A role for LRIG1 in the regulation of malignant glioma aggressiveness

  • Authors:
    • Feng Mao
    • Baofeng Wang
    • Qungen Xiao
    • Guifa Xi
    • Wei Sun
    • Huaqiu Zhang
    • Fei Ye
    • Feng Wan
    • Dongsheng Guo
    • Ting Lei
    • Xiaoping Chen
  • View Affiliations

  • Published online on: January 16, 2013     https://doi.org/10.3892/ijo.2013.1776
  • Pages: 1081-1087
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The molecular mechanisms that drive the develop­ment and aggressive progression of malignant astrocytic tumors remain obscure. Recently, in the search for endo­genous negative regulators of EGF receptor, LRIG1 was cloned and characterized as a putative tumor suppressor gene often downregulated in various human tumors, including astrocytic tumors. Although several studies have implicated the function of LRIG1 in the inhibition of tumorigenesis, its precise role and potential underlying mechanisms remain obscure. Therefore, we generated a full-length expression vector to overexpress LRIG1 in the U251 malignant glioma cell line. Introduction of exogenous LRIG1 into glioma cells inhibited cell proliferation manifested by MTT and soft agar clone assay in vitro and subcutaneously tumor xenografts. On the other hand, LRIG1 overexpression inhibited glioma growth by significantly changing the expression pattern of cyclins, resulting in delayed cell cycle. Employing transwell invasion and wound scratch assay and gelatin zymography, LRIG1 inhibited U-251 MG cell invasion and migration by attenuating MMP2 and MMP9 production. Under ligand-stimulated conditions, p-ERK levels did not change, whereas p-AKT levels were inhibited in cells with LRIG1 upregulation, indicating that LRIG1 exerts more inhibiting effects on the PI3K/AKT pathway. Our findings suggest that LRIG1 restricted glioma aggressiveness by inhibiting cell proliferation, migration and invasion. Restoration of LRIG1 to glioma cells could offer a novel therapeutic strategy.

Introduction

Malignant astrocytic tumors are the most common devastating primary neoplasms of the central nervous system with hallmark characteristic of aggressive biological behavior, such as rapid proliferation potential and diffused invasive capability (1). The strong capacity of these tumors to invade and migrate into surrounding normal brain tissue in early stage usually prevents complete removal of the tumor during operation even with radical resection approach and these tumors inevitably recur with no effective treatment (2). Same as in other solid tumors, genetic alterations that result in activation of oncogenes and/or inactivation of tumor suppressor genes are the underlying causes of glioma (3). Sequential gain of oncogenes, such as epidermal growth factor receptor (EGFR) and/or loss of tumor suppressor genes, such as p53, phosphatase and tensin homolog (PTEN), provides the necessary foundation for step-wise progression of malignant glioma from initiation to transformation and aggressive progression (4,5).

Among various identified oncogenes associated with aggressiveness of glioma, EGFR is best characterized. EGFR is amplified in a range of 30–50% of high-grade malignant astrocytoma cases (6). Enhanced EGFR signaling was proven to contribute to glioma initiation, progression, treatment-resistance and poor survival (7). In a search for negative regulator of EGFR signaling, we had previously cloned and characterized the human leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) gene (8). LRIG1 belongs to the LRIG gene family, which also includes LRIG2 and LRIG3 (9). LRIG1 is located at chromosome 3p14.3 (10), a region frequently deleted in various human cancers (11). Previous studies demonstrated that LRIG1 was downregulated in conventional renal cell carcinoma, squamous cell carcinoma of the skin and lung (12), breast cancer (13), nasopharyngeal carcinoma (14) and in various human cancer cell lines (10,1517). LRIG1 negatively regulates EGFR, both by inducing receptor degradation (18,19) and by inhibiting signaling (20,21). Thus, ectopic expression of LRIG1 suppresses glioma cell proliferation (21,22) and invasion (23). However, the exact mechanisms behind these suppressive effects have not been thoroughly analyzed.

In this study, we investigated the effect of LRIG1 over-expression on the malignant glioma cell line U-251 MG and analyzed the dynamic cell cycle and screened its potential target cyclins. Next we investigated the effects of LRIG1 on glioma cell invasion and production of MMP2 and MMP9. Moreover, we found that LRIG1 can modulate EGFR downstream signaling pathway-PI3K/AKT expression. Our findings demonstrate LRIG1 may exert its tumor suppressor function via targeting EGFR and its down signaling pathway.

Materials and methods

Cell culture and transfection

The human glioma cell line U-251 MG purchased from American Type Culture Collection was cultured in Dulbecco’s modified Eagle’s medium containing 10% (v/v) fetal bovine serum (Hyclone, USA) in a humidified (37°C, 5% CO2) incubator. Cells were transfected with expression vector pLRIG1-GFP, encoding an LRIG1-GFP fusion protein (24) or, as vector control, pEGFP-N1 (Clontech), encoding GFP, using Lipofectamine 2000 (Invitrogen, USA) according to the manufacturer’s instructions. G418-resistant clones that represented possible stably transfected cells were ring-cloned and expanded for further experiments.

RNA isolation and qRT-PCR

Total RNAs from cultured cells were isolated using TRIzol reagent (Invitrogen) according to the manufacturer’s instructions. qRT-PCR was used to evaluate the expression levels of mRNAs as described before (25). Oligonucleotide primer sequences used were as follows: MMP2 sense ATGGATCCTGGCTTTCCC-3′ and antisense 5′-GCTTCCAAACTTCACGCTC-3′; MMP9 sense 5′-TGAC AGCGACAAGAAGTG-3′ and antisense 5′-CAGTGAAGCGG TACATAGG-3′; LRIG1, 18S primers used were the same as described in a previously published report (16).

Western blot analysis and immunohistochemical staining

Cells were lysed and analyzed by Western blotting as described (26). The following antibodies were used: LRIG1 (Santa Cruz, USA); anti-EGFR, anti-phospho-EGFR (Upstate, USA); anti-phospho-ERK, anti-ERK, anti-AKT, anti-phospho-AKT (Cell Signaling, USA); anti-cyclin A, anti-cyclin E, GAPDH (Boster, China); anti-cyclin D1 (Neomarker, USA).

MTT assay and soft agar colony formation assay

The proliferation rate of various cells were measured by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) cell viability/proliferation assay as described (26). Soft agar colony formation assay was performed as described (27).

Cell cycle synchronization and dynamic cell cycle analysis

Cells grown to 80% confluency in 10-cm dishes were synchronized in culture medium containing 250 ng/ml nocodazole for 24 h. Then, detached cells were shaken-off and collected and washed twice with phosphate buffered saline and then, replated subconfluently in complete growth medium. Cells were collected at indicated time-points after nocodazole washout. Dynamic cell cycle were analyzed by flow cytometry using propidium iodide (PI) staining and was performed as previously described (28). Cell lysates were collected at indicated time-points and immunoblotting was performed using anti-cyclin antibodies.

In vitro invasion assay and cell migration assay

In vitro invasion capabilities were measured in transwell chamber assay as previous described (26). Cell migration activities were examined by two-dimensional wound healing assay or in vitro scratch assay as previous described (25).

Gelatin zymography

Serum-free conditioned media were used in SDS-PAGE gelatin-substrate zymography to detect the activity of MMP2 and 9 as described (29). Briefly, 3×105 transfected U-251 MG cells were plated in 6-well plates, cultured in 1 ml serum-free DMEM and incubated for 24 h. Twenty microliters of serum-free culture medium per sample were prepared in non-denaturating loading buffer and were size-fractionated in 10% SDS-polyacrylamide gel impregnated with 0.1% gelatin. The gels were incubated in a developing buffer for 42 h at 37°C. Direct comparisons between separate gels were not made, because the intensity of background staining was variable. Experiments were repeated three times.

In vivo tumor model

Pooled populations of transfected U-251 MG cells were used for subcutaneous tumor growth experiments. Briefly, 1×106 glioma cells were injected into both flanks of female BALB/c athymic nude mouse at 4–6 week of age. Tumor volumes were calculated using the following formula: tumor volume = 1/2 × (shortest diameter)2 × longest diameter (mm3). These experiments were performed in accordance with the guidelines of the Animal Experimental Committee, China Institutes for Biological Sciences.

Statistical analysis

Results are expressed as means ± standard deviation (SD). Statistical analyses were performed using SPSS statistical software (SPSS Inc., Chicago, IL). Student’s t-test was used. Significance was defined as *p<0.05 and **p<0.01.

Results

Transformation of U-251 MG cells with LRIG1 expression vector

U-251 MG cells were transfected with pLRIG1-GFP, encoding an LRIG1-GFP fusion protein, or as a vector control, with pEGFP-N1, encoding GFP (green fluorescent protein). Quantitative reverse transcriptase PCR (qRT-PCR) showed that LRIG1 mRNA levels were increased 33-fold in the pLRIG1-GFP transformed cells compared to the control cells (Fig. 1A). Western blotting confirmed expression of the LRIG1-GFP fusion protein in the pLRIG1-GFP transformed cells but not in the control cells (Fig. 1B). As previously described (24,30), the LRIG1-GFP fusion protein was mainly located in the plasma membrane (Fig. 1C).

LRIG1 represses cell proliferation by delaying cell cycle progression

Next, we test dynamic cell cycle change in synchronized clonal cell line after exogenous LRIG1 introduction. Most of the control cells left G0/G1 phase and entered S phase at 16-h time-point and entered S phase at 20-h time-point (Fig. 2B, low panel), while most of the LRIG1 overexpression cells started to leave G0/G1 phase and entered S phase at 20 h and then, entered S phase at 24 h (Fig. 2B, upper panel). This experiment demonstrated that exogenous introduction of LRIG1 into U-251 MG cells caused cell cycle arrest at G0/G1 phase and delayed cell cycle to enter S phase (Fig. 2A and B).

As ectopic expression of LRIG1 in U-251 MG cells delayed cell cycle, we tested whether LRIG1 expression alters the expression of cyclins. As shown in Fig. 2C, the expression of cyclin D1 lasts apparently longer in U-251 MG-LRIG1 cells than control cells, indicating LRIG1 overexpression lengthened the time of the cell cycle. The peak expression level of cyclin E was at 4-h for control group, while it was at 12–16-h in LRIG1 overexpressing cells, indicating the cell cycle was arrested at G0/G1 phase and delayed to enter S phase. The time course expression pattern of cyclin A was not significantly altered, indicating the entry to M phase was not affected by LRIG1 overexpression. These results were consistent with the dynamic cell cycle analysis shown in Fig. 2A. We next evaluated the effect of LRIG1 on the growth of glioma cells. The results of cell proliferation assay showed that ectopic expression of LRIG1 led to significant inhibition of cell proliferation compared to control cells (Fig. 2D). These results indicate a growth-inhibitory role of LRIG1 on glioma cell line U-251 MG.

LRIG1 inhibits colony formation in vitro and tumorigenicity in vivo

To examine the potential role of LRIG1 in tumorigenesis, the capacity of colony formation was evaluated on LRIG1 overexpressing and control cells. Notably, LRIG1 overexpression cells displayed obviously fewer and smaller colonies compared with control cells (Fig. 3A). This result indicated a growth-inhibitory role of LRIG1 on glioma cells. To further determine the antitumor effects of LRIG1, LRIG1 overexpression and control cells were injected into five nude mice as described. Although tumor volume between LRIG1 overexpression and control groups was not statistically significant (p= 0.28, data not shown), there was a trend for loss of LRIG1 with a higher risk with increasing tumor size. The average tumor volume was 106.4 mm3 in control group vs 6.89 mm3 in LRIG1 overexpression group at the end of eighth week after implantation. The tumor incidence was 80% (8 tumors/10 injection sites) in control group vs 30% (3 tumors/10 injection sites) in LRIG1 overexpression group (Fig. 3B and C). The results indicate that overexpression of LRIG1 inhibited tumorigenicity of U-251 MG cells in the nude mouse xenograft model.

LRIG1 inhibits U-251 MG cell migration and invasion and decreases secretion and activity of MMP2 and MMP9

In the following part of this study, we investigated whether over-expression of LRIG1 could affect cell invasion and migration. We found that invasion cells in the LRIG1 overexpressing cells were decreased to 12% of that of the control cells (Fig. 4A). Cell migration capability was further tested by wound scratch assay. The relative migrated distances of LRIG1 overexpressing cells were 45% of the distances migrated by the control cells at 48 h (Fig. 4B).

Gelatin zymography assay was performed to investigate the effect of the LRIG1 on the activity of MMP2 and MMP9. After exogenous introduction of LRIG1 into U-251 MG cells, MMP9 activity was significantly decreased, while MMP2 activity was modestly decreased (Fig. 4C). To further confirm the effect of LRIG1 on MMPs, quantitative real-time PCR results showed that after exogenous introduction of LRIG1 in to U-251 MG cell, the MMP2 mRNA levels were decreased to 44.8% and MMP9 mRNA levels were decreased to 39.5%, compared to control group (Fig. 4D). These results suggest that LRIG1 plays an important role in suppressing MMP2 and 9 production and is involved in invasion and migration in U-251 MG cells.

LRIG1 exerts more effect on PI3K/AKT pathway than MAPK/ERK1/2 pathway

In the next experiments, we confirmed that LRIG1 negatively regulated the levels of EGFR and phosphorylated EGFR in our experimental system (Fig. 5) (19). We then analyzed the expressions and phosphorylation of two key intracellular signal molecules, AKT and ERK1/2. The basal expression levels of AKT and ERK were not altered. Further, we showed the levels of phosphorylated AKT decreased in LRIG1 overexpressing cells, while the phosphorylated ERK level was not altered, indicating the LRIG1 exerts more influence on intracellular PI3K/AKT pathway than MAPK/ERK1/2 pathway (Fig. 5).

Discussion

In this study, we provided that LRIG1 functioned as a negative growth regulator for glioma cells and showed that over-expression of LRIG1 inhibited glioma to form clonies in vitro and to develop tumors in vivo. Ectopic expression of LRIG1 suppressed the invasion and migration of cultured glioma cells by regulating secreation of MMP2 and MMP9. These observations provide further understanding of the molecular mechanisms of inhibitory effects of LRIG1 on glioma cell aggressiveness.

Malignant astrocytic tumors are the most aggressive and highly lethal type of brain tumor with a strong ability of rapid proliferation and diffuse invasion into surrounding normal brain tissue. While the molecular events involved in the initiation of glioblastoma are not fully understood, it is believed that, deregulated tumor cell proliferation appears to be a critical early event in glioma development. To assess the potential role of LRIG1 in glioma cell growth, we over-expressed LRIG1 in glioma cell line U-251 MG. Our study revealed that LRIG1 overexpression resulted in decreased tumor cell growth confirmed by MTT and soft agar colony assay. We further showed that LRIG1 overexpression inhibits glioma cell proliferation in vivo. Cellular proliferation is tightly regulated by progression through the cell cycle of DNA synthesis and mitosis by the formation and activity of cyclin-cyclin-dependent-kinase (CDK) complexes (31). We demonstrated that enhanced expression of LRIG1 in glioma cells significantly changed cell cycle control protein cyclin D1, E expression pattern, resulting in cell cycle delay at G0/G1. The strong exogenous LRIG1 regulation over cell cycle control further implies that fundamental alterations of cyclins and tumor suppressors result in deregulated cell cycle and unrestrained cell proliferation (32).

In addition to its function as a potent negative growth regulator, the present and our previous study showed that LRIG1 overexpression led to a significant decrease of glioblastoma cell migration and invasion (23), while the molecular mechanisms of its inhibitory effects on glioma cell metastasis still remain obscure. Matrix metalloproteinases (MMPs) are a family of extracellular zinc-dependent endopeptidases that selectively cleave the protein components of the extracellular matrix (33). Numerous studies have demonstrated that malignant glioma cells secrete MMPs to facilitate their migration and invasion (34). We found that overexpression of LRIG1 inhibited glioma cell invasion and migration by suppressing MMP2 and 9 production. Because EGFR activation can promote MMP2 (35) and MMP9 expression (36,37) and LRIG1 negatively regulated the level of EGFR, we deduced that LRIG1 regulated glioma cell metastasis by attenuating EGFR signaling pathway, at least in part.

Accumulating evidence has identified the EGFR and its downstream signal networks as commonly deregulated components in the oncogenesis of glioma, especially primary GBM (1,38). Once activated and subsequent dimerization, EGFR activates a variety of intracellular phosphorylation-mediated signal transduction events (38). The important events in glioma biology include the RAS/mitogen activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)/AKT pathways that regulate cell proliferation, survival and invasion (1,38). Upregulation of LRIG1 influenced expression levels of p-AKT rather than p-ERK in response to ligand stimulation. Constitutive activation of PI3K/AKT signaling pathway plays a pivotal role in the pathogenesis and malignant progression of several human cancers, including glioblastoma (39). Herein, we found that overexpression of LRIG1 negatively regulated glioma cell proliferation, migration and invasion, possibly by inhibiting EGFR activation and its downstream signaling PI3K/AKT pathway.

Collectively, we identified that LRIG1 overexpression resulted in decreased cell growth in vitro and in vivo and delay in cell cycle by changing the expression pattern of cyclins. LRIG1 also attenuated invasion of glioma cells by decreasing production of MMP2 and 9. Analysis of the signaling pathways revealed that LRIG1 regulated EGFR signaling pathway transduction involved in the regulation of tumour cell proliferation and migration and invasion. It was demonstrated that LRIG1 exerts its tumor suppressor function and implicates its potential targets for future brain tumor treatments.

Acknowledgements

This study was supported by National Natural and Science Foundation of China (No. 81001116).

References

1 

Furnari FB, Fenton T, Bachoo RM, et al: Malignant astrocytic glioma: genetics, biology and paths to treatment. Genes Dev. 21:2683–2710. 2007. View Article : Google Scholar : PubMed/NCBI

2 

MacDonald TJ, Aguilera D and Kramm CM: Treatment of high-grade glioma in children and adolescents. Neurooncology. 13:1049–1058. 2011.PubMed/NCBI

3 

Sathornsumetee S, Reardon DA, Desjardins A, Quinn JA, Vredenburgh JJ and Rich JN: Molecularly targeted therapy for malignant glioma. Cancer. 110:13–24. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Smith JS, Tachibana I, Passe SM, et al: PTEN mutation, EGFR amplification and outcome in patients with anaplastic astrocytoma and glioblastoma multiforme. J Natl Cancer Inst. 93:1246–1256. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Reifenberger G and Collins VP: Pathology and molecular genetics of astrocytic gliomas. J Mol Med. 82:656–670. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Frederick L, Wang XY, Eley G and James CD: Diversity and frequency of epidermal growth factor receptor mutations in human glioblastomas. Cancer Res. 60:1383–1387. 2000.PubMed/NCBI

7 

Heimberger AB, Hlatky R, Suki D, et al: Prognostic effect of epidermal growth factor receptor and EGFRvIII in glioblastoma multiforme patients. Clin Cancer Res. 11:1462–1466. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Nilsson J, Vallbo C, Guo D, et al: Cloning, characterization and expression of human LIG1. Biochem Biophys Res Commun. 284:1155–1161. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Guo D, Holmlund C, Henriksson R and Hedman H: The LRIG gene family has three vertebrate paralogs widely expressed in human and mouse tissues and a homolog in Ascidiacea. Genomics. 84:157–165. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Hedman H, Nilsson J, Guo D and Henriksson R: Is LRIG1 a tumour suppressor gene at chromosome 3p14.3? Acta Oncol. 41:352–354. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Knuutila S, Aalto Y, Autio K, et al: DNA copy number losses in human neoplasms. Am J Pathol. 155:683–694. 1999. View Article : Google Scholar : PubMed/NCBI

12 

Boelens MC, van den Berg A, Fehrmann RS, et al: Current smoking-specific gene expression signature in normal bronchial epithelium is enhanced in squamous cell lung cancer. J Pathol. 218:182–191. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Miller JK, Shattuck DL, Ingalla EQ, et al: Suppression of the negative regulator LRIG1 contributes to ErbB2 overexpression in breast cancer. Cancer Res. 68:8286–8294. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Sheu JJ, Lee CH, Ko JY, et al: Chromosome 3p12.3–p14.2 and 3q26.2–q26.32 are genomic markers for prognosis of advanced nasopharyngeal carcinoma. Cancer Epidemiol Biomarkers Prev. 18:2709–2716. 2009.

15 

Tanemura A, Nagasawa T, Inui S and Itami S: LRIG-1 provides a novel prognostic predictor in squamous cell carcinoma of the skin: immunohistochemical analysis for 38 cases. Dermatol Surg. 31:423–430. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Thomasson M, Hedman H, Guo D, Ljungberg B and Henriksson R: LRIG1 and epidermal growth factor receptor in renal cell carcinoma: a quantitative RT-PCR and immunohisto-chemical analysis. Br J Cancer. 89:1285–1289. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Yang WM, Yan ZJ, Ye ZQ and Guo DS: LRIG1, a candidate tumour-suppressor gene in human bladder cancer cell line BIU87. BJU Int. 98:898–902. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Gur G, Rubin C, Katz M, et al: LRIG1 restricts growth factor signaling by enhancing receptor ubiquitylation and degradation. EMBO J. 23:3270–3281. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Laederich MB, Funes-Duran M, Yen L, et al: The leucine-rich repeat protein LRIG1 is a negative regulator of ErbB family receptor tyrosine kinases. J Biol Chem. 279:47050–47056. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Goldoni S, Iozzo RA, Kay P, et al: A soluble ectodomain of LRIG1 inhibits cancer cell growth by attenuating basal and ligand-dependent EGFR activity. Oncogene. 26:368–381. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Yi W, Holmlund C, Nilsson J, et al: Paracrine regulation of growth factor signaling by shed leucine-rich repeats and immunoglobulin-like domains 1. Exp Cell Res. 317:504–512. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Stutz MA, Shattuck DL, Laederich MB, Carraway KL III and Sweeney C: LRIG1 negatively regulates the oncogenic EGF receptor mutant EGFRvIII. Oncogene. 27:5741–5752. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Ye F, Gao Q, Xu T, et al: Upregulation of LRIG1 suppresses malignant glioma cell growth by attenuating EGFR activity. J Neurooncol. 94:183–194. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Nilsson J, Starefeldt A, Henriksson R and Hedman H: LRIG1 protein in human cells and tissues. Cell Tissue Res. 312:65–71. 2003.PubMed/NCBI

25 

Song H, Li Y, Lee J, Schwartz AL and Bu G: Low-density lipoprotein receptor-related protein 1 promotes cancer cell migration and invasion by inducing the expression of matrix metalloproteinases 2 and 9. Cancer Res. 69:879–886. 2009. View Article : Google Scholar

26 

Wang B, Han L, Chen R, et al: Downregulation of LRIG2 expression by RNA interference inhibits glioblastoma cell (GL15) growth, causes cell cycle redistribution, increases cell apoptosis and enhances cell adhesion and invasion in vitro. Cancer Biol Ther. 8:1018–1023. 2009. View Article : Google Scholar

27 

Lau YK, Murray LB, Houshmandi SS, Xu Y, Gutmann DH and Yu Q: Merlin is a potent inhibitor of glioma growth. Cancer Res. 68:5733–5742. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Cai M, Han L, Chen R, et al: Inhibition of LRIG3 gene expression via RNA interference modulates the proliferation, cell cycle, cell apoptosis, adhesion and invasion of glioblastoma cell (GL15). Cancer Lett. 278:104–112. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Galis ZS, Muszynski M, Sukhova GK, et al: Cytokine-stimulated human vascular smooth muscle cells synthesize a complement of enzymes required for extracellular matrix digestion. Circ Res. 75:181–189. 1994. View Article : Google Scholar

30 

Guo D, Nilsson J, Haapasalo H, et al: Perinuclear leucine-rich repeats and immunoglobulin-like domain proteins (LRIG1-3) as prognostic indicators in astrocytic tumors. Acta Neuropathol. 111:238–246. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Malumbres M and Barbacid M: Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. 9:153–166. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Vermeulen K, Van Bockstaele DR and Berneman ZN: The cell cycle: a review of regulation, deregulation and therapeutic targets in cancer. Cell Prolif. 36:131–149. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Stamenkovic I: Extracellular matrix remodelling: the role of matrix metalloproteinases. J Pathol. 200:448–464. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Nakada M, Okada Y and Yamashita J: The role of matrix metalloproteinases in glioma invasion. Front Biosci. 8:e261–269. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Kodali R, Hajjou M, Berman AB, et al: Chemokines induce matrix metalloproteinase-2 through activation of epidermal growth factor receptor in arterial smooth muscle cells. Cardiovasc Res. 69:706–715. 2006. View Article : Google Scholar

36 

Alper O, Bergmann-Leitner ES, Bennett TA, Hacker NF, Stromberg K and Stetler-Stevenson WG: Epidermal growth factor receptor signaling and the invasive phenotype of ovarian carcinoma cells. J Natl Cancer Inst. 93:1375–1384. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Cox G, Jones JL and O’Byrne KJ: Matrix metalloproteinase 9 and the epidermal growth factor signal pathway in operable non-small cell lung cancer. Clin Cancer Res. 6:2349–2355. 2000.PubMed/NCBI

38 

Huang PH, Xu AM and White FM: Oncogenic EGFR signaling networks in glioma. Sci Signal. 2:re62009.PubMed/NCBI

39 

Knobbe CB, Trampe-Kieslich A and Reifenberger G: Genetic alteration and expression of the phosphoinositol-3-kinase/Akt pathway genes PIK3CA and PIKE in human glioblastomas. Neuropathol Appl Neurobiol. 31:486–490. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March 2013
Volume 42 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Mao F, Wang B, Xiao Q, Xi G, Sun W, Zhang H, Ye F, Wan F, Guo D, Lei T, Lei T, et al: A role for LRIG1 in the regulation of malignant glioma aggressiveness. Int J Oncol 42: 1081-1087, 2013
APA
Mao, F., Wang, B., Xiao, Q., Xi, G., Sun, W., Zhang, H. ... Chen, X. (2013). A role for LRIG1 in the regulation of malignant glioma aggressiveness. International Journal of Oncology, 42, 1081-1087. https://doi.org/10.3892/ijo.2013.1776
MLA
Mao, F., Wang, B., Xiao, Q., Xi, G., Sun, W., Zhang, H., Ye, F., Wan, F., Guo, D., Lei, T., Chen, X."A role for LRIG1 in the regulation of malignant glioma aggressiveness". International Journal of Oncology 42.3 (2013): 1081-1087.
Chicago
Mao, F., Wang, B., Xiao, Q., Xi, G., Sun, W., Zhang, H., Ye, F., Wan, F., Guo, D., Lei, T., Chen, X."A role for LRIG1 in the regulation of malignant glioma aggressiveness". International Journal of Oncology 42, no. 3 (2013): 1081-1087. https://doi.org/10.3892/ijo.2013.1776