Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 827
Filtrar
1.
Brief Bioinform ; 25(2)2024 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-38385873

RESUMO

Lysine lactylation (Kla) is a newly discovered posttranslational modification that is involved in important life activities, such as glycolysis-related cell function, macrophage polarization and nervous system regulation, and has received widespread attention due to the Warburg effect in tumor cells. In this work, we first design a natural language processing method to automatically extract the 3D structural features of Kla sites, avoiding potential biases caused by manually designed structural features. Then, we establish two Kla prediction frameworks, Attention-based feature fusion Kla model (ABFF-Kla) and EBFF-Kla, to integrate the sequence features and the structure features based on the attention layer and embedding layer, respectively. The results indicate that ABFF-Kla and Embedding-based feature fusion Kla model (EBFF-Kla), which fuse features from protein sequences and spatial structures, have better predictive performance than that of models that use only sequence features. Our work provides an approach for the automatic extraction of protein structural features, as well as a flexible framework for Kla prediction. The source code and the training data of the ABFF-Kla and the EBFF-Kla are publicly deposited at: https://github.com/ispotato/Lactylation_model.


Assuntos
Lisina , Processamento de Linguagem Natural , Sequência de Aminoácidos , Domínios Proteicos , Processamento de Proteína Pós-Traducional
2.
Brief Bioinform ; 25(3)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38706321

RESUMO

Antiviral peptides (AVPs) have shown potential in inhibiting viral attachment, preventing viral fusion with host cells and disrupting viral replication due to their unique action mechanisms. They have now become a broad-spectrum, promising antiviral therapy. However, identifying effective AVPs is traditionally slow and costly. This study proposed a new two-stage computational framework for AVP identification. The first stage identifies AVPs from a wide range of peptides, and the second stage recognizes AVPs targeting specific families or viruses. This method integrates contrastive learning and multi-feature fusion strategy, focusing on sequence information and peptide characteristics, significantly enhancing predictive ability and interpretability. The evaluation results of the model show excellent performance, with accuracy of 0.9240 and Matthews correlation coefficient (MCC) score of 0.8482 on the non-AVP independent dataset, and accuracy of 0.9934 and MCC score of 0.9869 on the non-AMP independent dataset. Furthermore, our model can predict antiviral activities of AVPs against six key viral families (Coronaviridae, Retroviridae, Herpesviridae, Paramyxoviridae, Orthomyxoviridae, Flaviviridae) and eight viruses (FIV, HCV, HIV, HPIV3, HSV1, INFVA, RSV, SARS-CoV). Finally, to facilitate user accessibility, we built a user-friendly web interface deployed at https://awi.cuhk.edu.cn/∼dbAMP/AVP/.


Assuntos
Antivirais , Biologia Computacional , Peptídeos , Antivirais/farmacologia , Peptídeos/química , Biologia Computacional/métodos , Humanos , Vírus , Aprendizado de Máquina , Algoritmos
3.
Brief Bioinform ; 25(3)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38605642

RESUMO

MicroRNAs (miRNAs) synergize with various biomolecules in human cells resulting in diverse functions in regulating a wide range of biological processes. Predicting potential disease-associated miRNAs as valuable biomarkers contributes to the treatment of human diseases. However, few previous methods take a holistic perspective and only concentrate on isolated miRNA and disease objects, thereby ignoring that human cells are responsible for multiple relationships. In this work, we first constructed a multi-view graph based on the relationships between miRNAs and various biomolecules, and then utilized graph attention neural network to learn the graph topology features of miRNAs and diseases for each view. Next, we added an attention mechanism again, and developed a multi-scale feature fusion module, aiming to determine the optimal fusion results for the multi-view topology features of miRNAs and diseases. In addition, the prior attribute knowledge of miRNAs and diseases was simultaneously added to achieve better prediction results and solve the cold start problem. Finally, the learned miRNA and disease representations were then concatenated and fed into a multi-layer perceptron for end-to-end training and predicting potential miRNA-disease associations. To assess the efficacy of our model (called MUSCLE), we performed 5- and 10-fold cross-validation (CV), which got average the Area under ROC curves of 0.966${\pm }$0.0102 and 0.973${\pm }$0.0135, respectively, outperforming most current state-of-the-art models. We then examined the impact of crucial parameters on prediction performance and performed ablation experiments on the feature combination and model architecture. Furthermore, the case studies about colon cancer, lung cancer and breast cancer also fully demonstrate the good inductive capability of MUSCLE. Our data and code are free available at a public GitHub repository: https://github.com/zht-code/MUSCLE.git.


Assuntos
Neoplasias do Colo , Neoplasias Pulmonares , MicroRNAs , Humanos , Músculos , Aprendizagem , MicroRNAs/genética , Algoritmos , Biologia Computacional
4.
Brief Bioinform ; 24(1)2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36502435

RESUMO

Protein-protein interactions (PPIs) are a major component of the cellular biochemical reaction network. Rich sequence information and machine learning techniques reduce the dependence of exploring PPIs on wet experiments, which are costly and time-consuming. This paper proposes a PPI prediction model, multi-scale architecture residual network for PPIs (MARPPI), based on dual-channel and multi-feature. Multi-feature leverages Res2vec to obtain the association information between residues, and utilizes pseudo amino acid composition, autocorrelation descriptors and multivariate mutual information to achieve the amino acid composition and order information, physicochemical properties and information entropy, respectively. Dual channel utilizes multi-scale architecture improved ResNet network which extracts protein sequence features to reduce protein feature loss. Compared with other advanced methods, MARPPI achieves 96.03%, 99.01% and 91.80% accuracy in the intraspecific datasets of Saccharomyces cerevisiae, Human and Helicobacter pylori, respectively. The accuracy on the two interspecific datasets of Human-Bacillus anthracis and Human-Yersinia pestis is 97.29%, and 95.30%, respectively. In addition, results on specific datasets of disease (neurodegenerative and metabolic disorders) demonstrate the ability to detect hidden interactions. To better illustrate the performance of MARPPI, evaluations on independent datasets and PPIs network suggest that MARPPI can be used to predict cross-species interactions. The above shows that MARPPI can be regarded as a concise, efficient and accurate tool for PPI datasets.


Assuntos
Biologia Computacional , Mapeamento de Interação de Proteínas , Humanos , Mapeamento de Interação de Proteínas/métodos , Biologia Computacional/métodos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Mapas de Interação de Proteínas , Aminoácidos/metabolismo
5.
Brief Bioinform ; 24(6)2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37930024

RESUMO

Development of robust and effective strategies for synthesizing new compounds, drug targeting and constructing GEnome-scale Metabolic models (GEMs) requires a deep understanding of the underlying biological processes. A critical step in achieving this goal is accurately identifying the categories of pathways in which a compound participated. However, current machine learning-based methods often overlook the multifaceted nature of compounds, resulting in inaccurate pathway predictions. Therefore, we present a novel framework on Multi-View Multi-Label Learning for Metabolic Pathway Inference, hereby named MVML-MPI. First, MVML-MPI learns the distinct compound representations in parallel with corresponding compound encoders to fully extract features. Subsequently, we propose an attention-based mechanism that offers a fusion module to complement these multi-view representations. As a result, MVML-MPI accurately represents and effectively captures the complex relationship between compounds and metabolic pathways and distinguishes itself from current machine learning-based methods. In experiments conducted on the Kyoto Encyclopedia of Genes and Genomes pathways dataset, MVML-MPI outperformed state-of-the-art methods, demonstrating the superiority of MVML-MPI and its potential to utilize the field of metabolic pathway design, which can aid in optimizing drug-like compounds and facilitating the development of GEMs. The code and data underlying this article are freely available at https://github.com/guofei-tju/MVML-MPI. Contact:  jtang@cse.sc.edu, guofei@csu.edu.com or wuxi_dyj@csj.uestc.edu.cn.


Assuntos
Aprendizado de Máquina , Redes e Vias Metabólicas
6.
Brief Bioinform ; 24(6)2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37930025

RESUMO

Drug combination therapy has gradually become a promising treatment strategy for complex or co-existing diseases. As drug-drug interactions (DDIs) may cause unexpected adverse drug reactions, DDI prediction is an important task in pharmacology and clinical applications. Recently, researchers have proposed several deep learning methods to predict DDIs. However, these methods mainly exploit the chemical or biological features of drugs, which is insufficient and limits the performances of DDI prediction. Here, we propose a new deep multimodal feature fusion framework for DDI prediction, DMFDDI, which fuses drug molecular graph, DDI network and the biochemical similarity features of drugs to predict DDIs. To fully extract drug molecular structure, we introduce an attention-gated graph neural network for capturing the global features of the molecular graph and the local features of each atom. A sparse graph convolution network is introduced to learn the topological structure information of the DDI network. In the multimodal feature fusion module, an attention mechanism is used to efficiently fuse different features. To validate the performance of DMFDDI, we compare it with 10 state-of-the-art methods. The comparison results demonstrate that DMFDDI achieves better performance in DDI prediction. Our method DMFDDI is implemented in Python using the Pytorch machine-learning library, and it is freely available at https://github.com/DHUDEBLab/DMFDDI.git.


Assuntos
Aprendizado de Máquina , Redes Neurais de Computação , Interações Medicamentosas , Estrutura Molecular , Biblioteca Gênica
7.
Brief Bioinform ; 24(5)2023 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-37615358

RESUMO

Non-coding RNA (ncRNA) plays a critical role in biology. ncRNAs from the same family usually have similar functions, as a result, it is essential to predict ncRNA families before identifying their functions. There are two primary methods for predicting ncRNA families, namely, traditional biological methods and computational methods. In traditional biological methods, a lot of manpower and resources are required to predict ncRNA families. Therefore, this paper proposed a new ncRNA family prediction method called MFPred based on computational methods. MFPred identified ncRNA families by extracting sequence features of ncRNAs, and it possessed three primary modules, including (1) four ncRNA sequences encoding and feature extraction module, which encoded ncRNA sequences and extracted four different features of ncRNA sequences, (2) dynamic Bi_GRU and feature fusion module, which extracted contextual information features of the ncRNA sequence and (3) ResNet_SE module that extracted local information features of the ncRNA sequence. In this study, MFPred was compared with the previously proposed ncRNA family prediction methods using two frequently used public ncRNA datasets, NCY and nRC. The results showed that MFPred outperformed other prediction methods in the two datasets.


Assuntos
Biologia Computacional , RNA não Traduzido , Humanos , Biologia Computacional/métodos , RNA não Traduzido/genética
8.
Brief Bioinform ; 24(4)2023 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-37401369

RESUMO

As the volume of protein sequence and structure data grows rapidly, the functions of the overwhelming majority of proteins cannot be experimentally determined. Automated annotation of protein function at a large scale is becoming increasingly important. Existing computational prediction methods are typically based on expanding the relatively small number of experimentally determined functions to large collections of proteins with various clues, including sequence homology, protein-protein interaction, gene co-expression, etc. Although there has been some progress in protein function prediction in recent years, the development of accurate and reliable solutions still has a long way to go. Here we exploit AlphaFold predicted three-dimensional structural information, together with other non-structural clues, to develop a large-scale approach termed PredGO to annotate Gene Ontology (GO) functions for proteins. We use a pre-trained language model, geometric vector perceptrons and attention mechanisms to extract heterogeneous features of proteins and fuse these features for function prediction. The computational results demonstrate that the proposed method outperforms other state-of-the-art approaches for predicting GO functions of proteins in terms of both coverage and accuracy. The improvement of coverage is because the number of structures predicted by AlphaFold is greatly increased, and on the other hand, PredGO can extensively use non-structural information for functional prediction. Moreover, we show that over 205 000 ($\sim $100%) entries in UniProt for human are annotated by PredGO, over 186 000 ($\sim $90%) of which are based on predicted structure. The webserver and database are available at http://predgo.denglab.org/.


Assuntos
Biologia Computacional , Proteínas , Humanos , Biologia Computacional/métodos , Proteínas/química , Sequência de Aminoácidos , Redes Neurais de Computação , Bases de Dados Factuais , Bases de Dados de Proteínas
9.
Brief Bioinform ; 25(1)2023 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-38216539

RESUMO

In the drug development process, approximately 30% of failures are attributed to drug safety issues. In particular, the first-in-human (FIH) trial of a new drug represents one of the highest safety risks, and initial dose selection is crucial for ensuring safety in clinical trials. With traditional dose estimation methods, which extrapolate data from animals to humans, catastrophic events have occurred during Phase I clinical trials due to interspecies differences in compound sensitivity and unknown molecular mechanisms. To address this issue, this study proposes a CrossFuse-extreme gradient boosting (XGBoost) method that can directly predict the maximum recommended daily dose of a compound based on existing human research data, providing a reference for FIH dose selection. This method not only integrates multiple features, including molecular representations, physicochemical properties and compound-protein interactions, but also improves feature selection based on cross-validation. The results demonstrate that the CrossFuse-XGBoost method not only improves prediction accuracy compared to that of existing local weighted methods [k-nearest neighbor (k-NN) and variable k-NN (v-NN)] but also solves the low prediction coverage issue of v-NN, achieving full coverage of the external validation set and enabling more reliable predictions. Furthermore, this study offers a high level of interpretability by identifying the importance of different features in model construction. The 241 features with the most significant impact on the maximum recommended daily dose were selected, providing references for optimizing the structure of new compounds and guiding experimental research. The datasets and source code are freely available at https://github.com/cqmu-lq/CrossFuse-XGBoost.


Assuntos
Projetos de Pesquisa , Software , Animais , Humanos , Análise por Conglomerados
10.
Brief Bioinform ; 24(1)2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36642410

RESUMO

Anticancer peptides (ACPs) are the types of peptides that have been demonstrated to have anticancer activities. Using ACPs to prevent cancer could be a viable alternative to conventional cancer treatments because they are safer and display higher selectivity. Due to ACP identification being highly lab-limited, expensive and lengthy, a computational method is proposed to predict ACPs from sequence information in this study. The process includes the input of the peptide sequences, feature extraction in terms of ordinal encoding with positional information and handcrafted features, and finally feature selection. The whole model comprises of two modules, including deep learning and machine learning algorithms. The deep learning module contained two channels: bidirectional long short-term memory (BiLSTM) and convolutional neural network (CNN). Light Gradient Boosting Machine (LightGBM) was used in the machine learning module. Finally, this study voted the three models' classification results for the three paths resulting in the model ensemble layer. This study provides insights into ACP prediction utilizing a novel method and presented a promising performance. It used a benchmark dataset for further exploration and improvement compared with previous studies. Our final model has an accuracy of 0.7895, sensitivity of 0.8153 and specificity of 0.7676, and it was increased by at least 2% compared with the state-of-the-art studies in all metrics. Hence, this paper presents a novel method that can potentially predict ACPs more effectively and efficiently. The work and source codes are made available to the community of researchers and developers at https://github.com/khanhlee/acp-ope/.


Assuntos
Aprendizado Profundo , Peptídeos/uso terapêutico , Aprendizado de Máquina , Algoritmos , Redes Neurais de Computação
11.
Brief Bioinform ; 24(4)2023 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-37291761

RESUMO

Adverse drug-drug interactions (DDIs) have become an increasingly serious problem in the medical and health system. Recently, the effective application of deep learning and biomedical knowledge graphs (KGs) have improved the DDI prediction performance of computational models. However, the problems of feature redundancy and KG noise also arise, bringing new challenges for researchers. To overcome these challenges, we proposed a Multi-Channel Feature Fusion model for multi-typed DDI prediction (MCFF-MTDDI). Specifically, we first extracted drug chemical structure features, drug pairs' extra label features, and KG features of drugs. Then, these different features were effectively fused by a multi-channel feature fusion module. Finally, multi-typed DDIs were predicted through the fully connected neural network. To our knowledge, we are the first to integrate the extra label information into KG-based multi-typed DDI prediction; besides, we innovatively proposed a novel KG feature learning method and a State Encoder to obtain target drug pairs' KG-based features which contained more abundant and more key drug-related KG information with less noise; furthermore, a Gated Recurrent Unit-based multi-channel feature fusion module was proposed in an innovative way to yield more comprehensive feature information about drug pairs, effectively alleviating the problem of feature redundancy. We experimented with four datasets in the multi-class and the multi-label prediction tasks to comprehensively evaluate the performance of MCFF-MTDDI for predicting interactions of known-known drugs, known-new drugs and new-new drugs. In addition, we further conducted ablation studies and case studies. All the results fully demonstrated the effectiveness of MCFF-MTDDI.


Assuntos
Sistemas de Liberação de Medicamentos , Redes Neurais de Computação , Humanos , Interações Medicamentosas , Pesquisadores
12.
Methods ; 229: 125-132, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38964595

RESUMO

DNase I hypersensitive sites (DHSs) are chromatin regions highly sensitive to DNase I enzymes. Studying DHSs is crucial for understanding complex transcriptional regulation mechanisms and localizing cis-regulatory elements (CREs). Numerous studies have indicated that disease-related loci are often enriched in DHSs regions, underscoring the importance of identifying DHSs. Although wet experiments exist for DHSs identification, they are often labor-intensive. Therefore, there is a strong need to develop computational methods for this purpose. In this study, we used experimental data to construct a benchmark dataset. Seven feature extraction methods were employed to capture information about human DHSs. The F-score was applied to filter the features. By comparing the prediction performance of various classification algorithms through five-fold cross-validation, random forest was proposed to perform the final model construction. The model could produce an overall prediction accuracy of 0.859 with an AUC value of 0.837. We hope that this model can assist scholars conducting DNase research in identifying these sites.

13.
BMC Genomics ; 25(1): 466, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38741045

RESUMO

BACKGROUND: Protein-protein interactions (PPIs) hold significant importance in biology, with precise PPI prediction as a pivotal factor in comprehending cellular processes and facilitating drug design. However, experimental determination of PPIs is laborious, time-consuming, and often constrained by technical limitations. METHODS: We introduce a new node representation method based on initial information fusion, called FFANE, which amalgamates PPI networks and protein sequence data to enhance the precision of PPIs' prediction. A Gaussian kernel similarity matrix is initially established by leveraging protein structural resemblances. Concurrently, protein sequence similarities are gauged using the Levenshtein distance, enabling the capture of diverse protein attributes. Subsequently, to construct an initial information matrix, these two feature matrices are merged by employing weighted fusion to achieve an organic amalgamation of structural and sequence details. To gain a more profound understanding of the amalgamated features, a Stacked Autoencoder (SAE) is employed for encoding learning, thereby yielding more representative feature representations. Ultimately, classification models are trained to predict PPIs by using the well-learned fusion feature. RESULTS: When employing 5-fold cross-validation experiments on SVM, our proposed method achieved average accuracies of 94.28%, 97.69%, and 84.05% in terms of Saccharomyces cerevisiae, Homo sapiens, and Helicobacter pylori datasets, respectively. CONCLUSION: Experimental findings across various authentic datasets validate the efficacy and superiority of this fusion feature representation approach, underscoring its potential value in bioinformatics.


Assuntos
Biologia Computacional , Mapeamento de Interação de Proteínas , Mapeamento de Interação de Proteínas/métodos , Biologia Computacional/métodos , Algoritmos , Helicobacter pylori/metabolismo , Helicobacter pylori/genética , Máquina de Vetores de Suporte , Proteínas/metabolismo , Proteínas/química , Humanos , Mapas de Interação de Proteínas , Bases de Dados de Proteínas
14.
Brief Bioinform ; 23(5)2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-35945157

RESUMO

RNA 5-hydroxymethylcytosine (5hmC) is a kind of RNA modification, which is related to the life activities of many organisms. Studying its distribution is very important to reveal its biological function. Previously, high-throughput sequencing was used to identify 5hmC, but it is expensive and inefficient. Therefore, machine learning is used to identify 5hmC sites. Here, we design a model called R5hmCFDV, which is mainly divided into feature representation, feature fusion and classification. (i) Pseudo dinucleotide composition, dinucleotide binary profile and frequency, natural vector and physicochemical property are used to extract features from four aspects: nucleotide composition, coding, natural language and physical and chemical properties. (ii) To strengthen the relevance of features, we construct a novel feature fusion method. Firstly, the attention mechanism is employed to process four single features, stitch them together and feed them to the convolution layer. After that, the output data are processed by BiGRU and BiLSTM, respectively. Finally, the features of these two parts are fused by the multiply function. (iii) We design the deep voting algorithm for classification by imitating the soft voting mechanism in the Python package. The base classifiers contain deep neural network (DNN), convolutional neural network (CNN) and improved gated recurrent unit (GRU). And then using the principle of soft voting, the corresponding weights are assigned to the predicted probabilities of the three classifiers. The predicted probability values are multiplied by the corresponding weights and then summed to obtain the final prediction results. We use 10-fold cross-validation to evaluate the model, and the evaluation indicators are significantly improved. The prediction accuracy of the two datasets is as high as 95.41% and 93.50%, respectively. It demonstrates the stronger competitiveness and generalization performance of our model. In addition, all datasets and source codes can be found at https://github.com/HongyanShi026/R5hmCFDV.


Assuntos
Redes Neurais de Computação , RNA , 5-Metilcitosina/análogos & derivados , Aprendizado de Máquina , Nucleotídeos , RNA/genética
15.
Brief Bioinform ; 23(1)2022 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-34671814

RESUMO

One of the main problems with the joint use of multiple drugs is that it may cause adverse drug interactions and side effects that damage the body. Therefore, it is important to predict potential drug interactions. However, most of the available prediction methods can only predict whether two drugs interact or not, whereas few methods can predict interaction events between two drugs. Accurately predicting interaction events of two drugs is more useful for researchers to study the mechanism of the interaction of two drugs. In the present study, we propose a novel method, MDF-SA-DDI, which predicts drug-drug interaction (DDI) events based on multi-source drug fusion, multi-source feature fusion and transformer self-attention mechanism. MDF-SA-DDI is mainly composed of two parts: multi-source drug fusion and multi-source feature fusion. First, we combine two drugs in four different ways and input the combined drug feature representation into four different drug fusion networks (Siamese network, convolutional neural network and two auto-encoders) to obtain the latent feature vectors of the drug pairs, in which the two auto-encoders have the same structure, and their main difference is the number of neurons in the input layer of the two auto-encoders. Then, we use transformer blocks that include self-attention mechanism to perform latent feature fusion. We conducted experiments on three different tasks with two datasets. On the small dataset, the area under the precision-recall-curve (AUPR) and F1 scores of our method on task 1 reached 0.9737 and 0.8878, respectively, which were better than the state-of-the-art method. On the large dataset, the AUPR and F1 scores of our method on task 1 reached 0.9773 and 0.9117, respectively. In task 2 and task 3 of two datasets, our method also achieved the same or better performance as the state-of-the-art method. More importantly, the case studies on five DDI events are conducted and achieved satisfactory performance. The source codes and data are available at https://github.com/ShenggengLin/MDF-SA-DDI.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Redes Neurais de Computação , Interações Medicamentosas , Humanos , Oligossacarídeos , Software
16.
Brief Bioinform ; 23(1)2022 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-34472593

RESUMO

Phage therapy has become one of the most promising alternatives to antibiotics in the treatment of bacterial diseases, and identifying phage-host interactions (PHIs) helps to understand the possible mechanism through which a phage infects bacteria to guide the development of phage therapy. Compared with wet experiments, computational methods of identifying PHIs can reduce costs and save time and are more effective and economic. In this paper, we propose a PHI prediction method with a generative adversarial network (GAN)-based data augmentation and sequence-based feature fusion (PHIAF). First, PHIAF applies a GAN-based data augmentation module, which generates pseudo PHIs to alleviate the data scarcity. Second, PHIAF fuses the features originated from DNA and protein sequences for better performance. Third, PHIAF utilizes an attention mechanism to consider different contributions of DNA/protein sequence-derived features, which also provides interpretability of the prediction model. In computational experiments, PHIAF outperforms other state-of-the-art PHI prediction methods when evaluated via 5-fold cross-validation (AUC and AUPR are 0.88 and 0.86, respectively). An ablation study shows that data augmentation, feature fusion and an attention mechanism are all beneficial to improve the prediction performance of PHIAF. Additionally, four new PHIs with the highest PHIAF score in the case study were verified by recent literature. In conclusion, PHIAF is a promising tool to accelerate the exploration of phage therapy.


Assuntos
Bacteriófagos , Processamento de Imagem Assistida por Computador , Bacteriófagos/genética , Processamento de Imagem Assistida por Computador/métodos
17.
Brief Bioinform ; 23(1)2022 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-34953462

RESUMO

More than 6000 human diseases have been recorded to be caused by non-synonymous single nucleotide polymorphisms (nsSNPs). Rapid and accurate prediction of pathogenic nsSNPs can improve our understanding of the principle and design of new drugs, which remains an unresolved challenge. In the present work, a new computational approach, termed MSRes-MutP, is proposed based on ResNet blocks with multi-scale kernel size to predict disease-associated nsSNPs. By feeding the serial concatenation of the extracted four types of features, the performance of MSRes-MutP does not obviously improve. To address this, a second model FFMSRes-MutP is developed, which utilizes deep feature fusion strategy and multi-scale 2D-ResNet and 1D-ResNet blocks to extract relevant two-dimensional features and physicochemical properties. FFMSRes-MutP with the concatenated features achieves a better performance than that with individual features. The performance of FFMSRes-MutP is benchmarked on five different datasets. It achieves the Matthew's correlation coefficient (MCC) of 0.593 and 0.618 on the PredictSNP and MMP datasets, which are 0.101 and 0.210 higher than that of the existing best method PredictSNP1. When tested on the HumDiv and HumVar datasets, it achieves MCC of 0.9605 and 0.9507, and area under curve (AUC) of 0.9796 and 0.9748, which are 0.1747 and 0.2669, 0.0853 and 0.1335, respectively, higher than the existing best methods PolyPhen-2 and FATHMM (weighted). In addition, on blind test using a third-party dataset, FFMSRes-MutP performs as the second-best predictor (with MCC and AUC of 0.5215 and 0.7633, respectively), when compared with the other four predictors. Extensive benchmarking experiments demonstrate that FFMSRes-MutP achieves effective feature fusion and can be explored as a useful approach for predicting disease-associated nsSNPs. The webserver is freely available at http://csbio.njust.edu.cn/bioinf/ffmsresmutp/ for academic use.


Assuntos
Aprendizado Profundo , Doença/genética , Polimorfismo de Nucleotídeo Único , Algoritmos , Área Sob a Curva , Microambiente Celular , Biologia Computacional/métodos , Humanos , Mutação , Preparações Farmacêuticas
18.
Anal Biochem ; 690: 115510, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38513769

RESUMO

Phosphorylation is indispensable in comprehending biological processes, while biological experimental methods for identifying phosphorylation sites are tedious and arduous. With the rapid growth of biotechnology, deep learning methods have made significant progress in site prediction tasks. Nevertheless, most existing predictors only consider protein sequence information, that limits the capture of protein spatial information. Building upon the latest advancement in protein structure prediction by AlphaFold2, a novel integrated deep learning architecture PhosAF is developed to predict phosphorylation sites in human proteins by integrating CMA-Net and MFC-Net, which considers sequence and structure information predicted by AlphaFold2. Here, CMA-Net module is composed of multiple convolutional neural network layers and multi-head attention is appended to obtaining the local and long-term dependencies of sequence features. Meanwhile, the MFC-Net module composed of deep neural network layers is used to capture the complex representations of evolutionary and structure features. Furthermore, different features are combined to predict the final phosphorylation sites. In addition, we put forward a new strategy to construct reliable negative samples via protein secondary structures. Experimental results on independent test data and case study indicate that our model PhosAF surpasses the current most advanced methods in phosphorylation site prediction.

19.
Anal Biochem ; 689: 115492, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38458307

RESUMO

DNA 4 mC plays a crucial role in the genetic expression process of organisms. However, existing deep learning algorithms have shortcomings in the ability to represent DNA sequence features. In this paper, we propose a 4 mC site identification algorithm, DNABert-4mC, based on a fusion of the pruned pre-training DNABert-Pruning model and artificial feature encoding to identify 4 mC sites. The algorithm prunes and compresses the DNABert model, resulting in the pruned pre-training model DNABert-Pruning. This model reduces the number of parameters and removes redundancy from output features, yielding more precise feature representations while upholding accuracy.Simultaneously, the algorithm constructs an artificial feature encoding module to assist the DNABert-Pruning model in feature representation, effectively supplementing the information that is missing from the pre-trained features. The algorithm also introduces the AFF-4mC fusion strategy, which combines artificial feature encoding with the DNABert-Pruning model, to improve the feature representation capability of DNA sequences in multi-semantic spaces and better extract 4 mC sites and the distribution of nucleotide importance within the sequence. In experiments on six independent test sets, the DNABert-4mC algorithm achieved an average AUC value of 93.81%, outperforming seven other advanced algorithms with improvements of 2.05%, 5.02%, 11.32%, 5.90%, 12.02%, 2.42% and 2.34%, respectively.


Assuntos
Algoritmos , DNA , DNA/genética , Nucleotídeos
20.
Environ Sci Technol ; 58(14): 6149-6157, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38556993

RESUMO

The global management for persistent, mobile, and toxic (PMT) and very persistent and very mobile (vPvM) substances has been further strengthened with the rapid increase of emerging contaminants. The development of a ready-to-use and publicly available tool for the high-throughput screening of PMT/vPvM substances is thus urgently needed. However, the current model building with the coupling of conventional algorithms, small-scale data set, and simplistic features hinders the development of a robust model for screening PMT/vPvM with wide application domains. Here, we construct a graph convolutional network (GCN)-enhanced model with feature fusion of a molecular graph and molecular descriptors to effectively utilize the significant correlation between critical descriptors and PMT/vPvM substances. The model is built with 213,084 substances following the latest PMT classification criteria. The application domains of the GCN-enhanced model assessed by kernel density estimation demonstrate the high suitability for high-throughput screening PMT/vPvM substances with both a high accuracy rate (86.6%) and a low false-negative rate (6.8%). An online server named PMT/vPvM profiler is further developed with a user-friendly web interface (http://www.pmt.zj.cn/). Our study facilitates a more efficient evaluation of PMT/vPvM substances with a globally accessible screening platform.


Assuntos
Algoritmos , Ensaios de Triagem em Larga Escala
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa