Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 971
Filter
Add more filters

Publication year range
1.
Brief Bioinform ; 25(3)2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38706316

ABSTRACT

Protein-ligand interactions (PLIs) are essential for cellular activities and drug discovery. But due to the complexity and high cost of experimental methods, there is a great demand for computational approaches to recognize PLI patterns, such as protein-ligand docking. In recent years, more and more models based on machine learning have been developed to directly predict the root mean square deviation (RMSD) of a ligand docking pose with reference to its native binding pose. However, new scoring methods are pressingly needed in methodology for more accurate RMSD prediction. We present a new deep learning-based scoring method for RMSD prediction of protein-ligand docking poses based on a Graphormer method and Shell-like graph architecture, named GSScore. To recognize near-native conformations from a set of poses, GSScore takes atoms as nodes and then establishes the docking interface of protein-ligand into multiple bipartite graphs within different shell ranges. Benefiting from the Graphormer and Shell-like graph architecture, GSScore can effectively capture the subtle differences between energetically favorable near-native conformations and unfavorable non-native poses without extra information. GSScore was extensively evaluated on diverse test sets including a subset of PDBBind version 2019, CASF2016 as well as DUD-E, and obtained significant improvements over existing methods in terms of RMSE, $R$ (Pearson correlation coefficient), Spearman correlation coefficient and Docking power.


Subject(s)
Molecular Docking Simulation , Proteins , Ligands , Proteins/chemistry , Proteins/metabolism , Protein Binding , Software , Algorithms , Computational Biology/methods , Protein Conformation , Databases, Protein , Deep Learning
2.
Brief Bioinform ; 25(3)2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38600667

ABSTRACT

Human leukocyte antigen (HLA) recognizes foreign threats and triggers immune responses by presenting peptides to T cells. Computationally modeling the binding patterns between peptide and HLA is very important for the development of tumor vaccines. However, it is still a big challenge to accurately predict HLA molecules binding peptides. In this paper, we develop a new model TripHLApan for predicting HLA molecules binding peptides by integrating triple coding matrix, BiGRU + Attention models, and transfer learning strategy. We have found the main interaction site regions between HLA molecules and peptides, as well as the correlation between HLA encoding and binding motifs. Based on the discovery, we make the preprocessing and coding closer to the natural biological process. Besides, due to the input being based on multiple types of features and the attention module focused on the BiGRU hidden layer, TripHLApan has learned more sequence level binding information. The application of transfer learning strategies ensures the accuracy of prediction results under special lengths (peptides in length 8) and model scalability with the data explosion. Compared with the current optimal models, TripHLApan exhibits strong predictive performance in various prediction environments with different positive and negative sample ratios. In addition, we validate the superiority and scalability of TripHLApan's predictive performance using additional latest data sets, ablation experiments and binding reconstitution ability in the samples of a melanoma patient. The results show that TripHLApan is a powerful tool for predicting the binding of HLA-I and HLA-II molecular peptides for the synthesis of tumor vaccines. TripHLApan is publicly available at https://github.com/CSUBioGroup/TripHLApan.git.


Subject(s)
Cancer Vaccines , Humans , Protein Binding , Peptides/chemistry , HLA Antigens/chemistry , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class I/chemistry , Machine Learning
3.
J Virol ; 98(5): e0021224, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38591886

ABSTRACT

Porcine rotaviruses (PoRVs) cause severe economic losses in the swine industry. P[7] and P[23] are the predominant genotypes circulating on farms, but no vaccine is yet available. Here, we developed a bivalent subunit PoRV vaccine using truncated versions (VP4*) of the VP4 proteins from P[7] and P[23]. The vaccination of mice with the bivalent subunit vaccine elicited more robust neutralizing antibodies (NAbs) and cellular immune responses than its components, even at high doses. The bivalent subunit vaccine and inactivated bivalent vaccine prepared from strains PoRVs G9P[7] and G9P[23] were used to examine their protective efficacy in sows and suckling piglets after passive immunization. The immunized sows showed significantly elevated NAbs in the serum and colostrum, and the suckling piglets acquired high levels of sIgA antibodies from the colostrum. Challenging subunit-vaccinated or inactivated-vaccinated piglets with homologous virulent strains did not induce diarrhea, except in one or two piglets, which had mild diarrhea. Immunization with the bivalent subunit vaccine and inactivated vaccine also alleviated the microscopic lesions in the intestinal tissues caused by the challenge with the corresponding homologous virulent strain. However, all the piglets in the challenged group displayed mild to watery diarrhea and high levels of viral shedding, whereas the feces and intestines of the piglets in the bivalent subunit vaccine and inactivated vaccine groups had lower viral loads. In summary, our data show for the first time that a bivalent subunit vaccine combining VP4*P[7] and VP4*P[23] effectively protects piglets against the diarrhea caused by homologous virulent strains.IMPORTANCEPoRVs are the main causes of diarrhea in piglets worldwide. The multisegmented genome of PoRVs allows the reassortment of VP4 and VP7 genes from different RV species and strains. The P[7] and P[23] are the predominant genotypes circulating in pig farms, but no vaccine is available at present in China. Subunit vaccines, as nonreplicating vaccines, are an option to cope with variable genotypes. Here, we have developed a bivalent subunit candidate vaccine based on a truncated VP4 protein, which induced robust humoral and cellular immune responses and protected piglets against challenge with homologous PoRV. It also appears to be safe. These data show that the truncated VP4-protein-based subunit vaccine is a promising candidate for the prevention of PoRV diarrhea.


Subject(s)
Rotavirus Vaccines , Vaccines, Subunit , Animals , Female , Mice , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Diarrhea/prevention & control , Diarrhea/virology , Diarrhea/veterinary , Diarrhea/immunology , Genotype , Immunity, Cellular , Mice, Inbred BALB C , Rotavirus/immunology , Rotavirus Infections/prevention & control , Rotavirus Infections/veterinary , Rotavirus Infections/immunology , Rotavirus Infections/virology , Rotavirus Vaccines/immunology , Rotavirus Vaccines/administration & dosage , Swine , Swine Diseases/prevention & control , Swine Diseases/virology , Swine Diseases/immunology , Vaccination , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage
4.
Brief Bioinform ; 24(1)2023 01 19.
Article in English | MEDLINE | ID: mdl-36511222

ABSTRACT

Circular RNAs (circRNAs) are reverse-spliced and covalently closed RNAs. Their interactions with RNA-binding proteins (RBPs) have multiple effects on the progress of many diseases. Some computational methods are proposed to identify RBP binding sites on circRNAs but suffer from insufficient accuracy, robustness and explanation. In this study, we first take the characteristics of both RNA and RBP into consideration. We propose a method for discriminating circRNA-RBP binding sites based on multi-scale characterizing sequence and structure features, called CRMSS. For circRNAs, we use sequence ${k}\hbox{-}{mer}$ embedding and the forming probabilities of local secondary structures as features. For RBPs, we combine sequence and structure frequencies of RNA-binding domain regions to generate features. We capture binding patterns with multi-scale residual blocks. With BiLSTM and attention mechanism, we obtain the contextual information of high-level representation for circRNA-RBP binding. To validate the effectiveness of CRMSS, we compare its predictive performance with other methods on 37 RBPs. Taking the properties of both circRNAs and RBPs into account, CRMSS achieves superior performance over state-of-the-art methods. In the case study, our model provides reliable predictions and correctly identifies experimentally verified circRNA-RBP pairs. The code of CRMSS is freely available at https://github.com/BioinformaticsCSU/CRMSS.


Subject(s)
RNA, Circular , RNA , RNA, Circular/genetics , Binding Sites , RNA/metabolism , RNA-Binding Proteins/metabolism
5.
Brief Bioinform ; 24(1)2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36572658

ABSTRACT

Emerging evidence has proved that circular RNAs (circRNAs) are implicated in pathogenic processes. They are regarded as promising biomarkers for diagnosis due to covalently closed loop structures. As opposed to traditional experiments, computational approaches can identify circRNA-disease associations at a lower cost. Aggregating multi-source pathogenesis data helps to alleviate data sparsity and infer potential associations at the system level. The majority of computational approaches construct a homologous network using multi-source data, but they lose the heterogeneity of the data. Effective methods that use the features of multi-source data are considered as a matter of urgency. In this paper, we propose a model (CDHGNN) based on edge-weighted graph attention and heterogeneous graph neural networks for potential circRNA-disease association prediction. The circRNA network, micro RNA network, disease network and heterogeneous network are constructed based on multi-source data. To reflect association probabilities between nodes, an edge-weighted graph attention network model is designed for node features. To assign attention weights to different types of edges and learn contextual meta-path, CDHGNN infers potential circRNA-disease association based on heterogeneous neural networks. CDHGNN outperforms state-of-the-art algorithms in terms of accuracy. Edge-weighted graph attention networks and heterogeneous graph networks have both improved performance significantly. Furthermore, case studies suggest that CDHGNN is capable of identifying specific molecular associations and investigating biomolecular regulatory relationships in pathogenesis. The code of CDHGNN is freely available at https://github.com/BioinformaticsCSU/CDHGNN.


Subject(s)
MicroRNAs , RNA, Circular , RNA, Circular/genetics , Neural Networks, Computer , MicroRNAs/genetics , Algorithms , Computational Biology/methods
6.
Brief Bioinform ; 24(1)2023 01 19.
Article in English | MEDLINE | ID: mdl-36611256

ABSTRACT

Accumulating evidences demonstrate that circular RNA (circRNA) plays an important role in human diseases. Identification of circRNA-disease associations can help for the diagnosis of human diseases, while the traditional method based on biological experiments is time-consuming. In order to address the limitation, a series of computational methods have been proposed in recent years. However, few works have summarized these methods or compared the performance of them. In this paper, we divided the existing methods into three categories: information propagation, traditional machine learning and deep learning. Then, the baseline methods in each category are introduced in detail. Further, 5 different datasets are collected, and 14 representative methods of each category are selected and compared in the 5-fold, 10-fold cross-validation and the de novo experiment. In order to further evaluate the effectiveness of these methods, six common cancers are selected to compare the number of correctly identified circRNA-disease associations in the top-10, top-20, top-50, top-100 and top-200. In addition, according to the results, the observation about the robustness and the character of these methods are concluded. Finally, the future directions and challenges are discussed.


Subject(s)
Neoplasms , RNA, Circular , Humans , RNA, Circular/genetics , Benchmarking , Machine Learning , Neoplasms/genetics , Computational Biology/methods
7.
Bioinformatics ; 40(4)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38530778

ABSTRACT

MOTIVATION: Studying the molecular heterogeneity of cancer is essential for achieving personalized therapy. At the same time, understanding the biological processes that drive cancer development can lead to the identification of valuable therapeutic targets. Therefore, achieving accurate and interpretable clinical predictions requires paramount attention to thoroughly characterizing patients at both the molecular and biological pathway levels. RESULTS: Here, we present GraphPath, a biological knowledge-driven graph neural network with multi-head self-attention mechanism that implements the pathway-pathway interaction network. We train GraphPath to classify the cancer status of patients with prostate cancer based on their multi-omics profiling. Experiment results show that our method outperforms P-NET and other baseline methods. Besides, two external cohorts are used to validate that the model can be generalized to unseen samples with adequate predictive performance. We reduce the dimensionality of latent pathway embeddings and visualize corresponding classes to further demonstrate the optimal performance of the model. Additionally, since GraphPath's predictions are interpretable, we identify target cancer-associated pathways that significantly contribute to the model's predictions. Such a robust and interpretable model has the potential to greatly enhance our understanding of cancer's biological mechanisms and accelerate the development of targeted therapies. AVAILABILITY AND IMPLEMENTATION: https://github.com/amazingma/GraphPath.


Subject(s)
Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/genetics , Neural Networks, Computer
8.
Bioinformatics ; 40(Supplement_1): i418-i427, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38940145

ABSTRACT

MOTIVATION: Mutations are the crucial driving force for biological evolution as they can disrupt protein stability and protein-protein interactions which have notable impacts on protein structure, function, and expression. However, existing computational methods for protein mutation effects prediction are generally limited to single point mutations with global dependencies, and do not systematically take into account the local and global synergistic epistasis inherent in multiple point mutations. RESULTS: To this end, we propose a novel spatial and sequential message passing neural network, named DDAffinity, to predict the changes in binding affinity caused by multiple point mutations based on protein 3D structures. Specifically, instead of being on the whole protein, we perform message passing on the k-nearest neighbor residue graphs to extract pocket features of the protein 3D structures. Furthermore, to learn global topological features, a two-step additive Gaussian noising strategy during training is applied to blur out local details of protein geometry. We evaluate DDAffinity on benchmark datasets and external validation datasets. Overall, the predictive performance of DDAffinity is significantly improved compared with state-of-the-art baselines on multiple point mutations, including end-to-end and pre-training based methods. The ablation studies indicate the reasonable design of all components of DDAffinity. In addition, applications in nonredundant blind testing, predicting mutation effects of SARS-CoV-2 RBD variants, and optimizing human antibody against SARS-CoV-2 illustrate the effectiveness of DDAffinity. AVAILABILITY AND IMPLEMENTATION: DDAffinity is available at https://github.com/ak422/DDAffinity.


Subject(s)
Point Mutation , SARS-CoV-2 , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Computational Biology/methods , Protein Conformation , Humans , Neural Networks, Computer , Protein Binding , COVID-19/virology , Proteins/chemistry , Proteins/metabolism , Algorithms
9.
Bioinformatics ; 40(Supplement_1): i511-i520, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38940121

ABSTRACT

MOTIVATION: Identifying cancer genes remains a significant challenge in cancer genomics research. Annotated gene sets encode functional associations among multiple genes, and cancer genes have been shown to cluster in hallmark signaling pathways and biological processes. The knowledge of annotated gene sets is critical for discovering cancer genes but remains to be fully exploited. RESULTS: Here, we present the DIsease-Specific Hypergraph neural network (DISHyper), a hypergraph-based computational method that integrates the knowledge from multiple types of annotated gene sets to predict cancer genes. First, our benchmark results demonstrate that DISHyper outperforms the existing state-of-the-art methods and highlight the advantages of employing hypergraphs for representing annotated gene sets. Second, we validate the accuracy of DISHyper-predicted cancer genes using functional validation results and multiple independent functional genomics data. Third, our model predicts 44 novel cancer genes, and subsequent analysis shows their significant associations with multiple types of cancers. Overall, our study provides a new perspective for discovering cancer genes and reveals previously undiscovered cancer genes. AVAILABILITY AND IMPLEMENTATION: DISHyper is freely available for download at https://github.com/genemine/DISHyper.


Subject(s)
Neoplasms , Neural Networks, Computer , Humans , Neoplasms/genetics , Computational Biology/methods , Genomics/methods , Genes, Neoplasm , Molecular Sequence Annotation/methods , Databases, Genetic
10.
Bioinformatics ; 40(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38889266

ABSTRACT

MOTIVATION: Nanopore direct RNA sequencing (DRS) enables the detection of RNA N6-methyladenosine (m6A) without extra laboratory techniques. A number of supervised or comparative approaches have been developed to identify m6A from Nanopore DRS reads. However, existing methods typically utilize either statistical features of the current signals or basecalling-error features, ignoring the richer information of the raw signals of DRS reads. RESULTS: Here, we propose RedNano, a deep-learning method designed to detect m6A from Nanopore DRS reads by utilizing both raw signals and basecalling errors. RedNano processes the raw-signal feature and basecalling-error feature through residual networks. We validated the effectiveness of RedNano using synthesized, Arabidopsis, and human DRS data. The results demonstrate that RedNano surpasses existing methods by achieving higher area under the ROC curve (AUC) and area under the precision-recall curve (AUPRs) in all three datasets. Furthermore, RedNano performs better in cross-species validation, demonstrating its robustness. Additionally, when detecting m6A from an independent dataset of Populus trichocarpa, RedNano achieves the highest AUC and AUPR, which are 3.8%-9.9% and 5.5%-13.8% higher than other methods, respectively. AVAILABILITY AND IMPLEMENTATION: The source code of RedNano is freely available at https://github.com/Derryxu/RedNano.


Subject(s)
Arabidopsis , Arabidopsis/genetics , Humans , Sequence Analysis, RNA/methods , Adenosine/analogs & derivatives , Adenosine/analysis , Nanopore Sequencing/methods , Deep Learning , RNA/chemistry , Nanopores
11.
Bioinformatics ; 40(2)2024 02 01.
Article in English | MEDLINE | ID: mdl-38317025

ABSTRACT

MOTIVATION: Dropout events bring challenges in analyzing single-cell RNA sequencing data as they introduce noise and distort the true distributions of gene expression profiles. Recent studies focus on estimating dropout probability and imputing dropout events by leveraging information from similar cells or genes. However, the number of dropout events differs in different cells, due to the complex factors, such as different sequencing protocols, cell types, and batch effects. The dropout event differences are not fully considered in assessing the similarities between cells and genes, which compromises the reliability of downstream analysis. RESULTS: This work proposes a hybrid Generative Adversarial Network with dropouts identification to impute single-cell RNA sequencing data, named AGImpute. First, the numbers of dropout events in different cells in scRNA-seq data are differentially estimated by using a dynamic threshold estimation strategy. Next, the identified dropout events are imputed by a hybrid deep learning model, combining Autoencoder with a Generative Adversarial Network. To validate the efficiency of the AGImpute, it is compared with seven state-of-the-art dropout imputation methods on two simulated datasets and seven real single-cell RNA sequencing datasets. The results show that AGImpute imputes the least number of dropout events than other methods. Moreover, AGImpute enhances the performance of downstream analysis, including clustering performance, identifying cell-specific marker genes, and inferring trajectory in the time-course dataset. AVAILABILITY AND IMPLEMENTATION: The source code can be obtained from https://github.com/xszhu-lab/AGImpute.


Subject(s)
Single-Cell Analysis , Single-Cell Gene Expression Analysis , Sequence Analysis, RNA/methods , Reproducibility of Results , Single-Cell Analysis/methods , Transcriptome , Software , Cluster Analysis , Gene Expression Profiling
12.
Methods ; 226: 21-27, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38608849

ABSTRACT

Knowledge graph intent graph attention mechanism Predicting drug-target interactions (DTIs) plays a crucial role in drug discovery and drug development. Considering the high cost and risk of biological experiments, developing computational approaches to explore the interactions between drugs and targets can effectively reduce the time and cost of drug development. Recently, many methods have made significant progress in predicting DTIs. However, existing approaches still suffer from the high sparsity of DTI datasets and the cold start problem. In this paper, we develop a new model to predict drug-target interactions via a knowledge graph and intent graph named DTKGIN. Our method can effectively capture biological environment information for targets and drugs by mining their associated relations in the knowledge graph and considering drug-target interactions at a fine-grained level in the intent graph. DTKGIN learns the representation of drugs and targets from the knowledge graph and the intent graph. Then the probabilities of interactions between drugs and targets are obtained through the inner product of the representation of drugs and targets. Experimental results show that our proposed method outperforms other state-of-the-art methods in 10-fold cross-validation, especially in cold-start experimental settings. Furthermore, the case studies demonstrate the effectiveness of DTKGIN in predicting potential drug-target interactions. The code is available on GitHub: https://github.com/Royluoyi123/DTKGIN.


Subject(s)
Drug Discovery , Drug Discovery/methods , Humans , Algorithms , Computational Biology/methods , Drug Development/methods
13.
Proc Natl Acad Sci U S A ; 119(43): e2208506119, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36256824

ABSTRACT

DNA-damaging treatments such as radiotherapy (RT) have become promising to improve the efficacy of immune checkpoint inhibitors by enhancing tumor immunogenicity. However, accompanying treatment-related detrimental events in normal tissues have posed a major obstacle to radioimmunotherapy and present new challenges to the dose delivery mode of clinical RT. In the present study, ultrahigh dose rate FLASH X-ray irradiation was applied to counteract the intestinal toxicity in the radioimmunotherapy. In the context of programmed cell death ligand-1 (PD-L1) blockade, FLASH X-ray minimized mouse enteritis by alleviating CD8+ T cell-mediated deleterious immune response compared with conventional dose rate (CONV) irradiation. Mechanistically, FLASH irradiation was less efficient than CONV X-ray in eliciting cytoplasmic double-stranded DNA (dsDNA) and in activating cyclic GMP-AMP synthase (cGAS) in the intestinal crypts, resulting in the suppression of the cascade feedback consisting of CD8+ T cell chemotaxis and gasdermin E-mediated intestinal pyroptosis in the case of PD-L1 blocking. Meanwhile, FLASH X-ray was as competent as CONV RT in boosting the antitumor immune response initiated by cGAS activation and achieved equal tumor control in metastasis burdens when combined with anti-PD-L1 administration. Together, the present study revealed an encouraging protective effect of FLASH X-ray upon the normal tissue without compromising the systemic antitumor response when combined with immunological checkpoint inhibitors, providing the rationale for testing this combination as a clinical application in radioimmunotherapy.


Subject(s)
Neoplasms , Radioimmunotherapy , Mice , Animals , X-Rays , Pyroptosis , Immune Checkpoint Inhibitors , Ligands , Nucleotidyltransferases/metabolism
14.
J Am Chem Soc ; 146(15): 10357-10366, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38574191

ABSTRACT

Electrochemical reduction of carbon dioxide to organic chemicals provides a value-added route for mitigating greenhouse gas emissions. We report a family of carbon-supported Sn electrocatalysts with the tin size varying from single atom, ultrasmall clusters to nanocrystallites. High single-product Faradaic efficiency (FE) and low onset potential of CO2 conversion to acetate (FE = 90% @ -0.6 V), ethanol (FE = 92% @ -0.4 V), and formate (FE = 91% @ -0.6 V) were achieved over the catalysts of different active site dimensions. The CO2 conversion mechanism behind these highly selective, size-modulated p-block element catalysts was elucidated by structural characterization and computational modeling, together with kinetic isotope effect investigation.

15.
Gastroenterology ; 165(5): 1219-1232, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37507075

ABSTRACT

BACKGROUND & AIMS: BiTE (bispecific T-cell engager) immune therapy has demonstrated clinical activity in multiple tumor indications, but its influence in the tumor microenvironment remains unclear. CLDN18.2 is overexpressed in solid tumors including gastric cancer (GC) and pancreatic ductal adenocarcinoma (PDAC), both of which are characterized by the presence of immunosuppressive cells, including regulatory T cells (Tregs) and few effector T cells (Teffs). METHODS: We evaluated the activity of AMG 910, a CLDN18.2-targeted half-life extended (HLE) BiTE molecule, in GC and PDAC preclinical models and cocultured Tregs and Teffs in the presence of CLDN18.2-HLE-BiTE. RESULTS: AMG 910 induced potent, specific cytotoxicity in GC and PDAC cell lines. In GSU and SNU-620 GC xenograft models, AMG 910 engaged human CD3+ T cells with tumor cells, resulting in significant antitumor activity. AMG 910 monotherapy, in combination with a programmed death-1 (PD-1) inhibitor, suppressed tumor growth and enhanced survival in an orthotopic Panc4.14 PDAC model. Moreover, Treg infusion enhanced the antitumor efficacy of AMG 910 in the Panc4.14 model. In syngeneic KPC models of PDAC, treatment with a mouse surrogate CLDN18.2-HLE-BiTE (muCLDN18.2-HLE-BiTE) or the combination with an anti-PD-1 antibody significantly inhibited tumor growth. Tregs isolated from mice bearing KPC tumors that were treated with muCLDN18.2-HLE-BiTE showed decreased T cell suppressive activity and enhanced Teff cytotoxic activity, associated with increased production of type I cytokines and expression of Teff gene signatures. CONCLUSIONS: Our data suggest that BiTE molecule treatment converts Treg function from immunosuppressive to immune enhancing, leading to antitumor activity in immunologically "cold" tumors.


Subject(s)
Antibodies, Bispecific , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Animals , Mice , T-Lymphocytes, Regulatory/metabolism , Antibodies, Bispecific/genetics , Antibodies, Bispecific/pharmacology , Pancreatic Neoplasms/drug therapy , Cell Adhesion Molecules , Carcinoma, Pancreatic Ductal/drug therapy , Immunity , Tumor Microenvironment , Claudins
16.
Brief Bioinform ; 23(4)2022 07 18.
Article in English | MEDLINE | ID: mdl-35653713

ABSTRACT

Proteins maintain the functional order of cell in life by interacting with other proteins. Determination of protein complex structural information gives biological insights for the research of diseases and drugs. Recently, a breakthrough has been made in protein monomer structure prediction. However, due to the limited number of the known protein structure and homologous sequences of complexes, the prediction of residue-residue contacts on hetero-dimer interfaces is still a challenge. In this study, we have developed a deep learning framework for inferring inter-protein residue contacts from sequential information, called HDIContact. We utilized transfer learning strategy to produce Multiple Sequence Alignment (MSA) two-dimensional (2D) embedding based on patterns of concatenated MSA, which could reduce the influence of noise on MSA caused by mismatched sequences or less homology. For MSA 2D embedding, HDIContact took advantage of Bi-directional Long Short-Term Memory (BiLSTM) with two-channel to capture 2D context of residue pairs. Our comprehensive assessment on the Escherichia coli (E. coli) test dataset showed that HDIContact outperformed other state-of-the-art methods, with top precision of 65.96%, the Area Under the Receiver Operating Characteristic curve (AUROC) of 83.08% and the Area Under the Precision Recall curve (AUPR) of 25.02%. In addition, we analyzed the potential of HDIContact for human-virus protein-protein complexes, by achieving top five precision of 80% on O75475-P04584 related to Human Immunodeficiency Virus. All experiments indicated that our method was a valuable technical tool for predicting inter-protein residue contacts, which would be helpful for understanding protein-protein interaction mechanisms.


Subject(s)
Escherichia coli , Proteins , Computational Biology/methods , Humans , Machine Learning , Proteins/chemistry , Sequence Alignment
17.
Brief Bioinform ; 23(1)2022 01 17.
Article in English | MEDLINE | ID: mdl-34874989

ABSTRACT

Investigating differentially methylated regions (DMRs) presented in different tissues or cell types can help to reveal the mechanisms behind the tissue-specific gene expression. The identified tissue-/disease-specific DMRs also can be used as feature markers for spotting the tissues-of-origins of cell-free DNA (cfDNA) in noninvasive diagnosis. In recent years, many methods have been proposed to detect DMRs. However, due to the lack of benchmark DMRs, it is difficult for researchers to choose proper methods and select desirable DMR sets for downstream studies. The application of DMRs, used as feature markers, can be benefited by the longer length of DMRs containing more CpG sites when a threshold is given for the methylation differences of DMRs. According to this, two metrics ($Qn$ and $Ql$), in which the CpG numbers and lengths of DMRs with different methylation differences are weighted differently, are proposed in this paper to evaluate the DMR sets predicted by different methods on BS-seq data. DMR sets predicted by eight methods on both simulated datasets and real BS-seq datasets are evaluated by the proposed metrics, the benchmark-based metrics, and the enrichment analysis of biological data, including genomic features, transcription factors and histones. The rank correlation analysis shows that the $Qn$ and $Ql$ are highly correlated to the benchmark metrics for simulated datasets and the biological data enrichment analysis for real BS-seq data. Therefore, with no need for additional biological data, the proposed metrics can help researchers selecting a more suitable DMR set on a certain BS-seq dataset.


Subject(s)
Benchmarking , DNA Methylation , CpG Islands , Genome , Genomics , Sequence Analysis, DNA
18.
Brief Bioinform ; 23(3)2022 05 13.
Article in English | MEDLINE | ID: mdl-35289352

ABSTRACT

Determining drug indications is a critical part of the drug development process. However, traditional drug discovery is expensive and time-consuming. Drug repositioning aims to find potential indications for existing drugs, which is considered as an important alternative to the traditional drug discovery. In this article, we propose a multi-view learning with matrix completion (MLMC) method to predict the potential associations between drugs and diseases. Specifically, MLMC first learns the comprehensive similarity matrices from five drug similarity matrices and two disease similarity matrices based on the multi-view learning (ML) with Laplacian graph regularization, and updates the drug-disease association matrix simultaneously. Then, we introduce matrix completion (MC) to add some positive entries in original association matrix based on low-rank structure, and re-execute the multi-view learning algorithm for association prediction. At last, the prediction results of the above two operations are integrated as the final output. Evaluated by 10-fold cross-validation and de novo tests, MLMC achieves higher prediction accuracy than the current state-of-the-art methods. Moreover, case studies confirm the ability of our method in novel drug-disease association discovery. The codes of MLMC are available at https://github.com/BioinformaticsCSU/MLMC. Contact: jxwang@mail.csu.edu.cn.


Subject(s)
Computational Biology , Drug Repositioning , Algorithms , Computational Biology/methods , Drug Discovery , Drug Repositioning/methods
19.
Brief Bioinform ; 23(1)2022 01 17.
Article in English | MEDLINE | ID: mdl-34619757

ABSTRACT

Long-read sequencing technology enables significant progress in de novo genome assembly. However, the high error rate and the wide error distribution of raw reads result in a large number of errors in the assembly. Polishing is a procedure to fix errors in the draft assembly and improve the reliability of genomic analysis. However, existing methods treat all the regions of the assembly equally while there are fundamental differences between the error distributions of these regions. How to achieve very high accuracy in genome assembly is still a challenging problem. Motivated by the uneven errors in different regions of the assembly, we propose a novel polishing workflow named BlockPolish. In this method, we divide contigs into blocks with low complexity and high complexity according to statistics of aligned nucleotide bases. Multiple sequence alignment is applied to realign raw reads in complex blocks and optimize the alignment result. Due to the different distributions of error rates in trivial and complex blocks, two multitask bidirectional Long short-term memory (LSTM) networks are proposed to predict the consensus sequences. In the whole-genome assemblies of NA12878 assembled by Wtdbg2 and Flye using Nanopore data, BlockPolish has a higher polishing accuracy than other state-of-the-arts including Racon, Medaka and MarginPolish & HELEN. In all assemblies, errors are predominantly indels and BlockPolish has a good performance in correcting them. In addition to the Nanopore assemblies, we further demonstrate that BlockPolish can also reduce the errors in the PacBio assemblies. The source code of BlockPolish is freely available on Github (https://github.com/huangnengCSU/BlockPolish).


Subject(s)
High-Throughput Nucleotide Sequencing , Software , High-Throughput Nucleotide Sequencing/methods , Reproducibility of Results , Sequence Alignment , Sequence Analysis, DNA/methods
20.
Brief Bioinform ; 23(1)2022 01 17.
Article in English | MEDLINE | ID: mdl-34718402

ABSTRACT

The side effects of drugs present growing concern attention in the healthcare system. Accurately identifying the side effects of drugs is very important for drug development and risk assessment. Some computational models have been developed to predict the potential side effects of drugs and provided satisfactory performance. However, most existing methods can only predict whether side effects will occur and cannot determine the frequency of side effects. Although a few existing methods can predict the frequency of drug side effects, they strongly depend on the known drug-side effect relationships. Therefore, they cannot be applied to new drugs without known side effect frequency information. In this paper, we develop a novel similarity-based deep learning method, named SDPred, for determining the frequencies of drug side effects. Compared with the existing state-of-the-art models, SDPred integrates rich features and can be applied to predict the side effect frequencies of new drugs without any known drug-side effect association or frequency information. To our knowledge, this is the first work that can predict the side effect frequencies of new drugs in the population. The comparison results indicate that SDPred is much superior to all previously reported models. In addition, some case studies also demonstrate the effectiveness of our proposed method in practical applications. The SDPred software and data are freely available at https://github.com/zhc940702/SDPred, https://zenodo.org/record/5112573 and https://hub.docker.com/r/zhc940702/sdpred.


Subject(s)
Deep Learning , Drug-Related Side Effects and Adverse Reactions , Algorithms , Computational Biology/methods , Humans , Software
SELECTION OF CITATIONS
SEARCH DETAIL