Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.926
Filter
1.
NPJ Syst Biol Appl ; 10(1): 90, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39147782

ABSTRACT

YAP/TAZ signaling pathway is regulated by a multiplicity of feedback loops, crosstalk with other pathways, and both mechanical and biochemical stimuli. Computational modeling serves as a powerful tool to unravel how these different factors can regulate YAP/TAZ, emphasizing biophysical modeling as an indispensable tool for deciphering mechanotransduction and its regulation of cell fate. We provide a critical review of the current state-of-the-art of computational models focused on YAP/TAZ signaling.


Subject(s)
Computer Simulation , Mechanotransduction, Cellular , Transcription Factors , Mechanotransduction, Cellular/physiology , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , YAP-Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Signal Transduction/physiology , Models, Biological , Animals
2.
J Vis Exp ; (209)2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39141554

ABSTRACT

We present an innovative in vitro model aimed at investigating the combined effects of tissue rigidity and shear stress on endothelial cell (EC) function, which are crucial for understanding vascular health and the onset of diseases such as atherosclerosis. Traditionally, studies have explored the impacts of shear stress and substrate stiffness on ECs, independently. However, this integrated system combines these factors to provide a more precise simulation of the mechanical environment of the vasculature. The objective is to examine EC mechanotransduction across various tissue stiffness levels and flow conditions using human ECs. We detail the protocol for synthesizing gelatin methacrylate (GelMA) hydrogels with tunable stiffness and seeding them with ECs to achieve confluency. Additionally, we describe the design and assembly of a cost-effective flow chamber, supplemented by computational fluid dynamics simulations, to generate physiological flow conditions characterized by laminar flow and appropriate shear stress levels. The protocol also incorporates fluorescence labeling for confocal microscopy, enabling the assessment of EC responses to both tissue compliance and flow conditions. By subjecting cultured ECs to multiple integrated mechanical stimuli, this model enables comprehensive investigations into how factors such as hypertension and aging may affect EC function and EC-mediated vascular diseases. The insights gained from these investigations will be instrumental in elucidating the mechanisms underlying vascular diseases and in developing effective treatment strategies.


Subject(s)
Endothelial Cells , Hydrogels , Humans , Hydrogels/chemistry , Endothelial Cells/cytology , Gelatin/chemistry , Human Umbilical Vein Endothelial Cells , Mechanotransduction, Cellular/physiology , Methacrylates/chemistry , Stress, Mechanical , Microscopy, Confocal/methods , Hydrodynamics
3.
FASEB J ; 38(15): e23849, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39096133

ABSTRACT

Living cells navigate a complex landscape of mechanical cues that influence their behavior and fate, originating from both internal and external sources. At the molecular level, the translation of these physical stimuli into cellular responses relies on the intricate coordination of mechanosensors and transducers, ultimately impacting chromatin compaction and gene expression. Notably, epigenetic modifications on histone tails govern the accessibility of gene-regulatory sites, thereby regulating gene expression. Among these modifications, histone acetylation emerges as particularly responsive to the mechanical microenvironment, exerting significant control over cellular activities. However, the precise role of histone acetylation in mechanosensing and transduction remains elusive due to the complexity of the acetylation network. To address this gap, our aim is to systematically explore the key regulators of histone acetylation and their multifaceted roles in response to biomechanical stimuli. In this review, we initially introduce the ubiquitous force experienced by cells and then explore the dynamic alterations in histone acetylation and its associated co-factors, including HDACs, HATs, and acetyl-CoA, in response to these biomechanical cues. Furthermore, we delve into the intricate interactions between histone acetylation and mechanosensors/mechanotransducers, offering a comprehensive analysis. Ultimately, this review aims to provide a holistic understanding of the nuanced interplay between histone acetylation and mechanical forces within an academic framework.


Subject(s)
Histones , Histones/metabolism , Acetylation , Humans , Animals , Mechanotransduction, Cellular/physiology , Epigenesis, Genetic , Protein Processing, Post-Translational , Biomechanical Phenomena
4.
Int J Oral Sci ; 16(1): 52, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39085217

ABSTRACT

Compared with teenage patients, adult patients generally show a slower rate of tooth movement and more pronounced alveolar bone loss during orthodontic treatment, indicating the maladaptation of alveolar bone homeostasis under orthodontic force. However, this phenomenon is not well-elucidated to date, leading to increased treatment difficulties and unsatisfactory treatment outcomes in adult orthodontics. Aiming to provide a comprehensive knowledge and further inspire insightful understanding towards this issue, this review summarizes the current evidence and underlying mechanisms. The age-related abatements in mechanosensing and mechanotransduction in adult cells and periodontal tissue may contribute to retarded and unbalanced bone metabolism, thus hindering alveolar bone reconstruction during orthodontic treatment. To this end, periodontal surgery, physical and chemical cues are being developed to reactivate or rejuvenate the aging periodontium and restore the dynamic equilibrium of orthodontic-mediated alveolar bone metabolism. We anticipate that this review will present a general overview of the role that aging plays in orthodontic alveolar bone metabolism and shed new light on the prospective ways out of the impasse.


Subject(s)
Adaptation, Physiological , Aging , Alveolar Bone Loss , Alveolar Process , Humans , Adult , Aging/physiology , Adaptation, Physiological/physiology , Tooth Movement Techniques , Mechanotransduction, Cellular/physiology , Bone Remodeling/physiology
5.
Integr Biol (Camb) ; 162024 Jan 23.
Article in English | MEDLINE | ID: mdl-38952079

ABSTRACT

Mechanical forces are of major importance in regulating vascular homeostasis by influencing endothelial cell behavior and functions. Adherens junctions are critical sites for mechanotransduction in endothelial cells. ß-catenin, a component of adherens junctions and the canonical Wnt signaling pathway, plays a role in mechanoactivation. Evidence suggests that ß-catenin is involved in flow sensing and responds to tensional forces, impacting junction dynamics. The mechanoregulation of ß-catenin signaling is context-dependent, influenced by the type and duration of mechanical loads. In endothelial cells, ß-catenin's nuclear translocation and signaling are influenced by shear stress and strain, affecting endothelial permeability. The study investigates how shear stress, strain, and surface topography impact adherens junction dynamics, regulate ß-catenin localization, and influence endothelial barrier properties. Insight box Mechanical loads are potent regulators of endothelial functions through not completely elucidated mechanisms. Surface topography, wall shear stress and cyclic wall deformation contribute overlapping mechanical stimuli to which endothelial monolayer respond to adapt and maintain barrier functions. The use of custom developed flow chamber and bioreactor allows quantifying the response of mature human endothelial to well-defined wall shear stress and gradients of strain. Here, the mechanoregulation of ß-catenin by substrate topography, wall shear stress, and cyclic stretch is analyzed and linked to the monolayer control of endothelial permeability.


Subject(s)
Adherens Junctions , Endothelial Cells , Human Umbilical Vein Endothelial Cells , Mechanotransduction, Cellular , Stress, Mechanical , beta Catenin , beta Catenin/metabolism , Humans , Mechanotransduction, Cellular/physiology , Adherens Junctions/metabolism , Endothelial Cells/metabolism , Shear Strength , Wnt Signaling Pathway , Biomechanical Phenomena
6.
PLoS Biol ; 22(7): e3002729, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39024405

ABSTRACT

Sensory neurons specialize in detecting and signaling the presence of diverse environmental stimuli. Neuronal injury or disease may undermine such signaling, diminishing the availability of crucial information. Can animals distinguish between a stimulus not being present and the inability to sense that stimulus in the first place? To address this question, we studied Caenorhabditis elegans nematode worms that lack gentle body touch sensation due to genetic mechanoreceptor dysfunction. We previously showed that worms can compensate for the loss of touch by enhancing their sense of smell, via an FLP-20 neuropeptide pathway. Here, we find that touch-deficient worms exhibit, in addition to sensory compensation, also cautious-like behavior, as if preemptively avoiding potential undetectable hazards. Intriguingly, these behavioral adjustments are abolished when the touch neurons are removed, suggesting that touch neurons are required for signaling the unavailability of touch information, in addition to their conventional role of signaling touch stimulation. Furthermore, we found that the ASE taste neurons, which similarly to the touch neurons, express the FLP-20 neuropeptide, exhibit altered FLP-20 expression levels in a touch-dependent manner, thus cooperating with the touch circuit. These results imply a novel form of neuronal signaling that enables C. elegans to distinguish between lack of touch stimulation and loss of touch sensation, producing adaptive behavioral adjustments that could overcome the inability to detect potential threats.


Subject(s)
Behavior, Animal , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Mechanoreceptors , Touch , Animals , Caenorhabditis elegans/physiology , Mechanoreceptors/physiology , Mechanoreceptors/metabolism , Touch/physiology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Behavior, Animal/physiology , Neuropeptides/metabolism , Neuropeptides/genetics , Mechanotransduction, Cellular/physiology , Smell/physiology , Sensory Receptor Cells/physiology , Sensory Receptor Cells/metabolism
7.
Curr Biol ; 34(14): 3133-3151.e10, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38964319

ABSTRACT

The sense of touch is conferred by the conjoint function of somatosensory neurons and skin cells. These cells meet across a gap filled by a basal lamina, an ancient structure found in metazoans. Using Caenorhabditis elegans, we investigate the composition and ultrastructure of the extracellular matrix at the epidermis and touch receptor neuron (TRN) interface. We show that membrane-matrix complexes containing laminin, nidogen, and the MEC-4 mechano-electrical transduction channel reside at this interface and are central to proper touch sensation. Interestingly, the dimensions and spacing of these complexes correspond with the discontinuous beam-like extracellular matrix structures observed in serial-section transmission electron micrographs. These complexes fail to coalesce in touch-insensitive extracellular matrix mutants and in dissociated neurons. Loss of nidogen reduces the density of mechanoreceptor complexes and the amplitude of the touch-evoked currents they carry. Thus, neuron-epithelium cell interfaces are instrumental in mechanosensory complex assembly and function. Unlike the basal lamina ensheathing the pharynx and body wall muscle, nidogen recruitment to the puncta along TRNs is not dependent upon laminin binding. MEC-4, but not laminin or nidogen, is destabilized by point mutations in the C-terminal Kunitz domain of the extracellular matrix component, MEC-1. These findings imply that somatosensory neurons secrete proteins that actively repurpose the basal lamina to generate special-purpose mechanosensory complexes responsible for vibrotactile sensing.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Mechanoreceptors , Mechanotransduction, Cellular , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Mechanoreceptors/metabolism , Mechanoreceptors/physiology , Mechanotransduction, Cellular/physiology , Touch/physiology , Basement Membrane/metabolism , Basement Membrane/physiology , Extracellular Matrix/metabolism , Laminin/metabolism , Membrane Glycoproteins , Membrane Proteins
8.
Proc Natl Acad Sci U S A ; 121(28): e2404210121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38954541

ABSTRACT

Mesenchymal stem cells (MSCs) are essential in regenerative medicine. However, conventional expansion and harvesting methods often fail to maintain the essential extracellular matrix (ECM) components, which are crucial for their functionality and efficacy in therapeutic applications. Here, we introduce a bone marrow-inspired macroporous hydrogel designed for the large-scale production of MSC-ECM spheroids. Through a soft-templating approach leveraging liquid-liquid phase separation, we engineer macroporous hydrogels with customizable features, including pore size, stiffness, bioactive ligand distribution, and enzyme-responsive degradability. These tailored environments are conducive to optimal MSC proliferation and ease of harvesting. We find that soft hydrogels enhance mechanotransduction in MSCs, establishing a standard for hydrogel-based 3D cell culture. Within these hydrogels, MSCs exist as both cohesive spheroids, preserving their innate vitality, and as migrating entities that actively secrete functional ECM proteins. Additionally, we also introduce a gentle, enzymatic harvesting method that breaks down the hydrogels, allowing MSCs and secreted ECM to naturally form MSC-ECM spheroids. These spheroids display heightened stemness and differentiation capacity, mirroring the benefits of a native ECM milieu. Our research underscores the significance of sophisticated materials design in nurturing distinct MSC subpopulations, facilitating the generation of MSC-ECM spheroids with enhanced therapeutic potential.


Subject(s)
Extracellular Matrix , Hydrogels , Mesenchymal Stem Cells , Spheroids, Cellular , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Hydrogels/chemistry , Extracellular Matrix/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Humans , Cell Differentiation , Cell Culture Techniques/methods , Cell Proliferation , Porosity , Mechanotransduction, Cellular/physiology , Cells, Cultured
9.
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
10.
Integr Biol (Camb) ; 162024 Jan 23.
Article in English | MEDLINE | ID: mdl-38900169

ABSTRACT

Cells dynamically remodel their internal structures by modulating the arrangement of actin filaments (AFs). In this process, individual AFs exhibit stochastic behavior without knowing the macroscopic higher-order structures they are meant to create or disintegrate, but the mechanism allowing for such stochastic process-driven remodeling of subcellular structures remains incompletely understood. Here we employ percolation theory to explore how AFs interacting only with neighboring ones without recognizing the overall configuration can nonetheless create a substantial structure referred to as stress fibers (SFs) at particular locations. We determined the interaction probabilities of AFs undergoing cellular tensional homeostasis, a fundamental property maintaining intracellular tension. We showed that the duration required for the creation of SFs is shortened by the increased amount of preexisting actin meshwork, while the disintegration occurs independently of the presence of actin meshwork, suggesting that the coexistence of tension-bearing and non-bearing elements allows cells to promptly transition to new states in accordance with transient environmental changes. The origin of this asymmetry between creation and disintegration, consistently observed in actual cells, is elucidated through a minimal model analysis by examining the intrinsic nature of mechano-signal transmission. Specifically, unlike the symmetric case involving biochemical communication, physical communication to sense environmental changes is facilitated via AFs under tension, while other free AFs dissociated from tension-bearing structures exhibit stochastic behavior. Thus, both the numerical and minimal models demonstrate the essence of intracellular percolation, in which macroscopic asymmetry observed at the cellular level emerges not from microscopic asymmetry in the interaction probabilities of individual molecules, but rather only as a consequence of the manner of the mechano-signal transmission. These results provide novel insights into the role of the mutual interplay between distinct subcellular structures with and without tension-bearing capability. Insight: Cells continuously remodel their internal elements or structural proteins in response to environmental changes. Despite the stochastic behavior of individual structural proteins, which lack awareness of the larger subcellular structures they are meant to create or disintegrate, this self-assembly process somehow occurs to enable adaptation to the environment. Here we demonstrated through percolation simulations and minimal model analyses that there is an asymmetry in the response between the creation and disintegration of subcellular structures, which can aid environmental adaptation. This asymmetry inherently arises from the nature of mechano-signal transmission through structural proteins, namely tension-mediated information exchange within cells, despite the stochastic behavior of individual proteins lacking asymmetric characters in themselves.


Subject(s)
Actin Cytoskeleton , Models, Biological , Stochastic Processes , Stress Fibers , Actin Cytoskeleton/chemistry , Stress Fibers/physiology , Stress Fibers/metabolism , Computer Simulation , Mechanotransduction, Cellular/physiology , Stress, Mechanical , Humans , Animals , Actins/metabolism , Actins/chemistry
11.
STAR Protoc ; 5(2): 103098, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38823011

ABSTRACT

Here, we present a protocol to detect mechanosensitive responses of proteins in cells under compressive stress. We describe steps for preparing elastic gels to compress cells grown on an imaging chamber. We then detail procedures for imaging proteins at the cell cortex using high-resolution confocal microscopy. The protocol can be applied to examine the mechanosensitive response of fluorescently tagged proteins in mitotic cells or round interphase cells adhering to the imaging surface. For complete details on the use and execution of this protocol, please refer to Wang et al.1.


Subject(s)
Mechanotransduction, Cellular , Stress, Mechanical , Mechanotransduction, Cellular/physiology , Microscopy, Confocal/methods , Humans , Cells, Cultured , Proteins/metabolism , Proteins/analysis , Animals
12.
J Speech Lang Hear Res ; 67(7): 2128-2138, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38865255

ABSTRACT

PURPOSE: The vocal fold tissues undergo nearly continuous and repeated cycles of injury and repair throughout the course of an individual's lifetime. It is well established that certain individuals are at greater risk of lesion development based on personality and behavioral classification. However, these characteristics alone do not wholly predict or explain lesion development or severity. In this review, we discuss current knowledge of mechanotransduction proteins and their potential relevance to tissue homeostasis in the vocal folds. METHOD: A review of literature surrounding mechanotransduction and tissue homeostasis as it relates to the vocal folds was conducted. Review of the literature included searches of PubMed, Google Scholar, and other various online peer-reviewed sources. Search terms pertained to mechanosensation, mechanotransduction, mechanically activated channels, mechanical cellular regulation, and other associated concepts and terms. Additional literature was identified through the reference lists of identified papers. Findings of this literature review were then applied to known physiology and pathophysiology of the vocal folds in order to speculate on the contribution of mechanically mediated mechanisms within the vocal fold. DISCUSSION AND CONCLUSION: Because the vocal folds are such mechanically active structures, withstanding nearly constant external forces, there is strong support for the idea that mechanically sensitive molecular pathways within the vocal fold tissue play a major role in tissue homeostasis in the presence of these considerable forces. As such, mechanotransduction within the vocal fold should be considered and targeted in future biological studies of vocal fold physiology.


Subject(s)
Mechanotransduction, Cellular , Vocal Cords , Humans , Vocal Cords/physiology , Vocal Cords/physiopathology , Mechanotransduction, Cellular/physiology , Animals , Homeostasis/physiology
13.
J Biomech ; 171: 112179, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38852482

ABSTRACT

Cell volume and shape changes play a pivotal role in cellular mechanotransduction, governing cellular responses to external loading. Understanding the dynamics of cell behavior under loading conditions is essential to elucidate cell adaptation mechanisms in physiological and pathological contexts. In this study, we investigated the effects of dynamic cyclic compression loading on cell volume and shape changes, comparing them with static conditions. Using a custom-designed platform which allowed for simultaneous loading and imaging of cartilage tissue, tissues were subjected to 100 cycles of mechanical loading while measuring cell volume and shape alterations during the unloading phase at specific time points. The findings revealed a transient decrease in cell volume (13%) during the early cycles, followed by a gradual recovery to baseline levels after approximately 20 cycles, despite the cartilage tissue not being fully recovered at the unloading phase. This observed pattern indicates a temporal cell volume response that may be associated with cellular adaptation to the mechanical stimulus through mechanisms related to active cell volume regulation. Additionally, this study demonstrated that cell volume and shape responses during dynamic loading were significantly distinct from those observed under static conditions. Such findings suggest that cells in their natural tissue environment perceive and respond differently to dynamic compared to static mechanical cues, highlighting the significance of considering dynamic loading environments in studies related to cellular mechanics. Overall, this research contributes to the broader understanding of cellular behavior under mechanical stimuli, providing valuable insights into their ability to adapt to dynamic mechanical loading.


Subject(s)
Chondrocytes , Weight-Bearing , Animals , Chondrocytes/physiology , Weight-Bearing/physiology , Stress, Mechanical , Cell Size , Mechanotransduction, Cellular/physiology , Compressive Strength/physiology , Cattle , Cartilage, Articular/physiology , Cell Shape/physiology
14.
Int J Mol Sci ; 25(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38891829

ABSTRACT

It is remarkable how teeth maintain their healthy condition under exceptionally high levels of mechanical loading. This suggests the presence of inherent mechanical adaptation mechanisms within their structure to counter constant stress. Dentin, situated between enamel and pulp, plays a crucial role in mechanically supporting tooth function. Its intermediate stiffness and viscoelastic properties, attributed to its mineralized, nanofibrous extracellular matrix, provide flexibility, strength, and rigidity, enabling it to withstand mechanical loading without fracturing. Moreover, dentin's unique architectural features, such as odontoblast processes within dentinal tubules and spatial compartmentalization between odontoblasts in dentin and sensory neurons in pulp, contribute to a distinctive sensory perception of external stimuli while acting as a defensive barrier for the dentin-pulp complex. Since dentin's architecture governs its functions in nociception and repair in response to mechanical stimuli, understanding dentin mechanobiology is crucial for developing treatments for pain management in dentin-associated diseases and dentin-pulp regeneration. This review discusses how dentin's physical features regulate mechano-sensing, focusing on mechano-sensitive ion channels. Additionally, we explore advanced in vitro platforms that mimic dentin's physical features, providing deeper insights into fundamental mechanobiological phenomena and laying the groundwork for effective mechano-therapeutic strategies for dentinal diseases.


Subject(s)
Dentin , Dentin/physiology , Dentin/metabolism , Humans , Animals , Odontoblasts/physiology , Odontoblasts/metabolism , Odontoblasts/cytology , Mechanotransduction, Cellular/physiology , Biomechanical Phenomena , Dental Pulp/physiology , Extracellular Matrix/metabolism , Extracellular Matrix/physiology
15.
CNS Neurosci Ther ; 30(6): e14809, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38923822

ABSTRACT

BACKGROUND: As physical signals, mechanical cues regulate the neural cells in the brain. The mechanosensitive channels (MSCs) perceive the mechanical cues and transduce them by permeating specific ions or molecules across the plasma membrane, and finally trigger a series of intracellular bioelectrical and biochemical signals. Emerging evidence supports that wide-distributed, high-expressed MSCs like Piezo1 play important roles in several neurophysiological processes and neurological disorders. AIMS: To systematically conclude the functions of MSCs in the brain and provide a novel mechanobiological perspective for brain diseases. METHOD: We summarized the mechanical cues and MSCs detected in the brain and the research progress on the functional roles of MSCs in physiological conditions. We then concluded the pathological activation and downstream pathways triggered by MSCs in two categories of brain diseases, neurodegenerative diseases and place-occupying damages. Finally, we outlined the methods for manipulating MSCs and discussed their medical potential with some crucial outstanding issues. RESULTS: The MSCs present underlying common mechanisms in different brain diseases by acting as the "transportation hubs" to transduce the distinct signal patterns: the upstream mechanical cues and the downstream intracellular pathways. Manipulating the MSCs is feasible to alter the complicated downstream processes, providing them promising targets for clinical treatment. CONCLUSIONS: Recent research on MSCs provides a novel insight into brain diseases. The common mechanisms mediated by MSCs inspire a wide range of therapeutic potentials targeted on MSCs in different brain diseases.


Subject(s)
Brain Diseases , Ion Channels , Mechanotransduction, Cellular , Humans , Animals , Ion Channels/metabolism , Ion Channels/physiology , Brain Diseases/metabolism , Brain Diseases/physiopathology , Mechanotransduction, Cellular/physiology , Brain/metabolism
16.
ACS Appl Mater Interfaces ; 16(21): 27164-27176, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38750662

ABSTRACT

Macrophages are involved in every stage of the innate/inflammatory immune responses in the body tissues, including the resolution of the reaction, and they do so in close collaboration with the extracellular matrix (ECM). Simplified substrates with nanotopographical features attempt to mimic the structural properties of the ECM to clarify the functional features of the interaction of the ECM with macrophages. We still have a limited understanding of the macrophage behavior upon interaction with disordered nanotopography, especially with features smaller than 10 nm. Here, we combine atomic force microscopy (AFM), finite element modeling (FEM), and quantitative biochemical approaches in order to understand the mechanotransduction from the nanostructured surface into cellular responses. AFM experiments show a decrease of macrophage stiffness, measured with the Young's modulus, as a biomechanical response to a nanostructured (ns-) ZrOx surface. FEM experiments suggest that ZrOx surfaces with increasing roughness represent weaker mechanical boundary conditions. The mechanical cues from the substrate are transduced into the cell through the formation of integrin-regulated focal adhesions and cytoskeletal reorganization, which, in turn, modulate cell biomechanics by downregulating cell stiffness. Surface nanotopography and consequent biomechanical response impact the overall behavior of macrophages by increasing movement and phagocytic ability without significantly influencing their inflammatory behavior. Our study suggests a strong potential of surface nanotopography for the regulation of macrophage functions, which implies a prospective application relative to coating technology for biomedical devices.


Subject(s)
Macrophages , Surface Properties , Macrophages/cytology , Mice , Animals , Microscopy, Atomic Force , Nanostructures/chemistry , RAW 264.7 Cells , Extracellular Matrix/chemistry , Finite Element Analysis , Biomechanical Phenomena , Mechanotransduction, Cellular/physiology , Phagocytosis , Elastic Modulus
17.
Nat Methods ; 21(6): 1063-1073, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38802520

ABSTRACT

The dynamics of cellular membrane tension and its role in mechanosensing, which is the ability of cells to respond to physical stimuli, remain incompletely understood, mainly due to the lack of appropriate tools. Here, we report a force-controlled nanopipette-based method that combines fluidic force microscopy with fluorescence imaging for precise manipulation of the cellular membrane tension while monitoring the impact on single-cell mechanosensitivity. The force-controlled nanopipette enables control of the indentation force imposed on the cell cortex as well as of the aspiration pressure applied to the plasma membrane. We show that this setup can be used to concurrently monitor the activation of Piezo1 mechanosensitive ion channels via calcium imaging. Moreover, the spatiotemporal behavior of the tension propagation is assessed with the fluorescent membrane tension probe Flipper-TR, and further dissected using molecular dynamics modeling. Finally, we demonstrate that aspiration and indentation act independently on the cellular mechanobiological machinery, that indentation induces a local pre-tension in the membrane, and that membrane tension stays confined by links to the cytoskeleton.


Subject(s)
Cell Membrane , Ion Channels , Mechanotransduction, Cellular , Ion Channels/metabolism , Cell Membrane/metabolism , Mechanotransduction, Cellular/physiology , Humans , Molecular Dynamics Simulation , Calcium/metabolism , Animals
19.
Physiol Rep ; 12(9): e16043, 2024 May.
Article in English | MEDLINE | ID: mdl-38724885

ABSTRACT

The epithelial cells that line the kidneys and lower urinary tract are exposed to mechanical forces including shear stress and wall tension; however, the mechanosensors that detect and respond to these stimuli remain obscure. Candidates include the OSCA/TMEM63 family of ion channels, which can function as mechanosensors and osmosensors. Using Tmem63bHA-fl/HA-fl reporter mice, we assessed the localization of HA-tagged-TMEM63B within the urinary tract by immunofluorescence coupled with confocal microscopy. In the kidneys, HA-TMEM63B was expressed by proximal tubule epithelial cells, by the intercalated cells of the collecting duct, and by the epithelial cells lining the thick ascending limb of the medulla. In the urinary tract, HA-TMEM63B was expressed by the urothelium lining the renal pelvis, ureters, bladder, and urethra. HA-TMEM63B was also expressed in closely allied organs including the epithelial cells lining the seminal vesicles, vas deferens, and lateral prostate glands of male mice and the vaginal epithelium of female mice. Our studies reveal that TMEM63B is expressed by subsets of kidney and lower urinary tract epithelial cells, which we hypothesize are sites of TMEM63B mechanosensation or osmosensation, or both.


Subject(s)
Calcium Channels , Urinary Tract , Animals , Female , Male , Mice , Calcium Channels/genetics , Calcium Channels/metabolism , Epithelial Cells/metabolism , Mechanotransduction, Cellular/physiology , Mice, Inbred C57BL , Urinary Tract/metabolism , Urothelium/metabolism , Urothelium/cytology
20.
Trends Neurosci ; 47(7): 478-479, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38762363

ABSTRACT

Members of both the Piezo and transmembrane channel-like (TMC) families are bona fide mammalian mechanotransducers. In a recent study, Zhang, Shao et al. discovered that TMC7, a non-mechanosensitive TMC, inhibits Piezo2-dependent mechanosensation, with implications for the importance of cellular context for Piezo2 channels in normal and pathological responses to mechanical pain.


Subject(s)
Ion Channels , Mechanotransduction, Cellular , Nociceptors , Animals , Humans , Ion Channels/metabolism , Mechanotransduction, Cellular/physiology , Nociceptors/metabolism , Nociceptors/physiology , Pain/metabolism , Pain/physiopathology , Rodentia
SELECTION OF CITATIONS
SEARCH DETAIL