Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
EMBO J ; 43(13): 2715-2732, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38769437

RESUMEN

Microtubules regulate cell polarity and migration via local activation of focal adhesion turnover, but the mechanism of this process is insufficiently understood. Molecular complexes containing KANK family proteins connect microtubules with talin, the major component of focal adhesions. Here, local optogenetic activation of KANK1-mediated microtubule/talin linkage promoted microtubule targeting to an individual focal adhesion and subsequent withdrawal, resulting in focal adhesion centripetal sliding and rapid disassembly. This sliding is preceded by a local increase of traction force due to accumulation of myosin-II and actin in the proximity of the focal adhesion. Knockdown of the Rho activator GEF-H1 prevented development of traction force and abolished sliding and disassembly of focal adhesions upon KANK1 activation. Other players participating in microtubule-driven, KANK-dependent focal adhesion disassembly include kinases ROCK, PAK, and FAK, as well as microtubules/focal adhesion-associated proteins kinesin-1, APC, and αTAT. Based on these data, we develop a mathematical model for a microtubule-driven focal adhesion disruption involving local GEF-H1/RhoA/ROCK-dependent activation of contractility, which is consistent with experimental data.


Asunto(s)
Adhesiones Focales , Cinesinas , Microtúbulos , Factores de Intercambio de Guanina Nucleótido Rho , Adhesiones Focales/metabolismo , Microtúbulos/metabolismo , Humanos , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/genética , Cinesinas/metabolismo , Cinesinas/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/genética , Miosina Tipo II/metabolismo , Talina/metabolismo , Talina/genética , Animales
2.
Phys Rev Lett ; 132(18): 188402, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38759206

RESUMEN

Cell adhesion receptors are transmembrane proteins that bind cells to their environment. These proteins typically cluster into disk-shaped or linear structures. Here, we show that such clustering patterns spontaneously emerge when the receptor senses the membrane deformation gradient, for example, by reaching a lower-energy conformation when the membrane is tilted relative to the underlying binding substrate. Increasing the strength of the membrane gradient-sensing mechanism first yields isolated disk-shaped clusters and then long linear structures. Our theory is coherent with experimental estimates of the parameters, suggesting that a tilt-induced clustering mechanism is relevant in the context of cell adhesion.


Asunto(s)
Membrana Celular , Membrana Celular/metabolismo , Modelos Biológicos , Adhesión Celular/fisiología , Separación de Fases , Complejo GPIb-IX de Glicoproteína Plaquetaria
3.
Biophys J ; 122(18): 3704-3721, 2023 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-37301982

RESUMEN

Adherent filopodia are elongated finger-like membrane protrusions, extending from the edges of diverse cell types and participating in cell adhesion, spreading, migration, and environmental sensing. The formation and elongation of filopodia are driven by the polymerization of parallel actin filaments, comprising the filopodia cytoskeletal core. Here, we report that adherent filopodia, formed during the spreading of cultured cells on galectin-8-coated substrates, tend to change the direction of their extension in a chiral fashion, acquiring a left-bent shape. Cryoelectron tomography examination indicated that turning of the filopodia tip to the left is accompanied by the displacement of the actin core bundle to the right of the filopodia midline. Reduction of the adhesion to galectin-8 by treatment with thiodigalactoside abolished this filopodia chirality. By modulating the expression of a variety of actin-associated filopodia proteins, we identified myosin-X and formin DAAM1 as major filopodia chirality promoting factors. Formin mDia1, actin filament elongation factor VASP, and actin filament cross-linker fascin were also shown to be involved. Thus, the simple actin cytoskeleton of filopodia, together with a small number of associated proteins are sufficient to drive a complex navigation process, manifested by the development of left-right asymmetry in these cellular protrusions.


Asunto(s)
Actinas , Seudópodos , Actinas/metabolismo , Forminas/metabolismo , Seudópodos/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto/metabolismo
4.
J Cell Sci ; 134(8)2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33589498

RESUMEN

The small molecular inhibitor of formin FH2 domains, SMIFH2, is widely used in cell biological studies. It inhibits formin-driven actin polymerization in vitro, but not polymerization of pure actin. It is active against several types of formin from different species. Here, we found that SMIFH2 inhibits retrograde flow of myosin 2 filaments and contraction of stress fibers. We further checked the effect of SMIFH2 on non-muscle myosin 2A and skeletal muscle myosin 2 in vitro, and found that SMIFH2 inhibits activity of myosin ATPase and the ability to translocate actin filaments in the gliding actin in vitro motility assay. Inhibition of non-muscle myosin 2A in vitro required a higher concentration of SMIFH2 compared with that needed to inhibit retrograde flow and stress fiber contraction in cells. We also found that SMIFH2 inhibits several other non-muscle myosin types, including bovine myosin 10, Drosophila myosin 7a and Drosophila myosin 5, more efficiently than it inhibits formins. These off-target inhibitions demand additional careful analysis in each case when solely SMIFH2 is used to probe formin functions. This article has an associated First Person interview with Yukako Nishimura, joint first author of the paper.


Asunto(s)
Citoesqueleto de Actina , Miosinas , Actinas/genética , Animales , Bovinos , Forminas , Miosinas/genética
5.
J Cell Sci ; 134(8)2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33722978

RESUMEN

The mechanisms underlying the cellular response to extracellular matrices (ECMs) that consist of multiple adhesive ligands are still poorly understood. Here, we address this topic by monitoring specific cellular responses to two different extracellular adhesion molecules - the main integrin ligand fibronectin and galectin-8, a lectin that binds ß-galactoside residues  - as well as to mixtures of the two proteins. Compared with cell spreading on fibronectin, cell spreading on galectin-8-coated substrates resulted in increased projected cell area, more-pronounced extension of filopodia and, yet, the inability to form focal adhesions and stress fibers. These differences can be partially reversed by experimental manipulations of small G-proteins of the Rho family and their downstream targets, such as formins, the Arp2/3 complex and Rho kinase. We also show that the physical adhesion of cells to galectin-8 was stronger than adhesion to fibronectin. Notably, galectin-8 and fibronectin differently regulate cell spreading and focal adhesion formation, yet act synergistically to upregulate the number and length of filopodia. The physiological significance of the coherent cellular response to a molecularly complex matrix is discussed. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Adhesivos , Fibronectinas , Adhesión Celular , Galectinas , Seudópodos
6.
Nat Mater ; 21(3): 366-377, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34663953

RESUMEN

Mechanotransduction is a process by which cells sense the mechanical properties of their surrounding environment and adapt accordingly to perform cellular functions such as adhesion, migration and differentiation. Integrin-mediated focal adhesions are major sites of mechanotransduction and their connection with the actomyosin network is crucial for mechanosensing as well as for the generation and transmission of forces onto the substrate. Despite having emerged as major regulators of cell adhesion and migration, the contribution of microtubules to mechanotransduction still remains elusive. Here, we show that talin- and actomyosin-dependent mechanosensing of substrate rigidity controls microtubule acetylation (a tubulin post-translational modification) by promoting the recruitment of α-tubulin acetyltransferase 1 (αTAT1) to focal adhesions. Microtubule acetylation tunes the mechanosensitivity of focal adhesions and Yes-associated protein (YAP) translocation. Microtubule acetylation, in turn, promotes the release of the guanine nucleotide exchange factor GEF-H1 from microtubules to activate RhoA, actomyosin contractility and traction forces. Our results reveal a fundamental crosstalk between microtubules and actin in mechanotransduction that contributes to mechanosensitive cell adhesion and migration.


Asunto(s)
Mecanotransducción Celular , Microtúbulos , Citoesqueleto de Actina/metabolismo , Adhesión Celular , Adhesiones Focales/metabolismo , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo
7.
Semin Cell Dev Biol ; 102: 73-80, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31813767

RESUMEN

The actomyosin cytoskeleton network plays a key role in a variety of fundamental cellular processes such as cell division, migration, and cell adhesion. The functions of cytoskeleton rely on its capability to receive, generate, respond to and transmit mechanical signals throughout the cytoskeleton network within the cells and throughout the tissue via cell-extracellular matrix and cell-cell adhesions. Crucial to the cytoskeleton's functions is actin polymerization that is regulated by many cellular factors. Among these factors, the formin family proteins, which bind the barbed end of an actin filament (F-actin), are known to be a major actin polymerization promoting factor. Mounting evidence from single-molecule mechanical manipulation experiments have suggested that formin-dependent actin polymerization is sensitively regulated by the force and torque applied to the F-actin, making the formin family an emerging mechanosensing factor that selectively promotes elongation of the F-actin under tensile forces. In this review, we will focus on the current understanding of the mechanical regulation of formin-mediated actin polymerization, the key technologies that have enabled quantification of formin-mediated actin polymerization under mechanical constraints, and future perspectives and studies on molecular mechanisms involved in the mechanosensing of actin dynamics.


Asunto(s)
Actinas/metabolismo , Forminas/metabolismo , Polimerizacion , Actomiosina/metabolismo , Citoesqueleto/metabolismo , Humanos
8.
J Cell Sci ; 132(5)2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30787030

RESUMEN

Actin cytoskeleton self-organization in two cell types, fibroblasts and epitheliocytes, was studied in cells confined to isotropic adhesive islands. In fibroblasts plated onto islands of optimal size, an initially circular actin pattern evolves into a radial pattern of actin bundles that undergo asymmetric chiral swirling before finally producing parallel linear stress fibers. Epitheliocytes, however, did not exhibit succession through all the actin patterns described above. Upon confinement, the actin cytoskeleton in non-keratinocyte epitheliocytes was arrested at the circular stage, while in keratinocytes it progressed as far as the radial pattern but still could not break symmetry. Epithelial-mesenchymal transition pushed actin cytoskeleton development from circular towards radial patterns but remained insufficient to cause chirality. Knockout of cytokeratins also did not promote actin chirality development in keratinocytes. Left-right asymmetric cytoskeleton swirling could, however, be induced in keratinocytes by treatment with small doses of the G-actin sequestering drug, latrunculin A in a transcription-independent manner. Both the nucleus and the cytokeratin network followed the induced chiral swirling. Development of chirality in keratinocytes was controlled by DIAPH1 (mDia1) and VASP, proteins involved in regulation of actin polymerization.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Células Epiteliales/fisiología , Fibroblastos/fisiología , Actinas/antagonistas & inhibidores , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Adhesión Celular , Moléculas de Adhesión Celular/metabolismo , Forma de la Célula , Células Cultivadas , Transición Epitelial-Mesenquimal , Forminas/metabolismo , Humanos , Queratinocitos/fisiología , Proteínas de Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Multimerización de Proteína , Tiazolidinas/farmacología
9.
Nat Rev Mol Cell Biol ; 10(1): 21-33, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19197329

RESUMEN

Recent progress in the design and application of artificial cellular microenvironments and nanoenvironments has revealed the extraordinary ability of cells to adjust their cytoskeletal organization, and hence their shape and motility, to minute changes in their immediate surroundings. Integrin-based adhesion complexes, which are tightly associated with the actin cytoskeleton, comprise the cellular machinery that recognizes not only the biochemical diversity of the extracellular neighbourhood, but also its physical and topographical characteristics, such as pliability, dimensionality and ligand spacing. Here, we discuss the mechanisms of such environmental sensing, based on the finely tuned crosstalk between the assembly of one type of integrin-based adhesion complex, namely focal adhesions, and the forces that are at work in the associated cytoskeletal network owing to actin polymerization and actomyosin contraction.


Asunto(s)
Citoesqueleto/fisiología , Ambiente , Adhesiones Focales/fisiología , Actinas/metabolismo , Animales , Humanos , Integrinas/metabolismo , Transducción de Señal
11.
Nat Mater ; 18(6): 638-649, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31114072

RESUMEN

The interrelationship between microtubules and the actin cytoskeleton in mechanoregulation of integrin-mediated adhesions is poorly understood. Here, we show that the effects of microtubules on two major types of cell-matrix adhesion, focal adhesions and podosomes, are mediated by KANK family proteins connecting the adhesion protein talin with microtubule tips. Both total microtubule disruption and microtubule uncoupling from adhesions by manipulations with KANKs trigger a massive assembly of myosin IIA filaments, augmenting focal adhesions and disrupting podosomes. Myosin IIA filaments are indispensable effectors in the microtubule-driven regulation of integrin-mediated adhesions. Myosin IIA filament assembly depends on Rho activation by the RhoGEF GEF-H1, which is trapped by microtubules when they are connected with integrin-mediated adhesions via KANK proteins but released after their disconnection. Thus, microtubule capture by integrin-mediated adhesions modulates the GEF-H1-dependent effect of microtubules on the assembly of myosin IIA filaments. Subsequent actomyosin reorganization then remodels the focal adhesions and podosomes, closing the regulatory loop.


Asunto(s)
Adhesiones Focales/metabolismo , Integrinas/metabolismo , Microtúbulos/metabolismo , Miosina Tipo IIA no Muscular/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Proteínas del Citoesqueleto , Humanos , Mecanotransducción Celular , Podosomas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo
12.
Biophys J ; 117(5): 856-866, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31427069

RESUMEN

Actin filaments associated with myosin motors constitute the cytoskeletal force-generating machinery for many types of adherent cells. These actomyosin units are structurally ordered in muscle cells and, in particular, may be spatially registered across neighboring actin bundles. Such registry or stacking of myosin filaments have been recently observed in ordered actin bundles of even fibroblasts with super-resolution microscopy techniques. We introduce here a model for the dynamics of stacking arising from long-range mechanical interactions between actomyosin units through mutual contractile deformations of the intervening cytoskeletal network. The dynamics of registry involve two key processes: 1) polymerization and depolymerization of actin filaments and 2) remodeling of cross-linker-rich actin adhesion zones, both of which are, in principle, mechanosensitive. By calculating the elastic forces that drive registry and their effect on actin polymerization rates, we estimate a characteristic timescale of tens of minutes for registry to be established, in agreement with experimentally observed timescales for individual kinetic processes involved in myosin stack formation, which we track and quantify. This model elucidates the role of actin turnover dynamics in myosin stacking and explains the loss of stacks seen when actin assembly or disassembly and cross-linking is experimentally disrupted in fibroblasts.


Asunto(s)
Actinas/metabolismo , Miosinas/metabolismo , Animales , Fenómenos Biomecánicos , Elasticidad , Cinética , Polimerizacion , Ratas , Fibras de Estrés/metabolismo
13.
Nano Lett ; 18(8): 5239-5247, 2018 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-29976069

RESUMEN

Self-assembling actin filaments not only form the basis of the cytoskeleton network in cells but also are utilized as nanosized building blocks to make novel active matter in which the dynamic polymerization and depolymerization of actin filaments play a key role. Formins belong to a main family of actin nucleation factors that bind to the barbed end of actin filaments and regulate actin polymerization through an interaction with profilin. Due to actomyosin contractility and relative rotation between formin and actin filaments, formin-dependent actin polymerization is subject to force and rotation constraints. However, it remains unclear how force and rotation constraints affect formin-dependent actin polymerization in the presence of profilin. Here, we show that for rotation-unconstrained actin filaments, elongation is accelerated by both force and profilin. The combined effect leads to surprisingly fast actin elongation that can approach the diffusion-limited rate at forces of a few piconewtons. The elongation of rotation-constrained filaments is also accelerated by profilin but is insensitive to applied force. We show that FH2, the main actin binding domain, plays the primary mechanosensing role. Together, the findings not only significantly advance our understanding of the mechanochemical regulation of formin-mediated actin polymerization in cells but also can potentially be utilized to make novel actin-based active matter.

14.
Proc Natl Acad Sci U S A ; 112(20): E2595-601, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25941386

RESUMEN

Cells constantly sense and respond to mechanical signals by reorganizing their actin cytoskeleton. Although a number of studies have explored the effects of mechanical stimuli on actin dynamics, the immediate response of actin after force application has not been studied. We designed a method to monitor the spatiotemporal reorganization of actin after cell stimulation by local force application. We found that force could induce transient actin accumulation in the perinuclear region within ∼ 2 min. This actin reorganization was triggered by an intracellular Ca(2+) burst induced by force application. Treatment with the calcium ionophore A23187 recapitulated the force-induced perinuclear actin remodeling. Blocking of actin polymerization abolished this process. Overexpression of Klarsicht, ANC-1, Syne Homology (KASH) domain to displace nesprins from the nuclear envelope did not abolish Ca(2+)-dependent perinuclear actin assembly. However, the endoplasmic reticulum- and nuclear membrane-associated inverted formin-2 (INF2), a potent actin polymerization activator (mutations of which are associated with several genetic diseases), was found to be important for perinuclear actin assembly. The perinuclear actin rim structure colocalized with INF2 on stimulation, and INF2 depletion resulted in attenuation of the rim formation. Our study suggests that cells can respond rapidly to external force by remodeling perinuclear actin in a unique Ca(2+)- and INF2-dependent manner.


Asunto(s)
Citoesqueleto de Actina/fisiología , Núcleo Celular/fisiología , Mecanotransducción Celular/fisiología , Citoesqueleto de Actina/ultraestructura , Animales , Fenómenos Biomecánicos , Calcio/metabolismo , Técnica del Anticuerpo Fluorescente , Forminas , Immunoblotting , Ratones , Proteínas de Microfilamentos/metabolismo , Microscopía de Fuerza Atómica , Células 3T3 NIH , Estimulación Física
15.
Nano Lett ; 16(9): 5951-61, 2016 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-27559755

RESUMEN

To understand how cells form tissues, we need to understand how the tyrosine kinases are involved in controlling cell mechanics, whether they act directly as parts of mechanosensing machines or indirectly. Cells test the critical parameter of matrix rigidity by locally contracting ("pinching") matrices and measuring forces, and the depletion of contractile units causes transformation. We report here that knocking down the receptor tyrosine kinases (RTKs), AXL, and ROR2, alters rigidity sensing and increases the magnitude or duration of local contraction events, respectively. Phospho-AXL and ROR2 localize to contraction units and bind major contractile components, tropomyosin 2.1 (AXL), myosin IIA (AXL), and filamin A (ROR2). At a molecular level, phosphorylated AXL localizes to active myosin filaments and phosphorylates tropomyosin at a tyrosine critical for adhesion formation. ROR2 binding of ligand is unnecessary, but binding filamin A helps function. Thus, AXL and ROR2 alter rigidity sensing and consequently morphogenic processes by directly controlling local mechanosensory contractions without ligands.


Asunto(s)
Fibroblastos/citología , Mecanotransducción Celular , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/fisiología , Células Cultivadas , Técnicas de Silenciamiento del Gen , Humanos , Tirosina Quinasa del Receptor Axl
17.
J Cell Sci ; 124(Pt 9): 1425-32, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21486952

RESUMEN

Focal adhesions (FAs) have key roles in the interaction of cells with the extracellular matrix (ECM) and in adhesion-mediated signaling. These dynamic, multi-protein structures sense the ECM both chemically and physically, and respond to external and internal forces by changing their size and signaling activity. However, this mechanosensitivity is still poorly understood at the molecular level. Here, we present direct evidence that actomyosin contractility regulates the molecular kinetics of FAs. We show that the molecular turnover of proteins within FAs is primarily regulated by their dissociation rate constant (k(off)), which is sensitive to changes in forces applied to the FA. We measured the early changes in k(off) values for three FA proteins (vinculin, paxillin and zyxin) upon inhibition of actomyosin-generated forces using two methods - high temporal resolution FRAP and direct measurement of FA protein dissociation in permeabilized cells. When myosin II contractility was inhibited, the k(off) values for all three proteins changed rapidly, in a highly protein-specific manner: dissociation of vinculin from FAs was facilitated, whereas dissociation of paxillin and zyxin was attenuated. We hypothesize that these early kinetic changes initiate FA disassembly by affecting the molecular turnover of FAs and altering their composition.


Asunto(s)
Actomiosina/metabolismo , Adhesiones Focales/metabolismo , Línea Celular Tumoral , Proteínas del Citoesqueleto/metabolismo , Recuperación de Fluorescencia tras Fotoblanqueo , Adhesiones Focales/efectos de los fármacos , Glicoproteínas/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Toxinas Marinas , Oxazoles/farmacología , Paxillin/metabolismo , Vinculina/metabolismo , Zixina
18.
Curr Opin Cell Biol ; 18(5): 472-81, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16930976

RESUMEN

Adhesion-mediated signaling provides cells with information about multiple parameters of their microenvironment, including mechanical characteristics. Often, such signaling is based on a unique feature of adhesion structures: their ability to grow and strengthen when force is applied to them, either from within the cell or from the outside. Such adhesion reinforcement is characteristic of integrin-mediated cell-matrix adhesions, but may also operate in other types of adhesion structures. Though the amount of knowledge about adhesion-mediated signaling is growing rapidly, the mechanisms underlying force-dependent regulation of junction assembly are largely unknown. Experiments have been carried out that have started to uncover the major signaling pathways involved in the response of adhesion sites to force. Theoretical models have also been used to address the physical mechanisms underlying adhesion-mediated mechanosensing.


Asunto(s)
Adhesión Celular/fisiología , Uniones Célula-Matriz/metabolismo , Mecanotransducción Celular/fisiología , Transducción de Señal/fisiología , Animales , Modelos Biológicos , Estrés Mecánico , Termodinámica
19.
Nat Commun ; 14(1): 776, 2023 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-36774346

RESUMEN

Deviations from mirror symmetry in the development of bilateral organisms are common but the mechanisms of initial symmetry breaking are insufficiently understood. The actin cytoskeleton of individual cells self-organises in a chiral manner, but the molecular players involved remain essentially unidentified and the relationship between chirality of an individual cell and cell collectives is unclear. Here, we analysed self-organisation of the chiral actin cytoskeleton in individual cells on circular or elliptical patterns, and collective cell alignment in confined microcultures. Screening based on deep-learning analysis of actin patterns identified actin polymerisation regulators, depletion of which suppresses chirality (mDia1) or reverses chirality direction (profilin1 and CapZß). The reversed chirality  is mDia1-independent but requires the function of actin-crosslinker α-actinin1. A robust correlation between the effects of a variety of actin assembly regulators on chirality of individual cells and cell collectives is revealed. Thus, actin-driven cell chirality may underlie tissue and organ asymmetry.


Asunto(s)
Citoesqueleto de Actina , Actinas
20.
Biophys J ; 102(8): 1746-56, 2012 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-22768930

RESUMEN

Cell motion is driven by interplay between the actin cytoskeleton and the cell adhesions in the front part of the cell. The actin network segregates into lamellipodium and lamellum, whereas the adhesion complexes are characteristically distributed underneath the actin system. Here, we suggest a computational model for this characteristic organization of the actin-adhesion system. The model is based on the ability of the adhesion complexes to sense mechanical forces, the stick-slip character of the interaction between the adhesions and the moving actin network, and a hypothetical propensity of the actin network to disintegrate upon sufficiently strong stretching stresses. We identify numerically three possible types of system organization, all observed in living cells: two states in which the actin network exhibits segregation into lamellipodium and lamellum, whereas the cell edge either remains stationary or moves, and a state where the actin network does not undergo segregation. The model recovers the asynchronous fluctuations and outward bulging of the cell edge, and the dependence of the edge protrusion velocity on the rate of the nascent adhesion generation, the membrane tension, and the substrate rigidity.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Modelos Biológicos , Actinas/química , Actinas/metabolismo , Adhesión Celular , Adhesiones Focales/metabolismo , Cinética , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Seudópodos/metabolismo , Fibras de Estrés/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA