Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
Add more filters










Publication year range
1.
Cell Stem Cell ; 31(5): 640-656.e8, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38701758

ABSTRACT

Post-implantation, the pluripotent epiblast in a human embryo forms a central lumen, paving the way for gastrulation. Osmotic pressure gradients are considered the drivers of lumen expansion across development, but their role in human epiblasts is unknown. Here, we study lumenogenesis in a pluripotent-stem-cell-based epiblast model using engineered hydrogels. We find that leaky junctions prevent osmotic pressure gradients in early epiblasts and, instead, forces from apical actin polymerization drive lumen expansion. Once the lumen reaches a radius of ∼12 µm, tight junctions mature, and osmotic pressure gradients develop to drive further growth. Computational modeling indicates that apical actin polymerization into a stiff network mediates initial lumen expansion and predicts a transition to pressure-driven growth in larger epiblasts to avoid buckling. Human epiblasts show transcriptional signatures consistent with these mechanisms. Thus, actin polymerization drives lumen expansion in the human epiblast and may serve as a general mechanism of early lumenogenesis.


Subject(s)
Actins , Germ Layers , Osmotic Pressure , Polymerization , Humans , Actins/metabolism , Germ Layers/metabolism , Germ Layers/cytology , Models, Biological , Tight Junctions/metabolism
2.
Commun Biol ; 7(1): 658, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811770

ABSTRACT

The cytoskeleton is a complex network of interconnected biopolymers consisting of actin filaments, microtubules, and intermediate filaments. These biopolymers work in concert to transmit cell-generated forces to the extracellular matrix required for cell motility, wound healing, and tissue maintenance. While we know cell-generated forces are driven by actomyosin contractility and balanced by microtubule network resistance, the effect of intermediate filaments on cellular forces is unclear. Using a combination of theoretical modeling and experiments, we show that vimentin intermediate filaments tune cell stress by assisting in both actomyosin-based force transmission and reinforcement of microtubule networks under compression. We show that the competition between these two opposing effects of vimentin is regulated by the microenvironment stiffness. These results reconcile seemingly contradictory results in the literature and provide a unified description of vimentin's effects on the transmission of cell contractile forces to the extracellular matrix.


Subject(s)
Actomyosin , Mechanotransduction, Cellular , Microtubules , Vimentin , Microtubules/metabolism , Actomyosin/metabolism , Vimentin/metabolism , Humans , Extracellular Matrix/metabolism , Animals
3.
bioRxiv ; 2024 May 03.
Article in English | MEDLINE | ID: mdl-38746096

ABSTRACT

Cells regulate their shape and metabolic activity in response to the mechano-chemical properties of their microenvironment. To elucidate the impact of matrix stiffness and ligand density on a cell's bioenergetics, we developed a non-equilibrium, active chemo-mechanical model that accounts for mechanical energy of the cell and matrix, chemical energy from ATP hydrolysis, interfacial energy, and mechano-sensitive regulation of stress fiber assembly through signaling. By integrating the kinetics and energetics of these processes we introduce the concept of the metabolic potential of the cell that, when minimized, gives experimentally testable predictions of the cell contractility, shape, and the ATP consumption. Specifically, we show that MDA-MB-231 breast cancer cells in 3D collagen gels follow a spherical to spindle to spherical change in morphology with increasing matrix stiffness consistent with experimental observations. This biphasic transition in cell shape emerges from a competition between increased contractility accompanied by ATP hydrolysis enabled by mechano-sensitive signaling, which lowers the volumetric contribution to the metabolic potential of elongated cells and the interfacial energy which is lower for spherical shapes. On 2D hydrogels, our model predicts a hemispherical to spindle to disc shape transition with increasing gel stiffness. In both cases, we show that increasing matrix stiffness monotonically increases the cell's contractility as well as ATP consumption. Our model also predicts how the increased energy demand in stiffer microenvironments is met by AMPK activation, which is confirmed through experimental measurement of activated AMPK levels as a function of matrix stiffness carried out here in both 2D and 3D micro-environments. Further, model predictions of increased AMPK activation on stiffer micro-environments are found to correlate strongly with experimentally measured upregulation of mitochondrial potential, glucose uptake and ATP levels. The insights from our model can be used to understand mechanosensitive regulation of metabolism in physiological events such as metastasis and tumor progression during which cells experience dynamic changes in their microenvironment and metabolic state.

4.
Nat Commun ; 15(1): 4338, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773126

ABSTRACT

In interphase nuclei, chromatin forms dense domains of characteristic sizes, but the influence of transcription and histone modifications on domain size is not understood. We present a theoretical model exploring this relationship, considering chromatin-chromatin interactions, histone modifications, and chromatin extrusion. We predict that the size of heterochromatic domains is governed by a balance among the diffusive flux of methylated histones sustaining them and the acetylation reactions in the domains and the process of loop extrusion via supercoiling by RNAPII at their periphery, which contributes to size reduction. Super-resolution and nano-imaging of five distinct cell lines confirm the predictions indicating that the absence of transcription leads to larger heterochromatin domains. Furthermore, the model accurately reproduces the findings regarding how transcription-mediated supercoiling loss can mitigate the impacts of excessive cohesin loading. Our findings shed light on the role of transcription in genome organization, offering insights into chromatin dynamics and potential therapeutic targets.


Subject(s)
Chromatin , Epigenesis, Genetic , Heterochromatin , Histones , Transcription, Genetic , Humans , Histones/metabolism , Heterochromatin/metabolism , Heterochromatin/genetics , Chromatin/metabolism , Chromatin/genetics , RNA Polymerase II/metabolism , Cohesins , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Histone Code , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/genetics , Acetylation , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Interphase
5.
bioRxiv ; 2023 Oct 28.
Article in English | MEDLINE | ID: mdl-37961689

ABSTRACT

Mechanical properties of the extracellular matrices (ECMs) critically regulate a number of important cell function including growth, differentiation and migration. Type I collagen and glycosaminoglycans (GAGs) are two primary components of ECMs that contribute to tissue mechanics with the collagen fiber network sustaining tension and GAGs withstanding compression. Collagen stiffness as well as its architecture are known to be important role players in cell-ECM mechanical interactions, however, much less is known about how GAGs within ECMs regulate cell force generation and invasion. Inspired by a recent theoretical work from the Shenoy lab that GAGs play important roles in cell - ECM interactions, we hereby present experimental studies on the role of hyaluronic acid (HA, an unsulfated GAG) in single tumor cell traction force generation within HA collagen cogels using a recently developed 3D cell traction force microscopy. Our work revealed that CD44, a cell surface adhesion receptor to HA, was engaged in cell traction force generation in conjunction with ß1-integrin. Furthermore, we found that HA significantly modified the architecture and mechanics of the collagen fiber network, decreased tumor cells' propensity to remodel the collagen network, decreased traction force generation and transmission distance, and attenuated tumor invasion in agreement with theoretical predictions. Our findings highlighted the significance of CD44 and HA engagement in cell-ECM mechanical interactions, providing new insights on the mechanical model of cellular force transmission.

6.
bioRxiv ; 2023 Nov 11.
Article in English | MEDLINE | ID: mdl-37986921

ABSTRACT

The cell nucleus is continuously exposed to external signals, of both chemical and mechanical nature. To ensure proper cellular response, cells need to regulate not only the transmission of these signals, but also their timing and duration. Such timescale regulation is well described for fluctuating chemical signals, but if and how it applies to mechanical signals reaching the nucleus is still unknown. Here we demonstrate that the formation of fibrillar adhesions locks the nucleus in a mechanically deformed conformation, setting the mechanical response timescale to that of fibrillar adhesion remodelling (~1 hour). This process encompasses both mechanical deformation and associated mechanotransduction (such as via YAP), in response to both increased and decreased mechanical stimulation. The underlying mechanism is the anchoring of the vimentin cytoskeleton to fibrillar adhesions and the extracellular matrix through plectin 1f, which maintains nuclear deformation. Our results reveal a mechanism to regulate the timescale of mechanical adaptation, effectively setting a low pass filter to mechanotransduction.

7.
Nat Commun ; 14(1): 2902, 2023 05 22.
Article in English | MEDLINE | ID: mdl-37217555

ABSTRACT

Immune cells, such as macrophages and dendritic cells, can utilize podosomes, mechanosensitive actin-rich protrusions, to generate forces, migrate, and patrol for foreign antigens. Individual podosomes probe their microenvironment through periodic protrusion and retraction cycles (height oscillations), while oscillations of multiple podosomes in a cluster are coordinated in a wave-like fashion. However, the mechanisms governing both the individual oscillations and the collective wave-like dynamics remain unclear. Here, by integrating actin polymerization, myosin contractility, actin diffusion, and mechanosensitive signaling, we develop a chemo-mechanical model for podosome dynamics in clusters. Our model reveals that podosomes show oscillatory growth when actin polymerization-driven protrusion and signaling-associated myosin contraction occur at similar rates, while the diffusion of actin monomers drives wave-like coordination of podosome oscillations. Our theoretical predictions are validated by different pharmacological treatments and the impact of microenvironment stiffness on chemo-mechanical waves. Our proposed framework can shed light on the role of podosomes in immune cell mechanosensing within the context of wound healing and cancer immunotherapy.


Subject(s)
Podosomes , Podosomes/metabolism , Actins/metabolism , Macrophages/metabolism
8.
Proc Natl Acad Sci U S A ; 120(16): e2216811120, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37036981

ABSTRACT

Matrix stiffening and external mechanical stress have been linked to disease and cancer development in multiple tissues, including the liver, where cirrhosis (which increases stiffness markedly) is the major risk factor for hepatocellular carcinoma. Patients with nonalcoholic fatty liver disease and lipid droplet-filled hepatocytes, however, can develop cancer in noncirrhotic, relatively soft tissue. Here, by treating primary human hepatocytes with the monounsaturated fatty acid oleate, we show that lipid droplets are intracellular mechanical stressors with similar effects to tissue stiffening, including nuclear deformation, chromatin condensation, and impaired hepatocyte function. Mathematical modeling of lipid droplets as inclusions that have only mechanical interactions with other cellular components generated results consistent with our experiments. These data show that lipid droplets are intracellular sources of mechanical stress and suggest that nuclear membrane tension integrates cell responses to combined internal and external stresses.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Humans , Lipid Droplets/metabolism , Hepatocytes/pathology , Carcinoma, Hepatocellular/pathology , Non-alcoholic Fatty Liver Disease/pathology , Liver Neoplasms/pathology , Lipid Metabolism/physiology
9.
Nat Biomed Eng ; 7(2): 177-191, 2023 02.
Article in English | MEDLINE | ID: mdl-35996026

ABSTRACT

Changes in the micro-environment of fibrous connective tissue can lead to alterations in the phenotypes of tissue-resident cells, yet the underlying mechanisms are poorly understood. Here, by visualizing the dynamics of histone spatial reorganization in tenocytes and mesenchymal stromal cells from fibrous tissue of human donors via super-resolution microscopy, we show that physiological and pathological chemomechanical cues can directly regulate the spatial nanoscale organization and density of chromatin in these tissue-resident cell populations. Specifically, changes in substrate stiffness, altered oxygen tension and the presence of inflammatory signals drive chromatin relocalization and compaction into the nuclear boundary, mediated by the activity of the histone methyltransferase EZH2 and an intact cytoskeleton. In healthy cells, chemomechanically triggered changes in the spatial organization and density of chromatin are reversible and can be attenuated by dynamically stiffening the substrate. In diseased human cells, however, the link between mechanical or chemical inputs and chromatin remodelling is abrogated. Our findings suggest that aberrant chromatin organization in fibrous connective tissue may be a hallmark of disease progression that could be leveraged for therapeutic intervention.


Subject(s)
Chromatin , Cues , Humans , Histones/genetics , Cytoskeleton , Connective Tissue
10.
Sci Adv ; 8(47): eabq5944, 2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36417537

ABSTRACT

Small-molecule adsorption energies correlate with energy barriers of catalyzed intermediate reaction steps, determining the dominant microkinetic mechanism. Straining the catalyst can alter adsorption energies and break scaling relationships that inhibit reaction engineering, but identifying desirable strain patterns using density functional theory is intractable because of the high-dimensional search space. We train a graph neural network to predict the adsorption energy response of a catalyst/adsorbate system under a proposed surface strain pattern. The training data are generated by randomly straining and relaxing Cu-based binary alloy catalyst complexes taken from the Open Catalyst Project. The trained model successfully predicts the adsorption energy response for 85% of strains in unseen test data, outperforming ensemble linear baselines. Using ammonia synthesis as an example, we identify Cu-S alloy catalysts as promising candidates for strain engineering. Our approach can locate strain patterns that break adsorption energy scaling relations to improve catalyst performance.

11.
Nat Commun ; 13(1): 7089, 2022 11 19.
Article in English | MEDLINE | ID: mdl-36402771

ABSTRACT

The formation and recovery of gaps in the vascular endothelium governs a wide range of physiological and pathological phenomena, from angiogenesis to tumor cell extravasation. However, the interplay between the mechanical and signaling processes that drive dynamic behavior in vascular endothelial cells is not well understood. In this study, we propose a chemo-mechanical model to investigate the regulation of endothelial junctions as dependent on the feedback between actomyosin contractility, VE-cadherin bond turnover, and actin polymerization, which mediate the forces exerted on the cell-cell interface. Simulations reveal that active cell tension can stabilize cadherin bonds, but excessive RhoA signaling can drive bond dissociation and junction failure. While actin polymerization aids gap closure, high levels of Rac1 can induce junction weakening. Combining the modeling framework with experiments, our model predicts the influence of pharmacological treatments on the junction state and identifies that a critical balance between RhoA and Rac1 expression is required to maintain junction stability. Our proposed framework can help guide the development of therapeutics that target the Rho family of GTPases and downstream active mechanical processes.


Subject(s)
Actins , Endothelial Cells , Endothelial Cells/metabolism , Actins/metabolism , Feedback , Signal Transduction , Actin Cytoskeleton/metabolism
12.
ACS Nano ; 16(5): 6949-6955, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35512182

ABSTRACT

The COVID-19 pandemic has been inflicted upon humanity by the SARS-CoV-2 virus, the latest insidious incarnation of the coronaviruses group. While in its wake intense scientific research has produced breakthrough vaccines and cures, there still exists an immediate need to further understand the origin, mechanobiology and biochemistry, and destiny of this virus so that future pandemics arising from similar coronaviruses may be contained more effectively. In this Perspective, we discuss the various evidential findings of virus propagation and connect them to respective underpinning cellular biomechanical states leading to corresponding manifestations of the viral activity. We further propose avenues to tackle the virus, including from a "musical" vantage point, and contain its relentless strides that are currently afflicting the global populace.


Subject(s)
COVID-19 , Music , Humans , Pandemics , SARS-CoV-2 , Antiviral Agents/therapeutic use
13.
ACS Nano ; 16(6): 9452-9460, 2022 Jun 28.
Article in English | MEDLINE | ID: mdl-35617052

ABSTRACT

Two-dimensional (2D) dilute magnetic semiconductors (DMSs) are attractive material platforms for applications in multifunctional nanospintronics due to the prospect of embedding controllable magnetic order within nanoscale semiconductors. Identifying candidate host material and dopant systems requires consideration of doping formation energies, magnetic ordering, and the tendency for dopants to form clustered domains. In this work, we consider the defect thermodynamics and the dilute magnetic properties across charge states of 2D-MoS2 and 2D-WS2 with Mn magnetic dopants as candidate systems for 2D-DMSs. Using hybrid density functional calculations, we study the magnetic and electronic properties of these systems across configurations with thermodynamically favorable defects: 2D-MoS2 doped with Mn atoms at sulfur site (MnS), at two Mo sites (2MnMo), on top of a Mo atom (Mn-top), and at a Mo site (MnMo). While the majority of the Mn-defect complexes provide trap states, MnMo and MnW are amphoteric, although previously predicted to be donor defects. The impact of cluster formation of these amphoteric defects on magnetic ordering is also considered; both MnMo-MnMo (2Mn2Mo) and MnW-MnW (2Mn2W) clusters are found to be stable in ferromagnetic (FM) ordering. Interestingly, we observed the defect charge state dependent magnetic behavior of 2Mn2Mo and 2Mn2W clusters in 2D-TMDs. We investigate that the FM coupling of 2Mn2Mo and 2Mn2W clusters is stable in only a neutral charge state; however, the antiferromagnetic (AFM) coupling is stable in the +1 charge state. 2Mn2Mo clusters provide shallow donor levels in AFM coupling and deep donor levels in FM coupling. 2Mn2W clusters lead to trap states in the FM and AFM coupling. We demonstrate the AFM to FM phase transition at a critical electron density nce = 3.5 × 1013 cm-2 in 2D-MoS2 and 2D-WS2. At a 1.85% concentration of Mn, we calculate the Curie temperature of 580 K in the mean-field approximation.

14.
Proc Natl Acad Sci U S A ; 119(15): e2116718119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35394874

ABSTRACT

Cells can sense and respond to mechanical forces in fibrous extracellular matrices (ECMs) over distances much greater than their size. This phenomenon, termed long-range force transmission, is enabled by the realignment (buckling) of collagen fibers along directions where the forces are tensile (compressive). However, whether other key structural components of the ECM, in particular glycosaminoglycans (GAGs), can affect the efficiency of cellular force transmission remains unclear. Here we developed a theoretical model of force transmission in collagen networks with interpenetrating GAGs, capturing the competition between tension-driven collagen fiber alignment and the swelling pressure induced by GAGs. Using this model, we show that the swelling pressure provided by GAGs increases the stiffness of the collagen network by stretching the fibers in an isotropic manner. We found that the GAG-induced swelling pressure can help collagen fibers resist buckling as the cells exert contractile forces. This mechanism impedes the alignment of collagen fibers and decreases long-range cellular mechanical communication. We experimentally validated the theoretical predictions by comparing the intensity of collagen fiber alignment between cellular spheroids cultured on collagen gels versus collagen­GAG cogels. We found significantly lower intensities of aligned collagen in collagen­GAG cogels, consistent with the prediction that GAGs can prevent collagen fiber alignment. The role of GAGs in modulating force transmission uncovered in this work can be extended to understand pathological processes such as the formation of fibrotic scars and cancer metastasis, where cells communicate in the presence of abnormally high concentrations of GAGs.


Subject(s)
Cell Communication , Extracellular Matrix , Glycosaminoglycans , Biomechanical Phenomena , Cell Physiological Phenomena , Collagen/metabolism , Extracellular Matrix/metabolism , Fibrosis , Glycosaminoglycans/metabolism , Humans , Neoplasms
15.
Sci Adv ; 7(46): eabi8157, 2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34757787

ABSTRACT

The natural extracellular matrix (ECM) within tissues is physically contracted and remodeled by cells, allowing the collective shaping of functional tissue architectures. Synthetic materials that facilitate self-assembly similar to natural ECM are needed for cell culture, tissue engineering, and in vitro models of development and disease. To address this need, we develop fibrous hydrogel assemblies that are stabilized with photocrosslinking and display fiber density­dependent strain-responsive properties (strain stiffening and alignment). Encapsulated mesenchymal stromal cells locally contract low fiber density assemblies, resulting in macroscopic volumetric changes with increased cell densities and moduli. Because of properties such as shear-thinning and self-healing, assemblies can be processed into microtissues with aligned ECM deposition or through extrusion bioprinting and photopatterning to fabricate constructs with programmed shape changes due to cell contraction. These materials provide a synthetic approach to mimic features of natural ECM, which can now be processed for applications in biofabrication and tissue engineering.

16.
Biophys J ; 120(22): 5074-5089, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34627766

ABSTRACT

Mechanotransduction describes activation of gene expression by changes in the cell's physical microenvironment. Recent experiments show that mechanotransduction can lead to long-term "mechanical memory," in which cells cultured on stiff substrates for sufficient time (priming phase) maintain altered phenotype after switching to soft substrates (dissipation phase) as compared to unprimed controls. The timescale of memory acquisition and retention is orders of magnitude larger than the timescale of mechanosensitive cellular signaling, and memory retention time changes continuously with priming time. We develop a model that captures these features by accounting for positive reinforcement in mechanical signaling. The sensitivity of reinforcement represents the dynamic transcriptional state of the cell composed of protein lifetimes and three-dimensional chromatin organization. Our model provides a single framework connecting microenvironment mechanical history to cellular outcomes ranging from no memory to terminal differentiation. Predicting cellular memory of environmental changes can help engineer cellular dynamics through changes in culture environments.


Subject(s)
Mechanotransduction, Cellular , Reinforcement, Psychology , Gene Expression , Phenotype
17.
Sci Rep ; 11(1): 16478, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34389738

ABSTRACT

Here we present a microengineered soft-robotic in vitro platform developed by integrating a pneumatically regulated novel elastomeric actuator with primary culture of human cells. This system is capable of generating dynamic bending motion akin to the constriction of tubular organs that can exert controlled compressive forces on cultured living cells. Using this platform, we demonstrate cyclic compression of primary human endothelial cells, fibroblasts, and smooth muscle cells to show physiological changes in their morphology due to applied forces. Moreover, we present mechanically actuatable organotypic models to examine the effects of compressive forces on three-dimensional multicellular constructs designed to emulate complex tissues such as solid tumors and vascular networks. Our work provides a preliminary demonstration of how soft-robotics technology can be leveraged for in vitro modeling of complex physiological tissue microenvironment, and may enable the development of new research tools for mechanobiology and related areas.


Subject(s)
Robotics , Tissue Engineering , Compressive Strength , Endothelial Cells/physiology , Fibroblasts/physiology , Humans , In Vitro Techniques , Myocytes, Smooth Muscle/physiology , Neoplasm Invasiveness , Robotics/instrumentation , Robotics/methods
18.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article in English | MEDLINE | ID: mdl-34234016

ABSTRACT

Damage to the microtubule lattice, which serves as a rigid cytoskeletal backbone for the axon, is a hallmark mechanical initiator of pathophysiology after concussion. Understanding the mechanical stress transfer from the brain tissue to the axonal cytoskeleton is essential to determine the microtubule lattice's vulnerability to mechanical injury. Here, we develop an ultrastructural model of the axon's cytoskeletal architecture to identify the components involved in the dynamic load transfer during injury. Corroborative in vivo studies were performed using a gyrencephalic swine model of concussion via single and repetitive head rotational acceleration. Computational analysis of the load transfer mechanism demonstrates that the myelin sheath and the actin/spectrin cortex play a significant role in effectively shielding the microtubules from tissue stress. We derive failure maps in the space spanned by tissue stress and stress rate to identify physiological conditions in which the microtubule lattice can rupture. We establish that a softer axonal cortex leads to a higher susceptibility of the microtubules to failure. Immunohistochemical examination of tissue from the swine model of single and repetitive concussion confirms the presence of postinjury spectrin degradation, with more extensive pathology observed following repetitive injury. Because the degradation of myelin and spectrin occurs over weeks following the first injury, we show that softening of the myelin layer and axonal cortex exposes the microtubules to higher stress during repeated incidences of traumatic brain injuries. Our predictions explain how mechanical injury predisposes axons to exacerbated responses to repeated injuries, as observed in vitro and in vivo.


Subject(s)
Axons/metabolism , Brain Concussion/pathology , Brain Injuries/pathology , Models, Biological , Myelin Sheath/metabolism , Spectrin/metabolism , Animals , Humans , Male , Microtubules/metabolism , Middle Aged , Proteolysis , Swine , White Matter/pathology
19.
Cell Rep ; 35(4): 109047, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33909999

ABSTRACT

Most extracellular matrices (ECMs) are known to be dissipative, exhibiting viscoelastic and often plastic behaviors. However, the influence of dissipation, in particular mechanical plasticity in 3D confining microenvironments, on cell motility is not clear. In this study, we develop a chemo-mechanical model for dynamics of invadopodia, the protrusive structures that cancer cells use to facilitate invasion, by considering myosin recruitment, actin polymerization, matrix deformation, and mechano-sensitive signaling pathways. We demonstrate that matrix dissipation facilitates invadopodia growth by softening ECMs over repeated cycles, during which plastic deformation accumulates via cyclic ratcheting. Our model reveals that distinct protrusion patterns, oscillatory or monotonic, emerge from the interplay of timescales for polymerization-associated extension and myosin recruitment dynamics. Our model predicts the changes in invadopodia dynamics upon inhibition of myosin, adhesions, and the Rho-Rho-associated kinase (ROCK) pathway. Altogether, our work highlights the role of matrix plasticity in invadopodia dynamics and can help design dissipative biomaterials to modulate cancer cell motility.


Subject(s)
Extracellular Matrix/metabolism , Podosomes/metabolism , Cell Movement , Feedback , Humans , Signal Transduction
20.
Cell Rep ; 35(3): 109019, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882318

ABSTRACT

Reversible differentiation of vascular smooth muscle cells (VSMCs) plays a critical role in vascular biology and disease. Changes in VSMC differentiation correlate with stiffness of the arterial extracellular matrix (ECM), but causal relationships remain unclear. We show that VSMC plasticity is mechanosensitive and that both the de-differentiated and differentiated fates are promoted by the same ECM stiffness. Differential equations developed to model this behavior predicted that a null VSMC state generates the dual fates in response to ECM stiffness. Direct measurements of cellular forces, proliferation, and contractile gene expression validated these predictions and showed that fate outcome is mediated by Rac-Rho homeostasis. Rac, through distinct effects on YAP and TAZ, is required for both fates. Rho drives the contractile state alone, so its level of activity, relative to Rac, drives phenotypic choice. Our results show how the cellular response to a single ECM stiffness generates bi-stability and VSMC plasticity.


Subject(s)
Adaptation, Physiological , Mechanotransduction, Cellular/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neuropeptides/genetics , rac1 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics , Actins/genetics , Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Differentiation , Cell Proliferation , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Neuropeptides/metabolism , Phenotype , Primary Cell Culture , Promoter Regions, Genetic , Single-Cell Analysis , Transcription, Genetic , YAP-Signaling Proteins/genetics , YAP-Signaling Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...