首页 > 最新文献

Bioinformatics最新文献

英文 中文
scAce: an adaptive embedding and clustering method for single-cell gene expression data. scAce:单细胞基因表达数据的自适应嵌入和聚类方法。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2023-09-02 DOI: 10.1093/bioinformatics/btad546
Xinwei He, Kun Qian, Ziqian Wang, Shirou Zeng, Hongwei Li, Wei Vivian Li

Motivation: Since the development of single-cell RNA sequencing (scRNA-seq) technologies, clustering analysis of single-cell gene expression data has been an essential tool for distinguishing cell types and identifying novel cell types. Even though many methods have been available for scRNA-seq clustering analysis, the majority of them are constrained by the requirement on predetermined cluster numbers or the dependence on selected initial cluster assignment.

Results: In this article, we propose an adaptive embedding and clustering method named scAce, which constructs a variational autoencoder to simultaneously learn cell embeddings and cluster assignments. In the scAce method, we develop an adaptive cluster merging approach which achieves improved clustering results without the need to estimate the number of clusters in advance. In addition, scAce provides an option to perform clustering enhancement, which can update and enhance cluster assignments based on previous clustering results from other methods. Based on computational analysis of both simulated and real datasets, we demonstrate that scAce outperforms state-of-the-art clustering methods for scRNA-seq data, and achieves better clustering accuracy and robustness.

Availability and implementation: The scAce package is implemented in python 3.8 and is freely available from https://github.com/sldyns/scAce.

动因:自单细胞RNA测序(scRNA-seq)技术发展以来,单细胞基因表达数据的聚类分析一直是区分细胞类型和识别新型细胞类型的重要工具。尽管目前已有许多用于 scRNA-seq 聚类分析的方法,但大多数方法都受限于对预定聚类数量的要求或对选定的初始聚类分配的依赖:在本文中,我们提出了一种名为 scAce 的自适应嵌入和聚类方法,它构建了一个变异自动编码器来同时学习细胞嵌入和聚类分配。在 scAce 方法中,我们开发了一种自适应聚类合并方法,无需提前估计聚类数量,就能获得更好的聚类结果。此外,scAce 还提供了执行聚类增强的选项,可以根据其他方法的聚类结果更新和增强聚类分配。基于对模拟数据集和真实数据集的计算分析,我们证明了在 scRNA-seq 数据方面,scAce 优于最先进的聚类方法,并实现了更好的聚类准确性和鲁棒性:scAce 软件包由 python 3.8 实现,可从 https://github.com/sldyns/scAce 免费获取。
{"title":"scAce: an adaptive embedding and clustering method for single-cell gene expression data.","authors":"Xinwei He, Kun Qian, Ziqian Wang, Shirou Zeng, Hongwei Li, Wei Vivian Li","doi":"10.1093/bioinformatics/btad546","DOIUrl":"10.1093/bioinformatics/btad546","url":null,"abstract":"<p><strong>Motivation: </strong>Since the development of single-cell RNA sequencing (scRNA-seq) technologies, clustering analysis of single-cell gene expression data has been an essential tool for distinguishing cell types and identifying novel cell types. Even though many methods have been available for scRNA-seq clustering analysis, the majority of them are constrained by the requirement on predetermined cluster numbers or the dependence on selected initial cluster assignment.</p><p><strong>Results: </strong>In this article, we propose an adaptive embedding and clustering method named scAce, which constructs a variational autoencoder to simultaneously learn cell embeddings and cluster assignments. In the scAce method, we develop an adaptive cluster merging approach which achieves improved clustering results without the need to estimate the number of clusters in advance. In addition, scAce provides an option to perform clustering enhancement, which can update and enhance cluster assignments based on previous clustering results from other methods. Based on computational analysis of both simulated and real datasets, we demonstrate that scAce outperforms state-of-the-art clustering methods for scRNA-seq data, and achieves better clustering accuracy and robustness.</p><p><strong>Availability and implementation: </strong>The scAce package is implemented in python 3.8 and is freely available from https://github.com/sldyns/scAce.</p>","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2023-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10500084/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10649377","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Automated machine learning for genome wide association studies. 用于全基因组关联研究的自动化机器学习。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2023-09-02 DOI: 10.1093/bioinformatics/btad545
Kleanthi Lakiotaki, Zaharias Papadovasilakis, Vincenzo Lagani, Stefanos Fafalios, Paulos Charonyktakis, Michail Tsagris, Ioannis Tsamardinos

Motivation: Genome-wide association studies (GWAS) present several computational and statistical challenges for their data analysis, including knowledge discovery, interpretability, and translation to clinical practice.

Results: We develop, apply, and comparatively evaluate an automated machine learning (AutoML) approach, customized for genomic data that delivers reliable predictive and diagnostic models, the set of genetic variants that are important for predictions (called a biosignature), and an estimate of the out-of-sample predictive power. This AutoML approach discovers variants with higher predictive performance compared to standard GWAS methods, computes an individual risk prediction score, generalizes to new, unseen data, is shown to better differentiate causal variants from other highly correlated variants, and enhances knowledge discovery and interpretability by reporting multiple equivalent biosignatures.

Availability and implementation: Code for this study is available at: https://github.com/mensxmachina/autoML-GWAS. JADBio offers a free version at: https://jadbio.com/sign-up/. SNP data can be downloaded from the EGA repository (https://ega-archive.org/). PRS data are found at: https://www.aicrowd.com/challenges/opensnp-height-prediction. Simulation data to study population structure can be found at: https://easygwas.ethz.ch/data/public/dataset/view/1/.

动机:全基因组关联研究(GWAS)对其数据分析提出了一些计算和统计挑战,包括知识发现、可解释性和转化为临床实践。结果:我们开发、应用并比较评估了一种自动机器学习(AutoML)方法,该方法是为提供可靠预测和诊断模型的基因组数据定制的,对预测很重要的一组遗传变异(称为生物信号),以及对样本外预测能力的估计。与标准GWAS方法相比,这种AutoML方法发现具有更高预测性能的变体,计算个人风险预测得分,推广到新的、看不见的数据,被证明可以更好地区分因果变体和其他高度相关的变体,并通过报告多个等效的生物特征来增强知识发现和可解释性。可用性和实施:本研究的代码可在:https://github.com/mensxmachina/autoML-GWAS.JADBio提供免费版本,网址为:https://jadbio.com/sign-up/.SNP数据可从EGA存储库下载(https://ega-archive.org/)。PRS数据位于:https://www.aicrowd.com/challenges/opensnp-height-prediction.研究人口结构的模拟数据可在以下网址找到:https://easygwas.ethz.ch/data/public/dataset/view/1/.
{"title":"Automated machine learning for genome wide association studies.","authors":"Kleanthi Lakiotaki, Zaharias Papadovasilakis, Vincenzo Lagani, Stefanos Fafalios, Paulos Charonyktakis, Michail Tsagris, Ioannis Tsamardinos","doi":"10.1093/bioinformatics/btad545","DOIUrl":"10.1093/bioinformatics/btad545","url":null,"abstract":"<p><strong>Motivation: </strong>Genome-wide association studies (GWAS) present several computational and statistical challenges for their data analysis, including knowledge discovery, interpretability, and translation to clinical practice.</p><p><strong>Results: </strong>We develop, apply, and comparatively evaluate an automated machine learning (AutoML) approach, customized for genomic data that delivers reliable predictive and diagnostic models, the set of genetic variants that are important for predictions (called a biosignature), and an estimate of the out-of-sample predictive power. This AutoML approach discovers variants with higher predictive performance compared to standard GWAS methods, computes an individual risk prediction score, generalizes to new, unseen data, is shown to better differentiate causal variants from other highly correlated variants, and enhances knowledge discovery and interpretability by reporting multiple equivalent biosignatures.</p><p><strong>Availability and implementation: </strong>Code for this study is available at: https://github.com/mensxmachina/autoML-GWAS. JADBio offers a free version at: https://jadbio.com/sign-up/. SNP data can be downloaded from the EGA repository (https://ega-archive.org/). PRS data are found at: https://www.aicrowd.com/challenges/opensnp-height-prediction. Simulation data to study population structure can be found at: https://easygwas.ethz.ch/data/public/dataset/view/1/.</p>","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2023-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10562960/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10161763","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Accessibility of covariance information creates vulnerability in Federated Learning frameworks. 协方差信息的可访问性在联合学习框架中造成漏洞。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2023-09-02 DOI: 10.1093/bioinformatics/btad531
Manuel Huth, Jonas Arruda, Roy Gusinow, Lorenzo Contento, Evelina Tacconelli, Jan Hasenauer

Motivation: Federated Learning (FL) is gaining traction in various fields as it enables integrative data analysis without sharing sensitive data, such as in healthcare. However, the risk of data leakage caused by malicious attacks must be considered. In this study, we introduce a novel attack algorithm that relies on being able to compute sample means, sample covariances, and construct known linearly independent vectors on the data owner side.

Results: We show that these basic functionalities, which are available in several established FL frameworks, are sufficient to reconstruct privacy-protected data. Additionally, the attack algorithm is robust to defense strategies that involve adding random noise. We demonstrate the limitations of existing frameworks and propose potential defense strategies analyzing the implications of using differential privacy. The novel insights presented in this study will aid in the improvement of FL frameworks.

Availability and implementation: The code examples are provided at GitHub (https://github.com/manuhuth/Data-Leakage-From-Covariances.git). The CNSIM1 dataset, which we used in the manuscript, is available within the DSData R package (https://github.com/datashield/DSData/tree/main/data).

动机:联合学习(FL)在各个领域都越来越受欢迎,因为它能够在不共享敏感数据的情况下进行综合数据分析,例如在医疗保健领域。但是,必须考虑恶意攻击导致数据泄露的风险。在这项研究中,我们介绍了一种新的攻击算法,该算法依赖于能够计算样本均值、样本协方差,并在数据所有者侧构造已知的线性无关向量。结果:我们表明,这些基本功能在几个已建立的FL框架中可用,足以重建受隐私保护的数据。此外,该攻击算法对涉及添加随机噪声的防御策略是鲁棒的。我们展示了现有框架的局限性,并提出了潜在的防御策略,分析了使用差异隐私的含义。本研究中提出的新见解将有助于FL框架的改进。可用性和实现:代码示例在GitHub上提供(https://github.com/manuhuth/Data-Leakage-From-Covariances.git)。我们在手稿中使用的CNSIM1数据集在DSData R包中可用(https://github.com/datashield/DSData/tree/main/data)。
{"title":"Accessibility of covariance information creates vulnerability in Federated Learning frameworks.","authors":"Manuel Huth,&nbsp;Jonas Arruda,&nbsp;Roy Gusinow,&nbsp;Lorenzo Contento,&nbsp;Evelina Tacconelli,&nbsp;Jan Hasenauer","doi":"10.1093/bioinformatics/btad531","DOIUrl":"10.1093/bioinformatics/btad531","url":null,"abstract":"<p><strong>Motivation: </strong>Federated Learning (FL) is gaining traction in various fields as it enables integrative data analysis without sharing sensitive data, such as in healthcare. However, the risk of data leakage caused by malicious attacks must be considered. In this study, we introduce a novel attack algorithm that relies on being able to compute sample means, sample covariances, and construct known linearly independent vectors on the data owner side.</p><p><strong>Results: </strong>We show that these basic functionalities, which are available in several established FL frameworks, are sufficient to reconstruct privacy-protected data. Additionally, the attack algorithm is robust to defense strategies that involve adding random noise. We demonstrate the limitations of existing frameworks and propose potential defense strategies analyzing the implications of using differential privacy. The novel insights presented in this study will aid in the improvement of FL frameworks.</p><p><strong>Availability and implementation: </strong>The code examples are provided at GitHub (https://github.com/manuhuth/Data-Leakage-From-Covariances.git). The CNSIM1 dataset, which we used in the manuscript, is available within the DSData R package (https://github.com/datashield/DSData/tree/main/data).</p>","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2023-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10516515/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10176920","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
GraphCpG: imputation of single-cell methylomes based on locus-aware neighboring subgraphs. GraphCpG:基于位点感知相邻子图的单细胞甲基组插补。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2023-09-02 DOI: 10.1093/bioinformatics/btad533
Yuzhong Deng, Jianxiong Tang, Jiyang Zhang, Jianxiao Zou, Que Zhu, Shicai Fan

Motivation: Single-cell DNA methylation sequencing can assay DNA methylation at single-cell resolution. However, incomplete coverage compromises related downstream analyses, outlining the importance of imputation techniques. With a rising number of cell samples in recent large datasets, scalable and efficient imputation models are critical to addressing the sparsity for genome-wide analyses.

Results: We proposed a novel graph-based deep learning approach to impute methylation matrices based on locus-aware neighboring subgraphs with locus-aware encoding orienting on one cell type. Merely using the CpGs methylation matrix, the obtained GraphCpG outperforms previous methods on datasets containing more than hundreds of cells and achieves competitive performance on smaller datasets, with subgraphs of predicted sites visualized by retrievable bipartite graphs. Besides better imputation performance with increasing cell number, it significantly reduces computation time and demonstrates improvement in downstream analysis.

Availability and implementation: The source code is freely available at https://github.com/yuzhong-deng/graphcpg.git.

动机:单细胞DNA甲基化测序可以以单细胞分辨率测定DNA甲基化。然而,不完全覆盖损害了相关的下游分析,概述了插补技术的重要性。随着最近大型数据集中细胞样本数量的增加,可扩展和高效的插补模型对于解决全基因组分析的稀疏性至关重要。结果:我们提出了一种新的基于图的深度学习方法,以基于位点感知相邻子图的甲基化矩阵,其中位点感知编码面向一种细胞类型。仅使用CpGs甲基化矩阵,所获得的GraphCpG在包含数百个以上细胞的数据集上优于以前的方法,并在较小的数据集中实现了竞争性能,预测位点的子图通过可检索的二分图可视化。除了随着细胞数量的增加而获得更好的插补性能外,它还显著减少了计算时间,并证明了下游分析的改进。可用性和实现:源代码可在https://github.com/yuzhong-deng/graphcpg.git.
{"title":"GraphCpG: imputation of single-cell methylomes based on locus-aware neighboring subgraphs.","authors":"Yuzhong Deng,&nbsp;Jianxiong Tang,&nbsp;Jiyang Zhang,&nbsp;Jianxiao Zou,&nbsp;Que Zhu,&nbsp;Shicai Fan","doi":"10.1093/bioinformatics/btad533","DOIUrl":"10.1093/bioinformatics/btad533","url":null,"abstract":"<p><strong>Motivation: </strong>Single-cell DNA methylation sequencing can assay DNA methylation at single-cell resolution. However, incomplete coverage compromises related downstream analyses, outlining the importance of imputation techniques. With a rising number of cell samples in recent large datasets, scalable and efficient imputation models are critical to addressing the sparsity for genome-wide analyses.</p><p><strong>Results: </strong>We proposed a novel graph-based deep learning approach to impute methylation matrices based on locus-aware neighboring subgraphs with locus-aware encoding orienting on one cell type. Merely using the CpGs methylation matrix, the obtained GraphCpG outperforms previous methods on datasets containing more than hundreds of cells and achieves competitive performance on smaller datasets, with subgraphs of predicted sites visualized by retrievable bipartite graphs. Besides better imputation performance with increasing cell number, it significantly reduces computation time and demonstrates improvement in downstream analysis.</p><p><strong>Availability and implementation: </strong>The source code is freely available at https://github.com/yuzhong-deng/graphcpg.git.</p>","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2023-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10516632/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10121589","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction to: Optimal adjustment sets for causal query estimation in partially observed biomolecular networks. 修正:部分观察到的生物分子网络中因果查询估计的最优调整集。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2023-09-02 DOI: 10.1093/bioinformatics/btad559
This is a correction to: Sara Mohammad-Taheri and others, Optimal adjustment sets for causal query estimation in partially observed biomolecular networks, Bioinformatics, Volume 39, Issue Supplement_1, June 2023, Pages i494–i503, https://doi. org/10.1093/bioinformatics/btad270 In the originally published version of this manuscript, the sixth author’s name was incorrectly spelled as Charles Taply Hoyt. It should be Charles Tapley Hoyt.
{"title":"Correction to: Optimal adjustment sets for causal query estimation in partially observed biomolecular networks.","authors":"","doi":"10.1093/bioinformatics/btad559","DOIUrl":"https://doi.org/10.1093/bioinformatics/btad559","url":null,"abstract":"This is a correction to: Sara Mohammad-Taheri and others, Optimal adjustment sets for causal query estimation in partially observed biomolecular networks, Bioinformatics, Volume 39, Issue Supplement_1, June 2023, Pages i494–i503, https://doi. org/10.1093/bioinformatics/btad270 In the originally published version of this manuscript, the sixth author’s name was incorrectly spelled as Charles Taply Hoyt. It should be Charles Tapley Hoyt.","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2023-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10500085/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10262499","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
DrForna: visualization of cotranscriptional folding. 共同转录折叠的可视化。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2023-09-02 DOI: 10.1093/bioinformatics/btad555
Anda Ramona Tănasie, Peter Kerpedjiev, Stefan Hammer, Stefan Badelt

Motivation: Understanding RNA folding at the level of secondary structures can give important insights concerning the function of a molecule. We are interested to learn how secondary structures change dynamically during transcription, as well as whether particular secondary structures form already during or only after transcription. While different approaches exist to simulate cotranscriptional folding, the current strategies for visualization are lagging behind. New, more suitable approaches are necessary to help with exploring the generated data from cotranscriptional folding simulations.

Results: We present DrForna, an interactive visualization app for viewing the time course of a cotranscriptional RNA folding simulation. Specifically, users can scroll along the time axis and see the population of structures that are present at any particular time point.

Availability and implementation: DrForna is a JavaScript project available on Github at https://github.com/ViennaRNA/drforna and deployed at https://viennarna.github.io/drforna.

动机:在二级结构水平上理解RNA折叠可以提供有关分子功能的重要见解。我们有兴趣了解二级结构在转录过程中是如何动态变化的,以及特定的二级结构是在转录过程中已经形成还是在转录之后才形成。虽然存在不同的方法来模拟共转录折叠,但目前的可视化策略是滞后的。新的,更合适的方法是必要的,以帮助探索从共转录折叠模拟生成的数据。结果:我们提出了DrForna,一个交互式可视化应用程序,用于查看共转录RNA折叠模拟的时间过程。具体来说,用户可以沿着时间轴滚动,查看在任何特定时间点出现的结构的总体。可用性和实现:DrForna是一个JavaScript项目,可以在Github上获得https://github.com/ViennaRNA/drforna,部署在https://viennarna.github.io/drforna。
{"title":"DrForna: visualization of cotranscriptional folding.","authors":"Anda Ramona Tănasie,&nbsp;Peter Kerpedjiev,&nbsp;Stefan Hammer,&nbsp;Stefan Badelt","doi":"10.1093/bioinformatics/btad555","DOIUrl":"https://doi.org/10.1093/bioinformatics/btad555","url":null,"abstract":"<p><strong>Motivation: </strong>Understanding RNA folding at the level of secondary structures can give important insights concerning the function of a molecule. We are interested to learn how secondary structures change dynamically during transcription, as well as whether particular secondary structures form already during or only after transcription. While different approaches exist to simulate cotranscriptional folding, the current strategies for visualization are lagging behind. New, more suitable approaches are necessary to help with exploring the generated data from cotranscriptional folding simulations.</p><p><strong>Results: </strong>We present DrForna, an interactive visualization app for viewing the time course of a cotranscriptional RNA folding simulation. Specifically, users can scroll along the time axis and see the population of structures that are present at any particular time point.</p><p><strong>Availability and implementation: </strong>DrForna is a JavaScript project available on Github at https://github.com/ViennaRNA/drforna and deployed at https://viennarna.github.io/drforna.</p>","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2023-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10504468/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10357864","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Phylogenetic inference using generative adversarial networks. 使用生成对抗网络的系统发育推理。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2023-09-02 DOI: 10.1093/bioinformatics/btad543
Megan L Smith, Matthew W Hahn

Motivation: The application of machine learning approaches in phylogenetics has been impeded by the vast model space associated with inference. Supervised machine learning approaches require data from across this space to train models. Because of this, previous approaches have typically been limited to inferring relationships among unrooted quartets of taxa, where there are only three possible topologies. Here, we explore the potential of generative adversarial networks (GANs) to address this limitation. GANs consist of a generator and a discriminator: at each step, the generator aims to create data that is similar to real data, while the discriminator attempts to distinguish generated and real data. By using an evolutionary model as the generator, we use GANs to make evolutionary inferences. Since a new model can be considered at each iteration, heuristic searches of complex model spaces are possible. Thus, GANs offer a potential solution to the challenges of applying machine learning in phylogenetics.

Results: We developed phyloGAN, a GAN that infers phylogenetic relationships among species. phyloGAN takes as input a concatenated alignment, or a set of gene alignments, and infers a phylogenetic tree either considering or ignoring gene tree heterogeneity. We explored the performance of phyloGAN for up to 15 taxa in the concatenation case and 6 taxa when considering gene tree heterogeneity. Error rates are relatively low in these simple cases. However, run times are slow and performance metrics suggest issues during training. Future work should explore novel architectures that may result in more stable and efficient GANs for phylogenetics.

Availability and implementation: phyloGAN is available on github: https://github.com/meganlsmith/phyloGAN/.

动机:与推理相关的巨大模型空间阻碍了机器学习方法在系统发育中的应用。监督式机器学习方法需要来自整个领域的数据来训练模型。正因为如此,以前的方法通常仅限于推断分类群的无根四分之一之间的关系,其中只有三种可能的拓扑结构。在这里,我们探索生成对抗网络(gan)解决这一限制的潜力。gan由生成器和鉴别器组成:在每一步中,生成器的目标是创建与真实数据相似的数据,而鉴别器则试图区分生成的数据和真实数据。通过使用进化模型作为生成器,我们使用gan进行进化推理。由于每次迭代都可以考虑一个新的模型,因此可以对复杂模型空间进行启发式搜索。因此,gan为在系统发育学中应用机器学习的挑战提供了一个潜在的解决方案。结果:我们开发了phyloGAN,这是一个推断物种之间系统发育关系的GAN。phyloGAN将一个串联的比对或一组基因比对作为输入,并推断出考虑或忽略基因树异质性的系统发育树。在基因树异质性的情况下,我们探索了多达15个分类群和6个分类群的phyloGAN的性能。在这些简单的情况下,错误率相对较低。然而,运行时间很慢,性能指标在训练期间显示出问题。未来的工作应该探索新的架构,可能导致更稳定和高效的系统发育gan。可用性和实现:phyloGAN可在github上获得:https://github.com/meganlsmith/phyloGAN/。
{"title":"Phylogenetic inference using generative adversarial networks.","authors":"Megan L Smith,&nbsp;Matthew W Hahn","doi":"10.1093/bioinformatics/btad543","DOIUrl":"https://doi.org/10.1093/bioinformatics/btad543","url":null,"abstract":"<p><strong>Motivation: </strong>The application of machine learning approaches in phylogenetics has been impeded by the vast model space associated with inference. Supervised machine learning approaches require data from across this space to train models. Because of this, previous approaches have typically been limited to inferring relationships among unrooted quartets of taxa, where there are only three possible topologies. Here, we explore the potential of generative adversarial networks (GANs) to address this limitation. GANs consist of a generator and a discriminator: at each step, the generator aims to create data that is similar to real data, while the discriminator attempts to distinguish generated and real data. By using an evolutionary model as the generator, we use GANs to make evolutionary inferences. Since a new model can be considered at each iteration, heuristic searches of complex model spaces are possible. Thus, GANs offer a potential solution to the challenges of applying machine learning in phylogenetics.</p><p><strong>Results: </strong>We developed phyloGAN, a GAN that infers phylogenetic relationships among species. phyloGAN takes as input a concatenated alignment, or a set of gene alignments, and infers a phylogenetic tree either considering or ignoring gene tree heterogeneity. We explored the performance of phyloGAN for up to 15 taxa in the concatenation case and 6 taxa when considering gene tree heterogeneity. Error rates are relatively low in these simple cases. However, run times are slow and performance metrics suggest issues during training. Future work should explore novel architectures that may result in more stable and efficient GANs for phylogenetics.</p><p><strong>Availability and implementation: </strong>phyloGAN is available on github: https://github.com/meganlsmith/phyloGAN/.</p>","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2023-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10500083/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10631514","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 3
DeepMHCI: an anchor position-aware deep interaction model for accurate MHC-I peptide binding affinity prediction. DeepMHCI:一个锚定位置感知的深度相互作用模型,用于准确预测MHC-I肽结合亲和力。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2023-09-02 DOI: 10.1093/bioinformatics/btad551
Wei Qu, Ronghui You, Hiroshi Mamitsuka, Shanfeng Zhu

Motivation: Computationally predicting major histocompatibility complex class I (MHC-I) peptide binding affinity is an important problem in immunological bioinformatics, which is also crucial for the identification of neoantigens for personalized therapeutic cancer vaccines. Recent cutting-edge deep learning-based methods for this problem cannot achieve satisfactory performance, especially for non-9-mer peptides. This is because such methods generate the input by simply concatenating the two given sequences: a peptide and (the pseudo sequence of) an MHC class I molecule, which cannot precisely capture the anchor positions of the MHC binding motif for the peptides with variable lengths. We thus developed an anchor position-aware and high-performance deep model, DeepMHCI, with a position-wise gated layer and a residual binding interaction convolution layer. This allows the model to control the information flow in peptides to be aware of anchor positions and model the interactions between peptides and the MHC pseudo (binding) sequence directly with multiple convolutional kernels.

Results: The performance of DeepMHCI has been thoroughly validated by extensive experiments on four benchmark datasets under various settings, such as 5-fold cross-validation, validation with the independent testing set, external HPV vaccine identification, and external CD8+ epitope identification. Experimental results with visualization of binding motifs demonstrate that DeepMHCI outperformed all competing methods, especially on non-9-mer peptides binding prediction.

Availability and implementation: DeepMHCI is publicly available at https://github.com/ZhuLab-Fudan/DeepMHCI.

动机:计算预测主要组织相容性复合物I类(MHC-I)肽结合亲和力是免疫学生物信息学中的一个重要问题,这对于鉴定用于个性化治疗性癌症疫苗的新抗原也至关重要。针对这一问题,最近基于深度学习的前沿方法无法获得令人满意的性能,尤其是对于非9-聚体肽。这是因为这种方法通过简单地连接两个给定的序列来产生输入:肽和MHC I类分子的(伪序列),这不能精确地捕捉可变长度肽的MHC结合基序的锚定位置。因此,我们开发了一个锚位置感知和高性能的深度模型DeepMHCI,该模型具有位置门控层和残余结合相互作用卷积层。这允许该模型控制肽中的信息流以了解锚定位置,并直接用多个卷积核对肽和MHC伪(结合)序列之间的相互作用进行建模。结果:DeepMHCI的性能已通过在四个基准数据集上进行的广泛实验在各种设置下得到了彻底验证,如5倍交叉验证、独立测试集验证、外部HPV疫苗鉴定和外部CD8+表位鉴定。结合基序可视化的实验结果表明,DeepMHCI优于所有竞争方法,尤其是在非9-聚体肽结合预测方面。可用性和实施:DeepMHCI可在https://github.com/ZhuLab-Fudan/DeepMHCI.
{"title":"DeepMHCI: an anchor position-aware deep interaction model for accurate MHC-I peptide binding affinity prediction.","authors":"Wei Qu,&nbsp;Ronghui You,&nbsp;Hiroshi Mamitsuka,&nbsp;Shanfeng Zhu","doi":"10.1093/bioinformatics/btad551","DOIUrl":"10.1093/bioinformatics/btad551","url":null,"abstract":"<p><strong>Motivation: </strong>Computationally predicting major histocompatibility complex class I (MHC-I) peptide binding affinity is an important problem in immunological bioinformatics, which is also crucial for the identification of neoantigens for personalized therapeutic cancer vaccines. Recent cutting-edge deep learning-based methods for this problem cannot achieve satisfactory performance, especially for non-9-mer peptides. This is because such methods generate the input by simply concatenating the two given sequences: a peptide and (the pseudo sequence of) an MHC class I molecule, which cannot precisely capture the anchor positions of the MHC binding motif for the peptides with variable lengths. We thus developed an anchor position-aware and high-performance deep model, DeepMHCI, with a position-wise gated layer and a residual binding interaction convolution layer. This allows the model to control the information flow in peptides to be aware of anchor positions and model the interactions between peptides and the MHC pseudo (binding) sequence directly with multiple convolutional kernels.</p><p><strong>Results: </strong>The performance of DeepMHCI has been thoroughly validated by extensive experiments on four benchmark datasets under various settings, such as 5-fold cross-validation, validation with the independent testing set, external HPV vaccine identification, and external CD8+ epitope identification. Experimental results with visualization of binding motifs demonstrate that DeepMHCI outperformed all competing methods, especially on non-9-mer peptides binding prediction.</p><p><strong>Availability and implementation: </strong>DeepMHCI is publicly available at https://github.com/ZhuLab-Fudan/DeepMHCI.</p>","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2023-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10516514/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10217795","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
An extensive benchmark study on biomedical text generation and mining with ChatGPT. 利用ChatGPT对生物医学文本生成和挖掘进行了广泛的基准研究。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2023-09-02 DOI: 10.1093/bioinformatics/btad557
Qijie Chen, Haotong Sun, Haoyang Liu, Yinghui Jiang, Ting Ran, Xurui Jin, Xianglu Xiao, Zhimin Lin, Hongming Chen, Zhangmin Niu

Motivation: In recent years, the development of natural language process (NLP) technologies and deep learning hardware has led to significant improvement in large language models (LLMs). The ChatGPT, the state-of-the-art LLM built on GPT-3.5 and GPT-4, shows excellent capabilities in general language understanding and reasoning. Researchers also tested the GPTs on a variety of NLP-related tasks and benchmarks and got excellent results. With exciting performance on daily chat, researchers began to explore the capacity of ChatGPT on expertise that requires professional education for human and we are interested in the biomedical domain.

Results: To evaluate the performance of ChatGPT on biomedical-related tasks, this article presents a comprehensive benchmark study on the use of ChatGPT for biomedical corpus, including article abstracts, clinical trials description, biomedical questions, and so on. Typical NLP tasks like named entity recognization, relation extraction, sentence similarity, question and answering, and document classification are included. Overall, ChatGPT got a BLURB score of 58.50 while the state-of-the-art model had a score of 84.30. Through a series of experiments, we demonstrated the effectiveness and versatility of ChatGPT in biomedical text understanding, reasoning and generation, and the limitation of ChatGPT build on GPT-3.5.

Availability and implementation: All the datasets are available from BLURB benchmark https://microsoft.github.io/BLURB/index.html. The prompts are described in the article.

动机:近年来,自然语言处理(NLP)技术和深度学习硬件的发展导致了大型语言模型(LLM)的显著改进。ChatGPT是建立在GPT-3.5和GPT-4基础上的最先进的LLM,在一般语言理解和推理方面表现出出色的能力。研究人员还在各种与NLP相关的任务和基准测试中测试了GPT,并获得了优异的结果。随着在日常聊天中令人兴奋的表现,研究人员开始探索ChatGPT在需要对人类进行专业教育的专业知识方面的能力,我们对生物医学领域感兴趣。结果:为了评估ChatGPT在生物医学相关任务中的性能,本文对ChatGPT用于生物医学语料库进行了全面的基准研究,包括文章摘要、临床试验描述、生物医学问题等。典型的NLP任务包括命名实体识别、关系提取、句子相似性、问答,以及文档分类。总体而言,ChatGPT的BLURB得分为58.50,而最先进的模型得分为84.30。通过一系列实验,我们证明了ChatGPT在生物医学文本理解、推理和生成方面的有效性和通用性,以及基于GPT-3.5的ChatGPT的局限性。可用性和实现:所有数据集都可以从BLURB基准中获得https://microsoft.github.io/BLURB/index.html.文章中介绍了提示。
{"title":"An extensive benchmark study on biomedical text generation and mining with ChatGPT.","authors":"Qijie Chen,&nbsp;Haotong Sun,&nbsp;Haoyang Liu,&nbsp;Yinghui Jiang,&nbsp;Ting Ran,&nbsp;Xurui Jin,&nbsp;Xianglu Xiao,&nbsp;Zhimin Lin,&nbsp;Hongming Chen,&nbsp;Zhangmin Niu","doi":"10.1093/bioinformatics/btad557","DOIUrl":"10.1093/bioinformatics/btad557","url":null,"abstract":"<p><strong>Motivation: </strong>In recent years, the development of natural language process (NLP) technologies and deep learning hardware has led to significant improvement in large language models (LLMs). The ChatGPT, the state-of-the-art LLM built on GPT-3.5 and GPT-4, shows excellent capabilities in general language understanding and reasoning. Researchers also tested the GPTs on a variety of NLP-related tasks and benchmarks and got excellent results. With exciting performance on daily chat, researchers began to explore the capacity of ChatGPT on expertise that requires professional education for human and we are interested in the biomedical domain.</p><p><strong>Results: </strong>To evaluate the performance of ChatGPT on biomedical-related tasks, this article presents a comprehensive benchmark study on the use of ChatGPT for biomedical corpus, including article abstracts, clinical trials description, biomedical questions, and so on. Typical NLP tasks like named entity recognization, relation extraction, sentence similarity, question and answering, and document classification are included. Overall, ChatGPT got a BLURB score of 58.50 while the state-of-the-art model had a score of 84.30. Through a series of experiments, we demonstrated the effectiveness and versatility of ChatGPT in biomedical text understanding, reasoning and generation, and the limitation of ChatGPT build on GPT-3.5.</p><p><strong>Availability and implementation: </strong>All the datasets are available from BLURB benchmark https://microsoft.github.io/BLURB/index.html. The prompts are described in the article.</p>","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2023-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10562950/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10173923","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 2
MuTATE-an R package for comprehensive multi-objective molecular modeling. mutate -一个用于综合多目标分子建模的R包。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2023-09-02 DOI: 10.1093/bioinformatics/btad507
Sarah G Ayton, Víctor Treviño

Motivation: Comprehensive multi-omics studies have driven advances in disease modeling for effective precision medicine but pose a challenge for existing machine-learning approaches, which have limited interpretability across clinical endpoints. Automated, comprehensive disease modeling requires a machine-learning approach that can simultaneously identify disease subgroups and their defining molecular biomarkers by explaining multiple clinical endpoints. Current tools are restricted to individual endpoints or limited variable types, necessitate advanced computation skills, and require resource-intensive manual expert interpretation.

Results: We developed Multi-Target Automated Tree Engine (MuTATE) for automated and comprehensive molecular modeling, which enables user-friendly multi-objective decision tree construction and visualization of relationships between molecular biomarkers and patient subgroups characterized by multiple clinical endpoints. MuTATE incorporates multiple targets throughout model construction and allows for target weights, enabling construction of interpretable decision trees that provide insights into disease heterogeneity and molecular signatures. MuTATE eliminates the need for manual synthesis of multiple non-explainable models, making it highly efficient and accessible for bioinformaticians and clinicians. The flexibility and versatility of MuTATE make it applicable to a wide range of complex diseases, including cancer, where it can improve therapeutic decisions by providing comprehensive molecular insights for precision medicine. MuTATE has the potential to transform biomarker discovery and subtype identification, leading to more effective and personalized treatment strategies in precision medicine, and advancing our understanding of disease mechanisms at the molecular level.

Availability and implementation: MuTATE is freely available at GitHub (https://github.com/SarahAyton/MuTATE) under the GPLv3 license.

动机:全面的多组学研究推动了有效精准医学疾病建模的进步,但对现有的机器学习方法提出了挑战,这些方法在临床终点的可解释性有限。自动化、全面的疾病建模需要一种机器学习方法,该方法可以通过解释多个临床终点同时识别疾病亚组及其定义的分子生物标志物。当前的工具仅限于单个端点或有限的变量类型,需要高级计算技能,并且需要资源密集型的人工专家解释。结果:我们开发了多目标自动化树引擎(MuTATE),用于自动化和全面的分子建模,支持用户友好的多目标决策树构建和可视化分子生物标志物与具有多个临床终点特征的患者亚组之间的关系。MuTATE在整个模型构建过程中包含多个目标,并允许目标权重,从而能够构建可解释的决策树,从而深入了解疾病异质性和分子特征。MuTATE消除了人工合成多个不可解释模型的需要,使生物信息学家和临床医生能够高效地使用它。MuTATE的灵活性和多功能性使其适用于广泛的复杂疾病,包括癌症,它可以通过为精准医学提供全面的分子见解来改善治疗决策。MuTATE有可能改变生物标志物的发现和亚型鉴定,在精准医学中导致更有效和个性化的治疗策略,并在分子水平上推进我们对疾病机制的理解。可用性和实现:MuTATE在GPLv3许可下可在GitHub (https://github.com/SarahAyton/MuTATE)免费获得。
{"title":"MuTATE-an R package for comprehensive multi-objective molecular modeling.","authors":"Sarah G Ayton,&nbsp;Víctor Treviño","doi":"10.1093/bioinformatics/btad507","DOIUrl":"https://doi.org/10.1093/bioinformatics/btad507","url":null,"abstract":"<p><strong>Motivation: </strong>Comprehensive multi-omics studies have driven advances in disease modeling for effective precision medicine but pose a challenge for existing machine-learning approaches, which have limited interpretability across clinical endpoints. Automated, comprehensive disease modeling requires a machine-learning approach that can simultaneously identify disease subgroups and their defining molecular biomarkers by explaining multiple clinical endpoints. Current tools are restricted to individual endpoints or limited variable types, necessitate advanced computation skills, and require resource-intensive manual expert interpretation.</p><p><strong>Results: </strong>We developed Multi-Target Automated Tree Engine (MuTATE) for automated and comprehensive molecular modeling, which enables user-friendly multi-objective decision tree construction and visualization of relationships between molecular biomarkers and patient subgroups characterized by multiple clinical endpoints. MuTATE incorporates multiple targets throughout model construction and allows for target weights, enabling construction of interpretable decision trees that provide insights into disease heterogeneity and molecular signatures. MuTATE eliminates the need for manual synthesis of multiple non-explainable models, making it highly efficient and accessible for bioinformaticians and clinicians. The flexibility and versatility of MuTATE make it applicable to a wide range of complex diseases, including cancer, where it can improve therapeutic decisions by providing comprehensive molecular insights for precision medicine. MuTATE has the potential to transform biomarker discovery and subtype identification, leading to more effective and personalized treatment strategies in precision medicine, and advancing our understanding of disease mechanisms at the molecular level.</p><p><strong>Availability and implementation: </strong>MuTATE is freely available at GitHub (https://github.com/SarahAyton/MuTATE) under the GPLv3 license.</p>","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2023-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10500092/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10287680","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Bioinformatics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1