Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cell ; 155(2): 448-61, 2013 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-24120141

RESUMEN

Highly networked signaling hubs are often associated with disease, but targeting them pharmacologically has largely been unsuccessful in the clinic because of their functional pleiotropy. Motivated by the hypothesis that a dynamic signaling code confers functional specificity, we investigated whether dynamic features may be targeted pharmacologically to achieve therapeutic specificity. With a virtual screen, we identified combinations of signaling hub topologies and dynamic signal profiles that are amenable to selective inhibition. Mathematical analysis revealed principles that may guide stimulus-specific inhibition of signaling hubs, even in the absence of detailed mathematical models. Using the NFκB signaling module as a test bed, we identified perturbations that selectively affect the response to cytokines or pathogen components. Together, our results demonstrate that the dynamics of signaling may serve as a pharmacological target, and we reveal principles that delineate the opportunities and constraints of developing stimulus-specific therapeutic agents aimed at pleiotropic signaling hubs.


Asunto(s)
Terapia Molecular Dirigida , Transducción de Señal/efectos de los fármacos , Animales , Simulación por Computador , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Humanos , FN-kappa B/metabolismo
2.
Mol Cell ; 47(1): 111-21, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22633953

RESUMEN

Besides activating NFκB by phosphorylating IκBs, IKKα/IKKß kinases are also involved in regulating metabolic insulin signaling, the mTOR pathway, Wnt signaling, and autophagy. How IKKß enzymatic activity is targeted to stimulus-specific substrates has remained unclear. We show here that NEMO, known to be essential for IKKß activation by inflammatory stimuli, is also a specificity factor that directs IKKß activity toward IκBα. Physical interaction and functional competition studies with mutant NEMO and IκB proteins indicate that NEMO functions as a scaffold to recruit IκBα to IKKß. Interestingly, expression of NEMO mutants that allow for IKKß activation by the cytokine IL-1, but fail to recruit IκBs, results in hyperphosphorylation of alternative IKKß substrates. Furthermore IKK's function in autophagy, which is independent of NFκB, is significantly enhanced without NEMO as IκB scaffold. Our work establishes a role for scaffolds such as NEMO in determining stimulus-specific signal transduction via the pleiotropic signaling hub IKK.


Asunto(s)
Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Transducción de Señal , Células 3T3 , Animales , Autofagia/efectos de los fármacos , Western Blotting , Células HEK293 , Humanos , Quinasa I-kappa B/genética , Proteínas I-kappa B/genética , Interleucina-1/farmacología , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Microscopía Fluorescente , Complejos Multiproteicos/metabolismo , Inhibidor NF-kappaB alfa , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica
3.
Biophys J ; 114(6): 1377-1388, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29590595

RESUMEN

Receptor internalization by endocytosis regulates diverse cellular processes, from the rate of nutrient uptake to the timescale of essential signaling events. The established view is that internalization is tightly controlled by specific protein-binding interactions. However, recent work suggests that physical aspects of receptors influence the process in ways that cannot be explained by biochemistry alone. Specifically, work from several groups suggests that increasing the steric bulk of receptors may inhibit their uptake by multiple types of trafficking vesicles. How do biochemical and biophysical factors work together to control internalization? Here, we show that receptor uptake is well described by a thermodynamic trade-off between receptor-vesicle binding energy and the entropic cost of confining receptors within endocytic vesicles. Specifically, using large ligands to acutely increase the size of engineered variants of the transferrin receptor, we demonstrate that an increase in the steric bulk of a receptor dramatically decreases its probability of uptake by clathrin-coated structures. Further, in agreement with a simple thermodynamic analysis, all data collapse onto a single trend relating fractional occupancy of the endocytic structure to fractional occupancy of the surrounding plasma membrane, independent of receptor size. This fundamental scaling law provides a simple tool for predicting the impact of receptor expression level, steric bulk, and the size of endocytic structures on receptor uptake. More broadly, this work suggests that bulky ligands could be used to drive the accumulation of specific receptors at the plasma membrane surface, providing a biophysical tool for targeted modulation of signaling and metabolism from outside the cell.


Asunto(s)
Endocitosis , Entropía , Modelos Biológicos , Ingeniería de Proteínas , Línea Celular , Humanos , Cinética , Ligandos , Unión Proteica
4.
PLoS Biol ; 11(9): e1001647, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24019758

RESUMEN

NF-κB plays a vital role in cellular immune and inflammatory response, survival, and proliferation by regulating the transcription of various genes involved in these processes. To activate transcription, RelA (a prominent NF-κB family member) interacts with transcriptional co-activators like CREB-binding protein (CBP) and its paralog p300 in addition to its cognate κB sites on the promoter/enhancer regions of DNA. The RelA:CBP/p300 complex is comprised of two components--first, DNA binding domain of RelA interacts with the KIX domain of CBP/p300, and second, the transcriptional activation domain (TAD) of RelA binds to the TAZ1 domain of CBP/p300. A phosphorylation event of a well-conserved RelA(Ser276) is prerequisite for the former interaction to occur and is considered a decisive factor for the overall RelA:CBP/p300 interaction. The role of the latter interaction in the transcription of RelA-activated genes remains unclear. Here we provide the solution structure of the latter component of the RelA:CBP complex by NMR spectroscopy. The structure reveals the folding of RelA-TA2 (a section of TAD) upon binding to TAZ1 through its well-conserved hydrophobic sites in a series of grooves on the TAZ1 surface. The structural analysis coupled with the mechanistic studies by mutational and isothermal calorimetric analyses allowed the design of RelA-mutants that selectively abrogated the two distinct components of the RelA:CBP/p300 interaction. Detailed studies of these RelA mutants using cell-based techniques, mathematical modeling, and genome-wide gene expression analysis showed that a major set of the RelA-activated genes, larger than previously believed, is affected by this interaction. We further show how the RelA:CBP/p300 interaction controls the nuclear response of NF-κB through the negative feedback loop of NF-κB pathway. Additionally, chromatin analyses of RelA target gene promoters showed constitutive recruitment of CBP/p300, thus indicating a possible role of CBP/p300 in recruitment of RelA to its target promoter sites.


Asunto(s)
Proteína p300 Asociada a E1A/metabolismo , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Fosfoproteínas/metabolismo , Factor de Transcripción ReIA/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Diferenciación Celular/genética , Línea Celular , Proliferación Celular , Proteína p300 Asociada a E1A/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/inmunología , Interacciones Hidrofóbicas e Hidrofílicas , Espectroscopía de Resonancia Magnética , Proteínas de la Membrana/genética , Ratones , Datos de Secuencia Molecular , FN-kappa B/genética , Fosfoproteínas/genética , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína , Factor de Transcripción ReIA/genética , Transcripción Genética
5.
Mol Cell ; 30(5): 649-56, 2008 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-18538663

RESUMEN

Cell differentiation requires the ability to detect and respond appropriately to a variety of extracellular signals. Here we investigate a differentiation switch induced by changes in the concentration of a single stimulus. Yeast cells exposed to high doses of mating pheromone undergo cell division arrest. Cells at intermediate doses become elongated and divide in the direction of a pheromone gradient (chemotropic growth). Either of the pheromone-responsive MAP kinases, Fus3 and Kss1, promotes cell elongation, but only Fus3 promotes chemotropic growth. Whereas Kss1 is activated rapidly and with a graded dose-response profile, Fus3 is activated slowly and exhibits a steeper dose-response relationship (ultrasensitivity). Fus3 activity requires the scaffold protein Ste5; when binding to Ste5 is abrogated, Fus3 behaves like Kss1, and the cells no longer respond to a gradient or mate efficiently with distant partners. We propose that scaffold proteins serve to modulate the temporal and dose-response behavior of the MAP kinase.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/enzimología , Diferenciación Celular/efectos de los fármacos , Activación Enzimática , Feromonas/farmacología , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/crecimiento & desarrollo
6.
Immunol Rev ; 246(1): 221-38, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22435558

RESUMEN

Mathematical modeling has proved to be a critically important approach in the study of many complex networks and dynamic systems in physics, engineering, chemistry, and biology. The nuclear factor κB (NF-κB) system consists of more than 50 proteins and protein complexes and is both a highly networked and dynamic system. To date, mathematical modeling has only addressed a small fraction of the molecular species and their regulation, but when employed in conjunction with experimental analysis has already led to important insights. Here, we provide a personal account of studying how the NF-κB signaling system functions using mathematical descriptions of the molecular mechanisms. We focus on the insights gained about some of the key regulatory components: the control of the steady state, the signaling dynamics, and signaling crosstalk. We also discuss the biological relevance of these regulatory systems properties.


Asunto(s)
Modelos Biológicos , FN-kappa B/metabolismo , Transducción de Señal , Animales , Humanos , Subunidad p50 de NF-kappa B/metabolismo , Factor de Transcripción ReIA/metabolismo
7.
Biophys J ; 105(1): 231-41, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23823243

RESUMEN

The transcription factor NFκB, a key component of the immune system, shows intricate stimulus-specific temporal dynamics. Those dynamics are thought to play a role in controlling the physiological response to cytokines and pathogens. Biochemical evidence suggests that the NFκB inducing kinase, IKK, a signaling hub onto which many signaling pathways converge, is regulated via a regulatory cycle comprising a poised, an active, and an inactive state. We hypothesize that it operates as a modulator of signal dynamics, actively reshaping the signals generated at the receptor proximal level. Here we show that a regulatory cycle can function in at least three dynamical regimes, tunable by regulating a single kinetic parameter. In particular, the simplest three-state regulatory cycle can generate signals with two well-defined phases, each with distinct coding capabilities in terms of the information they can carry about the stimulus. We also demonstrate that such a kinase cycle can function as a signal categorizer classifying diverse incoming signals into outputs with a limited set of temporal activity profiles. Finally, we discuss the extension of the results to other regulatory motifs that could be understood in terms of the regimes of the three-state cycle.


Asunto(s)
Quinasa I-kappa B/metabolismo , Modelos Biológicos , Transducción de Señal , Activación Enzimática
8.
Adv Ther ; 39(7): 3225-3247, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35581423

RESUMEN

INTRODUCTION: Physicians are often required to make treatment decisions for patients with Crohn's disease on the basis of limited objective information about the state of the patient's gastrointestinal tissue while aiming to achieve mucosal healing. Tools to predict changes in mucosal health with treatment are needed. We evaluated a computational approach integrating a mechanistic model of Crohn's disease with a responder classifier to predict temporal changes in mucosal health. METHODS: A hybrid mechanistic-statistical platform was developed to predict biomarker and tissue health time courses in patients with Crohn's disease. Eligible patients from the VERSIFY study (n = 69) were classified into archetypical response cohorts using a decision tree based on early treatment data and baseline characteristics. A virtual patient matching algorithm assigned a digital twin to each patient from their corresponding response cohort. The digital twin was used to forecast response to treatment using the mechanistic model. RESULTS: The responder classifier predicted endoscopic remission and mucosal healing for treatment with vedolizumab over 26 weeks, with overall sensitivities of 80% and 75% and overall specificities of 69% and 70%, respectively. Predictions for changes in tissue damage over time in the validation set (n = 31), a measure of the overall performance of the platform, were considered good (at least 70% of data points matched), fair (at least 50%), and poor (less than 50%) for 71%, 23%, and 6% of patients, respectively. CONCLUSION: Hybrid computational tools including mechanistic components represent a promising form of decision support that can predict outcomes and patient progress in Crohn's disease.


Asunto(s)
Enfermedad de Crohn , Estudios de Cohortes , Enfermedad de Crohn/complicaciones , Enfermedad de Crohn/tratamiento farmacológico , Humanos , Mucosa Intestinal , Resultado del Tratamiento , Cicatrización de Heridas
9.
Curr Biol ; 17(8): 659-67, 2007 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-17363249

RESUMEN

BACKGROUND: A common property of signal transduction systems is that they rapidly lose their ability to respond to a given stimulus. For instance in yeast, the mitogen-activated protein (MAP) kinase Hog1 is activated and inactivated within minutes, even when the osmotic-stress stimulus is sustained. RESULTS: Here, we used a combination of experimental and computational analyses to investigate the dynamic behavior of Hog1 activation in vivo. Computational modeling suggested that a negative-feedback loop operates early in the pathway and leads to rapid attenuation of Hog1 signaling. Experimental analysis revealed that the membrane-bound osmosensor Sho1 is phosphorylated by Hog1 and that phosphorylation occurs on Ser-166. Moreover, Sho1 exists in a homo-oligomeric complex, and phosphorylation by Hog1 promotes a transition from the oligomeric to monomeric state. A phosphorylation-site mutation (Sho1(S166E)) diminishes the formation of Sho1-oligomers, dampens activation of the Hog1 kinase, and impairs growth in high-salt or sorbitol conditions. CONCLUSIONS: These findings reveal a novel phosphorylation-dependent feedback loop leading to diminished cellular responses to an osmotic-stress stimulus.


Asunto(s)
Retroalimentación Fisiológica , Proteínas de la Membrana/metabolismo , Presión Osmótica , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Transducción de Señal , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Biología de Sistemas
10.
PLoS Comput Biol ; 4(10): e1000197, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18846202

RESUMEN

The cellular response elicited by an environmental cue typically varies with the strength of the stimulus. For example, in the yeast Saccharomyces cerevisiae, the concentration of mating pheromone determines whether cells undergo vegetative growth, chemotropic growth, or mating. This implies that the signaling pathways responsible for detecting the stimulus and initiating a response must transmit quantitative information about the intensity of the signal. Our previous experimental results suggest that yeast encode pheromone concentration as the duration of the transmitted signal. Here we use mathematical modeling to analyze possible biochemical mechanisms for performing this "dose-to-duration" conversion. We demonstrate that modulation of signal duration increases the range of stimulus concentrations for which dose-dependent responses are possible; this increased dynamic range produces the counterintuitive result of "signaling beyond saturation" in which dose-dependent responses are still possible after apparent saturation of the receptors. We propose a mechanism for dose-to-duration encoding in the yeast pheromone pathway that is consistent with current experimental observations. Most previous investigations of information processing by signaling pathways have focused on amplitude encoding without considering temporal aspects of signal transduction. Here we demonstrate that dose-to-duration encoding provides cells with an alternative mechanism for processing and transmitting quantitative information about their surrounding environment. The ability of signaling pathways to convert stimulus strength into signal duration results directly from the nonlinear nature of these systems and emphasizes the importance of considering the dynamic properties of signaling pathways when characterizing their behavior. Understanding how signaling pathways encode and transmit quantitative information about the external environment will not only deepen our understanding of these systems but also provide insight into how to reestablish proper function of pathways that have become dysregulated by disease.


Asunto(s)
Modelos Biológicos , Transducción de Señal/fisiología , Biología Computacional , Relación Dosis-Respuesta a Droga , Retroalimentación Fisiológica , Feromonas/administración & dosificación , Feromonas/fisiología , Receptores de Superficie Celular/efectos de los fármacos , Receptores de Superficie Celular/fisiología , Receptores de Feromonas/efectos de los fármacos , Receptores de Feromonas/fisiología , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
11.
Cell Signal ; 55: 17-25, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30543861

RESUMEN

Small molecule approaches targeting the nuclear factor kappa B (NF-kB) pathway, a regulator of inflammation, have thus far proven unsuccessful in the clinic in part due to the complex pleiotropic nature of this network. Downstream effects depend on multiple factors including stimulus-specific temporal patterns of NF-kB activity. Despite considerable advances, genome-level impact of changes in temporal NF-kB activity caused by inhibitors and their stimulus dependency remains unexplored. This study evaluates the effects of pathway inhibitors on early NF-κB activity and downstream gene transcription. 3T3 fibroblasts were treated with SC-514, an inhibitor targeted to the NF-kB pathway, prior to stimulation with interleukin 1 beta (IL-1ß) or tumor necrosis factor alpha (TNF-α). Stimulus induced NF-κB activation was quantified using immunofluorescence imaging over 90-minutes and gene expression tracked over 6-hours using mRNA TagSeq. When stimulated with IL-1ß or TNF-α, significant differences (P < 0.05, two-way ANOVA), were observed in the temporal profiles of NF-κB activation between treated and untreated cells. Increasing numbers of differentially expressed genes (P < 0.01) were observed at higher inhibitor concentrations. Individual gene expression profiles varied in an inhibitor concentration and stimulus-dependent manner. The results in this study demonstrate small molecule inhibitors acting on pleiotropic pathway components can alter signal dynamics in a stimulus-dependent manner and affect gene response in complex ways.


Asunto(s)
Quinasa I-kappa B/antagonistas & inhibidores , Inflamación/metabolismo , Interleucina-1beta/metabolismo , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Células 3T3 , Animales , Expresión Génica , Regulación de la Expresión Génica , Ratones , Transducción de Señal
13.
Cell Syst ; 4(3): 330-343.e5, 2017 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-28237795

RESUMEN

Combinatorial control of gene expression is presumed to be mediated by molecular interactions between coincident transcription factors (TFs). While information on the genome-wide locations of TFs is available, the genes they regulate and whether they function combinatorially often remain open questions. Here, we developed a mechanistic, rather than statistical, modeling approach to elucidate TF control logic from gene expression data. Applying this approach to hundreds of genes in 85 datasets measuring the transcriptional responses of murine fibroblasts and macrophages to cytokines and pathogens, we found that stimulus-responsive TFs generally function sequentially in logical OR gates or singly. Logical AND gates were found between NF-κB-responsive mRNA synthesis and MAPKp38-responsive control of mRNA half-life, but not between temporally coincident TFs. Our analyses identified the functional target genes of each of the pathogen-responsive TFs and prompt a revision of the conceptual underpinnings of combinatorial control of gene expression to include sequentially acting molecular mechanisms that govern mRNA synthesis and decay.


Asunto(s)
Transcripción Genética/genética , Transcripción Genética/fisiología , Animales , Simulación por Computador , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Humanos , Modelos Biológicos , Unión Proteica , Factores de Transcripción/genética , Activación Transcripcional/genética , Activación Transcripcional/fisiología
14.
NPJ Syst Biol Appl ; 2: 16024, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28725478

RESUMEN

Transcription factor nuclear factor kappa B (NF-κB) shows cooperative switch-like activation followed by prolonged oscillatory nuclear translocation in response to extracellular stimuli. These dynamics are important for activation of the NF-κB transcriptional machinery, however, NF-κB activity regulated by coordinated actions of these dynamics has not been elucidated at the system level. Using a variety of B cells with artificially rewired NF-κB signaling networks, we show that oscillations and switch-like activation of NF-κB can be dissected and that, under some conditions, these two behaviors are separated upon antigen receptor activation. Comprehensive quantitative experiments and mathematical analysis showed that the functional role of switch activation in the NF-κB system is to overcome transient IKK (IκB kinase) activity to amplify nuclear translocation of NF-κB, thereby inducing the prolonged NF-κB oscillatory behavior necessary for target gene expression and B-cell activation.

15.
Artículo en Inglés | MEDLINE | ID: mdl-26015160

RESUMEN

Development and function of the immune system depends on cells exchanging information between themselves and with their environment. This information is processed and integrated by complex signal transduction and gene regulatory networks with rich temporal dynamics. A growing body of evidence points to a combination of network topology and temporal dynamics as a fundamental link between stimulus and function. Recent findings also bring cellular variability and stochastic events to the forefront as additional determinants of cell population responses to immune cues. In this article, we review examples of how the trinity of network topology, temporal dynamics, and cellular variability together determine the immune function. In particular we focus on Nuclear Factor kappa-B and T-cell receptor signaling networks as they have proven fertile ground for studying how function arises from the combination of topology, dynamics, and variability in a context of great clinical importance.


Asunto(s)
Redes Reguladoras de Genes/inmunología , Interacción Gen-Ambiente , FN-kappa B/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Animales , Humanos , FN-kappa B/genética , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal/genética
16.
J R Soc Interface ; 12(110): 0262, 2015 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-26311312

RESUMEN

The magnitude, duration and oscillation of cellular signalling pathway responses are often limited by negative feedback loops, defined as an 'activator-induced inhibitor' regulatory motif. Within the NFκB signalling pathway, a key negative feedback regulator is IκBα. We show here that, contrary to current understanding, NFκB-inducible expression is not sufficient for providing effective negative feedback. We then employ computational simulations of NFκB signalling to identify IκBα molecular properties that are critical for proper negative feedback control and test the resulting predictions in biochemical and single-cell live-imaging studies. We identified nuclear import and nuclear export of IκBα and the IκBα-NFκB complex, as well as the free IκBα half-life, as key determinants of post-induction repression of NFκB and the potential for subsequent reactivation. Our work emphasizes that negative feedback is an emergent systems property determined by multiple molecular and biophysical properties in addition to the required 'activator-induced inhibitor' relationship.


Asunto(s)
Simulación por Computador , Fibroblastos/metabolismo , Regulación de la Expresión Génica/fisiología , Quinasa I-kappa B/metabolismo , Modelos Biológicos , Transducción de Señal/fisiología , Secuencias de Aminoácidos , Animales , Fibroblastos/citología , Quinasa I-kappa B/genética , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo
17.
Science ; 344(6185): 760-4, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24833394

RESUMEN

A switchlike response in nuclear factor-κB (NF-κB) activity implies the existence of a threshold in the NF-κB signaling module. We show that the CARD-containing MAGUK protein 1 (CARMA1, also called CARD11)-TAK1 (MAP3K7)-inhibitor of NF-κB (IκB) kinase-ß (IKKß) module is a switch mechanism for NF-κB activation in B cell receptor (BCR) signaling. Experimental and mathematical modeling analyses showed that IKK activity is regulated by positive feedback from IKKß to TAK1, generating a steep dose response to BCR stimulation. Mutation of the scaffolding protein CARMA1 at serine-578, an IKKß target, abrogated not only late TAK1 activity, but also the switchlike activation of NF-κB in single cells, suggesting that phosphorylation of this residue accounts for the feedback.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Guanilato Ciclasa/metabolismo , Quinasa I-kappa B/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , FN-kappa B/agonistas , Receptores de Antígenos de Linfocitos B/metabolismo , Animales , Linfocitos B/metabolismo , Proteínas Adaptadoras de Señalización CARD/genética , Línea Celular , Pollos , Retroalimentación Fisiológica , Guanilato Ciclasa/genética , Quinasas Quinasa Quinasa PAM/genética , Ratones , Ratones Noqueados , Mutación , Fosforilación , Receptores de Antígenos de Linfocitos B/genética , Serina/genética , Serina/metabolismo , Transducción de Señal
18.
Sci Signal ; 4(186): ra54, 2011 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-21868361

RESUMEN

The maintenance and detection of signaling gradients are critical for proper development and cell migration. In single-cell organisms, gradient detection allows cells to orient toward a distant mating partner or nutrient source. Budding yeast expand their growth toward mating pheromone gradients through a process known as chemotropic growth. MATα cells secrete α-factor pheromone that stimulates chemotropism and mating differentiation in MATa cells and vice versa. Paradoxically, MATa cells secrete Bar1, a protease that degrades α-factor and that attenuates the mating response, yet is also required for efficient mating. We observed that MATa cells avoid each other during chemotropic growth. To explore this behavior, we developed a computational platform to simulate chemotropic growth. Our simulations indicated that the release of Bar1 enabled individual MATa cells to act as α-factor sinks. The simulations suggested that the resultant local reshaping of pheromone concentration created gradients that were directed away from neighboring MATa cells (self-avoidance) and that were increasingly amplified toward partners of the opposite sex during elongation. The behavior of Bar1-deficient cells in gradient chambers and mating assays supported these predictions from the simulations. Thus, budding yeast dynamically remodel their environment to ensure productive responses to an external stimulus and avoid nonproductive cell-cell interactions.


Asunto(s)
Péptidos/metabolismo , Proteolisis , Saccharomyces cerevisiae/metabolismo , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Factor de Apareamiento , Péptidos/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
19.
Curr Opin Genet Dev ; 20(6): 684-93, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20956081

RESUMEN

The health of organisms and cells depends on appropriate responses to diverse internal and external cues, stimuli, or challenges, such as changes in hormone or cytokine levels, or exposure to a pathogen. Cellular responses must be tailored to the identity and intensity of the stimulus and therefore intra-cellular signals must carry information about both. However, signaling mediators often form intricate networks that react to multiple stimuli yet manage to produce stimulus-specific responses. The multi-functionality ('functional pleiotropism') of signaling nodes suggests that biological networks have evolved ways of passing physiologically relevant stimulus information through shared channels. Increasing evidence supports the notion that this is achieved in part through temporal regulation of signaling mediators' activities. The present challenge is to identify the features of temporal activity profile that represent information about a given stimulus and understand how cells read the temporal codes to control their responses.


Asunto(s)
Espacio Intracelular , Transducción de Señal , Animales , Regulación de la Expresión Génica
20.
Biophys J ; 93(3): 806-21, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17513354

RESUMEN

We perform a systematic analysis of mechanisms of feedback regulation that underlie short-term adaptation in intracellular signaling systems. Upon receiving an external cue, these systems generate a transient response that quickly returns to basal levels even if the stimulus persists. Signaling pathways capable of short-term adaptation are found in systems as diverse as the high osmolarity response of yeast, gradient sensing in Dictyostelium, and the cytokine response in vertebrates. Using mathematical analysis and computational experiments, we compare different feedback architectures in terms of response amplitude and duration, ability to adapt, and response to variable stimulus levels. Our analysis reveals three important features of these systems: 1), multiple step signaling cascades improve sensitivity to low doses by an effect distinct from signal amplification; 2), some feedback architectures act as signal transducers converting stimulus strength into response duration; and 3), feedback deactivation acts as a dose-dependent switch between transient and sustained responses. Finally, we present characteristic features for each form of feedback regulation that can aid in their identification.


Asunto(s)
Retroalimentación , Modelos Teóricos , Activación Enzimática , Cinética , Matemática , Modelos Biológicos , Monoéster Fosfórico Hidrolasas/metabolismo , Fosfotransferasas/metabolismo , Transducción de Señal , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA