Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Sci Rep ; 14(1): 21543, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39278960

RESUMEN

This work initiates a concept of reduced reverse degree based RR D M -Polynomial for a graph, and differential and integral operators by using this RR D M -Polynomial. In this study twelve reduced reverse degree-based topological descriptors are formulated using the RR D M -Polynomial. The topological descriptors, denoted as T D 's, are numerical invariants that offer significant insights into the molecular topology of a molecular graph. These descriptors are essential for conducting QSPR investigations and accurately estimating physicochemical attributes. The structural and algebraic characteristics of the graphene and graphdiyne are studied to apply this methodology. The study involves the analysis and estimation of Reduced reverse degree-based topological descriptors and physicochemical features of graphene derivatives using best-fit quadratic regression models. This work opens up new directions for scientists and researchers to pursue, taking them into new fields of study.

2.
J Cheminform ; 16(1): 102, 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39160576

RESUMEN

Molecular fragmentation is an effective suite of approaches to reduce the formal computational complexity of quantum chemistry calculations while enhancing their algorithmic parallelisability. However, the practical applicability of fragmentation techniques remains hindered by a dearth of automation and effective metrics to assess the quality of a fragmentation scheme. In this article, we present the Quick Fragmentation via Automated Genetic Search (QFRAGS), a novel automated fragmentation algorithm that uses a genetic optimisation procedure to generate molecular fragments that yield low energy errors when adopted in Many Body Expansions (MBEs). Benchmark testing of QFRAGS on protein systems with less than 500 atoms, using two-body (MBE2) and three-body (MBE3) MBE calculations at the HF/6-31G* level, reveals mean absolute energy errors (MAEE) of 20.6 and 2.2 kJ  mol - 1 , respectively. For larger protein systems exceeding 500 atoms, MAEEs are 181.5 kJ  mol - 1 for MBE2 and 24.3 kJ  mol - 1 for MBE3. Furthermore, when compared to three manual fragmentation schemes on a 40-protein dataset, using both MBE and Fragment Molecular Orbital techniques, QFRAGS achieves comparable or often lower MAEEs. When applied to a 10-lipoglycan/glycolipid dataset, MAEs of 7.9 and 0.3 kJ  mol - 1 were observed at the MBE2 and MBE3 levels, respectively.Scientific Contribution This Article presents the Quick Fragmentation via Automated Genetic Search (QFRAGS), an innovative molecular fragmentation algorithm that significantly improves upon existing molecular fragmentation approaches by specifically addressing their lack of automation and effective fragmentation quality metrics. With an evolutionary optimisation strategy, QFRAGS actively pursues high quality fragments, generating fragmentation schemes that exhibit minimal energy errors on systems with hundreds to thousands of atoms. The advent of QFRAGS represents a significant advancement in molecular fragmentation, greatly improving the accessibility and computational feasibility of accurate quantum chemistry calculations.

3.
Heliyon ; 10(15): e34944, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39170540

RESUMEN

A carbon-based material with a broad scope of favourable developments is called graphene. Recently, a graphene nanoribbon with cove-edged was integrated by utilizing a bottom-up liquid-phase procedure, and it can be geometrically viewed as a hybrid of the armchair and the zigzag edges. It is indeed a type of nanoribbon containing asymmetric edges made up of sequential hexagons with impressive mechanical and electrical characteristics. Topological indices are numerical values associated with the structure of a chemical graph and are used to predict various physical, chemical, and biological properties of molecules. They are derived from the graph representation of molecules, where atoms are represented as vertices and bonds as edges. In this article, we derived the exact topological expressions of cove-edged graphene nanoribbons based on the graph-theoretical structural measures that help reduce the number of repetitive laboratory tasks necessary for studying the physicochemical characteristics of graphene nanoribbons with curved edges.

4.
J Comput Biol ; 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39082155

RESUMEN

The development of new drugs is a vital effort that has the potential to improve human health, well-being and life expectancy. Molecular property prediction is a crucial step in drug discovery, as it helps to identify potential therapeutic compounds. However, experimental methods for drug development can often be time-consuming and resource-intensive, with a low probability of success. To address such limitations, deep learning (DL) methods have emerged as a viable alternative due to their ability to identify high-discriminating patterns in molecular data. In particular, graph neural networks (GNNs) operate on graph-structured data to identify promising drug candidates with desirable molecular properties. These methods represent molecules as a set of node (atoms) and edge (chemical bonds) features to aggregate local information for molecular graph representation learning. Despite the availability of several GNN frameworks, each approach has its own shortcomings. Although, some GNNs may excel in certain tasks, they may not perform as well in others. In this work, we propose a hybrid approach that incorporates different graph-based methods to combine their strengths and mitigate their limitations to accurately predict molecular properties. The proposed approach consists in a multi-layered hybrid GNN architecture that integrates multiple GNN frameworks to compute graph embeddings for molecular property prediction. Furthermore, we conduct extensive experiments on multiple benchmark datasets to demonstrate that our hybrid approach significantly outperforms the state-of-the-art graph-based models. The data and code scripts to reproduce the results are available in the repository, https://github.com/pedro-quesado/HybridGNN.

5.
Front Pharmacol ; 15: 1369403, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38831885

RESUMEN

Accurately predicting Drug-Drug Interaction (DDI) is a critical and challenging aspect of the drug discovery process, particularly in preventing adverse reactions in patients undergoing combination therapy. However, current DDI prediction methods often overlook the interaction information between chemical substructures of drugs, focusing solely on the interaction information between drugs and failing to capture sufficient chemical substructure details. To address this limitation, we introduce a novel DDI prediction method: Multi-layer Adaptive Soft Mask Graph Neural Network (MASMDDI). Specifically, we first design a multi-layer adaptive soft mask graph neural network to extract substructures from molecular graphs. Second, we employ an attention mechanism to mine substructure feature information and update latent features. In this process, to optimize the final feature representation, we decompose drug-drug interactions into pairwise interaction correlations between the core substructures of each drug. Third, we use these features to predict the interaction probabilities of DDI tuples and evaluate the model using real-world datasets. Experimental results demonstrate that the proposed model outperforms state-of-the-art methods in DDI prediction. Furthermore, MASMDDI exhibits excellent performance in predicting DDIs of unknown drugs in two tasks that are more aligned with real-world scenarios. In particular, in the transductive scenario using the DrugBank dataset, the ACC and AUROC and AUPRC scores of MASMDDI are 0.9596, 0.9903, and 0.9894, which are 2% higher than the best performing baseline.

6.
J Adv Res ; 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38844122

RESUMEN

INTRODUCTION: With the escalating menace of organic compounds in environmental pollution imperiling the survival of aquatic organisms, the investigation of organic compound toxicity across diverse aquatic species assumes paramount significance for environmental protection. Understanding how different species respond to these compounds helps assess the potential ecological impact of pollution on aquatic ecosystems as a whole. Compared with traditional experimental methods, deep learning methods have higher accuracy in predicting aquatic toxicity, faster data processing speed and better generalization ability. OBJECTIVES: This article presents ATFPGT-multi, an advanced multi-task deep neural network prediction model for organic toxicity. METHODS: The model integrates molecular fingerprints and molecule graphs to characterize molecules, enabling the simultaneous prediction of acute toxicity for the same organic compound across four distinct fish species. Furthermore, to validate the advantages of multi-task learning, we independently construct prediction models, named ATFPGT-single, for each fish species. We employ cross-validation in our experiments to assess the performance and generalization ability of ATFPGT-multi. RESULTS: The experimental results indicate, first, that ATFPGT-multi outperforms ATFPGT-single on four fish datasets with AUC improvements of 9.8%, 4%, 4.8%, and 8.2%, respectively, demonstrating the superiority of multi-task learning over single-task learning. Furthermore, in comparison with previous algorithms, ATFPGT-multi outperforms comparative methods, emphasizing that our approach exhibits higher accuracy and reliability in predicting aquatic toxicity. Moreover, ATFPGT-multi utilizes attention scores to identify molecular fragments associated with fish toxicity in organic molecules, as demonstrated by two organic molecule examples in the main text, demonstrating the interpretability of ATFPGT-multi. CONCLUSION: In summary, ATFPGT-multi provides important support and reference for the further development of aquatic toxicity assessment. All of codes and datasets are freely available online at https://github.com/zhaoqi106/ATFPGT-multi.

7.
Brief Bioinform ; 25(3)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38706318

RESUMEN

Molecular property prediction faces the challenge of limited labeled data as it necessitates a series of specialized experiments to annotate target molecules. Data augmentation techniques can effectively address the issue of data scarcity. In recent years, Mixup has achieved significant success in traditional domains such as image processing. However, its application in molecular property prediction is relatively limited due to the irregular, non-Euclidean nature of graphs and the fact that minor variations in molecular structures can lead to alterations in their properties. To address these challenges, we propose a novel data augmentation method called Mix-Key tailored for molecular property prediction. Mix-Key aims to capture crucial features of molecular graphs, focusing separately on the molecular scaffolds and functional groups. By generating isomers that are relatively invariant to the scaffolds or functional groups, we effectively preserve the core information of molecules. Additionally, to capture interactive information between the scaffolds and functional groups while ensuring correlation between the original and augmented graphs, we introduce molecular fingerprint similarity and node similarity. Through these steps, Mix-Key determines the mixup ratio between the original graph and two isomers, thus generating more informative augmented molecular graphs. We extensively validate our approach on molecular datasets of different scales with several Graph Neural Network architectures. The results demonstrate that Mix-Key consistently outperforms other data augmentation methods in enhancing molecular property prediction on several datasets.


Asunto(s)
Algoritmos , Estructura Molecular , Biología Computacional/métodos , Programas Informáticos
8.
Mol Ther Nucleic Acids ; 35(2): 102186, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38706632

RESUMEN

Recent studies have highlighted the effectiveness of using antisense oligonucleotides (ASOs) for cellular RNA regulation, including targets that are considered undruggable; however, manually designing optimal ASO sequences can be labor intensive and time consuming, which potentially limits their broader application. To address this challenge, we introduce a platform, the ASOptimizer, a deep-learning-based framework that efficiently designs ASOs at a low cost. This platform not only selects the most efficient mRNA target sites but also optimizes the chemical modifications for enhanced performance. Indoleamine 2,3-dioxygenase 1 (IDO1) promotes cancer survival by depleting tryptophan and producing kynurenine, leading to immunosuppression through the aryl-hydrocarbon receptor (Ahr) pathway within the tumor microenvironment. We used ASOptimizer to identify ASOs that target IDO1 mRNA as potential cancer therapeutics. Our methodology consists of two stages: sequence engineering and chemical engineering. During the sequence-engineering stage, we optimized and predicted ASO sequences that could target IDO1 mRNA efficiently. In the chemical-engineering stage, we further refined these ASOs to enhance their inhibitory activity while reducing their potential cytotoxicity. In conclusion, our research demonstrates the potential of ASOptimizer for identifying ASOs with improved efficacy and safety.

9.
Interdiscip Sci ; 16(3): 712-726, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38683279

RESUMEN

The structures of fentanyl and its analogues are easy to be modified and few types have been included in database so far, which allow criminals to avoid the supervision of relevant departments. This paper introduces a molecular graph-based transformer model, which is combined with a data augmentation method based on substructure replacement to generate novel fentanyl analogues. 140,000 molecules were generated, and after a set of screening, 36,799 potential fentanyl analogues were finally obtained. We calculated the molecular properties of 36,799 potential fentanyl analogues. The results showed that the model could learn some properties of original fentanyl molecules. We compared the generated molecules from transformer model and data augmentation method based on substructure replacement with those generated by the other two molecular generation models based on deep learning, and found that the model in this paper can generate more novel potential fentanyl analogues. Finally, the findings of the paper indicate that transformer model based on molecular graph helps us explore the structure of potential fentanyl molecules as well as understand distribution of original molecules of fentanyl.


Asunto(s)
Fentanilo , Fentanilo/análogos & derivados , Fentanilo/química , Modelos Moleculares , Aprendizaje Profundo
10.
Interdiscip Sci ; 16(2): 361-377, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38457109

RESUMEN

Accurately predicting compound-protein interactions (CPI) is a critical task in computer-aided drug design. In recent years, the exponential growth of compound activity and biomedical data has highlighted the need for efficient and interpretable prediction approaches. In this study, we propose GraphsformerCPI, an end-to-end deep learning framework that improves prediction performance and interpretability. GraphsformerCPI treats compounds and proteins as sequences of nodes with spatial structures, and leverages novel structure-enhanced self-attention mechanisms to integrate semantic and graph structural features within molecules for deep molecule representations. To capture the vital association between compound atoms and protein residues, we devise a dual-attention mechanism to effectively extract relational features through .cross-mapping. By extending the powerful learning capabilities of Transformers to spatial structures and extensively utilizing attention mechanisms, our model offers strong interpretability, a significant advantage over most black-box deep learning methods. To evaluate GraphsformerCPI, extensive experiments were conducted on benchmark datasets including human, C. elegans, Davis and KIBA datasets. We explored the impact of model depth and dropout rate on performance and compared our model against state-of-the-art baseline models. Our results demonstrate that GraphsformerCPI outperforms baseline models in classification datasets and achieves competitive performance in regression datasets. Specifically, on the human dataset, GraphsformerCPI achieves an average improvement of 1.6% in AUC, 0.5% in precision, and 5.3% in recall. On the KIBA dataset, the average improvement in Concordance index (CI) and mean squared error (MSE) is 3.3% and 7.2%, respectively. Molecular docking shows that our model provides novel insights into the intrinsic interactions and binding mechanisms. Our research holds practical significance in effectively predicting CPIs and binding affinities, identifying key atoms and residues, enhancing model interpretability.


Asunto(s)
Aprendizaje Profundo , Proteínas , Humanos , Proteínas/química , Proteínas/metabolismo , Animales , Algoritmos , Caenorhabditis elegans/metabolismo , Diseño de Fármacos , Unión Proteica
11.
Molecules ; 29(2)2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38276570

RESUMEN

Existing formats based on the simplified molecular input line entry system (SMILES) encoding and molecular graph structure are designed to encode the complete semantic and structural information of molecules. However, the physicochemical properties of molecules are complex, and a single encoding of molecular features from SMILES sequences or molecular graph structures cannot adequately represent molecular information. Aiming to address this problem, this study proposes a sequence graph cross-attention (SG-ATT) representation architecture for a molecular property prediction model to efficiently use domain knowledge to enhance molecular graph feature encoding and combine the features of molecular SMILES sequences. The SG-ATT fuses the two-dimensional molecular features so that the current model input molecular information contains molecular structure information and semantic information. The SG-ATT was tested on nine molecular property prediction tasks. Among them, the biggest SG-ATT model performance improvement was 4.5% on the BACE dataset, and the average model performance improvement was 1.83% on the full dataset. Additionally, specific model interpretability studies were conducted to showcase the performance of the SG-ATT model on different datasets. In-depth analysis was provided through case studies of in vitro validation. Finally, network tools for molecular property prediction were developed for the use of researchers.

12.
BMC Bioinformatics ; 25(1): 39, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38262923

RESUMEN

BACKGROUND: Drug-drug interactions (DDI) are prevalent in combination therapy, necessitating the importance of identifying and predicting potential DDI. While various artificial intelligence methods can predict and identify potential DDI, they often overlook the sequence information of drug molecules and fail to comprehensively consider the contribution of molecular substructures to DDI. RESULTS: In this paper, we proposed a novel model for DDI prediction based on sequence and substructure features (SSF-DDI) to address these issues. Our model integrates drug sequence features and structural features from the drug molecule graph, providing enhanced information for DDI prediction and enabling a more comprehensive and accurate representation of drug molecules. CONCLUSION: The results of experiments and case studies have demonstrated that SSF-DDI significantly outperforms state-of-the-art DDI prediction models across multiple real datasets and settings. SSF-DDI performs better in predicting DDI involving unknown drugs, resulting in a 5.67% improvement in accuracy compared to state-of-the-art methods.


Asunto(s)
Inteligencia Artificial , Aprendizaje Profundo , Interacciones Farmacológicas
13.
Comb Chem High Throughput Screen ; 27(11): 1629-1641, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38213147

RESUMEN

BACKGROUND: Chemical graph theory has been used to mathematically model the various physical and biological aspects of chemical substances. A mathematical formulation that may be applied to any graph and can characterise a molecule structure is known as a topological index or molecular descriptor. METHOD: It is convenient and efficient to analyse the mathematical values and further research on various physical properties of a molecule based on these molecular descriptors. They provide useful alternatives to lengthy, expensive, and labour-intensive laboratory experiments. The topological indices can be used to predict the chemical structures, physicochemical properties, and biological activities using quantitative structure-activity relationships (QSARs) and quantitative structure-property relationships (QSPRs). RESULT: In this study, the molecular descriptors of the Dodeca-benzo-circumcorenene compounds are derived based on their corresponding molecular structures. CONCLUSION: The computed indices are then compared graphically to study their relationship with the molecular structure and with each other..


Asunto(s)
Relación Estructura-Actividad Cuantitativa , Estructura Molecular
14.
Methods ; 221: 18-26, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38040204

RESUMEN

Drug-induced liver injury (DILI) is a significant issue in drug development and clinical treatment due to its potential to cause liver dysfunction or damage, which, in severe cases, can lead to liver failure or even fatality. DILI has numerous pathogenic factors, many of which remain incompletely understood. Consequently, it is imperative to devise methodologies and tools for anticipatory assessment of DILI risk in the initial phases of drug development. In this study, we present DMFPGA, a novel deep learning predictive model designed to predict DILI. To provide a comprehensive description of molecular properties, we employ a multi-head graph attention mechanism to extract features from the molecular graphs, representing characteristics at the level of compound nodes. Additionally, we combine multiple fingerprints of molecules to capture features at the molecular level of compounds. The fusion of molecular fingerprints and graph features can more fully express the properties of compounds. Subsequently, we employ a fully connected neural network to classify compounds as either DILI-positive or DILI-negative. To rigorously evaluate DMFPGA's performance, we conduct a 5-fold cross-validation experiment. The obtained results demonstrate the superiority of our method over four existing state-of-the-art computational approaches, exhibiting an average AUC of 0.935 and an average ACC of 0.934. We believe that DMFPGA is helpful for early-stage DILI prediction and assessment in drug development.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Modelos Químicos , Humanos , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Desarrollo de Medicamentos , Aprendizaje Profundo
15.
Comput Biol Med ; 167: 107691, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37976819

RESUMEN

With the wide application of deep learning in Drug Discovery, deep generative model has shown its advantages in drug molecular generation. Generative adversarial networks can be used to learn the internal structure of molecules, but the training process may be unstable, such as gradient disappearance and model collapse, which may lead to the generation of molecules that do not conform to chemical rules or a single style. In this paper, a novel method called STAGAN was proposed to solve the difficulty of model training, by adding a new gradient penalty term in the discriminator and designing a parallel layer of batch normalization used in generator. As an illustration of method, STAGAN generated higher valid and unique molecules than previous models in training datasets from QM9 and ZINC-250K. This indicates that the proposed method can effectively solve the instability problem in the model training process, and can provide more instructive guidance for the further study of molecular graph generation.


Asunto(s)
Aprendizaje Profundo , Descubrimiento de Drogas , Modelos Químicos
16.
Environ Sci Technol ; 57(44): 16906-16917, 2023 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-37897806

RESUMEN

In silico models for predicting physicochemical properties and environmental fate parameters are necessary for the sound management of chemicals. This study employed graph attention network (GAT) algorithms to construct such models on 15 end points. The results showed that the GAT models outperformed the previous state-of-the-art models, and their performance was not influenced by the presence or absence of compounds with certain structures. Molecular similarity density (ρs) was found to be a key metrics characterizing data set modelability, in addition to the proportion of compounds at activity cliffs. By introducing molecular graph (MG) contrastive learning, MG-based ρs and molecular inconsistency in activities (IA) were calculated and employed for characterizing the structure-activity landscape (SAL)-based applicability domain ADSAL{ρs, IA}. The GAT models coupled with ADSAL{ρs, IA} significantly improved the prediction coefficient of determination (R2) on all the end points by an average of 14.4% and enabled all the end points to have R2 > 0.9, which could hardly be achieved previously. The models were employed to screen persistent, mobile, and/or bioaccumulative chemicals from inventories consisting of about 106 chemicals. Given the current state-of-the-art model performance and coverage of the various environmental end points, the constructed models with ADSAL{ρs, IA} may serve as benchmarks for future efforts to improve modeling efficacy.


Asunto(s)
Algoritmos , Benchmarking , Simulación por Computador
17.
J Cheminform ; 15(1): 81, 2023 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-37726841

RESUMEN

Graph neural networks have recently become a standard method for analyzing chemical compounds. In the field of molecular property prediction, the emphasis is now on designing new model architectures, and the importance of atom featurization is oftentimes belittled. When contrasting two graph neural networks, the use of different representations possibly leads to incorrect attribution of the results solely to the network architecture. To better understand this issue, we compare multiple atom representations by evaluating them on the prediction of free energy, solubility, and metabolic stability using graph convolutional networks. We discover that the choice of atom representation has a significant impact on model performance and that the optimal subset of features is task-specific. Additional experiments involving more sophisticated architectures, including graph transformers, support these findings. Moreover, we demonstrate that some commonly used atom features, such as the number of neighbors or the number of hydrogens, can be easily predicted using only information about bonds and atom type, yet their explicit inclusion in the representation has a positive impact on model performance. Finally, we explain the predictions of the best-performing models to better understand how they utilize the available atomic features.

18.
Molecules ; 28(15)2023 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-37570812

RESUMEN

Breast cancer ranks as the second leading cause of death among women, but early screening and self-awareness can help prevent it. Hormone therapy drugs that target estrogen levels offer potential treatments. However, conventional drug discovery entails extensive, costly processes. This study presents a framework for analyzing the quantitative structure-activity relationship (QSAR) of estrogen receptor alpha inhibitors. Our approach utilizes supervised learning, integrating self-attention Transformer and molecular graph information, to predict estrogen receptor alpha inhibitors. We established five classification models for predicting these inhibitors in breast cancer. Among these models, our proposed MATH model achieved remarkable precision, recall, F1 score, and specificity, with values of 0.952, 0.972, 0.960, and 0.922, respectively, alongside an ROC AUC of 0.977. MATH exhibited robust performance, suggesting its potential to assist pharmaceutical and health researchers in identifying candidate compounds for estrogen alpha inhibitors and guiding drug discovery pathways.


Asunto(s)
Neoplasias de la Mama , Aprendizaje Profundo , Femenino , Humanos , Receptor alfa de Estrógeno/metabolismo , Relación Estructura-Actividad Cuantitativa , Antagonistas de Estrógenos/farmacología , Estrógenos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico
19.
Brief Bioinform ; 24(5)2023 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-37507113

RESUMEN

Drug-drug interaction (DDI) identification is essential to clinical medicine and drug discovery. The two categories of drugs (i.e. chemical drugs and biotech drugs) differ remarkably in molecular properties, action mechanisms, etc. Biotech drugs are up-to-comers but highly promising in modern medicine due to higher specificity and fewer side effects. However, existing DDI prediction methods only consider chemical drugs of small molecules, not biotech drugs of large molecules. Here, we build a large-scale dual-modal graph database named CB-DB and customize a graph-based framework named CB-TIP to reason event-aware DDIs for both chemical and biotech drugs. CB-DB comprehensively integrates various interaction events and two heterogeneous kinds of molecular structures. It imports endogenous proteins founded on the fact that most drugs take effects by interacting with endogenous proteins. In the modality of molecular structure, drugs and endogenous proteins are two heterogeneous kinds of graphs, while in the modality of interaction, they are nodes connected by events (i.e. edges of different relationships). CB-TIP employs graph representation learning methods to generate drug representations from either modality and then contrastively mixes them to predict how likely an event occurs when a drug meets another in an end-to-end manner. Experiments demonstrate CB-TIP's great superiority in DDI prediction and the promising potential of uncovering novel DDIs.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Interacciones Farmacológicas , Descubrimiento de Drogas , Estructura Molecular , Proteínas
20.
Comput Biol Med ; 163: 107202, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37450964

RESUMEN

RNA N6-methyladenine (m6A), which is produced by the methylation of the N6 position of eukaryotic adenine, is a relatively common post-transcriptional modification on the surface of the molecule, which frequently plays a crucial role in biological processes. Biological experimental methods to identify m6A have been studied and implemented in recent years, but they cannot be promoted widely due to drawbacks such as the time and cost of reagents and equipment. Therefore, researchers have proposed computational strategies for identifying m6A sites, but these strategies do not account for the mechanism of methylation occurrence or the structure of RNA molecules. This study, therefore, proposed a novel deep learning model for predicting m6A sites, GR-m6A, which predicts m6A sites by extracting features from the physicochemical properties and spatial structure of molecules via residual networks. In GR-m6A, each RNA base string is represented by SMILES as two matrices comprising topology structural information and node attributes with molecular physicochemical characteristics. The feature encoding matrix was then obtained by fusing the topology matrix and the node matrix in accordance with the graphical convolutional network principle. Correspondingly, the more discriminative features were extracted from the encoding matrix using the residual neural network and predicted using a multilayer perceptron. As evident from the 5-fold cross-validation and independent validation, the GR-m6A model outperformed other existing methods. Thus, we hope that GR-m6A can aid researchers in predicting mammalian m6A loci. The source code and database are available at https://github.com/YingLiangjxau/GR-m6A.


Asunto(s)
Redes Neurales de la Computación , ARN , Animales , ARN/genética , ARN/metabolismo , Metilación , Mamíferos/genética , Mamíferos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA