Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Biol Cell ; 35(5): ar65, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38507238

RESUMO

α-catenin (α-cat) displays force-dependent unfolding and binding to actin filaments through direct and indirect means, but features of adherens junction structure and function most vulnerable to loss of these allosteric mechanisms have not been directly compared. By reconstituting an α-cat F-actin-binding domain unfolding mutant known to exhibit enhanced binding to actin (α-cat-H0-FABD+) into α-cat knockout Madin Darby Canine Kidney (MDCK) cells, we show that partial loss of the α-cat catch bond mechanism (via an altered H0 α-helix) leads to stronger epithelial sheet integrity with greater colocalization between the α-cat-H0-FABD+ mutant and actin. α-cat-H0-FABD+ -expressing cells are less efficient at closing scratch-wounds, suggesting reduced capacity for more dynamic cell-cell coordination. Evidence that α-cat-H0-FABD+ is equally accessible to the conformationally sensitive α18 antibody epitope as WT α-cat and shows similar vinculin recruitment suggests this mutant engages lower tension cortical actin networks, as its M-domain is not persistently open. Conversely, α-cat-M-domain salt-bridge mutants with persistent recruitment of vinculin and phosphorylated myosin light chain show only intermediate monolayer adhesive strengths, but display less directionally coordinated and thereby slower migration speeds during wound-repair. These data show α-cat M- and FABD-unfolding mutants differentially impact cell-cell cohesion and migration properties, and suggest signals favoring α-cat-cortical actin interaction without persistent M-domain opening may improve epithelial monolayer strength through enhanced coupling to lower tension actin networks.


Assuntos
Citoesqueleto de Actina , Actinas , Movimento Celular , Células Epiteliais , alfa Catenina , Cães , Animais , alfa Catenina/metabolismo , alfa Catenina/genética , Células Madin Darby de Rim Canino , Actinas/metabolismo , Células Epiteliais/metabolismo , Citoesqueleto de Actina/metabolismo , Ligação Proteica , Domínios Proteicos , Mutação , Junções Aderentes/metabolismo , Desdobramento de Proteína , Adesão Celular/fisiologia , Vinculina/metabolismo
2.
Biophys J ; 123(2): 157-171, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38062704

RESUMO

The actomyosin cytoskeleton generates mechanical forces that power important cellular processes, such as cell migration, cell division, and mechanosensing. Actomyosin self-assembles into contractile networks and bundles that underlie force generation and transmission in cells. A central step is the assembly of the myosin II filament from myosin monomers, regulation of which has been extensively studied. However, myosin filaments are almost always found as clusters within the cell cortex. While recent studies characterized cluster nucleation dynamics at the cell periphery, how myosin clusters grow on stress fibers remains poorly characterized. Here, we utilize a U2OS osteosarcoma cell line with endogenously tagged myosin II to measure the myosin cluster size distribution in the lamella of adherent cells. We find that myosin clusters can grow with Rho-kinase (ROCK) activity alone in the absence of myosin motor activity. Time-lapse imaging reveals that myosin clusters grow via increased myosin association to existing clusters, which is potentiated by ROCK-dependent myosin filament assembly. Enabling myosin motor activity allows further myosin cluster growth through myosin association that is dependent on F-actin architecture. Using a toy model, we show that myosin self-affinity is sufficient to recapitulate the experimentally observed myosin cluster size distribution, and that myosin cluster sizes are determined by the pool of myosin available for cluster growth. Together, our findings provide new insights into the regulation of myosin cluster sizes within the lamellar actomyosin cytoskeleton.


Assuntos
Actinas , Actomiosina , Actinas/metabolismo , Actomiosina/metabolismo , Miosinas/metabolismo , Citoesqueleto de Actina/metabolismo , Miosina Tipo II/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(42): e2305283120, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37819979

RESUMO

From flocks of birds to biomolecular assemblies, systems in which many individual components independently consume energy to perform mechanical work exhibit a wide array of striking behaviors. Methods to quantify the dynamics of these so-called active systems generally aim to extract important length or time scales from experimental fields. Because such methods focus on extracting scalar values, they do not wring maximal information from experimental data. We introduce a method to overcome these limitations. We extend the framework of correlation functions by taking into account the internal headings of displacement fields. The functions we construct represent the material response to specific types of active perturbation within the system. Utilizing these response functions we query the material response of disparate active systems composed of actin filaments and myosin motors, from model fluids to living cells. We show we can extract critical length scales from the turbulent flows of an active nematic, anticipate contractility in an active gel, distinguish viscous from viscoelastic dissipation, and even differentiate modes of contractility in living cells. These examples underscore the vast utility of this method which measures response functions from experimental observations of complex active systems.


Assuntos
Citoesqueleto de Actina , Miosinas , Actomiosina/fisiologia
4.
bioRxiv ; 2023 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-37333106

RESUMO

The actomyosin cytoskeleton generates mechanical forces that power important cellular processes, such as cell migration, cell division, and mechanosensing. Actomyosin self-assembles into contractile networks and bundles that underlie force generation and transmission in cells. A central step is the assembly of the myosin II filament from myosin monomers, regulation of which has been extensively studied. However, myosin filaments are almost always found as clusters within the cell cortex. While recent studies characterized cluster nucleation dynamics at the cell periphery, how myosin clusters grow on stress fibers remains poorly characterized. Here, we utilize a U2OS osteosarcoma cell line with endogenously tagged myosin II to measure the myosin cluster size distribution in the lamella of adherent cells. We find that myosin clusters can grow with Rho-kinase (ROCK) activity alone in the absence of myosin motor activity. Time-lapse imaging reveals that myosin clusters grow via increased myosin association to existing clusters, which is potentiated by ROCK-dependent myosin filament assembly. Enabling myosin motor activity allows further myosin cluster growth through myosin association that is dependent on F-actin architecture. Using a toy model, we show that myosin self-affinity is sufficient to recapitulate the experimentally observed myosin cluster size distribution, and that myosin cluster sizes are determined by the pool of myosin available for cluster growth. Together, our findings provide new insights into the regulation of myosin cluster sizes within the lamellar actomyosin cytoskeleton.

5.
Nat Immunol ; 24(6): 1007-1019, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37069398

RESUMO

Adoptive transfer of genetically engineered chimeric antigen receptor (CAR) T cells is becoming a promising treatment option for hematological malignancies. However, T cell immunotherapies have mostly failed in individuals with solid tumors. Here, with a CRISPR-Cas9 pooled library, we performed an in vivo targeted loss-of-function screen and identified ST3 ß-galactoside α-2,3-sialyltransferase 1 (ST3GAL1) as a negative regulator of the cancer-specific migration of CAR T cells. Analysis of glycosylated proteins revealed that CD18 is a major effector of ST3GAL1 in activated CD8+ T cells. ST3GAL1-mediated glycosylation induces the spontaneous nonspecific tissue sequestration of T cells by altering lymphocyte function-associated antigen-1 (LFA-1) endocytic recycling. Engineered CAR T cells with enhanced expression of ßII-spectrin, a central LFA-1-associated cytoskeleton molecule, reversed ST3GAL1-mediated nonspecific T cell migration and reduced tumor growth in mice by improving tumor-specific homing of CAR T cells. These findings identify the ST3GAL1-ßII-spectrin axis as a major cell-intrinsic program for cancer-targeting CAR T cell migration and as a promising strategy for effective T cell immunotherapy.


Assuntos
Receptores de Antígenos Quiméricos , Animais , Camundongos , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Movimento Celular , Imunoterapia Adotiva , Antígeno-1 Associado à Função Linfocitária , Espectrina , Humanos , Feminino
6.
Front Immunol ; 11: 1501, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32793204

RESUMO

The extracellular matrix (ECM) is extensively remodeled during inflammation providing essential guidance cues for immune cell migration and signals for cell activation and survival. There is increasing interest in the therapeutic targeting of ECM to mitigate chronic inflammatory diseases and enhance access to the tumor microenvironment. T cells utilize the ECM as a scaffold for interstitial migration, dependent on T cell expression of matrix-binding integrins αVß1/αVß3 and tissue display of the respective RGD-containing ligands. The specific ECM components that control T cell migration are unclear. Fibronectin (FN), a canonical RGD-containing matrix component, is heavily upregulated in inflamed tissues and in vitro can serve as a substrate for leukocyte migration. However, limited by lack of tools to intravitally visualize and manipulate FN, the specific role of FN in effector T cell migration in vivo is unknown. Here, we utilize fluorescently-tagged FN to probe for FN deposition, and intravital multiphoton microscopy to visualize T cell migration relative to FN in the inflamed ear dermis. Th1 cells were found to migrate along FN fibers, with T cells appearing to actively push or pull against flexible FN fibers. To determine the importance of T cell interactions with FN, we used a specific inhibitor of FN polymerization, pUR4. Intradermal delivery of pUR4 (but not the control peptide) to the inflamed skin resulted in a local reduction in FN deposition. We also saw a striking attenuation of Th1 effector T cell movement at the pUR4 injection site, suggesting FN plays a key role in T cell interstitial migration. In mechanistic studies, pUR4 incubation with FN in vitro resulted in enhanced tethering of T cells to FN matrix, limiting productive migration. In vivo, such tethering led to increased Th1 accumulation in the inflamed dermis. Enhanced Th1 accumulation exacerbated inflammation with increased Th1 activation and IFNγ cytokine production. Thus, our studies highlight the importance of ECM FN fibrils for T cell migration in inflamed tissues and suggest that manipulating local levels of ECM FN may prove beneficial in promoting T cell accumulation in tissues and enhancing local immunity to infection or cancer.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Mucosa Intestinal/imunologia , Pele/imunologia , Transferência Adotiva , Animais , Movimento Celular , Células Cultivadas , Matriz Extracelular/imunologia , Fibronectinas/química , Fibronectinas/imunologia , Inflamação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Fragmentos de Peptídeos/administração & dosagem , Polimerização , Receptores de Antígenos de Linfócitos T/genética
7.
Nat Commun ; 10(1): 1249, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30890704

RESUMO

Phagocytosis of invading pathogens or cellular debris requires a dramatic change in cell shape driven by actin polymerization. For antibody-covered targets, phagocytosis is thought to proceed through the sequential engagement of Fc-receptors on the phagocyte with antibodies on the target surface, leading to the extension and closure of the phagocytic cup around the target. We find that two actin-dependent molecular motors, class 1 myosins myosin 1e and myosin 1f, are specifically localized to Fc-receptor adhesions and required for efficient phagocytosis of antibody-opsonized targets. Using primary macrophages lacking both myosin 1e and myosin 1f, we find that without the actin-membrane linkage mediated by these myosins, the organization of individual adhesions is compromised, leading to excessive actin polymerization, slower adhesion turnover, and deficient phagocytic internalization. This work identifies a role for class 1 myosins in coordinated adhesion turnover during phagocytosis and supports a mechanism involving membrane-cytoskeletal crosstalk for phagocytic cup closure.


Assuntos
Actinas/metabolismo , Adesão Celular/fisiologia , Miosina Tipo I/metabolismo , Miosinas/metabolismo , Fagocitose/fisiologia , Actinas/ultraestrutura , Animais , Células da Medula Óssea , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Feminino , Microscopia Intravital , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica , Microscopia de Fluorescência , Miosina Tipo I/genética , Miosinas/genética , Cultura Primária de Células , Células RAW 264.7 , Receptores Fc/metabolismo , Receptores Fc/ultraestrutura , Imagem com Lapso de Tempo
8.
Nat Commun ; 9(1): 4948, 2018 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-30470750

RESUMO

The actin cytoskeleton is an active semi-flexible polymer network whose non-equilibrium properties coordinate both stable and contractile behaviors to maintain or change cell shape. While myosin motors drive the actin cytoskeleton out-of-equilibrium, the role of myosin-driven active stresses in the accumulation and dissipation of mechanical energy is unclear. To investigate this, we synthesize an actomyosin material in vitro whose active stress content can tune the network from stable to contractile. Each increment in activity determines a characteristic spectrum of actin filament fluctuations which is used to calculate the total mechanical work and the production of entropy in the material. We find that the balance of work and entropy does not increase monotonically and the entropy production rate is maximized in the non-contractile, stable state of actomyosin. Our study provides evidence that the origins of entropy production and activity-dependent dissipation relate to disorder in the molecular interactions between actin and myosin.


Assuntos
Actomiosina/química , Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Animais , Fenômenos Biomecânicos , Galinhas , Entropia , Humanos , Cinética , Miosinas/química , Miosinas/metabolismo
9.
J Cell Biol ; 217(4): 1485-1502, 2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29437785

RESUMO

Developing tissues change shape and tumors initiate spreading through collective cell motility. Conserved mechanisms by which tissues initiate motility into their surroundings are not known. We investigated cytoskeletal regulators during collective invasion by mouse tumor organoids and epithelial Madin-Darby canine kidney (MDCK) acini undergoing branching morphogenesis in collagen. Use of the broad-spectrum formin inhibitor SMIFH2 prevented the formation of migrating cell fronts in both cell types. Focusing on the role of the formin Dia1 in branching morphogenesis, we found that its depletion in MDCK cells does not alter planar cell motility either within the acinus or in two-dimensional scattering assays. However, Dia1 was required to stabilize protrusions extending into the collagen matrix. Live imaging of actin, myosin, and collagen in control acini revealed adhesions that deformed individual collagen fibrils and generated large traction forces, whereas Dia1-depleted acini exhibited unstable adhesions with minimal collagen deformation and lower force generation. This work identifies Dia1 as an essential regulator of tissue shape changes through its role in stabilizing focal adhesions.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Transporte/metabolismo , Adesão Celular , Movimento Celular , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/metabolismo , Actinas/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas de Transporte/genética , Forma Celular , Cães , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Feminino , Proteínas Fetais/metabolismo , Colágenos Fibrilares/metabolismo , Forminas , Fator de Crescimento de Hepatócito/farmacologia , Células Madin Darby de Rim Canino , Glândulas Mamárias Animais/patologia , Camundongos , Morfogênese , Miosinas/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais , Fatores de Tempo , Células Tumorais Cultivadas
10.
Sci Rep ; 7: 43764, 2017 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-28256617

RESUMO

Cholesterol regulates numerous cellular processes. Depleting its synthesis in skeletal myofibers induces vacuolization and contraction impairment. However, little is known about how cholesterol reduction affects cardiomyocyte behavior. Here, we deplete cholesterol by incubating neonatal cardiomyocytes with methyl-beta-cyclodextrin. Traction force microscopy shows that lowering cholesterol increases the rate of cell contraction and generates defects in cell relaxation. Cholesterol depletion also increases membrane tension, Ca2+ spikes frequency and intracellular Ca2+ concentration. These changes can be correlated with modifications in caveolin-3 and L-Type Ca2+ channel distributions across the sarcolemma. Channel regulation is also compromised since cAMP-dependent PKA activity is enhanced, increasing the probability of L-Type Ca2+ channel opening events. Immunofluorescence reveals that cholesterol depletion abrogates sarcomeric organization, changing spacing and alignment of α-actinin bands due to increase in proteolytic activity of calpain. We propose a mechanism in which cholesterol depletion triggers a signaling cascade, culminating with contraction impairment and myofibril disruption in cardiomyocytes.


Assuntos
Sinalização do Cálcio/fisiologia , Colesterol/metabolismo , Miócitos Cardíacos/fisiologia , Sarcolema/fisiologia , Actinina/metabolismo , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Caveolina 3/metabolismo , Células Cultivadas , Colesterol/deficiência , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ratos Wistar , Sarcolema/efeitos dos fármacos , Sarcolema/metabolismo , beta-Ciclodextrinas/metabolismo , beta-Ciclodextrinas/farmacologia
11.
PLoS Comput Biol ; 11(3): e1004076, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25748431

RESUMO

Adherent cells use forces at the cell-substrate interface to sense and respond to the physical properties of their environment. These cell forces can be measured with traction force microscopy which inverts the equations of elasticity theory to calculate them from the deformations of soft polymer substrates. We introduce a new type of traction force microscopy that in contrast to traditional methods uses additional image data for cytoskeleton and adhesion structures and a biophysical model to improve the robustness of the inverse procedure and abolishes the need for regularization. We use this method to demonstrate that ventral stress fibers of U2OS-cells are typically under higher mechanical tension than dorsal stress fibers or transverse arcs.


Assuntos
Citoesqueleto de Actina/química , Actinas/química , Modelos Biológicos , Fibras de Estresse/química , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Linhagem Celular Tumoral , Humanos , Processamento de Imagem Assistida por Computador , Microscopia de Força Atômica , Fibras de Estresse/metabolismo , Estresse Mecânico
12.
J Cell Biol ; 196(3): 363-74, 2012 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-22291038

RESUMO

Focal adhesion composition and size are modulated in a myosin II-dependent maturation process that controls adhesion, migration, and matrix remodeling. As myosin II activity drives stress fiber assembly and enhanced tension at adhesions simultaneously, the extent to which adhesion maturation is driven by tension or altered actin architecture is unknown. We show that perturbations to formin and α-actinin 1 activity selectively inhibited stress fiber assembly at adhesions but retained a contractile lamella that generated large tension on adhesions. Despite relatively unperturbed adhesion dynamics and force transmission, impaired stress fiber assembly impeded focal adhesion compositional maturation and fibronectin remodeling. Finally, we show that compositional maturation of focal adhesions could occur even when myosin II-dependent cellular tension was reduced by 80%. We propose that stress fiber assembly at the adhesion site serves as a structural template that facilitates adhesion maturation over a wide range of tensions. This work identifies the essential role of lamellar actin architecture in adhesion maturation.


Assuntos
Adesões Focais/fisiologia , Fibras de Estresse/fisiologia , Citoesqueleto de Actina/fisiologia , Actinina/metabolismo , Linhagem Celular Tumoral , Matriz Extracelular/metabolismo , Humanos , Miosina Tipo II/metabolismo
13.
Biophys J ; 100(12): 2883-93, 2011 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-21689521

RESUMO

Focal adhesions (FAs) are the predominant mechanism by which cells mechanically couple to and exert traction forces on their extracellular matrix (ECM). It is widely presumed that FA size is modulated by force to mediate changes in adhesion strength at different levels of cellular tension. However, previous studies seeking correlations between force and FA morphology have yielded variable and often conflicting results. Here we show that a strong correlation between adhesion size and traction force exists only during the initial stages of myosin-mediated adhesion maturation and growth. For mature adhesions, no correlation between traction stress and size is observed. Rather, the tension that is sustained at mature adhesions is more strongly influenced by proximity to the cell edge, with peripheral adhesions transmitting higher tension than adhesions near the cell center. Finally, we show that mature adhesions can withstand sixfold increases in tension without changes in size. Thus, although a strong correlation between adhesion size and mechanical tension is observed during the initial stages of myosin-mediated adhesion maturation, no correlation is observed in mature, elongated adhesions. This work places spatiotemporal constraints on the force-dependent growth of adhesions and provides insight into the mechanical regulation of cell-ECM adhesion.


Assuntos
Adesões Focais/fisiologia , Actinas/metabolismo , Animais , Fenômenos Biomecânicos/fisiologia , Linhagem Celular Tumoral , Extensões da Superfície Celular/metabolismo , Matriz Extracelular/fisiologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Miosina Tipo II/metabolismo , Células NIH 3T3 , Paxilina/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Estresse Mecânico , Fatores de Tempo
14.
Mol Biol Cell ; 22(8): 1330-9, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21307339

RESUMO

The regulation of cellular traction forces on the extracellular matrix is critical to cell adhesion, migration, proliferation, and differentiation. Diverse lamellar actin organizations ranging from contractile lamellar networks to stress fibers are observed in adherent cells. Although lamellar organization is thought to reflect the extent of cellular force generation, understanding of the physical behaviors of the lamellar actin cytoskeleton is lacking. To elucidate these properties, we visualized the actomyosin dynamics and organization in U2OS cells over a broad range of forces. At low forces, contractile lamellar networks predominate and force generation is strongly correlated to actomyosin retrograde flow dynamics with nominal change in organization. Lamellar networks build ∼60% of cellular tension over rapid time scales. At high forces, reorganization of the lamellar network into stress fibers results in moderate changes in cellular tension over slower time scales. As stress fibers build and tension increases, myosin band spacing decreases and α-actinin bands form. On soft matrices, force generation by lamellar networks is unaffected, whereas tension-dependent stress fiber assembly is abrogated. These data elucidate the dynamic and structural signatures of the actomyosin cytoskeleton at different levels of tension and set a foundation for quantitative models of cell and tissue mechanics.


Assuntos
Actinina/fisiologia , Actinas/fisiologia , Actomiosina/fisiologia , Miosinas/fisiologia , Fibras de Estresse/fisiologia , Fenômenos Biomecânicos , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Adesão Celular , Movimento Celular , Proliferação de Células , Matriz Extracelular/fisiologia , Análise de Fourier , Humanos , Microscopia de Força Atômica , Microscopia Confocal , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Células Tumorais Cultivadas
15.
J Vis Exp ; (46)2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21178972

RESUMO

The regulation of cellular adhesion to the extracellular matrix (ECM) is essential for cell migration and ECM remodeling. Focal adhesions are macromolecular assemblies that couple the contractile F-actin cytoskeleton to the ECM. This connection allows for the transmission of intracellular mechanical forces across the cell membrane to the underlying substrate. Recent work has shown the mechanical properties of the ECM regulate focal adhesion and F-actin morphology as well as numerous physiological processes, including cell differentiation, division, proliferation and migration. Thus, the use of cell culture substrates has become an increasingly prevalent method to precisely control and modulate ECM mechanical properties. To quantify traction forces at focal adhesions in an adherent cell, compliant substrates are used in conjunction with high-resolution imaging and computational techniques in a method termed traction force microscopy (TFM). This technique relies on measurements of the local magnitude and direction of substrate deformations induced by cellular contraction. In combination with high-resolution fluorescence microscopy of fluorescently tagged proteins, it is possible to correlate cytoskeletal organization and remodeling with traction forces. Here we present a detailed experimental protocol for the preparation of two-dimensional, compliant matrices for the purpose of creating a cell culture substrate with a well-characterized, tunable mechanical stiffness, which is suitable for measuring cellular contraction. These protocols include the fabrication of polyacrylamide hydrogels, coating of ECM proteins on such gels, plating cells on gels, and high-resolution confocal microscopy using a perfusion chamber. Additionally, we provide a representative sample of data demonstrating location and magnitude of cellular forces using cited TFM protocols.


Assuntos
Adesão Celular/fisiologia , Técnicas de Cultura de Células/métodos , Matriz Extracelular/fisiologia , Resinas Acrílicas/química , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proteínas da Matriz Extracelular/química , Adesões Focais/fisiologia , Humanos , Microscopia Confocal/métodos , Osteossarcoma/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA