Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(33): e2206888119, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35960842

RESUMEN

Self-organized pattern formation is vital for many biological processes. Reaction-diffusion models have advanced our understanding of how biological systems develop spatial structures, starting from homogeneity. However, biological processes inherently involve multiple spatial and temporal scales and transition from one pattern to another over time, rather than progressing from homogeneity to a pattern. To deal with such multiscale systems, coarse-graining methods are needed that allow the dynamics to be reduced to the relevant degrees of freedom at large scales, but without losing information about the patterns at small scales. Here, we present a semiphenomenological approach which exploits mass conservation in pattern formation, and enables reconstruction of information about patterns from the large-scale dynamics. The basic idea is to partition the domain into distinct regions (coarse grain) and determine instantaneous dispersion relations in each region, which ultimately inform about local pattern-forming instabilities. We illustrate our approach by studying the Min system, a paradigmatic model for protein pattern formation. By performing simulations, we first show that the Min system produces multiscale patterns in a spatially heterogeneous geometry. This prediction is confirmed experimentally by in vitro reconstitution of the Min system. Using a recently developed theoretical framework for mass-conserving reaction-diffusion systems, we show that the spatiotemporal evolution of the total protein densities on large scales reliably predicts the pattern-forming dynamics. Our approach provides an alternative and versatile theoretical framework for complex systems where analytical coarse-graining methods are not applicable, and can, in principle, be applied to a wide range of systems with an underlying conservation law.


Asunto(s)
Adenosina Trifosfatasas , Proteínas de Ciclo Celular , Proteínas de Escherichia coli , Adenosina Trifosfatasas/química , Proteínas de Ciclo Celular/química , Difusión , Proteínas de Escherichia coli/química , Modelos Teóricos
2.
Phys Rev Lett ; 126(10): 104101, 2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33784126

RESUMEN

Wavelength selection in reaction-diffusion systems can be understood as a coarsening process that is interrupted by counteracting processes at certain wavelengths. We first show that coarsening in mass-conserving systems is driven by self-amplifying mass transport between neighboring high-density domains. We derive a general coarsening criterion and show that coarsening is generically uninterrupted in two-component systems that conserve mass. The theory is then generalized to study interrupted coarsening and anticoarsening due to weakly broken mass conservation, providing a general path to analyze wavelength selection in pattern formation far from equilibrium.

3.
Proc Natl Acad Sci U S A ; 115(18): 4553-4558, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29666276

RESUMEN

Protein patterning is vital for many fundamental cellular processes. This raises two intriguing questions: Can such intrinsically complex processes be reduced to certain core principles and, if so, what roles do the molecular details play in individual systems? A prototypical example for protein patterning is the bacterial Min system, in which self-organized pole-to-pole oscillations of MinCDE proteins guide the cell division machinery to midcell. These oscillations are based on cycling of the ATPase MinD and its activating protein MinE between the membrane and the cytoplasm. Recent biochemical evidence suggests that MinE undergoes a reversible, MinD-dependent conformational switch from a latent to a reactive state. However, the functional relevance of this switch for the Min network and pattern formation remains unclear. By combining mathematical modeling and in vitro reconstitution of mutant proteins, we dissect the two aspects of MinE's switch, persistent membrane binding and a change in MinE's affinity for MinD. Our study shows that the MinD-dependent change in MinE's binding affinity for MinD is essential for patterns to emerge over a broad and physiological range of protein concentrations. Mechanistically, our results suggest that conformational switching of an ATPase-activating protein can lead to the spatial separation of its distinct functional states and thereby confer robustness on an intracellular protein network with vital roles in bacterial cell division.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiología , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/fisiología , Adenosina Trifosfato/metabolismo , División Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de la Membrana/metabolismo , Modelos Teóricos , Conformación Molecular , Unión Proteica/fisiología
4.
Bull Math Biol ; 82(10): 136, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33057872

RESUMEN

Reaction-diffusion processes across layered media arise in several scientific domains such as pattern-forming E. coli on agar substrates, epidermal-mesenchymal coupling in development, and symmetry-breaking in cell polarization. We develop a modeling framework for bilayer reaction-diffusion systems and relate it to a range of existing models. We derive conditions for diffusion-driven instability of a spatially homogeneous equilibrium analogous to the classical conditions for a Turing instability in the simplest nontrivial setting where one domain has a standard reaction-diffusion system, and the other permits only diffusion. Due to the transverse coupling between these two regions, standard techniques for computing eigenfunctions of the Laplacian cannot be applied, and so we propose an alternative method to compute the dispersion relation directly. We compare instability conditions with full numerical simulations to demonstrate impacts of the geometry and coupling parameters on patterning, and explore various experimentally relevant asymptotic regimes. In the regime where the first domain is suitably thin, we recover a simple modulation of the standard Turing conditions, and find that often the broad impact of the diffusion-only domain is to reduce the ability of the system to form patterns. We also demonstrate complex impacts of this coupling on pattern formation. For instance, we exhibit non-monotonicity of pattern-forming instabilities with respect to geometric and coupling parameters, and highlight an instability from a nontrivial interaction between kinetics in one domain and diffusion in the other. These results are valuable for informing design choices in applications such as synthetic engineering of Turing patterns, but also for understanding the role of stratified media in modulating pattern-forming processes in developmental biology and beyond.


Asunto(s)
Modelos Biológicos , Animales , Biología Evolutiva , Difusión , Escherichia coli , Humanos , Cinética , Conceptos Matemáticos
5.
Proc Natl Acad Sci U S A ; 113(3): 548-53, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26739566

RESUMEN

Protein patterns are known to adapt to cell shape and serve as spatial templates that choreograph downstream processes like cell polarity or cell division. However, how can pattern-forming proteins sense and respond to the geometry of a cell, and what mechanistic principles underlie pattern formation? Current models invoke mechanisms based on dynamic instabilities arising from nonlinear interactions between proteins but neglect the influence of the spatial geometry itself. Here, we show that patterns can emerge as a direct result of adaptation to cell geometry, in the absence of dynamical instability. We present a generic reaction module that allows protein densities robustly to adapt to the symmetry of the spatial geometry. The key component is an NTPase protein that cycles between nucleotide-dependent membrane-bound and cytosolic states. For elongated cells, we find that the protein dynamics generically leads to a bipolar pattern, which vanishes as the geometry becomes spherically symmetrical. We show that such a reaction module facilitates universal adaptation to cell geometry by sensing the local ratio of membrane area to cytosolic volume. This sensing mechanism is controlled by the membrane affinities of the different states. We apply the theory to explain AtMinD bipolar patterns in [Formula: see text] EcMinDE Escherichia coli. Due to its generic nature, the mechanism could also serve as a hitherto-unrecognized spatial template in many other bacterial systems. Moreover, the robustness of the mechanism enables self-organized optimization of protein patterns by evolutionary processes. Finally, the proposed module can be used to establish geometry-sensitive protein gradients in synthetic biological systems.


Asunto(s)
Proteínas/química , Polaridad Celular , Citosol/metabolismo , Modelos Biológicos , Conformación Proteica
6.
Mol Syst Biol ; 12(6): 873, 2016 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-27279643

RESUMEN

Cells owe their internal organization to self-organized protein patterns, which originate and adapt to growth and external stimuli via a process that is as complex as it is little understood. Here, we study the emergence, stability, and state transitions of multistable Min protein oscillation patterns in live Escherichia coli bacteria during growth up to defined large dimensions. De novo formation of patterns from homogenous starting conditions is observed and studied both experimentally and in simulations. A new theoretical approach is developed for probing pattern stability under perturbations. Quantitative experiments and simulations show that, once established, Min oscillations tolerate a large degree of intracellular heterogeneity, allowing distinctly different patterns to persist in different cells with the same geometry. Min patterns maintain their axes for hours in experiments, despite imperfections, expansion, and changes in cell shape during continuous cell growth. Transitions between multistable Min patterns are found to be rare events induced by strong intracellular perturbations. The instances of multistability studied here are the combined outcome of boundary growth and strongly nonlinear kinetics, which are characteristic of the reaction-diffusion patterns that pervade biology at many scales.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/crecimiento & desarrollo , Simulación por Computador , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Modelos Biológicos , Estabilidad Proteica
7.
Nat Commun ; 14(1): 6504, 2023 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-37845215

RESUMEN

How can a self-organized cellular function evolve, adapt to perturbations, and acquire new sub-functions? To make progress in answering these basic questions of evolutionary cell biology, we analyze, as a concrete example, the cell polarity machinery of Saccharomyces cerevisiae. This cellular module exhibits an intriguing resilience: it remains operational under genetic perturbations and recovers quickly and reproducibly from the deletion of one of its key components. Using a combination of modeling, conceptual theory, and experiments, we propose that multiple, redundant self-organization mechanisms coexist within the protein network underlying cell polarization and are responsible for the module's resilience and adaptability. Based on our mechanistic understanding of polarity establishment, we hypothesize that scaffold proteins, by introducing new connections in the existing network, can increase the redundancy of mechanisms and thus increase the evolvability of other network components. Moreover, our work gives a perspective on how a complex, redundant cellular module might have evolved from a more rudimental ancestral form.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomycetales , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomycetales/genética , Saccharomycetales/metabolismo , Variaciones en el Número de Copia de ADN , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Polaridad Celular/genética
9.
Nat Commun ; 12(1): 3312, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34083526

RESUMEN

Self-organisation of Min proteins is responsible for the spatial control of cell division in Escherichia coli, and has been studied both in vivo and in vitro. Intriguingly, the protein patterns observed in these settings differ qualitatively and quantitatively. This puzzling dichotomy has not been resolved to date. Using reconstituted proteins in laterally wide microchambers with a well-controlled height, we experimentally show that the Min protein dynamics on the membrane crucially depend on the micro chamber height due to bulk concentration gradients orthogonal to the membrane. A theoretical analysis shows that in vitro patterns at low microchamber height are driven by the same lateral oscillation mode as pole-to-pole oscillations in vivo. At larger microchamber height, additional vertical oscillation modes set in, marking the transition to a qualitatively different in vitro regime. Our work reveals the qualitatively different mechanisms of mass transport that govern Min protein-patterns for different bulk heights and thus shows that Min patterns in cells are governed by a different mechanism than those in vitro.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Proteínas de la Membrana/metabolismo , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Transporte Biológico Activo , Proteínas de Ciclo Celular/metabolismo , División Celular , Membrana Celular/metabolismo , Polaridad Celular , Escherichia coli/metabolismo , Escherichia coli/ultraestructura , Técnicas In Vitro , Modelos Biológicos , Dinámicas no Lineales
10.
Nat Commun ; 11(1): 539, 2020 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-31988277

RESUMEN

In the Caenorhabditis elegans zygote, PAR protein patterns, driven by mutual anatagonism, determine the anterior-posterior axis and facilitate the redistribution of proteins for the first cell division. Yet, the factors that determine the selection of the polarity axis remain unclear. We present a reaction-diffusion model in realistic cell geometry, based on biomolecular reactions and accounting for the coupling between membrane and cytosolic dynamics. We find that the kinetics of the phosphorylation-dephosphorylation cycle of PARs and the diffusive protein fluxes from the cytosol towards the membrane are crucial for the robust selection of the anterior-posterior axis for polarisation. The local ratio of membrane surface to cytosolic volume is the main geometric cue that initiates pattern formation, while the choice of the long-axis for polarisation is largely determined by the length of the aPAR-pPAR interface, and mediated by processes that minimise the diffusive fluxes of PAR proteins between cytosol and membrane.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Polaridad Celular , Animales , División Celular Asimétrica , Caenorhabditis elegans/citología , Caenorhabditis elegans/embriología , Biología Computacional , Citosol/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Desarrollo Embrionario , Cinética , Modelos Biológicos , Fosforilación , Transducción de Señal , Termodinámica
11.
Nat Commun ; 11(1): 5545, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33139718

RESUMEN

During development, cells gain positional information through the interpretation of dynamic morphogen gradients. A proposed mechanism for interpreting opposing morphogen gradients is mutual inhibition of downstream transcription factors, but isolating the role of this specific motif within a natural network remains a challenge. Here, we engineer a synthetic morphogen-induced mutual inhibition circuit in E. coli populations and show that mutual inhibition alone is sufficient to produce stable domains of gene expression in response to dynamic morphogen gradients, provided the spatial average of the morphogens falls within the region of bistability at the single cell level. When we add sender devices, the resulting patterning circuit produces theoretically predicted self-organised gene expression domains in response to a single gradient. We develop computational models of our synthetic circuits parameterised to timecourse fluorescence data, providing both a theoretical and experimental framework for engineering morphogen-induced spatial patterning in cell populations.


Asunto(s)
Escherichia coli/citología , Escherichia coli/crecimiento & desarrollo , Escherichia coli/genética , Simulación por Computador , Regulación Bacteriana de la Expresión Génica , Redes Reguladoras de Genes , Modelos Biológicos , Biología Sintética , Factores de Transcripción
12.
Elife ; 82019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31767054

RESUMEN

Although molecular self-organization and pattern formation are key features of life, only very few pattern-forming biochemical systems have been identified that can be reconstituted and studied in vitro under defined conditions. A systematic understanding of the underlying mechanisms is often hampered by multiple interactions, conformational flexibility and other complex features of the pattern forming proteins. Because of its compositional simplicity of only two proteins and a membrane, the MinDE system from Escherichia coli has in the past years been invaluable for deciphering the mechanisms of spatiotemporal self-organization in cells. Here, we explored the potential of reducing the complexity of this system even further, by identifying key functional motifs in the effector MinE that could be used to design pattern formation from scratch. In a combined approach of experiment and quantitative modeling, we show that starting from a minimal MinE-MinD interaction motif, pattern formation can be obtained by adding either dimerization or membrane-binding motifs. Moreover, we show that the pathways underlying pattern formation are recruitment-driven cytosolic cycling of MinE and recombination of membrane-bound MinE, and that these differ in their in vivo phenomenology.


Asunto(s)
Algoritmos , Proteínas de Escherichia coli/química , Modelos Teóricos , Motivos de Nucleótidos , Conformación Proteica , Multimerización de Proteína , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Secuencia de Bases , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , División Celular , Membrana Celular/química , Membrana Celular/metabolismo , Escherichia coli/citología , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo
14.
Cell Rep ; 1(6): 741-52, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-22813748

RESUMEN

Min-protein oscillations in Escherichia coli are characterized by the remarkable robustness with which spatial patterns dynamically adapt to variations of cell geometry. Moreover, adaption, and therefore proper cell division, is independent of temperature. These observations raise fundamental questions about the mechanisms establishing robust Min oscillations, and about the role of spatial cues, as they are at odds with present models. Here, we introduce a robust model based on experimental data, consistently explaining the mechanisms underlying pole-to-pole, striped, and circular patterns, as well as the observed temperature dependence of the oscillation period. Contrary to prior conjectures, the model predicts that MinD and cardiolipin domains are not colocalized. The transient sequestration of MinE and highly canalized transfer of MinD between polar zones are the key mechanisms underlying oscillations. MinD channeling enhances midcell localization and facilitates stripe formation, revealing the potential optimization process from which robust Min-oscillations originally arose.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/citología , Escherichia coli/metabolismo , Transducción de Señal , Adaptación Fisiológica , Cardiolipinas/química , Citosol/metabolismo , Hidrólisis , Modelos Biológicos , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA