首页 > 最新文献

Journal of Computational Biology最新文献

英文 中文
Estimating Haplotype Structure and Frequencies: A Bayesian Approach to Unknown Design in Pooled Genomic Data. 估计单倍型结构和频率:在集合基因组数据中进行未知设计的贝叶斯方法。
IF 1.4 4区 生物学 Q4 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-03 DOI: 10.1089/cmb.2023.0211
Yuexuan Wang, Ritabrata Dutta, Andreas Futschik

The estimation of haplotype structure and frequencies provides crucial information about the composition of genomes. Techniques, such as single-individual haplotyping, aim to reconstruct individual haplotypes from diploid genome sequencing data. However, our focus is distinct. We address the challenge of reconstructing haplotype structure and frequencies from pooled sequencing samples where multiple individuals are sequenced simultaneously. A frequentist method to address this issue has recently been proposed. In contrast to this and other methods that compute point estimates, our proposed Bayesian hierarchical model delivers a posterior that permits us to also quantify uncertainty. Since matching permutations in both haplotype structure and corresponding frequency matrix lead to the same reconstruction of their product, we introduce an order-preserving shrinkage prior that ensures identifiability with respect to permutations. For inference, we introduce a blocked Gibbs sampler that enforces the required constraints. In a simulation study, we assessed the performance of our method. Furthermore, by using our approach on two distinct sets of real data, we demonstrate that our Bayesian approach can reconstruct the dominant haplotypes in a challenging, high-dimensional set-up.

单倍型结构和频率的估计提供了有关基因组组成的重要信息。单体单倍型等技术旨在从二倍体基因组测序数据中重建单体单倍型。然而,我们的研究重点与众不同。我们要解决的难题是如何从多个个体同时测序的集合测序样本中重建单倍型结构和频率。最近有人提出了一种频数法来解决这个问题。与这种方法和其他计算点估计值的方法不同,我们提出的贝叶斯分层模型提供的后验值允许我们量化不确定性。由于单倍型结构和相应频率矩阵中的匹配排列会导致对其乘积的相同重构,因此我们引入了保序收缩先验,以确保排列的可识别性。在推断方面,我们引入了一个封锁式吉布斯采样器,以强制执行所需的约束条件。在模拟研究中,我们评估了我们方法的性能。此外,通过在两组不同的真实数据中使用我们的方法,我们证明了我们的贝叶斯方法可以在具有挑战性的高维环境中重建显性单倍型。
{"title":"Estimating Haplotype Structure and Frequencies: A Bayesian Approach to Unknown Design in Pooled Genomic Data.","authors":"Yuexuan Wang, Ritabrata Dutta, Andreas Futschik","doi":"10.1089/cmb.2023.0211","DOIUrl":"https://doi.org/10.1089/cmb.2023.0211","url":null,"abstract":"<p><p>The estimation of haplotype structure and frequencies provides crucial information about the composition of genomes. Techniques, such as single-individual haplotyping, aim to reconstruct individual haplotypes from diploid genome sequencing data. However, our focus is distinct. We address the challenge of reconstructing haplotype structure and frequencies from pooled sequencing samples where multiple individuals are sequenced simultaneously. A frequentist method to address this issue has recently been proposed. In contrast to this and other methods that compute point estimates, our proposed Bayesian hierarchical model delivers a posterior that permits us to also quantify uncertainty. Since matching permutations in both haplotype structure and corresponding frequency matrix lead to the same reconstruction of their product, we introduce an order-preserving shrinkage prior that ensures identifiability with respect to permutations. For inference, we introduce a blocked Gibbs sampler that enforces the required constraints. In a simulation study, we assessed the performance of our method. Furthermore, by using our approach on two distinct sets of real data, we demonstrate that our Bayesian approach can reconstruct the dominant haplotypes in a challenging, high-dimensional set-up.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.4,"publicationDate":"2024-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141492186","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
Detection and Segmentation of Glioma Tumors Utilizing a UNet Convolutional Neural Network Approach with Non-Subsampled Shearlet Transform. 利用 UNet 卷积神经网络方法和非子采样剪切力变换检测和分割胶质瘤肿瘤
IF 1.4 4区 生物学 Q4 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-06-27 DOI: 10.1089/cmb.2023.0339
Tamilarasi M, Kumarganesh S, K Martin Sagayam, Andrew J

The prompt and precise identification and delineation of tumor regions within glioma brain images are critical for mitigating the risks associated with this life-threatening ailment. In this study, we employ the UNet convolutional neural network (CNN) architecture for glioma tumor detection. Our proposed methodology comprises a transformation module, a feature extraction module, and a tumor segmentation module. The spatial domain representation of brain magnetic resonance imaging images undergoes decomposition into low- and high-frequency subbands via a non-subsampled shearlet transform. Leveraging the selective and directive characteristics of this transform enhances the classification efficacy of our proposed system. Shearlet features are extracted from both low- and high-frequency subbands and subsequently classified using the UNet-CNN architecture to identify tumor regions within glioma brain images. We validate our proposed glioma tumor detection methodology using publicly available datasets, namely Brain Tumor Segmentation (BRATS) 2019 and The Cancer Genome Atlas (TCGA). The mean classification rates achieved by our system are 99.1% for the BRATS 2019 dataset and 97.8% for the TCGA dataset. Furthermore, our system demonstrates notable performance metrics on the BRATS 2019 dataset, including 98.2% sensitivity, 98.7% specificity, 98.9% accuracy, 98.7% intersection over union, and 98.5% disc similarity coefficient. Similarly, on the TCGA dataset, our system achieves 97.7% sensitivity, 98.2% specificity, 98.7% accuracy, 98.6% intersection over union, and 98.4% disc similarity coefficient. Comparative analysis against state-of-the-art methods underscores the efficacy of our proposed glioma brain tumor detection approach.

及时、准确地识别和划分脑胶质瘤图像中的肿瘤区域,对于降低这种危及生命的疾病带来的风险至关重要。在本研究中,我们采用 UNet 卷积神经网络(CNN)架构来检测胶质瘤肿瘤。我们提出的方法包括转换模块、特征提取模块和肿瘤分割模块。脑磁共振成像图像的空间域表示通过非小样本剪切变换分解为低频和高频子带。利用这种变换的选择性和指向性特征,可提高我们所提系统的分类效率。从低频和高频子带中提取小剪切特征,然后使用 UNet-CNN 架构进行分类,从而识别脑胶质瘤脑图像中的肿瘤区域。我们使用公开可用的数据集(即脑肿瘤分割(BRATS)2019 和癌症基因组图谱(TCGA))验证了我们提出的胶质瘤肿瘤检测方法。我们的系统在 BRATS 2019 数据集上的平均分类率为 99.1%,在 TCGA 数据集上的平均分类率为 97.8%。此外,我们的系统还在 BRATS 2019 数据集上展示了显著的性能指标,包括 98.2% 的灵敏度、98.7% 的特异性、98.9% 的准确性、98.7% 的交集大于联合以及 98.5% 的圆盘相似系数。同样,在 TCGA 数据集上,我们的系统也达到了 97.7% 的灵敏度、98.2% 的特异性、98.7% 的准确性、98.6% 的交集大于联合以及 98.4% 的圆盘相似系数。与最先进方法的对比分析凸显了我们提出的胶质瘤脑肿瘤检测方法的功效。
{"title":"Detection and Segmentation of Glioma Tumors Utilizing a UNet Convolutional Neural Network Approach with Non-Subsampled Shearlet Transform.","authors":"Tamilarasi M, Kumarganesh S, K Martin Sagayam, Andrew J","doi":"10.1089/cmb.2023.0339","DOIUrl":"https://doi.org/10.1089/cmb.2023.0339","url":null,"abstract":"<p><p>The prompt and precise identification and delineation of tumor regions within glioma brain images are critical for mitigating the risks associated with this life-threatening ailment. In this study, we employ the UNet convolutional neural network (CNN) architecture for glioma tumor detection. Our proposed methodology comprises a transformation module, a feature extraction module, and a tumor segmentation module. The spatial domain representation of brain magnetic resonance imaging images undergoes decomposition into low- and high-frequency subbands via a non-subsampled shearlet transform. Leveraging the selective and directive characteristics of this transform enhances the classification efficacy of our proposed system. Shearlet features are extracted from both low- and high-frequency subbands and subsequently classified using the UNet-CNN architecture to identify tumor regions within glioma brain images. We validate our proposed glioma tumor detection methodology using publicly available datasets, namely Brain Tumor Segmentation (BRATS) 2019 and The Cancer Genome Atlas (TCGA). The mean classification rates achieved by our system are 99.1% for the BRATS 2019 dataset and 97.8% for the TCGA dataset. Furthermore, our system demonstrates notable performance metrics on the BRATS 2019 dataset, including 98.2% sensitivity, 98.7% specificity, 98.9% accuracy, 98.7% intersection over union, and 98.5% disc similarity coefficient. Similarly, on the TCGA dataset, our system achieves 97.7% sensitivity, 98.2% specificity, 98.7% accuracy, 98.6% intersection over union, and 98.4% disc similarity coefficient. Comparative analysis against state-of-the-art methods underscores the efficacy of our proposed glioma brain tumor detection approach.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.4,"publicationDate":"2024-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141457116","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
Estimating Enzyme Expression and Metabolic Pathway Activity in Borreliella-Infected and Uninfected Mice. 估计感染博雷利杆菌和未感染博雷利杆菌小鼠的酶表达和代谢途径活性
IF 1.4 4区 生物学 Q4 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-06-27 DOI: 10.1089/cmb.2024.0564
Filipp Martin Rondel, Hafsa Farooq, Roya Hosseini, Akshay Juyal, Sergey Knyazev, Serghei Mangul, Artem S Rogovskyy, Alexander Zelikovsky

Evaluating changes in metabolic pathway activity is essential for studying disease mechanisms and developing new treatments, with significant benefits extending to human health. Here, we propose EMPathways2, a maximum likelihood pipeline that is based on the expectation-maximization algorithm, which is capable of evaluating enzyme expression and metabolic pathway activity level. We first estimate enzyme expression from RNA-seq data that is used for simultaneous estimation of pathway activity levels using enzyme participation levels in each pathway. We implement the novel pipeline to RNA-seq data from several groups of mice, which provides a deeper look at the biochemical changes occurring as a result of bacterial infection, disease, and immune response. Our results show that estimated enzyme expression, pathway activity levels, and enzyme participation levels in each pathway are robust and stable across all samples. Estimated activity levels of a significant number of metabolic pathways strongly correlate with the infected and uninfected status of the respective rodent types.

评估代谢途径活性的变化对于研究疾病机制和开发新的治疗方法至关重要,对人类健康大有裨益。在此,我们提出了基于期望最大化算法的最大似然管道 EMPathways2,它能够评估酶的表达和代谢途径的活性水平。我们首先从 RNA-seq 数据中估算酶的表达量,然后利用酶在各通路中的参与度同时估算通路的活性水平。我们对几组小鼠的 RNA-seq 数据实施了这一新型管道,从而更深入地了解了细菌感染、疾病和免疫反应导致的生化变化。我们的研究结果表明,在所有样本中,估计的酶表达量、通路活性水平以及酶在每条通路中的参与水平都是稳健而稳定的。大量代谢途径的估计活性水平与相应啮齿类动物的感染和未感染状态密切相关。
{"title":"Estimating Enzyme Expression and Metabolic Pathway Activity in <i>Borreliella</i>-Infected and Uninfected Mice.","authors":"Filipp Martin Rondel, Hafsa Farooq, Roya Hosseini, Akshay Juyal, Sergey Knyazev, Serghei Mangul, Artem S Rogovskyy, Alexander Zelikovsky","doi":"10.1089/cmb.2024.0564","DOIUrl":"https://doi.org/10.1089/cmb.2024.0564","url":null,"abstract":"<p><p>Evaluating changes in metabolic pathway activity is essential for studying disease mechanisms and developing new treatments, with significant benefits extending to human health. Here, we propose EMPathways2, a maximum likelihood pipeline that is based on the expectation-maximization algorithm, which is capable of evaluating enzyme expression and metabolic pathway activity level. We first estimate enzyme expression from RNA-seq data that is used for simultaneous estimation of pathway activity levels using enzyme participation levels in each pathway. We implement the novel pipeline to RNA-seq data from several groups of mice, which provides a deeper look at the biochemical changes occurring as a result of bacterial infection, disease, and immune response. Our results show that estimated enzyme expression, pathway activity levels, and enzyme participation levels in each pathway are robust and stable across all samples. Estimated activity levels of a significant number of metabolic pathways strongly correlate with the infected and uninfected status of the respective rodent types.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.4,"publicationDate":"2024-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141457117","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
Nearly Instantaneous Time-Varying Reproduction Number for Contagious Diseases-a Direct Approach Based on Nonlinear Regression. 传染病的近瞬时时变繁殖数--基于非线性回归的直接方法
IF 1.4 4区 生物学 Q4 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-06-26 DOI: 10.1089/cmb.2023.0414
JūratĖ ŠaltytĖ Benth, Fred Espen Benth, Espen Rostrup Nakstad

While the world recovers from the COVID-19 pandemic, another outbreak of contagious disease remains the most likely future risk to public safety. Now is therefore the time to equip health authorities with effective tools to ensure they are operationally prepared for future events. We propose a direct approach to obtain reliable nearly instantaneous time-varying reproduction numbers for contagious diseases, using only the number of infected individuals as input and utilising the dynamics of the susceptible-infected-recovered (SIR) model. Our approach is based on a multivariate nonlinear regression model simultaneously assessing parameters describing the transmission and recovery rate as a function of the SIR model. Shortly after start of a pandemic, our approach enables estimation of daily reproduction numbers. It avoids numerous sources of additional variation and provides a generic tool for monitoring the instantaneous reproduction numbers. We use Norwegian COVID-19 data as case study and demonstrate that our results are well aligned with changes in the number of infected individuals and the change points following policy interventions. Our estimated reproduction numbers are notably less volatile, provide more credible short-time predictions for the number of infected individuals, and are thus clearly favorable compared with the results obtained by two other popular approaches used for monitoring a pandemic. The proposed approach contributes to increased preparedness to future pandemics of contagious diseases, as it can be used as a simple yet powerful tool to monitor the pandemics, provide short-term predictions, and thus support decision making regarding timely and targeted control measures.

