Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(9): e1011597, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37669278

RESUMEN

When infected with a virus, cells may secrete interferons (IFNs) that prompt nearby cells to prepare for upcoming infection. Reciprocally, viral proteins often interfere with IFN synthesis and IFN-induced signaling. We modeled the crosstalk between the propagating virus and the innate immune response using an agent-based stochastic approach. By analyzing immunofluorescence microscopy images we observed that the mutual antagonism between the respiratory syncytial virus (RSV) and infected A549 cells leads to dichotomous responses at the single-cell level and complex spatial patterns of cell signaling states. Our analysis indicates that RSV blocks innate responses at three levels: by inhibition of IRF3 activation, inhibition of IFN synthesis, and inhibition of STAT1/2 activation. In turn, proteins coded by IFN-stimulated (STAT1/2-activated) genes inhibit the synthesis of viral RNA and viral proteins. The striking consequence of these inhibitions is a lack of coincidence of viral proteins and IFN expression within single cells. The model enables investigation of the impact of immunostimulatory defective viral particles and signaling network perturbations that could potentially facilitate containment or clearance of the viral infection.


Asunto(s)
Virus Sincitial Respiratorio Humano , Virosis , Humanos , Inmunidad Innata , Interferones , Proteínas Virales
2.
PLoS Comput Biol ; 19(5): e1011155, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37216347

RESUMEN

Living cells utilize signaling pathways to sense, transduce, and process information. As the extracellular stimulation often has rich temporal characteristics which may govern dynamic cellular responses, it is important to quantify the rate of information flow through the signaling pathways. In this study, we used an epithelial cell line expressing a light-activatable FGF receptor and an ERK activity reporter to assess the ability of the MAPK/ERK pathway to transduce signal encoded in a sequence of pulses. By stimulating the cells with random light pulse trains, we demonstrated that the MAPK/ERK channel capacity is at least 6 bits per hour. The input reconstruction algorithm detects the light pulses with 1-min accuracy 5 min after their occurrence. The high information transmission rate may enable the pathway to coordinate multiple processes including cell movement and respond to rapidly varying stimuli such as chemoattracting gradients created by other cells.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Transducción de Señal , Línea Celular , Sistema de Señalización de MAP Quinasas/fisiología , Células Epiteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo
3.
J Virol ; 96(22): e0134122, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-36326278

RESUMEN

We observed the interference between two prevalent respiratory viruses, respiratory syncytial virus (RSV) and influenza A virus (IAV) (H1N1), and characterized its molecular underpinnings in alveolar epithelial cells (A549). We found that RSV induces higher levels of interferon beta (IFN-ß) production than IAV and that IFN-ß priming confers higher-level protection against infection with IAV than with RSV. Consequently, we focused on the sequential infection scheme of RSV and then IAV. Using A549 wild-type (WT), IFNAR1 knockout (KO), IFNLR1 KO, and IFNAR1-IFNLR1 double-KO cell lines, we found that both IFN-ß and IFN-λ are necessary for maximum protection against subsequent infection. Immunostaining revealed that preinfection with RSV partitions the cell population into a subpopulation susceptible to subsequent infection with IAV and an IAV-proof subpopulation. Strikingly, the susceptible cells turned out to be those already compromised and efficiently expressing RSV, whereas the bystander, interferon-primed cells are resistant to IAV infection. Thus, virus-virus exclusion at the cell population level is not realized through direct competition for a shared ecological niche (single cell) but rather is achieved with the involvement of specific cytokines induced by the host's innate immune response. IMPORTANCE Influenza A virus (IAV) and respiratory syncytial virus (RSV) are common recurrent respiratory infectants that show a relatively high coincidence. We demonstrated that preinfection with RSV partitions the cell population into a subpopulation susceptible to subsequent infection with IAV and an IAV-proof subpopulation. The susceptible cells are those already compromised and efficiently expressing RSV, whereas the bystander cells are resistant to IAV infection. The cross-protective effect critically depends on IFN-ß and IFN-λ signaling and thus ensues when the proportion of cells preinfected with RSV is relatively low yet sufficient to trigger a pervasive antiviral state in bystander cells. Our study suggests that mild, but not severe, respiratory infections may have a short-lasting protective role against more dangerous respiratory viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).


Asunto(s)
COVID-19 , Subtipo H1N1 del Virus de la Influenza A , Virus de la Influenza A , Gripe Humana , Virus Sincitial Respiratorio Humano , Humanos , SARS-CoV-2 , Interferones/metabolismo , Interferón lambda
4.
PLoS Comput Biol ; 18(2): e1009811, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35143483

RESUMEN

Nucleosomes are recognized as key regulators of transcription. However, the relationship between slow nucleosome unwrapping dynamics and bulk transcriptional properties has not been thoroughly explored. Here, an agent-based model that we call the dynamic defect Totally Asymmetric Simple Exclusion Process (ddTASEP) was constructed to investigate the effects of nucleosome-induced pausing on transcriptional dynamics. Pausing due to slow nucleosome dynamics induced RNAPII convoy formation, which would cooperatively prevent nucleosome rebinding leading to bursts of transcription. The mean first passage time (MFPT) and the variance of first passage time (VFPT) were analytically expressed in terms of the nucleosome rate constants, allowing for the direct quantification of the effects of nucleosome-induced pausing on pioneering polymerase dynamics. The mean first passage elongation rate γ(hc, ho) is inversely proportional to the MFPT and can be considered to be a new axis of the ddTASEP phase diagram, orthogonal to the classical αß-plane (where α and ß are the initiation and termination rates). Subsequently, we showed that, for ß = 1, there is a novel jamming transition in the αγ-plane that separates the ddTASEP dynamics into initiation-limited and nucleosome pausing-limited regions. We propose analytical estimates for the RNAPII density ρ, average elongation rate v, and transcription flux J and verified them numerically. We demonstrate that the intra-burst RNAPII waiting times tin follow the time-headway distribution of a max flux TASEP and that the average inter-burst interval [Formula: see text] correlates with the index of dispersion De. In the limit γ→0, the average burst size reaches a maximum set by the closing rate hc. When α≪1, the burst sizes are geometrically distributed, allowing large bursts even while the average burst size [Formula: see text] is small. Last, preliminary results on the relative effects of static and dynamic defects are presented to show that dynamic defects can induce equal or greater pausing than static bottle necks.


Asunto(s)
Nucleosomas , ARN Polimerasa II , ARN Polimerasa II/genética , Transcripción Genética
5.
PLoS Pathog ; 16(1): e1008247, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31917805

RESUMEN

In this study, we established a dynamic micromodel of urinary tract infection to analyze the impact of UT-segment-specific urinary outflow on the persistence of E. coli colonization. We found that the adherence of Dr+ E. coli to bladder T24 transitional cells and type IV collagen is maximal at lowest shear stress and is reduced by any increase in flow velocity. The analyzed adherence was effective in the whole spectrum of physiological shear stress and was almost irreversible over the entire range of generated shear force. Once Dr+ E. coli bound to host cells or collagen, they did not detach even in the presence of elevated shear stress or of chloramphenicol, a competitive inhibitor of binding. Investigating the role of epithelial surface architecture, we showed that the presence of budding cells-a model microarchitectural obstacle-promotes colonization of the urinary tract by E. coli. We report a previously undescribed phenomenon of epithelial cell "rolling-shedding" colonization, in which the detached epithelial cells reattach to the underlying cell line through a layer of adherent Dr+ E. coli. This rolling-shedding colonization progressed continuously due to "refilling" induced by the flow-perturbing obstacle. The shear stress of fluid containing free-floating bacteria fueled the rolling, while providing an uninterrupted supply of new bacteria to be trapped by the rolling cell. The progressive rolling allows for transfer of briefly attached bacteria onto the underlying monolayer in a repeating cascading event.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Escherichia coli/química , Escherichia coli/fisiología , Infecciones Urinarias/microbiología , Adhesinas de Escherichia coli/genética , Adhesinas de Escherichia coli/metabolismo , Adhesión Bacteriana , Escherichia coli/genética , Humanos , Estrés Mecánico
6.
Bioinformatics ; 33(22): 3667-3669, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-29036531

RESUMEN

SUMMARY: Rule-based modeling is a powerful approach for studying biomolecular site dynamics. Here, we present SPATKIN, a general-purpose simulator for rule-based modeling in two spatial dimensions. The simulation algorithm is a lattice-based method that tracks Brownian motion of individual molecules and the stochastic firing of rule-defined reaction events. Because rules are used as event generators, the algorithm is network-free, meaning that it does not require to generate the complete reaction network implied by rules prior to simulation. In a simulation, each molecule (or complex of molecules) is taken to occupy a single lattice site that cannot be shared with another molecule (or complex). SPATKIN is capable of simulating a wide array of membrane-associated processes, including adsorption, desorption and crowding. Models are specified using an extension of the BioNetGen language, which allows to account for spatial features of the simulated process. AVAILABILITY AND IMPLEMENTATION: The C ++ source code for SPATKIN is distributed freely under the terms of the GNU GPLv3 license. The source code can be compiled for execution on popular platforms (Windows, Mac and Linux). An installer for 64-bit Windows and a macOS app are available. The source code and precompiled binaries are available at the SPATKIN Web site (http://pmbm.ippt.pan.pl/software/spatkin). CONTACT: spatkin.simulator@gmail.com. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Biología Computacional/métodos , Simulación de Dinámica Molecular , Programas Informáticos , Algoritmos
7.
PLoS Comput Biol ; 13(4): e1005436, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28379950

RESUMEN

In this study a new computational method is developed to quantify decision making errors in cells, caused by noise and signaling failures. Analysis of tumor necrosis factor (TNF) signaling pathway which regulates the transcription factor Nuclear Factor κB (NF-κB) using this method identifies two types of incorrect cell decisions called false alarm and miss. These two events represent, respectively, declaring a signal which is not present and missing a signal that does exist. Using single cell experimental data and the developed method, we compute false alarm and miss error probabilities in wild-type cells and provide a formulation which shows how these metrics depend on the signal transduction noise level. We also show that in the presence of abnormalities in a cell, decision making processes can be significantly affected, compared to a wild-type cell, and the method is able to model and measure such effects. In the TNF-NF-κB pathway, the method computes and reveals changes in false alarm and miss probabilities in A20-deficient cells, caused by cell's inability to inhibit TNF-induced NF-κB response. In biological terms, a higher false alarm metric in this abnormal TNF signaling system indicates perceiving more cytokine signals which in fact do not exist at the system input, whereas a higher miss metric indicates that it is highly likely to miss signals that actually exist. Overall, this study demonstrates the ability of the developed method for modeling cell decision making errors under normal and abnormal conditions, and in the presence of transduction noise uncertainty. Compared to the previously reported pathway capacity metric, our results suggest that the introduced decision error metrics characterize signaling failures more accurately. This is mainly because while capacity is a useful metric to study information transmission in signaling pathways, it does not capture the overlap between TNF-induced noisy response curves.


Asunto(s)
Comunicación Celular/fisiología , Biología Computacional/métodos , Modelos Biológicos , Modelos Estadísticos , Transducción de Señal/fisiología , Teoría de las Decisiones , FN-kappa B/metabolismo , Procesamiento de Señales Asistido por Computador , Análisis de la Célula Individual , Factor de Necrosis Tumoral alfa/metabolismo
8.
PLoS Comput Biol ; 12(2): e1004787, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26928575

RESUMEN

The p53 transcription factor is a regulator of key cellular processes including DNA repair, cell cycle arrest, and apoptosis. In this theoretical study, we investigate how the complex circuitry of the p53 network allows for stochastic yet unambiguous cell fate decision-making. The proposed Markov chain model consists of the regulatory core and two subordinated bistable modules responsible for cell cycle arrest and apoptosis. The regulatory core is controlled by two negative feedback loops (regulated by Mdm2 and Wip1) responsible for oscillations, and two antagonistic positive feedback loops (regulated by phosphatases Wip1 and PTEN) responsible for bistability. By means of bifurcation analysis of the deterministic approximation we capture the recurrent solutions (i.e., steady states and limit cycles) that delineate temporal responses of the stochastic system. Direct switching from the limit-cycle oscillations to the "apoptotic" steady state is enabled by the existence of a subcritical Neimark-Sacker bifurcation in which the limit cycle loses its stability by merging with an unstable invariant torus. Our analysis provides an explanation why cancer cell lines known to have vastly diverse expression levels of Wip1 and PTEN exhibit a broad spectrum of responses to DNA damage: from a fast transition to a high level of p53 killer (a p53 phosphoform which promotes commitment to apoptosis) in cells characterized by high PTEN and low Wip1 levels to long-lasting p53 level oscillations in cells having PTEN promoter methylated (as in, e.g., MCF-7 cell line).


