首页 > 最新文献

Journal of Computational Biology最新文献

英文 中文
DERNA Enables Pareto Optimal RNA Design. DERNA 实现了帕累托最优 RNA 设计。
IF 1.7 4区 生物学 Q2 Mathematics Pub Date : 2024-03-01 Epub Date: 2024-02-27 DOI: 10.1089/cmb.2023.0283
Xinyu Gu, Yuanyuan Qi, Mohammed El-Kebir

The design of an RNA sequence v that encodes an input target protein sequence w is a crucial aspect of messenger RNA (mRNA) vaccine development. There are an exponential number of possible RNA sequences for a single target protein due to codon degeneracy. These potential RNA sequences can assume various secondary structure conformations, each with distinct minimum free energy (MFE), impacting thermodynamic stability and mRNA half-life. Furthermore, the presence of species-specific codon usage bias, quantified by the codon adaptation index (CAI), plays a vital role in translation efficiency. While earlier studies focused on optimizing either MFE or CAI, recent research has underscored the advantages of simultaneously optimizing both objectives. However, optimizing one objective comes at the expense of the other. In this work, we present the Pareto Optimal RNA Design problem, aiming to identify the set of Pareto optimal solutions for which no alternative solutions exist that exhibit better MFE and CAI values. Our algorithm DEsign RNA (DERNA) uses the weighted sum method to enumerate the Pareto front by optimizing convex combinations of both objectives. We use dynamic programming to solve each convex combination in O(|w|3) time and O(|w|2) space. Compared with a CDSfold, previous approach that only optimizes MFE, we show on a benchmark data set that DERNA obtains solutions with identical MFE but superior CAI. Moreover, we show that DERNA matches the performance in terms of solution quality of LinearDesign, a recent approach that similarly seeks to balance MFE and CAI. We conclude by demonstrating our method's potential for mRNA vaccine design for the SARS-CoV-2 spike protein.

设计能编码输入目标蛋白质序列 w 的 RNA 序列 v 是信使 RNA (mRNA) 疫苗开发的一个关键环节。由于密码子退化,单个目标蛋白质可能存在指数数量的 RNA 序列。这些潜在的 RNA 序列可以形成各种二级结构构象,每种构象都具有不同的最小自由能 (MFE),从而影响热力学稳定性和 mRNA 的半衰期。此外,以密码子适应指数(CAI)量化的物种特异性密码子使用偏差在翻译效率中起着至关重要的作用。早期的研究侧重于优化 MFE 或 CAI,而最近的研究则强调了同时优化这两个目标的优势。然而,优化一个目标会牺牲另一个目标。在这项工作中,我们提出了帕累托最优 RNA 设计问题,旨在找出一组帕累托最优解,对于这组最优解,不存在能表现出更好的 MFE 值和 CAI 值的替代方案。我们的算法 DEsign RNA (DERNA) 使用加权和方法,通过优化两个目标的凸组合来枚举帕累托前沿。我们使用动态编程在 O(|w|3) 时间和 O(|w|2) 空间内求解每个凸组合。与之前只优化 MFE 的 CDSfold 方法相比,我们在一个基准数据集上表明,DERNA 得到的解决方案具有相同的 MFE,但 CAI 更优。此外,我们还展示了 DERNA 在解决方案质量方面与 LinearDesign 的性能不相上下,后者是最近推出的一种方法,同样寻求 MFE 和 CAI 之间的平衡。最后,我们展示了我们的方法在 SARS-CoV-2 穗蛋白 mRNA 疫苗设计方面的潜力。
{"title":"DERNA Enables Pareto Optimal RNA Design.","authors":"Xinyu Gu, Yuanyuan Qi, Mohammed El-Kebir","doi":"10.1089/cmb.2023.0283","DOIUrl":"10.1089/cmb.2023.0283","url":null,"abstract":"<p><p>The design of an RNA sequence <math><mstyle><mi>v</mi></mstyle></math> that encodes an input target protein sequence <math><mstyle><mi>w</mi></mstyle></math> is a crucial aspect of messenger RNA (mRNA) vaccine development. There are an exponential number of possible RNA sequences for a single target protein due to codon degeneracy. These potential RNA sequences can assume various secondary structure conformations, each with distinct minimum free energy (MFE), impacting thermodynamic stability and mRNA half-life. Furthermore, the presence of species-specific codon usage bias, quantified by the codon adaptation index (CAI), plays a vital role in translation efficiency. While earlier studies focused on optimizing either MFE or CAI, recent research has underscored the advantages of simultaneously optimizing both objectives. However, optimizing one objective comes at the expense of the other. In this work, we present the Pareto Optimal RNA Design problem, aiming to identify the set of Pareto optimal solutions for which no alternative solutions exist that exhibit better MFE and CAI values. Our algorithm DEsign RNA (DERNA) uses the weighted sum method to enumerate the Pareto front by optimizing convex combinations of both objectives. We use dynamic programming to solve each convex combination in <math><mstyle><mi>O</mi></mstyle><mrow><mo>(</mo><mrow><mo>|</mo><mstyle><mi>w</mi></mstyle><msup><mrow><mo>|</mo></mrow><mrow><mn>3</mn></mrow></msup></mrow><mo>)</mo></mrow></math> time and <math><mstyle><mi>O</mi></mstyle><mrow><mo>(</mo><mrow><mo>|</mo><mstyle><mi>w</mi></mstyle><msup><mrow><mo>|</mo></mrow><mrow><mn>2</mn></mrow></msup></mrow><mo>)</mo></mrow></math> space. Compared with a CDSfold, previous approach that only optimizes MFE, we show on a benchmark data set that DERNA obtains solutions with identical MFE but superior CAI. Moreover, we show that DERNA matches the performance in terms of solution quality of LinearDesign, a recent approach that similarly seeks to balance MFE and CAI. We conclude by demonstrating our method's potential for mRNA vaccine design for the SARS-CoV-2 spike protein.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139990194","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
iGly-IDN: Identifying Lysine Glycation Sites in Proteins Based on Improved DenseNet. iGly-IDN:基于改进DenseNet的蛋白质中赖氨酸糖基化位点鉴定。
IF 1.7 4区 生物学 Q2 Mathematics Pub Date : 2024-02-01 Epub Date: 2023-11-28 DOI: 10.1089/cmb.2023.0112
Jianhua Jia, Genqiang Wu, Meifang Li

Lysine glycation is one of the most significant protein post-translational modifications, which changes the properties of the proteins and causes them to be dysfunctional. Accurately identifying glycation sites helps to understand the biological function and potential mechanism of glycation in disease treatments. Nonetheless, the experimental methods are ordinarily inefficient and costly, so effective computational methods need to be developed. In this study, we proposed the new model called iGly-IDN based on the improved densely connected convolutional networks (DenseNet). First, one hot encoding was adopted to obtain the original feature maps. Afterward, the improved DenseNet was adopted to capture feature information with the importance degrees during the feature learning. According to the experimental results, Acc reaches 66%, and Mathews correlation coefficient reaches 0.33 on the independent testing data set, which indicates that the iGly-IDN can provide more effective glycation site identification than the current predictors.

赖氨酸糖基化是蛋白质翻译后最重要的修饰之一,它改变了蛋白质的性质并导致它们功能失调。准确识别糖基化位点有助于了解糖基化在疾病治疗中的生物学功能和潜在机制。然而,实验方法通常效率低且成本高,因此需要开发有效的计算方法。在本研究中,我们提出了基于改进的密集连接卷积网络(DenseNet)的iGly-IDN新模型。首先,采用一种热编码方法获得原始特征映射;然后,在特征学习过程中,采用改进的DenseNet算法捕获具有重要度的特征信息。实验结果显示,在独立测试数据集上,Acc达到66%,Mathews相关系数达到0.33,表明iGly-IDN比现有的预测因子能够提供更有效的糖基化位点识别。
{"title":"iGly-IDN: Identifying Lysine Glycation Sites in Proteins Based on Improved DenseNet.","authors":"Jianhua Jia, Genqiang Wu, Meifang Li","doi":"10.1089/cmb.2023.0112","DOIUrl":"10.1089/cmb.2023.0112","url":null,"abstract":"<p><p>Lysine glycation is one of the most significant protein post-translational modifications, which changes the properties of the proteins and causes them to be dysfunctional. Accurately identifying glycation sites helps to understand the biological function and potential mechanism of glycation in disease treatments. Nonetheless, the experimental methods are ordinarily inefficient and costly, so effective computational methods need to be developed. In this study, we proposed the new model called iGly-IDN based on the improved densely connected convolutional networks (DenseNet). First, one hot encoding was adopted to obtain the original feature maps. Afterward, the improved DenseNet was adopted to capture feature information with the importance degrees during the feature learning. According to the experimental results, Acc reaches 66%, and Mathews correlation coefficient reaches 0.33 on the independent testing data set, which indicates that the iGly-IDN can provide more effective glycation site identification than the current predictors.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138451634","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Comparing the Performance of Three Computational Methods for Estimating the Effective Reproduction Number. 比较估算有效繁殖数的三种计算方法的性能
IF 1.7 4区 生物学 Q2 Mathematics Pub Date : 2024-02-01 Epub Date: 2024-01-16 DOI: 10.1089/cmb.2023.0065
Zihan Wang, Mengxia Xu, Zonglin Yang, Yu Jin, Yong Zhang

The effective reproduction number (Rt) is one of the most important epidemiological parameters, providing suggestions for monitoring the development trend of diseases and also for adjusting the prevention and control policies. However, a few studies have focused on the performance of some common computational methods for Rt. The purpose of this article is to compare the performance of three computational methods for Rt: the time-dependent (TD) method, the new time-varying (NT) method, and the sequential Bayesian (SB) method. Four evaluation methods-accuracy, correlation coefficient, similarity based on trend, and dynamic time warping distance-were used to compare the effectiveness of three computational methods for Rt under different time lags and time windows. The results showed that the NT method was a better choice for real-time monitoring and analysis of the epidemic in the middle and late stages of the infectious disease. The TD method could reflect the change of the number of cases stably and accurately, and was more suitable for monitoring the change of Rt during the whole process of the epidemic outbreak. When the data were relatively stable, the SB method could also provide a reliable estimate for Rt, while the error would increase when the fluctuation in the number of cases increased. The results would provide suggestions for selecting appropriate Rt estimation methods and making policy adjustments more timely and effectively according to the change of Rt.

有效繁殖数(Rt)是最重要的流行病学参数之一,可为监测疾病发展趋势和调整防控政策提供建议。本文旨在比较三种有效繁殖数计算方法的性能:时间相关(TD)方法、新时变(NT)方法和序列贝叶斯(SB)方法。四种评价方法--准确度、相关系数、基于趋势的相似性和动态时间扭曲距离--用于比较三种计算方法在不同时滞和时间窗口下对 Rt 的有效性。结果表明,NT 方法更适用于传染病中后期的疫情实时监测和分析。TD 方法能稳定、准确地反映病例数的变化,更适合监测疫情爆发全过程中 Rt 的变化。当数据相对稳定时,SB 方法也能提供可靠的 Rt 估计值,而当病例数波动增大时,误差就会增大。这些结果将为选择合适的 Rt 估算方法提供建议,并根据 Rt 的变化更及时有效地进行政策调整。
{"title":"Comparing the Performance of Three Computational Methods for Estimating the Effective Reproduction Number.","authors":"Zihan Wang, Mengxia Xu, Zonglin Yang, Yu Jin, Yong Zhang","doi":"10.1089/cmb.2023.0065","DOIUrl":"10.1089/cmb.2023.0065","url":null,"abstract":"<p><p>The effective reproduction number <math><mrow><mo>(</mo><mrow><msub><mrow><mi>R</mi></mrow><mrow><mi>t</mi></mrow></msub></mrow><mo>)</mo></mrow></math> is one of the most important epidemiological parameters, providing suggestions for monitoring the development trend of diseases and also for adjusting the prevention and control policies. However, a few studies have focused on the performance of some common computational methods for <i>R<sub>t</sub></i>. The purpose of this article is to compare the performance of three computational methods for <i>R<sub>t</sub></i>: the time-dependent (TD) method, the new time-varying (NT) method, and the sequential Bayesian (SB) method. Four evaluation methods-accuracy, correlation coefficient, similarity based on trend, and dynamic time warping distance-were used to compare the effectiveness of three computational methods for <i>R<sub>t</sub></i> under different time lags and time windows. The results showed that the NT method was a better choice for real-time monitoring and analysis of the epidemic in the middle and late stages of the infectious disease. The TD method could reflect the change of the number of cases stably and accurately, and was more suitable for monitoring the change of <i>R<sub>t</sub></i> during the whole process of the epidemic outbreak. When the data were relatively stable, the SB method could also provide a reliable estimate for <i>R<sub>t</sub></i>, while the error would increase when the fluctuation in the number of cases increased. The results would provide suggestions for selecting appropriate <i>R<sub>t</sub></i> estimation methods and making policy adjustments more timely and effectively according to the change of <i>R<sub>t</sub></i>.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139472540","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
DeepPPThermo: A Deep Learning Framework for Predicting Protein Thermostability Combining Protein-Level and Amino Acid-Level Features. DeepPPThermo:结合蛋白质级和氨基酸级特征预测蛋白质热稳定性的深度学习框架。
IF 1.7 4区 生物学 Q2 Mathematics Pub Date : 2024-02-01 Epub Date: 2023-12-13 DOI: 10.1089/cmb.2023.0097
Xiaoyang Xiang, Jiaxuan Gao, Yanrui Ding

Using wet experimental methods to discover new thermophilic proteins or improve protein thermostability is time-consuming and expensive. Machine learning methods have shown powerful performance in the study of protein thermostability in recent years. However, how to make full use of multiview sequence information to predict thermostability effectively is still a challenge. In this study, we proposed a deep learning-based classifier named DeepPPThermo that fuses features of classical sequence features and deep learning representation features for classifying thermophilic and mesophilic proteins. In this model, deep neural network (DNN) and bi-long short-term memory (Bi-LSTM) are used to mine hidden features. Furthermore, local attention and global attention mechanisms give different importance to multiview features. The fused features are fed to a fully connected network classifier to distinguish thermophilic and mesophilic proteins. Our model is comprehensively compared with advanced machine learning algorithms and deep learning algorithms, proving that our model performs better. We further compare the effects of removing different features on the classification results, demonstrating the importance of each feature and the robustness of the model. Our DeepPPThermo model can be further used to explore protein diversity, identify new thermophilic proteins, and guide directed mutations of mesophilic proteins.

使用湿实验方法发现新的嗜热蛋白质或改善蛋白质的热稳定性既耗时又昂贵。近年来,机器学习方法在蛋白质耐热性研究中表现出了强大的性能。然而,如何充分利用多视角序列信息来有效预测热稳定性仍是一个挑战。在这项研究中,我们提出了一种基于深度学习的分类器,名为 DeepPPThermo,它融合了经典序列特征和深度学习表示特征,用于对嗜热蛋白质和中嗜热蛋白质进行分类。在该模型中,深度神经网络(DNN)和双长短期记忆(Bi-LSTM)被用来挖掘隐藏特征。此外,局部注意力和全局注意力机制对多视角特征赋予了不同的重要性。融合后的特征被输入一个全连接网络分类器,以区分嗜热蛋白质和嗜中蛋白质。我们的模型与先进的机器学习算法和深度学习算法进行了综合比较,证明我们的模型性能更好。我们进一步比较了去除不同特征对分类结果的影响,证明了每个特征的重要性和模型的鲁棒性。我们的 DeepPPThermo 模型可进一步用于探索蛋白质多样性、识别新的嗜热蛋白质以及指导中嗜热蛋白质的定向突变。
{"title":"DeepPPThermo: A Deep Learning Framework for Predicting Protein Thermostability Combining Protein-Level and Amino Acid-Level Features.","authors":"Xiaoyang Xiang, Jiaxuan Gao, Yanrui Ding","doi":"10.1089/cmb.2023.0097","DOIUrl":"10.1089/cmb.2023.0097","url":null,"abstract":"<p><p>Using wet experimental methods to discover new thermophilic proteins or improve protein thermostability is time-consuming and expensive. Machine learning methods have shown powerful performance in the study of protein thermostability in recent years. However, how to make full use of multiview sequence information to predict thermostability effectively is still a challenge. In this study, we proposed a deep learning-based classifier named DeepPPThermo that fuses features of classical sequence features and deep learning representation features for classifying thermophilic and mesophilic proteins. In this model, deep neural network (DNN) and bi-long short-term memory (Bi-LSTM) are used to mine hidden features. Furthermore, local attention and global attention mechanisms give different importance to multiview features. The fused features are fed to a fully connected network classifier to distinguish thermophilic and mesophilic proteins. Our model is comprehensively compared with advanced machine learning algorithms and deep learning algorithms, proving that our model performs better. We further compare the effects of removing different features on the classification results, demonstrating the importance of each feature and the robustness of the model. Our DeepPPThermo model can be further used to explore protein diversity, identify new thermophilic proteins, and guide directed mutations of mesophilic proteins.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138805058","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Privacy-Preserving Identification of Cancer Subtype-Specific Driver Genes Based on Multigenomics Data with Privatedriver. 基于多基因组学数据的癌症亚型特异性驱动基因的隐私保护鉴定(Privatedriver)。
IF 1.7 4区 生物学 Q2 Mathematics Pub Date : 2024-02-01 Epub Date: 2024-01-25 DOI: 10.1089/cmb.2023.0115
Junrong Song, Zhiming Song, Jinpeng Zhang, Yuanli Gong

Identifying cancer subtype-specific driver genes from a large number of irrelevant passengers is crucial for targeted therapy in cancer treatment. Recently, the rapid accumulation of large-scale cancer genomics data from multiple institutions has presented remarkable opportunities for identification of cancer subtype-specific driver genes. However, the insufficient subtype samples, privacy issues, and heterogenous of aberration events pose great challenges in precisely identifying cancer subtype-specific driver genes. To address this, we introduce privatedriver, the first model for identifying subtype-specific driver genes that integrates genomics data from multiple institutions in a data privacy-preserving collaboration manner. The process of identifying subtype-specific cancer driver genes using privatedriver involves the following two steps: genomics data integration and collaborative training. In the integration process, the aberration events from multiple genomics data sources are combined for each institution using the forward and backward propagation method of NetICS. In the collaborative training process, each institution utilizes the federated learning framework to upload encrypted model parameters instead of raw data of all institutions to train a global model by using the non-negative matrix factorization algorithm. We applied privatedriver on head and neck squamous cell and colon cancer from The Cancer Genome Atlas website and evaluated it with two benchmarks using macro-Fscore. The comparison analysis demonstrates that privatedriver achieves comparable results to centralized learning models and outperforms most other nonprivacy preserving models, all while ensuring the confidentiality of patient information. We also demonstrate that, for varying predicted driver gene distributions in subtype, our model fully considers the heterogeneity of subtype and identifies subtype-specific driver genes corresponding to the given prognosis and therapeutic effect. The success of privatedriver reveals the feasibility and effectiveness of identifying cancer subtype-specific driver genes in a data protection manner, providing new insights for future privacy-preserving driver gene identification studies.

从大量无关基因中识别癌症亚型特异性驱动基因对于癌症治疗中的靶向治疗至关重要。最近,来自多个机构的大规模癌症基因组学数据的快速积累为鉴定癌症亚型特异性驱动基因提供了难得的机会。然而,亚型样本不足、隐私问题和畸变事件的异质性给精确鉴定癌症亚型特异性驱动基因带来了巨大挑战。为解决这一问题,我们引入了privatedriver,这是首个以数据隐私保护协作方式整合多个机构基因组学数据的亚型特异性驱动基因鉴定模型。使用 privatedriver 识别亚型特异性癌症驱动基因的过程包括以下两个步骤:基因组学数据整合和协作训练。在整合过程中,利用 NetICS 的前向和后向传播方法将来自多个基因组学数据源的畸变事件合并到每个机构。在协作训练过程中,各机构利用联合学习框架上传加密的模型参数,而不是所有机构的原始数据,通过非负矩阵因式分解算法训练全局模型。我们将privatedriver应用于癌症基因组图谱网站上的头颈部鳞状细胞癌和结肠癌,并用macro-Fscore与两个基准进行了评估。对比分析表明,privatedriver 在确保患者信息保密的前提下,取得了与集中式学习模型相当的结果,并优于大多数其他非隐私保护模型。我们还证明,对于亚型中不同的预测驱动基因分布,我们的模型充分考虑了亚型的异质性,并识别出与给定预后和治疗效果相对应的亚型特异性驱动基因。privatedriver 的成功揭示了以数据保护的方式识别癌症亚型特异性驱动基因的可行性和有效性,为未来保护隐私的驱动基因识别研究提供了新的启示。
{"title":"Privacy-Preserving Identification of Cancer Subtype-Specific Driver Genes Based on Multigenomics Data with Privatedriver.","authors":"Junrong Song, Zhiming Song, Jinpeng Zhang, Yuanli Gong","doi":"10.1089/cmb.2023.0115","DOIUrl":"10.1089/cmb.2023.0115","url":null,"abstract":"<p><p>Identifying cancer subtype-specific driver genes from a large number of irrelevant passengers is crucial for targeted therapy in cancer treatment. Recently, the rapid accumulation of large-scale cancer genomics data from multiple institutions has presented remarkable opportunities for identification of cancer subtype-specific driver genes. However, the insufficient subtype samples, privacy issues, and heterogenous of aberration events pose great challenges in precisely identifying cancer subtype-specific driver genes. To address this, we introduce privatedriver, the first model for identifying subtype-specific driver genes that integrates genomics data from multiple institutions in a data privacy-preserving collaboration manner. The process of identifying subtype-specific cancer driver genes using privatedriver involves the following two steps: genomics data integration and collaborative training. In the integration process, the aberration events from multiple genomics data sources are combined for each institution using the forward and backward propagation method of NetICS. In the collaborative training process, each institution utilizes the federated learning framework to upload encrypted model parameters instead of raw data of all institutions to train a global model by using the non-negative matrix factorization algorithm. We applied privatedriver on head and neck squamous cell and colon cancer from The Cancer Genome Atlas website and evaluated it with two benchmarks using macro-Fscore. The comparison analysis demonstrates that privatedriver achieves comparable results to centralized learning models and outperforms most other nonprivacy preserving models, all while ensuring the confidentiality of patient information. We also demonstrate that, for varying predicted driver gene distributions in subtype, our model fully considers the heterogeneity of subtype and identifies subtype-specific driver genes corresponding to the given prognosis and therapeutic effect. The success of privatedriver reveals the feasibility and effectiveness of identifying cancer subtype-specific driver genes in a data protection manner, providing new insights for future privacy-preserving driver gene identification studies.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139564179","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
MiCId GUI: The Graphical User Interface for MiCId, a Fast Microorganism Classification and Identification Workflow with Accurate Statistics and High Recall. MiCId GUI:MiCId 的图形用户界面,这是一种具有精确统计和高召回率的快速微生物分类和鉴定工作流程。
IF 1.7 4区 生物学 Q2 Mathematics Pub Date : 2024-02-01 Epub Date: 2024-02-02 DOI: 10.1089/cmb.2023.0149
Aleksey Ogurtsov, Gelio Alves, Alex Rubio, Brendan Joyce, Björn Andersson, Roger Karlsson, Edward R B Moore, Yi-Kuo Yu

Although many user-friendly workflows exist for identifications of peptides and proteins in mass-spectrometry-based proteomics, there is a need of easy to use, fast, and accurate workflows for identifications of microorganisms, antimicrobial resistant proteins, and biomass estimation. Identification of microorganisms is a computationally demanding task that requires querying thousands of MS/MS spectra in a database containing thousands to tens of thousands of microorganisms. Existing software can't handle such a task in a time efficient manner, taking hours to process a single MS/MS experiment. Another paramount factor to consider is the necessity of accurate statistical significance to properly control the proportion of false discoveries among the identified microorganisms, and antimicrobial-resistant proteins, and to provide robust biomass estimation. Recently, we have developed Microorganism Classification and Identification (MiCId) workflow that assigns accurate statistical significance to identified microorganisms, antimicrobial-resistant proteins, and biomass estimation. MiCId's workflow is also computationally efficient, taking about 6-17 minutes to process a tandem mass-spectrometry (MS/MS) experiment using computer resources that are available in most laptop and desktop computers, making it a portable workflow. To make data analysis accessible to a broader range of users, beyond users familiar with the Linux environment, we have developed a graphical user interface (GUI) for MiCId's workflow. The GUI brings to users all the functionality of MiCId's workflow in a friendly interface along with tools for data analysis, visualization, and to export results.

尽管在基于质谱的蛋白质组学中,有许多用户友好型工作流程可用于肽和蛋白质的鉴定,但在微生物、抗微生物蛋白和生物量估算的鉴定方面,仍需要简单易用、快速准确的工作流程。微生物鉴定是一项计算要求很高的任务,需要查询包含数千到数万种微生物的数据库中的数千个 MS/MS 图谱。现有软件无法高效地处理此类任务,处理一次 MS/MS 实验需要数小时。另一个需要考虑的重要因素是必须有准确的统计意义,以适当控制已鉴定微生物和抗微生物蛋白中的错误发现比例,并提供可靠的生物量估算。最近,我们开发了微生物分类和鉴定(MiCId)工作流程,该流程可为已鉴定的微生物、抗微生物蛋白和生物量估算赋予准确的统计意义。MiCId 工作流程的计算效率也很高,使用大多数笔记本电脑和台式电脑上的计算机资源处理串联质谱(MS/MS)实验大约需要 6-17 分钟,是一种便携式工作流程。为了让熟悉 Linux 环境的用户以外的更多用户也能使用数据分析,我们为 MiCId 的工作流程开发了图形用户界面(GUI)。图形用户界面以友好的界面为用户提供了 MiCId 工作流程的所有功能,以及数据分析、可视化和输出结果的工具。
{"title":"MiCId GUI: The Graphical User Interface for MiCId, a Fast Microorganism Classification and Identification Workflow with Accurate Statistics and High Recall.","authors":"Aleksey Ogurtsov, Gelio Alves, Alex Rubio, Brendan Joyce, Björn Andersson, Roger Karlsson, Edward R B Moore, Yi-Kuo Yu","doi":"10.1089/cmb.2023.0149","DOIUrl":"10.1089/cmb.2023.0149","url":null,"abstract":"<p><p>Although many user-friendly workflows exist for identifications of peptides and proteins in mass-spectrometry-based proteomics, there is a need of easy to use, fast, and accurate workflows for identifications of microorganisms, antimicrobial resistant proteins, and biomass estimation. Identification of microorganisms is a computationally demanding task that requires querying thousands of MS/MS spectra in a database containing thousands to tens of thousands of microorganisms. Existing software can't handle such a task in a time efficient manner, taking hours to process a single MS/MS experiment. Another paramount factor to consider is the necessity of accurate statistical significance to properly control the proportion of false discoveries among the identified microorganisms, and antimicrobial-resistant proteins, and to provide robust biomass estimation. Recently, we have developed Microorganism Classification and Identification (MiCId) workflow that assigns accurate statistical significance to identified microorganisms, antimicrobial-resistant proteins, and biomass estimation. MiCId's workflow is also computationally efficient, taking about 6-17 minutes to process a tandem mass-spectrometry (MS/MS) experiment using computer resources that are available in most laptop and desktop computers, making it a portable workflow. To make data analysis accessible to a broader range of users, beyond users familiar with the Linux environment, we have developed a graphical user interface (GUI) for MiCId's workflow. The GUI brings to users all the functionality of MiCId's workflow in a friendly interface along with tools for data analysis, visualization, and to export results.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10874827/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139671948","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Computing Minimal Boolean Models of Gene Regulatory Networks. 计算基因调控网络的最小布尔模型。
IF 1.7 4区 生物学 Q2 Mathematics Pub Date : 2024-02-01 Epub Date: 2023-10-27 DOI: 10.1089/cmb.2023.0122
Guy Karlebach, Peter N Robinson

Models of gene regulatory networks (GRNs) capture the dynamics of the regulatory processes that occur within the cell as a means to understanding the variability observed in gene expression between different conditions. Arguably the simplest mathematical construct used for modeling is the Boolean network, which dictates a set of logical rules for transition between states described as Boolean vectors. Due to the complexity of gene regulation and the limitations of experimental technologies, in most cases knowledge about regulatory interactions and Boolean states is partial. In addition, the logical rules themselves are not known a priori. Our goal in this work is to create an algorithm that finds the network that fits the data optimally, and identify the network states that correspond to the noise-free data. We present a novel methodology for integrating experimental data and performing a search for the optimal consistent structure via optimization of a linear objective function under a set of linear constraints. In addition, we extend our methodology into a heuristic that alleviates the computational complexity of the problem for datasets that are generated by single-cell RNA-Sequencing (scRNA-Seq). We demonstrate the effectiveness of these tools using simulated data, and in addition a publicly available scRNA-Seq dataset and the GRN that is associated with it. Our methodology will enable researchers to obtain a better understanding of the dynamics of GRNs and their biological role.

基因调控网络(GRN)模型捕捉细胞内发生的调控过程的动力学,作为理解不同条件下观察到的基因表达变异性的一种手段。可以说,用于建模的最简单的数学结构是布尔网络,它规定了一组逻辑规则,用于在被描述为布尔向量的状态之间转换。由于基因调控的复杂性和实验技术的局限性,在大多数情况下,关于调控相互作用和布尔状态的知识是部分的。此外,逻辑规则本身并不是先验已知的。我们在这项工作中的目标是创建一种算法,找到最适合数据的网络,并识别与无噪声数据相对应的网络状态。我们提出了一种新的方法,用于集成实验数据,并通过在一组线性约束下优化线性目标函数来搜索最优一致结构。此外,我们将我们的方法扩展为启发式方法,以减轻单细胞RNA测序(scRNA-Seq)生成的数据集的计算复杂性。我们使用模拟数据以及公开的scRNA-Seq数据集和与之相关的GRN来证明这些工具的有效性。我们的方法将使研究人员能够更好地了解GRN的动力学及其生物学作用。
{"title":"Computing Minimal Boolean Models of Gene Regulatory Networks.","authors":"Guy Karlebach, Peter N Robinson","doi":"10.1089/cmb.2023.0122","DOIUrl":"10.1089/cmb.2023.0122","url":null,"abstract":"<p><p>Models of gene regulatory networks (GRNs) capture the dynamics of the regulatory processes that occur within the cell as a means to understanding the variability observed in gene expression between different conditions. Arguably the simplest mathematical construct used for modeling is the Boolean network, which dictates a set of logical rules for transition between states described as Boolean vectors. Due to the complexity of gene regulation and the limitations of experimental technologies, in most cases knowledge about regulatory interactions and Boolean states is partial. In addition, the logical rules themselves are not known a priori. Our goal in this work is to create an algorithm that finds the network that fits the data optimally, and identify the network states that correspond to the noise-free data. We present a novel methodology for integrating experimental data and performing a search for the optimal consistent structure via optimization of a linear objective function under a set of linear constraints. In addition, we extend our methodology into a heuristic that alleviates the computational complexity of the problem for datasets that are generated by single-cell RNA-Sequencing (scRNA-Seq). We demonstrate the effectiveness of these tools using simulated data, and in addition a publicly available scRNA-Seq dataset and the GRN that is associated with it. Our methodology will enable researchers to obtain a better understanding of the dynamics of GRNs and their biological role.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"61562871","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Repeated Decision Stumping Distils Simple Rules from Single-Cell Data. 从单细胞数据中提取简单规则的重复决策难题
IF 1.7 4区 生物学 Q2 Mathematics Pub Date : 2024-01-01 Epub Date: 2024-01-04 DOI: 10.1089/cmb.2021.0613
Ivan A Croydon-Veleslavov, Michael P H Stumpf

Single-cell data afford unprecedented insights into molecular processes. But the complexity and size of these data sets have proved challenging and given rise to a large armory of statistical and machine learning approaches. The majority of approaches focuses on either describing features of these data, or making predictions and classifying unlabeled samples. In this study, we introduce repeated decision stumping (ReDX) as a method to distill simple models from single-cell data. We develop decision trees of depth one-hence "stumps"-to identify in an inductive manner, gene products involved in driving cell fate transitions, and in applications to published data we are able to discover the key players involved in these processes in an unbiased manner without prior knowledge. Our algorithm is deliberately targeting the simplest possible candidate hypotheses that can be extracted from complex high-dimensional data. There are three reasons for this: (1) the predictions become straightforwardly testable hypotheses; (2) the identified candidates form the basis for further mechanistic model development, for example, for engineering and synthetic biology interventions; and (3) this approach complements existing descriptive modeling approaches and frameworks. The approach is computationally efficient, has remarkable predictive power, including in simulation studies where the ground truth is known, and yields robust and statistically stable predictors; the same set of candidates is generated by applying the algorithm to different subsamples of experimental data.

单细胞数据提供了前所未有的分子过程洞察力。但事实证明,这些数据集的复杂性和规模具有挑战性,并催生了大量统计和机器学习方法。大多数方法都侧重于描述这些数据的特征,或对未标记的样本进行预测和分类。在这项研究中,我们引入了重复判定法(ReDX),作为一种从单细胞数据中提炼简单模型的方法。我们开发了深度为一的决策树--即 "树桩"--以归纳的方式识别参与驱动细胞命运转换的基因产物,在应用于已发表的数据时,我们能够在没有先验知识的情况下,以无偏见的方式发现参与这些过程的关键角色。我们的算法特意针对可以从复杂的高维数据中提取的最简单的候选假设。这样做有三个原因:(1) 预测成为可直接检验的假说;(2) 确定的候选假说为进一步的机理模型开发奠定了基础,例如用于工程和合成生物学干预;(3) 这种方法是对现有描述性建模方法和框架的补充。该方法计算效率高,预测能力强,包括在已知基本事实的模拟研究中,并能产生稳健且统计稳定的预测结果;将该算法应用于不同的实验数据子样本,可生成相同的候选集。
{"title":"Repeated Decision Stumping Distils Simple Rules from Single-Cell Data.","authors":"Ivan A Croydon-Veleslavov, Michael P H Stumpf","doi":"10.1089/cmb.2021.0613","DOIUrl":"10.1089/cmb.2021.0613","url":null,"abstract":"<p><p>Single-cell data afford unprecedented insights into molecular processes. But the complexity and size of these data sets have proved challenging and given rise to a large armory of statistical and machine learning approaches. The majority of approaches focuses on either describing features of these data, or making predictions and classifying unlabeled samples. In this study, we introduce repeated decision stumping (ReDX) as a method to distill simple models from single-cell data. We develop decision trees of depth one-hence \"stumps\"-to identify in an inductive manner, gene products involved in driving cell fate transitions, and in applications to published data we are able to discover the key players involved in these processes in an unbiased manner without prior knowledge. Our algorithm is deliberately targeting the simplest possible candidate hypotheses that can be extracted from complex high-dimensional data. There are three reasons for this: (1) the predictions become straightforwardly testable hypotheses; (2) the identified candidates form the basis for further mechanistic model development, for example, for engineering and synthetic biology interventions; and (3) this approach complements existing descriptive modeling approaches and frameworks. The approach is computationally efficient, has remarkable predictive power, including in simulation studies where the ground truth is known, and yields robust and statistically stable predictors; the same set of candidates is generated by applying the algorithm to different subsamples of experimental data.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139087092","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Density and Conservation Optimization of the Generalized Masked-Minimizer Sketching Scheme. 广义掩模最小草图方案的密度和守恒优化。
IF 1.7 4区 生物学 Q2 Mathematics Pub Date : 2024-01-01 Epub Date: 2023-11-17 DOI: 10.1089/cmb.2023.0212
Minh Hoang, Guillaume Marçais, Carl Kingsford

Minimizers and syncmers are sketching methods that sample representative k-mer seeds from a long string. The minimizer scheme guarantees a well-spread k-mer sketch (high coverage) while seeking to minimize the sketch size (low density). The syncmer scheme yields sketches that are more robust to base substitutions (high conservation) on random sequences, but do not have the coverage guarantee of minimizers. These sketching metrics are generally adversarial to one another, especially in the context of sketch optimization for a specific sequence, and thus are difficult to be simultaneously achieved. The parameterized syncmer scheme was recently introduced as a generalization of syncmers with more flexible sampling rules and empirically better coverage than the original syncmer variants. However, no approach exists to optimize parameterized syncmers. To address this shortcoming, we introduce a new scheme called masked minimizers that generalizes minimizers in manner analogous to how parameterized syncmers generalize syncmers and allows us to extend existing optimization techniques developed for minimizers. This results in a practical algorithm to optimize the masked minimizer scheme with respect to both density and conservation. We evaluate the optimization algorithm on various benchmark genomes and show that our algorithm finds sketches that are overall more compact, well-spread, and robust to substitutions than those found by previous methods. Our implementation is released at https://github.com/Kingsford-Group/maskedminimizer. This new technique will enable more efficient and robust genomic analyses in the many settings where minimizers and syncmers are used.

最小化器和同步器是从长串中采样代表性k-mer种子的素描方法。最小化方案保证了良好的k-mer草图(高覆盖率),同时寻求最小化草图尺寸(低密度)。synsynmer方案产生的草图对随机序列上的基替换(高守恒)具有更强的鲁棒性,但不具有最小化的覆盖保证。这些草图度量通常是相互对抗的,特别是在特定序列的草图优化环境中,因此很难同时实现。最近引入了参数化同步器方案,作为同步器的一种推广,它具有更灵活的采样规则和经验上比原始同步器变体更好的覆盖率。然而,目前还没有优化参数化同步器的方法。为了解决这个缺点,我们引入了一种名为掩码最小化器的新方案,它以类似于参数化同步器泛化同步器的方式泛化最小化器,并允许我们扩展为最小化器开发的现有优化技术。这导致了一个实用的算法来优化掩模最小化方案,同时考虑密度和守恒。我们在各种基准基因组上评估了优化算法,并表明我们的算法发现的草图总体上比以前的方法发现的草图更紧凑、分布良好、对替换更健壮。我们的实现发布在https://github.com/Kingsford-Group/maskedminimizer。这项新技术将在许多使用最小化和同步器的环境中实现更有效和健壮的基因组分析。
{"title":"Density and Conservation Optimization of the Generalized Masked-Minimizer Sketching Scheme.","authors":"Minh Hoang, Guillaume Marçais, Carl Kingsford","doi":"10.1089/cmb.2023.0212","DOIUrl":"10.1089/cmb.2023.0212","url":null,"abstract":"<p><p>Minimizers and syncmers are sketching methods that sample representative <i>k</i>-mer seeds from a long string. The minimizer scheme guarantees a well-spread <i>k</i>-mer sketch (high coverage) while seeking to minimize the sketch size (low density). The syncmer scheme yields sketches that are more robust to base substitutions (high conservation) on random sequences, but do not have the coverage guarantee of minimizers. These sketching metrics are generally adversarial to one another, especially in the context of sketch optimization for a specific sequence, and thus are difficult to be simultaneously achieved. The parameterized syncmer scheme was recently introduced as a generalization of syncmers with more flexible sampling rules and empirically better coverage than the original syncmer variants. However, no approach exists to optimize parameterized syncmers. To address this shortcoming, we introduce a new scheme called masked minimizers that generalizes minimizers in manner analogous to how parameterized syncmers generalize syncmers and allows us to extend existing optimization techniques developed for minimizers. This results in a practical algorithm to optimize the masked minimizer scheme with respect to both density and conservation. We evaluate the optimization algorithm on various benchmark genomes and show that our algorithm finds sketches that are overall more compact, well-spread, and robust to substitutions than those found by previous methods. Our implementation is released at https://github.com/Kingsford-Group/maskedminimizer. This new technique will enable more efficient and robust genomic analyses in the many settings where minimizers and syncmers are used.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10794853/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"136397678","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A Gene Selection Method Considering Measurement Errors. 考虑测量误差的基因选择方法。
IF 1.7 4区 生物学 Q2 Mathematics Pub Date : 2024-01-01 Epub Date: 2023-11-21 DOI: 10.1089/cmb.2023.0041
Hajoung Lee, Jaejik Kim

The analysis of gene expression data has made significant contributions to understanding disease mechanisms and developing new drugs and therapies. In such analysis, gene selection is often required for identifying informative and relevant genes and removing redundant and irrelevant ones. However, this is not an easy task as gene expression data have inherent challenges such as ultra-high dimensionality, biological noise, and measurement errors. This study focuses on the measurement errors in gene selection problems. Typically, high-throughput experiments have their own intrinsic measurement errors, which can result in an increase of falsely discovered genes. To alleviate this problem, this study proposes a gene selection method that takes into account measurement errors using generalized liner measurement error models. The method consists of iterative filtering and selection steps until convergence, leading to fewer false positives and providing stable results under measurement errors. The performance of the proposed method is demonstrated through simulation studies and applied to a lung cancer data set.

基因表达数据的分析对了解疾病机制和开发新的药物和治疗方法做出了重大贡献。在这种分析中,通常需要基因选择来识别信息丰富和相关的基因,并去除冗余和不相关的基因。然而,这并不是一项容易的任务,因为基因表达数据具有固有的挑战,如超高维度、生物噪声和测量误差。本文主要研究基因选择问题中的测量误差。通常,高通量实验有其固有的测量误差,这可能导致错误发现基因的增加。为了缓解这一问题,本研究提出了一种利用广义线性测量误差模型考虑测量误差的基因选择方法。该方法由迭代滤波和选择步骤组成,直到收敛,导致假阳性较少,并且在测量误差下提供稳定的结果。通过仿真研究证明了该方法的有效性,并将其应用于肺癌数据集。
{"title":"A Gene Selection Method Considering Measurement Errors.","authors":"Hajoung Lee, Jaejik Kim","doi":"10.1089/cmb.2023.0041","DOIUrl":"10.1089/cmb.2023.0041","url":null,"abstract":"<p><p>The analysis of gene expression data has made significant contributions to understanding disease mechanisms and developing new drugs and therapies. In such analysis, gene selection is often required for identifying informative and relevant genes and removing redundant and irrelevant ones. However, this is not an easy task as gene expression data have inherent challenges such as ultra-high dimensionality, biological noise, and measurement errors. This study focuses on the measurement errors in gene selection problems. Typically, high-throughput experiments have their own intrinsic measurement errors, which can result in an increase of falsely discovered genes. To alleviate this problem, this study proposes a gene selection method that takes into account measurement errors using generalized liner measurement error models. The method consists of iterative filtering and selection steps until convergence, leading to fewer false positives and providing stable results under measurement errors. The performance of the proposed method is demonstrated through simulation studies and applied to a lung cancer data set.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138444862","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Journal of Computational Biology
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
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