Abstract
Gastric cancer is the fourth most common type of cancer and the second highest leading cause of cancer-related deaths worldwide. It has already been established that miR-133a is involved in gastric cancer. In this study, we investigated the molecular mechanisms by which miR-133a inhibits the proliferation of gastric cancer cells. We analyzed the proliferative capacity of human gastric cancer cells SNU-1 using an MTT assay. Cell apoptosis was determined using flow cytometry. The expression levels of ERBB2, p-ERK1/2, and p-AKT in SNU-1 cells were determined using Western blot analysis. To confirm that ERBB2 is a direct target of miR-133a, a luciferase reporter assay was performed. Results showed that miR-133a overexpression inhibited SNU-1 cell proliferation and increased apoptosis. ERBB2 was a direct target of miR-133a, and it was negatively regulated by miR-133a. Interestingly, ERBB2 silencing has a similar impact to miR-133a overexpression, in that it significantly induced apoptosis and inhibited ERK and AKT activation. Our study showed that miR-133a inhibits the proliferation of gastric cancer cells by downregulating the expression of ERBB2 and its downstream signaling molecules p-ERK1/2 and p-AKT. Therefore, miR-133a might be used as a therapeutic target for treating gastric cancer.
Key words: MicroRNA-133a (miR-133a), Gastric cancer, ERBB2, p-ERK1/2, p-AKT
INTRODUCTION
Gastric cancer is the fourth most common type of cancer and the second highest leading cause of cancer-related deaths globally1. The common presenting symptoms of gastric cancer include weight loss, poor appetite, abdominal pain and discomfort, heartburn, nausea, vomiting, and pallor2. Generally, gastric cancer is diagnosed at an advanced stage, and the prognosis of patients with gastric cancer is often poor3. In addition, the molecular mechanisms involved in the development and progression of gastric cancer are not clear. It is very important to understand the different molecular pathways implicated in gastric cancer, as it would help in the identification of target biomarkers and therefore the development of novel drugs.
It has been shown that the deregulation of oncogenes or tumor suppressors, including microRNAs (miRNAs), leads to the development and progression of gastric cancer4,5. miRNAs are small short noncoding RNAs that are ∼22 nucleotides in length6. miRNAs act at the posttranscriptional level and modulate various cellular processes such as cell proliferation, invasion, and apoptosis7. Upregulation or downregulation of miRNAs affects the expression of various proteins and is therefore involved in tumorigenesis by facilitating proliferation and invasion8.
It has been found that aberrant expression of certain miRNAs, including miR-133a9, is associated with the development and progression of gastric cancer. In addition, the role of miR-133a as a tumor suppressor has been studied in other cancers including ovarian10, non-small cell lung11, osteosarcoma,12 and esophageal squamous cell carcinoma12. Recently, Qiu et al.9 reported that miR-133a inhibits cell proliferation, invasion, migration, and cell cycle progression via targeting transcription factors, especially protein 1, in gastric cancer. As miRNAs target a range of mRNAs, other oncogenes may also be involved in miR-133a-mediated cell proliferation and invasion in gastric cancer.
In the present study, we examined the role of miR-133a in gastric cancer cell proliferation and apoptosis, and the relationship between miR-133a and ERBB2 expressions in gastric cancer cells. ERBB2 (also known as HER2) is a transmembrane tyrosine kinase receptor and a member of the epidermal growth factor receptor (EGFR) family. ERBB2 is expressed in many tissues, including the stomach, breast, heart, and kidney. Overexpression of ERBB2 can promote cell proliferation and inhibit apoptosis, resulting in uncontrolled or excessive cell growth and tumorigenesis13. Thus, overexpression of ERBB2 plays an important role in the development and progression of human cancers.
MATERIALS AND METHODS
Cell Culture
Human gastric cancer cell line SNU-1 was grown in Roswell Park Memorial Institute (RPMI)-1640 medium (Invitrogen, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco, Grand Island, NY, USA), 100 U/ml penicillin sodium, and 100 μg/ml streptomycin sulfate (Sigma-Aldrich, St. Louis, MO USA) at 37°C in a humidified atmosphere of 5% CO2 14.
Cell Proliferation Analysis
The proliferative capacity of SNU-1 cells was determined using 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl-2H-tetrazolium bromide (MTT) assay. On each day of the assay, 100 μl of cells was placed into a 96-well plate, in triplicate, and incubated from day 1 to day 5 at 37°C. To assess cell proliferation, 50 μg of MTT (Sigma-Aldrich) was added to each well and then incubated for 4 h at 37°C. After incubation, 100 μl of 0.04 N HCl in 2-propanol was added and mixed thoroughly. The absorbance was detected at 570 nm using a Microplate Reader (Molecular Devices, Sunnyvale, CA, USA), with a subtraction of background reading at 650 nm15.
Cell Transfection
For precursor miRNA transfection, hsa-miR-183 pre-miR miRNA precursor was obtained from Ambion (No. A25576; Austin, TX, USA). Precursor molecules were transfected with SNU-1 cells using siPORT Amine transfection reagent (Ambion) according to the manufacturer’s instructions. Transfection effects were evaluated after 48 h. This experiment was performed in triplicate. Cells transfected with the Pre-miR™ miRNA precursor molecule (negative control) were used as a control in all transfection experiments.
For ERBB2 knockdown, siRNA-targeted ERBB2 (si-ERBB2) and siRNA negative control (si-NC) were purchased from Invitrogen. Cells were transfected with si-ERBB2 and si-NC using Lipofectamine 2000 transfection reagent (Invitrogen) according to the manufacturer’s instructions.
The efficiency of the overexpression of miR-133a and the silencing of ERBB2 were evaluated by quantitative RT-PCR (qRT-PCR).
Western Blotting
SNU-1 cells were washed twice with phosphate-buffered saline (PBS) and lysed with 1× sodium dodecyl sulfate (SDS)-loading buffer (50 mM Tris-Cl, pH 6.8, 100 mM DTT, 2% SDS, 10% glycerol, and 0.1% bromophenol blue). The protein samples [ERBB2, β-actin, p-AKT, and extracellular signal-regulated kinases 1 and 2 (ERK1/2)] were subjected to SDS-polyacrylamide gel electrophoresis (SDS-PAGE). Western blotting analysis was carried out with the respective primary antibodies: anti-ERBB2 and anti-β-actin (Santa Cruz Biotechnologies, Santa Cruz, CA, USA); anti-p-AKT and anti-ERK1/2 (Cell Signaling Technology, Danvers, MA, USA). The proteins were detected by enhanced chemiluminescence (ECL-plus; Amersham Pharmacia Biotech, Piscataway, NY, USA). In these analyses, β-actin served as a control for ERBB2, phosphorylated ERK1/2 (p-ERK1/2), and p-AKT16.
Apoptosis Analysis
The Dead Cell Apoptosis Kit with Annexin-V FITC and PI (V13242; Invitrogen) was used to perform cell apoptosis analysis according to the manufacturer’s instructions. Briefly, cells (3 × 106) were stained using the apoptosis kit and suspended in annexin V-binding buffer (Invitrogen). The suspended cells were used to perform flow cytometry. Cell apoptosis was determined using the FACSCalibur flow cytometer (BD Biosciences, Heidelberg, Germany). In total, 10,000 cells were analyzed per measurement. Data were analyzed using the FlowJo 10.0.7 software (Treestar Inc., Ashland, OR, USA)17.
qRT-PCR Analysis
Total RNA was extracted with TRIzol reagent (Invitrogen). RNA (500 ng) was polyadenylated and reverse transcribed to cDNA using an NCode miRNA First-Strand cDNA synthesis kit (Invitrogen). cDNA was used as a template for real-time PCR FastStart Universal SYBR green Master (Roche) with the universal reverse primer provided in the kit. RQ-PCR was performed on an Applied Biosystems real-time detection system (Applied Biosystems, Foster City, CA, USA), and the thermocycling parameters were 95°C for 3 min and 40 cycles of 95°C for 15 s followed by 60°C for 30 s. Each sample was run in triplicate and was normalized to U6 snRNA levels [U6 primers 5′-CTT CGG CAG CAC ATA TAC T-3′ (forward) and 5′-AAA ATA TGG AAC GCT TCA CG-3′ (reverse)]. Melting curve analysis was performed to confirm the specificity of the PCR products. The replicates were then averaged, and fold induction was determined by a ΔΔCT-based fold change calculation18.
Luciferase Activity Assay
The 3′-UTR segment of the ERBB2 gene, containing the miR-133a binding site, was amplified through PCR and inserted into the pmirGLO Dual-Luciferase miRNA Target Expression Vector (Promega, Madison, WI, USA). SNU-1 cells were cotransfected with ERBB2 3′-UTR and miR-133a or control using Lipofectamine 2000 (Invitrogen). The luciferase activity was analyzed at 48 h posttransfection using the Dual-Luciferase Reporter Assay System (Promega). For each transfection, the luciferase activity was averaged from three replicates19.
RESULTS
miR-133a Overexpression Inhibits Proliferation of Gastric Cancer Cells
Human gastric cancer cells (SNU-1) were transfected with miR-133a or control, and the expression level of miR-133a was determined using qRT-PCR. The expression level of miR-133a was higher than that of the control (p < 0.001) (Fig. 1A). We then used an MTT assay to determine the proliferative capacity of SNU-1 cells. Overexpression of miR-133a markedly inhibited the proliferation of SNU-1 cells compared to the control (Fig. 1B).
Figure 1.
miR-133a overexpression inhibits the proliferation of gastric cancer cells. (A) Quantitative RT-PCR (qRT-PCR) was performed to determine the relative expression of microRNA-133a (miR-133a) in SNU-1 cells transfected with miRNA-133a or control. (B) MTT assay was used to determine the proliferative capacity of SNU-1 cells, transfected with miR-133a or control, at day 1 to day 5. Data are presented as mean ± standard error. Control: Pre-miR™ miRNA precursor molecule. *p < 0.05; **p < 0.01; ***p < 0.001.
miR-133a Overexpression Promotes Apoptosis of Gastric Cancer Cells
Flow cytometry was used to determine the apoptosis of SNU-1 cells that were transfected with miR1-33a or control. The apoptotic cell rate was higher in the miR-133a-transfected group than those in the control group (p < 0.001) (Fig. 2), indicating that overexpression of miR-133a promoted apoptosis of SNU-1 cells.
Figure 2.
miR-133a overexpression promotes apoptosis of gastric cancer cells. A cell apoptosis assay was performed to examine the rate of apoptosis of SNU-1 cells transfected with miR-133a or control. (A) Flow cytometry analysis. (B) Data from the three samples were quantitated and expressed as mean ± standard error. Control: Pre-miR™ miRNA precursor molecule. ***p < 0.001.
miR-133a Overexpression Downregulates the Expression of ERBB2 in Gastric Cancer Cells
Western blot analysis was used to determine the expression levels of ERBB2 with β-actin expression as control in SNU-1 cells transfected with miR-133a or control. The expression of ERBB2 was fourfold lower in the miR-133a-transfected cells compared to that in control-transfected cells (p < 0.01) (Fig. 3). This indicates that overexpression of miR-133a reduced the expression level of ERBB2 in gastric cancer cells.
Figure 3.
miR-133a overexpression downregulates the expression of ERBB2 in gastric cancer cells. Western blotting was performed to examine the expression level of ERBB2 and β-actin in SNU-1 cells transfected with miR-133a or control. β-Actin served as control for ERBB2. (A) Western blot images. (B) Data from the three samples were quantitated and expressed as mean ± standard error. Control: Pre-miR™ miRNA precursor molecule. **p < 0.01.
ERBB2 Is a Direct Target of miR-133a in Gastric Cancer Cells
The putative seed sequence for miR-133a at the 3′-UTR of ERBB2 was indicated based on bioinformatics analysis (Fig. 4A). To further confirm that ERBB2 is a direct target of miR-133a, a luciferase reporter assay was performed using SNU-1 cells cotransfected with 3′-UTR of ERBB2 and miR-133a, control, or mock. Luciferase activity was markedly decreased in cells cotransfected with miR-133a and 3′-UTR of ERBB2 compared to the control (p < 0.001) and mock samples (p < 0.01) (Fig. 4B). This suggests that ERBB2 is a direct target of miR-133a in SNU-1 cells.
Figure 4.
ERBB2 is a direct target of miR-133a in gastric cancer cells. (A) Seed sequences of miR-133a in 3′-UTR of ERBB2 are indicated. (B) SNU-1 cells were cotransfected with 3′-UTR of ERBB2 and miR-133a, control, or mock, and then luciferase activities were measured. Data are presented as mean ± standard error. Control: Pre-miR™ miRNA precursor molecule. **p < 0.01; ***p < 0.001.
ERBB2 Silencing Promotes Apoptosis of Gastric Cancer Cells
Flow cytometry was performed again to assess the effects of ERBB2 silencing on SNU-1 cell apoptosis. ERBB2 silencing had a similar effect as miR-133a overexpression on SNU-1 cell apoptosis, in that it significantly enhanced apoptotic cell number (p < 0.001) (Fig. 5). These results indicating miR-133a overexpression enhanced gastric cancer cells apoptosis possibly via targeting ERBB2.
Figure 5.
ERBB2 silencing promotes apoptosis of gastric cancer cells. A cell apoptosis assay was performed to examine the rate of apoptosis of SNU-1 cells transfected with ERBB2 siRNA or negative control. (A) Flow cytometry analysis. (B) Data from the three samples were quantitated and expressed as mean ± standard error. ***p < 0.001.
miR-133a Overexpression Reduces the Expression of ERBB2 Downstream Signaling Molecules p-ERK and p-AKT
Western blot analysis was used to determine the expression levels of p-ERK1/2, ERK1/2, p-AKT, and AKT in miR-133a-transfected cells and ERBB2-silenced cells. The expression levels of p-ERK1/2 and p-AKT were markedly lower in the miR-133a-transfected cells compared to those in the control-transfected cells (p < 0.05 and p < 0.01) (Fig. 6A and B). In addition, the expression levels of p-ERK1/2 and p-AKT were both lower in the ERBB2-silenced cells than those in si-NC cells (p < 0.01) (Fig. 6C and D). This indicates that overexpression of miR-133a reduced the expression of ERBB2 and subsequently regulated p-ERK1/2 and p-AKT in SNU-1 cells.
Figure 6.
miR-133a overexpression reduces the expression of ERBB2 downstream signaling molecules phosphorylated extracellular signal-regulated kinase (p-ERK) and p-AKT. (A) Western blotting was performed to examine the expression level of p-ERK1/2, ERK1/2, p-AKT, AKT, and β-actin in SNU-1 cells transfected with miR-133a or control (Pre-miR™ miRNA precursor molecule). (B) The data from three samples were quantitated for p-ERK1/2 and p-AKT and expressed as mean ± standard error. (C) The expression of these proteins in SNU-1 cells transfected with ERBB2 siRNA or negative control was also detected. (D) Quantitative results from three samples were expressed as mean ± standard error. β-Actin served as control for other proteins. *p < 0.05; **p < 0.01.
DISCUSSION
To our knowledge, studies evaluating the association of miR-133a and ERBB2 in gastric cancer have not been conducted or published. In the present study, we showed that miR-133a and ERBB2 play an important role in gastric cancer. We demonstrated that miR-133a overexpression inhibits the proliferation of SNU-1 gastric cancer cells. Importantly, we identified that ERBB2 is a target of miR-133a in SNU-1 gastric cancer cells as overexpression of miR-133a downregulates the expression level of ERBB2 and its downstream signaling molecules p-ERK1/2 and p-AKT in gastric cancer cells. Considering these results, we strongly believe that the inhibitory effect of miR-133a on the proliferation of gastric cancer cells could be via the direct inhibition of ERBB2.
The effects of miR-133a on gastric cancer cell proliferation and apoptosis have been studied previously, albeit using different cell lines and biomarkers9,20. Qiu et al.9 showed that miR-133a inhibits proliferation, migration, invasion, and cell cycle progression in gastric cancer cells (SGC7901, MKN45, and BCG823) via targeting transcription factor Sp1. A similar finding was reported by Lai et al.20, who demonstrated that miR-133a inhibits proliferation and invasion and induces apoptosis in gastric cancer cells (HGC27, GC7901, and AGS) via targeting fascin actin-bundling protein 1 (FSCN1). Our findings are consistent with theirs, as we have demonstrated that overexpression of miR-133a inhibits proliferation and promotes apoptosis of SNU-1 gastric cancer cells by direct inhibition of ERBB2.
It has been shown that ERBB2 is overexpressed in 7% to 34% of gastric cancers21–24 and in many other cancers, including breast25, ovarian25, salivary26, bladder27, and lung cancers28. Furthermore, it has been reported that the expression level of ERBB2 is modulated by several miRNAs, including miR-20529, miR-125b30, and miR-472831. The association between miR-133a and ERBB2 has been reported in many cancers, including breast32, prostate33, and cervical cancer34. In the present study, we demonstrated that miR-133a overexpression downregulates the expression of ERBB2 in gastric cancer cells. In addition, we identified that ERBB2 is a direct target of miR-133a in gastric cancer cells.
To further understand the mechanism by which miR-133a inhibits cancer cells by targeting ERBB2, we analyzed the expression levels of the downstream signaling molecules of ERBB2, namely, p-ERK1/2 and p-AKT. Dimerization of ERBB results in subsequent phosphorylation of some downstream effector proteins, including the ERK1/2 and AKT pathways35. It has been shown that inhibition of ERBB2 phosphorylation results in the inhibition of downstream proteins p-ERK1/2 and p-AKT36. ERK1/2 are members of the mitogen-activated protein kinase super family37. ERK1/2 is activated by the phosphorylation of ERK1/2 (p-ERK1/2), which generally promotes cell proliferation and inhibits apoptosis38. It has been shown that p-ERK1/2 is activated in many cancers, including non-small cell lung39, oral40, and pancreatic cancer41. Protein kinase B (AKT) is a signal transduction protein involved in various cellular processes, including cell proliferation, invasion, and apoptosis. Several studies have revealed that AKT phosphorylation (p-AKT) plays a critical role in various cancers of various organs, including stomach, breast, prostate, and lung42. In the present study, we have demonstrated that miR-133a overexpression reduces the expression levels of p-ERK1/2 and p-AKT in gastric cancer cells. Similar findings have been reported by Zhang et al.43, who demonstrated that overexpression of miR-133a inhibits activation of the AKT and ERK signaling pathways, which contributes to the suppression of hepatocellular carcinoma cell growth. In addition, Wang et al.44 reported that miR-133a suppressed tumor growth and metastasis in colorectal cancer by inhibiting p-ERK.
In conclusion, our data demonstrate that miR-133a inhibits proliferation and induces apoptosis of gastric cancer cells by downregulating the expression of ERBB2 and its downstream signaling molecules p-ERK1/2 and p-AKT. Our findings suggest that miR-133a may be considered to be a potential therapeutic target for the treatment of gastric cancer.
REFERENCES
- 1. Piazuelo MB, Correa P. Gastric cáncer: Overview. Colombia Medica 2013;44:192–201. [PMC free article] [PubMed] [Google Scholar]
- 2. American Cancer Society. Signs and symptoms of stomach cancer; 2016. Available from http://www.cancer.org/cancer/stomachcancer/detailedguide/stomach-cancer-signs-symptoms
- 3. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:33–64. [DOI] [PubMed] [Google Scholar]
- 4. Ambros V. The functions of animal microRNAs. Nature 2004;431:350–5. [DOI] [PubMed] [Google Scholar]
- 5. Li PF, Chen SC, Xia T, Jiang XM, Shao YF, Xiao BX, Guo JM. Non-coding RNAs and gastric cancer. World J Gastroenterol. 2014;20:5411–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Slezak-Prochazka I, Durmus S, Kroesen BJ, Van dBA. MicroRNAs, macrocontrol: Regulation of miRNA processing. RNA 2010;16:1087–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Bartel DP. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 204;116:281–97. [DOI] [PubMed] [Google Scholar]
- 8. Hung CH, Chiu YC, Chen CH, Hu TH. MicroRNAs in hepatocellular carcinoma: Carcinogenesis, progression, and therapeutic target. Biomed Res Int. 2014;2014:486407. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 9. Qiu T, Xin Z, Jian W, Du Y, Xu J, Huang Z, Wei Z, Shu Y, Ping L. MiR-145, miR-133a and miR-133b inhibit proliferation, migration, invasion and cell cycle progression via targeting transcription factor Sp1 in gastric cancer. FEBS Lett. 2014;588:1168–77. [DOI] [PubMed] [Google Scholar]
- 10. Guo J, Xia B, Meng F, Lou G. miR-133a suppresses ovarian cancer cell proliferation by directly targeting insulin-like growth factor 1 receptor. Tumor Biol. 2014;35:1557–64. [DOI] [PubMed] [Google Scholar]
- 11. Wang LK, Hsiao TH, Hong TM, Chen HY, Kao SH, Wang WL, Yu SL, Lin CW, Yang PC. MicroRNA-133a suppresses multiple oncogenic membrane receptors and cell invasion in non-small cell lung carcinoma. PLoS One 2014;9:e96765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Fujiwara T, Katsuda T, Hagiwara K, Kosaka N, Yoshioka Y, Takahashi RU, Takeshita F, Kubota D, Kondo T, Ichikawa H. Clinical relevance and therapeutic significance of microRNA-133a expression profiles and functions in malignant osteosarcoma-initiating cells. Stem Cells 2014;32:959–73. [DOI] [PubMed] [Google Scholar]
- 13. Boku N. HER2-positive gastric cancer. Gastric Cancer 2014;17:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Sabine Schneider GC, Sahin Ugur, Hoy Benjamin, Rieder Gabriele, Wessler Silja. Complex cellular responses of Helicobacter pylori-colonized gastric adenocarcinoma cells. Infect Immun. 2011;79:2362–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Nasonburchenal K, Allopenna J, Bègue A, Stéhelin D, Dmitrovsky E, Martin P. Targeting of PML/RARalpha is lethal to retinoic acid-resistant promyelocytic leukemia cells. Blood 1998;92:1758–67. [PubMed] [Google Scholar]
- 16. Jin Y, Leung WK, Sung JJ, Wu JR. p53-independent pRB degradation contributes to a drug-induced apoptosis in AGS cells. Cell Res. 2005;15:695–703. [DOI] [PubMed] [Google Scholar]
- 17. Brauchle E, Thude S, Brucker SY, Schenkelayland K. Cell death stages in single apoptotic and necrotic cells monitored by Raman microspectroscopy. Sci Rep. 2014;4:4698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Motiño O, Francés DE, Mayoral R, Castro-Sánchez L, Fernández-Velasco M, Boscá L, García-Monzón C, Brea R, Casado M, Agra N. Regulation of microRNA 183 by cyclooxygenase 2 in liver is DEAD-box helicase p68 (DDX5) dependent: Role in insulin signaling. Mol Cell Biol. 2015;35:2554–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Cao LL, Xie JW, Lin Y, Zheng CH, Li P, Wang JB, Lin JX, Lu J, Chen QY, Huang CM. miR-183 inhibits invasion of gastric cancer by targeting Ezrin. Int J Clin Exp Pathol. 2014;7:5582–94. [PMC free article] [PubMed] [Google Scholar]
- 20. Lai C, Chen Z, Li R. MicroRNA-133a inhibits proliferation and invasion, and induces apoptosis in gastric carcinoma cells via targeting fascin actin-bundling protein 1. Mol Med Rep. 2015;12:25–27. [DOI] [PubMed] [Google Scholar]
- 21. Bang YJ, Van CE, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet 2010;376:687–97. [DOI] [PubMed] [Google Scholar]
- 22. Gravalos C, Jimeno A. HER2 in gastric cancer: A new prognostic factor and a novel therapeutic target. Ann Oncol. 2008;19:1523–9. [DOI] [PubMed] [Google Scholar]
- 23. Hofmann M, Stoss O, Shi D, Büttner R, Van dVM, Kim W, Ochiai A, Rüschoff J, Henkel T. Assessment of a HER2 scoring system for gastric cancer: Results from a validation study. Histopathology 2008;52:797–805. [DOI] [PubMed] [Google Scholar]
- 24. Tanner M, Hollmén M, Junttila TT, Kapanen AI, Tommola S, Soini Y, Helin H, Salo J, Joensuu H, Sihvo E. Amplification of in gastric carcinoma: Association with gene amplification, intestinal type, poor prognosis and sensitivity to trastuzumab. Ann Oncol. 2005;16:273–8. [DOI] [PubMed] [Google Scholar]
- 25. Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG, Keith DE, Levin WJ, Stuart SG, Udove J, Ullrich A. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Cancer Invest. 1990;8:219. [DOI] [PubMed] [Google Scholar]
- 26. Stenman G, Sandros J, Nordkvist A, Mark J, Sahlin P. Expression of the ERBB2 protein in benign and malignant salivary gland tumors. Genes Chromosomes Cancer 1991;3:128–35. [DOI] [PubMed] [Google Scholar]
- 27. Sauter G, Moch H, Moore D, Carroll P, Kerschmann R, Chew K, Mihatsch MJ, Gudat F, Waldman F. Heterogeneity of erbB-2 gene amplification in bladder cancer. Cancer Res. 1993;53(10 Suppl):2199–203. [PubMed] [Google Scholar]
- 28. Tateishi M, Ishida T, Mitsudomi T, Kaneko S, Sugimachi K. Prognostic value of c-erbB-2 protein expression in human lung adenocarcinoma and squamous cell carcinoma. Eur J Cancer 1991;27:1372–5. [DOI] [PubMed] [Google Scholar]
- 29. Adachi R, Horiuchi S, Sakurazawa Y, Hasegawa T, Sato K, Sakamaki T. ErbB2 down-regulates microRNA-205 in breast cancer. Biochem Biophys Res Commun. 2011;411:804–8. [DOI] [PubMed] [Google Scholar]
- 30. Shang C, Lu YM, Meng LR. MicroRNA-125b down-regulation mediates endometrial cancer invasion by targeting ERBB2. Med Sci Monit. 2012;18:149–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Schmitt DC, Silva LMD, Zhang W, Liu Z, Arora R, Lim S, Schuler AM, Mcclellan S, Andrews JF, Kahn AG. ErbB2-intronic MicroRNA-4728: A novel tumor suppressor and antagonist of oncogenic MAPK signaling. Cell Death Dis. 2015;6:e1742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Cui W, Zhang S, Shan C, Li Z, Zhou Z. microRNA-133a regulates the cell cycle and proliferation of breast cancer cells by targeting epidermal growth factor receptor through the EGFR/Akt signaling pathway. FEBS J. 2013;280:3962–74. [DOI] [PubMed] [Google Scholar]
- 33. Tao J, Wu D, Xu B, Qian W, Li P, Lu Q, Yin C, Zhang W. microRNA-133 inhibits cell proliferation, migration and invasion in prostate cancer cells by targeting the epidermal growth factor receptor. Oncol Rep. 2012;27:1967–75. [DOI] [PubMed] [Google Scholar]
- 34. Song X, Shi B, Huang K, Zhang W. miR-133a inhibits cervical cancer growth by targeting EGFR. Oncol Rep. 2015;34:1573–80. [DOI] [PubMed] [Google Scholar]
- 35. Akhtar S, Yousif MHM, Chandrasekhar B, Benter IF. Activation of EGFR/ERBB2 via pathways involving ERK1/2, P38 MAPK, AKT and FOXO enhances recovery of diabetic hearts from ischemia-reperfusion injury. PLoS One 2012;7:1–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Xia W, Liu LH, Ho P, Spector NL. Truncated ErbB2 receptor (p95ErbB2) is regulated by heregulin through heterodimer formation with ErbB3 yet remains sensitive to the dual EGFR/ErbB2 kinase inhibitor GW572016. Oncogene 2004;23:646–53. [DOI] [PubMed] [Google Scholar]
- 37. Mebratu Y, Tesfaigzi Y. How ERK1/2 activation controls cell proliferation and cell death is subcellular localization the answer? Bioorg Med Chem Lett. 2009;8:1168–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Lu Z, Xu S. ERK1/2 MAP kinases in cell survival and apoptosis. IUBMB Life 2006;58:621–31. [DOI] [PubMed] [Google Scholar]
- 39. Vicent S, Lópezpicazo JM, Toledo G, Lozano MD, Torre W, Garciacorchón C, Quero C, Soria JC, Martínalgarra S, Manzano RG. ERK1|[sol]|2 is activated in non-small-cell lung cancer and associated with advanced tumours. Br J Cancer 2004;90:1047–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Gkouveris I, Nikitakis N, Karanikou M, Rassidakis G, Sklavounou A. Erk1/2 activation and modulation of STAT3 signaling in oral cancer. Oncol Rep. 2014;32:2175–82. [DOI] [PubMed] [Google Scholar]
- 41. Wang M, Lu X, Dong X, Hao F, Liu Z, Ni G, Chen D. pERK1/2 silencing sensitizes pancreatic cancer BXPC-3 cell to gemcitabine-induced apoptosis via regulating Bax and Bcl-2 expression. World J Surg Oncol. 2015;13:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Cicenas J. The potential role of Akt phosphorylation in human cancers. Int J Biol Markers 2008;23:1–9. [DOI] [PubMed] [Google Scholar]
- 43. Zhang W, Liu K, Liu S, Ji B, Wang Y, Liu Y. MicroRNA-133a functions as a tumor suppressor by targeting IGF-1R in hepatocellular carcinoma. Tumor Biol. 2015;36:1–10. [DOI] [PubMed] [Google Scholar]
- 44. Wang H, An H, Wang B, Liao Q, Li W. miR-133a represses tumour growth and metastasis in colorectal cancer by targeting LIM and SH3 protein 1 and inhibiting the MAPK pathway. Eur J Cancer 2013;49:3924–35. [DOI] [PubMed] [Google Scholar]






