Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Brief Bioinform ; 23(3)2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35437577

RESUMO

Predicting protein properties from amino acid sequences is an important problem in biology and pharmacology. Protein-protein interactions among SARS-CoV-2 spike protein, human receptors and antibodies are key determinants of the potency of this virus and its ability to evade the human immune response. As a rapidly evolving virus, SARS-CoV-2 has already developed into many variants with considerable variation in virulence among these variants. Utilizing the proteomic data of SARS-CoV-2 to predict its viral characteristics will, therefore, greatly aid in disease control and prevention. In this paper, we review and compare recent successful prediction methods based on long short-term memory (LSTM), transformer, convolutional neural network (CNN) and a similarity-based topological regression (TR) model and offer recommendations about appropriate predictive methodology depending on the similarity between training and test datasets. We compare the effectiveness of these models in predicting the binding affinity and expression of SARS-CoV-2 spike protein sequences. We also explore how effective these predictive methods are when trained on laboratory-created data and are tasked with predicting the binding affinity of the in-the-wild SARS-CoV-2 spike protein sequences obtained from the GISAID datasets. We observe that TR is a better method when the sample size is small and test protein sequences are sufficiently similar to the training sequence. However, when the training sample size is sufficiently large and prediction requires extrapolation, LSTM embedding and CNN-based predictive model show superior performance.


Assuntos
COVID-19 , SARS-CoV-2 , Sequência de Aminoácidos , COVID-19/genética , Humanos , Ligação Proteica , Proteômica , SARS-CoV-2/genética , Análise de Sequência de Proteína , Glicoproteína da Espícula de Coronavírus/metabolismo
2.
J Biol Chem ; 297(3): 101023, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34343564

RESUMO

Ammonia is a cytotoxic molecule generated during normal cellular functions. Dysregulated ammonia metabolism, which is evident in many chronic diseases such as liver cirrhosis, heart failure, and chronic obstructive pulmonary disease, initiates a hyperammonemic stress response in tissues including skeletal muscle and in myotubes. Perturbations in levels of specific regulatory molecules have been reported, but the global responses to hyperammonemia are unclear. In this study, we used a multiomics approach to vertically integrate unbiased data generated using an assay for transposase-accessible chromatin with high-throughput sequencing, RNA-Seq, and proteomics. We then horizontally integrated these data across different models of hyperammonemia, including myotubes and mouse and human muscle tissues. Changes in chromatin accessibility and/or expression of genes resulted in distinct clusters of temporal molecular changes including transient, persistent, and delayed responses during hyperammonemia in myotubes. Known responses to hyperammonemia, including mitochondrial and oxidative dysfunction, protein homeostasis disruption, and oxidative stress pathway activation, were enriched in our datasets. During hyperammonemia, pathways that impact skeletal muscle structure and function that were consistently enriched were those that contribute to mitochondrial dysfunction, oxidative stress, and senescence. We made several novel observations, including an enrichment in antiapoptotic B-cell leukemia/lymphoma 2 family protein expression, increased calcium flux, and increased protein glycosylation in myotubes and muscle tissue upon hyperammonemia. Critical molecules in these pathways were validated experimentally. Human skeletal muscle from patients with cirrhosis displayed similar responses, establishing translational relevance. These data demonstrate complex molecular interactions during adaptive and maladaptive responses during the cellular stress response to hyperammonemia.


Assuntos
Genômica , Hiperamonemia/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Proteômica , Transcriptoma , Animais , Citometria de Fluxo , Humanos , Hiperamonemia/genética , Immunoblotting/métodos , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Reprodutibilidade dos Testes
3.
Bioinformatics ; 37(Suppl_1): i42-i50, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34252971

RESUMO

MOTIVATION: Anti-cancer drug sensitivity prediction using deep learning models for individual cell line is a significant challenge in personalized medicine. Recently developed REFINED (REpresentation of Features as Images with NEighborhood Dependencies) CNN (Convolutional Neural Network)-based models have shown promising results in improving drug sensitivity prediction. The primary idea behind REFINED-CNN is representing high dimensional vectors as compact images with spatial correlations that can benefit from CNN architectures. However, the mapping from a high dimensional vector to a compact 2D image depends on the a priori choice of the distance metric and projection scheme with limited empirical procedures guiding these choices. RESULTS: In this article, we consider an ensemble of REFINED-CNN built under different choices of distance metrics and/or projection schemes that can improve upon a single projection based REFINED-CNN model. Results, illustrated using NCI60 and NCI-ALMANAC databases, demonstrate that the ensemble approaches can provide significant improvement in prediction performance as compared to individual models. We also develop the theoretical framework for combining different distance metrics to arrive at a single 2D mapping. Results demonstrated that distance-averaged REFINED-CNN produced comparable performance as obtained from stacking REFINED-CNN ensemble but with significantly lower computational cost. AVAILABILITY AND IMPLEMENTATION: The source code, scripts, and data used in the paper have been deposited in GitHub (https://github.com/omidbazgirTTU/IntegratedREFINED). SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Aprendizado de Máquina , Neoplasias/tratamento farmacológico , Redes Neurais de Computação , Software
4.
Brief Bioinform ; 20(5): 1734-1753, 2019 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-31846027

RESUMO

Recent years have seen an increase in the availability of pharmacogenomic databases such as Genomics of Drug Sensitivity in Cancer (GDSC) and Cancer Cell Line Encyclopedia (CCLE) that provide genomic and functional characterization information for multiple cell lines. Studies have alluded to the fact that specific characterizations may be inconsistent between different databases. Analysis of the potential discrepancies in the different databases is highly significant, as these sources are frequently used to analyze and validate methodologies for personalized cancer therapies. In this article, we review the recent developments in investigating the correspondence between different pharmacogenomics databases and discuss the potential factors that require attention when incorporating these sources in any modeling analysis. Furthermore, we explored the consistency among these databases using copulas that can capture nonlinear dependencies between two sets of data.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/genética , Farmacogenética , Linhagem Celular Tumoral , Bases de Dados Genéticas , Humanos , Neoplasias/patologia
5.
Bioinformatics ; 35(17): 3143-3145, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30649230

RESUMO

SUMMARY: Biological processes are characterized by a variety of different genomic feature sets. However, often times when building models, portions of these features are missing for a subset of the dataset. We provide a modeling framework to effectively integrate this type of heterogeneous data to improve prediction accuracy. To test our methodology, we have stacked data from the Cancer Cell Line Encyclopedia to increase the accuracy of drug sensitivity prediction. The package addresses the dynamic regime of information integration involving sequential addition of features and samples. AVAILABILITY AND IMPLEMENTATION: The framework has been implemented as a R package Sstack, which can be downloaded from https://cran.r-project.org/web/packages/Sstack/index.html, where further explanation of the package is available. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Neoplasias , Software , Linhagem Celular Tumoral , Genoma , Genômica , Humanos
6.
BMC Bioinformatics ; 20(Suppl 12): 317, 2019 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-31216980

RESUMO

BACKGROUND: Clinical studies often track dose-response curves of subjects over time. One can easily model the dose-response curve at each time point with Hill equation, but such a model fails to capture the temporal evolution of the curves. On the other hand, one can use Gompertz equation to model the temporal behaviors at each dose without capturing the evolution of time curves across dosage. RESULTS: In this article, we propose a parametric model for dose-time responses that follows Gompertz law in time and Hill equation across dose approximately. We derive a recursion relation for dose-response curves over time capturing the temporal evolution and then specify a regression model connecting the parameters controlling the dose-time responses with individual level proteomic data. The resultant joint model allows us to predict the dose-response curves over time for new individuals. CONCLUSION: We have compared the efficacy of our proposed Recursive Hybrid model with individual dose-response predictive models at desired time points. We note that our proposed model exhibits a superior performance compared to the individual ones for both synthetic data and actual pharmacological data. For the desired dose-time varying genetic characterization and drug response values, we have used the HMS-LINCS database and demonstrated the effectiveness of our model for all available anticancer compounds.


Assuntos
Modelos Teóricos , Farmacologia , Simulação por Computador , Bases de Dados como Assunto , Relação Dose-Resposta a Droga , Humanos , Fatores de Tempo
7.
Bioinformatics ; 34(8): 1336-1344, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29267851

RESUMO

Motivation: Random forest (RF) has become a widely popular prediction generating mechanism. Its strength lies in its flexibility, interpretability and ability to handle large number of features, typically larger than the sample size. However, this methodology is of limited use if one wishes to identify statistically significant features. Several ranking schemes are available that provide information on the relative importance of the features, but there is a paucity of general inferential mechanism, particularly in a multi-variate set up. We use the conditional inference tree framework to generate a RF where features are deleted sequentially based on explicit hypothesis testing. The resulting sequential algorithm offers an inferentially justifiable, but model-free, variable selection procedure. Significant features are then used to generate predictive RF. An added advantage of our methodology is that both variable selection and prediction are based on conditional inference framework and hence are coherent. Results: We illustrate the performance of our Sequential Multi-Response Feature Selection approach through simulation studies and finally apply this methodology on Genomics of Drug Sensitivity for Cancer dataset to identify genetic characteristics that significantly impact drug sensitivities. Significant set of predictors obtained from our method are further validated from biological perspective. Availability and implementation: https://github.com/jomayer/SMuRF. Contact: souparno.ghosh@ttu.edu. Supplementary information: Supplementary data are available at Bioinformatics online.


Assuntos
Algoritmos , Genômica/métodos , Antineoplásicos/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética
8.
BMC Cancer ; 19(1): 593, 2019 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-31208434

RESUMO

BACKGROUND: Cancer patients with advanced disease routinely exhaust available clinical regimens and lack actionable genomic medicine results, leaving a large patient population without effective treatments options when their disease inevitably progresses. To address the unmet clinical need for evidence-based therapy assignment when standard clinical approaches have failed, we have developed a probabilistic computational modeling approach which integrates molecular sequencing data with functional assay data to develop patient-specific combination cancer treatments. METHODS: Tissue taken from a murine model of alveolar rhabdomyosarcoma was used to perform single agent drug screening and DNA/RNA sequencing experiments; results integrated via our computational modeling approach identified a synergistic personalized two-drug combination. Cells derived from the primary murine tumor were allografted into mouse models and used to validate the personalized two-drug combination. Computational modeling of single agent drug screening and RNA sequencing of multiple heterogenous sites from a single patient's epithelioid sarcoma identified a personalized two-drug combination effective across all tumor regions. The heterogeneity-consensus combination was validated in a xenograft model derived from the patient's primary tumor. Cell cultures derived from human and canine undifferentiated pleomorphic sarcoma were assayed by drug screen; computational modeling identified a resistance-abrogating two-drug combination common to both cell cultures. This combination was validated in vitro via a cell regrowth assay. RESULTS: Our computational modeling approach addresses three major challenges in personalized cancer therapy: synergistic drug combination predictions (validated in vitro and in vivo in a genetically engineered murine cancer model), identification of unifying therapeutic targets to overcome intra-tumor heterogeneity (validated in vivo in a human cancer xenograft), and mitigation of cancer cell resistance and rewiring mechanisms (validated in vitro in a human and canine cancer model). CONCLUSIONS: These proof-of-concept studies support the use of an integrative functional approach to personalized combination therapy prediction for the population of high-risk cancer patients lacking viable clinical options and without actionable DNA sequencing-based therapy.


Assuntos
Biologia Computacional/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Quimioterapia Combinada/métodos , Modelos Estatísticos , Medicina de Precisão/métodos , Rabdomiossarcoma Alveolar/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Cães , Sinergismo Farmacológico , Feminino , Xenoenxertos , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos NOD
9.
BMC Bioinformatics ; 19(Suppl 17): 497, 2018 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-30591023

RESUMO

BACKGROUND: In precision medicine, scarcity of suitable biological data often hinders the design of an appropriate predictive model. In this regard, large scale pharmacogenomics studies, like CCLE and GDSC hold the promise to mitigate the issue. However, one cannot directly employ data from multiple sources together due to the existing distribution shift in data. One way to solve this problem is to utilize the transfer learning methodologies tailored to fit in this specific context. RESULTS: In this paper, we present two novel approaches for incorporating information from a secondary database for improving the prediction in a target database. The first approach is based on latent variable cost optimization and the second approach considers polynomial mapping between the two databases. Utilizing CCLE and GDSC databases, we illustrate that the proposed approaches accomplish a better prediction of drug sensitivities for different scenarios as compared to the existing approaches. CONCLUSION: We have compared the performance of the proposed predictive models with database-specific individual models as well as existing transfer learning approaches. We note that our proposed approaches exhibit superior performance compared to the abovementioned alternative techniques for predicting sensitivity for different anti-cancer compounds, particularly the nonlinear mapping model shows the best overall performance.


Assuntos
Algoritmos , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Área Sob a Curva , Bases de Dados Factuais , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/genética
10.
BMC Bioinformatics ; 19(Suppl 3): 71, 2018 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29589559

RESUMO

BACKGROUND: A significant problem in precision medicine is the prediction of drug sensitivity for individual cancer cell lines. Predictive models such as Random Forests have shown promising performance while predicting from individual genomic features such as gene expressions. However, accessibility of various other forms of data types including information on multiple tested drugs necessitates the examination of designing predictive models incorporating the various data types. RESULTS: We explore the predictive performance of model stacking and the effect of stacking on the predictive bias and squared error. In addition we discuss the analytical underpinnings supporting the advantages of stacking in reducing squared error and inherent bias of random forests in prediction of outliers. The framework is tested on a setup including gene expression, drug target, physical properties and drug response information for a set of drugs and cell lines. CONCLUSION: The performance of individual and stacked models are compared. We note that stacking models built on two heterogeneous datasets provide superior performance to stacking different models built on the same dataset. It is also noted that stacking provides a noticeable reduction in the bias of our predictors when the dominant eigenvalue of the principle axis of variation in the residuals is significantly higher than the remaining eigenvalues.


Assuntos
Ensaios de Seleção de Medicamentos Antitumorais , Modelos Biológicos , Algoritmos , Área Sob a Curva , Viés , Linhagem Celular Tumoral , Aprendizado Profundo , Humanos , Neoplasias/tratamento farmacológico , Medicina de Precisão
11.
Bioinformatics ; 33(9): 1407-1410, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28334269

RESUMO

Summary: IntegratedMRF is an open-source R implementation for integrating drug response predictions from various genomic characterizations using univariate or multivariate random forests that includes various options for error estimation techniques. The integrated framework was developed following superior performance of random forest based methods in NCI-DREAM drug sensitivity prediction challenge. The computational framework can be applied to estimate mean and confidence interval of drug response prediction errors based on ensemble approaches with various combinations of genetic and epigenetic characterizations as inputs. The multivariate random forest implementation included in the package incorporates the correlations between output responses in the modeling and has been shown to perform better than existing approaches when the drug responses are correlated. Detailed analysis of the provided features is included in the Supplementary Material . Availability and Implementation: The framework has been implemented as a package IntegratedMRF , which can be downloaded from https://cran.r-project.org/web/packages/IntegratedMRF/index.html , where further explanation of the package is available. Contact: ranadip.pal@ttu.edu. Supplementary information: Supplementary data are available at Bioinformatics online.


Assuntos
Biomarcadores Farmacológicos , Genômica/métodos , Modelos Genéticos , Neoplasias/genética , Software , Estatística como Assunto/métodos , Antineoplásicos/uso terapêutico , Metilação de DNA , Regulação da Expressão Gênica , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Polimorfismo de Nucleotídeo Único , Medicina de Precisão/métodos , Transcriptoma
12.
BMC Bioinformatics ; 18(Suppl 4): 116, 2017 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-28361667

RESUMO

BACKGROUND: Design of personalized targeted therapies involve modeling of patient sensitivity to various drugs and drug combinations. Majority of studies evaluate the sensitivity of tumor cells to targeted drugs without modeling the effect of the drugs on normal cells. In this article, we consider the individual modeling of drug responses to tumor and normal cells and utilize them to design targeted combination therapies that maximize sensitivity over tumor cells and minimize toxicity over normal cells. RESULTS: The problem is formulated as maximizing sensitivity over tumor cell models while maintaining sensitivity below a threshold over normal cell models. We utilize the constrained structure of tumor proliferation models to design an accelerated lexicographic search algorithm for generating the optimal solution. For comparison purposes, we also designed two suboptimal search algorithms based on evolutionary algorithms and hill-climbing based techniques. Results over synthetic models and models generated from Genomics of Drug Sensitivity in Cancer database shows the ability of the proposed algorithms to arrive at optimal or close to optimal solutions in significantly lower number of steps as compared to exhaustive search. We also present the theoretical analysis of the expected number of comparisons required for the proposed Lexicographic search that compare favorably with the observed number of computations. CONCLUSIONS: The proposed algorithms provide a framework for design of combination therapy that tackles tumor heterogeneity while satisfying toxicity constraints.


Assuntos
Algoritmos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Modelos Teóricos , Neoplasias/tratamento farmacológico , Sobrevivência Celular , Genômica , Humanos
15.
Nat Commun ; 15(1): 5072, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38871711

RESUMO

Quantitative structure-activity relationship (QSAR) modeling is a powerful tool for drug discovery, yet the lack of interpretability of commonly used QSAR models hinders their application in molecular design. We propose a similarity-based regression framework, topological regression (TR), that offers a statistically grounded, computationally fast, and interpretable technique to predict drug responses. We compare the predictive performance of TR on 530 ChEMBL human target activity datasets against the predictive performance of deep-learning-based QSAR models. Our results suggest that our sparse TR model can achieve equal, if not better, performance than the deep learning-based QSAR models and provide better intuitive interpretation by extracting an approximate isometry between the chemical space of the drugs and their activity space.


Assuntos
Aprendizado Profundo , Relação Quantitativa Estrutura-Atividade , Humanos , Descoberta de Drogas/métodos , Análise de Regressão , Algoritmos
17.
BMC Bioinformatics ; 14: 239, 2013 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-23890326

RESUMO

BACKGROUND: The success of targeted anti-cancer drugs are frequently hindered by the lack of knowledge of the individual pathway of the patient and the extreme data requirements on the estimation of the personalized genetic network of the patient's tumor. The prediction of tumor sensitivity to targeted drugs remains a major challenge in the design of optimal therapeutic strategies. The current sensitivity prediction approaches are primarily based on genetic characterizations of the tumor sample. We propose a novel sensitivity prediction approach based on functional perturbation data that incorporates the drug protein interaction information and sensitivities to a training set of drugs with known targets. RESULTS: We illustrate the high prediction accuracy of our framework on synthetic data generated from the Kyoto Encyclopedia of Genes and Genomes (KEGG) and an experimental dataset of four canine osteosarcoma tumor cultures following application of 60 targeted small-molecule drugs. We achieve a low leave one out cross validation error of <10% for the canine osteosarcoma tumor cultures using a drug screen consisting of 60 targeted drugs. CONCLUSIONS: The proposed framework provides a unique input-output based methodology to model a cancer pathway and predict the effectiveness of targeted anti-cancer drugs. This framework can be developed as a viable approach for personalized cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Terapia de Alvo Molecular , Algoritmos , Animais , Antineoplásicos/uso terapêutico , Sobrevivência Celular , Cães , Redes Reguladoras de Genes , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Células Tumorais Cultivadas
18.
Curr Genomics ; 14(2): 91-110, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24082820

RESUMO

Until recently, understanding the regulatory behavior of cells has been pursued through independent analysis of the transcriptome or the proteome. Based on the central dogma, it was generally assumed that there exist a direct correspondence between mRNA transcripts and generated protein expressions. However, recent studies have shown that the correlation between mRNA and Protein expressions can be low due to various factors such as different half lives and post transcription machinery. Thus, a joint analysis of the transcriptomic and proteomic data can provide useful insights that may not be deciphered from individual analysis of mRNA or protein expressions. This article reviews the existing major approaches for joint analysis of transcriptomic and proteomic data. We categorize the different approaches into eight main categories based on the initial algorithm and final analysis goal. We further present analogies with other domains and discuss the existing research problems in this area.

19.
Bioinform Adv ; 3(1): vbad036, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37033467

RESUMO

Summary: Predictive learning from medical data incurs additional challenge due to concerns over privacy and security of personal data. Federated learning, intentionally structured to preserve high level of privacy, is emerging to be an attractive way to generate cross-silo predictions in medical scenarios. However, the impact of severe population-level heterogeneity on federated learners is not well explored. In this article, we propose a methodology to detect presence of population heterogeneity in federated settings and propose a solution to handle such heterogeneity by developing a federated version of Deep Regression Forests. Additionally, we demonstrate that the recently conceptualized REpresentation of Features as Images with NEighborhood Dependencies CNN framework can be combined with the proposed Federated Deep Regression Forests to provide improved performance as compared to existing approaches. Availability and implementation: The Python source code for reproducing the main results are available on GitHub: https://github.com/DanielNolte/FederatedDeepRegressionForests. Contact: ranadip.pal@ttu.edu. Supplementary information: Supplementary data are available at Bioinformatics Advances online.

20.
BMC Genomics ; 13 Suppl 6: S9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23134816

RESUMO

BACKGROUND: Numerous approaches exist for modeling of genetic regulatory networks (GRNs) but the low sampling rates often employed in biological studies prevents the inference of detailed models from experimental data. In this paper, we analyze the issues involved in estimating a model of a GRN from single cell line time series data with limited time points. RESULTS: We present an inference approach for a Boolean Network (BN) model of a GRN from limited transcriptomic or proteomic time series data based on prior biological knowledge of connectivity, constraints on attractor structure and robust design. We applied our inference approach to 6 time point transcriptomic data on Human Mammary Epithelial Cell line (HMEC) after application of Epidermal Growth Factor (EGF) and generated a BN with a plausible biological structure satisfying the data. We further defined and applied a similarity measure to compare synthetic BNs and BNs generated through the proposed approach constructed from transitions of various paths of the synthetic BNs. We have also compared the performance of our algorithm with two existing BN inference algorithms. CONCLUSIONS: Through theoretical analysis and simulations, we showed the rarity of arriving at a BN from limited time series data with plausible biological structure using random connectivity and absence of structure in data. The framework when applied to experimental data and data generated from synthetic BNs were able to estimate BNs with high similarity scores. Comparison with existing BN inference algorithms showed the better performance of our proposed algorithm for limited time series data. The proposed framework can also be applied to optimize the connectivity of a GRN from experimental data when the prior biological knowledge on regulators is limited or not unique.


Assuntos
Algoritmos , Redes Reguladoras de Genes , Linhagem Celular , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Humanos , Glândulas Mamárias Humanas/metabolismo , Modelos Genéticos , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA