Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Brief Bioinform ; 24(2)2023 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-36790856

RESUMEN

Potential miRNA-disease associations (MDA) play an important role in the discovery of complex human disease etiology. Therefore, MDA prediction is an attractive research topic in the field of biomedical machine learning. Recently, several models have been proposed for this task, but their performance limited by over-reliance on relevant network information with noisy graph structure connections. However, the application of self-supervised graph structure learning to MDA tasks remains unexplored. Our study is the first to use multi-view self-supervised contrastive learning (MSGCL) for MDA prediction. Specifically, we generated a learner view without association labels of miRNAs and diseases as input, and utilized the known association network to generate an anchor view that provides guiding signals for the learner view. The graph structure was optimized by designing a contrastive loss to maximize the consistency between the anchor and learner views. Our model is similar to a pre-trained model that continuously optimizes upstream tasks for high-quality association graph topology, thereby enhancing the latent representation of association predictions. The experimental results show that our proposed method outperforms state-of-the-art methods by 2.79$\%$ and 3.20$\%$ in area under the receiver operating characteristic curve (AUC) and area under the precision/recall curve (AUPR), respectively.


Asunto(s)
Aprendizaje Automático , MicroARNs , Humanos , Área Bajo la Curva , MicroARNs/genética , Curva ROC
2.
Brief Bioinform ; 24(6)2023 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-37974508

RESUMEN

Current methods of molecular image-based drug discovery face two major challenges: (1) work effectively in absence of labels, and (2) capture chemical structure from implicitly encoded images. Given that chemical structures are explicitly encoded by molecular graphs (such as nitrogen, benzene rings and double bonds), we leverage self-supervised contrastive learning to transfer chemical knowledge from graphs to images. Specifically, we propose a novel Contrastive Graph-Image Pre-training (CGIP) framework for molecular representation learning, which learns explicit information in graphs and implicit information in images from large-scale unlabeled molecules via carefully designed intra- and inter-modal contrastive learning. We evaluate the performance of CGIP on multiple experimental settings (molecular property prediction, cross-modal retrieval and distribution similarity), and the results show that CGIP can achieve state-of-the-art performance on all 12 benchmark datasets and demonstrate that CGIP transfers chemical knowledge in graphs to molecular images, enabling image encoder to perceive chemical structures in images. We hope this simple and effective framework will inspire people to think about the value of image for molecular representation learning.


Asunto(s)
Benchmarking , Aprendizaje , Humanos , Descubrimiento de Drogas
3.
Bioinformatics ; 40(4)2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38530779

RESUMEN

MOTIVATION: Molecular representation learning plays an indispensable role in crucial tasks such as property prediction and drug design. Despite the notable achievements of molecular pre-training models, current methods often fail to capture both the structural and feature semantics of molecular graphs. Moreover, while graph contrastive learning has unveiled new prospects, existing augmentation techniques often struggle to retain their core semantics. To overcome these limitations, we propose a gradient-compensated encoder parameter perturbation approach, ensuring efficient and stable feature augmentation. By merging enhancement strategies grounded in attribute masking and parameter perturbation, we introduce MoleMCL, a new MOLEcular pre-training model based on multi-level contrastive learning. RESULTS: Experimental results demonstrate that MoleMCL adeptly dissects the structure and feature semantics of molecular graphs, surpassing current state-of-the-art models in molecular prediction tasks, paving a novel avenue for molecular modeling. AVAILABILITY AND IMPLEMENTATION: The code and data underlying this work are available in GitHub at https://github.com/BioSequenceAnalysis/MoleMCL.


Asunto(s)
Diseño de Fármacos , Semántica
4.
Bioinformatics ; 40(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38905501

RESUMEN

MOTIVATION: In the field of drug discovery, accurately and effectively predicting the binding affinity between proteins and ligands is crucial for drug screening and optimization. However, current research primarily utilizes representations based on sequence or structure to predict protein-ligand binding affinity, with relatively less study on protein surface information, which is crucial for protein-ligand interactions. Moreover, when dealing with multimodal information of proteins, traditional approaches typically concatenate features from different modalities in a straightforward manner without considering the heterogeneity among them, which results in an inability to effectively exploit the complementary between modalities. RESULTS: We introduce a novel multimodal feature extraction (MFE) framework that, for the first time, incorporates information from protein surfaces, 3D structures, and sequences, and uses cross-attention mechanism for feature alignment between different modalities. Experimental results show that our method achieves state-of-the-art performance in predicting protein-ligand binding affinity. Furthermore, we conduct ablation studies that demonstrate the effectiveness and necessity of protein surface information and multimodal feature alignment within the framework. AVAILABILITY AND IMPLEMENTATION: The source code and data are available at https://github.com/Sultans0fSwing/MFE.


Asunto(s)
Unión Proteica , Proteínas , Ligandos , Proteínas/metabolismo , Proteínas/química , Biología Computacional/métodos , Descubrimiento de Drogas/métodos , Algoritmos , Sitios de Unión , Bases de Datos de Proteínas , Conformación Proteica
5.
BMC Bioinformatics ; 25(1): 216, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890584

RESUMEN

BACKGROUND: Recognition of enhancer-promoter Interactions (EPIs) is crucial for human development. EPIs in the genome play a key role in regulating transcription. However, experimental approaches for classifying EPIs are too expensive in terms of effort, time, and resources. Therefore, more and more studies are being done on developing computational techniques, particularly using deep learning and other machine learning techniques, to address such problems. Unfortunately, the majority of current computational methods are based on convolutional neural networks, recurrent neural networks, or a combination of them, which don't take into consideration contextual details and the long-range interactions between the enhancer and promoter sequences. A new transformer-based model called EPI-Trans is presented in this study to overcome the aforementioned limitations. The multi-head attention mechanism in the transformer model automatically learns features that represent the long interrelationships between enhancer and promoter sequences. Furthermore, a generic model is created with transferability that can be utilized as a pre-trained model for various cell lines. Moreover, the parameters of the generic model are fine-tuned using a particular cell line dataset to improve performance. RESULTS: Based on the results obtained from six benchmark cell lines, the average AUROC for the specific, generic, and best models is 94.2%, 95%, and 95.7%, while the average AUPR is 80.5%, 66.1%, and 79.6% respectively. CONCLUSIONS: This study proposed a transformer-based deep learning model for EPI prediction. The comparative results on certain cell lines show that EPI-Trans outperforms other cutting-edge techniques and can provide superior performance on the challenge of recognizing EPI.


Asunto(s)
Aprendizaje Profundo , Elementos de Facilitación Genéticos , Regiones Promotoras Genéticas , Humanos , Biología Computacional/métodos , Línea Celular , Redes Neurales de la Computación
6.
Brief Bioinform ; 23(3)2022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35348595

RESUMEN

Identifying new lead molecules to treat cancer requires more than a decade of dedicated effort. Before selected drug candidates are used in the clinic, their anti-cancer activity is generally validated by in vitro cellular experiments. Therefore, accurate prediction of cancer drug response is a critical and challenging task for anti-cancer drugs design and precision medicine. With the development of pharmacogenomics, the combination of efficient drug feature extraction methods and omics data has made it possible to use computational models to assist in drug response prediction. In this study, we propose DeepTTA, a novel end-to-end deep learning model that utilizes transformer for drug representation learning and a multilayer neural network for transcriptomic data prediction of the anti-cancer drug responses. Specifically, DeepTTA uses transcriptomic gene expression data and chemical substructures of drugs for drug response prediction. Compared to existing methods, DeepTTA achieved higher performance in terms of root mean square error, Pearson correlation coefficient and Spearman's rank correlation coefficient on multiple test sets. Moreover, we discovered that anti-cancer drugs bortezomib and dactinomycin provide a potential therapeutic option with multiple clinical indications. With its excellent performance, DeepTTA is expected to be an effective method in cancer drug design.


Asunto(s)
Antineoplásicos , Neoplasias , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Redes Neurales de la Computación , Medicina de Precisión/métodos , Transcriptoma
7.
Brief Bioinform ; 23(1)2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-34850810

RESUMEN

The interaction between microribonucleic acid and long non-coding ribonucleic acid plays a very important role in biological processes, and the prediction of the one is of great significance to the study of its mechanism of action. Due to the limitations of traditional biological experiment methods, more and more computational methods are applied to this field. However, the existing methods often have problems, such as inadequate acquisition of potential features of the sequence due to simple coding and the need to manually extract features as input. We propose a deep learning model, preMLI, based on rna2vec pre-training and deep feature mining mechanism. We use rna2vec to train the ribonucleic acid (RNA) dataset and to obtain the RNA word vector representation and then mine the RNA sequence features separately and finally concatenate the two feature vectors as the input of the prediction task. The preMLI performs better than existing methods on benchmark datasets and has cross-species prediction capabilities. Experiments show that both pre-training and deep feature mining mechanisms have a positive impact on the prediction performance of the model. To be more specific, pre-training can provide more accurate word vector representations. The deep feature mining mechanism also improves the prediction performance of the model. Meanwhile, The preMLI only needs RNA sequence as the input of the model and has better cross-species prediction performance than the most advanced prediction models, which have reference value for related research.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Biología Computacional/métodos , MicroARNs/genética , ARN Largo no Codificante/genética
8.
Brief Bioinform ; 23(2)2022 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-35043158

RESUMEN

Drug-target interactions (DTIs) prediction research presents important significance for promoting the development of modern medicine and pharmacology. Traditional biochemical experiments for DTIs prediction confront the challenges including long time period, high cost and high failure rate, and finally leading to a low-drug productivity. Chemogenomic-based computational methods can realize high-throughput prediction. In this study, we develop a deep collaborative filtering prediction model with multiembeddings, named DCFME (deep collaborative filtering prediction model with multiembeddings), which can jointly utilize multiple feature information from multiembeddings. Two different representation learning algorithms are first employed to extract heterogeneous network features. DCFME uses the generated low-dimensional dense vectors as input, and then simulates the drug-target relationship from the perspective of both couplings and heterogeneity. In addition, the model employs focal loss that concentrates the loss on sparse and hard samples in the training process. Comparative experiments with five baseline methods show that DCFME achieves more significant performance improvement on sparse datasets. Moreover, the model has better robustness and generalization capacity under several harder prediction scenarios.


Asunto(s)
Algoritmos , Desarrollo de Medicamentos , Desarrollo de Medicamentos/métodos
9.
Brief Bioinform ; 23(5)2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-35953081

RESUMEN

Posttranslational modification of lysine residues, K-PTM, is one of the most popular PTMs. Some lysine residues in proteins can be continuously or cascaded covalently modified, such as acetylation, crotonylation, methylation and succinylation modification. The covalent modification of lysine residues may have some special functions in basic research and drug development. Although many computational methods have been developed to predict lysine PTMs, up to now, the K-PTM prediction methods have been modeled and learned a single class of K-PTM modification. In view of this, this study aims to fill this gap by building a multi-label computational model that can be directly used to predict multiple K-PTMs in proteins. In this study, a multi-label prediction model, MLysPRED, is proposed to identify multiple lysine sites using features generated from human protein sequences. In MLysPRED, three kinds of multi-label sequence encoding algorithms (MLDBPB, MLPSDAAP, MLPSTAAP) are proposed and combined with three encoding strategies (CHHAA, DR and Kmer) to convert preprocessed lysine sequences into effective numerical features. A multidimensional normal distribution oversampling technique and graph-based multi-view clustering under-sampling algorithm were first proposed and incorporated to reduce the proportion of the original training samples, and multi-label nearest neighbor algorithm is used for classification. It is observed that MLysPRED achieved an Aiming of 92.21%, Coverage of 94.98%, Accuracy of 89.63%, Absolute-True of 81.46% and Absolute-False of 0.0682 on the independent datasets. Additionally, comparison of results with five existing predictors also indicated that MLysPRED is very promising and encouraging to predict multiple K-PTMs in proteins. For the convenience of the experimental scientists, 'MLysPRED' has been deployed as a user-friendly web-server at http://47.100.136.41:8181.


Asunto(s)
Lisina , Proteínas , Algoritmos , Análisis por Conglomerados , Biología Computacional/métodos , Humanos , Lisina/metabolismo , Distribución Normal , Procesamiento Proteico-Postraduccional , Proteínas/química
10.
PLoS Comput Biol ; 19(11): e1011597, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37956212

RESUMEN

The powerful combination of large-scale drug-related interaction networks and deep learning provides new opportunities for accelerating the process of drug discovery. However, chemical structures that play an important role in drug properties and high-order relations that involve a greater number of nodes are not tackled in current biomedical networks. In this study, we present a general hypergraph learning framework, which introduces Drug-Substructures relationship into Molecular interaction Networks to construct the micro-to-macro drug centric heterogeneous network (DSMN), and develop a multi-branches HyperGraph learning model, called HGDrug, for Drug multi-task predictions. HGDrug achieves highly accurate and robust predictions on 4 benchmark tasks (drug-drug, drug-target, drug-disease, and drug-side-effect interactions), outperforming 8 state-of-the-art task specific models and 6 general-purpose conventional models. Experiments analysis verifies the effectiveness and rationality of the HGDrug model architecture as well as the multi-branches setup, and demonstrates that HGDrug is able to capture the relations between drugs associated with the same functional groups. In addition, our proposed drug-substructure interaction networks can help improve the performance of existing network models for drug-related prediction tasks.


Asunto(s)
Algoritmos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Benchmarking , Sistemas de Liberación de Medicamentos , Descubrimiento de Drogas
11.
Brief Bioinform ; 22(4)2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-33096548

RESUMEN

Enhancer-promoter interactions (EPIs) play an important role in transcriptional regulation. Recently, machine learning-based methods have been widely used in the genome-scale identification of EPIs due to their promising predictive performance. In this paper, we propose a novel method, termed EPI-DLMH, for predicting EPIs with the use of DNA sequences only. EPI-DLMH consists of three major steps. First, a two-layer convolutional neural network is used to learn local features, and an bidirectional gated recurrent unit network is used to capture long-range dependencies on the sequences of promoters and enhancers. Second, an attention mechanism is used for focusing on relatively important features. Finally, a matching heuristic mechanism is introduced for the exploration of the interaction between enhancers and promoters. We use benchmark datasets in evaluating and comparing the proposed method with existing methods. Comparative results show that our model is superior to currently existing models in multiple cell lines. Specifically, we found that the matching heuristic mechanism introduced into the proposed model mainly contributes to the improvement of performance in terms of overall accuracy. Additionally, compared with existing models, our model is more efficient with regard to computational speed.


Asunto(s)
Aprendizaje Profundo , Elementos de Facilitación Genéticos , Modelos Genéticos , Regiones Promotoras Genéticas , Biología Computacional , Células HeLa , Heurística , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células K562
12.
Brief Bioinform ; 22(6)2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34037687

RESUMEN

As the best substitute for antibiotics, antimicrobial peptides (AMPs) have important research significance. Due to the high cost and difficulty of experimental methods for identifying AMPs, more and more researches are focused on using computational methods to solve this problem. Most of the existing calculation methods can identify AMPs through the sequence itself, but there is still room for improvement in recognition accuracy, and there is a problem that the constructed model cannot be universal in each dataset. The pre-training strategy has been applied to many tasks in natural language processing (NLP) and has achieved gratifying results. It also has great application prospects in the field of AMP recognition and prediction. In this paper, we apply the pre-training strategy to the model training of AMP classifiers and propose a novel recognition algorithm. Our model is constructed based on the BERT model, pre-trained with the protein data from UniProt, and then fine-tuned and evaluated on six AMP datasets with large differences. Our model is superior to the existing methods and achieves the goal of accurate identification of datasets with small sample size. We try different word segmentation methods for peptide chains and prove the influence of pre-training steps and balancing datasets on the recognition effect. We find that pre-training on a large number of diverse AMP data, followed by fine-tuning on new data, is beneficial for capturing both new data's specific features and common features between AMP sequences. Finally, we construct a new AMP dataset, on which we train a general AMP recognition model.


Asunto(s)
Algoritmos , Péptidos Antimicrobianos/química , Biología Computacional/métodos , Procesamiento de Lenguaje Natural , Programas Informáticos , Péptidos Antimicrobianos/farmacología , Bases de Datos Genéticas , Reproducibilidad de los Resultados
13.
Brief Bioinform ; 22(2): 1902-1917, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-32363401

RESUMEN

The increase in biological data and the formation of various biomolecule interaction databases enable us to obtain diverse biological networks. These biological networks provide a wealth of raw materials for further understanding of biological systems, the discovery of complex diseases and the search for therapeutic drugs. However, the increase in data also increases the difficulty of biological networks analysis. Therefore, algorithms that can handle large, heterogeneous and complex data are needed to better analyze the data of these network structures and mine their useful information. Deep learning is a branch of machine learning that extracts more abstract features from a larger set of training data. Through the establishment of an artificial neural network with a network hierarchy structure, deep learning can extract and screen the input information layer by layer and has representation learning ability. The improved deep learning algorithm can be used to process complex and heterogeneous graph data structures and is increasingly being applied to the mining of network data information. In this paper, we first introduce the used network data deep learning models. After words, we summarize the application of deep learning on biological networks. Finally, we discuss the future development prospects of this field.


Asunto(s)
Aprendizaje Profundo , Redes Neurales de la Computación , Algoritmos , Minería de Datos , Sistemas de Liberación de Medicamentos , Descubrimiento de Drogas , Interacciones Farmacológicas , Genotipo , Humanos , Microbiota , Fenotipo , Mapas de Interacción de Proteínas , Proteínas/química
14.
Bioinformatics ; 38(24): 5406-5412, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36271850

RESUMEN

MOTIVATION: Drug-drug interaction (DDI) prediction is a challenging problem in pharmacology and clinical applications. With the increasing availability of large biomedical databases, large-scale biological knowledge graphs containing drug information have been widely used for DDI prediction. However, large knowledge graphs inevitably suffer from data noise problems, which limit the performance and interpretability of models based on the knowledge graph. Recent studies attempt to improve models by introducing inductive bias through an attention mechanism. However, they all only depend on the topology of entity nodes independently to generate fixed attention pathways, without considering the semantic diversity of entity nodes in different drug pair links. This makes it difficult for models to select more meaningful nodes to overcome data quality limitations and make more interpretable predictions. RESULTS: To address this issue, we propose a Link-aware Graph Attention method for DDI prediction, called LaGAT, which is able to generate different attention pathways for drug entities based on different drug pair links. For a drug pair link, the LaGAT uses the embedding representation of one of the drugs as a query vector to calculate the attention weights, thereby selecting the appropriate topological neighbor nodes to obtain the semantic information of the other drug. We separately conduct experiments on binary and multi-class classification and visualize the attention pathways generated by the model. The results prove that LaGAT can better capture semantic relationships and achieves remarkably superior performance over both the classical and state-of-the-art models on DDI prediction. AVAILABILITYAND IMPLEMENTATION: The source code and data are available at https://github.com/Azra3lzz/LaGAT. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Exactitud de los Datos , Semántica , Interacciones Farmacológicas , Bases de Datos Factuales , Programas Informáticos
15.
Langmuir ; 39(34): 12053-12062, 2023 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-37594209

RESUMEN

Biocompatible polymers with nontraditional intrinsic luminescence (NTIL) possess the advantages of environmental friendliness and facile structural regulation. To regulate the emission wavelength of polymers with NTIL, the alkane chain lengths of hyperbranched polysiloxane (HBPSi) are adjusted. Optical investigation shows that the emission wavelength of HBPSi is closely related to the alkane chain lengths; namely, short alkane chains will generate relatively long-wavelength emission. Electronic communication among functional groups is responsible for the emission. In a concentrated solution, HBPSi molecules aggregate together due to the strong hydrogen bond and amphiphilicity, and the functional groups in the aggregate are so close that their electron clouds are overlapped and generate spatial electronic delocalizations. HBPSi with shorter alkane chains will generate larger electronic delocalizations and emit longer-wavelength emissions. Moreover, these polymers show excellent applications in the fabrication of fluorescent films and chemical sensing. This work could provide a strategy for regulating the emission wavelengths of unconventional fluorescent polymers.

16.
Breast J ; 2023: 6282654, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38075552

RESUMEN

Breast cancer is considered the most prevalent malignancy due to its high incidence rate, recurrence, and metastasis in women that makes it one of the deadliest cancers. The current study aimed to predict the genes associated with the recurrence and metastasis of breast cancer and to validate their effect on MDA-MB-231 cells. Through the bioinformatics analysis, the transcription factor 7 cofactor (MLLT11) as the target gene was obtained. MLLT11-specific siRNA was synthesized and transfected into MDA-MB-231 cells. The results demonstrated that the siRNA significantly reduced the MLLT11 mRNA levels. Moreover, cell migration and invasion, as well as the protein levels of phosphatidylinositol 3-kinase (PI3K), AKT, matrix metalloproteinase (MMP) 2, and MMP9, were significantly lower in the groups treated with siRNA while the apoptosis was augmented. Collectively, MLLT11 siRNA elicited ameliorative properties on breast cancer cells, possibly via the inhibition of the PI3K/AKT signaling pathway.


Asunto(s)
Neoplasias de la Mama , Femenino , Humanos , Apoptosis , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Células MDA-MB-231 , Invasividad Neoplásica/patología , Proteínas de Neoplasias/farmacología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Factores de Transcripción
17.
Mikrochim Acta ; 190(6): 224, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37184606

RESUMEN

Nitrogen-doped carbon dots (NCDs) have been constructed in which coal washing wastewater is used as carbon precursor, tryptophan is added for nitrogen doping and surface functional together with polyethylene glycol. The nitrogen doping and surface functional with electron rich groups resulted in excellent fluorescent properties regarding stability, reversibility, printability with high quantum yield which not only enable the NCDs as fluorescent ink for advanced message encryption, but also realize specific on-off-on fluorescent sensing of Hg2+ and GSH as solution, hydrogel and filter paper sensors. The NCDs had a linear range of 0.01-100 µM and a detection limit of 6.27 nM (RSD 0.33%) for Hg2+ and the NCDs@Hg2+ had a linear range of 0.01-60 µM and a detection limit of 3.53 nM (RSD 1.53%) for GSH in sensing studies with aqueous solutions. In addition, with the low cytotoxicity and good biocompatibility NCDs have been successfully used for imaging Hg2+ and GSH in living MG-63 cells. The presented NCDs recycle waste coal washing water into worthwhile material which can be implemented as promising anti-counterfeiting and message encryption candidates as well as effective Hg2+ and GSH sensing, tracking and removing tools in complicated environmental and biological systems.


Asunto(s)
Mercurio , Puntos Cuánticos , Carbono , Colorantes Fluorescentes , Glutatión , Mercurio/análisis , Nitrógeno
18.
Brief Bioinform ; 21(1): 1-10, 2020 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-30239587

RESUMEN

Sequence clustering is a basic bioinformatics task that is attracting renewed attention with the development of metagenomics and microbiomics. The latest sequencing techniques have decreased costs and as a result, massive amounts of DNA/RNA sequences are being produced. The challenge is to cluster the sequence data using stable, quick and accurate methods. For microbiome sequencing data, 16S ribosomal RNA operational taxonomic units are typically used. However, there is often a gap between algorithm developers and bioinformatics users. Different software tools can produce diverse results and users can find them difficult to analyze. Understanding the different clustering mechanisms is crucial to understanding the results that they produce. In this review, we selected several popular clustering tools, briefly explained the key computing principles, analyzed their characters and compared them using two independent benchmark datasets. Our aim is to assist bioinformatics users in employing suitable clustering tools effectively to analyze big sequencing data. Related data, codes and software tools were accessible at the link http://lab.malab.cn/∼lg/clustering/.

19.
Brief Bioinform ; 21(2): 486-497, 2020 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-30753282

RESUMEN

A biological network is complex. A group of critical nodes determines the quality and state of such a network. Increasing studies have shown that diseases and biological networks are closely and mutually related and that certain diseases are often caused by errors occurring in certain nodes in biological networks. Thus, studying biological networks and identifying critical nodes can help determine the key targets in treating diseases. The problem is how to find the critical nodes in a network efficiently and with low cost. Existing experimental methods in identifying critical nodes generally require much time, manpower and money. Accordingly, many scientists are attempting to solve this problem by researching efficient and low-cost computing methods. To facilitate calculations, biological networks are often modeled as several common networks. In this review, we classify biological networks according to the network types used by several kinds of common computational methods and introduce the computational methods used by each type of network.


Asunto(s)
Biología Computacional/métodos , Algoritmos , Biología Computacional/economía , Costos y Análisis de Costo , Genes Esenciales , Proteínas/metabolismo
20.
PLoS Biol ; 17(6): e3000330, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31226122

RESUMEN

The repair of white matter damage is of paramount importance for functional recovery after brain injuries. Here, we report that interleukin-4 (IL-4) promotes oligodendrocyte regeneration and remyelination. IL-4 receptor expression was detected in a variety of glial cells after ischemic brain injury, including oligodendrocyte lineage cells. IL-4 deficiency in knockout mice resulted in greater deterioration of white matter over 14 d after stroke. Consistent with these findings, intranasal delivery of IL-4 nanoparticles after stroke improved white matter integrity and attenuated long-term sensorimotor and cognitive deficits in wild-type mice, as revealed by histological immunostaining, electron microscopy, diffusion tensor imaging, and electrophysiology. The selective effect of IL-4 on remyelination was verified in an ex vivo organotypic model of demyelination. By leveraging primary oligodendrocyte progenitor cells (OPCs), microglia-depleted mice, and conditional OPC-specific peroxisome proliferator-activated receptor gamma (PPARγ) knockout mice, we discovered a direct salutary effect of IL-4 on oligodendrocyte differentiation that was mediated by the PPARγ axis. Our findings reveal a new regenerative role of IL-4 in the central nervous system (CNS), which lies beyond its known immunoregulatory functions on microglia/macrophages or peripheral lymphocytes. Therefore, intranasal IL-4 delivery may represent a novel therapeutic strategy to improve white matter integrity in stroke and other brain injuries.


Asunto(s)
Interleucina-4/metabolismo , Oligodendroglía/metabolismo , PPAR gamma/metabolismo , Animales , Lesiones Encefálicas , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatología , Diferenciación Celular/fisiología , Enfermedades Desmielinizantes/metabolismo , Interleucina-4/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Vaina de Mielina/metabolismo , Regeneración Nerviosa , Neurogénesis , Oligodendroglía/fisiología , PPAR gamma/fisiología , Recuperación de la Función , Remielinización/fisiología , Transducción de Señal , Accidente Cerebrovascular , Sustancia Blanca
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA