Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
PLoS Comput Biol ; 20(7): e1011642, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38990984

RESUMO

The Virtual Epileptic Patient (VEP) refers to a computer-based representation of a patient with epilepsy that combines personalized anatomical data with dynamical models of abnormal brain activities. It is capable of generating spatio-temporal seizure patterns that resemble those recorded with invasive methods such as stereoelectro EEG data, allowing for the evaluation of clinical hypotheses before planning surgery. This study highlights the effectiveness of calibrating VEP models using a global optimization approach. The approach utilizes SaCeSS, a cooperative metaheuristic algorithm capable of parallel computation, to yield high-quality solutions without requiring excessive computational time. Through extensive benchmarking on synthetic data, our proposal successfully solved a set of different configurations of VEP models, demonstrating better scalability and superior performance against other parallel solvers. These results were further enhanced using a Bayesian optimization framework for hyperparameter tuning, with significant gains in terms of both accuracy and computational cost. Additionally, we added a scalable uncertainty quantification phase after model calibration, and used it to assess the variability in estimated parameters across different problems. Overall, this study has the potential to improve the estimation of pathological brain areas in drug-resistant epilepsy, thereby to inform the clinical decision-making process.


Assuntos
Algoritmos , Teorema de Bayes , Encéfalo , Biologia Computacional , Eletroencefalografia , Epilepsia , Modelos Neurológicos , Humanos , Epilepsia/fisiopatologia , Encéfalo/fisiopatologia , Eletroencefalografia/métodos , Biologia Computacional/métodos , Simulação por Computador , Rede Nervosa/fisiopatologia
2.
PLoS Comput Biol ; 20(1): e1011151, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38190398

RESUMO

The mammalian cell cycle is regulated by a well-studied but complex biochemical reaction system. Computational models provide a particularly systematic and systemic description of the mechanisms governing mammalian cell cycle control. By combining both state-of-the-art multiplexed experimental methods and powerful computational tools, this work aims at improving on these models along four dimensions: model structure, validation data, validation methodology and model reusability. We developed a comprehensive model structure of the full cell cycle that qualitatively explains the behaviour of human retinal pigment epithelial-1 cells. To estimate the model parameters, time courses of eight cell cycle regulators in two compartments were reconstructed from single cell snapshot measurements. After optimisation with a parallel global optimisation metaheuristic we obtained excellent agreements between simulations and measurements. The PEtab specification of the optimisation problem facilitates reuse of model, data and/or optimisation results. Future perturbation experiments will improve parameter identifiability and allow for testing model predictive power. Such a predictive model may aid in drug discovery for cell cycle-related disorders.


Assuntos
Descoberta de Drogas , Neurônios , Humanos , Animais , Divisão Celular , Ciclo Celular , Projetos de Pesquisa , Mamíferos
3.
Brief Bioinform ; 23(1)2022 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-34619769

RESUMO

Ordinary differential equation models are nowadays widely used for the mechanistic description of biological processes and their temporal evolution. These models typically have many unknown and nonmeasurable parameters, which have to be determined by fitting the model to experimental data. In order to perform this task, known as parameter estimation or model calibration, the modeller faces challenges such as poor parameter identifiability, lack of sufficiently informative experimental data and the existence of local minima in the objective function landscape. These issues tend to worsen with larger model sizes, increasing the computational complexity and the number of unknown parameters. An incorrectly calibrated model is problematic because it may result in inaccurate predictions and misleading conclusions. For nonexpert users, there are a large number of potential pitfalls. Here, we provide a protocol that guides the user through all the steps involved in the calibration of dynamic models. We illustrate the methodology with two models and provide all the code required to reproduce the results and perform the same analysis on new models. Our protocol provides practitioners and researchers in biological modelling with a one-stop guide that is at the same time compact and sufficiently comprehensive to cover all aspects of the problem.


Assuntos
Modelos Biológicos , Biologia de Sistemas , Calibragem , Biologia de Sistemas/métodos
4.
Bioinformatics ; 39(1)2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36416122

RESUMO

MOTIVATION: Dynamic mechanistic modelling in systems biology has been hampered by the complexity and variability associated with the underlying interactions, and by uncertain and sparse experimental measurements. Ensemble modelling, a concept initially developed in statistical mechanics, has been introduced in biological applications with the aim of mitigating those issues. Ensemble modelling uses a collection of different models compatible with the observed data to describe the phenomena of interest. However, since systems biology models often suffer from a lack of identifiability and observability, ensembles of models are particularly unreliable when predicting non-observable states. RESULTS: We present a strategy to assess and improve the reliability of a class of model ensembles. In particular, we consider kinetic models described using ordinary differential equations with a fixed structure. Our approach builds an ensemble with a selection of the parameter vectors found when performing parameter estimation with a global optimization metaheuristic. This technique enforces diversity during the sampling of parameter space and it can quantify the uncertainty in the predictions of state trajectories. We couple this strategy with structural identifiability and observability analysis, and when these tests detect possible prediction issues we obtain model reparameterizations that surmount them. The end result is an ensemble of models with the ability to predict the internal dynamics of a biological process. We demonstrate our approach with models of glucose regulation, cell division, circadian oscillations and the JAK-STAT signalling pathway. AVAILABILITY AND IMPLEMENTATION: The code that implements the methodology and reproduces the results is available at https://doi.org/10.5281/zenodo.6782638. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Modelos Biológicos , Biologia de Sistemas , Reprodutibilidade dos Testes , Biologia de Sistemas/métodos , Incerteza , Cinética
5.
Bioinformatics ; 39(11)2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37988145

RESUMO

MOTIVATION: One of the main causes hampering predictability during the model identification and automated design of gene circuits in synthetic biology is the effect of molecular noise. Stochasticity may significantly impact the dynamics and function of gene circuits, specially in bacteria and yeast due to low mRNA copy numbers. Standard stochastic simulation methods are too computationally costly in realistic scenarios to be applied to optimization-based design or parameter estimation. RESULTS: In this work, we present IDESS (Identification and automated Design of Stochastic gene circuitS), a software toolbox for optimization-based design and model identification of gene regulatory circuits in the stochastic regime. This software incorporates an efficient approximation of the Chemical Master Equation as well as a stochastic simulation algorithm-both with GPU and CPU implementations-combined with global optimization algorithms capable of solving Mixed Integer Nonlinear Programming problems. The toolbox efficiently addresses two types of problems relevant in systems and synthetic biology: the automated design of stochastic synthetic gene circuits, and the parameter estimation for model identification of stochastic gene regulatory networks. AVAILABILITY AND IMPLEMENTATION: IDESS runs under the MATLAB environment and it is available under GPLv3 license at https://doi.org/10.5281/zenodo.7788692.


Assuntos
Redes Reguladoras de Genes , Software , Simulação por Computador , Algoritmos , Biologia Sintética , Processos Estocásticos
6.
PLoS Comput Biol ; 19(10): e1011014, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37851682

RESUMO

Mechanistic dynamical models allow us to study the behavior of complex biological systems. They can provide an objective and quantitative understanding that would be difficult to achieve through other means. However, the systematic development of these models is a non-trivial exercise and an open problem in computational biology. Currently, many research efforts are focused on model discovery, i.e. automating the development of interpretable models from data. One of the main frameworks is sparse regression, where the sparse identification of nonlinear dynamics (SINDy) algorithm and its variants have enjoyed great success. SINDy-PI is an extension which allows the discovery of rational nonlinear terms, thus enabling the identification of kinetic functions common in biochemical networks, such as Michaelis-Menten. SINDy-PI also pays special attention to the recovery of parsimonious models (Occam's razor). Here we focus on biological models composed of sets of deterministic nonlinear ordinary differential equations. We present a methodology that, combined with SINDy-PI, allows the automatic discovery of structurally identifiable and observable models which are also mechanistically interpretable. The lack of structural identifiability and observability makes it impossible to uniquely infer parameter and state variables, which can compromise the usefulness of a model by distorting its mechanistic significance and hampering its ability to produce biological insights. We illustrate the performance of our method with six case studies. We find that, despite enforcing sparsity, SINDy-PI sometimes yields models that are unidentifiable. In these cases we show how our method transforms their equations in order to obtain a structurally identifiable and observable model which is also interpretable.


Assuntos
Modelos Biológicos , Dinâmica não Linear , Algoritmos , Biologia de Sistemas/métodos , Biologia Computacional
7.
PLoS Comput Biol ; 19(7): e1011264, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37410779

RESUMO

This work presents a methodology to evaluate the bioenergetic feasibility of alternative metabolic pathways for a given microbial conversion, optimising their energy yield and driving forces as a function of the concentration of metabolic intermediates. The tool, based on thermodynamic principles and multi-objective optimisation, accounts for pathway variants in terms of different electron carriers, as well as energy conservation (proton translocating) reactions within the pathway. The method also accommodates other constraints, some of them non-linear, such as the balance of conserved moieties. The approach involves the transformation of the maximum energy yield problem into a multi-objective mixed-integer linear optimisation problem which is then subsequently solved using the epsilon-constraint method, highlighting the trade-off between yield and rate in metabolic reactions. The methodology is applied to analyse several pathway alternatives occurring during propionate oxidation in anaerobic fermentation processes, as well as to the reverse TCA cycle pathway occurring during autotrophic microbial CO2 fixation. The results obtained using the developed methodology match previously reported literature and bring about insights into the studied pathways.


Assuntos
Ciclo do Ácido Cítrico , Redes e Vias Metabólicas , Metabolismo Energético , Fermentação , Termodinâmica
8.
PLoS Comput Biol ; 17(1): e1008646, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33497393

RESUMO

Reproducibility and reusability of the results of data-based modeling studies are essential. Yet, there has been-so far-no broadly supported format for the specification of parameter estimation problems in systems biology. Here, we introduce PEtab, a format which facilitates the specification of parameter estimation problems using Systems Biology Markup Language (SBML) models and a set of tab-separated value files describing the observation model and experimental data as well as parameters to be estimated. We already implemented PEtab support into eight well-established model simulation and parameter estimation toolboxes with hundreds of users in total. We provide a Python library for validation and modification of a PEtab problem and currently 20 example parameter estimation problems based on recent studies.


Assuntos
Linguagens de Programação , Biologia de Sistemas/métodos , Algoritmos , Bases de Dados Factuais , Modelos Biológicos , Modelos Estatísticos , Reprodutibilidade dos Testes
9.
Annu Rev Control ; 51: 441-459, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33362427

RESUMO

The recent coronavirus disease (COVID-19) outbreak has dramatically increased the public awareness and appreciation of the utility of dynamic models. At the same time, the dissemination of contradictory model predictions has highlighted their limitations. If some parameters and/or state variables of a model cannot be determined from output measurements, its ability to yield correct insights - as well as the possibility of controlling the system - may be compromised. Epidemic dynamics are commonly analysed using compartmental models, and many variations of such models have been used for analysing and predicting the evolution of the COVID-19 pandemic. In this paper we survey the different models proposed in the literature, assembling a list of 36 model structures and assessing their ability to provide reliable information. We address the problem using the control theoretic concepts of structural identifiability and observability. Since some parameters can vary during the course of an epidemic, we consider both the constant and time-varying parameter assumptions. We analyse the structural identifiability and observability of all of the models, considering all plausible choices of outputs and time-varying parameters, which leads us to analyse 255 different model versions. We classify the models according to their structural identifiability and observability under the different assumptions and discuss the implications of the results. We also illustrate with an example several alternative ways of remedying the lack of observability of a model. Our analyses provide guidelines for choosing the most informative model for each purpose, taking into account the available knowledge and measurements.

10.
BMC Bioinformatics ; 21(1): 472, 2020 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-33087041

RESUMO

BACKGROUND: Optimality principles have been used to explain the structure and behavior of living matter at different levels of organization, from basic phenomena at the molecular level, up to complex dynamics in whole populations. Most of these studies have assumed a single-criteria approach. Such optimality principles have been justified from an evolutionary perspective. In the context of the cell, previous studies have shown how dynamics of gene expression in small metabolic models can be explained assuming that cells have developed optimal adaptation strategies. Most of these works have considered rather simplified representations, such as small linear pathways, or reduced networks with a single branching point, and a single objective for the optimality criteria. RESULTS: Here we consider the extension of this approach to more realistic scenarios, i.e. biochemical pathways of arbitrary size and structure. We first show that exploiting optimality principles for these networks poses great challenges due to the complexity of the associated optimal control problems. Second, in order to surmount such challenges, we present a computational framework which has been designed with scalability and efficiency in mind, including mechanisms to avoid the most common pitfalls. Third, we illustrate its performance with several case studies considering the central carbon metabolism of S. cerevisiae and B. subtilis. In particular, we consider metabolic dynamics during nutrient shift experiments. CONCLUSIONS: We show how multi-objective optimal control can be used to predict temporal profiles of enzyme activation and metabolite concentrations in complex metabolic pathways. Further, we also show how to consider general cost/benefit trade-offs. In this study we have considered metabolic pathways, but this computational framework can also be applied to analyze the dynamics of other complex pathways, such as signal transduction or gene regulatory networks.


Assuntos
Biologia Computacional/métodos , Redes e Vias Metabólicas , Redes Reguladoras de Genes , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/metabolismo , Transdução de Sinais , Transcriptoma
11.
Bioinformatics ; 35(5): 830-838, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30816929

RESUMO

MOTIVATION: Kinetic models contain unknown parameters that are estimated by optimizing the fit to experimental data. This task can be computationally challenging due to the presence of local optima and ill-conditioning. While a variety of optimization methods have been suggested to surmount these issues, it is difficult to choose the best one for a given problem a priori. A systematic comparison of parameter estimation methods for problems with tens to hundreds of optimization variables is currently missing, and smaller studies provided contradictory findings. RESULTS: We use a collection of benchmarks to evaluate the performance of two families of optimization methods: (i) multi-starts of deterministic local searches and (ii) stochastic global optimization metaheuristics; the latter may be combined with deterministic local searches, leading to hybrid methods. A fair comparison is ensured through a collaborative evaluation and a consideration of multiple performance metrics. We discuss possible evaluation criteria to assess the trade-off between computational efficiency and robustness. Our results show that, thanks to recent advances in the calculation of parametric sensitivities, a multi-start of gradient-based local methods is often a successful strategy, but a better performance can be obtained with a hybrid metaheuristic. The best performer combines a global scatter search metaheuristic with an interior point local method, provided with gradients estimated with adjoint-based sensitivities. We provide an implementation of this method to render it available to the scientific community. AVAILABILITY AND IMPLEMENTATION: The code to reproduce the results is provided as Supplementary Material and is available at Zenodo https://doi.org/10.5281/zenodo.1304034. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Benchmarking , Software , Algoritmos , Cinética , Modelos Biológicos
12.
Bioprocess Biosyst Eng ; 43(9): 1671-1688, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32377941

RESUMO

Data integration to model-based description of biological systems incorporating gene dynamics improves the performance of microbial systems. Bioprocess performance, typically predicted using empirical Monod-type models, is essential for a sustainable bioeconomy. To replace empirical models, we updated a hybrid gene regulatory network-growth kinetic model, predicting aromatic pollutants degradation and biomass growth in Pseudomonas putida mt-2. We modeled a complex biological system including extensive information to understand the role of the regulatory elements in toluene biodegradation and biomass growth. The updated model exhibited extra complications such as the existence of oscillations and discontinuities. As parameter estimation of complex biological models remains a key challenge, we used the updated model to present a dual-parameter identification approach (the 'dual approach') combining two independent methodologies. Approach I handled the complexity by incorporation of demonstrated biological knowledge in the model-development process and combination of global sensitivity analysis and optimisation. Approach II complemented Approach I handling multimodality, ill-conditioning and overfitting through regularisation estimation, global optimisation, and identifiability analysis. To systematically quantify the biological system, we used a vast amount of high-quality time-course data. The dual approach resulted in an accurately calibrated kinetic model (NRMSE: 0.17055) efficiently handling the additional model complexity. We tested model validation using three independent experimental data sets, achieving greater predictive power (NRMSE: 0.18776) than the individual approaches (NRMSE I: 0.25322, II: 0.25227) and increasing model robustness. These results demonstrated data-driven predictive modeling potentially leading to bioprocess' model-based control and optimisation.


Assuntos
Redes Reguladoras de Genes , Pseudomonas putida , Tolueno/metabolismo , Biodegradação Ambiental , Pseudomonas putida/genética , Pseudomonas putida/metabolismo
13.
BMC Bioinformatics ; 20(1): 82, 2019 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-30770736

RESUMO

BACKGROUND: Dynamic modelling is a core element in the systems biology approach to understanding complex biosystems. Here, we consider the problem of parameter estimation in models of biological oscillators described by deterministic nonlinear differential equations. These problems can be extremely challenging due to several common pitfalls: (i) a lack of prior knowledge about parameters (i.e. massive search spaces), (ii) convergence to local optima (due to multimodality of the cost function), (iii) overfitting (fitting the noise instead of the signal) and (iv) a lack of identifiability. As a consequence, the use of standard estimation methods (such as gradient-based local ones) will often result in wrong solutions. Overfitting can be particularly problematic, since it produces very good calibrations, giving the impression of an excellent result. However, overfitted models exhibit poor predictive power. Here, we present a novel automated approach to overcome these pitfalls. Its workflow makes use of two sequential optimisation steps incorporating three key algorithms: (1) sampling strategies to systematically tighten the parameter bounds reducing the search space, (2) efficient global optimisation to avoid convergence to local solutions, (3) an advanced regularisation technique to fight overfitting. In addition, this workflow incorporates tests for structural and practical identifiability. RESULTS: We successfully evaluate this novel approach considering four difficult case studies regarding the calibration of well-known biological oscillators (Goodwin, FitzHugh-Nagumo, Repressilator and a metabolic oscillator). In contrast, we show how local gradient-based approaches, even if used in multi-start fashion, are unable to avoid the above-mentioned pitfalls. CONCLUSIONS: Our approach results in more efficient estimations (thanks to the bounding strategy) which are able to escape convergence to local optima (thanks to the global optimisation approach). Further, the use of regularisation allows us to avoid overfitting, resulting in more generalisable calibrated models (i.e. models with greater predictive power).


Assuntos
Algoritmos , Relógios Biológicos , Redes e Vias Metabólicas , Modelos Biológicos , Biologia de Sistemas/métodos , Calibragem , Humanos , Transdução de Sinais
14.
Bioinformatics ; 34(14): 2433-2440, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29522196

RESUMO

Motivation: Optimality principles have been used to explain many biological processes and systems. However, the functions being optimized are in general unknown a priori. Here we present an inverse optimal control framework for modeling dynamics in systems biology. The objective is to identify the underlying optimality principle from observed time-series data and simultaneously estimate unmeasured time-dependent inputs and time-invariant model parameters. As a special case, we also consider the problem of optimal simultaneous estimation of inputs and parameters from noisy data. After presenting a general statement of the inverse optimal control problem, and discussing special cases of interest, we outline numerical strategies which are scalable and robust. Results: We discuss the existence, relevance and implications of identifiability issues in the above problems. We present a robust computational approach based on regularized cost functions and the use of suitable direct numerical methods based on the control-vector parameterization approach. To avoid convergence to local solutions, we make use of hybrid global-local methods. We illustrate the performance and capabilities of this approach with several challenging case studies, including simulated and real data. We pay particular attention to the computational scalability of our approach (with the objective of considering large numbers of inputs and states). We provide a software implementation of both the methods and the case studies. Availability and implementation: The code used to obtain the results reported here is available at https://zenodo.org/record/1009541. Supplementary information: Supplementary data are available at Bioinformatics online.


Assuntos
Modelos Biológicos , Software , Biologia de Sistemas/métodos , Algoritmos , Janus Quinase 2/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais
15.
Bioinformatics ; 34(8): 1421-1423, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29206901

RESUMO

Motivation: Mathematical modeling using ordinary differential equations is used in systems biology to improve the understanding of dynamic biological processes. The parameters of ordinary differential equation models are usually estimated from experimental data. To analyze a priori the uniqueness of the solution of the estimation problem, structural identifiability analysis methods have been developed. Results: We introduce GenSSI 2.0, an advancement of the software toolbox GenSSI (Generating Series for testing Structural Identifiability). GenSSI 2.0 is the first toolbox for structural identifiability analysis to implement Systems Biology Markup Language import, state/parameter transformations and multi-experiment structural identifiability analysis. In addition, GenSSI 2.0 supports a range of MATLAB versions and is computationally more efficient than its previous version, enabling the analysis of more complex models. Availability and implementation: GenSSI 2.0 is an open-source MATLAB toolbox and available at https://github.com/genssi-developer/GenSSI. Contact: thomas.ligon@physik.uni-muenchen.de or jan.hasenauer@helmholtz-muenchen.de. Supplementary information: Supplementary data are available at Bioinformatics online.


Assuntos
Modelos Biológicos , Software , Biologia de Sistemas/métodos
16.
PLoS Comput Biol ; 13(11): e1005878, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29186132

RESUMO

The concept of dynamical compensation has been recently introduced to describe the ability of a biological system to keep its output dynamics unchanged in the face of varying parameters. However, the original definition of dynamical compensation amounts to lack of structural identifiability. This is relevant if model parameters need to be estimated, as is often the case in biological modelling. Care should we taken when using an unidentifiable model to extract biological insight: the estimated values of structurally unidentifiable parameters are meaningless, and model predictions about unmeasured state variables can be wrong. Taking this into account, we explore alternative definitions of dynamical compensation that do not necessarily imply structural unidentifiability. Accordingly, we show different ways in which a model can be made identifiable while exhibiting dynamical compensation. Our analyses enable the use of the new concept of dynamical compensation in the context of parameter identification, and reconcile it with the desirable property of structural identifiability.


Assuntos
Retroalimentação Fisiológica , Modelos Biológicos , Biologia de Sistemas , Humanos
17.
PLoS Comput Biol ; 13(2): e1005379, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28166222

RESUMO

Despite significant efforts and remarkable progress, the inference of signaling networks from experimental data remains very challenging. The problem is particularly difficult when the objective is to obtain a dynamic model capable of predicting the effect of novel perturbations not considered during model training. The problem is ill-posed due to the nonlinear nature of these systems, the fact that only a fraction of the involved proteins and their post-translational modifications can be measured, and limitations on the technologies used for growing cells in vitro, perturbing them, and measuring their variations. As a consequence, there is a pervasive lack of identifiability. To overcome these issues, we present a methodology called SELDOM (enSEmbLe of Dynamic lOgic-based Models), which builds an ensemble of logic-based dynamic models, trains them to experimental data, and combines their individual simulations into an ensemble prediction. It also includes a model reduction step to prune spurious interactions and mitigate overfitting. SELDOM is a data-driven method, in the sense that it does not require any prior knowledge of the system: the interaction networks that act as scaffolds for the dynamic models are inferred from data using mutual information. We have tested SELDOM on a number of experimental and in silico signal transduction case-studies, including the recent HPN-DREAM breast cancer challenge. We found that its performance is highly competitive compared to state-of-the-art methods for the purpose of recovering network topology. More importantly, the utility of SELDOM goes beyond basic network inference (i.e. uncovering static interaction networks): it builds dynamic (based on ordinary differential equation) models, which can be used for mechanistic interpretations and reliable dynamic predictions in new experimental conditions (i.e. not used in the training). For this task, SELDOM's ensemble prediction is not only consistently better than predictions from individual models, but also often outperforms the state of the art represented by the methods used in the HPN-DREAM challenge.


Assuntos
Algoritmos , Engenharia Metabólica/métodos , Modelos Biológicos , Proteoma/metabolismo , Transdução de Sinais/fisiologia , Animais , Simulação por Computador , Humanos , Análise do Fluxo Metabólico
18.
BMC Bioinformatics ; 18(1): 52, 2017 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-28109249

RESUMO

BACKGROUND: The development of large-scale kinetic models is one of the current key issues in computational systems biology and bioinformatics. Here we consider the problem of parameter estimation in nonlinear dynamic models. Global optimization methods can be used to solve this type of problems but the associated computational cost is very large. Moreover, many of these methods need the tuning of a number of adjustable search parameters, requiring a number of initial exploratory runs and therefore further increasing the computation times. Here we present a novel parallel method, self-adaptive cooperative enhanced scatter search (saCeSS), to accelerate the solution of this class of problems. The method is based on the scatter search optimization metaheuristic and incorporates several key new mechanisms: (i) asynchronous cooperation between parallel processes, (ii) coarse and fine-grained parallelism, and (iii) self-tuning strategies. RESULTS: The performance and robustness of saCeSS is illustrated by solving a set of challenging parameter estimation problems, including medium and large-scale kinetic models of the bacterium E. coli, bakerés yeast S. cerevisiae, the vinegar fly D. melanogaster, Chinese Hamster Ovary cells, and a generic signal transduction network. The results consistently show that saCeSS is a robust and efficient method, allowing very significant reduction of computation times with respect to several previous state of the art methods (from days to minutes, in several cases) even when only a small number of processors is used. CONCLUSIONS: The new parallel cooperative method presented here allows the solution of medium and large scale parameter estimation problems in reasonable computation times and with small hardware requirements. Further, the method includes self-tuning mechanisms which facilitate its use by non-experts. We believe that this new method can play a key role in the development of large-scale and even whole-cell dynamic models.


Assuntos
Biologia Computacional/métodos , Biologia de Sistemas/métodos , Animais , Células CHO , Cricetulus/genética , Drosophila melanogaster/genética , Escherichia coli/genética , Regulação da Expressão Gênica , Modelos Teóricos , Dinâmica não Linear , Saccharomyces cerevisiae/genética , Transdução de Sinais , Transcrição Gênica
19.
Bioinformatics ; 32(21): 3360-3362, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27402908

RESUMO

MOTIVATION: The design of de novo circuits with predefined performance specifications is a challenging problem in Synthetic Biology. Computational models and tools have proved to be crucial for a successful wet lab implementation. Natural gene circuits are complex, subject to evolutionary tradeoffs and playing multiple roles. However, most synthetic designs implemented to date are simple and perform a single task. As the field progresses, advanced computational tools are needed in order to handle greater levels of circuit complexity in a more flexible way and considering multiple design criteria. RESULTS: This works presents SYNBADm (SYNthetic Biology Automated optimal Design in Matlab), a software toolbox for the automatic optimal design of gene circuits with targeted functions from libraries of components. This tool makes use of global optimization to simultaneously search the space of structures and kinetic parameters. SYNBADm can efficiently handle high levels of circuit complexity and can consider multiple design criteria through multiobjective optimization. Further, it provides flexible design capabilities, i.e. the user can define the modeling framework, library of components and target performance function(s). AVAILABILITY AND IMPLEMENTATION: SYNBADm runs under the popular MATLAB computational environment, and is available under GPLv3 license at https://sites.google.com/site/synbadm CONTACT: ireneotero@iim.csic.es or julio@iim.csic.es.


Assuntos
Genes Sintéticos , Software , Redes Reguladoras de Genes , Biologia Sintética
20.
Bioinformatics ; 32(21): 3357-3359, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27378288

RESUMO

MOTIVATION: Many problems of interest in dynamic modeling and control of biological systems can be posed as non-linear optimization problems subject to algebraic and dynamic constraints. In the context of modeling, this is the case of, e.g. parameter estimation, optimal experimental design and dynamic flux balance analysis. In the context of control, model-based metabolic engineering or drug dose optimization problems can be formulated as (multi-objective) optimal control problems. Finding a solution to those problems is a very challenging task which requires advanced numerical methods. RESULTS: This work presents the AMIGO2 toolbox: the first multiplatform software tool that automatizes the solution of all those problems, offering a suite of state-of-the-art (multi-objective) global optimizers and advanced simulation approaches. AVAILABILITY AND IMPLEMENTATION: The toolbox and its documentation are available at: sites.google.com/site/amigo2toolbox CONTACT: ebalsa@iim.csic.esSupplementary information: Supplementary data are available at Bioinformatics online.


Assuntos
Software , Biologia de Sistemas , Algoritmos , Animais , Humanos , Engenharia Metabólica , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA