首页 > 最新文献

bioRxiv - Bioinformatics最新文献

英文 中文
Pangenomics to understand prophage dynamics in the Pectobacterium genus and the radiating lineages of P. brasiliense 通过泛基因组学了解果胶杆菌属和巴西果胶杆菌放射系的噬菌体动态
Pub Date : 2024-09-18 DOI: 10.1101/2024.09.02.610764
Lakhansing A. Pardeshi, Inge van Duivenbode, Michiel J.C. Pel, Eef M. Jonkheer, Anne Kupczok, Dick de Ridder, Sandra Smit, Theo van der Lee
Bacterial pathogens of the genus Pectobacterium are responsible for soft rot and blackleg disease in a wide range of crops and have a global impact on food production. The emergence of new lineages and their competitive succession is frequently observed in Pectobacterium species, in particular in P. brasiliense. With a focus on one such recently emerged P. brasiliense lineage in the Netherlands that causes blackleg in potatoes, we studied genome evolution in this genus using a reference-free graph-based pangenome approach. We clustered 1,977,865 proteins from 454 Pectobacterium spp. genomes into 30,156 homology groups. The Pectobacterium genus pangenome is open and its growth is mainly contributed by the accessory genome. Bacteriophage genes were enriched in the accessory genome and contributed 16% of the pangenome. Blackleg-causing P. brasiliense isolates had increased genome size with high levels of prophage integration. To study the diversity and dynamics of these prophages across the pangenome, we developed an approach to trace prophages across genomes using pangenome homology group signatures. We identified lineage-specific as well as generalist bacteriophages infecting Pectobacterium species. Our results capture the ongoing dynamics of mobile genetic elements, even in the clonal lineages. The observed lineage-specific prophage dynamics provide mechanistic insights into Pectobacterium pangenome growth and contribution to the radiating lineages of P. brasiliense.
果胶杆菌属细菌病原体是多种农作物软腐病和黑胫病的罪魁祸首,对全球粮食生产具有重要影响。在果胶杆菌物种中,尤其是在巴西果胶杆菌中,经常可以观察到新品系的出现及其竞争性演替。我们重点研究了最近在荷兰出现的导致马铃薯黑腿病的 P. brasiliense 品系,并采用基于无参照图的庞基因组方法研究了该菌属的基因组进化。我们将 454 个果胶杆菌属基因组中的 1,977,865 个蛋白质聚类为 30,156 个同源组。果胶杆菌属的庞基因组是开放的,其生长主要由附属基因组贡献。噬菌体基因富集在附属基因组中,占庞大基因组的 16%。引起黑腿病的 P. brasiliense 分离物的基因组规模增大,噬菌体整合程度较高。为了研究这些噬菌体在整个泛基因组中的多样性和动态,我们开发了一种方法,利用泛基因组同源组特征追踪噬菌体在基因组中的分布。我们发现了感染果胶杆菌的特异性噬菌体和通性噬菌体。我们的研究结果捕捉到了移动遗传因子的持续动态变化,即使在克隆品系中也是如此。观察到的特定品系的噬菌体动态为了解果胶杆菌泛基因组的生长和对巴西鹅膏菌辐射品系的贡献提供了机制性的见解。
{"title":"Pangenomics to understand prophage dynamics in the Pectobacterium genus and the radiating lineages of P. brasiliense","authors":"Lakhansing A. Pardeshi, Inge van Duivenbode, Michiel J.C. Pel, Eef M. Jonkheer, Anne Kupczok, Dick de Ridder, Sandra Smit, Theo van der Lee","doi":"10.1101/2024.09.02.610764","DOIUrl":"https://doi.org/10.1101/2024.09.02.610764","url":null,"abstract":"Bacterial pathogens of the genus Pectobacterium are responsible for soft rot and blackleg disease in a wide range of crops and have a global impact on food production. The emergence of new lineages and their competitive succession is frequently observed in Pectobacterium species, in particular in P. brasiliense. With a focus on one such recently emerged P. brasiliense lineage in the Netherlands that causes blackleg in potatoes, we studied genome evolution in this genus using a reference-free graph-based pangenome approach. We clustered 1,977,865 proteins from 454 Pectobacterium spp. genomes into 30,156 homology groups. The Pectobacterium genus pangenome is open and its growth is mainly contributed by the accessory genome. Bacteriophage genes were enriched in the accessory genome and contributed 16% of the pangenome. Blackleg-causing P. brasiliense isolates had increased genome size with high levels of prophage integration. To study the diversity and dynamics of these prophages across the pangenome, we developed an approach to trace prophages across genomes using pangenome homology group signatures. We identified lineage-specific as well as generalist bacteriophages infecting Pectobacterium species. Our results capture the ongoing dynamics of mobile genetic elements, even in the clonal lineages. The observed lineage-specific prophage dynamics provide mechanistic insights into Pectobacterium pangenome growth and contribution to the radiating lineages of P. brasiliense.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"29 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250168","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
Mouse-Geneformer: A Deep Learning Model for Mouse Single-Cell Transcriptome and Its Cross-Species Utility Mouse-Geneformer:小鼠单细胞转录组深度学习模型及其跨物种实用性
Pub Date : 2024-09-18 DOI: 10.1101/2024.09.09.611960
Keita Ito, Tsubasa Hirakawa, Shuji Shigenobu, Hironobu Fujiyoshi, Takayoshi Yamashita
Deep learning techniques are increasingly utilized to analyze large-scale single-cell RNA sequencing (scRNA-seq) data, offering valuable insights from complex transcriptome datasets. Geneformer, a pre-trained model using a Transformer Encoder architecture and human scRNA-seq datasets, has demonstrated remarkable success in human transcriptome analysis. However, given the prominence of the mouse, Mus musculus, as a primary mammalian model in biological and medical research, there is an acute need for a mouse-specific version of Geneformer. In this study, we developed a mouse-specific Geneformer (mouse-Geneformer) by constructing a large transcriptome dataset consisting of 21 million mouse scRNA-seq profiles and pre-training Geneformer on this dataset. The mouse-Geneformer effectively models the mouse transcriptome and, upon fine-tuning for downstream tasks, enhances the accuracy of cell type classification. In silico perturbation experiments using mouse-Geneformer successfully identified disease-causing genes that have been validated in in vivo experiments. These results demonstrate the feasibility of analyzing mouse data with mouse-Geneformer and highlight the robustness of the Geneformer architecture, applicable to any species with large-scale transcriptome data available. Furthermore, we found that mouse-Geneformer can analyze human transcriptome data in a cross-species manner. After the ortholog-based gene name conversion, the analysis of human scRNA-seq data using mouse-Geneformer, followed by fine-tuning with human data, achieved cell type classification accuracy comparable to that obtained using the original human Geneformer. In in silico simulation experiments using human disease models, we obtained results similar to human-Geneformer for the myocardial infarction model but only partially consistent results for the COVID-19 model, a trait unique to humans (laboratory mice are not susceptible to SARS-CoV-2). These findings suggest the potential for cross-species application of the Geneformer model while emphasizing the importance of species-specific models for capturing the full complexity of disease mechanisms. Despite the existence of the original Geneformer tailored for humans, human research could benefit from mouse-Geneformer due to its inclusion of samples that are ethically or technically inaccessible for humans, such as embryonic tissues and certain disease models. Additionally, this cross-species approach indicates potential use for non-model organisms, where obtaining large-scale single-cell transcriptome data is challenging.
深度学习技术越来越多地被用于分析大规模单细胞 RNA 测序(scRNA-seq)数据,从而为复杂的转录组数据集提供有价值的见解。Geneformer 是一种使用 Transformer 编码器架构和人类 scRNA-seq 数据集进行预训练的模型,在人类转录组分析中取得了显著的成功。然而,鉴于小鼠(Mus musculus)作为主要哺乳动物模型在生物和医学研究中的突出地位,我们迫切需要一个小鼠专用版的 Geneformer。在这项研究中,我们构建了一个由 2100 万个小鼠 scRNA-seq 图谱组成的大型转录组数据集,并在该数据集上对 Geneformer 进行了预训练,从而开发出了小鼠专用的 Geneformer(小鼠-Geneformer)。小鼠基因改造器有效地模拟了小鼠转录组,并根据下游任务进行了微调,提高了细胞类型分类的准确性。使用小鼠基因改造器进行的硅学扰动实验成功鉴定出了致病基因,这些基因已在体内实验中得到验证。这些结果证明了用小鼠-Geneformer 分析小鼠数据的可行性,并突出了 Geneformer 架构的稳健性,它适用于任何有大规模转录组数据的物种。此外,我们还发现小鼠-Geneformer 可以跨物种分析人类转录组数据。在基于直向同源物的基因名称转换之后,使用小鼠-Geneformer 分析人类 scRNA-seq 数据,再根据人类数据进行微调,其细胞类型分类准确率与使用原始人类 Geneformer 所获得的准确率相当。在使用人类疾病模型进行的硅学模拟实验中,我们在心肌梗塞模型中得到了与人类基因改造器相似的结果,但在 COVID-19 模型中只得到了部分一致的结果,这是人类特有的特征(实验鼠对 SARS-CoV-2 病毒不敏感)。这些发现表明,Geneformer 模型具有跨物种应用的潜力,同时也强调了物种特异性模型对于捕捉疾病机制全部复杂性的重要性。尽管最初的 Geneformer 是为人类量身定做的,但由于小鼠-Geneformer 包含了人类在伦理或技术上无法获得的样本,如胚胎组织和某些疾病模型,因此人类研究可以从小鼠-Geneformer 中获益。此外,这种跨物种方法还显示了在非模式生物体中的潜在用途,因为在非模式生物体中获取大规模单细胞转录组数据具有挑战性。
{"title":"Mouse-Geneformer: A Deep Learning Model for Mouse Single-Cell Transcriptome and Its Cross-Species Utility","authors":"Keita Ito, Tsubasa Hirakawa, Shuji Shigenobu, Hironobu Fujiyoshi, Takayoshi Yamashita","doi":"10.1101/2024.09.09.611960","DOIUrl":"https://doi.org/10.1101/2024.09.09.611960","url":null,"abstract":"Deep learning techniques are increasingly utilized to analyze large-scale single-cell RNA sequencing (scRNA-seq) data, offering valuable insights from complex transcriptome datasets. Geneformer, a pre-trained model using a Transformer Encoder architecture and human scRNA-seq datasets, has demonstrated remarkable success in human transcriptome analysis. However, given the prominence of the mouse, Mus musculus, as a primary mammalian model in biological and medical research, there is an acute need for a mouse-specific version of Geneformer. In this study, we developed a mouse-specific Geneformer (mouse-Geneformer) by constructing a large transcriptome dataset consisting of 21 million mouse scRNA-seq profiles and pre-training Geneformer on this dataset. The mouse-Geneformer effectively models the mouse transcriptome and, upon fine-tuning for downstream tasks, enhances the accuracy of cell type classification. In silico perturbation experiments using mouse-Geneformer successfully identified disease-causing genes that have been validated in in vivo experiments. These results demonstrate the feasibility of analyzing mouse data with mouse-Geneformer and highlight the robustness of the Geneformer architecture, applicable to any species with large-scale transcriptome data available. Furthermore, we found that mouse-Geneformer can analyze human transcriptome data in a cross-species manner. After the ortholog-based gene name conversion, the analysis of human scRNA-seq data using mouse-Geneformer, followed by fine-tuning with human data, achieved cell type classification accuracy comparable to that obtained using the original human Geneformer. In in silico simulation experiments using human disease models, we obtained results similar to human-Geneformer for the myocardial infarction model but only partially consistent results for the COVID-19 model, a trait unique to humans (laboratory mice are not susceptible to SARS-CoV-2). These findings suggest the potential for cross-species application of the Geneformer model while emphasizing the importance of species-specific models for capturing the full complexity of disease mechanisms. Despite the existence of the original Geneformer tailored for humans, human research could benefit from mouse-Geneformer due to its inclusion of samples that are ethically or technically inaccessible for humans, such as embryonic tissues and certain disease models. Additionally, this cross-species approach indicates potential use for non-model organisms, where obtaining large-scale single-cell transcriptome data is challenging.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"50 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250171","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
Network-based estimation of therapeutic efficacy and adverse reaction potential for prioritisation of anti-cancer drug combinations 基于网络的疗效和不良反应可能性评估,用于确定抗癌药物组合的优先次序
Pub Date : 2024-09-18 DOI: 10.1101/2024.09.17.613439
Arindam Ghosh, Vittorio Fortino
Drug combinations, although a key therapeutic agent against cancer, are yet to reach their full applicability potential due to the challenges involved in the identification of effective and safe drug pairs. In vitro or in vivo screening would have been the optimal approach if combinatorial explosion was not an issue. In silico methods, on the other hand, can enable rapid screening of drug pairs to prioritise for experimental validation. Here we present a novel network medicine approach that systematically models the proximity of drug targets to disease-associated genes and adverse effect-associated genes, through the combination of network propagation algorithm and gene set enrichment analysis. The proposed approach is applied in the context of identifying effective drug combinations for cancer treatment starting from a training set of drug combinations curated from DrugComb and DrugBank databases. We observed that effective drug combinations usually enrich disease-related gene sets while adverse drug combinations enrich adverse-effect gene sets. We use this observation to systematically train classifiers distinguishing drug combinations with higher therapeutic effects and no known adverse reaction from combinations with lower therapeutic effects and potential adverse reactions in six cancer types. The approach is tested and validated using drug combinations curated from in vitro screening data and clinical reports. Trained classification models are also used to identify novel potential anti-cancer drug combinations for experimental validation. We believe our framework would be a key addition to the anti-cancer drug combination identification pipeline by enabling rapid yet robust estimation of therapeutic efficacy or adverse reaction potential.
联合用药虽然是治疗癌症的关键药物,但由于在确定有效和安全的药物配对方面存在挑战,因此尚未充分发挥其应用潜力。如果组合爆炸不是问题,体外或体内筛选将是最佳方法。硅学方法则可以快速筛选药物配对,为实验验证确定优先次序。在这里,我们提出了一种新的网络医学方法,通过结合网络传播算法和基因组富集分析,系统地模拟药物靶点与疾病相关基因和不良反应相关基因的接近程度。我们将所提出的方法应用于从 DrugComb 和 DrugBank 数据库中收集的药物组合训练集开始,识别治疗癌症的有效药物组合。我们观察到,有效的药物组合通常会丰富与疾病相关的基因集,而不良的药物组合则会丰富不良反应基因集。我们利用这一观察结果系统地训练了分类器,以区分六种癌症类型中疗效较高且无已知不良反应的药物组合与疗效较低且有潜在不良反应的药物组合。我们使用从体外筛选数据和临床报告中筛选出的药物组合对该方法进行了测试和验证。经过训练的分类模型还用于识别新的潜在抗癌药物组合,以进行实验验证。我们相信,我们的框架将成为抗癌药物组合识别管道的重要补充,能够快速而稳健地估计疗效或不良反应的可能性。
{"title":"Network-based estimation of therapeutic efficacy and adverse reaction potential for prioritisation of anti-cancer drug combinations","authors":"Arindam Ghosh, Vittorio Fortino","doi":"10.1101/2024.09.17.613439","DOIUrl":"https://doi.org/10.1101/2024.09.17.613439","url":null,"abstract":"Drug combinations, although a key therapeutic agent against cancer, are yet to reach their full applicability potential due to the challenges involved in the identification of effective and safe drug pairs. In vitro or in vivo screening would have been the optimal approach if combinatorial explosion was not an issue. In silico methods, on the other hand, can enable rapid screening of drug pairs to prioritise for experimental validation. Here we present a novel network medicine approach that systematically models the proximity of drug targets to disease-associated genes and adverse effect-associated genes, through the combination of network propagation algorithm and gene set enrichment analysis. The proposed approach is applied in the context of identifying effective drug combinations for cancer treatment starting from a training set of drug combinations curated from DrugComb and DrugBank databases. We observed that effective drug combinations usually enrich disease-related gene sets while adverse drug combinations enrich adverse-effect gene sets. We use this observation to systematically train classifiers distinguishing drug combinations with higher therapeutic effects and no known adverse reaction from combinations with lower therapeutic effects and potential adverse reactions in six cancer types. The approach is tested and validated using drug combinations curated from in vitro screening data and clinical reports. Trained classification models are also used to identify novel potential anti-cancer drug combinations for experimental validation. We believe our framework would be a key addition to the anti-cancer drug combination identification pipeline by enabling rapid yet robust estimation of therapeutic efficacy or adverse reaction potential.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"15 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250170","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 Guide for Active Learning in Synergistic Drug Discovery 协同药物发现中的主动学习指南
Pub Date : 2024-09-18 DOI: 10.1101/2024.09.13.612819
Shuhui Wang, Alexandre Allauzen, Philippe Nghe, Vaitea Opuu
Synergistic drug combination screening is a promising strategy in drug discovery, but it involves navigating a costly and complex search space. While AI, particularly deep learning, has advanced synergy predictions, its effectiveness is limited by the low occurrence of synergistic drug pairs. Active learning, which integrates experimental testing into the learning process, has been proposed to address this challenge. In this work, we explore the key components of active learning to provide recommendations for its implementation. We find that molecular encoding has a limited impact on performance, while the cellular environment features significantly enhance predictions. Additionally, active learning can discover 60% of synergistic drug pairs with only exploring 10% of combinatorial space. The synergy yield ratio is observed to be even higher with smaller batch sizes, where dynamic tuning of the exploration-exploitation strategy can further enhance performance.
协同药物组合筛选是一种前景广阔的药物发现策略,但需要在昂贵而复杂的搜索空间中进行导航。虽然人工智能,尤其是深度学习,已经推进了协同作用的预测,但其有效性受到协同药物配对出现率低的限制。为了应对这一挑战,有人提出了将实验测试融入学习过程的主动学习。在这项工作中,我们探讨了主动学习的关键要素,为其实施提供了建议。我们发现,分子编码对性能的影响有限,而细胞环境特征则能显著提高预测能力。此外,主动学习只需探索 10%的组合空间,就能发现 60% 的协同药物配对。据观察,在批量规模较小的情况下,协同收益率甚至更高,而动态调整探索-开发策略可进一步提高性能。
{"title":"A Guide for Active Learning in Synergistic Drug Discovery","authors":"Shuhui Wang, Alexandre Allauzen, Philippe Nghe, Vaitea Opuu","doi":"10.1101/2024.09.13.612819","DOIUrl":"https://doi.org/10.1101/2024.09.13.612819","url":null,"abstract":"Synergistic drug combination screening is a promising strategy in drug discovery, but it involves navigating a costly and complex search space. While AI, particularly deep learning, has advanced synergy predictions, its effectiveness is limited by the low occurrence of synergistic drug pairs. Active learning, which integrates experimental testing into the learning process, has been proposed to address this challenge. In this work, we explore the key components of active learning to provide recommendations for its implementation. We find that molecular encoding has a limited impact on performance, while the cellular environment features significantly enhance predictions. Additionally, active learning can discover 60% of synergistic drug pairs with only exploring 10% of combinatorial space. The synergy yield ratio is observed to be even higher with smaller batch sizes, where dynamic tuning of the exploration-exploitation strategy can further enhance performance.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"37 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250460","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
Pan-Cancer Genetic Analysis of Mitochondrial DNA Repair Gene Set 线粒体 DNA 修复基因组的泛癌症遗传分析
Pub Date : 2024-09-17 DOI: 10.1101/2024.09.14.613048
Angela Dong, Ayana Meegol Rasteh, Hengrui Liu
Background: The mitochondrial DNA repair has gained attention for its potential impact on pan-cancer genetic analysis. This study investigates the clinical relevance of mitochondrial DNA repair genes: PARP1, DNA 2, PRIMPOL, TP53, MGME1. Methods: Using multi-omics profiling data and Gene Set Cancer Analysis (GSCA) with normalized SEM mRNA expression, this research analyzes differential expression, gene mutation, and drug correlation. Results: TP53 was the most commonly mutated mitochondrial-related gene in cancer, with UCS and OV having the highest mutation rates. CPG mutations linked to lowest survival rates. Breast cancer, with various subtypes, was potentially influenced by mitochondrial DNA repair genes. ACC was shown to be high in gene survival analysis. BRCA, USC, LUCS, COAD, and OV showed CNV levels impacting survival. A negative gene expression-methylation correlation was observed and was weakest in KIRC. Mitochondrial DNA repair genes were linked to Cell cycle_A activation. A weak correlation was found between immune infiltration and mitochondrial genes. Few drug compounds were shown to be affected by mitochondrial-related genes. Conclusion: Understanding mitochondrial-related genes could redefine cancer diagnosis, and prognosis, and serve as therapeutic biomarkers, potentially altering cancer cell behavior and treatment outcomes.
背景:线粒体 DNA 修复因其对泛癌症基因分析的潜在影响而备受关注。本研究调查了线粒体 DNA 修复基因的临床相关性:PARP1、DNA 2、PRIMPOL、TP53、MGME1。研究方法本研究利用多组学剖析数据和基因组癌症分析(GSCA)与归一化 SEM mRNA 表达,分析差异表达、基因突变和药物相关性。结果发现TP53是癌症中最常见的线粒体相关基因突变,其中UCS和OV的突变率最高。CPG突变与最低生存率有关。不同亚型的乳腺癌可能受到线粒体 DNA 修复基因的影响。在基因存活率分析中,ACC 的存活率较高。BRCA、USC、LUCS、COAD和OV显示出影响生存的CNV水平。基因表达与甲基化呈负相关,KIRC的相关性最弱。线粒体 DNA 修复基因与细胞周期_A 的激活有关。免疫渗透与线粒体基因之间存在微弱的相关性。很少有药物化合物对线粒体相关基因产生影响。结论了解线粒体相关基因可重新定义癌症诊断和预后,并可作为治疗生物标志物,从而有可能改变癌细胞行为和治疗效果。
{"title":"Pan-Cancer Genetic Analysis of Mitochondrial DNA Repair Gene Set","authors":"Angela Dong, Ayana Meegol Rasteh, Hengrui Liu","doi":"10.1101/2024.09.14.613048","DOIUrl":"https://doi.org/10.1101/2024.09.14.613048","url":null,"abstract":"Background: The mitochondrial DNA repair has gained attention for its potential impact on pan-cancer genetic analysis. This study investigates the clinical relevance of mitochondrial DNA repair genes: PARP1, DNA 2, PRIMPOL, TP53, MGME1. Methods: Using multi-omics profiling data and Gene Set Cancer Analysis (GSCA) with normalized SEM mRNA expression, this research analyzes differential expression, gene mutation, and drug correlation. Results: TP53 was the most commonly mutated mitochondrial-related gene in cancer, with UCS and OV having the highest mutation rates. CPG mutations linked to lowest survival rates. Breast cancer, with various subtypes, was potentially influenced by mitochondrial DNA repair genes. ACC was shown to be high in gene survival analysis. BRCA, USC, LUCS, COAD, and OV showed CNV levels impacting survival. A negative gene expression-methylation correlation was observed and was weakest in KIRC. Mitochondrial DNA repair genes were linked to Cell cycle_A activation. A weak correlation was found between immune infiltration and mitochondrial genes. Few drug compounds were shown to be affected by mitochondrial-related genes. Conclusion: Understanding mitochondrial-related genes could redefine cancer diagnosis, and prognosis, and serve as therapeutic biomarkers, potentially altering cancer cell behavior and treatment outcomes.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"138 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250179","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 Comparison of Tokenization Impact in Attention Based and State Space Genomic Language Models 基于注意力和状态空间的基因组语言模型中标记化影响的比较
Pub Date : 2024-09-17 DOI: 10.1101/2024.09.09.612081
LeAnn M Lindsey, Nicole L Pershing, Anisa Habib, W. Zac Stephens, Anne J Blaschke, Hari Sundar
Genomic language models have recently emerged as powerful tools to decode and interpret genetic sequences. Existing genomic language models have utilized various tokenization methods including character tokenization, overlapping and non-overlapping k-mer tokenization, and byte-pair encoding, a method widely used in natural language models. Genomic models have significant differences from natural language and protein language models because of their low character variability, complex and overlapping features, and inconsistent directionality. These differences make sub-word tokenization in genomic language models significantly different from traditional language models. This study explores the impact of tokenization in attention-based and state-space genomic language models by evaluating their downstream performance on various fine-tuning tasks. We propose new definitions for fertility, the token per word ratio, in the context of genomic language models, and introduce tokenization parity, which measures how consistently a tokenizer parses homologous sequences. We also perform an ablation study on the state-space model, Mamba, to evaluate the impact of character-based tokenization compared to byte-pair encoding. Our results indicate that the choice of tokenizer significantly impacts model performance and that when experiments control for input sequence length, character tokenization is the best choice in state-space models for all evaluated task categories except epigenetic mark prediction.
基因组语言模型近来已成为解码和解释基因序列的强大工具。现有的基因组语言模型采用了各种标记化方法,包括字符标记化、重叠和非重叠 k-mer 标记化以及字节对编码(一种广泛用于自然语言模型的方法)。基因组模型与自然语言和蛋白质语言模型有很大不同,因为它们的字符变异性低、特征复杂且相互重叠、方向性不一致。这些差异使得基因组语言模型中的子词标记化与传统语言模型有很大不同。本研究通过评估基于注意力的基因组语言模型和状态空间基因组语言模型在各种微调任务中的下游性能,探讨了标记化对它们的影响。我们为基因组语言模型中的 "生育率"(token per word ratio)提出了新的定义,并引入了标记化奇偶性(tokenization parity),以衡量标记化器解析同源序列的一致性。我们还对状态空间模型 Mamba 进行了消融研究,以评估基于字符的标记化与字节对编码相比所产生的影响。我们的结果表明,标记化器的选择对模型性能有显著影响,而且当实验控制输入序列长度时,字符标记化是状态空间模型中除表观遗传标记预测外所有评估任务类别的最佳选择。
{"title":"A Comparison of Tokenization Impact in Attention Based and State Space Genomic Language Models","authors":"LeAnn M Lindsey, Nicole L Pershing, Anisa Habib, W. Zac Stephens, Anne J Blaschke, Hari Sundar","doi":"10.1101/2024.09.09.612081","DOIUrl":"https://doi.org/10.1101/2024.09.09.612081","url":null,"abstract":"Genomic language models have recently emerged as powerful tools to decode and interpret genetic sequences. Existing genomic language models have utilized various tokenization methods including character tokenization, overlapping and non-overlapping k-mer tokenization, and byte-pair encoding, a method widely used in natural language models. Genomic models have significant differences from natural language and protein language models because of their low character variability, complex and overlapping features, and inconsistent directionality. These differences make sub-word tokenization in genomic language models significantly different from traditional language models. This study explores the impact of tokenization in attention-based and state-space genomic language models by evaluating their downstream performance on various fine-tuning tasks. We propose new definitions for fertility, the token per word ratio, in the context of genomic language models, and introduce tokenization parity, which measures how consistently a tokenizer parses homologous sequences. We also perform an ablation study on the state-space model, Mamba, to evaluate the impact of character-based tokenization compared to byte-pair encoding. Our results indicate that the choice of tokenizer significantly impacts model performance and that when experiments control for input sequence length, character tokenization is the best choice in state-space models for all evaluated task categories except epigenetic mark prediction.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"24 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250175","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
Accelerating Ligand Discovery for Insect Odorant Receptors 加速发现昆虫气味受体的配体
Pub Date : 2024-09-17 DOI: 10.1101/2024.09.12.612620
Arthur Comte, Maxence Lalis, Ludivine Brajon, Riccardo Moracci, Nicolas Montagné, Jérémie Topin, Emmanuelle Jacquin Joly, Sébastien Fiorucci
Odorant receptors (ORs) are main actors of the insects peripheral olfactory system, making them prime targets for pest control through olfactory disruption. Traditional methods employed in the context of chemical ecology for identifying OR ligands rely on analyzing compounds present in the insect′s environment or screening molecules with structures similar to known ligands. However, these approaches can be time-consuming and constrained by the limited chemical space they explore. Recent advances in OR structural understanding, coupled with scientific breakthroughs in protein structure prediction, have facilitated the application of structure-based virtual screening (SBVS) techniques for accelerated ligand discovery. Here, we report the first successful application of SBVS to insect ORs. We developed a unique workflow that combines molecular docking predictions, in vivo validation and behavioral assays to identify new behaviorally active volatiles for non-pheromonal receptors. This work serves as a proof of concept, laying the groundwork for future studies and highlighting the need for improved computational approaches. Finally, we propose a simple model for predicting receptor response spectra based on the hypothesis that the binding pocket properties partially encode this information, as suggested by our results on Spodoptera littoralis ORs.
气味受体(ORs)是昆虫外周嗅觉系统的主要角色,因此成为通过嗅觉干扰控制害虫的主要目标。在化学生态学背景下,识别气味受体配体的传统方法依赖于分析昆虫环境中存在的化合物或筛选与已知配体结构相似的分子。然而,这些方法耗时长,而且受限于其探索的有限化学空间。近来对 OR 结构理解的进步,加上蛋白质结构预测方面的科学突破,促进了基于结构的虚拟筛选(SBVS)技术在加速配体发现方面的应用。在此,我们报告了 SBVS 在昆虫 ORs 中的首次成功应用。我们开发了一种独特的工作流程,将分子对接预测、体内验证和行为试验结合在一起,为非外激素受体鉴定新的行为活性挥发物。这项工作可作为概念验证,为今后的研究奠定基础,并强调了改进计算方法的必要性。最后,我们提出了一个预测受体反应谱的简单模型,该模型基于这样一个假设:结合口袋的特性部分地编码了这一信息,正如我们对滨海蝶ORs的研究结果所表明的那样。
{"title":"Accelerating Ligand Discovery for Insect Odorant Receptors","authors":"Arthur Comte, Maxence Lalis, Ludivine Brajon, Riccardo Moracci, Nicolas Montagné, Jérémie Topin, Emmanuelle Jacquin Joly, Sébastien Fiorucci","doi":"10.1101/2024.09.12.612620","DOIUrl":"https://doi.org/10.1101/2024.09.12.612620","url":null,"abstract":"Odorant receptors (ORs) are main actors of the insects peripheral olfactory system, making them prime targets for pest control through olfactory disruption. Traditional methods employed in the context of chemical ecology for identifying OR ligands rely on analyzing compounds present in the insect′s environment or screening molecules with structures similar to known ligands. However, these approaches can be time-consuming and constrained by the limited chemical space they explore. Recent advances in OR structural understanding, coupled with scientific breakthroughs in protein structure prediction, have facilitated the application of structure-based virtual screening (SBVS) techniques for accelerated ligand discovery. Here, we report the first successful application of SBVS to insect ORs. We developed a unique workflow that combines molecular docking predictions, in vivo validation and behavioral assays to identify new behaviorally active volatiles for non-pheromonal receptors. This work serves as a proof of concept, laying the groundwork for future studies and highlighting the need for improved computational approaches. Finally, we propose a simple model for predicting receptor response spectra based on the hypothesis that the binding pocket properties partially encode this information, as suggested by our results on Spodoptera littoralis ORs.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250178","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 Evolutionary Statistics Toolkit for Simplified Sequence Analysis on Web with Client-Side Processing 利用客户端处理简化网络序列分析的进化统计工具包
Pub Date : 2024-09-17 DOI: 10.1101/2024.08.01.606148
Alper Karagöl, Taner Karagöl
We present the Evolutionary Statistics Toolkit, a user-friendly web-based platform designed for specialized analysis of genetic sequences, which integrates multiple evolutionary statistics. The toolkit focuses on a selection of specialized tools, including Tajima's D calculator with Site Frequency Spectrum (SFS), Shannon's Entropy (H), alignment re-formatting, HGSV to FASTA conversion, pair-wise frequency analysis, FASTA to SEQRES, RNA 2D structure alignment, Kyte-Doolittle hydrophilicity plot tool and kurtosis coefficient calculator. Tajima's D is calculated using the reference formula: D = (π - θW)/sqrt(VD), where π corresponds to the average number of differences, θW is Watterson's estimator of θ, and VD is the variance of π - θW. Shannon's Entropy is defined as H = -∑ pi* log2(pi), where pi is the probability of occurrence of each unique character (nucleotide or amino acid) in the sequence. The toolkit facilitates streamlined workflows for early researchers in evolutionary biology, genomics, and related fields. With comparing with existing codes, we propose it also emerges as an educational interactive website for beginners in evolutionary statistics. The source code for each tool in the toolkit is available through GitHub links provided on the website. This open-source approach allows users to inspect the code, suggest improvements, or further adapt the tools for their specific usage and research needs. This article describes the functionalities, and validation of each tool within the platform, along with comparison with accessible existing statistical utilities. The toolkit is freely accessible on: https://www.alperkaragol.com/toolkit
我们推出了进化统计工具包(Evolutionary Statistics Toolkit),这是一个用户友好型网络平台,专为遗传序列的专业分析而设计,集成了多种进化统计功能。该工具包侧重于精选的专业工具,包括田岛D计算器与位点频率谱(SFS)、香农熵(H)、排列重格式化、HGSV到FASTA转换、成对频率分析、FASTA到SEQRES、RNA二维结构排列、Kyte-Doolittle亲水性绘图工具和峰度系数计算器。田岛 D 使用参考公式计算:D = (π - θW)/sqrt(VD),其中 π 对应于平均差异数,θW 是 Watterson 对 θ 的估计值,VD 是 π - θW 的方差。香农熵的定义为 H = -∑ pi* log2(pi),其中 pi 是序列中每个唯一特征(核苷酸或氨基酸)出现的概率。该工具包为进化生物学、基因组学及相关领域的早期研究人员简化了工作流程。与现有代码相比,我们建议该工具包还可作为进化统计初学者的教育互动网站。工具包中每个工具的源代码都可以通过网站上提供的 GitHub 链接获取。这种开源方法允许用户检查代码、提出改进建议或进一步调整工具,以满足其特定的使用和研究需求。本文介绍了该平台中每个工具的功能和验证,以及与现有统计工具的比较。该工具包可在以下网址免费访问: https://www.alperkaragol.com/toolkit
{"title":"An Evolutionary Statistics Toolkit for Simplified Sequence Analysis on Web with Client-Side Processing","authors":"Alper Karagöl, Taner Karagöl","doi":"10.1101/2024.08.01.606148","DOIUrl":"https://doi.org/10.1101/2024.08.01.606148","url":null,"abstract":"We present the Evolutionary Statistics Toolkit, a user-friendly web-based platform designed for specialized analysis of genetic sequences, which integrates multiple evolutionary statistics. The toolkit focuses on a selection of specialized tools, including Tajima's D calculator with Site Frequency Spectrum (SFS), Shannon's Entropy (H), alignment re-formatting, HGSV to FASTA conversion, pair-wise frequency analysis, FASTA to SEQRES, RNA 2D structure alignment, Kyte-Doolittle hydrophilicity plot tool and kurtosis coefficient calculator. Tajima's D is calculated using the reference formula: D = (π - θ<sub>W</sub>)/sqrt(V<sub>D</sub>), where π corresponds to the average number of differences, θ<sub>W</sub> is Watterson's estimator of θ, and V<sub>D</sub> is the variance of π - θ<sub>W</sub>. Shannon's Entropy is defined as H = -∑ p<sub>i</sub>* log<sub>2</sub>(p<sub>i</sub>), where p<sub>i</sub> is the probability of occurrence of each unique character (nucleotide or amino acid) in the sequence. The toolkit facilitates streamlined workflows for early researchers in evolutionary biology, genomics, and related fields. With comparing with existing codes, we propose it also emerges as an educational interactive website for beginners in evolutionary statistics. The source code for each tool in the toolkit is available through GitHub links provided on the website. This open-source approach allows users to inspect the code, suggest improvements, or further adapt the tools for their specific usage and research needs. This article describes the functionalities, and validation of each tool within the platform, along with comparison with accessible existing statistical utilities. The toolkit is freely accessible on: https://www.alperkaragol.com/toolkit","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"15 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250176","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
SVbyEye: A visual tool to characterize structural variation among whole-genome assemblies SVbyEye:表征全基因组组装结构变异的可视化工具
Pub Date : 2024-09-17 DOI: 10.1101/2024.09.11.612418
David Porubsky, Xavi Guitart, DongAhn Yoo, Philip C. Dishuck, William T. Harvey, Evan E. Eichler
MotivationWe are now in the era of being able to routinely generate highly contiguous (near telomere-to-telomere) genome assemblies of human and nonhuman species. Complex structural variation and regions of rapid evolutionary turnover are being discovered for the first time. Thus, efficient and informative visualization tools are needed to evaluate and directly observe structural differences between two or more genomes.ResultsWe developed SVbyEye, an open-source R package to visualize and annotate sequence-to-sequence alignments along with various functionalities to process alignments in PAF format. The tool facilitates the characterization of complex structural variants in the context of sequence homology helping resolve the mechanisms underlying their formation.Availability and implementationSVbyEye is available at https://github.com/daewoooo/SVbyEye.
动机我们现在正处于能够常规生成高度连续(接近端粒到端粒)的人类和非人类物种基因组组装的时代。我们首次发现了复杂的结构变异和快速进化更替的区域。因此,我们需要高效且信息丰富的可视化工具来评估和直接观察两个或多个基因组之间的结构差异。结果我们开发了 SVbyEye,这是一个开源的 R 软件包,用于可视化和注释序列到序列的比对以及处理 PAF 格式比对的各种功能。该工具有助于在序列同源性的背景下描述复杂的结构变异,帮助解决其形成的机制问题。可用性和实现SVbyEye可在https://github.com/daewoooo/SVbyEye。
{"title":"SVbyEye: A visual tool to characterize structural variation among whole-genome assemblies","authors":"David Porubsky, Xavi Guitart, DongAhn Yoo, Philip C. Dishuck, William T. Harvey, Evan E. Eichler","doi":"10.1101/2024.09.11.612418","DOIUrl":"https://doi.org/10.1101/2024.09.11.612418","url":null,"abstract":"Motivation\u0000We are now in the era of being able to routinely generate highly contiguous (near telomere-to-telomere) genome assemblies of human and nonhuman species. Complex structural variation and regions of rapid evolutionary turnover are being discovered for the first time. Thus, efficient and informative visualization tools are needed to evaluate and directly observe structural differences between two or more genomes.\u0000Results\u0000We developed SVbyEye, an open-source R package to visualize and annotate sequence-to-sequence alignments along with various functionalities to process alignments in PAF format. The tool facilitates the characterization of complex structural variants in the context of sequence homology helping resolve the mechanisms underlying their formation.\u0000Availability and implementation\u0000SVbyEye is available at https://github.com/daewoooo/SVbyEye.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"95 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250177","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
CRAK-Velo: Chromatin Accessibility Kinetics integration improves RNA Velocity estimation CRAK-Velo:染色质可及性动力学集成改进了 RNA 速度估算
Pub Date : 2024-09-17 DOI: 10.1101/2024.09.12.612736
Nour El Kazwini, Mingze Gao, Idris Kouadri Boudjelthia, Fangxin Cai, Yuanhua Huang, Guido Sanguinetti
RNA velocity has recently emerged as a key tool in the analysis of single-cell transcriptomic data, yet connecting RNA velocity analyses to underlying regulatory processes has proved challenging. Here we propose CRAK-Velo, a semi-mechanistic model which integrates chromatin accessibility data in the estimation of RNA velocities. CRAK-Velo provides biologically consistent estimates of developmental flows and enables accurate cell-type deconvolution, while additionally shining light on regulatory processes at the level of interactions between genes and chromatin regions.
近来,RNA 速度已成为分析单细胞转录组数据的重要工具,然而将 RNA 速度分析与潜在调控过程联系起来却很有挑战性。在这里,我们提出了 CRAK-Velo,这是一种半机制模型,它将染色质可及性数据整合到 RNA 速度的估算中。CRAK-Velo 提供了生物学上一致的发育流估计值,并实现了准确的细胞类型解旋,同时还揭示了基因与染色质区域之间相互作用水平上的调控过程。
{"title":"CRAK-Velo: Chromatin Accessibility Kinetics integration improves RNA Velocity estimation","authors":"Nour El Kazwini, Mingze Gao, Idris Kouadri Boudjelthia, Fangxin Cai, Yuanhua Huang, Guido Sanguinetti","doi":"10.1101/2024.09.12.612736","DOIUrl":"https://doi.org/10.1101/2024.09.12.612736","url":null,"abstract":"RNA velocity has recently emerged as a key tool in the analysis of single-cell transcriptomic data, yet connecting RNA velocity analyses to underlying regulatory processes has proved challenging. Here we propose CRAK-Velo, a semi-mechanistic model which integrates chromatin accessibility data in the estimation of RNA velocities. CRAK-Velo provides biologically consistent estimates of developmental flows and enables accurate cell-type deconvolution, while additionally shining light on regulatory processes at the level of interactions between genes and chromatin regions.","PeriodicalId":501307,"journal":{"name":"bioRxiv - Bioinformatics","volume":"65 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250172","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