Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Biophys J ; 123(13): 1792-1803, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38783602

RESUMEN

Hydra vulgaris, long known for its remarkable regenerative capabilities, is also a long-standing source of inspiration for models of spontaneous patterning. Recently it became clear that early patterning during Hydra regeneration is an integrated mechanochemical process whereby morphogen dynamics is influenced by tissue mechanics. One roadblock to understanding Hydra self-organization is our lack of knowledge about the mechanical properties of these organisms. In this study, we combined microfluidic developments to perform parallelized microaspiration rheological experiments and numerical simulations to characterize these mechanical properties. We found three different behaviors depending on the applied stresses: an elastic response, a viscoelastic response, and tissue rupture. Using models of deformable shells, we quantify their Young's modulus, shear viscosity, and the critical stresses required to switch between behaviors. Based on these experimental results, we propose a description of the tissue mechanics during normal regeneration. Our results provide a first step toward the development of original mechanochemical models of patterning grounded in quantitative experimental data.


Asunto(s)
Hydra , Regeneración , Animales , Hydra/fisiología , Fenómenos Biomecánicos , Modelos Biológicos , Viscosidad , Módulo de Elasticidad , Estrés Mecánico , Reología
2.
Phys Rev Lett ; 125(12): 128103, 2020 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-33016731

RESUMEN

While many cellular mechanisms leading to chemotherapeutic resistance have been identified, there is an increasing realization that tumor-stroma interactions also play an important role. In particular, mechanical alterations are inherent to solid cancer progression and profoundly impact cell physiology. Here, we explore the influence of compressive stress on the efficacy of chemotherapeutics in pancreatic cancer spheroids. We find that increased compressive stress leads to decreased drug efficacy. Theoretical modeling and experiments suggest that mechanical stress decreases cell proliferation which in turn reduces the efficacy of chemotherapeutics that target proliferating cells. Our work highlights a mechanical form of drug resistance and suggests new strategies for therapy.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Modelos Biológicos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Resistencia a Antineoplásicos , Humanos , Estrés Mecánico , Gemcitabina
3.
Proc Natl Acad Sci U S A ; 114(51): 13465-13470, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29187529

RESUMEN

Cells that proliferate within a confined environment build up mechanical compressive stress. For example, mechanical pressure emerges in the naturally space-limited tumor environment. However, little is known about how cells sense and respond to mechanical compression. We developed microfluidic bioreactors to enable the investigation of the effects of compressive stress on the growth of the genetically tractable model organism Saccharomyces cerevisiae We used this system to determine that compressive stress is partly sensed through a module consisting of the mucin Msb2 and the cell wall protein Sho1, which act together as a sensor module in one of the two major osmosensing pathways in budding yeast. This signal is transmitted via the MAPKKK kinase Ste11. Thus, we term this mechanosensitive pathway the "SMuSh" pathway, for Ste11 through Mucin/Sho1 pathway. The SMuSh pathway delays cells in the G1 phase of the cell cycle and improves cell survival in response to growth-induced pressure. We also found that the cell wall integrity (CWI) pathway contributes to the response to mechanical compressive stress. These latter results are confirmed in complimentary experiments in Mishra et al. [Mishra R, et al. (2017) Proc Natl Acad Sci USA, 10.1073/pnas.1709079114]. When both the SMuSh and the CWI pathways are deleted, cells fail to adapt to compressive stress, and all cells lyse at relatively low pressure when grown in confinement. Thus, we define a network that is essential for cell survival during growth under pressure. We term this mechanosensory system the SCWISh (survival through the CWI and SMuSh) network.


Asunto(s)
Pared Celular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Estrés Mecánico , Citoesqueleto de Actina/metabolismo , Ciclo Celular , Péptidos y Proteínas de Señalización Intracelular/genética , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Mecanotransducción Celular , Proteínas de la Membrana/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
4.
Methods ; 94: 114-9, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26210402

RESUMEN

There is increasing evidence that multicellular structures respond to mechanical cues, such as the confinement and compression exerted by the surrounding environment. In order to understand the response of tissues to stress, we investigate the effect of an isotropic stress on different biological systems. The stress is generated using the osmotic pressure induced by a biocompatible polymer. We compare the response of multicellular spheroids, individual cells and matrigel to the same osmotic perturbation. Our findings indicate that the osmotic pressure occasioned by polymers acts on these systems like an isotropic mechanical stress. When submitted to this pressure, the volume of multicellular spheroids decreases much more than one could expect from the behavior of individual cells.


Asunto(s)
Esferoides Celulares/fisiología , Resinas Acrílicas/química , Animales , Agregación Celular , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Tamaño de la Célula , Mecanotransducción Celular , Ratones , Presión Osmótica
5.
Biophys J ; 107(8): 1821-1828, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25418163

RESUMEN

In most instances, the growth of solid tumors occurs in constrained environments and requires a competition for space. A mechanical crosstalk can arise from this competition. In this article, we dissect the biomechanical sequence caused by a controlled compressive stress on multicellular spheroids (MCSs) used as a tumor model system. On timescales of minutes, we show that a compressive stress causes a reduction of the MCS volume, linked to a reduction of the cell volume in the core of the MCS. On timescales of hours, we observe a reversible induction of the proliferation inhibitor, p27Kip1, from the center to the periphery of the spheroid. On timescales of days, we observe that cells are blocked in the cell cycle at the late G1 checkpoint, the restriction point. We show that the effect of pressure on the proliferation can be antagonized by silencing p27Kip1. Finally, we quantify a clear correlation between the pressure-induced volume change and the growth rate of the spheroid. The compression-induced proliferation arrest that we studied is conserved for five cell lines, and is completely reversible. It demonstrates a generic crosstalk between mechanical stresses and the key players of cell cycle regulation. Our results suggest a role of volume change in the sensitivity to pressure, and that p27Kip1 is strongly influenced by this change.


Asunto(s)
Proliferación Celular , Tamaño de la Célula , Fuerza Compresiva , Esferoides Celulares/fisiología , Animales , Puntos de Control de la Fase G1 del Ciclo Celular , Células HT29 , Humanos , Ratones , Esferoides Celulares/citología
6.
APL Bioeng ; 8(2): 026122, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38894959

RESUMEN

Micropipette aspiration (MPA) is one of the gold standards for quantifying biological samples' mechanical properties, which are crucial from the cell membrane scale to the multicellular tissue. However, relying on the manipulation of individual home-made glass pipettes, MPA suffers from low throughput and no automation. Here, we introduce the sliding insert micropipette aspiration method, which permits parallelization and automation, thanks to the insertion of tubular pipettes, obtained by photolithography, within microfluidic channels. We show its application both at the lipid bilayer level, by probing vesicles to measure membrane bending and stretching moduli, and at the tissue level by quantifying the viscoelasticity of 3D cell aggregates. This approach opens the way to high-throughput, quantitative mechanical testing of many types of biological samples, from vesicles and individual cells to cell aggregates and explants, under dynamic physico-chemical stimuli.

7.
bioRxiv ; 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38585850

RESUMEN

The crowded bacterial cytoplasm is comprised of biomolecules that span several orders of magnitude in size and electrical charge. This complexity has been proposed as the source of the rich spatial organization and apparent anomalous diffusion of intracellular components, although this has not been tested directly. Here, we use biplane microscopy to track the 3D motion of self-assembled bacterial Genetically Encoded Multimeric nanoparticles (bGEMs) with tunable size (20 to 50 nm) and charge (-2160 to +1800 e) in live Escherichia coli cells. To probe intermolecular details at spatial and temporal resolutions beyond experimental limits, we also developed a colloidal whole-cell model that explicitly represents the size and charge of cytoplasmic macromolecules and the porous structure of the bacterial nucleoid. Combining these techniques, we show that bGEMs spatially segregate by size, with small 20-nm particles enriched inside the nucleoid, and larger and/or positively charged particles excluded from this region. Localization is driven by entropic and electrostatic forces arising from cytoplasmic polydispersity, nucleoid structure, geometrical confinement, and interactions with other biomolecules including ribosomes and DNA. We observe that at the timescales of traditional single molecule tracking experiments, motion appears sub-diffusive for all particle sizes and charges. However, using computer simulations with higher temporal resolution, we find that the apparent anomalous exponents are governed by the region of the cell in which bGEMs are located. Molecular motion does not display anomalous diffusion on short time scales and the apparent sub-diffusion arises from geometrical confinement within the nucleoid and by the cell boundary.

8.
Phys Rev Lett ; 110(13): 138103, 2013 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-23581378

RESUMEN

Collective cell motion is observed in a wide range of biological processes. In tumors, physiological gradients of nutrients, growth factors, or even oxygen give rise to gradients of proliferation. We show using fluorescently labeled particles that these gradients drive a velocity field resulting in a cellular flow in multicellular spheroids. Under mechanical stress, the cellular flow is drastically reduced. We describe the results with a hydrodynamic model that considers only convection of the particles by the cellular flow.


Asunto(s)
Movimiento Celular/fisiología , Modelos Biológicos , Esferoides Celulares/citología , Animales , Compuestos Inorgánicos de Carbono/química , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Neoplasias del Colon/patología , Medios de Cultivo , Dextranos/química , Colorantes Fluorescentes/química , Hidrodinámica , Ratones , Nanopartículas/química , Dióxido de Silicio/química , Estrés Mecánico , Sulfuros/química
9.
Curr Opin Cell Biol ; 85: 102269, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37897928

RESUMEN

All living cells are crowded with macromolecules. Crowding can directly modulate biochemical reactions to various degrees depending on the sizes, shapes, and binding affinities of the reactants. Here, we explore the possibility that cells can sense and adapt to changes in crowding through the widespread modulation of biochemical reactions without the need for a dedicated sensor. Additionally, we explore phase separation as a general physicochemical response to changes in crowding, and a mechanism to both transduce information and physically restore crowding homeostasis.


Asunto(s)
Fenómenos Fisiológicos Celulares , Sustancias Macromoleculares/metabolismo
10.
Lab Chip ; 23(20): 4445-4455, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37740366

RESUMEN

Conventional culture conditions are oftentimes insufficient to study tissues, organisms, or 3D multicellular assemblies. They lack both dynamic chemical and mechanical control over the microenvironment. While specific microfluidic devices have been developed to address chemical control, they often do not allow the control of compressive forces emerging when cells proliferate in a confined environment. Here, we present a generic microfluidic device to control both chemical and mechanical compressive forces. This device relies on the use of sliding elements consisting of microfabricated rods that can be inserted inside a microfluidic device. Sliding elements enable the creation of reconfigurable closed culture chambers for the study of whole organisms or model micro-tissues. By confining the micro-tissues, we studied the biophysical impact of growth-induced pressure and showed that this mechanical stress is associated with an increase in macromolecular crowding, shedding light on this understudied type of mechanical stress. Our mechano-chemostat allows the long-term culture of biological samples and can be used to study both the impact of specific conditions as well as the consequences of mechanical compression.


Asunto(s)
Microfluídica , Estrés Mecánico , Presión
11.
Nat Phys ; 18(4): 411-416, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37152719

RESUMEN

Cells that grow in confined spaces eventually build up mechanical compressive stress. This growth-induced pressure (GIP) decreases cell growth. GIP is important in a multitude of contexts from cancer, to microbial infections, to biofouling, yet our understanding of its origin and molecular consequences remains limited. Here, we combine microfluidic confinement of the yeast Saccharomyces cerevisiae, with rheological measurements using genetically encoded multimeric nanoparticles (GEMs) to reveal that growth-induced pressure is accompanied with an increase in a key cellular physical property: macromolecular crowding. We develop a fully calibrated model that predicts how increased macromolecular crowding hinders protein expression and thus diminishes cell growth. This model is sufficient to explain the coupling of growth rate to pressure without the need for specific molecular sensors or signaling cascades. As molecular crowding is similar across all domains of life, this could be a deeply conserved mechanism of biomechanical feedback that allows environmental sensing originating from the fundamental physical properties of cells.

12.
Phys Rev Lett ; 107(18): 188102, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-22107677

RESUMEN

The precise role of the microenvironment on tumor growth is poorly understood. Whereas the tumor is in constant competition with the surrounding tissue, little is known about the mechanics of this interaction. Using a novel experimental procedure, we study quantitatively the effect of an applied mechanical stress on the long-term growth of a spheroid cell aggregate. We observe that a stress of 10 kPa is sufficient to drastically reduce growth by inhibition of cell proliferation mainly in the core of the spheroid. We compare the results to a simple numerical model developed to describe the role of mechanics in cancer progression.


Asunto(s)
Esferoides Celulares/patología , Estrés Fisiológico , Apoptosis , Proliferación Celular , Simulación por Computador , Humanos , Modelos Biológicos , Células Tumorales Cultivadas
13.
Adv Biol (Weinh) ; 5(7): e2100484, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33969641

RESUMEN

Microdevices composed of microwell arrays integrating nanoelectrodes (OptoElecWell) are developed to achieve dual high-resolution optical and electrochemical detections on single Saccharomyces cerevisiae yeast cells. Each array consists of 1.6 × 105 microwells measuring 8 µm in diameter and 5 µm height, with a platinum nanoring electrode for in situ electrochemistry, all integrated on a transparent thin wafer for further high-resolution live-cell imaging. After optimizing the filling rate, 32% of cells are effectively trapped within microwells. This allows to analyse S. cerevisiae metabolism associated with basal respiration while simultaneously measuring optically other cellular parameters. In this study, the impact of glucose concentration on respiration and intracellular rheology is focused. It is found that while the oxygen uptake rate decreases with increasing glucose concentration, diffusion of tracer nanoparticles increases. The OptoElecWell-based respiration methodology provides similar results compared to the commercial gold-standard Seahorse XF analyzer, while using 20 times fewer biological samples, paving the way to achieve single cell metabolomics. In addition, it facilitates an optical route to monitor the contents within single cells. The proposed device, in combination with the dual detection analysis, opens up new avenues for measuring cellular metabolism, and relating it to cellular physiological indicators at single cell level.


Asunto(s)
Saccharomyces cerevisiae , Saccharomycetales , Electrodos , Oxígeno , Reología
14.
Chemphyschem ; 11(8): 1667-72, 2010 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-20373509

RESUMEN

We report on a new, original and efficient method for pi-stacking functionalization of single-wall carbon nanotubes. This method is applied to the synthesis of a high-yield light-harvesting system combining single-wall carbon nanotubes and porphyrin molecules. We developed a micelle-swelling technique that leads to controlled and stable complexes presenting an efficient energy transfer. We demonstrate the key role of the organic solvent in the functionalization mechanism. By swelling the micelles, the solvent helps the non-water-soluble porphyrins to reach the micelle core and allows a strong enhancement of the interaction between porphyrins and nanotubes. This technique opens new avenues for the functionalization of carbon nanostructures.

15.
PLoS One ; 12(8): e0182633, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28813456

RESUMEN

Increasingly accurate and massive data have recently shed light on the fundamental question of how cells maintain a stable size trajectory as they progress through the cell cycle. Microbes seem to use strategies ranging from a pure sizer, where the end of a given phase is triggered when the cell reaches a critical size, to pure adder, where the cell adds a constant size during a phase. Yet the biological origins of the observed spectrum of behavior remain elusive. We analyze a molecular size-control mechanism, based on experimental data from the yeast S. cerevisiae, that gives rise to behaviors smoothly interpolating between adder and sizer. The size-control is obtained from the accumulation of an activator protein that titrates an inhibitor protein. Strikingly, the size-control is composed of two different regimes: for small initial cell size, the size-control is a sizer, whereas for larger initial cell size, it is an imperfect adder, in agreement with recent experiments. Our model thus indicates that the adder and critical size behaviors may just be different dynamical regimes of a single simple biophysical mechanism.


Asunto(s)
Tamaño de la Célula , Modelos Biológicos , Algoritmos , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/fisiología
16.
Methods Mol Biol ; 1612: 269-279, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28634950

RESUMEN

During tumor progression, cancer cells acquire the ability to escape the primary tumor and invade adjacent tissues. They migrate through the stroma to reach blood or lymphatics vessels that will allow them to disseminate throughout the body and form metastasis at distant organs. To assay invasion capacity of cells in vitro, multicellular spheroids of cancer cells, mimicking primary tumor, are commonly embedded in collagen I extracellular matrix, which mimics the stroma. However, due to their higher density, spheroids tend to sink at the bottom of the collagen droplets, resulting in the spreading of the cells on two dimensions. We developed an innovative method based on droplet microfluidics to embed and control the position of multicellular spheroids inside spherical droplets of collagen. In this method cancer cells are exposed to a uniform three-dimensional (3D) collagen environment resulting in 3D cell invasion.


Asunto(s)
Colágeno/química , Microfluídica/métodos , Invasividad Neoplásica , Esferoides Celulares/citología , Animales , Línea Celular Tumoral , Movimiento Celular , Humanos , Ratones , Microfluídica/instrumentación , Modelos Biológicos , Células 3T3 NIH , Esferoides Celulares/patología
17.
Nat Phys ; 12(8): 762-766, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27642362

RESUMEN

In natural settings, microbes tend to grow in dense populations [1-4] where they need to push against their surroundings to accommodate space for new cells. The associated contact forces play a critical role in a variety of population-level processes, including biofilm formation [5-7], the colonization of porous media [8, 9], and the invasion of biological tissues [10-12]. Although mechanical forces have been characterized at the single cell level [13-16], it remains elusive how collective pushing forces result from the combination of single cell forces. Here, we reveal a collective mechanism of confinement, which we call self-driven jamming, that promotes the build-up of large mechanical pressures in microbial populations. Microfluidic experiments on budding yeast populations in space-limited environments show that self-driven jamming arises from the gradual formation and sudden collapse of force chains driven by microbial proliferation, extending the framework of driven granular matter [17-20]. The resulting contact pressures can become large enough to slow down cell growth, to delay the cell cycle in the G1 phase, and to strain or even destroy the microenvironment through crack propagation. Our results suggest that self-driven jamming and build-up of large mechanical pressures is a natural tendency of microbes growing in confined spaces, contributing to microbial pathogenesis and biofouling [21-26].

18.
Interface Focus ; 4(6): 20140033, 2014 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-25485084

RESUMEN

We discuss the short-time response of a multicellular spheroid to an external pressure jump. Our experiments show that 5 min after the pressure jump, the cell density increases in the centre of the spheroid but does not change appreciably close to the surface of the spheroid. This result can be explained if the cells are polarized which we show to be the case. Motivated by the experimental results, we develop a theory for polarized spheroids where the cell polarity is radial (except in a thin shell close to the spheroid surface). The theory takes into account the dependence of cell division and apoptosis rates on the local stress, the cell polarity and active stress generated by the cells and the dependence of active stress on the local pressure. We find a short-time increase of the cell density after a pressure jump that decays as a power law from the spheroid centre, which is in reasonable agreement with the experimental results. By comparing our theory to experiments, we can estimate the isotropic compression modulus of the tissue.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA