Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(13): e2314588121, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38502691

RESUMEN

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


Asunto(s)
Tenascina , Pez Cebra , Animales , Tenascina/genética , Pez Cebra/genética , Pez Cebra/metabolismo , Axones/metabolismo , Colágeno/metabolismo , Matriz Extracelular/metabolismo
2.
J Cell Sci ; 134(2)2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33495358

RESUMEN

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


Asunto(s)
Citoesqueleto , Proteómica , Transducción de Señal , Familia-src Quinasas , Animales , Células Cultivadas , Simulación de Dinámica Molecular , Fosforilación , Dominios Homologos src , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
3.
EMBO Rep ; 20(9): e48084, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31368207

RESUMEN

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


Asunto(s)
Núcleo Celular/metabolismo , Mecanotransducción Celular/fisiología , Membrana Nuclear/metabolismo , Factores de Transcripción/metabolismo , Ciclo Celular/genética , Ciclo Celular/fisiología , División Celular/genética , División Celular/fisiología , Línea Celular , Núcleo Celular/genética , Citometría de Flujo , Fase G1/genética , Fase G1/fisiología , Células HeLa , Humanos , Mecanotransducción Celular/genética , Microscopía de Fuerza Atómica , Membrana Nuclear/genética , Plásmidos/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Fase S/genética , Fase S/fisiología , Factores de Transcripción/genética
4.
J Cell Sci ; 131(15)2018 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-30030370

RESUMEN

Endothelial integrity relies on a mechanical crosstalk between intercellular and cell-matrix interactions. This crosstalk is compromised in hemorrhagic vascular lesions of patients carrying loss-of-function mutations in cerebral cavernous malformation (CCM) genes. RhoA/ROCK-dependent cytoskeletal remodeling is central to the disease, as it causes unbalanced cell adhesion towards increased cell-extracellular matrix adhesions and destabilized cell-cell junctions. This study reveals that CCM proteins directly orchestrate ROCK1 and ROCK2 complementary roles on the mechanics of the endothelium. CCM proteins act as a scaffold, promoting ROCK2 interactions with VE-cadherin and limiting ROCK1 kinase activity. Loss of CCM1 (also known as KRIT1) produces excessive ROCK1-dependent actin stress fibers and destabilizes intercellular junctions. Silencing of ROCK1 but not ROCK2 restores the adhesive and mechanical homeostasis of CCM1 and CCM2-depleted endothelial monolayers, and rescues the cardiovascular defects of ccm1 mutant zebrafish embryos. Conversely, knocking down Rock2 but not Rock1 in wild-type zebrafish embryos generates defects reminiscent of the ccm1 mutant phenotypes. Our study uncovers the role of the CCM1-CCM2 complex in controlling ROCK1 and ROCK2 to preserve endothelial integrity and drive heart morphogenesis. Moreover, it solely identifies the ROCK1 isoform as a potential therapeutic target for the CCM disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Células Endoteliales/metabolismo , Proteína KRIT1/metabolismo , Quinasas Asociadas a rho/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Western Blotting , Cadherinas/genética , Cadherinas/metabolismo , Proteínas Portadoras/genética , Bovinos , Células Endoteliales/citología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inmunoprecipitación , Proteína KRIT1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Pez Cebra , Quinasas Asociadas a rho/genética
5.
Exp Cell Res ; 378(1): 113-117, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30794802

RESUMEN

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


Asunto(s)
Mecanorreceptores/metabolismo , Mecanotransducción Celular , Animales , Retroalimentación Fisiológica , Adhesiones Focales/metabolismo , Humanos , Mecanorreceptores/fisiología
6.
Proc Natl Acad Sci U S A ; 114(8): E1413-E1421, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28174275

RESUMEN

Fibroblasts are a main player in the tumor-inhibitory microenvironment. Upon tumor initiation and progression, fibroblasts can lose their tumor-inhibitory capacity and promote tumor growth. The molecular mechanisms that underlie this switch have not been defined completely. Previously, we identified four proteins overexpressed in cancer-associated fibroblasts and linked to Rho GTPase signaling. Here, we show that knocking out the Ras homolog family member A (RhoA) gene in normal fibroblasts decreased their tumor-inhibitory capacity, as judged by neighbor suppression in vitro and accompanied by promotion of tumor growth in vivo. This also induced PC3 cancer cell motility and increased colony size in 2D cultures. RhoA knockout in fibroblasts induced vimentin intermediate filament reorganization, accompanied by reduced contractile force and increased stiffness of cells. There was also loss of wide F-actin stress fibers and large focal adhesions. In addition, we observed a significant loss of α-smooth muscle actin, which indicates a difference between RhoA knockout fibroblasts and classic cancer-associated fibroblasts. In 3D collagen matrix, RhoA knockout reduced fibroblast branching and meshwork formation and resulted in more compactly clustered tumor-cell colonies in coculture with PC3 cells, which might boost tumor stem-like properties. Coculturing RhoA knockout fibroblasts and PC3 cells induced expression of proinflammatory genes in both. Inflammatory mediators may induce tumor cell stemness. Network enrichment analysis of transcriptomic changes, however, revealed that the Rho signaling pathway per se was significantly triggered only after coculturing with tumor cells. Taken together, our findings in vivo and in vitro indicate that Rho signaling governs the inhibitory effects by fibroblasts on tumor-cell growth.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Proliferación Celular/fisiología , Neoplasias/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Actinas/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Células Cultivadas , Colágeno/metabolismo , Femenino , Adhesiones Focales/metabolismo , Células HEK293 , Humanos , Ratones , Ratones SCID , Transducción de Señal/fisiología , Fibras de Estrés/metabolismo , Quinasas Asociadas a rho/metabolismo
7.
J Cell Sci ; 130(3): 626-636, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28049720

RESUMEN

Cell migration is a complex process requiring density and rigidity sensing of the microenvironment to adapt cell migratory speed through focal adhesion and actin cytoskeleton regulation. ICAP-1 (also known as ITGB1BP1), a ß1 integrin partner, is essential for ensuring integrin activation cycle and focal adhesion formation. We show that ICAP-1 is monoubiquitylated by Smurf1, preventing ICAP-1 binding to ß1 integrin. The non-ubiquitylatable form of ICAP-1 modifies ß1 integrin focal adhesion organization and interferes with fibronectin density sensing. ICAP-1 is also required for adapting cell migration in response to substrate stiffness in a ß1-integrin-independent manner. ICAP-1 monoubiquitylation regulates rigidity sensing by increasing MRCKα (also known as CDC42BPA)-dependent cell contractility through myosin phosphorylation independently of substrate rigidity. We provide evidence that ICAP-1 monoubiquitylation helps in switching from ROCK2-mediated to MRCKα-mediated cell contractility. ICAP-1 monoubiquitylation serves as a molecular switch to coordinate extracellular matrix density and rigidity sensing thus acting as a crucial modulator of cell migration and mechanosensing.


Asunto(s)
Movimiento Celular , Matriz Extracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Quinasa de Distrofia Miotónica/metabolismo , Ubiquitinación , Quinasas Asociadas a rho/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Sitios de Unión , Fenómenos Biomecánicos , Adhesión Celular , Línea Celular , Fibronectinas/metabolismo , Adhesiones Focales/metabolismo , Humanos , Integrina beta1/química , Integrina beta1/metabolismo , Ratones , Modelos Biológicos , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo
8.
Phys Rev Lett ; 122(16): 168101, 2019 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-31075005

RESUMEN

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


Asunto(s)
Movimiento Celular , Modelos Biológicos , Animales , Perros , Fibronectinas/metabolismo , Células de Riñón Canino Madin Darby
9.
Biol Cell ; 109(3): 127-137, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27990663

RESUMEN

BACKGROUND INFORMATION: Integrins are key receptors that allow cells to sense and respond to their mechanical environment. Although they bind the same ligand, ß1 and ß3 integrins have distinct and cooperative roles in mechanotransduction. RESULTS: Using traction force microscopy on unconstrained cells, we show that deleting ß3 causes traction forces to increase, whereas the deletion of ß1 integrin results in a strong decrease of contractile forces. Consistently, loss of ß3 integrin also induces an increase in ß1 integrin activation. Using a genetic approach, we identified the phosphorylation of the distal NPXY domain as an essential process for ß3 integrin to be able to modulate traction forces. Loss of ß3 integrins also impacted cell shape and the spatial distribution of traction forces, by causing forces to be generated closer to the cell edge, and the cell shape. CONCLUSIONS: Our results emphasize the role of ß3 integrin in spatial distribution of cellular forces. We speculate that, by modulating its affinity with kindlin, ß3 integrins may be able to locate near the cell edge where it can control ß1 integrin activation and clustering. SIGNIFICANCE: Tensional homeostasis at the single cell level is performed by the ability of ß3 adhesions to negatively regulate the activation degree and spatial localization of ß1 integrins. By combining genetic approaches and new tools to analyze traction distribution and cell morphology on a population of cells we were able to identify the molecular partners involved in cellular forces regulation.


Asunto(s)
Proteínas Portadoras/genética , Fibroblastos/metabolismo , Integrina alfaVbeta3/genética , Integrina beta1/genética , Integrina beta3/genética , Mecanotransducción Celular , Secuencia de Aminoácidos , Animales , Fenómenos Biomecánicos , Proteínas Portadoras/metabolismo , Adhesión Celular , Línea Celular , Fibroblastos/ultraestructura , Eliminación de Gen , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Integrina alfaVbeta3/metabolismo , Integrina beta1/metabolismo , Integrina beta3/metabolismo , Ratones , Fosforilación , Unión Proteica , Dominios Proteicos
10.
Biophys J ; 110(2): 470-480, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26789769

RESUMEN

The cytoskeleton plays a key role in the ability of cells to both resist mechanical stress and generate force, but the precise involvement of intermediate filaments in these processes remains unclear. We focus here on desmin, a type III intermediate filament, which is specifically expressed in muscle cells and serves as a skeletal muscle differentiation marker. By using several complementary experimental techniques, we have investigated the impact of overexpressing desmin and expressing a mutant desmin on the passive and active mechanical properties of C2C12 myoblasts. We first show that the overexpression of wild-type-desmin increases the overall rigidity of the cells, whereas the expression of a mutated E413K desmin does not. This mutation in the desmin gene is one of those leading to desminopathies, a subgroup of myopathies associated with progressive muscular weakness that are characterized by the presence of desmin aggregates and a disorganization of sarcomeres. We show that the expression of this mutant desmin in C2C12 myoblasts induces desmin network disorganization, desmin aggregate formation, and a small decrease in the number and total length of stress fibers. We finally demonstrate that expression of the E413K mutant desmin also alters the traction forces generation of single myoblasts lacking organized sarcomeres.


Asunto(s)
Desmina/metabolismo , Mutación Missense , Mioblastos/metabolismo , Animales , Línea Celular , Desmina/genética , Ratones , Movimiento (Física) , Estructura Terciaria de Proteína , Fibras de Estrés/genética , Fibras de Estrés/metabolismo , Estrés Mecánico
12.
Proc Natl Acad Sci U S A ; 109(5): 1506-11, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22307605

RESUMEN

The organization of cells into epithelium depends on cell interaction with both the extracellular matrix (ECM) and adjacent cells. The role of cell-cell adhesion in the regulation of epithelial topology is well-described. ECM is better known to promote cell migration and provide a structural scaffold for cell anchoring, but its contribution to multicellular morphogenesis is less well-understood. We developed a minimal model system to investigate how ECM affects the spatial organization of intercellular junctions. Fibronectin micropatterns were used to constrain the location of cell-ECM adhesion. We found that ECM affects the degree of stability of intercellular junction positioning and the magnitude of intra- and intercellular forces. Intercellular junctions were permanently displaced, and experienced large perpendicular tensional forces as long as they were positioned close to ECM. They remained stable solely in regions deprived of ECM, where they were submitted to lower tensional forces. The heterogeneity of the spatial organization of ECM induced anisotropic distribution of mechanical constraints in cells, which seemed to adapt their position to minimize both intra- and intercellular forces. These results uncover a morphogenetic role for ECM in the mechanical regulation of cells and intercellular junction positioning.


Asunto(s)
Matriz Extracelular/fisiología , Uniones Intercelulares/fisiología , Línea Celular Tumoral , Humanos
14.
Nat Commun ; 14(1): 717, 2023 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-36759504

RESUMEN

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


Asunto(s)
Colágeno , Fibroblastos , Reología , Ingeniería de Tejidos/métodos
15.
Elife ; 122023 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-37548995

RESUMEN

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


Asunto(s)
Células Epiteliales , Fenómenos Mecánicos , Células Epiteliales/fisiología , Epitelio , Adhesión Celular/fisiología , Elasticidad , Estrés Mecánico
16.
J Cell Biol ; 222(1)2023 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-36250940

RESUMEN

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


Asunto(s)
Clatrina , Endocitosis , Adhesiones Focales , Integrina beta1 , Integrina beta3 , Clatrina/metabolismo , Endocitosis/fisiología , Integrina beta1/genética , Mecanotransducción Celular , Talina/genética
17.
Cancer Gene Ther ; 29(10): 1429-1438, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35379907

RESUMEN

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


Asunto(s)
Actinas , Adhesiones Focales , Factores Despolimerizantes de la Actina/metabolismo , Actinas/genética , Actinas/metabolismo , Movimiento Celular/fisiología , Adhesiones Focales/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptor Relacionado con Estrógeno ERRalfa
18.
Elife ; 112022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35416768

RESUMEN

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


Asunto(s)
Actinas , Actinas/metabolismo , Membrana Celular/metabolismo , Forma de la Célula , Tamaño de la Célula , Retroalimentación , Presión Osmótica
19.
Nat Commun ; 13(1): 6059, 2022 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-36229487

RESUMEN

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


Asunto(s)
Ciclina D1 , Integrinas , División Celular , Ciclina D1/metabolismo , Integrinas/metabolismo , Mecanotransducción Celular/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Proteína de Unión al GTP rac1/metabolismo
20.
Cell Rep ; 36(8): 109616, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34433061

RESUMEN

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


Asunto(s)
Citoesqueleto/metabolismo , Fibronectinas/metabolismo , Integrinas/metabolismo , Neovascularización Fisiológica/fisiología , Angiomotinas/metabolismo , Animales , Membrana Celular/metabolismo , Movimiento Celular/fisiología , Endotelio/metabolismo , Ratones Transgénicos , Sustitutos del Plasma/farmacología , Seudópodos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA