Cargando…

Preferential, enhanced breast cancer cell migration on biomimetic electrospun nanofiber ‘cell highways’

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 screeni...

Descripción completa

Detalles Bibliográficos
Autores principales: Nelson, Mark Tyler, Short, Aaron, Cole, Sara L, Gross, Amy C, Winter, Jessica, Eubank, Tim D, Lannutti, John J
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4236463/
https://www.ncbi.nlm.nih.gov/pubmed/25385001
http://dx.doi.org/10.1186/1471-2407-14-825
_version_ 1782345168080338944
author Nelson, Mark Tyler
Short, Aaron
Cole, Sara L
Gross, Amy C
Winter, Jessica
Eubank, Tim D
Lannutti, John J
author_facet Nelson, Mark Tyler
Short, Aaron
Cole, Sara L
Gross, Amy C
Winter, Jessica
Eubank, Tim D
Lannutti, John J
author_sort Nelson, Mark Tyler
collection PubMed
description 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. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/1471-2407-14-825) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-4236463
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-42364632014-11-19 Preferential, enhanced breast cancer cell migration on biomimetic electrospun nanofiber ‘cell highways’ Nelson, Mark Tyler Short, Aaron Cole, Sara L Gross, Amy C Winter, Jessica Eubank, Tim D Lannutti, John J BMC Cancer Research Article 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. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/1471-2407-14-825) contains supplementary material, which is available to authorized users. BioMed Central 2014-11-10 /pmc/articles/PMC4236463/ /pubmed/25385001 http://dx.doi.org/10.1186/1471-2407-14-825 Text en © Nelson et al.; licensee BioMed Central Ltd. 2014 This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research Article
Nelson, Mark Tyler
Short, Aaron
Cole, Sara L
Gross, Amy C
Winter, Jessica
Eubank, Tim D
Lannutti, John J
Preferential, enhanced breast cancer cell migration on biomimetic electrospun nanofiber ‘cell highways’
title Preferential, enhanced breast cancer cell migration on biomimetic electrospun nanofiber ‘cell highways’
title_full Preferential, enhanced breast cancer cell migration on biomimetic electrospun nanofiber ‘cell highways’
title_fullStr Preferential, enhanced breast cancer cell migration on biomimetic electrospun nanofiber ‘cell highways’
title_full_unstemmed Preferential, enhanced breast cancer cell migration on biomimetic electrospun nanofiber ‘cell highways’
title_short Preferential, enhanced breast cancer cell migration on biomimetic electrospun nanofiber ‘cell highways’
title_sort preferential, enhanced breast cancer cell migration on biomimetic electrospun nanofiber ‘cell highways’
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4236463/
https://www.ncbi.nlm.nih.gov/pubmed/25385001
http://dx.doi.org/10.1186/1471-2407-14-825
work_keys_str_mv AT nelsonmarktyler preferentialenhancedbreastcancercellmigrationonbiomimeticelectrospunnanofibercellhighways
AT shortaaron preferentialenhancedbreastcancercellmigrationonbiomimeticelectrospunnanofibercellhighways
AT colesaral preferentialenhancedbreastcancercellmigrationonbiomimeticelectrospunnanofibercellhighways
AT grossamyc preferentialenhancedbreastcancercellmigrationonbiomimeticelectrospunnanofibercellhighways
AT winterjessica preferentialenhancedbreastcancercellmigrationonbiomimeticelectrospunnanofibercellhighways
AT eubanktimd preferentialenhancedbreastcancercellmigrationonbiomimeticelectrospunnanofibercellhighways
AT lannuttijohnj preferentialenhancedbreastcancercellmigrationonbiomimeticelectrospunnanofibercellhighways