Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.614
Filter
1.
J Biomed Opt ; 29(Suppl 2): S22708, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38872791

ABSTRACT

Significance: The ability to observe and monitor cell density and morphology has been imperative for assessing the health of a cell culture and for producing high quality, high yield cell cultures for decades. Microcarrier-based cultures, used for large-scale cellular expansion processes, are not compatible with traditional visualization-based methods, such as widefield microscopy, due to their thickness and material composition. Aim: Here, we assess the optical imaging compatibilities of commercial polystyrene microcarriers versus custom-fabricated gelatin methacryloyl (gelMA) microcarriers for non-destructive and non-invasive visualization of the entire microcarrier surface, direct cell enumeration, and sub-cellular visualization of mesenchymal stem/stromal cells. Approach: Mie scattering and wavefront error simulations of the polystyrene and gelMA microcarriers were performed to assess the potential for elastic scattering-based imaging of adherent cells. A Zeiss Z.1 light-sheet microscope was adapted to perform light-sheet tomography using label-free elastic scattering contrast from planar side illumination to achieve optical sectioning and permit non-invasive and non-destructive, in toto, three-dimensional, high-resolution visualization of cells cultured on microcarriers. Results: The polystyrene microcarrier prevents visualization of cells on the distal half of the microcarrier using either fluorescence or elastic scattering contrast, whereas the gelMA microcarrier allows for high fidelity visualization of cell morphology and quantification of cell density using light-sheet fluorescence microscopy and tomography. Conclusions: The combination of optical-quality gelMA microcarriers and label-free light-sheet tomography will facilitate enhanced control of bioreactor-microcarrier cell culture processes.


Subject(s)
Cell Adhesion , Hydrogels , Mesenchymal Stem Cells , Polystyrenes , Polystyrenes/chemistry , Mesenchymal Stem Cells/cytology , Hydrogels/chemistry , Cell Adhesion/physiology , Optical Imaging/methods , Optical Imaging/instrumentation , Humans , Gelatin/chemistry , Cell Culture Techniques/methods , Cell Culture Techniques/instrumentation , Cells, Cultured , Animals
2.
Sensors (Basel) ; 24(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38894171

ABSTRACT

Adherent cells perceive mechanical feedback from the underlying matrix and convert it into biochemical signals through a process known as mechanotransduction. The response to changes in the microenvironment relies on the cell's mechanical properties, including elasticity, which was recently identified as a biomarker for various diseases. Here, we propose the design, development, and characterization of a new system for the measurement of adherent cells' strain drop, a parameter correlated with cells' elasticity. To consider the interplay between adherent cells and the host extracellular matrix, cell stretching was combined with adhesion on substrates with different stiffnesses. The technique is based on the linear stretching of silicone chambers, high-speed image acquisition, and feedback for image centering. The system was characterized in terms of the strain homogeneity, impact of collagen coating, centering capability, and sensitivity. Subsequently, it was employed to measure the strain drop of two osteosarcoma cell lines, low-aggressive osteoblast-like SaOS-2 and high-aggressive 143B, cultured on two different substrates to recall the stiffness of the bone and lung extracellular matrices. Results demonstrated good substrate homogeneity, a negligible effect of the collagen coating, and an accurate image centering. Finally, the experimental results showed an average strain drop that was lower in the 143B cells in comparison with the SaOS-2 cells in all the tested conditions.


Subject(s)
Osteosarcoma , Osteosarcoma/pathology , Humans , Cell Line, Tumor , Extracellular Matrix/metabolism , Mechanotransduction, Cellular/physiology , Cell Adhesion/physiology , Elasticity , Stress, Mechanical , Bone Neoplasms/pathology , Collagen/chemistry , Collagen/metabolism , Osteoblasts/cytology , Osteoblasts/physiology
3.
PLoS Comput Biol ; 20(6): e1012112, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38861575

ABSTRACT

Cell sedimentation in 3D hydrogel cultures refers to the vertical migration of cells towards the bottom of the space. Understanding this poorly examined phenomenon may allow us to design better protocols to prevent it, as well as provide insights into the mechanobiology of cancer development. We conducted a multiscale experimental and mathematical examination of 3D cancer growth in triple negative breast cancer cells. Migration was examined in the presence and absence of Paclitaxel, in high and low adhesion environments and in the presence of fibroblasts. The observed behaviour was modeled by hypothesizing active migration due to self-generated chemotactic gradients. Our results did not reject this hypothesis, whereby migration was likely to be regulated by the MAPK and TGF-ß pathways. The mathematical model enabled us to describe the experimental data in absence (normalized error<40%) and presence of Paclitaxel (normalized error<10%), suggesting inhibition of random motion and advection in the latter case. Inhibition of sedimentation in low adhesion and co-culture experiments further supported the conclusion that cells actively migrated downwards due to the presence of signals produced by cells already attached to the adhesive glass surface.


Subject(s)
Cell Adhesion , Cell Movement , Paclitaxel , Humans , Cell Adhesion/physiology , Cell Movement/physiology , Paclitaxel/pharmacology , Cell Line, Tumor , Models, Biological , Cell Culture Techniques, Three Dimensional/methods , Triple Negative Breast Neoplasms/pathology , Computational Biology , Fibroblasts/physiology , Chemotaxis/physiology
4.
Cell Immunol ; 401-402: 104843, 2024.
Article in English | MEDLINE | ID: mdl-38905771

ABSTRACT

Monocyte migration is an important process in inflammation and atherogenesis. Identification of the key signalling pathways that regulate monocyte migration can provide prospective targets for prophylactic treatments in inflammatory diseases. Previous research showed that the focal adhesion kinase Pyk2, Src kinase and MAP kinases play an important role in MCP-1-induced monocyte migration. In this study, we demonstrate that MCP-1 induces iPLA2 activity, which is regulated by PKCß and affects downstream activation of Rac1 and Pyk2. Rac1 interacts directly with iPLA2 and Pyk2, and plays a crucial role in MCP-1-mediated monocyte migration by modulating downstream Pyk2 and p38 MAPK activation. Furthermore, Rac1 is necessary for cell spreading and F-actin polymerization during monocyte adhesion to fibronectin. Finally, we provide evidence that Rac1 controls the secretion of inflammatory mediator vimentin from MCP-1-stimulated monocytes. Altogether, this study demonstrates that the PKCß/iPLA2/Rac1/Pyk2/p38 MAPK signalling cascade is essential for MCP-1-induced monocyte adhesion and migration.


Subject(s)
Cell Adhesion , Cell Movement , Chemokine CCL2 , Focal Adhesion Kinase 2 , Monocytes , Signal Transduction , p38 Mitogen-Activated Protein Kinases , rac1 GTP-Binding Protein , Humans , Monocytes/metabolism , Monocytes/immunology , Chemokine CCL2/metabolism , Cell Adhesion/physiology , rac1 GTP-Binding Protein/metabolism , Focal Adhesion Kinase 2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C beta/metabolism , Actins/metabolism
5.
Phys Rev Lett ; 132(18): 188402, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38759206

ABSTRACT

Cell adhesion receptors are transmembrane proteins that bind cells to their environment. These proteins typically cluster into disk-shaped or linear structures. Here, we show that such clustering patterns spontaneously emerge when the receptor senses the membrane deformation gradient, for example, by reaching a lower-energy conformation when the membrane is tilted relative to the underlying binding substrate. Increasing the strength of the membrane gradient-sensing mechanism first yields isolated disk-shaped clusters and then long linear structures. Our theory is coherent with experimental estimates of the parameters, suggesting that a tilt-induced clustering mechanism is relevant in the context of cell adhesion.


Subject(s)
Cell Membrane , Cell Membrane/metabolism , Models, Biological , Cell Adhesion/physiology , Phase Separation , Platelet Glycoprotein GPIb-IX Complex
6.
J R Soc Interface ; 21(214): 20240105, 2024 May.
Article in English | MEDLINE | ID: mdl-38774959

ABSTRACT

During mesenchymal migration, F-actin protrusion at the leading edge and actomyosin contraction determine the retrograde flow of F-actin within the lamella. The coupling of this flow to integrin-based adhesions determines the force transmitted to the extracellular matrix and the net motion of the cell. In tissues, motion may also arise from convection, driven by gradients in tissue-scale surface tensions and pressures. However, how migration coordinates with convection to determine the net motion of cellular ensembles is unclear. To explore this, we study the spreading of cell aggregates on adhesive micropatterns on compliant substrates. During spreading, a cell monolayer expands from the aggregate towards the adhesive boundary. However, cells are unable to stabilize the protrusion beyond the adhesive boundary, resulting in retraction of the protrusion and detachment of cells from the matrix. Subsequently, the cells move upwards and rearwards, yielding a bulk convective flow towards the centre of the aggregate. The process is cyclic, yielding a steady-state balance between outward (protrusive) migration along the surface, and 'retrograde' (contractile) flows above the surface. Modelling the cell aggregates as confined active droplets, we demonstrate that the interplay between surface tension-driven flows within the aggregate, radially outward monolayer flow and conservation of mass leads to an internal circulation.


Subject(s)
Cell Adhesion , Cell Movement , Models, Biological , Cell Movement/physiology , Cell Adhesion/physiology , Cell Aggregation/physiology , Animals , Humans , Actins/metabolism
7.
Mol Immunol ; 171: 12-21, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38735126

ABSTRACT

Macrophages are critical in mediating immune and inflammatory responses, while monocyte-to-macrophage differentiation is one of the main macrophage resources that involves various matrix proteins. Matrix remodeling associated 7 (MXRA7) was recently discovered to affect a variety of physiological and pathological processes related to matrix biology. In the present study, we investigated the role of MXRA7 in monocyte-to-macrophage differentiation in vitro. We found that knockdown of MXRA7 inhibited the proliferation of THP-1 human monocytic cells. Knockdown of MXRA7 increased the adhesion ability of THP-1 cells through upregulation the expression of adhesion molecules VCAM-1 and ICAM1. Knockdown of MXRA7 alone could promoted the differentiation of THP-1 cells to macrophages. Furthermore, the MXRA7-knockdown THP-1 cells produced a more significant upregulation pattern with M1-type cytokines (TNF-α, IL-1ß and IL-6) than with those M2-type molecules (TGF-ß1 and IL-1RA) upon PMA stimulation, indicating that knockdown of MXRA7 facilitated THP-1 cells differentiation toward M1 macrophages. RNA sequencing analysis revealed the potential biological roles of MXRA7 in cell adhesion, macrophage and monocyte differentiation. Moreover, MXRA7 knockdown promoted the expression of NF-κB p52/p100, while PMA stimulation could increase the expression of NF-κB p52/p100 and activating MAPK signaling pathways in MXRA7 knockdown cells. In conclusion, MXRA7 affected the differentiation of THP-1 cells toward macrophages possibly through NF-κB signaling pathways.


Subject(s)
Cell Differentiation , Macrophages , Monocytes , Humans , Cell Adhesion/physiology , Cell Differentiation/immunology , Cell Differentiation/genetics , Cell Proliferation , Cytokines/metabolism , Gene Knockdown Techniques , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/genetics , Macrophages/metabolism , Macrophages/immunology , Monocytes/metabolism , NF-kappa B/metabolism , Signal Transduction , THP-1 Cells , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/genetics
8.
Mol Biol Cell ; 35(7): ar102, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38809584

ABSTRACT

Interferon Regulatory Factor 6 (IRF6) is a transcription factor essential for keratinocyte cell-cell adhesions. Previously, we found that recycling of E-cadherin was defective in the absence of IRF6, yet total E-cadherin levels were not altered, suggesting a previously unknown, nontranscriptional function for IRF6. IRF6 protein contains a DNA binding domain (DBD) and a protein binding domain (PBD). The transcriptional function of IRF6 depends on its DBD and PBD, however, whether the PBD is necessary for the interaction with cytoplasmic proteins has yet to be demonstrated. Here, we show that an intact PBD is required for recruitment of cell-cell adhesion proteins at the plasma membrane, including the recycling of E-cadherin. Colocalizations and coimmunoprecipitations reveal that IRF6 forms a complex in recycling endosomes with Rab11, Myosin Vb, and E-cadherin, and that the PBD is required for this interaction. These data indicate that IRF6 is a novel effector of the endosomal recycling of E-cadherin and demonstrate a non-transcriptional function for IRF6 in regulating cell-cell adhesions.


Subject(s)
Cadherins , Cell Adhesion , Endosomes , Interferon Regulatory Factors , Animals , Humans , Mice , Cadherins/metabolism , Cell Adhesion/physiology , Cell Membrane/metabolism , Endosomes/metabolism , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Keratinocytes/metabolism , Protein Binding , Protein Domains , Protein Transport , rab GTP-Binding Proteins/metabolism
9.
J Vis Exp ; (207)2024 May 03.
Article in English | MEDLINE | ID: mdl-38767378

ABSTRACT

Ultrashort self-assembling peptides (SAPs) can spontaneously form nanofibers that resemble the extracellular matrix. These fibers allow the formation of hydrogels that are biocompatible, biodegradable, and non-immunogenic. We have previously proven that SAPs, when biofunctionalized with protein-derived motifs, can mimic the extracellular matrix characteristics that support colorectal organoid formation. These biofunctional peptide hydrogels retain the original parent peptide's mechanical properties, tunability, and printability while incorporating cues that allow cell-matrix interactions to increase cell adhesion. This paper presents the protocols needed to evaluate and characterize the effects of various biofunctional peptide hydrogels on cell adhesion and lumen formation using an adenocarcinoma cancer cell line able to form colorectal cancer organoids cost-effectively. These protocols will help evaluate biofunctional peptide hydrogel effects on cell adhesion and luminal formation using immunostaining and fluorescence image analysis. The cell line used in this study has been previously utilized for generating organoids in animal-derived matrices.


Subject(s)
Colorectal Neoplasms , Hydrogels , Organoids , Peptides , Organoids/cytology , Humans , Colorectal Neoplasms/pathology , Cell Line, Tumor , Hydrogels/chemistry , Peptides/chemistry , Nanofibers/chemistry , Adenocarcinoma/pathology , Extracellular Matrix/chemistry , Cell Adhesion/physiology
10.
Proc Natl Acad Sci U S A ; 121(22): e2318248121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38787878

ABSTRACT

For eukaryotic cells to heal wounds, respond to immune signals, or metastasize, they must migrate, often by adhering to extracellular matrix (ECM). Cells may also deposit ECM components, leaving behind a footprint that influences their crawling. Recent experiments showed that some epithelial cell lines on micropatterned adhesive stripes move persistently in regions they have previously crawled over, where footprints have been formed, but barely advance into unexplored regions, creating an oscillatory migration of increasing amplitude. Here, we explore through mathematical modeling how footprint deposition and cell responses to footprint combine to allow cells to develop oscillation and other complex migratory motions. We simulate cell crawling with a phase field model coupled to a biochemical model of cell polarity, assuming local contact with the deposited footprint activates Rac1, a protein that establishes the cell's front. Depending on footprint deposition rate and response to the footprint, cells on micropatterned lines can display many types of motility, including confined, oscillatory, and persistent motion. On two-dimensional (2D) substrates, we predict a transition between cells undergoing circular motion and cells developing an exploratory phenotype. Small quantitative changes in a cell's interaction with its footprint can completely alter exploration, allowing cells to tightly regulate their motion, leading to different motility phenotypes (confined vs. exploratory) in different cells when deposition or sensing is variable from cell to cell. Consistent with our computational predictions, we find in earlier experimental data evidence of cells undergoing both circular and exploratory motion.


Subject(s)
Cell Movement , Extracellular Matrix , Cell Movement/physiology , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , rac1 GTP-Binding Protein/metabolism , Humans , Cell Polarity/physiology , Models, Biological , Animals , Cell Adhesion/physiology , Epithelial Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/physiology
11.
Invest Ophthalmol Vis Sci ; 65(5): 4, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38691089

ABSTRACT

Purpose: To investigate the adhesion of Acanthamoeba to scleral contact lens (ScCL) surface according to lens shape. Methods: Two strains of A. polyphaga (CDC:V062 and ATCC 30461) and one clinical Acanthamoeba isolate, were inoculated onto five contact lens (CL): one first-generation silicone hydrogel (SHCL; lotrafilcon B; adhesion control) containing plasma surface treatment; two ScCL (fluorosilicone acrylate) one containing surface treatment composed of plasma and the other containing plasma with Hydra-PEG, and two CL designed with a flat shape having the same material and surface treatments of the ScCL. Trophozoites that adhered to the lens's surfaces were counted by inverted optical light microscopy. Possible alterations of the lens surface that could predispose amoeba adhesion and Acanthamoeba attached to these lens surfaces were evaluated by scanning electron microscopy (SEM). Results: All strains revealed greater adhesion to the ScCL when compared with the flat lenses (P < 0.001). The clinical isolate and the ATCC 30461 had a higher adhesion (P < 0.001) when compared with the CDC:V062. A rough texture was observed on the surface of the lenses that have been examined by SEM. Also, SEM revealed that the isolates had a rounded appearance on the surface of the ScCL in contrast with an elongated appearance on the surface of the silicone hydrogel. Conclusions: The findings revealed that the curved shape of the ScCL favors amoeba adhesion.


Subject(s)
Acanthamoeba , Microscopy, Electron, Scanning , Acanthamoeba/physiology , Acanthamoeba/ultrastructure , Sclera , Humans , Contact Lenses, Hydrophilic/parasitology , Cell Adhesion/physiology , Contact Lenses/parasitology , Trophozoites/ultrastructure , Trophozoites/physiology , Hydrogels , Animals
12.
J R Soc Interface ; 21(214): 20240022, 2024 May.
Article in English | MEDLINE | ID: mdl-38715321

ABSTRACT

Using a three-dimensional model of cell monolayers, we study the spatial organization of active stress chains as the monolayer transitions from a solid to a liquid state. The critical exponents that characterize this transition map the isotropic stress percolation onto the two-dimensional random percolation universality class, suggesting short-range stress correlations near this transition. This mapping is achieved via two distinct, independent pathways: (i) cell-cell adhesion and (ii) active traction forces. We unify our findings by linking the nature of this transition to high-stress fluctuations, distinctly linked to each pathway. The results elevate the importance of the transmission of mechanical information in dense active matter and provide a new context for understanding the non-equilibrium statistical physics of phase transition in active systems.


Subject(s)
Cell Adhesion , Models, Biological , Cell Adhesion/physiology , Stress, Mechanical , Phase Transition
13.
J Phys Condens Matter ; 36(29)2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38574682

ABSTRACT

Cell-matrix adhesions connect the cytoskeleton to the extracellular environment and are essential for maintaining the integrity of tissue and whole organisms. Remarkably, cell adhesions can adapt their size and composition to an applied force such that their size and strength increases proportionally to the load. Mathematical models for the clutch-like force transmission at adhesions are frequently based on the assumption that mechanical load is applied tangentially to the adhesion plane. Recently, we suggested a molecular mechanism that can explain adhesion growth under load for planar cell adhesions. The mechanism is based on conformation changes of adhesion molecules that are dynamically exchanged with a reservoir. Tangential loading drives the occupation of some states out of equilibrium, which for thermodynamic reasons, leads to the association of further molecules with the cluster, which we refer to as self-stabilization. Here, we generalize this model to forces that pull at an oblique angle to the plane supporting the cell, and examine if this idealized model also predicts self-stabilization. We also allow for a variable distance between the parallel planes representing cytoskeletal F-actin and transmembrane integrins. Simulation results demonstrate that the binding mechanism and the geometry of the cluster have a strong influence on the response of adhesion clusters to force. For oblique angles smaller than about 40∘, we observe a growth of the adhesion site under force. However this self-stabilization is reduced as the angle between the force and substrate plane increases, with vanishing self-stabilization for normal pulling. Overall, these results highlight the fundamental difference between the assumption of pulling and shearing forces in commonly used models of cell adhesion.


Subject(s)
Extracellular Matrix , Focal Adhesions , Focal Adhesions/metabolism , Extracellular Matrix/metabolism , Cell Adhesion/physiology , Actins , Integrins/metabolism
14.
Nat Comput Sci ; 4(4): 299-309, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38594592

ABSTRACT

The three-dimensional (3D) organization of cells determines tissue function and integrity, and changes markedly in development and disease. Cell-based simulations have long been used to define the underlying mechanical principles. However, high computational costs have so far limited simulations to either simplified cell geometries or small tissue patches. Here, we present SimuCell3D, an efficient open-source program to simulate large tissues in three dimensions with subcellular resolution, growth, proliferation, extracellular matrix, fluid cavities, nuclei and non-uniform mechanical properties, as found in polarized epithelia. Spheroids, vesicles, sheets, tubes and other tissue geometries can readily be imported from microscopy images and simulated to infer biomechanical parameters. Doing so, we show that 3D cell shapes in layered and pseudostratified epithelia are largely governed by a competition between surface tension and intercellular adhesion. SimuCell3D enables the large-scale in silico study of 3D tissue organization in development and disease at a great level of detail.


Subject(s)
Cell Polarity , Computer Simulation , Models, Biological , Biomechanical Phenomena/physiology , Cell Adhesion/physiology , Cell Polarity/physiology , Cell Shape/physiology , Epithelial Cells/physiology , Epithelial Cells/cytology , Extracellular Matrix/physiology , Extracellular Matrix/chemistry , Imaging, Three-Dimensional/methods , Software
15.
Mol Biol Cell ; 35(5): ar65, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38507238

ABSTRACT

α-catenin (α-cat) displays force-dependent unfolding and binding to actin filaments through direct and indirect means, but features of adherens junction structure and function most vulnerable to loss of these allosteric mechanisms have not been directly compared. By reconstituting an α-cat F-actin-binding domain unfolding mutant known to exhibit enhanced binding to actin (α-cat-H0-FABD+) into α-cat knockout Madin Darby Canine Kidney (MDCK) cells, we show that partial loss of the α-cat catch bond mechanism (via an altered H0 α-helix) leads to stronger epithelial sheet integrity with greater colocalization between the α-cat-H0-FABD+ mutant and actin. α-cat-H0-FABD+ -expressing cells are less efficient at closing scratch-wounds, suggesting reduced capacity for more dynamic cell-cell coordination. Evidence that α-cat-H0-FABD+ is equally accessible to the conformationally sensitive α18 antibody epitope as WT α-cat and shows similar vinculin recruitment suggests this mutant engages lower tension cortical actin networks, as its M-domain is not persistently open. Conversely, α-cat-M-domain salt-bridge mutants with persistent recruitment of vinculin and phosphorylated myosin light chain show only intermediate monolayer adhesive strengths, but display less directionally coordinated and thereby slower migration speeds during wound-repair. These data show α-cat M- and FABD-unfolding mutants differentially impact cell-cell cohesion and migration properties, and suggest signals favoring α-cat-cortical actin interaction without persistent M-domain opening may improve epithelial monolayer strength through enhanced coupling to lower tension actin networks.


Subject(s)
Actin Cytoskeleton , Actins , Cell Movement , Epithelial Cells , alpha Catenin , Dogs , Animals , alpha Catenin/metabolism , alpha Catenin/genetics , Madin Darby Canine Kidney Cells , Actins/metabolism , Epithelial Cells/metabolism , Actin Cytoskeleton/metabolism , Protein Binding , Protein Domains , Mutation , Adherens Junctions/metabolism , Protein Unfolding , Cell Adhesion/physiology , Vinculin/metabolism
16.
Science ; 383(6689): 1374-1379, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38513010

ABSTRACT

Cells connect with their environment through surface receptors and use physical tension in receptor-ligand bonds for various cellular processes. Single-molecule techniques have revealed bond strength by measuring "rupture force," but it has long been recognized that rupture force is dependent on loading rate-how quickly force is ramped up. Thus, the physiological loading rate needs to be measured to reveal the mechanical strength of individual bonds in their functional context. We have developed an overstretching tension sensor (OTS) to allow more accurate force measurement in physiological conditions with single-molecule detection sensitivity even in mechanically active regions. We used serially connected OTSs to show that the integrin loading rate ranged from 0.5 to 4 piconewtons per second and was about three times higher in leukocytes than in epithelial cells.


Subject(s)
Biosensing Techniques , Cell Adhesion , Integrins , Mechanotransduction, Cellular , Cell Adhesion/physiology , Integrins/chemistry , Integrins/metabolism , Single Molecule Imaging , Humans , Cell Line, Tumor , Tensile Strength , Oligonucleotide Probes , Nucleic Acid Hybridization
17.
Endocrinology ; 165(5)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38518755

ABSTRACT

Seminal extracellular vesicles (EVs) contain different subgroups that have diverse effects on sperm function. However, the effect of seminal EVs-especially their subgroups-on endometrial receptivity is largely unknown. Here, we found that seminal EVs could be divided into high-density EVs (EV-H), medium density EVs, and low-density EVs after purification using iodixanol. We demonstrated that EV-H could promote the expression and secretion of leukemia inhibitor factor (LIF) in human endometrial cells. In EV-H-treated endometrial cells, we identified 1274 differentially expressed genes (DEGs). DEGs were enriched in cell adhesion and AKT and STAT3 pathways. Therefore, we illustrated that EV-H enhanced the adhesion of human choriocarcinoma JAr cell spheroids to endometrial cells through the LIF-STAT3 pathway. Collectively, our findings indicated that seminal EV-H could regulate endometrial receptivity through the LIF pathway, which could provide novel insights into male fertility.


Subject(s)
Embryo Implantation , Extracellular Vesicles , Female , Humans , Male , Pregnancy , Cell Adhesion/physiology , Embryo Implantation/physiology , Endometrium/metabolism , Extracellular Vesicles/metabolism , Leukemia Inhibitory Factor/metabolism , Semen/metabolism
18.
ACS Appl Mater Interfaces ; 16(11): 13622-13639, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38466038

ABSTRACT

The design of implantable biomaterials involves precise tuning of surface features because the early cellular fate on such engineered surfaces is highly influenced by many physicochemical factors [roughness, hydrophilicity, reactive oxygen species (ROS) responsiveness, etc.]. Herein, to enhance soft tissue integration for successful implantation, Ti substrates decorated with uniform layers of nanoceria (Ce), called Ti@Ce, were optimally developed by a simple and cost-effective in situ immersion coating technique. The characterization of Ti@Ce shows a uniform Ce distribution with enhanced roughness (∼3-fold increase) and hydrophilicity (∼4-fold increase) and adopted ROS-scavenging capacity by nanoceria coating. When human gingival fibroblasts were seeded on Ti@Ce under oxidative stress conditions, Ti@Ce supported cellular adhesion, spreading, and survivability by its cellular ROS-scavenging capacity. Mechanistically, the unique nanocoating resulted in higher expression of amphiphysin (a nanotopology sensor), paxillin (a focal adhesion protein), and cell adhesive proteins (collagen-1 and fibronectin). Ti@Ce also led to global chromatin condensation by decreasing histone 3 acetylation as an early differentiation feature. Transcriptome analysis by RNA sequencing confirmed the chromatin remodeling, antiapoptosis, antioxidant, cell adhesion, and TGF-ß signaling-related gene signatures in Ti@Ce. As key fibroblast transcription (co)factors, Ti@Ce promotes serum response factor and MRTF-α nucleus localization. Considering all of this, it is proposed that the surface engineering approach using Ce could improve the biological properties of Ti implants, supporting their functioning at soft tissue interfaces and utilization as a bioactive implant for clinical conditions such as peri-implantitis.


Subject(s)
Cerium , Fibroblasts , Titanium , Humans , Reactive Oxygen Species/metabolism , Titanium/pharmacology , Titanium/chemistry , Cells, Cultured , Surface Properties , Cell Adhesion/physiology , Fibroblasts/metabolism
19.
Biofabrication ; 16(3)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38471164

ABSTRACT

Cells sense mechanical signals from the surrounding environment and transmit them to the nucleus through mechanotransduction to regulate cellular behavior. Microcontact printing, which utilizes elastomer stamps, is an effective method for simulating the cellular microenvironment and manipulating cell morphology. However, the conventional fabrication process of silicon masters and elastomer stamps requires complex procedures and specialized equipment, which restricts the widespread application of micropatterning in cell biology and hinders the investigation of the role of cell geometry in regulating cell behavior. In this study, we present an innovative method for convenient resin stamp microfabrication based on digital micromirror device planar lithography. Using this method, we generated a series of patterns ranging from millimeter to micrometer scales and validated their effectiveness in controlling adhesion at both collective and individual cell levels. Additionally, we investigated mechanotransduction and cell behavior on elongated micropatterned substrates. We then examined the effects of cell elongation on cytoskeleton organization, nuclear deformation, focal adhesion formation, traction force generation, nuclear mechanics, and the growth of HeLa cells. Our findings reveal a positive correlation between cell length and mechanotransduction. Interestingly, HeLa cells with moderate length exhibit the highest cell division and proliferation rates. These results highlight the regulatory role of cell elongation in mechanotransduction and its significant impact on cancer cell growth. Furthermore, our methodology for controlling cell adhesion holds the potential for addressing fundamental questions in both cell biology and biomedical engineering.


Subject(s)
Elastomers , Mechanotransduction, Cellular , Humans , HeLa Cells , Cell Adhesion/physiology , Cell Division
20.
Proc Natl Acad Sci U S A ; 121(13): e2314947121, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38513099

ABSTRACT

Protein kinase A (PKA) is a ubiquitous, promiscuous kinase whose activity is specified through subcellular localization mediated by A-kinase anchoring proteins (AKAPs). PKA has complex roles as both an effector and a regulator of integrin-mediated cell adhesion to extracellular matrix (ECM). Recent observations demonstrate that PKA is an active component of focal adhesions (FA), suggesting the existence of one or more FA AKAPs. Using a promiscuous biotin ligase fused to PKA type-IIα regulatory (RIIα) subunits and subcellular fractionation, we identify the archetypal FA protein talin1 as an AKAP. Talin is a large, mechanosensitive scaffold that directly links integrins to actin filaments and promotes FA assembly by recruiting additional components in a force-dependent manner. The rod region of talin1 consists of 62 α-helices bundled into 13 rod domains, R1 to R13. Direct binding assays and NMR spectroscopy identify helix41 in the R9 subdomain of talin as the PKA binding site. PKA binding to helix41 requires unfolding of the R9 domain, which requires the linker region between R9 and R10. Experiments with single molecules and in cells manipulated to alter actomyosin contractility demonstrate that the PKA-talin interaction is regulated by mechanical force across the talin molecule. Finally, talin mutations that disrupt PKA binding also decrease levels of total and phosphorylated PKA RII subunits as well as phosphorylation of VASP, a known PKA substrate, within FA. These observations identify a mechanically gated anchoring protein for PKA, a force-dependent binding partner for talin1, and a potential pathway for adhesion-associated mechanotransduction.


Subject(s)
A Kinase Anchor Proteins , Focal Adhesions , Focal Adhesions/metabolism , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , Talin/metabolism , Mechanotransduction, Cellular , Cell Adhesion/physiology , Integrins/metabolism , Protein Binding , Cyclic AMP-Dependent Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...