在全球从 COVID-19 大流行中恢复过来的同时,传染性疾病的再次爆发仍然是未来公共安全最可能面临的风险。因此,现在正是为卫生部门提供有效工具的时候,以确保他们为未来事件做好行动准备。我们提出了一种直接方法,仅使用受感染个体的数量作为输入,并利用易感-感染-恢复(SIR)模型的动态变化,就能获得可靠的近乎瞬时的传染病时变繁殖数。我们的方法基于一个多变量非线性回归模型,同时评估作为 SIR 模型函数的描述传播率和恢复率的参数。大流行开始后不久,我们的方法就能估算出每天的繁殖数量。它避免了许多额外的变化来源,为监测瞬时繁殖数量提供了通用工具。我们使用挪威 COVID-19 数据作为案例研究,并证明我们的结果与感染人数的变化以及政策干预后的变化点非常吻合。我们估计的繁殖数量波动明显较小,对感染者数量的短时预测更加可信,因此与其他两种用于监测大流行病的流行方法相比,我们的结果明显更有优势。所提出的方法有助于提高对未来传染病大流行的防范能力,因为它可以作为一种简单而强大的工具来监测大流行,提供短期预测,从而为及时采取有针对性的控制措施提供决策支持。
{"title":"Nearly Instantaneous Time-Varying Reproduction Number for Contagious Diseases-a Direct Approach Based on Nonlinear Regression.","authors":"JūratĖ ŠaltytĖ Benth, Fred Espen Benth, Espen Rostrup Nakstad","doi":"10.1089/cmb.2023.0414","DOIUrl":"https://doi.org/10.1089/cmb.2023.0414","url":null,"abstract":"<p><p>While the world recovers from the COVID-19 pandemic, another outbreak of contagious disease remains the most likely future risk to public safety. Now is therefore the time to equip health authorities with effective tools to ensure they are operationally prepared for future events. We propose a direct approach to obtain reliable nearly instantaneous time-varying reproduction numbers for contagious diseases, using only the number of infected individuals as input and utilising the dynamics of the susceptible-infected-recovered (SIR) model. Our approach is based on a multivariate nonlinear regression model simultaneously assessing parameters describing the transmission and recovery rate as a function of the SIR model. Shortly after start of a pandemic, our approach enables estimation of daily reproduction numbers. It avoids numerous sources of additional variation and provides a generic tool for monitoring the instantaneous reproduction numbers. We use Norwegian COVID-19 data as case study and demonstrate that our results are well aligned with changes in the number of infected individuals and the change points following policy interventions. Our estimated reproduction numbers are notably less volatile, provide more credible short-time predictions for the number of infected individuals, and are thus clearly favorable compared with the results obtained by two other popular approaches used for monitoring a pandemic. The proposed approach contributes to increased preparedness to future pandemics of contagious diseases, as it can be used as a simple yet powerful tool to monitor the pandemics, provide short-term predictions, and thus support decision making regarding timely and targeted control measures.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.4,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141457118","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
NPI-DCGNN: An Accurate Tool for Identifying ncRNA-Protein Interactions Using a Dual-Channel Graph Neural Network. NPI-DCGNN:利用双通道图神经网络识别 ncRNA 与蛋白质相互作用的精确工具
IF 1.4 4区 生物学 Q4 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-06-26 DOI: 10.1089/cmb.2023.0449
Xin Zhang, Liangwei Zhao, Ziyi Chai, Hao Wu, Wei Yang, Chen Li, Yu Jiang, Quanzhong Liu

Noncoding RNA (NcRNA)-protein interactions (NPIs) play fundamentally important roles in carrying out cellular activities. Although various predictors based on molecular features and graphs have been published to boost the identification of NPIs, most of them often ignore the information between known NPIs or exhibit insufficient learning ability from graphs, posing a significant challenge in effectively identifying NPIs. To develop a more reliable and accurate predictor for NPIs, in this article, we propose NPI-DCGNN, an end-to-end NPI predictor based on a dual-channel graph neural network (DCGNN). NPI-DCGNN initially treats the known NPIs as an ncRNA-protein bipartite graph. Subsequently, for each ncRNA-protein pair, NPI-DCGNN extracts two local subgraphs centered around the ncRNA and protein, respectively, from the bipartite graph. After that, it utilizes a dual-channel graph representation learning layer based on GNN to generate high-level feature representations for the ncRNA-protein pair. Finally, it employs a fully connected network and output layer to predict whether an interaction exists between the pair of ncRNA and protein. Experimental results on four experimentally validated datasets demonstrate that NPI-DCGNN outperforms several state-of-the-art NPI predictors. Our case studies on the NPInter database further demonstrate the prediction power of NPI-DCGNN in predicting NPIs. With the availability of the source codes (https://github.com/zhangxin11111/NPI-DCGNN), we anticipate that NPI-DCGNN could facilitate the studies of ncRNA interactome by providing highly reliable NPI candidates for further experimental validation.

非编码 RNA(NcRNA)-蛋白质相互作用(NPIs)在细胞活动中发挥着极其重要的作用。虽然目前已有多种基于分子特征和图谱的预测方法来促进 NPIs 的鉴定,但大多数预测方法往往忽略了已知 NPIs 之间的信息,或者对图谱的学习能力不足,这给有效鉴定 NPIs 带来了巨大挑战。为了开发更可靠、更准确的 NPI 预测器,本文提出了基于双通道图神经网络(DCGNN)的端到端 NPI 预测器 NPI-DCGNN。NPI-DCGNN 最初将已知 NPI 视为 ncRNA 蛋白双向图。随后,对于每一对 ncRNA 蛋白,NPI-DCGNN 分别从双方图中提取以 ncRNA 和蛋白质为中心的两个局部子图。然后,它利用基于 GNN 的双通道图表示学习层为 ncRNA 蛋白对生成高级特征表示。最后,它利用全连接网络和输出层来预测 ncRNA 和蛋白质对之间是否存在相互作用。在四个经过实验验证的数据集上的实验结果表明,NPI-DCGNN 的性能优于几种最先进的 NPI 预测器。我们在 NPInter 数据库上进行的案例研究进一步证明了 NPI-DCGNN 在预测 NPI 方面的预测能力。随着源代码(https://github.com/zhangxin11111/NPI-DCGNN)的提供,我们预计 NPI-DCGNN 可以为进一步的实验验证提供高度可靠的 NPI 候选者,从而促进 ncRNA 相互作用组的研究。
{"title":"NPI-DCGNN: An Accurate Tool for Identifying ncRNA-Protein Interactions Using a Dual-Channel Graph Neural Network.","authors":"Xin Zhang, Liangwei Zhao, Ziyi Chai, Hao Wu, Wei Yang, Chen Li, Yu Jiang, Quanzhong Liu","doi":"10.1089/cmb.2023.0449","DOIUrl":"https://doi.org/10.1089/cmb.2023.0449","url":null,"abstract":"<p><p>Noncoding RNA (NcRNA)-protein interactions (NPIs) play fundamentally important roles in carrying out cellular activities. Although various predictors based on molecular features and graphs have been published to boost the identification of NPIs, most of them often ignore the information between known NPIs or exhibit insufficient learning ability from graphs, posing a significant challenge in effectively identifying NPIs. To develop a more reliable and accurate predictor for NPIs, in this article, we propose NPI-DCGNN, an end-to-end NPI predictor based on a dual-channel graph neural network (DCGNN). NPI-DCGNN initially treats the known NPIs as an ncRNA-protein bipartite graph. Subsequently, for each ncRNA-protein pair, NPI-DCGNN extracts two local subgraphs centered around the ncRNA and protein, respectively, from the bipartite graph. After that, it utilizes a dual-channel graph representation learning layer based on GNN to generate high-level feature representations for the ncRNA-protein pair. Finally, it employs a fully connected network and output layer to predict whether an interaction exists between the pair of ncRNA and protein. Experimental results on four experimentally validated datasets demonstrate that NPI-DCGNN outperforms several state-of-the-art NPI predictors. Our case studies on the NPInter database further demonstrate the prediction power of NPI-DCGNN in predicting NPIs. With the availability of the source codes (https://github.com/zhangxin11111/NPI-DCGNN), we anticipate that NPI-DCGNN could facilitate the studies of ncRNA interactome by providing highly reliable NPI candidates for further experimental validation.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.4,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141457119","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
Combining Complementarity and Binding Energetics in the Assessment of Protein Interactions: EnCPdock-A Practical Manual. 结合互补性和结合能评估蛋白质相互作用:EnCPdock - 实用手册
IF 1.7 4区 生物学 Q2 Mathematics Pub Date : 2024-06-17 DOI: 10.1089/cmb.2024.0554
Gargi Biswas, Debasish Mukherjee, Sankar Basu

The combined effect of shape and electrostatic complementarities (Sc, EC) at the interface of the interacting protein partners (PPI) serves as the physical basis for such associations and is a strong determinant of their binding energetics. EnCPdock (https://www.scinetmol.in/EnCPdock/) presents a comprehensive web platform for the direct conjoint comparative analyses of complementarity and binding energetics in PPIs. It elegantly interlinks the dual nature of local (Sc) and nonlocal complementarity (EC) in PPIs using the complementarity plot. It further derives an AI-based ΔGbinding with a prediction accuracy comparable to the state of the art. This book chapter presents a practical manual to conceptualize and implement EnCPdock with its various features and functionalities, collectively having the potential to serve as a valuable protein engineering tool in the design of novel protein interfaces.

在相互作用的蛋白质伙伴(PPI)界面上,形状和静电互补性(Sc、EC)的共同作用是这种结合的物理基础,也是其结合能量的重要决定因素。EnCPdock (https://www.scinetmol.in/EnCPdock/) 提供了一个综合网络平台,用于直接联合比较分析互补性和 PPI 的结合能量。它利用互补图将 PPI 中的局部互补性(Sc)和非局部互补性(EC)的双重性质巧妙地联系在一起。它还进一步推导出了基于人工智能的 ΔG结合,其预测准确度可媲美目前的技术水平。本书的这一章介绍了一份实用手册,用于构思和实施 EnCPdock 及其各种特性和功能,这些特性和功能有可能成为设计新型蛋白质界面的重要蛋白质工程工具。
{"title":"Combining Complementarity and Binding Energetics in the Assessment of Protein Interactions: EnCPdock-A Practical Manual.","authors":"Gargi Biswas, Debasish Mukherjee, Sankar Basu","doi":"10.1089/cmb.2024.0554","DOIUrl":"https://doi.org/10.1089/cmb.2024.0554","url":null,"abstract":"<p><p>The combined effect of shape and electrostatic complementarities (Sc, EC) at the interface of the interacting protein partners (PPI) serves as the physical basis for such associations and is a strong determinant of their binding energetics. EnCPdock (https://www.scinetmol.in/EnCPdock/) presents a comprehensive web platform for the direct conjoint comparative analyses of complementarity and binding energetics in PPIs. It elegantly interlinks the dual nature of local (Sc) and nonlocal complementarity (EC) in PPIs using the complementarity plot. It further derives an AI-based ΔG<sub>binding</sub> with a prediction accuracy comparable to the <i>state of the art</i>. This book chapter presents a practical manual to conceptualize and implement EnCPdock with its various features and functionalities, collectively having the potential to serve as a valuable protein engineering tool in the design of novel protein interfaces.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2024-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141419324","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
QMix: An Efficient Program to Automatically Estimate Multi-Matrix Mixture Models for Amino Acid Substitution Process. QMix:自动估算氨基酸替代过程多矩阵混合物模型的高效程序
IF 1.7 4区 生物学 Q2 Mathematics Pub Date : 2024-06-11 DOI: 10.1089/cmb.2023.0403
Nguyen Huy Tinh, Cuong Cao Dang, Le Sy Vinh

The single-matrix amino acid (AA) substitution models are widely used in phylogenetic analyses; however, they are unable to properly model the heterogeneity of AA substitution rates among sites. The multi-matrix mixture models can handle the site rate heterogeneity and outperform the single-matrix models. Estimating multi-matrix mixture models is a complex process and no computer program is available for this task. In this study, we implemented a computer program of the so-called QMix based on the algorithm of LG4X and LG4M with several enhancements to automatically estimate multi-matrix mixture models from large datasets. QMix employs QMaker algorithm instead of XRATE algorithm to accurately and rapidly estimate the parameters of models. It is able to estimate mixture models with different number of matrices and supports multi-threading computing to efficiently estimate models from thousands of genes. We re-estimate mixture models LG4X and LG4M from 1471 HSSP alignments. The re-estimated models (HP4X and HP4M) are slightly better than LG4X and LG4M in building maximum likelihood trees from HSSP and TreeBASE datasets. QMix program required about 10 hours on a computer with 18 cores to estimate a mixture model with four matrices from 200 HSSP alignments. It is easy to use and freely available for researchers.

单矩阵氨基酸(AA)替换模型被广泛应用于系统发育分析中,但它们无法正确模拟不同位点间 AA 替换率的异质性。多矩阵混合模型可以处理位点率异质性,其效果优于单矩阵模型。估计多矩阵混合模型是一个复杂的过程,目前还没有计算机程序可以完成这项任务。在本研究中,我们在 LG4X 和 LG4M 算法的基础上进行了一些改进,实现了所谓的 QMix 计算机程序,可以从大型数据集中自动估计多矩阵混合模型。QMix 采用 QMaker 算法而不是 XRATE 算法来准确、快速地估计模型参数。它能估计不同矩阵数的混合模型,并支持多线程计算,能从数千个基因中高效地估计模型。我们从 1471 个 HSSP 对齐中重新估计了混合模型 LG4X 和 LG4M。在从 HSSP 和 TreeBASE 数据集构建最大似然树方面,重新估计的模型(HP4X 和 HP4M)略优于 LG4X 和 LG4M。QMix 程序需要在一台有 18 个内核的计算机上运行约 10 个小时,才能从 200 个 HSSP 数据集中估计出一个有四个矩阵的混合模型。它易于使用,可供研究人员免费使用。
{"title":"QMix: An Efficient Program to Automatically Estimate Multi-Matrix Mixture Models for Amino Acid Substitution Process.","authors":"Nguyen Huy Tinh, Cuong Cao Dang, Le Sy Vinh","doi":"10.1089/cmb.2023.0403","DOIUrl":"https://doi.org/10.1089/cmb.2023.0403","url":null,"abstract":"<p><p>The single-matrix amino acid (AA) substitution models are widely used in phylogenetic analyses; however, they are unable to properly model the heterogeneity of AA substitution rates among sites. The multi-matrix mixture models can handle the site rate heterogeneity and outperform the single-matrix models. Estimating multi-matrix mixture models is a complex process and no computer program is available for this task. In this study, we implemented a computer program of the so-called QMix based on the algorithm of LG4X and LG4M with several enhancements to automatically estimate multi-matrix mixture models from large datasets. QMix employs QMaker algorithm instead of XRATE algorithm to accurately and rapidly estimate the parameters of models. It is able to estimate mixture models with different number of matrices and supports multi-threading computing to efficiently estimate models from thousands of genes. We re-estimate mixture models LG4X and LG4M from 1471 HSSP alignments. The re-estimated models (HP4X and HP4M) are slightly better than LG4X and LG4M in building maximum likelihood trees from HSSP and TreeBASE datasets. QMix program required about 10 hours on a computer with 18 cores to estimate a mixture model with four matrices from 200 HSSP alignments. It is easy to use and freely available for researchers.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.7,"publicationDate":"2024-06-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141300786","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
Toward the Reconciliation of Inconsistent Molecular Structures from Biochemical Databases. 努力调和生化数据库中不一致的分子结构。
IF 1.4 4区 生物学 Q4 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-06-01 Epub Date: 2024-05-17 DOI: 10.1089/cmb.2024.0520
Casper Asbjørn Eriksen, Jakob Lykke Andersen, Rolf Fagerberg, Daniel Merkle

Information on the structure of molecules, retrieved via biochemical databases, plays a pivotal role in various disciplines, including metabolomics, systems biology, and drug discovery. No such database can be complete and it is often necessary to incorporate data from several sources. However, the molecular structure for a given compound is not necessarily consistent between databases. This article presents StructRecon, a novel tool for resolving unique molecular structures from database identifiers. Currently, identifiers from BiGG, ChEBI, Escherichia coli Metabolome Database (ECMDB), MetaNetX, and PubChem are supported. StructRecon traverses the cross-links between entries in different databases to construct what we call identifier graphs. The goal of these graphs is to offer a more complete view of the total information available on a given compound across all the supported databases. To reconcile discrepancies met during the traversal of the databases, we develop an extensible model for molecular structure supporting multiple independent levels of detail, which allows standardization of the structure to be applied iteratively. In some cases, our standardization approach results in multiple candidate structures for a given compound, in which case a random walk-based algorithm is used to select the most likely structure among incompatible alternatives. As a case study, we applied StructRecon to the EColiCore2 model. We found at least one structure for 98.66% of its compounds, which is more than twice as many as possible when using the databases in more standard ways not considering the complex network of cross-database references captured by our identifier graphs. StructRecon is open-source and modular, which enables support for more databases in the future.

通过生化数据库获取的分子结构信息在代谢组学、系统生物学和药物发现等多个学科中发挥着举足轻重的作用。任何此类数据库都不可能是完整的,通常需要结合多个来源的数据。然而,不同数据库中给定化合物的分子结构并不一定一致。本文介绍的 StructRecon 是一种从数据库标识符解析独特分子结构的新型工具。目前,该工具支持来自 BiGG、ChEBI、大肠杆菌代谢组数据库(ECMDB)、MetaNetX 和 PubChem 的标识符。StructRecon 会遍历不同数据库中条目之间的交叉链接,以构建我们所说的标识符图。这些图谱的目的是提供一个更完整的视图,显示特定化合物在所有支持数据库中可用的全部信息。为了调和在遍历数据库过程中遇到的差异,我们开发了一个可扩展的分子结构模型,支持多个独立的细节级别,从而可以反复应用结构标准化。在某些情况下,我们的标准化方法会为给定化合物生成多个候选结构,在这种情况下,我们会使用一种基于随机漫步的算法,从不相容的备选结构中选择最有可能的结构。作为案例研究,我们将 StructRecon 应用于 EColiCore2 模型。我们为其中 98.66% 的化合物找到了至少一种结构,这比以更标准的方式使用数据库而不考虑我们的标识符图捕捉到的复杂的跨数据库引用网络所能找到的结构数量高出一倍多。StructRecon 是开源和模块化的,因此未来可以支持更多数据库。
{"title":"Toward the Reconciliation of Inconsistent Molecular Structures from Biochemical Databases.","authors":"Casper Asbjørn Eriksen, Jakob Lykke Andersen, Rolf Fagerberg, Daniel Merkle","doi":"10.1089/cmb.2024.0520","DOIUrl":"10.1089/cmb.2024.0520","url":null,"abstract":"<p><p><b>Information on the structure of molecules, retrieved via biochemical databases, plays a pivotal role in various disciplines, including metabolomics, systems biology, and drug discovery. No such database can be complete and it is often necessary to incorporate data from several sources. However, the molecular structure for a given compound is not necessarily consistent between databases. This article presents StructRecon, a novel tool for resolving unique molecular structures from database identifiers. Currently, identifiers from BiGG, ChEBI,</b> <i>Escherichia coli</i> Metabolome Database <b>(ECMDB), MetaNetX, and PubChem are supported. StructRecon traverses the cross-links between entries in different databases to construct what we call identifier graphs. The goal of these graphs is to offer a more complete view of the total information available on a given compound across all the supported databases. To reconcile discrepancies met during the traversal of the databases, we develop an extensible model for molecular structure supporting multiple independent levels of detail, which allows standardization of the structure to be applied iteratively. In some cases, our standardization approach results in multiple candidate structures for a given compound, in which case a random walk-based algorithm is used to select the most likely structure among incompatible alternatives. As a case study, we applied StructRecon to the <i>EColiCore2</i> model. We found at least one structure for 98.66% of its compounds, which is more than twice as many as possible when using the databases in more standard ways not considering the complex network of cross-database references captured by our identifier graphs. StructRecon is open-source and modular, which enables support for more databases in the future.</b></p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.4,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140957701","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
Transcriptional Hubs Within Cliques in Ensemble Hi-C Chromatin Interaction Networks. Hi-C 染色质相互作用网络中小群内的转录枢纽
IF 1.4 4区 生物学 Q4 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-06-01 Epub Date: 2024-05-20 DOI: 10.1089/cmb.2024.0515
Gatis Melkus, Andrejs Sizovs, Peteris Rucevskis, Sandra Silina

Chromatin conformation capture technologies permit the study of chromatin spatial organization on a genome-wide scale at a variety of resolutions. Despite the increasing precision and resolution of high-throughput chromatin conformation capture (Hi-C) methods, it remains challenging to conclusively link transcriptional activity to spatial organizational phenomena. We have developed a clique-based approach for analyzing Hi-C data that helps identify chromosomal hotspots that feature considerable enrichment of chromatin annotations for transcriptional start sites and, building on previously published work, show that these chromosomal hotspots are not only significantly enriched in RNA polymerase II binding sites as identified by the ENCODE project, but also identify a noticeable increase in FANTOM5 and GTEx transcription within our identified cliques across a variety of tissue types. From the obtained data, we surmise that our cliques are a suitable method for identifying transcription factories in Hi-C data, and outline further extensions to the method that may make it useful for locating regions of increased transcriptional activity in datasets where in-depth expression or polymerase data may not be available.

染色质构象捕获技术允许以各种分辨率研究全基因组范围的染色质空间组织。尽管高通量染色质构象捕获(Hi-C)方法的精确度和分辨率不断提高,但要将转录活动与空间组织现象确凿地联系起来仍具有挑战性。我们开发了一种基于集群的方法来分析 Hi-C 数据,这种方法有助于识别染色体热点,这些热点的特点是染色质注释对转录起始位点有相当大的富集作用,并且在以前发表的工作基础上,我们发现这些染色体热点不仅在 ENCODE 项目确定的 RNA 聚合酶 II 结合位点中有显著富集,而且在我们确定的集群内,各种组织类型的 FANTOM5 和 GTEx 转录也有明显增加。根据所获得的数据,我们推测我们的集群是在 Hi-C 数据中识别转录工厂的一种合适方法,并概述了该方法的进一步扩展,这可能会使它在无法获得深度表达或聚合酶数据的数据集中用于定位转录活动增加的区域。
{"title":"Transcriptional Hubs Within Cliques in Ensemble Hi-C Chromatin Interaction Networks.","authors":"Gatis Melkus, Andrejs Sizovs, Peteris Rucevskis, Sandra Silina","doi":"10.1089/cmb.2024.0515","DOIUrl":"10.1089/cmb.2024.0515","url":null,"abstract":"<p><p>\u0000 <b>Chromatin conformation capture technologies permit the study of chromatin spatial organization on a genome-wide scale at a variety of resolutions. Despite the increasing precision and resolution of high-throughput chromatin conformation capture (Hi-C) methods, it remains challenging to conclusively link transcriptional activity to spatial organizational phenomena. We have developed a clique-based approach for analyzing Hi-C data that helps identify chromosomal hotspots that feature considerable enrichment of chromatin annotations for transcriptional start sites and, building on previously published work, show that these chromosomal hotspots are not only significantly enriched in RNA polymerase II binding sites as identified by the ENCODE project, but also identify a noticeable increase in FANTOM5 and GTEx transcription within our identified cliques across a variety of tissue types. From the obtained data, we surmise that our cliques are a suitable method for identifying transcription factories in Hi-C data, and outline further extensions to the method that may make it useful for locating regions of increased transcriptional activity in datasets where in-depth expression or polymerase data may not be available.</b>\u0000 </p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.4,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141071020","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
Enhanced Compression of k-Mer Sets with Counters via de Bruijn Graphs. 通过 de Bruijn 图增强带有计数器的 k-Mer 集的压缩。
IF 1.4 4区 生物学 Q4 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-06-01 Epub Date: 2024-05-31 DOI: 10.1089/cmb.2024.0530
Enrico Rossignolo, Matteo Comin

An essential task in computational genomics involves transforming input sequences into their constituent k-mers. The quest for an efficient representation of k-mer sets is crucial for enhancing the scalability of bioinformatic analyses. One widely used method involves converting the k-mer set into a de Bruijn graph (dBG), followed by seeking a compact graph representation via the smallest path cover. This study introduces USTAR* (Unitig STitch Advanced constRuction), a tool designed to compress both a set of k-mers and their associated counts. USTAR leverages the connectivity and density of dBGs, enabling a more efficient path selection for constructing the path cover. The efficacy of USTAR is demonstrated through its application in compressing real read data sets. USTAR improves the compression achieved by UST (Unitig STitch), the best algorithm, by percentages ranging from 2.3% to 26.4%, depending on the k-mer size, and it is up to 7× times faster.

计算基因组学的一项基本任务是将输入序列转化为其组成的 k-聚合物。要提高生物信息分析的可扩展性,就必须寻求一种高效的 k-mer 集表示方法。一种广泛使用的方法是将 k-mer 集转换成 de Bruijn 图(dBG),然后通过最小路径覆盖寻求紧凑的图表示。本研究介绍了 USTAR*(Unitig STitch Advanced constRuction),这是一种旨在压缩 k-聚合物集及其相关计数的工具。USTAR 利用 dBGs 的连接性和密度,为构建路径覆盖提供了更有效的路径选择。USTAR 在压缩真实读取数据集中的应用证明了它的功效。USTAR 比最佳算法 UST(Unitig STitch)的压缩率提高了 2.3% 到 26.4%,具体取决于 k-mer 的大小,而且速度快达 7 倍。
{"title":"Enhanced Compression of <i>k</i>-Mer Sets with Counters via de Bruijn Graphs.","authors":"Enrico Rossignolo, Matteo Comin","doi":"10.1089/cmb.2024.0530","DOIUrl":"10.1089/cmb.2024.0530","url":null,"abstract":"<p><p>An essential task in computational genomics involves transforming input sequences into their constituent <i>k</i>-mers. The quest for an efficient representation of <i>k</i>-mer sets is crucial for enhancing the scalability of bioinformatic analyses. One widely used method involves converting the <i>k</i>-mer set into a de Bruijn graph (dBG), followed by seeking a compact graph representation via the smallest path cover. This study introduces USTAR* (Unitig STitch Advanced constRuction), a tool designed to compress both a set of <i>k</i>-mers and their associated counts. USTAR leverages the connectivity and density of dBGs, enabling a more efficient path selection for constructing the path cover. The efficacy of USTAR is demonstrated through its application in compressing real read data sets. USTAR improves the compression achieved by UST (Unitig STitch), the best algorithm, by percentages ranging from 2.3% to 26.4%, depending on the <i>k</i>-mer size, and it is up to <math><mn>7</mn><mo>×</mo></math> times faster.</p>","PeriodicalId":15526,"journal":{"name":"Journal of Computational Biology","volume":null,"pages":null},"PeriodicalIF":1.4,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141183858","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