ADAM9 functions as a promoter of gastric cancer growth which is negatively and post-transcriptionally regulated by miR-126

  • Authors:
    • Junqing Wang
    • Yunyun Zhou
    • Xiaochun Fei
    • Xuehua Chen
    • Jiqi Yan
    • Bingya Liu
    • Zhenggang Zhu
  • View Affiliations

  • Published online on: February 16, 2017     https://doi.org/10.3892/or.2017.5460
  • Pages: 2033-2040
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

A disintegrin and metalloproteinase domain 9 (ADAM9) is a membrane-anchored protein implicated in cell-cell and cell-matrix interactions, including the process of tumorigenesis. However, the role of ADAM9 in gastric cancer (GC) has not been clearly illustrated. In the present study, we found aberrant overexpression of ADAM9 in both GC tissues and cell lines. The expression of ADAM9 was significantly correlated with patient clinicopathological features including tumor size, local invasion, lymph node metastasis and tumor‑node‑metastasis (TNM) stage. Knockdown of ADAM9 in GC SGC-7901 cells, which presented the highest ADAM9 expression among the cell lines, induced a dramatic suppression of cell proliferation along with the arrest of the cell cycle in the G0/G1 phase. Furthermore, we validated that the 3' untranslated region of ADAM9 mRNA could be bound by miR-126, a suppressor in GC, and overexpression of miR-126 significantly downregulated ADAM9 in the GC cells. In conclusion, ADAM9 functions as a tumor promoter in GC by modulating GC cell proliferation. ADAM9 could possibly be regarded as a biomarker for GC diagnosis and prevention. Moreover, as directly targeted by miR-126 in GC, ADAM9 may be a potential target for GC therapeutic treatment which warrants intensive study.

Introduction

As one of the most common digestive malignancies in humans worldwide, gastric cancer (GC) exhibits aggressive malignant behavior. GC patients have a current 5-year survival rate of only ~24% (1). At present, most GC patients are clinically diagnosed in the advanced stages and the median survival time of GC patients with local invasion or metastasis is less than 12 months (2). The challenges of discovering associated tumor markers and understanding the mechanisms of GC initiation, progression and metastasis, are the keys to diagnose, prevent and treat GC appropriately in the early stages as well as develop targeted treatment (3)

A disintegrin and metalloproteinases (ADAMs), are a family of proteins which play a pivotal role in the proteolytic process implicated in cell-cell and cell-matrix interactions (4). Members of the ADAM family are divided into two groups: membrane-anchored and the secreted-type proteins (ADAMST) (5). Noteworthy, membrane-anchored ADAMs belong to type I trans-membrane proteins, which consist of a disintegrin-containing extracellular domain and a metalloproteinase domain (6). The functions of ADAMs are multiple and they are mainly involved in the proteolytic processing of trans-membrane proteins, contributing to various pathologies, including cell adhesion, cell signaling pathways and human tumors (7). Ectodomain shedding is a critical process conducted through proteolytic cleavage of membrane-anchored molecules into the extracellular microenvironment, and is related with tumorigenesis (8). ADAMs also participate in the ectodomain shedding process by undergoing cleavage close to the trans-membrane domains (9).

A disintegrin and metalloproteinase domain 9 (ADAM9), one of the ADAM family members, has been found and described in a variety of solid tumors with overexpression and dysregulation, in glioma, prostate, colon and breast cancer, which suggest ADAM9 as an important molecule involved in tumorigenesis (1013). However, in GC, the role of ADAM9 is still elusive and deserves to be elucidated.

MicroRNAs (miRs) are a class of non-coding RNAs consisting of 22 nucleotides, which recognize a specific sequence of messenger RNAs (mRNAs) on the 3′ untranslated region (3′UTR) as targets, and consequentially induce either inhibition of mRNA translation or degeneration of the targeted mRNAs (14). miRs functionally regulate and control various pivotal pathophysiological processes post-transcriptionally, including tumor initiation and progression (15,16). For instance, miR-146 suppresses gallbladder cancer cell proliferation by targeting epidermal growth factor (EGF) (17); miR-99a inhibits cell growth in osteosarcoma by negatively regulating TNFAIP8 (18). In GC, numerous miRs exhibit complex and marked effects either by suppressing or promoting tumor progression. For example, miR-30a reportedly targets RPA1 in GC cells and consequently suppresses the growth of GC cells with cell cycle arrest (19); on the contrary, by decreasing the expression of FBW7 through direct post-transcriptional regulation, miR-363 significantly promotes the cell proliferation and chemotherapy resistance in GC (20). Certainly, miRs provide us with enormous possibilities to discover new targets in GC prevention, diagnosis and therapeutic treatment.

