Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Mol Biol Cell ; 30(26): 3104-3111, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31693433

ABSTRACT

Recent studies indicate that adherent cells are keenly sensitive to external physical environment, such as substrate rigidity and topography, and internal physical states, such as cell shape and spreading area. Many of these responses are believed to involve coupled output and input of mechanical forces, which may constitute the key sensing mechanism to generate downstream regulatory signals for cell growth and differentiation. Here, we show that the state of cell migration also plays a regulatory role. Compared with migrating cells, stationary cells generate stronger, less dynamic, and more peripherally localized traction forces. These changes are coupled to reduced focal adhesion turnover and enhanced paxillin phosphorylation. Further, using cells migrating along checkerboard micropatterns, we show that the appearance of new focal adhesions directly in front of existing focal adhesions is associated with the down-regulation of existing focal adhesions and associated traction forces. Together, our results imply a mechanism where cell migration regulates traction forces by promoting dynamic turnover of focal adhesions, which may then regulate processes such as wound healing and embryogenesis where cell differentiation must coordinate with migration state and proper localization.


Subject(s)
Biomechanical Phenomena/physiology , Cell Movement/physiology , Focal Adhesions/physiology , 3T3 Cells , Animals , Cell Adhesion/physiology , Cell Line , Cell Shape/physiology , Mice , Paxillin/metabolism , Phosphorylation
2.
Nat Commun ; 6: 8129, 2015 Sep 09.
Article in English | MEDLINE | ID: mdl-26350361

ABSTRACT

It remains extremely challenging to dissect the cooperative influence of multiple extracellular matrix (ECM) parameters on cell behaviour. This stems in part from a lack of easily deployable strategies for the combinatorial variation of matrix biochemical and biophysical properties. Here we describe a simple, high-throughput platform based on light-modulated hyaluronic acid hydrogels that enables imposition of mutually independent and spatially continuous gradients of ligand density and substrate stiffness. We validate this system by showing that it can support mechanosensitive differentiation of mesenchymal stem cells. We also use it to show that the oncogenic microRNA, miR18a, is nonlinearly regulated by matrix stiffness and fibronectin density in glioma cells. The parallelization of experiments enabled by this platform allows condensation of studies that would normally require hundreds of independent hydrogels to a single substrate. This system is a highly accessible, high-throughput technique to study the combinatorial variation of biophysical and biochemical signals in a single experimental paradigm.


Subject(s)
Extracellular Matrix/metabolism , Fibronectins/metabolism , Glioma/genetics , High-Throughput Screening Assays/instrumentation , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Mesenchymal Stem Cells/metabolism , Methacrylates/chemistry , MicroRNAs/genetics , Adipose Tissue/cytology , Cell Differentiation , Cell Line, Tumor , Glioma/metabolism , High-Throughput Screening Assays/methods , Humans , Mesenchymal Stem Cells/cytology , Microscopy, Phase-Contrast , Reverse Transcriptase Polymerase Chain Reaction
3.
J Cell Biol ; 210(4): 583-94, 2015 Aug 17.
Article in English | MEDLINE | ID: mdl-26261182

ABSTRACT

Non-muscle myosin II (NMII) is reported to play multiple roles during cell migration and invasion. However, the exact biophysical roles of different NMII isoforms during these processes remain poorly understood. We analyzed the contributions of NMIIA and NMIIB in three-dimensional (3D) migration and in generating the forces required for efficient invasion by mammary gland carcinoma cells. Using traction force microscopy and microfluidic invasion devices, we demonstrated that NMIIA is critical for generating force during active protrusion, and NMIIB plays a major role in applying force on the nucleus to facilitate nuclear translocation through tight spaces. We further demonstrate that the nuclear membrane protein nesprin-2 is a possible linker coupling NMIIB-based force generation to nuclear translocation. Together, these data reveal a central biophysical role for NMIIB in nuclear translocation during 3D invasive migration, a result with relevance not only to cancer metastasis but for 3D migration in other settings such as embryonic cell migration and wound healing.


Subject(s)
Cell Movement , Cell Nucleus/physiology , Nonmuscle Myosin Type IIB/physiology , Active Transport, Cell Nucleus , Animals , Biomechanical Phenomena , Cell Line, Tumor , Mice , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism
4.
Adv Drug Deliv Rev ; 79-80: 172-83, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25174308

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and deadly brain tumor, with a mean survival time of only 21months. Despite the dramatic improvements in our understanding of GBM fueled by recent revolutions in molecular and systems biology, treatment advances for GBM have progressed inadequately slowly, which is due in part to the wide cellular and molecular heterogeneity both across tumors and within a single tumor. Thus, there is increasing clinical interest in targeting cell-extrinsic factors as way of slowing or halting the progression of GBM. These cell-extrinsic factors, collectively termed the microenvironment, include the extracellular matrix, blood vessels, stromal cells that surround tumor cells, and all associated soluble and scaffold-bound signals. In this review, we will first describe the regulation of GBM tumors by these microenvironmental factors. Next, we will discuss the various in vitro approaches that have been exploited to recapitulate and model the GBM tumor microenvironment in vitro. We conclude by identifying future challenges and opportunities in this field, including the development of microenvironmental platforms amenable to high-throughput discovery and screening. We anticipate that these ongoing efforts will prove to be valuable both as enabling tools for accelerating our understanding of microenvironmental regulation in GBM and as foundations for next-generation molecular screening platforms that may serve as a conceptual bridge between traditional reductionist systems and animal or clinical studies.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Tumor Microenvironment/physiology , Animals , Extracellular Matrix/metabolism , High-Throughput Screening Assays/methods , Humans , Models, Biological , Molecular Biology/methods , Survival Rate , Systems Biology/methods , Tissue Engineering/methods
5.
PLoS One ; 9(7): e101771, 2014.
Article in English | MEDLINE | ID: mdl-25000176

ABSTRACT

The aggressive and rapidly lethal brain tumor glioblastoma (GBM) is associated with profound tissue stiffening and genomic lesions in key members of the epidermal growth factor receptor (EGFR) pathway. Previous studies from our laboratory have shown that increasing microenvironmental stiffness in culture can strongly enhance glioma cell behaviors relevant to tumor progression, including proliferation, yet it has remained unclear whether stiffness and EGFR regulate proliferation through common or independent signaling mechanisms. Here we test the hypothesis that microenvironmental stiffness regulates cell cycle progression and proliferation in GBM tumor cells by altering EGFR-dependent signaling. We began by performing an unbiased reverse phase protein array screen, which revealed that stiffness modulates expression and phosphorylation of a broad range of signals relevant to proliferation, including members of the EGFR pathway. We subsequently found that culturing human GBM tumor cells on progressively stiffer culture substrates both dramatically increases proliferation and facilitates passage through the G1/S checkpoint of the cell cycle, consistent with an EGFR-dependent process. Western Blots showed that increasing microenvironmental stiffness enhances the expression and phosphorylation of EGFR and its downstream effector Akt. Pharmacological loss-of-function studies revealed that the stiffness-sensitivity of proliferation is strongly blunted by inhibition of EGFR, Akt, or PI3 kinase. Finally, we observed that stiffness strongly regulates EGFR clustering, with phosphorylated EGFR condensing into vinculin-positive focal adhesions on stiff substrates and dispersing as microenvironmental stiffness falls to physiological levels. Our findings collectively support a model in which tissue stiffening promotes GBM proliferation by spatially and biochemically amplifying EGFR signaling.


Subject(s)
ErbB Receptors/metabolism , Glioblastoma/pathology , Mechanical Phenomena , Signal Transduction , Tumor Microenvironment , Biomechanical Phenomena , Cell Proliferation , Focal Adhesions/metabolism , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Humans , Mechanotransduction, Cellular , Nitrogen/metabolism , Phosphorylation , Protein Transport , S Phase Cell Cycle Checkpoints
6.
Biomaterials ; 35(31): 8846-8853, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25047626

ABSTRACT

Glioblastoma multiforme (GBM), the most prevalent primary brain cancer, is characterized by diffuse infiltration of tumor cells into brain tissue, which severely complicates surgical resection and contributes to tumor recurrence. The most rapid mode of tissue infiltration occurs along blood vessels or white matter tracts, which represent topological interfaces thought to serve as "tracks" that speed cell migration. Despite this observation, the field lacks experimental paradigms that capture key features of these tissue interfaces and allow reductionist dissection of mechanisms of this interfacial motility. To address this need, we developed a culture system in which tumor cells are sandwiched between a fibronectin-coated ventral surface representing vascular basement membrane and a dorsal hyaluronic acid (HA) surface representing brain parenchyma. We find that inclusion of the dorsal HA surface induces formation of adhesive complexes and significantly slows cell migration relative to a free fibronectin-coated surface. This retardation is amplified by inclusion of integrin binding peptides in the dorsal layer and expression of CD44, suggesting that the dorsal surface slows migration through biochemically specific mechanisms rather than simple steric hindrance. Moreover, both the reduction in migration speed and assembly of dorsal adhesions depend on myosin activation and the stiffness of the ventral layer, implying that mechanochemical feedback directed by the ventral layer can influence adhesive signaling at the dorsal surface.


Subject(s)
Brain Neoplasms/pathology , Cell Culture Techniques/methods , Cell Movement , Coated Materials, Biocompatible/metabolism , Glioblastoma/pathology , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Acrylic Resins/chemistry , Acrylic Resins/metabolism , Brain/metabolism , Brain/pathology , Brain Neoplasms/metabolism , Cell Adhesion , Cell Line , Coated Materials, Biocompatible/chemistry , Glioblastoma/metabolism , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Integrins/metabolism , Myosin Type II/metabolism
7.
Methods Mol Biol ; 1066: 147-56, 2013.
Article in English | MEDLINE | ID: mdl-23955741

ABSTRACT

Cell shape and substrate rigidity play critical roles in regulating cell behaviors and fate. Controlling cell shape on elastic adhesive materials holds great promise for creating a physiologically relevant culture environment for basic and translational research and clinical applications. However, it has been technically challenging to create high-quality adhesive patterns on compliant substrates. We have developed an efficient and economical method to create precise micron-scaled adhesive patterns on the surface of a hydrogel (Rape et al., Biomaterials 32:2043-2051, 2011). This method will facilitate the research on traction force generation, cellular mechanotransduction, and tissue engineering, where precise controls of both materials rigidity and adhesive patterns are important.


Subject(s)
Acrylic Resins/metabolism , Cell Adhesion/physiology , Cell Communication/physiology , Hydrogels/metabolism , 3T3 Cells , Animals , Cell Line , Cell Movement/physiology , Mechanotransduction, Cellular/physiology , Mice , Stress, Mechanical
8.
Integr Biol (Camb) ; 5(3): 569-77, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23370891

ABSTRACT

Cell migration through tight interstitial spaces in three dimensional (3D) environments impacts development, wound healing and cancer metastasis and is altered by the aging process. The stiffness of the extracellular matrix (ECM) increases with aging and affects the cells and cytoskeletal processes involved in cell migration. However, the nucleus, which is the largest and densest organelle, has not been widely studied during cell migration through the ECM. Additionally, the nucleus is stiffened during the aging process through the accumulation of a mutant nucleoskeleton protein lamin A, progerin. By using microfabricated substrates to mimic the confined environment of surrounding tissues, we characterized nuclear movements and deformation during cell migration into micropillars where interspacing can be tuned to vary nuclear confinement. Cell motility decreased with decreased micropillar (µP) spacing and correlated with increased dysmorphic shapes of nuclei. We examined the effects of increased nuclear stiffness which correlates with cellular aging by studying Hutchinson-Gilford progeria syndrome cells which are known to accumulate progerin. With the expression of progerin, cells showed a threshold response to decreased µP spacing. Cells became trapped in the close spacing, possibly from visible micro-defects in the nucleoskeleton induced by cell crawling through the µP and from reduced force generation, measured independently. We suggest that ECM changes during aging could be compounded by the increasing stiffness of the nucleus and thus changes in cell migration through 3D tissues.


Subject(s)
Cell Movement , Cell Nucleus/metabolism , Progeria/physiopathology , Actins/metabolism , Animals , Extracellular Matrix/metabolism , Humans , Imaging, Three-Dimensional , Lamin Type A/metabolism , Mice , Models, Biological , NIH 3T3 Cells , Neoplasm Metastasis , Progeria/metabolism , Time Factors , Wound Healing
9.
J Funct Biomater ; 3(2): 398-417, 2012 May 23.
Article in English | MEDLINE | ID: mdl-24955540

ABSTRACT

With a range of desirable mechanical and optical properties, single wall carbon nanotubes (SWCNTs) are a promising material for nanobiotechnologies. SWCNTs also have potential as biomaterials for modulation of cellular structures. Previously, we showed that highly purified, dispersed SWCNTs grossly alter F-actin inside cells. F-actin plays critical roles in the maintenance of cell structure, force transduction, transport and cytokinesis. Thus, quantification of SWCNT-actin interactions ranging from molecular, sub-cellular and cellular levels with both structure and function is critical for developing SWCNT-based biotechnologies. Further, this interaction can be exploited, using SWCNTs as a unique actin-altering material. Here, we utilized molecular dynamics simulations to explore the interactions of SWCNTs with actin filaments. Fluorescence lifetime imaging microscopy confirmed that SWCNTs were located within ~5 nm of F-actin in cells but did not interact with G-actin. SWCNTs did not alter myosin II sub-cellular localization, and SWCNT treatment in cells led to significantly shorter actin filaments. Functionally, cells with internalized SWCNTs had greatly reduced cell traction force. Combined, these results demonstrate direct, specific SWCNT alteration of F-actin structures which can be exploited for SWCNT-based biotechnologies and utilized as a new method to probe fundamental actin-related cellular processes and biophysics.

10.
J Cell Sci ; 124(Pt 24): 4233-40, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22193960

ABSTRACT

Traction forces increase after microtubule depolymerization; however, the signaling mechanisms underlying this, in particular the dependence upon myosin II, remain unclear. We investigated the mechanism of traction force increase after nocodazole-induced microtubule depolymerization by applying traction force microscopy to cells cultured on micropatterned polyacrylamide hydrogels to obtain samples of homogeneous shape and size. Control cells and cells treated with a focal adhesion kinase (FAK) inhibitor showed similar increases in traction forces, indicating that the response is independent of FAK. Surprisingly, pharmacological inhibition of myosin II did not prevent the increase of residual traction forces upon nocodazole treatment. This increase was abolished upon pharmacological inhibition of FAK. These results suggest two distinct pathways for the regulation of traction forces. First, microtubule depolymerization activates a myosin-II-dependent mechanism through a FAK-independent pathway. Second, microtubule depolymerization also enhances traction forces through a myosin-II-independent, FAK-regulated pathway. Traction forces are therefore regulated by a complex network of complementary signals and force-generating mechanisms.


Subject(s)
Microtubules/physiology , Signal Transduction , Animals , Cell Shape/drug effects , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions , Mice , Microtubules/drug effects , Myosin Type II/antagonists & inhibitors , NIH 3T3 Cells , Nocodazole/pharmacology , Tubulin Modulators/pharmacology
11.
Biomaterials ; 32(8): 2043-51, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21163521

ABSTRACT

Mechanical forces provide critical inputs for proper cellular functions. The interplay between the generation of, and response to, mechanical forces regulate such cellular processes as differentiation, proliferation, and migration. We postulate that adherent cells respond to a number of physical and topographical factors, including cell size and shape, by detecting the magnitude and/or distribution of traction forces under different conditions. To address this possibility we introduce a new simple method for precise micropatterning of hydrogels, and then apply the technique to systematically investigate the relationship between cell geometry, focal adhesions, and traction forces in cells with a series of spread areas and aspect ratios. Contrary to previous findings, we find that traction force is not determined primarily by the cell spreading area but by the distance from cell center to the perimeter. This distance in turn controls traction forces by regulating the size of focal adhesions, such that constraining the size of focal adhesions by micropatterning can override the effect of geometry. We propose that the responses of traction forces to center-periphery distance, possibly through a positive feedback mechanism that regulates focal adhesions, provide the cell with the information on its own shape and size. A similar positive feedback control may allow cells to respond to a variety of physical or topographical signals via a unified mechanism.


Subject(s)
Cell Shape/physiology , Focal Adhesions/physiology , Hydrogels/chemistry , Stress, Mechanical , Acrylic Resins/chemistry , Animals , Cell Adhesion/physiology , Cell Culture Techniques , Cells, Cultured , Mice , NIH 3T3 Cells
12.
ACS Nano ; 4(8): 4872-8, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20669976

ABSTRACT

The ability of globular actin to form filaments and higher-order network structures of the cytoskeleton is essential for cells to maintain their shape and perform essential functions such as force generation, motility, and division. Alterations of actin structures can dramatically change a cell's ability to function. We found that purified and dispersed single wall carbon nanotubes (SWCNTs) can induce actin bundling in cells and in purified model actin systems. SWCNTs do not induce acute cell death, but cell proliferation is greatly reduced in SWCNT-treated cells with an increase in actin-related division defects. Actin, normally present in basal stress fibers in control cells, is located in heterogeneous structures throughout the SWCNT-treated cell. These SWCNT-induced changes in actin structures are seen functionally in multinucleated cells and with reduced force generation. Ex vivo, purified actin filaments cross-linked with alpha-actinin and formed isotropic networks, whereas SWCNTs caused purified actin filaments to assemble into bundles. While purified, isolated SWCNTs do not appear acutely toxic, this subcellular reorganization may cause chronic changes to cellular functions.


Subject(s)
Actins/chemistry , Actins/metabolism , Nanotubes, Carbon , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , HeLa Cells , Humans , Microscopy, Confocal , Nanotubes, Carbon/toxicity , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL