Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 573
Filtrar
1.
Development ; 148(10)2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33999996

RESUMEN

Movement of epithelial cells in a tissue occurs through neighbor exchange and drives tissue shape changes. It requires intercellular junction remodeling, a process typically powered by the contractile actomyosin cytoskeleton. This has been investigated mainly in homogeneous epithelia, where intercalation takes minutes. However, in some tissues, intercalation involves different cell types and can take hours. Whether slow and fast intercalation share the same mechanisms remains to be examined. To address this issue, we used the fly eye, where the cone cells exchange neighbors over ∼10 h to shape the lens. We uncovered three pathways regulating this slow mode of cell intercalation. First, we found a limited requirement for MyosinII. In this case, mathematical modeling predicts an adhesion-dominant intercalation mechanism. Genetic experiments support this prediction, revealing a role for adhesion through the Nephrin proteins Roughest and Hibris. Second, we found that cone cell intercalation is regulated by the Notch pathway. Third, we show that endocytosis is required for membrane removal and Notch activation. Taken together, our work indicates that adhesion, endocytosis and Notch can direct slow cell intercalation during tissue morphogenesis.


Asunto(s)
Adhesión Celular/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/embriología , Endocitosis/fisiología , Receptores Notch/metabolismo , Retina/embriología , Células Fotorreceptoras Retinianas Conos/metabolismo , Actomiosina/metabolismo , Uniones Adherentes/fisiología , Animales , Tipificación del Cuerpo/fisiología , Moléculas de Adhesión Celular Neuronal/metabolismo , Comunicación Celular , Proteínas de Drosophila/genética , Células Epiteliales/citología , Proteínas del Ojo/metabolismo , Adhesiones Focales/fisiología , Proteínas de la Membrana/metabolismo , Miosina Tipo II/metabolismo , Receptores Notch/genética , Transducción de Señal/fisiología
2.
Nano Lett ; 23(7): 2467-2475, 2023 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-36975035

RESUMEN

Mechanical signals establish two-way communication between mammalian cells and their environment. Cells contacting a surface exert forces via contractility and transmit them at the areas of focal adhesions. External stimuli, such as compressive and pulling forces, typically affect the adhesion-free cell surface. Here, we demonstrate the collaborative employment of Fluidic Force Microscopy and confocal Traction Force Microscopy supported by the Cellogram solver to enable a powerful integrated force probing approach, where controlled vertical forces are applied to the free surface of individual cells, while the concomitant deformations are used to map their transmission to the substrate. Force transmission across human cells is measured with unprecedented temporal and spatial resolution, enabling the investigation of the cellular mechanisms involved in the adaptation, or maladaptation, to external mechanical stimuli. Altogether, the system enables facile and precise force interrogation of individual cells, with the capacity to perform population-based analysis.


Asunto(s)
Adhesión Celular , Matriz Extracelular , Adhesiones Focales , Mecanotransducción Celular , Animales , Humanos , Adhesión Celular/fisiología , Membrana Celular/fisiología , Adhesiones Focales/metabolismo , Adhesiones Focales/fisiología , Mamíferos/anatomía & histología , Mamíferos/fisiología , Fenómenos Mecánicos , Mecanotransducción Celular/fisiología , Microscopía de Fuerza Atómica/métodos , Matriz Extracelular/fisiología
3.
Development ; 147(7)2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-32108024

RESUMEN

Endothelial cell adhesion is implicated in blood vessel sprout formation, yet how adhesion controls angiogenesis, and whether it occurs via rapid remodeling of adherens junctions or focal adhesion assembly, or both, remains poorly understood. Furthermore, how endothelial cell adhesion is controlled in particular tissues and under different conditions remains unexplored. Here, we have identified an unexpected role for spatiotemporal c-Src activity in sprouting angiogenesis in the retina, which is in contrast to the dominant focus on the role of c-Src in the maintenance of vascular integrity. Thus, mice specifically deficient in endothelial c-Src displayed significantly reduced blood vessel sprouting and loss in actin-rich filopodial protrusions at the vascular front of the developing retina. In contrast to what has been observed during vascular leakage, endothelial cell-cell adhesion was unaffected by loss of c-Src. Instead, decreased angiogenic sprouting was due to loss of focal adhesion assembly and cell-matrix adhesion, resulting in loss of sprout stability. These results demonstrate that c-Src signaling at specified endothelial cell membrane compartments (adherens junctions or focal adhesions) control vascular processes in a tissue- and context-dependent manner.


Asunto(s)
Adhesión Celular/genética , Células Endoteliales/fisiología , Adhesiones Focales/genética , Genes src/fisiología , Neovascularización Fisiológica/genética , Retina/embriología , Animales , Células Cultivadas , Embrión de Mamíferos , Endotelio Vascular/embriología , Endotelio Vascular/metabolismo , Femenino , Adhesiones Focales/metabolismo , Adhesiones Focales/fisiología , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Noqueados , Retina/metabolismo
4.
PLoS Pathog ; 17(1): e1009065, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33508039

RESUMEN

Bartonella T4SS effector BepC was reported to mediate internalization of big Bartonella aggregates into host cells by modulating F-actin polymerization. After that, BepC was indicated to induce host cell fragmentation, an interesting cell phenotype that is characterized by failure of rear-end retraction during cell migration, and subsequent dragging and fragmentation of cells. Here, we found that expression of BepC resulted in significant stress fiber formation and contractile cell morphology, which depended on combination of the N-terminus FIC (filamentation induced by c-AMP) domain and C-terminus BID (Bartonella intracellular delivery) domain of BepC. The FIC domain played a key role in BepC-induced stress fiber formation and cell fragmentation because deletion of FIC signature motif or mutation of two conserved amino acid residues abolished BepC-induced cell fragmentation. Immunoprecipitation confirmed the interaction of BepC with GEF-H1 (a microtubule-associated RhoA guanosine exchange factor), and siRNA-mediated depletion of GEF-H1 prevented BepC-induced stress fiber formation. Interaction with BepC caused the dissociation of GEF-H1 from microtubules and activation of RhoA to induce formation of stress fibers. The ROCK (Rho-associated protein kinase) inhibitor Y27632 completely blocked BepC effects on stress fiber formation and cell contractility. Moreover, stress fiber formation by BepC increased the stability of focal adhesions, which consequently impeded rear-edge detachment. Overall, our study revealed that BepC-induced stress fiber formation was achieved through the GEF-H1/RhoA/ROCK pathway.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Bartonella/metabolismo , Membrana Celular/metabolismo , Adhesiones Focales/fisiología , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Fibras de Estrés/fisiología , Sistemas de Secreción Tipo IV/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Movimiento Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Microtúbulos/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/genética , Sistemas de Secreción Tipo IV/genética
5.
Proc Natl Acad Sci U S A ; 117(51): 32413-32422, 2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33262280

RESUMEN

Integrin-dependent adhesions mediate reciprocal exchange of force and information between the cell and the extracellular matrix. These effects are attributed to the "focal adhesion clutch," in which moving actin filaments transmit force to integrins via dynamic protein interactions. To elucidate these processes, we measured force on talin together with actin flow speed. While force on talin in small lamellipodial adhesions correlated with actin flow, talin tension in large adhesions further from the cell edge was mainly flow-independent. Stiff substrates shifted force transfer toward the flow-independent mechanism. Flow-dependent force transfer required talin's C-terminal actin binding site, ABS3, but not vinculin. Flow-independent force transfer initially required vinculin and at later times the central actin binding site, ABS2. Force transfer through integrins thus occurs not through a continuous clutch but through a series of discrete states mediated by distinct protein interactions, with their ratio modulated by substrate stiffness.


Asunto(s)
Actinas/metabolismo , Integrinas/metabolismo , Actinas/genética , Animales , Sitios de Unión , Transferencia Resonante de Energía de Fluorescencia , Adhesiones Focales/fisiología , Ratones , Mutación , Células 3T3 NIH , Talina/genética , Talina/metabolismo , Imagen de Lapso de Tiempo , Vinculina/genética , Vinculina/metabolismo
6.
Biophys J ; 121(9): 1777-1786, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35306023

RESUMEN

Tissue stiffness (Young's modulus) is a key control parameter in cell behavior and bioengineered gels where defined mechanical properties have become an essential part of the toolkit for interrogating mechanotransduction. Here, we show using a mechanical cell model that the effective substrate stiffness experienced by a cell depends, not just on the engineered mechanical properties of the substrate but critically also on the particular arrangement of adhesions between cell and substrate. In particular, we find that cells with different adhesion patterns can experience two different gel stiffnesses as equivalent and will generate the same mean cell deformations. In considering small patches of adhesion, which mimic focal adhesion complexes, we show how the experimentally observed focal adhesion growth and elongation on stiff substrates can be explained by energy considerations. Relatedly, energy arguments also provide a reason why nascent adhesions do not establish into focal adhesions on soft substrates, as has been commonly observed. Fewer and larger adhesions are predicted to be preferred over more and smaller, an effect enhanced by random spot placing with the simulations predicting qualitatively realistic cell shapes in this case.


Asunto(s)
Adhesiones Focales , Mecanotransducción Celular , Adhesión Celular/fisiología , Forma de la Célula , Módulo de Elasticidad , Adhesiones Focales/fisiología , Mecanotransducción Celular/fisiología
7.
Dev Biol ; 469: 125-134, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33096063

RESUMEN

Collective cell migration is a process where cohorts of cells exhibit coordinated migratory behavior. During individual and collective cellular migration, cells must extend protrusions to interact with the extracellular environment, sense chemotactic cues, and act as points of attachment. The mechanisms and regulators of protrusive behavior have been widely studied in individually migrating cells; however, how this behavior is regulated throughout collectives is not well understood. To address this, we used the zebrafish posterior lateral line primordium (pLLP) as a model. The pLLP is a cluster of ~150 â€‹cells that migrates along the zebrafish trunk, depositing groups of cells that will become sensory organs. To define protrusive behavior, we performed mosaic analysis to sparsely label pLLP cells with a transgene marking filamentous actin. This approach revealed an abundance of brush-like protrusions throughout the pLLP that orient in the direction of migration. Formation of these protrusions depends on the Arp2/3 complex, a regulator of dendritic actin. This argues that these brush-like protrusions are an in vivo example of lamellipodia. Mosaic analysis demonstrated that these lamellipodia-like protrusions are located in a close proximity to the overlying skin. Immunostaining revealed an abundance of focal adhesion complexes surrounding the pLLP. Disruption of these complexes specifically in pLLP cells led to impaired pLLP migration. Finally, we show that Erk signaling, a known regulator of focal adhesions, is required for proper formation of lamellipodia-like protrusions and pLLP migration. Altogether, our results suggest a model where the coordinated dynamics of lamellipodia-like protrusions, making contact with either the overlying skin or the extracellular matrix through focal adhesions, promotes migration of pLLP cells.


Asunto(s)
Movimiento Celular , Adhesiones Focales/fisiología , Seudópodos/fisiología , Pez Cebra/embriología , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Actinas/análisis , Animales , Sistema de Señalización de MAP Quinasas , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Seudópodos/enzimología , Seudópodos/metabolismo , Pez Cebra/fisiología
8.
J Biol Chem ; 296: 100481, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33647313

RESUMEN

The extracellular matrix (ECM) plays an important role in maintaining tissue homeostasis and poses a significant physical barrier to in vivo cell migration. Accordingly, as a means of enhancing tissue invasion, tumor cells use matrix metalloproteinases to degrade ECM proteins. However, the in vivo ECM is comprised not only of proteins but also of a variety of nonprotein components. Hyaluronan (HA), one of the most abundant nonprotein components of the interstitial ECM, forms a gel-like antiadhesive barrier that is impenetrable to particulate matter and cells. Mechanisms by which tumor cells penetrate the HA barrier have not been addressed. Here, we demonstrate that transmembrane protein 2 (TMEM2), the only known transmembrane hyaluronidase, is the predominant mediator of contact-dependent HA degradation and subsequent integrin-mediated cell-substrate adhesion. We show that a variety of tumor cells are able to eliminate substrate-bound HA in a tightly localized pattern corresponding to the distribution of focal adhesions (FAs) and stress fibers. This FA-targeted HA degradation is mediated by TMEM2, which itself is localized at site of FAs. TMEM2 depletion inhibits the ability of tumor cells to attach and migrate in an HA-rich environment. Importantly, TMEM2 directly binds at least two integrins via interaction between extracellular domains. Our findings demonstrate a critical role for TMEM2-mediated HA degradation in the adhesion and migration of cells on HA-rich ECM substrates and provide novel insight into the early phase of FA formation.


Asunto(s)
Ácido Hialurónico/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Adhesión Celular/fisiología , Línea Celular Tumoral , Membrana Celular/metabolismo , Movimiento Celular/fisiología , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Adhesiones Focales/fisiología , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/fisiología , Hialuronoglucosaminidasa/metabolismo , Integrinas/metabolismo , Proteínas de la Membrana/fisiología , Ratones
9.
PLoS Biol ; 17(3): e3000057, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30917109

RESUMEN

Cells in developing organisms are subjected to particular mechanical forces that shape tissues and instruct cell fate decisions. How these forces are sensed and transmitted at the molecular level is therefore an important question, one that has mainly been investigated in cultured cells in vitro. Here, we elucidate how mechanical forces are transmitted in an intact organism. We studied Drosophila muscle attachment sites, which experience high mechanical forces during development and require integrin-mediated adhesion for stable attachment to tendons. Therefore, we quantified molecular forces across the essential integrin-binding protein Talin, which links integrin to the actin cytoskeleton. Generating flies expressing 3 Förster resonance energy transfer (FRET)-based Talin tension sensors reporting different force levels between 1 and 11 piconewton (pN) enabled us to quantify physiologically relevant molecular forces. By measuring primary Drosophila muscle cells, we demonstrate that Drosophila Talin experiences mechanical forces in cell culture that are similar to those previously reported for Talin in mammalian cell lines. However, in vivo force measurements at developing flight muscle attachment sites revealed that average forces across Talin are comparatively low and decrease even further while attachments mature and tissue-level tension remains high. Concomitantly, the Talin concentration at attachment sites increases 5-fold as quantified by fluorescence correlation spectroscopy (FCS), suggesting that only a small proportion of Talin molecules are mechanically engaged at any given time. Reducing Talin levels at late stages of muscle development results in muscle-tendon rupture in the adult fly, likely as a result of active muscle contractions. We therefore propose that a large pool of adhesion molecules is required to share high tissue forces. As a result, less than 15% of the molecules experience detectable forces at developing muscle attachment sites at the same time. Our findings define an important new concept of how cells can adapt to changes in tissue mechanics to prevent mechanical failure in vivo.


Asunto(s)
Desarrollo de Músculos/fisiología , Sarcómeros/metabolismo , Talina/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Animales , Western Blotting , Células Cultivadas , Drosophila , Matriz Extracelular/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Adhesiones Focales/metabolismo , Adhesiones Focales/fisiología , Integrinas/genética , Integrinas/metabolismo , Masculino , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/metabolismo , Unión Proteica , Talina/genética , Tendones/metabolismo
10.
Biol Pharm Bull ; 45(2): 207-212, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35110508

RESUMEN

Octa-arginine (R8) has been extensively studied as a cell-penetrating peptide. R8 binds to diverse transmembrane heparan sulfate proteoglycans (HSPGs), including syndecans, and is internalized by cells. R8 is also reported to bind to integrin ß1. In this study, we evaluated the biological activities of R8 and octa-lysine (K8), a peptide similar to R8, with a focus on cell adhesion. R8 and K8 were immobilized on aldehyde-agarose matrices via covalent conjugation, and the effect of these peptides on cell attachment, spreading, and proliferation was examined using human dermal fibroblasts. The results indicated that R8- and K8-matrices mediate cell adhesion mainly via HSPGs. Moreover, R8- and K8-matrices interacted with integrin ß1 and promote cell spreading and proliferation. These results are useful for further understanding of the R8-membrane interactions and the cellular uptake mechanisms. In addition, the R8- and K8-matrices may potentially be used as a multi-functional biomaterial to promote cell adhesion, spreading, and proliferation.


Asunto(s)
Fibroblastos/efectos de los fármacos , Proteoglicanos de Heparán Sulfato/metabolismo , Integrinas/metabolismo , Lisina/química , Lisina/farmacología , Oligopéptidos/farmacología , Actinas/fisiología , Adhesión Celular/efectos de los fármacos , Proliferación Celular , Ácido Edético/farmacología , Fibroblastos/fisiología , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/fisiología , Heparina/farmacología , Humanos
11.
Proc Natl Acad Sci U S A ; 116(44): 22004-22013, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31570575

RESUMEN

The ability of cells to perceive the mechanical identity of extracellular matrix, generally known as mechanosensing, is generally depicted as a consequence of an intricate balance between pulling forces actuated by the actin fibers on the adhesion plaques and the mechanical reaction of the supporting material. However, whether the cell is sensitive to the stiffness or to the energy required to deform the material remains unclear. To address this important issue, here the cytoskeleton mechanics of BALB/3T3 and MC3T3 cells seeded on linearly elastic substrates under different levels of deformation were studied. In particular, the effect of prestrain on cell mechanics was evaluated by seeding cells both on substrates with no prestrain and on substrates with different levels of prestrain. Results indicated that cells recognize the existence of prestrain, exhibiting a stiffer cytoskeleton on stretched material compared to cells seeded on unstretched substrate. Cytoskeleton mechanics of cells seeded on stretched material were, in addition, comparable to those measured after the stretching of the substrate and cells together to the same level of deformation. This observation clearly suggests that cell mechanosensing is not mediated only by the stiffness of the substrate, as widely assumed in the literature, but also by the deformation energy associated with the substrate. Indeed, the clutch model, based on the exclusive dependence of cell mechanics upon substrate stiffness, fails to describe our experimental results. By modifying the clutch model equations to incorporate the dependence on the strain energy, we were able to correctly interpret the experimental evidence.


Asunto(s)
Mecanotransducción Celular/fisiología , Animales , Línea Celular , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiología , Adhesiones Focales/metabolismo , Adhesiones Focales/fisiología , Ratones , Ratones Endogámicos BALB C , Células 3T3 NIH
12.
J Cell Sci ; 132(8)2019 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-30890648

RESUMEN

Heterodimeric integrin receptors control cell adhesion, migration and extracellular matrix assembly. While the α integrin subunit determines extracellular ligand specificity, the ß integrin chain binds to an acidic residue of the ligand, and cytoplasmic adapter protein families such as talins, kindlins and paxillin, to form mechanosensing cell matrix adhesions. Alternative splicing of the ß1 integrin cytoplasmic tail creates ubiquitously expressed ß1A, and the heart and skeletal muscle-specific ß1D form. To study the physiological difference between these forms, we developed fluorescent ß1 integrins and analyzed their dynamics, localization, and cytoplasmic adapter recruitment and effects on cell proliferation. On fibronectin, GFP-tagged ß1A integrin showed dynamic exchange in peripheral focal adhesions, and long, central fibrillar adhesions. In contrast, GFP-ß1D integrins exchanged slowly, forming immobile and short central adhesions. While adhesion recruitment of GFP-ß1A integrin was sensitive to C-terminal tail mutagenesis, GFP-ß1D integrin was recruited independently of the distal NPXY motif. In addition, a P786A mutation in the proximal, talin-binding NPXY783 motif switched ß1D to a highly dynamic integrin. In contrast, the inverse A786P mutation in ß1A integrin interfered with paxillin recruitment and proliferation. Thus, differential ß1 integrin splicing controls integrin-dependent adhesion signaling, to adapt to the specific physiological needs of differentiated muscle cells.


Asunto(s)
Empalme Alternativo , Integrina beta1/metabolismo , Paxillin/metabolismo , Transducción de Señal , Animales , Proliferación Celular , Citoplasma/metabolismo , Citoesqueleto/metabolismo , Fibronectinas/fisiología , Adhesiones Focales/fisiología , Ratones , Músculo Esquelético/metabolismo , Células 3T3 NIH
13.
J Cell Sci ; 132(11)2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31152052

RESUMEN

Cancer cells are softer than the normal cells, and metastatic cells are even softer. These changes in biomechanical properties contribute to cancer progression by facilitating cell movement through physically constraining environments. To identify properties that enabled passage through physical constraints, cells that were more efficient at moving through narrow membrane micropores were selected from established cell lines. By examining micropore-selected human MDA MB 231 breast cancer and MDA MB 435 melanoma cancer cells, membrane fluidity and nuclear elasticity were excluded as primary contributors. Instead, reduced actin cytoskeleton anisotropy, focal adhesion density and cell stiffness were characteristics associated with efficient passage through constraints. By comparing transcriptomic profiles between the parental and selected populations, increased Ras/MAPK signalling was linked with cytoskeleton rearrangements and cell softening. MEK inhibitor treatment reversed the transcriptional, cytoskeleton, focal adhesion and elasticity changes. Conversely, expression of oncogenic KRas in parental MDA MB 231 cells, or oncogenic BRaf in parental MDA MB 435 cells, significantly reduced cell stiffness. These results reveal that MAPK signalling, in addition to tumour cell proliferation, has a significant role in regulating cell biomechanics.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Citoesqueleto de Actina/fisiología , Fenómenos Biomecánicos/fisiología , Movimiento Celular/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Melanoma/fisiopatología , Anisotropía , Línea Celular Tumoral , Plasticidad de la Célula/fisiología , Proliferación Celular , Adhesiones Focales/fisiología , Humanos , Filtros Microporos , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/patología , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
14.
Nanotechnology ; 32(21)2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33596559

RESUMEN

The procedure commonly adopted to characterize cell materials using atomic force microscopy neglects the stress state induced in the cell by the adhesion structures that anchor it to the substrate. In several studies, the cell is considered as made from a single material and no specific information is provided regarding the mechanical properties of subcellular components. Here we present an optimization algorithm to determine separately the material properties of subcellular components of mesenchymal stem cells subjected to nanoindentation measurements. We assess how these properties change if the adhesion structures at the cell-substrate interface are considered or not in the algorithm. In particular, among the adhesion structures, the focal adhesions and the stress fibers were simulated. We found that neglecting the adhesion structures leads to underestimate the cell mechanical properties thus making errors up to 15%. This result leads us to conclude that the action of adhesion structures should be taken into account in nanoindentation measurements especially for cells that include a large number of adhesions to the substrate.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Microscopía de Fuerza Atómica/métodos , Modelos Biológicos , Algoritmos , Fenómenos Biomecánicos , Adhesión Celular , Análisis de Elementos Finitos , Adhesiones Focales/fisiología , Humanos , Fibras de Estrés/fisiología
15.
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
16.
Bioessays ; 41(1): e1800165, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30485463

RESUMEN

Focal adhesions disassemble during mitosis, but surprisingly little is known about how these structures respond to other phases of the cell cycle. Three recent papers reveal unexpected results as they examine adhesions through the cell cycle. A biphasic response is detected where focal adhesions grow during S phase before disassembly begins early in G2. In M phase, activated integrins at the tips of retraction fibers anchor mitotic cells, but these adhesions lack the defining components of focal adhesions, such as talin, paxillin, and zyxin. Re-examining cell-matrix adhesion reveals reticular adhesions, a new class of adhesion. These αVß5 integrin-mediated adhesions also lack conventional focal adhesion components and anchor mitotic cells to the extracellular matrix. As reviewed here, these studies present insight into how adhesion complexes vary through the cell cycle, and how unconventional adhesions maintain attachment during mitosis while providing spatial memory to guide daughter cell re-spreading after cell division.


Asunto(s)
Puntos de Control del Ciclo Celular , Adhesiones Focales/fisiología , Animales , Adhesión Celular , Adhesiones Focales/metabolismo , Humanos
17.
Proc Natl Acad Sci U S A ; 115(41): 10339-10344, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30254158

RESUMEN

Multicellular organisms have well-defined, tightly regulated mechanisms for cell adhesion. Heterodimeric αß integrin receptors play central roles in this function and regulate processes for normal cell functions, including signaling, cell migration, and development, binding to the extracellular matrix, and senescence. They are involved in hemostasis and the immune response, participate in leukocyte function, and have biological implications in angiogenesis and cancer. Proper control of integrin activation for cellular communication with the external environment requires several physiological processes. Perturbation of these equilibria may lead to constitutive integrin activation that results in bleeding disorders. Furthermore, integrins play key roles in cancer progression and metastasis in which certain tumor types exhibit higher levels of various integrins. Thus, the integrin-associated signaling complex is important for cancer therapy development. During inside-out signaling, the cytoskeletal protein talin plays a key role in regulating integrin affinity whereby the talin head domain activates integrin by binding to the cytoplasmic tail of ß-integrin and acidic membrane phospholipids. To understand the mechanism of integrin activation by talin, we determined the crystal structure of the talin head domain bound to the acidic phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2), allowing us to design a lipid-binding-deficient talin mutant. Our confocal microscopy with talin knockout cells suggests that the talin-cell membrane interaction seems essential for focal adhesion formation and stabilization. Basal integrin activation in Chinese hamster ovary cells suggests that the lipid-binding-deficient talin mutant inhibits integrin activation. Thus, membrane attachment of talin seems necessary for integrin activation and focal adhesion formation.


Asunto(s)
Membrana Celular/metabolismo , Adhesiones Focales/fisiología , Integrinas/metabolismo , Talina/química , Talina/metabolismo , Regulación Alostérica , Animales , Sitios de Unión , Células CHO , Cricetulus , Cristalografía por Rayos X , Modelos Moleculares , Fosfatidilinositol 4,5-Difosfato/química , Fosfatidilinositol 4,5-Difosfato/metabolismo , Conformación Proteica , Dominios Proteicos , Talina/genética
18.
Proc Natl Acad Sci U S A ; 115(25): E5696-E5705, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29866846

RESUMEN

Recent studies have revealed pronounced effects of the spatial distribution of EphA2 receptors on cellular response to receptor activation. However, little is known about molecular mechanisms underlying this spatial sensitivity, in part due to lack of experimental systems. Here, we introduce a hybrid live-cell patterned supported lipid bilayer experimental platform in which the sites of EphA2 activation and integrin adhesion are spatially controlled. Using a series of live-cell imaging and single-molecule tracking experiments, we map the transmission of signals from ephrinA1:EphA2 complexes. Results show that ligand-dependent EphA2 activation induces localized myosin-dependent contractions while simultaneously increasing focal adhesion dynamics throughout the cell. Mechanistically, Src kinase is activated at sites of ephrinA1:EphA2 clustering and subsequently diffuses on the membrane to focal adhesions, where it up-regulates FAK and paxillin tyrosine phosphorylation. EphrinA1:EphA2 signaling triggers multiple cellular responses with differing spatial dependencies to enable a directed migratory response to spatially resolved contact with ephrinA1 ligands.


Asunto(s)
Adhesión Celular/fisiología , Movimiento Celular/fisiología , Efrina-A1/metabolismo , Adhesiones Focales/metabolismo , Adhesiones Focales/fisiología , Receptor EphA2/metabolismo , Transducción de Señal/fisiología , Línea Celular Tumoral , Humanos , Ligandos , Membrana Dobles de Lípidos/metabolismo , Miosinas/metabolismo , Paxillin/metabolismo , Fosforilación/fisiología , Regulación hacia Arriba/fisiología , Familia-src Quinasas/metabolismo
19.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34768998

RESUMEN

Mechanical cues are crucial for survival, adaptation, and normal homeostasis in virtually every cell type. The transduction of mechanical messages into intracellular biochemical messages is termed mechanotransduction. While significant advances in biochemical signaling have been made in the last few decades, the role of mechanotransduction in physiological and pathological processes has been largely overlooked until recently. In this review, the role of interactions between the cytoskeleton and cell-cell/cell-matrix adhesions in transducing mechanical signals is discussed. In addition, mechanosensors that reside in the cell membrane and the transduction of mechanical signals to the nucleus are discussed. Finally, we describe two examples in which mechanotransduction plays a significant role in normal physiology and disease development. The first example is the role of mechanotransduction in the proliferation and metastasis of cancerous cells. In this system, the role of mechanotransduction in cellular processes, including proliferation, differentiation, and motility, is described. In the second example, the role of mechanotransduction in a mechanically active organ, the gastrointestinal tract, is described. In the gut, mechanotransduction contributes to normal physiology and the development of motility disorders.


Asunto(s)
Membrana Celular/fisiología , Citoesqueleto/fisiología , Mecanotransducción Celular/fisiología , Animales , Núcleo Celular/fisiología , Adhesiones Focales/fisiología , Humanos
20.
Int J Mol Sci ; 22(12)2021 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-34205295

RESUMEN

Mesenchymal stem cells (MSCs) maintain the musculoskeletal system by differentiating into multiple lineages, including osteoblasts and adipocytes. Mechanical signals, including strain and low-intensity vibration (LIV), are important regulators of MSC differentiation via control exerted through the cell structure. Lamin A/C is a protein vital to the nuclear architecture that supports chromatin organization and differentiation and contributes to the mechanical integrity of the nucleus. We investigated whether lamin A/C and mechanoresponsiveness are functionally coupled during adipogenesis in MSCs. siRNA depletion of lamin A/C increased the nuclear area, height, and volume and decreased the circularity and stiffness. Lamin A/C depletion significantly decreased markers of adipogenesis (adiponectin, cellular lipid content) as did LIV treatment despite depletion of lamin A/C. Phosphorylation of focal adhesions in response to mechanical challenge was also preserved during loss of lamin A/C. RNA-seq showed no major adipogenic transcriptome changes resulting from LIV treatment, suggesting that LIV regulation of adipogenesis may not occur at the transcriptional level. We observed that during both lamin A/C depletion and LIV, interferon signaling was downregulated, suggesting potentially shared regulatory mechanism elements that could regulate protein translation. We conclude that the mechanoregulation of adipogenesis and the mechanical activation of focal adhesions function independently from those of lamin A/C.


Asunto(s)
Adipogénesis , Adhesiones Focales/fisiología , Lamina Tipo A/fisiología , Células Madre Mesenquimatosas/fisiología , Animales , Módulo de Elasticidad , Interferones/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Transducción de Señal , Proteínas de Unión a Telómeros/metabolismo , Vibración
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA