Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cancer Res ; 81(13): 3649-3663, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33975882

ABSTRACT

Although intratumoral genomic heterogeneity can impede cancer research and treatment, less is known about the effects of phenotypic heterogeneities. To investigate the role of cell migration heterogeneities in metastasis, we phenotypically sorted metastatic breast cancer cells into two subpopulations based on migration ability. Although migration is typically considered to be associated with metastasis, when injected orthotopically in vivo, the weakly migratory subpopulation metastasized significantly more than the highly migratory subpopulation. To investigate the mechanism behind this observation, both subpopulations were assessed at each stage of the metastatic cascade, including dissemination from the primary tumor, survival in the circulation, extravasation, and colonization. Although both subpopulations performed each step successfully, weakly migratory cells presented as circulating tumor cell (CTC) clusters in the circulation, suggesting clustering as one potential mechanism behind the increased metastasis of weakly migratory cells. RNA sequencing revealed weakly migratory subpopulations to be more epithelial and highly migratory subpopulations to be more mesenchymal. Depletion of E-cadherin expression from weakly migratory cells abrogated metastasis. Conversely, induction of E-cadherin expression in highly migratory cells increased metastasis. Clinical patient data and blood samples showed that CTC clustering and E-cadherin expression are both associated with worsened patient outcome. This study demonstrates that deconvolving phenotypic heterogeneities can reveal fundamental insights into metastatic progression. More specifically, these results indicate that migratory ability does not necessarily correlate with metastatic potential and that E-cadherin promotes metastasis in phenotypically sorted breast cancer cell subpopulations by enabling CTC clustering. SIGNIFICANCE: This study employs phenotypic cell sorting for migration to reveal a weakly migratory, highly metastatic breast cancer cell subpopulation regulated by E-cadherin, highlighting the dichotomy between cancer cell migration and metastasis.


Subject(s)
Antigens, CD/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Neoplastic Cells, Circulating/pathology , Animals , Antigens, CD/genetics , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cadherins/genetics , Cell Proliferation , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Neoplastic Cells, Circulating/metabolism , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Sci Rep ; 7: 42088, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28186196

ABSTRACT

A critical step in breast cancer progression is local tissue invasion, during which cells pass from the epithelial compartment to the stromal compartment. We recently showed that malignant leader cells can promote the invasion of otherwise non-invasive epithelial follower cells, but the effects of this induced-invasion phenomenon on follower cell phenotype remain unclear. Notably, this process can expose epithelial cells to the stromal extracellular matrix (ECM), which is distinct from the ECM within the normal epithelial microenvironment. Here, we used a 3D epithelial morphogenesis model in which cells were cultured in biochemically and mechanically defined matrices to examine matrix-mediated gene expression and the associated phenotypic response. We found that 3D collagen matrix promoted expression of mesenchymal genes including MT1-MMP, which was required for collagen-stimulated invasive behavior. Epithelial invasion required matrix anchorage as well as signaling through Src, PI3K, and Rac1, and increasingly stiff collagen promoted dispersive epithelial cell invasion. These results suggest that leader cell-facilitated access to the stromal ECM may trigger an invasive phenotype in follower epithelial cells that could enable them to actively participate in local tissue invasion.


Subject(s)
Cell Movement , Collagen/metabolism , Epithelial Cells/physiology , Extracellular Matrix/metabolism , Tumor Microenvironment , Cell Culture Techniques , Humans , Models, Biological , Phenotype
3.
Integr Biol (Camb) ; 8(8): 821-35, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27384462

ABSTRACT

Cell migration within 3D interstitial microenvironments is sensitive to extracellular matrix (ECM) properties, but the mechanisms that regulate migration guidance by 3D matrix features remain unclear. To examine the mechanisms underlying the cell migration response to aligned ECM, which is prevalent at the tumor-stroma interface, we utilized time-lapse microscopy to compare the behavior of MDA-MB-231 breast adenocarcinoma cells within randomly organized and well-aligned 3D collagen ECM. We developed a novel experimental system in which cellular morphodynamics during initial 3D cell spreading served as a reductionist model for the complex process of matrix-directed 3D cell migration. Using this approach, we found that ECM alignment induced spatial anisotropy of cells' matrix probing by promoting protrusion frequency, persistence, and lengthening along the alignment axis and suppressing protrusion dynamics orthogonal to alignment. Preference for on-axis behaviors was dependent upon FAK and Rac1 signaling and translated across length and time scales such that cells within aligned ECM exhibited accelerated elongation, front-rear polarization, and migration relative to cells in random ECM. Together, these findings indicate that adhesive and protrusive signaling allow cells to respond to coordinated physical cues in the ECM, promoting migration efficiency and cell migration guidance by 3D matrix structure.


Subject(s)
Cell Movement , Cell Surface Extensions , Extracellular Matrix/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Anisotropy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion , Cell Line, Tumor , Collagen/chemistry , Female , Humans , Mammary Neoplasms, Animal/metabolism , Mice , Neoplasm Invasiveness , Signal Transduction
4.
Mol Biol Cell ; 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26960796

ABSTRACT

During metastasis, cells can use proteolytic activity to form tube-like "microtracks" within the extracellular matrix (ECM). Using these microtracks, cells can migrate unimpeded through the stroma. To investigate the molecular mechanisms of microtrack migration, we developed an in vitro 3D micromolded collagen platform. When in microtracks, cells tend to migrate unidirectionally. Since focal adhesions are the primary mechanism by which cells interact with the ECM, we examined the roles of several focal adhesion molecules in driving unidirectional motion. Vinculin knockdown results in the repeated reversal of migration direction compared with control cells. Tracking the position of the Golgi centroid relative to the position of the nucleus centroid reveals that vinculin knockdown disrupts cell polarity in microtracks. Vinculin also directs migration on 2D substrates and in 3D uniform collagen matrices, indicated by reduced speed, shorter net displacement and decreased directionality in vinculin-deficient cells. In addition, vinculin is necessary for Focal Adhesion Kinase (FAK) activation in 3D as vinculin knockdown results in reduced FAK activation in both 3D uniform collagen matrices and microtracks, but not on 2D substrates, and accordingly, FAK inhibition halts cell migration in 3D microtracks. Together, these data indicate that vinculin plays a key role in polarization during migration.

5.
Cell Mol Bioeng ; 8(1): 76-85, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25866589

ABSTRACT

As cancer progresses, cells must adapt to a new and stiffer environment, which can ultimately alter how normal cells within the tumor behave. In turn, these cells are known to further aid tumor progression. Therefore, there is potentially a unique avenue to better understand metastatic potential through single-cell biophysical assays performed on patient-derived cells. Here, we perform biophysical characterization of primary human fibroblastic cells obtained from mammary carcinoma and normal contralateral tissue. Through a series of tissue dissociation, differential centrifugation and trypsinization steps, we isolate an adherent fibroblastic population viable for biomechanical testing. 2D TFM and 3D migration measurements in a collagen matrix show that fibroblasts obtained from patient tumors generate more traction forces and display improved migration potential than their counterparts from normal tissue. Moreover, through the use of an embedded spheroid model, we confirmed the extracellular matrix (ECM) remodeling behavior of primary cells isolated from carcinoma. Overall, correlating biophysical characterization of normal- and carcinoma-derived samples from individual patient along with patient outcome may become a powerful approach to further our comprehension of metastasis and ultimately design drug targets on a patient-specific basis.

6.
PLoS One ; 10(3): e0119008, 2015.
Article in English | MEDLINE | ID: mdl-25785437

ABSTRACT

Vascular smooth muscle cell (VSMC) migration and matrix degradation occurs with intimal hyperplasia associated with atherosclerosis, vascular injury, and restenosis. One proposed mechanism by which VSMCs degrade matrix is through the use of podosomes, transient actin-based structures that are thought to play a role in extracellular matrix degradation by creating localized sites of matrix metalloproteinase (MMP) secretion. To date, podosomes in VSMCs have largely been studied by stimulating cells with phorbol esters, such as phorbol 12,13-dibutyrate (PDBu), however little is known about the physiological cues that drive podosome formation. We present the first evidence that physiological, physical stimuli mimicking cues present within the microenvironment of diseased arteries can induce podosome formation in VSMCs. Both microtopographical cues and imposed pressure mimicking stage II hypertension induce podosome formation in A7R5 rat aortic smooth muscle cells. Moreover, wounding using a scratch assay induces podosomes at the leading edge of VSMCs. Notably the effect of each of these biophysical stimuli on podosome stimulation can be inhibited using a Src inhibitor. Together, these data indicate that physical cues can induce podosome formation in VSMCs.


Subject(s)
Muscle, Smooth, Vascular/cytology , Podosomes/physiology , Actins/metabolism , Animals , Biophysical Phenomena/physiology , Cell Line , Muscle, Smooth, Vascular/metabolism , Phosphorylation , Rats , src-Family Kinases/metabolism
7.
Am J Physiol Cell Physiol ; 308(6): C436-47, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25500742

ABSTRACT

Tumor cell invasion through the stromal extracellular matrix (ECM) is a key feature of cancer metastasis, and understanding the cellular mechanisms of invasive migration is critical to the development of effective diagnostic and therapeutic strategies. Since cancer cell migration is highly adaptable to physiochemical properties of the ECM, it is critical to define these migration mechanisms in a context-specific manner. Although extensive work has characterized cancer cell migration in two- and three-dimensional (3D) matrix environments, the migration program employed by cells to move through native and cell-derived microtracks within the stromal ECM remains unclear. We previously reported the development of an in vitro model of patterned type I collagen microtracks that enable matrix metalloproteinase-independent microtrack migration. Here we show that collagen microtracks closely resemble channel-like gaps in native mammary stroma ECM and examine the extracellular and intracellular mechanisms underlying microtrack migration. Cell-matrix mechanocoupling, while critical for migration through 3D matrix, is not necessary for microtrack migration. Instead, cytoskeletal dynamics, including actin polymerization, cortical tension, and microtubule turnover, enable persistent, polarized migration through physiological microtracks. These results indicate that tumor cells employ context-specific mechanisms to migrate and suggest that selective targeting of cytoskeletal dynamics, but not adhesion, proteolysis, or cell traction forces, may effectively inhibit cancer cell migration through preformed matrix microtracks within the tumor stroma.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Collagen/metabolism , Extracellular Matrix/metabolism , Mammary Neoplasms, Experimental/metabolism , Tumor Microenvironment , Actins/metabolism , Actomyosin/metabolism , Animals , Breast Neoplasms/pathology , Cell Adhesion , Cell Line, Tumor , Cell Shape , Cytoskeleton/metabolism , Extracellular Matrix/pathology , Female , Humans , Integrin beta1/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mechanotransduction, Cellular , Mice, Inbred NOD , Mice, Transgenic , Neoplasm Invasiveness , Time Factors
8.
Integr Biol (Camb) ; 5(3): 606-16, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23388698

ABSTRACT

While the mechanisms employed by metastatic cancer cells to migrate remain poorly understood, it has been widely accepted that metastatic cancer cells can invade the tumor stroma by degrading the extracellular matrix (ECM) with matrix metalloproteinases (MMPs). Although MMP inhibitors showed early promise in preventing metastasis in animal models, they have largely failed clinically. Recently, studies have shown that some cancer cells can use proteolysis to mechanically rearrange their ECM to form tube-like "microtracks" which other cells can follow without using MMPs themselves. We speculate that this mode of migration in the secondary cells may be one example of migration which can occur without endogenous protease activity in the secondary cells. Here we present a technique to study this migration in a 3D, collagen-based environment which mimics the size and topography of the tracks produced by proteolytically active cancer cells. Using time-lapse phase-contrast microscopy, we find that these microtracks permit the rapid and persistent migration of noninvasive MCF10A mammary epithelial cells, which are unable to otherwise migrate in 3D collagen. Additionally, while highly metastatic MDAMB231 breast cancer cells are able to invade a 3D collagen matrix, seeding within the patterned microtracks induced significantly increased cell migration speed, which was not decreased by pharmacological MMP inhibition. Together, these data suggest that microtracks within a 3D ECM may facilitate the migration of cells in an MMP-independent fashion, and may reveal novel insight into the clinical challenges facing MMP inhibitors.


Subject(s)
Cell Culture Techniques , Collagen/chemistry , Extracellular Matrix/metabolism , Matrix Metalloproteinases/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Culture Media , Female , Fluorescent Dyes , Humans , Microscopy, Phase-Contrast , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , RNA Interference
9.
Clin Exp Metastasis ; 30(5): 615-30, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23328900

ABSTRACT

Solid tumors consist of genetically and phenotypically diverse subpopulations of cancer cells with unique capacities for growth, differentiation, and invasion. While the molecular and microenvironmental bases for heterogeneity are increasingly appreciated, the outcomes of such intratumor heterogeneity, particularly in the context of tumor invasion and metastasis, remain poorly understood. To study heterotypic cell-cell interactions and elucidate the biological consequences of intratumor heterogeneity, we developed a tissue-engineered multicellular spheroid (MCS) co-culture model that recapitulates the cellular diversity and fully three-dimensional cell-cell and cell-matrix interactions that characterize human carcinomas. We found that "invasion-competent" malignant cells induced the collective invasion of otherwise "invasion-incompetent" epithelial cells, and that these two cell types consistently exhibited distinct leader and follower roles during invasion. Analysis of extracellular matrix (ECM) microarchitecture revealed that malignant cell invasion was accompanied by extensive ECM remodeling including matrix alignment and proteolytic track-making. Inhibition of cell contractility- and proteolysis-mediated matrix reorganization prevented leader-follower behavior and malignant cell-induced epithelial cell invasion. These results indicate that heterogeneous subpopulations within a tumor may possess specialized roles during tumor progression and suggest that complex interactions among the various subpopulations of cancer cells within a tumor may regulate critical aspects of tumor biology and affect clinical outcome.


Subject(s)
Epithelial Cells/parasitology , Models, Biological , Neoplasm Invasiveness , Spheroids, Cellular , Cell Line, Tumor , Extracellular Matrix/pathology , Female , Humans , Tumor Microenvironment
10.
Methods Cell Biol ; 110: 139-78, 2012.
Article in English | MEDLINE | ID: mdl-22482948

ABSTRACT

Contractile force generation plays a critical role in cell adhesion, migration, and extracellular matrix reorganization in both 2D and 3D environments. Characterization of cellular forces has led to a greater understanding of cell migration, cellular mechanosensing, tissue formation, and disease progression. Methods to characterize cellular traction stresses now date back over 30 years, and they have matured from qualitative comparisons of cell-mediated substrate movements to high-resolution, highly quantitative measures of cellular force. Here, we will provide an overview of common methods used to measure forces in both 2D and 3D microenvironments. Specific focus will be placed on traction force microscopy, which measures the force exerted by cells on 2D planar substrates, and the use of confocal reflectance microscopy, which can be used to quantify collagen fibril compaction as a metric for 3D traction forces. In addition to providing experimental methods to analyze cellular forces, we discuss the application of these techniques to a large range of biomedical problems and some of the significant challenges that still remain in this field.


Subject(s)
Mechanotransduction, Cellular/physiology , Single-Cell Analysis/methods , Stress, Mechanical , Acrylic Resins/chemistry , Cell Adhesion , Cell Communication , Cell Movement , Cells, Cultured , Collagen/chemistry , Elastic Modulus , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Fibronectins/chemistry , Humans , Laminin/chemistry , Microscopy, Confocal
11.
Biomaterials ; 33(16): 4157-65, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22405848

ABSTRACT

Fibrillar collagen gels, which are used extensively in vitro to study tumor-microenvironment interactions, are composed of a cell-instructive network of interconnected fibers and pores whose organization is sensitive to polymerization conditions such as bulk concentration, pH, and temperature. Using confocal reflectance microscopy and image autocorrelation analysis to quantitatively assess gel microarchitecture, we show that additional polymerization parameters including culture media formulation and gel thickness significantly affect the dimensions and organization of fibers and pores in collagen gels. These findings enabled the development of a three-dimensional culture system in which cell-scale gel microarchitecture was decoupled from bulk gel collagen concentration. Interestingly, morphology and migration characteristics of embedded MDA-MB-231 cells were sensitive to gel microarchitecture independently of collagen gel concentration. Cells adopted a polarized, motile phenotype in gels with larger fibers and pores and a rounded or stellate, less motile phenotype in gels with small fibers and pores regardless of bulk gel density. Conversely, cell proliferation was sensitive to gel concentration but not microarchitecture. These results indicate that cell-scale gel microarchitecture may trump bulk-scale gel density in controlling specific cell behaviors, underscoring the biophysical role of gel microarchitecture in influencing cell behavior.


Subject(s)
Breast Neoplasms/pathology , Extracellular Matrix , Biophysics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Collagen/metabolism , Culture Media , Female , Humans , Microscopy, Confocal
12.
Crit Rev Oncol Hematol ; 83(2): 170-83, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22178415

ABSTRACT

The physical sciences and engineering have introduced novel perspectives into the study of cancer through model systems, tools, and metrics that enable integration of basic science observations with clinical data. These methods have contributed to the identification of several overarching mechanisms that drive processes during cancer progression including tumor growth, angiogenesis, and metastasis. During tumor cell invasion - the first clinically observable step of metastasis - cells demonstrate diverse and evolving physical phenotypes that cannot typically be defined by any single molecular mechanism, and mechanobiology has been used to study the physical cell behaviors that comprise the "invasive phenotype". In this review, we discuss the continually evolving pathological characterization and in vitro mechanobiological characterization of tumor invasion, with emphasis on emerging physical biology and mechanobiology strategies that have contributed to a more robust mechanistic understanding of tumor cell invasion. These physical approaches may ultimately help to better predict and identify tumor metastasis.


Subject(s)
Biophysical Phenomena , Neoplasm Invasiveness/pathology , Neoplasms/pathology , Animals , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Neoplasms/metabolism
13.
Macromol Biosci ; 12(1): 12-20, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22021131

ABSTRACT

Both substrate topography and substrate mechanical properties are known to influence cell behavior, but little is known about how they act in concert. Here, a method is presented to introduce topographical features into PA hydrogel substrates that span a wide range of physiological E values. Gel swelling plays a significant role in the fidelity of protruding micromolded features, with the most efficient pattern transfer occurring at a crosslinking concentration equal to or greater than ≈5%. In contrast, swelling does not influence the spacing fidelity of microcontact printed islands of collagen on 2D PA substrates. BAECs cultured on micromolded PA substrates exhibit contact guidance along ridges patterned for all E tested.


Subject(s)
Acrylamide/chemistry , Acrylic Resins/chemistry , Cell Movement/physiology , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Biocompatible Materials , Biomechanical Phenomena , Cell Adhesion/physiology , Cells, Cultured , Elasticity , Endothelial Cells/metabolism , Materials Testing , Surface Properties
14.
Phys Biol ; 8(1): 015009, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21301071

ABSTRACT

To adhere and migrate, cells generate forces through the cytoskeleton that are transmitted to the surrounding matrix. While cellular force generation has been studied on 2D substrates, less is known about cytoskeletal-mediated traction forces of cells embedded in more in vivo-like 3D matrices. Recent studies have revealed important differences between the cytoskeletal structure, adhesion, and migration of cells in 2D and 3D. Because the cytoskeleton mediates force, we sought to directly compare the role of the cytoskeleton in modulating cell force in 2D and 3D. MDA-MB-231 cells were treated with agents that perturbed actin, microtubules, or myosin, and analyzed for changes in cytoskeletal organization and force generation in both 2D and 3D. To quantify traction stresses in 2D, traction force microscopy was used; in 3D, force was assessed based on single cell-mediated collagen fibril reorganization imaged using confocal reflectance microscopy. Interestingly, even though previous studies have observed differences in cell behaviors like migration in 2D and 3D, our data indicate that forces generated on 2D substrates correlate with forces within 3D matrices. Disruption of actin, myosin or microtubules in either 2D or 3D microenvironments disrupts cell-generated force. These data suggest that despite differences in cytoskeletal organization in 2D and 3D, actin, microtubules and myosin contribute to contractility and matrix reorganization similarly in both microenvironments.


Subject(s)
Cytoskeleton/ultrastructure , Microscopy, Atomic Force/methods , Tissue Scaffolds/chemistry , Actins/ultrastructure , Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Cytoskeleton/drug effects , Female , Humans , Microtubules/ultrastructure , Myosins/ultrastructure , Single-Cell Analysis/methods , Tubulin Modulators/pharmacology
15.
J Biomed Mater Res A ; 96(2): 301-12, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21171149

ABSTRACT

We developed a method to produce discrete fibrin microthreads, which can be seeded with human mesenchymal stem cells (hMSCs) and used as a suture to enhance the efficiency and localization of cell delivery. To assess the efficacy of fibrin microthreads to support hMSC attachment, proliferation, and survival, microthreads (100 µm diameter per microthread) were bundled together, seeded with 50,000 hMSCs for 2 h, and cultured for 5 days. Cell density on microthread bundles increased over time in culture to a maximum average density of 731 ± 101 cells/mm(2) after 5 days. A LIVE/DEAD assay confirmed that the cells were viable, and Ki-67 staining verified hMSC proliferation. In addition, functional differentiation assays demonstrated that hMSCs cultured on microthreads retained their ability to differentiate into adipocytes and osteocytes. The results of this study demonstrate that fibrin microthreads support hMSC viability and proliferation, while maintaining their multipotency. We anticipate that these cell-seeded fibrin microthreads will serve as a platform technology to improve localized delivery and engraftment of viable cells to damaged tissue.


Subject(s)
Cell Differentiation/drug effects , Fibrin/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Biomarkers/metabolism , Cattle , Cell Adhesion/drug effects , Cell Count , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Collagen/pharmacology , Gels , Humans , Ki-67 Antigen/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Osteogenesis/drug effects , Rats , Sus scrofa , Time Factors
16.
Article in English | MEDLINE | ID: mdl-22255298

ABSTRACT

Cells use cytoskeletally-generated force to adhere, migrate and remodel their environment. While cellular forces generated by cells plated on 2D substrates is well-studied, much less is known about the forces generated by cells in 3D matrices, which more closely mimic the in vivo environment. Here, an approach to characterize cellular forces in 3D using confocal reflectance microscopy is presented. Remodeling of collagen fibrils due to the forces exerted by embedded cells was imaged in real-time as cells adhere to and contract the matrix. We implemented this approach in conjunction with 2D Traction Force Microscopy to compare cytoskeletally-mediated forces of cells in 3D collagen matrices to forces exerted by cells on 2D collagen-coated hydrogel substrates. Our results indicate that confocal reflectance microscopy of collagen fibrils can provide semi-quantitative information regarding cellular force in 3D matrices, and that the actin cytoskeleton plays a similar role in regulating cell contractility in both 2D and 3D microenvironments.


Subject(s)
Collagen/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...