Asunto(s)
Retroalimentación Fisiológica/fisiología , Modelos Biológicos , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/fisiología , Apoptosis/fisiología , Puntos de Control del Ciclo Celular/fisiología , Regulación Neoplásica de la Expresión Génica , Humanos
9.
Nature ; 466(7303): 267-71, 2010 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-20581820

RESUMEN

Cells operate in dynamic environments using extraordinary communication capabilities that emerge from the interactions of genetic circuitry. The mammalian immune response is a striking example of the coordination of different cell types. Cell-to-cell communication is primarily mediated by signalling molecules that form spatiotemporal concentration gradients, requiring cells to respond to a wide range of signal intensities. Here we use high-throughput microfluidic cell culture and fluorescence microscopy, quantitative gene expression analysis and mathematical modelling to investigate how single mammalian cells respond to different concentrations of the signalling molecule tumour-necrosis factor (TNF)-alpha, and relay information to the gene expression programs by means of the transcription factor nuclear factor (NF)-kappaB. We measured NF-kappaB activity in thousands of live cells under TNF-alpha doses covering four orders of magnitude. We find, in contrast to population-level studies with bulk assays, that the activation is heterogeneous and is a digital process at the single-cell level with fewer cells responding at lower doses. Cells also encode a subtle set of analogue parameters to modulate the outcome; these parameters include NF-kappaB peak intensity, response time and number of oscillations. We developed a stochastic mathematical model that reproduces both the digital and analogue dynamics as well as most gene expression profiles at all measured conditions, constituting a broadly applicable model for TNF-alpha-induced NF-kappaB signalling in various types of cells. These results highlight the value of high-throughput quantitative measurements with single-cell resolution in understanding how biological systems operate.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Células 3T3 , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Técnicas de Cultivo de Célula , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Ratones , Técnicas Analíticas Microfluídicas , Microscopía Fluorescente , Modelos Biológicos , Procesos Estocásticos , Especificidad por Sustrato , Factores de Tiempo
10.
J Chem Phys ; 143(21): 215102, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26646890

RESUMEN

Biological signals in cells are transmitted with the use of reaction cycles, such as the phosphorylation-dephosphorylation cycle, in which substrate is modified by antagonistic enzymes. An appreciable share of such reactions takes place in crowded environments of two-dimensional structures, such as plasma membrane or intracellular membranes, and is expected to be diffusion-controlled. In this work, starting from the microscopic bimolecular reaction rate constants and using estimates of the mean first-passage time for an enzyme-substrate encounter, we derive diffusion-dependent effective macroscopic reaction rate coefficients (EMRRC) for a generic reaction cycle. Each EMRRC was found to be half of the harmonic average of the microscopic rate constant (phosphorylation c or dephosphorylation d), and the effective (crowding-dependent) motility divided by a slowly decreasing logarithmic function of the sum of the enzyme concentrations. This implies that when c and d differ, the two EMRRCs scale differently with the motility, rendering the steady-state fraction of phosphorylated substrate molecules diffusion-dependent. Analytical predictions are verified using kinetic Monte Carlo simulations on the two-dimensional triangular lattice at the single-molecule resolution. It is demonstrated that the proposed formulas estimate the steady-state concentrations and effective reaction rates for different sets of microscopic reaction rates and concentrations of reactants, including a non-trivial example where with increasing diffusivity the fraction of phosphorylated substrate molecules changes from 10% to 90%.


Asunto(s)
Biocatálisis , Simulación por Computador , Modelos Biológicos , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Quinasas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Difusión , Humanos , Cinética , Método de Montecarlo , Fosforilación
11.
J Immunol ; 189(2): 646-58, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22711887

RESUMEN

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.


Asunto(s)
Simulación por Computador , Modelos Inmunológicos , Proteínas Proto-Oncogénicas c-fyn/fisiología , Receptores de Antígenos de Linfocitos B/fisiología , Transducción de Señal/inmunología , Familia-src Quinasas/fisiología , Animales , Señalización del Calcio/inmunología , Retroalimentación Fisiológica , Humanos , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Familia-src Quinasas/deficiencia , Familia-src Quinasas/metabolismo
12.
Phys Biol ; 10(3): 035006, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23735655

RESUMEN

The Raf/MEK/ERK cascade is one of the most studied and important signal transduction pathways. However, existing models largely ignore the existence of isoforms of the constituent kinases and their interactions. Here, we propose a model of the ERK cascade that includes heretofore neglected differences between isoforms of MEK. In particular, MEK1 is subject to a negative feedback from activated ERK, which is further conferred to MEK2 via hetero-dimerization. Specifically, ERK phosphorylates MEK1 at the residue Thr292, hypothetically creating an additional phosphatase binding site, accelerating MEK1 and MEK2 dephosphorylation. We incorporated these recently discovered interactions into a mathematical model of the ERK cascade that reproduces the experimental results of Catalanotti et al (2009 Nature Struct. Mol. Biol. 16 294-303) and Kamioka et al (2010 J. Biol. Chem. 285 33540-8). Furthermore, the model allows for predictions regarding the differences in the catalytic activity and function of the MEK isoforms. We propose that the MEK1/MEK2 ratio regulates the duration of the response, which increases with the level of MEK2 and decreases with the level of MEK1. In turn, the amplitude of the response is controlled by the total amount of the two isoforms. We confirm the proposed model structure performing a random parameter sampling, which led us to the conclusion that the sampled parameters, selected to properly reproduce wild-type (WT) cell behavior, to allow for qualitative reproduction of differences in behavior WT cells and cell mutants studied experimentally.


Asunto(s)
MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Sistema de Señalización de MAP Quinasas , Animales , Simulación por Computador , Humanos , Modelos Biológicos , Fosforilación , Mapas de Interacción de Proteínas , Isoformas de Proteínas/metabolismo
13.
Phys Biol ; 10(3): 035007, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23735670

RESUMEN

Bistable regulatory elements enhance heterogeneity in cell populations and, in multicellular organisms, allow cells to specialize and specify their fate. Our study demonstrates that in a system of bistable genetic switch, the noise characteristics control in which of the two epigenetic attractors the cell population will settle. We focus on two types of noise: the gene switching noise and protein dimerization noise. We found that the change of magnitudes of these noise components for one of the two competing genes introduces a large asymmetry of the protein stationary probability distribution and changes the relative probability of individual gene activation. Interestingly, an increase of noise associated with a given gene can either promote or suppress the activation of the gene, depending on the type of noise. Namely, each gene is repressed by an increase of its gene switching noise and activated by an increase of its protein-product dimerization noise. The observed effect was found robust to the large, up to fivefold deviations of the model parameters. In summary, we demonstrated that noise itself may determine the relative strength of the epigenetic attractors, which may provide a unique mode of control of cell fate decisions.


Asunto(s)
Regulación de la Expresión Génica , Modelos Genéticos , Proteínas/genética , Proteínas/metabolismo , Procesos Estocásticos , Animales , Simulación por Computador , Epigénesis Genética , Redes Reguladoras de Genes , Humanos , Biosíntesis de Proteínas , Multimerización de Proteína , Proteínas/química , ARN Mensajero/genética , Transcripción Genética
14.
J Theor Biol ; 317: 140-51, 2013 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-23063780

RESUMEN

The aim of this study is to demonstrate that in molecular dynamical systems with the underlying bi- or multistability, the type of noise determines the most strongly attracting steady state or stochastic attractor. As an example we consider a simple stochastic model of autoregulatory gene with a nonlinear positive feedback, which in the deterministic approximation has two stable steady state solutions. Three types of noise are considered: transcriptional and translational - due to the small number of gene product molecules and the gene switching noise - due to gene activation and inactivation transitions. We demonstrate that the type of noise in addition to the noise magnitude dictates the allocation of probability mass between the two stable steady states. In particular, we found that when the gene switching noise dominates over the transcriptional and translational noise (which is characteristic of eukaryotes), the gene preferentially activates, while in the opposite case, when the transcriptional noise dominates (which is characteristic of prokaryotes) the gene preferentially remains inactive. Moreover, even in the zero-noise limit, when the probability mass generically concentrates in the vicinity of one of two steady states, the choice of the most strongly attracting steady state is noise type-dependent. Although the epigenetic attractors are defined with the aid of the deterministic approximation of the stochastic regulatory process, their relative attractivity is controlled by the type of noise, in addition to noise magnitude. Since noise characteristics vary during the cell cycle and development, such mode of regulation can be potentially employed by cells to switch between alternative epigenetic attractors.


Asunto(s)
Redes Reguladoras de Genes , Biosíntesis de Proteínas/genética , Transcripción Genética , Activación Transcripcional , Animales , Simulación por Computador , Humanos , Modelos Genéticos , Método de Montecarlo , Procesos Estocásticos
15.
Sci Rep ; 13(1): 11143, 2023 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-37429934

RESUMEN

Resolving practical non-identifiability of computational models typically requires either additional data or non-algorithmic model reduction, which frequently results in models containing parameters lacking direct interpretation. Here, instead of reducing models, we explore an alternative, Bayesian approach, and quantify the predictive power of non-identifiable models. We considered an example biochemical signalling cascade model as well as its mechanical analogue. For these models, we demonstrated that by measuring a single variable in response to a properly chosen stimulation protocol, the dimensionality of the parameter space is reduced, which allows for predicting the measured variable's trajectory in response to different stimulation protocols even if all model parameters remain unidentified. Moreover, one can predict how such a trajectory will transform in the case of a multiplicative change of an arbitrary model parameter. Successive measurements of remaining variables further reduce the dimensionality of the parameter space and enable new predictions. We analysed potential pitfalls of the proposed approach that can arise when the investigated model is oversimplified, incorrect, or when the training protocol is inadequate. The main advantage of the suggested iterative approach is that the predictive power of the model can be assessed and practically utilised at each step.

16.
PLoS One ; 18(6): e0286416, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37267242

RESUMEN

An overwhelming majority of mathematical models of regulatory pathways, including the intensively studied NF-κB pathway, remains non-identifiable, meaning that their parameters may not be determined by existing data. The existing NF-κB models that are capable of reproducing experimental data contain non-identifiable parameters, whereas simplified models with a smaller number of parameters exhibit dynamics that differs from that observed in experiments. Here, we reduced an existing model of the canonical NF-κB pathway by decreasing the number of equations from 15 to 6. The reduced model retains two negative feedback loops mediated by IκBα and A20, and in response to both tonic and pulsatile TNF stimulation exhibits dynamics that closely follow that of the original model. We carried out the sensitivity-based linear analysis and Monte Carlo-based analysis to demonstrate that the resulting model is both structurally and practically identifiable given measurements of 5 model variables from a simple TNF stimulation protocol. The reduced model is capable of reproducing different types of responses that are characteristic to regulatory motifs controlled by negative feedback loops: nearly-perfect adaptation as well as damped and sustained oscillations. It can serve as a building block of more comprehensive models of the immune response and cancer, where NF-κB plays a decisive role. Our approach, although may not be automatically generalized, suggests that models of other regulatory pathways can be transformed to identifiable, while retaining their dynamical features.


Asunto(s)
FN-kappa B , Transducción de Señal , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Inhibidor NF-kappaB alfa/metabolismo
17.
Biology (Basel) ; 12(12)2023 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-38132287

RESUMEN

A cell constantly receives signals and takes different fates accordingly. Given the uncertainty rendered by signal transduction noise, a cell may incorrectly perceive these signals. It may mistakenly behave as if there is a signal, although there is none, or may miss the presence of a signal that actually exists. In this paper, we consider a signaling system with two outputs, and introduce and develop methods to model and compute key cell decision-making parameters based on the two outputs and in response to the input signal. In the considered system, the tumor necrosis factor (TNF) regulates the two transcription factors, the nuclear factor κB (NFκB) and the activating transcription factor-2 (ATF-2). These two system outputs are involved in important physiological functions such as cell death and survival, viral replication, and pathological conditions, such as autoimmune diseases and different types of cancer. Using the introduced methods, we compute and show what the decision thresholds are, based on the single-cell measured concentration levels of NFκB and ATF-2. We also define and compute the decision error probabilities, i.e., false alarm and miss probabilities, based on the concentration levels of the two outputs. By considering the joint response of the two outputs of the signaling system, one can learn more about complex cellular decision-making processes, the corresponding decision error rates, and their possible involvement in the development of some pathological conditions.

18.
Sci Signal ; 16(815): eabq1173, 2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-38085817

RESUMEN

Type I interferons (IFNs) are key coordinators of the innate immune response to viral infection, which, through activation of the transcriptional regulators STAT1 and STAT2 (STAT1/2) in bystander cells, induce the expression of IFN-stimulated genes (ISGs). Here, we showed that in cells transfected with poly(I:C), an analog of viral RNA, the transcriptional activity of STAT1/2 was terminated because of depletion of the interferon-ß (IFN-ß) receptor, IFNAR. Activation of RNase L and PKR, products of two ISGs, not only hindered the replenishment of IFNAR but also suppressed negative regulators of IRF3 and NF-κB, consequently promoting IFNB transcription. We incorporated these findings into a mathematical model of innate immunity. By coupling signaling through the IRF3-NF-κB and STAT1/2 pathways with the activities of RNase L and PKR, the model explains how poly(I:C) switches the transcriptional program from being STAT1/2 induced to being IRF3 and NF-κB induced, which converts IFN-ß-responding cells to IFN-ß-secreting cells.


Asunto(s)
Interferón beta , ARN , Interferón beta/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Inmunidad Innata , Modelos Teóricos , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo
19.
Phys Biol ; 9(5): 055002, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23011381

RESUMEN

Living cells may be considered as biochemical reactors of multiple steady states. Transitions between these states are enabled by noise, or, in spatially extended systems, may occur due to the traveling wave propagation. We analyze a one-dimensional bistable stochastic birth-death process by means of potential and temperature fields. The potential is defined by the deterministic limit of the process, while the temperature field is governed by noise. The stable steady state in which the potential has its global minimum defines the global deterministic attractor. For the stochastic system, in the low noise limit, the stationary probability distribution becomes unimodal, concentrated in one of two stable steady states, defined in this study as the global stochastic attractor. Interestingly, these two attractors may be located in different steady states. This observation suggests that the asymptotic behavior of spatially extended stochastic systems depends on the substrate diffusivity and size of the reactor. We confirmed this hypothesis within kinetic Monte Carlo simulations of a bistable reaction- diffusion model on the hexagonal lattice. In particular, we found that although the kinase-phosphatase system remains inactive in a small domain, the activatory traveling wave may propagate when a larger domain is considered.


Asunto(s)
Modelos Biológicos , Monoéster Fosfórico Hidrolasas/metabolismo , Fosfotransferasas/metabolismo , Procesos Estocásticos , Activación Enzimática , Cadenas de Markov , Método de Montecarlo , Temperatura , Termodinámica
20.
J Theor Biol ; 295: 116-24, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22123371

RESUMEN

The MAPK cascades are principal kinase transduction pathways in eukaryotic cells. This family includes RAF/ERK, JNK, and p38 pathways. In the MAPK cascade, the signal is transmitted through three layers of sequentially activated kinases, MAP3K, MAP2K, and MAPK. The latter two kinases require dual phosphorylation for activation. The dual phosphorylation requirement has been implicated in bringing about bistability and switch-like responses in the cascade. MAPK signaling has been known to involve scaffolds-multidomain proteins that can assemble protein complexes; in this case the three MAPK components. Scaffolds are thought to increase the specificity of signaling by pairing enzymes and substrates. Scaffolds have been shown to biphasically control the response (the level of activated MAPK) and amplify it at a certain scaffold concentration range. In order to understand the interplay of scaffolding and multisite phosphorylation, in this study we analyze simplified MAPK signaling models in which we assume that either mono- or double phosphorylation of MAP2K and MAPK is required for activation. We demonstrate that the requirement for double phosphorylation directs signaling through scaffolds. In the hypothetical scenario in which mono-phosphorylation suffices for kinase activity, the presence of scaffolds has little effect on the response. This suggests that double phosphorylation in MAPK pathways, although not as efficient as mono-phosphorylation, evolved together with scaffolds to assure the tighter control and higher specificity in signaling, by enabling scaffolds to function as response amplifiers.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Modelos Biológicos , Animales , Citoplasma/enzimología , Células Eucariotas/enzimología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA