Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Nat Rev Mol Cell Biol ; 18(12): 743-757, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29115298

RESUMEN

The way in which cells coordinate their behaviours during various biological processes, including morphogenesis, cancer progression and tissue remodelling, largely depends on the mechanical properties of the external environment. In contrast to single cells, collective cell behaviours rely on the cellular interactions not only with the surrounding extracellular matrix but also with neighbouring cells. Collective dynamics is not simply the result of many individually moving blocks. Instead, cells coordinate their movements by actively interacting with each other. These mechanisms are governed by mechanosensitive adhesion complexes at the cell-substrate interface and cell-cell junctions, which respond to but also further transmit physical signals. The mechanosensitivity and mechanotransduction at adhesion complexes are important for regulating tissue cohesiveness and thus are important for collective cell behaviours. Recent studies have shown that the physical properties of the cellular environment, which include matrix stiffness, topography, geometry and the application of external forces, can alter collective cell behaviours, tissue organization and cell-generated forces. On the basis of these findings, we can now start building our understanding of the mechanobiology of collective cell movements that span over multiple length scales from the molecular to the tissue level.


Asunto(s)
Comunicación Celular/fisiología , Movimiento Celular/fisiología , Microambiente Celular/fisiología , Matriz Extracelular/fisiología , Mecanotransducción Celular/fisiología , Animales , Humanos
2.
Nucleic Acids Res ; 50(8): 4389-4413, 2022 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-35474385

RESUMEN

Imbalance in the finely orchestrated system of chromatin-modifying enzymes is a hallmark of many pathologies such as cancers, since causing the affection of the epigenome and transcriptional reprogramming. Here, we demonstrate that a loss-of-function mutation (LOF) of the major histone lysine methyltransferase SETDB1 possessing oncogenic activity in lung cancer cells leads to broad changes in the overall architecture and mechanical properties of the nucleus through genome-wide redistribution of heterochromatin, which perturbs chromatin spatial compartmentalization. Together with the enforced activation of the epithelial expression program, cytoskeleton remodeling, reduced proliferation rate and restricted cellular migration, this leads to the reversed oncogenic potential of lung adenocarcinoma cells. These results emphasize an essential role of chromatin architecture in the determination of oncogenic programs and illustrate a relationship between gene expression, epigenome, 3D genome and nuclear mechanics.


Asunto(s)
Cromatina , Neoplasias Pulmonares , Humanos , Cromatina/genética , Epigenoma , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Heterocromatina , Fenotipo , Neoplasias Pulmonares/genética
3.
J Proteome Res ; 22(3): 996-1002, 2023 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-36748112

RESUMEN

The simple light isotope metabolic-labeling technique relies on the in vivo biosynthesis of amino acids from U-[12C]-labeled molecules provided as the sole carbon source. The incorporation of the resulting U-[12C]-amino acids into proteins presents several key advantages for mass-spectrometry-based proteomics analysis, as it results in more intense monoisotopic ions, with a better signal-to-noise ratio in bottom-up analysis. In our initial studies, we developed the simple light isotope metabolic (SLIM)-labeling strategy using prototrophic eukaryotic microorganisms, the yeasts Candida albicans and Saccharomyces cerevisiae, as well as strains with genetic markers that lead to amino-acid auxotrophy. To extend the range of SLIM-labeling applications, we evaluated (i) the incorporation of U-[12C]-glucose into proteins of human cells grown in a complex RPMI-based medium containing the labeled molecule, considering that human cell lines require a large number of essential amino-acids to support their growth, and (ii) an indirect labeling strategy in which the nematode Caenorhabditis elegans grown on plates was fed U-[12C]-labeled bacteria (Escherichia coli) and the worm proteome analyzed for 12C incorporation into proteins. In both cases, we were able to demonstrate efficient incorporation of 12C into the newly synthesized proteins, opening the way for original approaches in quantitative proteomics.


Asunto(s)
Caenorhabditis elegans , Proteoma , Animales , Humanos , Caenorhabditis elegans/genética , Proteoma/análisis , Escherichia coli/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Aminoácidos/metabolismo , Línea Celular , Isótopos , Marcaje Isotópico/métodos
4.
Proc Natl Acad Sci U S A ; 117(23): 12817-12825, 2020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32444491

RESUMEN

Morphogenesis, tumor formation, and wound healing are regulated by tissue rigidity. Focal adhesion behavior is locally regulated by stiffness; however, how cells globally adapt, detect, and respond to rigidity remains unknown. Here, we studied the interplay between the rheological properties of the cytoskeleton and matrix rigidity. We seeded fibroblasts onto flexible microfabricated pillar arrays with varying stiffness and simultaneously measured the cytoskeleton organization, traction forces, and cell-rigidity responses at both the adhesion and cell scale. Cells adopted a rigidity-dependent phenotype whereby the actin cytoskeleton polarized on stiff substrates but not on soft. We further showed a crucial role of active and passive cross-linkers in rigidity-sensing responses. By reducing myosin II activity or knocking down α-actinin, we found that both promoted cell polarization on soft substrates, whereas α-actinin overexpression prevented polarization on stiff substrates. Atomic force microscopy indentation experiments showed that this polarization response correlated with cell stiffness, whereby cell stiffness decreased when active or passive cross-linking was reduced and softer cells polarized on softer matrices. Theoretical modeling of the actin network as an active gel suggests that adaptation to matrix rigidity is controlled by internal mechanical properties of the cytoskeleton and puts forward a universal scaling between nematic order of the actin cytoskeleton and the substrate-to-cell elastic modulus ratio. Altogether, our study demonstrates the implication of cell-scale mechanosensing through the internal stress within the actomyosin cytoskeleton and its coupling with local rigidity sensing at focal adhesions in the regulation of cell shape changes and polarity.


Asunto(s)
Citoesqueleto/metabolismo , Módulo de Elasticidad , Mecanotransducción Celular , Andamios del Tejido/química , Actinina/metabolismo , Polaridad Celular , Reactivos de Enlaces Cruzados/química , Citoesqueleto/ultraestructura , Fibroblastos/metabolismo , Humanos , Modelos Teóricos , Miosinas/metabolismo
5.
Nat Mater ; 20(8): 1156-1166, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33603188

RESUMEN

Actomyosin machinery endows cells with contractility at a single-cell level. However, within a monolayer, cells can be contractile or extensile based on the direction of pushing or pulling forces exerted by their neighbours or on the substrate. It has been shown that a monolayer of fibroblasts behaves as a contractile system while epithelial or neural progentior monolayers behave as an extensile system. Through a combination of cell culture experiments and in silico modelling, we reveal the mechanism behind this switch in extensile to contractile as the weakening of intercellular contacts. This switch promotes the build-up of tension at the cell-substrate interface through an increase in actin stress fibres and traction forces. This is accompanied by mechanotransductive changes in vinculin and YAP activation. We further show that contractile and extensile differences in cell activity sort cells in mixtures, uncovering a generic mechanism for pattern formation during cell competition, and morphogenesis.


Asunto(s)
Actomiosina/metabolismo , Fenómenos Mecánicos , Fenómenos Biomecánicos , Movimiento Celular , Simulación por Computador , Modelos Biológicos
6.
Biol Cell ; 113(11): 441-449, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34287982

RESUMEN

BACKGROUND INFORMATION: Actin cytoskeleton contractility plays a critical role in morphogenetic processes by generating forces that are then transmitted to cell-cell and cell-ECM adhesion complexes. In turn, mechanical properties of the environment are sensed and transmitted to the cytoskeleton at cell adhesion sites, influencing cellular processes such as cell migration, differentiation and survival. Anchoring of the actomyosin cytoskeleton to adhesion sites is mediated by adaptor proteins such as talin or α-catenin that link F-actin to transmembrane cell adhesion receptors, thereby allowing mechanical coupling between the intracellular and extracellular compartments. Thus, a key issue is to be able to measure the forces generated by actomyosin and transmitted to the adhesion complexes. Approaches developed in cells and those probing single molecule mechanical properties of α-catenin molecules allowed to identify α-catenin, an F-actin binding protein which binds to the cadherin complexes as a major player in cadherin-based mechanotransduction. However, it is still very difficult to bridge intercellular forces measured at cellular levels and those measured at the single-molecule level. RESULTS: Here, we applied an intermediate approach allowing reconstruction of the actomyosin-α-catenin complex in acellular conditions to probe directly the transmitted forces. For this, we combined micropatterning of purified α-catenin and spontaneous actomyosin network assembly in the presence of G-actin and Myosin II with microforce sensor arrays used so far to measure cell-generated forces. CONCLUSIONS: Using this method, we show that self-organizing actomyosin bundles bound to micrometric α-catenin patches can apply near-nano-Newton forces. SIGNIFICANCE: Our results pave the way for future studies on molecular/cellular mechanotransduction and mechanosensing.


Asunto(s)
Actomiosina , Mecanotransducción Celular , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Cadherinas , Adhesión Celular , alfa Catenina/metabolismo
7.
J Cell Sci ; 131(24)2018 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-30573527

RESUMEN

In various physiological processes, the cell collective is organized in a monolayer, such as seen in a simple epithelium. The advances in the understanding of mechanical behavior of the monolayer and its underlying cellular and molecular mechanisms will help to elucidate the properties of cell collectives. In this Review, we discuss recent in vitro studies on monolayer mechanics and their implications on collective dynamics, regulation of monolayer mechanics by physical confinement and geometrical cues and the effect of tissue mechanics on biological processes, such as cell division and extrusion. In particular, we focus on the active nematic property of cell monolayers and the emerging approach to view biological systems in the light of liquid crystal theory. We also highlight the mechanosensing and mechanotransduction mechanisms at the sub-cellular and molecular level that are mediated by the contractile actomyosin cytoskeleton and cell-cell adhesion proteins, such as E-cadherin and α-catenin. To conclude, we argue that, in order to have a holistic understanding of the cellular response to biophysical environments, interdisciplinary approaches and multiple techniques - from large-scale traction force measurements to molecular force protein sensors - must be employed.


Asunto(s)
Uniones Adherentes/metabolismo , Cadherinas/metabolismo , Adhesión Celular/fisiología , Mecanotransducción Celular/fisiología , Actomiosina/metabolismo , Animales , Citoesqueleto/metabolismo , Humanos
8.
Soft Matter ; 16(7): 1825-1839, 2020 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-31970382

RESUMEN

The physical cues from the extracellular environment mediates cell signaling spatially and temporally. Cells respond to physical cues from their environment in a non-monotonic fashion. Despite our understanding of the role of substrate rigidity on single cell migration, how cells respond collectively to increasing extracellular matrix stiffness is not well established. Here we patterned multicellular epithelial Madin-Darby canine kidney (MDCK) islands on polyacrylamide gels of varying stiffness and studied their expansion. Our findings show that the MDCK islands expanded faster with increasing stiffness only up to an optimum stiffness, over which the expansion plateaued. We then focused on the expansion of the front of the assemblies and the formation of leader cells. We observed cell front destabilization only above substrate stiffness of a few kPa. The extension of multicellular finger-like structures at the edges of the colonies for intermediate and high stiffnesses from 6 to 60 kPa responded to higher substrate stiffness by increasing focal adhesion areas and actin cable assembly. Additionally, the number of leader cells at the finger-like protrusions increased with stiffness in correlation with an increase of the area of these multicellular protrusions. Consequently, the force profile along the epithelial fingers in the parallel and transverse directions of migration showed an unexpected relationship leading to a global force decrease with the increase of stiffness. Taken together, our findings show that epithelial cell colonies respond to substrate stiffness but in a non-trivial manner that may be of importance to understand morphogenesis and collective cell invasion during tumour progression.


Asunto(s)
Carcinogénesis/genética , Movimiento Celular/genética , Adhesiones Focales/genética , Neoplasias/genética , Actinas/química , Actinas/genética , Animales , Perros , Células Epiteliales/metabolismo , Humanos , Células de Riñón Canino Madin Darby , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Neoplasias/patología , Especificidad por Sustrato
10.
Biophys J ; 117(3): 464-478, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31307676

RESUMEN

Morphological changes during development, tissue repair, and disease largely rely on coordinated cell movements and are controlled by the tissue environment. Epithelial cell sheets are often subjected to large-scale deformation during tissue formation. The active mechanical environment in which epithelial cells operate have the ability to promote collective oscillations, but how these cellular movements are generated and relate to collective migration remains unclear. Here, combining in vitro experiments and computational modeling, we describe a form of collective oscillations in confined epithelial tissues in which the oscillatory motion is the dominant contribution to the cellular movements. We show that epithelial cells exhibit large-scale coherent oscillations when constrained within micropatterns of varying shapes and sizes and that their period and amplitude are set by the smallest confinement dimension. Using molecular perturbations, we then demonstrate that force transmission at cell-cell junctions and its coupling to cell polarity are pivotal for the generation of these collective movements. We find that the resulting tissue deformations are sufficient to trigger osillatory mechanotransduction of YAP within cells, potentially affecting a wide range of cellular processes.


Asunto(s)
Movimiento Celular , Células Epiteliales/citología , Actinas/metabolismo , Animales , Fenómenos Biomecánicos , Células CACO-2 , Adhesión Celular , Simulación por Computador , Perros , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Queratinocitos/citología , Células de Riñón Canino Madin Darby , Mecanotransducción Celular , Modelos Biológicos
11.
Soft Matter ; 15(13): 2798-2810, 2019 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-30888391

RESUMEN

Biological systems integrate dynamics at many scales, from molecules, protein complexes and genes, to cells, tissues and organisms. At every step of the way, mechanics, biochemistry and genetics offer complementary approaches to understand these dynamics. At the tissue scale, in vitro monolayers of epithelial cells provide a model to capture the influence of various factors on the motions of the tissue, in order to understand in vivo processes from morphogenesis, cancer progression and tissue remodelling. Ongoing efforts include research aimed at deciphering the roles of the cytoskeleton, of cell-substrate and cell-cell adhesions, and of cell proliferation-the point we investigate here. We show that confined to adherent strips, and on the time scale of a day or two, monolayers move with a characteristic front speed independent of proliferation, but that the motion is accompanied by persistent velocity waves, only in the absence of cell divisions. Here we show that the long-range transmission of physical signals is strongly coupled to cell density and proliferation. We interpret our results from a kinematic and mechanical perspective. Our study provides a framework to understand density-driven mechanisms of collective cell migration.

12.
Nano Lett ; 17(1): 399-406, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-27990827

RESUMEN

Focal adhesions (FAs) are important mediators of cell-substrate interactions. One of their key functions is the transmission of forces between the intracellular acto-myosin network and the substrate. However, the relationships between cell traction forces, FA architecture, and molecular forces within FAs are poorly understood. Here, by combining Förster resonance energy transfer (FRET)-based molecular force biosensors with micropillar-based traction force sensors and high-resolution fluorescence microscopy, we simultaneously map molecular tension across vinculin, a key protein in FAs, and traction forces at FAs. Our results reveal strong spatiotemporal correlations between vinculin tension and cell traction forces at FAs throughout a wide range of substrate stiffnesses. Furthermore, we find that molecular tension within individual FAs follows a biphasic distribution from the proximal (toward the cell nucleus) to distal end (toward the cell edge). Using super-resolution imaging, we show that such a distribution relates to that of FA proteins. On the basis of our experimental data, we propose a model in which FA dynamics results from tension changes along the FAs.

14.
J Math Biol ; 74(7): 1657-1678, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27783151

RESUMEN

We present a phenomenological model intended to describe at the protein population level the formation of cell-cell junctions by the local recruitment of homophilic cadherin adhesion receptors. This modeling may have a much wider implication in biological processes since many adhesion receptors, channel proteins and other membrane-born proteins associate in clusters or oligomers at the cell surface. Mathematically, it consists in a degenerate reaction-diffusion system of two partial differential equations modeling the time-space evolution of two cadherin populations over a surface: the first one represents the diffusing cadherins and the second one concerns the fixed ones. After discussing the stability of the solutions of the model, we perform numerical simulations and show relevant analogies with experimental results. In particular, we show patterns or aggregates formation for a certain set of parameters. Moreover, perturbing the stationary solution, both density populations converge in large times to some saturation level. Finally, an exponential rate of convergence is numerically obtained and is shown to be in agreement, for a suitable set of parameters, with the one obtained in some in vitro experiments.


Asunto(s)
Cadherinas/metabolismo , Adhesión Celular/fisiología , Modelos Biológicos , Difusión , Uniones Intercelulares
15.
J Cell Sci ; 127(Pt 8): 1660-71, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24522194

RESUMEN

Adhesive interactions of cadherins induce crosstalk between adhesion complexes and the actin cytoskeleton, allowing strengthening of adhesions and cytoskeletal organization. The underlying mechanisms are not completely understood, and microtubules (MTs) might be involved, as for integrin-mediated cell-extracellular-matrix adhesions. Therefore, we investigated the relationship between N-cadherin and MTs by analyzing the influence of N-cadherin engagement on MT distribution and dynamics. MTs progressed less, with a lower elongation rate, towards cadherin adhesions than towards focal adhesions. Increased actin treadmilling and the presence of an actomyosin contractile belt, suggested that actin relays inhibitory signals from cadherin adhesions to MTs. The reduced rate of MT elongation, associated with reduced recruitment of end-binding (EB) proteins to plus ends, was alleviated by expression of truncated N-cadherin, but was only moderately affected when actomyosin was disrupted. By contrast, destabilizing actomyosin fibers allowed MTs to enter the adhesion area, suggesting that tangential actin bundles impede MT growth independently of MT dynamics. Blocking MT penetration into the adhesion area strengthened cadherin adhesions. Taken together, these results establish a crosstalk between N-cadherin, F-actin and MTs. The opposing effects of cadherin and integrin engagement on actin organization and MT distribution might induce bias of the MT network during cell polarization.


Asunto(s)
Actomiosina/metabolismo , Cadherinas/metabolismo , Adhesión Celular , Microtúbulos/metabolismo , Animales , Perros , Células de Riñón Canino Madin Darby , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Estabilidad Proteica , Transporte de Proteínas
17.
J Neurosci ; 33(46): 18149-60, 2013 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-24227724

RESUMEN

In the developing brain, cortical GABAergic interneurons migrate long distances from the medial ganglionic eminence (MGE) in which they are generated, to the cortex in which they settle. MGE cells express the cell adhesion molecule N-cadherin, a homophilic cell-cell adhesion molecule that regulates numerous steps of brain development, from neuroepithelium morphogenesis to synapse formation. N-cadherin is also expressed in embryonic territories crossed by MGE cells during their migration. In this study, we demonstrate that N-cadherin is a key player in the long-distance migration of future cortical interneurons. Using N-cadherin-coated substrate, we show that N-cadherin-dependent adhesion promotes the migration of mouse MGE cells in vitro. Conversely, mouse MGE cells electroporated with a construct interfering with cadherin function show reduced cell motility, leading process instability, and impaired polarization associated with abnormal myosin IIB dynamics. In vivo, the capability of electroporated MGE cells to invade the developing cortical plate is altered. Using genetic ablation of N-cadherin in mouse embryos, we show that N-cadherin-depleted MGEs are severely disorganized. MGE cells hardly exit the disorganized proliferative area. N-cadherin ablation at the postmitotic stage, which does not affect MGE morphogenesis, alters MGE cell motility and directionality. The tangential migration to the cortex of N-cadherin ablated MGE cells is delayed, and their radial migration within the cortical plate is perturbed. Altogether, these results identify N-cadherin as a pivotal adhesion substrate that activates cell motility in future cortical interneurons and maintains cell polarity over their long-distance migration to the developing cortex.


Asunto(s)
Cadherinas/metabolismo , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Corteza Cerebral/metabolismo , Interneuronas/metabolismo , Neurogénesis/fisiología , Animales , Cadherinas/deficiencia , Células Cultivadas , Corteza Cerebral/citología , Técnicas de Cocultivo , Femenino , Predicción , Humanos , Masculino , Ratones , Ratones Transgénicos , Embarazo
18.
J Biol Chem ; 288(7): 4957-69, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23266828

RESUMEN

Maintaining cell cohesiveness within tissues requires that intercellular adhesions develop sufficient strength to support traction forces applied by myosin motors and by neighboring cells. Cadherins are transmembrane receptors that mediate intercellular adhesion. The cadherin cytoplasmic domain recruits several partners, including catenins and vinculin, at sites of cell-cell adhesion. Our study used force measurements to address the role of αE-catenin and vinculin in the regulation of the strength of E-cadherin-based adhesion. αE-catenin-deficient cells display only weak aggregation and fail to strengthen intercellular adhesion over time, a process rescued by the expression of αE-catenin or chimeric E-cadherin·αE-catenins, including a chimera lacking the αE-catenin dimerization domain. Interestingly, an αE-catenin mutant lacking the modulation and actin-binding domains restores cadherin-dependent cell-cell contacts but cannot strengthen intercellular adhesion. The expression of αE-catenin mutated in its vinculin-binding site is defective in its ability to rescue cadherin-based adhesion strength in cells lacking αE-catenin. Vinculin depletion or the overexpression of the αE-catenin modulation domain strongly decreases E-cadherin-mediated adhesion strength. This supports the notion that both molecules are required for intercellular contact maturation. Furthermore, stretching of cell doublets increases vinculin recruitment and α18 anti-αE-catenin conformational epitope immunostaining at cell-cell contacts. Taken together, our results indicate that αE-catenin and vinculin cooperatively support intercellular adhesion strengthening, probably via a mechanoresponsive link between the E-cadherin·ß-catenin complexes and the underlying actin cytoskeleton.


Asunto(s)
Cadherinas/metabolismo , Regulación de la Expresión Génica , Vinculina/metabolismo , alfa Catenina/metabolismo , Actinas/metabolismo , Animales , Adhesión Celular , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Eliminación de Gen , Ratones , Microscopía Fluorescente/métodos , Modelos Biológicos , Modelos Genéticos , Mutación , Unión Proteica , Factores de Tiempo
19.
Methods Mol Biol ; 2764: 145-156, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38393593

RESUMEN

Ectodermal organ development, including lacrimal gland, is characterized by an interaction between an epithelium and a mesenchyme. Murine lacrimal gland is a good model to study non-stereotypical branching morphogenesis. In vitro cultures allow the study of morphogenesis events with easy access to high-resolution imaging. Particularly, embryonic lacrimal gland organotypic 3D cell cultures enable the follow-up of branching morphogenesis thanks to the analysis of territories organization by immunohistochemistry. In this chapter, we describe a method to culture primary epithelial fragments together with primary mesenchymal cells, isolated from embryonic day 17 lacrimal glands.


Asunto(s)
Aparato Lagrimal , Ratones , Animales , Epitelio , Morfogénesis , Técnicas de Cultivo Tridimensional de Células , Técnicas de Cultivo de Órganos
20.
Curr Opin Cell Biol ; 18(5): 541-8, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16905303

RESUMEN

A major form of animal cell-cell adhesion results from the dynamic association of cadherin molecules, cytosolic catenins and actin microfilaments. Cadherins dynamically regulate the cytoskeleton. In turn, the actin cytoskeleton contributes to cadherin molecule oligomerization at cell contacts and to cell reshaping in response to environmental changes. Over the past two years, this evolutionarily conserved adhesion system has been intensively revisited in both its structural and functional aspects; this is illustrated by the remarkable progress in the determination of physical parameters of cadherin bonds (including force measurement) and the new insights into the role of alpha-catenin and the regulation of actin dynamics at cadherin contacts. Other recent studies uncover the important contribution of acto-myosin, microtubules and cell tension to adherens junction formation, cell differentiation and tissue reshaping/remodeling. An open challenge is now to integrate these new data with the diversity of cadherin adhesive complexes.


Asunto(s)
Cadherinas/metabolismo , Citoesqueleto/metabolismo , Uniones Intercelulares/fisiología , Actinas/metabolismo , Animales , Membrana Celular/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Microtúbulos/metabolismo , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA