首页 > 最新文献

Briefings in bioinformatics最新文献

英文 中文
Evolutionary algorithms simulating molecular evolution: a new field proposal. 模拟分子进化的进化算法:一个新的领域建议。
IF 6.8 2区 生物学 Q1 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-25 DOI: 10.1093/bib/bbae360
James S L Browning, Daniel R Tauritz, John Beckmann

The genetic blueprint for the essential functions of life is encoded in DNA, which is translated into proteins-the engines driving most of our metabolic processes. Recent advancements in genome sequencing have unveiled a vast diversity of protein families, but compared with the massive search space of all possible amino acid sequences, the set of known functional families is minimal. One could say nature has a limited protein "vocabulary." A major question for computational biologists, therefore, is whether this vocabulary can be expanded to include useful proteins that went extinct long ago or have never evolved (yet). By merging evolutionary algorithms, machine learning, and bioinformatics, we can develop highly customized "designer proteins." We dub the new subfield of computational evolution, which employs evolutionary algorithms with DNA string representations, biologically accurate molecular evolution, and bioinformatics-informed fitness functions, Evolutionary Algorithms Simulating Molecular Evolution.

生命基本功能的遗传蓝图由 DNA 编码,然后转化为蛋白质--驱动我们大部分新陈代谢过程的引擎。基因组测序技术的最新进展揭示了蛋白质家族的巨大多样性,但与所有可能氨基酸序列的巨大搜索空间相比,已知功能家族的数量微乎其微。可以说,自然界的蛋白质 "词汇量 "是有限的。因此,计算生物学家面临的一个主要问题是,能否扩大这一词汇量,以包括那些早已灭绝或从未进化过的有用蛋白质。通过融合进化算法、机器学习和生物信息学,我们可以开发出高度定制化的 "设计师蛋白质"。我们将这一计算进化的新子领域命名为 "模拟分子进化的进化算法"(Evolutionary Algorithms Simulating Molecular Evolution)。
{"title":"Evolutionary algorithms simulating molecular evolution: a new field proposal.","authors":"James S L Browning, Daniel R Tauritz, John Beckmann","doi":"10.1093/bib/bbae360","DOIUrl":"10.1093/bib/bbae360","url":null,"abstract":"<p><p>The genetic blueprint for the essential functions of life is encoded in DNA, which is translated into proteins-the engines driving most of our metabolic processes. Recent advancements in genome sequencing have unveiled a vast diversity of protein families, but compared with the massive search space of all possible amino acid sequences, the set of known functional families is minimal. One could say nature has a limited protein \"vocabulary.\" A major question for computational biologists, therefore, is whether this vocabulary can be expanded to include useful proteins that went extinct long ago or have never evolved (yet). By merging evolutionary algorithms, machine learning, and bioinformatics, we can develop highly customized \"designer proteins.\" We dub the new subfield of computational evolution, which employs evolutionary algorithms with DNA string representations, biologically accurate molecular evolution, and bioinformatics-informed fitness functions, Evolutionary Algorithms Simulating Molecular Evolution.</p>","PeriodicalId":9209,"journal":{"name":"Briefings in bioinformatics","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11317538/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141916159","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Computational methods and biomarker discovery strategies for spatial proteomics: a review in immuno-oncology. 空间蛋白质组学的计算方法和生物标记物发现策略:免疫肿瘤学综述。
IF 6.8 2区 生物学 Q1 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-25 DOI: 10.1093/bib/bbae421
Haoyang Mi, Shamilene Sivagnanam, Won Jin Ho, Shuming Zhang, Daniel Bergman, Atul Deshpande, Alexander S Baras, Elizabeth M Jaffee, Lisa M Coussens, Elana J Fertig, Aleksander S Popel

Advancements in imaging technologies have revolutionized our ability to deeply profile pathological tissue architectures, generating large volumes of imaging data with unparalleled spatial resolution. This type of data collection, namely, spatial proteomics, offers invaluable insights into various human diseases. Simultaneously, computational algorithms have evolved to manage the increasing dimensionality of spatial proteomics inherent in this progress. Numerous imaging-based computational frameworks, such as computational pathology, have been proposed for research and clinical applications. However, the development of these fields demands diverse domain expertise, creating barriers to their integration and further application. This review seeks to bridge this divide by presenting a comprehensive guideline. We consolidate prevailing computational methods and outline a roadmap from image processing to data-driven, statistics-informed biomarker discovery. Additionally, we explore future perspectives as the field moves toward interfacing with other quantitative domains, holding significant promise for precision care in immuno-oncology.

成像技术的进步彻底改变了我们深入剖析病理组织结构的能力,产生了大量具有无与伦比的空间分辨率的成像数据。这种数据收集方式,即空间蛋白质组学,为我们深入了解各种人类疾病提供了宝贵的资料。与此同时,计算算法也在不断发展,以管理空间蛋白质组学在这一进展中所固有的不断增加的维度。许多基于成像的计算框架,如计算病理学,已被提出用于研究和临床应用。然而,这些领域的发展需要不同领域的专业知识,这给它们的整合和进一步应用造成了障碍。本综述旨在通过提供一份全面的指南来弥合这一鸿沟。我们整合了当前流行的计算方法,并勾勒出从图像处理到数据驱动、统计信息生物标记物发现的路线图。此外,我们还探讨了该领域与其他定量领域对接的未来前景,为免疫肿瘤学的精准治疗带来了巨大希望。
{"title":"Computational methods and biomarker discovery strategies for spatial proteomics: a review in immuno-oncology.","authors":"Haoyang Mi, Shamilene Sivagnanam, Won Jin Ho, Shuming Zhang, Daniel Bergman, Atul Deshpande, Alexander S Baras, Elizabeth M Jaffee, Lisa M Coussens, Elana J Fertig, Aleksander S Popel","doi":"10.1093/bib/bbae421","DOIUrl":"10.1093/bib/bbae421","url":null,"abstract":"<p><p>Advancements in imaging technologies have revolutionized our ability to deeply profile pathological tissue architectures, generating large volumes of imaging data with unparalleled spatial resolution. This type of data collection, namely, spatial proteomics, offers invaluable insights into various human diseases. Simultaneously, computational algorithms have evolved to manage the increasing dimensionality of spatial proteomics inherent in this progress. Numerous imaging-based computational frameworks, such as computational pathology, have been proposed for research and clinical applications. However, the development of these fields demands diverse domain expertise, creating barriers to their integration and further application. This review seeks to bridge this divide by presenting a comprehensive guideline. We consolidate prevailing computational methods and outline a roadmap from image processing to data-driven, statistics-informed biomarker discovery. Additionally, we explore future perspectives as the field moves toward interfacing with other quantitative domains, holding significant promise for precision care in immuno-oncology.</p>","PeriodicalId":9209,"journal":{"name":"Briefings in bioinformatics","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11343572/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142046352","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
A dual-scale fused hypergraph convolution-based hyperedge prediction model for predicting missing reactions in genome-scale metabolic networks. 基于双尺度融合超图卷积的超edge 预测模型,用于预测基因组尺度代谢网络中的缺失反应。
IF 6.8 2区 生物学 Q1 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-25 DOI: 10.1093/bib/bbae383
Weihong Huang, Feng Yang, Qiang Zhang, Juan Liu

Genome-scale metabolic models (GEMs) are powerful tools for predicting cellular metabolic and physiological states. However, there are still missing reactions in GEMs due to incomplete knowledge. Recent gaps filling methods suggest directly predicting missing responses without relying on phenotypic data. However, they do not differentiate between substrates and products when constructing the prediction models, which affects the predictive performance of the models. In this paper, we propose a hyperedge prediction model that distinguishes substrates and products based on dual-scale fused hypergraph convolution, DSHCNet, for inferring the missing reactions to effectively fill gaps in the GEM. First, we model each hyperedge as a heterogeneous complete graph and then decompose it into three subgraphs at both homogeneous and heterogeneous scales. Then we design two graph convolution-based models to, respectively, extract features of the vertices in two scales, which are then fused via the attention mechanism. Finally, the features of all vertices are further pooled to generate the representative feature of the hyperedge. The strategy of graph decomposition in DSHCNet enables the vertices to engage in message passing independently at both scales, thereby enhancing the capability of information propagation and making the obtained product and substrate features more distinguishable. The experimental results show that the average recovery rate of missing reactions obtained by DSHCNet is at least 11.7% higher than that of the state-of-the-art methods, and that the gap-filled GEMs based on our DSHCNet model achieve the best prediction performance, demonstrating the superiority of our method.

基因组尺度代谢模型(GEM)是预测细胞代谢和生理状态的强大工具。然而,由于知识不完整,GEM 中仍然存在缺失反应。最新的缺口填补方法建议直接预测缺失反应,而无需依赖表型数据。然而,这些方法在构建预测模型时没有区分底物和产物,从而影响了模型的预测性能。本文提出了一种基于双尺度融合超图卷积(DSHCNet)的区分底物和产物的超边缘预测模型,用于推断缺失反应,以有效填补 GEM 的空白。首先,我们将每个超边缘建模为一个异构完整图,然后将其分解为三个同构和异构尺度的子图。然后,我们设计了两个基于图卷积的模型,分别提取两个尺度上的顶点特征,然后通过注意力机制将其融合。最后,进一步汇集所有顶点的特征,生成超edge 的代表特征。DSHCNet 中的图分解策略使顶点在两个尺度上都能独立进行信息传递,从而增强了信息传播能力,并使得到的乘积特征和基底特征更易区分。实验结果表明,DSHCNet 所获得的缺失反应平均恢复率比最先进的方法至少高出 11.7%,而且基于 DSHCNet 模型的间隙填充 GEM 获得了最好的预测性能,证明了我们的方法的优越性。
{"title":"A dual-scale fused hypergraph convolution-based hyperedge prediction model for predicting missing reactions in genome-scale metabolic networks.","authors":"Weihong Huang, Feng Yang, Qiang Zhang, Juan Liu","doi":"10.1093/bib/bbae383","DOIUrl":"10.1093/bib/bbae383","url":null,"abstract":"<p><p>Genome-scale metabolic models (GEMs) are powerful tools for predicting cellular metabolic and physiological states. However, there are still missing reactions in GEMs due to incomplete knowledge. Recent gaps filling methods suggest directly predicting missing responses without relying on phenotypic data. However, they do not differentiate between substrates and products when constructing the prediction models, which affects the predictive performance of the models. In this paper, we propose a hyperedge prediction model that distinguishes substrates and products based on dual-scale fused hypergraph convolution, DSHCNet, for inferring the missing reactions to effectively fill gaps in the GEM. First, we model each hyperedge as a heterogeneous complete graph and then decompose it into three subgraphs at both homogeneous and heterogeneous scales. Then we design two graph convolution-based models to, respectively, extract features of the vertices in two scales, which are then fused via the attention mechanism. Finally, the features of all vertices are further pooled to generate the representative feature of the hyperedge. The strategy of graph decomposition in DSHCNet enables the vertices to engage in message passing independently at both scales, thereby enhancing the capability of information propagation and making the obtained product and substrate features more distinguishable. The experimental results show that the average recovery rate of missing reactions obtained by DSHCNet is at least 11.7% higher than that of the state-of-the-art methods, and that the gap-filled GEMs based on our DSHCNet model achieve the best prediction performance, demonstrating the superiority of our method.</p>","PeriodicalId":9209,"journal":{"name":"Briefings in bioinformatics","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11299038/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141888485","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
GA-GBLUP: leveraging the genetic algorithm to improve the predictability of genomic selection. GA-GBLUP:利用遗传算法提高基因组选择的可预测性。
IF 6.8 2区 生物学 Q1 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-25 DOI: 10.1093/bib/bbae385
Yang Xu, Yuxiang Zhang, Yanru Cui, Kai Zhou, Guangning Yu, Wenyan Yang, Xin Wang, Furong Li, Xiusheng Guan, Xuecai Zhang, Zefeng Yang, Shizhong Xu, Chenwu Xu

Genomic selection (GS) has emerged as an effective technology to accelerate crop hybrid breeding by enabling early selection prior to phenotype collection. Genomic best linear unbiased prediction (GBLUP) is a robust method that has been routinely used in GS breeding programs. However, GBLUP assumes that markers contribute equally to the total genetic variance, which may not be the case. In this study, we developed a novel GS method called GA-GBLUP that leverages the genetic algorithm (GA) to select markers related to the target trait. We defined four fitness functions for optimization, including AIC, BIC, R2, and HAT, to improve the predictability and bin adjacent markers based on the principle of linkage disequilibrium to reduce model dimension. The results demonstrate that the GA-GBLUP model, equipped with R2 and HAT fitness function, produces much higher predictability than GBLUP for most traits in rice and maize datasets, particularly for traits with low heritability. Moreover, we have developed a user-friendly R package, GAGBLUP, for GS, and the package is freely available on CRAN (https://CRAN.R-project.org/package=GAGBLUP).

基因组选择(GS)通过在收集表型之前进行早期选择,已成为加速作物杂交育种的有效技术。基因组最佳线性无偏预测(GBLUP)是一种稳健的方法,已被常规用于基因组选择育种项目。然而,GBLUP 假设标记对总遗传变异的贡献相同,而实际情况可能并非如此。在本研究中,我们开发了一种名为 GA-GBLUP 的新型基因组学方法,利用遗传算法(GA)来选择与目标性状相关的标记。我们定义了 AIC、BIC、R2 和 HAT 等四种适合度函数进行优化,以提高可预测性,并根据连锁不平衡原理将相邻标记进行分选,以减少模型维度。结果表明,对于水稻和玉米数据集中的大多数性状,尤其是遗传率较低的性状,配备 R2 和 HAT 健身函数的 GA-GBLUP 模型比 GBLUP 产生的预测性要高得多。此外,我们还为 GS 开发了一个用户友好型 R 软件包 GAGBLUP,该软件包可在 CRAN(https://CRAN.R-project.org/package=GAGBLUP)上免费获取。
{"title":"GA-GBLUP: leveraging the genetic algorithm to improve the predictability of genomic selection.","authors":"Yang Xu, Yuxiang Zhang, Yanru Cui, Kai Zhou, Guangning Yu, Wenyan Yang, Xin Wang, Furong Li, Xiusheng Guan, Xuecai Zhang, Zefeng Yang, Shizhong Xu, Chenwu Xu","doi":"10.1093/bib/bbae385","DOIUrl":"10.1093/bib/bbae385","url":null,"abstract":"<p><p>Genomic selection (GS) has emerged as an effective technology to accelerate crop hybrid breeding by enabling early selection prior to phenotype collection. Genomic best linear unbiased prediction (GBLUP) is a robust method that has been routinely used in GS breeding programs. However, GBLUP assumes that markers contribute equally to the total genetic variance, which may not be the case. In this study, we developed a novel GS method called GA-GBLUP that leverages the genetic algorithm (GA) to select markers related to the target trait. We defined four fitness functions for optimization, including AIC, BIC, R2, and HAT, to improve the predictability and bin adjacent markers based on the principle of linkage disequilibrium to reduce model dimension. The results demonstrate that the GA-GBLUP model, equipped with R2 and HAT fitness function, produces much higher predictability than GBLUP for most traits in rice and maize datasets, particularly for traits with low heritability. Moreover, we have developed a user-friendly R package, GAGBLUP, for GS, and the package is freely available on CRAN (https://CRAN.R-project.org/package=GAGBLUP).</p>","PeriodicalId":9209,"journal":{"name":"Briefings in bioinformatics","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11299030/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141888487","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Enhancer-driven gene regulatory networks inference from single-cell RNA-seq and ATAC-seq data. 从单细胞 RNA-seq 和 ATAC-seq 数据推断增强子驱动的基因调控网络。
IF 6.8 2区 生物学 Q1 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-25 DOI: 10.1093/bib/bbae369
Yang Li, Anjun Ma, Yizhong Wang, Qi Guo, Cankun Wang, Hongjun Fu, Bingqiang Liu, Qin Ma

Deciphering the intricate relationships between transcription factors (TFs), enhancers, and genes through the inference of enhancer-driven gene regulatory networks (eGRNs) is crucial in understanding gene regulatory programs in a complex biological system. This study introduces STREAM, a novel method that leverages a Steiner forest problem model, a hybrid biclustering pipeline, and submodular optimization to infer eGRNs from jointly profiled single-cell transcriptome and chromatin accessibility data. Compared to existing methods, STREAM demonstrates enhanced performance in terms of TF recovery, TF-enhancer linkage prediction, and enhancer-gene relation discovery. Application of STREAM to an Alzheimer's disease dataset and a diffuse small lymphocytic lymphoma dataset reveals its ability to identify TF-enhancer-gene relations associated with pseudotime, as well as key TF-enhancer-gene relations and TF cooperation underlying tumor cells.

通过推断增强子驱动的基因调控网络(eGRN)来破译转录因子(TFs)、增强子和基因之间错综复杂的关系,对于理解复杂生物系统中的基因调控程序至关重要。本研究介绍了一种新方法 STREAM,它利用斯坦纳森林问题模型、混合双聚类管道和亚模块优化,从联合剖析的单细胞转录组和染色质可及性数据中推断出 eGRN。与现有方法相比,STREAM在TF恢复、TF-增强子关联预测和增强子-基因关系发现方面表现出更强的性能。将 STREAM 应用于阿尔茨海默病数据集和弥漫性小淋巴细胞淋巴瘤数据集显示,它有能力识别与假时间相关的 TF-增强子-基因关系,以及肿瘤细胞底层的关键 TF-增强子-基因关系和 TF 合作。
{"title":"Enhancer-driven gene regulatory networks inference from single-cell RNA-seq and ATAC-seq data.","authors":"Yang Li, Anjun Ma, Yizhong Wang, Qi Guo, Cankun Wang, Hongjun Fu, Bingqiang Liu, Qin Ma","doi":"10.1093/bib/bbae369","DOIUrl":"10.1093/bib/bbae369","url":null,"abstract":"<p><p>Deciphering the intricate relationships between transcription factors (TFs), enhancers, and genes through the inference of enhancer-driven gene regulatory networks (eGRNs) is crucial in understanding gene regulatory programs in a complex biological system. This study introduces STREAM, a novel method that leverages a Steiner forest problem model, a hybrid biclustering pipeline, and submodular optimization to infer eGRNs from jointly profiled single-cell transcriptome and chromatin accessibility data. Compared to existing methods, STREAM demonstrates enhanced performance in terms of TF recovery, TF-enhancer linkage prediction, and enhancer-gene relation discovery. Application of STREAM to an Alzheimer's disease dataset and a diffuse small lymphocytic lymphoma dataset reveals its ability to identify TF-enhancer-gene relations associated with pseudotime, as well as key TF-enhancer-gene relations and TF cooperation underlying tumor cells.</p>","PeriodicalId":9209,"journal":{"name":"Briefings in bioinformatics","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11289686/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141854883","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
MetaPredictor: in silico prediction of drug metabolites based on deep language models with prompt engineering. MetaPredictor:基于深度语言模型和提示工程的药物代谢物硅学预测。
IF 6.8 2区 生物学 Q1 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-25 DOI: 10.1093/bib/bbae374
Keyun Zhu, Mengting Huang, Yimeng Wang, Yaxin Gu, Weihua Li, Guixia Liu, Yun Tang

Metabolic processes can transform a drug into metabolites with different properties that may affect its efficacy and safety. Therefore, investigation of the metabolic fate of a drug candidate is of great significance for drug discovery. Computational methods have been developed to predict drug metabolites, but most of them suffer from two main obstacles: the lack of model generalization due to restrictions on metabolic transformation rules or specific enzyme families, and high rate of false-positive predictions. Here, we presented MetaPredictor, a rule-free, end-to-end and prompt-based method to predict possible human metabolites of small molecules including drugs as a sequence translation problem. We innovatively introduced prompt engineering into deep language models to enrich domain knowledge and guide decision-making. The results showed that using prompts that specify the sites of metabolism (SoMs) can steer the model to propose more accurate metabolite predictions, achieving a 30.4% increase in recall and a 16.8% reduction in false positives over the baseline model. The transfer learning strategy was also utilized to tackle the limited availability of metabolic data. For the adaptation to automatic or non-expert prediction, MetaPredictor was designed as a two-stage schema consisting of automatic identification of SoMs followed by metabolite prediction. Compared to four available drug metabolite prediction tools, our method showed comparable performance on the major enzyme families and better generalization that could additionally identify metabolites catalyzed by less common enzymes. The results indicated that MetaPredictor could provide a more comprehensive and accurate prediction of drug metabolism through the effective combination of transfer learning and prompt-based learning strategies.

代谢过程可将药物转化为具有不同性质的代谢物,这些性质可能会影响药物的疗效和安全性。因此,研究候选药物的代谢命运对药物发现具有重要意义。目前已开发出预测药物代谢物的计算方法,但大多数方法都存在两个主要障碍:一是受代谢转化规则或特定酶家族的限制,模型缺乏通用性;二是假阳性预测率较高。在此,我们提出了一种无规则、端到端和基于提示的方法--MetaPredictor,用于预测作为序列翻译问题的小分子(包括药物)可能的人类代谢物。我们创新性地将提示工程引入深度语言模型,以丰富领域知识并指导决策。结果表明,使用指定代谢位点(SoMs)的提示可以引导模型提出更准确的代谢物预测,与基线模型相比,召回率提高了 30.4%,误报率降低了 16.8%。迁移学习策略还被用来解决代谢数据有限的问题。为了适应自动或非专家预测,MetaPredictor 被设计成一个两阶段模式,包括自动识别 SoMs,然后进行代谢物预测。与现有的四种药物代谢物预测工具相比,我们的方法在主要酶家族上表现出了相当的性能,并且具有更好的通用性,可以额外识别由不太常见的酶催化的代谢物。结果表明,通过有效结合迁移学习和基于提示的学习策略,MetaPredictor 可以提供更全面、更准确的药物代谢预测。
{"title":"MetaPredictor: in silico prediction of drug metabolites based on deep language models with prompt engineering.","authors":"Keyun Zhu, Mengting Huang, Yimeng Wang, Yaxin Gu, Weihua Li, Guixia Liu, Yun Tang","doi":"10.1093/bib/bbae374","DOIUrl":"10.1093/bib/bbae374","url":null,"abstract":"<p><p>Metabolic processes can transform a drug into metabolites with different properties that may affect its efficacy and safety. Therefore, investigation of the metabolic fate of a drug candidate is of great significance for drug discovery. Computational methods have been developed to predict drug metabolites, but most of them suffer from two main obstacles: the lack of model generalization due to restrictions on metabolic transformation rules or specific enzyme families, and high rate of false-positive predictions. Here, we presented MetaPredictor, a rule-free, end-to-end and prompt-based method to predict possible human metabolites of small molecules including drugs as a sequence translation problem. We innovatively introduced prompt engineering into deep language models to enrich domain knowledge and guide decision-making. The results showed that using prompts that specify the sites of metabolism (SoMs) can steer the model to propose more accurate metabolite predictions, achieving a 30.4% increase in recall and a 16.8% reduction in false positives over the baseline model. The transfer learning strategy was also utilized to tackle the limited availability of metabolic data. For the adaptation to automatic or non-expert prediction, MetaPredictor was designed as a two-stage schema consisting of automatic identification of SoMs followed by metabolite prediction. Compared to four available drug metabolite prediction tools, our method showed comparable performance on the major enzyme families and better generalization that could additionally identify metabolites catalyzed by less common enzymes. The results indicated that MetaPredictor could provide a more comprehensive and accurate prediction of drug metabolism through the effective combination of transfer learning and prompt-based learning strategies.</p>","PeriodicalId":9209,"journal":{"name":"Briefings in bioinformatics","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11289679/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141854886","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
CatLearning: highly accurate gene expression prediction from histone mark. CatLearning:根据组蛋白标记进行高精度基因表达预测。
IF 6.8 2区 生物学 Q1 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-25 DOI: 10.1093/bib/bbae373
Weining Lu, Yin Tang, Yu Liu, Shiyi Lin, Qifan Shuai, Bin Liang, Rongqing Zhang, Yu Cheng, Dong Fang

Histone modifications, known as histone marks, are pivotal in regulating gene expression within cells. The vast array of potential combinations of histone marks presents a considerable challenge in decoding the regulatory mechanisms solely through biological experimental approaches. To overcome this challenge, we have developed a method called CatLearning. It utilizes a modified convolutional neural network architecture with a specialized adaptation Residual Network to quantitatively interpret histone marks and predict gene expression. This architecture integrates long-range histone information up to 500Kb and learns chromatin interaction features without 3D information. By using only one histone mark, CatLearning achieves a high level of accuracy. Furthermore, CatLearning predicts gene expression by simulating changes in histone modifications at enhancers and throughout the genome. These findings help comprehend the architecture of histone marks and develop diagnostic and therapeutic targets for diseases with epigenetic changes.

组蛋白修饰,即组蛋白标记,是调节细胞内基因表达的关键。组蛋白标记的潜在组合种类繁多,这给仅通过生物实验方法解码调控机制带来了巨大挑战。为了克服这一挑战,我们开发了一种名为 CatLearning 的方法。它利用改进的卷积神经网络架构和专门的适应残差网络来定量解释组蛋白标记和预测基因表达。该架构整合了长达 500Kb 的长程组蛋白信息,并在没有三维信息的情况下学习染色质相互作用特征。通过只使用一个组蛋白标记,CatLearning 实现了高水平的准确性。此外,CatLearning 还能通过模拟增强子和整个基因组中组蛋白修饰的变化来预测基因表达。这些发现有助于理解组蛋白标记的结构,并开发出针对表观遗传变化疾病的诊断和治疗目标。
{"title":"CatLearning: highly accurate gene expression prediction from histone mark.","authors":"Weining Lu, Yin Tang, Yu Liu, Shiyi Lin, Qifan Shuai, Bin Liang, Rongqing Zhang, Yu Cheng, Dong Fang","doi":"10.1093/bib/bbae373","DOIUrl":"10.1093/bib/bbae373","url":null,"abstract":"<p><p>Histone modifications, known as histone marks, are pivotal in regulating gene expression within cells. The vast array of potential combinations of histone marks presents a considerable challenge in decoding the regulatory mechanisms solely through biological experimental approaches. To overcome this challenge, we have developed a method called CatLearning. It utilizes a modified convolutional neural network architecture with a specialized adaptation Residual Network to quantitatively interpret histone marks and predict gene expression. This architecture integrates long-range histone information up to 500Kb and learns chromatin interaction features without 3D information. By using only one histone mark, CatLearning achieves a high level of accuracy. Furthermore, CatLearning predicts gene expression by simulating changes in histone modifications at enhancers and throughout the genome. These findings help comprehend the architecture of histone marks and develop diagnostic and therapeutic targets for diseases with epigenetic changes.</p>","PeriodicalId":9209,"journal":{"name":"Briefings in bioinformatics","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11285185/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141787300","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
BioM2: biologically informed multi-stage machine learning for phenotype prediction using omics data. BioM2:利用omics数据进行表型预测的生物信息多级机器学习。
IF 6.8 2区 生物学 Q1 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-25 DOI: 10.1093/bib/bbae384
Shunjie Zhang, Pan Li, Shenghan Wang, Jijun Zhu, Zhongting Huang, Fuqiang Cai, Sebastian Freidel, Fei Ling, Emanuel Schwarz, Junfang Chen

Navigating the complex landscape of high-dimensional omics data with machine learning models presents a significant challenge. The integration of biological domain knowledge into these models has shown promise in creating more meaningful stratifications of predictor variables, leading to algorithms that are both more accurate and generalizable. However, the wider availability of machine learning tools capable of incorporating such biological knowledge remains limited. Addressing this gap, we introduce BioM2, a novel R package designed for biologically informed multistage machine learning. BioM2 uniquely leverages biological information to effectively stratify and aggregate high-dimensional biological data in the context of machine learning. Demonstrating its utility with genome-wide DNA methylation and transcriptome-wide gene expression data, BioM2 has shown to enhance predictive performance, surpassing traditional machine learning models that operate without the integration of biological knowledge. A key feature of BioM2 is its ability to rank predictor variables within biological categories, specifically Gene Ontology pathways. This functionality not only aids in the interpretability of the results but also enables a subsequent modular network analysis of these variables, shedding light on the intricate systems-level biology underpinning the predictive outcome. We have proposed a biologically informed multistage machine learning framework termed BioM2 for phenotype prediction based on omics data. BioM2 has been incorporated into the BioM2 CRAN package (https://cran.r-project.org/web/packages/BioM2/index.html).

利用机器学习模型浏览复杂的高维 omics 数据是一项重大挑战。将生物领域知识整合到这些模型中,有望对预测变量进行更有意义的分层,从而使算法更准确、更具有普适性。然而,能够将此类生物知识融入机器学习工具的可用性仍然有限。为了填补这一空白,我们推出了 BioM2,这是一个新颖的 R 软件包,专为生物信息多阶段机器学习而设计。BioM2 可独特地利用生物信息,在机器学习中有效地分层和聚合高维生物数据。BioM2 在全基因组 DNA 甲基化和全转录组基因表达数据中的应用表明,它能提高预测性能,超越了没有整合生物知识的传统机器学习模型。BioM2 的一个主要特点是它能在生物类别(特别是基因本体论途径)内对预测变量进行排序。这一功能不仅有助于结果的可解释性,还能对这些变量进行后续的模块化网络分析,从而揭示支撑预测结果的错综复杂的系统级生物学。我们提出了一种基于生物信息的多阶段机器学习框架,称为 BioM2,用于基于 omics 数据的表型预测。BioM2 已被纳入 BioM2 CRAN 软件包 (https://cran.r-project.org/web/packages/BioM2/index.html)。
{"title":"BioM2: biologically informed multi-stage machine learning for phenotype prediction using omics data.","authors":"Shunjie Zhang, Pan Li, Shenghan Wang, Jijun Zhu, Zhongting Huang, Fuqiang Cai, Sebastian Freidel, Fei Ling, Emanuel Schwarz, Junfang Chen","doi":"10.1093/bib/bbae384","DOIUrl":"10.1093/bib/bbae384","url":null,"abstract":"<p><p>Navigating the complex landscape of high-dimensional omics data with machine learning models presents a significant challenge. The integration of biological domain knowledge into these models has shown promise in creating more meaningful stratifications of predictor variables, leading to algorithms that are both more accurate and generalizable. However, the wider availability of machine learning tools capable of incorporating such biological knowledge remains limited. Addressing this gap, we introduce BioM2, a novel R package designed for biologically informed multistage machine learning. BioM2 uniquely leverages biological information to effectively stratify and aggregate high-dimensional biological data in the context of machine learning. Demonstrating its utility with genome-wide DNA methylation and transcriptome-wide gene expression data, BioM2 has shown to enhance predictive performance, surpassing traditional machine learning models that operate without the integration of biological knowledge. A key feature of BioM2 is its ability to rank predictor variables within biological categories, specifically Gene Ontology pathways. This functionality not only aids in the interpretability of the results but also enables a subsequent modular network analysis of these variables, shedding light on the intricate systems-level biology underpinning the predictive outcome. We have proposed a biologically informed multistage machine learning framework termed BioM2 for phenotype prediction based on omics data. BioM2 has been incorporated into the BioM2 CRAN package (https://cran.r-project.org/web/packages/BioM2/index.html).</p>","PeriodicalId":9209,"journal":{"name":"Briefings in bioinformatics","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11316398/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141911770","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
TME-NET: an interpretable deep neural network for predicting pan-cancer immune checkpoint inhibitor responses. TME-NET:用于预测泛癌症免疫检查点抑制剂反应的可解释深度神经网络。
IF 6.8 2区 生物学 Q1 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-25 DOI: 10.1093/bib/bbae410
Xiaobao Ding, Lin Zhang, Ming Fan, Lihua Li

Immunotherapy with immune checkpoint inhibitors (ICIs) is increasingly used to treat various tumor types. Determining patient responses to ICIs presents a significant clinical challenge. Although components of the tumor microenvironment (TME) are used to predict patient outcomes, comprehensive assessments of the TME are frequently overlooked. Using a top-down approach, the TME was divided into five layers-outcome, immune role, cell, cellular component, and gene. Using this structure, a neural network called TME-NET was developed to predict responses to ICIs. Model parameter weights and cell ablation studies were used to investigate the influence of TME components. The model was developed and evaluated using a pan-cancer cohort of 948 patients across four cancer types, with Area Under the Curve (AUC) and accuracy as performance metrics. Results show that TME-NET surpasses established models such as support vector machine and k-nearest neighbors in AUC and accuracy. Visualization of model parameter weights showed that at the cellular layer, Th1 cells enhance immune responses, whereas myeloid-derived suppressor cells and M2 macrophages show strong immunosuppressive effects. Cell ablation studies further confirmed the impact of these cells. At the gene layer, the transcription factors STAT4 in Th1 cells and IRF4 in M2 macrophages significantly affect TME dynamics. Additionally, the cytokine-encoding genes IFNG from Th1 cells and ARG1 from M2 macrophages are crucial for modulating immune responses within the TME. Survival data from immunotherapy cohorts confirmed the prognostic ability of these markers, with p-values <0.01. In summary, TME-NET performs well in predicting immunotherapy responses and offers interpretable insights into the immunotherapy process. It can be customized at https://immbal.shinyapps.io/TME-NET.

免疫检查点抑制剂(ICIs)免疫疗法越来越多地用于治疗各种类型的肿瘤。确定患者对 ICIs 的反应是一项重大的临床挑战。虽然肿瘤微环境(TME)的组成部分可用于预测患者的预后,但对TME的全面评估却经常被忽视。采用自上而下的方法,TME 被分为五层--结果、免疫作用、细胞、细胞成分和基因。利用这种结构,开发出了一种名为 TME-NET 的神经网络,用于预测对 ICIs 的反应。模型参数权重和细胞消融研究用于研究 TME 成分的影响。该模型以曲线下面积(AUC)和准确性作为性能指标,使用四种癌症类型的 948 名患者组成的泛癌症队列进行了开发和评估。结果表明,TME-NET 的 AUC 和准确率超过了支持向量机和 k 近邻等成熟模型。对模型参数权重的可视化显示,在细胞层,Th1 细胞增强了免疫反应,而髓源性抑制细胞和 M2 巨噬细胞则表现出强烈的免疫抑制效应。细胞消减研究进一步证实了这些细胞的影响。在基因层,Th1 细胞中的转录因子 STAT4 和 M2 巨噬细胞中的转录因子 IRF4 对 TME 动态有显著影响。此外,Th1 细胞的细胞因子编码基因 IFNG 和 M2 巨噬细胞的 ARG1 对于调节 TME 内的免疫反应也至关重要。来自免疫疗法队列的生存数据证实了这些标记物的预后能力,p 值为
{"title":"TME-NET: an interpretable deep neural network for predicting pan-cancer immune checkpoint inhibitor responses.","authors":"Xiaobao Ding, Lin Zhang, Ming Fan, Lihua Li","doi":"10.1093/bib/bbae410","DOIUrl":"10.1093/bib/bbae410","url":null,"abstract":"<p><p>Immunotherapy with immune checkpoint inhibitors (ICIs) is increasingly used to treat various tumor types. Determining patient responses to ICIs presents a significant clinical challenge. Although components of the tumor microenvironment (TME) are used to predict patient outcomes, comprehensive assessments of the TME are frequently overlooked. Using a top-down approach, the TME was divided into five layers-outcome, immune role, cell, cellular component, and gene. Using this structure, a neural network called TME-NET was developed to predict responses to ICIs. Model parameter weights and cell ablation studies were used to investigate the influence of TME components. The model was developed and evaluated using a pan-cancer cohort of 948 patients across four cancer types, with Area Under the Curve (AUC) and accuracy as performance metrics. Results show that TME-NET surpasses established models such as support vector machine and k-nearest neighbors in AUC and accuracy. Visualization of model parameter weights showed that at the cellular layer, Th1 cells enhance immune responses, whereas myeloid-derived suppressor cells and M2 macrophages show strong immunosuppressive effects. Cell ablation studies further confirmed the impact of these cells. At the gene layer, the transcription factors STAT4 in Th1 cells and IRF4 in M2 macrophages significantly affect TME dynamics. Additionally, the cytokine-encoding genes IFNG from Th1 cells and ARG1 from M2 macrophages are crucial for modulating immune responses within the TME. Survival data from immunotherapy cohorts confirmed the prognostic ability of these markers, with p-values <0.01. In summary, TME-NET performs well in predicting immunotherapy responses and offers interpretable insights into the immunotherapy process. It can be customized at https://immbal.shinyapps.io/TME-NET.</p>","PeriodicalId":9209,"journal":{"name":"Briefings in bioinformatics","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11337220/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142016418","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Harnessing large language models' zero-shot and few-shot learning capabilities for regulatory research. 利用大型语言模型的零点学习和少量学习能力开展监管研究。
IF 6.8 2区 生物学 Q1 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-25 DOI: 10.1093/bib/bbae354
Hamed Meshkin, Joel Zirkle, Ghazal Arabidarrehdor, Anik Chaturbedi, Shilpa Chakravartula, John Mann, Bradlee Thrasher, Zhihua Li

Large language models (LLMs) are sophisticated AI-driven models trained on vast sources of natural language data. They are adept at generating responses that closely mimic human conversational patterns. One of the most notable examples is OpenAI's ChatGPT, which has been extensively used across diverse sectors. Despite their flexibility, a significant challenge arises as most users must transmit their data to the servers of companies operating these models. Utilizing ChatGPT or similar models online may inadvertently expose sensitive information to the risk of data breaches. Therefore, implementing LLMs that are open source and smaller in scale within a secure local network becomes a crucial step for organizations where ensuring data privacy and protection has the highest priority, such as regulatory agencies. As a feasibility evaluation, we implemented a series of open-source LLMs within a regulatory agency's local network and assessed their performance on specific tasks involving extracting relevant clinical pharmacology information from regulatory drug labels. Our research shows that some models work well in the context of few- or zero-shot learning, achieving performance comparable, or even better than, neural network models that needed thousands of training samples. One of the models was selected to address a real-world issue of finding intrinsic factors that affect drugs' clinical exposure without any training or fine-tuning. In a dataset of over 700 000 sentences, the model showed a 78.5% accuracy rate. Our work pointed to the possibility of implementing open-source LLMs within a secure local network and using these models to perform various natural language processing tasks when large numbers of training examples are unavailable.

大型语言模型(LLM)是在大量自然语言数据基础上训练而成的复杂人工智能驱动模型。它们善于生成近似人类对话模式的回复。最著名的例子之一是 OpenAI 的 ChatGPT,它已被广泛应用于各个领域。尽管它们具有灵活性,但由于大多数用户必须将数据传输到运营这些模型的公司的服务器上,因此也带来了巨大的挑战。在线使用 ChatGPT 或类似模型可能会无意中将敏感信息暴露在数据泄露的风险之下。因此,在安全的本地网络中实施开源且规模较小的 LLM,对于确保数据隐私和保护具有最高优先级的组织(如监管机构)来说是至关重要的一步。作为可行性评估,我们在监管机构的本地网络中实施了一系列开源 LLM,并评估了它们在从监管药物标签中提取相关临床药理信息的特定任务中的表现。我们的研究表明,一些模型在很少或零次学习的情况下也能很好地工作,其性能可与需要数千个训练样本的神经网络模型相媲美,甚至更好。我们选择了其中一个模型来解决现实世界中的一个问题,即在没有任何训练或微调的情况下找到影响药物临床暴露的内在因素。在一个包含 70 多万个句子的数据集中,该模型的准确率达到了 78.5%。我们的工作表明,可以在安全的本地网络中实施开源 LLM,并在没有大量训练实例的情况下使用这些模型执行各种自然语言处理任务。
{"title":"Harnessing large language models' zero-shot and few-shot learning capabilities for regulatory research.","authors":"Hamed Meshkin, Joel Zirkle, Ghazal Arabidarrehdor, Anik Chaturbedi, Shilpa Chakravartula, John Mann, Bradlee Thrasher, Zhihua Li","doi":"10.1093/bib/bbae354","DOIUrl":"10.1093/bib/bbae354","url":null,"abstract":"<p><p>Large language models (LLMs) are sophisticated AI-driven models trained on vast sources of natural language data. They are adept at generating responses that closely mimic human conversational patterns. One of the most notable examples is OpenAI's ChatGPT, which has been extensively used across diverse sectors. Despite their flexibility, a significant challenge arises as most users must transmit their data to the servers of companies operating these models. Utilizing ChatGPT or similar models online may inadvertently expose sensitive information to the risk of data breaches. Therefore, implementing LLMs that are open source and smaller in scale within a secure local network becomes a crucial step for organizations where ensuring data privacy and protection has the highest priority, such as regulatory agencies. As a feasibility evaluation, we implemented a series of open-source LLMs within a regulatory agency's local network and assessed their performance on specific tasks involving extracting relevant clinical pharmacology information from regulatory drug labels. Our research shows that some models work well in the context of few- or zero-shot learning, achieving performance comparable, or even better than, neural network models that needed thousands of training samples. One of the models was selected to address a real-world issue of finding intrinsic factors that affect drugs' clinical exposure without any training or fine-tuning. In a dataset of over 700 000 sentences, the model showed a 78.5% accuracy rate. Our work pointed to the possibility of implementing open-source LLMs within a secure local network and using these models to perform various natural language processing tasks when large numbers of training examples are unavailable.</p>","PeriodicalId":9209,"journal":{"name":"Briefings in bioinformatics","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11342240/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142035262","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Briefings in 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