Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Biomater Sci Eng ; 4(2): 368-377, 2018 Feb 12.
Article in English | MEDLINE | ID: mdl-33418731

ABSTRACT

Many tools from the field of tissue engineering can be used to develop novel model systems to study cancer. We have utilized biomimetic synthetic hydrogels, based on poly(ethylene glycol) (PEG) modified with cell adhesive peptides (RGDS) and peptides sensitive to degradation by matrix metalloproteinases 2 and 9 (GGGPQGIWGQGK), as highly controlled 3D substrates for cell culture. We have previously shown that this hydrogel can support growth of tumor cells and also growth and assembly of microvascular networks. Based on this technology, a 3D in vitro tumor angiogenesis model was developed using a dual layer PEG-based hydrogel comprised of vascular cells (endothelial cells, pericytes) and lung adenocarcinoma cells in separate layers to support recapitulation of the vessel recruitment process as it occurs in vivo. This model was previously used to study highly metastatic murine 344SQ cells and in this paper was used to investigate 2 additional types of lung adenocarcinoma cells: nonmetastatic murine 393P cells and somewhat metastatic human A549 cells. All three cell types readily formed spheroid structures in the 3D hydrogels. When cultured in the dual layer format, where tumor cell spheroids were adjacent to a hydrogel layer with microvascular tubule networks, all three tumor cell types recruited vascular cells into the cancer cell layer. Interactions between vessels invading the cancer layer and the cancer cell structures was nearly twice as high for the highly metastatic 344SQ cells as for the other two cell types. Secretion of angiogenic growth factors by the tumor cells was evaluated. 344SQ cells produced the greatest amount of VEGF and FGFb, which probably accounts for the greater degree of vessel recruitment observed. Upon interaction with vessel structures, the 344SQ spheroids underwent a dramatic change in morphology, increasing in size and adopting highly irregular shapes, suggestive of invasive phenotype. This behavior was observed to a much lesser degree for A549 cells and 393P cells.

2.
Sci Rep ; 6: 32726, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27596933

ABSTRACT

Tumor angiogenesis is critical to tumor growth and metastasis, yet much is unknown about the role vascular cells play in the tumor microenvironment. In vitro models that mimic in vivo tumor neovascularization facilitate exploration of this role. Here we investigated lung adenocarcinoma cancer cells (344SQ) and endothelial and pericyte vascular cells encapsulated in cell-adhesive, proteolytically-degradable poly(ethylene) glycol-based hydrogels. 344SQ in hydrogels formed spheroids and secreted proangiogenic growth factors that significantly increased with exposure to transforming growth factor beta 1 (TGF-ß1), a potent tumor progression-promoting factor. Vascular cells in hydrogels formed tubule networks with localized activated TGF-ß1. To study cancer cell-vascular cell interactions, we engineered a 2-layer hydrogel with 344SQ and vascular cell layers. Large, invasive 344SQ clusters (area > 5,000 µm(2), circularity < 0.25) developed at the interface between the layers, and were not evident further from the interface or in control hydrogels without vascular cells. A modified model with spatially restricted 344SQ and vascular cell layers confirmed that observed cluster morphological changes required close proximity to vascular cells. Additionally, TGF-ß1 inhibition blocked endothelial cell-driven 344SQ migration. Our findings suggest vascular cells contribute to tumor progression and establish this culture system as a platform for studying tumor vascularization.


Subject(s)
Blood Cells/drug effects , Hydrogels/pharmacology , Lung Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Polyethylene Glycols/chemistry , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Blood Cells/pathology , Cell Communication/drug effects , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Disease Progression , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells , Humans , Lung/drug effects , Lung/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/physiopathology , Neovascularization, Pathologic/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Microenvironment/drug effects
3.
Adv Drug Deliv Rev ; 97: 250-9, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26571106

ABSTRACT

Tumor angiogenesis is a hallmark of cancer that has been identified as a critical component of cancer progression, facilitating rapid tumor growth and metastasis. Anti-angiogenic therapies have exhibited only modest clinical success, highlighting a need for better models that can be used to gain a more thorough understanding of tumor angiogenesis and screen potential therapeutics more accurately. This review explores how recent progress in in vitro cancer and vascular models individually can be applied to the development of in vitro tumor angiogenesis models. Current in vitro tumor angiogenesis models are also discussed, with a focus on aspects of the process that have been successfully recapitulated and opportunities for applying new technologies to expand model complexity to better represent the tumor microenvironment. Continued advances in vascularized tumor models will provide tools to identify novel therapeutic targets and validate their therapeutic benefit.


Subject(s)
Neoplasms/pathology , Neovascularization, Pathologic , Animals , Cell Culture Techniques , Humans , Organ Culture Techniques , Tissue Engineering
4.
Cancer Res ; 72(22): 6013-23, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22952217

ABSTRACT

Better understanding of the biophysical and biochemical cues of the tumor extracellular matrix environment that influence metastasis may have important implications for new cancer therapeutics. Initial exploration into this question has used naturally derived protein matrices that suffer from variability, poor control over matrix biochemistry, and inability to modify the matrix biochemistry and mechanics. Here, we report the use of a synthetic polymer-based scaffold composed primarily of poly(ethylene glycol), or PEG, modified with bioactive peptides to study murine models of lung adenocarcinoma. In this study, we focus on matrix-derived influences on epithelial morphogenesis of a metastatic cell line (344SQ) that harbors mutations in Kras and p53 (trp53) and is prone to a microRNA-200 (miR-200)-dependent epithelial-mesenchymal transition (EMT) and metastasis. The modified PEG hydrogels feature biospecific cell adhesion and cell-mediated proteolytic degradation with independently adjustable matrix stiffness. 344SQ encapsulated in bioactive peptide-modified, matrix metalloproteinase-degradable PEG hydrogels formed lumenized epithelial spheres comparable to that seen with three-dimensional culture in Matrigel. Altering both matrix stiffness and the concentration of cell-adhesive ligand significantly influenced epithelial morphogenesis as manifest by differences in the extent of lumenization, in patterns of intrasphere apoptosis and proliferation, and in expression of epithelial polarity markers. Regardless of matrix composition, exposure to TGF-ß induced a loss of epithelial morphologic features, shift in expression of EMT marker genes, and decrease in mir-200 levels consistent with EMT. Our findings help illuminate matrix-derived cues that influence epithelial morphogenesis and highlight the potential utility that this synthetic matrix-mimetic tool has for cancer biology.


Subject(s)
Adenocarcinoma/pathology , Cell Culture Techniques/methods , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Hydrogels/chemistry , Lung Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , Disease Models, Animal , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Genes, p53 , Genes, ras , Lung Neoplasms/genetics , Mice , Mutation , Oligopeptides/chemistry , Polyethylene Glycols/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...