首页 > 最新文献

Bioinformatics最新文献

英文 中文
PQSDC: a parallel lossless compressor for quality scores data via sequences partition and Run-Length prediction mapping. PQSDC:通过序列分区和运行长度预测映射的并行无损质量分数数据压缩器。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2024-05-17 DOI: 10.1093/bioinformatics/btae323
Hui Sun, Yingfeng Zheng, Haonan Xie, Huidong Ma, Cheng Zhong, Meng Yan, Xiaoguang Liu, Gang Wang
MOTIVATIONThe quality scores data (QSD) account for 70% in compressed FastQ files obtained from the short and long reads sequencing technologies. Designing effective compressors for QSD that counterbalance compression ratio, time cost, and memory consumption is essential in scenarios such as large-scale genomics data sharing and long-term data backup. This study presents a novel parallel lossless QSD-dedicated compression algorithm named PQSDC, which fulfills the above requirements well. PQSDC is based on two core components: a parallel sequences-partition model designed to reduce peak memory consumption and time cost during compression and decompression processes, as well as a parallel four-level run-length prediction mapping model to enhance compression ratio. Besides, the PQSDC algorithm is also designed to be highly concurrent using multi-core CPU clusters.RESULTSWe evaluate PQSDC and 4 state-of-the-art compression algorithms on 27 real-world datasets, including 61.857 billion QSD characters and 632.908 million QSD sequences. (1) For short reads, compared to baselines, the maximum improvement of PQSDC reaches 7.06% in average compression ratio, and 8.01% in weighted average compression ratio. During compression and decompression, the maximum total time savings of PQSDC are 79.96% and 84.56%, respectively; the maximum average memory savings are 68.34% and 77.63%, respectively. (2) For long reads, the maximum improvement of PQSDC reaches 12.51% and 13.42% in average and weighted average compression ratio, respectively. The maximum total time savings during compression and decompression are 53.51% and 72.53%, respectively; the maximum average memory savings are 19.44% and 17.42%, respectively. (3) Furthermore, PQSDC ranks second in compression robustness among the tested algorithms, indicating that it is less affected by the probability distribution of the QSD collections. Overall, our work provides a promising solution for QSD parallel compression, which balances storage cost, time consumption, and memory occupation primely.AVAILABILITYThe proposed PQSDC compressor can be downloaded from https://github.com/fahaihi/PQSDC.SUPPLEMENTARY INFORMATIONSupplementary data are available at Bioinformatics online.
动机在长短读数测序技术获得的 FastQ 压缩文件中,质量分数数据(QSD)占 70%。在大规模基因组学数据共享和长期数据备份等应用场景中,设计有效的 QSD 压缩器以平衡压缩率、时间成本和内存消耗至关重要。本研究提出了一种名为 PQSDC 的新型并行无损 QSD 专用压缩算法,很好地满足了上述要求。PQSDC 基于两个核心组件:旨在减少压缩和解压缩过程中峰值内存消耗和时间成本的并行序列分区模型,以及旨在提高压缩比的并行四级运行长度预测映射模型。我们在 27 个实际数据集(包括 618.57 亿个 QSD 字符和 6.32908 亿个 QSD 序列)上评估了 PQSDC 和 4 种最先进的压缩算法。(1) 与基线算法相比,PQSDC 对短读取数据的平均压缩率最大提高了 7.06%,加权平均压缩率提高了 8.01%。在压缩和解压缩过程中,PQSDC 最大节省的总时间分别为 79.96% 和 84.56%;最大节省的平均内存分别为 68.34% 和 77.63%。(2) 对于长读取,PQSDC 在平均压缩率和加权平均压缩率方面的最大改进分别达到 12.51% 和 13.42%。压缩和解压缩过程中节省的总时间最大值分别为 53.51% 和 72.53%;节省的平均内存最大值分别为 19.44% 和 17.42%。(3) 此外,PQSDC 的压缩鲁棒性在测试算法中排名第二,表明它受 QSD 集合概率分布的影响较小。总之,我们的工作为 QSD 并行压缩提供了一种很有前途的解决方案,它主要平衡了存储成本、时间消耗和内存占用。AVAILABILITY拟议的 PQSDC 压缩器可从 https://github.com/fahaihi/PQSDC.SUPPLEMENTARY 下载。
{"title":"PQSDC: a parallel lossless compressor for quality scores data via sequences partition and Run-Length prediction mapping.","authors":"Hui Sun, Yingfeng Zheng, Haonan Xie, Huidong Ma, Cheng Zhong, Meng Yan, Xiaoguang Liu, Gang Wang","doi":"10.1093/bioinformatics/btae323","DOIUrl":"https://doi.org/10.1093/bioinformatics/btae323","url":null,"abstract":"MOTIVATION\u0000The quality scores data (QSD) account for 70% in compressed FastQ files obtained from the short and long reads sequencing technologies. Designing effective compressors for QSD that counterbalance compression ratio, time cost, and memory consumption is essential in scenarios such as large-scale genomics data sharing and long-term data backup. This study presents a novel parallel lossless QSD-dedicated compression algorithm named PQSDC, which fulfills the above requirements well. PQSDC is based on two core components: a parallel sequences-partition model designed to reduce peak memory consumption and time cost during compression and decompression processes, as well as a parallel four-level run-length prediction mapping model to enhance compression ratio. Besides, the PQSDC algorithm is also designed to be highly concurrent using multi-core CPU clusters.\u0000\u0000\u0000RESULTS\u0000We evaluate PQSDC and 4 state-of-the-art compression algorithms on 27 real-world datasets, including 61.857 billion QSD characters and 632.908 million QSD sequences. (1) For short reads, compared to baselines, the maximum improvement of PQSDC reaches 7.06% in average compression ratio, and 8.01% in weighted average compression ratio. During compression and decompression, the maximum total time savings of PQSDC are 79.96% and 84.56%, respectively; the maximum average memory savings are 68.34% and 77.63%, respectively. (2) For long reads, the maximum improvement of PQSDC reaches 12.51% and 13.42% in average and weighted average compression ratio, respectively. The maximum total time savings during compression and decompression are 53.51% and 72.53%, respectively; the maximum average memory savings are 19.44% and 17.42%, respectively. (3) Furthermore, PQSDC ranks second in compression robustness among the tested algorithms, indicating that it is less affected by the probability distribution of the QSD collections. Overall, our work provides a promising solution for QSD parallel compression, which balances storage cost, time consumption, and memory occupation primely.\u0000\u0000\u0000AVAILABILITY\u0000The proposed PQSDC compressor can be downloaded from https://github.com/fahaihi/PQSDC.\u0000\u0000\u0000SUPPLEMENTARY INFORMATION\u0000Supplementary data are available at Bioinformatics online.","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-05-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140962816","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
MUSE-XAE: MUtational Signature Extraction with eXplainable AutoEncoder enhances tumour types classification. MUSE-XAE:MUtational Signature Extraction with eXplainable AutoEncoder 可增强肿瘤类型分类。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2024-05-16 DOI: 10.1093/bioinformatics/btae320
Corrado Pancotti, Cesare Rollo, Francesco Codicè, G. Birolo, Piero Fariselli, T. Sanavia
MOTIVATIONMutational signatures are a critical component in deciphering the genetic alterations that underlie cancer development and have become a valuable resource to understand the genomic changes during tumorigenesis. Therefore, it is essential to employ precise and accurate methods for their extraction to ensure that the underlying patterns are reliably identified and can be effectively utilized in new strategies for diagnosis, prognosis and treatment of cancer patients.RESULTSWe present MUSE-XAE, a novel method for mutational signature extraction from cancer genomes using an explainable autoencoder. Our approach employs a hybrid architecture consisting of a nonlinear encoder that can capture nonlinear interactions among features, and a linear decoder which ensures the interpretability of the active signatures. We evaluated and compared MUSE-XAE with other available tools on both synthetic and real cancer datasets and demonstrated that it achieves superior performance in terms of precision and sensitivity in recovering mutational signature profiles. MUSE-XAE extracts highly discriminative mutational signature profiles by enhancing the classification of primary tumour types and subtypes in real world settings. This approach could facilitate further research in this area, with neural networks playing a critical role in advancing our understanding of cancer genomics.AVAILABILITYMUSE-XAE software is freely available at https://github.com/compbiomed-unito/MUSE-XAE.SUPPLEMENTARY INFORMATIONSupplementary data are available at Bioinformatics online.
动机突变特征是破译癌症发生的基因改变的重要组成部分,已成为了解肿瘤发生过程中基因组变化的宝贵资源。因此,必须采用精确的方法来提取突变特征,以确保可靠地识别基本模式,并有效地用于癌症患者的诊断、预后和治疗新策略。我们的方法采用了一种混合架构,由一个非线性编码器和一个线性解码器组成,前者可捕捉特征间的非线性相互作用,后者可确保主动特征的可解释性。我们在合成和真实癌症数据集上对 MUSE-XAE 和其他可用工具进行了评估和比较,结果表明它在恢复突变特征图谱的精确度和灵敏度方面都表现出色。MUSE-XAE 在真实环境中提高了原发性肿瘤类型和亚型的分类能力,从而提取出具有高度鉴别性的突变特征图谱。这种方法可以促进该领域的进一步研究,神经网络在推动我们对癌症基因组学的理解方面发挥着至关重要的作用。AVAILABILITYMUSE-XAE 软件可在 https://github.com/compbiomed-unito/MUSE-XAE.SUPPLEMENTARY 免费获取信息补充数据可在 Bioinformatics online 上获取。
{"title":"MUSE-XAE: MUtational Signature Extraction with eXplainable AutoEncoder enhances tumour types classification.","authors":"Corrado Pancotti, Cesare Rollo, Francesco Codicè, G. Birolo, Piero Fariselli, T. Sanavia","doi":"10.1093/bioinformatics/btae320","DOIUrl":"https://doi.org/10.1093/bioinformatics/btae320","url":null,"abstract":"MOTIVATION\u0000Mutational signatures are a critical component in deciphering the genetic alterations that underlie cancer development and have become a valuable resource to understand the genomic changes during tumorigenesis. Therefore, it is essential to employ precise and accurate methods for their extraction to ensure that the underlying patterns are reliably identified and can be effectively utilized in new strategies for diagnosis, prognosis and treatment of cancer patients.\u0000\u0000\u0000RESULTS\u0000We present MUSE-XAE, a novel method for mutational signature extraction from cancer genomes using an explainable autoencoder. Our approach employs a hybrid architecture consisting of a nonlinear encoder that can capture nonlinear interactions among features, and a linear decoder which ensures the interpretability of the active signatures. We evaluated and compared MUSE-XAE with other available tools on both synthetic and real cancer datasets and demonstrated that it achieves superior performance in terms of precision and sensitivity in recovering mutational signature profiles. MUSE-XAE extracts highly discriminative mutational signature profiles by enhancing the classification of primary tumour types and subtypes in real world settings. This approach could facilitate further research in this area, with neural networks playing a critical role in advancing our understanding of cancer genomics.\u0000\u0000\u0000AVAILABILITY\u0000MUSE-XAE software is freely available at https://github.com/compbiomed-unito/MUSE-XAE.\u0000\u0000\u0000SUPPLEMENTARY INFORMATION\u0000Supplementary data are available at Bioinformatics online.","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140966527","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CopyVAE: a variational autoencoder-based approach for copy number variation inference using single-cell transcriptomics CopyVAE:基于变异自动编码器的单细胞转录组学拷贝数变异推断方法
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2024-04-27 DOI: 10.1093/bioinformatics/btae284
Semih Kurt, Mandi Chen, Hosein Toosi, Xinsong Chen, Camilla Engblom, Jeff Mold, Johan Hartman, Jens Lagergren
Copy number variations (CNVs) are common genetic alterations in tumour cells. The delineation of CNVs holds promise for enhancing our comprehension of cancer progression. Moreover, accurate inference of CNVs from single-cell sequencing data is essential for unravelling intratumoral heterogeneity. However, existing inference methods face limitations in resolution and sensitivity. To address these challenges, we present CopyVAE, a deep learning framework based on a variational autoencoder architecture. Through experiments, we demonstrated that CopyVAE can accurately and reliably detect copy number variations (CNVs) from data obtained using single-cell RNA sequencing. CopyVAE surpasses existing methods in terms of sensitivity and specificity. We also discussed CopyVAE’s potential to advance our understanding of genetic alterations and their impact on disease advancement. CopyVAE is implemented and freely available under MIT license at https://github.com/kurtsemih/copyVAE Supplementary data are available at Bioinformatics online.
拷贝数变异(CNV)是肿瘤细胞中常见的基因改变。描述 CNVs 有助于加深我们对癌症进展的理解。此外,从单细胞测序数据中准确推断 CNV 对于揭示瘤内异质性至关重要。然而,现有的推断方法在分辨率和灵敏度方面存在局限性。 为了应对这些挑战,我们提出了基于变异自动编码器架构的深度学习框架 CopyVAE。通过实验,我们证明 CopyVAE 可以从单细胞 RNA 测序获得的数据中准确可靠地检测拷贝数变异(CNV)。在灵敏度和特异性方面,CopyVAE 超越了现有方法。我们还讨论了 CopyVAE 在推动我们了解基因改变及其对疾病发展的影响方面的潜力。 CopyVAE 在 MIT 许可下实现并免费提供,网址是 https://github.com/kurtsemih/copyVAE 补充数据可在 Bioinformatics online 上获取。
{"title":"CopyVAE: a variational autoencoder-based approach for copy number variation inference using single-cell transcriptomics","authors":"Semih Kurt, Mandi Chen, Hosein Toosi, Xinsong Chen, Camilla Engblom, Jeff Mold, Johan Hartman, Jens Lagergren","doi":"10.1093/bioinformatics/btae284","DOIUrl":"https://doi.org/10.1093/bioinformatics/btae284","url":null,"abstract":"\u0000 \u0000 \u0000 Copy number variations (CNVs) are common genetic alterations in tumour cells. The delineation of CNVs holds promise for enhancing our comprehension of cancer progression. Moreover, accurate inference of CNVs from single-cell sequencing data is essential for unravelling intratumoral heterogeneity. However, existing inference methods face limitations in resolution and sensitivity.\u0000 \u0000 \u0000 \u0000 To address these challenges, we present CopyVAE, a deep learning framework based on a variational autoencoder architecture. Through experiments, we demonstrated that CopyVAE can accurately and reliably detect copy number variations (CNVs) from data obtained using single-cell RNA sequencing. CopyVAE surpasses existing methods in terms of sensitivity and specificity. We also discussed CopyVAE’s potential to advance our understanding of genetic alterations and their impact on disease advancement.\u0000 \u0000 \u0000 \u0000 CopyVAE is implemented and freely available under MIT license at https://github.com/kurtsemih/copyVAE\u0000 \u0000 \u0000 \u0000 Supplementary data are available at Bioinformatics online.\u0000","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-04-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140651944","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
LMCrot: An enhanced protein crotonylation site predictor by leveraging an interpretable window-level embedding from a transformer-based protein language model. LMCrot:通过利用基于转换器的蛋白质语言模型的可解释窗口级嵌入,增强蛋白质巴豆酰化位点预测器。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2024-04-25 DOI: 10.1093/bioinformatics/btae290
Pawel Pratyush, Soufia Bahmani, Suresh Pokharel, Hamid D Ismail, Dukka B Kc
MOTIVATIONRecent advancements in natural language processing have highlighted the effectiveness of global contextualized representations from Protein Language Models (pLMs) in numerous downstream tasks. Nonetheless, strategies to encode the site-of-interest leveraging pLMs for per-residue prediction tasks, such as crotonylation (Kcr) prediction, remain largely uncharted.RESULTSHerein, we adopt a range of approaches for utilizing pLMs by experimenting with different input sequence types (full-length protein sequence versus window sequence), assessing the implications of utilizing per-residue embedding of the site-of-interest as well as embeddings of window residues centered around it. Building upon these insights, we developed a novel residual ConvBiLSTM network designed to process window-level embeddings of the site-of-interest generated by the ProtT5-XL-UniRef50 pLM using full-length sequences as input. This model, termed T5ResConvBiLSTM, surpasses existing state-of-the-art Kcr predictors in performance across three diverse datasets. To validate our approach of utilizing full sequence-based window-level embeddings, we also delved into the interpretability of ProtT5-derived embedding tensors in two ways: firstly, by scrutinizing the attention weights obtained from the transformer's encoder block; and secondly, by computing SHAP values for these tensors, providing a model-agnostic interpretation of the prediction results. Additionally, we enhance the latent representation of ProtT5 by incorporating two additional local representations, one derived from amino acid properties and the other from supervised embedding layer, through an intermediate-fusion stacked generalization approach, using an n-mer window sequence (or, peptide fragment). The resultant stacked model, dubbed LMCrot, exhibits a more pronounced improvement in predictive performance across the tested datasets.AVAILABILITY AND IMPLEMENTATIONLMCrot is publicly available at https://github.com/KCLabMTU/LMCrot.
动机自然语言处理领域的最新进展凸显了蛋白质语言模型(pLMs)的全局上下文表征在众多下游任务中的有效性。结果在本文中,我们采用了一系列利用 pLM 的方法,尝试了不同的输入序列类型(全长蛋白质序列和窗口序列),评估了利用兴趣点的每残基嵌入以及以兴趣点为中心的窗口残基嵌入的影响。基于这些见解,我们开发了一种新型残差 ConvBiLSTM 网络,旨在处理 ProtT5-XL-UniRef50 pLM 使用全长序列作为输入生成的兴趣点窗口级嵌入。这个被称为 T5ResConvBiLSTM 的模型在三个不同数据集上的性能超过了现有的最先进 Kcr 预测器。为了验证我们利用基于全序列的窗口级嵌入的方法,我们还通过两种方式深入研究了 ProtT5 衍生的嵌入张量的可解释性:首先,我们仔细研究了从变换器编码器块中获得的注意力权重;其次,我们计算了这些张量的 SHAP 值,为预测结果提供了与模型无关的解释。此外,我们还通过中间融合堆叠泛化方法,使用 n 聚体窗口序列(或肽片段),加入了两个额外的局部表征,一个来自氨基酸特性,另一个来自监督嵌入层,从而增强了 ProtT5 的潜在表征。由此产生的堆叠模型被命名为 LMCrot,在测试数据集的预测性能方面有了更明显的提高。可用性和实施LMCrot 可在 https://github.com/KCLabMTU/LMCrot 网站上公开获取。
{"title":"LMCrot: An enhanced protein crotonylation site predictor by leveraging an interpretable window-level embedding from a transformer-based protein language model.","authors":"Pawel Pratyush, Soufia Bahmani, Suresh Pokharel, Hamid D Ismail, Dukka B Kc","doi":"10.1093/bioinformatics/btae290","DOIUrl":"https://doi.org/10.1093/bioinformatics/btae290","url":null,"abstract":"MOTIVATION\u0000Recent advancements in natural language processing have highlighted the effectiveness of global contextualized representations from Protein Language Models (pLMs) in numerous downstream tasks. Nonetheless, strategies to encode the site-of-interest leveraging pLMs for per-residue prediction tasks, such as crotonylation (Kcr) prediction, remain largely uncharted.\u0000\u0000\u0000RESULTS\u0000Herein, we adopt a range of approaches for utilizing pLMs by experimenting with different input sequence types (full-length protein sequence versus window sequence), assessing the implications of utilizing per-residue embedding of the site-of-interest as well as embeddings of window residues centered around it. Building upon these insights, we developed a novel residual ConvBiLSTM network designed to process window-level embeddings of the site-of-interest generated by the ProtT5-XL-UniRef50 pLM using full-length sequences as input. This model, termed T5ResConvBiLSTM, surpasses existing state-of-the-art Kcr predictors in performance across three diverse datasets. To validate our approach of utilizing full sequence-based window-level embeddings, we also delved into the interpretability of ProtT5-derived embedding tensors in two ways: firstly, by scrutinizing the attention weights obtained from the transformer's encoder block; and secondly, by computing SHAP values for these tensors, providing a model-agnostic interpretation of the prediction results. Additionally, we enhance the latent representation of ProtT5 by incorporating two additional local representations, one derived from amino acid properties and the other from supervised embedding layer, through an intermediate-fusion stacked generalization approach, using an n-mer window sequence (or, peptide fragment). The resultant stacked model, dubbed LMCrot, exhibits a more pronounced improvement in predictive performance across the tested datasets.\u0000\u0000\u0000AVAILABILITY AND IMPLEMENTATION\u0000LMCrot is publicly available at https://github.com/KCLabMTU/LMCrot.","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140656696","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CORDAX web server: An online platform for the prediction and 3D visualization of aggregation motifs in protein sequences. CORDAX 网络服务器:用于预测蛋白质序列中聚集图案并将其三维可视化的在线平台。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2024-04-25 DOI: 10.1093/bioinformatics/btae279
Nikolaos N. Louros, F. Rousseau, J. Schymkowitz
MOTIVATIONProteins, the molecular workhorses of biological systems, execute a multitude of critical functions dictated by their precise three-dimensional structures. In a complex and dynamic cellular environment, proteins can undergo misfolding, leading to the formation of aggregates that take up various forms, including amorphous and ordered aggregation in the shape of amyloid fibrils. This phenomenon is closely linked to a spectrum of widespread debilitating pathologies, such as Alzheimer's disease, Parkinson's disease, type-II diabetes, and several other proteinopathies, but also hampers the engineering of soluble agents, as in the case of antibody development. As such, the accurate prediction of aggregation propensity within protein sequences has become pivotal due to profound implications in understanding disease mechanisms, as well as in improving biotechnological and therapeutic applications.RESULTSWe previously developed Cordax, a structure-based predictor that utilizes logistic regression to detect aggregation motifs in protein sequences based on their structural complementarity to the amyloid cross-beta architecture. Here, we present a dedicated web server interface for Cordax. This online platform combines several features including detailed scoring of sequence aggregation propensity, as well as 3D visualization with several customization options for topology models of the structural cores formed by predicted aggregation motifs. In addition, information is provided on experimentally determined aggregation-prone regions that exhibit sequence similarity to predicted motifs, scores, and links to other predictor outputs, as well as simultaneous predictions of relevant sequence propensities, such as solubility, hydrophobicity, and secondary structure propensity.AVAILABILITYThe Cordax webserver is freely accessible at https://cordax.switchlab.org/.
动因蛋白质是生物系统的分子主力,根据其精确的三维结构执行多种关键功能。在复杂多变的细胞环境中,蛋白质会发生错误折叠,从而形成各种形式的聚集体,包括淀粉样纤维状的无定形聚集体和有序聚集体。这种现象与阿尔茨海默病、帕金森病、II 型糖尿病和其他几种蛋白质疾病等一系列广泛的衰弱性病症密切相关,同时也阻碍了可溶性制剂的工程设计,例如抗体的开发。因此,准确预测蛋白质序列中的聚集倾向已变得至关重要,因为这对了解疾病机制以及改进生物技术和治疗应用具有深远影响。在此,我们介绍了 Cordax 的专用网络服务器界面。该在线平台集多种功能于一体,包括序列聚集倾向的详细评分,以及三维可视化,并为预测聚集图案形成的结构核心拓扑模型提供了多种自定义选项。此外,该平台还提供实验确定的易聚集区域的信息,这些区域与预测图案的序列具有相似性、得分、与其他预测器输出结果的链接,以及相关序列倾向性的同步预测,如溶解性、疏水性和二级结构倾向性。
{"title":"CORDAX web server: An online platform for the prediction and 3D visualization of aggregation motifs in protein sequences.","authors":"Nikolaos N. Louros, F. Rousseau, J. Schymkowitz","doi":"10.1093/bioinformatics/btae279","DOIUrl":"https://doi.org/10.1093/bioinformatics/btae279","url":null,"abstract":"MOTIVATION\u0000Proteins, the molecular workhorses of biological systems, execute a multitude of critical functions dictated by their precise three-dimensional structures. In a complex and dynamic cellular environment, proteins can undergo misfolding, leading to the formation of aggregates that take up various forms, including amorphous and ordered aggregation in the shape of amyloid fibrils. This phenomenon is closely linked to a spectrum of widespread debilitating pathologies, such as Alzheimer's disease, Parkinson's disease, type-II diabetes, and several other proteinopathies, but also hampers the engineering of soluble agents, as in the case of antibody development. As such, the accurate prediction of aggregation propensity within protein sequences has become pivotal due to profound implications in understanding disease mechanisms, as well as in improving biotechnological and therapeutic applications.\u0000\u0000\u0000RESULTS\u0000We previously developed Cordax, a structure-based predictor that utilizes logistic regression to detect aggregation motifs in protein sequences based on their structural complementarity to the amyloid cross-beta architecture. Here, we present a dedicated web server interface for Cordax. This online platform combines several features including detailed scoring of sequence aggregation propensity, as well as 3D visualization with several customization options for topology models of the structural cores formed by predicted aggregation motifs. In addition, information is provided on experimentally determined aggregation-prone regions that exhibit sequence similarity to predicted motifs, scores, and links to other predictor outputs, as well as simultaneous predictions of relevant sequence propensities, such as solubility, hydrophobicity, and secondary structure propensity.\u0000\u0000\u0000AVAILABILITY\u0000The Cordax webserver is freely accessible at https://cordax.switchlab.org/.","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140654240","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CASCC: a co-expression assisted single-cell RNA-seq data clustering method. CASCC:共表达辅助单细胞 RNA-seq 数据聚类方法。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2024-04-25 DOI: 10.1093/bioinformatics/btae283
Lingyi Cai, Dimitris Anastassiou
SUMMARYExisting clustering methods for characterizing cell populations from single-cell RNA sequencing are constrained by several limitations stemming from the fact that clusters often cannot be homogeneous, particularly for transitioning populations. On the other hand, dominant cell populations within samples can be identified independently by their strong gene co-expression signatures using methods unrelated to partitioning. Here, we introduce a clustering method, CASCC, designed to improve biological accuracy using gene co-expression features identified using an unsupervised adaptive attractor algorithm. CASCC outperformed other methods as evidenced by multiple evaluation metrics, and our results suggest that CASCC can improve the analysis of single-cell transcriptomics, enabling potential new discoveries related to underlying biological mechanisms.AVAILABILITY AND IMPLEMENTATIONThe CASCC R package is publicly available at https://github.com/LingyiC/CASCC and https://zenodo.org/doi/10.5281/zenodo.10648327.SUPPLEMENTARY INFORMATIONSupplementary data are available at Bioinformatics online.
摘要从单细胞 RNA 测序中描述细胞群特征的现有聚类方法受到一些限制,这些限制源于聚类通常不可能是同质的,尤其是对于过渡细胞群。另一方面,利用与分区无关的方法,可以通过强大的基因共表达特征独立识别样本中的优势细胞群。在此,我们介绍一种聚类方法 CASCC,旨在利用无监督自适应吸引子算法识别的基因共表达特征提高生物学准确性。我们的研究结果表明,CASCC 可以改进单细胞转录组学分析,从而实现与潜在生物机制相关的潜在新发现。AVAILABILITY AND IMPLEMENTATIONThe CASCC R package is publicly available at https://github.com/LingyiC/CASCC and https://zenodo.org/doi/10.5281/zenodo.10648327.SUPPLEMENTARY INFORMATIONSupplementary data are available at Bioinformatics online.
{"title":"CASCC: a co-expression assisted single-cell RNA-seq data clustering method.","authors":"Lingyi Cai, Dimitris Anastassiou","doi":"10.1093/bioinformatics/btae283","DOIUrl":"https://doi.org/10.1093/bioinformatics/btae283","url":null,"abstract":"SUMMARY\u0000Existing clustering methods for characterizing cell populations from single-cell RNA sequencing are constrained by several limitations stemming from the fact that clusters often cannot be homogeneous, particularly for transitioning populations. On the other hand, dominant cell populations within samples can be identified independently by their strong gene co-expression signatures using methods unrelated to partitioning. Here, we introduce a clustering method, CASCC, designed to improve biological accuracy using gene co-expression features identified using an unsupervised adaptive attractor algorithm. CASCC outperformed other methods as evidenced by multiple evaluation metrics, and our results suggest that CASCC can improve the analysis of single-cell transcriptomics, enabling potential new discoveries related to underlying biological mechanisms.\u0000\u0000\u0000AVAILABILITY AND IMPLEMENTATION\u0000The CASCC R package is publicly available at https://github.com/LingyiC/CASCC and https://zenodo.org/doi/10.5281/zenodo.10648327.\u0000\u0000\u0000SUPPLEMENTARY INFORMATION\u0000Supplementary data are available at Bioinformatics online.","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140657084","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
AmplificationTimeR: An R Package for Timing Sequential Amplification Events. AmplificationTimeR:用于为顺序放大事件计时的 R 软件包。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2024-04-24 DOI: 10.1093/bioinformatics/btae281
G. M. Jakobsdottir, Stefan C Dentro, Robert G Bristow, David C Wedge
MOTIVATIONFew methods exist for timing individual amplification events in regions of focal amplification. Current methods are also limited in the copy number states that they are able to time. Here we introduce AmplificationTimeR, a method for timing higher level copy number gains and inferring the most parsimonious order of events for regions that have undergone both single gains and whole genome duplication. Our method is an extension of established approaches for timing genomic gains.RESULTSWe can time more copy number states, and in states covered by other methods our results are comparable to previously published methods.AVAILABILITYAmplificationTimer is freely available as an R package hosted at https://github.com/Wedge-lab/AmplificationTimeR.SUPPLEMENTARY INFORMATIONSupplementary data are available at Bioinformatics online.
动机目前很少有方法能对病灶扩增区域的单个扩增事件进行计时。目前的方法在能够计时的拷贝数状态方面也受到限制。在此,我们介绍 AmplificationTimeR,这是一种为更高水平的拷贝数增殖计时的方法,并能推断出经历过单个增殖和全基因组复制的区域的最合理的事件顺序。我们的方法是对已有的基因组增殖计时方法的扩展。结果我们可以对更多的拷贝数状态进行计时,而且在其他方法所涵盖的状态下,我们的结果与之前发表的方法相当。AVILABILITYAmplificationTimer 是一个 R 包,可免费获取,托管在 https://github.com/Wedge-lab/AmplificationTimeR.SUPPLEMENTARY INFORMATIONSupplementary data are available at Bioinformatics online.
{"title":"AmplificationTimeR: An R Package for Timing Sequential Amplification Events.","authors":"G. M. Jakobsdottir, Stefan C Dentro, Robert G Bristow, David C Wedge","doi":"10.1093/bioinformatics/btae281","DOIUrl":"https://doi.org/10.1093/bioinformatics/btae281","url":null,"abstract":"MOTIVATION\u0000Few methods exist for timing individual amplification events in regions of focal amplification. Current methods are also limited in the copy number states that they are able to time. Here we introduce AmplificationTimeR, a method for timing higher level copy number gains and inferring the most parsimonious order of events for regions that have undergone both single gains and whole genome duplication. Our method is an extension of established approaches for timing genomic gains.\u0000\u0000\u0000RESULTS\u0000We can time more copy number states, and in states covered by other methods our results are comparable to previously published methods.\u0000\u0000\u0000AVAILABILITY\u0000AmplificationTimer is freely available as an R package hosted at https://github.com/Wedge-lab/AmplificationTimeR.\u0000\u0000\u0000SUPPLEMENTARY INFORMATION\u0000Supplementary data are available at Bioinformatics online.","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140659774","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
DIMet: An open-source tool for Differential analysis of targeted Isotope-labeled Metabolomics data. DIMet:用于靶向同位素标记代谢组学数据差异分析的开源工具。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2024-04-24 DOI: 10.1093/bioinformatics/btae282
Johanna Galvis, J. Guyon, Benjamin Dartigues, Helge Hecht, Björn Grüning, Florian Specque, Hayssam Soueidan, S. Karkar, Thomas Daubon, M. Nikolski
MOTIVATIONMany diseases, such as cancer, are characterized by an alteration of cellular metabolism allowing cells to adapt to changes in the microenvironment. Stable isotope-resolved metabolomics and downstream data analyses are widely used techniques for unraveling cells' metabolic activity to understand the altered functioning of metabolic pathways in the diseased state. While a number of bioinformatic solutions exist for the differential analysis of Stable Isotope-Resolved Metabolomics data, there is currently no available resource providing a comprehensive toolbox.RESULTSIn this work, we present DIMet, a one-stop comprehensive tool for differential analysis of targeted tracer data. DIMet accepts metabolite total abundances, isotopologue contributions, and isotopic mean enrichment, and supports differential comparison (pairwise and multi-group), time-series analyses, and labeling profile comparison. Moreover, it integrates transcriptomics and targeted metabolomics data through network-based metabolograms. We illustrate the use of DIMet in real SIRM datasets obtained from Glioblastoma P3 cell-line samples. DIMet is open-source, and is readily available for routine downstream analysis of isotope-labeled targeted metabolomics data, as it can be used both in the command line interface or as a complete toolkit in the public Galaxy Europe and Workfow4Metabolomics web platforms.AVAILABILITYDIMet is freely available at https://github.com/cbib/DIMet, and through https://usegalaxy.eu and https://workflow4metabolomics.usegalaxy.fr. All the datasets are available at Zenodo https://zenodo.org/records/10925786.SUPPLEMENTARY INFORMATIONSupplementary data are available at Bioinformatics online.
动机许多疾病(如癌症)都以细胞代谢改变为特征,使细胞能够适应微环境的变化。稳定同位素分辨代谢组学和下游数据分析是广泛使用的技术,用于揭示细胞的代谢活动,以了解代谢途径在患病状态下的功能变化。虽然有许多生物信息学解决方案可用于稳定同位素分辨代谢组学数据的差异分析,但目前还没有提供综合工具箱的可用资源。DIMet 可接受代谢物总丰度、同位素贡献和同位素平均富集度,并支持差异比较(成对和多组)、时间序列分析和标记曲线比较。此外,它还通过基于网络的代谢全图整合了转录组学和靶向代谢组学数据。我们在从胶质母细胞瘤 P3 细胞系样本中获得的真实 SIRM 数据集中演示了 DIMet 的使用。DIMet 是开源的,可随时用于同位素标记的靶向代谢组学数据的常规下游分析,因为它既可以在命令行界面中使用,也可以作为一个完整的工具包在公共的 Galaxy Europe 和 Workfow4Metabolomics 网络平台中使用。AVAILABILITYDIMet 可在 https://github.com/cbib/DIMet 免费获取,也可通过 https://usegalaxy.eu 和 https://workflow4metabolomics.usegalaxy.fr 获取。所有数据集均可在 Zenodo https://zenodo.org/records/10925786.SUPPLEMENTARY 上获取信息补充数据可在 Bioinformatics online 上获取。
{"title":"DIMet: An open-source tool for Differential analysis of targeted Isotope-labeled Metabolomics data.","authors":"Johanna Galvis, J. Guyon, Benjamin Dartigues, Helge Hecht, Björn Grüning, Florian Specque, Hayssam Soueidan, S. Karkar, Thomas Daubon, M. Nikolski","doi":"10.1093/bioinformatics/btae282","DOIUrl":"https://doi.org/10.1093/bioinformatics/btae282","url":null,"abstract":"MOTIVATION\u0000Many diseases, such as cancer, are characterized by an alteration of cellular metabolism allowing cells to adapt to changes in the microenvironment. Stable isotope-resolved metabolomics and downstream data analyses are widely used techniques for unraveling cells' metabolic activity to understand the altered functioning of metabolic pathways in the diseased state. While a number of bioinformatic solutions exist for the differential analysis of Stable Isotope-Resolved Metabolomics data, there is currently no available resource providing a comprehensive toolbox.\u0000\u0000\u0000RESULTS\u0000In this work, we present DIMet, a one-stop comprehensive tool for differential analysis of targeted tracer data. DIMet accepts metabolite total abundances, isotopologue contributions, and isotopic mean enrichment, and supports differential comparison (pairwise and multi-group), time-series analyses, and labeling profile comparison. Moreover, it integrates transcriptomics and targeted metabolomics data through network-based metabolograms. We illustrate the use of DIMet in real SIRM datasets obtained from Glioblastoma P3 cell-line samples. DIMet is open-source, and is readily available for routine downstream analysis of isotope-labeled targeted metabolomics data, as it can be used both in the command line interface or as a complete toolkit in the public Galaxy Europe and Workfow4Metabolomics web platforms.\u0000\u0000\u0000AVAILABILITY\u0000DIMet is freely available at https://github.com/cbib/DIMet, and through https://usegalaxy.eu and https://workflow4metabolomics.usegalaxy.fr. All the datasets are available at Zenodo https://zenodo.org/records/10925786.\u0000\u0000\u0000SUPPLEMENTARY INFORMATION\u0000Supplementary data are available at Bioinformatics online.","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140659536","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
MammalMethylClock R package: Software for DNA Methylation-Based epigenetic clocks in mammals. MammalMethylClock R 软件包:哺乳动物基于 DNA 甲基化的表观遗传时钟软件。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2024-04-24 DOI: 10.1093/bioinformatics/btae280
J. Zoller, Steve Horvath
MOTIVATIONEpigenetic clocks are prediction methods based on DNA methylation levels in a given species or set of species. Defined as multivariate regression models, these DNA methylation-based biomarkers of age or mortality risk are useful in species conservation efforts and in preclinical studies.RESULTSWe present an R package called MammalMethylClock for the construction, assessment, and application of epigenetic clocks in different mammalian species. The R package includes the utility for implementing pre-existing mammalian clocks from the Mammalian Methylation Consortium.AVAILABILITYThe source code and documentation manual for MammalMethylClock, and clock coefficient .csv files that are included within this software package, can be found on Zenodo at https://doi.org/10.5281/zenodo.10971037.SUPPLEMENTARY INFORMATIONSupplementary data are available at Bioinformatics online.
动机表观遗传时钟是一种基于特定物种或物种集 DNA 甲基化水平的预测方法。这些基于 DNA 甲基化的年龄或死亡风险生物标志物被定义为多变量回归模型,在物种保护工作和临床前研究中非常有用。结果我们提出了一个名为 MammalMethylClock 的 R 软件包,用于构建、评估和应用不同哺乳动物物种的表观遗传时钟。该 R 软件包包括用于实现哺乳动物甲基化联盟(Mammalian Methylation Consortium)已有哺乳动物时钟的实用程序。可获得性MammalMethylClock 的源代码和文档手册以及该软件包中包含的时钟系数 .csv 文件可在 Zenodo 上找到,网址是:https://doi.org/10.5281/zenodo.10971037.SUPPLEMENTARY 信息补充数据可在 Bioinformatics online 上获得。
{"title":"MammalMethylClock R package: Software for DNA Methylation-Based epigenetic clocks in mammals.","authors":"J. Zoller, Steve Horvath","doi":"10.1093/bioinformatics/btae280","DOIUrl":"https://doi.org/10.1093/bioinformatics/btae280","url":null,"abstract":"MOTIVATION\u0000Epigenetic clocks are prediction methods based on DNA methylation levels in a given species or set of species. Defined as multivariate regression models, these DNA methylation-based biomarkers of age or mortality risk are useful in species conservation efforts and in preclinical studies.\u0000\u0000\u0000RESULTS\u0000We present an R package called MammalMethylClock for the construction, assessment, and application of epigenetic clocks in different mammalian species. The R package includes the utility for implementing pre-existing mammalian clocks from the Mammalian Methylation Consortium.\u0000\u0000\u0000AVAILABILITY\u0000The source code and documentation manual for MammalMethylClock, and clock coefficient .csv files that are included within this software package, can be found on Zenodo at https://doi.org/10.5281/zenodo.10971037.\u0000\u0000\u0000SUPPLEMENTARY INFORMATION\u0000Supplementary data are available at Bioinformatics online.","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140660126","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Large-scale Structure-Informed multiple sequence alignment of proteins with SIMSApiper. 利用 SIMSApiper 对蛋白质进行大规模结构信息多序列比对。
IF 5.8 3区 生物学 Q1 Mathematics Pub Date : 2024-04-22 DOI: 10.1093/bioinformatics/btae276
Charlotte Crauwels, Sophie-Luise Heidig, Adrián Díaz, Wim F Vranken
SUMMARYSIMSApiper is a Nextflow pipeline that creates reliable, structure-informed MSAs of thousands of protein sequences in time-frames faster than standard structure-based alignment methods. Structural information can be provided by the user or collected by the pipeline from online resources. Parallelization with sequence identity based subsets can be activated to significantly speed up the alignment process. Finally, the number of gaps in the final alignment can be reduced by leveraging the position of conserved secondary structure elements.AVAILABILITY AND IMPLEMENTATIONThe pipeline is implemented using Nextflow, Python3 and Bash. It is publicly available on github.com/Bio2Byte/simsapiper.SUPPLEMENTARY INFORMATIONAll data is available on GitHub.
摘要SIMSApiper 是一种 Nextflow 管道,它能以比标准的基于结构的比对方法更快的速度,为成千上万的蛋白质序列创建可靠的、基于结构的 MSAs。结构信息可以由用户提供,也可以由管道从在线资源中收集。基于序列同一性子集的并行化可以被激活,从而显著加快比对过程。最后,利用保守二级结构元素的位置,可以减少最终比对中的间隙数量。它可在 github.com/Bio2Byte/simsapiper 上公开获取。补充信息所有数据均可在 GitHub 上获取。
{"title":"Large-scale Structure-Informed multiple sequence alignment of proteins with SIMSApiper.","authors":"Charlotte Crauwels, Sophie-Luise Heidig, Adrián Díaz, Wim F Vranken","doi":"10.1093/bioinformatics/btae276","DOIUrl":"https://doi.org/10.1093/bioinformatics/btae276","url":null,"abstract":"SUMMARY\u0000SIMSApiper is a Nextflow pipeline that creates reliable, structure-informed MSAs of thousands of protein sequences in time-frames faster than standard structure-based alignment methods. Structural information can be provided by the user or collected by the pipeline from online resources. Parallelization with sequence identity based subsets can be activated to significantly speed up the alignment process. Finally, the number of gaps in the final alignment can be reduced by leveraging the position of conserved secondary structure elements.\u0000\u0000\u0000AVAILABILITY AND IMPLEMENTATION\u0000The pipeline is implemented using Nextflow, Python3 and Bash. It is publicly available on github.com/Bio2Byte/simsapiper.\u0000\u0000\u0000SUPPLEMENTARY INFORMATION\u0000All data is available on GitHub.","PeriodicalId":8903,"journal":{"name":"Bioinformatics","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140676036","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","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
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