Open Access

Human giant larvae‑1 promotes migration and invasion of malignant glioma cells by regulating N‑cadherin

  • Authors:
    • Yan Wang
    • Yu Zhang
    • Ben Sang
    • Xianlong Zhu
    • Rutong Yu
    • Xiuping Zhou
  • View Affiliations

  • Published online on: January 4, 2021     https://doi.org/10.3892/ol.2021.12428
  • Article Number: 167
  • Copyright: © Wang 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

Human giant larvae‑1 (Hugl‑1) is a human homologue of Drosophila tumor suppressor lethal (2)‑giant larvae and has been reported to be involved in the development of human malignancies. Previous studies performed by our group demonstrated that Hugl‑1 inhibits glioma cell proliferation in an intracranial model of nude mice. However, the exact molecular mechanisms underlying the participation of Hugl‑1 in glioma invasion and migration, and in the depolarizing process remain largely unknown. Utilizing the U251‑MG cells with stable expression of Hugl‑1, the present study used wound healing, Transwell invasion and western blot assays to explore the role and specific mechanism of Hugl‑1 in glioma invasion and migration. The results of the present study demonstrated that overexpression of Hugl‑1 decreased cell‑cell adhesion and increased cell‑cell extracellular matrix adhesion. In addition, overexpression of Hugl‑1 promoted pseudopodia formation, glioma cell migration and invasion. The molecular mechanism of action involved the negative regulation of N‑cadherin protein levels by Hugl‑1. Overexpression or knockdown of N‑cadherin partially suppressed or enhanced the effects of Hugl‑1 on glioma cell migration and invasion, respectively. Furthermore, Hugl‑1 inhibited cell proliferation, while promoting cell migration, which suggests that it may serve a two‑sided biological role in cellular processes. Taken together, these results suggest that Hugl‑1 promotes the migration and invasion of malignant glioma cells by decreasing N‑cadherin expression. Thus, Hugl‑1 may be applied in the development of targeted and personalized treatment.

Introduction

Glioblastoma (GBM) is the most common malignant primary central nervous system tumor, with an average survival time of 12–15 months (1). The failure of conventional treatments is attributed to its highly invasive and diffusely infiltrative nature (2). Thus, the identification of novel therapeutic targets and strategies to improve the efficacy of existing forms of treatment is urgently required.

Cell polarity and intercellular adhesion play a key role in regulating normal tissue structure and function (3). The disruption of cell polarity and cell adhesion is usually associated with tumor formation (4). Lethal (2)-giant larvae (Lgl) is a cortical cytoskeletal protein, which was initially identified in Drosophila and exhibits notable effects in the establishment and maintenance of apical-basal epithelial polarity, asymmetric cell division, tissue integrity and cell proliferation (5). The human homologues of Lgl1 and Lgl2 are termed human giant larvae (Hugl)-1 and Hugl-2. Mutations that cause loss of function of Lgl have been demonstrated to result in tissue overgrowth and neoplastic tumor formation (6,7). The Hugl-1 protein shares 62.5% similarity with Lgl (810). A previous study indicated that hepatocellular carcinoma (HCC) contains frequent mutations of Hugl-1, whereas overexpression of HCC-derived aberrant Hugl-1 variants significantly promote HCC cell migration and invasion (11). In addition, Hugl-1 expression is downregulated in different types of human cancer, including colorectal cancer, melanoma, prostate cancer, breast cancer, endometrial cancer, lung cancer and esophageal carcinoma (1215). Hugl-1 expression is positively associated with a higher survival rate in patients with pancreatic carcinoma, suggesting its use as a reliable prognostic marker (16). The majority of previous studies have focused on epithelial-derived tumors (1115), thus the role of Hugl-1 in gliomas (glia-derived tumors) has not yet been fully elucidated. A previous study performed by our group has demonstrated that Hugl-1 protein levels decrease in human glioma tissues, whereas overexpression of Hugl-1 attenuates glioma cell proliferation in an intracranial model of nude mice; however, it does not affect glioma cell proliferation in vitro (17). As a regulator of cell polarity, Hugl-1 exhibits important properties that are closely associated with cell adhesion and cytoskeletal function and structure (18). However, the role of Hugl-1 in glioma migration and invasion has not yet been fully investigated.

Cell surface adhesion molecules are the main mediators of cell-cell interactions, which are essential for tumor malignant biological behaviors. Reorganization of the cell cytoskeleton and alteration of cell-cell adhesion are required prior to cell migration (19,20). These processes are mainly mediated by cadherin family members. It is reported that E-cadherin is essential for the normal migration of cranial neural crest cells in vivo, while P-cadherin, also known as placental cadherin, is associated with malignant invasion of esophageal squamous cells (2124). In most tumors, N-cadherin expression is often upregulated and can be used as a promoter of tumor invasion (25,26). N-cadherin expression in epithelial cells can induce morphological changes of fibroblast phenotype and orchestrate cell-cell communication during cell movement (27). N-cadherin is also known as an epithelial-to-mesenchymal transition marker and exhibits several functions according to the cell environment that can promote adhesion or induce migration (28,29). However, increasing evidence suggests that N-cadherin exhibits tumor-inhibitory roles in non-epithelial derived neoplasms, such as osteosarcoma and glioma (27,30). Thus, the functions of N-cadherin may be tumor-type specific (27).

The present study aimed to investigate the role and molecular mechanism of Hugl-1 on the motility of malignant glioma cells.

Materials and methods

Cell culture

The U251-MG glioma cell line was purchased from the Shanghai Cell Bank, Type Culture Collection Committee, Chinese Academy of Sciences. Cells were maintained in DMEM/F-12 media (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (FBS; Biological Industries), at 37°C in 5% CO2.

Stable transfection of Hugl-1 into U251-MG cells

The pEGFP-C1 vector alone or the pEGFP-C1-Hugl-1 construct (provided by Professor Zhengjun Chen, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences) was transfected into U251-MG cells (GFP-Vector or GFP-Hugl-1 cells, respectively) using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. Briefly, 9 µl Lipofectamine® 2000 and 3 µg of the Hugl-1 expression plasmid were added to 1 ml Opti-MEM (Invitrogen; Thermo Fisher Scientific, Inc.) and incubated for 10 min at room temperature. The plasmid and Lipofectamine were mixed together and incubated for 30 min before adding them to the U251-MG cells. The transfectants were subsequently selected using G418 (1,200 µg/ml), and single-cell clones were obtained following 3–4 weeks of growth for expansion. The G418-resistant cells were used for subsequent experiments. DsRed-C1 or DsRed-N-cadherin plasmids were kindly provided by the Laboratory of Cell Biology of Northeast Normal University (Changchun, China). DsRed-C1 or DsRed-N-cadherin plasmids were transfected into Hugl-1 overexpressing U251-MG cells. The specific transfection procedure was the same as that of Hugl-1.

Digestion assay

Cultured GFP-Vector or GFP-Hugl-1 cells were digested with trypsin simultaneously (Gibco; Thermo Fisher Scientific, Inc.). Briefly, cells were digested with trypsin at room temperature for 8 min and observed at designated time points (0, 2, 4 and 8 min) under an inverted light microscope during trypsinization at ×200 magnification (Olympus Corporation; IX71).

Attachment assay

The attachment assay was performed using 12-well plates. The cell suspension was added into the plates and cell images were obtained at 3, 6, 9 and 24 h using an inverted light microscope at ×400 magnification (Olympus Corporation; IX71).

Wound healing assay

Cell migration was assessed via the wound healing assay, as previously described (31). Briefly, cells were seeded into 6-well plates and cultured until they reached ~80% confluence. The cell monolayers were scratched using a 10 µl sterile pipette tip. Cells were subsequently washed twice with PBS to remove floating cells and serum-free DMEM/F-12 medium (Gibco; Thermo Fisher Scientific, Inc.) was added. Cell wound healing was observed at 0, 24 or 48 h using an inverted light microscope at ×200 magnification (Olympus Corporation; IX71).

Transwell invasion assay

The cell invasion assay was performed as previously described (31). Briefly, Transwell membranes were precoated with DMEM-diluted Matrigel® (BD Biosciences) for 3 h at 37°C. Cells (2×104) were plated in the upper chambers of Transwell plates in 200 µl serum-free culture DMEM/F-12 medium (Gibco; Thermo Fisher Scientific, Inc.). A total of 500 µl DMEM/F-12 medium supplemented with 10% FBS was plated in the lower chambers. Following incubation at room temperature for 24 h, the invasive cells were fixed in methanol for 15 min at room temperature and subsequently stained for 15 min at room temperature with 0.1% crystal violet. Invasive cells were viewed and counted under an inverted light microscope at ×200 magnification (Olympus Corporation; IX71).

Western blotting

U251-MG cells were lysed with RIPA lysis buffer (50 mM Tris-HCl, 150 mM NaCl, 0.5% sodium deoxycholate and 0.1% SDS) supplemented with protease inhibitor cocktail, and total proteins were quantified using a BCA kit (Beyotime Institute of Biotechnology) according to the manufacturer's protocol. Western blotting was performed as previously described (32). Briefly, equal amounts of protein (20 µg/lane) were separated by SDS-PAGE on 8 or 10% gels, transferred onto polyvinylidene fluoride membranes and blocked using 3% BSA (Sangon Biotech Co., Ltd.) for 2 h at room temperature. The membranes were then incubated with primary antibodies against Hugl-1 (1:500) [kindly gifted by Dr ZG Luo from the Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (33)], N-cadherin (1:2,000; cat. no. ab76011; Abcam), β-catenin (1:5,000; cat. no. ab32572; Abcam), integrinβ1 (1:1,000; cat. no. ab134179; Abcam) and β-actin (1:1,000; cat. no. MABT523; EMD Millipore). Following the primary antibody incubation at 4°C overnight, membranes were probed with HRP-labelled goat anti-rabbit or anti-mouse IgG secondary antibodies (1:4,000; cat. nos. sc2004 and sc2005; Santa Cruz Biotechnology, Inc.) at room temperature for 2 h. The signal was detected using the Pierce ECL Plus Western Blotting Substrate (Pierce; Thermo Fisher Scientific, Inc.) and exposed to ChemiDoc Touch (Bio-Rad Laboratories, Inc.). Finally, gray analysis was performed using ImageJ 1.48V (National Institutes of Health) to compare the level of each protein.

Phalloidin staining

U251-MG cells were incubated for 24 h at 37°C and cultured in DMEM/F-12 medium supplemented with 10% FBS for 30 min at 37°C. Subsequently, cells were fixed with 4% paraformaldehyde for 10 min at room temperature, washed twice with PBS, and 0.5% Triton X-100 was added for 5 min at room temperature. Finally, 200 µl of the diluted phalloidin (cat. no. 94072; Sigma-Aldrich; Merck KGaA) was added and incubated at room temperature in the dark for 30 min. Actin filaments were observed using an inverted fluorescence microscope at ×400 magnification (Olympus Corporation; IX71).

Statistical analysis

Statistical analysis was performed using SPSS 13.0 software (SPSS, Inc.). All experiments were performed in triplicate and data are presented as the mean ± standard error of the mean. Student's unpaired t-test was used to compare differences between two groups, while one-way AVONA followed by Tukey's post hoc test were used to compare differences between multiple groups. P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of Hugl-1 on cell adhesive activity

It has been demonstrated that Hugl-1 protein expression is downregulated in human glioma tissues compared with normal brain tissues (17). Given that Hugl-1 acts as a tumor suppressor in several human tumors, and its expressed at low levels in gliomas, the effect of Hugl-1 downregulation on this basis may be negligible. In addition, we detected six different glioma cell lines in the previous study and demonstrated that Hugl-1 protein expression was extremely low in U251-MG cells (17). Thus, to investigate the role of Hugl-1 in glioma, stable GFP-Hugl-1 overexpression was established in U251-MG glioma cells, and constitutive expression was assessed via western blotting (Fig. 1A and B). Due to the high molecular weight of Hugl-1 (115 kDa) (13), the molecular weight of GFP-Hugl-1 fusing protein was 141 kDa (115 kDa + 26 kDa), which caused weaker GFP signaling in the GFP-Hugl-1 group compared with the GFP group. To determine the difference in the adhesive ability between the GFP-Hugl-1 and GFP groups, cells were trypsinized, and the results demonstrated that Hugl-1 overexpressing cells retracted more slowly compared with the GFP control cells (Fig. 1C), suggesting a better cell-extracellular matrix adhesive ability. Conversely, Hugl-1 overexpressing cells extended pseudopodia faster than GFP cells following plating (Fig. 1D). Notably, GFP cells formed cell aggregates unlike Hugl-1 overexpressing cells (Fig. 1D, black arrow), suggesting that upregulation of Hugl-1 decreases cell-cell adhesive activity. The cells presented in Fig. 1D exhibited complete roundness and good refraction, indicating that the cells were healthy. In addition, the digested cells were placed in a culture dish and gradually expanded.

N-cadherin is a member of the calcium-dependent adhesion molecule family, which mediates adhesion between homotypic cells (34). Thus, N-cadherin protein expression was detected at 3, 6 and 24 h following plating. The results demonstrated that N-cadherin expression was lower in Hugl-1 overexpressing cells compared with GFP cells at 3 and 6 h following plating (Fig. 1E and F).

Previous studies have demonstrated that adhesion molecules play an important role in the early stage of cell adhesion (35,36), which gradually decreases overtime (37). In the present study, no significant difference was observed in N-cadherin expression between the two groups 24 h after plating. Taken together, these results suggest that overexpression of Hugl-1 decreases cell-cell adhesion, while increasing cell-extracellular matrix adhesion by regulating N-cadherin expression.

Hugl-1 accelerates cytoskeletal remodeling

To fully characterize the intercellular adhesion defects observed in Hugl-1 overexpressing cells, the intracellular organization of the cytoskeleton was assessed. Cells were incubated for 24 h and cultured in media supplemented with 10% FBS for 30 min. Subsequently, cells were stained with phalloidin-conjugated actin to assess actin reassembling. The results demonstrated that Hugl-1 overexpressing cells expanded their lamellipodia earlier, which contained concentrated F-actin, and stretched faster than GFP cells (Fig. 2A). Previous studies have established β-catenin as a promoter signal, which is not only a key transcription factor in the Wnt signaling pathway but also a structural adaptor between cadherin and actin skeleton during cell adhesion (38,39). The present study hypothesized that β-catenin may mediate cytoskeleton remodeling by overexpressing Hugl-1. The results of the present study demonstrated that β-catenin expression decreased in Hugl-1 overexpressing cells, while the expression levels of integrin β1, another important molecule involved in cytoskeleton remodeling and adhesion (40), remained unchanged (Fig. 2B and C). Collectively, these results suggest that Hugl-1 may accelerate cell cytoskeleton reorganization by regulating the N-cadherin-β-catenin complex.

Hugl-1 promotes the migration and invasion of glioma cells

The effect of overexpressing Hugl-1 on the migration of glioma cells was assessed via the wound healing assay. The results demonstrated that Hugl-1 overexpressing glioma cells exhibited faster wound healing than GFP cells after 48 h (Fig. 3A and B), whereby the number of migratory cells significantly increased to 30±7% (P<0.01; Fig. 3A and B). In addition, the effect of overexpressing Hugl-1 on the invasion of glioma cells was assessed via the Transwell assay. The results demonstrated that the number of invasive cells significantly increased to 139±5% following overexpression of Hugl-1 (P<0.01; Fig. 3C and D), which confirms that Hugl-1 promotes the invasive ability of U251-MG cells.

N-cadherin partially mediates the effects of Hugl-1 expression on glioma cell migration

N-cadherin plays a key role in regulating cell polarity and motility (27). Based on the results presented in Fig. 1E, whether N-cadherin mediates the effects of Hugl-1 expression on glioma cell migration was subsequently assessed. N-cadherin was overexpressed in GFP and Hugl-1 overexpressing cells (Fig. 4A), and the results demonstrated that upregulation of Hugl-1 promoted cell migration, the effects of which were reversed following overexpression of N-cadherin (Fig. 4B and E).

Generally, the exogenous protein level is higher than that of the endogenous level. However, considering the high molecular weight of GFP-Hugl-1 (141 kDa) and N-cadherin (127 kDa), and the relatively low transit transfection efficiency (12,13), the exogenous protein level was lower than the endogenous level in the present study (Fig. 4A). Notably, the induction effect of Hugl-1 on glioma cell migration was partially abolished following overexpression of N-cadherin, compared with that of the Hugl-1 overexpressing cells (Fig. 4B and E). Conversely, N-cadherin knockdown promoted cell migration, and the effects were similar to those noted in the Hugl-1 overexpressing cells (Fig. 4C, D and F). Furthermore, the increased migratory ability induced by Hugl-1 upregulation was significantly enhanced following downregulation of N-cadherin in glioma cells (Fig. 4D and F). Taken together, these results suggest that N-cadherin partially mediates the effects of Hugl-1 expression on glioma cell migration.

Discussion

Intercellular adhesion plays a crucial role in the maintenance of cell polarity to regulate normal tissue architecture and function (41,42). This process is often disrupted in neoplastic tumors (43,44). Loss of polarity is considered one of the trigger signals for tumorigenesis and invasion of surrounding tissues (45,46). As a cell polarity regulator, Hugl-1 expression is downregulated in several types of human cancer, such as squamous cell lung carcinoma (15), esophageal carcinoma (47), pancreatic carcinoma (16), endometrial cancer (14) and malignant melanoma (12), and is inversely associated with patient prognosis (1115). We previously demonstrated that Hugl-1 can inhibit tumor progression in vivo, while no significant effects on cell proliferation were observed in vitro (17).

The results of the present study demonstrated that overexpression of Hugl-1 decreased cell-cell adhesion, probably by regulating N-cadherin protein expression. In addition, overexpression of Hugl-1 promoted glioma cell migration and invasion. Notably, overexpression or knockdown of N-cadherin partially suppressed or enhanced the induction effect of Hugl-1 expression on glioma cell migration and invasion, respectively. Taken together, these results suggest that Hugl-1 promotes migration and invasion of malignant glioma cells by decreasing N-cadherin expression, thus Hugl-1 may act as a novel therapeutic target in patients with GBM, and function as a marker of GBM prognosis.

Schimanski et al (13) demonstrated that Hugl-1 expression is lost in 75% of tumor samples and that these deletions are associated with advanced disease stage, particularly with lymph node metastasis. Similarly, loss of Hugl-1 expression in endometrial cancer may contribute to lymph node metastasis (14). Notably, overexpression of wild-type Hugl-1 inhibits HCC migration and invasion (11). Kuphal et al (12) reported that upregulation of Hugl-1 increases cell adhesion and decreases cell migration in malignant melanoma. However, the results of the present study demonstrated that overexpression of Hugl-1 promoted glioma cell migration and invasion. Although Hugl-1 expression decreases in malignant melanoma, HCC and gliomas, it exhibits opposite effects on cell migration and invasion (promotion versus inhibition) in different types of tumors (11,12,17). These differences may be due to the different cell types used in each experiment under specific conditions. Kuphal et al (12) and Lu et al (11) used Mel Im or SK-HEP-1 cells, which are epithelial cells, while the present study used U251-MG glioma cells, a cell type that belongs to glia-derived cells (48). However, whether the functions of Hugl-1 are cell-type specific remains unknown and should be investigated in prospective studies.

Cell migration and invasion include multiple processes, such as extracellular matrix degradation, cytoskeletal reorganization, de-adhesion and adhesion (49,50). Cytoskeletal reorganization is an important process that affects assembly and disassembly of cell-cell adhesions and leads to morphological and motility changes of tumor cells (50). It is well-known that low expression levels of Hugl-1 in gliomas decrease cell-cell adhesion, promote cell migration and ultimately contribute to cancer cell dissemination and tumor progression (51). However, the results of the present study are not consistent with these conclusions, suggesting that the balance of polar proteins may be the optimum condition for maintaining cell homeostasis (52). In addition, the results of the present study demonstrated that overexpression of Hugl-1 significantly promoted pseudopodia formation and supported the enhanced cell-extracellular matrix adhesion. Asano et al (53) reported that N-cadherin expression is negatively associated with tumor invasion. The results of the present study demonstrated that overexpression of Hugl-1 decreased cell-cell adhesion and increased cell migration, which was consistent with the decreased protein levels of N-cadherin. Recently, Jossin et al (54) reported that LLGL1 directly binds to N-cadherin and is able to promote its internalization, while disrupting the N-cadherin-LLGL1 interaction, which results in cortical heterotopias. The results of the present study are consistent with these findings.

In conclusion, the results of the present study demonstrated that Hugl-1 promoted glioma cell migration and invasion by decreasing N-cadherin expression. Combined with our previous studies, the results presented here provide a novel role for Hugl-1, which includes inhibition of cell proliferation, while promoting cell migration in glioma, suggesting that Hugl-1 may play two-sided roles in malignant biological processes. In addition, the results presented here provide useful information for the clinical diagnosis of malignant GBM and the prognosis of patients with GBM. Further studies are required to determine the exact role and precise molecular mechanism of the cell polarity molecule, Hugl-1, for the effective treatment of glioma. Although the present study investigated the role of Hugl-1 in glioma cells, it still presented some limitations. The current experiments were all completed at the cellular level; therefore, there is a lack of detection in animal experiments, which should be further explored in future studies to confirm the present findings.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant nos. 81672489, 81872053 and 81902526) and the Postgraduate Research & Practice Innovation Program of Jiangsu Province (grant no. KYCX20_2460).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YW wrote the manuscript and contributed to data analysis. YZ, BS and XZ performed the experiments. RY contributed to the study design. XZ contributed to the study design, reviewed and edited the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Gould J: Breaking down the epidemiology of brain cancer. Nature. 561:S40–S41. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Meyer MA: Malignant gliomas in adults. N Engl J Med. 359:18502008. View Article : Google Scholar : PubMed/NCBI

3 

Osswald M and Morais-de-Sá E: Dealing with apical-basal polarity and intercellular junctions: A multidimensional challenge for epithelial cell division. Curr Opin Cell Biol. 60:75–83. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Martin-Belmonte F and Perez-Moreno M: Epithelial cell polarity, stem cells and cancer. Nat Rev Cancer. 12:23–38. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Hariharan IK and Bilder D: Regulation of imaginal disc growth by tumor-suppressor genes in Drosophila. Annu Rev Genet. 40:335–361. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Kashyap A, Zimmerman T, Ergül N, Bosserhoff A, Hartman U, Alla V, Bataille F, Galle PR, Strand S and Strand D: The human Lgl polarity gene, Hugl-2, induces MET and suppresses Snail tumorigenesis. Oncogene. 32:1396–1407. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Zimmermann T, Kashyap A, Hartmann U, Otto G, Galle PR, Strand S and Strand D: Cloning and characterization of the promoter of Hugl-2, the human homologue of Drosophila lethal giant larvae (lgl) polarity gene. Biochem Biophys Res Commun. 366:1067–1073. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Grifoni D, Garoia F, Bellosta P, Parisi F, De Biase D, Collina G, Strand D, Cavicchi S and Pession A: aPKCzeta cortical loading is associated with Lgl cytoplasmic release and tumor growth in Drosophila and human epithelia. Oncogene. 26:5960–5965. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Ohshiro T, Yagami T, Zhang C and Matsuzaki F: Role of cortical tumour-suppressor proteins in asymmetric division of Drosophila neuroblast. Nature. 408:593–596. 2000. View Article : Google Scholar : PubMed/NCBI

10 

Vasioukhin V: Lethal giant puzzle of Lgl. Dev Neurosci. 28:13–24. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Lu X, Feng X, Man X, Yang G, Tang L, Du D, Zhang F, Yuan H, Huang Q, Zhang Z, et al: Aberrant splicing of Hugl-1 is associated with hepatocellular carcinoma progression. Clin Cancer Res. 15:3287–3296. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Kuphal S, Wallner S, Schimanski CC, Bataille F, Hofer P, Strand S, Strand D and Bosserhoff AK: Expression of Hugl-1 is strongly reduced in malignant melanoma. Oncogene. 25:103–110. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Schimanski CC, Schmitz G, Kashyap A, Bosserhoff AK, Bataille F, Schäfer SC, Lehr HA, Berger MR, Galle PR, Strand S and Strand D: Reduced expression of Hugl-1, the human homologue of Drosophila tumour suppressor gene lgl, contributes to progression of colorectal cancer. Oncogene. 24:3100–3109. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Tsuruga T, Nakagawa S, Watanabe M, Takizawa S, Matsumoto Y, Nagasaka K, Sone K, Hiraike H, Miyamoto Y, Hiraike O, et al: Loss of Hugl-1 expression associates with lymph node metastasis in endometrial cancer. Oncol Res. 16:431–435. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Matsuzaki T, Takekoshi S, Toriumi K, Kitatani K, Nitou M, Imamura N, Ogura G, Masuda R, Nakamura N and Iwazaki M: Reduced expression of Hugl 1 contributes to the progression of lung squamous cell carcinoma. Tokai J Exp Clin Med. 40:169–177. 2015.PubMed/NCBI

16 

Biesterfeld S, Kauhausen A, Kost C, Gockel I, Schimanski CC and Galle PR: Preservation of HUGL-1 expression as a favourable prognostic factor in pancreatic carcinoma. Anticancer Res. 32:3153–3159. 2012.PubMed/NCBI

17 

Liu X, Lu D, Ma P, Liu H, Cao Y, Sang B, Zhu X, Shi Q, Hu J, Yu R and Zhou X: Hugl-1 inhibits glioma cell growth in intracranial model. J Neurooncol. 125:113–121. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Massimi P, Narayan N, Thomas M, Gammoh N, Strand S, Strand D and Banks L: Regulation of the hDlg/hScrib/Hugl-1 tumour suppressor complex. Exp Cell Res. 314:3306–3317. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Zhong XL and Rescorla FJ: Cell surface adhesion molecules and adhesion-initiated signaling: Understanding of anoikis resistance mechanisms and therapeutic opportunities. Cell Signal. 24:393–401. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Fife CM, McCarroll JA and Kavallaris M: Movers and shakers: Cell cytoskeleton in cancer metastasis. Brit J Pharmacol. 171:5507–5523. 2014. View Article : Google Scholar

21 

Gloushankova NA, Rubtsova SN and Zhitnyak IY: Cadherin-mediated cell-cell interactions in normal and cancer cells. Tissue Barriers. 5:e13569002017. View Article : Google Scholar : PubMed/NCBI

22 

Pal M, Bhattacharya S, Kalyan G and Hazra S: Cadherin profiling for therapeutic interventions in Epithelial Mesenchymal Transition (EMT) and tumorigenesis. Exp Cell Res. 368:137–146. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Nair KS, Naidoo R and Chetty R: Expression of cell adhesion molecules in oesophageal carcinoma and its prognostic value. J Clin Pathol. 58:343–351. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Huang C, Kratzer MC, Wedlich D and Kashef J: E-cadherin is required for cranial neural crest migration in Xenopus laevis. Dev Biol. 411:159–171. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Bremmer F, Schallenberg S, Jarry H, Küffer S, Kaulfuss S, Burfeind P, Strauß A, Thelen P, Radzun HJ, Ströbel P, et al: Role of N-cadherin in proliferation, migration, and invasion of germ cell tumours. Oncotarget. 6:33426–33437. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Mrozik KM, Blaschuk OW, Cheong CM, Zannettino ACW and Vandyke K: N-cadherin in cancer metastasis, its emerging role in haematological malignancies and potential as a therapeutic target in cancer. BMC Cancer. 18:9392018. View Article : Google Scholar : PubMed/NCBI

27 

Camand E, Peglion F, Osmani N, Sanson M and Etienne-Manneville S: N-cadherin expression level modulates integrin-mediated polarity and strongly impacts on the speed and directionality of glial cell migration. J Cell Sci. 125:844–857. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Gheldof A and Berx G: Cadherins and epithelial-to-mesenchymal transition. Prog Mol Biol Transl Sci. 116:317–336. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Kourtidis A, Lu R, Pence LJ and Anastasiadis PZ: A central role for cadherin signaling in cancer. Exp Cell Res. 358:78–85. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Kashima T, Kawaguchi J, Takeshita S, Kuroda M, Takanashi M, Horiuchi H, Imamura T, Ishikawa Y, Ishida T, Mori S, et al: Anomalous cadherin expression in osteosarcoma. Possible relationships to metastasis and morphogenesis. Am J Pathol. 155:1549–1555. 1999. View Article : Google Scholar : PubMed/NCBI

31 

Han ZX, Wang XX, Zhang SN, Wu JX, Qian HY, Wen YY, Tian H, Pei DS and Zheng JN: Downregulation of PAK5 inhibits glioma cell migration and invasion potentially through the PAK5-Egr1-MMP2 signaling pathway. Brain Tumor Pathol. 31:234–241. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Li F, Jin D, Guan L, Zhang CC, Wu T, Wang YJ and Gao DS: CEP55 promoted the migration, invasion and neuroshpere formation of the glioma cell line U251. Neurosci Lett. 705:80–86. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Wang T, Liu Y, Xu XH, Deng CY, Wu KY, Zhu J, Fu XQ, He M and Luo ZG: Lgl1 activation of rab10 promotes axonal membrane trafficking underlying neuronal polarization. Dev Cell. 21:431–444. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Wicki A, Lehembre F, Wick N, Hantusch B, Kerjaschki D and Christofori G: Tumor invasion in the absence of epithelial-mesenchymal transition: Podoplanin-mediated remodeling of the actin cytoskeleton. Cancer Cell. 9:261–272. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Li N, Chen G, Liu J, Xia Y, Chen H, Tang H, Zhang F and Gu N: Effect of surface topography and bioactive properties on early adhesion and growth behavior of mouse preosteoblast MC3T3-E1 cells. ACS Appl Mater Interfaces. 6:17134–17143. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Lewczuk Ł, Pryczynicz A and Guzińska-Ustymowicz K: Cell adhesion molecules in endometrial cancer-A systematic review. Adv Med Sci. 64:423–429. 2019. View Article : Google Scholar : PubMed/NCBI

37 

McKeown SJ, Wallace AS and Anderson RB: Expression and function of cell adhesion molecules during neural crest migration. Dev Biol. 373:244–257. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Zhan T, Rindtorff N and Boutros M: Wnt signaling in cancer. Oncogene. 36:1461–1473. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Taciak B, Pruszynska I, Kiraga L, Bialasek M and Krol M: Wnt signaling pathway in development and cancer. J Physiol Pharmacol. 69:2018.PubMed/NCBI

40 

Li ZH, Zhou Y, Ding YX, Guo QL and Zhao L: Roles of integrin in tumor development and the target inhibitors. Chin J Nat Med. 17:241–251. 2019.PubMed/NCBI

41 

Li JC, Cheng LC and Jiang HY: Cell shape and intercellular adhesion regulate mitotic spindle orientation. Mol Biol Cell. 30:2458–2468. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Mack NA and Georgiou M: The interdependence of the Rho GTPases and apicobasal cell polarity. Small GTPases. 5:102014. View Article : Google Scholar : PubMed/NCBI

43 

Bonastre E, Brambilla E and Sanchez-Cespedes M: Cell adhesion and polarity in squamous cell carcinoma of the lung. J Pathol. 238:606–616. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Waghmare I and Kango-Singh M: Loss of cell adhesion increases tumorigenic potential of polarity deficient scribble mutant cells. PLoS One. 11:e01580812016. View Article : Google Scholar : PubMed/NCBI

45 

Wan S, Meyer AS, Weiler SME, Rupp C, Tóth M, Sticht C, Singer S, Thomann S, Roessler S, Schorpp-Kistner M, et al: Cytoplasmic localization of the cell polarity factor scribble supports liver tumor formation and tumor cell invasiveness. Hepatology. 67:1842–1856. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Bhattacharya S: Cell polarity: A link to epithelial-mesenchymal transition and vascular mimicry. Crit Rev Eukar Gene. 28:101–105. 2018. View Article : Google Scholar

47 

Song J, Peng XL, Ji MY, Ai MH, Zhang JX and Dong WG: Hugl-1 induces apoptosis in esophageal carcinoma cells both in vitro and in vivo. World J Gastroenterol. 19:4127–4136. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Ke LD, Shi YX and Yung WK: VEGF(121), VEGF(165) overexpression enhances tumorigenicity in U251 MG but not in NG-1 glioma cells. Cancer Res. 62:1854–1861. 2002.PubMed/NCBI

49 

Painter KJ, Armstrong NJ and Sherratt JA: The impact of adhesion on cellular invasion processes in cancer and development. J Theor Biol. 264:1057–1067. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Cavallaro U and Christofori G: Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat Rev Cancer. 4:118–132. 2004. View Article : Google Scholar : PubMed/NCBI

51 

Grifoni D, Garoia F, Schimanski CC, Schmitz G, Laurenti E, Galle PR, Pession A, Cavicchi S and Strand D: The human protein Hugl-1 substitutes for Drosophila lethal giant larvae tumour suppressor function in vivo. Oncogene. 23:8688–8694. 2004. View Article : Google Scholar : PubMed/NCBI

52 

Yu FX, Zhao B and Guan KL: Hippo pathway in organ size control, tissue homeostasis, and cancer. Cell. 163:811–828. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Asano K, Duntsch CD, Zhou Q, Weimar JD, Bordelon D, Robertson JH and Pourmotabbed T: Correlation of N-cadherin expression in high grade gliomas with tissue invasion. J Neurooncol. 70:3–15. 2004. View Article : Google Scholar : PubMed/NCBI

54 

Jossin Y, Lee M, Klezovitch O, Kon E, Cossard A, Lien WH, Fernandez TE, Cooper JA and Vasioukhin V: Llgl1 connects cell polarity with cell-cell adhesion in embryonic neural stem cells. Dev Cell. 41:481–495.e5. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2021
Volume 21 Issue 2

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
Wang Y, Zhang Y, Sang B, Zhu X, Yu R and Zhou X: Human giant larvae‑1 promotes migration and invasion of malignant glioma cells by regulating N‑cadherin. Oncol Lett 21: 167, 2021
APA
Wang, Y., Zhang, Y., Sang, B., Zhu, X., Yu, R., & Zhou, X. (2021). Human giant larvae‑1 promotes migration and invasion of malignant glioma cells by regulating N‑cadherin. Oncology Letters, 21, 167. https://doi.org/10.3892/ol.2021.12428
MLA
Wang, Y., Zhang, Y., Sang, B., Zhu, X., Yu, R., Zhou, X."Human giant larvae‑1 promotes migration and invasion of malignant glioma cells by regulating N‑cadherin". Oncology Letters 21.2 (2021): 167.
Chicago
Wang, Y., Zhang, Y., Sang, B., Zhu, X., Yu, R., Zhou, X."Human giant larvae‑1 promotes migration and invasion of malignant glioma cells by regulating N‑cadherin". Oncology Letters 21, no. 2 (2021): 167. https://doi.org/10.3892/ol.2021.12428