Open Access

Melanoma differentiation‑associated gene‑7 suppresses human gastric cancer cell invasion and migration

  • Authors:
    • Li Xu
    • Jinyan Chen
    • Wei Lin
    • Jinkun Chen
    • Zhiwei Chen
  • View Affiliations

  • Published online on: September 27, 2017     https://doi.org/10.3892/ol.2017.7086
  • Pages: 7139-7144
  • Copyright: © Xu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Gastric cancer is one of the most common types of cancer in the world. Patients with gastric cancer often respond poorly to conventional chemotherapies, therefore more comprehensive therapy is required. Melanoma differentiation‑associated gene‑7 (MDA‑7), also termed interleukin‑24, is a potent tumor suppressor gene. Numerous studies have demonstrated that MDA‑7 suppresses the growth and induces the apoptosis of cancer cells. In the present study, the MDA‑7 gene was transfected into human gastric cancer AGS cells using adenovirus. Transwell and wound healing assays were performed to evaluate AGS cell invasion and migration, respectively. Western blotting was used to detect the expression of epithelial (E)‑cadherin, cluster of differentiation (CD)44 and matrix metalloproteinase (MMP)‑2 and MMP‑9 proteins. A recombinant virus package was successfully constructed, and it was verified using western blotting that exogenous MDA‑7 was highly expressed in the AGS cells. MDA‑7 overexpression inhibited invasion and migration, decreased CD44, MMP‑2 and MMP‑9 expression, and increased epithelial (E‑)cadherin expression in the AGS cells. Results of the present study revealed that MDA‑7 inhibits gastric cancer invasion and metastasis by inhibiting CD44, MMP‑2 and MMP‑9 expression and by promoting E‑cadherin expression.

Introduction

Gastric cancer is the third most common type of cancer and one of the leading causes of cancer-associated mortality in China (1,2). Patients with gastric cancer often respond poorly to conventional chemotherapies, and, therefore, more comprehensive therapy is required (3). In general, gastric cancer remains difficult to cure, primarily due to gastric cancer cells possessing high invasion and metastasis capability. Melanoma differentiation-associated gene-7 (MDA-7), also termed interleukin (IL)-24, is a member of the IL-10 gene family, and in vitro and in vivo studies have indicated that MDA-7 overexpression suppresses tumor growth and causes tumor cell apoptosis in several types of human cancer, including mesothelioma, osteosarcoma, melanoma, lung cancer, breast cancer, pancreatic cancer, glioblastoma and prostate cancer (46). It is known that MDA-7, a cytokine-tumor suppressor gene and the only tumor suppressor gene in the IL-10 family, not only inhibits tumor growth but also stimulates the immune system and has an antitumor effect; these features render the MDA-7 gene a promising option for the treatment of cancer (7). Currently, basic studies regarding MDA-7/IL-24 in gastric cancer have been limited; to the best of our knowledge, whether MDA-7/IL-24 inhibits gastric cancer cell invasion and metastasis and the potential underlying mechanisms of action have not been reported.

The present study evaluated the effect of MDA-7/IL-24 inhibition on the invasive and metastatic capability of human gastric cancer AGS cells. Western blotting was used to detect the expression of epithelial (E)-cadherin, cluster of differentiation (CD)44, matrix metalloproteinase (MMP)-2 and MMP-9 proteins.

Materials and methods

Cell culture

AGS cells were purchased from the Chinese Academy of Sciences (Shanghai Institute for Biological Science, Shanghai, China) and were cultured in Ham's F12 medium (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at 37°C and 5% CO2.

Plasmid construction

Total RNA from AGS cells was isolated using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. The RNA concentration was quantified by assessment of the optical density; only RNA samples with an A260-A280 ratio between 1.8 and 2.0 were used to obtain complementary DNA (cDNA). A total of 2 µg of each RNA was reverse-transcribed into cDNA by using the first-strand cDNA synthesis kit (Promega Corporation, Madison, WI, USA), and the complete MDA-7 sequence was amplified from the coding region of the gene (GenBank accession no. NM_001185156.1). The PCR cycling conditions were as follows: 94°C for 2 min, 25 cycles of 94°C for 30 sec, 72°C for 30 sec and 56°C for 1 min, followed by a final extension 72°C for 7 min. The forward and reverse primers used were 5′-ACGCGTCGACGCATGAATTTTCAACAGAGGCTG-3′ (SalI restriction site underlined) and 5′-CCGCTCGAGGAGCTTGTAGAATTTCTGCATC-3′ (XhoI restriction site underlined), respectively. The SalI and XhoI sites were cloned into the vector pShuttle-IRES-hrGFP-1 (Stratagene; Agilent Technologies Inc., USA) and verified by DNA sequencing. Subsequently, they underwent homologous recombination with PmeI-linearized plasmid with the BJ5183 strain backbone carrier pAdEasy-1 (Stratagene, Agilent Technologies Inc.), and positive recombinants were identified using the PacI restriction product, termed pAd-MDA-7.

Transfection of MDA-7/IL-24

Following PacI linearization, the recombinant adenovirus vector pAd-MDA-7 and the empty vector were transfected with the aid of Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) into a 293A cell line (low passage; Chinese Academy of Sciences Cell Bank) and were transfected with the intracellular adenovirus packages Ad-MDA-7 or Ad-Control (empty negative control). The virus was repeatedly amplified in the 293A cells, and virus titer was determined by the green fluorescent protein counting method (8). AGS cells were subsequently infected with Ad-MDA-7 or Ad-Control using adenovirus (Stratagene; Agilent Technologies Inc.), and total cellular protein was collected from the lysed cells after 48 h. Western blotting was used to detect MDA-7 expression in 30 µg protein. The primary antibody used was a fusion protein tag antibody: Anti-FLAG M2 antibody (dilution, 1:500; cat. no. F3165; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany), and then incubated with anti-Mouse IgG for 1.5 h at room temperature (dilution, 1:3,000; cat. no. A9044; Sigma-Aldrich; Merck KGaA). The experiment was performed according to the Press Western breeze kit (Invitrogen; Thermo Fisher Scientific, Inc.) protocol.

Matrigel Transwell invasion assay

AGS cells were infected with Ad-MDA-7 or Ad-Control. After 6 h, the cells were harvested by centrifugation at 300 × g at room temperature for 5 min, and resuspended in serum-free F12 medium, and then transferred to the upper chambers of a Matrigel-coated Transwell system (25,000 cells/well); the bottom chambers contained F12 medium with 20% FBS. The cells were incubated for 24 and 48 h at 37°C, and then invaded to the bottom surface of the membrane. The membranes were fixed in 4% paraformaldehyde at room temperature for 30 min, stained with hematoxylin at room temperature for 1 min and counted using a light microscope (magnification, ×100); the relative cell number was calculated. The average number of cells in four random fields/membrane was used to calculate the relative cell number.

Wound healing assay

AGS cells were infected with Ad-MDA-7 or Ad-Control. After 24 h, a pipette tip was used to scratch a straight line down the middle of the cell monolayer. The cells were observed and images were captured under a microscope at 72 h using light microscope (magnification, ×100).

Western blot analysis

AGS cells were infected with Ad-MDA-7 or Ad-Control over 48 h, harvested and lysed by adding ice-cold lysis buffer containing 1 mM phenylmethylsulphone fluoride to extract the total proteins. Protein concentrations were determined using the bicinchoninic acid method. For western blot analysis, 30 µg protein were separated by 12% SDS-PAGE (110 V, 1.5 h), and the membranes were blotted by wet transfer (110 V, 1.5 h, 4°C) onto polyvinylidene fluoride membranes (EMD Millipore, Billerica, MA, USA). The membranes were then blocked in 5% non-fat milk solution in TBS for 1 h at room temperature, and then incubated overnight at 4°C with primary antibodies against CD44 (cat. no. 3578), epithelial (E-)cadherin (cat. no. 5296), MMP-2 (cat. no. 4022) and MMP-9 (cat. no. 3852) (dilution for all, 1:1,000; all Cell Signaling Technology, Inc., Danvers, MA, USA). The membranes were washed with TBS-Tween (TBST), and then incubated for 1.5 h at room temperature with a goat anti-Mouse IgG horseradish peroxidase-conjugated secondary antibody (dilution, 1:1,000; cat. no. 62-6520; Invitrogen; Thermo Fisher Scientific, Inc.). Following washing with TBST, the membranes were exposed to X-ray film (1–15 min) to visualize the immunoreactive bands. Densitometric analysis of specific bands was performed using Image Lab software version 5.0 (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The quantity of the target protein was calibrated with respect to β-actin, and the control value and relative intensities were obtained.

Statistical analysis

The results are presented as the mean ± standard error of the mean. The significance between experimental values was determined using Student's paired t-tests; one-way analysis of variance using the statistical software SPSS16.0 (SPSS, Inc., Chicago, IL, USA) was used to test differences in repeated measures across experiments. P<0.05 was considered to indicate a statistically significant difference. Values were analyzed using the statistical package SPSS (version 16.0; SPSS, Inc., Chicago, IL, USA).

Results

Ad-Control or Ad-MDA-7 infection of AGS cells

AGS cells infected with Ad-MDA7 or Ad-Control for 48 h were observed under a fluorescence microscope. In total >95% of cells emitted strong green fluorescence (Fig. 1). Western blotting using the anti-FLAG M2 antibody revealed that cells infected with Ad-MDA-7 expressed MDA-7 protein, whereas the cells infected with Ad-Control did not (Fig. 2).

Ad-MDA-7 inhibits AGS cell invasion and migration

The in vitro Transwell invasion assay demonstrated that, after 24 and 48 h, there was significantly lower invasion and metastasis by the AGS cells transfected with the MDA-7 gene in comparison with the normal cells and the Ad-Control group (P<0.05; Fig. 3).

In the wound healing assay, gap closure after 48 h by AGS cells transfected with the MDA-7 gene was markedly prolonged as compared with the normal and Ad-Control groups (Fig. 4).

Ad-MDA-7 decreases the expression of CD44 and E-cadherin proteins

To understand the underlying molecular mechanism of MDA-7 inhibition on cell invasion and metastasis, CD44 and E-cadherin expression was examined in the cells (Fig. 5). Compared with the normal and Ad-Control groups, MDA-7 gene transfection decreased CD44 protein expression (P<0.05) and increased E-cadherin protein expression after 48 h (P<0.05).

Ad-MDA-7 decreases the expression of MMP-2 and MMP-9 proteins

Compared with the normal and Ad-Control groups, MDA-7 gene transfection decreased MMP-2 and MMP-9 expression after 48 h (P<0.05; Fig. 6).

Discussion

Invasion and metastasis are the primary biological characteristics of malignant tumors and are the primary causes of surgical, radiotherapy and chemotherapy failure. Invasion and metastasis are the most important causes of mortality in patients with cancer (9). The incidence of gastric cancer is high (10) and the current means of treating gastric cancer in the clinic is surgery post-operatively; however, its effect on progressive patients is not ideal (11). Distal metastasis of gastric cancer cells is the primary cause of mortality in patients with gastric cancer; a biological characteristic of gastric cancer, it is a major obstacle to long-term survival and to improving prognosis (12).

Tumor metastasis involves multi-gene participation and is a complex multi-stage evolution of the biological processes. It was revealed that MDA-7 expression levels were negatively associated with tumor cell proliferation (13). Jiang et al (14) reported that MDA-7 consists of melanoma cells and megakaryocytes, and it was demonstrated that the gene sequence and protein structure of MDA-7 have IL-10 homology domains belonging to the IL-10 family (15). MDA-7 inhibits the growth of a variety of tumor cells and induces features of apoptosis, but does not affect normal cells. The present study suggests that, as a novel tumor suppressor gene, MDA-7 is capable of inhibiting tumor cell growth and angiogenesis, and inducing apoptosis, while stimulating immune cell cytokine expression (1618). A number of studies have demonstrated MDA-7 expression in vitro and in vivo in liver cancer, lung cancer, pancreatic cancer, breast cancer and esophageal cancer, where it significantly inhibited tumor growth and induced apoptosis (1922).

To study the effect of MDA-7 on invasion and metastasis in gastric cancer, the MDA-7 gene was inserted into an adenovirus vector-expressing recombinant adenovirus. The recombinant adenovirus was used to infect gastric cancer AGS cells to obtain increased expression of MDA-7. Matrigel, wound healing and Transwell assays were used to investigate gastric cancer cell adhesion, migration and invasion ability, respectively. The assays demonstrated that MDA-7 inhibits gastric cancer cell invasion and metastasis.

To investigate the underlying molecular mechanism, the expression of E-cadherin, CD44, MMP-2 and MMP-9 proteins was examined in gastric cancer cells transfected with MDA-7.

Tumor invasion and metastasis is a complex multi-step process involving cancer cells shed from the primary site and the degradation of basement membrane binding. Cancer cells invade the blood vessels and lymphatic vessels, eventually colonizing them, and the proliferation forms metastases. Tumor cells shedding from the primary site, which is associated with decreased cell adhesion, is the first step in metastasis. Therefore, mediating cell-cell adhesion of the calcium-dependent cell adhesion molecule E-cadherin serves an important role in tumor invasion and metastasis (23). A study on breast cancer, colorectal cancer, bladder cancer and other malignant tumors (24) revealed that E-cadherin is an inhibitory factor in tumor malignant transformation, invasion and metastasis. E-cadherin downregulation or loss of function is significantly associated with tumor differentiation, invasive growth, metastasis and poor prognosis.

Another important step in tumor invasion and metastasis is the degradation and destruction of the extracellular matrix (ECM) and basement membrane structure barrier, which creates favorable conditions for tumor cell metastasis. MMPs are key enzymes of ECM catabolism, and MMP-2 and MMP-9 serve an important role in tumor invasion and metastasis (25). The actions of MMPs are facilitated by the degradation of ECM and basement membrane type IV collagen fibers, and they promote tumor angiogenesis and cancer cell invasion and metastasis (26,27).

Following infiltration from the blood vessels and lymph vessels by tumor cells and the development of metastases, cell adhesion is subsequently involved in the process and performs an important role. CD44 belongs to the adhesion molecule family and mediates ECM-cell adhesion; its expression is increased in a variety of tumor cells, promoting tumor cell invasion and metastasis, and performing an important role in tumor occurrence, development and metastasis (28,29).

In the present study, CD44, MMP-2 and MMP-9 protein expression was decreased and E-cadherin protein expression was increased in gastric cancer cells transfected with the MDA-7 gene. It was considered that MDA-7 inhibited CD44, MMP-2 and MMP-9 protein expression and promoted E-cadherin protein expression, consequently inhibiting the invasion and metastasis of gastric cancer cells. These features render the MDA-7 gene a promising cancer treatment approach. However, for MDA-7 gene therapy as well as the means of gene transfer to be effective, in-depth study of the antitumor capacity, efficiency and persistence of gene expression is required. The antitumor effect of MDA-7 in Stage I clinical trials has been demonstrated (30,31); in the future, the MDA-7 gene may be a novel tool in cancer gene therapy.

Acknowledgements

The present study was sponsored by the Natural Science Foundation of Fujian Province (grant nos. 2012J01430, 2016J01403 and 2014J01405), Fujian Provincial Health Systems Young Backbone Personnel Training Project Funding Plan (grant no. 2013-ZQN-ZD-30), National Health and Family Planning Commission to Build a Scientific Research Fund - the third round of health education in Fujian joint research projects (grant no. WKJ-FJ-37).

References

1 

Hou R, Cao B and Chen Z, Li Y, Ning T, Li C, Xu C and Chen Z: Association of cytotoxic T lymphocyte-associated antigen-4 gene haplotype with the susceptibility to gastric cancer. Mol Biol Rep. 37:515–520. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Chen WQ, Zheng RS, Zhang SW, Zeng HM and Zou XN: The incidences and mortalities of major cancers in China, 2010. Chin J Cancer. 33:402–405. 2014.PubMed/NCBI

3 

Lissoni P, Brivio F, Ardizzoia A, Tancini G and Barni S: Subcutaneous therapy with low-dose interleukin-2 plus the neurohormone melatonin in metastatic gastric cancer patients with low performance status. Tumori. 79:401–404. 1993.PubMed/NCBI

4 

Bhutia SK, Das SK, Azab B, Menezes ME, Dent P, Wang XY, Sarkar D and Fisher PB: Targeting breast cancer-initiating/stem cells with melanoma differentiation-associated gene-7/interleukin-24. Int J Cancer. 133:2726–2736. 2013.PubMed/NCBI

5 

Menezes ME, Shen XN, Das SK, Emdad L, Guo C, Yuan F, Li YJ, Archer MC, Zacksenhaus E, Windle JJ, et al: MDA-7/IL-24 functions as a tumor suppressor gene in vivo in transgenic mouse models of breast cancer. Oncotarget. 6:36928–36942. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Sauane M, Gupta P, Lebedeva IV, Su ZZ, Sarkar D, Randolph A, Valerie K, Gopalkrishnan RV and Fisher PB: N-glycosylation of MDA-7/IL-24 is dispensable for tumor cell-specific apoptosis and ‘bystander’ antitumor activity. Cancer Res. 66:11869–11877. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Tong AW, Nemunaitis J, Su D, Zhang Y, Cunningham C, Senzer N, Netto G, Rich D, Mhashilkar A, Parker K, et al: Intratumoral injection of INGN 241, a nonreplicating adenovector expressing the melanoma-differentiation associated gene-7 (mda-7/IL24): Biologic outcome in advanced cancer patients. Mol Ther. 11:160–172. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Chen X, Liu D, Wang J, Su Q, Zhou P, Liu J, Luan M and Xu X: Suppression effect of recombinant adenovirus vector containing hIL-24 on Hep-2 laryngeal carcinoma cells. Oncol Lett. 7:771–777. 2014.PubMed/NCBI

9 

Hurst DR and Welch DR: Metastasis suppressor genes at the interface between the environment and tumor cell growth. Int Rev Cell Mol Biol. 286:107–180. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Yao JC, Mansfield PF, Pisters PW, Feig BW, Janjan NA, Crane C and Ajani JA: Combined modality the therapy for gastric cancer. Semin Surg Oncol. 21:223–227. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Qu Y, Zhou C, Zhang J, Cai Q, Li J, Du T, Zhu Z, Cui X and Liu B: The metastasis suppressor SOX11 is an independent prognostic factor for improved survival in gastric cancer. Int J Oncol. 44:1512–1520. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Panneerselvam J, Munshi A and Ramesh R: Molecular targets and signaling pathways regulated by interleukin (IL)-24 in mediating its antitumor activities. J Mol Signal. 8:152013. View Article : Google Scholar : PubMed/NCBI

14 

Jiang H, Lin JJ, Su ZZ, Goldstein NI and Fisher PB: Subtraction hybridization identifies a novel melanoma differentiation associated gene, mda-7, modulated during human melanoma differentiation, growth and progression. Oncogene. 11:2477–2486. 1995.PubMed/NCBI

15 

Chada S, Sutton RB, Ekmekcioglu S, Ellerhorst J, Mumm JB, Leitner WW, Yang HY, Sahin AA, Hunt KK, Fuson KL, et al: Mda-7/IL-24 is a unique cytokine-tumor suppressor in the IL-10 family. Int Immunopharmacol. 4:649–667. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Lebedeva IV, Emdad L, Su ZZ, Gupta P, Sauane M, Sarkar D, Staudt MR, Liu SJ, Taher MM, Xiao R, et al: mda-7/IL-24, novel anticancer cytokine: Focus on bystander antitumor, radiosensitization and antiangiogenic properties and overview of the phase I clinical experience (Review). Int J Oncol. 31:985–1007. 2007.PubMed/NCBI

17 

Menezes ME, Bhatia S, Bhoopathi P, Das SK, Emdad L, Dasgupta S, Dent P, Wang XY, Sarkar D and Fisher PB: MDA-7/IL-24: Multifunctional cancer killing cytokine. Adv Exp Med Biol. 818:127–153. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Xie Y, Sheng W, Xiang J, Ye Z, Zhu Y, Chen X and Yang J: Recombinant human IL-24 suppresses lung carcinoma cell growth via induction of cell apoptosis and inhibition of tumor angiogenesis. Cancer Biother Radiopharm. 23:310–320. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Ma G, Kawamura K, Shan Y, Okamoto S, Li Q, Namba M, Shingyoji M, Tada Y, Tatsumi K, Hiroshima K, et al: Combination of adenoviruses expressing melanoma differentiation-associated gene-7 and chemotherapeutic agents produces enhanced cytotoxicity on esophageal carcinoma. Cancer Gene Ther. 21:31–37. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Pan X, Sheng W, Zhu Q, Xie Y, Ye Z, Xiang J, Li D and Yang J: Inhibition of pancreatic carcinoma growth by adenovirus-mediated human interleukin-24 expression in animal model. Cancer Biother Radiopharm. 23:425–434. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Chen X, Liu D, Wang J, Su Q, Zhou P, Liu J, Luan M and Xu X: Suppression effect of recombinant adenovirus vector containing hIL-24 on Hep-2 laryngeal carcinoma cells. Oncol Lett. 7:771–777. 2014.PubMed/NCBI

22 

Patani N, Douglas-Jones A, Mansel R, Jiang W and Mokbel K: Tumour suppressor function of MDA-7/IL-24 in human breast cancer. Cancer Cell Int. 10:292010.PubMed/NCBI

23 

Shamir ER and Ewald AJ: Adhesion in mammary development: Novel roles for E-cadherin in individual and collective cell migration. Curr Top Dev Biol. 112:353–382. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Sarrió D, Palacios J, Hergueta-Redondo M, Gómez-López G, Cano A and Moreno-Bueno G: Functional characterization of E-and P-cadherin in invasive breast cancer cells. BMC Cancer. 9:742009. View Article : Google Scholar : PubMed/NCBI

25 

Sounni NE and Noel A: Membrane type-matrix metalloproteinases and tumor progression. Biochimie. 87:329–342. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Tao L, Li Z, Lin L, Lei Y, Hongyuan Y, Hongwei J, Yang L and Chuize K: MMP1, 2, 3, 7, and 9 gene polymorphisms and urinary cancer risk: A meta-analysis. Genet Test Mol Biomarkers. 19:548–555. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Belotti D, Paganoni P, Manenti L, Garofalo A, Marchini S, Taraboletti G and Giavazzi R: Matrix metalloproteinases (MMP9 and MMP2) induce the release of vascular endothelial growth factor (VEGF) by ovarian carcinoma cells: Implications for ascites formation. Cancer Res. 63:5224–5229. 2003.PubMed/NCBI

28 

Bánky B, Rásó-Barnett L, Barbai T, Tímár J, Becságh P and Rásó E: Characteristics of CD44 alternative splice pattern in the course of human colorectal adenocarcinoma progression. Mol Cancer. 11:832012. View Article : Google Scholar : PubMed/NCBI

29 

Paulis YW, Huijbers EJ, van der Schaft DW, Soetekouw PM, Pauwels P, Tjan-Heijnen VC and Griffioen AW: CD44 enhances tumor aggressiveness by promoting tumor cell plasticity. Oncotarget. 6:19634–19646. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Dent P, Yacoub A, Grant S, Curiel DT and Fisher PB: MDA-7/IL-24 regulates proliferation, invasion and tumor cell radiosensitivity: A new cancer therapy? J Cell Biochem. 95:712–719. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Cunningham CC, Chada S, Merritt JA, Tong A, Senzer N, Zhang Y, Mhashilkar A, Parker K, Vukelja S, Richards D, et al: Clinical and local biological effects of an intratumoral injection of mda-7 (IL24; INGN 241) in patients with advanced carcinoma: A phase I study. Mol Ther. 11:149–159. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2017
Volume 14 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu L, Chen J, Lin W, Chen J and Chen Z: Melanoma differentiation‑associated gene‑7 suppresses human gastric cancer cell invasion and migration. Oncol Lett 14: 7139-7144, 2017
APA
Xu, L., Chen, J., Lin, W., Chen, J., & Chen, Z. (2017). Melanoma differentiation‑associated gene‑7 suppresses human gastric cancer cell invasion and migration. Oncology Letters, 14, 7139-7144. https://doi.org/10.3892/ol.2017.7086
MLA
Xu, L., Chen, J., Lin, W., Chen, J., Chen, Z."Melanoma differentiation‑associated gene‑7 suppresses human gastric cancer cell invasion and migration". Oncology Letters 14.6 (2017): 7139-7144.
Chicago
Xu, L., Chen, J., Lin, W., Chen, J., Chen, Z."Melanoma differentiation‑associated gene‑7 suppresses human gastric cancer cell invasion and migration". Oncology Letters 14, no. 6 (2017): 7139-7144. https://doi.org/10.3892/ol.2017.7086