Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters








Publication year range
1.
Cell ; 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39389057

ABSTRACT

Current metagenomic tools can fail to identify highly divergent RNA viruses. We developed a deep learning algorithm, termed LucaProt, to discover highly divergent RNA-dependent RNA polymerase (RdRP) sequences in 10,487 metatranscriptomes generated from diverse global ecosystems. LucaProt integrates both sequence and predicted structural information, enabling the accurate detection of RdRP sequences. Using this approach, we identified 161,979 potential RNA virus species and 180 RNA virus supergroups, including many previously poorly studied groups, as well as RNA virus genomes of exceptional length (up to 47,250 nucleotides) and genomic complexity. A subset of these novel RNA viruses was confirmed by RT-PCR and RNA/DNA sequencing. Newly discovered RNA viruses were present in diverse environments, including air, hot springs, and hydrothermal vents, with virus diversity and abundance varying substantially among ecosystems. This study advances virus discovery, highlights the scale of the virosphere, and provides computational tools to better document the global RNA virome.

2.
Int J Biol Macromol ; 281(Pt 2): 136147, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39357703

ABSTRACT

Protein-DNA interactions play critical roles in various biological processes and are essential for drug discovery. However, traditional experimental methods are labor-intensive and unable to keep pace with the increasing volume of protein sequences, leading to a substantial number of proteins lacking DNA-binding annotations. Therefore, developing an efficient computational method to identify protein-DNA binding sites is crucial. Unfortunately, most existing computational methods rely on manually selected features or protein structure information, making these methods inapplicable to large-scale prediction tasks. In this study, we introduced PDNAPred, a sequence-based method that combines two pre-trained protein language models with a designed CNN-GRU network to identify DNA-binding sites. Additionally, to tackle the issue of imbalanced dataset samples, we employed focal loss. Our comprehensive experiments demonstrated that PDNAPred significantly improved the accuracy of DNA-binding site prediction, outperforming existing state-of-the-art sequence-based methods. Remarkably, PDNAPred also achieved results comparable to advanced structure-based methods. The designed CNN-GRU network enhances its capability to detect DNA-binding sites accurately. Furthermore, we validated the versatility of PDNAPred by training it on RNA-binding site datasets, showing its potential as a general framework for amino acid binding site prediction. Finally, we conducted model interpretability analysis to elucidate the reasons behind PDNAPred's outstanding performance.

3.
Int J Biol Macromol ; 280(Pt 3): 135762, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39322150

ABSTRACT

Allergy is a prevalent phenomenon, involving allergens such as nuts and milk. Avoiding exposure to allergens is the most effective preventive measure against allergic reactions. However, current homology-based methods for identifying allergenic proteins encounter challenges when dealing with non-homologous data. Traditional machine learning approaches rely on manually extracted features, which lack important protein functional characteristics, including evolutionary information. Consequently, there is still considerable room for improvement in existing methods. In this study, we present PreAlgPro, a method for identifying allergenic proteins based on pre-trained protein language models and deep learning techniques. Specifically, we employed the ProtT5 model to extract protein embedding features, replacing the manual feature extraction step. Furthermore, we devised an Attention-CNN neural network architecture to identify potential features that contribute to the classification of allergenic proteins. The performance of our model was evaluated on four independent test sets, and the experimental results demonstrate that PreAlgPro surpasses existing state-of-the-art methods. Additionally, we collected allergenic protein samples to validate the robustness of the model and conducted an analysis of model interpretability.

4.
Int J Biol Macromol ; 280(Pt 3): 136048, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39332561

ABSTRACT

Vesicular transport is a critical cellular process responsible for the proper organization and functioning of eukaryotic cells. This mechanism relies on specialized vesicles that shuttle macromolecules, such as proteins, across the cellular landscape, a process pivotal to maintaining cellular homeostasis. Disruptions in vesicular transport have been linked to various disease mechanisms, including cancer and neurodegenerative disorders. In this study, we present vesiMCNN, a novel computational approach that integrates pre-trained protein language models with a multi-window scanning convolutional neural network architecture to accurately identify vesicular transport proteins. To the best of our knowledge, this is the first study to leverage the power of pre-trained language models in combination with the multi-window scanning technique for this task. Our method achieved a Matthews Correlation Coefficient (MCC) of 0.558 and an Area Under the Receiver Operating Characteristic (AUC-ROC) of 0.933, outperforming existing state-of-the-art approaches. Additionally, we have curated a comprehensive benchmark dataset for the study of vesicular transport proteins, which can facilitate further research in this field. The remarkable performance of our model, combined with the comprehensive dataset and novel deep learning model, marks a significant advancement in the field of vesicular transport protein research.

5.
Brief Bioinform ; 25(6)2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39316944

ABSTRACT

As a main subtype of post-translational modification (PTM), protein lysine acylations (PLAs) play crucial roles in regulating diverse functions of proteins. With recent advancements in proteomics technology, the identification of PTM is becoming a data-rich field. A large amount of experimentally verified data is urgently required to be translated into valuable biological insights. With computational approaches, PLA can be accurately detected across the whole proteome, even for organisms with small-scale datasets. Herein, a comprehensive summary of 166 in silico PLA prediction methods is presented, including a single type of PLA site and multiple types of PLA sites. This recapitulation covers important aspects that are critical for the development of a robust predictor, including data collection and preparation, sample selection, feature representation, classification algorithm design, model evaluation, and method availability. Notably, we discuss the application of protein language models and transfer learning to solve the small-sample learning issue. We also highlight the prediction methods developed for functionally relevant PLA sites and species/substrate/cell-type-specific PLA sites. In conclusion, this systematic review could potentially facilitate the development of novel PLA predictors and offer useful insights to researchers from various disciplines.


Subject(s)
Computational Biology , Lysine , Protein Processing, Post-Translational , Proteins , Humans , Acylation , Algorithms , Computational Biology/methods , Databases, Protein , Lysine/metabolism , Lysine/chemistry , Proteins/metabolism , Proteins/chemistry , Software
6.
Int J Biol Macromol ; 280(Pt 1): 135741, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39293623

ABSTRACT

Post-translational modifications (PTMs) diversify protein functions by adding/removing chemical groups to certain amino acid. As a newly-reported PTM, lysine ß-hydroxybutyrylation (Kbhb) presents a new avenue to functional proteomics. Therefore, accurate and efficient prediction of Kbhb sites is imperative. However, the current experimental methods for identifying PTM sites are often expensive and time-consuming. Up to now, there is no computational method proposed for Kbhb sites detection. To this end, we present the first deep learning-based method, termed SLAM, to in silico identify lysine ß-hydroxybutyrylation sites. The performance of SLAM is evaluated on both 5-fold cross-validation and independent test, achieving 0.890, 0.899, 0.907 and 0.923 in terms of AUROC values, on the general and species-specific independent test sets, respectively. As one example, we predicted the potential Kbhb sites in human S-adenosyl-L-homocysteine hydrolase, which is in agreement with experimentally-verified Kbhb sites. In summary, our method could enable accurate and efficient characterization of novel Kbhb sites that are crucial for the function and stability of proteins and could be applied in the structure-guided identification of other important PTM sites. The SLAM online service and source code is available at https://ai4bio.online/SLAM and https://github.com/Gabriel-QIN/SLAM, respectively.

7.
Int J Biol Macromol ; 280(Pt 1): 135599, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39276905

ABSTRACT

The computational identification of nucleic acid-binding proteins (NABP) is of great significance for understanding the mechanisms of these biological activities and drug discovery. Although a bunch of sequence-based methods have been proposed to predict NABP and achieved promising performance, the structure information is often overlooked. On the other hand, the power of popular protein language models (pLM) has seldom been harnessed for predicting NABPs. In this study, we propose a novel framework called GraphNABP, to predict NABP by integrating sequence and predicted 3D structure information. Specifically, sequence embeddings and protein molecular graphs were first obtained from ProtT5 protein language model and predicted 3D structures, respectively. Then, graph attention (GAT) and bidirectional long short-term memory (BiLSTM) neural networks were used to enhance feature representations. Finally, a fully connected layer is used to predict NABPs. To the best of our knowledge, this is the first time to integrate AlphaFold and protein language models for the prediction of NABPs. The performances on multiple independent test sets indicate that GraphNABP outperforms other state-of-the-art methods. Our results demonstrate the effectiveness of pLM embeddings and structural information for NABP prediction. The codes and data used in this study are available at https://github.com/lixiangli01/GraphNABP.

8.
Metab Eng Commun ; 19: e00248, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39310048

ABSTRACT

Plastic waste has caused a global environmental crisis. Biocatalytic depolymerization mediated by enzymes has emerged as an efficient and sustainable alternative for plastic treatment and recycling. However, it is challenging and time-consuming to discover novel plastic-degrading enzymes using conventional cultivation-based or omics methods. There is a growing interest in developing effective computational methods to identify new enzymes with desirable plastic degradation functionalities by exploring the ever-increasing databases of protein sequences. In this study, we designed an innovative machine learning-based framework, named PEZy-Miner, to mine for enzymes with high potential in degrading plastics of interest. Two datasets integrating information from experimentally verified enzymes and homologs with unknown plastic-degrading activity were created respectively, covering eleven types of plastic substrates. Protein language models and binary classification models were developed to predict enzymatic degradation of plastics along with confidence and uncertainty estimation. PEZy-Miner exhibited high prediction accuracy and stability when validated on experimentally verified enzymes. Furthermore, by masking the experimentally verified enzymes and blending them into homolog dataset, PEZy-Miner effectively concentrated the experimentally verified entries by 14∼30 times while shortlisting promising plastic-degrading enzyme candidates. We applied PEZy-Miner to 0.1 million putative sequences, out of which 27 new sequences were identified with high confidence. This study provided a new computational tool for mining and recommending promising new plastic-degrading enzymes.

9.
Antibodies (Basel) ; 13(3)2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39311379

ABSTRACT

Therapeutic antibodies such as monoclonal antibodies (mAbs), bispecific and multispecific antibodies are pivotal in therapeutic protein development and have transformed disease treatments across various therapeutic areas. The integrity of therapeutic antibodies, however, is compromised by sequence liabilities, notably deamidation, where asparagine (N) and glutamine (Q) residues undergo chemical degradations. Deamidation negatively impacts the efficacy, stability, and safety of diverse classes of antibodies, thus necessitating the critical need for the early and accurate identification of vulnerable sites. In this article, a comprehensive antibody deamidation-specific dataset (n = 2285) of varied modalities was created by using high-throughput automated peptide mapping followed by supervised machine learning to predict the deamidation propensities, as well as the extents, throughout the entire antibody sequences. We propose a novel chimeric deep learning model, integrating protein language model (pLM)-derived embeddings with local sequence information for enhanced deamidation predictions. Remarkably, this model requires only sequence inputs, eliminating the need for laborious feature engineering. Our approach demonstrates state-of-the-art performance, offering a streamlined workflow for high-throughput automated peptide mapping and deamidation prediction, with the potential of broader applicability to other antibody sequence liabilities.

10.
Adv Sci (Weinh) ; : e2406305, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39319609

ABSTRACT

Antimicrobial peptides (AMPs) are a promising solution for treating antibiotic-resistant pathogens. However, efficient generation of diverse AMPs without prior knowledge of peptide structures or sequence alignments remains a challenge. Here, ProT-Diff is introduced, a modularized deep generative approach that combines a pretrained protein language model with a diffusion model for the de novo generation of AMPs sequences. ProT-Diff generates thousands of AMPs with diverse lengths and structures within a few hours. After silico physicochemical screening, 45 peptides are selected for experimental validation. Forty-four peptides showed antimicrobial activity against both gram-positive or gram-negative bacteria. Among broad-spectrum peptides, AMP_2 exhibited potent antimicrobial activity, low hemolysis, and minimal cytotoxicity. An in vivo assessment demonstrated its effectiveness against a drug-resistant E. coli strain in acute peritonitis. This study not only introduces a viable and user-friendly strategy for de novo generation of antimicrobial peptides, but also provides potential antimicrobial drug candidates with excellent activity. It is believed that this study will facilitate the development of other peptide-based drug candidates in the future, as well as proteins with tailored characteristics.

11.
J Comput Biol ; 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39246251

ABSTRACT

The identification of intrinsically disordered proteins and their functional roles is largely dependent on the performance of computational predictors, necessitating a high standard of accuracy in these tools. In this context, we introduce a novel series of computational predictors, termed PDFll (Predictors of Disorder and Function of proteins from the Language of Life), which are designed to offer precise predictions of protein disorder and associated functional roles based on protein sequences. PDFll is developed through a two-step process. Initially, it leverages large-scale protein language models (pLMs), trained on an extensive dataset comprising billions of protein sequences. Subsequently, the embeddings derived from pLMs are integrated into streamlined, yet sophisticated, deep-learning models to generate predictions. These predictions notably surpass the performance of existing state-of-the-art predictors, particularly those that forecast disorder and function without utilizing evolutionary information.

12.
Comput Biol Med ; 181: 109048, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39182368

ABSTRACT

Neuropeptides are the most ubiquitous neurotransmitters in the immune system, regulating various biological processes. Neuropeptides play a significant role for the discovery of new drugs and targets for nervous system disorders. Traditional experimental methods for identifying neuropeptides are time-consuming and costly. Although several computational methods have been developed to predict the neuropeptides, the accuracy is still not satisfactory due to the representability of the extracted features. In this work, we propose an efficient and interpretable model, NeuroPpred-SHE, for predicting neuropeptides by selecting the optimal feature subset from both hand-crafted features and embeddings of a protein language model. Specially, we first employed a pre-trained T5 protein language model to extract embedding features and twelve other encoding methods to extract hand-crafted features from peptide sequences, respectively. Secondly, we fused both embedding features and hand-crafted features to enhance the feature representability. Thirdly, we utilized random forest (RF), Max-Relevance and Min-Redundancy (mRMR) and eXtreme Gradient Boosting (XGBoost) methods to select the optimal feature subset from the fused features. Finally, we employed five machine learning methods (GBDT, XGBoost, SVM, MLP, and LightGBM) to build the models. Our results show that the model based on GBDT achieves the best performance. Furthermore, our final model was compared with other state-of-the-art methods on an independent test set, the results indicate that our model achieves an AUROC of 97.8 % which is higher than all the other state-of-the-art predictors. Our model is available at: https://github.com/wenjean/NeuroPpred-SHE.


Subject(s)
Neuropeptides , Computational Biology/methods , Humans , Databases, Protein , Machine Learning , Sequence Analysis, Protein/methods
13.
Genes (Basel) ; 15(8)2024 Aug 18.
Article in English | MEDLINE | ID: mdl-39202449

ABSTRACT

Protein and nucleic acid binding site prediction is a critical computational task that benefits a wide range of biological processes. Previous studies have shown that feature selection holds particular significance for this prediction task, making the generation of more discriminative features a key area of interest for many researchers. Recent progress has shown the power of protein language models in handling protein sequences, in leveraging the strengths of attention networks, and in successful applications to tasks such as protein structure prediction. This naturally raises the question of the applicability of protein language models in predicting protein and nucleic acid binding sites. Various approaches have explored this potential. This paper first describes the development of protein language models. Then, a systematic review of the latest methods for predicting protein and nucleic acid binding sites is conducted by covering benchmark sets, feature generation methods, performance comparisons, and feature ablation studies. These comparisons demonstrate the importance of protein language models for the prediction task. Finally, the paper discusses the challenges of protein and nucleic acid binding site prediction and proposes possible research directions and future trends. The purpose of this survey is to furnish researchers with actionable suggestions for comprehending the methodologies used in predicting protein-nucleic acid binding sites, fostering the creation of protein-centric language models, and tackling real-world obstacles encountered in this field.


Subject(s)
Protein Binding , Binding Sites , Computational Biology/methods , Proteins/chemistry , Proteins/metabolism , Proteins/genetics , Nucleic Acids/metabolism , Nucleic Acids/chemistry , Humans
14.
Int J Biol Macromol ; 278(Pt 4): 135064, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39182884

ABSTRACT

Enzyme specificity towards cofactors like NAD(P)H is crucial for applications in bioremediation and eco-friendly chemical synthesis. Despite their role in converting pollutants and creating sustainable products, predicting enzyme specificity faces challenges due to sparse data and inadequate models. To bridge this gap, we developed the cutting-edge INSIGHT platform to enhance the prediction of coenzyme specificity in NAD(P)-dependent enzymes. INSIGHT integrates extensive data from principal bioinformatics resources, concentrating on both NADH and NADPH specificities, and utilizes advanced protein language models to refine the predictions. This integration not only strengthens computational predictions but also meets the practical demands of high-throughput screening and optimization. Experimental validation confirms INSIGHT's effectiveness, boosting our ability to engineer enzymes for efficient, sustainable industrial and environmental processes. This work advances the practical use of computational tools in enzyme research, addressing industrial needs and offering scalable solutions for environmental challenges.


Subject(s)
NADP , NAD , Protein Engineering , NADP/metabolism , NADP/chemistry , Substrate Specificity , NAD/metabolism , NAD/chemistry , Protein Engineering/methods , Computational Biology/methods , Models, Molecular , Coenzymes/metabolism , Coenzymes/chemistry
15.
bioRxiv ; 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39131363

ABSTRACT

Exponential increases in microbial and viral genomic data demand transformational advances in scalable, generalizable frameworks for their interpretation. Standard homology-based functional analyses are hindered by the rapid divergence of microbial and especially viral genomes and proteins that significantly decreases the volume of usable data. Here, we present Protein Set Transformer (PST), a protein-based genome language model that models genomes as sets of proteins without considering sparsely available functional labels. Trained on >100k viruses, PST outperformed other homology- and language model-based approaches for relating viral genomes based on shared protein content. Further, PST demonstrated protein structural and functional awareness by clustering capsid-fold-containing proteins with known capsid proteins and uniquely clustering late gene proteins within related viruses. Our data establish PST as a valuable method for diverse viral genomics, ecology, and evolutionary applications. We posit that the PST framework can be a foundation model for microbial genomics when trained on suitable data.

16.
J Cheminform ; 16(1): 92, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095917

ABSTRACT

Protein language models (PLMs) play a dominant role in protein representation learning. Most existing PLMs regard proteins as sequences of 20 natural amino acids. The problem with this representation method is that it simply divides the protein sequence into sequences of individual amino acids, ignoring the fact that certain residues often occur together. Therefore, it is inappropriate to view amino acids as isolated tokens. Instead, the PLMs should recognize the frequently occurring combinations of amino acids as a single token. In this study, we use the byte-pair-encoding algorithm and unigram to construct advanced residue vocabularies for protein sequence tokenization, and we have shown that PLMs pre-trained using these advanced vocabularies exhibit superior performance on downstream tasks when compared to those trained with simple vocabularies. Furthermore, we introduce PETA, a comprehensive benchmark for systematically evaluating PLMs. We find that vocabularies comprising 50 and 200 elements achieve optimal performance. Our code, model weights, and datasets are available at https://github.com/ginnm/ProteinPretraining . SCIENTIFIC CONTRIBUTION: This study introduces advanced protein sequence tokenization analysis, leveraging the byte-pair-encoding algorithm and unigram. By recognizing frequently occurring combinations of amino acids as single tokens, our proposed method enhances the performance of PLMs on downstream tasks. Additionally, we present PETA, a new comprehensive benchmark for the systematic evaluation of PLMs, demonstrating that vocabularies of 50 and 200 elements offer optimal performance.

17.
J Cell Biochem ; : e30642, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164870

ABSTRACT

The Type III secretion effectors (T3SEs) are bacterial proteins synthesized by Gram-negative pathogens and delivered into host cells via the Type III secretion system (T3SS). These effectors usually play a pivotal role in the interactions between bacteria and hosts. Hence, the precise identification of T3SEs aids researchers in exploring the pathogenic mechanisms of bacterial infections. Since the diversity and complexity of T3SE sequences often make traditional experimental methods time-consuming, it is imperative to explore more efficient and convenient computational approaches for T3SE prediction. Inspired by the promising potential exhibited by pre-trained language models in protein recognition tasks, we proposed a method called PLM-T3SE that utilizes protein language models (PLMs) for effective recognition of T3SEs. First, we utilized PLM embeddings and evolutionary features from the position-specific scoring matrix (PSSM) profiles to transform protein sequences into fixed-length vectors for model training. Second, we employed the extreme gradient boosting (XGBoost) algorithm to rank these features based on their importance. Finally, a MLP neural network model was used to predict T3SEs based on the selected optimal feature set. Experimental results from the cross-validation and independent test demonstrated that our model exhibited superior performance compared to the existing models. Specifically, our model achieved an accuracy of 98.1%, which is 1.8%-42.4% higher than the state-of-the-art predictors based on the same independent data set test. These findings highlight the superiority of the PLM-T3SE and the remarkable characterization ability of PLM embeddings for T3SE prediction.

18.
Interdiscip Sci ; 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39155325

ABSTRACT

X-ray diffraction crystallography has been most widely used for protein three-dimensional (3D) structure determination for which whether proteins are crystallizable is a central prerequisite. Yet, there are a number of procedures during protein crystallization, including protein material production, purification, and crystal production, which take turns affecting the crystallization outcome. Due to the expensive and laborious nature of this multi-stage process, various computational tools have been developed to predict protein crystallization propensity, which is then used to guide the experimental determination. In this study, we presented a novel deep learning framework, PLMC, to improve multi-stage protein crystallization propensity prediction by leveraging a pre-trained protein language model. To effectively train PLMC, two groups of features of each protein were integrated into a more comprehensive representation, including protein language embeddings from the large-scale protein sequence database and a handcrafted feature set consisting of physicochemical, sequence-based and disordered-related information. These features were further separately embedded for refinement, and then concatenated for the final prediction. Notably, our extensive benchmarking tests demonstrate that PLMC greatly outperforms other state-of-the-art methods by achieving AUC scores of 0.773, 0.893, and 0.913, respectively, at the aforementioned individual stages, and 0.982 at the final crystallization stage. Furthermore, PLMC is shown to be superior for predicting the crystallization of both globular and membrane proteins, as demonstrated by an AUC score of 0.991 for the latter. These results suggest the significant potential of PLMC in assisting researchers with the experimental design of crystallizable protein variants.

19.
Comput Struct Biotechnol J ; 23: 3020-3029, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39171252

ABSTRACT

Colorectal cancer (CRC) is the third most diagnosed cancer and the second deadliest cancer worldwide representing a major public health problem. In recent years, increasing evidence has shown that microRNA (miRNA) can control the expression of targeted human messenger RNA (mRNA) by reducing their abundance or translation, acting as oncogenes or tumor suppressors in various cancers, including CRC. Due to the significant up-regulation of oncogenic miRNAs in CRC, elucidating the underlying mechanism and identifying dysregulated miRNA targets may provide a basis for improving current therapeutic interventions. In this paper, we proposed Gra-CRC-miRTar, a pre-trained nucleotide-to-graph neural network framework, for identifying potential miRNA targets in CRC. Different from previous studies, we constructed two pre-trained models to encode RNA sequences and transformed them into de Bruijn graphs. We employed different graph neural networks to learn the latent representations. The embeddings generated from de Bruijn graphs were then fed into a Multilayer Perceptron (MLP) to perform the prediction tasks. Our extensive experiments show that Gra-CRC-miRTar achieves better performance than other deep learning algorithms and existing predictors. In addition, our analyses also successfully revealed 172 out of 201 functional interactions through experimentally validated miRNA-mRNA pairs in CRC. Collectively, our effort provides an accurate and efficient framework to identify potential miRNA targets in CRC, which can also be used to reveal miRNA target interactions in other malignancies, facilitating the development of novel therapeutics. The Gra-CRC-miRTar web server can be found at: http://gra-crc-mirtar.com/.

20.
Anal Biochem ; 694: 115637, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39121938

ABSTRACT

Accurate identifications of protein-peptide binding residues are essential for protein-peptide interactions and advancing drug discovery. To address this problem, extensive research efforts have been made to design more discriminative feature representations. However, extracting these explicit features usually depend on third-party tools, resulting in low computational efficacy and suffering from low predictive performance. In this study, we design an end-to-end deep learning-based method, E2EPep, for protein-peptide binding residue prediction using protein sequence only. E2EPep first employs and fine-tunes two state-of-the-art pre-trained protein language models that can extract two different high-latent feature representations from protein sequences relevant for protein structures and functions. A novel feature fusion module is then designed in E2EPep to fuse and optimize the above two feature representations of binding residues. In addition, we have also design E2EPep+, which integrates E2EPep and PepBCL models, to improve the prediction performance. Experimental results on two independent testing data sets demonstrate that E2EPep and E2EPep + could achieve the average AUC values of 0.846 and 0.842 while achieving an average Matthew's correlation coefficient value that is significantly higher than that of existing most of sequence-based methods and comparable to that of the state-of-the-art structure-based predictors. Detailed data analysis shows that the primary strength of E2EPep lies in the effectiveness of feature representation using cross-attention mechanism to fuse the embeddings generated by two fine-tuned protein language models. The standalone package of E2EPep and E2EPep + can be obtained at https://github.com/ckx259/E2EPep.git for academic use only.


Subject(s)
Peptides , Protein Binding , Proteins , Proteins/chemistry , Proteins/metabolism , Peptides/chemistry , Peptides/metabolism , Deep Learning , Binding Sites , Databases, Protein , Computational Biology/methods
SELECTION OF CITATIONS
SEARCH DETAIL