Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 137(8)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38563084

RESUMEN

Angiogenesis is a tightly controlled dynamic process demanding a delicate equilibrium between pro-angiogenic signals and factors that promote vascular stability. The spatiotemporal activation of the transcriptional co-factors YAP (herein referring to YAP1) and TAZ (also known WWTR1), collectively denoted YAP/TAZ, is crucial to allow for efficient collective endothelial migration in angiogenesis. The focal adhesion protein deleted-in-liver-cancer-1 (DLC1) was recently described as a transcriptional downstream target of YAP/TAZ in endothelial cells. In this study, we uncover a negative feedback loop between DLC1 expression and YAP activity during collective migration and sprouting angiogenesis. In particular, our study demonstrates that signaling via the RhoGAP domain of DLC1 reduces nuclear localization of YAP and its transcriptional activity. Moreover, the RhoGAP activity of DLC1 is essential for YAP-mediated cellular processes, including the regulation of focal adhesion turnover, traction forces, and sprouting angiogenesis. We show that DLC1 restricts intracellular cytoskeletal tension by inhibiting Rho signaling at the basal adhesion plane, consequently reducing nuclear YAP localization. Collectively, these findings underscore the significance of DLC1 expression levels and its function in mitigating intracellular tension as a pivotal mechanotransductive feedback mechanism that finely tunes YAP activity throughout the process of sprouting angiogenesis.


Asunto(s)
Adhesiones Focales , Proteínas Activadoras de GTPasa , Mecanotransducción Celular , Proteínas Supresoras de Tumor , Proteínas Señalizadoras YAP , Animales , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Movimiento Celular , Retroalimentación Fisiológica , Adhesiones Focales/metabolismo , Adhesiones Focales/genética , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Activadoras de GTPasa/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Mecanotransducción Celular/genética , Neovascularización Fisiológica , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Señalizadoras YAP/metabolismo
2.
Nature ; 579(7797): 111-117, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32103177

RESUMEN

The avascular nature of cartilage makes it a unique tissue1-4, but whether and how the absence of nutrient supply regulates chondrogenesis remain unknown. Here we show that obstruction of vascular invasion during bone healing favours chondrogenic over osteogenic differentiation of skeletal progenitor cells. Unexpectedly, this process is driven by a decreased availability of extracellular lipids. When lipids are scarce, skeletal progenitors activate forkhead box O (FOXO) transcription factors, which bind to the Sox9 promoter and increase its expression. Besides initiating chondrogenesis, SOX9 acts as a regulator of cellular metabolism by suppressing oxidation of fatty acids, and thus adapts the cells to an avascular life. Our results define lipid scarcity as an important determinant of chondrogenic commitment, reveal a role for FOXO transcription factors during lipid starvation, and identify SOX9 as a critical metabolic mediator. These data highlight the importance of the nutritional microenvironment in the specification of skeletal cell fate.


Asunto(s)
Huesos/citología , Microambiente Celular , Condrogénesis , Metabolismo de los Lípidos , Factor de Transcripción SOX9/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Huesos/irrigación sanguínea , Condrocitos/citología , Condrocitos/metabolismo , Ácidos Grasos/metabolismo , Femenino , Privación de Alimentos , Factores de Transcripción Forkhead/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Osteogénesis , Oxidación-Reducción , Factor de Transcripción SOX9/genética , Transducción de Señal , Cicatrización de Heridas
3.
Proc Natl Acad Sci U S A ; 120(15): e2216934120, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37011188

RESUMEN

Cells continuously sense external forces from their microenvironment, the extracellular matrix (ECM). In turn, they generate contractile forces, which stiffen and remodel this matrix. Although this bidirectional mechanical exchange is crucial for many cell functions, it remains poorly understood. Key challenges are that the majority of available matrices for such studies, either natural or synthetic, are difficult to control or lack biological relevance. Here, we use a synthetic, yet highly biomimetic hydrogel based on polyisocyanide (PIC) polymers to investigate the effects of the fibrous architecture and the nonlinear mechanics on cell-matrix interactions. Live-cell rheology was combined with advanced microscopy-based approaches to understand the mechanisms behind cell-induced matrix stiffening and plastic remodeling. We demonstrate how cell-mediated fiber remodeling and the propagation of fiber displacements are modulated by adjusting the biological and mechanical properties of this material. Moreover, we validate the biological relevance of our results by demonstrating that cellular tractions in PIC gels develop analogously to those in the natural ECM. This study highlights the potential of PIC gels to disentangle complex bidirectional cell-matrix interactions and to improve the design of materials for mechanobiology studies.


Asunto(s)
Matriz Extracelular , Hidrogeles , Matriz Extracelular/fisiología , Comunicación Celular
4.
Angiogenesis ; 24(4): 843-860, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34342749

RESUMEN

Cerebral cavernous malformation (CCM) is a cerebrovascular disease in which stacks of dilated haemorrhagic capillaries form focally in the brain. Whether and how defective mechanotransduction, cellular mosaicism and inflammation interplay to sustain the progression of CCM disease is unknown. Here, we reveal that CCM1- and CCM2-silenced endothelial cells expanded in vitro enter into senescence-associated secretory phenotype (SASP) that they use to invade the extracellular matrix and attract surrounding wild-type endothelial and immune cells. Further, we demonstrate that this SASP is driven by the cytoskeletal, molecular and transcriptomic disorders provoked by ROCK dysfunctions. By this, we propose that CCM2 and ROCK could be parts of a scaffold controlling senescence, bringing new insights into the emerging field of the control of ageing by cellular mechanics. These in vitro findings reconcile the known dysregulated traits of CCM2-deficient endothelial cells into a unique endothelial fate. Based on these in vitro results, we propose that a SASP could link the increased ROCK-dependent cell contractility in CCM2-deficient endothelial cells with microenvironment remodelling and long-range chemo-attraction of endothelial and immune cells.


Asunto(s)
Células Endoteliales , Hemangioma Cavernoso del Sistema Nervioso Central , Proteínas Portadoras/genética , Células Endoteliales/metabolismo , Humanos , Mecanotransducción Celular , Fenotipo , Fenotipo Secretor Asociado a la Senescencia , Microambiente Tumoral
5.
PLoS Comput Biol ; 16(8): e1007874, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32822340

RESUMEN

Shear stress induces directed endothelial cell (EC) migration in blood vessels leading to vessel diameter increase and induction of vascular maturation. Other factors, such as EC elongation and interaction between ECs and non-vascular areas are also important. Computational models have previously been used to study collective cell migration. These models can be used to predict EC migration and its effect on vascular remodelling during embryogenesis. We combined live time-lapse imaging of the remodelling vasculature of the quail embryo yolk sac with flow quantification using a combination of micro-Particle Image Velocimetry and computational fluid dynamics. We then used the flow and remodelling data to inform a model of EC migration during remodelling. To obtain the relation between shear stress and velocity in vitro for EC cells, we developed a flow chamber to assess how confluent sheets of ECs migrate in response to shear stress. Using these data as an input, we developed a multiphase, self-propelled particles (SPP) model where individual agents are driven to migrate based on the level of shear stress while maintaining appropriate spatial relationship to nearby agents. These agents elongate, interact with each other, and with avascular agents at each time-step of the model. We compared predicted vascular shape to real vascular shape after 4 hours from our time-lapse movies and performed sensitivity analysis on the various model parameters. Our model shows that shear stress has the largest effect on the remodelling process. Importantly, however, elongation played an especially important part in remodelling. This model provides a powerful tool to study the input of different biological processes on remodelling.


Asunto(s)
Hidrodinámica , Remodelación Vascular , Animales , Circulación Sanguínea , Movimiento Celular/fisiología , Forma de la Célula , Biología Computacional , Células Endoteliales/fisiología , Codorniz/anatomía & histología , Codorniz/embriología , Estrés Mecánico
6.
PLoS Comput Biol ; 16(1): e1007250, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31929522

RESUMEN

Actin protrusion dynamics plays an important role in the regulation of three-dimensional (3D) cell migration. Cells form protrusions that adhere to the surrounding extracellular matrix (ECM), mechanically probe the ECM and contract in order to displace the cell body. This results in cell migration that can be directed by the mechanical anisotropy of the ECM. However, the subcellular processes that regulate protrusion dynamics in 3D cell migration are difficult to investigate experimentally and therefore not well understood. Here, we present a computational model of cell migration through a degradable viscoelastic ECM. This model is a 2D representation of 3D cell migration. The cell is modeled as an active deformable object that captures the viscoelastic behavior of the actin cortex and the subcellular processes underlying 3D cell migration. The ECM is regarded as a viscoelastic material, with or without anisotropy due to fibrillar strain stiffening, and modeled by means of the meshless Lagrangian smoothed particle hydrodynamics (SPH) method. ECM degradation is captured by local fluidization of the material and permits cell migration through the ECM. We demonstrate that changes in ECM stiffness and cell strength affect cell migration and are accompanied by changes in number, lifetime and length of protrusions. Interestingly, directly changing the total protrusion number or the average lifetime or length of protrusions does not affect cell migration. A stochastic variability in protrusion lifetime proves to be enough to explain differences in cell migration velocity. Force-dependent adhesion disassembly does not result in faster migration, but can make migration more efficient. We also demonstrate that when a number of simultaneous protrusions is enforced, the optimal number of simultaneous protrusions is one or two, depending on ECM anisotropy. Together, the model provides non-trivial new insights in the role of protrusions in 3D cell migration and can be a valuable contribution to increase the understanding of 3D cell migration mechanics.


Asunto(s)
Actinas , Movimiento Celular/fisiología , Matriz Extracelular , Modelos Biológicos , Actinas/química , Actinas/metabolismo , Actinas/fisiología , Biología Computacional , Simulación por Computador , Elasticidad/fisiología , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Viscosidad
7.
Biophys J ; 119(2): 243-257, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32621867

RESUMEN

The interplay between cell-cell and cell-substrate interactions is complex yet necessary for the formation and healthy functioning of tissues. The same mechanosensing mechanisms used by the cell to sense its extracellular matrix also play a role in intercellular interactions. We used the discrete element method to develop a computational model of a deformable cell that includes subcellular components responsible for mechanosensing. We modeled a three-dimensional cell pair on a patterned (two-dimensional) substrate, a simple laboratory setup to study intercellular interactions. We explicitly modeled focal adhesions and adherens junctions. These mechanosensing adhesions matured, becoming stabilized by force. We also modeled contractile stress fibers that bind the discrete adhesions. The mechanosensing fibers strengthened upon stalling. Traction exerted on the substrate was used to generate traction maps (along the cell-substrate interface). These simulated maps are compared to experimental maps obtained via traction force microscopy. The model recreates the dependence on substrate stiffness of the tractions' spatial distribution, contractile moment of the cell pair, intercellular force, and number of focal adhesions. It also recreates the phenomenon of cell decoupling, in which cells exert forces separately when substrate stiffness increases. More importantly, the model provides viable molecular explanations for decoupling: mechanosensing mechanisms are responsible for competition between different fiber-adhesion configurations present in the cell pair. The point at which an increasing substrate stiffness becomes as high as that of the cell-cell interface is the tipping point at which configurations that favor cell-substrate adhesion dominate over those favoring cell-cell adhesion. This competition is responsible for decoupling.


Asunto(s)
Matriz Extracelular , Adhesiones Focales , Adhesión Celular , Fenómenos Mecánicos , Mecanotransducción Celular , Fibras de Estrés
8.
Angiogenesis ; 23(3): 315-324, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31997048

RESUMEN

Angiogenesis is the formation of new blood vessels from the pre-existing vasculature. It is essential for normal tissue growth and regeneration, and also plays a key role in many diseases [Carmeliet in Nat Med 9:653-660, 2003]. Cytoskeletal components have been shown to be important for angiogenic sprout initiation and maintenance [Kniazeva and Putnam in Am J Physiol 297:C179-C187, 2009] as well as endothelial cell shape control during invasion [Elliott et al. in Nat Cell Biol 17:137-147, 2015]. The exact nature of cytoskeleton-mediated forces for sprout initiation and progression, however, remains poorly understood. Questions on the importance of tip cell pulling versus stalk cell pushing are to a large extent unanswered, which among others has to do with the difficulty of quantifying and resolving those forces in time and space. We developed methods based on time-lapse confocal microscopy and image processing-further termed 4D displacement microscopy-to acquire detailed, spatially and temporally resolved extracellular matrix (ECM) deformations, indicative of cell-ECM mechanical interactions around invading sprouts. We demonstrate that matrix deformations dependent on actin-mediated force generation are spatio-temporally correlated with sprout morphological dynamics. Furthermore, sprout tips were found to exert radially pulling forces on the extracellular matrix, which were quantified by means of a computational model of collagen ECM mechanics. Protrusions from extending sprouts mostly increase their pulling forces, while retracting protrusions mainly reduce their pulling forces. Displacement microscopy analysis further unveiled a characteristic dipole-like deformation pattern along the sprout direction that was consistent among seemingly very different sprout shapes-with oppositely oriented displacements at sprout tip versus sprout base and a transition zone of negligible displacements in between. These results demonstrate that sprout-ECM interactions are dominated by pulling forces and underline the key role of tip cell pulling for sprouting angiogenesis.


Asunto(s)
Simulación por Computador , Citoesqueleto/metabolismo , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Modelos Cardiovasculares , Neovascularización Fisiológica , Humanos
9.
J Acoust Soc Am ; 145(2): 1048, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30823826

RESUMEN

Bone healing process is a complicated phenomenon regulated by biochemical and mechanical signals. Experimental studies have shown that ultrasound (US) accelerates bone ossification and has a multiple influence on cell differentiation and angiogenesis. In a recent work of the authors, a bioregulatory model for providing bone-healing predictions was addressed, taking into account for the first time the salutary effect of US on the involved angiogenesis. In the present work, a mechanobioregulatory model of bone solidification under the US presence incorporating also the mechanical environment on the regeneration process, which is known to affect cellular processes, is presented. An iterative procedure is adopted, where the finite element method is employed to compute the mechanical stimuli at the linear elastic phases of the poroelastic callus region and a coupled system of partial differential equations to simulate the enhancement by the US cell angiogenesis process and thus the oxygen concentration in the fractured area. Numerical simulations with and without the presence of US that illustrate the influence of progenitor cells' origin in the healing pattern and the healing rate and simultaneously demonstrate the salutary effect of US on bone repair are presented and discussed.


Asunto(s)
Fenómenos Biomecánicos/efectos de la radiación , Huesos , Curación de Fractura/efectos de la radiación , Modelos Biológicos , Ondas Ultrasónicas , Animales , Huesos/citología , Huesos/efectos de la radiación , Simulación por Computador , Curación de Fractura/fisiología , Fracturas Óseas/fisiopatología , Osteogénesis/efectos de la radiación
10.
J Nanobiotechnology ; 16(1): 82, 2018 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-30368242

RESUMEN

BACKGROUND: The continuously growing human exposure to combustion-derived particles (CDPs) drives in depth investigation of the involved complex toxicological mechanisms of those particles. The current study evaluated the hypothesis that CDPs could affect cell-induced remodeling of the extracellular matrix due to their underlying toxicological mechanisms. The effects of two ultrafine and one fine form of CDPs on human lung fibroblasts (MRC-5 cell line) were investigated, both in 2D cell culture and in 3D collagen type I hydrogels. A multi-parametric analysis was employed. RESULTS: In vitro dynamic 3D analysis of collagen matrices showed that matrix displacement fields induced by human lung fibroblasts are disturbed when exposed to carbonaceous particles, resulting in inhibition of matrix remodeling. In depth analysis using general toxicological assays revealed that a plausible explanation comprises a cascade of numerous detrimental effects evoked by the carbon particles, including oxidative stress, mitochondrial damage and energy storage depletion. Also, ultrafine particles revealed stronger toxicological and inhibitory effects compared to their larger counterparts. The inhibitory effects can be almost fully restored when treating the impaired cells with antioxidants like vitamin C. CONCLUSIONS: The unraveled in vitro pathway, by which ultrafine particles alter the fibroblasts' vital role of matrix remodeling, extends our knowledge about the contribution of these biologically active particles in impaired lung tissue repair mechanisms, and development and exacerbation of chronic lung diseases. The new insights may even pave the way to precautionary actions. The results provide justification for toxicological assessments to include mechanism-linked assays besides the traditional in vitro toxicological screening assays.


Asunto(s)
Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Pulmón/citología , Material Particulado/toxicidad , Adenosina Trifosfato/metabolismo , Antioxidantes/metabolismo , Colágeno Tipo I/metabolismo , Matriz Extracelular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo
11.
BMC Bioinformatics ; 18(1): 365, 2017 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-28797233

RESUMEN

BACKGROUND: Traction Force Microscopy (TFM) is a widespread technique to estimate the tractions that cells exert on the surrounding substrate. To recover the tractions, it is necessary to solve an inverse problem, which is ill-posed and needs regularization to make the solution stable. The typical regularization scheme is given by the minimization of a cost functional, which is divided in two terms: the error present in the data or data fidelity term; and the regularization or penalty term. The classical approach is to use zero-order Tikhonov or L2-regularization, which uses the L2-norm for both terms in the cost function. Recently, some studies have demonstrated an improved performance using L1-regularization (L1-norm in the penalty term) related to an increase in the spatial resolution and sensitivity of the recovered traction field. In this manuscript, we present a comparison between the previous two regularization schemes (relying in the L2-norm for the data fidelity term) and the full L1-regularization (using the L1-norm for both terms in the cost function) for synthetic and real data. RESULTS: Our results reveal that L1-regularizations give an improved spatial resolution (more important for full L1-regularization) and a reduction in the background noise with respect to the classical zero-order Tikhonov regularization. In addition, we present an approximation, which makes feasible the recovery of cellular tractions over whole cells on typical full-size microscope images when working in the spatial domain. CONCLUSIONS: The proposed full L1-regularization improves the sensitivity to recover small stress footprints. Moreover, the proposed method has been validated to work on full-field microscopy images of real cells, what certainly demonstrates it is a promising tool for biological applications.


Asunto(s)
Microscopía Fluorescente , Algoritmos , Animales , Fenómenos Biomecánicos , Células CHO , Cricetinae , Cricetulus , Hidrogeles
12.
J Theor Biol ; 365: 247-64, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25452136

RESUMEN

A timely restoration of the ruptured blood vessel network in order to deliver oxygen and nutrients to the fracture zone is crucial for successful bone healing. Indeed, oxygen plays a key role in the aerobic metabolism of cells, in the activity of a myriad of enzymes as well as in the regulation of several (angiogenic) genes. In this paper, a previously developed model of bone fracture healing is further improved with a detailed description of the influence of oxygen on various cellular processes that occur during bone fracture healing. Oxygen ranges of the cell-specific oxygen-dependent processes were established based on the state-of-the art experimental knowledge through a rigorous literature study. The newly developed oxygen model is compared with previously published experimental and in silico results. An extensive sensitivity analysis was also performed on the newly introduced oxygen thresholds, indicating the robustness of the oxygen model. Finally, the oxygen model was applied to the challenging clinical case of a critical sized defect (3mm) where it predicted the formation of a fracture non-union. Further model analyses showed that the harsh hypoxic conditions in the central region of the callus resulted in cell death and disrupted bone healing thereby indicating the importance of a timely vascularization for the successful healing of a large bone defect. In conclusion, this work demonstrates that the oxygen model is a powerful tool to further unravel the complex spatiotemporal interplay of oxygen delivery, diffusion and consumption with the several healing steps, each occurring at distinct, optimal oxygen tensions during the bone repair process.


Asunto(s)
Curación de Fractura/efectos de los fármacos , Fracturas Óseas/patología , Modelos Biológicos , Oxígeno/farmacología , Callo Óseo/efectos de los fármacos , Callo Óseo/patología , Simulación por Computador , Matriz Extracelular/metabolismo , Humanos , Factores de Tiempo
13.
PLoS Comput Biol ; 10(11): e1003888, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25375821

RESUMEN

Although bone has a unique restorative capacity, i.e., it has the potential to heal scarlessly, the conditions for spontaneous bone healing are not always present, leading to a delayed union or a non-union. In this work, we use an integrative in vivo-in silico approach to investigate the occurrence of non-unions, as well as to design possible treatment strategies thereof. The gap size of the domain geometry of a previously published mathematical model was enlarged in order to study the complex interplay of blood vessel formation, oxygen supply, growth factors and cell proliferation on the final healing outcome in large bone defects. The multiscale oxygen model was not only able to capture the essential aspects of in vivo non-unions, it also assisted in understanding the underlying mechanisms of action, i.e., the delayed vascularization of the central callus region resulted in harsh hypoxic conditions, cell death and finally disrupted bone healing. Inspired by the importance of a timely vascularization, as well as by the limited biological potential of the fracture hematoma, the influence of the host environment on the bone healing process in critical size defects was explored further. Moreover, dependent on the host environment, several treatment strategies were designed and tested for effectiveness. A qualitative correspondence between the predicted outcomes of certain treatment strategies and experimental observations was obtained, clearly illustrating the model's potential. In conclusion, the results of this study demonstrate that due to the complex non-linear dynamics of blood vessel formation, oxygen supply, growth factor production and cell proliferation and the interactions thereof with the host environment, an integrative in silico-in vivo approach is a crucial tool to further unravel the occurrence and treatments of challenging critical sized bone defects.


Asunto(s)
Curación de Fractura/fisiología , Fracturas Óseas/fisiopatología , Modelos Biológicos , Animales , Cartílago/metabolismo , Células Cultivadas , Simulación por Computador , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno/metabolismo , Periostio/citología , Ingeniería de Tejidos , Andamios del Tejido
14.
PLoS Comput Biol ; 9(10): e1003267, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24146605

RESUMEN

Adhesion governs to a large extent the mechanical interaction between a cell and its microenvironment. As initial cell spreading is purely adhesion driven, understanding this phenomenon leads to profound insight in both cell adhesion and cell-substrate interaction. It has been found that across a wide variety of cell types, initial spreading behavior universally follows the same power laws. The simplest cell type providing this scaling of the radius of the spreading area with time are modified red blood cells (RBCs), whose elastic responses are well characterized. Using a mechanistic description of the contact interaction between a cell and its substrate in combination with a deformable RBC model, we are now able to investigate in detail the mechanisms behind this universal power law. The presented model suggests that the initial slope of the spreading curve with time results from a purely geometrical effect facilitated mainly by dissipation upon contact. Later on, the spreading rate decreases due to increasing tension and dissipation in the cell's cortex as the cell spreads more and more. To reproduce this observed initial spreading, no irreversible deformations are required. Since the model created in this effort is extensible to more complex cell types and can cope with arbitrarily shaped, smooth mechanical microenvironments of the cells, it can be useful for a wide range of investigations where forces at the cell boundary play a decisive role.


Asunto(s)
Fenómenos Biomecánicos/fisiología , Fenómenos Fisiológicos Celulares/fisiología , Eritrocitos/citología , Eritrocitos/fisiología , Modelos Biológicos , Forma de la Célula/fisiología , Biología Computacional , Elasticidad/fisiología
15.
Polymers (Basel) ; 16(9)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38732733

RESUMEN

The thermo-responsive behavior of Poly(N-isopropylacrylamide) makes it an ideal candidate to easily embed cells and allows the polymer mixture to be injected. However, P(NiPAAm) hydrogels possess minor mechanical properties. To increase the mechanical properties, a covalent bond is introduced into the P(NIPAAm) network through a biocompatible thiol-ene click-reaction by mixing two polymer solutions. Co-polymers with variable thiol or acrylate groups to thermo-responsive co-monomer ratios, ranging from 1% to 10%, were synthesized. Precise control of the crosslink density allowed customization of the hydrogel's mechanical properties to match different tissue stiffness levels. Increasing the temperature of the hydrogel above its transition temperature of 31 °C induced the formation of additional physical interactions. These additional interactions both further increased the stiffness of the material and impacted its relaxation behavior. The developed optimized hydrogels reach stiffnesses more than ten times higher compared to the state of the art using similar polymers. Furthermore, when adding cells to the precursor polymer solutions, homogeneous thermo-responsive hydrogels with good cell viability were created upon mixing. In future work, the influence of the mechanical micro-environment on the cell's behavior can be studied in vitro in a continuous manner by changing the incubation temperature.

16.
PLoS Comput Biol ; 8(10): e1002724, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23071433

RESUMEN

The healing of a fracture depends largely on the development of a new blood vessel network (angiogenesis) in the callus. During angiogenesis tip cells lead the developing sprout in response to extracellular signals, amongst which vascular endothelial growth factor (VEGF) is critical. In order to ensure a correct development of the vasculature, the balance between stalk and tip cell phenotypes must be tightly controlled, which is primarily achieved by the Dll4-Notch1 signaling pathway. This study presents a novel multiscale model of osteogenesis and sprouting angiogenesis, incorporating lateral inhibition of endothelial cells (further denoted MOSAIC model) through Dll4-Notch1 signaling, and applies it to fracture healing. The MOSAIC model correctly predicted the bone regeneration process and recapitulated many experimentally observed aspects of tip cell selection: the salt and pepper pattern seen for cell fates, an increased tip cell density due to the loss of Dll4 and an excessive number of tip cells in high VEGF environments. When VEGF concentration was even further increased, the MOSAIC model predicted the absence of a vascular network and fracture healing, thereby leading to a non-union, which is a direct consequence of the mutual inhibition of neighboring cells through Dll4-Notch1 signaling. This result was not retrieved for a more phenomenological model that only considers extracellular signals for tip cell migration, which illustrates the importance of implementing the actual signaling pathway rather than phenomenological rules. Finally, the MOSAIC model demonstrated the importance of a proper criterion for tip cell selection and the need for experimental data to further explore this. In conclusion, this study demonstrates that the MOSAIC model creates enhanced capabilities for investigating the influence of molecular mechanisms on angiogenesis and its relation to bone formation in a more mechanistic way and across different time and spatial scales.


Asunto(s)
Células Endoteliales/fisiología , Modelos Biológicos , Neovascularización Fisiológica/fisiología , Osteogénesis/fisiología , Animales , Callo Óseo/metabolismo , Biología Computacional , Simulación por Computador , Células Endoteliales/metabolismo , Curación de Fractura/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratas , Receptor Notch1/metabolismo , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
17.
PLoS Comput Biol ; 6(9): e1000915, 2010 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-20824125

RESUMEN

Recently developed atrophic non-union models are a good representation of the clinical situation in which many non-unions develop. Based on previous experimental studies with these atrophic non-union models, it was hypothesized that in order to obtain successful fracture healing, blood vessels, growth factors, and (proliferative) precursor cells all need to be present in the callus at the same time. This study uses a combined in vivo-in silico approach to investigate these different aspects (vasculature, growth factors, cell proliferation). The mathematical model, initially developed for the study of normal fracture healing, is able to capture essential aspects of the in vivo atrophic non-union model despite a number of deviations that are mainly due to simplifications in the in silico model. The mathematical model is subsequently used to test possible treatment strategies for atrophic non-unions (i.e. cell transplant at post-osteotomy, week 3). Preliminary in vivo experiments corroborate the numerical predictions. Finally, the mathematical model is applied to explain experimental observations and identify potentially crucial steps in the treatments and can thereby be used to optimize experimental and clinical studies in this area. This study demonstrates the potential of the combined in silico-in vivo approach and its clinical implications for the early treatment of patients with problematic fractures.


Asunto(s)
Regeneración Ósea/fisiología , Huesos/fisiología , Fracturas Óseas/terapia , Modelos Biológicos , Cicatrización de Heridas/fisiología , Animales , Células de la Médula Ósea/fisiología , Movimiento Celular , Proliferación Celular , Trasplante de Células , Condrogénesis/fisiología , Simulación por Computador , Modelos Animales de Enfermedad , Fracturas Óseas/fisiopatología , Histocitoquímica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neovascularización Fisiológica/fisiología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Endogámicas WKY , Estadísticas no Paramétricas
18.
Carbohydr Polym ; 252: 117183, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33183630

RESUMEN

The elucidation of cell-surface interactions and the development of model platforms to help uncover their underlying mechanisms remains vital to the design of effective biomaterials. To this end, dextran palmitates with varying degrees of substitution were synthesised with a multipurpose functionality: an ability to modulate surface energy through surface chemistry, and an ideal thermal behaviour for patterning. Herein, dextran palmitate films are produced by spin coating, and patterned by thermal nanoimprint lithography with nano-to-microscale topographies. These films of moderately hydrophobic polysaccharide esters with low nanoscale roughness performed as well as fibronectin coatings in the culture of bovine aortic endothelial cells. Upon patterning, they display distinct regions of roughness, restricting cell adhesion to the smoothest surfaces, while guiding multicellular arrangements in the patterned topographies. The development of biomaterial interfaces through topochemical fabrication such as this could prove useful in understanding protein and cell-surface interactions.


Asunto(s)
Materiales Biocompatibles/química , Adhesión Celular , Dextranos/química , Células Endoteliales/citología , Ésteres/química , Andamios del Tejido , Animales , Bovinos , Técnicas de Cultivo de Célula , Propiedades de Superficie
19.
Sci Rep ; 11(1): 5687, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33707502

RESUMEN

Tailored hydrogels mimicking the native extracellular environment could help overcome the high variability in outcomes within regenerative endodontics. This study aimed to evaluate the effect of the chemokine-binding and antimicrobial polymer, chlorite-oxidized oxyamylose (COAM), on the microstructural properties of fibrin and self-assembling peptide (SAP) hydrogels. A further goal was to assess the influence of the microstructural differences between the hydrogels on the in vitro behavior of human dental pulp stem cells (hDPSCs). Structural and mechanical characterization of the hydrogels with and without COAM was performed by atomic force microscopy and scanning electron microscopy to characterize their microstructure (roughness and fiber length, diameter, straightness, and alignment) and by nanoindentation to measure their stiffness (elastic modulus). Then, hDPSCs were encapsulated in hydrogels with and without COAM. Cell viability and circularity were determined using confocal microscopy, and proliferation was determined using DNA quantification. Inclusion of COAM did not alter the microstructure of the fibrin hydrogels at the fiber level while affecting the SAP hydrogel microstructure (homogeneity), leading to fiber aggregation. The stiffness of the SAP hydrogels was sevenfold higher than the fibrin hydrogels. The viability and attachment of hDPSCs were significantly higher in fibrin hydrogels than in SAP hydrogels. The DNA content was significantly affected by the hydrogel type and the presence of COAM. The microstructural stability after COAM inclusion and the favorable hDPSCs' response observed in fibrin hydrogels suggest this system as a promising carrier for COAM and application in endodontic regeneration.


Asunto(s)
Amilosa/análogos & derivados , Cloruros/farmacología , Pulpa Dental/citología , Fibrina/química , Hidrogeles/química , Péptidos/química , Células Madre/citología , Adolescente , Amilosa/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , ADN/análisis , Femenino , Fibrina/ultraestructura , Humanos , Masculino , Microscopía de Fuerza Atómica , Oxidación-Reducción/efectos de los fármacos , Células Madre/efectos de los fármacos , Adulto Joven
20.
Nat Commun ; 12(1): 3192, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-34045434

RESUMEN

Tissues achieve their complex spatial organization through an interplay between gene regulatory networks, cell-cell communication, and physical interactions mediated by mechanical forces. Current strategies to generate in-vitro tissues have largely failed to implement such active, dynamically coordinated mechanical manipulations, relying instead on extracellular matrices which respond to, rather than impose mechanical forces. Here, we develop devices that enable the actuation of organoids. We show that active mechanical forces increase growth and lead to enhanced patterning in an organoid model of the neural tube derived from single human pluripotent stem cells (hPSC). Using a combination of single-cell transcriptomics and immunohistochemistry, we demonstrate that organoid mechanoregulation due to actuation operates in a temporally restricted competence window, and that organoid response to stretch is mediated extracellularly by matrix stiffness and intracellularly by cytoskeleton contractility and planar cell polarity. Exerting active mechanical forces on organoids using the approaches developed here is widely applicable and should enable the generation of more reproducible, programmable organoid shape, identity and patterns, opening avenues for the use of these tools in regenerative medicine and disease modelling applications.


Asunto(s)
Tubo Neural/citología , Organoides/fisiología , Ingeniería de Tejidos/métodos , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Línea Celular , Matriz Extracelular/fisiología , Humanos , Hidrogeles/química , Mecanotransducción Celular/fisiología , Células Madre Pluripotentes , Polietilenglicoles/química , RNA-Seq , Medicina Regenerativa/métodos , Análisis de la Célula Individual , Ingeniería de Tejidos/instrumentación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA