Journal Home > Volume 9 , Issue 4

Glioma is the most common primary malignant tumor of the adult central nervous system. It has high morbidity and poor survival. Myelin protein zero-like protein 1 (MPZL1) is a cell surface glycoprotein that activates numerous adhesion-dependent signaling pathways. MPZL1 plays important roles in human cancers that include metastatic process; however, it is not clear if MPZL1 plays a role in human glioma. Therefore, this study aimed to determine if silencing MPZL1 impacted the cell proliferative features of human glioma cells. First, MPZL1 expression was investigated in human glioma samples and tumor cell lines. Then the effects of small interfering RNA (siRNA)-targeting MPZL1 were analyzed on proliferation, colony formation, cell cycle progression, and invasion of human glioma cells. The results from this study demonstrated that MPZL1 was highly expressed in human glioma tissues and glioma cell lines. In addition, knockdown of MPZL1 significantly inhibited cell proliferation, colony formation, and invasiveness of glioma cells, and effectively induced cell cycle arrest at the G1 phase. Western blotting analysis indicated that silencing MPZL1 expression downregulated the expression of matrix metalloproteinase-2 (MMP-2), WNT1, caspase-3, cyclin A1, epidermal growth factor receptor (EGFR), and signal transducer and activator of transcription 3 (STAT3), and upregulated p53. The results from this study suggest that MPZL1 might be a marker for tumors and could be a potential therapeutic target for human glioma.


menu
Abstract
Full text
Outline
About this article

Silencing myelin protein zero-like 1 expression suppresses cell proliferation and invasiveness of human glioma cells by inhibiting multiple cancer-associated signal pathways

Show Author's information Simiao Zhang1,§Sandian Zhang2,§Hongzhen Wang3,§Xuege Huang4Jinzhi Wang4Jingna Li1Dan Cheng1Hongwei Wang5Daru Lu6,7( )Yunliang Wang1,8( )
Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, Henan, China
Department of Radiation Oncology, Li Huili Hospital of Ningbo Medical Center, Ningbo 315040, Zhejiang, China
Department of Orthopaedics, the First People's Hospital of Kunshan City, Kunshan 215300, Jiangsu, China
Department of Cell Biology, School of Medicine, Soochow University, Suzhou 215123, Jiangsu, China
Shanghai Realgen Biotech Inc., Pudong New District, Shanghai 203215, China
State Key Laboratory of Genetic Engineering and MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200032, China
NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning, Science and Technology Research Institute, Chongqing 400020, China
Department of Neurology, The 960 Central Hospital of PLA, Zibo 255300, Shangdong, China

§ These authors contributed equally to this work.

Abstract

Glioma is the most common primary malignant tumor of the adult central nervous system. It has high morbidity and poor survival. Myelin protein zero-like protein 1 (MPZL1) is a cell surface glycoprotein that activates numerous adhesion-dependent signaling pathways. MPZL1 plays important roles in human cancers that include metastatic process; however, it is not clear if MPZL1 plays a role in human glioma. Therefore, this study aimed to determine if silencing MPZL1 impacted the cell proliferative features of human glioma cells. First, MPZL1 expression was investigated in human glioma samples and tumor cell lines. Then the effects of small interfering RNA (siRNA)-targeting MPZL1 were analyzed on proliferation, colony formation, cell cycle progression, and invasion of human glioma cells. The results from this study demonstrated that MPZL1 was highly expressed in human glioma tissues and glioma cell lines. In addition, knockdown of MPZL1 significantly inhibited cell proliferation, colony formation, and invasiveness of glioma cells, and effectively induced cell cycle arrest at the G1 phase. Western blotting analysis indicated that silencing MPZL1 expression downregulated the expression of matrix metalloproteinase-2 (MMP-2), WNT1, caspase-3, cyclin A1, epidermal growth factor receptor (EGFR), and signal transducer and activator of transcription 3 (STAT3), and upregulated p53. The results from this study suggest that MPZL1 might be a marker for tumors and could be a potential therapeutic target for human glioma.

Keywords: brain tumor, glioma, RNA interference, MPZL1

References(54)

[1]
Jansen M, Yip S, Louis DN. Molecular pathology in adult gliomas: diagnostic, prognostic, and predictive markers. Lancet Neurol 2010, 9(7): 717-726.
[2]
Sun Z, Zhang B, Wang C, et al. Forkhead box P3 regulates ARHGAP15 expression and affects migration of glioma cells through the Rac1 signaling pathway. Cancer Sci 2017, 108(1): 61-72.
[3]
Cuddapah VA, Turner KL, Seifert S, et al. Bradykinin- induced chemotaxis of human gliomas requires the activation of KCa3.1 and ClC-3. J Neurosci 2013, 33(4): 1427-1440.
[4]
Habela CW, Ernest NJ, Swindall AF, et al. Chloride accumulation drives volume dynamics underlying cell proliferation and migration. J Neurophysiol 2009, 101(2): 750-757.
[5]
Cuddapah VA, Robel S, Watkins S, et al. A neurocentric perspective on glioma invasion. Nature Rev Neurosci 2014, 15(7): 455-465.
[6]
Joester A, Faissner A. The structure and function of tenascins in the nervous system. Matrix Biol 2001, 20(1): 13-22.
[7]
Deng Z, Yu H, Wang N, et al. Impact of preoperative Karnofsky Performance Scale (KPS) and American Society of Anesthesiologists (ASA) scores on perioperative complications in patients with recurrent glioma undergoing repeated operation. J Neurorestoratol 2019, 7(3): 143-152.
[8]
Huang HY, Sharma HS, Chen L, et al. 2018 yearbook of neurorestoratology. J Neurorestoratol 2019, 7(1): 11-20.
[9]
Huang HY, Chen L, Mao GS, et al. The 2019 yearbook of neurorestoratology. J Neurorestoratol 2020, 8(1): 1-11.
[10]
Zhao ZJ, Zhao R. Purification and cloning of PZR, a binding protein and putative physiological substrate of tyrosine phosphatase SHP-2. J Biol Chem 1998, 273(45): 29367-29372.
[11]
Kusano K, Thomas TN, Fujiwara K. Phosphorylation and localization of protein-zero related (PZR) in cultured endothelial cells. Endothelium 2008, 15(3): 127-136.
[12]
Jia DS, Jing Y, Zhang ZF, et al. Amplification of MPZL1/PZR promotes tumor cell migration through Src-mediated phosphorylation of cortactin in hepatocellular carcinoma. Cell Res 2014, 24(2): 204-217.
[13]
Chen DN, Cao L, Wang XJ. MPZL1 promotes tumor cell proliferation and migration via activation of Src kinase in ovarian cancer. Oncol Rep 2019, 42(2): 679-687.
[14]
Beigbeder A, Chartier FJM, Bisson N. MPZL1 forms a signalling complex with GRB2 adaptor and PTPN11 phosphatase in HER2-positive breast cancer cells. Sci Rep 2017, 7(1): 11514.
[15]
Zannettino ACW, Roubelakis M, Welldon KJ, et al. Novel mesenchymal and haematopoietic cell isoforms of the SHP-2 docking receptor, PZR: identification, molecular cloning and effects on cell migration. Biochem J 2003, 370(2): 537-549.
[16]
Lund CV, Nguyen MT, Owens GC, et al. Reduced glioma infiltration in Src-deficient mice. J Neurooncol 2006, 78(1): 19-29.
[17]
Summy JM, Gallick GE. Treatment for advanced tumors: Src reclaims center stage. Clin Cancer Res 2006, 12(5): 1398-1401.
[18]
Stedt H, Alasaarela L, Samaranayake H, et al. Specific inhibition of Src kinase impairs malignant glioma growth in vitro and in vivo. Mol Ther Nucleic Acids 2012, 1(5): e19.
[19]
Sherry MM, Reeves A, Wu JK, et al. STAT3 is required for proliferation and maintenance of multipotency in glioblastoma stem cells. Stem Cells 2009, 27(10): 2383-2392.
[20]
Priester M, Copanaki E, Vafaizadeh V, et al. STAT3 silencing inhibits glioma single cell infiltration and tumor growth. Neuro Oncol 2013, 15(7): 840-852.
[21]
Zielske SP, Stevenson M. Importin 7 may be dispensable for human immunodeficiency virus type 1 and Simian immunodeficiency virus infection of primary macrophages. J Virol 2005, 79(17): 11541-11546.
[22]
Huse JT, Holland EC. Targeting brain cancer: advances in the molecular pathology of malignant glioma and medulloblastoma. Nat Rev Cancer 2010, 10(5): 319-331.
[23]
Taniguchi K, Karin M. IL-6 and related cytokines as the critical lynchpins between inflammation and cancer. Semin Immunol 2014, 26(1): 54-74.
[24]
Labbé DP, Hardy S, Tremblay ML. Protein tyrosine phosphatases in cancer: friends and foes! Prog Mol Biol Transl Sci 2012, 106: 253-306.
[25]
Huang W-Q, Lin Q, Zhuang X, et al. Structure, function, and pathogenesis of SHP2 in developmental disorders and tumorigenesis. Curr Cancer Drug Targets 2014, 14(6): 567-588.
[26]
Grossmann KS, Rosário M, Birchmeier C, et al. The tyrosine phosphatase Shp2 in development and cancer. Adv Cancer Res 2010, 106: 53-89.
[27]
Reya T, Clevers H. Wnt signalling in stem cells and cancer. Nature 2005, 434(7035): 843-850.
[28]
Clevers H. Wnt/β-catenin signaling in development and disease. Cell 2006, 127(3): 469-480.
[29]
Chen G, Shukeir N, Potti A, et al. Up-regulation of Wnt-1 and β-catenin production in patients with advanced metastatic prostate carcinoma: Potential pathogenetic and prognostic implications. Cancer 2004, 101(6): 1345-1356.
[30]
Zhang WM, Muzio L, Rubini C, et al. Effect of WNT-1 on β-catenin expression and its relation to Ki-67 and tumor differentiation in oral squamous cell carcinoma. Oncol Rep 2005: 1095-1099.
[31]
Zhang K, Zhang J, Han L, et al. Wnt/beta-catenin signaling in glioma. J Neuroimmune Pharmacol 2012, 7(4): 740-749.
[32]
Wu T, Wang LN, Tang DR, et al. SOST silencing promotes proliferation and invasion and reduces apoptosis of retinoblastoma cells by activating Wnt/β-catenin signaling pathway. Gene Ther 2017, 24(7): 399-407.
[33]
Wolfenson H, Lavelin I, Geiger B. Dynamic regulation of the structure and functions of integrin adhesions. Dev Cell 2013, 24(5): 447-458.
[34]
Demuth T, Berens ME. Molecular mechanisms of glioma cell migration and invasion. J Neurooncol 2004, 70(2): 217-228.
[35]
Zhou M, Qin S, Chu Y, et al. Immunolocalization of MMP-2 and MMP-9 in human rheumatoid synovium. Int J Clin Exp Pathol 2014, 7(6): 3048-3056.
[36]
Komatsu K, Nakanishi Y, Nemoto N, et al. Expression and quantitative analysis of matrix metalloproteinase-2 and -9 in human gliomas. Brain Tumor Pathol 2004, 21(3): 105-112.
[37]
Wang M, Wang T, Liu S, et al. The expression of matrix metalloproteinase-2 and -9 in human gliomas of different pathological grades. Brain Tumor Pathol 2003, 20(2): 65-72.
[38]
Huang R, Wallqvist A, Covell DG. Anticancer metal compounds in NCI’s tumor-screening database: putative mode of action. Biochem Pharmacol 2005, 69(7): 1009-1039.
[39]
Liu DZ, Tian Z, Yan ZH, et al. Design, synthesis and evaluation of 1,2-benzisothiazol-3-one derivatives as potent caspase-3 inhibitors. Biorg Med Chem 2013, 21(11): 2960-2967.
[40]
Strasser A, Cory S, Adams JM. Deciphering the rules of programmed cell death to improve therapy of cancer and other diseases. EMBO J 2011, 30(18): 3667-3683.
[41]
Bruyère C, Meijer L. Targeting cyclin-dependent kinases in anti-neoplastic therapy. Curr Opin Cell Biol 2013, 25(6): 772-779.
[42]
Vivanco I, Robins HI, Rohle D, et al. Differential sensitivity of glioma-versus lung cancer-specific EGFR mutations to EGFR kinase inhibitors. Cancer Discov 2012, 2(5): 458-471.
[43]
Guha D, Saha T, Bose S, et al. Integrin-EGFR interaction regulates anoikis resistance in colon cancer cells. Apoptosis 2019, 24(11/12): 958-971.
[44]
Sullivan KD, Gallant-Behm CL, Henry RE, et al. The p53 circuit board. Biochim Biophys Acta 2012, 1825(2): 229-244.
[45]
Zhang YX, Na RH, Wang XL. LncRNA WT1-AS up-regulates p53 to inhibit the proliferation of cervical squamous carcinoma cells. BMC Cancer 2019, 19(1): 1052.
[46]
Sabapathy K, Lane DP. Understanding p53 functions through p53 antibodies. J Mol Cell Biol 2019, 11(4): 317-329.
[47]
Silvennoinen O, Schindler C, Schlessinger J, et al. Ras-independent growth factor signaling by transcription factor tyrosine phosphorylation. Science 1993, 261(5129): 1736-1739.
[48]
Zhong Z, Wen Z, Darnell JE Jr. Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science 1994, 264(5155): 95-98.
[49]
Ruff-Jamison S, Zhong Z, Wen Z, et al. Epidermal growth factor and lipopolysaccharide activate Stat3 transcription factor in mouse liver. J Biol Chem 1994, 269(35): 21933-21935.
[50]
Wojcik EJ, Sharifpoor S, Miller NA, et al. A novel activating function of c-Src and Stat3 on HGF transcription in mammary carcinoma cells. Oncogene 2006, 25(19): 2773-2784.
[51]
Xie TX, Wei DY, Liu MG, et al. Stat3 activation regulates the expression of matrix metalloproteinase-2 and tumor invasion and metastasis. Oncogene 2004, 23(20): 3550-3560.
[52]
Mandal T, Bhowmik A, Chatterjee A, et al. Reduced phosphorylation of Stat3 at Ser-727 mediated by casein kinase 2—protein phosphatase 2A enhances Stat3 Tyr-705 induced tumorigenic potential of glioma cells. Cell Signal 2014, 26(8): 1725-1734.
[53]
Faruqi TR, Gomez D, Bustelo XR, et al. Rac1 mediates STAT3 activation by autocrine IL-6. PNAS 2001, 98(16): 9014-9019.
[54]
Senft C, Priester M, Polacin M, et al. Inhibition of the JAK-2/STAT3 signaling pathway impedes the migratory and invasive potential of human glioblastoma cells. J Neurooncol 2011, 101(3): 393-403.
Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 27 June 2021
Revised: 29 July 2021
Accepted: 16 August 2021
Published: 04 January 2022
Issue date: December 2021

Copyright

© The authors 2021

Acknowledgements

The authors thank Dr. Wenxiang Wei from Soochow University, Suzhou, China for his valuable advices and technical supports.

Rights and permissions

This article is published with open access at http://jnr.tsinghuajournals.com

Return