Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 18(1): 220-236, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36525964

ABSTRACT

Titin-truncating variants (TTNtv) are the single largest genetic cause of dilated cardiomyopathy (DCM). In this study we modeled disease phenotypes of A-band TTNtv-induced DCM in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) using genome editing and tissue engineering technologies. Transcriptomic, cellular, and micro-tissue studies revealed that A-band TTNtv hiPSC-CMs exhibit pathogenic proteinopathy, sarcomere defects, aberrant Na+ channel activities, and contractile dysfunction. These phenotypes establish a dual mechanism of poison peptide effect and haploinsufficiency that collectively contribute to DCM pathogenesis. However, TTNtv cellular defects did not interfere with the function of the core contractile machinery, the actin-myosin-troponin-Ca2+ complex, and preserved the therapeutic mechanism of sarcomere modulators. Treatment of TTNtv cardiac micro-tissues with investigational sarcomere modulators augmented contractility and resulted in sustained transcriptomic changes that promote reversal of DCM disease signatures. Together, our findings elucidate the underlying pathogenic mechanisms of A-band TTNtv-induced DCM and demonstrate the validity of sarcomere modulators as potential therapeutics.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Humans , Myocytes, Cardiac/pathology , Sarcomeres , Induced Pluripotent Stem Cells/pathology , Connectin/genetics , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Myocardial Contraction
2.
Pharmacol Res ; 125(Pt B): 258-265, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28918173

ABSTRACT

Within the short few years since the report of its application in precise genome editing, CRISPR technology has become the method of choice to modify and modulate gene expression in biomedical research and therapeutic development. Subsequently, a variety of research, diagnostic, and therapeutic tools have been developed based upon CRISPR's mechanism of action. Such tools have helped to deepen the understanding of fundamental biology and broaden the horizon in the search for treatments for diseases that have been considered hard or impossible to cure. As CRISPR technology advances closer to clinical applications, its short comings are becoming more apparent, thus creating opportunities to improve the technology's efficacy, specificity, and safety profile in this setting. We will summarize the current status of CRISPR technology and discuss its future impact in this review.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Drug Discovery , Animals , Biomedical Research , Humans
3.
Autophagy ; 12(4): 713-26, 2016.
Article in English | MEDLINE | ID: mdl-27050463

ABSTRACT

Autophagy is the primary process for recycling cellular constituents through lysosomal degradation. In addition to nonselective autophagic engulfment of cytoplasm, autophagosomes can recognize specific cargo by interacting with ubiquitin-binding autophagy receptors such as SQSTM1/p62 (sequestosome 1). This selective form of autophagy is important for degrading aggregation-prone proteins prominent in many neurodegenerative diseases. We carried out a high content image-based siRNA screen (4 to 8 siRNA per gene) for modulators of autophagic flux by monitoring fluorescence of GFP-SQSTM1 as well as colocalization of GFP-SQSTM1 with LAMP2 (lysosomal-associated membrane protein 2)-positive lysosomal vesicles. GFP-SQSTM1 and LAMP2 phenotypes of primary screen hits were confirmed in 2 cell types and profiled with image-based viability and MTOR signaling assays. Common seed analysis guided siRNA selection for these assays to reduce bias toward off-target effects. Confirmed hits were further validated in a live-cell assay to monitor fusion of autophagosomes with lysosomes. Knockdown of 10 targets resulted in phenotypic profiles across multiple assays that were consistent with upregulation of autophagic flux. These hits include modulators of transcription, lysine acetylation, and ubiquitination. Two targets, KAT8 (K[lysine] acetyltransferase 8) and CSNK1A1 (casein kinase 1, α 1), have been implicated in autophagic regulatory feedback loops. We confirmed that CSNK1A1 knockout (KO) cell lines have accelerated turnover of long-lived proteins labeled with (14)C-leucine in a pulse-chase assay as additional validation of our screening assays. Data from this comprehensive autophagy screen point toward novel regulatory pathways that might yield new therapeutic targets for neurodegeneration.


Subject(s)
Autophagy , High-Throughput Screening Assays/methods , Imaging, Three-Dimensional , RNA, Small Interfering/metabolism , Animals , Cell Line, Tumor , Cell Survival , Gene Knockdown Techniques , Green Fluorescent Proteins/metabolism , Humans , Reproducibility of Results
4.
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
5.
Nat Protoc ; 7(1): 155-70, 2012 Jan 05.
Article in English | MEDLINE | ID: mdl-22222790

ABSTRACT

High-throughput ballistic injection nanorheology is a method for the quantitative study of cell mechanics. Cell mechanics are measured by ballistic injection of submicron particles into the cytoplasm of living cells and tracking the spontaneous displacement of the particles at high spatial resolution. The trajectories of the cytoplasm-embedded particles are transformed into mean-squared displacements, which are subsequently transformed into frequency-dependent viscoelastic moduli and time-dependent creep compliance of the cytoplasm. This method allows for the study of a wide range of cellular conditions, including cells inside a 3D matrix, cell subjected to shear flows and biochemical stimuli, and cells in a live animal. Ballistic injection lasts <1 min and is followed by overnight incubation. Multiple particle tracking for one cell lasts <1 min. Forty cells can be examined in <1 h.


Subject(s)
Caenorhabditis elegans/cytology , Nanotechnology/methods , Rheology/methods , Animals , Biomechanical Phenomena , Cells, Cultured , Humans , Mice , Nanoparticles/analysis , Swiss 3T3 Cells , Viscoelastic Substances
6.
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
7.
Biophys J ; 101(8): 1880-6, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-22004741

ABSTRACT

The organization of chromatin in the cell nucleus is crucial for gene expression regulation. However, physically probing the nuclear interior is challenging because high forces have to be applied using minimally invasive techniques. Here, magnetic nanorods embedded in the nucleus of living cells are subjected to controlled rotational forces, producing micron-sized displacements in the nuclear interior. The resulting time-dependent rotation of the nanorods is analyzed in terms of viscoelastic parameters of the nucleus, in wild-type and Lamin A/C deficient cells. This method and analysis reveal that Lamin A/C knockout, together perhaps with other changes that result from the knockout, induce significant decreases in the nuclear viscosity and elasticity.


Subject(s)
Cell Nucleus , Magnetic Fields , Nanotubes , Animals , Biomechanical Phenomena , Cell Line , Cell Nucleus/metabolism , Cell Survival , Chromatin/metabolism , Elasticity , Gene Knockout Techniques , Lamin Type A/deficiency , Lamin Type A/genetics , Mice , Rotation , Viscosity
8.
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
9.
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
10.
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
11.
PLoS One ; 4(9): e7054, 2009 Sep 16.
Article in English | MEDLINE | ID: mdl-19756147

ABSTRACT

Einstein's original description of Brownian motion established a direct relationship between thermally-excited random forces and the transport properties of a submicron particle in a viscous liquid. Recent work based on reconstituted actin filament networks suggests that nonthermal forces driven by the motor protein myosin II can induce large non-equilibrium fluctuations that dominate the motion of particles in cytoskeletal networks. Here, using high-resolution particle tracking, we find that thermal forces, not myosin-induced fluctuating forces, drive the motion of submicron particles embedded in the cytoskeleton of living cells. These results resolve the roles of myosin II and contractile actomyosin structures in the motion of nanoparticles lodged in the cytoplasm, reveal the biphasic mechanical architecture of adherent cells-stiff contractile stress fibers interdigitating in a network at the cell cortex and a soft actin meshwork in the body of the cell, validate the method of particle tracking-microrheology, and reconcile seemingly disparate atomic force microscopy (AFM) and particle-tracking microrheology measurements of living cells.


Subject(s)
Actomyosin/chemistry , Actomyosin/physiology , Myosin Type II/physiology , 3T3 Cells , Actins/chemistry , Animals , Cytoplasm/metabolism , Cytoskeleton/metabolism , Endocytosis , Mice , Microscopy, Atomic Force/methods , Microscopy, Fluorescence/methods , Muscle Contraction , Nanoparticles/chemistry , Rheology , Stress Fibers/metabolism
12.
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
13.
Exp Cell Res ; 314(8): 1892-905, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18396275

ABSTRACT

The evolutionary-conserved interactions between KASH and SUN domain-containing proteins within the perinuclear space establish physical connections, called LINC complexes, between the nucleus and the cytoskeleton. Here, we show that the KASH domains of Nesprins 1, 2 and 3 interact promiscuously with luminal domains of Sun1 and Sun2. These constructs disrupt endogenous LINC complexes as indicated by the displacement of endogenous Nesprins from the nuclear envelope. We also provide evidence that KASH domains most probably fit a pocket provided by SUN domains and that post-translational modifications are dispensable for that interaction. We demonstrate that the disruption of endogenous LINC complexes affect cellular mechanical stiffness to an extent that compares to the loss of mechanical stiffness previously reported in embryonic fibroblasts derived from mouse lacking A-type lamins, a mouse model of muscular dystrophies and cardiomyopathies. These findings support a model whereby physical connections between the nucleus and the cytoskeleton are mediated by interactions between diverse combinations of Sun proteins and Nesprins through their respective evolutionary-conserved domains. Furthermore, they emphasize, for the first time, the relevance of LINC complexes in cellular mechanical stiffness suggesting a possible involvement of their disruption in various laminopathies, a group of human diseases linked to mutations of A-type lamins.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Membrane Proteins/chemistry , Microfilament Proteins/chemistry , Microtubule-Associated Proteins/chemistry , Nerve Tissue Proteins/chemistry , Nuclear Proteins/chemistry , Amino Acid Sequence , Animals , Binding Sites , Biomechanical Phenomena , Cell Line , Cytoskeletal Proteins , Cytoskeleton/physiology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Nuclear Lamina/chemistry , Nuclear Lamina/physiology , Nuclear Proteins/metabolism , Protein Structure, Tertiary
14.
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
15.
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
...