Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
PLoS Comput Biol ; 18(2): e1009156, 2022 02.
Article in English | MEDLINE | ID: mdl-35157694

ABSTRACT

Lymphocytes have been described to perform different motility patterns such as Brownian random walks, persistent random walks, and Lévy walks. Depending on the conditions, such as confinement or the distribution of target cells, either Brownian or Lévy walks lead to more efficient interaction with the targets. The diversity of these motility patterns may be explained by an adaptive response to the surrounding extracellular matrix (ECM). Indeed, depending on the ECM composition, lymphocytes either display a floating motility without attaching to the ECM, or sliding and stepping motility with respectively continuous or discontinuous attachment to the ECM, or pivoting behaviour with sustained attachment to the ECM. Moreover, on the long term, lymphocytes either perform a persistent random walk or a Brownian-like movement depending on the ECM composition. How the ECM affects cell motility is still incompletely understood. Here, we integrate essential mechanistic details of the lymphocyte-matrix adhesions and lymphocyte intrinsic cytoskeletal induced cell propulsion into a Cellular Potts model (CPM). We show that the combination of de novo cell-matrix adhesion formation, adhesion growth and shrinkage, adhesion rupture, and feedback of adhesions onto cell propulsion recapitulates multiple lymphocyte behaviours, for different lymphocyte subsets and various substrates. With an increasing attachment area and increased adhesion strength, the cells' speed and persistence decreases. Additionally, the model predicts random walks with short-term persistent but long-term subdiffusive properties resulting in a pivoting type of motility. For small adhesion areas, the spatial distribution of adhesions emerges as a key factor influencing cell motility. Small adhesions at the front allow for more persistent motility than larger clusters at the back, despite a similar total adhesion area. In conclusion, we present an integrated framework to simulate the effects of ECM proteins on cell-matrix adhesion dynamics. The model reveals a sufficient set of principles explaining the plasticity of lymphocyte motility.


Subject(s)
Cell-Matrix Junctions , Extracellular Matrix , Cell Adhesion/physiology , Cell Movement/physiology , Cell-Matrix Junctions/physiology , Computer Simulation , Extracellular Matrix/metabolism
2.
Nat Cell Biol ; 23(7): 745-757, 2021 07.
Article in English | MEDLINE | ID: mdl-34155382

ABSTRACT

Intestinal organoids capture essential features of the intestinal epithelium such as crypt folding, cellular compartmentalization and collective movements. Each of these processes and their coordination require patterned forces that are at present unknown. Here we map three-dimensional cellular forces in mouse intestinal organoids grown on soft hydrogels. We show that these organoids exhibit a non-monotonic stress distribution that defines mechanical and functional compartments. The stem cell compartment pushes the extracellular matrix and folds through apical constriction, whereas the transit amplifying zone pulls the extracellular matrix and elongates through basal constriction. The size of the stem cell compartment depends on the extracellular-matrix stiffness and endogenous cellular forces. Computational modelling reveals that crypt shape and force distribution rely on cell surface tensions following cortical actomyosin density. Finally, cells are pulled out of the crypt along a gradient of increasing tension. Our study unveils how patterned forces enable compartmentalization, folding and collective migration in the intestinal epithelium.


Subject(s)
Cell Movement , Epithelial Cells/physiology , Intestinal Mucosa/physiology , Mechanotransduction, Cellular , Animals , Cell Communication , Cell-Matrix Junctions/physiology , Cells, Cultured , Computer Simulation , Epithelial Cells/metabolism , Female , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Male , Mice, Transgenic , Microscopy, Confocal , Models, Biological , Organoids , Stress, Mechanical , Surface Tension , Time Factors
3.
Mol Biol Cell ; 32(5): 402-412, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33405954

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a morphogenetic process that endows epithelial cells with migratory and invasive potential. Mechanical and chemical signals from the tumor microenvironment can activate the EMT program, thereby permitting cancer cells to invade the surrounding stroma and disseminate to distant organs. Transforming growth factor ß1 (TGFß1) is a potent inducer of EMT that can also induce apoptosis depending on the microenvironmental context. In particular, stiff microenvironments promote EMT while softer ones promote apoptosis. Here, we investigated the molecular signaling downstream of matrix stiffness that regulates the phenotypic switch in response to TGFß1 and uncovered a critical role for integrin-linked kinase (ILK). Specifically, depleting ILK from mammary epithelial cells precludes their ability to sense the stiffness of their microenvironment. In response to treatment with TGFß1, ILK-depleted cells undergo apoptosis on both soft and stiff substrata. We found that knockdown of ILK decreases focal adhesions and increases cell-cell adhesions, thus shifting the balance from cell-matrix to cell-cell adhesion. High cell-matrix adhesion promotes EMT whereas high cell-cell adhesion promotes apoptosis downstream of TGFß1. These results highlight an important role for ILK in controlling cell phenotype by regulating adhesive connections to the local microenvironment.


Subject(s)
Cell Adhesion/physiology , Epithelial-Mesenchymal Transition/physiology , Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis/physiology , Cell Adhesion/drug effects , Cell Line , Cell Movement/drug effects , Cell-Matrix Junctions/physiology , Epithelial Cells/drug effects , Focal Adhesions , Mice , Protein Serine-Threonine Kinases/physiology , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology
4.
Sci Rep ; 10(1): 14124, 2020 08 24.
Article in English | MEDLINE | ID: mdl-32839490

ABSTRACT

The present study shows chronic adjustments in the myotendinous junction (MTJ) in response to different ladder-based resistance training (LRT) protocols. Thirty adult male Wistar rats were divided into groups: sedentary (S), calisthenics (LRT without additional load [C]), and resistance-trained (LRT with extra weight [R]). We demonstrated longer lengths of sarcoplasmatic invaginations in the trained groups; however, evaginations were seen mainly in group R. We showed a greater thickness of sarcoplasmatic invaginations in groups C and R, in addition to greater evaginations in R. We also observed thinner basal lamina in trained groups. The support collagen layer (SCL) adjacent to the MTJ and the diameters of the transverse fibrils were larger in R. We also discovered a niche of telocytes in the MTJ with electron micrographs of the plantar muscle and with immunostaining with CD34+ in the gastrocnemius muscle near the blood vessels and pericytes. We concluded that the continuous adjustments in the MTJ ultrastructure were the result of tissue plasticity induced by LRT, which is causally related to muscle hypertrophy and, consequently, to the remodeling of the contact interface. Also, we reveal the existence of a collagen layer adjacent to MTJ and discover a new micro anatomic location of telocytes.


Subject(s)
Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiology , Physical Conditioning, Animal/physiology , Resistance Training/methods , Sarcoplasmic Reticulum/physiology , Telocytes/physiology , Adaptation, Physiological/physiology , Adherens Junctions/physiology , Animals , Basement Membrane/physiology , Cell Adhesion , Cell Movement/physiology , Cell-Matrix Junctions/physiology , Collagen/metabolism , Male , Rats , Rats, Wistar , Sedentary Behavior
5.
Mol Biol Cell ; 31(12): 1273-1288, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32267210

ABSTRACT

Forces generated by heart muscle contraction must be balanced by adhesion to the extracellular matrix (ECM) and to other cells for proper heart function. Decades of data have suggested that cell-ECM adhesions are important for sarcomere assembly. However, the relationship between cell-ECM adhesions and sarcomeres assembling de novo remains untested. Sarcomeres arise from muscle stress fibers (MSFs) that are translocating on the top (dorsal) surface of cultured cardiomyocytes. Using an array of tools to modulate cell-ECM adhesion, we established a strong positive correlation between the extent of cell-ECM adhesion and sarcomere assembly. On the other hand, we found a strong negative correlation between the extent of cell-ECM adhesion and the rate of MSF translocation, a phenomenon also observed in nonmuscle cells. We further find a conserved network architecture that also exists in nonmuscle cells. Taken together, our results show that cell-ECM adhesions mediate coupling between the substrate and MSFs, allowing their maturation into sarcomere-containing myofibrils.


Subject(s)
Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Stress Fibers/metabolism , Actins/metabolism , Actins/physiology , Cell Culture Techniques/methods , Cell-Matrix Junctions/physiology , Extracellular Matrix/physiology , Humans , Myocytes, Cardiac/physiology , Myofibrils/physiology , Sarcomeres/physiology , Stress Fibers/physiology
6.
Biophys J ; 117(10): 1795-1804, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31706566

ABSTRACT

Cells in vivo encounter and exert forces as they interact with the extracellular matrix (ECM) and neighboring cells during migration. These mechanical forces play crucial roles in regulating cell migratory behaviors. Although a variety of studies have focused on describing single-cell or the collective cell migration behaviors, a fully mechanistic understanding of how the cell-cell (intercellular) and cell-ECM (extracellular) traction forces individually and cooperatively regulate single-cell migration and coordinate multicellular movement in a cellular monolayer is still lacking. Here, we developed an integrated experimental and analytical system to examine both the intercellular and extracellular traction forces acting on individual cells within an endothelial cell colony as well as their roles in guiding cell migratory behaviors (i.e., cell translation and rotation). Combined with force, multipole, and moment analysis, our results revealed that traction force dominates in regulating cell active translation, whereas intercellular force actively modulates cell rotation. Our findings advance the understanding of the intricacies of cell-cell and cell-ECM forces in regulating cellular migratory behaviors that occur during the monolayer development and may yield deeper insights into the single-cell dynamic behaviors during tissue development, embryogenesis, and wound healing.


Subject(s)
Cell Movement/physiology , Cell-Matrix Junctions/physiology , Animals , Biomechanical Phenomena , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Mice , Rotation , Torque
7.
Nat Commun ; 10(1): 1518, 2019 04 03.
Article in English | MEDLINE | ID: mdl-30944331

ABSTRACT

When migrating in vivo, cells are exposed to numerous conflicting signals: chemokines, repellents, extracellular matrix, growth factors. The roles of several of these molecules have been studied individually in vitro or in vivo, but we have yet to understand how cells integrate them. To start addressing this question, we used the cephalic neural crest as a model system and looked at the roles of its best examples of positive and negative signals: stromal-cell derived factor 1 (Sdf1/Cxcl12) and class3-Semaphorins. Here we show that Sdf1 and Sema3A antagonistically control cell-matrix adhesion via opposite effects on Rac1 activity at the single cell level. Directional migration at the population level emerges as a result of global Semaphorin-dependent confinement and broad activation of adhesion by Sdf1 in the context of a biased Fibronectin distribution. These results indicate that uneven in vivo topology renders the need for precise distribution of secreted signals mostly dispensable.


Subject(s)
Cell Movement/physiology , Cell-Matrix Junctions/physiology , Neural Crest/cytology , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Communication/physiology , Cell Line , Cell Shape/drug effects , Cell Surface Extensions/drug effects , Cell-Matrix Junctions/drug effects , Cell-Matrix Junctions/metabolism , Chemokine CXCL12/metabolism , Female , Fibronectins/metabolism , Male , Manganese/metabolism , Mice , Nerve Tissue Proteins/physiology , Neural Crest/drug effects , Neural Crest/metabolism , Receptors, CXCR4/metabolism , Semaphorins/metabolism , Xenopus laevis/embryology , rac1 GTP-Binding Protein/metabolism
8.
PLoS One ; 14(4): e0215122, 2019.
Article in English | MEDLINE | ID: mdl-30995271

ABSTRACT

BACKGROUND AND AIMS: The goal of the study was to assess calcium alone and Aquamin, a multi-mineral natural product that contains magnesium and detectable levels of 72 trace elements in addition to calcium, for capacity to affect growth and differentiation in colonoid cultures derived from histologically-normal human colon tissue. METHODS: Colonoid cultures were maintained in a low-calcium (0.25 mM) medium or in medium supplemented with an amount of calcium (1.5-3.0 mM), either from calcium alone or Aquamin for a period of two weeks. This was shown in a previous study to induce differentiation in colonoids derived from large adenomas. Changes in growth, morphological features and protein expression profile were assessed at the end of the incubation period using a combination of phase-contrast and scanning electron microscopy, histology and immunohistology, proteomic assessment and transmission electron microscopy. RESULTS: Unlike the previously-studied tumor-derived colonoids (which remained un-differentiated in the absence of calcium-supplementation), normal tissue colonoids underwent differentiation as indicated by gross and microscopic appearance, a low proliferative index and high-level expression of cytokeratin 20 in the absence of intervention (i.e., in control condition). Only modest additional changes were seen in these parameters with either calcium alone or Aquamin (providing up to 3.0 mM calcium). In spite of this, proteomic analysis and immunohistochemistry revealed that both interventions induced strong up-regulation of proteins that promote cell-cell and cell-matrix adhesive functions, barrier formation and tissue integrity. Transmission electron microscopy revealed an increase in desmosomes in response to intervention. CONCLUSIONS: These findings demonstrate that colonoids derived from histologically normal human tissue can undergo differentiation in the presence of a low ambient calcium concentration. However, higher calcium levels induce elaboration of proteins that promote cell-cell and cell-matrix adhesion. These changes could lead to improved barrier function and improved colon tissue health.


Subject(s)
Adenoma/pathology , Calcium/pharmacology , Cell Adhesion/drug effects , Cell Communication/drug effects , Cell Differentiation/drug effects , Cell-Matrix Junctions/physiology , Colon/cytology , Adenoma/metabolism , Cell Culture Techniques , Cell Proliferation/drug effects , Cells, Cultured , Colon/drug effects , Colon/metabolism , Humans , Minerals/pharmacology , Organoids/cytology , Organoids/metabolism , Proteome/analysis
9.
J Cell Sci ; 131(16)2018 08 17.
Article in English | MEDLINE | ID: mdl-30054383

ABSTRACT

Cell-matrix adhesion regulates membrane trafficking controlling anchorage-dependent signaling. While a dynamic Golgi complex can contribute to this pathway, its regulation by adhesion remains unclear. Here we report that loss of adhesion dramatically disorganized the Golgi in mouse and human fibroblast cells. Golgi integrity is restored rapidly upon integrin-mediated re-adhesion to FN and is disrupted by integrin blocking antibody. In suspended cells, the cis, cis-medial and trans-Golgi networks differentially disorganize along the microtubule network but show no overlap with the ER, making this disorganization distinct from known Golgi fragmentation. This pathway is regulated by an adhesion-dependent reduction and recovery of Arf1 activation. Constitutively active Arf1 disrupts this regulation and prevents Golgi disorganization due to loss of adhesion. Adhesion-dependent Arf1 activation regulates its binding to the microtubule minus-end motor protein dynein to control Golgi reorganization, which is blocked by ciliobrevin. Adhesion-dependent Golgi organization controls its function, regulating cell surface glycosylation due to loss of adhesion, which is blocked by constitutively active Arf1. This study, hence, identified integrin-dependent cell-matrix adhesion to be a novel regulator of Arf1 activation, controlling Golgi organization and function in anchorage-dependent cells. This article has an associated First Person interview with the first author of the paper.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Cell Adhesion/physiology , Cell-Matrix Junctions/physiology , Golgi Apparatus/metabolism , Golgi Apparatus/physiology , ADP-Ribosylation Factor 1/genetics , Animals , Cell Membrane/metabolism , Cells, Cultured , Embryo, Mammalian , Humans , Integrins/metabolism , Mice , Signal Transduction/physiology , trans-Golgi Network/metabolism , trans-Golgi Network/physiology
10.
Nat Cell Biol ; 20(6): 646-654, 2018 06.
Article in English | MEDLINE | ID: mdl-29802405

ABSTRACT

It has long been proposed that the cell cycle is regulated by physical forces at the cell-cell and cell-extracellular matrix (ECM) interfaces1-12. However, the evolution of these forces during the cycle has never been measured in a tissue, and whether this evolution affects cell cycle progression is unknown. Here, we quantified cell-cell tension and cell-ECM traction throughout the complete cycle of a large cell population in a growing epithelium. These measurements unveil temporal mechanical patterns that span the entire cell cycle and regulate its duration, the G1-S transition and mitotic rounding. Cells subjected to higher intercellular tension exhibit a higher probability to transition from G1 to S, as well as shorter G1 and S-G2-M phases. Moreover, we show that tension and mechanical energy are better predictors of the duration of G1 than measured geometric properties. Tension increases during the cell cycle but decreases 3 hours before mitosis. Using optogenetic control of contractility, we show that this tension drop favours mitotic rounding. Our results establish that cell cycle progression is regulated cooperatively by forces between the dividing cell and its neighbours.


Subject(s)
Cell Communication , Cell Cycle , Cell Proliferation , Cell-Matrix Junctions/physiology , Epithelial Cells/physiology , Extracellular Matrix/physiology , Mechanotransduction, Cellular , Animals , Cadherins/metabolism , Cell-Matrix Junctions/metabolism , Dogs , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Madin Darby Canine Kidney Cells , Mitosis , Stress, Mechanical , Time Factors
11.
Dev Cell ; 44(1): 87-96.e5, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29316444

ABSTRACT

Truncating mutations in the sarcomere protein titin cause dilated cardiomyopathy due to sarcomere insufficiency. However, it remains mechanistically unclear how these mutations decrease sarcomere content in cardiomyocytes. Utilizing human induced pluripotent stem cell-derived cardiomyocytes, CRISPR/Cas9, and live microscopy, we characterize the fundamental mechanisms of human cardiac sarcomere formation. We observe that sarcomerogenesis initiates at protocostameres, sites of cell-extracellular matrix adhesion, where nucleation and centripetal assembly of α-actinin-2-containing fibers provide a template for the fusion of Z-disk precursors, Z bodies, and subsequent striation. We identify that ß-cardiac myosin-titin-protocostamere form an essential mechanical connection that transmits forces required to direct α-actinin-2 centripetal fiber assembly and sarcomere formation. Titin propagates diastolic traction stresses from ß-cardiac myosin, but not α-cardiac myosin or non-muscle myosin II, to protocostameres during sarcomerogenesis. Ablating protocostameres or decoupling titin from protocostameres abolishes sarcomere assembly. Together these results identify the mechanical and molecular components critical for human cardiac sarcomerogenesis.


Subject(s)
Actinin/metabolism , Cell-Matrix Junctions/physiology , Connectin/metabolism , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Sarcomeres/physiology , Ventricular Myosins/metabolism , Actinin/genetics , Adolescent , Adult , Cells, Cultured , Connectin/genetics , Humans , Induced Pluripotent Stem Cells/physiology , Male , Middle Aged , Myocytes, Cardiac/physiology , Ventricular Myosins/genetics
12.
Bull Exp Biol Med ; 163(4): 510-514, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28853071

ABSTRACT

Multiple-day distraction regenerate contains a number of foci or zones merging into one another with different structural organization of the fibrocellular basis that reflects successive stages of reparative regeneration. At the moment of the last traction, the central part of the regenerate represents a zone of fibrogenesis (growth zone), which is common for the proximal and distal parts of the distraction regenerate. The other zones are paired and symmetrical: zones of angiogenesis, fibrous osteogenesis (primary), and osteogenesis (secondary). The latter include subzones of resorption, secondary osteogenesis, and functional remodeling.


Subject(s)
Bone Regeneration/physiology , Osteogenesis/physiology , Animals , Cell-Matrix Junctions/metabolism , Cell-Matrix Junctions/physiology , Microscopy, Atomic Force , Osteoblasts/metabolism , Osteoblasts/ultrastructure , Osteogenesis, Distraction , Sheep
13.
Biomech Model Mechanobiol ; 16(4): 1207-1224, 2017 08.
Article in English | MEDLINE | ID: mdl-28213831

ABSTRACT

Cell adhesion is crucial for cells to not only physically interact with each other but also sense their microenvironment and respond accordingly. In fact, adherent cells can generate physical forces that are transmitted to the surrounding matrix, regulating the formation of cell-matrix adhesions. The main purpose of this work is to develop a computational model to simulate the dynamics of cell-matrix adhesions through a cohesive formulation within the framework of the finite element method and based on the principles of continuum damage mechanics. This model enables the simulation of the mechanical adhesion between cell and extracellular matrix (ECM) as regulated by local multidirectional forces and thus predicts the onset and growth of the adhesion. In addition, this numerical approach allows the simulation of the cell as a whole, as it models the complete mechanical interaction between cell and ECM. As a result, we can investigate and quantify how different mechanical conditions in the cell (e.g., contractile forces, actin cytoskeletal properties) or in the ECM (e.g., stiffness, external forces) can regulate the dynamics of cell-matrix adhesions.


Subject(s)
Computer Simulation , Models, Biological , Cell Adhesion/physiology , Cell-Matrix Junctions/physiology , Cytoskeleton/metabolism , Extracellular Matrix/physiology , Humans
14.
Med. oral patol. oral cir. bucal (Internet) ; 21(5): e601-e607, sept. 2016. ilus
Article in English | IBECS | ID: ibc-155772

ABSTRACT

BACKGROUND: Leukocyte-platelet rich fibrin (L-PRF) is a second generation platelet concentrate clinically used to accelerate tissue healing and bone regeneration. Achieving reduced implant osseointegration time could provide immediate or early loading of implants. The aim of this study was to evaluate the L-PRF-induced osseointegration and bone-implant contact (BIC) in an experimental animal model. MATERIAL AND METHODS: Twelve 4-month-old New Zealand white rabbits were used. Following general anesthesia, 3-5 mL of blood was obtained from the central artery in rabbit ear and L-PRF was prepared. Two implant cavities (5 mm long and 3 mm in diameter) were created in each tibia with a total of four cavities in each animal. Two of these cavities were selected and covered with PRF (test group). The remaining L-PRF was used to soak the implants placed into the L-PRF covered sockets. Other cavities were left as controls. In total, 48 implants were placed. Animals were sacrificed after two, three, or four weeks. Histological samples were obtained and periimplant tissues were histomorphometrically evaluated for bone-to-implant contact and new bone formation. RESULTS: Histomorphometric analyses of the defects revealed that the L-PRF was detectable up to the second week. Application of L-PRF increased the rate and amount of new bone formation in the experimental group compared to the control group. Bone-to-implant contact was enhanced when the surface was prewetted with LPRF (p < 0.01). CONCLUSIONS: The results of this study demonstrated that L-PRF application may increases amount and rate of new bone formation during the early healing period and provides a faster osseointegration around implants


Subject(s)
Humans , Platelet-Rich Plasma , Fibrin Tissue Adhesive/therapeutic use , Osseointegration/physiology , Dental Implantation, Endosseous/methods , Bone Regeneration/physiology , Cell-Matrix Junctions/physiology , Intercellular Signaling Peptides and Proteins/therapeutic use
15.
PLoS One ; 11(2): e0148254, 2016.
Article in English | MEDLINE | ID: mdl-26840835

ABSTRACT

Many cell types remodel the extracellular matrix of the tissues they inhabit in response to a wide range of environmental stimuli, including mechanical cues. Such is the case in dermal wound healing, where fibroblast migrate into and remodel the provisional fibrin matrix in a complex manner that depends in part on the local mechanical environment and the evolving multi-scale mechanical interactions of the system. In this study, we report on the development of an image-based multi-scale mechanical model that predicts the short-term (24 hours), structural reorganization of a fibrin gel by fibroblasts. These predictive models are based on an in vitro experimental system where clusters of fibroblasts (i.e., explants) were spatially arranged into a triangular geometry onto the surface of fibrin gels that were subjected to either Fixed or Free in-plane mechanical constraints. Experimentally, regional differences in short-term structural remodeling and cell migration were observed for the two gel boundary conditions. A pilot experiment indicated that these small differences in the short-term remodeling of the fibrin gel translate into substantial differences in long-term (4 weeks) remodeling, particularly in terms of collagen production. The multi-scale models were able to predict some regional differences in remodeling and qualitatively similar reorganization patterns for the two boundary conditions. However, other aspects of the model, such as the magnitudes and rates of deformation of gel, did not match the experiments. These discrepancies between model and experiment provide fertile ground for challenging model assumptions and devising new experiments to enhance our understanding of how this multi-scale system functions. These efforts will ultimately improve the predictions of the remodeling process, particularly as it relates to dermal wound healing and the reduction of patient scarring. Such models could be used to recommend patient-specific mechanical-based treatment dependent on parameters such as wound geometry, location, age, and health.


Subject(s)
Cell-Matrix Junctions/physiology , Cicatrix/metabolism , Extracellular Matrix/physiology , Skin Physiological Phenomena , Wound Healing/physiology , Cell Communication , Cell Movement , Cells, Cultured , Collagen/metabolism , Computer Simulation , Fibrin/metabolism , Fibroblasts/physiology , Gels/metabolism , Microspheres , Skin/injuries
17.
Exp Cell Res ; 343(1): 60-66, 2016 04 10.
Article in English | MEDLINE | ID: mdl-26524505

ABSTRACT

The extracellular matrix (ECM) microenvironment plays a central role in cell migration by providing physiochemical information that influences overall cell behavior. Much of this external information is accessed by direct interaction of the cell with ECM ligands and structures via integrin-based adhesions that are hypothesized to act as mechanosensors for testing the surrounding microenvironment. Our current understanding of these mechanical complexes is derived primarily from studies of cellular adhesions formed on two-dimensional (2D) substrates in vitro. Yet the rules of cell/ECM engagement and mechanosensing in three-dimensional (3D) microenvironments are invariably more complex under both in vitro and in vivo conditions. Here we review the current understanding of how cellular mechanosensing occurs through adhesion complexes within 3D microenvironments and discuss how these mechanisms can vary and differ from interactions on 2D substrates.


Subject(s)
Cell Adhesion/physiology , Cell-Matrix Junctions/physiology , Mechanotransduction, Cellular , Animals , Collagen Type I/chemistry , Collagen Type I/metabolism , Humans , Molecular Conformation
18.
J Theor Biol ; 384: 84-94, 2015 Nov 07.
Article in English | MEDLINE | ID: mdl-26235289

ABSTRACT

Cell-matrix adhesions are crucial in different biological processes like tissue morphogenesis, cell motility, and extracellular matrix remodeling. These interactions that link cell cytoskeleton and matrix fibers are built through protein clutches, generally known as adhesion complexes. The adhesion formation process has been deeply studied in two-dimensional (2D) cases; however, the knowledge is limited for three-dimensional (3D) cases. In this work, we simulate different local extracellular matrix properties in order to unravel the fundamental mechanisms that regulate the formation of cell-matrix adhesions in 3D. We aim to study the mechanical interaction of these biological structures through a three dimensional discrete approach, reproducing the transmission pattern force between the cytoskeleton and a single extracellular matrix fiber. This numerical model provides a discrete analysis of the proteins involved including spatial distribution, interaction between them, and study of the different phenomena, such as protein clutches unbinding or protein unfolding.


Subject(s)
Cell-Matrix Junctions/physiology , Cytoskeleton/physiology , Extracellular Matrix/physiology , Models, Biological , Actin Cytoskeleton/physiology , Animals , Myosins/physiology , Protein Refolding , Protein Unfolding
19.
Eur J Dermatol ; 25 Suppl 1: 4-11, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26287030

ABSTRACT

We review the functions of peroxisome proliferator activated receptor (PPAR) ß/δ in skin wound healing and cancer. In particular, we highlight the roles of PPARß/δ in inhibiting keratinocyte apoptosis at wound edges via activation of the PI3K/PKBα/Akt1 pathway and its role during re-epithelialization in regulating keratinocyte adhesion and migration. In fibroblasts, PPARß/δ controls IL-1 signalling and thereby contributes to the homeostatic control of keratinocyte proliferation. We discuss its therapeutic potential for treating diabetic wounds and inflammatory skin diseases such as psoriasis and acne vulgaris. PPARß/δ is classified as a tumour growth modifier; it is activated by chronic low-grade inflammation, which promotes the production of lipids that, in turn, enhance PPARß/δ transcription activity. Our earlier work unveiled a cascade of events triggered by PPARß/δ that involve the oncogene Src, which promotes ultraviolet-induced skin cancer in mice via enhanced EGFR/Erk1/2 signalling and the expression of epithelial-to-mesenchymal transition (EMT) markers. Interestingly, PPARß/δ expression is correlated with the expression of SRC and EMT markers in human skin squamous cell carcinoma. Furthermore, there is a positive interaction between PPARß/δ, SRC, and TGFß1 at the transcriptional level in various human epithelial cancers. Taken together, these observations suggest the need for evaluating PPARß/δ modulators that attenuate or increase its activity, depending on the therapeutic target.


Subject(s)
PPAR delta/physiology , Skin Neoplasms/physiopathology , Wound Healing/physiology , Animals , Cell Communication , Cell-Matrix Junctions/physiology , Humans , Keratinocytes/physiology , PPAR-beta/physiology
20.
Mol Biol Cell ; 26(13): 2456-65, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25971797

ABSTRACT

Mechanical linkage between cell-cell and cell-extracellular matrix (ECM) adhesions regulates cell shape changes during embryonic development and tissue homoeostasis. We examined how the force balance between cell-cell and cell-ECM adhesions changes with cell spread area and aspect ratio in pairs of MDCK cells. We used ECM micropatterning to drive different cytoskeleton strain energy states and cell-generated traction forces and used a Förster resonance energy transfer tension biosensor to ask whether changes in forces across cell-cell junctions correlated with E-cadherin molecular tension. We found that continuous peripheral ECM adhesions resulted in increased cell-cell and cell-ECM forces with increasing spread area. In contrast, confining ECM adhesions to the distal ends of cell-cell pairs resulted in shorter junction lengths and constant cell-cell forces. Of interest, each cell within a cell pair generated higher strain energies than isolated single cells of the same spread area. Surprisingly, E-cadherin molecular tension remained constant regardless of changes in cell-cell forces and was evenly distributed along cell-cell junctions independent of cell spread area and total traction forces. Taken together, our results showed that cell pairs maintained constant E-cadherin molecular tension and regulated total forces relative to cell spread area and shape but independently of total focal adhesion area.


Subject(s)
Cadherins/metabolism , Cell Adhesion/physiology , Cell-Matrix Junctions/physiology , Extracellular Matrix/metabolism , Animals , Biomechanical Phenomena , Cell Shape/physiology , Dogs , Madin Darby Canine Kidney Cells , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL