Open Access

High expression of GALNT7 promotes invasion and proliferation of glioma cells

  • Authors:
    • Shi Hua
    • Hongyan Li
    • Yuguang Liu
    • Jian Zhang
    • Yanhao Cheng
    • Chao Dai
  • View Affiliations

  • Published online on: September 25, 2018     https://doi.org/10.3892/ol.2018.9498
  • Pages: 6307-6314
  • Copyright: © Hua 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

Polypeptide‑N‑acetyl‑galactosaminlytransferase 7 (GALNT7), a member of the GalNAc‑transferase family, has not been previously evaluated as a prognostic factor of glioblastoma (GBM) or low‑grade glioma (LGG). Based on The Cancer Genome Atlas database and bioinformatics analyses, the expression of GALNT7 was demosntrated to be higher in GBM and LGG tissues than in normal brain tissue. The expression levels of GANLT7 were associated with age, tumor grade, survival rate, disease‑free survival time and overall survival time. Gene correlation and gene‑set enrichment analyses suggested that GALNT7 may affect the proliferative and invasive abilities of glioma cells through multiple signaling pathways, including regulation of the actin cytoskeleton, natural killer cell‑mediated cytotoxicity, the janus kinase‑signal transducer and activator of transcription (STAT) signaling pathway, cell adhesion molecules and extracellular matrix receptor interaction pathways. Furthermore, 5 target genes of GALNT7 involved in these signaling pathways were identified, including Crk, Rac family small GTPase 1 , STAT3, poliovirus receptor and Tenascin C. In summary, high expression of GALNT7 was associated with poor prognosis of glioma, and may be used as an effective biomarker of glioma.

Introduction

Glioma is the most common and aggressive type of tumor of the human central nervous system (CNS). The annual incidence of glioma has been reported as 30~80/1 million worldwide (1). Regarding pathological grade, glioma includes glioblastoma (GBM) and low-grade glioma (LGG). Currently, standard treatments for glioma include chemoradiotherapy and targeted therapy, which are administered following maximum surgical resection (25). The prognosis of glioma was reported to have a 5-year survival rate of ~10–20% (6). Therapeutic techniques for increasing the survival rate and improving quality of life of patients with glioma in the neurosurgical field (7,8).

Polypeptide-N-acetyl-galactosaminlytransferase 7 (GALNT7) is a member of the N-acetyl-D-galactosamine-transferase family. The enzyme encoded by this gene controls the initiation step of mucin-type O-linked protein glycosylation and transfers N-acetyl galactosamine to serine and threonine amino acid residues (9). Previous studies have indicated that GALNT7 can influence the prognoses of multiple types of malignant tumor, including cervical (10), esophageal (11) and liver cancer (12), as well as renal cell carcinoma (13), by affecting cell proliferation, metastasis, apoptosis, migration, differentiation and invasion (1417).

The Cancer Genome Atlas (TCGA) is a publicly funded project that aims to catalogue and identify major cancer-causing genomic alterations, to create a comprehensive ‘atlas’ of genomic profiles of various types of cancer. To date, the program has investigated >30 types of human tumor, including GBM and LGG, through large-scale genome sequencing and integrated multi-dimensional analyses (18). To the best of our knowledge, the present study is the first to investigate the role of GALNT7 in glioma, using GBM and LGG RNA-seq data downloaded from the TCGA database.

Materials and methods

Data collection

The RNA-seq expression and clinical data of 174 patients with GBM and 529 patients with LGG were obtained from TCGA (using the search terms: GBM and LGG, level 3 and RNA-seq; http://cancergenome.nih.gov/). Complete gene expression data, clinical data and definite pathological diagnosis was available for all included patients. RNA-seq expression data for 114 normal brain tissues were download from the GTEx project (https://www.gtexportal.org/home/; Brain-Cortex-GTEx, RNA-seq) and used as the control group (19).

Statistical analysis

SPSS 21.0 (IBM Corp., Armonk, NY, USA) was used for data processing. Quantitative data are expressed as the mean ± standard deviation (x¯±s). Two samples were compared by t-test and multiple comparisons were performed by one-way analysis of variance followed by Least-Significant-Difference test. Qualitative data were expressed as a percentage (%) and analyzed by χ2. Survival analysis was performed using Cox regression and Kaplan-Meier analysis (P<0.05). Gene-set enrichment analysis was performed using GSEA (version, 2.2.1; http://software.broadinstitute.org/gsea/downloads.jsp) from the Molecular Signatures Database (v6.3 MsigDB; Broad Institute, Inc., Massachusetts Institute of Technology, and Regents of the University of California). According to default-weighted enrichment, the cut-offs for the number of random combinations was 1,000, for nominal P-values (NOM P-val) <0.05, and false discovery rates (FDR), <0.25. Pearson correlation coefficient was used to assess the association between the expression of 2 genes. P<0.05 was considered to indicate a statistically significant difference.

Results

GALNT7 expression is positively associated with glioma malignancy

GALNT7 expression levels were significantly higher in glioma, particularly in GBM, compared with the negative control samples (P<0.05). This indicates a positive association between GALNT7 expression and glioma malignancy (Table I; Fig. 1A).

Table I.

Differential analysis of GLANT7 RNA-seq expression.

Table I.

Differential analysis of GLANT7 RNA-seq expression.

GroupNo.mean ± standard deviationFP-value
GBM1743.46±1.3815.37<0.05
LGG5292.92±1.58
Normal1142.46±1.58

[i] GBM, glioblastoma; LGG, low-grade glioma. F, one-way ANOVA value.

Association between GALNT7 expression and clinical characteristics

GALNT7 was expressed at different levels in normal brain tissues and glioma tissues. In order to investigate the effect of its expression in glioma, GALNT7 expression in normal brain tissue from GTEx was employed as the cut-off value, and TCGA data regarding expression of GALNT7 in glioma tissue were divided into high (HIGH) and low (LOW) groups. The association between GALNT7 expression levels and clinical characteristics of patients with glioma was analyzed using t-test. The results demonstrated that expression level was not associated with sex, but it was associated with age, tumor grade and survival rate (Table II).

Table II.

Clinical characteristics of patients with glioma.

Table II.

Clinical characteristics of patients with glioma.

CharacteristicsNo.Low GALNT7 expression (%)High GALNT7 expression (%)χ2P-value
Age (years) 10.46<0.05
  <40274132 (48.2)142 (51.8)
  ≥42414149 (36.0)265 (64.0)
Sex 0.050.82
  Male393162 (41.2)231 (58.8)
  Female295119 (40.3)176 (59.7)
Grade 38.37<0.05
  G2257138 (53.7)119 (46.3)
  G3263104 (39.5)159 (60.5)
  G416839 (23.2)129 (76.8)
Survival status 38.456<0.05
  Not alive416209 (50.2)207 (49.8)
  Alive27272 (26.5)200 (73.5)

[i] GLANT7, polypeptide-N-acetyl-galactosaminlytransferase 7. Part of the follow up data from TCGA was missing, including 6 patients with GBM and 9 patients with LGG. Therefore these patients were excluded from form Table II.

Negative association between GALNT7 expression and survival time

Using the median GALNT7 expression value as the standard, GALNT7 expression data were divided into HIGH GALNT7 transcripts per million (TPM) and LOW GALNT7 TPM groups. Cox survival analysis demonstrated a regression coefficient (B) of 0.102 (P<0.05; Table III). Together with Kaplan-Meier survival analysis, this suggests that patients with glioma exhibiting high expression of GALNT7 have relatively shorter disease-free (Fig. 1B) and overall (Fig. 1C) survival times than those exhibiting low expression.

Table III.

Cox regression analysis of GALNT7 expression.

Table III.

Cox regression analysis of GALNT7 expression.

GALNT7BSEWalddfP-valueHR95% Confidence intervals
0.100.0230.941<0.051.11(1.07,1.15)

[i] GLANT7, polypeptide-N-acetyl-galactosaminlytransferase 7; B, regression coefficient; SE, standard error; Wald, χ2 value; df, degree of freedom; HR, hazard ratio.

GSEA and prediction of co-expressed genes

Based on the KEGG database (20), the results of GSEA (NOM P-val <0.05; FDR<0.25) indicated 6 pathways associated with glioma, including ‘regulation of the actin cytoskeleton’ (Fig. 2A), ‘natural killer cell-mediated cytotoxicity’ (Fig. 2B), ‘JAK-STAT signaling’ (Fig. 2C), ‘cell adhesion molecules’ (‘CAMs’) (Fig. 2D) and ‘ECM-receptor interaction’ (Fig. 2E). In order to predict genes that are co-expressed, Pearson's correlation analysis was used to determine the correlation between GALNT7 expression and all genes associated with these signaling pathways. As a result, Crk (Fig. 2F), RAC1 (Fig. 2G), STAT3 (Fig. 2H), PVR (Fig. 2I) and Tnc (Fig. 2J) were identified as potential target genes. Crk participates in regulating the ‘actin cytoskeleton’ pathway, RAC1 is involved in the ‘natural killer cell mediated cytotoxicity’ pathway and STAT3 is associated with the ‘JAK-STAT’ signaling pathway. Also, PVR participates in the ‘CAMs’ pathway and Tnc is involved in the ‘ECM-receptor interaction’ pathway.

Differential and survival analysis of co-expressed genes in glioma

Differential analyses demonstrated that Crk (Fig. 3A) expression is positively associated with the malignancy of glioma; positive association was also observed with RAC1 (Fig. 3B), STAT3 (Fig. 3C), and Tnc (Fig. 3E). PVR (Fig. 3D) highly expressed in GBM, but expressed lower than normal brain tissue in LGG (P<0.05). Survival analyses demonstrate that disease-free time was negatively correlated with the expression of RAC1 (Fig. 4B), STAT3 (Fig. 4C), PVR (Fig. 4D) and Tnc (Fig. 4E), but not Crk (Fig. 4A). Meanwhile, the expression levels of Crk (Fig. 4F), RAC1 (Fig. 4G), STAT3 (Fig. 4H), PVR (Fig. 4I) and Tnc (Fig. 4J) were negatively correlated with overall survival time (P<0.05).

Discussion

Glioma is divided into 4 grades according to the World Health Organization standard (1): Grade I, pilocytic astrocytoma, which manifests as a benign tumor and patients may have a full recovery following total tumor resection; Grade II, includes oligoastrocytoma and diffuse astrocytoma, it has a poorer prognosis compared with Grade I, but is still considered to be LGG (2); Grade III, includes anaplastic astrocytoma, and Grade IV, GBM. Grade III and IV tumors are associated with high degrees of malignancy, strong invasive abilities, poor prognosis and multiple differentiation potentials (3,21). The present study aimed to reveal the association between GALNT7 expression and glioma by identifying co-expressed genes and relevant signaling pathways.

Firstly, a positive correlation between GALNT7 expression and the malignancy of glioma was assessed by analyzing TCGA glioma datasets. Cox and Kaplan-Meier survival analyses demonstrated that patients with glioma exhibiting high expression levels of GALNT7 had relatively short disease-free and overall survival times. Therefore, high expression levels of GALNT7 may affect the prognosis of glioma.

Correlation analyses suggested that Crk, RAC1, STAT3, PVR and Tnc are genes that are co-expressed with GALNT7. Survival and differential analyses demonstrated that the expression of Crk, RAC1, STAT3, PVR and Tnc are positively associated with the malignancy of glioma, and are negatively associated with survival time. These results were consistent with those of previous studies, which indicated that high expression levels of Crk, RAC1, STAT3, PVR and Tnc were associated with poor prognosis by promoting the invasion, proliferation and other biological functions of glioma (2227). Therefore, we speculate that GALNT7 may affect the progression of glioma by upregulating the expression of target genes through relevant signaling pathways. Based on this hypothesis, GSEA (28,29) was used to identify such relevant signaling pathways. As a result, 5 signaling pathways associated with GALNT7 and glioma were identified, including ‘regulation of the actin cytoskeleton’, ‘natural killer cell-mediated cytotoxicity’, ‘JAK-STAT signaling’, ‘CAMs’ and ‘ECM-receptor interaction’ pathways. Specific target genes of GALNT7 participate in each signaling pathway, and were subsequently evaluated.

There are a variety of intracellular actin binding proteins that regulate multiple cellular functions, including changes in the structure of the actin cytoskeleton, which occur through the binding or dissociating of the proteins with actin (30). Regulation of the actin cytoskeleton pathway governs the cell motility of glioma by regulating the locomotion of the cytoskeleton, and slows the recovery of patients with glioma (31,32). Crk participates in regulation of the ‘actin cytoskeleton pathway’, and its expression is associated with overall survival time of patients with glioma. Tsuda et al (22) demonstrate that overexpression of Crk can increase the invasive potential of cancer cells by notably inducing tyrosine phosphorylation of scaffolding molecules, including p130 (Cas) and paxillin through Src family tyrosine kinases, and stimulating the activation loop of intracellular signaling. Therefore, as a gene that is co-expressed with Crk, GALNT7 promotes the invasion of glioma by upregulating Crk expression through regulation of the actin cytoskeleton pathway.

NK cells form the first line of immune defense against tumors (33,34). Such cells destroy tumors by controlling cell proliferation, cytotoxicity and cytokine production in the early stages of tumor formation (35,36). RAC1 has been reported to be involved in the NK cell-mediated cytotoxicity pathway (37). Former research suggests that high expression levels of RAC1 inhibit the cytotoxic effects of NK cells by two mechanisms: Decreased interaction between NK and target cells; NK cells that do interact have a reduced ability to polarize their effector molecules towards target cells (38). Based on these results, coexpression with RAC1 and high expression of GALNT7 inhibits the cytotoxic effects of NK cells by regulating the natural killer cell-mediated cytotoxicity pathway, which leads to loss of control of glioma cell proliferation and poor prognosis.

The JAK-STAT signaling pathway is mainly formed of the tyrosine kinase associated receptor, JAK and STAT (39). Previous studies reported that the activation of the JAK-STAT signaling pathway is associated with poor prognosis of glioma (40,41). Additionally, a previous study verified that high expression levels of STAT3 enhance the proliferation of glioma cells (24). In the present study, GALNT7 was coexpressed with STAT3, and high expression levels of STAT3 were associated with poor prognosis of glioma. This suggests that GALNT7 may trigger and increase in glioma cell proliferation by activating the JAK-STAT signaling pathway through upregulation of STAT3 expression.

CAMs are membrane and transmembrane glycoproteins that regulate intercellular, cell and extracellular matrix interactions, which are closely associated with cell adhesion, migration, differentiation and signal transduction (42,43). CAMs are widely expressed in glioma and are regulated by the CAMs signaling pathway (44). PVR is involved in the CAMs signaling pathway, and can enhance the metastasis of glioblastoma through its over-expression (45). As GALNT7 is co-expressed with PVR, GALNT7 may regulate the CAMs signaling pathway by up-regulating PVR, thereby influencing tumor cell migration and affecting prognosis.

ECM is mainly formed of insoluble components that contribute to the behavior and structure of stromal cells and epithelial vessels, respectively. The ECM constitutes collagen, elastin, proteoglycan and glycoprotein. ECM can influence cell differentiation, proliferation, adhesion, morphogenesis and phenotypic expression (46). Brösicke et al (47,48) proposed Tnc as a key gene in the ECM-receptor interaction pathway and that high coexpression with GALNT7 occurs within glioma tissue, which is consistent with our research. Accumulating evidence also suggests that Tnc serves a crucial role in cell migration and invasion, the most malignant characteristics of glioma. Therefore, GALNT7 can affect the ECM-receptor interaction pathway by upregulating the expression of Tnc; this mechanism may affect the invasion of glioma cells and lead to a poor prognosis of glioma.

In conclusion, high expression of GALNT7 was demonstrated to be associated with poor prognosis of glioma, likely via promoting invasion and proliferation through multiple signaling pathways. Therefore, GALNT7 may be employed as a novel molecular target for the early detection, diagnosis and treatment of glioma.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets generated or analyzed during the current study are available in the TCGA/KEGG/GTEx repository, [https://cancergenome.nih.gov/] [https://www.kegg.jp/] [https://www.gtexportal.org/home/].

Authors' contributions

SH contributed to writing the manuscript, and was responsible for project design and data interpretation; YC and CD were responsible for data collection and preliminary analysis; HL was responsible for all data analyses; YL and JZ conceived and designed the present study given final approval of the version to be published.

Ethical approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

GALNT7

polypeptide-N-acetyl-galactosaminlytransferase 7

TCGA

The Cancer Genome Atlas

KEGG

Kyoto Encyclopedia of Genes and Genomes database

GTEx

Genotype-Tissue Expression project

GBM

glioblastoma

LGG

low grade glioma

NOM P-val

nominal P-values

FDR

false discovery rates

MsigDB

Molecular Signatures Database

NK

natural killer

CAMS

cell adhesion molecules

CNS

central nervous system

MIF

macrophage migration inhibitory factor

JAK

janus kinase

STAT

signal transducer and activator of transcription

GSEA

gene-set enrichment analysis

References

1 

Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P and Ellison DW: The 2016 world health organization classification of tumors of the central nervous system: A summary. Acta Neuropathol. 131:803–820. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Forst DA, Nahed BV, Loeffler JS and Batchelor TT: Low-grade gliomas. Oncologist. 19:403–413. 2014. View Article : Google Scholar : PubMed/NCBI

3 

de Groot JF: High-grade gliomas. Continuum (Minneap Minn. ). 21:332–344. 2015.PubMed/NCBI

4 

Kim IA, Yang YJ, Yoon SC, Choi IB, Kay CS, Kwon HC, Kim CM, Joe YA, Kang JK and Hong YK: Potential of adenoviral p53 gene therapy and irradiation for the treatment of malignant gliomas. Int J Oncol. 19:1041–1047. 2001.PubMed/NCBI

5 

Maris D, Nica D, Mohan D, Moisa H and Ciurea AV: Multidisciplinary management of adult low grade gliomas. Chirurgia (Bucur). 109:590–599. 2014.PubMed/NCBI

6 

Pekmezci M and Perry A: Genetic markers in adult high-grade gliomas. Semin Radiat Oncol. 24:235–239. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Duffy A, Le J, Sausville E and Emadi A: Autophagy modulation: A target for cancer treatment development. Cancer Chemother Pharmacol. 75:439–447. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Lin Q, Mao KL, Tian FR, Yang JJ, Chen PP, Xu J, Fan ZL, Zhao YP, Li WF, Zheng L, et al: Brain tumor-targeted delivery and therapy by focused ultrasound introduced doxorubicin-loaded cationic liposomes. Cancer Chemother Pharmacol. 77:269–280. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Bennett EP, Hassan H, Hollingsworth MA and Clausen H: A novel human UDP-N-acetyl-D-galactosamine: Polypeptide N-acetylgalactosaminyltransferase, GalNAc-T7, with specificity for partial GalNAc-glycosylated acceptor substrates. FEBS Lett. 460:226–230. 1999. View Article : Google Scholar : PubMed/NCBI

10 

Wu H, Chen J, Li D, Liu X, Li L and Wang K: MicroRNA-30e functions as a tumor suppressor in cervical carcinoma cells through targeting GALNT7. Transl Oncol. 10:876–885. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Lu Q, Xu L, Li C, Yuan Y, Huang S and Chen H: miR-214 inhibits invasion and migration via downregulating GALNT7 in esophageal squamous cell cancer. Tumour Biol. 37:14605–14614. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Shan SW, Fang L, Shatseva T, Rutnam ZJ, Yang X, Du W, Lu WY, Xuan JW, Deng Z and Yang BB: Mature miR-17-5p and passenger miR-17-3p induce hepatocellular carcinoma by targeting PTEN, GalNT7 and vimentin in different signal pathways. J Cell Sci. 126:1517–1530. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Li Y, Li Y, Chen D, Jin L, Su Z, Liu J, Duan H, Li X, Qi Z, Shi M, et al: miR30a5p in the tumorigenesis of renal cell carcinoma: A tumor suppressive microRNA. Mol Med Rep. 13:4085–4094. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Duan HF, Li XQ, Hu HY, Li YC, Cai Z, Mei XS, Yu P, Nie LP, Zhang W, Yu ZD and Nie GH: Functional elucidation of miR-494 in the tumorigenesis of nasopharyngeal carcinoma. Tumour Biol. 36:6679–6689. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Li W, Ma H and Sun J: MicroRNA34a/c function as tumor suppressors in Hep2 laryngeal carcinoma cells and may reduce GALNT7 expression. Mol Med Rep. 9:1293–1298. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Gaziel-Sovran A, Segura MF, Di Micco R, Collins MK, Hanniford D, de Miera Vega-Saenz E, Rakus JF, Dankert JF, Shang S, Kerbel RS, et al: miR-30b/30d regulation of GalNAc transferases enhances invasion and immunosuppression during metastasis. Cancer Cell. 20:104–118. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Mayer A, Schneider F, Vaupel P, Sommer C and Schmidberger H: Differential expression of HIF-1 in glioblastoma multiforme and anaplastic astrocytoma. Int J Oncol. 41:1260–1270. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Gonda DD, Cheung VJ, Muller KA, Goyal A, Carter BS and Chen CC: The cancer genome atlas expression profiles of low-grade gliomas. Neurosurg Focus. 36:E232014. View Article : Google Scholar : PubMed/NCBI

19 

The Genotype-Tissue Expression (GTEx) project. Nat Genet. 45:580–585. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Du J, Yuan Z, Ma Z, Song J, Xie X and Chen Y: KEGG-PATH: Kyoto encyclopedia of genes and genomes-based pathway analysis using a path analysis model. Mol Biosyst. 10:2441–2447. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Pouleau HB, Sadeghi N, Baleriaux D, Melot C, De Witte O and Lefranc F: High levels of cellular proliferation predict pseudoprogression in glioblastoma patients. Int J Oncol. 40:923–928. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Tsuda M and Tanaka S: Roles for crk in cancer metastasis and invasion. Genes Cancer. 3:334–340. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Yukinaga H, Shionyu C, Hirata E, Ui-Tei K, Nagashima T, Kondo S, Okada-Hatakeyama M, Naoki H and Matsuda M: Fluctuation of Rac1 activity is associated with the phenotypic and transcriptional heterogeneity of glioma cells. J Cell Sci. 127:1805–1815. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Priester M, Copanaki E, Vafaizadeh V, Hensel S, Bernreuther C, Glatzel M, Seifert V, Groner B, Kögel D and Weissenberger J: STAT3 silencing inhibits glioma single cell infiltration and tumor growth. Neuro Oncol. 15:840–852. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Chandramohan V, Bryant JD, Piao H, Keir ST, Lipp ES, Lefaivre M, Perkinson K, Bigner DD, Gromeier M and McLendon RE: Validation of an Immunohistochemistry Assay for Detection of CD155, the poliovirus receptor, in malignant gliomas. Arch Pathol Lab Med. 141:1697–1704. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Xia S, Lal B, Tung B, Wang S, Goodwin CR and Laterra J: Tumor microenvironment tenascin-C promotes glioblastoma invasion and negatively regulates tumor proliferation. Neuro Oncol. 18:507–517. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Brosicke N, van Landeghem FK, Scheffler B and Faissner A: Tenascin-C is expressed by human glioma in vivo and shows a strong association with tumor blood vessels. Cell Tissue Res. 354:409–430. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Machado CM, Freitas AT and Couto FM: Enrichment analysis applied to disease prognosis. J Biomed Semantics. 4:212013. View Article : Google Scholar : PubMed/NCBI

29 

Mueller C, deCarvalho AC, Mikkelsen T, Lehman NL, Calvert V, Espina V, Liotta LA and Petricoin EF III: Glioblastoma cell enrichment is critical for analysis of phosphorylated drug targets and proteomic-genomic correlations. Cancer Res. 74:818–828. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Hou M, Liu X, Cao J and Chen B: SEPT7 overexpression inhibits glioma cell migration by targeting the actin cytoskeleton pathway. Oncol Rep. 35:2003–2010. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Klopocka W, Korczynski J and Pomorski P: Cytoskeleton and nucleotide signaling in glioma C6 cells. Adv Exp Med Biol. 986:103–119. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Uzdensky A, Kristiansen B, Moan J and Juzeniene A: Dynamics of signaling, cytoskeleton and cell cycle regulation proteins in glioblastoma cells after sub-lethal photodynamic treatment: Antibody microarray study. Biochim Biophys Acta. 1820:795–803. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Huang BY, Zhan YP, Zong WJ, Yu CJ, Li JF, Qu YM and Han S: The PD-1/B7-H1 pathway modulates the natural killer cells versus mouse glioma stem cells. PLoS One. 10:e01347152015. View Article : Google Scholar : PubMed/NCBI

34 

Miller JS: Biology of natural killer cells in cancer and infection. Cancer Invest. 20:405–419. 2002. View Article : Google Scholar : PubMed/NCBI

35 

He W, Kuang Y, Xing X, Simpson RJ, Huang H, Yang T, Chen J, Yang L, Liu E, He W and Gu J: Proteomic comparison of 3D and 2D glioma models reveals increased HLA-E expression in 3D models is associated with resistance to NK cell-mediated cytotoxicity. J Proteome Res. 13:2272–2281. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Orozco-Morales M, Sanchez-Garcia FJ, Golan-Cancela I, Hernández-Pedro N, Costoya JA, de la Cruz VP, Moreno-Jiménez S, Sotelo J and Pineda B: RB mutation and RAS overexpression induce resistance to NK cell-mediated cytotoxicity in glioma cells. Cancer Cell Int. 15:572015. View Article : Google Scholar : PubMed/NCBI

37 

Mainiero F, Soriani A, Strippoli R, Jacobelli J, Gismondi A, Piccoli M, Frati L and Santoni A: RAC1/P38 MAPK signaling pathway controls beta1 integrin-induced interleukin-8 production in human natural killer cells. Immunity. 12:7–16. 2000. View Article : Google Scholar : PubMed/NCBI

38 

Billadeau DD, Brumbaugh KM, Dick CJ, Schoon RA, Bustelo XR and Leibson PJ: The Vav-Rac1 pathway in cytotoxic lymphocytes regulates the generation of cell-mediated killing. J Exp Med. 188:549–559. 1998. View Article : Google Scholar : PubMed/NCBI

39 

Nicolas CS, Amici M, Bortolotto ZA, Doherty A, Csaba Z, Fafouri A, Dournaud P, Gressens P, Collingridge GL and Peineau S: The role of JAK-STAT signaling within the CNS. JAKSTAT. 2:e229252013.PubMed/NCBI

40 

Tu Y, Zhong Y, Fu J, Cao Y, Fu G, Tian X and Wang B: Activation of JAK/STAT signal pathway predicts poor prognosis of patients with gliomas. Med Oncol. 28:15–23. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Zheng Q, Han L, Dong Y, Tian J, Huang W, Liu Z, Jia X, Jiang T, Zhang J, Li X, et al: JAK2/STAT3 targeted therapy suppresses tumor invasion via disruption of the EGFRvIII/JAK2/STAT3 axis and associated focal adhesion in EGFRvIII-expressing glioblastoma. Neuro Oncol. 16:1229–1243. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Zhao WJ and Schachner M: Neuregulin 1 enhances cell adhesion molecule l1 expression in human glioma cells and promotes their migration as a function of malignancy. J Neuropathol Exp Neurol. 72:244–255. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Li J, Liu X, Duan Y, Liu Y, Wang H, Lian S, Zhuang G and Fan Y: Combined blockade of T cell immunoglobulin and mucin domain 3 and carcinoembryonic antigen-related cell adhesion molecule 1 results in durable therapeutic efficacy in mice with intracranial gliomas. Med Sci Monit. 23:3593–3602. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Chen X, Ma WY, Xu SC, Liang Y, Fu YB, Pang B, Xin T, Fan HT, Zhang R, Luo JG, et al: The overexpression of epithelial cell adhesion molecule (EpCAM) in glioma. J Neurooncol. 119:39–47. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Sloan KE, Stewart JK, Treloar AF, Matthews RT and Jay DG: CD155/PVR enhances glioma cell dispersal by regulating adhesion signaling and focal adhesion dynamics. Cancer Res. 65:10930–10937. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Brösicke N and Faissner A: Role of tenascins in the ECM of gliomas. Cell Adh Migr. 9:131–140. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Brosicke N, Sallouh M, Prior LM, Job A, Weberskirch R and Faissner A: Extracellular matrix glycoprotein-derived synthetic peptides differentially modulate glioma and sarcoma cell migration. Cell Mol Neurobiol. 35:741–753. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Shimizu T, Kurozumi K, Ishida J, Ichikawa T and Date I: Adhesion molecules and the extracellular matrix as drug targets for glioma. Brain Tumor Pathol. 33:97–106. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2018
Volume 16 Issue 5

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
Hua S, Li H, Liu Y, Zhang J, Cheng Y and Dai C: High expression of GALNT7 promotes invasion and proliferation of glioma cells. Oncol Lett 16: 6307-6314, 2018
APA
Hua, S., Li, H., Liu, Y., Zhang, J., Cheng, Y., & Dai, C. (2018). High expression of GALNT7 promotes invasion and proliferation of glioma cells. Oncology Letters, 16, 6307-6314. https://doi.org/10.3892/ol.2018.9498
MLA
Hua, S., Li, H., Liu, Y., Zhang, J., Cheng, Y., Dai, C."High expression of GALNT7 promotes invasion and proliferation of glioma cells". Oncology Letters 16.5 (2018): 6307-6314.
Chicago
Hua, S., Li, H., Liu, Y., Zhang, J., Cheng, Y., Dai, C."High expression of GALNT7 promotes invasion and proliferation of glioma cells". Oncology Letters 16, no. 5 (2018): 6307-6314. https://doi.org/10.3892/ol.2018.9498