首页 > 最新文献

Briefings in Functional Genomics最新文献

英文 中文
DBPMod: a supervised learning model for computational recognition of DNA-binding proteins in model organisms. DBPMod:用于计算识别模式生物中 DNA 结合蛋白的监督学习模型。
IF 2.5 3区 生物学 Q3 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elad039
Upendra K Pradhan, Prabina K Meher, Sanchita Naha, Nitesh K Sharma, Aarushi Agarwal, Ajit Gupta, Rajender Parsad

DNA-binding proteins (DBPs) play critical roles in many biological processes, including gene expression, DNA replication, recombination and repair. Understanding the molecular mechanisms underlying these processes depends on the precise identification of DBPs. In recent times, several computational methods have been developed to identify DBPs. However, because of the generic nature of the models, these models are unable to identify species-specific DBPs with higher accuracy. Therefore, a species-specific computational model is needed to predict species-specific DBPs. In this paper, we introduce the computational DBPMod method, which makes use of a machine learning approach to identify species-specific DBPs. For prediction, both shallow learning algorithms and deep learning models were used, with shallow learning models achieving higher accuracy. Additionally, the evolutionary features outperformed sequence-derived features in terms of accuracy. Five model organisms, including Caenorhabditis elegans, Drosophila melanogaster, Escherichia coli, Homo sapiens and Mus musculus, were used to assess the performance of DBPMod. Five-fold cross-validation and independent test set analyses were used to evaluate the prediction accuracy in terms of area under receiver operating characteristic curve (auROC) and area under precision-recall curve (auPRC), which was found to be ~89-92% and ~89-95%, respectively. The comparative results demonstrate that the DBPMod outperforms 12 current state-of-the-art computational approaches in identifying the DBPs for all five model organisms. We further developed the web server of DBPMod to make it easier for researchers to detect DBPs and is publicly available at https://iasri-sg.icar.gov.in/dbpmod/. DBPMod is expected to be an invaluable tool for discovering DBPs, supplementing the current experimental and computational methods.

DNA 结合蛋白(DBPs)在基因表达、DNA 复制、重组和修复等许多生物过程中发挥着关键作用。对这些过程的分子机制的了解取决于对 DBPs 的精确鉴定。近来,人们开发了多种计算方法来识别 DBPs。然而,由于模型的通用性,这些模型无法更准确地识别物种特异性 DBPs。因此,需要一种针对特定物种的计算模型来预测特定物种的 DBPs。本文介绍了计算 DBPMod 方法,该方法利用机器学习方法来识别物种特异性 DBPs。在预测方面,我们使用了浅层学习算法和深度学习模型,其中浅层学习模型的准确率更高。此外,在准确性方面,进化特征优于序列衍生特征。五种模式生物,包括秀丽隐杆线虫、黑腹果蝇、大肠杆菌、智人和麝,被用来评估DBPMod的性能。通过五倍交叉验证和独立测试集分析,以接收者操作特征曲线下面积(auROC)和精确度-召回曲线下面积(auPRC)来评估预测精确度,结果发现预测精确度分别为~89-92%和~89-95%。比较结果表明,DBPMod 在识别所有五种模式生物的 DBPs 方面优于目前最先进的 12 种计算方法。我们进一步开发了 DBPMod 的网络服务器,使研究人员更容易检测 DBPs,该服务器已在 https://iasri-sg.icar.gov.in/dbpmod/ 上公开发布。DBPMod 预计将成为发现 DBPs 的宝贵工具,补充当前的实验和计算方法。
{"title":"DBPMod: a supervised learning model for computational recognition of DNA-binding proteins in model organisms.","authors":"Upendra K Pradhan, Prabina K Meher, Sanchita Naha, Nitesh K Sharma, Aarushi Agarwal, Ajit Gupta, Rajender Parsad","doi":"10.1093/bfgp/elad039","DOIUrl":"10.1093/bfgp/elad039","url":null,"abstract":"<p><p>DNA-binding proteins (DBPs) play critical roles in many biological processes, including gene expression, DNA replication, recombination and repair. Understanding the molecular mechanisms underlying these processes depends on the precise identification of DBPs. In recent times, several computational methods have been developed to identify DBPs. However, because of the generic nature of the models, these models are unable to identify species-specific DBPs with higher accuracy. Therefore, a species-specific computational model is needed to predict species-specific DBPs. In this paper, we introduce the computational DBPMod method, which makes use of a machine learning approach to identify species-specific DBPs. For prediction, both shallow learning algorithms and deep learning models were used, with shallow learning models achieving higher accuracy. Additionally, the evolutionary features outperformed sequence-derived features in terms of accuracy. Five model organisms, including Caenorhabditis elegans, Drosophila melanogaster, Escherichia coli, Homo sapiens and Mus musculus, were used to assess the performance of DBPMod. Five-fold cross-validation and independent test set analyses were used to evaluate the prediction accuracy in terms of area under receiver operating characteristic curve (auROC) and area under precision-recall curve (auPRC), which was found to be ~89-92% and ~89-95%, respectively. The comparative results demonstrate that the DBPMod outperforms 12 current state-of-the-art computational approaches in identifying the DBPs for all five model organisms. We further developed the web server of DBPMod to make it easier for researchers to detect DBPs and is publicly available at https://iasri-sg.icar.gov.in/dbpmod/. DBPMod is expected to be an invaluable tool for discovering DBPs, supplementing the current experimental and computational methods.</p>","PeriodicalId":55323,"journal":{"name":"Briefings in Functional Genomics","volume":" ","pages":"363-372"},"PeriodicalIF":2.5,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10483304","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Improving cell type identification with Gaussian noise-augmented single-cell RNA-seq contrastive learning. 利用高斯噪声增强单细胞 RNA-seq 对比学习改进细胞类型识别。
IF 2.5 3区 生物学 Q3 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elad059
Ibrahim Alsaggaf, Daniel Buchan, Cen Wan

Cell type identification is an important task for single-cell RNA-sequencing (scRNA-seq) data analysis. Many prediction methods have recently been proposed, but the predictive accuracy of difficult cell type identification tasks is still low. In this work, we proposed a novel Gaussian noise augmentation-based scRNA-seq contrastive learning method (GsRCL) to learn a type of discriminative feature representations for cell type identification tasks. A large-scale computational evaluation suggests that GsRCL successfully outperformed other state-of-the-art predictive methods on difficult cell type identification tasks, while the conventional random genes masking augmentation-based contrastive learning method also improved the accuracy of easy cell type identification tasks in general.

细胞类型鉴定是单细胞 RNA 序列(scRNA-seq)数据分析的一项重要任务。最近提出了许多预测方法,但对困难的细胞类型鉴定任务的预测准确率仍然很低。在这项工作中,我们提出了一种新颖的基于高斯噪声增强的 scRNA-seq 对比学习方法(GsRCL),以学习一种用于细胞类型鉴定任务的判别特征表征。大规模计算评估表明,GsRCL 在高难度细胞类型鉴定任务中的表现成功地超越了其他最先进的预测方法,而传统的基于随机基因掩蔽增强的对比学习方法也普遍提高了简单细胞类型鉴定任务的准确率。
{"title":"Improving cell type identification with Gaussian noise-augmented single-cell RNA-seq contrastive learning.","authors":"Ibrahim Alsaggaf, Daniel Buchan, Cen Wan","doi":"10.1093/bfgp/elad059","DOIUrl":"10.1093/bfgp/elad059","url":null,"abstract":"<p><p>Cell type identification is an important task for single-cell RNA-sequencing (scRNA-seq) data analysis. Many prediction methods have recently been proposed, but the predictive accuracy of difficult cell type identification tasks is still low. In this work, we proposed a novel Gaussian noise augmentation-based scRNA-seq contrastive learning method (GsRCL) to learn a type of discriminative feature representations for cell type identification tasks. A large-scale computational evaluation suggests that GsRCL successfully outperformed other state-of-the-art predictive methods on difficult cell type identification tasks, while the conventional random genes masking augmentation-based contrastive learning method also improved the accuracy of easy cell type identification tasks in general.</p>","PeriodicalId":55323,"journal":{"name":"Briefings in Functional Genomics","volume":" ","pages":"441-451"},"PeriodicalIF":2.5,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139503121","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Genomics in Clinical trials for Breast Cancer. 乳腺癌临床试验中的基因组学。
IF 2.5 3区 生物学 Q3 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elad054
David Enoma

Breast cancer (B.C.) still has increasing incidences and mortality rates globally. It is known that B.C. and other cancers have a very high rate of genetic heterogeneity and genomic mutations. Traditional oncology approaches have not been able to provide a lasting solution. Targeted therapeutics have been instrumental in handling the complexity and resistance associated with B.C. However, the progress of genomic technology has transformed our understanding of the genetic landscape of breast cancer, opening new avenues for improved anti-cancer therapeutics. Genomics is critical in developing tailored therapeutics and identifying patients most benefit from these treatments. The next generation of breast cancer clinical trials has incorporated next-generation sequencing technologies into the process, and we have seen benefits. These innovations have led to the approval of better-targeted therapies for patients with breast cancer. Genomics has a role to play in clinical trials, including genomic tests that have been approved, patient selection and prediction of therapeutic response. Multiple clinical trials in breast cancer have been done and are still ongoing, which have applied genomics technology. Precision medicine can be achieved in breast cancer therapy with increased efforts and advanced genomic studies in this domain. Genomics studies assist with patient outcomes improvement and oncology advancement by providing a deeper understanding of the biology behind breast cancer. This article will examine the present state of genomics in breast cancer clinical trials.

在全球范围内,乳腺癌(B.C.)的发病率和死亡率仍在不断上升。众所周知,乳腺癌和其他癌症具有很高的遗传异质性和基因组突变率。传统的肿瘤学方法无法提供持久的解决方案。然而,基因组学技术的进步改变了我们对乳腺癌基因状况的认识,为改进抗癌疗法开辟了新途径。基因组学对于开发有针对性的疗法和确定最受益于这些疗法的患者至关重要。下一代乳腺癌临床试验已将新一代测序技术纳入其中,我们已从中获益。这些创新已使针对乳腺癌患者的更佳疗法获得批准。基因组学可在临床试验中发挥作用,包括已获批准的基因组检测、患者选择和治疗反应预测。应用基因组学技术的多项乳腺癌临床试验已经完成并仍在进行中。随着在乳腺癌治疗领域加大努力和开展先进的基因组研究,可以实现精准医疗。基因组学研究通过深入了解乳腺癌背后的生物学原理,有助于改善患者的治疗效果,推动肿瘤学的发展。本文将探讨基因组学在乳腺癌临床试验中的应用现状。
{"title":"Genomics in Clinical trials for Breast Cancer.","authors":"David Enoma","doi":"10.1093/bfgp/elad054","DOIUrl":"10.1093/bfgp/elad054","url":null,"abstract":"<p><p>Breast cancer (B.C.) still has increasing incidences and mortality rates globally. It is known that B.C. and other cancers have a very high rate of genetic heterogeneity and genomic mutations. Traditional oncology approaches have not been able to provide a lasting solution. Targeted therapeutics have been instrumental in handling the complexity and resistance associated with B.C. However, the progress of genomic technology has transformed our understanding of the genetic landscape of breast cancer, opening new avenues for improved anti-cancer therapeutics. Genomics is critical in developing tailored therapeutics and identifying patients most benefit from these treatments. The next generation of breast cancer clinical trials has incorporated next-generation sequencing technologies into the process, and we have seen benefits. These innovations have led to the approval of better-targeted therapies for patients with breast cancer. Genomics has a role to play in clinical trials, including genomic tests that have been approved, patient selection and prediction of therapeutic response. Multiple clinical trials in breast cancer have been done and are still ongoing, which have applied genomics technology. Precision medicine can be achieved in breast cancer therapy with increased efforts and advanced genomic studies in this domain. Genomics studies assist with patient outcomes improvement and oncology advancement by providing a deeper understanding of the biology behind breast cancer. This article will examine the present state of genomics in breast cancer clinical trials.</p>","PeriodicalId":55323,"journal":{"name":"Briefings in Functional Genomics","volume":" ","pages":"325-334"},"PeriodicalIF":2.5,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139038236","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
DeepPRMS: advanced deep learning model to predict protein arginine methylation sites. DeepPRMS:预测蛋白质精氨酸甲基化位点的高级深度学习模型。
IF 2.5 3区 生物学 Q3 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elae001
Monika Khandelwal, Ranjeet Kumar Rout

Protein methylation is a form of post-translational modifications of protein, which is crucial for various cellular processes, including transcription activity and DNA repair. Correctly predicting protein methylation sites is fundamental for research and drug discovery. Some experimental techniques, such as methyl-specific antibodies, chromatin immune precipitation and mass spectrometry, exist for predicting protein methylation sites, but these techniques are time-consuming and costly. The ability to predict methylation sites using in silico techniques may help researchers identify potential candidate sites for future examination and make it easier to carry out site-specific investigations and downstream characterizations. In this research, we proposed a novel deep learning-based predictor, named DeepPRMS, to identify protein methylation sites in primary sequences. The DeepPRMS utilizes the gated recurrent unit (GRU) and convolutional neural network (CNN) algorithms to extract the sequential and spatial information from the primary sequences. GRU is used to extract sequential information, while CNN is used for spatial information. We combined the latent representation of GRU and CNN models to have a better interaction among them. Based on the independent test data set, DeepPRMS obtained an accuracy of 85.32%, a specificity of 84.94%, Matthew's correlation coefficient of 0.71 and a sensitivity of 85.80%. The results indicate that DeepPRMS can predict protein methylation sites with high accuracy and outperform the state-of-the-art models. The DeepPRMS is expected to effectively guide future research experiments for identifying potential methylated protein sites. The web server is available at http://deepprms.nitsri.ac.in/.

蛋白质甲基化是蛋白质翻译后修饰的一种形式,对包括转录活动和 DNA 修复在内的各种细胞过程至关重要。正确预测蛋白质甲基化位点是研究和药物发现的基础。目前已有一些实验技术,如甲基特异性抗体、染色质免疫沉淀和质谱技术,可用于预测蛋白质甲基化位点,但这些技术耗时长、成本高。利用硅学技术预测甲基化位点的能力可帮助研究人员确定潜在的候选位点,以便今后进行研究,并使位点特异性研究和下游表征更容易进行。在这项研究中,我们提出了一种基于深度学习的新型预测器,名为 DeepPRMS,用于识别原始序列中的蛋白质甲基化位点。DeepPRMS 利用门控递归单元(GRU)和卷积神经网络(CNN)算法从原始序列中提取序列和空间信息。GRU 用于提取序列信息,而 CNN 则用于提取空间信息。我们将 GRU 模型和 CNN 模型的潜表征结合起来,使它们之间有更好的互动。基于独立测试数据集,DeepPRMS 的准确率为 85.32%,特异性为 84.94%,马修相关系数为 0.71,灵敏度为 85.80%。这些结果表明,DeepPRMS 可以高精度地预测蛋白质甲基化位点,其结果优于最先进的模型。DeepPRMS有望有效指导未来的研究实验,识别潜在的甲基化蛋白质位点。网络服务器的网址是 http://deepprms.nitsri.ac.in/。
{"title":"DeepPRMS: advanced deep learning model to predict protein arginine methylation sites.","authors":"Monika Khandelwal, Ranjeet Kumar Rout","doi":"10.1093/bfgp/elae001","DOIUrl":"10.1093/bfgp/elae001","url":null,"abstract":"<p><p>Protein methylation is a form of post-translational modifications of protein, which is crucial for various cellular processes, including transcription activity and DNA repair. Correctly predicting protein methylation sites is fundamental for research and drug discovery. Some experimental techniques, such as methyl-specific antibodies, chromatin immune precipitation and mass spectrometry, exist for predicting protein methylation sites, but these techniques are time-consuming and costly. The ability to predict methylation sites using in silico techniques may help researchers identify potential candidate sites for future examination and make it easier to carry out site-specific investigations and downstream characterizations. In this research, we proposed a novel deep learning-based predictor, named DeepPRMS, to identify protein methylation sites in primary sequences. The DeepPRMS utilizes the gated recurrent unit (GRU) and convolutional neural network (CNN) algorithms to extract the sequential and spatial information from the primary sequences. GRU is used to extract sequential information, while CNN is used for spatial information. We combined the latent representation of GRU and CNN models to have a better interaction among them. Based on the independent test data set, DeepPRMS obtained an accuracy of 85.32%, a specificity of 84.94%, Matthew's correlation coefficient of 0.71 and a sensitivity of 85.80%. The results indicate that DeepPRMS can predict protein methylation sites with high accuracy and outperform the state-of-the-art models. The DeepPRMS is expected to effectively guide future research experiments for identifying potential methylated protein sites. The web server is available at http://deepprms.nitsri.ac.in/.</p>","PeriodicalId":55323,"journal":{"name":"Briefings in Functional Genomics","volume":" ","pages":"452-463"},"PeriodicalIF":2.5,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139547623","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
GAM-MDR: probing miRNA-drug resistance using a graph autoencoder based on random path masking. GAM-MDR:使用基于随机路径屏蔽的图自动编码器探测 miRNA-耐药性。
IF 2.5 3区 生物学 Q3 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elae005
Zhecheng Zhou, Zhenya Du, Xin Jiang, Linlin Zhuo, Yixin Xu, Xiangzheng Fu, Mingzhe Liu, Quan Zou

MicroRNAs (miRNAs) are found ubiquitously in biological cells and play a pivotal role in regulating the expression of numerous target genes. Therapies centered around miRNAs are emerging as a promising strategy for disease treatment, aiming to intervene in disease progression by modulating abnormal miRNA expressions. The accurate prediction of miRNA-drug resistance (MDR) is crucial for the success of miRNA therapies. Computational models based on deep learning have demonstrated exceptional performance in predicting potential MDRs. However, their effectiveness can be compromised by errors in the data acquisition process, leading to inaccurate node representations. To address this challenge, we introduce the GAM-MDR model, which combines the graph autoencoder (GAE) with random path masking techniques to precisely predict potential MDRs. The reliability and effectiveness of the GAM-MDR model are mainly reflected in two aspects. Firstly, it efficiently extracts the representations of miRNA and drug nodes in the miRNA-drug network. Secondly, our designed random path masking strategy efficiently reconstructs critical paths in the network, thereby reducing the adverse impact of noisy data. To our knowledge, this is the first time that a random path masking strategy has been integrated into a GAE to infer MDRs. Our method was subjected to multiple validations on public datasets and yielded promising results. We are optimistic that our model could offer valuable insights for miRNA therapeutic strategies and deepen the understanding of the regulatory mechanisms of miRNAs. Our data and code are publicly available at GitHub:https://github.com/ZZCrazy00/GAM-MDR.

微小核糖核酸(miRNA)遍布于生物细胞中,在调节众多靶基因的表达方面发挥着关键作用。以 miRNA 为中心的疗法正在成为一种前景广阔的疾病治疗策略,旨在通过调节异常的 miRNA 表达来干预疾病的进展。准确预测 miRNA 耐药性(MDR)对于 miRNA 疗法的成功至关重要。基于深度学习的计算模型在预测潜在 MDR 方面表现出色。然而,数据采集过程中的错误可能会影响其有效性,导致节点表示不准确。为了应对这一挑战,我们引入了 GAM-MDR 模型,该模型将图自动编码器(GAE)与随机路径掩蔽技术相结合,以精确预测潜在的 MDR。GAM-MDR 模型的可靠性和有效性主要体现在两个方面。首先,它能有效提取 miRNA 药物网络中 miRNA 和药物节点的表征。其次,我们设计的随机路径掩蔽策略能有效重建网络中的关键路径,从而降低了噪声数据的不利影响。据我们所知,这是首次将随机路径屏蔽策略集成到 GAE 中来推断 MDR。我们的方法在公共数据集上进行了多次验证,并取得了令人满意的结果。我们相信,我们的模型能为 miRNA 治疗策略提供有价值的见解,并加深对 miRNA 调控机制的理解。我们的数据和代码可在 GitHub:https://github.com/ZZCrazy00/GAM-MDR 上公开获取。
{"title":"GAM-MDR: probing miRNA-drug resistance using a graph autoencoder based on random path masking.","authors":"Zhecheng Zhou, Zhenya Du, Xin Jiang, Linlin Zhuo, Yixin Xu, Xiangzheng Fu, Mingzhe Liu, Quan Zou","doi":"10.1093/bfgp/elae005","DOIUrl":"10.1093/bfgp/elae005","url":null,"abstract":"<p><p>MicroRNAs (miRNAs) are found ubiquitously in biological cells and play a pivotal role in regulating the expression of numerous target genes. Therapies centered around miRNAs are emerging as a promising strategy for disease treatment, aiming to intervene in disease progression by modulating abnormal miRNA expressions. The accurate prediction of miRNA-drug resistance (MDR) is crucial for the success of miRNA therapies. Computational models based on deep learning have demonstrated exceptional performance in predicting potential MDRs. However, their effectiveness can be compromised by errors in the data acquisition process, leading to inaccurate node representations. To address this challenge, we introduce the GAM-MDR model, which combines the graph autoencoder (GAE) with random path masking techniques to precisely predict potential MDRs. The reliability and effectiveness of the GAM-MDR model are mainly reflected in two aspects. Firstly, it efficiently extracts the representations of miRNA and drug nodes in the miRNA-drug network. Secondly, our designed random path masking strategy efficiently reconstructs critical paths in the network, thereby reducing the adverse impact of noisy data. To our knowledge, this is the first time that a random path masking strategy has been integrated into a GAE to infer MDRs. Our method was subjected to multiple validations on public datasets and yielded promising results. We are optimistic that our model could offer valuable insights for miRNA therapeutic strategies and deepen the understanding of the regulatory mechanisms of miRNAs. Our data and code are publicly available at GitHub:https://github.com/ZZCrazy00/GAM-MDR.</p>","PeriodicalId":55323,"journal":{"name":"Briefings in Functional Genomics","volume":" ","pages":"475-483"},"PeriodicalIF":2.5,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139934372","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
ASACO: Automatic and Serial Analysis of CO-expression to discover gene modifiers with potential use in drug repurposing. ASACO:通过对 CO 表达进行自动和序列分析,发现可能用于药物再利用的基因修饰因子。
IF 2.5 3区 生物学 Q3 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elae006
Cristina Moral-Turón, Gualberto Asencio-Cortés, Francesc Rodriguez-Diaz, Alejandro Rubio, Alberto G Navarro, Ana M Brokate-Llanos, Andrés Garzón, Manuel J Muñoz, Antonio J Pérez-Pulido

Massive gene expression analyses are widely used to find differentially expressed genes under specific conditions. The results of these experiments are often available in public databases that are undergoing a growth similar to that of molecular sequence databases in the past. This now allows novel secondary computational tools to emerge that use such information to gain new knowledge. If several genes have a similar expression profile across heterogeneous transcriptomics experiments, they could be functionally related. These associations are usually useful for the annotation of uncharacterized genes. In addition, the search for genes with opposite expression profiles is useful for finding negative regulators and proposing inhibitory compounds in drug repurposing projects. Here we present a new web application, Automatic and Serial Analysis of CO-expression (ASACO), which has the potential to discover positive and negative correlator genes to a given query gene, based on thousands of public transcriptomics experiments. In addition, examples of use are presented, comparing with previous contrasted knowledge. The results obtained propose ASACO as a useful tool to improve knowledge about genes associated with human diseases and noncoding genes. ASACO is available at http://www.bioinfocabd.upo.es/asaco/.

大规模基因表达分析被广泛用于寻找特定条件下的差异表达基因。这些实验的结果通常可以在公共数据库中找到,而公共数据库的发展与过去分子序列数据库的发展类似。这使得利用这些信息获取新知识的新型二次计算工具应运而生。如果多个基因在不同的转录组学实验中具有相似的表达谱,那么它们在功能上可能是相关的。这些关联通常有助于对未定性基因进行注释。此外,在药物再利用项目中,搜索具有相反表达谱的基因对于寻找负调控因子和提出抑制性化合物也很有用。在此,我们介绍一种新的网络应用程序--CO-expression 自动和序列分析(ASACO),它可以根据成千上万的公开转录组学实验,发现与给定查询基因正相关和负相关的基因。此外,还介绍了使用实例,并与之前的知识进行了对比。研究结果表明,ASACO 是提高人类疾病相关基因和非编码基因知识的有用工具。ASACO 可在 http://www.bioinfocabd.upo.es/asaco/ 上查阅。
{"title":"ASACO: Automatic and Serial Analysis of CO-expression to discover gene modifiers with potential use in drug repurposing.","authors":"Cristina Moral-Turón, Gualberto Asencio-Cortés, Francesc Rodriguez-Diaz, Alejandro Rubio, Alberto G Navarro, Ana M Brokate-Llanos, Andrés Garzón, Manuel J Muñoz, Antonio J Pérez-Pulido","doi":"10.1093/bfgp/elae006","DOIUrl":"10.1093/bfgp/elae006","url":null,"abstract":"<p><p>Massive gene expression analyses are widely used to find differentially expressed genes under specific conditions. The results of these experiments are often available in public databases that are undergoing a growth similar to that of molecular sequence databases in the past. This now allows novel secondary computational tools to emerge that use such information to gain new knowledge. If several genes have a similar expression profile across heterogeneous transcriptomics experiments, they could be functionally related. These associations are usually useful for the annotation of uncharacterized genes. In addition, the search for genes with opposite expression profiles is useful for finding negative regulators and proposing inhibitory compounds in drug repurposing projects. Here we present a new web application, Automatic and Serial Analysis of CO-expression (ASACO), which has the potential to discover positive and negative correlator genes to a given query gene, based on thousands of public transcriptomics experiments. In addition, examples of use are presented, comparing with previous contrasted knowledge. The results obtained propose ASACO as a useful tool to improve knowledge about genes associated with human diseases and noncoding genes. ASACO is available at http://www.bioinfocabd.upo.es/asaco/.</p>","PeriodicalId":55323,"journal":{"name":"Briefings in Functional Genomics","volume":" ","pages":"484-494"},"PeriodicalIF":2.5,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139998292","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Integration tools for scRNA-seq data and spatial transcriptomics sequencing data. scRNA-seq 数据和空间转录组学测序数据的整合工具。
IF 2.5 3区 生物学 Q3 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elae002
Chaorui Yan, Yanxu Zhu, Miao Chen, Kainan Yang, Feifei Cui, Quan Zou, Zilong Zhang

Numerous methods have been developed to integrate spatial transcriptomics sequencing data with single-cell RNA sequencing (scRNA-seq) data. Continuous development and improvement of these methods offer multiple options for integrating and analyzing scRNA-seq and spatial transcriptomics data based on diverse research inquiries. However, each method has its own advantages, limitations and scope of application. Researchers need to select the most suitable method for their research purposes based on the actual situation. This review article presents a compilation of 19 integration methods sourced from a wide range of available approaches, serving as a comprehensive reference for researchers to select the suitable integration method for their specific research inquiries. By understanding the principles of these methods, we can identify their similarities and differences, comprehend their applicability and potential complementarity, and lay the foundation for future method development and understanding. This review article presents 19 methods that aim to integrate scRNA-seq data and spatial transcriptomics data. The methods are classified into two main groups and described accordingly. The article also emphasizes the incorporation of High Variance Genes in annotating various technologies, aiming to obtain biologically relevant information aligned with the intended purpose.

目前已开发出许多方法来整合空间转录组学测序数据和单细胞 RNA 测序(scRNA-seq)数据。这些方法的不断发展和改进为基于不同研究调查的 scRNA-seq 和空间转录组学数据的整合和分析提供了多种选择。然而,每种方法都有其自身的优势、局限性和应用范围。研究人员需要根据实际情况选择最适合自己研究目的的方法。本综述文章汇编了 19 种整合方法,这些方法来源广泛,可为研究人员选择适合其特定研究调查的整合方法提供全面参考。通过了解这些方法的原理,我们可以找出它们的异同,理解它们的适用性和潜在互补性,并为未来的方法开发和理解奠定基础。本综述文章介绍了 19 种旨在整合 scRNA-seq 数据和空间转录组学数据的方法。这些方法被分为两大类,并进行了相应的描述。文章还强调了高方差基因在各种技术注释中的应用,旨在获得与预期目的一致的生物相关信息。
{"title":"Integration tools for scRNA-seq data and spatial transcriptomics sequencing data.","authors":"Chaorui Yan, Yanxu Zhu, Miao Chen, Kainan Yang, Feifei Cui, Quan Zou, Zilong Zhang","doi":"10.1093/bfgp/elae002","DOIUrl":"10.1093/bfgp/elae002","url":null,"abstract":"<p><p>Numerous methods have been developed to integrate spatial transcriptomics sequencing data with single-cell RNA sequencing (scRNA-seq) data. Continuous development and improvement of these methods offer multiple options for integrating and analyzing scRNA-seq and spatial transcriptomics data based on diverse research inquiries. However, each method has its own advantages, limitations and scope of application. Researchers need to select the most suitable method for their research purposes based on the actual situation. This review article presents a compilation of 19 integration methods sourced from a wide range of available approaches, serving as a comprehensive reference for researchers to select the suitable integration method for their specific research inquiries. By understanding the principles of these methods, we can identify their similarities and differences, comprehend their applicability and potential complementarity, and lay the foundation for future method development and understanding. This review article presents 19 methods that aim to integrate scRNA-seq data and spatial transcriptomics data. The methods are classified into two main groups and described accordingly. The article also emphasizes the incorporation of High Variance Genes in annotating various technologies, aiming to obtain biologically relevant information aligned with the intended purpose.</p>","PeriodicalId":55323,"journal":{"name":"Briefings in Functional Genomics","volume":" ","pages":"295-302"},"PeriodicalIF":2.5,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139547636","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Integrating multi-omics data to analyze the potential pathogenic mechanism of CTSH gene involved in type 1 diabetes in the exocrine pancreas. 整合多组学数据,分析外分泌胰腺CTSH基因参与1型糖尿病的潜在致病机制。
IF 2.5 3区 生物学 Q3 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elad052
Zerun Song, Shuai Li, Zhenwei Shang, Wenhua Lv, Xiangshu Cheng, Xin Meng, Rui Chen, Shuhao Zhang, Ruijie Zhang

Type 1 diabetes (T1D) is an autoimmune disease caused by the destruction of insulin-producing pancreatic islet beta cells. Despite significant advancements, the precise pathogenesis of the disease remains unknown. This work integrated data from expression quantitative trait locus (eQTL) studies with Genome wide association study (GWAS) summary data of T1D and single-cell transcriptome data to investigate the potential pathogenic mechanisms of the CTSH gene involved in T1D in exocrine pancreas. Using the summary data-based Mendelian randomization (SMR) approach, we obtained four potential causative genes associated with T1D: BTN3A2, PGAP3, SMARCE1 and CTSH. To further investigate these genes'roles in T1D development, we validated them using a scRNA-seq dataset from pancreatic tissues of both T1D patients and healthy controls. The analysis showed a significantly high expression of the CTSH gene in T1D acinar cells, whereas the other three genes showed no significant changes in the scRNA-seq data. Moreover, single-cell WGCNA analysis revealed the strongest positive correlation between the module containing CTSH and T1D. In addition, we found cellular ligand-receptor interactions between the acinar cells and different cell types, especially ductal cells. Finally, based on functional enrichment analysis, we hypothesized that the CTSH gene in the exocrine pancreas enhances the antiviral response, leading to the overexpression of pro-inflammatory cytokines and the development of an inflammatory microenvironment. This process promotes β cells injury and ultimately the development of T1D. Our findings offer insights into the underlying pathogenic mechanisms of T1D.

1型糖尿病(T1D)是一种由产生胰岛素的胰岛细胞破坏引起的自身免疫性疾病。尽管取得了重大进展,但该疾病的确切发病机制仍不清楚。本研究结合表达数量性状位点(eQTL)研究数据、T1D基因组全关联研究(GWAS)汇总数据和单细胞转录组数据,探讨外分泌胰腺中CTSH基因参与T1D的潜在致病机制。采用基于汇总数据的孟德尔随机化(SMR)方法,我们获得了4个与T1D相关的潜在致病基因:BTN3A2、PGAP3、SMARCE1和CTSH。为了进一步研究这些基因在T1D发展中的作用,我们使用来自T1D患者和健康对照者胰腺组织的scRNA-seq数据集验证了它们。分析显示CTSH基因在T1D腺泡细胞中显著高表达,而其他三个基因在scRNA-seq数据中没有显著变化。此外,单细胞WGCNA分析显示,含有CTSH的模块与T1D之间存在最强的正相关。此外,我们发现腺泡细胞与不同类型的细胞,特别是导管细胞之间存在细胞配体-受体相互作用。最后,基于功能富集分析,我们假设外分泌胰腺中的CTSH基因增强了抗病毒反应,导致促炎细胞因子的过度表达和炎症微环境的形成。这一过程促进β细胞损伤并最终导致T1D的发生。我们的研究结果为T1D的潜在致病机制提供了见解。
{"title":"Integrating multi-omics data to analyze the potential pathogenic mechanism of CTSH gene involved in type 1 diabetes in the exocrine pancreas.","authors":"Zerun Song, Shuai Li, Zhenwei Shang, Wenhua Lv, Xiangshu Cheng, Xin Meng, Rui Chen, Shuhao Zhang, Ruijie Zhang","doi":"10.1093/bfgp/elad052","DOIUrl":"10.1093/bfgp/elad052","url":null,"abstract":"<p><p>Type 1 diabetes (T1D) is an autoimmune disease caused by the destruction of insulin-producing pancreatic islet beta cells. Despite significant advancements, the precise pathogenesis of the disease remains unknown. This work integrated data from expression quantitative trait locus (eQTL) studies with Genome wide association study (GWAS) summary data of T1D and single-cell transcriptome data to investigate the potential pathogenic mechanisms of the CTSH gene involved in T1D in exocrine pancreas. Using the summary data-based Mendelian randomization (SMR) approach, we obtained four potential causative genes associated with T1D: BTN3A2, PGAP3, SMARCE1 and CTSH. To further investigate these genes'roles in T1D development, we validated them using a scRNA-seq dataset from pancreatic tissues of both T1D patients and healthy controls. The analysis showed a significantly high expression of the CTSH gene in T1D acinar cells, whereas the other three genes showed no significant changes in the scRNA-seq data. Moreover, single-cell WGCNA analysis revealed the strongest positive correlation between the module containing CTSH and T1D. In addition, we found cellular ligand-receptor interactions between the acinar cells and different cell types, especially ductal cells. Finally, based on functional enrichment analysis, we hypothesized that the CTSH gene in the exocrine pancreas enhances the antiviral response, leading to the overexpression of pro-inflammatory cytokines and the development of an inflammatory microenvironment. This process promotes β cells injury and ultimately the development of T1D. Our findings offer insights into the underlying pathogenic mechanisms of T1D.</p>","PeriodicalId":55323,"journal":{"name":"Briefings in Functional Genomics","volume":" ","pages":"406-417"},"PeriodicalIF":2.5,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138483553","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Predicting the role of the human gut microbiome in type 1 diabetes using machine-learning methods. 利用机器学习方法预测人类肠道微生物组在 1 型糖尿病中的作用。
IF 2.5 3区 生物学 Q3 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elae004
Xiao-Wei Liu, Han-Lin Li, Cai-Yi Ma, Tian-Yu Shi, Tian-Yu Wang, Dan Yan, Hua Tang, Hao Lin, Ke-Jun Deng

Gut microbes is a crucial factor in the pathogenesis of type 1 diabetes (T1D). However, it is still unclear which gut microbiota are the key factors affecting T1D and their influence on the development and progression of the disease. To fill these knowledge gaps, we constructed a model to find biomarker from gut microbiota in patients with T1D. We first identified microbial markers using Linear discriminant analysis Effect Size (LEfSe) and random forest (RF) methods. Furthermore, by constructing co-occurrence networks for gut microbes in T1D, we aimed to reveal all gut microbial interactions as well as major beneficial and pathogenic bacteria in healthy populations and type 1 diabetic patients. Finally, PICRUST2 was used to predict Kyoto Encyclopedia of Genes and Genomes (KEGG) functional pathways and KO gene levels of microbial markers to investigate the biological role. Our study revealed that 21 identified microbial genera are important biomarker for T1D. Their AUC values are 0.962 and 0.745 on discovery set and validation set. Functional analysis showed that 10 microbial genera were significantly positively associated with D-arginine and D-ornithine metabolism, spliceosome in transcription, steroid hormone biosynthesis and glycosaminoglycan degradation. These genera were significantly negatively correlated with steroid biosynthesis, cyanoamino acid metabolism and drug metabolism. The other 11 genera displayed an inverse correlation. In summary, our research identified a comprehensive set of T1D gut biomarkers with universal applicability and have revealed the biological consequences of alterations in gut microbiota and their interplay. These findings offer significant prospects for individualized management and treatment of T1D.

肠道微生物是 1 型糖尿病(T1D)发病机制中的一个关键因素。 然而,目前仍不清楚哪些肠道微生物群是影响 T1D 的关键因素,也不清楚它们对疾病的发生和发展有何影响。为了填补这些知识空白,我们建立了一个模型,从 T1D 患者的肠道微生物群中寻找生物标志物。我们首先使用线性判别分析效应大小(LEfSe)和随机森林(RF)方法确定了微生物标记物。此外,通过构建 T1D 肠道微生物共现网络,我们旨在揭示健康人群和 1 型糖尿病患者的所有肠道微生物相互作用以及主要有益菌和致病菌。最后,我们利用 PICRUST2 预测了《京都基因与基因组百科全书》(KEGG)功能通路和微生物标记物的 KO 基因水平,以研究其生物学作用。我们的研究发现,21 个已识别的微生物属是 T1D 的重要生物标记物。它们在发现集和验证集上的AUC值分别为0.962和0.745。功能分析显示,10 个微生物属与 D-精氨酸和 D-鸟氨酸代谢、转录中的剪接体、类固醇激素生物合成和糖胺聚糖降解呈显著正相关。这些属与类固醇生物合成、氰基氨基酸代谢和药物代谢呈明显负相关。其他 11 个属呈反向相关。总之,我们的研究确定了一套具有普遍适用性的 T1D 肠道生物标志物,并揭示了肠道微生物群改变及其相互作用的生物学后果。这些发现为 T1D 的个体化管理和治疗提供了重要的前景。
{"title":"Predicting the role of the human gut microbiome in type 1 diabetes using machine-learning methods.","authors":"Xiao-Wei Liu, Han-Lin Li, Cai-Yi Ma, Tian-Yu Shi, Tian-Yu Wang, Dan Yan, Hua Tang, Hao Lin, Ke-Jun Deng","doi":"10.1093/bfgp/elae004","DOIUrl":"10.1093/bfgp/elae004","url":null,"abstract":"<p><p>Gut microbes is a crucial factor in the pathogenesis of type 1 diabetes (T1D). However, it is still unclear which gut microbiota are the key factors affecting T1D and their influence on the development and progression of the disease. To fill these knowledge gaps, we constructed a model to find biomarker from gut microbiota in patients with T1D. We first identified microbial markers using Linear discriminant analysis Effect Size (LEfSe) and random forest (RF) methods. Furthermore, by constructing co-occurrence networks for gut microbes in T1D, we aimed to reveal all gut microbial interactions as well as major beneficial and pathogenic bacteria in healthy populations and type 1 diabetic patients. Finally, PICRUST2 was used to predict Kyoto Encyclopedia of Genes and Genomes (KEGG) functional pathways and KO gene levels of microbial markers to investigate the biological role. Our study revealed that 21 identified microbial genera are important biomarker for T1D. Their AUC values are 0.962 and 0.745 on discovery set and validation set. Functional analysis showed that 10 microbial genera were significantly positively associated with D-arginine and D-ornithine metabolism, spliceosome in transcription, steroid hormone biosynthesis and glycosaminoglycan degradation. These genera were significantly negatively correlated with steroid biosynthesis, cyanoamino acid metabolism and drug metabolism. The other 11 genera displayed an inverse correlation. In summary, our research identified a comprehensive set of T1D gut biomarkers with universal applicability and have revealed the biological consequences of alterations in gut microbiota and their interplay. These findings offer significant prospects for individualized management and treatment of T1D.</p>","PeriodicalId":55323,"journal":{"name":"Briefings in Functional Genomics","volume":" ","pages":"464-474"},"PeriodicalIF":2.5,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139906960","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
EIEPCF: accurate inference of functional gene regulatory networks by eliminating indirect effects from confounding factors. EIEPCF:通过消除混杂因素的间接影响,准确推断功能基因调控网络。
IF 2.5 3区 生物学 Q3 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elad040
Huixiang Peng, Jing Xu, Kangchen Liu, Fang Liu, Aidi Zhang, Xiujun Zhang

Reconstructing functional gene regulatory networks (GRNs) is a primary prerequisite for understanding pathogenic mechanisms and curing diseases in animals, and it also provides an important foundation for cultivating vegetable and fruit varieties that are resistant to diseases and corrosion in plants. Many computational methods have been developed to infer GRNs, but most of the regulatory relationships between genes obtained by these methods are biased. Eliminating indirect effects in GRNs remains a significant challenge for researchers. In this work, we propose a novel approach for inferring functional GRNs, named EIEPCF (eliminating indirect effects produced by confounding factors), which eliminates indirect effects caused by confounding factors. This method eliminates the influence of confounding factors on regulatory factors and target genes by measuring the similarity between their residuals. The validation results of the EIEPCF method on simulation studies, the gold-standard networks provided by the DREAM3 Challenge and the real gene networks of Escherichia coli demonstrate that it achieves significantly higher accuracy compared to other popular computational methods for inferring GRNs. As a case study, we utilized the EIEPCF method to reconstruct the cold-resistant specific GRN from gene expression data of cold-resistant in Arabidopsis thaliana. The source code and data are available at https://github.com/zhanglab-wbgcas/EIEPCF.

重建功能基因调控网络(GRN)是了解动物致病机制和治愈疾病的首要前提,也为培育植物抗病抗腐蚀的蔬果品种奠定了重要基础。目前已开发出许多推断 GRN 的计算方法,但这些方法得到的基因间调控关系大多存在偏差。消除 GRN 中的间接效应仍是研究人员面临的一项重大挑战。在这项工作中,我们提出了一种推断功能性 GRN 的新方法,命名为 EIEPCF(消除混杂因素产生的间接效应),它可以消除混杂因素造成的间接效应。这种方法通过测量混杂因子和靶基因残差之间的相似性来消除混杂因子对调控因子和靶基因的影响。EIEPCF 方法在模拟研究、DREAM3 挑战赛提供的黄金标准网络和大肠杆菌真实基因网络上的验证结果表明,与其他推断 GRN 的流行计算方法相比,该方法的准确性明显更高。作为案例研究,我们利用 EIEPCF 方法从拟南芥抗寒基因表达数据中重建了抗寒特异性 GRN。源代码和数据见 https://github.com/zhanglab-wbgcas/EIEPCF。
{"title":"EIEPCF: accurate inference of functional gene regulatory networks by eliminating indirect effects from confounding factors.","authors":"Huixiang Peng, Jing Xu, Kangchen Liu, Fang Liu, Aidi Zhang, Xiujun Zhang","doi":"10.1093/bfgp/elad040","DOIUrl":"10.1093/bfgp/elad040","url":null,"abstract":"<p><p>Reconstructing functional gene regulatory networks (GRNs) is a primary prerequisite for understanding pathogenic mechanisms and curing diseases in animals, and it also provides an important foundation for cultivating vegetable and fruit varieties that are resistant to diseases and corrosion in plants. Many computational methods have been developed to infer GRNs, but most of the regulatory relationships between genes obtained by these methods are biased. Eliminating indirect effects in GRNs remains a significant challenge for researchers. In this work, we propose a novel approach for inferring functional GRNs, named EIEPCF (eliminating indirect effects produced by confounding factors), which eliminates indirect effects caused by confounding factors. This method eliminates the influence of confounding factors on regulatory factors and target genes by measuring the similarity between their residuals. The validation results of the EIEPCF method on simulation studies, the gold-standard networks provided by the DREAM3 Challenge and the real gene networks of Escherichia coli demonstrate that it achieves significantly higher accuracy compared to other popular computational methods for inferring GRNs. As a case study, we utilized the EIEPCF method to reconstruct the cold-resistant specific GRN from gene expression data of cold-resistant in Arabidopsis thaliana. The source code and data are available at https://github.com/zhanglab-wbgcas/EIEPCF.</p>","PeriodicalId":55323,"journal":{"name":"Briefings in Functional Genomics","volume":" ","pages":"373-383"},"PeriodicalIF":2.5,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10112267","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Briefings in Functional Genomics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1