Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 307
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 187(2): 481-494.e24, 2024 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-38194965

RESUMO

Cellular form and function emerge from complex mechanochemical systems within the cytoplasm. Currently, no systematic strategy exists to infer large-scale physical properties of a cell from its molecular components. This is an obstacle to understanding processes such as cell adhesion and migration. Here, we develop a data-driven modeling pipeline to learn the mechanical behavior of adherent cells. We first train neural networks to predict cellular forces from images of cytoskeletal proteins. Strikingly, experimental images of a single focal adhesion (FA) protein, such as zyxin, are sufficient to predict forces and can generalize to unseen biological regimes. Using this observation, we develop two approaches-one constrained by physics and the other agnostic-to construct data-driven continuum models of cellular forces. Both reveal how cellular forces are encoded by two distinct length scales. Beyond adherent cell mechanics, our work serves as a case study for integrating neural networks into predictive models for cell biology.


Assuntos
Proteínas do Citoesqueleto , Aprendizado de Máquina , Adesão Celular , Citoplasma/metabolismo , Proteínas do Citoesqueleto/metabolismo , Adesões Focais/metabolismo , Modelos Biológicos
2.
Annu Rev Biomed Eng ; 26(1): 93-118, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38316064

RESUMO

Cell traction force plays a critical role in directing cellular functions, such as proliferation, migration, and differentiation. Current understanding of cell traction force is largely derived from 2D measurements where cells are plated on 2D substrates. However, 2D measurements do not recapitulate a vital aspect of living systems; that is, cells actively remodel their surrounding extracellular matrix (ECM), and the remodeled ECM, in return, can have a profound impact on cell phenotype and traction force generation. This reciprocal adaptivity of living systems is encoded in the material properties of biological gels. In this review, we summarize recent progress in measuring cell traction force for cells embedded within 3D biological gels, with an emphasis on cell-ECM cross talk. We also provide perspectives on tools and techniques that could be adapted to measure cell traction force in complex biochemical and biophysical environments.


Assuntos
Matriz Extracelular , Esferoides Celulares , Humanos , Matriz Extracelular/metabolismo , Esferoides Celulares/citologia , Animais , Movimento Celular , Géis/química , Adesão Celular , Microscopia de Força Atômica/métodos , Análise de Célula Única/métodos , Hidrogéis/química
3.
Proc Natl Acad Sci U S A ; 119(11): e2106098119, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35259013

RESUMO

SignificanceThe pseudokinase integrin-linked kinase (ILK) is a central component of focal adhesions, cytoplasmic multiprotein complexes that integrate and transduce biochemical and mechanical signals from the extracellular environment into the cell and vice versa. However, the precise molecular functions, particularly the mechanosensory properties of ILK and the significance of retained adenosine triphosphate (ATP) binding, are still unclear. Combining molecular-dynamics simulations with cell biology, we establish a role for ATP binding to pseudokinases. We find that ATP promotes the structural stability of ILK, allosterically influences the interaction between ILK and its binding partner parvin at adhesions, and enhances the mechanoresistance of this complex. On the cellular level, ATP binding facilitates efficient traction force buildup, focal adhesion stabilization, and efficient cell migration.


Assuntos
Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Actomiosina/química , Actomiosina/metabolismo , Regulação Alostérica , Sítios de Ligação , Adesão Celular , Movimento Celular , Estabilidade Enzimática , Adesões Focais , Mecanotransdução Celular , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/metabolismo , Modelos Moleculares , Conformação Molecular , Mutação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Serina-Treonina Quinases/genética , Relação Estrutura-Atividade , Especificidade por Substrato
4.
Proc Natl Acad Sci U S A ; 119(10): e2115217119, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35235449

RESUMO

The cytoskeleton of eukaryotic cells is primarily composed of networks of filamentous proteins, F-actin, microtubules, and intermediate filaments. Interactions among the cytoskeletal components are important in determining cell structure and in regulating cell functions. For example, F-actin and microtubules work together to control cell shape and polarity, while the subcellular organization and transport of vimentin intermediate filament (VIF) networks depend on their interactions with microtubules. However, it is generally thought that F-actin and VIFs form two coexisting but separate networks that are independent due to observed differences in their spatial distribution and functions. In this paper, we present a closer investigation of both the structural and functional interplay between the F-actin and VIF cytoskeletal networks. We characterize the structure of VIFs and F-actin networks within the cell cortex using structured illumination microscopy and cryo-electron tomography. We find that VIFs and F-actin form an interpenetrating network (IPN) with interactions at multiple length scales, and VIFs are integral components of F-actin stress fibers. From measurements of recovery of cell contractility after transient stretching, we find that the IPN structure results in enhanced contractile forces and contributes to cell resilience. Studies of reconstituted networks and dynamic measurements in cells suggest direct and specific associations between VIFs and F-actin. From these results, we conclude that VIFs and F-actin work synergistically, both in their structure and in their function. These results profoundly alter our understanding of the contributions of the components of the cytoskeleton, particularly the interactions between intermediate filaments and F-actin.


Assuntos
Citoplasma/metabolismo , Filamentos Intermediários/metabolismo , Vimentina/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/química , Actinas/metabolismo , Animais , Biopolímeros/metabolismo , Células Cultivadas , Tomografia com Microscopia Eletrônica/métodos , Filamentos Intermediários/química , Camundongos , Vimentina/química
5.
J Cell Sci ; 135(10)2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35621127

RESUMO

Podosomes are mechanosensitive protrusive actin structures that are prominent in myeloid cells, and they have been linked to vascular extravasation. Recent studies have suggested that podosomes are hierarchically organized and have coordinated dynamics on the cell scale, which implies that the local force generation by single podosomes can be different from their global combined action. Complementary to previous studies focusing on individual podosomes, here we investigated the cell-wide force generation of podosome-bearing ER-Hoxb8 monocytes. We found that the occurrence of focal tractions accompanied by a cell-wide substrate indentation cannot be explained by summing the forces of single podosomes. Instead, our findings suggest that superimposed contraction on the cell scale gives rise to a buckling mechanism that can explain the measured cell-scale indentation. Specifically, the actomyosin network contraction causes peripheral in-plane substrate tractions, while the accumulated internal stress results in out-of-plane deformation in the central cell region via a buckling instability, producing the cell-scale indentation. Hence, we propose that contraction of the actomyosin network, which connects the podosomes, leads to a substrate indentation that acts in addition to the protrusion forces of individual podosomes. This article has an associated First Person interview with the first author of the paper.


Assuntos
Podossomos , Actomiosina , Extensões da Superfície Celular , Humanos , Monócitos , Tração
6.
Am J Physiol Heart Circ Physiol ; 326(5): H1180-H1192, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38457352

RESUMO

Endothelial cells (ECs) within the vascular system encounter fluid shear stress (FSS). High, laminar FSS promotes vasodilation and anti-inflammatory responses, whereas low or disturbed FSS induces dysfunction and inflammation. However, the adaptation of endothelial cells (ECs) to dynamically changing FSS patterns remains underexplored. Here, by combining traction force microscopy with a custom flow chamber, we examined human umbilical vein endothelial cells adapting their traction during transitions from short-term low shear to long-term high shear stress. We discovered that the initial low FSS elevates the traction by only half of the amount in response to direct high FSS even after flow changes to high FSS. However, in the long term under high FSS, the flow started with low FSS triggers a substantial second rise in traction for over 10 h. In contrast, the flow started directly with high FSS results in a quick traction surge followed by a huge reduction below the baseline traction in <30 min. Importantly, we find that the orientation of traction vectors is steered by initial shear exposure. Using Granger causality analysis, we show that the traction that aligns in the flow direction under direct high FSS functionally causes cell alignment toward the flow direction. However, EC traction that orients perpendicular to the flow that starts with temporary low FSS functionally causes cell orientation perpendicular to the flow. Taken together, our findings elucidate the significant influence of initial short-term low FSS on lasting changes in endothelial traction that induces EC alignment.NEW & NOTEWORTHY In our study, we uncover that preconditioning with low shear stress yields enduring impacts on endothelial cell traction and orientation, persisting even after transitioning to high-shear conditions. Using Granger causality analysis, we demonstrate a functional link between the direction of cell traction and subsequent cellular alignment across varying shear environments.


Assuntos
Células Endoteliais da Veia Umbilical Humana , Estresse Mecânico , Humanos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Cultivadas , Mecanotransdução Celular , Fatores de Tempo , Adesão Celular
7.
Small ; 20(28): e2400644, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38326079

RESUMO

Tissue development is mediated by a combination of mechanical and biological signals. Currently, there are many reports on biological signals regulating repair. However, insufficient attention is paid to the process of mechanical regulation, especially the active mechanical regulation in vivo, which has not been realized. Herein, a novel dynamically regulated repair system for both in vitro and in vivo applications is developed, which utilizes magnetic nanoparticles as non-contact actuators to activate hydrogels. The magnetic hydrogel can be periodically activated and deformed to different amplitudes by a dynamic magnetic system. An in vitro skin model is used to explore the impact of different dynamic stimuli on cellular mechano-transduction signal activation and cell differentiation. Specifically, the effect of mechanical stimulation on the phenotypic transition of fibroblasts to myofibroblasts is investigated. Furthermore, in vivo results verify that dynamic massage can simulate and enhance the traction effect in skin defects, thereby accelerating the wound healing process by promoting re-epithelialization and mediating dermal contraction.


Assuntos
Bandagens , Massagem , Cicatrização , Animais , Massagem/métodos , Fibroblastos , Humanos , Hidrogéis/química , Diferenciação Celular , Pele , Camundongos , Miofibroblastos/citologia
8.
Small ; 20(4): e2302404, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37735983

RESUMO

Strain gradients widely exist in development and physiological activities. The directional movement of cells is essential for proper cell localization, and directional cell migration in responses to gradients of chemicals, rigidity, density, and topography of extracellular matrices have been well-established. However; it is unclear whether strain gradients imposed on cells are sufficient to drive directional cell migration. In this work, a programmable uniaxial cell stretch device is developed that creates controllable strain gradients without changing substrate stiffness or ligand distributions. It is demonstrated that over 60% of the single rat embryonic fibroblasts migrate toward the lower strain side in static and the 0.1 Hz cyclic stretch conditions at ≈4% per mm strain gradients. It is confirmed that such responses are distinct from durotaxis or haptotaxis. Focal adhesion analysis confirms higher rates of contact area and protrusion formation on the lower strain side of the cell. A 2D extended motor-clutch model is developed to demonstrate that the strain-introduced traction force determines integrin fibronectin pairs' catch-release dynamics, which drives such directional migration. Together, these results establish strain gradient as a novel cue to regulate directional cell migration and may provide new insights in development and tissue repairs.


Assuntos
Quimiotaxia , Matriz Extracelular , Ratos , Animais , Movimento Celular , Adesões Focais , Adesão Celular
9.
Biol Chem ; 405(1): 67-77, 2024 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-37674311

RESUMO

Microtubules are highly polar structures and are characterized by high anisotropy and stiffness. In neurons, they play a key role in the directional transport of vesicles and organelles. In the neuronal projections called axons, they form parallel bundles, mostly oriented with the plus-end towards the axonal termination. Their physico-chemical properties have recently attracted attention as a potential candidate in sensing, processing and transducing physical signals generated by mechanical forces. Here, we discuss the main evidence supporting the role of microtubules as a signal hub for axon growth in response to a traction force. Applying a tension to the axon appears to stabilize the microtubules, which, in turn, coordinate a modulation of axonal transport, local translation and their cross-talk. We speculate on the possible mechanisms modulating microtubule dynamics under tension, based on evidence collected in neuronal and non-neuronal cell types. However, the fundamental question of the causal relationship between these mechanisms is still elusive because the mechano-sensitive element in this chain has not yet been identified.


Assuntos
Axônios , Microtúbulos , Microtúbulos/metabolismo , Axônios/metabolismo , Neurônios/metabolismo
10.
Biotechnol Bioeng ; 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38978386

RESUMO

It is of great importance to study the detachment/attachment behaviors of cells (cancer cell, immune cell, and epithelial cell), as they are closely related with tumor metastasis, immunoreaction, and tissue development at variety scales. To characterize the detachment/attachment during the interaction between cells and substrate, some researchers proposed using cell traction force (CTF) as the indicator. To date, various strategies have been developed to measure the CTF. However, these methods only realize the measurements of cell passive forces on flat cases. To quantify the active CTF on nonflat surfaces, which can better mimic the in vivo case, we employed elastic hydrogel microspheres as a force sensor. The microspheres were fabricated by microfluidic chips with controllable size and mechanical properties to mimic substrate. Cells were cultured on microsphere and the CTF led to the deformation of microsphere. By detecting the morphology information, the CTF exerted by attached cells can be calculated by the in-house numerical code. Using these microspheres, the CTF of various cells (including tumor cell, immunological cell, and epithelium cell) were successfully obtained on nonflat surfaces with different curvature radii. The proposed method provides a versatile platform to measure the CTF with high precision and to understand the detachment/attachment behaviors during physiology processes.

11.
Biotechnol Bioeng ; 121(6): 1820-1830, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38407981

RESUMO

Cell traction force (CTF) is a kind of active force that is a cell senses external environment and actively applies to the contact matrix which is currently a representative stress in cell-extracellular matrix (ECM) interaction. Studying the distribution and variation of CTF during cell-ECM interaction help to explain the impact of physical factors on cell behaviors from the perspective of mechanobiology. However, most of the strategies of characterizing CTF are still limited by the measurement needs in three-dimensional (3D), quantitative characteristics and in vivo condition. Microsphere stress sensor (MSS) as a new type of technology is capable of realizing the quantitative characterization of CTF in 3D and in vivo. Herein, we employed microfluidic platform to design and fabricate MSS which possesses adjustable fluorescent performances, physical properties, and size ranges for better applicable to different cells (3T3, A549). Focusing on the common tumor cells behaviors (adhesion, spreading, and migration) in the process of metastasis, we chose SH-SY5Y as the representative research object in this work. We calculated CTF with the profile and distribution to demonstrate that the normal and shear stress can determined different cell behaviors. Additionally, CTF can also regulate cell adhesion, spreading, and migration in different cell states. Based on this method, the quantitative characterization of CFT of health and disease cells can be achieved, which further help to study and explore the potential mechanism of cell-ECM interaction.


Assuntos
Matriz Extracelular , Hidrogéis , Microesferas , Humanos , Matriz Extracelular/química , Animais , Hidrogéis/química , Camundongos , Adesão Celular , Linhagem Celular Tumoral , Estresse Mecânico , Movimento Celular
12.
Surg Endosc ; 2024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-39046496

RESUMO

PURPOSE: In vesicourethral anastomosis (VUA), which is part of robot-assisted radical prostatectomy, surgeons must proceed carefully to avoid urethral damage. We developed and evaluated a VUA bench-top model that can measure the traction force on the urethra during robotic surgery. MATERIALS AND METHODS: The VUA model included the urethra, bladder, pelvic bones, and a small force sensor that was capable of measuring the traction force on the urethra. Eight skilled and eight novice urologists performed a VUA task in robotic surgery. The skilled surgeons assessed the model's realism and usefulness as a training tool using a 5-point Likert scale. The evaluation items [task time, maximum force, force volume, and length of time that specific excessive forces were applied to the urethra (2, 3, 4, and ≥ 5 N)] were compared between the skilled and novice surgeons using the Mann-Whitney U test. Measurements were conducted in four directions with respect to the maximum force on the urethra: 11-1, 2-4, 5-7, and 8-10 o'clock. RESULTS: The quality of the model was scored 3.7 to 4.9 points for all 16 items in 4 domains except for "Usefulness compared with animal models." There were differences in the task time and almost all force parameters between the skilled and novice surgeons. CONCLUSION: We developed a relatively high-quality VUA bench-top model that measures traction force on the urethra, and we have revealed differences in the forces of action on the urethra in two groups of surgeons with different skill levels.

13.
Nano Lett ; 23(7): 2467-2475, 2023 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-36975035

RESUMO

Mechanical signals establish two-way communication between mammalian cells and their environment. Cells contacting a surface exert forces via contractility and transmit them at the areas of focal adhesions. External stimuli, such as compressive and pulling forces, typically affect the adhesion-free cell surface. Here, we demonstrate the collaborative employment of Fluidic Force Microscopy and confocal Traction Force Microscopy supported by the Cellogram solver to enable a powerful integrated force probing approach, where controlled vertical forces are applied to the free surface of individual cells, while the concomitant deformations are used to map their transmission to the substrate. Force transmission across human cells is measured with unprecedented temporal and spatial resolution, enabling the investigation of the cellular mechanisms involved in the adaptation, or maladaptation, to external mechanical stimuli. Altogether, the system enables facile and precise force interrogation of individual cells, with the capacity to perform population-based analysis.


Assuntos
Adesão Celular , Matriz Extracelular , Adesões Focais , Mecanotransdução Celular , Animais , Humanos , Adesão Celular/fisiologia , Membrana Celular/fisiologia , Adesões Focais/metabolismo , Adesões Focais/fisiologia , Mamíferos/anatomia & histologia , Mamíferos/fisiologia , Fenômenos Mecânicos , Mecanotransdução Celular/fisiologia , Microscopia de Força Atômica/métodos , Matriz Extracelular/fisiologia
14.
Int J Mol Sci ; 25(11)2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38892386

RESUMO

The mechanical forces exerted by cells on their surrounding microenvironment are known as cellular traction forces. These forces play crucial roles in various biological processes, such as tissue development, wound healing and cell functions. However, it is hard for traditional techniques to measure cellular traction forces accurately because their magnitude (from pN to nN) and the length scales over which they occur (from nm to µm) are extremely small. In order to fully understand mechanotransduction, highly sensitive tools for measuring cellular forces are needed. Current powerful techniques for measuring traction forces include traction force microscopy (TFM) and fluorescent molecular force sensors (FMFS). In this review, we elucidate the force imaging principles of TFM and FMFS. Then we highlight the application of FMFS in a variety of biological processes and offer our perspectives and insights into the potential applications of FMFS.


Assuntos
Mecanotransdução Celular , Humanos , Animais , Microscopia de Força Atômica/métodos , Técnicas Biossensoriais/métodos
15.
Int J Mol Sci ; 25(7)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38612536

RESUMO

The endometrial epithelium and underlying stroma undergo profound changes to support and limit embryo adhesion and invasion, which occur in the secretory phase of the menstrual cycle during the window of implantation. This coincides with a peak in progesterone and estradiol production. We hypothesized that the interplay between hormone-induced changes in the mechanical properties of the endometrial epithelium and stroma supports this process. To study it, we used hormone-responsive endometrial adenocarcinoma-derived Ishikawa cells growing on substrates of different stiffness. We showed that Ishikawa monolayers on soft substrates are more tightly clustered and uniform than on stiff substrates. Probing for mechanical alterations, we found accelerated stress-relaxation after apical nanoindentation in hormone-stimulated monolayers on stiff substrates. Traction force microscopy furthermore revealed an increased number of foci with high traction in the presence of estradiol and progesterone on soft substrates. The detection of single cells and small cell clusters positive for the intermediate filament protein vimentin and the progesterone receptor further underscored monolayer heterogeneity. Finally, adhesion assays with trophoblast-derived AC-1M-88 spheroids were used to examine the effects of substrate stiffness and steroid hormones on endometrial receptivity. We conclude that the extracellular matrix and hormones act together to determine mechanical properties and, ultimately, embryo implantation.


Assuntos
Matriz Extracelular , Progesterona , Feminino , Humanos , Epitélio , Ciclo Menstrual , Estradiol
16.
Am J Physiol Cell Physiol ; 325(1): C314-C323, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37335028

RESUMO

Extracellular matrix (ECM) composition is important in a host of pathophysiological processes such as angiogenesis, atherosclerosis, and diabetes, and during each of these processes ECM composition has been reported to change over time. However, the impact ECM composition has on the ability of endothelium to respond mechanically is currently unknown. Therefore, in this study, we seeded human umbilical vein endothelial cells (HUVECs) onto soft hydrogels coated with an ECM concentration of 0.1 mg/mL at the following collagen I (Col-I) and fibronectin (FN) ratios: 100% Col-I, 75% Col-I-25% FN, 50% Col-I-50% FN, 25% Col-I-75% FN, and 100% FN. We subsequently measured tractions, intercellular stresses, strain energy, cell morphology, and cell velocity. Our results revealed that tractions and strain energy are maximal at 50% Col-I-50% FN and minimal at 100% Col-I and 100% FN. Intercellular stress response was maximal on 50% Col-I-50% FN and minimal on 25% Col-I-75% FN. Cell area and cell circularity displayed a divergent relationship for different Col-I and FN ratios. We believe that these results will be of great importance to the cardiovascular field, biomedical field, and cell mechanics.NEW & NOTEWORTHY The endothelium constitutes the innermost layer of all blood vessels and plays an important role in vascular physiology and pathology. During certain vascular diseases, the extracellular matrix has been suggested to transition from a collagen-rich matrix to a fibronectin-rich matrix. In this study, we demonstrate the impact various collagen and fibronectin ratios have on endothelial biomechanical and morphological response.


Assuntos
Células Endoteliais , Fibronectinas , Humanos , Matriz Extracelular/fisiologia , Colágeno , Colágeno Tipo I , Endotélio , Células Cultivadas
17.
J Biol Chem ; 298(5): 101867, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35351517

RESUMO

Traction force microscopy (TFM) has emerged as a versatile technique for the measurement of single-cell-generated forces. TFM has gained wide use among mechanobiology laboratories, and several variants of the original methodology have been proposed. However, issues related to the experimental setup and, most importantly, data analysis of cell traction datasets may restrain the adoption of TFM by a wider community. In this review, we summarize the state of the art in TFM-related research, with a focus on the analytical methods underlying data analysis. We aim to provide the reader with a friendly compendium underlying the potential of TFM and emphasizing the methodological framework required for a thorough understanding of experimental data. We also compile a list of data analytics tools freely available to the scientific community for the furtherance of knowledge on this powerful technique.


Assuntos
Tração , Biofísica , Adesão Celular , Microscopia de Força Atômica/métodos
18.
Biochem Biophys Res Commun ; 642: 154-161, 2023 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-36580826

RESUMO

The physicochemical properties of biomaterials influence cell adhesion, shape, and polarization of macrophages. In this study, we aimed to evaluate the polarization of macrophages in terms of the regulation of cell adhesion and how synthetic mimics for heparin and poly(sodium-4-styrenesulfonate) can regulate macrophage polarization by modulating cell shape, focal adhesion, cell traction force, and intracellular tension. Our initial findings showed that macrophages cultured with heparin-mimicking polymer-based hydrogel matrix showed reduced expression of cell adhesion markers such as integrins, vinculin, RhoA, and ROCK1/2 and reduced cell shape, elongation, cell-matrix traction force, and intracellular tension. Furthermore, we observed a significant decrease in cell adhesion in cells cultured on the hydrogel, resulting in the promotion of M1 polarization. These findings offer insights into the important roles of cell-matrix interactions in macrophage polarization and offer a platform for heparin-mimicking polymer-based hydrogel matrices to induce M1 polarization by inducing cell adhesion without classical activators.


Assuntos
Hidrogéis , Polímeros , Adesão Celular , Heparina/farmacologia , Heparina/metabolismo , Macrófagos/metabolismo , Polímeros/farmacologia , Polímeros/metabolismo , Materiais Biomiméticos
19.
J Microsc ; 290(2): 69-96, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36870022

RESUMO

The advent of atomic force microscopy (AFM) provides an amazing instrument for characterising the structures and properties of living biological systems under aqueous conditions with unprecedented spatiotemporal resolution. In addition to its own unique capabilities for applications in life sciences, AFM is highly compatible and has been widely integrated with various complementary techniques to simultaneously sense the multidimensional (biological, chemical and physical) properties of biological systems, offering novel possibilities for comprehensively revealing the underlying mechanisms guiding life activities particularly in the studies of single cells. Herein, typical combinations of AFM and complementary techniques (including optical microscopy, ultrasound, infrared spectroscopy, Raman spectroscopy, fluidic force microscopy and traction force microscopy) and their applications in single-cell analysis are reviewed. The future perspectives are also provided.


Assuntos
Análise Espectral Raman , Microscopia de Força Atômica/métodos
20.
J Biomech Eng ; 145(12)2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37715307

RESUMO

Within the aortic valve (AV) leaflet exists a population of interstitial cells (AVICs) that maintain the constituent tissues by extracellular matrix (ECM) secretion, degradation, and remodeling. AVICs can transition from a quiescent, fibroblast-like phenotype to an activated, myofibroblast phenotype in response to growth or disease. AVIC dysfunction has been implicated in AV disease processes, yet our understanding of AVIC function remains quite limited. A major characteristic of the AVIC phenotype is its contractile state, driven by contractile forces generated by the underlying stress fibers (SF). However, direct assessment of the AVIC SF contractile state and structure within physiologically mimicking three-dimensional environments remains technically challenging, as the size of single SFs are below the resolution of light microscopy. Therefore, in the present study, we developed a three-dimensional (3D) computational approach of AVICs embedded in 3D hydrogels to estimate their SF local orientations and contractile forces. One challenge with this approach is that AVICs will remodel the hydrogel, so that the gel moduli will vary spatially. We thus utilized our previous approach (Khang et al. 2023, "Estimation of Aortic Valve Interstitial Cell-Induced 3D Remodeling of Poly (Ethylene Glycol) Hydrogel Environments Using an Inverse Finite Element Approach," Acta Biomater., 160, pp. 123-133) to define local hydrogel mechanical properties. The AVIC SF model incorporated known cytosol and nucleus mechanical behaviors, with the cell membrane assumed to be perfectly bonded to the surrounding hydrogel. The AVIC SFs were first modeled as locally unidirectional hyperelastic fibers with a contractile force component. An adjoint-based inverse modeling approach was developed to estimate local SF orientation and contractile force. Substantial heterogeneity in SF force and orientations were observed, with the greatest levels of SF alignment and contractile forces occurring in AVIC protrusions. The addition of a dispersed SF orientation to the modeling approach did not substantially alter these findings. To the best of our knowledge, we report the first fully 3D computational contractile cell models which can predict locally varying stress fiber orientation and contractile force levels.


Assuntos
Valva Aórtica , Fibras de Estresse , Fenômenos Mecânicos , Contração Muscular , Hidrogéis/metabolismo , Células Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA