LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 44

Search options

  1. Article ; Online: Joint deep autoencoder and subgraph augmentation for inferring microbial responses to drugs.

    Zhou, Zhecheng / Zhuo, Linlin / Fu, Xiangzheng / Zou, Quan

    Briefings in bioinformatics

    2024  Volume 25, Issue 1

    Abstract: Exploring microbial stress responses to drugs is crucial for the advancement of new therapeutic methods. While current artificial intelligence methodologies have expedited our understanding of potential microbial responses to drugs, the models are ... ...

    Abstract Exploring microbial stress responses to drugs is crucial for the advancement of new therapeutic methods. While current artificial intelligence methodologies have expedited our understanding of potential microbial responses to drugs, the models are constrained by the imprecise representation of microbes and drugs. To this end, we combine deep autoencoder and subgraph augmentation technology for the first time to propose a model called JDASA-MRD, which can identify the potential indistinguishable responses of microbes to drugs. In the JDASA-MRD model, we begin by feeding the established similarity matrices of microbe and drug into the deep autoencoder, enabling to extract robust initial features of both microbes and drugs. Subsequently, we employ the MinHash and HyperLogLog algorithms to account intersections and cardinality data between microbe and drug subgraphs, thus deeply extracting the multi-hop neighborhood information of nodes. Finally, by integrating the initial node features with subgraph topological information, we leverage graph neural network technology to predict the microbes' responses to drugs, offering a more effective solution to the 'over-smoothing' challenge. Comparative analyses on multiple public datasets confirm that the JDASA-MRD model's performance surpasses that of current state-of-the-art models. This research aims to offer a more profound insight into the adaptability of microbes to drugs and to furnish pivotal guidance for drug treatment strategies. Our data and code are publicly available at: https://github.com/ZZCrazy00/JDASA-MRD.
    MeSH term(s) Artificial Intelligence ; Algorithms ; Neural Networks, Computer
    Language English
    Publishing date 2024-01-03
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2068142-2
    ISSN 1477-4054 ; 1467-5463
    ISSN (online) 1477-4054
    ISSN 1467-5463
    DOI 10.1093/bib/bbad483
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: StableDNAm: towards a stable and efficient model for predicting DNA methylation based on adaptive feature correction learning.

    Zhuo, Linlin / Wang, Rui / Fu, Xiangzheng / Yao, Xiaojun

    BMC genomics

    2023  Volume 24, Issue 1, Page(s) 742

    Abstract: Background: DNA methylation, instrumental in numerous life processes, underscores the paramount importance of its accurate prediction. Recent studies suggest that deep learning, due to its capacity to extract profound insights, provides a more precise ... ...

    Abstract Background: DNA methylation, instrumental in numerous life processes, underscores the paramount importance of its accurate prediction. Recent studies suggest that deep learning, due to its capacity to extract profound insights, provides a more precise DNA methylation prediction. However, issues related to the stability and generalization performance of these models persist.
    Results: In this study, we introduce an efficient and stable DNA methylation prediction model. This model incorporates a feature fusion approach, adaptive feature correction technology, and a contrastive learning strategy. The proposed model presents several advantages. First, DNA sequences are encoded at four levels to comprehensively capture intricate information across multi-scale and low-span features. Second, we design a sequence-specific feature correction module that adaptively adjusts the weights of sequence features. This improvement enhances the model's stability and scalability, or its generality. Third, our contrastive learning strategy mitigates the instability issues resulting from sparse data. To validate our model, we conducted multiple sets of experiments on commonly used datasets, demonstrating the model's robustness and stability. Simultaneously, we amalgamate various datasets into a single, unified dataset. The experimental outcomes from this combined dataset substantiate the model's robust adaptability.
    Conclusions: Our research findings affirm that the StableDNAm model is a general, stable, and effective instrument for DNA methylation prediction. It holds substantial promise for providing invaluable assistance in future methylation-related research and analyses.
    MeSH term(s) DNA Methylation ; Protein Processing, Post-Translational
    Language English
    Publishing date 2023-12-05
    Publishing country England
    Document type Journal Article
    ZDB-ID 2041499-7
    ISSN 1471-2164 ; 1471-2164
    ISSN (online) 1471-2164
    ISSN 1471-2164
    DOI 10.1186/s12864-023-09802-7
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article: An efficient model for predicting human diseases through miRNA based on multiple-types of contrastive learning.

    Liao, Qingquan / Fu, Xiangzheng / Zhuo, Linlin / Chen, Hao

    Frontiers in microbiology

    2023  Volume 14, Page(s) 1325001

    Abstract: Multiple studies have demonstrated that microRNA (miRNA) can be deeply involved in the regulatory mechanism of human microbiota, thereby inducing disease. Developing effective methods to infer potential associations between microRNAs (miRNAs) and ... ...

    Abstract Multiple studies have demonstrated that microRNA (miRNA) can be deeply involved in the regulatory mechanism of human microbiota, thereby inducing disease. Developing effective methods to infer potential associations between microRNAs (miRNAs) and diseases can aid early diagnosis and treatment. Recent methods utilize machine learning or deep learning to predict miRNA-disease associations (MDAs), achieving state-of-the-art performance. However, the problem of sparse neighborhoods of nodes due to lack of data has not been well solved. To this end, we propose a new model named MTCL-MDA, which integrates multiple-types of contrastive learning strategies into a graph collaborative filtering model to predict potential MDAs. The model adopts a contrastive learning strategy based on topology, which alleviates the damage to model performance caused by sparse neighborhoods. In addition, the model also adopts a semantic-based contrastive learning strategy, which not only reduces the impact of noise introduced by topology-based contrastive learning, but also enhances the semantic information of nodes. Experimental results show that our model outperforms existing models on all evaluation metrics. Case analysis shows that our model can more accurately identify potential MDA, which is of great significance for the screening and diagnosis of real-life diseases. Our data and code are publicly available at: https://github.com/Lqingquan/MTCL-MDA.
    Language English
    Publishing date 2023-12-14
    Publishing country Switzerland
    Document type Journal Article
    ZDB-ID 2587354-4
    ISSN 1664-302X
    ISSN 1664-302X
    DOI 10.3389/fmicb.2023.1325001
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: DlncRNALoc: A discrete wavelet transform-based model for predicting lncRNA subcellular localization.

    Fu, Xiangzheng / Chen, Yifan / Tian, Sha

    Mathematical biosciences and engineering : MBE

    2023  Volume 20, Issue 12, Page(s) 20648–20667

    Abstract: The prediction of long non-coding RNA (lncRNA) subcellular localization is essential to the understanding of its function and involvement in cellular regulation. Traditional biological experimental methods are costly and time-consuming, making ... ...

    Abstract The prediction of long non-coding RNA (lncRNA) subcellular localization is essential to the understanding of its function and involvement in cellular regulation. Traditional biological experimental methods are costly and time-consuming, making computational methods the preferred approach for predicting lncRNA subcellular localization (LSL). However, existing computational methods have limitations due to the structural characteristics of lncRNAs and the uneven distribution of data across subcellular compartments. We propose a discrete wavelet transform (DWT)-based model for predicting LSL, called DlncRNALoc. We construct a physicochemical property matrix of a 2-tuple bases based on lncRNA sequences, and we introduce a DWT lncRNA feature extraction method. We use the Synthetic Minority Over-sampling Technique (SMOTE) for oversampling and the local fisher discriminant analysis (LFDA) algorithm to optimize feature information. The optimized feature vectors are fed into support vector machine (SVM) to construct a predictive model. DlncRNALoc has been applied for a five-fold cross-validation on the three sets of benchmark datasets. Extensive experiments have demonstrated the superiority and effectiveness of the DlncRNALoc model in predicting LSL.
    MeSH term(s) RNA, Long Noncoding/genetics ; Wavelet Analysis ; Algorithms ; Support Vector Machine ; Computational Biology/methods
    Chemical Substances RNA, Long Noncoding
    Language English
    Publishing date 2023-11-14
    Publishing country United States
    Document type Journal Article
    ZDB-ID 2265126-3
    ISSN 1551-0018 ; 1551-0018
    ISSN (online) 1551-0018
    ISSN 1551-0018
    DOI 10.3934/mbe.2023913
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: MS-BACL: enhancing metabolic stability prediction through bond graph augmentation and contrastive learning.

    Wang, Tao / Li, Zhen / Zhuo, Linlin / Chen, Yifan / Fu, Xiangzheng / Zou, Quan

    Briefings in bioinformatics

    2024  Volume 25, Issue 3

    Abstract: Motivation: Accurately predicting molecular metabolic stability is of great significance to drug research and development, ensuring drug safety and effectiveness. Existing deep learning methods, especially graph neural networks, can reveal the molecular ...

    Abstract Motivation: Accurately predicting molecular metabolic stability is of great significance to drug research and development, ensuring drug safety and effectiveness. Existing deep learning methods, especially graph neural networks, can reveal the molecular structure of drugs and thus efficiently predict the metabolic stability of molecules. However, most of these methods focus on the message passing between adjacent atoms in the molecular graph, ignoring the relationship between bonds. This makes it difficult for these methods to estimate accurate molecular representations, thereby being limited in molecular metabolic stability prediction tasks.
    Results: We propose the MS-BACL model based on bond graph augmentation technology and contrastive learning strategy, which can efficiently and reliably predict the metabolic stability of molecules. To our knowledge, this is the first time that bond-to-bond relationships in molecular graph structures have been considered in the task of metabolic stability prediction. We build a bond graph based on 'atom-bond-atom', and the model can simultaneously capture the information of atoms and bonds during the message propagation process. This enhances the model's ability to reveal the internal structure of the molecule, thereby improving the structural representation of the molecule. Furthermore, we perform contrastive learning training based on the molecular graph and its bond graph to learn the final molecular representation. Multiple sets of experimental results on public datasets show that the proposed MS-BACL model outperforms the state-of-the-art model.
    Availability and implementation: The code and data are publicly available at https://github.com/taowang11/MS.
    MeSH term(s) Neural Networks, Computer
    Language English
    Publishing date 2024-03-30
    Publishing country England
    Document type Journal Article
    ZDB-ID 2068142-2
    ISSN 1477-4054 ; 1467-5463
    ISSN (online) 1477-4054
    ISSN 1467-5463
    DOI 10.1093/bib/bbae127
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: AEGNN-M:A 3D Graph-Spatial Co-Representation Model for Molecular Property Prediction.

    Cai, Lijun / He, Yuling / Fu, Xiangzheng / Zhuo, Linlin / Zou, Quan / Yao, Xiaojun

    IEEE journal of biomedical and health informatics

    2024  Volume PP

    Abstract: Improving the drug development process can expedite the introduction of more novel drugs that cater to the demands of precision medicine. Accurately predicting molecular properties remains a fundamental challenge in drug discovery and development. ... ...

    Abstract Improving the drug development process can expedite the introduction of more novel drugs that cater to the demands of precision medicine. Accurately predicting molecular properties remains a fundamental challenge in drug discovery and development. Currently, a plethora of computer-aided drug discovery (CADD) methods have been widely employed in the field of molecular prediction. However, most of these methods primarily analyze molecules using low-dimensional representations such as SMILES notations, molecular fingerprints, and molecular graph-based descriptors. Only a few approaches have focused on incorporating and utilizing high-dimensional spatial structural representations of molecules. In light of the advancements in artificial intelligence, we introduce a 3D graph-spatial co-representation model called AEGNN-M, which combines two graph neural networks, GAT and EGNN. AEGNN-M enables learning of information from both molecular graphs representations and 3D spatial structural representations to predict molecular properties accurately. We conducted experiments on seven public datasets, three regression datasets and 14 breast cancer cell line phenotype screening datasets, comparing the performance of AEGNN-M with state-of-the-art deep learning methods. Extensive experimental results demonstrate the satisfactory performance of the AEGNN-M model. Furthermore, we analyzed the performance impact of different modules within AEGNN-M and the influence of spatial structural representations on the model's performance. The interpretability analysis also revealed the significance of specific atoms in determining particular molecular properties.
    Language English
    Publishing date 2024-02-22
    Publishing country United States
    Document type Journal Article
    ZDB-ID 2695320-1
    ISSN 2168-2208 ; 2168-2194
    ISSN (online) 2168-2208
    ISSN 2168-2194
    DOI 10.1109/JBHI.2024.3368608
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Enhancing drug-food interaction prediction with precision representations through multilevel self-supervised learning.

    Wei, Jinhang / Li, Zhen / Zhuo, Linlin / Fu, Xiangzheng / Wang, Mingjing / Li, Keqin / Chen, Chengshui

    Computers in biology and medicine

    2024  Volume 171, Page(s) 108104

    Abstract: Drug-food interactions (DFIs) crucially impact patient safety and drug efficacy by modifying absorption, distribution, metabolism, and excretion. The application of deep learning for predicting DFIs is promising, yet the development of computational ... ...

    Abstract Drug-food interactions (DFIs) crucially impact patient safety and drug efficacy by modifying absorption, distribution, metabolism, and excretion. The application of deep learning for predicting DFIs is promising, yet the development of computational models remains in its early stages. This is mainly due to the complexity of food compounds, challenging dataset developers in acquiring comprehensive ingredient data, often resulting in incomplete or vague food component descriptions. DFI-MS tackles this issue by employing an accurate feature representation method alongside a refined computational model. It innovatively achieves a more precise characterization of food features, a previously daunting task in DFI research. This is accomplished through modules designed for perturbation interactions, feature alignment and domain separation, and inference feedback. These modules extract essential information from features, using a perturbation module and a feature interaction encoder to establish robust representations. The feature alignment and domain separation modules are particularly effective in managing data with diverse frequencies and characteristics. DFI-MS stands out as the first in its field to combine data augmentation, feature alignment, domain separation, and contrastive learning. The flexibility of the inference feedback module allows its application in various downstream tasks. Demonstrating exceptional performance across multiple datasets, DFI-MS represents a significant advancement in food presentations technology. Our code and data are available at https://github.com/kkkayle/DFI-MS.
    MeSH term(s) Humans ; Food-Drug Interactions ; Food ; Supervised Machine Learning
    Language English
    Publishing date 2024-02-06
    Publishing country United States
    Document type Journal Article
    ZDB-ID 127557-4
    ISSN 1879-0534 ; 0010-4825
    ISSN (online) 1879-0534
    ISSN 0010-4825
    DOI 10.1016/j.compbiomed.2024.108104
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Diff-AMP: tailored designed antimicrobial peptide framework with all-in-one generation, identification, prediction and optimization.

    Wang, Rui / Wang, Tao / Zhuo, Linlin / Wei, Jinhang / Fu, Xiangzheng / Zou, Quan / Yao, Xiaojun

    Briefings in bioinformatics

    2024  Volume 25, Issue 2

    Abstract: Antimicrobial peptides (AMPs), short peptides with diverse functions, effectively target and combat various organisms. The widespread misuse of chemical antibiotics has led to increasing microbial resistance. Due to their low drug resistance and toxicity, ...

    Abstract Antimicrobial peptides (AMPs), short peptides with diverse functions, effectively target and combat various organisms. The widespread misuse of chemical antibiotics has led to increasing microbial resistance. Due to their low drug resistance and toxicity, AMPs are considered promising substitutes for traditional antibiotics. While existing deep learning technology enhances AMP generation, it also presents certain challenges. Firstly, AMP generation overlooks the complex interdependencies among amino acids. Secondly, current models fail to integrate crucial tasks like screening, attribute prediction and iterative optimization. Consequently, we develop a integrated deep learning framework, Diff-AMP, that automates AMP generation, identification, attribute prediction and iterative optimization. We innovatively integrate kinetic diffusion and attention mechanisms into the reinforcement learning framework for efficient AMP generation. Additionally, our prediction module incorporates pre-training and transfer learning strategies for precise AMP identification and screening. We employ a convolutional neural network for multi-attribute prediction and a reinforcement learning-based iterative optimization strategy to produce diverse AMPs. This framework automates molecule generation, screening, attribute prediction and optimization, thereby advancing AMP research. We have also deployed Diff-AMP on a web server, with code, data and server details available in the Data Availability section.
    MeSH term(s) Antimicrobial Peptides ; Amino Acids ; Anti-Bacterial Agents ; Diffusion ; Kinetics
    Chemical Substances Antimicrobial Peptides ; Amino Acids ; Anti-Bacterial Agents
    Language English
    Publishing date 2024-03-06
    Publishing country England
    Document type Journal Article
    ZDB-ID 2068142-2
    ISSN 1477-4054 ; 1467-5463
    ISSN (online) 1477-4054
    ISSN 1467-5463
    DOI 10.1093/bib/bbae078
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Discovery of Novel Noncovalent KRAS G12D Inhibitors through Structure-Based Virtual Screening and Molecular Dynamics Simulations.

    Du, Zhenya / Tu, Gao / Gong, Yaguo / Fu, Xiangzheng / Wu, Qibiao / Long, Guankui

    Molecules (Basel, Switzerland)

    2024  Volume 29, Issue 6

    Abstract: The development of effective inhibitors targeting the Kirsten rat sarcoma viral proto-oncogene ( ... ...

    Abstract The development of effective inhibitors targeting the Kirsten rat sarcoma viral proto-oncogene (KRAS
    MeSH term(s) Humans ; Molecular Dynamics Simulation ; Proto-Oncogene Proteins p21(ras)/genetics ; Molecular Docking Simulation ; Mutation ; Neoplasms
    Chemical Substances Proto-Oncogene Proteins p21(ras) (EC 3.6.5.2) ; KRAS protein, human
    Language English
    Publishing date 2024-03-10
    Publishing country Switzerland
    Document type Journal Article
    ZDB-ID 1413402-0
    ISSN 1420-3049 ; 1431-5165 ; 1420-3049
    ISSN (online) 1420-3049
    ISSN 1431-5165 ; 1420-3049
    DOI 10.3390/molecules29061229
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: Revisiting Drug-Protein Interaction Prediction: A Novel Global-Local Perspective.

    Zhou, Zhecheng / Liao, Qingquan / Wei, Jinhang / Zhuo, Linlin / Wu, Xiaonan / Fu, Xiangzheng / Zou, Quan

    Bioinformatics (Oxford, England)

    2024  

    Abstract: Motivation: Accurate inference of potential Drug-protein interactions (DPIs) aids in understanding drug mechanisms and developing novel treatments. Existing deep learning models, however, struggle with accurate node representation in DPI prediction, ... ...

    Abstract Motivation: Accurate inference of potential Drug-protein interactions (DPIs) aids in understanding drug mechanisms and developing novel treatments. Existing deep learning models, however, struggle with accurate node representation in DPI prediction, limiting their performance.
    Results: We propose a new computational framework that integrates global and local features of nodes in the drug-protein bipartite graph for efficient DPI inference. Initially, we employ pre-trained models to acquire fundamental knowledge of drugs and proteins and to determine their initial features. Subsequently, the MinHash and HyperLogLog algorithms are utilized to estimate the similarity and set cardinality between drug and protein subgraphs, serving as their local features. Then, an energy-constrained diffusion mechanism is integrated into the transformer architecture, capturing interdependencies between nodes in the drug-protein bipartite graph and extracting their global features. Finally, we fuse the local and global features of nodes and employ multi-layer perceptrons (MLPs) to predict the likelihood of potential DPIs. A comprehensive and precise node representation guarantees efficient prediction of unknown DPIs by the model. Various experiments validate the accuracy and reliability of our model, with molecular docking results revealing its capability to identify potential DPIs not present in existing databases. This approach are expected to offer valuable insights for furthering drug repurposing and personalized medicine research.
    Availability and implementation: Our code and data are accessible at: https://github.com/ZZCrazy00/DPI.
    Supplementary information: Supplementary data are available at Bioinformatics online.
    Language English
    Publishing date 2024-04-22
    Publishing country England
    Document type Journal Article
    ZDB-ID 1422668-6
    ISSN 1367-4811 ; 1367-4803
    ISSN (online) 1367-4811
    ISSN 1367-4803
    DOI 10.1093/bioinformatics/btae271
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top