Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
BMC Cancer ; 14: 825, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25385001

ABSTRACT

BACKGROUND: Aggressive metastatic breast cancer cells seemingly evade surgical resection and current therapies, leading to colonization in distant organs and tissues and poor patient prognosis. Therefore, high-throughput in vitro tools allowing rapid, accurate, and novel anti-metastatic drug screening are grossly overdue. Conversely, aligned nanofiber constitutes a prominent component of the late-stage breast tumor margin extracellular matrix. This parallel suggests that the use of a synthetic ECM in the form of a nanoscale model could provide a convenient means of testing the migration potentials of cancer cells to achieve a long-term goal of providing clinicians an in vitro platform technology to test the efficacy of novel experimental anti-metastatic compounds. METHODS: Electrospinning produces highly aligned, cell-adhesive nanofiber matrices by applying a strong electric field to a polymer-containing solution. The resulting fibrous microstructure and morphology closely resembles in vivo tumor microenvironments suggesting their use in analysis of migratory potentials of metastatic cancer cells. Additionally, a novel interface with a gel-based delivery system creates CXCL12 chemotactic gradients to enhance CXCR4-expressing cell migration. RESULTS: Cellular dispersions of MCF-10A normal mammary epithelial cells or human breast cancer cells (MCF-7 and MDA-MB-231) seeded on randomly-oriented nanofiber exhibited no significant differences in total or net distance traveled as a result of the underlying topography. Cells traveled ~2-5 fold greater distances on aligned fiber. Highly-sensitive MDA-MB-231 cells displayed an 82% increase in net distance traversed in the presence of a CXCL12 gradient. In contrast, MCF-7 cells exhibited only 31% increase and MCF-10A cells showed no statistical difference versus control or vehicle conditions. MCF-10A cells displayed little sensitivity to CXCL12 gradients, while MCF-7 cells displayed early sensitivity when CXCL12 concentrations were higher. MDA-MB-231 cells displayed low relative expression levels of CXCR4, but high sensitivity resulting in 55-fold increase at late time points due to CXCL12 gradient dissipation. CONCLUSIONS: This model could create clinical impact as an in vitro diagnostic tool for rapid assessment of tumor needle biopsies to confirm metastatic tumors, their invasiveness, and allow high-throughput drug screening providing rapid development of personalized therapies.


Subject(s)
Biomimetic Materials , Breast Neoplasms/pathology , Cell Movement , Nanofibers/ultrastructure , Biomimetic Materials/chemical synthesis , Breast Neoplasms/chemistry , Cell Movement/drug effects , Chemokine CXCL12/pharmacology , Chemotactic Factors/pharmacology , Extracellular Matrix/ultrastructure , Female , High-Throughput Screening Assays , Humans , MCF-7 Cells , Neoplasm Invasiveness , Neoplasm Metastasis , RNA, Messenger/analysis , Receptors, CXCR4/genetics , Tumor Microenvironment
2.
Macromol Biosci ; 17(6)2017 06.
Article in English | MEDLINE | ID: mdl-28221720

ABSTRACT

3D hydrogels better replicate in vivo conditions, and yield different results from 2D substrates. However, imaging interactions between cells and the hydrogel microenvironment is challenging because of light diffraction and poor focal depth. Here, cryosectioning and vibrating microtomy methods and fixation protocols are compared. Collagen I/III hydrogel sections (20-100 µm) are fixed with paraformaldehyde (2%-4%) and structurally evaluated. Cryosectioning damaged hydrogels, and vibrating microtomy (100 µm, 2%) yielded the best preservation of microstructure and cell integrity. These results demonstrate a potential processing method that preserves hydrogel and cell integrity, permitting imaging of cell interactions with the microenvironment.


Subject(s)
Cell Culture Techniques , Collagen Type I/chemistry , Extracellular Matrix/drug effects , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Cell Communication/drug effects , Cell Proliferation/drug effects , Cellular Microenvironment/drug effects , Collagen Type I/therapeutic use , Formaldehyde/chemistry , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/therapeutic use , Polymers/chemistry
3.
J Comp Neurol ; 524(17): 3485-3502, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27418162

ABSTRACT

We sought to determine the contribution of scaffold topography to the migration and morphology of neural stem cells by mimicking anatomical features of scaffolds found in vivo. We mimicked two types of central nervous system scaffolds encountered by neural stem cells during development in vitro by constructing different diameter electrospun polycaprolactone (PCL) fiber mats, a substrate that we have shown to be topographically similar to brain scaffolds. We compared the effects of large fibers (made to mimic blood vessel topography) with those of small-diameter fibers (made to mimic radial glial process topography) on the migration and differentiation of neural stem cells. Neural stem cells showed differential migratory and morphological reactions with laminin in different topographical contexts. We demonstrate, for the first time, that neural stem cell biological responses to laminin are dependent on topographical context. Large-fiber topography without laminin prevented cell migration, which was partially reversed by treatment with rock inhibitor. Cell morphology complexity assayed by fractal dimension was inhibited in nocodazole- and cytochalasin-D-treated neural precursor cells in large-fiber topography, but was not changed in small-fiber topography with these inhibitors. These data indicate that cell morphology has different requirements on cytoskeletal proteins dependent on the topographical environment encountered by the cell. We propose that the physical structure of distinct scaffolds induces unique signaling cascades that regulate migration and morphology in embryonic neural precursor cells. J. Comp. Neurol. 524:3485-3502, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Movement/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Actins/antagonists & inhibitors , Actins/metabolism , Animals , Cell Movement/drug effects , Cells, Cultured , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Female , Humans , Laminin/metabolism , Male , Mice , Microtubules/drug effects , Microtubules/metabolism , Models, Neurological , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neurons/cytology , Neurons/drug effects , Polyesters , Surface Properties , Tissue Scaffolds , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
4.
Biotechnol Prog ; 31(5): 1406-15, 2015.
Article in English | MEDLINE | ID: mdl-26081199

ABSTRACT

Gliomas are highly invasive forms of brain cancer comprising more than 50% of brain tumor cases in adults, and astrocytomas account for ∼60-70% of all gliomas. As a result of multiple factors, including enhanced migratory properties and extracellular matrix remodeling, even with current standards of care, mean survival time for patients is only ∼12 months. Because glioblastoma multiforme (GBM) cells arise from astrocytes, there is great interest in elucidating the interactions of these two cell types in vivo. Previous work performed on two-dimensional assays (i.e., tissue culture plastic and Boyden chamber assays) utilizes substrates that lack the complexities of the natural microenvironment. Here, we employed a three-dimensional, electrospun poly-(caprolactone) (PCL) nanofiber system (NFS) to mimic some features of topographical properties evidenced in vivo. Co-cultures of human GBM cells and rat astrocytes, as performed on the NFS, showed a significant increase in astrocyte GFAP expression, particularly in the presence of extracellular matrix (ECM) deposited by GBM cells. In addition, GBM migration increased in the presence of astrocytes or soluble factors (i.e., conditioned media). However, the presence of fixed astrocytes acted as an antagonist, lowering GBM migration rates. This data suggests that astrocytes and GBM cells interact through a multitude of pathways, including soluble factors and direct contact. This work demonstrates the potential of the NFS to duplicate some topographical features of the GBM tumor microenvironment, permitting analysis of topographical effects in GBM migration.


Subject(s)
Astrocytes/metabolism , Biomimetics/methods , Glioblastoma/pathology , Nanofibers/chemistry , White Matter/metabolism , Animals , Astrocytes/cytology , Cell Line , Cell Line, Tumor , Cell Movement , Coculture Techniques , Extracellular Matrix/metabolism , Humans , Rats
5.
J Mater Chem B ; 3(40): 7818-7830, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26693013

ABSTRACT

Bone defects can originate from a variety of causes, including trauma, cancer, congenital deformity, and surgical reconstruction. Success of the current "gold standard" treatment (i.e., autologous bone grafts) is greatly influenced by insufficient or inappropriate bone stock. There is thus a critical need for the development of new, engineered materials for bone repair. This review describes the use of natural and synthetic hydrogels as scaffolds for bone tissue engineering. We discuss many of the advantages that hydrogels offer as bone repair materials, including their potential for osteoconductivity, biodegradability, controlled growth factor release, and cell encapsulation. We also discuss the use of hydrogels in composite devices with metals, ceramics, or polymers. These composites are useful because of the low mechanical moduli of hydrogels. Finally, the potential for thermosetting and photo-cross-linked hydrogels as three-dimensionally (3D) printed, patient-specific devices is highlighted. Three-dimensional printing enables controlled spatial distribution of scaffold materials, cells, and growth factors. Hydrogels, especially natural hydrogels present in bone matrix, have great potential to augment existing bone tissue engineering devices for the treatment of critical size bone defects.

6.
ACS Appl Mater Interfaces ; 5(19): 9276-84, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-24010546

ABSTRACT

Glioblastoma multiforme (GBM) tumors, which arise from glia in the central nervous system (CNS), are one of the most deadly forms of human cancer with a median survival time of ∼1 year. Their high infiltrative capacity makes them extremely difficult to treat, and even with aggressive multimodal clinical therapies, outcomes are dismal. To improve understanding of cell migration in these tumors, three-dimensional (3D) multicomponent composite hydrogels consisting of collagen and hyaluronic acid, or hyaluronan (HA), were developed. Collagen is a component of blood vessels known to be associated with GBM migration; whereas, HA is one of the major components of the native brain extracellular matrix (ECM). We characterized hydrogel microstructural features and utilized these materials to investigate patient tumor-derived, single cell morphology, spreading, and migration in 3D culture. GBM morphology was influenced by collagen type with cells adopting a rounded morphology in collagen-IV versus a spindle-shaped morphology in collagen-I/III. GBM spreading and migration were inversely dependent on HA concentration; with higher concentrations promoting little or no migration. Further, noncancerous astrocytes primarily displayed rounded morphologies at lower concentrations of HA; in contrast to the spindle-shaped (spread) morphologies of GBMs. These results suggest that GBM behaviors are sensitive to ECM mimetic materials in 3D and that these composite hydrogels could be used to develop 3D brain mimetic models for studying migration processes.


Subject(s)
Cell Movement , Collagen/chemistry , Glioblastoma/chemistry , Hydrogels/chemistry , Astrocytes/pathology , Cell Line, Tumor , Cell Proliferation , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Glioblastoma/pathology , Humans , Hyaluronic Acid/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL