Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(13): e2314588121, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38502691

ABSTRACT

During development, motor axons are guided toward muscle target by various extrinsic cues including extracellular matrix (ECM) proteins whose identities and cellular source remain poorly characterized. Here, using single-cell RNAseq of sorted GFP+ cells from smyhc1:gfp-injected zebrafish embryos, we unravel the slow muscle progenitors (SMP) pseudotemporal trajectory at the single-cell level and show that differentiating SMPs are a major source of ECM proteins. The SMP core-matrisome was characterized and computationally predicted to form a basement membrane-like structure tailored for motor axon guidance, including basement membrane-associated ECM proteins, as collagen XV-B, one of the earliest core-matrisome gene transcribed in differentiating SMPs and the glycoprotein Tenascin C. To investigate how contact-mediated guidance cues are organized along the motor path to exert their function in vivo, we used microscopy-based methods to analyze and quantify motor axon navigation in tnc and col15a1b knock-out fish. We show that motor axon shape and growth rely on the timely expression of the attractive cue Collagen XV-B that locally provides axons with a permissive soft microenvironment and separately organizes the repulsive cue Tenascin C into a unique functional dual topology. Importantly, bioprinted micropatterns that mimic this in vivo ECM topology were sufficient to drive directional motor axon growth. Our study offers evidence that not only the composition of ECM cues but their topology critically influences motor axon navigation in vertebrates with potential applications in regenerative medicine for peripheral nerve injury as regenerating nerves follow their original path.


Subject(s)
Tenascin , Zebrafish , Animals , Tenascin/genetics , Zebrafish/genetics , Zebrafish/metabolism , Axons/metabolism , Collagen/metabolism , Extracellular Matrix/metabolism
2.
Elife ; 122023 08 07.
Article in English | MEDLINE | ID: mdl-37548995

ABSTRACT

Cell-generated forces play a major role in coordinating the large-scale behavior of cell assemblies, in particular during development, wound healing, and cancer. Mechanical signals propagate faster than biochemical signals, but can have similar effects, especially in epithelial tissues with strong cell-cell adhesion. However, a quantitative description of the transmission chain from force generation in a sender cell, force propagation across cell-cell boundaries, and the concomitant response of receiver cells is missing. For a quantitative analysis of this important situation, here we propose a minimal model system of two epithelial cells on an H-pattern ('cell doublet'). After optogenetically activating RhoA, a major regulator of cell contractility, in the sender cell, we measure the mechanical response of the receiver cell by traction force and monolayer stress microscopies. In general, we find that the receiver cells show an active response so that the cell doublet forms a coherent unit. However, force propagation and response of the receiver cell also strongly depend on the mechano-structural polarization in the cell assembly, which is controlled by cell-matrix adhesion to the adhesive micropattern. We find that the response of the receiver cell is stronger when the mechano-structural polarization axis is oriented perpendicular to the direction of force propagation, reminiscent of the Poisson effect in passive materials. We finally show that the same effects are at work in small tissues. Our work demonstrates that cellular organization and active mechanical response of a tissue are key to maintain signal strength and lead to the emergence of elasticity, which means that signals are not dissipated like in a viscous system, but can propagate over large distances.


Subject(s)
Epithelial Cells , Mechanical Phenomena , Epithelial Cells/physiology , Epithelium , Cell Adhesion/physiology , Elasticity , Stress, Mechanical
3.
Nat Commun ; 14(1): 717, 2023 02 09.
Article in English | MEDLINE | ID: mdl-36759504

ABSTRACT

The mechanical properties of biological tissues are key to their physical integrity and function. Although external loading or biochemical treatments allow the estimation of these properties globally, it remains difficult to assess how such external stimuli compare with cell-generated contractions. Here we engineer microtissues composed of optogenetically-modified fibroblasts encapsulated within collagen. Using light to control the activity of RhoA, a major regulator of cellular contractility, we induce local contractions within microtissues, while monitoring microtissue stress and strain. We investigate the regulation of these local contractions and their spatio-temporal distribution. We demonstrate the potential of our technique for quantifying tissue elasticity and strain propagation, before examining the possibility of using light to create and map local anisotropies in mechanically heterogeneous microtissues. Altogether, our results open an avenue to guide the formation of tissues while non-destructively charting their rheology in real time, using their own constituting cells as internal actuators.


Subject(s)
Collagen , Fibroblasts , Rheology , Tissue Engineering/methods
4.
J Cell Biol ; 222(1)2023 01 02.
Article in English | MEDLINE | ID: mdl-36250940

ABSTRACT

Integrin endocytosis is essential for many fundamental cellular processes. Whether and how the internalization impacts cellular mechanics remains elusive. Whereas previous studies reported the contribution of the integrin activator, talin, in force development, the involvement of inhibitors is less documented. We identified ICAP-1 as an integrin inhibitor involved in mechanotransduction by co-working with NME2 to control clathrin-mediated endocytosis of integrins at the edge of focal adhesions (FA). Loss of ICAP-1 enables ß3-integrin-mediated force generation independently of ß1 integrin. ß3-integrin-mediated forces were associated with a decrease in ß3 integrin dynamics stemming from their reduced diffusion within adhesion sites and slow turnover of FA. The decrease in ß3 integrin dynamics correlated with a defect in integrin endocytosis. ICAP-1 acts as an adaptor for clathrin-dependent endocytosis of integrins. ICAP-1 controls integrin endocytosis by interacting with NME2, a key regulator of dynamin-dependent clathrin-coated pits fission. Control of clathrin-mediated integrin endocytosis by an inhibitor is an unprecedented mechanism to tune forces at FA.


Subject(s)
Clathrin , Endocytosis , Focal Adhesions , Integrin beta1 , Integrin beta3 , Clathrin/metabolism , Endocytosis/physiology , Integrin beta1/genetics , Mechanotransduction, Cellular , Talin/genetics
5.
Nat Commun ; 13(1): 6059, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36229487

ABSTRACT

Extracellular matrix (ECM) elasticity is perceived by cells via focal adhesion structures, which transduce mechanical cues into chemical signalling to conform cell behavior. Although the contribution of ECM compliance to the control of cell migration or division is extensively studied, little is reported regarding infectious processes. We study this phenomenon with the extraintestinal Escherichia coli pathogen UTI89. We show that UTI89 takes advantage, via its CNF1 toxin, of integrin mechanoactivation to trigger its invasion into cells. We identify the HACE1 E3 ligase-interacting protein Optineurin (OPTN) as a protein regulated by ECM stiffness. Functional analysis establishes a role of OPTN in bacterial invasion and integrin mechanical coupling and for stimulation of HACE1 E3 ligase activity towards the Rac1 GTPase. Consistent with a role of OPTN in cell mechanics, OPTN knockdown cells display defective integrin-mediated traction force buildup, associated with limited cellular invasion by UTI89. Nevertheless, OPTN knockdown cells display strong mechanochemical adhesion signalling, enhanced Rac1 activation and increased cyclin D1 translation, together with enhanced cell proliferation independent of ECM stiffness. Together, our data ascribe a new function to OPTN in mechanobiology.


Subject(s)
Cyclin D1 , Integrins , Cell Division , Cyclin D1/metabolism , Integrins/metabolism , Mechanotransduction, Cellular/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , rac1 GTP-Binding Protein/metabolism
6.
Elife ; 112022 04 13.
Article in English | MEDLINE | ID: mdl-35416768

ABSTRACT

Mechanics has been a central focus of physical biology in the past decade. In comparison, how cells manage their size is less understood. Here, we show that a parameter central to both the physics and the physiology of the cell, its volume, depends on a mechano-osmotic coupling. We found that cells change their volume depending on the rate at which they change shape, when they spontaneously spread or when they are externally deformed. Cells undergo slow deformation at constant volume, while fast deformation leads to volume loss. We propose a mechanosensitive pump and leak model to explain this phenomenon. Our model and experiments suggest that volume modulation depends on the state of the actin cortex and the coupling of ion fluxes to membrane tension. This mechano-osmotic coupling defines a membrane tension homeostasis module constantly at work in cells, causing volume fluctuations associated with fast cell shape changes, with potential consequences on cellular physiology.


Subject(s)
Actins , Actins/metabolism , Cell Membrane/metabolism , Cell Shape , Cell Size , Feedback , Osmotic Pressure
7.
Cancer Gene Ther ; 29(10): 1429-1438, 2022 10.
Article in English | MEDLINE | ID: mdl-35379907

ABSTRACT

Cell migration depends on the dynamic organisation of the actin cytoskeleton and assembly and disassembly of focal adhesions (FAs). However, the precise mechanisms coordinating these processes remain poorly understood. We previously identified the oestrogen-related receptor α (ERRα) as a major regulator of cell migration. Here, we show that loss of ERRα leads to abnormal accumulation of actin filaments that is associated with an increased level of inactive form of the actin-depolymerising factor cofilin. We further show that ERRα depletion decreases cell adhesion and results in defective FA formation and turnover. Interestingly, specific inhibition of the RhoA-ROCK-LIMK-cofilin pathway rescues the actin polymerisation defects resulting from ERRα silencing, but not cell adhesion. Instead, we found that MAP4K4 is a direct target of ERRα and down-regulation of its activity rescues cell adhesion and FA formation in the ERRα-depleted cells. Altogether, our results highlight a crucial role of ERRα in coordinating the dynamic of actin network and FAs through the independent regulation of the RhoA and MAP4K4 pathways.


Subject(s)
Actins , Focal Adhesions , Actin Depolymerizing Factors/metabolism , Actins/genetics , Actins/metabolism , Cell Movement/physiology , Focal Adhesions/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , ERRalpha Estrogen-Related Receptor
8.
Dev Cell ; 56(20): 2841-2855.e8, 2021 10 25.
Article in English | MEDLINE | ID: mdl-34559979

ABSTRACT

Glioblastoma are heterogeneous tumors composed of highly invasive and highly proliferative clones. Heterogeneity in invasiveness could emerge from discrete biophysical properties linked to specific molecular expression. We identified clones of patient-derived glioma propagating cells that were either highly proliferative or highly invasive and compared their cellular architecture, migratory, and biophysical properties. We discovered that invasiveness was linked to cellular fitness. The most invasive cells were stiffer, developed higher mechanical forces on the substrate, and moved stochastically. The mechano-chemical-induced expression of the formin FMN1 conferred invasive strength that was confirmed in patient samples. Moreover, FMN1 expression was also linked to motility in other cancer and normal cell lines, and its ectopic expression increased fitness parameters. Mechanistically, FMN1 acts from the microtubule lattice and promotes a robust mechanical cohesion, leading to highly invasive motility.


Subject(s)
Cell Movement/physiology , Formins/metabolism , Glioblastoma/metabolism , Neoplasm Invasiveness/pathology , Brain Neoplasms/pathology , Cell Line, Tumor , Fetal Proteins/metabolism , Glioblastoma/pathology , Humans , Microfilament Proteins/metabolism
9.
Cell Rep ; 36(8): 109616, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34433061

ABSTRACT

Vascular development is a complex multistep process involving the coordination of cellular functions such as migration, proliferation, and differentiation. How mechanical forces generated by cells and transmission of these physical forces control vascular development is poorly understood. Using an endothelial-specific genetic model in mice, we show that deletion of the scaffold protein Angiomotin (Amot) inhibits migration and expansion of the physiological and pathological vascular network. We further show that Amot is required for tip cell migration and the extension of cellular filopodia. Exploiting in vivo and in vitro molecular approaches, we show that Amot binds Talin and is essential for relaying forces between fibronectin and the cytoskeleton. Finally, we provide evidence that Amot is an important component of the endothelial integrin adhesome and propose that Amot integrates spatial cues from the extracellular matrix to form a functional vascular network.


Subject(s)
Cytoskeleton/metabolism , Fibronectins/metabolism , Integrins/metabolism , Neovascularization, Physiologic/physiology , Angiomotins/metabolism , Animals , Cell Membrane/metabolism , Cell Movement/physiology , Endothelium/metabolism , Mice, Transgenic , Plasma Substitutes/pharmacology , Pseudopodia/metabolism
10.
J Cell Sci ; 134(2)2021 01 25.
Article in English | MEDLINE | ID: mdl-33495358

ABSTRACT

Upon activation by different transmembrane receptors, the same signaling protein can induce distinct cellular responses. A way to decipher the mechanisms of such pleiotropic signaling activity is to directly manipulate the decision-making activity that supports the selection between distinct cellular responses. We developed an optogenetic probe (optoSRC) to control SRC signaling, an example of a pleiotropic signaling node, and we demonstrated its ability to generate different acto-adhesive structures (lamellipodia or invadosomes) upon distinct spatio-temporal control of SRC kinase activity. The occurrence of each acto-adhesive structure was simply dictated by the dynamics of optoSRC nanoclusters in adhesive sites, which were dependent on the SH3 and Unique domains of the protein. The different decision-making events regulated by optoSRC dynamics induced distinct downstream signaling pathways, which we characterized using time-resolved proteomic and network analyses. Collectively, by manipulating the molecular mobility of SRC kinase activity, these experiments reveal the pleiotropy-encoding mechanism of SRC signaling.


Subject(s)
Cytoskeleton , Proteomics , Signal Transduction , src-Family Kinases , Animals , Cells, Cultured , Molecular Dynamics Simulation , Phosphorylation , src Homology Domains , src-Family Kinases/genetics , src-Family Kinases/metabolism
11.
J Vis Exp ; (161)2020 07 23.
Article in English | MEDLINE | ID: mdl-32773764

ABSTRACT

Traction force microscopy (TFM) enables the measurement of forces produced by a cell on a substrate. This technique infers traction force measurements from an experimentally observed displacement field produced by a cell pulling on an elastic substrate. Here, we adapted TFM to investigate the spatial and temporal structure of the force field exerted by B cells when activated by antigen engagement of the B cell receptor. Gel rigidity, bead density, and protein functionalization must be optimized for the study of relatively small cells (~ 6 µm) that interact with, and respond specifically to ligands for cell surface receptors.


Subject(s)
Lymphocyte Activation/physiology , Microscopy, Atomic Force/methods , Humans
12.
ACS Nano ; 14(6): 7121-7139, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32432851

ABSTRACT

Among the eukaryotic cells that navigate through fully developed metazoan tissues, protozoans from the Apicomplexa phylum have evolved motile developmental stages that move much faster than the fastest crawling cells owing to a peculiar substrate-dependent type of motility, known as gliding. Best-studied models are the Plasmodium sporozoite and the Toxoplasma tachyzoite polarized cells for which motility is vital to achieve their developmental programs in the metazoan hosts. The gliding machinery is shared between the two parasites and is largely characterized. Localized beneath the cell surface, it includes actin filaments, unconventional myosin motors housed within a multimember glideosome unit, and apically secreted transmembrane adhesins. In contrast, less is known about the force mechanisms powering cell movement. Pioneered biophysical studies on the sporozoite and phenotypic analysis of tachyzoite actin-related mutants have added complexity to the general view that force production for parasite forward movement directly results from the myosin-driven rearward motion of the actin-coupled adhesion sites. Here, we have interrogated how forces and substrate adhesion-de-adhesion cycles operate and coordinate to allow the typical left-handed helical gliding mode of the tachyzoite. By combining quantitative traction force and reflection interference microscopy with micropatterning and expansion microscopy, we unveil at the millisecond and nanometer scales the integration of a critical apical anchoring adhesion with specific traction and spring-like forces. We propose that the acto-myoA motor directs the traction force which allows transient energy storage by the microtubule cytoskeleton and therefore sets the thrust force required for T. gondii tachyzoite vital helical gliding capacity.


Subject(s)
Parasites , Toxoplasma , Actins , Animals , Protozoan Proteins , Torque , Traction
13.
Mol Biol Cell ; 31(16): 1675-1690, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32348198

ABSTRACT

During the initial stages of cell division, the cytoskeleton is extensively reorganized so that a bipolar mitotic spindle can be correctly assembled. This process occurs through the action of molecular motors, cytoskeletal networks, and the nucleus. How the combined activity of these different components is spatiotemporally regulated to ensure efficient spindle assembly remains unclear. To investigate how cell shape, cytoskeletal organization, and molecular motors cross-talk to regulate initial spindle assembly, we use a combination of micropatterning with high-resolution imaging and 3D cellular reconstruction. We show that during prophase, centrosomes and nucleus reorient so that centrosomes are positioned on the shortest nuclear axis at nuclear envelope (NE) breakdown. We also find that this orientation depends on a combination of centrosome movement controlled by Arp2/3-mediated regulation of microtubule dynamics and Dynein-generated forces on the NE that regulate nuclear reorientation. Finally, we observe this centrosome configuration favors the establishment of an initial bipolar spindle scaffold, facilitating chromosome capture and accurate segregation, without compromising division plane orientation.


Subject(s)
Centrosome/metabolism , Mitosis , Spindle Apparatus/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Cell Adhesion , Cell Shape , Dyneins/metabolism , HEK293 Cells , HeLa Cells , Humans , Movement , Nuclear Envelope/metabolism , Prophase , Rotation
14.
Trends Biotechnol ; 38(2): 142-153, 2020 02.
Article in English | MEDLINE | ID: mdl-31601426

ABSTRACT

Traditionally, cancer-like scaffolds have been developed with tissue regeneration in mind and therefore designed to mimic the regenerative environment of otherwise healthy tissues. However, the tumoral niche exhibits specific biophysical cues far from being 'cell friendly', suggesting that a different approach should be taken to design these artificial neoplastic niches. From bare 2D surfaces to 3D and 1D microstructured platforms, this opinion article focuses on evolving approaches used to mimic in vitro the neoplastic niche, discussing why this pathology cannot be assessed with tissue engineering (TE) approaches (i.e., using scaffolds facilitating cell growth, migration, and matrix degradation in the absence of diffusional restrictions, among others), and suggests how to improve them with recent lessons learned from mechanobiology and topobiology.


Subject(s)
Neoplasms/pathology , Tissue Engineering/methods , Tissue Scaffolds , Animals , Cell Movement , Extracellular Matrix Proteins/metabolism , Humans , Mechanotransduction, Cellular , Tumor Microenvironment
15.
EMBO Rep ; 20(9): e48084, 2019 09.
Article in English | MEDLINE | ID: mdl-31368207

ABSTRACT

The shape of the cell nucleus can vary considerably during developmental and pathological processes; however, the impact of nuclear morphology on cell behavior is not known. Here, we observed that the nuclear envelope flattens as cells transit from G1 to S phase and inhibition of myosin II prevents nuclear flattening and impedes progression to S phase. Strikingly, we show that applying compressive force on the nucleus in the absence of myosin II-mediated tension is sufficient to restore G1 to S transition. Using a combination of tools to manipulate nuclear morphology, we observed that nuclear flattening activates a subset of transcription factors, including TEAD and AP1, leading to transcriptional induction of target genes that promote G1 to S transition. In addition, we found that nuclear flattening mediates TEAD and AP1 activation in response to ROCK-generated contractility or cell spreading. Our results reveal that the nuclear envelope can operate as a mechanical sensor whose deformation controls cell growth in response to tension.


Subject(s)
Cell Nucleus/metabolism , Mechanotransduction, Cellular/physiology , Nuclear Envelope/metabolism , Transcription Factors/metabolism , Cell Cycle/genetics , Cell Cycle/physiology , Cell Division/genetics , Cell Division/physiology , Cell Line , Cell Nucleus/genetics , Flow Cytometry , G1 Phase/genetics , G1 Phase/physiology , HeLa Cells , Humans , Mechanotransduction, Cellular/genetics , Microscopy, Atomic Force , Nuclear Envelope/genetics , Plasmids/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , S Phase/genetics , S Phase/physiology , Transcription Factors/genetics
16.
Nat Commun ; 10(1): 2870, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31253773

ABSTRACT

An important channel of cell-to-cell communication is direct contact. The immune synapse is a paradigmatic example of such type of interaction: it forms upon engagement of antigen receptors in lymphocytes by antigen-presenting cells and allows the local exchange of molecules and information. Although mechanics has been shown to play an important role in this process, how forces organize and impact on synapse function is unknown. We find that mechanical forces are spatio-temporally patterned at the immune synapse: global pulsatile myosin II-driven tangential forces are observed at the synapse periphery while localised forces generated by invadosome-like F-actin protrusions are detected at its centre. Noticeably, we observe that these force-producing actin protrusions constitute the main site of antigen extraction and endocytosis and require myosin II contractility to form. The interplay between global and local forces dictated by the organization of the actomyosin cytoskeleton therefore controls endocytosis at the immune synapse.


Subject(s)
Actin Cytoskeleton/physiology , Actomyosin/metabolism , B-Lymphocytes/physiology , Endocytosis/physiology , Myosin Type II/metabolism , Actomyosin/genetics , Animals , Cell Communication , Crosses, Genetic , Integrases/genetics , Integrases/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Myosin Type II/genetics , Receptors, Complement 3d
17.
Phys Rev Lett ; 122(16): 168101, 2019 Apr 26.
Article in English | MEDLINE | ID: mdl-31075005

ABSTRACT

The structural and functional organization of biological tissues relies on the intricate interplay between chemical and mechanical signaling. Whereas the role of constant and transient mechanical perturbations is generally accepted, several studies recently highlighted the existence of long-range mechanical excitations (i.e., waves) at the supracellular level. Here, we confine epithelial cell monolayers to quasi-one-dimensional geometries, to force the establishment of tissue-level waves of well-defined wavelength and period. Numerical simulations based on a self-propelled Voronoi model reproduce the observed waves and exhibit a phase transition between a global and a multinodal wave, controlled by the confinement size. We confirm experimentally the existence of such a phase transition, and show that wavelength and period are independent of the confinement length. Together, these results demonstrate the intrinsic origin of tissue oscillations, which could provide cells with a mechanism to accurately measure distances at the supracellular level.


Subject(s)
Cell Movement , Models, Biological , Animals , Dogs , Fibronectins/metabolism , Madin Darby Canine Kidney Cells
19.
Exp Cell Res ; 378(1): 113-117, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30794802

ABSTRACT

Since the emergence of mechanobiology, mechanical signals have been shown to influence almost every process in biology. Cells transduce mechanical signals into biochemical signaling pathways, adjust their behavior and/or phenotype before transmitting these signals to neighboring cells. Mechanical signals thus appear as information, which can be "written" by cells in the surrounding extracellular matrix, "transmitted" through it and "read" by other cells. This brief review summarizes our current understanding of the mechanisms regulating the tensional state of cells and tissues subjected to mechanical perturbations, prior to examining existing or potential experimental approaches to study these mechanisms.


Subject(s)
Mechanoreceptors/metabolism , Mechanotransduction, Cellular , Animals , Feedback, Physiological , Focal Adhesions/metabolism , Humans , Mechanoreceptors/physiology
20.
Nat Commun ; 10(1): 52, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30604763

ABSTRACT

The presence of aberrant number of centrioles is a recognized cause of aneuploidy and hallmark of cancer. Hence, centriole duplication needs to be tightly regulated. It has been proposed that centriole separation limits centrosome duplication. The mechanism driving centriole separation is poorly understood and little is known on how this is linked to centriole duplication. Here, we propose that actin-generated forces regulate centriole separation. By imposing geometric constraints via micropatterns, we were able to prove that precise acto-myosin force arrangements control direction, distance and time of centriole separation. Accordingly, inhibition of acto-myosin contractility impairs centriole separation. Alongside, we observed that organization of acto-myosin force modulates specifically the length of S-G2 phases of the cell cycle, PLK4 recruitment at the centrosome and centriole fidelity. These discoveries led us to suggest that acto-myosin forces might act in fundamental mechanisms of aneuploidy prevention.


Subject(s)
Actins/metabolism , Cell Cycle/physiology , Centrioles/metabolism , Myosins/metabolism , Protein Serine-Threonine Kinases/metabolism , Actins/physiology , Aneuploidy , Cell Cycle/drug effects , Centrioles/physiology , HeLa Cells , Humans , Intravital Microscopy/methods , Microscopy, Confocal , Myosins/physiology , Protein Serine-Threonine Kinases/physiology , Thymidine/pharmacology , Time-Lapse Imaging/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...