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










Publication year range
1.
Oncotarget ; 9(66): 32556-32569, 2018 Aug 24.
Article in English | MEDLINE | ID: mdl-30220965

ABSTRACT

Matrix metalloproteinases (MMPs) may play a critical role in metastatic cancers, yet multiple human clinical trials targeting MMPs have surprisingly failed. Cancer cell density changes dramatically during the early growth of a primary tumor and during the early seeding steps of secondary tumors and has been implicated in playing an important role in regulating metastasis and drug resistance. This study reveals that the expression of MMPs is tightly regulated by local tumor cell density through the synergistic signaling mechanism of Interleukin 6 (IL-6) and Interleukin 8 (IL-8) via the JAK2/STAT3 complex. Local tumor cell density also plays a role in the responsiveness of cells to matrix metalloproteinases inhibitors (MMPI), such as Batimastat, Marimastat, Bryostatin I, and Cipemastat, where different migratory phenotypes are observed in low and high cell density conditions. Cell density-dependent MMP regulation can be directly targeted by the simultaneous inhibition of IL-6 and IL-8 receptors via Tocilizumab and Reparixin to significantly decrease the expression of MMPs in mouse xenograft models and decrease effective metastasis. This study reveals a new strategy to decrease MMP expression through pharmacological intervention of the cognate receptors of IL-6 and IL-8 to decrease metastatic capacity of tumor cells.

2.
FASEB J ; 32(3): 1207-1221, 2018 03.
Article in English | MEDLINE | ID: mdl-29097501

ABSTRACT

Microtubules have long been implicated to play an integral role in metastatic disease, for which a critical step is the local invasion of tumor cells into the 3-dimensional (3D) collagen-rich stromal matrix. Here we show that cell migration of human cancer cells uses the dynamic formation of highly branched protrusions that are composed of a microtubule core surrounded by cortical actin, a cytoskeletal organization that is absent in cells on 2-dimensional (2D) substrates. Microtubule plus-end tracking protein End-binding 1 and motor protein dynein subunits light intermediate chain 2 and heavy chain 1, which do not regulate 2D migration, critically modulate 3D migration by affecting RhoA and thus regulate protrusion branching through differential assembly dynamics of microtubules. An important consequence of this observation is that the commonly used cancer drug paclitaxel is 100-fold more effective at blocking migration in a 3D matrix than on a 2D matrix. This work reveals the central role that microtubule dynamics plays in powering cell migration in a more pathologically relevant setting and suggests further testing of therapeutics targeting microtubules to mitigate migration.-Jayatilaka, H., Giri, A., Karl, M., Aifuwa, I., Trenton, N. J., Phillip, J. M., Khatau, S., Wirtz, D. EB1 and cytoplasmic dynein mediate protrusion dynamics for efficient 3-dimensional cell migration.


Subject(s)
Cell Culture Techniques/methods , Cell Movement , Cell Surface Extensions/physiology , Cytoplasmic Dyneins/metabolism , Fibrosarcoma/pathology , Microtubule-Associated Proteins/metabolism , Fibrosarcoma/metabolism , Humans , Microtubules/metabolism , Microtubules/pathology , Tumor Cells, Cultured
3.
Sci Rep ; 5: 18437, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26675084

ABSTRACT

Intratumoral heterogeneity greatly complicates the study of molecular mechanisms driving cancer progression and our ability to predict patient outcomes. Here we have developed an automated high-throughput cell-imaging platform (htCIP) that allows us to extract high-content information about individual cells, including cell morphology, molecular content and local cell density at single-cell resolution. We further develop a comprehensive visually-aided morpho-phenotyping recognition (VAMPIRE) tool to analyze irregular cellular and nuclear shapes in both 2D and 3D microenvironments. VAMPIRE analysis of ~39,000 cells from 13 previously sequenced patient-derived pancreatic cancer samples indicate that metastasized cells present significantly lower heterogeneity than primary tumor cells. We found the same morphological signature for metastasis for a cohort of 10 breast cancer cell lines. We further decipher the relative contributions to heterogeneity from cell cycle, cell-cell contact, cell stochasticity and heritable morphological variations.


Subject(s)
Computational Biology/methods , Neoplasms/pathology , Single-Cell Analysis/methods , Tumor Microenvironment , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Cell Count , Cell Line, Tumor , Cell Shape , Female , Humans , Male , Microscopy, Fluorescence , Middle Aged , Neoplasm Metastasis , Pancreatic Neoplasms/pathology , Reproducibility of Results , Young Adult
4.
Integr Biol (Camb) ; 5(3): 523-34, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23319145

ABSTRACT

Cell cycle distribution of adherent cells is typically assessed using flow cytometry, which precludes the measurements of many cell properties and their cycle phase in the same environment. Here we develop and validate a microscopy system to quantitatively analyze the cell-cycle phase of thousands of adherent cells and their associated cell properties simultaneously. This assay demonstrates that population-averaged cell phenotypes can be written as a linear combination of cell-cycle fractions and phase-dependent phenotypes. By perturbing the cell cycle through inhibition of cell-cycle regulators or changing nuclear morphology by depletion of structural proteins, our results reveal that cell cycle regulators and structural proteins can significantly interfere with each other's prima facie functions. This study introduces a high-throughput method to simultaneously measure the cell cycle and phenotypes at single-cell resolution, which reveals a complex functional interplay between the cell cycle and cell phenotypes.


Subject(s)
Cell Culture Techniques/methods , Cell Cycle , Microscopy, Fluorescence/methods , Actins/metabolism , Animals , Cell Adhesion , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cytoplasm/metabolism , DNA/metabolism , Flow Cytometry , Humans , Lamin Type A/metabolism , Mice , Models, Biological , Phenotype
5.
Sci Rep ; 3: 1087, 2013.
Article in English | MEDLINE | ID: mdl-23336069

ABSTRACT

Cells continuously sense and respond to external mechanical forces through their cytoskeleton. Here we show that only a small subset of actin fibers, those forming the perinuclear actin cap that wraps around the nucleus, form in response to low physiological mechanical stresses in adherent fibroblasts. While conventional basal stress fibers form only past a threshold shear stress of 0.5 dyn/cm(2), actin-cap fibers are formed at shear stresses 50 times lower and orders-of-magnitude faster than biochemical stimulation. This fast differential response is uniquely mediated by focal adhesion protein zyxin at low shear stress and actomyosin fibers of the actin cap. We identify additional roles for lamin A/C of the nuclear lamina and linkers of nucleus to cytoskeleton (LINC) molecules nesprin2giant and nesprin3, which anchor actin cap fibers to the nucleus. These results suggest an interconnected physical pathway for mechanotransduction, from the extracellular milieu to the nucleus.


Subject(s)
Actins/metabolism , Cell Nucleus/metabolism , Mechanotransduction, Cellular , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Actomyosin/metabolism , Animals , Cell Adhesion , Cell Line , Cells, Cultured , Cytoskeleton/metabolism , Embryo, Mammalian/cytology , Extracellular Space/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Lamin Type A/metabolism , Mice , Microscopy, Fluorescence , RNA Interference , Stress Fibers/metabolism , Stress, Mechanical , Talin/metabolism , Time Factors , Zyxin/genetics , Zyxin/metabolism
6.
Sci Rep ; 2: 555, 2012.
Article in English | MEDLINE | ID: mdl-22870384

ABSTRACT

The ability for cells to sense and adapt to different physical microenvironments plays a critical role in development, immune responses, and cancer metastasis. Here we identify a small subset of focal adhesions that terminate fibers in the actin cap, a highly ordered filamentous actin structure that is anchored to the top of the nucleus by the LINC complexes; these differ from conventional focal adhesions in morphology, subcellular organization, movements, turnover dynamics, and response to biochemical stimuli. Actin cap associated focal adhesions (ACAFAs) dominate cell mechanosensing over a wide range of matrix stiffness, an ACAFA-specific function regulated by actomyosin contractility in the actin cap, while conventional focal adhesions are restrictively involved in mechanosensing for extremely soft substrates. These results establish the perinuclear actin cap and associated ACAFAs as major mediators of cellular mechanosensing and a critical element of the physical pathway that transduce mechanical cues all the way to the nucleus.


Subject(s)
Actin Capping Proteins/metabolism , Cell Communication/physiology , Focal Adhesions/metabolism , Actinin/metabolism , Actins/metabolism , Animals , Cell Adhesion/physiology , Cell Nucleus/metabolism , Fibroblasts/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Myosin Type II/metabolism
7.
Sci Rep ; 2: 488, 2012.
Article in English | MEDLINE | ID: mdl-22761994

ABSTRACT

Cells often migrate in vivo in an extracellular matrix that is intrinsically three-dimensional (3D) and the role of actin filament architecture in 3D cell migration is less well understood. Here we show that, while recently identified linkers of nucleoskeleton to cytoskeleton (LINC) complexes play a minimal role in conventional 2D migration, they play a critical role in regulating the organization of a subset of actin filament bundles - the perinuclear actin cap - connected to the nucleus through Nesprin2giant and Nesprin3 in cells in 3D collagen I matrix. Actin cap fibers prolong the nucleus and mediate the formation of pseudopodial protrusions, which drive matrix traction and 3D cell migration. Disruption of LINC complexes disorganizes the actin cap, which impairs 3D cell migration. A simple mechanical model explains why LINC complexes and the perinuclear actin cap are essential in 3D migration by providing mechanical support to the formation of pseudopodial protrusions.


Subject(s)
Cell Movement/physiology , Cell Nucleus/metabolism , Cytoskeleton/metabolism , Actins/metabolism , Animals , Cell Movement/genetics , Cell Nucleus/genetics , Cytoskeleton/genetics , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Lamin Type A/genetics , Lamin Type A/metabolism , Mice , Mice, Knockout , Microfilament Proteins/metabolism , Multiprotein Complexes/metabolism , Phenotype , RNA Interference
8.
PLoS One ; 7(5): e36689, 2012.
Article in English | MEDLINE | ID: mdl-22574215

ABSTRACT

The actin filament cytoskeleton mediates cell motility and adhesion in somatic cells. However, whether the function and organization of the actin network are fundamentally different in pluripotent stem cells is unknown. Here we show that while conventional actin stress fibers at the basal surface of cells are present before and after onset of differentiation of mouse (mESCs) and human embryonic stem cells (hESCs), actin stress fibers of the actin cap, which wrap around the nucleus, are completely absent from undifferentiated mESCs and hESCs and their formation strongly correlates with differentiation. Similarly, the perinuclear actin cap is absent from human induced pluripotent stem cells (hiPSCs), while it is organized in the parental lung fibroblasts from which these hiPSCs are derived and in a wide range of human somatic cells, including lung, embryonic, and foreskin fibroblasts and endothelial cells. During differentiation, the formation of the actin cap follows the expression and proper localization of nuclear lamin A/C and associated linkers of nucleus and cytoskeleton (LINC) complexes at the nuclear envelope, which physically couple the actin cap to the apical surface of the nucleus. The differentiation of hESCs is accompanied by the progressive formation of a perinuclear actin cap while induced pluripotency is accompanied by the specific elimination of the actin cap, and that, through lamin A/C and LINC complexes, this actin cap is involved in progressively shaping the nucleus of hESCs undergoing differentiation. While, the localization of lamin A/C at the nuclear envelope is required for perinuclear actin cap formation, it is not sufficient to control nuclear shape.


Subject(s)
Cell Differentiation , Cell Nucleus/metabolism , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Stress Fibers/metabolism , Animals , Cell Line , Cell Nucleus Shape , Fibroblasts/cytology , Gene Expression Regulation , Humans , Lamin Type A/metabolism , Lung/cytology , Mice , Protein Transport
9.
J Cell Sci ; 124(Pt 24): 4267-85, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22193958

ABSTRACT

In several migratory cells, the microtubule-organizing center (MTOC) is repositioned between the leading edge and nucleus, creating a polarized morphology. Although our understanding of polarization has progressed as a result of various scratch-wound and cell migration studies, variations in culture conditions required for such assays have prevented a unified understanding of the intricacies of MTOC and nucleus positioning that result in cell polarization. Here, we employ a new SMRT (for sparse, monolayer, round, triangular) analysis that uses a universal coordinate system based on cell centroid to examine the pathways regulating MTOC and nuclear positions in cells plated in a variety of conditions. We find that MTOC and nucleus positioning are crucially and independently affected by cell shape and confluence; MTOC off-centering correlates with the polarization of single cells; acto-myosin contractility and microtubule dynamics are required for single-cell polarization; and end binding protein 1 and light intermediate chain 1, but not Par3 and light intermediate chain 2, are required for single-cell polarization and directional cell motility. Using various cellular geometries and conditions, we implement a systematic and reproducible approach to identify regulators of MTOC and nucleus positioning that depend on extracellular guidance cues.


Subject(s)
Cell Nucleus/physiology , Cell Polarity , Cytoplasmic Dyneins/physiology , Microtubule-Associated Proteins/physiology , Microtubule-Organizing Center/physiology , Actins/physiology , Adaptor Proteins, Signal Transducing , Animals , Cell Adhesion Molecules/physiology , Cell Cycle Proteins , Cell Movement , Cell Nucleus/ultrastructure , Cell Shape , Cells, Cultured , Cytoplasmic Dyneins/antagonists & inhibitors , Lamins/physiology , Mice , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Organizing Center/ultrastructure , Microtubules/physiology , Myosin Type II/physiology
10.
Biophys J ; 99(11): 3563-70, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21112280

ABSTRACT

Embryonic and adult fibroblasts can be returned to pluripotency by the expression of reprogramming genes. Multiple lines of evidence suggest that these human induced pluripotent stem (hiPS) cells and human embryonic stem (hES) cells are behaviorally, karyotypically, and morphologically similar. Here we sought to determine whether the physical properties of hiPS cells, including their micromechanical properties, are different from those of hES cells. To this end, we use the method of particle tracking microrheology to compare the viscoelastic properties of the cytoplasm of hES cells, hiPS cells, and the terminally differentiated parental human fibroblasts from which our hiPS cells are derived. Our results indicate that although the cytoplasm of parental fibroblasts is both viscous and elastic, the cytoplasm of hiPS cells does not exhibit any measurable elasticity and is purely viscous over a wide range of timescales. The viscous phenotype of hiPS cells is recapitulated in parental cells with disassembled actin filament network. The cytoplasm of hES cells is predominantly viscous but contains subcellular regions that are also elastic. This study supports the hypothesis that intracellular elasticity correlates with the degree of cellular differentiation and reveals significant differences in the mechanical properties of hiPS cells and hES cells. Because mechanical stimuli have been shown to mediate the precise fate of differentiating stem cells, our results support the concept that stem cell "softness" is a key feature of force-mediated differentiation of stem cells and suggest there may be subtle functional differences between force-mediated differentiation of hiPS cells and hES cells.


Subject(s)
Embryonic Stem Cells/physiology , Induced Pluripotent Stem Cells/physiology , Rheology , Actin Cytoskeleton/metabolism , Actins/metabolism , Cell Line , Diffusion , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Molecular Dynamics Simulation , Nanoparticles/chemistry , Viscosity
12.
Nucleus ; 1(4): 337-42, 2010.
Article in English | MEDLINE | ID: mdl-21327082

ABSTRACT

We recently demonstrated the existence of a previously uncharacterized subset of actomyosin fibers that form the perinuclear actin cap, a cytoskeletal structure that tightly wraps around the nucleus of a wide range of somatic cells. Fibers in the actin cap are distinct from well-characterized, conventional actin fibers at the basal and dorsal surfaces of adherent cells in their subcellular location, internal organization, dynamics, ability to generate contractile forces, response to cytoskeletal pharmacological treatments, response to biochemical stimuli, regulation by components of the linkers of nucleoskeleton and cytoskeleton (LINC) complexes, and response to disease-associated mutations in LMNA, the gene that encodes for the nuclear lamin component lamin A/C. The perinuclear actin cap precisely shapes the nucleus in interphase cells. The perinuclear actin cap may also be a mediator of microenvironment mechanosensing and mechanotransduction, as well as a regulator of cell motility, polarization and differentiation.


Subject(s)
Actins/metabolism , Cell Nucleus/metabolism , Actin Cytoskeleton/metabolism , Animals , Cell Line , Cell Shape , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , Mice , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
13.
Proc Natl Acad Sci U S A ; 106(45): 19017-22, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19850871

ABSTRACT

Defects in nuclear morphology often correlate with the onset of disease, including cancer, progeria, cardiomyopathy, and muscular dystrophy. However, the mechanism by which a cell controls its nuclear shape is unknown. Here, we use adhesive micropatterned surfaces to control the overall shape of fibroblasts and find that the shape of the nucleus is tightly regulated by the underlying cell adhesion geometry. We found that this regulation occurs through a dome-like actin cap that covers the top of the nucleus. This cap is composed of contractile actin filament bundles containing phosphorylated myosin, which form a highly organized, dynamic, and oriented structure in a wide variety of cells. The perinuclear actin cap is specifically disorganized or eliminated by inhibition of actomyosin contractility and rupture of the LINC complexes, which connect the nucleus to the actin cap. The organization of this actin cap and its nuclear shape-determining function are disrupted in cells from mouse models of accelerated aging (progeria) and muscular dystrophy with distorted nuclei caused by alterations of A-type lamins. These results highlight the interplay between cell shape, nuclear shape, and cell adhesion mediated by the perinuclear actin cap.


Subject(s)
Actins/metabolism , Cell Adhesion/physiology , Cell Nucleus Shape/physiology , Cell Shape/physiology , Myosins/metabolism , Animals , Mice , Microscopy, Fluorescence , Muscular Dystrophies/pathology , Progeria/pathology
14.
Biophys J ; 95(11): 5462-75, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18790843

ABSTRACT

Laminopathies encompass a wide array of human diseases associated to scattered mutations along LMNA, a single gene encoding A-type lamins. How such genetic alterations translate to cellular defects and generate such diverse disease phenotypes remains enigmatic. Recent work has identified nuclear envelope proteins--emerin and the linker of the nucleoskeleton and cytoskeleton (LINC) complex--which connect the nuclear lamina to the cytoskeleton. Here we quantitatively examine the composition of the nuclear envelope, as well as the architecture and functions of the cytoskeleton in cells derived from two laminopathic mouse models, including Hutchinson-Gilford progeria syndrome (Lmna(L530P/L530P)) and Emery-Dreifuss muscular dystrophy (Lmna(-/-)). Cells derived from the overtly aphenotypical model of X-linked Emery-Dreifuss muscular dystrophy (Emd(-/y)) were also included. We find that the centrosome is detached from the nucleus, preventing centrosome polarization in cells under flow--defects that are mediated by the loss of emerin from the nuclear envelope. Moreover, while basal actin and focal adhesion structure are mildly affected, RhoA activation, cell-substratum adhesion, and cytoplasmic elasticity are greatly lowered, exclusively in laminopathic models in which the LINC complex is disrupted. These results indicate a new function for emerin in cell polarization and suggest that laminopathies are not directly associated with cells' inability to polarize, but rather with cytoplasmic softening and weakened adhesion mediated by the disruption of the LINC complex across the nuclear envelope.


Subject(s)
Actins/metabolism , Cell Nucleus/metabolism , Microtubules/metabolism , Muscular Dystrophy, Emery-Dreifuss/pathology , Progeria/pathology , Animals , Biomechanical Phenomena , Cell Adhesion , Cell Line , Cell Movement , Cytoplasm/metabolism , Cytoskeleton/metabolism , Disease Models, Animal , Humans , Membrane Proteins/metabolism , Mice , Muscular Dystrophy, Emery-Dreifuss/genetics , Muscular Dystrophy, Emery-Dreifuss/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Phenotype , Progeria/genetics , Progeria/metabolism , Rats , rhoA GTP-Binding Protein/metabolism
15.
Biophys J ; 93(7): 2542-52, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17631533

ABSTRACT

Lamin A/C is a major constituent of the nuclear lamina, a thin filamentous protein layer that lies beneath the nuclear envelope. Here we show that lamin A/C deficiency in mouse embryonic fibroblasts (Lmna(-/-) MEFs) diminishes the ability of these cells to polarize at the edge of a wound and significantly reduces cell migration speed into the wound. Moreover, lamin A/C deficiency induces significant separation of the microtubule organizing center (MTOC) from the nuclear envelope. Investigations using ballistic intracellular nanorheology reveal that lamin A/C deficiency also dramatically affects the micromechanical properties of the cytoplasm. Both the elasticity (stretchiness) and the viscosity (propensity of a material to flow) of the cytoplasm in Lmna(-/-) MEFs are significantly reduced. Disassembly of either the actin filament or microtubule networks in Lmna(+/+) MEFs results in decrease of cytoplasmic elasticity and viscosity down to levels found in Lmna(-/-) MEFs. Together these results show that both the mechanical properties of the cytoskeleton and cytoskeleton-based processes, including cell motility, coupled MTOC and nucleus dynamics, and cell polarization, depend critically on the integrity of the nuclear lamina, which suggest the existence of a functional mechanical connection between the nucleus and the cytoskeleton. These results also suggest that cell polarization during cell migration requires tight mechanical coupling between MTOC and nucleus, which is mediated by lamin A/C.


Subject(s)
Lamin Type A/chemistry , Lamin Type A/deficiency , Animals , Cell Movement , Cell Nucleus/metabolism , Cytoskeleton/metabolism , Fibroblasts/metabolism , Mice , Mice, Transgenic , Microtubules/metabolism , Nanoparticles , Nuclear Lamina/metabolism , Rheology/methods , Stress, Mechanical , Wound Healing
16.
J Cell Sci ; 119(Pt 9): 1760-8, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16636071

ABSTRACT

Cells in vivo are constantly subjected to mechanical shear stresses that play important regulatory roles in various physiological and pathological processes. Cytoskeletal reorganizations that occur in response to shear flow have been studied extensively, but whether the cytoplasm of an adherent cell adapts its mechanical properties to respond to shear is largely unknown. Here we develop a new method where fluorescent nanoparticles are ballistically injected into the cells to probe, with high resolution, possible local viscoelastic changes in the cytoplasm of individual cells subjected to fluid flow. This new assay, ballistic intracellular nanorheology (BIN), reveals that shear flow induces a dramatic sustained 25-fold increase in cytoplasmic viscosity in serum-starved Swiss 3T3 fibroblasts. By contrast, cells stimulated with the actin contractile agonist LPA show highly transient stiffening of much lower amplitude, despite the formation of similar cytoskeletal structures. Shear-induced cytoplasmic stiffening is attenuated by inhibiting actomyosin interactions and is entirely eliminated by specific Rho-kinase (ROCK) inhibition. Together, these results show that biochemical and biophysical stimuli may elicit the formation of qualitatively similar cytoskeleton structures (i.e. stress fibers and focal adhesions), but induces quantitatively different micromechanical responses. Our results suggest that when an adherent cell is subjected to shear stresses, its first order of action is to prevent detachment from its substratum by greatly stiffening its cytoplasm through enhanced actin assembly and Rho-kinase mediated contractility.


Subject(s)
Cytoskeleton/metabolism , Cytosol , Fluorescent Dyes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nanotechnology/methods , Protein Serine-Threonine Kinases/metabolism , Rheology/methods , 3T3 Cells , Actins/metabolism , Animals , Cell Adhesion/physiology , Cytosol/chemistry , Cytosol/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lysophospholipids/metabolism , Mice , Microinjections/instrumentation , Microinjections/methods , Myosins/metabolism , Nanoparticles , Nanotechnology/instrumentation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rheology/instrumentation , Shear Strength , Signal Transduction/physiology , Stress, Mechanical , rho-Associated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...