Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Math Biosci ; 370: 109158, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38373479

ABSTRACT

Fibroblasts in a confluent monolayer are known to adopt elongated morphologies in which cells are oriented parallel to their neighbors. We collected and analyzed new microscopy movies to show that confluent fibroblasts are motile and that neighboring cells often move in anti-parallel directions in a collective motion phenomenon we refer to as "fluidization" of the cell population. We used machine learning to perform cell tracking for each movie and then leveraged topological data analysis (TDA) to show that time-varying point-clouds generated by the tracks contain significant topological information content that is driven by fluidization, i.e., the anti-parallel movement of individual neighboring cells and neighboring groups of cells over long distances. We then utilized the TDA summaries extracted from each movie to perform Bayesian parameter estimation for the D'Orsgona model, an agent-based model (ABM) known to produce a wide array of different patterns, including patterns that are qualitatively similar to fluidization. Although the D'Orsgona ABM is a phenomenological model that only describes inter-cellular attraction and repulsion, the estimated region of D'Orsogna model parameter space was consistent across all movies, suggesting that a specific level of inter-cellular repulsion force at close range may be a mechanism that helps drive fluidization patterns in confluent mesenchymal cell populations.


Subject(s)
Movement , Systems Analysis , Bayes Theorem , Cell Movement
2.
Chembiochem ; 25(1): e202300551, 2024 01 02.
Article in English | MEDLINE | ID: mdl-37856284

ABSTRACT

Stress-activated signaling pathways orchestrate cellular behaviors and fates. Studying the precise role(s) of stress-activated protein kinases is challenging, because stress conditions induce adaptation and impose selection pressure. To meet this challenge, we have applied an optogenetic system with a single plasmid to express light-activated p38α or its upstream activator, MKK6, in conjunction with live-cell fluorescence microscopy. In starved cells, decaging of constitutively active p38α or MKK6 by brief exposure to UV light elicits rapid p38-mediated signaling, release of cytochrome c from mitochondria, and apoptosis with different kinetics. In parallel, light activation of p38α also suppresses autophagosome formation, similarly to stimulation with growth factors that activate PI3K/Akt/mTORC1 signaling. Active MKK6 negatively regulates serum-induced ERK activity, which is p38-independent as previously reported. Here, we reproduce that result with the one plasmid system and show that although decaging active p38α does not reduce basal ERK activity in our cells, it can block growth factor-stimulated ERK signaling in serum-starved cells. These results clarify the roles of MKK6 and p38α in dynamic signaling programs, which act in concert to actuate apoptotic death while suppressing cell survival mechanisms.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases , Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases , p38 Mitogen-Activated Protein Kinases , MAP Kinase Kinase 6/genetics
3.
RSC Chem Biol ; 4(10): 765-773, 2023 Oct 04.
Article in English | MEDLINE | ID: mdl-37799579

ABSTRACT

Evolution has diversified the mammalian proteome by the generation of protein isoforms that originate from identical genes, e.g., through alternative gene splicing or post-translational modifications, or very similar genes found in gene families. Protein isoforms can have either overlapping or unique functions and traditional chemical, biochemical, and genetic techniques are often limited in their ability to differentiate between isoforms due to their high similarity. This is particularly true in the context of highly dynamic cell signaling cascades, which often require acute spatiotemporal perturbation to assess mechanistic details. To that end, we describe a method for the selective perturbation of the individual protein isoforms of the mitogen-activated protein kinase (MAPK) p38. The genetic installation of a photocaging group at a conserved active site lysine enables the precise light-controlled initiation of kinase signaling, followed by investigation of downstream events. Through optical control, we have identified a novel point of crosstalk between two major signaling cascades: the p38/MAPK pathway and the extracellular signal-regulated kinase (ERK)/MAPK pathway. Specifically, using the photoactivated p38 isoforms, we have found the p38γ and p38δ variants to be positive regulators of the ERK signaling cascade, while confirming the p38α and p38ß variants as negative regulators.

4.
Biophys J ; 122(18): 3816-3829, 2023 09 19.
Article in English | MEDLINE | ID: mdl-37644720

ABSTRACT

To generate forces that drive migration of a eukaryotic cell, arrays of actin filaments (F-actin) are assembled at the cell's leading membrane edge. To maintain cell propulsion and respond to dynamic external cues, actin filaments must be disassembled to regenerate the actin monomers (G-actin), and transport of G-actin from sites of disassembly back to the leading edge completes the treadmilling cycle and limits the flux of F-actin assembly. Whether or not molecular diffusion is sufficient for G-actin transport has been a long-standing topic of debate, in part because the dynamic nature of cell motility and migration hinders the estimation of transport parameters. In this work, we applied an experimental system in which cells adopt an approximately constant and symmetrical shape; they cannot migrate but exhibit fast, steady treadmilling in the thin region protruding from the cell. Using fluorescence recovery after photobleaching, we quantified the relative concentrations and corresponding fluxes of F- and G-actin in this system. In conjunction with mathematical modeling, constrained by measured features of each region of interest, this approach revealed that diffusion alone cannot account for the transport of G-actin to the leading edge. Although G-actin diffusion and vectorial transport might vary with position in the protruding region, good agreement with the fluorescence recovery after photobleaching measurements was achieved by a model with constant G-actin diffusivity ∼2 µm2/s and anterograde G-actin velocity less than 1 µm/s.


Subject(s)
Actin Cytoskeleton , Actins , Cell Movement , Diffusion , Fluorescence
5.
J Theor Biol ; 568: 111506, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37094713

ABSTRACT

Proper wound healing relies on invasion of fibroblasts via directed migration. While the related experimental and mathematical modeling literature has mainly focused on cell migration directed by soluble cues (chemotaxis), there is ample evidence that fibroblast migration is also directed by insoluble, matrix-bound cues (haptotaxis). Furthermore, numerous studies indicate that fibronectin (FN), a haptotactic ligand for fibroblasts, is present and dynamic in the provisional matrix throughout the proliferative phase of wound healing. In the present work, we show the plausibility of a hypothesis that fibroblasts themselves form and maintain haptotactic gradients in a semi-autonomous fashion. As a precursor to this, we examine the positive control scenario where FN is pre-deposited in the wound matrix, and fibroblasts maintain haptotaxis by removing FN at an appropriate rate. After developing conceptual and quantitative understanding of this scenario, we consider two cases in which fibroblasts activate the latent form of a matrix-loaded cytokine, TGFß, which upregulates the fibroblasts' own secretion of FN. In the first of these, the latent cytokine is pre-patterned and released by the fibroblasts. In the second, fibroblasts in the wound produce the latent TGFß, with the presence of the wound providing the only instruction. In all cases, wound invasion is more effective than a negative control model with haptotaxis disabled; however, there is a trade-off between the degree of fibroblast autonomy and the rate of invasion.


Subject(s)
Chemotaxis , Cues , Cell Movement/physiology , Chemotaxis/physiology , Wound Healing/physiology , Fibroblasts , Cytokines
6.
Mol Biol Cell ; 34(6): ar60, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36884295

ABSTRACT

The extracellular signal-regulated kinase (ERK) signaling pathway plays prominent roles in cell growth, proliferation, and differentiation. ERK signaling is dynamic, involving phosphorylation/dephosphorylation, nucleocytoplasmic shuttling, and interactions with scores of protein substrates in the cytosol and in the nucleus. Live-cell fluorescence microscopy using genetically encoded ERK biosensors offers the potential to infer those dynamics in individual cells. In this study, we have monitored ERK signaling using four commonly used translocation- and Förster resonance energy transfer-based biosensors in a common cell stimulation context. Consistent with previous reports, we found that each biosensor responds with unique kinetics; it is clear that there is not a single dynamic signature characterizing the complexity of ERK phosphorylation, translocation, and kinase activity. In particular, the widely adopted ERK Kinase Translocation Reporter (ERKKTR) gives a readout that reflects ERK activity in both compartments. Mathematical modeling offers an interpretation of the measured ERKKTR kinetics, in relation to cytosolic and nuclear ERK activity, and suggests that biosensor-specific dynamics substantially influence the measured output.


Subject(s)
Biosensing Techniques , Extracellular Signal-Regulated MAP Kinases , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Phosphorylation , Signal Transduction
7.
J Biol Chem ; 298(5): 101886, 2022 05.
Article in English | MEDLINE | ID: mdl-35367415

ABSTRACT

Phospholipase C-γ1 (PLC-γ1) is a receptor-proximal enzyme that promotes signal transduction through PKC in mammalian cells. Because of the complexity of PLC-γ1 regulation, a two-state (inactive/active) model does not account for the intricacy of activation and inactivation steps at the plasma membrane. Here, we introduce a structure-based kinetic model of PLC-γ1, considering interactions of its regulatory Src homology 2 (SH2) domains and perturbation of those dynamics upon phosphorylation of Tyr783, a hallmark of activation. For PLC-γ1 phosphorylation to dramatically enhance enzyme activation as observed, we found that high intramolecular affinity of the C-terminal SH2 (cSH2) domain-pTyr783 interaction is critical, but this affinity need not outcompete the autoinhibitory interaction of the cSH2 domain. Under conditions for which steady-state PLC-γ1 activity is sensitive to the rate of Tyr783 phosphorylation, maintenance of the active state is surprisingly insensitive to the phosphorylation rate, since pTyr783 is well protected by the cSH2 domain while the enzyme is active. In contrast, maintenance of enzyme activity is sensitive to the rate of PLC-γ1 membrane (re)binding. Accordingly, we found that hypothetical PLC-γ1 mutations that either weaken autoinhibition or strengthen membrane binding influence the activation kinetics differently, which could inform the characterization of oncogenic variants. Finally, we used this newly informed kinetic scheme to refine a spatial model of PLC/PKC polarization during chemotaxis. The refined model showed improved stability of the polarized pattern while corroborating previous qualitative predictions. As demonstrated here for PLC-γ1, this approach may be adapted to model the dynamics of other receptor- and membrane-proximal enzymes.


Subject(s)
Isoenzymes , Type C Phospholipases , Animals , Carrier Proteins/metabolism , Isoenzymes/metabolism , Kinetics , Mammals/metabolism , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Type C Phospholipases/metabolism , src Homology Domains/genetics
8.
Biophys J ; 121(1): 102-118, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34861242

ABSTRACT

Orchestration of cell migration is essential for development, tissue regeneration, and the immune response. This dynamic process integrates adhesion, signaling, and cytoskeletal subprocesses across spatial and temporal scales. In mesenchymal cells, adhesion complexes bound to extracellular matrix mediate both biochemical signal transduction and physical interaction with the F-actin cytoskeleton. Here, we present a mathematical model that offers insight into both aspects, considering spatiotemporal dynamics of nascent adhesions, active signaling molecules, mechanical clutching, actin treadmilling, and nonmuscle myosin II contractility. At the core of the model is a positive feedback loop, whereby adhesion-based signaling promotes generation of barbed ends at, and protrusion of, the cell's leading edge, which in turn promotes formation and stabilization of nascent adhesions. The model predicts a switch-like transition and optimality of membrane protrusion, determined by the balance of actin polymerization and retrograde flow, with respect to extracellular matrix density. The model, together with new experimental measurements, explains how protrusion can be modulated by mechanical effects (nonmuscle myosin II contractility and adhesive bond stiffness) and F-actin turnover.


Subject(s)
Actins , Myosin Type II , Actin Cytoskeleton/metabolism , Actins/metabolism , Cell Surface Extensions , Myosin Type II/metabolism , Signal Transduction
9.
Biomicrofluidics ; 15(4): 044101, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34290842

ABSTRACT

Microfluidics approaches have gained popularity in the field of directed cell migration, enabling control of the extracellular environment and integration with live-cell microscopy; however, technical hurdles remain. Among the challenges are the stability and predictability of the environment, which are especially critical for the observation of fibroblasts and other slow-moving cells. Such experiments require several hours and are typically plagued by the introduction of bubbles and other disturbances that naturally arise in standard microfluidics protocols. Here, we report on the development of a passive pumping strategy, driven by the high capillary pressure and evaporative capacity of paper, and its application to study fibroblast chemotaxis. The paper pumps-flowvers (flow + clover)-are inexpensive, compact, and scalable, and they allow nearly bubble-free operation, with a predictable volumetric flow rate on the order of µl/min, for several hours. To demonstrate the utility of this approach, we combined the flowver pumping strategy with a Y-junction microfluidic device to generate a chemoattractant gradient landscape that is both stable (6+ h) and predictable (by finite-element modeling calculations). Integrated with fluorescence microscopy, we were able to recapitulate previous, live-cell imaging studies of fibroblast chemotaxis to platelet derived growth factor (PDGF), with an order-of-magnitude gain in throughput. The increased throughput of single-cell analysis allowed us to more precisely define PDGF gradient conditions conducive for chemotaxis; we were also able to interpret how the orientation of signaling through the phosphoinositide 3-kinase pathway affects the cells' sensing of and response to conducive gradients.

10.
J Biol Chem ; 295(25): 8494-8504, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32371393

ABSTRACT

The selective pressure imposed by extrinsic death signals and stressors adds to the challenge of isolating and interpreting the roles of proteins in stress-activated signaling networks. By expressing a kinase with activating mutations and a caged lysine blocking the active site, we can rapidly switch on catalytic activity with light and monitor the ensuing dynamics. Applying this approach to MAP kinase 6 (MKK6), which activates the p38 subfamily of MAPKs, we found that decaging active MKK6 in fibroblasts is sufficient to trigger apoptosis in a p38-dependent manner. Both in fibroblasts and in a murine melanoma cell line expressing mutant B-Raf, MKK6 activation rapidly and potently inhibited the pro-proliferative extracellular signal-regulated kinase (ERK) pathway; to our surprise, this negative cross-regulation was equally robust when all p38 isoforms were inhibited. These results position MKK6 as a new pleiotropic signal transducer that promotes both pro-apoptotic and anti-proliferative signaling, and they highlight the utility of caged, light-activated kinases for dissecting stress-activated signaling networks.


Subject(s)
MAP Kinase Kinase 6/metabolism , Signal Transduction/radiation effects , Ultraviolet Rays , Animals , Apoptosis/radiation effects , Cell Line , Cell Proliferation/radiation effects , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , MAP Kinase Kinase 6/genetics , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutagenesis , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
11.
PLoS Comput Biol ; 16(4): e1007708, 2020 04.
Article in English | MEDLINE | ID: mdl-32255775

ABSTRACT

Chemotaxis of fibroblasts and other mesenchymal cells is critical for embryonic development and wound healing. Fibroblast chemotaxis directed by a gradient of platelet-derived growth factor (PDGF) requires signaling through the phospholipase C (PLC)/protein kinase C (PKC) pathway. Diacylglycerol (DAG), the lipid product of PLC that activates conventional PKCs, is focally enriched at the up-gradient leading edge of fibroblasts responding to a shallow gradient of PDGF, signifying polarization. To explain the underlying mechanisms, we formulated reaction-diffusion models including as many as three putative feedback loops based on known biochemistry. These include the previously analyzed mechanism of substrate-buffering by myristoylated alanine-rich C kinase substrate (MARCKS) and two newly considered feedback loops involving the lipid, phosphatidic acid (PA). DAG kinases and phospholipase D, the enzymes that produce PA, are identified as key regulators in the models. Paradoxically, increasing DAG kinase activity can enhance the robustness of DAG/active PKC polarization with respect to chemoattractant concentration while decreasing their whole-cell levels. Finally, in simulations of wound invasion, efficient collective migration is achieved with thresholds for chemotaxis matching those of polarization in the reaction-diffusion models. This multi-scale modeling framework offers testable predictions to guide further study of signal transduction and cell behavior that affect mesenchymal chemotaxis.


Subject(s)
Phosphatidic Acids/metabolism , Protein Kinase C/metabolism , Type C Phospholipases/metabolism , Animals , Chemotaxis/physiology , Diglycerides/metabolism , Fibroblasts/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Models, Theoretical , Myristoylated Alanine-Rich C Kinase Substrate/metabolism , Phosphatidic Acids/physiology , Phospholipase D/metabolism , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Protein Kinase C/physiology , Signal Transduction/physiology , Type C Phospholipases/physiology
12.
Integr Biol (Camb) ; 11(6): 280-292, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31365063

ABSTRACT

We used particle-based computer simulations to study the emergent properties of the actomyosin cytoskeleton. Our model accounted for biophysical interactions between filamentous actin and non-muscle myosin II and was motivated by recent experiments demonstrating that spatial regulation of myosin activity is required for fibroblasts responding to spatial gradients of platelet derived growth factor (PDGF) to undergo chemotaxis. Our simulations revealed the spontaneous formation of actin asters, consistent with the punctate actin structures observed in chemotacting fibroblasts. We performed a systematic analysis of model parameters to identify biochemical steps in myosin activity that significantly affect aster formation and performed simulations in which model parameter values vary spatially to investigate how the model responds to chemical gradients. Interestingly, spatial variations in motor stiffness generated time-dependent behavior of the actomyosin network, in which actin asters continued to spontaneously form and dissociate in different regions of the gradient. Our results should serve as a guide for future experimental investigations.


Subject(s)
Actin Cytoskeleton/metabolism , Actomyosin/physiology , Chemotaxis , Computer Simulation , Fibroblasts/cytology , Animals , Cell Movement , Humans , Models, Biological , Myosin Type II/metabolism , Pattern Recognition, Automated , Platelet-Derived Growth Factor/metabolism , Signal Transduction
13.
Sci Signal ; 12(584)2019 06 04.
Article in English | MEDLINE | ID: mdl-31164479

ABSTRACT

Live-cell fluorescence microscopy is broadly applied to study the dynamics of receptor-mediated cell signaling, but the availability of intracellular biosensors is limited. A biosensor based on the tandem SH2 domains from phospholipase C-γ1 (PLCγ1), tSH2-WT, has been used to measure phosphorylation of the epidermal growth factor receptor (EGFR). Here, we found that tSH2-WT lacked specificity for phosphorylated EGFR, consistent with the known promiscuity of SH2 domains. Further, EGF-stimulated membrane recruitment of tSH2-WT differed qualitatively from the expected kinetics of EGFR phosphorylation. Analysis of a mathematical model suggested, and experiments confirmed, that the high avidity of tSH2-WT resulted in saturation of its target and interference with EGFR endocytosis. To overcome the apparent target specificity and saturation issues, we implemented two protein engineering strategies. In the first approach, we screened a combinatorial library generated by random mutagenesis of the C-terminal SH2 domain (cSH2) of PLCγ1 and isolated a mutant form (mSH2) with enhanced specificity for phosphorylated Tyr992 (pTyr992) of EGFR. A biosensor based on mSH2 closely reported the kinetics of EGFR phosphorylation but retained cross-reactivity similar to tSH2-WT. In the second approach, we isolated a pTyr992-binding protein (SPY992) from a combinatorial library generated by mutagenesis of the Sso7d protein scaffold. Compared to tSH2-WT and mSH2, SPY992 exhibited superior performance as a specific, moderate-affinity biosensor. We extended this approach to isolate a biosensor for EGFR pTyr1148 (SPY1148). This approach of integrating theoretical considerations with protein engineering strategies can be generalized to design and evaluate suitable biosensors for various phospho-specific targets.


Subject(s)
Biosensing Techniques , ErbB Receptors/metabolism , Animals , Cell Line , Endocytosis , ErbB Receptors/genetics , Mice , Microscopy, Fluorescence , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Phosphorylation/genetics , src Homology Domains
14.
Biophys J ; 113(1): 185-194, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-28700916

ABSTRACT

During the proliferative phase of cutaneous wound healing, dermal fibroblasts are recruited into the clotted wound by a concentration gradient of platelet-derived growth factor (PDGF), together with other spatial cues. Despite the importance of this chemotactic process, the mechanisms controlling the directed migration of slow-moving mesenchymal cells such as fibroblasts are not well understood. Here, we develop and analyze a reaction-diffusion model of phospholipase C/protein kinase C (PKC) signaling, which was recently identified as a requisite PDGF-gradient-sensing pathway, with the goal of identifying mechanisms that can amplify its sensitivity in the shallow external gradients typical of chemotaxis experiments. We show that phosphorylation of myristoylated alanine-rich C kinase substrate by membrane-localized PKC constitutes a positive feedback that is sufficient for local pathway amplification. The release of phosphorylated myristoylated alanine-rich C kinase substrate and its subsequent diffusion and dephosphorylation in the cytosol also serves to suppress the pathway in down-gradient regions of the cell. By itself, this mechanism only weakly amplifies signaling in a shallow PDGF gradient, but it synergizes with other feedback mechanisms to enhance amplification. This model offers a framework for a mechanistic understanding of phospholipase C/PKC signaling in chemotactic gradient sensing and can guide the design of experiments to assess the roles of putative feedback loops.


Subject(s)
Chemotaxis/physiology , Fibroblasts/enzymology , Models, Biological , Protein Kinase C/metabolism , Type C Phospholipases/metabolism , Animals , Cell Membrane/metabolism , Diffusion , Feedback, Physiological/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Myristoylated Alanine-Rich C Kinase Substrate , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Signal Transduction/physiology
15.
J Biol Chem ; 292(7): 2866-2872, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28069808

ABSTRACT

Mechanistic target of rapamycin complex 1 (mTORC1) controls biosynthesis and has been implicated in uncontrolled cell growth in cancer. Although many details of mTORC1 regulation are well understood, a systems-level, predictive framework synthesizing those details is currently lacking. We constructed various mathematical models of mTORC1 activation mediated by Akt and aligned the model outputs to kinetic data acquired for growth factor-stimulated cells. A model based on a putative feedforward loop orchestrated by Akt consistently predicted how the pathway was altered by depletion of key regulatory proteins. Analysis of the successful model also elucidates two dynamical motifs: neutralization of a negative regulator, which characterizes how Akt indirectly activates mTORC1, and seesaw enzyme regulation, which describes how activated and inhibited states of mTORC1 are controlled in concert to produce a nonlinear, ultrasensitive response. Such insights lend quantitative understanding of signaling networks and their precise manipulation in various contexts.


Subject(s)
Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Kinetics , Mechanistic Target of Rapamycin Complex 1 , Mice , Models, Biological , NIH 3T3 Cells
16.
Biophys J ; 111(9): 1827-1830, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27712827

ABSTRACT

Filopodia are thin, fingerlike structures that contain bundled actin filaments and project from the cell periphery. These structures are dogmatically endowed with the ability to sense cues in the microenvironment, implying that filopodia foster local signal transduction, yet their small diameter hampers the imaging of dynamic processes therein. To overcome this challenge, we analyzed total internal reflection fluorescence images of migrating fibroblasts coexpressing either a plasma membrane marker or tagged AktPH domain, a translocation biosensor for signaling through the phosphoinositide 3-kinase pathway, along with a cytosolic volume marker. We devised a scheme to estimate the radii of filopodia using either the membrane marker or volume marker data, and we used that information to account for geometry effects in the biosensor data. With conservative estimates of relative target molecule abundance, it is revealed that filopodia typically harbor higher densities of 3' phosphoinositides than adjacent regions at the cell periphery. In this context at least, the analysis supports the filopodial signaling hypothesis.


Subject(s)
Cell Movement , Pseudopodia/metabolism , Signal Transduction , Animals , Mice , NIH 3T3 Cells , Optical Imaging
17.
Integr Biol (Camb) ; 8(8): 894-903, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27477203

ABSTRACT

B-lymphocyte migration, directed by chemokine gradients, is essential for homing to sites of antigen presentation. B cells move rapidly, exhibiting amoeboid morphology like other leukocytes, yet quantitative studies addressing B-cell migration are currently lacking relative to neutrophils, macrophages, and T cells. Here, we used total internal reflection fluorescence (TIRF) microscopy to characterize the changes in shape (morphodynamics) of primary, murine B cells as they migrated on surfaces with adsorbed chemokine, CXCL13, and the adhesive ligand, ICAM-1. B cells exhibited frequent, spontaneous dilation and shrinking events at the sides of the leading membrane edge, a phenomenon that was predictive of turning versus directional persistence. To characterize directed B-cell migration, a microfluidic device was implemented to generate gradients of adsorbed CXCL13 gradients. Haptotaxis assays revealed a modest yet consistently positive bias of the cell's persistent random walk behavior towards CXCL13 gradients. Quantification of tactic fidelity showed that bias is optimized by steeper gradients without excessive midpoint density of adsorbed chemokine. Under these conditions, B-cell migration is more persistent when the direction of migration is better aligned with the gradient.


Subject(s)
B-Lymphocytes/cytology , Chemokine CXCL13/metabolism , Adsorption , Animals , Cell Movement , Computational Biology , Computer Simulation , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred C57BL , Microfluidics , Microscopy, Fluorescence , Phenotype
18.
J Cell Sci ; 129(12): 2329-42, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27173494

ABSTRACT

Haptotaxis is the process by which cells respond to gradients of substrate-bound cues, such as extracellular matrix proteins (ECM); however, the cellular mechanism of this response remains poorly understood and has mainly been studied by comparing cell behavior on uniform ECMs with different concentrations of components. To study haptotaxis in response to gradients, we utilized microfluidic chambers to generate gradients of the ECM protein fibronectin, and imaged the cell migration response. Lamellipodia are fan-shaped protrusions that are common in migrating cells. Here, we define a new function for lamellipodia and the cellular mechanism required for haptotaxis - differential actin and lamellipodial protrusion dynamics lead to biased cell migration. Modest differences in lamellipodial dynamics occurring over time periods of seconds to minutes are summed over hours to produce differential whole cell movement towards higher concentrations of fibronectin. We identify a specific subset of lamellipodia regulators as being crucial for haptotaxis. Numerous studies have linked components of this pathway to cancer metastasis and, consistent with this, we find that expression of the oncogenic Rac1 P29S mutation abrogates haptotaxis. Finally, we show that haptotaxis also operates through this pathway in 3D environments.


Subject(s)
Chemotaxis , Fibronectins/pharmacology , Pseudopodia/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Animals , Chemotaxis/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Integrin beta1/metabolism , Mice , Models, Biological , Signal Transduction/drug effects , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , Wiskott-Aldrich Syndrome Protein/metabolism , Wiskott-Aldrich Syndrome Protein Family/metabolism , rac GTP-Binding Proteins/metabolism , src-Family Kinases/metabolism
19.
J Cell Biol ; 209(6): 803-12, 2015 Jun 22.
Article in English | MEDLINE | ID: mdl-26101216

ABSTRACT

The lamellipodium is an important structure for cell migration containing branched actin nucleated via the Arp2/3 complex. The formation of branched actin is relatively well studied, but less is known about its disassembly and how this influences migration. GMF is implicated in both Arp2/3 debranching and inhibition of Arp2/3 activation. Modulation of GMFß, a ubiquitous GMF isoform, by depletion or overexpression resulted in changes in lamellipodial dynamics, branched actin content, and migration. Acute pharmacological inhibition of Arp2/3 by CK-666, coupled to quantitative live-cell imaging of the complex, showed that depletion of GMFß decreased the rate of branched actin disassembly. These data, along with mutagenesis studies, suggest that debranching (not inhibition of Arp2/3 activation) is a primary activity of GMFß in vivo. Furthermore, depletion or overexpression of GMFß disrupted the ability of cells to directionally migrate to a gradient of fibronectin (haptotaxis). These data suggest that debranching by GMFß plays an important role in branched actin regulation, lamellipodial dynamics, and directional migration.


Subject(s)
Actins/biosynthesis , Cell Movement/physiology , Glia Maturation Factor/physiology , Pseudopodia/physiology , Actin-Related Protein 2-3 Complex/antagonists & inhibitors , Actin-Related Protein 2-3 Complex/metabolism , Animals , Cell Line , Enzyme Activation , Fibroblasts/physiology , Fibronectins/pharmacology , Indoles/pharmacology , Mice , Protein Isoforms/biosynthesis
20.
J R Soc Interface ; 12(106)2015 May 06.
Article in English | MEDLINE | ID: mdl-25904526

ABSTRACT

T cells play a central role in the adaptive immune response, and their directed migration is essential for homing to sites of antigen presentation. Like neutrophils, T lymphocytes are rapidly moving cells that exhibit amoeboid movement, characterized by a definitive polarity with F-actin concentrated at the front and myosin II elsewhere. In this study, we used total internal reflection fluorescence (TIRF) microscopy to monitor the cells' areas of contact with a surface presenting adhesive ICAM-1 and the chemokine, CXCL12/SDF-1. Our analysis reveals that T-cell migration and reorientation are achieved by bifurcation and lateral separation of protrusions along the leading membrane edge, followed by cessation of one of the protrusions, which acts as a pivot for cell turning. We show that the distribution of bifurcation frequencies exhibits characteristics of a random, spontaneous process; yet, the waiting time between bifurcation events depends on whether or not the pivot point remains on the same side of the migration axis. Our analysis further suggests that switching of the dominant protrusion between the two sides of the migration axis is associated with persistent migration, whereas the opposite is true of cell turning. To help explain the bifurcation phenomenon and how distinct migration behaviours might arise, a spatio-temporal, stochastic model describing F-actin dynamics is offered.


Subject(s)
Actins/physiology , Cell Movement/physiology , Membrane Fluidity/physiology , Models, Biological , Molecular Motor Proteins/physiology , T-Lymphocytes/physiology , Animals , Cell Polarity/physiology , Cell Size , Cells, Cultured , Computer Simulation , Humans , Mice , Models, Statistical , Stochastic Processes , T-Lymphocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...