Based on our previous research and studies from other authors, microRNA-126 (miR-126) appears to be an extremely important microRNA that functions as a suppressor in GC progression and which is frequently downregulated in both GC tissues and cell lines (2124). As interactions between miRs and their targets show complex networking, an individual miR could target various mRNAs in multiple pathophysiological processes and an individual mRNA could possibly be targeted by different miRs simultaneously. Intensive detection of miR-126-targeted molecules in GC is valuable to provide us with a clear sense of how this suppressor functions in GC and also to provide us with sufficient evidence in order to find new antitumor targets.

In the present study, by evaluating 76 pairs of GC tissues compared with the adjacent non-cancerous tissues and 4 GC cell lines (SGC-7901, MKN-45, MKN-28 and SUN-16), we ascertained that ADAM9 was aberrantly overexpressed in GC. High levels of ADAM9 were significantly correlated with GC clinicopathological features, such as tumor size, local invasion, lymph node metastasis and tumor stage, which suggest a poorer prognosis for patients with a high ADAM9 level. Knockdown of ADAM9 expression in SGC-7901 cells significantly suppressed cell proliferation and arrested the cell cycle at the G0/G1 phase. Moreover, by applying a dual-luciferase reporter assay, we discovered that miR-126 could directly bind to the 3′UTR of ADAM9 mRNA and markedly downregulate ADAM9 expression. The promotional effect of ADAM9 on GC cell proliferation was revealed through overexpression of miR-126. All the aforementioned findings illustrate that ADAM9 functions as a tumor promoter in GC and exerts a tumor-suppressive function.

Materials and methods

Cell culture and surgical specimens

The immortalized gastric epithelium cell line and 4 GC cell lines (SGC-7901, MKN-45, MKN-28 and SUN-16) were purchased from the Shanghai Institutes for Biological Sciences, Chinese Academy of Science (Shanghai, China). SGC-7901 cells overexpressing miR-126 (SGC-7901/miR-126) and the negative control (NigmiR) were constructed in our previous study (25). All cells were cultured in RPMI-1640 medium supplemented with 10% heat-inactivated fetal bovine serum (FBS), 100 µg/ml streptomycin and 100 U/ml penicillin in a humidified cell incubator at 37°C with an atmosphere of 5% CO2.

Seventy-six pairs of GC specimens and adjacent non-cancerous tissues were collected from GC patients who had undergone a radical gastrectomy without preoperative therapy at the Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine during 2012–2014. Ethical approval was obtained from the Research Medical Ethics Committee of Rujin Hospital, Shanghai Jiao Tong University School of Medicine.

Immunohistochemistry and western blot analyses

Antibodies against ADAM9 and GAPDH (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), and horseradish peroxidase-conjugated secondary antibody (Abcam, Cambridge, MA, USA) were prepared. Immunohistochemical analysis was carried out using antibody against ADAM9 following the manufacturers instructions (1:50), and the tissues were individually examined by two professional pathologists. GAPDH was used as a loading control.

RIPA buffer containing a protease inhibitor cocktail was used to lyse the cells, and the protein concentration was measured by BCA Protein Assay kit (both from Pierce, Rockford, IL, USA). Proteins were electrophoresed and electrotransferred. Antibodies against ADAM9 (1:1,000) and GAPDH (1:5,000) were probed, and a horseradish peroxidase-conjugated secondary antibody was used for further probing. The protein quantity was detected using GAPDH as a loading control.

RNA isolation and real-time qPCR assay

Total RNA was extracted from the cell lines using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturers instructions. The first-strand cDNA was synthesized using a HighCapacity cDNA Reverse Transcription kit (ABI, Foster City CA, USA). RT-primers of ADAM9 mRNAs were synthesized as follows: forward, 5′-GGAAGAGTGTGACTGTGGTAC-3′ and reverse, 5′-CCTCGGCATAAAGTACCTCC-3′ by Sangon Biotech Co. (Shanghai, China). Real-time quantitative polymerase chain reaction (qRT-PCR) was performed according to TaqMan Gene Expression Assays protocol (ABI).

Cell transfection

SGC-7901 cells were transfected with pGU6/Neo vectors (GenePharma, Shanghai, China) containing shRNA suppressing ADAM9 translation or non-containing ones. Cells were cultured and selected in medium containing 400 µg/ml G418 (Santa Cruz Biotechnology, Inc.). The stable transfected cells aforementioned were assessed by qRT-PCR and western blot analysis compared with the negative control cells. All cells were cultured and maintained in medium containing 200 µg/ml G418.

Recombinant adenovirus Ad5/F35 (Ad5/F35-ADAM9) was constructed for overexpressing ADAM9 and Ad5/F35-Null was used as a negative control (GenePharma). SGC-7901 cells overexpressing miR-126 (SGC-7901/miR-126) and the negative control (NigmiR) were further transfected with Ad5/F35-ADAM9 or Ad5/F35-Null, and were assessed.

Cell proliferation assay and cell cycle analysis

SGC-7901 cells (1×106) stably transfected or the negative control cells were cultured in 96-well microtiter plates in triplicate and incubated for 5 days at 37°C with an atmosphere of 5% CO2. The OD was measured using microplate computer software (Bio-Rad Laboratories, Inc., Hercules, CA, USA) according to the protocol from the Cell Counting Kit-8 (CCK-8) assay kit (Dojindo, Tokyo, Japan). The curves for cell proliferation were plotted.

The aforementioned cells were treated with ethanol fixation, RNase A treatment and propidium iodide staining, and were then detected under flow cytometry by FACSCalibur (Becton-Dickinson, Franklin Lakes, NJ, USA). Cell populations at the G0/G1, S and G2/M phases were quantified by ModFit software (Becton-Dickinson) excluding a calculation of cell debris and fixation artifacts.

Dual-luciferase reporter assay

ADAM9 was predicted as a potential target of miR-126 by bioinformatics analysis (microcosm, http://mirecords.biolead.org). A 206 bp sequence from the 3′UTR of ADAM9 mRNA including the putative miR-126 binding site was selected as follows: 5′-uagagaaauuaauuuaaau uagaauuucuauuaugaaucaugugaaagcaugacauucguucacaauagca cuauuuuaaauaaauuauaagcuuuaagguacgaaguauuuaaugaucuaau caaauauguugauucauggcuauaauaaagcaggagcaauuauaaaaucuuc aaucaauugaacuuuuacaaaaccacuug-3′. The corresponding mutant sequence was constructed by Sangon Biotech Co., as follows: 5′-aacacauaauuaauaauuuuaacaauuacaaauuucauugaag aguauggaagucuuaccuacucuaaaccucaaauauuauuuauuaaaauggu auuacgaagguacuuuauuaaacaacaauugauaaaagaucaaugaagccaaa auuuauggacguggauuaaaauauuguacuaacuaaucaucauauucuauag cucauc-3′. These sequences were cloned into pMIR-REPORT luciferase vectors (Promega, Madison, WI, USA), containing Firefly luciferase, and pRL-TK vectors containg Renilla luciferase which were used as a control. SGC-7901 cells overexpressing miR-126 or the negative control were co-transfected with the aforementioned vectors and the luciferase activity was measured using a Dual-Glo Luciferase assay system (Promega) 48 h post-transfection.

Statistical analysis

Statistical analysis was carried out using SPSS 18.0. P-values were calculated using a paired t-test and Fisher's exact test. P-values <0.05 were considered to indicate a statistically significant result.

Results

ADAM9 is overexpressed in GC tumor specimens

Seventy-six paired specimens from GC tumors and adjacent non-cancerous tissues were examined using IHC. According to the expression intensity of ADAM9, these cases were separated into two groups: an ADAM9 low expression and an ADAM9 high expression group. Among the tumor specimens, 72.3% (55/76) of the cases presented high levels of ADAM9 expression, and only 27.6% (21/76) of the cases expressed a relatively lower level of ADAM9. On the contrary, in non-cancerous tissues, high expression of ADAM9 was detected in a small portion of the cases (13.2%, 10/76) and 86.8% (66/76) of the specimens presented a low level of ADAM9 expression. This proves that ADAM9 is expressed frequently higher in GC tissues when compared with adjacent non-cancerous tissues (Fig. 1).

ADAM9 expression is upregulated and miR-126 is downregulated in GC cell lines

Analysis of the transcription levels of miR-126 in the 4 GC cell lines (SGC-7901, MKN-28, MKN-45 and SUN-16) using qRT-PCR revealed that the expression levels of miR-126 were significantly downregulated in the GC cell lines compared with the GES-1 cells (P<0.05) (Fig. 2A). In contrast, the mRNA levels of ADAM9 were upregulated in the 4 different GC cell lines compared with the GES-1 cells (P<0.05) (Fig. 2B). Similarly, western blot analysis demonstrated that the protein levels of ADAM9 were significantly higher in the GC cell lines than that in the GES-1 cells (Fig. 2C). These results were consistent with the observations obtained in the IHC analysis of the tumor tissues.

High expression of ADAM9 is correlated with GC clinicopathological features

The correlation between the expression of ADAM9 and the clinicopathological features of the 76 GC cases was analyzed. According to Table I, there was no significant correlation between ADAM9 expression and patient age, gender or tumor location. However, a significant trend towards a larger tumor size (P<0.05), deeper local invasion (P<0.05), more frequent lymph node metastasis (P<0.05) and more advanced tumor-node-metastasis (TNM) stage (P<0.05) in cases with higher expression levels indicates a correlation between ADAM9 overexpression and certain GC clinicopathological features.

Table I.

Correlation between ADAM9 expression and clinicopathological features of the 76 GC cases.

Table I.

Correlation between ADAM9 expression and clinicopathological features of the 76 GC cases.

ADAM9 expression

Clinicopathological parametersLow (n=21)High (n=55) P-valuea
Age (years) 0.592
  ≤60817
  >601338
Gender 0.196
  Male1221
  Female934
Tumor diameter (cm) 0.023
  ≤51519
  >5836
Location 0.778
  Distal third1639
  Middle or proximal third516
Histological classification 0.792
  Poorly-differentiated adenocarcinoma722
  Middle/well-differentiated adenocarcinoma46
  Signet ring cell carcinoma36
  Mucinous adenocarcinoma13
Local invasion 0.016
  T1,T21316
  T3,T4839
Lymph node metastasis 0.030
  No1215
  Yes940
TNM stage 0.046
  I,II1012
  III,IV1143

[i] ADAM9 expression level associated with clinicopathological features in 76 GC patients, including age, gender, tumor size, tumor location, histological classification, local invasion, lymph node metastasis and TNM stage. Statistical significance was assessed by Fisher's exact test. ADAM9, a disintegrin and metalloproteinase domain 9; GC, gastric cancer; TNM, tumor-node-metastasis.

Knockdown of ADAM9 suppresses cell proliferation and arrests the cell cycle in SGC-7901 cells

SGC-7901 cells, which expressed the highest level of ADAM9 among the 4 GC cell lines, were selected and transfected with pGU6/Neo vectors to knock down the expression of ADAM9. We verified the transfection effect through qRT-PCR and western blot analysis (Fig. 3).

We conducted flow cytometric analysis and found that, the cell cycle of SGC-7901 cells was significantly arrested at the G0/G1 phase when ADAM9 was knocked down (Fig. 4A and B). The percentage of the SGC-7901 cells in the G0/G1 phase was increased from 50.89 to 64.78% (P<0.01). The S phase was decreased from 30.63 to 18.05%, and the G2/M phase was decreased from 28.16 to 17.16% (Fig. 4B). Meanwhile, as the CCK-8 assay demonstrated, we observed a significant decrease in cell proliferation in the ADAM9-knockdown SGC-7901 cells as the P-value was <0.01 for day 1 and the P-value was <0.05 for days 2–4 (Fig. 4C). These results indicate that knockdown of ADAM9 in SGC-7901 cells significantly impacts tumor cell growth.

ADAM9 is a direct target post-transcriptionally regulated by miR-126 in GC cells

We used microcosm, bioinformatics analysis employing an online prediction software, to predict ADAM9 as a potential target of miR-126 (Fig. 5A). Demonstration of the direct interaction between ADAM9 mRNA and miR-126 was carried out using a dual-luciferase reporter assay. Luciferase reporter vectors containing a 206-bp 3′UTR sequence of ADAM9 (WT-UTR) and the corresponding control luciferase vectors containing a mutated miR-126 target site (MUT-UTR) were constructed. As shown in Fig. 5B, overexpression of miR-126 in the SGC-7901 cells (SGC-7901/miR-126) significantly decreased the luciferase signal of ADAM9/pMIR/WT, compared with the negative control (SGC-7901/NigmiR). In addition, this suppressive effect induced by miR-126 was significantly abolished in the SGC-7901 cells with the putative binding site of mutated miR-126 (Fig. 5B). Moreover, both the mRNA level and the protein expression of ADAM9 were significantly decreased in SGC-7901/miR-126 cells (Fig. 5C and D). Collectively, these results revealed that ADAM9 is a direct target of miR-126 in GC and was post-transcriptionally downregulated by miR-126.

Introduction of ADAM9 in SGC-7901 cells reverses the phenotype of growth arrest induced by overexpression of miR-126

The aforementioned results indicate that ADAM9 is one of the direct targets suppressed by miR-126 in GC. Based on this, we assumed that introducing ADAM9 in the miR-126-overexpressing SGC-7901 cells would at least relatively reverse the phenotypes caused by overexpression of miR-126. Recombinant adenovirus Ad5/F35 was applied in the present study to upregulate the expression of ADAM9 in the SGC-7901 cells. As shown in Fig. 6, the inhibitory effect on SGC-7901 cell proliferation by miR-126 was significantly reversed when ADAM9 expression was increased. Meanwhile, the cell cycle arrest effect induced by miR-126 was also reverses after ADAM9 introduction. Thus, ADAM9 is a molecule that promotes GC cell growth, which may be targeted by miR-126 as a part of its post-transcriptional mechanism for suppressing GC.

Discussion

The initiation and development of human tumors are under the control of a multitude of factors. With respect to gastric cancer (GC), a large number of molecules and their relative mechanisms, which are involved in GC tumorigenesis have been discovered. A variety of molecules participate in the process of GC with different types of mechanisms such as signaling pathways and post-transcriptional regulation, which are accompanied by huge networking between the molecules discovered or those which need to be further studied. For example, nucleophosmin (NPM)/B23 was found to be aberrantly overexpressed and regulated in GC, functioning as an indicator of GC associated with advanced TNM stage, poor prognosis and recurrence (26). Plant homeodomain finger protein 10 (PHF10) was ascertained as a promoter of GC enhancing the ability of cell proliferation (27). In addition, concerning non-coding RNAs, for example, miR-223 targets EPB41L3 in GC and promotes tumor cell invasion and migration (28). miR-107 downregulates CDK6 mRNA, and induces inhibition of GC cell invasion (29).

ADAM9 is a member of the ADAM family anchored to the membrane, and is related to various human tumors as we previously mentioned. In pancreatic ductal adenocarcinoma, ADAM9 is upregulated at the mRNA level and over 70% of pancreatic carcinomas present high protein levels (30). In prostate cancer, ADAM9 appears to be regulated, and inhibition of ADAM9 in vivo significantly suppressed tumor growth (10). ADAM9 was also found to modulate tumor-stromal cell interaction and sequentially promote cell motility in human hepatocellular carcinoma and lung cancer (31,32). Moreover, several reports have demonstrated high expression of ADAM9 in GC (33,34). However, the role of ADAM9 in GC and its relative upstream regulatory mechanisms remain unclear.

In the present study, we validated the expression of ADAM9 in 76 GC tumor tissues and cell lines. ADAM9 exhibited an obvious high expression level in GC tissues. By analyzing the clinicopathological features of the 76 patients, we found that high levels of ADAM9 showed a significant correlation with larger tumor size, deeper local invasion, more frequent lymph node metastasis and more advanced tumor stages. Thus, ADAM9 is an independent factor correlated with poor GC outcomes and prognosis.

Simultaneously, qRT-PCR and western blot analysis showed that expression of ADAM9 was significantly higher at both the mRNA and protein levels in GC cells than levels in GES-1 cells. Among the 4 GC cell lines, SGC-7901 cells presented the highest level of ADAM9. Considering what we observed from the specimens and cells, we believe that ADAM9 is a potential functional molecule in GC progression. To verify the function of ADAM9 in GC cells, we selected SGC-7901 cells and knocked down the ADAM9 expression by stable transfection. An in vitro cellular functional experiment was carried out. As we had hypothesized, when ADAM9 was knocked down in the SGC-7901 cells, the cell proliferation ability was markedly suppressed. Additionally, flow cytometric analysis demonstrated an obvious arrest of the cell cycle in GC cells at the G0/G1 phase, indicating that inhibition of ADAM9 effectively suppressed the growth of GC cells.

Furthermore, we speculated the mechanism by which ADAM9 promotes the cell growth of GC. Through the use of online bioinformatics tools, we found that a potential binding site for miR-126 exists in the 3′UTR of ADAM9 mRNA. miR-126 is an important non-coding RNA, which has been confirmed as a suppressor of GC growth. In our recent study we found that miR-126 exerts its tumor-suppressive function in various types of cancer by targeting different mRNAs, and in GC, CRKL, LAT-1, VEGF-A and CADM-1, are all targets of miR-126. We further speculated as to whether ADAM9 is a functional target of miR-126 in GC. We then conducted a dual-luciferase reporter assay to verify the direct interaction between ADAM9 and miR-126. A combination of miR with a specific 3′UTR of the target mRNA could cause an impact on luciferase gene expression. As the results showed, overexpression of miR-126 significantly decreased the luciferase signal intensity. On the contrary, mutated 3′UTR of ADAM9 mRNA failed to bind with miR-126 and presented no significant change in luciferase signal intensity. Thus, there is a direct correlation between miR-126 and the 3′UTR of ADAM9 mRNA.

To further understand whether miR-126 suppresses GC through ADAM9, we ectopically expressed ADAM9 in SGC-7901 cells overexpressing miR-126. As expected, by introducing ADAM9, the cell proliferation suppression induced by miR-126 was significantly reversed. In addition, the percentage of the cells arrested in the G0/G1 phase was notably decreased when ADAM9 was overexpressed. All the aforementioned results suggest that ADAM9 is one of the direct targets regulated by miR-126 in GC cells and by which miR-126 conducts its potential tumor suppressive function in GC.

In conclusion, according to the findings in the present study, we conclude that ADAM9 is one of the direct targets post-transcriptionally modulated by miR-126, which helps us to understand the tumor-suppressive mechanism of miR-126. High levels of ADAM9 in GC are correlated with a poor prognosis and aberrant overexpression of ADAM9 leads to the promotion of GC cell growth. ADAM9 should be considered as a potential target for GC prevention, diagnosis and therapeutic treatment.

Acknowledgements

The authors thank Qucai, Minmin Shi, Hui Ye and Jun Ji for providing valuable technical supports and assistance. The present study was kindly supported by grants from the following: the National Natural Science Foundation of China (nos. 81602544 and 81372187), and the Liu Haoqing Foundation for Medicine, Ruijin Hospital Shanghai.

References

1 

Russo F, Linsalata M and Orlando A: Probiotics against neoplastic transformation of gastric mucosa: Effects on cell proliferation and polyamine metabolism. World J Gastroenterol. 20:13258–13272. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Wang W, Sun XW, Li CF, Lv L, Li YF, Chen YB, Xu DZ, Kesari R, Huang CY, Li W, et al: Comparison of the 6th and 7th editions of the UICC TNM staging system for gastric cancer: Results of a Chinese single-institution study of 1,503 patients. Ann Surg Oncol. 18:1060–1067. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Bringeland EA, Wasmuth HH, Fougner R, Mjønes P and Grønbech JE: Impact of perioperative chemotherapy on oncological outcomes after gastric cancer surgery. Br J Surg. 101:1712–1720. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Bahudhanapati H, Bhattacharya S and Wei S: Evolution of vertebrate Adam genes; Duplication of testicular Adams from ancient Adam9/9-like loci. PLoS One. 10:e01362812015. View Article : Google Scholar : PubMed/NCBI

5 

Kelwick R, Desanlis I, Wheeler GN and Edwards DR: The ADAMTS (A Disintegrin and Metalloproteinase with Thrombospondin motifs) family. Genome Biol. 16:1132015. View Article : Google Scholar : PubMed/NCBI

6 

Amendola RS, Martin AC, Selistre-de-Araújo HS, Paula-Neto HA, Saldanha-Gama R and Barja-Fidalgo C: ADAM9 disintegrin domain activates human neutrophils through an autocrine circuit involving integrins and CXCR2. J Leukoc Biol. Mar 12–2015.(Epub ahead of print). pii: jlb.3A0914-455R. View Article : Google Scholar : PubMed/NCBI

7 

Peduto L: ADAM9 as a potential target molecule in cancer. Curr Pharm Des. 15:2282–2287. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Quach HT, Hirano S, Fukuhara S, Watanabe T, Kanoh N, Iwabuchi Y, Usui T and Kataoka T: Irciniastatin A induces potent and sustained activation of extracellular signal-regulated kinase and thereby promotes ectodomain shedding of tumor necrosis factor receptor 1 in human lung carcinoma A549 cells. Biol Pharm Bull. 38:941–946. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Jones JC, Rustagi S and Dempsey PJ: ADAM proteases and gastrointestinal function. Annu Rev Physiol. 78:243–276. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Liu CM, Hsieh CL, He YC, Lo SJ, Liang JA, Hsieh TF, Josson S, Chung LW, Hung MC and Sung SY: In vivo targeting of ADAM9 gene expression using lentivirus-delivered shRNA suppresses prostate cancer growth by regulating REG4 dependent cell cycle progression. PLoS One. 8:e537952013. View Article : Google Scholar : PubMed/NCBI

11 

Li J, Ji Z, Qiao C, Qi Y and Shi W: Overexpression of ADAM9 promotes colon cancer cells invasion. J Invest Surg. 26:127–133. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Micocci KC, Martin AC, Montenegro CF, Durante AC, Pouliot N, Cominetti MR and Selistre-de-Araujo HS: ADAM9 silencing inhibits breast tumor cell invasion in vitro. Biochimie. 95:1371–1378. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Chen CM, Hsieh YH, Hwang JM, Jan HJ, Hsieh SC, Lin SH and Lai CY: Fisetin suppresses ADAM9 expression and inhibits invasion of glioma cancer cells through increased phosphorylation of ERK1/2. Tumour Biol. 36:3407–3415. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Kiselev FL: MicroRNA and cancer. Mol Biol. 48:232–242. 2014.(In Russian).

15 

Kong YW, Ferland-McCollough D, Jackson TJ and Bushell M: microRNAs in cancer management. Lancet Oncol. 13:e249–e258. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Connerty P, Ahadi A and Hutvagner G: RNA Binding proteins in the miRNA pathway. Int J Mol Sci. 17:E312015. View Article : Google Scholar : PubMed/NCBI

17 

Cai J, Xu L, Cai Z, Wang J, Zhou B and Hu H: MicroRNA-146b-5p inhibits the growth of gallbladder carcinoma by targeting epidermal growth factor receptor. Mol Med Rep. 12:1549–1555. 2015.PubMed/NCBI

18 

Xing B and Ren C: Tumor-suppressive miR-99a inhibits cell proliferation via targeting of TNFAIP8 in osteosarcoma cells. Am J Transl Res. 8:1082–1090. 2016.PubMed/NCBI

19 

Zou Z, Ni M, Zhang J, Chen Y, Ma H, Qian S, Tang L, Tang J, Yao H, Zhao C, et al: miR-30a can inhibit DNA replication by targeting RPA1 thus slows down the proliferation of cancer cells. Biochem J. 473:2131–2139. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Zhang PF, Sheng LL, Wang G, Tian M, Zhu LY, Zhang R, Zhang J and Zhu JS: miR-363 promotes proliferation and chemo-resistance of human gastric cancer via targeting of FBW7 ubiquitin ligase expression. Oncotarget. 7:35284–35292. 2016.PubMed/NCBI

21 

Wang J, Chen X, Li P, Su L, Yu B, Cai Q, Li J, Yu Y, Liu B and Zhu Z: CRKL promotes cell proliferation in gastric cancer and is negatively regulated by miR-126. Chem Biol Interact. 206:230–238. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Wang J, Chen X, Su L, Li P, Cai Q, Liu B, Wu W and Zhu Z: MicroRNA-126 inhibits cell proliferation in gastric cancer by targeting LAT-1. Biomed Pharmacother. 72:66–73. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Yang Z, Wang R, Zhang T and Dong X: MicroRNA-126 regulates migration and invasion of gastric cancer by targeting CADM1. Int J Clin Exp Pathol. 8:8869–8880. 2015.PubMed/NCBI

24 

Chen H, Li L, Wang S, Lei Y, Ge Q, Lv N, Zhou X and Chen C: Reduced miR-126 expression facilitates angiogenesis of gastric cancer through its regulation on VEGF-A. Oncotarget. 5:11873–11885. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Feng R, Chen X, Yu Y, Su L, Yu B, Li J, Cai Q, Yan M, Liu B and Zhu Z: miR-126 functions as a tumour suppressor in human gastric cancer. Cancer Lett. 298:50–63. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Wong JC, Hasan MR, Rahman M, Yu AC, Chan SK, Schaeffer DF, Kennecke HF, Lim HJ, Owen D and Tai IT: Nucleophosmin 1, upregulated in adenomas and cancers of the colon, inhibits p53-mediated cellular senescence. Int J Cancer. 133:1567–1577. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Wet M, Liu JY, Lv X, Nei H, Liu BY, Zhu ZG, Yang ZY and Gu QL: Preparation of PHF10 antibody and analysis of PHF10 expression gastric cancer tissues. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 26:874–876. 2010.PubMed/NCBI

28 

Li X, Zhang Y, Zhang H, Liu X, Gong T, Li M, Sun L, Ji G, Shi Y, Han Z, et al: miRNA-223 promotes gastric cancer invasion and metastasis by targeting tumor suppressor EPB41L3. Mol Cancer Res. 9:824–833. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Feng L, Xie Y, Zhang H and Wu Y: miR-107 targets cyclin-dependent kinase 6 expression, induces cell cycle G1 arrest and inhibits invasion in gastric cancer cells. Med Oncol. 29:856–863. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Yamada D, Ohuchida K, Mizumoto K, Ohhashi S, Yu J, Egami T, Fujita H, Nagai E and Tanaka M: Increased expression of ADAM 9 and ADAM 15 mRNA in pancreatic cancer. Anticancer Res. 27:793–799. 2007.PubMed/NCBI

31 

Zhang J, Chen N, Qi J, Zhou B and Qiu X: HDGF and ADAM9 are novel molecular staging biomarkers, prognostic biomarkers and predictive biomarkers for adjuvant chemotherapy in surgically resected stage I non-small cell lung cancer. J Cancer Res Clin Oncol. 140:1441–1449. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Tao K, Qian N, Tang Y, Ti Z, Song W, Cao D and Dou K: Increased expression of a disintegrin and metalloprotease-9 in hepatocellular carcinoma: Implications for tumor progression and prognosis. Jpn J Clin Oncol. 40:645–651. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Kim JM, Jeung HC, Rha SY, Yu EJ, Kim TS, Shin YK, Zhang X, Park KH, Park SW, Chung HC, et al: The effect of disintegrin-metalloproteinase ADAM9 in gastric cancer progression. Mol Cancer Ther. 13:3074–3085. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Carl-McGrath S, Lendeckel U, Ebert M, Roessner A and Röcken C: The disintegrin-metalloproteinases ADAM9, ADAM12, and ADAM15 are upregulated in gastric cancer. Int J Oncol. 26:17–24. 2005.PubMed/NCBI

Related Articles

Journal Cover

April-2017
Volume 37 Issue 4

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
Wang J, Zhou Y, Fei X, Chen X, Yan J, Liu B and Zhu Z: ADAM9 functions as a promoter of gastric cancer growth which is negatively and post-transcriptionally regulated by miR-126. Oncol Rep 37: 2033-2040, 2017
APA
Wang, J., Zhou, Y., Fei, X., Chen, X., Yan, J., Liu, B., & Zhu, Z. (2017). ADAM9 functions as a promoter of gastric cancer growth which is negatively and post-transcriptionally regulated by miR-126. Oncology Reports, 37, 2033-2040. https://doi.org/10.3892/or.2017.5460
MLA
Wang, J., Zhou, Y., Fei, X., Chen, X., Yan, J., Liu, B., Zhu, Z."ADAM9 functions as a promoter of gastric cancer growth which is negatively and post-transcriptionally regulated by miR-126". Oncology Reports 37.4 (2017): 2033-2040.
Chicago
Wang, J., Zhou, Y., Fei, X., Chen, X., Yan, J., Liu, B., Zhu, Z."ADAM9 functions as a promoter of gastric cancer growth which is negatively and post-transcriptionally regulated by miR-126". Oncology Reports 37, no. 4 (2017): 2033-2040. https://doi.org/10.3892/or.2017.5460