Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Front Physiol ; 14: 1130925, 2023.
Article in English | MEDLINE | ID: mdl-37334053

ABSTRACT

Intrathecal administration is an important mode for delivering biological agents targeting central nervous system (CNS) diseases. However, current clinical practices lack a sound theorical basis for a quantitative understanding of the variables and conditions that govern the delivery efficiency and specific tissue targeting especially in the brain. This work presents a distributed mechanistic pharmacokinetic model (DMPK) for predictive analysis of intrathecal drug delivery to CNS. The proposed DMPK model captures the spatiotemporal dispersion of antisense oligonucleotides (ASO) along the neuraxis over clinically relevant time scales of days and weeks as a function of infusion, physiological and molecular properties. We demonstrate its prediction capability using biodistribution data of antisense oligonucleotide (ASO) administration in non-human primates. The results are in close agreement with the observed ASO pharmacokinetics in all key compartments of the central nervous system. The model enables determination of optimal injection parameters such as intrathecal infusion volume and duration for maximum ASO delivery to the brain. Our quantitative model-guided analysis is suitable for identifying optimal parameter settings to target specific brain regions with therapeutic drugs such as ASOs.

2.
Sci Rep ; 10(1): 9628, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32541819

ABSTRACT

Analysis of the topology of transcriptional regulatory networks (TRNs) is an effective way to study the regulatory interactions between the transcription factors (TFs) and the target genes. TRNs are characterized by the abundance of motifs such as feed forward loops (FFLs), which contribute to their structural and functional properties. In this paper, we focus on the role of motifs (specifically, FFLs) in signal propagation in TRNs and the organization of the TRN topology with FFLs as building blocks. To this end, we classify nodes participating in FFLs (termed motif central nodes) into three distinct roles (namely, roles A, B and C), and contrast them with TRN nodes having high connectivity on the basis of their potential for information dissemination, using metrics such as network efficiency, path enumeration, epidemic models and standard graph centrality measures. We also present the notion of a three tier architecture and how it can help study the structural properties of TRN based on connectivity and clustering tendency of motif central nodes. Finally, we motivate the potential implication of the structural properties of motif centrality in design of efficient protocols of information routing in communication networks as well as their functional properties in global regulation and stress response to study specific disease conditions and identification of drug targets.


Subject(s)
Gene Expression Regulation , Gene Regulatory Networks , Transcription Factors/metabolism , Gene Expression Regulation/physiology , Gene Regulatory Networks/physiology , Genes , Transcription Factors/physiology
3.
Biophys J ; 118(7): 1526-1536, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32101713

ABSTRACT

Individual cells in a solution display variable uptake of nanomaterials, peptides, and nutrients. Such variability reflects their heterogeneity in endocytic capacity. In a recent work, we have shown that the endocytic capacity of a cell depends on its size and surface density of endocytic components (transporters). We also demonstrated that in MDA-MB-231 breast cancer cells, the cell-surface transporter density (n) may decay with cell radius (r) following the power rule n ∼ rα, where α ≈ -1. In this work, we investigate how n and r may independently contribute to the endocytic heterogeneity of a cell population. Our analysis indicates that the smaller cells display more heterogeneity because of the higher stochastic variations in n. By contrast, the larger cells display a more uniform uptake, reflecting less-stochastic variations in n. We provide analyses of these dependencies by establishing a stochastic model. Our analysis reveals that the exponent α in the above relationship is not a constant; rather, it is a random variable whose distribution depends on cell size r. Using Bayesian analysis, we characterize the cell-size-dependent distributions of α that accurately capture the particle uptake heterogeneity of MDA-MB-231 cells.


Subject(s)
Bayes Theorem , Biological Transport , Cell Line, Tumor , Cell Size
4.
J Theor Biol ; 480: 34-42, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31374284

ABSTRACT

Fn14 is a transmembrane receptor protein belonging to the tumor necrosis factor receptor (TNFR) superfamily. Many experimental reports have shown that crosslinking of the receptor by its extracellular ligand TWEAK induces prolonged activation of transcription factor NF-κB. This behavior is distinct from TNF-α receptor, which is a more well-characterized member of the TNFR family. TNF-α receptor, despite sharing many similar molecular interactions with Fn14, only transiently activates NF-κB in response to TNF-α stimulation. Here, we investigate molecular mechanisms that enable Fn14 to display such distinctive behavior. In particular, we focus on two specific features of the Fn14 pathway that potentially give rise to a positive feedback regulation and differentiate it from the TNF-α receptor signaling. By developing a mechanistic model, we analyze how these features may determine the dynamics of an Fn14-NF-κB response. Our analysis reveals that stimulation of Fn14 by TWEAK may generate highly non-linear dynamics, including stable limit cycles and bistable responses. The type of response depends both on the strength and duration of a TWEAK signal. Our predictions and analyses also show that the molecular interactions underlying the positive feedback explain the prolonged activation of NF-κB under certain parameter regimes. In light of the model predictions, we propose possible deregulations of Fn14 leading to its overexpression in solid tumors and tissue injuries.


Subject(s)
Models, Biological , NF-kappa B/metabolism , Signal Transduction , TWEAK Receptor/metabolism , Animals , Cell Nucleus/metabolism , Feedback, Physiological , Gene Expression Regulation , Humans
5.
PLoS Comput Biol ; 15(1): e1006706, 2019 01.
Article in English | MEDLINE | ID: mdl-30653502

ABSTRACT

Receptor tyrosine kinases (RTKs) typically contain multiple autophosphorylation sites in their cytoplasmic domains. Once activated, these autophosphorylation sites can recruit downstream signaling proteins containing Src homology 2 (SH2) and phosphotyrosine-binding (PTB) domains, which recognize phosphotyrosine-containing short linear motifs (SLiMs). These domains and SLiMs have polyspecific or promiscuous binding activities. Thus, multiple signaling proteins may compete for binding to a common SLiM and vice versa. To investigate the effects of competition on RTK signaling, we used a rule-based modeling approach to develop and analyze models for ligand-induced recruitment of SH2/PTB domain-containing proteins to autophosphorylation sites in the insulin-like growth factor 1 (IGF1) receptor (IGF1R). Models were parameterized using published datasets reporting protein copy numbers and site-specific binding affinities. Simulations were facilitated by a novel application of model restructuration, to reduce redundancy in rule-derived equations. We compare predictions obtained via numerical simulation of the model to those obtained through simple prediction methods, such as through an analytical approximation, or ranking by copy number and/or KD value, and find that the simple methods are unable to recapitulate the predictions of numerical simulations. We created 45 cell line-specific models that demonstrate how early events in IGF1R signaling depend on the protein abundance profile of a cell. Simulations, facilitated by model restructuration, identified pairs of IGF1R binding partners that are recruited in anti-correlated and correlated fashions, despite no inclusion of cooperativity in our models. This work shows that the outcome of competition depends on the physicochemical parameters that characterize pairwise interactions, as well as network properties, including network connectivity and the relative abundances of competitors.


Subject(s)
Models, Biological , Receptor, IGF Type 1/metabolism , Signal Transduction/physiology , Animals , Binding Sites , Cell Line , Cluster Analysis , Computational Biology , Humans , Mice , Phosphorylation , Protein Binding , Proteins/chemistry , Proteins/metabolism , src Homology Domains
6.
Biophys J ; 116(2): 347-359, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30580920

ABSTRACT

The size of a cell is central to many functions, including cellular communication and exchange of materials with the environment. This modeling and experimental study focused on understanding how the size of a cell determines its ability to uptake nanometer-scale extracellular materials from the environment. Several mechanisms in the cell plasma membrane mediate cellular uptake of nutrients, biomolecules, and particles. These mechanisms involve recognition and internalization of the extracellular molecules via endocytic components, such as clathrin-coated pits, vacuoles, and micropinocytic vesicles. Because the demand for an external resource could be different for cells of different sizes, the collective actions of these various endocytic routes should also vary based on the cell size. Here, using a reaction-diffusion model, we analyze single-cell data to interrogate the one/one mapping between the size of the MDA-MB 231 breast cancer cells and their ability to uptake nanoparticles. Our analysis indicates that under both reaction- and diffusion-controlled regimes, cellular uptake follows a linear relationship with the cell radius. Furthermore, this linear dependency is insensitive to particle size variation within 20-200 nm range. This result is counterintuitive because the general perception is that cellular uptake is proportional to the cell volume (mass) or surface area and hence follow a cubic or square relationship with the cell radius. A further analysis using our model reveals a potential mechanism underlying this linear relationship.


Subject(s)
Cell Size , Endocytosis , Nanoparticles/metabolism , Cell Line, Tumor , Cytoplasmic Vesicles/metabolism , Humans , Nonlinear Dynamics
7.
J R Soc Interface ; 15(140)2018 03.
Article in English | MEDLINE | ID: mdl-29593085

ABSTRACT

Tissue targeting is a critical challenge for systemic delivery of drug nanocarriers. To overcome this challenge, major research efforts have been undertaken to design ligand-conjugated nanoparticles. However, limited work has been done to quantitatively assess the effectiveness of such approach. In this work, using a mechanistic spatio-temporal model, I investigate the effectiveness of ligand-directed tissue targeting. By applying an approach from the colloidal filtration theory, I develop a Brownian dynamics model of nanoparticle-cell interaction. The model incorporates a single cell and its surrounding flow field. It considers both specific (receptor-mediated) and non-specific (bare cell surface-mediated) recognition of nanoparticles subject to convective and diffusive motion. Using the model, I investigate how the specific and non-specific interactions compare in determining the overall targeting efficacy. My analysis provides some interesting findings that contradict the general notion that effective targeting is possible based upon the differential receptor expression in cancer and non-cancer cells. I show that such strategy may yield only a marginal gain in the targeting efficacy. Moreover, non-specific interaction may have an important influence on particle recognition by cells even at high receptor expression levels.


Subject(s)
Drug Carriers , Models, Biological , Nanoparticles , Neoplasms , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/therapeutic use , Humans , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Particle Size
8.
R Soc Open Sci ; 5(9): 180190, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30839725

ABSTRACT

Aggregation of cell surface receptor proteins by multivalent antigens is an essential early step for immune cell signalling. A number of experimental and modelling studies in the past have investigated multivalent ligand-mediated aggregation of IgE receptors (FcɛRI) in the plasma membrane of mast cells. However, understanding of the mechanisms of FcɛRI aggregation remains incomplete. Experimental reports indicate that FcɛRI forms relatively small and finite-sized clusters when stimulated by a multivalent ligand. By contrast, modelling studies have shown that receptor cross-linking by a trivalent ligand may lead to the formation of large receptor superaggregates that may potentially give rise to hyperactive cellular responses. In this work, we have developed a Brownian dynamics-based spatio-temporal model to analyse FcɛRI aggregation by a trivalent antigen. Unlike the existing models, which implemented non-spatial simulation approaches, our model explicitly accounts for the coarse-grained site-specific features of the multivalent species (molecules and complexes). The model incorporates membrane diffusion, steric collisions and sub-nanometre-scale site-specific interaction of the time-evolving species of arbitrary structures. Using the model, we investigated temporal evolution of the species and their diffusivities. Consistent with a recent experimental report, our model predicted sharp decay in species mobility in the plasma membrane in response receptor cross-linking by a multivalent antigen. We show that, due to such decay in the species mobility, post-stimulation receptor aggregation may become self-limiting. Our analysis reveals a potential regulatory mechanism suppressing hyperactivation of immune cells in response to multivalent antigens.

9.
J Comput Biol ; 24(12): 1275-1283, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29099235

ABSTRACT

This article introduces a multiscale framework for spatiotemporal modeling of protein-protein interaction. Cellular protein molecules represent multivalent species that contain modular features, such as binding domains and phosphorylation motifs. The binding and transformations of these features occur at a small time and spatial scale. On the other hand, space and time involved in protein diffusion, colocalization, and formation of complexes could be relatively large. Here, we present an agent-based framework integrated with a multiscale Brownian Dynamics (BD) simulation algorithm. The framework employs spatial graphs to describe multivalent molecules and complexes with their site-specific details. By implementing a time-adaptive feature, the BD algorithm enables efficient computation while capturing the site-specific interactions of the diffusing species at the sub-nanometer scale. We demonstrate these capabilities by modeling two multivalent molecules, one representing a ligand and the other a receptor, in a two-dimensional plane (cell membrane). Using the model, we show that the algorithm can accelerate computation by orders of magnitudes in both concentrated and dilute regimes. We also show that the algorithm enables robust model predictions against a wide range of selection of time step sizes.


Subject(s)
Algorithms , Models, Theoretical , Protein Interaction Maps , Proteins/metabolism , Spatio-Temporal Analysis , Humans , Molecular Dynamics Simulation
10.
BMC Syst Biol ; 11(1): 113, 2017 Nov 25.
Article in English | MEDLINE | ID: mdl-29178887

ABSTRACT

BACKGROUND: Particle size is a key parameter for drug-delivery nanoparticle design. It is believed that the size of a nanoparticle may have important effects on its ability to overcome the transport barriers in biological tissues. Nonetheless, such effects remain poorly understood. Using a multiscale model, this work investigates particle size effects on the tissue distribution and penetration efficacy of drug-delivery nanoparticles. RESULTS: We have developed a multiscale spatiotemporal model of nanoparticle transport in biological tissues. The model implements a time-adaptive Brownian Dynamics algorithm that links microscale particle-cell interactions and adhesion dynamics to tissue-scale particle dispersion and penetration. The model accounts for the advection, diffusion, and cellular uptakes of particles. Using the model, we have analyzed how particle size affects the intra-tissue dispersion and penetration of drug delivery nanoparticles. We focused on two published experimental works that investigated particle size effects in in vitro and in vivo tissue conditions. By analyzing experimental data reported in these two studies, we show that particle size effects may appear pronounced in an in vitro cell-free tissue system, such as collagen matrix. In an in vivo tissue system, the effects of particle size could be relatively modest. We provide a detailed analysis on how particle-cell interactions may determine distribution and penetration of nanoparticles in a biological tissue. CONCLUSION: Our work suggests that the size of a nanoparticle may play a less significant role in its ability to overcome the intra-tissue transport barriers. We show that experiments involving cell-free tissue systems may yield misleading observations of particle size effects due to the absence of advective transport and particle-cell interactions.


Subject(s)
Drug Carriers/pharmacokinetics , Models, Biological , Nanoparticles , Drug Delivery Systems , Particle Size , Tissue Distribution
11.
Nanotechnology ; 28(4): 045601, 2017 Jan 27.
Article in English | MEDLINE | ID: mdl-27977417

ABSTRACT

Ineffective drug release at the target site is among the top challenges for cancer treatment. This reflects the facts that interaction with the physiological condition can denature active ingredients of drugs, and low delivery to the disease microenvironment leads to poor therapeutic outcomes. We hypothesize that depositing a thin layer of bioresponsive polymer on the surface of drug nanoparticles would not only protect drugs from degradation but also allow the release of drugs at the target site. Here, we report a one-step process to prepare bioresponsive polymer coated drug nanorods (NRs) from liquid precursors using the solvent diffusion method. A thin layer (10.3 ± 1.4 nm) of poly(ε-caprolactone) (PCL) polymer coating was deposited on the surface of camptothecin (CPT) anti-cancer drug NRs. The mean size of PCL-coated CPT NRs was 500.9 ± 91.3 nm length × 122.7 ± 10.1 nm width. The PCL polymer coating was biodegradable at acidic pH 6 as determined by Fourier transform infrared spectroscopy. CPT drugs were released up to 51.5% when PCL coating dissolved into non-toxic carboxyl and hydroxyl groups. Trastuzumab (TTZ), a humanized IgG monoclonal antibody, was conjugated to the NR surface for breast cancer cell targeting. Combination treatments using CPT and TTZ decreased the HER-2 positive BT-474 breast cancer cell growth by 66.9 ± 5.3% in vitro. These results suggest effective combination treatments of breast cancer cells using bioresponsive polymer coated drug delivery.


Subject(s)
Breast Neoplasms/drug therapy , Coated Materials, Biocompatible/chemistry , Nanotubes/chemistry , Polymers/chemistry , Camptothecin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Liberation , Female , Humans , Image Processing, Computer-Assisted , Nanotubes/ultrastructure , Polyesters/chemistry , Spectroscopy, Fourier Transform Infrared , Trastuzumab/pharmacology , Trastuzumab/therapeutic use
12.
Bioinformatics ; 32(21): 3366-3368, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27402907

ABSTRACT

: BioNetGen is an open-source software package for rule-based modeling of complex biochemical systems. Version 2.2 of the software introduces numerous new features for both model specification and simulation. Here, we report on these additions, discussing how they facilitate the construction, simulation and analysis of larger and more complex models than previously possible. AVAILABILITY AND IMPLEMENTATION: Stable BioNetGen releases (Linux, Mac OS/X and Windows), with documentation, are available at http://bionetgen.org Source code is available at http://github.com/RuleWorld/bionetgen CONTACT: bionetgen.help@gmail.comSupplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Biochemistry , Software , Humans , Models, Theoretical , Programming Languages
13.
ACS Chem Biol ; 9(7): 1508-19, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24784318

ABSTRACT

To investigate why responses of mast cells to antigen-induced IgE receptor (FcεRI) aggregation depend nonlinearly on antigen dose, we characterized a new artificial ligand, DF3, through complementary modeling and experimentation. This ligand is a stable trimer of peptides derived from bacteriophage T4 fibritin, each conjugated to a hapten (DNP). We found low and high doses of DF3 at which degranulation of mast cells sensitized with DNP-specific IgE is minimal, but ligand-induced receptor aggregation is comparable to aggregation at an intermediate dose, optimal for degranulation. This finding makes DF3 an ideal reagent for studying the balance of negative and positive signaling in the FcεRI pathway. We find that the lipid phosphatase SHIP and the protein tyrosine phosphatase SHP-1 negatively regulate mast cell degranulation over all doses considered. In contrast, SHP-2 promotes degranulation. With high DF3 doses, relatively rapid recruitment of SHIP to the plasma membrane may explain the reduced degranulation response. Our results demonstrate that optimal secretory responses of mast cells depend on the formation of receptor aggregates that promote sufficient positive signaling by Syk to override phosphatase-mediated negative regulatory signals.


Subject(s)
Antigens/immunology , Cell Degranulation , Immunoglobulin E/immunology , Mast Cells/immunology , Phosphoric Monoester Hydrolases/immunology , Receptors, IgE/immunology , Viral Proteins/immunology , Animals , Antigens/chemistry , Humans , Ligands , Mast Cells/cytology , Models, Molecular , Peptides/chemistry , Peptides/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/immunology , Rats , Signal Transduction , Viral Proteins/chemistry
14.
Biochemistry ; 53(16): 2594-604, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24697349

ABSTRACT

Adaptor protein Grb2 binds phosphotyrosines in the epidermal growth factor (EGF) receptor (EGFR) and thereby links receptor activation to intracellular signaling cascades. Here, we investigated how recruitment of Grb2 to EGFR is affected by the spatial organization and quaternary state of activated EGFR. We used the techniques of image correlation spectroscopy (ICS) and lifetime-detected Förster resonance energy transfer (also known as FLIM-based FRET or FLIM-FRET) to measure ligand-induced receptor clustering and Grb2 binding to activated EGFR in BaF/3 cells. BaF/3 cells were stably transfected with fluorescently labeled forms of Grb2 (Grb2-mRFP) and EGFR (EGFR-eGFP). Following stimulation of the cells with EGF, we detected nanometer-scale association of Grb2-mRFP with EGFR-eGFP clusters, which contained, on average, 4 ± 1 copies of EGFR-eGFP per cluster. In contrast, the pool of EGFR-eGFP without Grb2-mRFP had an average cluster size of 1 ± 0.3 EGFR molecules per punctum. In the absence of EGF, there was no association between EGFR-eGFP and Grb2-mRFP. To interpret these data, we extended our recently developed model for EGFR activation, which considers EGFR oligomerization up to tetramers, to include recruitment of Grb2 to phosphorylated EGFR. The extended model, with adjustment of one new parameter (the ratio of the Grb2 and EGFR copy numbers), is consistent with a cluster size distribution where 2% of EGFR monomers, 5% of EGFR dimers, <1% of EGFR trimers, and 94% of EGFR tetramers are associated with Grb2. Together, our experimental and modeling results further implicate tetrameric EGFR as the key signaling unit and call into question the widely held view that dimeric EGFR is the predominant signaling unit.


Subject(s)
ErbB Receptors/metabolism , GRB2 Adaptor Protein/metabolism , Animals , ErbB Receptors/chemistry , ErbB Receptors/genetics , Fluorescence Resonance Energy Transfer , GRB2 Adaptor Protein/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Models, Molecular , Models, Theoretical , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
15.
PLoS Comput Biol ; 9(9): e1003217, 2013.
Article in English | MEDLINE | ID: mdl-24086117

ABSTRACT

In colorectal cancer cells, APC, a tumor suppressor protein, is commonly expressed in truncated form. Truncation of APC is believed to disrupt degradation of ß-catenin, which is regulated by a multiprotein complex called the destruction complex. The destruction complex comprises APC, Axin, ß-catenin, serine/threonine kinases, and other proteins. The kinases CK1α and GSK -3ß, which are recruited by Axin, mediate phosphorylation of ß-catenin, which initiates its ubiquitination and proteosomal degradation. The mechanism of regulation of ß-catenin degradation by the destruction complex and the role of truncation of APC in colorectal cancer are not entirely understood. Through formulation and analysis of a rule-based computational model, we investigated the regulation of ß-catenin phosphorylation and degradation by APC and the effect of APC truncation on function of the destruction complex. The model integrates available mechanistic knowledge about site-specific interactions and phosphorylation of destruction complex components and is consistent with an array of published data. We find that the phosphorylated truncated form of APC can outcompete Axin for binding to ß-catenin, provided that Axin is limiting, and thereby sequester ß-catenin away from Axin and the Axin-recruited kinases CK1α and GSK -3ß. Full-length APC also competes with Axin for binding to ß-catenin; however, full-length APC is able, through its SAMP repeats, which bind Axin and which are missing in truncated oncogenic forms of APC, to bring ß-catenin into indirect association with Axin and Axin-recruited kinases. Because our model indicates that the positive effects of truncated APC on ß-catenin levels depend on phosphorylation of APC, at the first 20-amino acid repeat, and because phosphorylation of this site is mediated by CK1ε, we suggest that CK1ε is a potential target for therapeutic intervention in colorectal cancer. Specific inhibition of CK1ε is predicted to limit binding of ß-catenin to truncated APC and thereby to reverse the effect of APC truncation.


Subject(s)
Colorectal Neoplasms/genetics , Genes, APC , Humans , Hydrolysis , Mutation , Phosphorylation , Up-Regulation , beta Catenin/genetics , beta Catenin/metabolism
16.
Mol Biosyst ; 9(7): 1849-63, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23629589

ABSTRACT

The epidermal growth factor receptor (EGFR) kinase is generally considered to be activated by either ligand-induced dimerisation or a ligand-induced conformational change within pre-formed dimers. Ligand-induced higher-order EGFR oligomerisation or clustering has been reported but it is not clear how EGFR oligomers, as distinct from EGFR dimers, influence signaling outputs. To address this question, we combined measures of receptor clustering (microscopy; image correlation spectroscopy) and phosphorylation (Western blots) with modelling of mass-action chemical kinetics. A stable BaF/3 cell-line that contains a high proportion (>90%) of inactive dimers of EGFR-eGFP but no secreted ligand and no other detectable ErbB receptors was used as the model cell system. EGF at concentrations of greater than 1 nM was found to cluster EGFR-eGFP dimers into higher-order complexes and cause parallel increases in EGFR phosphorylation. The kinetics of EGFR clustering and phosphorylation were both rapid, plateauing within 2 minutes after stimulation with 30 nM EGF. A rule-based model was formulated to interpret the data. This model took into account ligand binding, ligand-induced conformational changes in the cytosolic tail, monomer-dimer-trimer-tetramer transitions via ectodomain- and kinase-mediated interactions, and phosphorylation. The model predicts that cyclic EGFR tetramers are the predominant phosphorylated species, in which activated receptor dimers adopt a cyclic side-by-side orientation, and that receptor kinase activation is stabilised by the intramolecular interactions responsible for cyclic tetramerization.


Subject(s)
ErbB Receptors/chemistry , ErbB Receptors/metabolism , Models, Biological , Protein Multimerization , Animals , Cell Line , Computer Simulation , Epidermal Growth Factor/metabolism , Kinetics , Ligands , Mice , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs
17.
PLoS One ; 8(3): e60159, 2013.
Article in English | MEDLINE | ID: mdl-23544131

ABSTRACT

Heterogeneity in responses of cells to a stimulus, such as a pathogen or allergen, can potentially play an important role in deciding the fate of the responding cell population and the overall systemic response. Measuring heterogeneous responses requires tools capable of interrogating individual cells. Cell signaling studies commonly do not have single-cell resolution because of the limitations of techniques used such as Westerns, ELISAs, mass spectrometry, and DNA microarrays. Microfluidics devices are increasingly being used to overcome these limitations. Here, we report on a microfluidic platform for cell signaling analysis that combines two orthogonal single-cell measurement technologies: on-chip flow cytometry and optical imaging. The device seamlessly integrates cell culture, stimulation, and preparation with downstream measurements permitting hands-free, automated analysis to minimize experimental variability. The platform was used to interrogate IgE receptor (FcεRI) signaling, which is responsible for triggering allergic reactions, in RBL-2H3 cells. Following on-chip crosslinking of IgE-FcεRI complexes by multivalent antigen, we monitored signaling events including protein phosphorylation, calcium mobilization and the release of inflammatory mediators. The results demonstrate the ability of our platform to produce quantitative measurements on a cell-by-cell basis from just a few hundred cells. Model-based analysis of the Syk phosphorylation data suggests that heterogeneity in Syk phosphorylation can be attributed to protein copy number variations, with the level of Syk phosphorylation being particularly sensitive to the copy number of Lyn.


Subject(s)
Immunoglobulin E/metabolism , Microfluidics/methods , Receptors, IgE/metabolism , Signal Transduction , Single-Cell Analysis/methods , Animals , Calcium/metabolism , Cell Degranulation , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Models, Immunological , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Rats , Signal Transduction/immunology , Syk Kinase , Time Factors , Tumor Necrosis Factor-alpha/biosynthesis , beta-N-Acetylhexosaminidases/metabolism
18.
Autophagy ; 9(1): 74-92, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23196898

ABSTRACT

Macroautophagy (autophagy) is a cellular recycling program essential for homeostasis and survival during cytotoxic stress. This process, which has an emerging role in disease etiology and treatment, is executed in four stages through the coordinated action of more than 30 proteins. An effective strategy for studying complicated cellular processes, such as autophagy, involves the construction and analysis of mathematical or computational models. When developed and refined from experimental knowledge, these models can be used to interrogate signaling pathways, formulate novel hypotheses about systems, and make predictions about cell signaling changes induced by specific interventions. Here, we present the development of a computational model describing autophagic vesicle dynamics in a mammalian system. We used time-resolved, live-cell microscopy to measure the synthesis and turnover of autophagic vesicles in single cells. The stochastically simulated model was consistent with data acquired during conditions of both basal and chemically-induced autophagy. The model was tested by genetic modulation of autophagic machinery and found to accurately predict vesicle dynamics observed experimentally. Furthermore, the model generated an unforeseen prediction about vesicle size that is consistent with both published findings and our experimental observations. Taken together, this model is accurate and useful and can serve as the foundation for future efforts aimed at quantitative characterization of autophagy.


Subject(s)
Autophagy/physiology , Computer Simulation , Models, Biological , Autophagy/drug effects , Autophagy/genetics , Autophagy-Related Proteins , Cell Line , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/physiology , Green Fluorescent Proteins/metabolism , Humans , Macrolides/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Microtubule-Associated Proteins/metabolism , Morpholines/pharmacology , Recombinant Fusion Proteins/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Vesicular Transport Proteins
19.
J Immunol ; 189(2): 646-58, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22711887

ABSTRACT

BCR signaling regulates the activities and fates of B cells. BCR signaling encompasses two feedback loops emanating from Lyn and Fyn, which are Src family protein tyrosine kinases (SFKs). Positive feedback arises from SFK-mediated trans phosphorylation of BCR and receptor-bound Lyn and Fyn, which increases the kinase activities of Lyn and Fyn. Negative feedback arises from SFK-mediated cis phosphorylation of the transmembrane adapter protein PAG1, which recruits the cytosolic protein tyrosine kinase Csk to the plasma membrane, where it acts to decrease the kinase activities of Lyn and Fyn. To study the effects of the positive and negative feedback loops on the dynamical stability of BCR signaling and the relative contributions of Lyn and Fyn to BCR signaling, we consider in this study a rule-based model for early events in BCR signaling that encompasses membrane-proximal interactions of six proteins, as follows: BCR, Lyn, Fyn, Csk, PAG1, and Syk, a cytosolic protein tyrosine kinase that is activated as a result of SFK-mediated phosphorylation of BCR. The model is consistent with known effects of Lyn and Fyn deletions. We find that BCR signaling can generate a single pulse or oscillations of Syk activation depending on the strength of Ag signal and the relative levels of Lyn and Fyn. We also show that bistability can arise in Lyn- or Csk-deficient cells.


Subject(s)
Computer Simulation , Models, Immunological , Proto-Oncogene Proteins c-fyn/physiology , Receptors, Antigen, B-Cell/physiology , Signal Transduction/immunology , src-Family Kinases/physiology , Animals , Calcium Signaling/immunology , Feedback, Physiological , Humans , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, Antigen, B-Cell/metabolism , src-Family Kinases/deficiency , src-Family Kinases/metabolism
20.
PLoS One ; 7(12): e51669, 2012.
Article in English | MEDLINE | ID: mdl-23284735

ABSTRACT

We present a model of the early events in mast cell signaling mediated by FcεRI where the plasma membrane is composed of many small ordered lipid domains (rafts), surrounded by a non-order region of lipids consisting of the remaining plasma membrane. The model treats the rafts as transient structures that constantly form and breakup, but that maintain a fixed average number per cell. The rafts have a high propensity for harboring Lyn kinase, aggregated, but not unaggregated receptors, and the linker for the activation of T cells (LAT). Phosphatase activity in the rafts is substantially reduced compared to the nonraft region. We use the model to analyze published experiments on the rat basophilic leukemia (RBL)-2H3 cell line that seem to contradict the notion that rafts offer protection. In these experiments IgE was cross-linked with a multivalent antigen and then excess monovalent hapten was added to break-up cross-links. The dephosphorylation of the unaggregated receptor (nonraft associated) and of LAT (raft associated) were then monitored in time and found to decay at similar rates, leading to the conclusion that rafts offer no protection from dephosphorylation. In the model, because the rafts are transient, a protein that is protected while in a raft will be subject to dephosphorylation when the raft breaks up and the protein finds itself in the nonraft region of the membrane. We show that the model is consistent with the receptor and LAT dephosphorylation experiments while still allowing rafts to enhance signaling by providing substantial protection from phosphatases.


Subject(s)
Immunoglobulin E/metabolism , Leukemia, Basophilic, Acute/metabolism , Mast Cells/metabolism , Membrane Microdomains/metabolism , Models, Biological , Receptors, IgE/metabolism , src-Family Kinases/metabolism , Animals , Calcium/metabolism , Cell Membrane/metabolism , Computer Simulation , Haptens/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia, Basophilic, Acute/pathology , Lipoylation , Mast Cells/pathology , Membrane Microdomains/pathology , Mutation/genetics , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Rats , Signal Transduction , Syk Kinase , Tumor Cells, Cultured , Tyrosine/metabolism , src-Family Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...