Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
FASEB J ; 26(10): 4045-56, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22707566

ABSTRACT

Cell migration on planar surfaces is driven by cycles of actin protrusion, integrin-mediated adhesion, and myosin-mediated contraction; however, this mechanism may not accurately describe movement in 3-dimensional (3D) space. By subjecting cells to restrictive 3D environments, we demonstrate that physical confinement constitutes a biophysical stimulus that alters cell morphology and suppresses mesenchymal motility in human breast carcinoma (MDA-MB-231). Dorsoventral polarity, stress fibers, and focal adhesions are markedly attenuated by confinement. Inhibitors of myosin, Rho/ROCK, or ß1-integrins do not impair migration through 3-µm-wide channels (confinement), even though these treatments repress motility in 50-µm-wide channels (unconfined migration) by ≥50%. Strikingly, confined migration persists even when F-actin is disrupted, but depends largely on microtubule (MT) dynamics. Interfering with MT polymerization/depolymerization causes confined cells to undergo frequent directional changes, thereby reducing the average net displacement by ≥80% relative to vehicle controls. Live-cell EB1-GFP imaging reveals that confinement redirects MT polymerization toward the leading edge, where MTs continuously impact during advancement of the cell front. These results demonstrate that physical confinement can induce cytoskeletal alterations that reduce the dependence of migrating cells on adhesion-contraction force coupling. This mechanism may explain why integrins can exhibit reduced or altered function during migration in 3D environments.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , Actins/metabolism , Amides/pharmacology , Azepines/pharmacology , Cell Line, Tumor , Cytoskeleton/metabolism , Humans , Microtubules/metabolism , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/metabolism , Naphthalenes/pharmacology , Paclitaxel/pharmacology , Pyridines/pharmacology , Tubulin Modulators/pharmacology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
2.
PLoS One ; 7(1): e29211, 2012.
Article in English | MEDLINE | ID: mdl-22279529

ABSTRACT

Cell migration is crucial for both physiological and pathological processes. Current in vitro cell motility assays suffer from various drawbacks, including insufficient temporal and/or optical resolution, or the failure to include a controlled chemotactic stimulus. Here, we address these limitations with a migration chamber that utilizes a self-sustaining chemotactic gradient to induce locomotion through confined environments that emulate physiological settings. Dynamic real-time analysis of both population-scale and single-cell movement are achieved at high resolution. Interior surfaces can be functionalized through adsorption of extracellular matrix components, and pharmacological agents can be administered to cells directly, or indirectly through the chemotactic reservoir. Direct comparison of multiple cell types can be achieved in a single enclosed system to compare inherent migratory potentials. Our novel microfluidic design is therefore a powerful tool for the study of cellular chemotaxis, and is suitable for a wide range of biological and biomedical applications.


Subject(s)
Cell Movement/physiology , Chemotaxis/physiology , Microfluidic Analytical Techniques/methods , Single-Cell Analysis/methods , Actins/genetics , Actins/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Chemotaxis/drug effects , Dimethylpolysiloxanes/chemistry , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Microfluidic Analytical Techniques/instrumentation , Microscopy, Confocal , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Paclitaxel/pharmacology , Reproducibility of Results , Single-Cell Analysis/instrumentation , Thiazolidines/pharmacology , Transfection
3.
Article in English | MEDLINE | ID: mdl-21913338

ABSTRACT

Cell-cell adhesion molecules (CAMs) comprise a broad class of linker proteins that are crucial for the development of multicellular organisms, and for the continued maintenance of organ and tissue structure. Because of its pivotal function in tissue homeostasis, the deregulation of intercellular adhesion is linked to the onset of most solid tumors. The breakdown of homeostatic cell adhesions in highly ordered epithelial sheets is directly implicated in carcinogenesis, while continued changes in the adhesion profile of the primary tumor mass facilitate growth and expansion into adjacent tissue. Intercellular adhesion molecules are also involved in each subsequent phase of metastasis, including transendothelial migration, transit through the bloodstream or lymphatics, and renewed proliferation in secondary sites. This review addresses various roles of cadherin- and selectin-mediated intercellular adhesion in tumor initiation and malignant transformation, and discusses the mechanisms for the arrest and adhesion of circulating tumor cells to the vessel endothelium. Considering the contributions of these CAMs to cancer progression in the context of a systematic biological framework may prove valuable in identifying new ways to diagnose and treat cancer.


Subject(s)
Cell Adhesion Molecules/metabolism , Neoplasm Metastasis , Neoplasms/metabolism , Animals , Cadherins/deficiency , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion , Epithelial-Mesenchymal Transition , Epithelium/metabolism , Epithelium/physiopathology , Humans , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Signal Transduction
4.
FASEB J ; 26(3): 1349-59, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22159147

ABSTRACT

Selectins promote metastasis by mediating specific interactions between selectin ligands on tumor cells and selectin-expressing host cells in the microvasculature. Using affinity chromatography in conjunction with tandem mass spectrometry and bioinformatics tools, we identified mucin 16 (MUC16) as a novel selectin ligand expressed by metastatic pancreatic cancer cells. While up-regulated in many pancreatic cancers, the biological function of sialofucosylated MUC16 has yet to be fully elucidated. To address this, we employed blot rolling and cell-free flow-based adhesion assays using MUC16 immunopurified from pancreatic cancer cells and found that it efficiently binds E- and L- but not P-selectin. The selectin-binding determinants are sialofucosylated structures displayed on O- and N-linked glycans. Silencing MUC16 expression by RNAi markedly reduces pancreatic cancer cell binding to E- and L-selectin under flow. These findings provide a novel integrated perspective on the enhanced metastatic potential associated with MUC16 overexpression and the role of selectins in metastasis.


Subject(s)
CA-125 Antigen/metabolism , E-Selectin/metabolism , L-Selectin/metabolism , Membrane Proteins/metabolism , P-Selectin/metabolism , Animals , Binding Sites , Blotting, Western , CA-125 Antigen/genetics , CA-125 Antigen/isolation & purification , CHO Cells , Cell Line , Cell Line, Tumor , Chromatography, Affinity , Cricetinae , Cricetulus , E-Selectin/genetics , Flow Cytometry , Fucose/metabolism , Humans , Immunoprecipitation , L-Selectin/genetics , Ligands , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , N-Acetylneuraminic Acid/metabolism , Neoplasm Metastasis , P-Selectin/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Polysaccharides/metabolism , Protein Binding , RNA Interference , Tandem Mass Spectrometry
5.
Phys Biol ; 8(1): 015013, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21301059

ABSTRACT

Selectins (L-, E- and P-selectin) are calcium-dependent transmembrane glycoproteins that are expressed on the surface of circulating leukocytes, activated platelets, and inflamed endothelial cells. Selectins bind predominantly to sialofucosylated glycoproteins and glycolipids (E-selectin only) present on the surface of apposing cells, and mediate transient adhesive interactions pertinent to inflammation and cancer metastasis. The rapid turnover of selectin-ligand bonds, due to their fast on- and off-rates along with their remarkably high tensile strengths, enables them to mediate cell tethering and rolling in shear flow. This paper presents the current body of knowledge regarding the role of selectins in inflammation and cancer metastasis, and discusses experimental methodologies and mathematical models used to resolve the biophysics of selectin-mediated cell adhesion. Understanding the biochemistry and biomechanics of selectin-ligand interactions pertinent to inflammatory disorders and cancer metastasis may provide insights for developing promising therapies and/or diagnostic tools to combat these disorders.


Subject(s)
Inflammation/metabolism , Neoplasms/metabolism , Selectins/metabolism , Biophysical Phenomena , Cell Adhesion , Humans , Models, Biological , Neoplasm Metastasis/pathology , Neoplasms/pathology , Protein Binding
6.
Breast Cancer Res Treat ; 129(3): 691-701, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21069453

ABSTRACT

Detached breast tumor cells produce dynamic microtubule protrusions that promote reattachment of cells and are termed tubulin microtentacles (McTNs) due to their mechanistic distinctions from actin-based filopodia/invadopodia and tubulin-based cilia. McTNs are enriched with vimentin and detyrosinated α-tubulin, (Glu-tubulin). Evidence suggests that vimentin and Glu-tubulin are cross-linked by kinesin motor proteins. Using known kinesin inhibitors, Lidocaine and Tetracaine, the roles of kinesins in McTN formation and function were tested. Live-cell McTN counts, adhesion assays, immunofluorescence, and video microscopy were performed to visualize inhibitor effects on McTNs. Viability and apoptosis assays were used to confirm the non-toxicity of the inhibitors. Treatments of human non-tumorigenic mammary epithelial and breast tumor cells with Lidocaine or Tetracaine caused rapid collapse of vimentin filaments. Live-cell video microscopy demonstrated that Tetracaine reduces motility of intracellular GFP-kinesin and causes centripetal collapse of McTNs. Treatment with Tetracaine inhibited the extension of McTNs and their ability to promote tumor cell aggregation and reattachment. Lidocaine showed similar effects but to a lesser degree. Our current data support a model in which the inhibition of kinesin motor proteins by Tetracaine leads to the reductions in McTNs, and provides a novel mechanism for the ability of this anesthetic to decrease metastatic progression.


