Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Comput Biol ; 20(4): e1011945, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38578805

ABSTRACT

Early identification of safe and efficacious disease targets is crucial to alleviating the tremendous cost of drug discovery projects. However, existing experimental methods for identifying new targets are generally labor-intensive and failure-prone. On the other hand, computational approaches, especially machine learning-based frameworks, have shown remarkable application potential in drug discovery. In this work, we propose Progeni, a novel machine learning-based framework for target identification. In addition to fully exploiting the known heterogeneous biological networks from various sources, Progeni integrates literature evidence about the relations between biological entities to construct a probabilistic knowledge graph. Graph neural networks are then employed in Progeni to learn the feature embeddings of biological entities to facilitate the identification of biologically relevant target candidates. A comprehensive evaluation of Progeni demonstrated its superior predictive power over the baseline methods on the target identification task. In addition, our extensive tests showed that Progeni exhibited high robustness to the negative effect of exposure bias, a common phenomenon in recommendation systems, and effectively identified new targets that can be strongly supported by the literature. Moreover, our wet lab experiments successfully validated the biological significance of the top target candidates predicted by Progeni for melanoma and colorectal cancer. All these results suggested that Progeni can identify biologically effective targets and thus provide a powerful and useful tool for advancing the drug discovery process.


Subject(s)
Computational Biology , Drug Discovery , Machine Learning , Neural Networks, Computer , Humans , Computational Biology/methods , Drug Discovery/methods , Algorithms , Melanoma , Probability , Colorectal Neoplasms
3.
Nat Commun ; 14(1): 8459, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38123534

ABSTRACT

Single-cell technologies enable the dynamic analyses of cell fate mapping. However, capturing the gene regulatory relationships and identifying the driver factors that control cell fate decisions are still challenging. We present CEFCON, a network-based framework that first uses a graph neural network with attention mechanism to infer a cell-lineage-specific gene regulatory network (GRN) from single-cell RNA-sequencing data, and then models cell fate dynamics through network control theory to identify driver regulators and the associated gene modules, revealing their critical biological processes related to cell states. Extensive benchmarking tests consistently demonstrated the superiority of CEFCON in GRN construction, driver regulator identification, and gene module identification over baseline methods. When applied to the mouse hematopoietic stem cell differentiation data, CEFCON successfully identified driver regulators for three developmental lineages, which offered useful insights into their differentiation from a network control perspective. Overall, CEFCON provides a valuable tool for studying the underlying mechanisms of cell fate decisions from single-cell RNA-seq data.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Animals , Mice , Cell Differentiation/genetics , Cell Lineage/genetics , Gene Regulatory Networks , Single-Cell Analysis/methods
4.
Pac Symp Biocomput ; 28: 157-168, 2023.
Article in English | MEDLINE | ID: mdl-36540973

ABSTRACT

Identifying effective target-disease associations (TDAs) can alleviate the tremendous cost incurred by clinical failures of drug development. Although many machine learning models have been proposed to predict potential novel TDAs rapidly, their credibility is not guaranteed, thus requiring extensive experimental validation. In addition, it is generally challenging for current models to predict meaningful associations for entities with less information, hence limiting the application potential of these models in guiding future research. Based on recent advances in utilizing graph neural networks to extract features from heterogeneous biological data, we develop CreaTDA, an end-to-end deep learning-based framework that effectively learns latent feature representations of targets and diseases to facilitate TDA prediction. We also propose a novel way of encoding credibility information obtained from literature to enhance the performance of TDA prediction and predict more novel TDAs with real evidence support from previous studies. Compared with state-of-the-art baseline methods, CreaTDA achieves substantially better prediction performance on the whole TDA network and its sparse sub-networks containing the proteins associated with few known diseases. Our results demonstrate that CreaTDA can provide a powerful and helpful tool for identifying novel target-disease associations, thereby facilitating drug discovery.


Subject(s)
Computational Biology , Neural Networks, Computer , Humans , Computational Biology/methods , Machine Learning , Drug Discovery , Proteins
5.
Nat Commun ; 12(1): 5465, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526500

ABSTRACT

Peptide-protein interactions are involved in various fundamental cellular functions and their identification is crucial for designing efficacious peptide therapeutics. Recently, a number of computational methods have been developed to predict peptide-protein interactions. However, most of the existing prediction approaches heavily depend on high-resolution structure data. Here, we present a deep learning framework for multi-level peptide-protein interaction prediction, called CAMP, including binary peptide-protein interaction prediction and corresponding peptide binding residue identification. Comprehensive evaluation demonstrated that CAMP can successfully capture the binary interactions between peptides and proteins and identify the binding residues along the peptides involved in the interactions. In addition, CAMP outperformed other state-of-the-art methods on binary peptide-protein interaction prediction. CAMP can serve as a useful tool in peptide-protein interaction prediction and identification of important binding residues in the peptides, which can thus facilitate the peptide drug discovery process.


Subject(s)
Algorithms , Computational Biology/methods , Deep Learning , Peptides/metabolism , Proteins/metabolism , Binding Sites , Models, Molecular , Peptides/chemistry , Protein Binding , Protein Domains , Proteins/chemistry , Reproducibility of Results
6.
Signal Transduct Target Ther ; 6(1): 165, 2021 04 24.
Article in English | MEDLINE | ID: mdl-33895786

ABSTRACT

The global spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) requires an urgent need to find effective therapeutics for the treatment of coronavirus disease 2019 (COVID-19). In this study, we developed an integrative drug repositioning framework, which fully takes advantage of machine learning and statistical analysis approaches to systematically integrate and mine large-scale knowledge graph, literature and transcriptome data to discover the potential drug candidates against SARS-CoV-2. Our in silico screening followed by wet-lab validation indicated that a poly-ADP-ribose polymerase 1 (PARP1) inhibitor, CVL218, currently in Phase I clinical trial, may be repurposed to treat COVID-19. Our in vitro assays revealed that CVL218 can exhibit effective inhibitory activity against SARS-CoV-2 replication without obvious cytopathic effect. In addition, we showed that CVL218 can interact with the nucleocapsid (N) protein of SARS-CoV-2 and is able to suppress the LPS-induced production of several inflammatory cytokines that are highly relevant to the prevention of immunopathology induced by SARS-CoV-2 infection.


Subject(s)
Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , COVID-19/metabolism , Computer Simulation , Drug Repositioning , Models, Biological , SARS-CoV-2/metabolism , Humans
7.
Front Pharmacol ; 11: 112, 2020.
Article in English | MEDLINE | ID: mdl-32184722

ABSTRACT

Synthetic lethality (SL), an important type of genetic interaction, can provide useful insight into the target identification process for the development of anticancer therapeutics. Although several well-established SL gene pairs have been verified to be conserved in humans, most SL interactions remain cell-line specific. Here, we demonstrated that the cell-line-specific gene expression profiles derived from the shRNA perturbation experiments performed in the LINCS L1000 project can provide useful features for predicting SL interactions in human. In this paper, we developed a semi-supervised neural network-based method called EXP2SL to accurately identify SL interactions from the L1000 gene expression profiles. Through a systematic evaluation on the SL datasets of three different cell lines, we demonstrated that our model achieved better performance than the baseline methods and verified the effectiveness of using the L1000 gene expression features and the semi-supervise training technique in SL prediction.

SELECTION OF CITATIONS
SEARCH DETAIL
...