首页 > 最新文献

bioRxiv - Bioinformatics最新文献

英文 中文
Flexible and robust cell type annotation for highly multiplexed tissue images 为高度复用的组织图像提供灵活、稳健的细胞类型标注
Pub Date : 2024-09-16 DOI: 10.1101/2024.09.12.612510
Huangqingbo Sun, Shiqiu Yu, Anna Martinez Casals, Anna Bäckström, Yuxin Lu, Cecilia Lindskog, Emma Lundberg, Robert F. Murphy
Identifying cell types in highly multiplexed images is essential for understanding tissue spatial organization. Current cell type annotation methods often rely on extensive reference images and manual adjustments. In this work, we present a tool, Robust Image-Based Cell Annotator (RIBCA), that enables accurate, automated, unbiased, and fine-grained cell type annotation for images with a wide range of antibody panels, without requiring additional model training or human intervention. Our tool has successfully annotated over 1 million cells, revealing the spatial organization of various cell types across more than 40 different human tissues. It is open-source and features a modular design, allowing for easy extension to additional cell types.
在高度复用的图像中识别细胞类型对于了解组织的空间组织至关重要。目前的细胞类型标注方法通常依赖于大量的参考图像和人工调整。在这项工作中,我们提出了一种名为 "基于强大图像的细胞注释器"(RIBCA)的工具,无需额外的模型训练或人工干预,就能对具有各种抗体面板的图像进行准确、自动、无偏见和精细的细胞类型注释。我们的工具已成功注释了 100 多万个细胞,揭示了 40 多种不同人体组织中各种细胞类型的空间组织。该工具是开源的,采用模块化设计,可轻松扩展到其他细胞类型。
{"title":"Flexible and robust cell type annotation for highly multiplexed tissue images","authors":"Huangqingbo Sun, Shiqiu Yu, Anna Martinez Casals, Anna Bäckström, Yuxin Lu, Cecilia Lindskog, Emma Lundberg, Robert F. Murphy","doi":"10.1101/2024.09.12.612510","DOIUrl":"https://doi.org/10.1101/2024.09.12.612510","url":null,"abstract":"Identifying cell types in highly multiplexed images is essential for understanding tissue spatial organization. Current cell type annotation methods often rely on extensive reference images and manual adjustments. In this work, we present a tool, Robust Image-Based Cell Annotator (RIBCA), that enables accurate, automated, unbiased, and fine-grained cell type annotation for images with a wide range of antibody panels, without requiring additional model training or human intervention. Our tool has successfully annotated over 1 million cells, revealing the spatial organization of various cell types across more than 40 different human tissues. It is open-source and features a modular design, allowing for easy extension to additional cell types.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"26 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250182","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Escaping the drug-bias trap: using debiasing design to improve interpretability and generalization of drug-target interaction prediction 摆脱药物偏倚陷阱:利用去杂设计提高药物-靶点相互作用预测的可解释性和通用性
Pub Date : 2024-09-15 DOI: 10.1101/2024.09.12.612771
Pei-Dong Zhang, Jianzhu Ma, Ting Chen
Considering the high cost associated with determining reaction affinities through in-vitro experiments, virtual screening of potential drugs bound with specific protein pockets from vast compounds is critical in AI-assisted drug discovery. Deep-leaning approaches have been proposed for Drug-Target Interaction (DTI) prediction. However, they have shown overestimated accuracy because of the drug-bias trap, a challenge that results from excessive reliance on the drug branch in the traditional drug-protein dual-branch network approach. This casts doubt on the interpretability and generalizability of existing Drug-Target Interaction (DTI) models. Therefore, we introduce UdanDTI, an innovative deep-learning architecture designed specifically for predicting drug-protein interactions. UdanDTI applies an unbalanced dual-branch system and an attentive aggregation module to enhance interpretability from a biological perspective. Across various public datasets, UdanDTI demonstrates outstanding performance, outperforming state-of-the-art models under in-domain, cross-domain, and structural interpretability settings. Notably, it demonstrates exceptional accuracy in predicting drug responses of two crucial subgroups of Epidermal Growth Factor Receptor (EGFR) mutations associated with non-small cell lung cancer, consistent with experimental results. Meanwhile, UdanDTI could complement the advanced molecular docking software DiffDock. The codes and datasets of UdanDTI are available at https://github.com/CQ-zhang-2016/UdanDTI.
考虑到通过体外实验确定反应亲和力的成本较高,从大量化合物中虚拟筛选出与特定蛋白质口袋结合的潜在药物对于人工智能辅助药物发现至关重要。有人提出了用于药物-靶点相互作用(DTI)预测的深度倾斜方法。然而,由于传统的药物-蛋白质双分支网络方法过度依赖药物分支而导致的药物偏倚陷阱(drug-bias trap),这些方法的准确性被高估了。这使人们对现有药物-靶点相互作用(DTI)模型的可解释性和可推广性产生了怀疑。因此,我们引入了 UdanDTI,这是一种专为预测药物-蛋白质相互作用而设计的创新型深度学习架构。UdanDTI 采用不平衡双分支系统和贴心的聚合模块,从生物学角度提高了可解释性。在各种公共数据集上,UdanDTI 都表现出了卓越的性能,在域内、跨域和结构可解释性设置下都优于最先进的模型。值得注意的是,它在预测与非小细胞肺癌相关的表皮生长因子受体(EGFR)突变的两个关键亚组的药物反应方面表现出了极高的准确性,这与实验结果是一致的。同时,UdanDTI 可以与先进的分子对接软件 DiffDock 相辅相成。UdanDTI的代码和数据集可在https://github.com/CQ-zhang-2016/UdanDTI。
{"title":"Escaping the drug-bias trap: using debiasing design to improve interpretability and generalization of drug-target interaction prediction","authors":"Pei-Dong Zhang, Jianzhu Ma, Ting Chen","doi":"10.1101/2024.09.12.612771","DOIUrl":"https://doi.org/10.1101/2024.09.12.612771","url":null,"abstract":"Considering the high cost associated with determining reaction affinities through in-vitro experiments, virtual screening of potential drugs bound with specific protein pockets from vast compounds is critical in AI-assisted drug discovery. Deep-leaning approaches have been proposed for Drug-Target Interaction (DTI) prediction. However, they have shown overestimated accuracy because of the drug-bias trap, a challenge that results from excessive reliance on the drug branch in the traditional drug-protein dual-branch network approach. This casts doubt on the interpretability and generalizability of existing Drug-Target Interaction (DTI) models. Therefore, we introduce UdanDTI, an innovative deep-learning architecture designed specifically for predicting drug-protein interactions. UdanDTI applies an unbalanced dual-branch system and an attentive aggregation module to enhance interpretability from a biological perspective. Across various public datasets, UdanDTI demonstrates outstanding performance, outperforming state-of-the-art models under in-domain, cross-domain, and structural interpretability settings. Notably, it demonstrates exceptional accuracy in predicting drug responses of two crucial subgroups of Epidermal Growth Factor Receptor (EGFR) mutations associated with non-small cell lung cancer, consistent with experimental results. Meanwhile, UdanDTI could complement the advanced molecular docking software DiffDock. The codes and datasets of UdanDTI are available at https://github.com/CQ-zhang-2016/UdanDTI.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"19 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250372","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Variant Evolution Graph: Can We Infer How SARS-CoV-2 Variants are Evolving? 变异体进化图:我们能推断出 SARS-CoV-2 变异是如何演变的吗?
Pub Date : 2024-09-15 DOI: 10.1101/2024.09.13.612805
Badhan Das, Lenwood S. Heath
The SARS-CoV-2 virus has undergone mutations over time, leading to genetic diversity among circulating viral strains. This genetic diversity can affect the characteristics of the virus, including its transmissibility and the severity of symptoms in infected individuals. During the pandemic, this frequent mutation creates an enormous cloud of variants known as viral quasispecies. Most variation is lost due to the tight bottlenecks imposed by transmission and survival. Advancements in next-generation sequencing have facilitated the rapid and cost-effective production of complete viral genomes, enabling the ongoing monitoring of the evolution of the SARS-CoV-2 genome. However, inferring a reliable phylogeny from GISAID (the Global Initiative on Sharing All Influenza Data) is daunting due to the vast number of sequences. In the face of this complexity, this research proposes a new method of representing the evolutionary and epidemiological relationships among the SARS-CoV-2 variants inspired by quasispecies theory. We aim to build a Variant Evolution Graph (VEG), a novel way to model viral evolution in a local pandemic region based on the mutational distance of the genotypes of the variants. VEG is a directed acyclic graph and not necessarily a tree because a variant can evolve from more than one variant; here, the vertices represent the genotypes of the variants associated with their human hosts, and the edges represent the evolutionary relationships among these variants. A disease transmission network, DTN, which represents the transmission relationships among the hosts, is also proposed and derived from the VEG. We downloaded the genotypes of the variants recorded in GISAID, which are complete, have high coverage, and have a complete collection date from five countries: Somalia (22), Bhutan (102), Hungary (581), Iran (1334), and Nepal (1719). We ran our algorithm on these datasets to get the evolution history of the variants, build the variant evolution graph represented by the adjacency matrix, and infer the disease transmission network. Our research represents a novel and unprecedented contribution to the field of viral evolution, offering new insights and approaches not explored in prior studies.
随着时间的推移,SARS-CoV-2 病毒发生了变异,导致循环病毒株之间的遗传多样性。这种基因多样性会影响病毒的特性,包括其传播性和感染者症状的严重程度。在大流行期间,这种频繁的变异会产生大量变异株,被称为病毒准种。由于传播和存活的瓶颈限制,大多数变异都已消失。下一代测序技术的进步促进了完整病毒基因组的快速和低成本生产,使我们能够持续监测 SARS-CoV-2 基因组的演变。然而,由于序列数量庞大,要从 GISAID(全球流感数据共享计划)中推断出可靠的系统发育过程非常困难。面对这种复杂性,本研究受准种群理论的启发,提出了一种新的方法来表示 SARS-CoV-2 变种之间的进化和流行病学关系。我们的目标是建立一个变体进化图(VEG),这是一种基于变体基因型突变距离的新方法,用于模拟局部大流行区域的病毒进化。VEG 是有向无环图,不一定是树,因为一个变种可以从多个变种演化而来;在这里,顶点代表与人类宿主相关的变种基因型,边代表这些变种之间的演化关系。我们还提出了一个疾病传播网络(DTN),它代表宿主之间的传播关系,并从 VEG 中衍生出来。我们下载了 GISAID 中记录的变异体的基因型,这些变异体来自五个国家,具有完整、高覆盖率和完整的收集日期:索马里(22 个)、不丹(102 个)、匈牙利(581 个)、伊朗(1334 个)和尼泊尔(1719 个)。我们在这些数据集上运行我们的算法,以获得变异体的进化历史,构建由邻接矩阵表示的变异体进化图,并推断疾病传播网络。我们的研究为病毒进化领域做出了前所未有的新贡献,提供了以往研究未曾探索过的新见解和新方法。
{"title":"Variant Evolution Graph: Can We Infer How SARS-CoV-2 Variants are Evolving?","authors":"Badhan Das, Lenwood S. Heath","doi":"10.1101/2024.09.13.612805","DOIUrl":"https://doi.org/10.1101/2024.09.13.612805","url":null,"abstract":"The SARS-CoV-2 virus has undergone mutations over time, leading to genetic diversity among circulating viral strains. This genetic diversity can affect the characteristics of the virus, including its transmissibility and the severity of symptoms in infected individuals. During the pandemic, this frequent mutation creates an enormous cloud of variants known as viral quasispecies. Most variation is lost due to the tight bottlenecks imposed by transmission and survival. Advancements in next-generation sequencing have facilitated the rapid and cost-effective production of complete viral genomes, enabling the ongoing monitoring of the evolution of the SARS-CoV-2 genome. However, inferring a reliable phylogeny from GISAID (the Global Initiative on Sharing All Influenza Data) is daunting due to the vast number of sequences. In the face of this complexity, this research proposes a new method of representing the evolutionary and epidemiological relationships among the SARS-CoV-2 variants inspired by quasispecies theory. We aim to build a Variant Evolution Graph (VEG), a novel way to model viral evolution in a local pandemic region based on the mutational distance of the genotypes of the variants. VEG is a directed acyclic graph and not necessarily a tree because a variant can evolve from more than one variant; here, the vertices represent the genotypes of the variants associated with their human hosts, and the edges represent the evolutionary relationships among these variants. A disease transmission network, DTN, which represents the transmission relationships among the hosts, is also proposed and derived from the VEG. We downloaded the genotypes of the variants recorded in GISAID, which are complete, have high coverage, and have a complete collection date from five countries: Somalia (22), Bhutan (102), Hungary (581), Iran (1334), and Nepal (1719). We ran our algorithm on these datasets to get the evolution history of the variants, build the variant evolution graph represented by the adjacency matrix, and infer the disease transmission network. Our research represents a novel and unprecedented contribution to the field of viral evolution, offering new insights and approaches not explored in prior studies.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"65 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250370","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
An updated AL-Base reveals ranked enrichment of immunoglobulin light chain variable genes in AL amyloidosis 更新后的 AL-Base 显示,免疫球蛋白轻链可变基因在 AL 淀粉样变性病中富集排列
Pub Date : 2024-09-15 DOI: 10.1101/2024.09.11.612490
Gareth John Morgan, Allison N Nau, Sherry Wong, Brian H Spencer, Yun Shen, Axin Hua, Matthew J Bullard, Vaishali Sanchorawala, Tatiana Prokaeva
Background: Each monoclonal antibody light chain associated with AL amyloidosis has a unique sequence. Defining how these sequences lead to amyloid deposition could facilitate faster diagnosis and lead to new treatments. Methods: Light chain sequences are collected in the Boston University AL-Base repository. Monoclonal sequences from AL amyloidosis, multiple myeloma and the healthy polyclonal immune repertoire were compared to identify differences in precursor gene use, mutation frequency and physicochemical properties. Results: AL-Base now contains 2,193 monoclonal light chain sequences from plasma cell dyscrasias. Sixteen germline precursor genes were enriched in AL amyloidosis, relative to multiple myeloma and the polyclonal repertoire. Two genes, IGKV1-16 and IGLV1-36, were infrequently observed but highly enriched in AL amyloidosis. The number of mutations varied widely between light chains. AL-associated κ light chains harbored significantly more mutations compared to multiple myeloma and polyclonal sequences, whereas AL-associated λ light chains had fewer mutations. Machine learning tools designed to predict amyloid propensity were less accurate for new sequences than their original training data.Conclusions: Rarely-observed light chain variable genes may carry a high risk of AL amyloidosis. New approaches are needed to define sequence-associated risk factors for AL amyloidosis. AL-Base is a foundational resource for such studies.
背景:与 AL 淀粉样变性相关的每种单克隆抗体轻链都有独特的序列。确定这些序列是如何导致淀粉样蛋白沉积的,有助于更快地诊断并找到新的治疗方法。方法:波士顿大学 AL-Base 资料库收集了轻链序列。比较了 AL 淀粉样变性、多发性骨髓瘤和健康多克隆免疫复合物的单克隆序列,以确定前体基因使用、突变频率和理化性质的差异。结果:AL-Base目前包含2193个来自浆细胞性疾病的单克隆轻链序列。相对于多发性骨髓瘤和多克隆序列,16个种系前体基因在AL淀粉样变性中富集。IGKV1-16和IGLV1-36这两个基因在AL淀粉样变性病中并不常见,但却高度富集。不同轻链的突变数量差异很大。与多发性骨髓瘤和多克隆序列相比,AL相关的κ轻链突变明显较多,而AL相关的λ轻链突变较少。旨在预测淀粉样蛋白倾向的机器学习工具对新序列的准确性低于其原始训练数据:结论:罕见的轻链变异基因可能具有高发AL淀粉样变性病的风险。结论:罕见的轻链可变基因可能具有高风险,需要采用新方法来确定与序列相关的 AL 淀粉样变性风险因素。AL-Base 是此类研究的基础资源。
{"title":"An updated AL-Base reveals ranked enrichment of immunoglobulin light chain variable genes in AL amyloidosis","authors":"Gareth John Morgan, Allison N Nau, Sherry Wong, Brian H Spencer, Yun Shen, Axin Hua, Matthew J Bullard, Vaishali Sanchorawala, Tatiana Prokaeva","doi":"10.1101/2024.09.11.612490","DOIUrl":"https://doi.org/10.1101/2024.09.11.612490","url":null,"abstract":"Background: Each monoclonal antibody light chain associated with AL amyloidosis has a unique sequence. Defining how these sequences lead to amyloid deposition could facilitate faster diagnosis and lead to new treatments. Methods: Light chain sequences are collected in the Boston University AL-Base repository. Monoclonal sequences from AL amyloidosis, multiple myeloma and the healthy polyclonal immune repertoire were compared to identify differences in precursor gene use, mutation frequency and physicochemical properties. Results: AL-Base now contains 2,193 monoclonal light chain sequences from plasma cell dyscrasias. Sixteen germline precursor genes were enriched in AL amyloidosis, relative to multiple myeloma and the polyclonal repertoire. Two genes, IGKV1-16 and IGLV1-36, were infrequently observed but highly enriched in AL amyloidosis. The number of mutations varied widely between light chains. AL-associated κ light chains harbored significantly more mutations compared to multiple myeloma and polyclonal sequences, whereas AL-associated λ light chains had fewer mutations. Machine learning tools designed to predict amyloid propensity were less accurate for new sequences than their original training data.\u0000Conclusions: Rarely-observed light chain variable genes may carry a high risk of AL amyloidosis. New approaches are needed to define sequence-associated risk factors for AL amyloidosis. AL-Base is a foundational resource for such studies.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"15 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250369","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Associations on the Fly, a new feature aiming to facilitate exploration of the Open Targets Platform evidence 即时关联 "是一项新功能,旨在促进对 "开放目标平台 "证据的探索
Pub Date : 2024-09-14 DOI: 10.1101/2024.09.10.612089
Carlos Cruz-Castillo, Luca Fumis, Chintan Mehta, Ricardo Esteban Martinez-Osorio, Juan Maria Roldan-Romero, Helena Cornu, Prashant Uniyal, Antonio Solano-Roman, Miguel Carmona, David Ochoa, Ellen M McDonagh, Annalisa Buniello
The Open Targets Platform (https://platform.opentargets.org) is a unique, comprehensive, open-source resource supporting systematic identification and prioritisation of targets for drug discovery. The Platform combines, harmonises and integrates data from >20 diverse sources to provide target-disease associations, covering evidence derived from genetic associations, somatic mutations, known drugs, differential expression, animal models, pathways and systems biology. An in-house target identification scoring framework weighs the evidence from each data source and type, contributing to an overall score for each of the 7.8M target-disease associations. However, the previous infrastructure did not allow user-led dynamic adjustments in the contribution of different evidence types for target prioritisation, a limitation frequently raised by our user community. Furthermore, the previous Platform user interface did not support navigation and exploration of the underlying target-disease evidence on the same page, occasionally making the user journey counterintuitive. Here, we describe Associations on the Fly (AOTF), a new Platform feature - developed as part of a wider product refactoring project - to enable formulation of more flexible and impactful therapeutic hypotheses through dynamic adjustment of the weight of contributing evidence from each source, altering the prioritisation of targets.
开放靶点平台 (https://platform.opentargets.org) 是一个独特、全面的开源资源,支持系统识别和优先排序药物发现的靶点。该平台结合、协调和整合了来自 20 个不同来源的数据,提供靶点与疾病的关联,涵盖来自遗传关联、体细胞突变、已知药物、差异表达、动物模型、途径和系统生物学的证据。一个内部靶点识别评分框架对来自每个数据源和类型的证据进行权衡,为 780 万个靶点-疾病关联中的每一个给出一个总分。然而,以前的基础架构不允许用户主导动态调整不同证据类型对目标优先级的贡献,这是我们的用户社区经常提出的一个局限性。此外,以前的平台用户界面不支持在同一页面上导航和探索底层目标-疾病证据,有时会使用户的使用过程有违直觉。在此,我们将介绍 "即时关联"(AOTF)这一平台新功能,它是作为更广泛的产品重构项目的一部分而开发的,通过动态调整各来源证据的贡献权重,改变目标的优先级,从而提出更灵活、更有影响力的治疗假设。
{"title":"Associations on the Fly, a new feature aiming to facilitate exploration of the Open Targets Platform evidence","authors":"Carlos Cruz-Castillo, Luca Fumis, Chintan Mehta, Ricardo Esteban Martinez-Osorio, Juan Maria Roldan-Romero, Helena Cornu, Prashant Uniyal, Antonio Solano-Roman, Miguel Carmona, David Ochoa, Ellen M McDonagh, Annalisa Buniello","doi":"10.1101/2024.09.10.612089","DOIUrl":"https://doi.org/10.1101/2024.09.10.612089","url":null,"abstract":"The Open Targets Platform (https://platform.opentargets.org) is a unique, comprehensive, open-source resource supporting systematic identification and prioritisation of targets for drug discovery. The Platform combines, harmonises and integrates data from >20 diverse sources to provide target-disease associations, covering evidence derived from genetic associations, somatic mutations, known drugs, differential expression, animal models, pathways and systems biology. An in-house target identification scoring framework weighs the evidence from each data source and type, contributing to an overall score for each of the 7.8M target-disease associations. However, the previous infrastructure did not allow user-led dynamic adjustments in the contribution of different evidence types for target prioritisation, a limitation frequently raised by our user community. Furthermore, the previous Platform user interface did not support navigation and exploration of the underlying target-disease evidence on the same page, occasionally making the user journey counterintuitive. Here, we describe Associations on the Fly (AOTF), a new Platform feature - developed as part of a wider product refactoring project - to enable formulation of more flexible and impactful therapeutic hypotheses through dynamic adjustment of the weight of contributing evidence from each source, altering the prioritisation of targets.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"77 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142268545","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
LoVis4u: Locus Visualisation tool for comparative genomics LoVis4u:用于比较基因组学的基因座可视化工具
Pub Date : 2024-09-14 DOI: 10.1101/2024.09.11.612399
Artyom A. Egorov, Gemma C. Atkinson
Summary: Comparative genomic analysis often involves visualisation of alignments of genomic loci. While several software tools are available for this task, ranging from Python and R libraries to standalone graphical user interfaces, there is lack of a tool that offers fast, automated usage and the production of publication-ready vector images. Here we present LoVis4u, a command-line tool and Python API designed for highly customizable and fast visualisation of multiple genomic loci. LoVis4u generates vector images in PDF format based on annotation data from GenBank or GFF files. It is capable of visualising entire genomes of bacteriophages as well as plasmids and user-defined regions of longer prokaryotic genomes. Additionally, LoVis4u offers optional data processing steps to identify and highlight accessory and core genes in input sequences. Availability and Implementation: LoVis4u is implemented in Python3 and runs on Linux and MacOS. The command-line interface covers most practical use cases, while the provided Python API allows usage within a Python program, integration into external tools, and additional customisation. Source code is available at the GitHub page: github.com/art-egorov/lovis4u. Detailed documentation that includes an example-driven guide is available from the software home page: art-egorov.github.io/lovis4u.
摘要:比较基因组分析通常涉及基因组位点排列的可视化。虽然有多种软件工具可用于这一任务,从 Python 和 R 库到独立的图形用户界面,但仍缺乏一种工具可提供快速、自动化的使用并生成可用于发表的矢量图像。我们在此介绍 LoVis4u,它是一种命令行工具和 Python 应用程序接口,专为高度定制化和快速可视化多个基因组位点而设计。LoVis4u 可根据 GenBank 或 GFF 文件中的注释数据生成 PDF 格式的矢量图像。它能够可视化噬菌体的整个基因组以及质粒和用户定义的较长原核生物基因组区域。此外,LoVis4u 还提供可选的数据处理步骤,用于识别和突出显示输入序列中的附属基因和核心基因。可用性和实施:LoVis4u 由 Python3 实现,可在 Linux 和 MacOS 上运行。命令行界面涵盖了大多数实际用例,而提供的 Python API 允许在 Python 程序中使用、集成到外部工具中以及进行额外的定制。源代码可在 GitHub 页面获取:github.com/art-egorov/lovis4u。详细的文档(包括示例驱动指南)可从软件主页获取:art-egorov.github.io/lovis4u。
{"title":"LoVis4u: Locus Visualisation tool for comparative genomics","authors":"Artyom A. Egorov, Gemma C. Atkinson","doi":"10.1101/2024.09.11.612399","DOIUrl":"https://doi.org/10.1101/2024.09.11.612399","url":null,"abstract":"<strong>Summary:</strong> Comparative genomic analysis often involves visualisation of alignments of genomic loci. While several software tools are available for this task, ranging from Python and R libraries to standalone graphical user interfaces, there is lack of a tool that offers fast, automated usage and the production of publication-ready vector images. Here we present LoVis4u, a command-line tool and Python API designed for highly customizable and fast visualisation of multiple genomic loci. LoVis4u generates vector images in PDF format based on annotation data from GenBank or GFF files. It is capable of visualising entire genomes of bacteriophages as well as plasmids and user-defined regions of longer prokaryotic genomes. Additionally, LoVis4u offers optional data processing steps to identify and highlight accessory and core genes in input sequences. <strong>Availability and Implementation:</strong> LoVis4u is implemented in Python3 and runs on Linux and MacOS. The command-line interface covers most practical use cases, while the provided Python API allows usage within a Python program, integration into external tools, and additional customisation. Source code is available at the GitHub page: github.com/art-egorov/lovis4u. Detailed documentation that includes an example-driven guide is available from the software home page: art-egorov.github.io/lovis4u.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"147 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250375","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A high-throughput phenotypic screen combined with an ultra-large-scale deep learning-based virtual screening reveals novel scaffolds of antibacterial compounds 高通量表型筛选与基于深度学习的超大规模虚拟筛选相结合,揭示了新型抗菌化合物支架
Pub Date : 2024-09-14 DOI: 10.1101/2024.09.11.612340
Gabriele Scalia, Steven T Rutherford, Ziqing Lu, Kerry R Buchholz, Nicholas Skelton, Kangway Chuang, Nathaniel Diamant, Jan-Christian Huetter, Jerome Luescher, Ahn Miu, Jeff Blaney, Leo Gendelev, Elizabeth Skippington, Greg Zynda, Nia Dickson, Michal Koziarski, Yoshua Bengio, Aviv Regev, Man-Wah Tan, Tommaso Biancalani
The proliferation of multi-drug-resistant bacteria underscores an urgent need for novel antibiotics. Traditional discovery methods face challenges due to limited chemical diversity, high costs, and difficulties in identifying structurally novel compounds. Here, we explore the integration of small molecule high-throughput screening with a deep learning-based virtual screening approach to uncover new antibacterial compounds. Leveraging a diverse library of nearly 2 million small molecules, we conducted comprehensive phenotypic screening against a sensitized Escherichia coli strain that, at a low hit rate, yielded thousands of hits. We trained a deep learning model, GNEprop, to predict antibacterial activity, ensuring robustness through out-of-distribution generalization techniques. Virtual screening of over 1.4 billion compounds identified potential candidates, of which 82 exhibited antibacterial activity, illustrating a 90X improved hit rate over the high-throughput screening experiment GNEprop was trained on. Importantly, a significant portion of these newly identified compounds exhibited high dissimilarity to known antibiotics, indicating promising avenues for further exploration in antibiotic discovery.
多重耐药细菌的扩散凸显了对新型抗生素的迫切需求。由于化学多样性有限、成本高昂以及难以确定结构新颖的化合物,传统的发现方法面临着挑战。在这里,我们探讨了如何将小分子高通量筛选与基于深度学习的虚拟筛选方法相结合,以发现新的抗菌化合物。我们利用由近 200 万个小分子组成的多样化文库,针对敏化大肠杆菌菌株进行了全面的表型筛选,在低命中率的情况下,产生了数千个命中。我们训练了一个深度学习模型 GNEprop 来预测抗菌活性,并通过分布外泛化技术确保其稳健性。对超过 14 亿个化合物进行的虚拟筛选确定了潜在的候选化合物,其中 82 个具有抗菌活性,这表明与训练 GNEprop 的高通量筛选实验相比,命中率提高了 90 倍。重要的是,在这些新发现的化合物中,有很大一部分与已知抗生素具有很高的相似性,这为进一步探索抗生素发现提供了广阔的前景。
{"title":"A high-throughput phenotypic screen combined with an ultra-large-scale deep learning-based virtual screening reveals novel scaffolds of antibacterial compounds","authors":"Gabriele Scalia, Steven T Rutherford, Ziqing Lu, Kerry R Buchholz, Nicholas Skelton, Kangway Chuang, Nathaniel Diamant, Jan-Christian Huetter, Jerome Luescher, Ahn Miu, Jeff Blaney, Leo Gendelev, Elizabeth Skippington, Greg Zynda, Nia Dickson, Michal Koziarski, Yoshua Bengio, Aviv Regev, Man-Wah Tan, Tommaso Biancalani","doi":"10.1101/2024.09.11.612340","DOIUrl":"https://doi.org/10.1101/2024.09.11.612340","url":null,"abstract":"The proliferation of multi-drug-resistant bacteria underscores an urgent need for novel antibiotics. Traditional discovery methods face challenges due to limited chemical diversity, high costs, and difficulties in identifying structurally novel compounds. Here, we explore the integration of small molecule high-throughput screening with a deep learning-based virtual screening approach to uncover new antibacterial compounds. Leveraging a diverse library of nearly 2 million small molecules, we conducted comprehensive phenotypic screening against a sensitized Escherichia coli strain that, at a low hit rate, yielded thousands of hits. We trained a deep learning model, GNEprop, to predict antibacterial activity, ensuring robustness through out-of-distribution generalization techniques. Virtual screening of over 1.4 billion compounds identified potential candidates, of which 82 exhibited antibacterial activity, illustrating a 90X improved hit rate over the high-throughput screening experiment GNEprop was trained on. Importantly, a significant portion of these newly identified compounds exhibited high dissimilarity to known antibiotics, indicating promising avenues for further exploration in antibiotic discovery.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"13 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250376","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Leveraging Pretrained Deep Protein Language Model to Predict Peptide Collision Cross Section 利用预训练的深度蛋白质语言模型预测多肽碰撞截面
Pub Date : 2024-09-14 DOI: 10.1101/2024.09.11.612388
Ayano Nakai-Kasai, Kosuke Ogata, Yasushi Ishihama, Toshiyuki Tanaka
Collision cross section (CCS) of peptide ions provides an important separation dimension in liquid chromatography/tandem mass spectrometry-based proteomics that incorporates ion mobility spectrometry (IMS), and its accurate prediction is the basis for advanced proteomics workflows. This paper describes novel experimental data and a novel prediction model for challenging CCS prediction tasks including longer peptides that tend to have higher charge states. The proposed model is based on a pretrained deep protein language model. While the conventional prediction model requires training from scratch, the proposed model enables training with less amount of time owing to the use of the pretrained model as a feature extractor. Results of experiments with the novel experimental data show that the proposed model succeeds in drastically reducing the training time while maintaining the same or even better prediction performance compared with the conventional method. Our approach presents the possibility of prediction in a greener manner of various peptide properties in proteomic liquid chromatography/tandem mass spectrometry experiments.
肽离子的碰撞截面(CCS)是基于液相色谱/串联质谱(IMS)的蛋白质组学的一个重要分离维度,其准确预测是高级蛋白质组学工作流程的基础。本文介绍了新的实验数据和一种新的预测模型,该模型适用于具有挑战性的 CCS 预测任务,包括往往具有较高电荷状态的长肽。所提出的模型基于预训练的深度蛋白质语言模型。传统的预测模型需要从头开始训练,而所提出的模型由于使用了预训练模型作为特征提取器,因此训练时间更短。新实验数据的实验结果表明,与传统方法相比,所提出的模型成功地大幅缩短了训练时间,同时保持了相同甚至更好的预测性能。我们的方法为以更绿色的方式预测蛋白质组液相色谱/串联质谱实验中的各种肽特性提供了可能。
{"title":"Leveraging Pretrained Deep Protein Language Model to Predict Peptide Collision Cross Section","authors":"Ayano Nakai-Kasai, Kosuke Ogata, Yasushi Ishihama, Toshiyuki Tanaka","doi":"10.1101/2024.09.11.612388","DOIUrl":"https://doi.org/10.1101/2024.09.11.612388","url":null,"abstract":"Collision cross section (CCS) of peptide ions provides an important separation dimension in liquid chromatography/tandem mass spectrometry-based proteomics that incorporates ion mobility spectrometry (IMS), and its accurate prediction is the basis for advanced proteomics workflows. This paper describes novel experimental data and a novel prediction model for challenging CCS prediction tasks including longer peptides that tend to have higher charge states. The proposed model is based on a pretrained deep protein language model. While the conventional prediction model requires training from scratch, the proposed model enables training with less amount of time owing to the use of the pretrained model as a feature extractor. Results of experiments with the novel experimental data show that the proposed model succeeds in drastically reducing the training time while maintaining the same or even better prediction performance compared with the conventional method. Our approach presents the possibility of prediction in a greener manner of various peptide properties in proteomic liquid chromatography/tandem mass spectrometry experiments.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"7 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142268539","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
ScReNI: single-cell regulatory network inference through integrating scRNA-seq and scATAC-seq data ScReNI:通过整合 scRNA-seq 和 scATAC-seq 数据推断单细胞调控网络
Pub Date : 2024-09-14 DOI: 10.1101/2024.09.10.612385
Xueli Xu, Yanran Liang, Miaoxiu Tang, Jiongliang Wang, Xi Wang, Yixue Li, Jie Wang
Single cells exhibit heterogeneous gene expression profiles and chromatin accessibility, measurable separately via single-cell RNA sequencing (scRNA-seq) and single-cell transposase chromatin accessibility sequencing (scATAC-seq). Consequently, each cell possesses a unique gene regulatory network. However, limited methods exist for inferring cell-specific regulatory networks, particularly through the integration of scRNA-seq and scATAC-seq data. Here, we develop a novel algorithm named single-cell regulatory network inference (ScReNI), which leverages k-nearest neighbors and random forest algorithms to integrate scRNA-seq and scATAC-seq data for inferring gene regulatory networks at the single-cell level. ScReNI is built to analyze both paired and unpaired datasets for scRNA-seq and scATAC-seq. Using these two types of single-cell sequencing datasets, we validate that a higher fraction of regulatory relationships inferred by ScReNI are detected by chromatin immunoprecipitation sequencing (ChIP-seq) data. ScReNI shows superior performance in network-based cell clustering when compared to existing single-cell network inference methods. Importantly, ScReNI offers the unique function of identifying cell-enriched regulators based on each cell-specific network. In summary, ScReNI facilitates the inferences of cell-specific regulatory networks and cell-enriched regulators.
单细胞表现出异质性的基因表达谱和染色质可及性,可通过单细胞 RNA 测序(scRNA-seq)和单细胞转座酶染色质可及性测序(scATAC-seq)分别测量。因此,每个细胞都拥有独特的基因调控网络。然而,目前推断细胞特异性调控网络的方法有限,特别是通过整合 scRNA-seq 和 scATAC-seq 数据。在这里,我们开发了一种名为单细胞调控网络推断(ScReNI)的新算法,它利用k-近邻和随机森林算法整合scRNA-seq和scATAC-seq数据,推断单细胞水平的基因调控网络。ScReNI 可用于分析 scRNA-seq 和 scATAC-seq 的配对和非配对数据集。利用这两种类型的单细胞测序数据集,我们验证了染色质免疫沉淀测序(ChIP-seq)数据能检测到更多由 ScReNI 推断的调控关系。与现有的单细胞网络推断方法相比,ScReNI 在基于网络的细胞聚类方面表现出更优越的性能。重要的是,ScReNI 具有根据每个细胞特异性网络识别细胞富集调控因子的独特功能。总之,ScReNI 有助于推断细胞特异性调控网络和细胞富集调控因子。
{"title":"ScReNI: single-cell regulatory network inference through integrating scRNA-seq and scATAC-seq data","authors":"Xueli Xu, Yanran Liang, Miaoxiu Tang, Jiongliang Wang, Xi Wang, Yixue Li, Jie Wang","doi":"10.1101/2024.09.10.612385","DOIUrl":"https://doi.org/10.1101/2024.09.10.612385","url":null,"abstract":"Single cells exhibit heterogeneous gene expression profiles and chromatin accessibility, measurable separately via single-cell RNA sequencing (scRNA-seq) and single-cell transposase chromatin accessibility sequencing (scATAC-seq). Consequently, each cell possesses a unique gene regulatory network. However, limited methods exist for inferring cell-specific regulatory networks, particularly through the integration of scRNA-seq and scATAC-seq data. Here, we develop a novel algorithm named single-cell regulatory network inference (ScReNI), which leverages k-nearest neighbors and random forest algorithms to integrate scRNA-seq and scATAC-seq data for inferring gene regulatory networks at the single-cell level. ScReNI is built to analyze both paired and unpaired datasets for scRNA-seq and scATAC-seq. Using these two types of single-cell sequencing datasets, we validate that a higher fraction of regulatory relationships inferred by ScReNI are detected by chromatin immunoprecipitation sequencing (ChIP-seq) data. ScReNI shows superior performance in network-based cell clustering when compared to existing single-cell network inference methods. Importantly, ScReNI offers the unique function of identifying cell-enriched regulators based on each cell-specific network. In summary, ScReNI facilitates the inferences of cell-specific regulatory networks and cell-enriched regulators.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"3 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142268540","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A new framework for SubtiWiki, the database for the model organism Bacillus subtilis 模式生物枯草杆菌数据库 SubtiWiki 的新框架
Pub Date : 2024-09-14 DOI: 10.1101/2024.09.10.612211
Christoph Elfmann, Vincenz Dumann, Tim van den Berg, Jorg Stulke
Bacillus subtilis is a Gram-positive model bacterium and one of the most-studied and best understood organisms. The complex information resulting from its investigation is compiled in the database SubtiWiki (https://subtiwiki.uni-goettingen.de/v5) in an integrated and intuitive manner. To enhance the utility of SubtiWiki, we have added novel features such as a viewer to interrogate conserved genomic organization, a widget that shows mutant fitness data for all non-essential genes, and a widget showing protein structures, structure predictions and complex structures. Moreover, we have integrated metabolites as new entities. The new framework also includes a documented API, enabling programmatic access to data for computational tasks. Here we present the recent developments of SubtiWiki and the current state of the data for this organism.
枯草芽孢杆菌是一种革兰氏阳性模式菌,也是研究最多、最易理解的生物之一。对它的研究产生的复杂信息以综合、直观的方式编入数据库 SubtiWiki (https://subtiwiki.uni-goettingen.de/v5)。为了提高 SubtiWiki 的实用性,我们添加了一些新功能,如用于查询保守基因组组织的查看器、显示所有非必要基因突变适配性数据的小工具,以及显示蛋白质结构、结构预测和复杂结构的小工具。此外,我们还将代谢物整合为新的实体。新框架还包括一个文档化的应用程序接口(API),可以通过编程访问数据来完成计算任务。在此,我们将介绍 SubtiWiki 的最新进展以及该生物体的数据现状。
{"title":"A new framework for SubtiWiki, the database for the model organism Bacillus subtilis","authors":"Christoph Elfmann, Vincenz Dumann, Tim van den Berg, Jorg Stulke","doi":"10.1101/2024.09.10.612211","DOIUrl":"https://doi.org/10.1101/2024.09.10.612211","url":null,"abstract":"Bacillus subtilis is a Gram-positive model bacterium and one of the most-studied and best understood organisms. The complex information resulting from its investigation is compiled in the database SubtiWiki (https://subtiwiki.uni-goettingen.de/v5) in an integrated and intuitive manner. To enhance the utility of SubtiWiki, we have added novel features such as a viewer to interrogate conserved genomic organization, a widget that shows mutant fitness data for all non-essential genes, and a widget showing protein structures, structure predictions and complex structures. Moreover, we have integrated metabolites as new entities. The new framework also includes a documented API, enabling programmatic access to data for computational tasks. Here we present the recent developments of SubtiWiki and the current state of the data for this organism.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"1 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142268544","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
bioRxiv - 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