Subject(s)
Anesthetics, Local/pharmacology , Breast Neoplasms/pathology , Kinesins/antagonists & inhibitors , Kinesins/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Kinesins/genetics , Lidocaine/pharmacology , Mammary Glands, Human/cytology , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Tetracaine/pharmacology , Tubulin/metabolism , Vimentin/metabolism
7.
Cancer Res ; 70(20): 8127-37, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20924103

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is associated with increased breast tumor metastasis; however, the specific mechanisms by which EMT promotes metastasis remain somewhat unclear. Despite the importance of cytoskeletal dynamics during both EMT and metastasis, very few current studies examine the cytoskeleton of detached and circulating tumor cells. Specific posttranslational α-tubulin modifications are critical for adherent cell motility and implicated in numerous pathologies, but also remain understudied in detached cells. We report here that EMT induced through ectopic expression of Twist or Snail promotes α-tubulin detyrosination and the formation of tubulin-based microtentacles in detached HMLEs. Mechanistically, EMT downregulates the tubulin tyrosine ligase enzyme, resulting in an accumulation of detyrosinated α-tubulin (Glu-tubulin), and increases microtentacles that penetrate endothelial layers to facilitate tumor cell reattachment. Confocal microscopy shows that microtentacles are capable of penetrating the junctions between endothelial cells. Suppression of endogenous Twist in metastatic human breast tumor cells is capable of reducing both tubulin detyrosination and microtentacles. Clinical breast tumor samples display high concordance between Glu-tubulin and Twist expression levels, emphasizing the coupling between EMT and tubulin detyrosination in vivo. Coordinated elevation of Twist and Glu-tubulin at invasive tumor fronts, particularly within ductal carcinoma in situ samples, establishes that EMT-induced tubulin detyrosination occurs at the earliest stages of tumor invasion. These data support a novel model where the EMT that occurs during tumor invasion downregulates tubulin tyrosine ligase, increasing α-tubulin detyrosination and promoting microtentacles that could enhance the reattachment of circulating tumor cells to the vascular endothelium during metastasis.


Subject(s)
Breast Neoplasms/pathology , Epithelium/physiology , Mesoderm/physiology , Tubulin/metabolism , Biopsy , Breast Neoplasms/surgery , Cell Adhesion , Cytoskeleton/physiology , Epithelium/pathology , Female , Humans , Immunohistochemistry , Mesoderm/pathology , Mesoderm/ultrastructure , Neoplasm Invasiveness , Neoplasm Metastasis/pathology , Nuclear Proteins/physiology , Twist-Related Protein 1/physiology , Tyrosine/metabolism
8.
Cancer Res ; 70(20): 7737-41, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20924109

ABSTRACT

Detection of circulating tumor cells (CTC) is advancing as an effective predictor of patient outcome and therapeutic response. Unfortunately, our knowledge of CTC biology remains limited, and the impact of drug treatments on CTC metastatic potential is currently unclear. Improved CTC imaging in vivo and analysis of free-floating tumor cells now show that cytoskeletal regulation in CTCs contrasts starkly with tumor cells attached to extracellular matrix. In this review, we examine how persistent microtubule stabilization promotes the formation of microtentacles on the surface of detached breast tumor cells and enhances metastatic potential.


Subject(s)
Cytoskeleton/pathology , Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Cell Aggregation , Homeostasis , Humans , Intermediate Filaments/metabolism , Intermediate Filaments/pathology , Microtubules/metabolism , Microtubules/pathology , Neoplasm Metastasis , Tubulin/metabolism , Vimentin/metabolism
9.
Breast Cancer Res Treat ; 121(1): 65-78, 2010 May.
Article in English | MEDLINE | ID: mdl-19593636

ABSTRACT

In the clinical treatment of breast cancer, antimitotic cytotoxic agents are one of the most commonly employed chemotherapies, owing largely to their antiproliferative effects on the growth and survival of adherent cells in studies that model primary tumor growth. Importantly, the manner in which these chemotherapeutics impact the metastatic process remains unclear. Furthermore, since dissemination of tumor cells through the systemic circulation and lymphatics necessitates periods of detached survival, it is equally important to consider how circulating tumor cells respond to such compounds. To address this question, we exposed both nontumorigenic and tumor-derived epithelial cell lines to two antitumor compounds, jasplakinolide and paclitaxel (Taxol), in a series of attached and detached states. We report here that jasplakinolide promoted the extension of microtubule-based projections and microtentacle protrusions in adherent and suspended cells, respectively. These protrusions were specifically enriched by upregulation of a stable post-translationally modified form of alpha-tubulin, and this occurred prior to, and independently of any reductions in cellular viability. Microtubule stabilization with Taxol significantly enhanced these effects. Additionally, Taxol promoted the attachment and spreading of suspended tumor cell populations on extracellular matrix. While the antiproliferative effects of these compounds are well recognized and clinically valuable, our findings that microfilament and microtubule binding chemotherapeutics rapidly increase the mechanisms that promote endothelial adhesion of circulating tumor cells warrant caution to avoid inadvertently enhancing metastatic potential, while targeting cell division.


Subject(s)
Antimitotic Agents/adverse effects , Breast Neoplasms/metabolism , Breast Neoplasms/ultrastructure , Neoplastic Cells, Circulating/drug effects , Cell Adhesion/drug effects , Cell Line, Tumor , Depsipeptides/adverse effects , Female , Fluorescent Antibody Technique , Humans , Immunoblotting , Microscopy, Confocal , Microtubules/drug effects , Paclitaxel/adverse effects
10.
Cancer Biol Ther ; 9(1): 66-76, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20009567

ABSTRACT

The centrosome is the major organelle responsible for the nucleation and organization of microtubules into arrays. Recent studies demonstrate that microtubules can nucleate outside the centrosome. The molecular mechanisms controlling acentrosomal microtubule nucleation are currently poorly defined, and the function of this type of microtubule regulation in tumor cell biology is particularly unclear. Since microtubule nucleation is initiated by the gamma-tubulin protein, we examined the regulation of gamma-tubulin in a panel of human breast tumor cell lines, ranging from non-tumorigenic to highly aggressive. We have identified a more dispersive subcellular localization of gamma-tubulin in aggressive breast cancer cell lines, while gamma-tubulin localization remains largely centrosomal in non-aggressive cell lines. Delocalization of gamma-tubulin occurs independently from changes in protein expression and is therefore regulated at the post-translational level. Subcellular fractionation revealed that tumor cell lines show an aberrantly increased release of gamma-tubulin into a soluble cytoplasmic fraction, with the most dramatic changes observed in tumor cell lines of greater aggressiveness. Extraction of soluble gamma-tubulin revealed acentrosomal incorporation of gamma-tubulin in cytoplasmic microtubules and along cell junctions. Moreover, acentrosomal delocalization of gamma-tubulin yielded resistance to colchicine-mediated microtubule collapse. These findings support a model where the solubility of gamma-tubulin can be altered through post-translational modification and provides a new mechanism for microtubule dysregulation in breast cancer. Gamma-tubulin that is delocalized from the centrosome can still clearly be incorporated into filaments, and defines a novel mechanism for tumor cells to develop resistance to microtubule-targeted chemotherapies.


Subject(s)
Protein Processing, Post-Translational , Tubulin/metabolism , Benzimidazoles/metabolism , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Cellular Structures/metabolism , Centrosome/metabolism , Cytosol/metabolism , Female , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes/metabolism , Humans , Microtubules/metabolism , Organelles/metabolism , Solubility , Subcellular Fractions/metabolism
11.
Cancer Res ; 68(14): 5678-88, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18632620

ABSTRACT

Solid tumor metastasis often involves detachment of epithelial carcinoma cells into the vasculature or lymphatics. However, most studies of cytoskeletal rearrangement in solid tumors focus on attached cells. In this study, we report for the first time that human breast tumor cells produce unique tubulin-based protrusions when detached from extracellular matrix. Tumor cell lines of high metastatic potential show significantly increased extension and frequency of microtubule protrusions, which we have termed tubulin microtentacles. Our previous studies in nontumorigenic mammary epithelial cells showed that such detachment-induced microtentacles are enriched in detyrosinated alpha-tubulin. However, amounts of detyrosinated tubulin were similar in breast tumor cell lines despite varying microtentacle levels. Because detyrosinated alpha-tubulin associates strongly with intermediate filament proteins, we examined the contribution of cytokeratin and vimentin filaments to tumor cell microtentacles. Increased microtentacle frequency and extension correlated strongly with loss of cytokeratin expression and up-regulation of vimentin, as is often observed during tumor progression. Moreover, vimentin filaments coaligned with microtentacles, whereas cytokeratin did not. Disruption of vimentin with PP1/PP2A-specific inhibitors significantly reduced microtentacles and inhibited cell reattachment to extracellular matrix. Furthermore, expression of a dominant-negative vimentin mutant disrupted endogenous vimentin filaments and significantly reduced microtentacles, providing specific genetic evidence that vimentin supports microtentacles. Our results define a novel model in which coordination of vimentin and detyrosinated microtubules provides structural support for the extensive microtentacles observed in detached tumor cells and a possible mechanism to promote successful metastatic spread.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Tubulin/metabolism , Vimentin/chemistry , Cell Adhesion , Cell Line, Tumor , Cloning, Molecular , Cytoskeleton/metabolism , Extracellular Matrix , Humans , Models, Biological , Mutation , Neoplasm Metastasis , Tubulin/chemistry , Tumor Cells, Cultured , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...