Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Cell Sci ; 136(8)2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-37083041

RESUMEN

Focal adhesions are composed of transmembrane integrins, linking the extracellular matrix to the actomyosin cytoskeleton, via cytoplasmic proteins. Adhesion depends on the activation of integrins. Talin and kindlin proteins are intracellular activators of integrins that bind to ß-integrin cytoplasmic tails. Integrin activation and clustering through extracellular ligands guide the organization of adhesion complexes. However, the roles of talin and kindlin in this process are poorly understood. To determine the contribution of talin, kindlin, lipids and actomyosin in integrin clustering, we used a biomimetic in vitro system, made of giant unilamellar vesicles, containing transmembrane integrins (herein αIIbß3), with purified talin (talin-1), kindlin (kindlin-2, also known as FERMT2) and actomyosin. Here, we show that talin and kindlin individually have the ability to cluster integrins. Talin and kindlin synergize to induce the formation of larger integrin clusters containing the three proteins. Comparison of protein density reveals that kindlin increases talin and integrin density, whereas talin does not affect kindlin and integrin density. Finally, kindlin increases integrin-talin-actomyosin coupling. Our study unambiguously demonstrates how kindlin and talin cooperate to induce integrin clustering, which is a major parameter for cell adhesion.


Asunto(s)
Integrinas , Talina , Integrinas/metabolismo , Talina/genética , Talina/metabolismo , Actomiosina , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Adhesión Celular
2.
Nat Methods ; 16(12): 1263-1268, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31636458

RESUMEN

Super-resolution microscopy offers tremendous opportunities to unravel the complex and dynamic architecture of living cells. However, current super-resolution microscopes are well suited for revealing protein distributions or cell morphology, but not both. We present a super-resolution platform that permits correlative single-molecule imaging and stimulated emission depletion microscopy in live cells. It gives nanoscale access to the positions and movements of synaptic proteins within the morphological context of growth cones and dendritic spines.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Microscopía Fluorescente/métodos , Imagen Individual de Molécula/métodos , Animales , Células Cultivadas , Femenino , Humanos , Ratones , Ratas , Ratas Sprague-Dawley
3.
Int J Mol Sci ; 22(12)2021 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-34199292

RESUMEN

Giant unilamellar vesicles (GUV) are powerful tools to explore physics and biochemistry of the cell membrane in controlled conditions. For example, GUVs were extensively used to probe cell adhesion, but often using non-physiological linkers, due to the difficulty of incorporating transmembrane adhesion proteins into model membranes. Here we describe a new protocol for making GUVs incorporating the transmembrane protein integrin using gel-assisted swelling. We report an optimised protocol, enumerating the pitfalls encountered and precautions to be taken to maintain the robustness of the protocol. We characterise intermediate steps of small proteoliposome formation and the final formed GUVs. We show that the integrin molecules are successfully incorporated and are functional.


Asunto(s)
Geles/química , Integrinas/metabolismo , Liposomas Unilamelares/química , Adhesión Celular , Fluorescencia , Humanos , Membrana Dobles de Lípidos/metabolismo , Lípidos/química , Tamaño de la Partícula
4.
Nat Methods ; 14(12): 1184-1190, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29083400

RESUMEN

Single-molecule localization microscopy techniques have proven to be essential tools for quantitatively monitoring biological processes at unprecedented spatial resolution. However, these techniques are very low throughput and are not yet compatible with fully automated, multiparametric cellular assays. This shortcoming is primarily due to the huge amount of data generated during imaging and the lack of software for automation and dedicated data mining. We describe an automated quantitative single-molecule-based super-resolution methodology that operates in standard multiwell plates and uses analysis based on high-content screening and data-mining software. The workflow is compatible with fixed- and live-cell imaging and allows extraction of quantitative data like fluorophore photophysics, protein clustering or dynamic behavior of biomolecules. We demonstrate that the method is compatible with high-content screening using 3D dSTORM and DNA-PAINT based super-resolution microscopy as well as single-particle tracking.


Asunto(s)
Bases de Datos Factuales , Procesamiento de Imagen Asistido por Computador/métodos , Imagen Individual de Molécula/métodos , Animales , Células COS , Chlorocebus aethiops , Minería de Datos , Colorantes Fluorescentes , Células HeLa , Humanos , Proteínas de la Membrana/análisis , Transporte de Proteínas , Receptores de Neurotransmisores/análisis , Programas Informáticos , Flujo de Trabajo
5.
Exp Cell Res ; 379(2): 235-244, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30943383

RESUMEN

Cells are mechanical living machines that remodel their microenvironment by adhering and generating forces on the extracellular matrix (ECM) using integrin-dependent adhesion sites (IAS). In return, the biochemical and physical nature of the ECM determines cellular behavior and morphology during proliferation, differentiation and migration. IAS come in different shapes and forms. They have specific compositions, morphologies, mechanical and biochemical signaling activities, which serve different cellular functions. Proteomic studies showed that IAS are composed of a large repertoire of proteins that could be linked to different functional activities, including signaling, force-transmission and force-sensing. Thanks to recent technological advances in microscopy and protein engineering, it is now possible to localize single proteins in three dimensions inside IAS, determine their diffusive behaviors, orientations, and how much mechanical force is transmitted across individual components. Here, we review how researchers have used those tools to investigate how IAS components assemble and dynamically interact to produce diverse functions of adhesive structures.


Asunto(s)
Adhesión Celular/fisiología , Matriz Extracelular/metabolismo , Integrinas/metabolismo , Sustancias Macromoleculares/metabolismo , Animales , Humanos , Mecanotransducción Celular/fisiología , Estrés Mecánico
6.
Nature ; 511(7509): 319-25, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25030168

RESUMEN

Malignancy is associated with altered expression of glycans and glycoproteins that contribute to the cellular glycocalyx. We constructed a glycoprotein expression signature, which revealed that metastatic tumours upregulate expression of bulky glycoproteins. A computational model predicted that these glycoproteins would influence transmembrane receptor spatial organization and function. We tested this prediction by investigating whether bulky glycoproteins in the glycocalyx promote a tumour phenotype in human cells by increasing integrin adhesion and signalling. Our data revealed that a bulky glycocalyx facilitates integrin clustering by funnelling active integrins into adhesions and altering integrin state by applying tension to matrix-bound integrins, independent of actomyosin contractility. Expression of large tumour-associated glycoproteins in non-transformed mammary cells promoted focal adhesion assembly and facilitated integrin-dependent growth factor signalling to support cell growth and survival. Clinical studies revealed that large glycoproteins are abundantly expressed on circulating tumour cells from patients with advanced disease. Thus, a bulky glycocalyx is a feature of tumour cells that could foster metastasis by mechanically enhancing cell-surface receptor function.


Asunto(s)
Glicocálix/metabolismo , Glicoproteínas/metabolismo , Integrinas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Animales , Mama/citología , Mama/metabolismo , Mama/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Fibroblastos , Glicocálix/química , Humanos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/metabolismo , Integrinas/química , Ratones , Terapia Molecular Dirigida , Mucina-1/metabolismo , Metástasis de la Neoplasia/patología , Células Neoplásicas Circulantes , Unión Proteica , Receptores de Superficie Celular
7.
Exp Cell Res ; 343(1): 28-34, 2016 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-26571074

RESUMEN

Cells adjust their adhesive and cytoskeletal organizations according to changes in the biochemical and physical nature of their surroundings. In return, by adhering and generating forces on the extracellular matrix (ECM) cells organize their microenvironment. Integrin-dependent focal adhesions (FAs) are the converging zones integrating biochemical and biomechanical signals arising from the ECM and the actin cytoskeleton. Thus, integrin-mediated adhesion and mechanotransduction, the conversion of mechanical forces into biochemical signals, are involved in critical cellular functions such as migration, proliferation and differentiation, and their deregulation contributes to pathologies including cancer. A challenging problem is to decipher how stochastic protein movements and interactions lead to formation of dynamic architecture such as integrin-dependent adhesive structures. In this review, we will describe recent advances made possible by super-resolution microscopies and single molecule tracking approaches that provided new understanding on the organization and the dynamics of integrins and intracellular regulators at the nanoscale in living cells.


Asunto(s)
Integrinas/metabolismo , Adhesión Celular , Humanos , Mecanotransducción Celular , Microscopía , Nanotecnología , Transporte de Proteínas , Estrés Mecánico
8.
EMBO J ; 29(6): 1055-68, 2010 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-20150894

RESUMEN

Extracellular matrices in vivo are heterogeneous structures containing gaps that cells bridge with an actomyosin network. To understand the basis of bridging, we plated cells on surfaces patterned with fibronectin (FN)-coated stripes separated by non-adhesive regions. Bridges developed large tensions where concave cell edges were anchored to FN by adhesion sites. Actomyosin complexes assembled near those sites (both actin and myosin filaments) and moved towards the centre of the non-adhesive regions in a treadmilling network. Inhibition of myosin-II (MII) or Rho-kinase collapsed bridges, whereas extension continued over adhesive areas. Inhibition of actin polymerization (latrunculin-A, jasplakinolide) also collapsed the actomyosin network. We suggest that MII has distinct functions at different bridge regions: (1) at the concave edges of bridges, MIIA force stimulates actin filament assembly at adhesions and (2) in the body of bridges, myosin cross-links actin filaments and stimulates actomyosin network healing when breaks occur. Both activities ensure turnover of actin networks needed to maintain stable bridges from one adhesive region to another.


Asunto(s)
Actomiosina/química , Contracción Muscular/fisiología , Actinas/metabolismo , Actomiosina/metabolismo , Citoesqueleto/metabolismo , Cinética , Miosinas/química , Miosinas/metabolismo , Quinasas Asociadas a rho/química , Quinasas Asociadas a rho/metabolismo
9.
Trends Cell Biol ; 33(3): 204-220, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36055943

RESUMEN

Detection and conversion of mechanical forces into biochemical signals is known as mechanotransduction. From cells to tissues, mechanotransduction regulates migration, proliferation, and differentiation in processes such as immune responses, development, and cancer progression. Mechanosensitive structures such as integrin adhesions, the actin cortex, ion channels, caveolae, and the nucleus sense and transmit forces. In vitro approaches showed that mechanosensing is based on force-dependent protein deformations and reorganizations. However, the mechanisms in cells remained unclear since cell imaging techniques lacked molecular resolution. Thanks to recent developments in super-resolution microscopy (SRM) and molecular force sensors, it is possible to obtain molecular insight of mechanosensing in live cells. We discuss how understanding of molecular mechanotransduction was revolutionized by these innovative approaches, focusing on integrin adhesions, actin structures, and the plasma membrane.


Asunto(s)
Actinas , Mecanotransducción Celular , Humanos , Mecanotransducción Celular/fisiología , Actinas/metabolismo , Citoesqueleto/metabolismo , Integrinas/metabolismo , Microscopía , Adhesión Celular , Citoesqueleto de Actina/metabolismo
10.
J Cell Biol ; 222(3)2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36562751

RESUMEN

Septins are cytoskeletal proteins conserved from algae and protists to mammals. A unique feature of septins is their presence as heteromeric complexes that polymerize into filaments in solution and on lipid membranes. Although animal septins associate extensively with actin-based structures in cells, whether septins organize as filaments in cells and if septin organization impacts septin function is not known. Customizing a tripartite split-GFP complementation assay, we show that all septins decorating actin stress fibers are octamer-containing filaments. Depleting octamers or preventing septins from polymerizing leads to a loss of stress fibers and reduced cell stiffness. Super-resolution microscopy revealed septin fibers with widths compatible with their organization as paired septin filaments. Nanometer-resolved distance measurements and single-protein tracking further showed that septin filaments are membrane bound and largely immobilized. Finally, reconstitution assays showed that septin filaments mediate actin-membrane anchoring. We propose that septin organization as octamer-based filaments is essential for septin function in anchoring and stabilizing actin filaments at the plasma membrane.


Asunto(s)
Actinas , Septinas , Humanos , Actinas/metabolismo , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Microscopía , Septinas/análisis
11.
J Cell Sci ; 123(Pt 3): 413-23, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20067993

RESUMEN

Maintaining a physical connection across cytoplasm is crucial for many biological processes such as matrix force generation, cell motility, cell shape and tissue development. However, in the absence of stress fibers, the coherent structure that transmits force across the cytoplasm is not understood. We find that nonmuscle myosin-II (NMII) contraction of cytoplasmic actin filaments establishes a coherent cytoskeletal network irrespective of the nature of adhesive contacts. When NMII activity is inhibited during cell spreading by Rho kinase inhibition, blebbistatin, caldesmon overexpression or NMIIA RNAi, the symmetric traction forces are lost and cell spreading persists, causing cytoplasm fragmentation by membrane tension that results in 'C' or dendritic shapes. Moreover, local inactivation of NMII by chromophore-assisted laser inactivation causes local loss of coherence. Actin filament polymerization is also required for cytoplasmic coherence, but microtubules and intermediate filaments are dispensable. Loss of cytoplasmic coherence is accompanied by loss of circumferential actin bundles. We suggest that NMIIA creates a coherent actin network through the formation of circumferential actin bundles that mechanically link elements of the peripheral actin cytoskeleton where much of the force is generated during spreading.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Miosina Tipo IIA no Muscular/fisiología , Animales , Western Blotting , Células Cultivadas , Citoesqueleto/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Ratones , Células 3T3 NIH , Miosina Tipo IIA no Muscular/genética , Miosina Tipo IIA no Muscular/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores
12.
J Cell Biol ; 221(12)2022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36205720

RESUMEN

The spatial organization of cell-surface receptors is fundamental for the coordination of biological responses to physical and biochemical cues of the extracellular matrix. How serine/threonine kinase receptors, ALK3-BMPRII, cooperate with integrins upon BMP2 to drive cell migration is unknown. Whether the dynamics between integrins and BMP receptors intertwine in space and time to guide adhesive processes is yet to be elucidated. We found that BMP2 stimulation controls the spatial organization of BMPRs by segregating ALK3 from BMPRII into ß3 integrin-containing focal adhesions. The selective recruitment of ALK3 to focal adhesions requires ß3 integrin engagement and ALK3 activation. BMP2 controls the partitioning of immobilized ALK3 within and outside focal adhesions according to single-protein tracking and super-resolution imaging. The spatial control of ALK3 in focal adhesions by optogenetics indicates that ALK3 acts as an adhesive receptor by eliciting cell spreading required for cell migration. ALK3 segregation from BMPRII in integrin-based adhesions is a key aspect of the spatio-temporal control of BMPR signaling.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1 , Integrina beta3 , Proteína Morfogenética Ósea 2/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Adhesión Celular , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Adhesiones Focales/metabolismo , Integrina beta3/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo
13.
Biophys J ; 100(4): 845-57, 2011 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-21320428

RESUMEN

Cell spreading is regulated by signaling from the integrin receptors that activate intracellular signaling pathways to control actin filament regulatory proteins. We developed a hybrid model of whole-cell spreading in which we modeled the integrin signaling network as ordinary differential equations in multiple compartments, and cell spreading as a three-dimensional stochastic model. The computed activity of the signaling network, represented as time-dependent activity levels of the actin filament regulatory proteins, is used to drive the filament dynamics. We analyzed the hybrid model to understand the role of signaling during the isotropic phase of fibroblasts spreading on fibronectin-coated surfaces. Simulations showed that the isotropic phase of spreading depends on integrin signaling to initiate spreading but not to maintain the spreading dynamics. Simulations predicted that signal flow in the absence of Cdc42 or WASP would reduce the spreading rate but would not affect the shape evolution of the spreading cell. These predictions were verified experimentally. Computational analyses showed that the rate of spreading and the evolution of cell shape are largely controlled by the membrane surface load and membrane bending rigidity, and changing information flow through the integrin signaling network has little effect. Overall, the plasma membrane acts as a damper such that only ∼5% of the actin dynamics capability is needed for isotropic spreading. Thus, the biophysical properties of the plasma membrane can condense varying levels of signaling network activities into a single cohesive macroscopic cellular behavior.


Asunto(s)
Actinas/metabolismo , Membrana Celular/metabolismo , Movimiento Celular , Citoesqueleto/metabolismo , Transducción de Señal , Animales , Simulación por Computador , Técnicas de Inactivación de Genes , Ratones , Modelos Biológicos , Polimerizacion , Propiedades de Superficie , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Proteína de Unión al GTP cdc42/metabolismo
14.
Methods Mol Biol ; 2217: 85-113, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33215379

RESUMEN

Cell migration is a complex biophysical process which involves the coordination of molecular assemblies including integrin-dependent adhesions, signaling networks and force-generating cytoskeletal structures incorporating both actin polymerization and myosin activity. During the last decades, proteomic studies have generated impressive protein-protein interaction maps, although the subcellular location, duration, strength, sequence, and nature of these interactions are still concealed. In this chapter we describe how recent developments in superresolution microscopy (SRM) and single-protein tracking (SPT) start to unravel protein interactions and actions in subcellular molecular assemblies driving cell migration.


Asunto(s)
Movimiento Celular , Integrinas/metabolismo , Microscopía/métodos , Optogenética/métodos , Mapeo de Interacción de Proteínas/métodos , Imagen Individual de Molécula/métodos , Actinas/genética , Actinas/metabolismo , Animales , Adhesión Celular , Línea Celular Transformada , Criptocromos/genética , Criptocromos/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Expresión Génica , Integrinas/genética , Ratones , Microscopía/instrumentación , Miosinas/genética , Miosinas/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Unión Proteica , Seudópodos/metabolismo , Seudópodos/ultraestructura , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/genética , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
15.
Nat Commun ; 12(1): 3104, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34035280

RESUMEN

Focal adhesions (FAs) initiate chemical and mechanical signals involved in cell polarity, migration, proliferation and differentiation. Super-resolution microscopy revealed that FAs are organized at the nanoscale into functional layers from the lower plasma membrane to the upper actin cytoskeleton. Yet, how FAs proteins are guided into specific nano-layers to promote interaction with given targets is unknown. Using single protein tracking, super-resolution microscopy and functional assays, we link the molecular behavior and 3D nanoscale localization of kindlin with its function in integrin activation inside FAs. We show that immobilization of integrins in FAs depends on interaction with kindlin. Unlike talin, kindlin displays free diffusion along the plasma membrane outside and inside FAs. We demonstrate that the kindlin Pleckstrin Homology domain promotes membrane diffusion and localization to the membrane-proximal integrin nano-layer, necessary for kindlin enrichment and function in FAs. Using kindlin-deficient cells, we show that kindlin membrane localization and diffusion are crucial for integrin activation, cell spreading and FAs formation. Thus, kindlin uses a different route than talin to reach and activate integrins, providing a possible molecular basis for their complementarity during integrin activation.


Asunto(s)
Membrana Celular/metabolismo , Adhesiones Focales/metabolismo , Integrinas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Sitios de Unión , Adhesión Celular , Células Cultivadas , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Humanos , Integrinas/genética , Proteínas de la Membrana/genética , Ratones Noqueados , Movimiento (Física) , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutación , Proteínas de Neoplasias/genética , Unión Proteica
16.
Nat Cell Biol ; 23(11): 1148-1162, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34737443

RESUMEN

Actin filaments generate mechanical forces that drive membrane movements during trafficking, endocytosis and cell migration. Reciprocally, adaptations of actin networks to forces regulate their assembly and architecture. Yet, a demonstration of forces acting on actin regulators at actin assembly sites in cells is missing. Here we show that local forces arising from actin filament elongation mechanically control WAVE regulatory complex (WRC) dynamics and function, that is, Arp2/3 complex activation in the lamellipodium. Single-protein tracking revealed WRC lateral movements along the lamellipodium tip, driven by elongation of actin filaments and correlating with WRC turnover. The use of optical tweezers to mechanically manipulate functional WRC showed that piconewton forces, as generated by single-filament elongation, dissociated WRC from the lamellipodium tip. WRC activation correlated with its trapping, dwell time and the binding strength at the lamellipodium tip. WRC crosslinking, hindering its mechanical dissociation, increased WRC dwell time and Arp2/3-dependent membrane protrusion. Thus, forces generated by individual actin filaments on their regulators can mechanically tune their turnover and hence activity during cell migration.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Movimiento Celular , Fibroblastos/metabolismo , Mecanotransducción Celular , Seudópodos/metabolismo , Citoesqueleto de Actina/genética , Complejo 2-3 Proteico Relacionado con la Actina/genética , Animales , Línea Celular Transformada , Ratones , Microscopía Fluorescente , Pinzas Ópticas , Imagen Individual de Molécula , Estrés Mecánico , Factores de Tiempo
17.
Biophys J ; 98(10): 2136-46, 2010 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-20483321

RESUMEN

Cell motility is important for many developmental and physiological processes. Motility arises from interactions between physical forces at the cell surface membrane and the biochemical reactions that control the actin cytoskeleton. To computationally analyze how these factors interact, we built a three-dimensional stochastic model of the experimentally observed isotropic spreading phase of mammalian fibroblasts. The multiscale model is composed at the microscopic levels of three actin filament remodeling reactions that occur stochastically in space and time, and these reactions are regulated by the membrane forces due to membrane surface resistance (load) and bending energy. The macroscopic output of the model (isotropic spreading of the whole cell) occurs due to the movement of the leading edge, resulting solely from membrane force-constrained biochemical reactions. Numerical simulations indicate that our model qualitatively captures the experimentally observed isotropic cell-spreading behavior. The model predicts that increasing the capping protein concentration will lead to a proportional decrease in the spread radius of the cell. This prediction was experimentally confirmed with the use of Cytochalasin D, which caps growing actin filaments. Similarly, the predicted effect of actin monomer concentration was experimentally verified by using Latrunculin A. Parameter variation analyses indicate that membrane physical forces control cell shape during spreading, whereas the biochemical reactions underlying actin cytoskeleton dynamics control cell size (i.e., the rate of spreading). Thus, during cell spreading, a balance between the biochemical and biophysical properties determines the cell size and shape. These mechanistic insights can provide a format for understanding how force and chemical signals together modulate cellular regulatory networks to control cell motility.


Asunto(s)
Movimiento Celular/fisiología , Forma de la Célula/fisiología , Citocalasina D/farmacología , Fibroblastos/fisiología , Movimiento/fisiología , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Citoesqueleto de Actina/fisiología , Actinas , Adenosina Difosfato/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Membrana Celular/fisiología , Polaridad Celular/fisiología , Forma de la Célula/efectos de los fármacos , Tamaño de la Célula , Células Cultivadas , Estructuras Celulares/efectos de los fármacos , Citoesqueleto/fisiología , Células Epiteliales/fisiología , Fluidez de la Membrana/fisiología , Proteínas Motoras Moleculares
18.
Nat Cell Biol ; 22(8): 1011-1023, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32719553

RESUMEN

Detection and conversion of mechanical forces into biochemical signals controls cell functions during physiological and pathological processes. Mechanosensing is based on protein deformations and reorganizations, yet the molecular mechanisms are still unclear. Using a cell-stretching device compatible with super-resolution microscopy and single-protein tracking, we explored the nanoscale deformations and reorganizations of individual proteins inside mechanosensitive structures. We achieved super-resolution microscopy after live stretching on intermediate filaments, microtubules and integrin adhesions. Simultaneous single-protein tracking and stretching showed that while integrins followed the elastic deformation of the substrate, actin filaments and talin also displayed lagged and transient inelastic responses associated with active acto-myosin remodelling and talin deformations. Capturing acute reorganizations of single molecules during stretching showed that force-dependent vinculin recruitment is delayed and depends on the maturation of integrin adhesions. Thus, cells respond to external forces by amplifying transiently and locally cytoskeleton displacements, enabling protein deformation and recruitment in mechanosensitive structures.


Asunto(s)
Actinas/fisiología , Forma de la Célula , Animales , Fenómenos Biomecánicos , Células Cultivadas , Técnicas Citológicas , Humanos , Integrinas/metabolismo , Ratones , Microscopía/métodos , Nanoestructuras , Pliegue de Proteína , Transporte de Proteínas , Talina/metabolismo , Vinculina/metabolismo
19.
Curr Biol ; 29(17): 2852-2866.e5, 2019 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-31422887

RESUMEN

The spatiotemporal coordination of actin regulators in the lamellipodium determines the dynamics and architecture of branched F-actin networks during cell migration. The WAVE regulatory complex (WRC), an effector of Rac1 during cell protrusion, is concentrated at the lamellipodium tip. Thus, activated Rac1 should operate at this location to activate WRC and trigger membrane protrusion. Yet correlation of Rho GTPase activation with cycles of membrane protrusion previously revealed complex spatiotemporal patterns of Rac1 and RhoA activation in the lamellipodium. Combining single protein tracking (SPT) and super-resolution imaging with loss- or gain-of-function mutants of Rho GTPases, we show that Rac1 immobilizations at the lamellipodium tip correlate with its activation, in contrast to RhoA. Using Rac1 effector loop mutants and wild-type versus mutant variants of WRC, we show that selective immobilizations of activated Rac1 at the lamellipodium tip depend on effector binding, including WRC. In contrast, wild-type Rac1 only displays slower diffusion at the lamellipodium tip, suggesting transient activations. Local optogenetic activation of Rac1, triggered by membrane recruitment of Tiam1, shows that Rac1 activation must occur close to the lamellipodium tip and not behind the lamellipodium to trigger efficient membrane protrusion. However, coupling tracking with optogenetic activation of Rac1 demonstrates that diffusive properties of wild-type Rac1 are unchanged despite enhanced lamellipodium protrusion. Taken together, our results support a model whereby transient activations of Rac1 occurring close to the lamellipodium tip trigger WRC binding. This short-lived activation ensures a local and rapid control of Rac1 actions on its effectors to trigger actin-based protrusion.


Asunto(s)
Movimiento Celular , Extensiones de la Superficie Celular/metabolismo , Fibroblastos/metabolismo , Neuropéptidos/metabolismo , Seudópodos/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Embrión de Mamíferos/metabolismo , Ratones , Proteína de Unión al GTP rhoA/metabolismo
20.
Methods Mol Biol ; 1749: 291-311, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29526005

RESUMEN

To get a complete understanding of cell migration, it is critical to study its orchestration at the molecular level. Since the recent developments in single-molecule imaging, it is now possible to study molecular phenomena at the single-molecule level inside living cells. In this chapter, we describe how such approaches have been and can be used to decipher molecular mechanisms involved in cell migration.


Asunto(s)
Movimiento Celular/fisiología , Transporte de Proteínas/fisiología , Animales , Línea Celular , Humanos , Ratones , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda