Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Chem Phys ; 159(18)2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-37937934

RESUMO

Stochastic chemical kinetics is a widely used formalism for studying stochasticity of chemical reactions inside single cells. Experimental studies of reaction networks are generally performed with cells that are part of a growing population, yet the population context is rarely taken into account when models are developed. Models that neglect the population context lose their validity whenever the studied system influences traits of cells that can be selected in the population, a property that naturally arises in the complex interplay between single-cell and population dynamics of cell fate decision systems. Here, we represent such systems as absorbing continuous-time Markov chains. We show that conditioning on non-absorption allows one to derive a modified master equation that tracks the time evolution of the expected population composition within a growing population. This allows us to derive consistent population dynamics models from a specification of the single-cell process. We use this approach to classify cell fate decision systems into two types that lead to different characteristic phases in emerging population dynamics. Subsequently, we deploy the gained insights to experimentally study a recurrent problem in biology: how to link plasmid copy number fluctuations and plasmid loss events inside single cells to growth of cell populations in dynamically changing environments.


Assuntos
Modelos Biológicos , Processos Estocásticos , Cadeias de Markov , Diferenciação Celular , Cinética , Simulação por Computador
2.
Phys Biol ; 20(5)2023 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-37343568

RESUMO

This study describes a method for controlling the production of protein in individual cells using stochastic models of gene expression. By combining modern microscopy platforms with optogenetic gene expression, experimentalists are able to accurately apply light to individual cells, which can induce protein production. Here we use a finite state projection based stochastic model of gene expression, along with Bayesian state estimation to control protein copy numbers within individual cells. We compare this method to previous methods that use population based approaches. We also demonstrate the ability of this control strategy to ameliorate discrepancies between the predictions of a deterministic model and stochastic switching system.


Assuntos
Proteínas , Humanos , Processos Estocásticos , Teorema de Bayes , Expressão Gênica
3.
J Math Biol ; 86(3): 43, 2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36745224

RESUMO

Cell-to-cell variability, born of stochastic chemical kinetics, persists even in large isogenic populations. In the study of single-cell dynamics this is typically accounted for. However, on the population level this source of heterogeneity is often sidelined to avoid the inevitable complexity it introduces. The homogeneous models used instead are more tractable but risk disagreeing with their heterogeneous counterparts and may thus lead to severely suboptimal control of bioproduction. In this work, we introduce a comprehensive mathematical framework for solving bioproduction optimal control problems in the presence of heterogeneity. We study population-level models in which such heterogeneity is retained, and propose order-reduction approximation techniques. The reduced-order models take forms typical of homogeneous bioproduction models, making them a useful benchmark by which to study the importance of heterogeneity. Moreover, the derivation from the heterogeneous setting sheds light on parameter selection in ways a direct homogeneous outlook cannot, and reveals the source of approximation error. With view to optimally controlling bioproduction in microbial communities, we ask the question: when does optimising the reduced-order models produce strategies that work well in the presence of population heterogeneity? We show that, in some cases, homogeneous approximations provide remarkably accurate surrogate models. Nevertheless, we also demonstrate that this is not uniformly true: overlooking the heterogeneity can lead to significantly suboptimal control strategies. In these cases, the heterogeneous tools and perspective are crucial to optimise bioproduction.


Assuntos
Modelos Biológicos , Humanos , Dinâmica Populacional
4.
Math Biosci ; 350: 108866, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35753520

RESUMO

Stochastic chemical kinetics at the single-cell level give rise to heterogeneous populations of cells even when all individuals are genetically identical. This heterogeneity can lead to nonuniform behaviour within populations, including different growth characteristics, cell-fate dynamics, and response to stimuli. Ultimately, these diverse behaviours lead to intricate population dynamics that are inherently multiscale: the population composition evolves based on population-level processes that interact with stochastically distributed single-cell states. Therefore, descriptions that account for this heterogeneity are essential to accurately model and control chemical processes. However, for real-world systems such models are computationally expensive to simulate, which can make optimisation problems, such as optimal control or parameter inference, prohibitively challenging. Here, we consider a class of multiscale population models that incorporate population-level mechanisms while remaining faithful to the underlying stochasticity at the single-cell level and the interplay between these two scales. To address the complexity, we study an order-reduction approximations based on the distribution moments. Since previous moment-closure work has focused on the single-cell kinetics, extending these techniques to populations models prompts us to revisit old observations as well as tackle new challenges. In this extended multiscale context, we encounter the previously established observation that the simplest closure techniques can lead to non-physical system trajectories. Despite their poor performance in some systems, we provide an example where these simple closures outperform more sophisticated closure methods in accurately, efficiently, and robustly solving the problem of optimal control of bioproduction in a microbial consortium model.


Assuntos
Modelos Biológicos , Simulação por Computador , Humanos , Dinâmica Populacional , Processos Estocásticos
5.
Nat Commun ; 13(1): 2199, 2022 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-35459274

RESUMO

Microscopy image analysis has recently made enormous progress both in terms of accuracy and speed thanks to machine learning methods and improved computational resources. This greatly facilitates the online adaptation of microscopy experimental plans using real-time information of the observed systems and their environments. Applications in which reactiveness is needed are multifarious. Here we report MicroMator, an open and flexible software for defining and driving reactive microscopy experiments. It provides a Python software environment and an extensible set of modules that greatly facilitate the definition of events with triggers and effects interacting with the experiment. We provide a pedagogic example performing dynamic adaptation of fluorescence illumination on bacteria, and demonstrate MicroMator's potential via two challenging case studies in yeast to single-cell control and single-cell recombination, both requiring real-time tracking and light targeting at the single-cell level.


Assuntos
Microscopia , Software , Processamento de Imagem Assistida por Computador/métodos , Aprendizado de Máquina , Saccharomyces cerevisiae
6.
PLoS Comput Biol ; 18(3): e1009950, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35303737

RESUMO

Understanding and characterising biochemical processes inside single cells requires experimental platforms that allow one to perturb and observe the dynamics of such processes as well as computational methods to build and parameterise models from the collected data. Recent progress with experimental platforms and optogenetics has made it possible to expose each cell in an experiment to an individualised input and automatically record cellular responses over days with fine time resolution. However, methods to infer parameters of stochastic kinetic models from single-cell longitudinal data have generally been developed under the assumption that experimental data is sparse and that responses of cells to at most a few different input perturbations can be observed. Here, we investigate and compare different approaches for calculating parameter likelihoods of single-cell longitudinal data based on approximations of the chemical master equation (CME) with a particular focus on coupling the linear noise approximation (LNA) or moment closure methods to a Kalman filter. We show that, as long as cells are measured sufficiently frequently, coupling the LNA to a Kalman filter allows one to accurately approximate likelihoods and to infer model parameters from data even in cases where the LNA provides poor approximations of the CME. Furthermore, the computational cost of filtering-based iterative likelihood evaluation scales advantageously in the number of measurement times and different input perturbations and is thus ideally suited for data obtained from modern experimental platforms. To demonstrate the practical usefulness of these results, we perform an experiment in which single cells, equipped with an optogenetic gene expression system, are exposed to various different light-input sequences and measured at several hundred time points and use parameter inference based on iterative likelihood evaluation to parameterise a stochastic model of the system.


Assuntos
Fenômenos Bioquímicos , Cinética , Probabilidade , Processos Estocásticos
7.
Proc Natl Acad Sci U S A ; 119(11): e2114438119, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35271387

RESUMO

SignificanceAt the single-cell level, biochemical processes are inherently stochastic. For many natural systems, the resulting cell-to-cell variability is exploited by microbial populations. In synthetic biology, however, the interplay of cell-to-cell variability and population processes such as selection or growth often leads to circuits not functioning as predicted by simple models. Here we show how multiscale stochastic kinetic models that simultaneously track single-cell and population processes can be obtained based on an augmentation of the chemical master equation. These models enable us to quantitatively predict complex population dynamics of a yeast optogenetic differentiation system from a specification of the circuit's components and to demonstrate how cell-to-cell variability can be exploited to purposefully create unintuitive circuit functionality.


Assuntos
Variação Biológica da População , Redes Reguladoras de Genes , Optogenética , Saccharomyces cerevisiae , Análise de Célula Única , Optogenética/métodos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Análise de Célula Única/métodos , Processos Estocásticos , Biologia Sintética
8.
Nat Commun ; 12(1): 5829, 2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34611168

RESUMO

Artificial microbial consortia seek to leverage division-of-labour to optimize function and possess immense potential for bioproduction. Co-culturing approaches, the preferred mode of generating a consortium, remain limited in their ability to give rise to stable consortia having finely tuned compositions. Here, we present an artificial differentiation system in budding yeast capable of generating stable microbial consortia with custom functionalities from a single strain at user-defined composition in space and in time based on optogenetically-driven genetic rewiring. Owing to fast, reproducible, and light-tunable dynamics, our system enables dynamic control of consortia composition in continuous cultures for extended periods. We further demonstrate that our system can be extended in a straightforward manner to give rise to consortia with multiple subpopulations. Our artificial differentiation strategy establishes a novel paradigm for the creation of complex microbial consortia that are simple to implement, precisely controllable, and versatile to use.


Assuntos
Saccharomyces cerevisiae/crescimento & desenvolvimento , Consórcios Microbianos/fisiologia
9.
PLoS Comput Biol ; 17(7): e1009214, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34319979

RESUMO

The chemical master equation and its continuum approximations are indispensable tools in the modeling of chemical reaction networks. These are routinely used to capture complex nonlinear phenomena such as multimodality as well as transient events such as first-passage times, that accurately characterise a plethora of biological and chemical processes. However, some mechanisms, such as heterogeneous cellular growth or phenotypic selection at the population level, cannot be represented by the master equation and thus have been tackled separately. In this work, we propose a unifying framework that augments the chemical master equation to capture such auxiliary dynamics, and we develop and analyse a numerical solver that accurately simulates the system dynamics. We showcase these contributions by casting a diverse array of examples from the literature within this framework and applying the solver to both match and extend previous studies. Analytical calculations performed for each example validate our numerical results and benchmark the solver implementation.


Assuntos
Modelos Biológicos , Modelos Químicos , Fenômenos Químicos , Biologia Computacional , Simulação por Computador , Regulação da Expressão Gênica , Cinética , Conceitos Matemáticos , Redes e Vias Metabólicas , Dinâmica não Linear , Fenótipo , Seleção Genética , Análise de Célula Única , Processos Estocásticos , Biologia de Sistemas
10.
Epidemics ; 34: 100428, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33444928

RESUMO

Contact tracing via smartphone applications is expected to be of major importance for maintaining control of the COVID-19 pandemic. However, viable deployment demands a minimal quarantine burden on the general public. That is, consideration must be given to unnecessary quarantining imposed by a contact tracing policy. Previous studies have modeled the role of contact tracing, but have not addressed how to balance these two competing needs. We propose a modeling framework that captures contact heterogeneity. This allows contact prioritization: contacts are only notified if they were acutely exposed to individuals who eventually tested positive. The framework thus allows us to address the delicate balance of preventing disease spread while minimizing the social and economic burdens of quarantine. This optimal contact tracing strategy is studied as a function of limitations in testing resources, partial technology adoption, and other intervention methods such as social distancing and lockdown measures. The framework is globally applicable, as the distribution describing contact heterogeneity is directly adaptable to any digital tracing implementation.


Assuntos
Busca de Comunicante/métodos , Pandemias/prevenção & controle , Quarentena , COVID-19 , Humanos , Aplicativos Móveis , Modelos Teóricos , Distanciamento Físico , Smartphone
11.
PLoS Comput Biol ; 15(9): e1007290, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31479447

RESUMO

Across diverse biological systems-ranging from neural networks to intracellular signaling and genetic regulatory networks-the information about changes in the environment is frequently encoded in the full temporal dynamics of the network nodes. A pressing data-analysis challenge has thus been to efficiently estimate the amount of information that these dynamics convey from experimental data. Here we develop and evaluate decoding-based estimation methods to lower bound the mutual information about a finite set of inputs, encoded in single-cell high-dimensional time series data. For biological reaction networks governed by the chemical Master equation, we derive model-based information approximations and analytical upper bounds, against which we benchmark our proposed model-free decoding estimators. In contrast to the frequently-used k-nearest-neighbor estimator, decoding-based estimators robustly extract a large fraction of the available information from high-dimensional trajectories with a realistic number of data samples. We apply these estimators to previously published data on Erk and Ca2+ signaling in mammalian cells and to yeast stress-response, and find that substantial amount of information about environmental state can be encoded by non-trivial response statistics even in stationary signals. We argue that these single-cell, decoding-based information estimates, rather than the commonly-used tests for significant differences between selected population response statistics, provide a proper and unbiased measure for the performance of biological signaling networks.


Assuntos
Biologia Computacional/métodos , Modelos Biológicos , Transdução de Sinais/fisiologia , Animais , Sinalização do Cálcio/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Mamíferos/fisiologia , Análise de Célula Única , Fatores de Tempo , Leveduras/fisiologia
12.
PLoS Comput Biol ; 15(7): e1007168, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31265463

RESUMO

Mathematical models have been used successfully at diverse scales of biological organization, ranging from ecology and population dynamics to stochastic reaction events occurring between individual molecules in single cells. Generally, many biological processes unfold across multiple scales, with mutations being the best studied example of how stochasticity at the molecular scale can influence outcomes at the population scale. In many other contexts, however, an analogous link between micro- and macro-scale remains elusive, primarily due to the challenges involved in setting up and analyzing multi-scale models. Here, we employ such a model to investigate how stochasticity propagates from individual biochemical reaction events in the bacterial innate immune system to the ecology of bacteria and bacterial viruses. We show analytically how the dynamics of bacterial populations are shaped by the activities of immunity-conferring enzymes in single cells and how the ecological consequences imply optimal bacterial defense strategies against viruses. Our results suggest that bacterial populations in the presence of viruses can either optimize their initial growth rate or their population size, with the first strategy favoring simple immunity featuring a single restriction modification system and the second strategy favoring complex bacterial innate immunity featuring several simultaneously active restriction modification systems.


Assuntos
Bactérias/imunologia , Bactérias/virologia , Bacteriófagos/imunologia , Biologia Computacional , Enzimas de Restrição-Modificação do DNA/imunologia , Ecossistema , Imunidade Inata , Consórcios Microbianos/imunologia , Modelos Biológicos , Modelos Imunológicos , Análise de Célula Única , Processos Estocásticos
13.
Nat Commun ; 8(1): 1535, 2017 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-29142298

RESUMO

Bacteria in groups vary individually, and interact with other bacteria and the environment to produce population-level patterns of gene expression. Investigating such behavior in detail requires measuring and controlling populations at the single-cell level alongside precisely specified interactions and environmental characteristics. Here we present an automated, programmable platform that combines image-based gene expression and growth measurements with on-line optogenetic expression control for hundreds of individual Escherichia coli cells over days, in a dynamically adjustable environment. This integrated platform broadly enables experiments that bridge individual and population behaviors. We demonstrate: (i) population structuring by independent closed-loop control of gene expression in many individual cells, (ii) cell-cell variation control during antibiotic perturbation, (iii) hybrid bio-digital circuits in single cells, and freely specifiable digital communication between individual bacteria. These examples showcase the potential for real-time integration of theoretical models with measurement and control of many individual cells to investigate and engineer microbial population behavior.


Assuntos
Biologia Computacional/métodos , Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Software , Escherichia coli/citologia , Genética Populacional , Modelos Genéticos , Optogenética
14.
J Chem Phys ; 146(12): 124122, 2017 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-28388123

RESUMO

Determining the sensitivity of certain system states or outputs to variations in parameters facilitates our understanding of the inner working of that system and is an essential design tool for the de novo construction of robust systems. In cell biology, the output of interest is often the response of a certain reaction network to some input (e.g., stressors or nutrients) and one aims to quantify the sensitivity of this response in the presence of parameter heterogeneity. We argue that for such applications, parametric sensitivities in their standard form do not paint a complete picture of a system's robustness since one assumes that all cells in the population have the same parameters and are perturbed in the same way. Here, we consider stochasticreaction networks in which the parameters are randomly distributed over the population and propose a new sensitivity index that captures the robustness of system outputs upon changes in the characteristics of the parameter distribution, rather than the parameters themselves. Subsequently, we make use of Girsanov's likelihood ratio method to construct a Monte Carlo estimator of this sensitivity index. However, it turns out that this estimator has an exceedingly large variance. To overcome this problem, we propose a novel estimation algorithm that makes use of a marginalization of the path distribution of stochasticreaction networks and leads to Rao-Blackwellized estimators with reduced variance.

15.
Biosystems ; 149: 15-25, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27461396

RESUMO

Continuous-time Markov chain (CTMC) models have become a central tool for understanding the dynamics of complex reaction networks and the importance of stochasticity in the underlying biochemical processes. When such models are employed to answer questions in applications, in order to ensure that the model provides a sufficiently accurate representation of the real system, it is of vital importance that the model parameters are inferred from real measured data. This, however, is often a formidable task and all of the existing methods fail in one case or the other, usually because the underlying CTMC model is high-dimensional and computationally difficult to analyze. The parameter inference methods that tend to scale best in the dimension of the CTMC are based on so-called moment closure approximations. However, there exists a large number of different moment closure approximations and it is typically hard to say a priori which of the approximations is the most suitable for the inference procedure. Here, we propose a moment-based parameter inference method that automatically chooses the most appropriate moment closure method. Accordingly, contrary to existing methods, the user is not required to be experienced in moment closure techniques. In addition to that, our method adaptively changes the approximation during the parameter inference to ensure that always the best approximation is used, even in cases where different approximations are best in different regions of the parameter space.


Assuntos
Fenômenos Bioquímicos , Redes Reguladoras de Genes , Modelos Teóricos , Animais , Fenômenos Bioquímicos/fisiologia , Humanos , Cadeias de Markov , Processos Estocásticos
16.
Proc Natl Acad Sci U S A ; 112(26): 8148-53, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26085136

RESUMO

Systems biology rests on the idea that biological complexity can be better unraveled through the interplay of modeling and experimentation. However, the success of this approach depends critically on the informativeness of the chosen experiments, which is usually unknown a priori. Here, we propose a systematic scheme based on iterations of optimal experiment design, flow cytometry experiments, and Bayesian parameter inference to guide the discovery process in the case of stochastic biochemical reaction networks. To illustrate the benefit of our methodology, we apply it to the characterization of an engineered light-inducible gene expression circuit in yeast and compare the performance of the resulting model with models identified from nonoptimal experiments. In particular, we compare the parameter posterior distributions and the precision to which the outcome of future experiments can be predicted. Moreover, we illustrate how the identified stochastic model can be used to determine light induction patterns that make either the average amount of protein or the variability in a population of cells follow a desired profile. Our results show that optimal experiment design allows one to derive models that are accurate enough to precisely predict and regulate the protein expression in heterogeneous cell populations over extended periods of time.


Assuntos
Regulação da Expressão Gênica , Luz , Processos Estocásticos , Biologia de Sistemas
17.
J Chem Phys ; 143(24): 244103, 2015 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-26723647

RESUMO

Many stochastic models of biochemical reaction networks contain some chemical species for which the number of molecules that are present in the system can only be finite (for instance due to conservation laws), but also other species that can be present in arbitrarily large amounts. The prime example of such networks are models of gene expression, which typically contain a small and finite number of possible states for the promoter but an infinite number of possible states for the amount of mRNA and protein. One of the main approaches to analyze such models is through the use of equations for the time evolution of moments of the chemical species. Recently, a new approach based on conditional moments of the species with infinite state space given all the different possible states of the finite species has been proposed. It was argued that this approach allows one to capture more details about the full underlying probability distribution with a smaller number of equations. Here, I show that the result that less moments provide more information can only stem from an unnecessarily complicated description of the system in the classical formulation. The foundation of this argument will be the derivation of moment equations that describe the complete probability distribution over the finite state space but only low-order moments over the infinite state space. I will show that the number of equations that is needed is always less than what was previously claimed and always less than the number of conditional moment equations up to the same order. To support these arguments, a symbolic algorithm is provided that can be used to derive minimal systems of unconditional moment equations for models with partially finite state space.


Assuntos
Algoritmos , Proteínas/química , RNA Mensageiro/química , Redes Reguladoras de Genes , Cinética , Modelos Moleculares , Regiões Promotoras Genéticas , Proteínas/genética , RNA Mensageiro/genética , Processos Estocásticos
18.
J R Soc Interface ; 10(88): 20130588, 2013 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-23985733

RESUMO

Exploiting the information provided by the molecular noise of a biological process has proved to be valuable in extracting knowledge about the underlying kinetic parameters and sources of variability from single-cell measurements. However, quantifying this additional information a priori, to decide whether a single-cell experiment might be beneficial, is currently only possible in systems where either the chemical master equation is computationally tractable or a Gaussian approximation is appropriate. Here, we provide formulae for computing the information provided by measured means and variances from the first four moments and the parameter derivatives of the first two moments of the underlying process. For stochastic kinetic models for which these moments can be either computed exactly or approximated efficiently, the derived formulae can be used to approximate the information provided by single-cell distribution experiments. Based on this result, we propose an optimal experimental design framework which we employ to compare the utility of dual-reporter and perturbation experiments for quantifying the different noise sources in a simple model of gene expression. Subsequently, we compare the information content of a set of experiments which have been performed in an engineered light-switch gene expression system in yeast and show that well-chosen gene induction patterns may allow one to identify features of the system which remain hidden in unplanned experiments.


Assuntos
Simulação por Computador , Modelos Biológicos , Projetos de Pesquisa , Processos Estocásticos
19.
Proc Natl Acad Sci U S A ; 109(21): 8340-5, 2012 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-22566653

RESUMO

Recent computational studies indicate that the molecular noise of a cellular process may be a rich source of information about process dynamics and parameters. However, accessing this source requires stochastic models that are usually difficult to analyze. Therefore, parameter estimation for stochastic systems using distribution measurements, as provided for instance by flow cytometry, currently remains limited to very small and simple systems. Here we propose a new method that makes use of low-order moments of the measured distribution and thereby keeps the essential parts of the provided information, while still staying applicable to systems of realistic size. We demonstrate how cell-to-cell variability can be incorporated into the analysis obviating the need for the ubiquitous assumption that the measurements stem from a homogeneous cell population. We demonstrate the method for a simple example of gene expression using synthetic data generated by stochastic simulation. Subsequently, we use time-lapsed flow cytometry data for the osmo-stress induced transcriptional response in budding yeast to calibrate a stochastic model, which is then used as a basis for predictions. Our results show that measurements of the mean and the variance can be enough to determine the model parameters, even if the measured distributions are not well-characterized by low-order moments only--e.g., if they are bimodal.


Assuntos
Regulação Fúngica da Expressão Gênica/fisiologia , Glicerol/metabolismo , Modelos Genéticos , Saccharomyces cerevisiae/genética , Estresse Fisiológico/genética , Equilíbrio Hidroeletrolítico/genética , Simulação por Computador , Citometria de Fluxo , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas de Saccharomyces cerevisiae/genética , Transdução de Sinais/genética , Processos Estocásticos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA