Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Protein Sci ; 33(6): e4988, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38757367

RESUMEN

Identifying unknown functional properties of proteins is essential for understanding their roles in both health and disease states. The domain composition of a protein can reveal critical information in this context, as domains are structural and functional units that dictate how the protein should act at the molecular level. The expensive and time-consuming nature of wet-lab experimental approaches prompted researchers to develop computational strategies for predicting the functions of proteins. In this study, we proposed a new method called Domain2GO that infers associations between protein domains and function-defining gene ontology (GO) terms, thus redefining the problem as domain function prediction. Domain2GO uses documented protein-level GO annotations together with proteins' domain annotations. Co-annotation patterns of domains and GO terms in the same proteins are examined using statistical resampling to obtain reliable associations. As a use-case study, we evaluated the biological relevance of examples selected from the Domain2GO-generated domain-GO term mappings via literature review. Then, we applied Domain2GO to predict unknown protein functions by propagating domain-associated GO terms to proteins annotated with these domains. For function prediction performance evaluation and comparison against other methods, we employed Critical Assessment of Function Annotation 3 (CAFA3) challenge datasets. The results demonstrated the high potential of Domain2GO, particularly for predicting molecular function and biological process terms, along with advantages such as producing interpretable results and having an exceptionally low computational cost. The approach presented here can be extended to other ontologies and biological entities to investigate unknown relationships in complex and large-scale biological data. The source code, datasets, results, and user instructions for Domain2GO are available at https://github.com/HUBioDataLab/Domain2GO. Additionally, we offer a user-friendly online tool at https://huggingface.co/spaces/HUBioDataLab/Domain2GO, which simplifies the prediction of functions of previously unannotated proteins solely using amino acid sequences.


Asunto(s)
Anotación de Secuencia Molecular , Dominios Proteicos , Proteínas , Proteínas/química , Proteínas/metabolismo , Proteínas/genética , Bases de Datos de Proteínas , Biología Computacional/métodos , Ontología de Genes , Humanos , Programas Informáticos
2.
Comput Biol Med ; 169: 107810, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38134749

RESUMEN

Non-silent single nucleotide genetic variants, like nonsense changes and insertion-deletion variants, that affect protein function and length substantially are prevalent and are frequently misclassified. The low sensitivity and specificity of existing variant effect predictors for nonsense and indel variations restrict their use in clinical applications. We propose the Pathogenic Mutation Prediction (PMPred) method to predict the pathogenicity of single nucleotide variations, which impair protein function by prematurely terminating a protein's elongation during its synthesis. The prediction starts by monitoring functional effects (Gene Ontology annotation changes) of the change in sequence, using an existing ensemble machine learning model (UniGOPred). This, in turn, reveals the mutations that significantly deviate functionally from the wild-type sequence. We have identified novel harmful mutations in patient data and present them as motivating case studies. We also show that our method has increased sensitivity and specificity compared to state-of-the-art, especially in single nucleotide variations that produce large functional changes in the final protein. As further validation, we have done a comparative docking study on such a variation that is misclassified by existing methods and, using the altered binding affinities, show how PMPred can correctly predict the pathogenicity when other tools miss it. PMPred is freely accessible as a web service at https://pmpred.kansil.org/, and the related code is available at https://github.com/kansil/PMPred.


Asunto(s)
Exoma , Descubrimiento del Conocimiento , Humanos , Secuenciación del Exoma , Mutación , Nucleótidos , Biología Computacional/métodos
3.
Comput Struct Biotechnol J ; 21: 4743-4758, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37822561

RESUMEN

Background: Genomic variations may cause deleterious effects on protein functionality and perturb biological processes. Elucidating the effects of variations is critical for developing novel treatment strategies for diseases of genetic origin. Computational approaches have been aiding the work in this field by modeling and analyzing the mutational landscape. However, new approaches are required, especially for accurate representation and data-centric analysis of sequence variations. Method: In this study, we propose ASCARIS (Annotation and StruCture-bAsed RepresentatIon of Single amino acid variations), a method for the featurization (i.e., quantitative representation) of single amino acid variations (SAVs), which could be used for a variety of purposes, such as predicting their functional effects or building multi-omics-based integrative models. ASCARIS utilizes the direct and spatial correspondence between the location of the SAV on the sequence/structure and 30 different types of positional feature annotations (e.g., active/lipidation/glycosylation sites; calcium/metal/DNA binding, inter/transmembrane regions, etc.), along with structural features and physicochemical properties. The main novelty of this method lies in constructing reusable numerical representations of SAVs via functional annotations. Results: We statistically analyzed the relationship between these features and the consequences of variations and found that each carries information in this regard. To investigate potential applications of ASCARIS, we trained variant effect prediction models that utilize our SAV representations as input. We carried out an ablation study and a comparison against the state-of-the-art methods and observed that ASCARIS has a competing and complementary performance against widely-used predictors. ASCARIS can be used alone or in combination with other approaches to represent SAVs from a functional perspective. ASCARIS is available as a programmatic tool at https://github.com/HUBioDataLab/ASCARIS and as a web-service at https://huggingface.co/spaces/HUBioDataLab/ASCARIS.

5.
Bioinformatics ; 39(39 Suppl 1): i103-i110, 2023 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-37387156

RESUMEN

MOTIVATION: Utilizing AI-driven approaches for drug-target interaction (DTI) prediction require large volumes of training data which are not available for the majority of target proteins. In this study, we investigate the use of deep transfer learning for the prediction of interactions between drug candidate compounds and understudied target proteins with scarce training data. The idea here is to first train a deep neural network classifier with a generalized source training dataset of large size and then to reuse this pre-trained neural network as an initial configuration for re-training/fine-tuning purposes with a small-sized specialized target training dataset. To explore this idea, we selected six protein families that have critical importance in biomedicine: kinases, G-protein-coupled receptors (GPCRs), ion channels, nuclear receptors, proteases, and transporters. In two independent experiments, the protein families of transporters and nuclear receptors were individually set as the target datasets, while the remaining five families were used as the source datasets. Several size-based target family training datasets were formed in a controlled manner to assess the benefit provided by the transfer learning approach. RESULTS: Here, we present a systematic evaluation of our approach by pre-training a feed-forward neural network with source training datasets and applying different modes of transfer learning from the pre-trained source network to a target dataset. The performance of deep transfer learning is evaluated and compared with that of training the same deep neural network from scratch. We found that when the training dataset contains fewer than 100 compounds, transfer learning outperforms the conventional strategy of training the system from scratch, suggesting that transfer learning is advantageous for predicting binders to under-studied targets. AVAILABILITY AND IMPLEMENTATION: The source code and datasets are available at https://github.com/cansyl/TransferLearning4DTI. Our web-based service containing the ready-to-use pre-trained models is accessible at https://tl4dti.kansil.org.


Asunto(s)
Redes Neurales de la Computación , Péptido Hidrolasas , Programas Informáticos , Aprendizaje Automático
6.
J Cheminform ; 15(1): 16, 2023 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-36747300

RESUMEN

The identification of drug/compound-target interactions (DTIs) constitutes the basis of drug discovery, for which computational predictive approaches have been developed. As a relatively new data-driven paradigm, proteochemometric (PCM) modeling utilizes both protein and compound properties as a pair at the input level and processes them via statistical/machine learning. The representation of input samples (i.e., proteins and their ligands) in the form of quantitative feature vectors is crucial for the extraction of interaction-related properties during the artificial learning and subsequent prediction of DTIs. Lately, the representation learning approach, in which input samples are automatically featurized via training and applying a machine/deep learning model, has been utilized in biomedical sciences. In this study, we performed a comprehensive investigation of different computational approaches/techniques for protein featurization (including both conventional approaches and the novel learned embeddings), data preparation and exploration, machine learning-based modeling, and performance evaluation with the aim of achieving better data representations and more successful learning in DTI prediction. For this, we first constructed realistic and challenging benchmark datasets on small, medium, and large scales to be used as reliable gold standards for specific DTI modeling tasks. We developed and applied a network analysis-based splitting strategy to divide datasets into structurally different training and test folds. Using these datasets together with various featurization methods, we trained and tested DTI prediction models and evaluated their performance from different angles. Our main findings can be summarized under 3 items: (i) random splitting of datasets into train and test folds leads to near-complete data memorization and produce highly over-optimistic results, as a result, should be avoided, (ii) learned protein sequence embeddings work well in DTI prediction and offer high potential, despite interaction-related properties (e.g., structures) of proteins are unused during their self-supervised model training, and (iii) during the learning process, PCM models tend to rely heavily on compound features while partially ignoring protein features, primarily due to the inherent bias in DTI data, indicating the requirement for new and unbiased datasets. We hope this study will aid researchers in designing robust and high-performing data-driven DTI prediction systems that have real-world translational value in drug discovery.

7.
Brief Bioinform ; 24(2)2023 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-36736370

RESUMEN

As the number of protein sequences increases in biological databases, computational methods are required to provide accurate functional annotation with high coverage. Although several machine learning methods have been proposed for this purpose, there are still two main issues: (i) construction of reliable positive and negative training and validation datasets, and (ii) fair evaluation of their performances based on predefined experimental settings. To address these issues, we have developed ProFAB: Open Protein Functional Annotation Benchmark, which is a platform providing an infrastructure for a fair comparison of protein function prediction methods. ProFAB provides filtered and preprocessed protein annotation datasets and enables the training and evaluation of function prediction methods via several options. We believe that ProFAB will be useful for both computational and experimental researchers by enabling the utilization of ready-to-use datasets and machine learning algorithms for protein function prediction based on Gene Ontology terms and Enzyme Commission numbers. ProFAB is available at https://github.com/kansil/ProFAB and https://profab.kansil.org.


Asunto(s)
Benchmarking , Programas Informáticos , Anotación de Secuencia Molecular , Algoritmos , Proteínas/metabolismo , Biología Computacional/métodos
8.
Expert Opin Drug Discov ; 17(12): 1425-1441, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36444655

RESUMEN

BACKGROUND: Drug development productivity has been declining lately due to elevated costs and reduced discovery rates. Therefore, pharmaceutical companies have been seeking alternative ways to determine and evaluate drug candidates. RESEARCH DESIGN AND METHODS: In this work, we proposed a new computational approach to directly predict the regulatory approval of drug candidates, and implemented it as a method called 'DrugApp.' To accomplish this task, we employed multiple types of features including molecular and physicochemical properties of drug candidates, together with clinical trial and patent-related features, which are then processed by random forest classifiers to train our disease group-specific approval prediction models. RESULTS: Our evaluations indicated DrugApp has a high and robust prediction performance. Within a use-case study, we showed our method can predict phase IV trial drugs that are later withdrawn from the market due to severe side effects. Finally, we used DrugApp models to forecast the approval of drug candidates that are currently in phases I/II/III of clinical trials. CONCLUSIONS: We hope that our study will aid the research community in terms of evaluating and improving the process of drug development. The datasets, source code, results, and pre-trained models of DrugApp are freely available at https://github.com/HUBioDataLab/DrugApp.


Asunto(s)
Aprobación de Drogas , Programas Informáticos , Humanos , Desarrollo de Medicamentos , Aprendizaje Automático
9.
Bioinformatics ; 38(17): 4226-4229, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-35801913

RESUMEN

SUMMARY: Accurate prediction of the subcellular locations (SLs) of proteins is a critical topic in protein science. In this study, we present SLPred, an ensemble-based multi-view and multi-label protein subcellular localization prediction tool. For a query protein sequence, SLPred provides predictions for nine main SLs using independent machine-learning models trained for each location. We used UniProtKB/Swiss-Prot human protein entries and their curated SL annotations as our source data. We connected all disjoint terms in the UniProt SL hierarchy based on the corresponding term relationships in the cellular component category of Gene Ontology and constructed a training dataset that is both reliable and large scale using the re-organized hierarchy. We tested SLPred on multiple benchmarking datasets including our-in house sets and compared its performance against six state-of-the-art methods. Results indicated that SLPred outperforms other tools in the majority of cases. AVAILABILITY AND IMPLEMENTATION: SLPred is available both as an open-access and user-friendly web-server (https://slpred.kansil.org) and a stand-alone tool (https://github.com/kansil/SLPred). All datasets used in this study are also available at https://slpred.kansil.org. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Biología Computacional , Proteínas , Humanos , Bases de Datos de Proteínas , Ontología de Genes , Proteínas/genética , Secuencia de Aminoácidos , Transporte de Proteínas , Biología Computacional/métodos
11.
J Gastrointest Cancer ; 52(4): 1266-1276, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34910274

RESUMEN

PURPOSE: Computational approaches have been used at different stages of drug development with the purpose of decreasing the time and cost of conventional experimental procedures. Lately, techniques mainly developed and applied in the field of artificial intelligence (AI), have been transferred to different application domains such as biomedicine. METHODS: In this study, we conducted an investigative analysis via data-driven evaluation of potential hepatocellular carcinoma (HCC) therapeutics in the context of AI-assisted drug discovery/repurposing. First, we discussed basic concepts, computational approaches, databases, modeling approaches, and featurization techniques in drug discovery/repurposing. In the analysis part, we automatically integrated HCC-related biological entities such as genes/proteins, pathways, phenotypes, drugs/compounds, and other diseases with similar implications, and represented these heterogeneous relationships via a knowledge graph using the CROssBAR system. RESULTS: Following the system-level evaluation and selection of critical genes/proteins and pathways to target, our deep learning-based drug/compound-target protein interaction predictors DEEPScreen and MDeePred have been employed for predicting new bioactive drugs and compounds for these critical targets. Finally, we embedded ligands of selected HCC-associated proteins which had a significant enrichment with the CROssBAR system into a 2-D space to identify and repurpose small molecule inhibitors as potential drug candidates based on their molecular similarities to known HCC drugs. CONCLUSIONS: We expect that these series of data-driven analyses can be used as a roadmap to propose early-stage potential inhibitors (from database-scale sets of compounds) to both HCC and other complex diseases, which may subsequently be analyzed with more targeted in silico and experimental approaches.


Asunto(s)
Antineoplásicos/farmacología , Inteligencia Artificial , Carcinoma Hepatocelular/tratamiento farmacológico , Desarrollo de Medicamentos/métodos , Neoplasias Hepáticas/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Biología Computacional , Humanos , Neoplasias Hepáticas/patología , Terapia Molecular Dirigida
12.
PLoS Comput Biol ; 17(11): e1009171, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34843456

RESUMEN

Predictive approaches such as virtual screening have been used in drug discovery with the objective of reducing developmental time and costs. Current machine learning and network-based approaches have issues related to generalization, usability, or model interpretability, especially due to the complexity of target proteins' structure/function, and bias in system training datasets. Here, we propose a new method "DRUIDom" (DRUg Interacting Domain prediction) to identify bio-interactions between drug candidate compounds and targets by utilizing the domain modularity of proteins, to overcome problems associated with current approaches. DRUIDom is composed of two methodological steps. First, ligands/compounds are statistically mapped to structural domains of their target proteins, with the aim of identifying their interactions. As such, other proteins containing the same mapped domain or domain pair become new candidate targets for the corresponding compounds. Next, a million-scale dataset of small molecule compounds, including those mapped to domains in the previous step, are clustered based on their molecular similarities, and their domain associations are propagated to other compounds within the same clusters. Experimentally verified bioactivity data points, obtained from public databases, are meticulously filtered to construct datasets of active/interacting and inactive/non-interacting drug/compound-target pairs (~2.9M data points), and used as training data for calculating parameters of compound-domain mappings, which led to 27,032 high-confidence associations between 250 domains and 8,165 compounds, and a finalized output of ~5 million new compound-protein interactions. DRUIDom is experimentally validated by syntheses and bioactivity analyses of compounds predicted to target LIM-kinase proteins, which play critical roles in the regulation of cell motility, cell cycle progression, and differentiation through actin filament dynamics. We showed that LIMK-inhibitor-2 and its derivatives significantly block the cancer cell migration through inhibition of LIMK phosphorylation and the downstream protein cofilin. One of the derivative compounds (LIMKi-2d) was identified as a promising candidate due to its action on resistant Mahlavu liver cancer cells. The results demonstrated that DRUIDom can be exploited to identify drug candidate compounds for intended targets and to predict new target proteins based on the defined compound-domain relationships. Datasets, results, and the source code of DRUIDom are fully-available at: https://github.com/cansyl/DRUIDom.


Asunto(s)
Quinasas Lim/antagonistas & inhibidores , Quinasas Lim/química , Factores Despolimerizantes de la Actina/química , Factores Despolimerizantes de la Actina/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Biología Computacional , Simulación por Computador , Desarrollo de Medicamentos , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Humanos , Técnicas In Vitro , Ligandos , Quinasas Lim/metabolismo , Aprendizaje Automático , Simulación del Acoplamiento Molecular , Invasividad Neoplásica/prevención & control , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Farmacología en Red/estadística & datos numéricos , Fosforilación/efectos de los fármacos , Dominios Proteicos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Interfaz Usuario-Computador
13.
Nucleic Acids Res ; 49(16): e96, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34181736

RESUMEN

Systemic analysis of available large-scale biological/biomedical data is critical for studying biological mechanisms, and developing novel and effective treatment approaches against diseases. However, different layers of the available data are produced using different technologies and scattered across individual computational resources without any explicit connections to each other, which hinders extensive and integrative multi-omics-based analysis. We aimed to address this issue by developing a new data integration/representation methodology and its application by constructing a biological data resource. CROssBAR is a comprehensive system that integrates large-scale biological/biomedical data from various resources and stores them in a NoSQL database. CROssBAR is enriched with the deep-learning-based prediction of relationships between numerous data entries, which is followed by the rigorous analysis of the enriched data to obtain biologically meaningful modules. These complex sets of entities and relationships are displayed to users via easy-to-interpret, interactive knowledge graphs within an open-access service. CROssBAR knowledge graphs incorporate relevant genes-proteins, molecular interactions, pathways, phenotypes, diseases, as well as known/predicted drugs and bioactive compounds, and they are constructed on-the-fly based on simple non-programmatic user queries. These intensely processed heterogeneous networks are expected to aid systems-level research, especially to infer biological mechanisms in relation to genes, proteins, their ligands, and diseases.


Asunto(s)
Biología Computacional/métodos , Programas Informáticos , Bases de Datos de Compuestos Químicos , Bases de Datos Genéticas , Aprendizaje Profundo , Humanos
14.
Chem Sci ; 11(9): 2531-2557, 2020 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-33209251

RESUMEN

The identification of physical interactions between drug candidate compounds and target biomolecules is an important process in drug discovery. Since conventional screening procedures are expensive and time consuming, computational approaches are employed to provide aid by automatically predicting novel drug-target interactions (DTIs). In this study, we propose a large-scale DTI prediction system, DEEPScreen, for early stage drug discovery, using deep convolutional neural networks. One of the main advantages of DEEPScreen is employing readily available 2-D structural representations of compounds at the input level instead of conventional descriptors that display limited performance. DEEPScreen learns complex features inherently from the 2-D representations, thus producing highly accurate predictions. The DEEPScreen system was trained for 704 target proteins (using curated bioactivity data) and finalized with rigorous hyper-parameter optimization tests. We compared the performance of DEEPScreen against the state-of-the-art on multiple benchmark datasets to indicate the effectiveness of the proposed approach and verified selected novel predictions through molecular docking analysis and literature-based validation. Finally, JAK proteins that were predicted by DEEPScreen as new targets of a well-known drug cladribine were experimentally demonstrated in vitro on cancer cells through STAT3 phosphorylation, which is the downstream effector protein. The DEEPScreen system can be exploited in the fields of drug discovery and repurposing for in silico screening of the chemogenomic space, to provide novel DTIs which can be experimentally pursued. The source code, trained "ready-to-use" prediction models, all datasets and the results of this study are available at ; https://github.com/cansyl/DEEPscreen.

16.
Bioinformatics ; 36(14): 4227-4230, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32407491

RESUMEN

SUMMARY: iBioProVis is an interactive tool for visual analysis of the compound bioactivity space in the context of target proteins, drugs and drug candidate compounds. iBioProVis tool takes target protein identifiers and, optionally, compound SMILES as input, and uses the state-of-the-art non-linear dimensionality reduction method t-Distributed Stochastic Neighbor Embedding (t-SNE) to plot the distribution of compounds embedded in a 2D map, based on the similarity of structural properties of compounds and in the context of compounds' cognate targets. Similar compounds, which are embedded to proximate points on the 2D map, may bind the same or similar target proteins. Thus, iBioProVis can be used to easily observe the structural distribution of one or two target proteins' known ligands on the 2D compound space, and to infer new binders to the same protein, or to infer new potential target(s) for a compound of interest, based on this distribution. Principal component analysis (PCA) projection of the input compounds is also provided, Hence the user can interactively observe the same compound or a group of selected compounds which is projected by both PCA and embedded by t-SNE. iBioProVis also provides detailed information about drugs and drug candidate compounds through cross-references to widely used and well-known databases, in the form of linked table views. Two use-case studies were demonstrated, one being on angiotensin-converting enzyme 2 (ACE2) protein which is Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Spike protein receptor. ACE2 binding compounds and seven antiviral drugs were closely embedded in which two of them have been under clinical trial for Coronavirus disease 19 (COVID-19). AVAILABILITY AND IMPLEMENTATION: iBioProVis and its carefully filtered dataset are available at https://ibpv.kansil.org/ for public use. CONTACT: vatalay@metu.edu.tr. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Modelos Moleculares , Peptidil-Dipeptidasa A/química , Programas Informáticos , Glicoproteína de la Espiga del Coronavirus/química , Enzima Convertidora de Angiotensina 2 , Inhibidores de la Enzima Convertidora de Angiotensina/química , Antivirales/química , Betacoronavirus , COVID-19 , Infecciones por Coronavirus , Humanos , Internet , Pandemias , Neumonía Viral , Análisis de Componente Principal , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 3/química , SARS-CoV-2 , Interfaz Usuario-Computador
17.
Genome Biol ; 20(1): 244, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31744546

RESUMEN

BACKGROUND: The Critical Assessment of Functional Annotation (CAFA) is an ongoing, global, community-driven effort to evaluate and improve the computational annotation of protein function. RESULTS: Here, we report on the results of the third CAFA challenge, CAFA3, that featured an expanded analysis over the previous CAFA rounds, both in terms of volume of data analyzed and the types of analysis performed. In a novel and major new development, computational predictions and assessment goals drove some of the experimental assays, resulting in new functional annotations for more than 1000 genes. Specifically, we performed experimental whole-genome mutation screening in Candida albicans and Pseudomonas aureginosa genomes, which provided us with genome-wide experimental data for genes associated with biofilm formation and motility. We further performed targeted assays on selected genes in Drosophila melanogaster, which we suspected of being involved in long-term memory. CONCLUSION: We conclude that while predictions of the molecular function and biological process annotations have slightly improved over time, those of the cellular component have not. Term-centric prediction of experimental annotations remains equally challenging; although the performance of the top methods is significantly better than the expectations set by baseline methods in C. albicans and D. melanogaster, it leaves considerable room and need for improvement. Finally, we report that the CAFA community now involves a broad range of participants with expertise in bioinformatics, biological experimentation, biocuration, and bio-ontologies, working together to improve functional annotation, computational function prediction, and our ability to manage big data in the era of large experimental screens.


Asunto(s)
Anotación de Secuencia Molecular/tendencias , Animales , Biopelículas , Candida albicans/genética , Drosophila melanogaster/genética , Genoma Bacteriano , Genoma Fúngico , Humanos , Locomoción , Memoria a Largo Plazo , Anotación de Secuencia Molecular/métodos , Pseudomonas aeruginosa/genética
18.
Sci Rep ; 9(1): 7344, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31089211

RESUMEN

Automated protein function prediction is critical for the annotation of uncharacterized protein sequences, where accurate prediction methods are still required. Recently, deep learning based methods have outperformed conventional algorithms in computer vision and natural language processing due to the prevention of overfitting and efficient training. Here, we propose DEEPred, a hierarchical stack of multi-task feed-forward deep neural networks, as a solution to Gene Ontology (GO) based protein function prediction. DEEPred was optimized through rigorous hyper-parameter tests, and benchmarked using three types of protein descriptors, training datasets with varying sizes and GO terms form different levels. Furthermore, in order to explore how training with larger but potentially noisy data would change the performance, electronically made GO annotations were also included in the training process. The overall predictive performance of DEEPred was assessed using CAFA2 and CAFA3 challenge datasets, in comparison with the state-of-the-art protein function prediction methods. Finally, we evaluated selected novel annotations produced by DEEPred with a literature-based case study considering the 'biofilm formation process' in Pseudomonas aeruginosa. This study reports that deep learning algorithms have significant potential in protein function prediction; particularly when the source data is large. The neural network architecture of DEEPred can also be applied to the prediction of the other types of ontological associations. The source code and all datasets used in this study are available at: https://github.com/cansyl/DEEPred .


Asunto(s)
Redes Neurales de la Computación , Proteínas/metabolismo , Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Minería de Datos , Aprendizaje Profundo , Ontología de Genes , Humanos , Modelos Biológicos , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/fisiología , Programas Informáticos
19.
Brief Bioinform ; 20(5): 1878-1912, 2019 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-30084866

RESUMEN

The identification of interactions between drugs/compounds and their targets is crucial for the development of new drugs. In vitro screening experiments (i.e. bioassays) are frequently used for this purpose; however, experimental approaches are insufficient to explore novel drug-target interactions, mainly because of feasibility problems, as they are labour intensive, costly and time consuming. A computational field known as 'virtual screening' (VS) has emerged in the past decades to aid experimental drug discovery studies by statistically estimating unknown bio-interactions between compounds and biological targets. These methods use the physico-chemical and structural properties of compounds and/or target proteins along with the experimentally verified bio-interaction information to generate predictive models. Lately, sophisticated machine learning techniques are applied in VS to elevate the predictive performance. The objective of this study is to examine and discuss the recent applications of machine learning techniques in VS, including deep learning, which became highly popular after giving rise to epochal developments in the fields of computer vision and natural language processing. The past 3 years have witnessed an unprecedented amount of research studies considering the application of deep learning in biomedicine, including computational drug discovery. In this review, we first describe the main instruments of VS methods, including compound and protein features (i.e. representations and descriptors), frequently used libraries and toolkits for VS, bioactivity databases and gold-standard data sets for system training and benchmarking. We subsequently review recent VS studies with a strong emphasis on deep learning applications. Finally, we discuss the present state of the field, including the current challenges and suggest future directions. We believe that this survey will provide insight to the researchers working in the field of computational drug discovery in terms of comprehending and developing novel bio-prediction methods.


Asunto(s)
Sistemas de Administración de Bases de Datos , Aprendizaje Profundo , Descubrimiento de Drogas , Simulación por Computador
20.
BMC Bioinformatics ; 19(1): 334, 2018 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-30241466

RESUMEN

BACKGROUND: The automated prediction of the enzymatic functions of uncharacterized proteins is a crucial topic in bioinformatics. Although several methods and tools have been proposed to classify enzymes, most of these studies are limited to specific functional classes and levels of the Enzyme Commission (EC) number hierarchy. Besides, most of the previous methods incorporated only a single input feature type, which limits the applicability to the wide functional space. Here, we proposed a novel enzymatic function prediction tool, ECPred, based on ensemble of machine learning classifiers. RESULTS: In ECPred, each EC number constituted an individual class and therefore, had an independent learning model. Enzyme vs. non-enzyme classification is incorporated into ECPred along with a hierarchical prediction approach exploiting the tree structure of the EC nomenclature. ECPred provides predictions for 858 EC numbers in total including 6 main classes, 55 subclass classes, 163 sub-subclass classes and 634 substrate classes. The proposed method is tested and compared with the state-of-the-art enzyme function prediction tools by using independent temporal hold-out and no-Pfam datasets constructed during this study. CONCLUSIONS: ECPred is presented both as a stand-alone and a web based tool to provide probabilistic enzymatic function predictions (at all five levels of EC) for uncharacterized protein sequences. Also, the datasets of this study will be a valuable resource for future benchmarking studies. ECPred is available for download, together with all of the datasets used in this study, at: https://github.com/cansyl/ECPred . ECPred webserver can be accessed through http://cansyl.metu.edu.tr/ECPred.html .


Asunto(s)
Biología Computacional/métodos , Enzimas/clasificación , Enzimas/metabolismo , Análisis de Secuencia de Proteína/métodos , Programas Informáticos , Terminología como Asunto , Algoritmos , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...