Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nat Mater ; 22(2): 249-259, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36357687

RESUMEN

While mechanical stimulation is known to regulate a wide range of biological processes at the cellular and tissue levels, its medical use for tissue regeneration and rehabilitation has been limited by the availability of suitable devices. Here we present a mechanically active gel-elastomer-nitinol tissue adhesive (MAGENTA) that generates and delivers muscle-contraction-mimicking stimulation to a target tissue with programmed strength and frequency. MAGENTA consists of a shape memory alloy spring that enables actuation up to 40% strain, and an adhesive that efficiently transmits the actuation to the underlying tissue. MAGENTA activates mechanosensing pathways involving yes-associated protein and myocardin-related transcription factor A, and increases the rate of muscle protein synthesis. Disuse muscles treated with MAGENTA exhibit greater size and weight, and generate higher forces compared to untreated muscles, demonstrating the prevention of atrophy. MAGENTA thus has promising applications in the treatment of muscle atrophy and regenerative medicine.


Asunto(s)
Músculo Esquelético , Adhesivos Tisulares , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Adhesivos Tisulares/metabolismo , Colorantes de Rosanilina/metabolismo , Atrofia Muscular/prevención & control , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Contracción Muscular
2.
Chem Rev ; 121(18): 11336-11384, 2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-33507740

RESUMEN

Polymeric tissue adhesives provide versatile materials for wound management and are widely used in a variety of medical settings ranging from minor to life-threatening tissue injuries. Compared to the traditional methods of wound closure (i.e., suturing and stapling), they are relatively easy to use, enable rapid application, and introduce minimal tissue damage. Furthermore, they can act as hemostats to control bleeding and provide a tissue-healing environment at the wound site. Despite their numerous current applications, tissue adhesives still face several limitations and unresolved challenges (e.g., weak adhesion strength and poor mechanical properties) that limit their use, leaving ample room for future improvements. Successful development of next-generation adhesives will likely require a holistic understanding of the chemical and physical properties of the tissue-adhesive interface, fundamental mechanisms of tissue adhesion, and requirements for specific clinical applications. In this review, we discuss a set of rational guidelines for design of adhesives, recent progress in the field along with examples of commercially available adhesives and those under development, tissue-specific considerations, and finally potential functions for future adhesives. Advances in tissue adhesives will open new avenues for wound care and potentially provide potent therapeutics for various medical applications.


Asunto(s)
Adhesivos Tisulares , Polímeros/química , Adhesivos Tisulares/uso terapéutico , Cicatrización de Heridas
3.
Nat Mater ; 20(9): 1290-1299, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33875851

RESUMEN

Cell migration on two-dimensional substrates is typically characterized by lamellipodia at the leading edge, mature focal adhesions and spread morphologies. These observations result from adherent cell migration studies on stiff, elastic substrates, because most cells do not migrate on soft, elastic substrates. However, many biological tissues are soft and viscoelastic, exhibiting stress relaxation over time in response to a deformation. Here, we have systematically investigated the impact of substrate stress relaxation on cell migration on soft substrates. We observed that cells migrate minimally on substrates with an elastic modulus of 2 kPa that are elastic or exhibit slow stress relaxation, but migrate robustly on 2-kPa substrates that exhibit fast stress relaxation. Strikingly, migrating cells were not spread out and did not extend lamellipodial protrusions, but were instead rounded, with filopodia protrusions extending at the leading edge, and exhibited small nascent adhesions. Computational models of cell migration based on a motor-clutch framework predict the observed impact of substrate stress relaxation on cell migration and filopodia dynamics. Our findings establish substrate stress relaxation as a key requirement for robust cell migration on soft substrates and uncover a mode of two-dimensional cell migration marked by round morphologies, filopodia protrusions and weak adhesions.


Asunto(s)
Movimiento Celular , Seudópodos/metabolismo , Membrana Basal/metabolismo , Fenómenos Biomecánicos , Adhesión Celular , Línea Celular , Línea Celular Tumoral , Elasticidad , Humanos
4.
Proc Natl Acad Sci U S A ; 113(20): 5492-7, 2016 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-27140623

RESUMEN

The extracellular matrix (ECM) is a complex assembly of structural proteins that provides physical support and biochemical signaling to cells in tissues. The mechanical properties of the ECM have been found to play a key role in regulating cell behaviors such as differentiation and malignancy. Gels formed from ECM protein biopolymers such as collagen or fibrin are commonly used for 3D cell culture models of tissue. One of the most striking features of these gels is that they exhibit nonlinear elasticity, undergoing strain stiffening. However, these gels are also viscoelastic and exhibit stress relaxation, with the resistance of the gel to a deformation relaxing over time. Recent studies have suggested that cells sense and respond to both nonlinear elasticity and viscoelasticity of ECM, yet little is known about the connection between nonlinear elasticity and viscoelasticity. Here, we report that, as strain is increased, not only do biopolymer gels stiffen but they also exhibit faster stress relaxation, reducing the timescale over which elastic energy is dissipated. This effect is not universal to all biological gels and is mediated through weak cross-links. Mechanistically, computational modeling and atomic force microscopy (AFM) indicate that strain-enhanced stress relaxation of collagen gels arises from force-dependent unbinding of weak bonds between collagen fibers. The broader effect of strain-enhanced stress relaxation is to rapidly diminish strain stiffening over time. These results reveal the interplay between nonlinear elasticity and viscoelasticity in collagen gels, and highlight the complexity of the ECM mechanics that are likely sensed through cellular mechanotransduction.


Asunto(s)
Colágeno/química , Simulación por Computador , Elasticidad , Proteínas de la Matriz Extracelular/química , Geles/química , Estrés Mecánico , Viscosidad
5.
Biophys J ; 114(2): 450-461, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29401442

RESUMEN

Contractile cells can reorganize fibrous extracellular matrices and form dense tracts of fibers between neighboring cells. These tracts guide the development of tubular tissue structures and provide paths for the invasion of cancer cells. Here, we studied the mechanisms of the mechanical plasticity of collagen tracts formed by contractile premalignant acinar cells and fibroblasts. Using fluorescence microscopy and second harmonic generation, we quantified the collagen densification, fiber alignment, and strains that remain within the tracts after cellular forces are abolished. We explained these observations using a theoretical fiber network model that accounts for the stretch-dependent formation of weak cross-links between nearby fibers. We tested the predictions of our model using shear rheology experiments. Both our model and rheological experiments demonstrated that increasing collagen concentration leads to substantial increases in plasticity. We also considered the effect of permanent elongation of fibers on network plasticity and derived a phase diagram that classifies the dominant mechanisms of plasticity based on the rate and magnitude of deformation and the mechanical properties of individual fibers. Plasticity is caused by the formation of new cross-links if moderate strains are applied at small rates or due to permanent fiber elongation if large strains are applied over short periods. Finally, we developed a coarse-grained model for plastic deformation of collagen networks that can be employed to simulate multicellular interactions in processes such as morphogenesis, cancer invasion, and fibrosis.


Asunto(s)
Colágeno/metabolismo , Fenómenos Mecánicos , Animales , Fenómenos Biomecánicos , Matriz Extracelular/metabolismo , Fibroblastos/citología , Ratones , Modelos Biológicos , Células 3T3 NIH , Ratas , Esferoides Celulares/metabolismo , Estrés Mecánico
6.
Biophys J ; 111(10): 2296-2308, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27851951

RESUMEN

Living tissues consist largely of cells and extracellular matrices (ECMs). The mechanical properties of ECM have been found to play a key role in regulating cell behaviors such as migration, proliferation, and differentiation. Although most studies to date have focused on elucidating the impact of matrix elasticity on cell behaviors, recent studies have revealed an impact of matrix viscoelasticity on cell behaviors and reported plastic remodeling of ECM by cells. In this study, we rigorously characterized the plasticity in materials commonly used for cell culture. This characterization of plasticity revealed time-dependent plasticity, or viscoplasticity, in collagen gels, reconstituted basement membrane matrix, agarose gels, alginate gels, and fibrin gels, but not in polyacrylamide gels. Viscoplasticity was associated with gels that contained weak bonds, and covalent cross-linking diminished viscoplasticity in collagen and alginate gels. Interestingly, the degree of plasticity was found to be nonlinear, or dependent on the magnitude of stress or strain, in collagen gels, but not in the other viscoplastic materials. Viscoplastic models were employed to describe plasticity in the viscoplastic materials. Relevance of matrix viscoplasticity to cell-matrix interactions was established through a quantitative assessment of plastic remodeling of collagen gels by cells. Plastic remodeling of collagen gels was found to be dependent on cellular force, mediated through integrin-based adhesions, and occurred even with inhibition of proteolytic degradation of the matrix. Together, these results reveal that matrix viscoplasticity facilitates plastic remodeling of matrix by cellular forces.


Asunto(s)
Elasticidad , Matriz Extracelular/metabolismo , Células 3T3 , Animales , Fenómenos Biomecánicos , Colágeno/química , Colágeno/metabolismo , Integrina beta1/metabolismo , Ensayo de Materiales , Ratones , Proteolisis , Viscosidad
7.
Phys Rev Lett ; 114(11): 114501, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25839275

RESUMEN

Direct evidence is provided for the transition from surface conduction (SC) to electro-osmotic flow (EOF) above a critical channel depth (d) of a nanofluidic device. The dependence of the overlimiting conductance (OLC) on d is consistent with theoretical predictions, scaling as d(-1) for SC and d(4/5) for EOF with a minimum around d=8 µm. The propagation of transient deionization shocks is also visualized, revealing complex patterns of EOF vortices and unstable convection with increasing d. This unified picture of surface-driven OLC can guide further advances in electrokinetic theory, as well as engineering applications of ion concentration polarization in microfluidics and porous media.


Asunto(s)
Técnicas Analíticas Microfluídicas/instrumentación , Técnicas Analíticas Microfluídicas/métodos , Modelos Teóricos , Técnicas Electroquímicas/instrumentación , Técnicas Electroquímicas/métodos , Nanotecnología/instrumentación , Nanotecnología/métodos , Presión Osmótica , Propiedades de Superficie
8.
Biomaterials ; 309: 122597, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38696944

RESUMEN

Wounds often necessitate the use of instructive biomaterials to facilitate effective healing. Yet, consistently filling the wound and retaining the material in place presents notable challenges. Here, we develop a new class of injectable tissue adhesives by leveraging the dynamic crosslinking chemistry of Schiff base reactions. These adhesives demonstrate outstanding mechanical properties, especially in regard to stretchability and self-healing capacity, and biodegradability. Furthermore, they also form robust adhesion to biological tissues. Their therapeutic potential was evaluated in a rodent model of volumetric muscle loss (VML). Ultrasound imaging confirmed that the adhesives remained within the wound site, effectively filled the void, and degraded at a rate comparable to the healing process. Histological analysis indicated that the adhesives facilitated muscle fiber and blood vessel formation, and induced anti-inflammatory macrophages. Notably, the injured muscles of mice treated with the adhesives displayed increased weight and higher force generation than the control groups. This approach to adhesive design paves the way for the next generation of medical adhesives in tissue repair.


Asunto(s)
Regeneración , Adhesivos Tisulares , Cicatrización de Heridas , Animales , Adhesivos Tisulares/química , Adhesivos Tisulares/farmacología , Cicatrización de Heridas/efectos de los fármacos , Regeneración/efectos de los fármacos , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/lesiones , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Ratones Endogámicos C57BL , Masculino
9.
Nat Biomed Eng ; 6(10): 1167-1179, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34980903

RESUMEN

Hydrogels that provide mechanical support and sustainably release therapeutics have been used to treat tendon injuries. However, most hydrogels are insufficiently tough, release drugs in bursts, and require cell infiltration or suturing to integrate with surrounding tissue. Here we report that a hydrogel serving as a high-capacity drug depot and combining a dissipative tough matrix on one side and a chitosan adhesive surface on the other side supports tendon gliding and strong adhesion (larger than 1,000 J m-2) to tendon on opposite surfaces of the hydrogel, as we show with porcine and human tendon preparations during cyclic-friction loadings. The hydrogel is biocompatible, strongly adheres to patellar, supraspinatus and Achilles tendons of live rats, boosted healing and reduced scar formation in a rat model of Achilles-tendon rupture, and sustainably released the corticosteroid triamcinolone acetonide in a rat model of patellar tendon injury, reducing inflammation, modulating chemokine secretion, recruiting tendon stem and progenitor cells, and promoting macrophage polarization to the M2 phenotype. Hydrogels with 'Janus' surfaces and sustained-drug-release functionality could be designed for a range of biomedical applications.


Asunto(s)
Tendón Calcáneo , Quitosano , Traumatismos de los Tendones , Ratas , Humanos , Porcinos , Animales , Hidrogeles , Quitosano/metabolismo , Adhesivos/metabolismo , Triamcinolona Acetonida/metabolismo , Traumatismos de los Tendones/tratamiento farmacológico , Traumatismos de los Tendones/metabolismo , Tendón Calcáneo/metabolismo , Quimiocinas/metabolismo
10.
Adv Sci (Weinh) ; 8(4): 2000403, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33643782

RESUMEN

Cell elongation along the division axis, or mitotic elongation, mediates proper segregation of chromosomes and other intracellular materials, and is required for completion of cell division. In three-dimensionally confining extracellular matrices, such as dense collagen gels, dividing cells must generate space to allow mitotic elongation to occur. In principle, cells can generate space for mitotic elongation during cell spreading, prior to mitosis, or via extracellular force generation or matrix degradation during mitosis. However, the processes by which cells drive mitotic elongation in collagen-rich extracellular matrices remains unclear. Here, it is shown that single cancer cells generate substantial pushing forces on the surrounding collagen extracellular matrix to drive cell division in confining collagen gels and allow mitotic elongation to proceed. Neither cell spreading, prior to mitosis, nor matrix degradation, during spreading or mitotic elongation, are found to be required for mitotic elongation. Mechanistically, laser ablation studies, pharmacological inhibition studies, and computational modeling establish that pushing forces generated during mitosis in collagen gels arise from a combination of interpolar spindle elongation and cytokinetic ring contraction. These results reveal a fundamental mechanism mediating cell division in confining extracellular matrices, providing insight into how tumor cells are able to proliferate in dense collagen-rich tissues.

11.
J Cell Biol ; 220(8)2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34132746

RESUMEN

Epithelial cells undergo striking morphological changes during division to ensure proper segregation of genetic and cytoplasmic materials. These morphological changes occur despite dividing cells being mechanically restricted by neighboring cells, indicating the need for extracellular force generation. Beyond driving cell division itself, forces associated with division have been implicated in tissue-scale processes, including development, tissue growth, migration, and epidermal stratification. While forces generated by mitotic rounding are well understood, forces generated after rounding remain unknown. Here, we identify two distinct stages of division force generation that follow rounding: (1) Protrusive forces along the division axis that drive division elongation, and (2) outward forces that facilitate postdivision spreading. Cytokinetic ring contraction of the dividing cell, but not activity of neighboring cells, generates extracellular forces that propel division elongation and contribute to chromosome segregation. Forces from division elongation are observed in epithelia across many model organisms. Thus, division elongation forces represent a universal mechanism that powers cell division in confining epithelia.


Asunto(s)
División Celular , Forma de la Célula , Células Epiteliales/fisiología , Mecanotransducción Celular , Animales , Animales Modificados Genéticamente , Comunicación Celular , Segregación Cromosómica , Simulación por Computador , Perros , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Células Epiteliales/metabolismo , Células de Riñón Canino Madin Darby , Microscopía Confocal , Microscopía Fluorescente , Modelos Biológicos , Estrés Mecánico , Factores de Tiempo , Imagen de Lapso de Tiempo
12.
Viruses ; 13(8)2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34452517

RESUMEN

Patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019, suffer from respiratory and non-respiratory symptoms. Among these symptoms, the loss of smell has attracted considerable attention. The objectives of this study were to determine which cells are infected, what happens in the olfactory system after viral infection, and how these pathologic changes contribute to olfactory loss. For this purpose, Syrian golden hamsters were used. First, we verified the olfactory structures in the nasal cavity of Syrian golden hamsters, namely the main olfactory epithelium, the vomeronasal organ, and their cellular components. Second, we found angiotensin-converting enzyme 2 expression, a receptor protein of SARS-CoV-2, in both structures and infections of supporting, microvillar, and solitary chemosensory cells. Third, we observed pathological changes in the infected epithelium, including reduced thickness of the mucus layer, detached epithelia, indistinct layers of epithelia, infiltration of inflammatory cells, and apoptotic cells in the overall layers. We concluded that a structurally and functionally altered microenvironment influences olfactory function. We observed the regeneration of the damaged epithelium, and found multilayers of basal cells, indicating that they were activated and proliferating to reconstitute the injured epithelium.


Asunto(s)
COVID-19/virología , Células Quimiorreceptoras/virología , Mucosa Olfatoria/virología , SARS-CoV-2 , Órgano Vomeronasal/virología , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/patología , Células Quimiorreceptoras/patología , Masculino , Mesocricetus , Cavidad Nasal/patología , Cavidad Nasal/virología , Mucosa Olfatoria/metabolismo , Mucosa Olfatoria/patología , Neuronas Receptoras Olfatorias/metabolismo , Neuronas Receptoras Olfatorias/patología , Neuronas Receptoras Olfatorias/virología , Receptores de Coronavirus/metabolismo , Regeneración , SARS-CoV-2/aislamiento & purificación , Órgano Vomeronasal/metabolismo , Órgano Vomeronasal/patología
13.
Sci Transl Med ; 13(614): eabe8868, 2021 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-34613813

RESUMEN

Mechanical stimulation (mechanotherapy) can promote skeletal muscle repair, but a lack of reproducible protocols and mechanistic understanding of the relation between mechanical cues and tissue regeneration limit progress in this field. To address these gaps, we developed a robotic device equipped with real-time force control and compatible with ultrasound imaging for tissue strain analysis. We investigated the hypothesis that specific mechanical loading improves tissue repair by modulating inflammatory responses that regulate skeletal muscle regeneration. We report that cyclic compressive loading within a specific range of forces substantially improves functional recovery of severely injured muscle in mice. This improvement is attributable in part to rapid clearance of neutrophil populations and neutrophil-mediated factors, which otherwise may impede myogenesis. Insights from this work will help advance therapeutic strategies for tissue regeneration broadly.


Asunto(s)
Procedimientos Quirúrgicos Robotizados , Robótica , Músculo Esquelético , Neutrófilos , Regeneración
14.
Biomaterials ; 200: 15-24, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30743050

RESUMEN

Hydrogels are commonly used as artificial extracellular matrices for 3D cell culture and for tissue engineering. Viscoelastic hydrogels with tunable stress relaxation have recently been developed, and stress relaxation in the hydrogels has been found to play a key role in regulating cell behaviors such as differentiation, spreading, and proliferation. Here we report a simple but precise materials approach to tuning stress relaxation of alginate hydrogels with polyethylene glycol (PEG) covalently grafted onto the alginate. Hydrogel relaxation was modulated independent of the initial elastic modulus by varying molecular weight and concentration of PEG along with calcium crosslinking of the alginate. Increased concentration and molecular weight of the PEG resulted in faster stress relaxation, a higher loss modulus, and increased creep. Interestingly, we found that stress relaxation of the hydrogels is determined by the total mass amount of PEG in the hydrogel, and not the molecular weight or concentration of PEG chains alone. We then evaluated the utility of these hydrogels for 3D cell culture. Faster relaxation in RGD-coupled alginate-PEG hydrogels led to increased spreading and proliferation of fibroblasts, and enhanced osteogenic differentiation of mesenchymal stem cells (MSCs). Thus, this work establishes a new materials approach to tuning stress relaxation in alginate hydrogels for 3D cell culture.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Hidrogeles/química , Polietilenglicoles/química , Estrés Mecánico , Células 3T3 , Alginatos/química , Animales , Diferenciación Celular , Integrina beta1/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Peso Molecular , Osteogénesis , Paxillin/metabolismo
15.
Cell Cycle ; 18(15): 1671-1675, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31234701

RESUMEN

The mitotic spindle has long been known to play a crucial role in mitosis, orchestrating the segregation of chromosomes into two daughter cells during mitosis with high fidelity. Intracellular forces generated by the mitotic spindle are increasingly well understood, and recent work has revealed that the efficiency and the accuracy of mitosis is ensured by the scaling of mitotic spindle size with cell size. However, the role of the spindle in cancer progression has largely been ignored. Two recent studies point toward the role of mitotic spindle evolution in cancer progression through extracellular force generation. Cancer cells with lengthened spindles exhibit highly increased metastatic potential. Further, interpolar spindle elongation drives protrusive extracellular force generation along the mitotic axis to allow mitotic elongation, a morphological change that is required for cell division. Together, these findings open a new research area studying the role of the mitotic spindle evolution in cancer metastasis.


Asunto(s)
Mitosis , Huso Acromático , Tamaño de la Célula , Segregación Cromosómica , Cromosomas
16.
Sci Rep ; 9(1): 17188, 2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31748579

RESUMEN

Yes-associated protein (YAP) is a transcriptional regulator and mechanotransducer, relaying extracellular matrix (ECM) stiffness into proliferative gene expression in 2D culture. Previous studies show that YAP activation is dependent on F-actin stress fiber mediated nuclear pore opening, however the protein mediators of YAP translocation remain unclear. Here, we show that YAP co-localizes with F-actin during activating conditions, such as sparse plating and culturing on stiff 2D substrates. To identify proteins mediating YAP translocation, we performed co-immunoprecipitation followed by mass spectrometry (co-IP/MS) for proteins that differentially associated with YAP under activating conditions. Interestingly, YAP preferentially associates with ß1 integrin under activating conditions, and ß4 integrin under inactivating conditions. In activating conditions, CRISPR/Cas9 knockout (KO) of ß1 integrin (ΔITGB1) resulted in decreased cell area, which correlated with decreased YAP nuclear localization. ΔITGB1 did not significantly affect the slope of the correlation between YAP nuclear localization with area, but did decrease overall nuclear YAP independently of cell spreading. In contrast, ß4 integrin KO (ΔITGB4) cells showed no change in cell area and similarly decreased nuclear YAP. These results reveal proteins that differentially associate with YAP during activation, which may aid in regulating YAP nuclear translocation.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Matriz Extracelular/metabolismo , Integrina beta1/metabolismo , Integrina beta4/metabolismo , Mecanotransducción Celular , Factores de Transcripción/metabolismo , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/genética , Células Cultivadas , Humanos , Integrina beta1/química , Integrina beta1/genética , Integrina beta4/química , Integrina beta4/genética , Fosforilación , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Transducción de Señal , Factores de Transcripción/genética
17.
Sci Adv ; 5(8): eaaw6171, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31457089

RESUMEN

In tissues, cells reside in confining microenvironments, which may mechanically restrict the ability of a cell to double in size as it prepares to divide. How confinement affects cell cycle progression remains unclear. We show that cells progressed through the cell cycle and proliferated when cultured in hydrogels exhibiting fast stress relaxation but were mostly arrested in the G0/G1 phase of the cell cycle when cultured in hydrogels that exhibit slow stress relaxation. In fast-relaxing gels, activity of stretch-activated channels (SACs), including TRPV4, promotes activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway, which in turn drives cytoplasmic localization of the cell cycle inhibitor p27Kip1, thereby allowing S phase entry and proliferation. Cell growth during G1 activated the TRPV4-PI3K/Akt-p27Kip1 signaling axis, but growth is inhibited in the confining slow-relaxing hydrogels. Thus, in confining microenvironments, cells sense when growth is sufficient for division to proceed through a growth-responsive signaling axis mediated by SACs.


Asunto(s)
Proliferación Celular , Puntos de Control de la Fase G1 del Ciclo Celular , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Canales Catiónicos TRPV/metabolismo , Alginatos/química , Animales , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Módulo de Elasticidad , Humanos , Hidrogeles/química , Presión Osmótica , Fosfatidilinositol 3-Quinasas/química , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Estrés Mecánico , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética
18.
Nat Commun ; 10(1): 1848, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31015465

RESUMEN

Increased tissue stiffness is a driver of breast cancer progression. The transcriptional regulator YAP is considered a universal mechanotransducer, based largely on 2D culture studies. However, the role of YAP during in vivo breast cancer remains unclear. Here, we find that mechanotransduction occurs independently of YAP in breast cancer patient samples and mechanically tunable 3D cultures. Mechanistically, the lack of YAP activity in 3D culture and in vivo is associated with the absence of stress fibers and an order of magnitude decrease in nuclear cross-sectional area relative to 2D culture. This work highlights the context-dependent role of YAP in mechanotransduction, and establishes that YAP does not mediate mechanotransduction in breast cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/patología , Matriz Extracelular/patología , Mecanotransducción Celular , Fosfoproteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Mama/patología , Densidad de la Mama , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Progresión de la Enfermedad , Matriz Extracelular/metabolismo , Femenino , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Invasividad Neoplásica/patología , Fosfoproteínas/genética , Factores de Transcripción , Proteínas Señalizadoras YAP
19.
Nat Biomed Eng ; 3(12): 1009-1019, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31285581

RESUMEN

In breast cancer, the increased stiffness of the extracellular matrix is a key driver of malignancy. Yet little is known about the epigenomic changes that underlie the tumorigenic impact of extracellular matrix mechanics. Here, we show in a three-dimensional culture model of breast cancer that stiff extracellular matrix induces a tumorigenic phenotype through changes in chromatin state. We found that increased stiffness yielded cells with more wrinkled nuclei and with increased lamina-associated chromatin, that cells cultured in stiff matrices displayed more accessible chromatin sites, which exhibited footprints of Sp1 binding, and that this transcription factor acts along with the histone deacetylases 3 and 8 to regulate the induction of stiffness-mediated tumorigenicity. Just as cell culture on soft environments or in them rather than on tissue-culture plastic better recapitulates the acinar morphology observed in mammary epithelium in vivo, mammary epithelial cells cultured on soft microenvironments or in them also more closely replicate the in vivo chromatin state. Our results emphasize the importance of culture conditions for epigenomic studies, and reveal that chromatin state is a critical mediator of mechanotransduction.


Asunto(s)
Neoplasias de la Mama , Cromatina , Epitelio , Fenotipo , Neoplasias de la Mama/patología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Células Epiteliales , Epitelio/patología , Matriz Extracelular/metabolismo , Femenino , Humanos , Mecanotransducción Celular , Factor de Transcripción Sp1 , Factores de Transcripción , Microambiente Tumoral
20.
Biomaterials ; 154: 213-222, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29132046

RESUMEN

The physical and architectural cues of the extracellular matrix (ECM) play a critical role in regulating important cellular functions such as spreading, migration, proliferation, and differentiation. Natural ECM is a complex viscoelastic scaffold composed of various distinct components that are often organized into a fibrillar microstructure. Hydrogels are frequently used as synthetic ECMs for 3D cell culture, but are typically elastic, due to covalent crosslinking, and non-fibrillar. Recent work has revealed the importance of stress relaxation in viscoelastic hydrogels in regulating biological processes such as spreading and differentiation, but these studies all utilize synthetic ECM hydrogels that are non-fibrillar. Key mechanotransduction events, such as focal adhesion formation, have only been observed in fibrillar networks in 3D culture to date. Here we present an interpenetrating network (IPN) hydrogel system based on HA crosslinked with dynamic covalent bonds and collagen I that captures the viscoelasticity and fibrillarity of ECM in tissues. The IPN hydrogels exhibit two distinct processes in stress relaxation, one from collagen and the other from HA crosslinking dynamics. Stress relaxation in the IPN hydrogels can be tuned by modulating HA crosslinker affinity, molecular weight of the HA, or HA concentration. Faster relaxation in the IPN hydrogels promotes cell spreading, fiber remodeling, and focal adhesion (FA) formation - behaviors often inhibited in other hydrogel-based materials in 3D culture. This study presents a new, broadly adaptable materials platform for mimicking key ECM features of viscoelasticity and fibrillarity in hydrogels for 3D cell culture and sheds light on how these mechanical and structural cues regulate cell behavior.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Movimiento Celular , Colágeno/farmacología , Adhesiones Focales/metabolismo , Ácido Hialurónico/farmacología , Hidrogeles/farmacología , Células Madre Mesenquimatosas/citología , Movimiento Celular/efectos de los fármacos , Colágeno/química , Reactivos de Enlaces Cruzados/química , Adhesiones Focales/efectos de los fármacos , Humanos , Ácido Hialurónico/química , Hidrogeles/química , Células Madre Mesenquimatosas/efectos de los fármacos , Reología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA