum301416
Tumour size (cm)   0.279
  <5401921
  ≥514  9  5
Distant metastasis   0.014
  No371522
  Yes1713  4
Lymph node metastasis   0.002
  No321121
  Yes2217  5
TNM stage <0.0001a
  I–II29  821
  III–IV2520  5

a Significant difference.

Moreover, miR-337 expression level was also detected in CRC cell lines (LoVo, HCT116, HT29, SW480, SW620) and normal human colon epithelium cell line FHC. The expression level of miR-337 was reduced in CRC cell lines compared with that in FHC (Fig. 1B, P<0.05). These results suggested that miR-337 may play important roles in CRC formation and progression.

miR-337 inhibits CRC cell proliferation, invasion and increases apoptosis

To observe the effects of miR-337 on CRC cells, miR-337 mimics or miR-NC were transfected into HCT116 and SW480 cells. After transfection, RT-qPCR was performed to evaluate the transfection efficiency and found that miR-337 was markedly upregulated in HCT116 and SW480 cells after transfection with miR-337 mimics (Fig. 2A, P<0.05). CCK-8 assay was utilised to examine the effect of miR-337 overexpression on CRC cell proliferation and revealed that upregulation of miR-337 obviously suppressed the HCT116 and SW480 cell proliferation (Fig. 2B, P<0.05). To evaluate the effect of miR-337 on the invasion capacity of CRC cells, cell invasion assay was applied in HCT116 and SW480 cells which were transfected with miR-337 mimic or miR-NC. As shown in Fig. 2C, miR-337 overexpression decreased the invasive abilities of HCT116 and SW480 cells (P<0.05). Moreover, we used flow cytometry analysis to explore the effect of miR-337 on CRC cell apoptosis. Results showed that the restoration expression of miR-337 increased cell apoptosis in HCT116 and SW480 cells (Fig. 2D, P<0.05). These data suggested that miR-337 may serve as a tumour suppressor in CRC.

KRAS is a direct target of miR-337 in CRC

To elucidate the underlying mechanisms by which miR-337 affects the biological functions of CRC cells, we investigated the potential targets of miR-337 using bioinformatics analysis. Among these candidate genes, KRAS was selected for further validation because it was aberrantly observed in CRC and plays key roles in CRC occurrence and development (23). As shown in Fig. 3A, two miR-337 putative binding sites were identified covering the nucleotides 3532-3528 (site 1) and 4448-4454 (site 2) of KRAS 3′-UTR. To assess whether miR-337 could directly target the 3′-UTR of KRAS, luciferase reporter assay was conducted in HEK293T cells cotransfected with luciferase reporter vector and miR-337 mimics or miR-NC. Results revealed that upregulation of miR-337 led to a significant decrease in pGL3-KRAS-3′-UTR site 1 Wt and pGL3-KRAS-3′-UTR site 2 Wt luciferase activity in HEK293T cells compared with miR-NC cells (Fig. 3B, P<0.05). Mutation of the two miR-337 binding sites (site 1 Mut or site 2 Mut) restored the normal luciferase activity of KRAS-3′-UTR in HEK293T cells.

To further confirm whether miR-337 can affect the endogenous level of KRAS, miR-337 mimics or miR-NC were transfected into HCT116 and SW480 cells to investigate the mRNA and protein levels of KRAS using RT-qPCR and western blot analysis. Results showed that enforced expression of miR-337 significantly reduced the mRNA (Fig. 3C, P<0.05) and protein (Fig. 3D, P<0.05) levels of KRAS in HCT116 and SW480 cells. Overall, KRAS gene is a direct target of miR-337 in CRC.

miR-337 was inversely correlated with KRAS expression in CRC tissues

To further examine the association between miR-337 and KRAS, we measured KRAS mRNA and protein expression in CRC tissues and normal adjacent tissues using RT-qPCR and western blot analysis. KRAS was dramatically upregulated at both mRNA (Fig. 4A, P<0.05) and protein (Fig. 4B, P<0.05) levels in CRC tissue compared with normal adjacent tissues. Spearman's correlation analysis further demonstrated that miR-337 expression level was negatively correlated with the expression level of KRAS mRNA in CRC tissues (Fig. 4C; r=-0.6648, P<0.0001).

KRAS knockdown recapitulates similar effects to miR-337 overexpression in CRC cells

KRAS was identified as a direct target of miR-337. Therefore, we hypothesised that miR-337 affecting the biological functions of CRC cells might be achieved by KRAS knockdown. To confirm this hypothesis, si-KRAS was used to decrease KRAS expression level in HCT116 and SW480 cells. After transfection, western blot analysis showed that KRAS protein expression was obviously downregulated in HCT116 and SW480 cells transfected with si-KRAS (Fig. 5A, P<0.05). Functional assays demonstrated that KRAS knockdown could repress proliferation (Fig. 5B, P<0.05) and invasion (Fig. 5C, P<0.05) and promoted apoptosis (Fig. 5D, P<0.05) of HCT116 and SW480 cells, which was similar to the effects induced by miR-337 overexpression. The tumour-suppressing effects of miR-337 on CRC cells depends, at least in part, on its direct target KRAS.

Overexpression of KRAS rescued the tumour-suppressive effects of miR-337 in CRC cells

To further evaluate whether KRAS is responsible for the functional roles of miR-337 in CRC, rescue experiments were performed in HCT116 and SW480 cells transfected with miR-337 mimics with or without pCMV-KRAS. Western blot analysis showed that KRAS protein was obviously inhibited by miR-337 overexpression in HCT116 and SW480 cells, whereas upregulation of KRAS by pCMV-KRAS abolished the suppression of KRAS caused by miR-337 mimics (Fig. 6A, P<0.05). Function investigation revealed that upregulation of KRAS reversed the tumour-suppressing effects of miR-337 on CRC cell proliferation (Fig. 6B, P<0.05), invasion (Fig. 6C, P<0.05) and apoptosis (Fig. 6D, P<0.05). miR-337 served as a tumour suppressor in CRC, at least in part, by directly targeting KRAS.

miR-337 inactivates the AKT and ERK signalling pathways in CRC

Previous studies reported that activation of KRAS can trigger several significant pathways, including the AKT and ERK pathways (24,25). Therefore, we measured the protein expression level of p-AKT, AKT, p-ERK and ERK in HCT116 and SW480 cells after transfection with miR-337 mimics or miR-NC. The data of western blot analysis indicated that ectopic expression of miR-337 reduced the p-AKT and p-ERK expression levels and negatively regulated AKT and ERK pathways (Fig. 7). These results suggested that miR-337 exerts tumour-suppressing roles in CRC cells by directly targeting KRAS and indirectly regulating AKT and ERK pathways.

Discussion

Tumour progression and development in CRC is a multi-step process involving a large number of genetic and epigenetic alterations (26,27). Considerable evidence indicates that abnormally expressed miRNAs play critical roles in CRC through regulation of oncogenic and tumour-suppressor genes (2830). Hence, investigating the function of miRNAs involved in CRC occurrence and development may provide novel therapeutic targets for CRC diagnosis and treatments. In this study, miR-337 was significantly downregulated in CRC tissues and cells. The low expression level of miR-337 was correlated with malignant clinical features of CRC patients, including lymph node metastasis, distant metastasis and TNM stage. In addition, upregulation of miR-337 inhibited cell proliferation and invasion and promoted apoptosis of CRC by directly targeting KRAS and regulating AKT and ERK pathways. These findings suggested that miR-337 could be potentially developed as effective therapeutic target for the therapy of CRC patients.

The abnormal expression of miR-337 has been reported in several kinds of human malignancies. For example, miR-337 was downregulated in gastric cancer tissues and correlated with lymph node metastasis (31,32). In neuroblastoma, miR-337 was expressed at low level in tumour tissues and cell lines. Neuroblastoma patients with high miR-337 expression had greater survival probability than patients with low miR-337 expression levels (19). In pancreatic cancer, the expression level of miR-337 decreased and was related to TNM stage, lymph node status and length of patient survival (20). In addition, low expression level of miR-337 was associated with longer survival in patients with pancreatic cancer (21). miR-337 was downregulated in lung cancer (33), breast cancer (34) and oesophageal squamous cell carcinoma (35). miR-337 may be a diagnostic and prognostic biomarker for these types of cancer.

miR-337 participated in cancer carcinogenesis and progression. For instance, Zheng et al found that expression restoration of miR-337 attenuated gastric cancer cell proliferation, invasion and angiogenesis in vitro (32). Xiang et al revealed that miR-337 overexpression repressed cell growth, metastasis and angiogenesis of neuroblastoma cells both in vitro and in vivo (19). Zhang et al reported that resumption of expression of miR-337 suppressed cell proliferation and invasion of pancreatic ductal adenocarcinoma (20). Du et al showed that miR-337 re-expression sensitised non-small cell lung cancer (NSCLC) cells to paclitaxel and enhanced paclitaxel-induced G2/M arrest (33). These studies suggested a fundamental role for miR-337 in tumourigenesis and development of malignant tumours and illustrate its potential as a therapeutic target for various types of cancer.

Identification of the miR-337 direct targets is important in understanding its roles in NSCLC and is also essential in the development of novel therapeutic strategies for patients with CRC. Several targets of miR-337 have been validated; these targets include MMP14 (19,32) in gastric cancer and neuroblastoma, TGFBR2 (36) in chondrogenesis, HOXB7 (20) in pancreatic cancer and STAT3 (33) and RAP1A (33) in NSCLC. In the present study, KRAS was identified as a direct target of miR-337 in CRC. Firstly, bioinformatic analysis predicted that KRAS was a potential target of miR-337. The luciferase reporter assays indicated that the 3′-UTR of KRAS could be directly targeted by miR-337. Subsequently, RT-qPCR and western blot analysis confirmed that miR-337 negatively regulated KRAS expression in CRC cells at both mRNA and protein levels. Furthermore, KRAS was upregulated in CRC tissues and inversely correlated with miR-337 expression. The biological roles of KRAS knockdown in CRC cells were similar to those induced by miR-337 overexpression. Besides, overexpression of KRAS rescued the tumour-suppressive effects of miR-337 in CRC cells. These results strongly demonstrated that KRAS is a direct and functional target of miR-337 in CRC.

KRAS, which is a member of the Ras family, is located at 12p12.1 and encodes a protein that is a member of the small GTPase superfamily (37). The expression levels of KRAS are overexpressed in an aggregate of human cancers, such as gastric cancer (38), bladder cancer (39), lung cancer (40) and pancreatic cancer (41). In CRC, KRAS was upregulated in tumour tissues compared with normal colorectal mucosa. The expression level of KRAS was correlated with the degree of differentiation of CRC. The average progression-free survival of patients with high KRAS levels was significantly lower than patients with high levels (23). Moreover, abnormal activation of the KRAS was involved in various biological behaviour of cells, including cell proliferation, survival, differentiation, migration, invasion, angiogenesis and apoptosis (42,43). Therefore, KRAS may be developed as a major target for potential CRC therapies and numerous strategies.

In conclusion, miR-337 plays a critical role in the occurrence and development of CRC by directly targeting KRAS and indirectly regulating AKT and ERK pathways, thereby suggesting a theoretical basis for its application in the treatment of patients with CRC.

References

1 

Jemal A, Siegel R, Xu J and Ward E: Cancer statistics, 2010. CA Cancer J Clin. 60:277–300. 2010. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

Botteri E, Iodice S, Bagnardi V, Raimondi S, Lowenfels AB and Maisonneuve P: Smoking and colorectal cancer: A meta-analysis. JAMA. 300:2765–2778. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Lee IM, Shiroma EJ, Lobelo F, Puska P, Blair SN and Katzmarzyk PT; Lancet Physical Activity Series Working Group, : Effect of physical inactivity on major non-communicable diseases worldwide: An analysis of burden of disease and life expectancy. Lancet. 380:219–229. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Meyerhardt JA and Mayer RJ: Systemic therapy for colorectal cancer. N Engl J Med. 352:476–487. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Juez I, Rubio C and Figueras J: Multidisciplinary approach of colorectal liver metastases. Clin Transl Oncol. 13:721–727. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Hayes J, Peruzzi PP and Lawler S: MicroRNAs in cancer: Biomarkers, functions and therapy. Trends Mol Med. 20:460–469. 2014. View Article : Google Scholar : PubMed/NCBI

8 

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

9 

Xue J, Niu J, Wu J and Wu ZH: MicroRNAs in cancer therapeutic response: Friend and foe. World J Clin Oncol. 5:730–743. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Tutar L, Tutar E, Özgür A and Tutar Y: Therapeutic targeting of microRNAs in cancer: Future perspectives. Drug Dev Res. 76:382–388. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Stahlhut C and Slack FJ: MicroRNAs and the cancer phenotype: Profiling, signatures and clinical implications. Genome Med. 5:1112013. View Article : Google Scholar : PubMed/NCBI

12 

Farazi TA, Hoell JI, Morozov P and Tuschl T: MicroRNAs in human cancer. Adv Exp Med Biol. 774:1–20. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Guo F, Luo Y, Mu YF, Qin SL, Qi Y, Qiu YE and Zhong M: miR-193b directly targets STMN1 and inhibits the malignant phenotype in colorectal cancer. Am J Cancer Res. 6:2463–2475. 2016.PubMed/NCBI

14 

Ding L, Zhang S, Xu M, Zhang R, Sui P and Yang Q: MicroRNA-27a contributes to the malignant behavior of gastric cancer cells by directly targeting PH domain and leucine-rich repeat protein phosphatase 2. J Exp Clin Cancer Res. 36:452017. View Article : Google Scholar : PubMed/NCBI

15 

Huang L, Li F, Deng P and Hu C: MicroRNA-223 promotes tumor progression in lung cancer A549 cells via activation of the NF-κB signaling pathway. Oncol Res. 24:405–413. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Kong Q, Chen XS, Tian T, Xia XY and Xu P: MicroRNA-194 suppresses prostate cancer migration and invasion by downregulating human nuclear distribution protein. Oncol Rep. 37:803–812. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Yang F, Li Y, Xu L, Zhu Y, Gao H, Zhen L and Fang L: miR-17 as a diagnostic biomarker regulates cell proliferation in breast cancer. Onco Targets Ther. 10:543–550. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Zhang B, Pan X, Cobb GP and Anderson TA: microRNAs as oncogenes and tumor suppressors. Dev Biol. 302:1–12. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Xiang X, Mei H, Zhao X, Pu J, Li D, Qu H, Jiao W, Zhao J, Huang K, Zheng L, et al: miRNA-337-3p suppresses neuroblastoma progression by repressing the transcription of matrix metalloproteinase 14. Oncotarget. 6:22452–22466. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Zhang R, Leng H, Huang J, Du Y, Wang Y, Zang W, Chen X and Zhao G: miR-337 regulates the proliferation and invasion in pancreatic ductal adenocarcinoma by targeting HOXB7. Diagn Pathol. 9:1712014. View Article : Google Scholar : PubMed/NCBI

21 

Zhang R, Zheng S, Du Y, Wang Y, Zang W and Zhao G: Levels of HOXB7 and miR-337 in pancreatic ductal adenocarcinoma patients. Diagn Pathol. 9:612014. View Article : Google Scholar : 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 

Chen S, Wang Y, Zhang Y and Wan Y: Low expression of PKCα and high expression of KRAS predict poor prognosis in patients with colorectal cancer. Oncol Lett. 12:1655–1660. 2016.PubMed/NCBI

24 

Hubbard PA, Moody CL and Murali R: Allosteric modulation of Ras and the PI3K/AKT/mTOR pathway: Emerging therapeutic opportunities. Front Physiol. 5:4782014. View Article : Google Scholar : PubMed/NCBI

25 

Calvo F, Agudo-Ibáñez L and Crespo P: The Ras-ERK pathway: Understanding site-specific signaling provides hope of new anti-tumor therapies. BioEssays. 32:412–421. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Guda K, Veigl ML, Varadan V, Nosrati A, Ravi L, Lutterbaugh J, Beard L, Willson JK, Sedwick WD, Wang ZJ, et al: Novel recurrently mutated genes in African American colon cancers. Proc Natl Acad Sci USA. 112:pp. 1149–1154. 2015; View Article : Google Scholar : PubMed/NCBI

27 

Bardhan K and Liu K: Epigenetics and colorectal cancer pathogenesis. Cancers (Basel). 5:676–713. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Kang X, Wang M, Wang H, Shen X and Guan W: MiR-4282 suppresses proliferation and mobility of human colorectal carcinoma cells by targeting semaphorin 3E. Panminerva Med. 58:197–205. 2016.PubMed/NCBI

29 

Fan C, Lin Y, Mao Y, Huang Z, Liu AY, Ma H, Yu D, Maitikabili A, Xiao H, Zhang C, et al: MicroRNA-543 suppresses colorectal cancer growth and metastasis by targeting KRAS, MTA1 and HMGA2. Oncotarget. 7:21825–21839. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Long HC, Gao X, Lei CJ, Zhu B, Li L, Zeng C, Huang JB and Feng JR: miR-542-3p inhibits the growth and invasion of colorectal cancer cells through targeted regulation of cortactin. Int J Mol Med. 37:1112–1118. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Wang Z, Wang J, Yang Y, Hao B, Wang R, Li Y and Wu Q: Loss of hsa-miR-337-3p expression is associated with lymph node metastasis of human gastric cancer. J Exp Clin Cancer Res. 32:762013. View Article : Google Scholar : PubMed/NCBI

32 

Zheng L, Jiao W, Mei H, Song H, Li D, Xiang X, Chen Y, Yang F, Li H, Huang K, et al: miRNA-337-3p inhibits gastric cancer progression through repressing myeloid zinc finger 1-facilitated expression of matrix metalloproteinase 14. Oncotarget. 7:40314–40328. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Du L, Subauste MC, DeSevo C, Zhao Z, Baker M, Borkowski R, Schageman JJ, Greer R, Yang CR, Suraokar M, et al: miR-337-3p and its targets STAT3 and RAP1A modulate taxane sensitivity in non-small cell lung cancers. PLoS One. 7:e391672012. View Article : Google Scholar : PubMed/NCBI

34 

Kang HS, Kim J, Jang SG, Kwon SY, Park YS, Green JE, Kim HK and Ro J: MicroRNA signature for HER2-positive breast and gastric cancer. Anticancer Res. 34:3807–3810. 2014.PubMed/NCBI

35 

Wu C, Wang C, Guan X, Liu Y, Li D, Zhou X, Zhang Y, Chen X, Wang J, Zen K, et al: Diagnostic and prognostic implications of a serum miRNA panel in oesophageal squamous cell carcinoma. PLoS One. 9:e922922014. View Article : Google Scholar : PubMed/NCBI

36 

Zhong N, Sun J, Min Z, Zhao W, Zhang R, Wang W, Tian J, Tian L, Ma J, Li D, et al: MicroRNA-337 is associated with chondrogenesis through regulating TGFBR2 expression. Osteoarthritis Cartilage. 20:593–602. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Janssen KP, Alberici P, Fsihi H, Gaspar C, Breukel C, Franken P, Rosty C, Abal M, El Marjou F, Smits R, et al: APC and oncogenic KRAS are synergistic in enhancing Wnt signaling in intestinal tumor formation and progression. Gastroenterology. 131:1096–1109. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Li M, Liu W, Zhu YF, Chen YL, Zhang BZ and Wang R: Correlation of COX-2 and K-ras expression to clinical outcome in gastric cancer. Acta Oncol. 45:1115–1119. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Przybojewska B, Jagiello A and Jalmuzna P: H-RAS, K-RAS, and N-RAS gene activation in human bladder cancers. Cancer Genet Cytogenet. 121:73–77. 2000. View Article : Google Scholar : PubMed/NCBI

40 

de Mello RA, Marques DS, Medeiros R and Araújo AM: Epidermal growth factor receptor and K-Ras in non-small cell lung cancer-molecular pathways involved and targeted therapies. World J Clin Oncol. 2:367–376. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Lisiansky V, Naumov I, Shapira S, Kazanov D, Starr A, Arber N and Kraus S: Gene therapy of pancreatic cancer targeting the K-Ras oncogene. Cancer Gene Ther. 19:862–869. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Kalikaki A, Politaki H, Souglakos J, Apostolaki S, Papadimitraki E, Georgoulia N, Tzardi M, Mavroudis D, Georgoulias V and Voutsina A: KRAS genotypic changes of circulating tumor cells during treatment of patients with metastatic colorectal cancer. PLoS One. 9:e1049022014. View Article : Google Scholar : PubMed/NCBI

43 

Campbell PM, Groehler AL, Lee KM, Ouellette MM, Khazak V and Der CJ: K-Ras promotes growth transformation and invasion of immortalized human pancreatic cells by Raf and phosphatidylinositol 3-kinase signaling. Cancer Res. 67:2098–2106. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 38 Issue 5

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
Liu X, Wang Y and Zhao J: MicroRNA‑337 inhibits colorectal cancer progression by directly targeting KRAS and suppressing the AKT and ERK pathways Retraction in /10.3892/or.2022.8394. Oncol Rep 38: 3187-3196, 2017
APA
Liu, X., Wang, Y., & Zhao, J. (2017). MicroRNA‑337 inhibits colorectal cancer progression by directly targeting KRAS and suppressing the AKT and ERK pathways Retraction in /10.3892/or.2022.8394. Oncology Reports, 38, 3187-3196. https://doi.org/10.3892/or.2017.5997
MLA
Liu, X., Wang, Y., Zhao, J."MicroRNA‑337 inhibits colorectal cancer progression by directly targeting KRAS and suppressing the AKT and ERK pathways Retraction in /10.3892/or.2022.8394". Oncology Reports 38.5 (2017): 3187-3196.
Chicago
Liu, X., Wang, Y., Zhao, J."MicroRNA‑337 inhibits colorectal cancer progression by directly targeting KRAS and suppressing the AKT and ERK pathways Retraction in /10.3892/or.2022.8394". Oncology Reports 38, no. 5 (2017): 3187-3196. https://doi.org/10.3892/or.2017.5997