Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Bioinformatics ; 40(2)2024 Feb 01.
Article En | MEDLINE | ID: mdl-38305458

MOTIVATION: Diabetes is a chronic metabolic disorder that has been a major cause of blindness, kidney failure, heart attacks, stroke, and lower limb amputation across the world. To alleviate the impact of diabetes, researchers have developed the next generation of anti-diabetic drugs, known as dipeptidyl peptidase IV inhibitory peptides (DPP-IV-IPs). However, the discovery of these promising drugs has been restricted due to the lack of effective peptide-mining tools. RESULTS: Here, we presented StructuralDPPIV, a deep learning model designed for DPP-IV-IP identification, which takes advantage of both molecular graph features in amino acid and sequence information. Experimental results on the independent test dataset and two wet experiment datasets show that our model outperforms the other state-of-art methods. Moreover, to better study what StructuralDPPIV learns, we used CAM technology and perturbation experiment to analyze our model, which yielded interpretable insights into the reasoning behind prediction results. AVAILABILITY AND IMPLEMENTATION: The project code is available at https://github.com/WeiLab-BioChem/Structural-DPP-IV.


Deep Learning , Diabetes Mellitus , Humans , Dipeptidyl Peptidase 4 , Amino Acids , Peptides
2.
J Chem Inf Model ; 64(7): 2807-2816, 2024 Apr 08.
Article En | MEDLINE | ID: mdl-37252890

Anticancer peptides (ACPs) recently have been receiving increasing attention in cancer therapy due to their low consumption, few adverse side effects, and easy accessibility. However, it remains a great challenge to identify anticancer peptides via experimental approaches, requiring expensive and time-consuming experimental studies. In addition, traditional machine-learning-based methods are proposed for ACP prediction mainly depending on hand-crafted feature engineering, which normally achieves low prediction performance. In this study, we propose CACPP (Contrastive ACP Predictor), a deep learning framework based on the convolutional neural network (CNN) and contrastive learning for accurately predicting anticancer peptides. In particular, we introduce the TextCNN model to extract the high-latent features based on the peptide sequences only and exploit the contrastive learning module to learn more distinguishable feature representations to make better predictions. Comparative results on the benchmark data sets indicate that CACPP outperforms all the state-of-the-art methods in the prediction of anticancer peptides. Moreover, to intuitively show that our model has good classification ability, we visualize the dimension reduction of the features from our model and explore the relationship between ACP sequences and anticancer functions. Furthermore, we also discuss the influence of data set construction on model prediction and explore our model performance on the data sets with verified negative samples.


Benchmarking , Drug-Related Side Effects and Adverse Reactions , Humans , Machine Learning , Neural Networks, Computer , Peptides/pharmacology
3.
Comput Biol Med ; 167: 107666, 2023 12.
Article En | MEDLINE | ID: mdl-37956623

Molecular representation learning (MRL) is a fundamental task for drug discovery. However, previous deep-learning (DL) methods focus excessively on learning robust inner-molecular representations by mask-dominated pretraining frameworks, neglecting abundant chemical reactivity molecular relationships that have been demonstrated as the determining factor for various molecular property prediction tasks. Here, we present MolCAP to promote MRL, a graph-pretraining Transformer based on chemical reactivity (IMR) knowledge with prompted finetuning. Results show that MolCAP outperforms comparative methods based on traditional molecular pretraining frameworks, in 13 publicly available molecular datasets across a diversity of biomedical tasks. Prompted by MolCAP, even basic graph neural networks are capable of achieving surprising performance that outperforms previous models, indicating the promising prospect of applying reactivity information to MRL. In addition, manually designed molecular templets are potential to uncover the dataset bias. All in all, we expect our MolCAP to gain more chemical meaningful insights for the entire process of drug discovery.


Drug Discovery , Learning , Neural Networks, Computer
4.
Nat Commun ; 14(1): 6155, 2023 Oct 03.
Article En | MEDLINE | ID: mdl-37788995

Automating retrosynthesis with artificial intelligence expedites organic chemistry research in digital laboratories. However, most existing deep-learning approaches are hard to explain, like a "black box" with few insights. Here, we propose RetroExplainer, formulizing the retrosynthesis task into a molecular assembly process, containing several retrosynthetic actions guided by deep learning. To guarantee a robust performance of our model, we propose three units: a multi-sense and multi-scale Graph Transformer, structure-aware contrastive learning, and dynamic adaptive multi-task learning. The results on 12 large-scale benchmark datasets demonstrate the effectiveness of RetroExplainer, which outperforms the state-of-the-art single-step retrosynthesis approaches. In addition, the molecular assembly process renders our model with good interpretability, allowing for transparent decision-making and quantitative attribution. When extended to multi-step retrosynthesis planning, RetroExplainer has identified 101 pathways, in which 86.9% of the single reactions correspond to those already reported in the literature. As a result, RetroExplainer is expected to offer valuable insights for reliable, high-throughput, and high-quality organic synthesis in drug development.

5.
Brief Bioinform ; 24(6)2023 09 22.
Article En | MEDLINE | ID: mdl-37861173

NcRNA-encoded small peptides (ncPEPs) have recently emerged as promising targets and biomarkers for cancer immunotherapy. Therefore, identifying cancer-associated ncPEPs is crucial for cancer research. In this work, we propose CoraL, a novel supervised contrastive meta-learning framework for predicting cancer-associated ncPEPs. Specifically, the proposed meta-learning strategy enables our model to learn meta-knowledge from different types of peptides and train a promising predictive model even with few labeled samples. The results show that our model is capable of making high-confidence predictions on unseen cancer biomarkers with only five samples, potentially accelerating the discovery of novel cancer biomarkers for immunotherapy. Moreover, our approach remarkably outperforms existing deep learning models on 15 cancer-associated ncPEPs datasets, demonstrating its effectiveness and robustness. Interestingly, our model exhibits outstanding performance when extended for the identification of short open reading frames derived from ncPEPs, demonstrating the strong prediction ability of CoraL at the transcriptome level. Importantly, our feature interpretation analysis discovers unique sequential patterns as the fingerprint for each cancer-associated ncPEPs, revealing the relationship among certain cancer biomarkers that are validated by relevant literature and motif comparison. Overall, we expect CoraL to be a useful tool to decipher the pathogenesis of cancer and provide valuable information for cancer research. The dataset and source code of our proposed method can be found at https://github.com/Johnsunnn/CoraL.


Anthozoa , Neoplasms , Animals , Anthozoa/genetics , Neoplasms/genetics , Biomarkers, Tumor/genetics , Immunotherapy , Peptides/genetics , RNA, Untranslated
6.
Comput Biol Med ; 164: 107260, 2023 09.
Article En | MEDLINE | ID: mdl-37557052

The promoter region, positioned proximal to the transcription start sites, exerts control over the initiation of gene transcription by modulating the interaction with RNA polymerase. Consequently, the accurate recognition of promoter regions represents a critical focus within the bioinformatics domain. Although some methods leveraging pre-trained language models (PLMs) for promoter prediction have been proposed, the full potential of such PLMs remains largely untapped. In this study, we introduce PLPMpro, a model that capitalizes on prompt-learning and the pre-trained language model to enhance the prediction of promoter sequences. PLPMpro effectively harnesses the prompt learning paradigm to fully exploit the inherent capacities of the PLM, resulting in substantial improvements in prediction performance. Experiment results unequivocally demonstrate the efficacy of prompt learning in bolstering the capabilities of the pre-trained model. Consequently, PLPMpro surpasses both typical pre-trained model-based methods for promoter prediction and typical deep learning methods. Furthermore, we conduct various experiments to meticulously scrutinize the effects of different prompt learning settings and different numbers of soft modules on the model performance. More importantly, the interpretation experiment reveals that the pre-trained model captures biological semantics. Collectively, this research imparts a novel perspective on the optimal utilization of PLMs for addressing biological problems.


Computational Biology , Semantics , Promoter Regions, Genetic/genetics , Computational Biology/methods
7.
Comput Biol Med ; 164: 107238, 2023 09.
Article En | MEDLINE | ID: mdl-37515874

Recent research has highlighted the pivotal role of RNA post-transcriptional modifications in the regulation of RNA expression and function. Accurate identification of RNA modification sites is important for understanding RNA function. In this study, we propose a novel RNA modification prediction method, namely Rm-LR, which leverages a long-range-based deep learning approach to accurately predict multiple types of RNA modifications using RNA sequences only. Rm-LR incorporates two large-scale RNA language pre-trained models to capture discriminative sequential information and learn local important features, which are subsequently integrated through a bilinear attention network. Rm-LR supports a total of ten RNA modification types (m6A, m1A, m5C, m5U, m6Am, Ψ, Am, Cm, Gm, and Um) and significantly outperforms the state-of-the-art methods in terms of predictive capability on benchmark datasets. Experimental results show the effectiveness and superiority of Rm-LR in prediction of various RNA modifications, demonstrating the strong adaptability and robustness of our proposed model. We demonstrate that RNA language pretrained models enable to learn dense biological sequential representations from large-scale long-range RNA corpus, and meanwhile enhance the interpretability of the models. This work contributes to the development of accurate and reliable computational models for RNA modification prediction, providing insights into the complex landscape of RNA modifications.


Deep Learning , RNA/genetics , RNA/metabolism , Sequence Analysis, RNA/methods
8.
Comput Biol Med ; 161: 106946, 2023 07.
Article En | MEDLINE | ID: mdl-37244151

Drug-target interactions (DTI) prediction is a crucial task in drug discovery. Existing computational methods accelerate the drug discovery in this respect. However, most of them suffer from low feature representation ability, significantly affecting the predictive performance. To address the problem, we propose a novel neural network architecture named DrugormerDTI, which uses Graph Transformer to learn both sequential and topological information through the input molecule graph and Resudual2vec to learn the underlying relation between residues from proteins. By conducting ablation experiments, we verify the importance of each part of the DrugormerDTI. We also demonstrate the good feature extraction and expression capabilities of our model via comparing the mapping results of the attention layer and molecular docking results. Experimental results show that our proposed model performs better than baseline methods on four benchmarks. We demonstrate that the introduction of Graph Transformer and the design of residue are appropriate for drug-target prediction.


Drug Development , Neural Networks, Computer , Molecular Docking Simulation , Drug Development/methods , Drug Discovery/methods , Proteins/chemistry , Drug Interactions
9.
Bioinformatics ; 39(3)2023 03 01.
Article En | MEDLINE | ID: mdl-36897030

MOTIVATION: Plant Small Secreted Peptides (SSPs) play an important role in plant growth, development, and plant-microbe interactions. Therefore, the identification of SSPs is essential for revealing the functional mechanisms. Over the last few decades, machine learning-based methods have been developed, accelerating the discovery of SSPs to some extent. However, existing methods highly depend on handcrafted feature engineering, which easily ignores the latent feature representations and impacts the predictive performance. RESULTS: Here, we propose ExamPle, a novel deep learning model using Siamese network and multi-view representation for the explainable prediction of the plant SSPs. Benchmarking comparison results show that our ExamPle performs significantly better than existing methods in the prediction of plant SSPs. Also, our model shows excellent feature extraction ability. Importantly, by utilizing in silicomutagenesis experiment, ExamPle can discover sequential characteristics and identify the contribution of each amino acid for the predictions. The key novel principle learned by our model is that the head region of the peptide and some specific sequential patterns are strongly associated with the SSPs' functions. Thus, ExamPle is expected to be a useful tool for predicting plant SSPs and designing effective plant SSPs. AVAILABILITY AND IMPLEMENTATION: Our codes and datasets are available at https://github.com/Johnsunnn/ExamPle.


Deep Learning , Peptides , Machine Learning , Amino Acids , Benchmarking
10.
Adv Sci (Weinh) ; 10(11): e2206151, 2023 04.
Article En | MEDLINE | ID: mdl-36794291

Accurately predicting peptide secondary structures remains a challenging task due to the lack of discriminative information in short peptides. In this study, PHAT is proposed, a deep hypergraph learning framework for the prediction of peptide secondary structures and the exploration of downstream tasks. The framework includes a novel interpretable deep hypergraph multi-head attention network that uses residue-based reasoning for structure prediction. The algorithm can incorporate sequential semantic information from large-scale biological corpus and structural semantic information from multi-scale structural segmentation, leading to better accuracy and interpretability even with extremely short peptides. The interpretable models are able to highlight the reasoning of structural feature representations and the classification of secondary substructures. The importance of secondary structures in peptide tertiary structure reconstruction and downstream functional analysis is further demonstrated, highlighting the versatility of our models. To facilitate the use of the model, an online server is established which is accessible via http://inner.wei-group.net/PHAT/. The work is expected to assist in the design of functional peptides and contribute to the advancement of structural biology research.


Algorithms , Peptides , Protein Structure, Secondary , Peptides/chemistry
11.
Nucleic Acids Res ; 51(7): 3017-3029, 2023 04 24.
Article En | MEDLINE | ID: mdl-36796796

Here, we present DeepBIO, the first-of-its-kind automated and interpretable deep-learning platform for high-throughput biological sequence functional analysis. DeepBIO is a one-stop-shop web service that enables researchers to develop new deep-learning architectures to answer any biological question. Specifically, given any biological sequence data, DeepBIO supports a total of 42 state-of-the-art deep-learning algorithms for model training, comparison, optimization and evaluation in a fully automated pipeline. DeepBIO provides a comprehensive result visualization analysis for predictive models covering several aspects, such as model interpretability, feature analysis and functional sequential region discovery. Additionally, DeepBIO supports nine base-level functional annotation tasks using deep-learning architectures, with comprehensive interpretations and graphical visualizations to validate the reliability of annotated sites. Empowered by high-performance computers, DeepBIO allows ultra-fast prediction with up to million-scale sequence data in a few hours, demonstrating its usability in real application scenarios. Case study results show that DeepBIO provides an accurate, robust and interpretable prediction, demonstrating the power of deep learning in biological sequence functional analysis. Overall, we expect DeepBIO to ensure the reproducibility of deep-learning biological sequence analysis, lessen the programming and hardware burden for biologists and provide meaningful functional insights at both the sequence level and base level from biological sequences alone. DeepBIO is publicly available at https://inner.wei-group.net/DeepBIO.


The development of next-generation sequencing techniques has led to an exponential increase in the amount of biological sequence data accessible. It naturally poses a fundamental challenge­how to build the relationships from such large-scale sequences to their functions. In this work, we present DeepBIO, the first-of-its-kind automated and interpretable deep-learning platform for high-throughput biological sequence functional analysis. It enables researchers to develop new deep-learning architectures to answer any biological question in a fully automated pipeline. We expect DeepBIO to ensure the reproducibility of deep-learning-based biological sequence analysis, lessen the programming and hardware burden for biologists and provide meaningful functional insights at both the sequence level and base level from biological sequences alone.


Deep Learning , Reproducibility of Results , Algorithms , High-Throughput Nucleotide Sequencing
12.
Brief Bioinform ; 24(1)2023 01 19.
Article En | MEDLINE | ID: mdl-36562719

BACKGROUND: Cell-penetrating peptides (CPPs) have received considerable attention as a means of transporting pharmacologically active molecules into living cells without damaging the cell membrane, and thus hold great promise as future therapeutics. Recently, several machine learning-based algorithms have been proposed for predicting CPPs. However, most existing predictive methods do not consider the agreement (disagreement) between similar (dissimilar) CPPs and depend heavily on expert knowledge-based handcrafted features. RESULTS: In this study, we present SiameseCPP, a novel deep learning framework for automated CPPs prediction. SiameseCPP learns discriminative representations of CPPs based on a well-pretrained model and a Siamese neural network consisting of a transformer and gated recurrent units. Contrastive learning is used for the first time to build a CPP predictive model. Comprehensive experiments demonstrate that our proposed SiameseCPP is superior to existing baseline models for predicting CPPs. Moreover, SiameseCPP also achieves good performance on other functional peptide datasets, exhibiting satisfactory generalization ability.


Cell-Penetrating Peptides , Cell-Penetrating Peptides/metabolism , Algorithms , Biological Transport , Neural Networks, Computer , Machine Learning
13.
Genome Biol ; 23(1): 219, 2022 10 17.
Article En | MEDLINE | ID: mdl-36253864

In this study, we propose iDNA-ABF, a multi-scale deep biological language learning model that enables the interpretable prediction of DNA methylations based on genomic sequences only. Benchmarking comparisons show that our iDNA-ABF outperforms state-of-the-art methods for different methylation predictions. Importantly, we show the power of deep language learning in capturing both sequential and functional semantics information from background genomes. Moreover, by integrating the interpretable analysis mechanism, we well explain what the model learns, helping us build the mapping from the discovery of important sequential determinants to the in-depth analysis of their biological functions.


DNA Methylation , Language , Genomics , Models, Biological
14.
Bioinformatics ; 38(13): 3351-3360, 2022 06 27.
Article En | MEDLINE | ID: mdl-35604077

SUMMARY: Identifying the protein-peptide binding residues is fundamentally important to understand the mechanisms of protein functions and explore drug discovery. Although several computational methods have been developed, most of them highly rely on third-party tools or complex data preprocessing for feature design, easily resulting in low computational efficacy and suffering from low predictive performance. To address the limitations, we propose PepBCL, a novel BERT (Bidirectional Encoder Representation from Transformers) -based contrastive learning framework to predict the protein-peptide binding residues based on protein sequences only. PepBCL is an end-to-end predictive model that is independent of feature engineering. Specifically, we introduce a well pre-trained protein language model that can automatically extract and learn high-latent representations of protein sequences relevant for protein structures and functions. Further, we design a novel contrastive learning module to optimize the feature representations of binding residues underlying the imbalanced dataset. We demonstrate that our proposed method significantly outperforms the state-of-the-art methods under benchmarking comparison, and achieves more robust performance. Moreover, we found that we further improve the performance via the integration of traditional features and our learnt features. Interestingly, the interpretable analysis of our model highlights the flexibility and adaptability of deep learning-based protein language model to capture both conserved and non-conserved sequential characteristics of peptide-binding residues. Finally, to facilitate the use of our method, we establish an online predictive platform as the implementation of the proposed PepBCL, which is now available at http://server.wei-group.net/PepBCL/. AVAILABILITY AND IMPLEMENTATION: https://github.com/Ruheng-W/PepBCL. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Deep Learning , Proteins/chemistry , Peptides , Protein Binding , Amino Acid Sequence
15.
Methods ; 204: 258-262, 2022 08.
Article En | MEDLINE | ID: mdl-35093537

DNA N4-methylcytosine (4mC) is an important DNA modification and plays a crucial role in a variety of biological processes. Accurate 4mC site identification is fundamental to improving the understanding of 4mC biological functions and mechanisms. However, lots of identification approaches are limited to traditional machine learning, which leads to weak learning ability and a complex feature extraction process. Here, we propose Mouse4mC-BGRU, an advanced deep learning model that utilizes adaptive embedding based on bidirectional gated recurrent units (BGRU). Benchmark results show that our model performs better than the state-of-the-art methods in the prediction of 4mC sites in the mouse genome. By using adaptive features to extract representation, Mouse4mC-BGRU can capture the latent biology information of input sequence, which effectively enhances model representation ability. In addition, we visualize the training process of Mouse4mC-BGRU with dim reduction tools and intuitively show the effectiveness of our model, demonstrating that Mouse4mC-BGRU has great potential to be a powerful and practically useful tool to accurately identify 4mC sites.


Deep Learning , Animals , DNA/genetics , Genome , Machine Learning , Mice
16.
Brief Bioinform ; 23(1)2022 01 17.
Article En | MEDLINE | ID: mdl-34882225

Recently, machine learning methods have been developed to identify various peptide bio-activities. However, due to the lack of experimentally validated peptides, machine learning methods cannot provide a sufficiently trained model, easily resulting in poor generalizability. Furthermore, there is no generic computational framework to predict the bioactivities of different peptides. Thus, a natural question is whether we can use limited samples to build an effective predictive model for different kinds of peptides. To address this question, we propose Mutual Information Maximization Meta-Learning (MIMML), a novel meta-learning-based predictive model for bioactive peptide discovery. Using few samples from various functional peptides, MIMML can sufficiently learn the discriminative information amongst various functions and characterize functional differences. Experimental results show excellent performance of MIMML though using far fewer training samples as compared to the state-of-the-art methods. We also decipher the latent relationships among different kinds of functions to understand what meta-model learned to improve a specific task. In summary, this study is a pioneering work in the field of functional peptide mining and provides the first-of-its-kind solution for few-sample learning problems in biological sequence analysis, accelerating the new functional peptide discovery. The source codes and datasets are available on https://github.com/TearsWaiting/MIMML.


Machine Learning , Peptides , Peptides/chemistry , Software
17.
Bioinformatics ; 37(24): 4603-4610, 2021 12 11.
Article En | MEDLINE | ID: mdl-34601568

MOTIVATION: DNA methylation plays an important role in epigenetic modification, the occurrence, and the development of diseases. Therefore, identification of DNA methylation sites is critical for better understanding and revealing their functional mechanisms. To date, several machine learning and deep learning methods have been developed for the prediction of different DNA methylation types. However, they still highly rely on manual features, which can largely limit the high-latent information extraction. Moreover, most of them are designed for one specific DNA methylation type, and therefore cannot predict multiple methylation sites in multiple species simultaneously. In this study, we propose iDNA-ABT, an advanced deep learning model that utilizes adaptive embedding based on Bidirectional Encoder Representations from Transformers (BERT) together with transductive information maximization (TIM). RESULTS: Benchmark results show that our proposed iDNA-ABT can automatically and adaptively learn the distinguishing features of biological sequences from multiple species, and thus perform significantly better than the state-of-the-art methods in predicting three different DNA methylation types. In addition, TIM loss is proven to be effective in dichotomous tasks via the comparison experiment. Furthermore, we verify that our features have strong adaptability and robustness to different species through comparison of adaptive embedding and six handcrafted feature encodings. Importantly, our model shows great generalization ability in different species, demonstrating that our model can adaptively capture the cross-species differences and improve the predictive performance. For the convenient use of our method, we further established an online webserver as the implementation of the proposed iDNA-ABT. AVAILABILITY AND IMPLEMENTATION: Our proposed iDNA-ABT and data are freely accessible via http://server.wei-group.net/iDNA_ABT and our source codes are available for downloading in the GitHub repository (https://github.com/YUYING07/iDNA_ABT). SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


DNA Methylation , Deep Learning , Software , Machine Learning , Epigenesis, Genetic
...