Behaviors of Glioblastoma Cells in in Vitro Microenvironments

Wenwen Diao, Xuezhi Tong, Cheng Yang, Fengrong Zhang, Chun Bao, Hao Chen, Liyu Liu, Ming Li, Fangfu Ye, Qihui Fan, Jiangfei Wang, Zhong-Can Ou-Yang, Wenwen Diao, Xuezhi Tong, Cheng Yang, Fengrong Zhang, Chun Bao, Hao Chen, Liyu Liu, Ming Li, Fangfu Ye, Qihui Fan, Jiangfei Wang, Zhong-Can Ou-Yang

Abstract

Glioblastoma (GBM) is the most malignant and highly aggressive brain tumor. In this study, four types of typical GBM cell lines (LN229, SNB19, U87, U251) were cultured in a microfabricated 3-D model to study their in vitro behaviors. The 3-D in vitro model provides hollow micro-chamber arrays containing a natural collagen interface and thus allows the GBM cells to grow in the 3-D chambers. The GBM cells in this model showed specific properties on the aspects of cell morphology, proliferation, migration, and invasion, some of which were rarely observed before. Furthermore, how the cells invaded into the surrounding ECM and the corresponding specific invasion patterns were observed in details, implying that the four types of cells have different features during their development in cancer. This complex in vitro model, if applied to patient derived cells, possesses the potential of becoming a clinically relevant predictive model.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
(A) 3D in vitro model with micro-chambers in collagen; (B) enlarged image showing one micro-chamber; (C) top view of the chip, captured in the bright field and processed with ImageJ, where the scale bar is 1000 μm. The size of the microchambers is 200 × 200 μm2, with the distance between nearest neighbor pairs being 400 μm.
Figure 2
Figure 2
Representative fluorescent images showing the morphology of the four types of GBM cells in micro-chambers within collagen during the four culture days.
Figure 3
Figure 3
Cell proliferation rate of LN229 (blue), SNB19 (purple), U87 (light pink), and U251 (dark pink) indicated by (a) normalized integrated intensity and (b) normalized cell area. Data are presented as mean ± SD of three independent experiments.
Figure 4
Figure 4
Cell migration speed, directionality ratio, and MSD of four types of GBM cells: (a) the average cell migration speed (N = 200 for each test); (b) directionality ratio (N = 20 for each test); (c) mean square displacement (MSD), plotted as function of time interval (N = 20 for each test). Data are presented as mean ± SD of three independent experiments.
Figure 5
Figure 5
(a) Average and (b) maximum protrusion length of four types of GBM cells; (c) normalized cell number (conjectured from normalized integrated fluorescent intensity) of SNB19 cells inside individual micro-chambers and those invading into the surrounding collagen gel for four consecutive days; (d) normalized cell number (integrated intensity) of U87 cells inside individual micro-chambers and those in the surrounding gel. Data are presented as mean ± SD of three independent experiments.
Figure 6
Figure 6
Confocal images showing representative cell filopodia protrusions (green fluorescent) in a single micro-chamber, where the top and bottom rows are, respectively, top-view and side-view images. (i,j) Locally amplified side-view images of (d) and (f). The collagen fibers (blue) were observed in reflection mode.

References

    1. Ostrom QT, et al. CBTRUS Statistical Report: Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2006-2010. Neuro-Oncology. 2013;15:1–56. doi: 10.1093/neuonc/not151.
    1. Furnari FB, et al. Malignant astrocytic glioma: genetics, biology, and paths to treatment. Gene Dev. 2007;21:2683–2710. doi: 10.1101/gad.1596707.
    1. Ho, I. A. W. & Shim, W. S. N. Contribution of the Microenvironmental Niche to Glioblastoma Heterogeneity. Biomed Res Int, 1155/2017/9634172 (2017).
    1. Xiao W, Sohrabi A, Seidlits SK. Integrating the glioblastoma microenvironment into engineered experimental models. Future science OA. 2017;3:FSO189. doi: 10.4155/fsoa-2016-0094.
    1. Gieryng A, Pszczolkowska D, Walentynowicz KA, Rajan WD, Kaminska B. Immune microenvironment of gliomas. Lab Invest. 2017;97:498–518. doi: 10.1038/labinvest.2017.19.
    1. Cuddapah VA, Robel S, Watkins S, Sontheimer H. A neurocentric perspective on glioma invasion. Nat Rev Neurosci. 2014;15:455–465. doi: 10.1038/nrn3765.
    1. Rutka JT. The role of fascin in the migration and invasiveness of malignant glioma cells. Neoplasia. 2008;10:149–159. doi: 10.1593/neo.07909.
    1. Calabrese C, et al. A perivascular niche for brain tumor stem cells. Cancer Cell. 2007;11:69–82. doi: 10.1016/j.ccr.2006.11.020.
    1. Zamecnik J. The extracellular space and matrix of gliomas. Acta Neuropathol. 2005;110:435–442. doi: 10.1007/s00401-005-1078-5.
    1. Cukierman E, Pankov R, Yamada KM. Cell interactions with three-dimensional matrices. Curr Opin Cell Biol. 2002;14:633–639. doi: 10.1016/S0955-0674(02)00364-2.
    1. Mammoto T, et al. Role of Collagen Matrix in Tumor Angiogenesis and Glioblastoma Multiforme Progression. Am J Pathol. 2013;183:1293–1305. doi: 10.1016/j.ajpath.2013.06.026.
    1. Hegedus B, Marga F, Jakab K, Sharpe-Timms KL, Forgacs G. The interplay of cell-cell and cell-matrix interactions in the invasive properties of brain tumors. Biophys J. 2006;91:2708–2716. doi: 10.1529/biophysj.105.077834.
    1. Liu AP, Chaudhuri O, Parekh SH. New advances in probing cell-extracellular matrix interactions. Integr Biol-Uk. 2017;9:383–405. doi: 10.1039/c6ib00251j.
    1. Xu, H. et al. A dynamic in vivo-like organotypic blood-brain barrier model to probe metastatic brain tumors. Sci Rep-Uk6, 1038/Srep36670 (2016).
    1. Wirtz D, Konstantopoulos K, Searson PC. The physics of cancer: the role of physical interactions and mechanical forces in metastasis. Nat Rev Cancer. 2011;11:512–522. doi: 10.1038/nrc3080.
    1. Stroka KM, et al. Water Permeation Drives Tumor Cell Migration in Confined Microenvironments. Cell. 2014;157:611–623. doi: 10.1016/j.cell.2014.02.052.
    1. Tosoni A, Ermani M, Brandes AA. The pathogenesis and treatment of brain metastases: a comprehensive review. Crit Rev Oncol Hemat. 2004;52:199–215. doi: 10.1016/j.critrevonc.2004.08.006.
    1. Allen, M., Bjerke, M., Edlund, H., Nelander, S. & Westermark, B. Origin of the U87MG glioma cell line: Good news and bad news. Sci Transl Med 8, 1126/scitranslmed.aaf6853 (2016).
    1. Zheng XY, et al. Proteomic analysis for the assessment of different lots of fetal bovine serum as a raw material for cell culture. Part IV. Application of proteomics to the manufacture of biological drugs. Biotechnol Progr. 2006;22:1294–1300. doi: 10.1021/bp060121o.
    1. Wakimoto H, et al. Maintenance of primary tumor phenotype and genotype in glioblastoma stem cells. Neuro-Oncology. 2012;14:132–144. doi: 10.1093/neuonc/nor195.
    1. Kaufman LJ, et al. Glioma expansion in collagen I matrices: Analyzing collagen concentration-dependent growth and motility patterns. Biophys J. 2005;89:635–650. doi: 10.1529/biophysj.105.061994.
    1. Wang C, Tong XM, Yang F. Bioengineered 3D Brain Tumor Model To Elucidate the Effects of Matrix Stiffness on Glioblastoma Cell Behavior Using PEG-Based Hydrogels. Mol Pharmaceut. 2014;11:2115–2125. doi: 10.1021/mp5000828.
    1. Yang YL, Motte S, Kaufman LJ. Pore size variable type I collagen gels and their interaction with glioma cells. Biomaterials. 2010;31:5678–5688. doi: 10.1016/j.biomaterials.2010.03.039.
    1. Lees JG, et al. Role of dynamin in elongated cell migration in a 3D matrix. Bba-Mol Cell Res. 2015;1853:611–618. doi: 10.1016/j.bbamcr.2014.12.008.
    1. Kim HD, et al. Epidermal Growth Factor-induced Enhancement of Glioblastoma Cell Migration in 3D Arises from an Intrinsic Increase in Speed But an Extrinsic Matrix- and Proteolysis-dependent Increase in Persistence. Mol Biol Cell. 2008;19:4249–4259. doi: 10.1091/mbc.E08-05-0501.
    1. Yamada KM, Cukierman E. Modeling tissue morphogenesis and cancer in 3D. Cell. 2007;130:601–610. doi: 10.1016/j.cell.2007.08.006.
    1. Short, A. R. et al. Imaging Cell-Matrix Interactions in 3D Collagen Hydrogel Culture Systems. Macromol Biosci17, 1002/Mabi.201600478 (2017).
    1. Kawamura A, Tamaki N, Kokunai T. Effect of dexamethasone on cell proliferation of neuroepithelial tumor cell lines. Neurol Med-Chir. 1998;38:633–638. doi: 10.2176/Nmc.38.633.
    1. Stepanenko AA, Kavsan VM. Karyotypically distinct U251, U373, and SNB19 glioma cell lines are of the same origin but have different drug treatment sensitivities. Gene. 2014;540:263–265. doi: 10.1016/j.gene.2014.02.053.
    1. Nelson CM, Inman JL, Bissell MJ. Three-dimensional lithographically defined organotypic tissue arrays for quantitative analysis of morphogenesis and neoplastic progression. Nature protocols. 2008;3:674–678. doi: 10.1038/nprot.2008.35.
    1. Manini, I. et al. Role of Microenvironment in Glioma Invasion: What We Learned from In Vitro Models. 19, 10.3390/ijms19010147 (2018).
    1. Cordelieres, F. P. et al. Automated Cell Tracking and Analysis in Phase-Contrast Videos (iTrack4U): Development of Java Software Based on Combined Mean-Shift Processes. Plos One8, 10.1371/journal.pone.0081266 (2013).
    1. Codling EA, Plank MJ, Benhamou S. Random walk models in biology. J R Soc Interface. 2008;5:813–834. doi: 10.1098/rsif.2008.0014.
    1. Feder TJ, BrustMascher I, Slattery JP, Baird B, Webb WW. Constrained diffusion or immobile fraction on cell surfaces: A new interpretation. Biophys J. 1996;70:2767–2773. doi: 10.1016/S0006-3495(96)79846-6.
    1. Friedl P, Gilmour D. Collective cell migration in morphogenesis, regeneration and cancer. Nat Rev Mol Cell Bio. 2009;10:445–457. doi: 10.1038/nrm2720.
    1. Gray RS, Cheung KJ, Ewald AJ. Cellular mechanisms regulating epithelial morphogenesis and cancer invasion. Curr Opin Cell Biol. 2010;22:640–650. doi: 10.1016/j.ceb.2010.08.019.
    1. Zhang, J. D. et al. TRIM45 functions as a tumor suppressor in the brain via its E3 ligase activity by stabilizing p53 through K63-linked ubiquitination. Cell Death Dis8, 10.1038/Cddis.2017.149 (2017).
    1. Duhart, J. M., Brocardo, L., Caldart, C. S., Marpegan, L. & Golombek, D. A. Circadian Alterations in a Murine Model of Hypothalamic Glioma. Front Physiol 8, 10.3389/Fphys.2017.00864 (2017).
    1. Liu, Y. et al. Efficacy of adenovirally expressed soluble TRAIL in human glioma organotypic slice culture and glioma xenografts. Cell Death Dis2, 10.1038/cddis.2010.95 (2011).
    1. Karpel-Massler G, et al. Combined inhibition of Bcl-2/Bcl-xL and Usp9X/Bag3 overcomes apoptotic resistance in glioblastoma in vitro and in vivo. Oncotarget. 2015;6:14507–14521. doi: 10.18632/oncotarget.3993.
    1. Nagaya, T. et al. Near Infrared Photoimmunotherapy Targeting EGFR Positive Triple Negative Breast Cancer: Optimizing the Conjugate-Light Regimen. Plos One10, 10.1371/journal.pone.0136829 (2015).
    1. Yu, K. K. H. et al. High content screening of patient-derived cell lines highlights the potential of non-standard chemotherapeutic agents for the treatment of glioblastoma. Plos One13, 10.1371/journal.pone.0193694 (2018).
    1. Bago JR, et al. Electrospun nanofibrous scaffolds increase the efficacy of stem cell-mediated therapy of surgically resected glioblastoma. Biomaterials. 2016;90:116–125. doi: 10.1016/j.biomaterials.2016.03.008.
    1. Zhang X, et al. Flexible Fabrication of Shape-Controlled Collagen Building Blocks for Self-Assembly of 3D Microtissues. Small. 2015;11:3666–3675. doi: 10.1002/smll.201500556.
    1. Fan QH, et al. A novel 3-D bio-microfluidic system mimicking in vivo heterogeneous tumour microstructures reveals complex tumour-stroma interactions. Lab Chip. 2017;17:2852–2860. doi: 10.1039/c7lc00191f.
    1. Brightman AO, et al. Time-lapse confocal reflection microscopy of collagen fibrillogenesis and extracellular matrix assembly in vitro. Biopolymers. 2000;54:222–234. doi: 10.1002/1097-0282(200009)54:3<222::AID-BIP80>;2-K.
    1. Gorelik R, Gautreau A. Quantitative and unbiased analysis of directional persistence in cell migration. Nat Protoc. 2014;9:1931–1943. doi: 10.1038/nprot.2014.131.

Source: PubMed

3
Subskrybuj