首页 > 最新文献

Current Bioinformatics最新文献

英文 中文
LBSA-DRIVER: A Novel Approach to Identifying Cancer Driver Genes Using List-Based Simulated Annealing LBSA-DRIVER:利用基于列表的模拟退火法识别癌症驱动基因的新方法
IF 4 3区 生物学 Q3 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-15 DOI: 10.2174/0115748936302984240604061302
Yilmaz Atay, Lionel Alangeh Ngobesing, Mustafa Ozgur Cingiz
Introduction: Cancer driver genes are genes responsible for cancer genesis; thus, identifying cancer-related genes is crucial in fostering cancer treatment. The accuracy in identifying cancer driver genes within the vast pool of normal passenger genes directly influences the efficacy of treatment approaches. Objective: This research aimed to effectively identify cancer driver genes using the List-based Simulated Annealing (LBSA) optimization technique. Method: The proposed model (LBSA-DRIVER) harnesses a list-based simulated annealing algorithm within a bipartite network to pinpoint cancer driver genes. The process begins with creating a bipartite graph that integrates gene mutations and expression data from carefully chosen datasets. The LBSA algorithm is then applied to the generated graph to identify and rank the genes, drawing insights from a biological interaction network. Result: Following the algorithm's development, rigorous experimental analyses have been conducted using four benchmark datasets from The Cancer Genome Atlas (TCGA) database. The datasets used were the Breast Cancer dataset (BRCA), Prostate Adenocarcinoma dataset (PRAD), Ovarian cancer dataset (OV), and Glioblastoma Multiforme dataset (GBM). Conclusion: Our findings, including precision, recall, F-score, and accuracy metrics, provide strong evidence of the effectiveness of the proposed model in identifying driver genes.
简介癌症驱动基因是导致癌症发生的基因,因此,识别癌症相关基因对促进癌症治疗至关重要。在大量正常客体基因中识别癌症驱动基因的准确性直接影响着治疗方法的效果。研究目的本研究旨在利用基于列表的模拟退火(LBSA)优化技术有效识别癌症驱动基因。方法:所提出的模型(LBSA-DRIVER)在双态网络中利用基于列表的模拟退火算法来精确定位癌症驱动基因。这一过程首先要创建一个双栅格图,将基因突变和表达数据从精心选择的数据集中整合到一起。然后将 LBSA 算法应用到生成的图中,对基因进行识别和排序,并从生物交互网络中汲取灵感。结果算法开发完成后,我们使用癌症基因组图谱(TCGA)数据库中的四个基准数据集进行了严格的实验分析。这些数据集包括乳腺癌数据集(BRCA)、前列腺癌数据集(PRAD)、卵巢癌数据集(OV)和多形性胶质母细胞瘤数据集(GBM)。结论我们的研究结果,包括精确度、召回率、F-分数和准确度指标,有力地证明了所提出的模型在识别驱动基因方面的有效性。
{"title":"LBSA-DRIVER: A Novel Approach to Identifying Cancer Driver Genes Using List-Based Simulated Annealing","authors":"Yilmaz Atay, Lionel Alangeh Ngobesing, Mustafa Ozgur Cingiz","doi":"10.2174/0115748936302984240604061302","DOIUrl":"https://doi.org/10.2174/0115748936302984240604061302","url":null,"abstract":"Introduction: Cancer driver genes are genes responsible for cancer genesis; thus, identifying cancer-related genes is crucial in fostering cancer treatment. The accuracy in identifying cancer driver genes within the vast pool of normal passenger genes directly influences the efficacy of treatment approaches. Objective: This research aimed to effectively identify cancer driver genes using the List-based Simulated Annealing (LBSA) optimization technique. Method: The proposed model (LBSA-DRIVER) harnesses a list-based simulated annealing algorithm within a bipartite network to pinpoint cancer driver genes. The process begins with creating a bipartite graph that integrates gene mutations and expression data from carefully chosen datasets. The LBSA algorithm is then applied to the generated graph to identify and rank the genes, drawing insights from a biological interaction network. Result: Following the algorithm's development, rigorous experimental analyses have been conducted using four benchmark datasets from The Cancer Genome Atlas (TCGA) database. The datasets used were the Breast Cancer dataset (BRCA), Prostate Adenocarcinoma dataset (PRAD), Ovarian cancer dataset (OV), and Glioblastoma Multiforme dataset (GBM). Conclusion: Our findings, including precision, recall, F-score, and accuracy metrics, provide strong evidence of the effectiveness of the proposed model in identifying driver genes.","PeriodicalId":10801,"journal":{"name":"Current Bioinformatics","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141720127","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
MFTP-Tool: A Wide & Deep Learning Framework for Multi-Functional Therapeutic Peptides Prediction MFTP 工具:用于多功能治疗肽预测的广泛深度学习框架
IF 4 3区 生物学 Q3 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-10 DOI: 10.2174/0115748936299646240625092734
Yang Lv, Ting Liu, YuChen Ma, Hongqiang Lyu, Ze Liu
Background: The identification and functional prediction of Multifunctional Therapeutic Peptides (MFTP) play a pivotal role in drug discovery, particularly for conditions such as inflammation and hyperglycemia. Current computational methods exhibit limitations in their ability to accurately predict the multifunctionality of these peptides. Methods: We propose a novel Wide and Deep Learning Framework that integrates both deep learning and machine learning approaches. The deep learning segment processes word vectors using a neural network model, while the wide segment utilizes the physicochemical properties of peptides in a random forest-based model. This hybrid approach aims to enhance the accuracy of MFTP function prediction. Results: Our framework outperformed the existing PrMFTP predictor in terms of precision, coverage, accuracy, and absolute true values. The evaluation was conducted on both training and independent testing datasets, demonstrating the robustness and generalizability of our model. Conclusion: The proposed Wide & Deep Learning Framework offers a significant advancement in the computational prediction of MFTP functions. The availability of our model through a userfriendly web interface at MFTP-Tool.m6aminer.cn provides a valuable tool for researchers in the field of therapeutic peptide-based drug discovery, potentially accelerating the development of new treatments.
背景:多功能治疗肽(MFTP)的鉴定和功能预测在药物发现,尤其是炎症和高血糖等疾病的药物发现中发挥着关键作用。目前的计算方法在准确预测这些多肽的多功能性方面存在局限性。方法:我们提出了一种新颖的广泛深度学习框架,它整合了深度学习和机器学习方法。深度学习部分使用神经网络模型处理词向量,而广度学习部分则在基于随机森林的模型中利用肽的物理化学特性。这种混合方法旨在提高 MFTP 功能预测的准确性。结果我们的框架在精确度、覆盖率、准确度和绝对真实值方面都优于现有的 PrMFTP 预测器。评估是在训练数据集和独立测试数据集上进行的,这证明了我们模型的鲁棒性和通用性。结论所提出的宽amp; 深度学习框架在 MFTP 功能的计算预测方面取得了重大进展。我们的模型可以通过MFTP-Tool.m6aminer.cn上的用户友好型网络界面获得,这为基于治疗肽的药物发现领域的研究人员提供了一个宝贵的工具,有可能加速新疗法的开发。
{"title":"MFTP-Tool: A Wide & Deep Learning Framework for Multi-Functional Therapeutic Peptides Prediction","authors":"Yang Lv, Ting Liu, YuChen Ma, Hongqiang Lyu, Ze Liu","doi":"10.2174/0115748936299646240625092734","DOIUrl":"https://doi.org/10.2174/0115748936299646240625092734","url":null,"abstract":"Background: The identification and functional prediction of Multifunctional Therapeutic Peptides (MFTP) play a pivotal role in drug discovery, particularly for conditions such as inflammation and hyperglycemia. Current computational methods exhibit limitations in their ability to accurately predict the multifunctionality of these peptides. Methods: We propose a novel Wide and Deep Learning Framework that integrates both deep learning and machine learning approaches. The deep learning segment processes word vectors using a neural network model, while the wide segment utilizes the physicochemical properties of peptides in a random forest-based model. This hybrid approach aims to enhance the accuracy of MFTP function prediction. Results: Our framework outperformed the existing PrMFTP predictor in terms of precision, coverage, accuracy, and absolute true values. The evaluation was conducted on both training and independent testing datasets, demonstrating the robustness and generalizability of our model. Conclusion: The proposed Wide & Deep Learning Framework offers a significant advancement in the computational prediction of MFTP functions. The availability of our model through a userfriendly web interface at MFTP-Tool.m6aminer.cn provides a valuable tool for researchers in the field of therapeutic peptide-based drug discovery, potentially accelerating the development of new treatments.","PeriodicalId":10801,"journal":{"name":"Current Bioinformatics","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141586134","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
Automatic Detection of Standard Planes in Fetal Ultrasound Images based on Convolutional Neural Networks and Ensemble Learning 基于卷积神经网络和集合学习的胎儿超声图像标准平面自动检测技术
IF 4 3区 生物学 Q3 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-10 DOI: 10.2174/0115748936295679240620094626
Baoping Zhu, Fan Yang, Hongliang Duan, Zhipeng Gao
aims: This study aims to leverage artificial intelligence for enhancing medical diagnosis, focusing on ultrasound evaluation of fetal development and detection of fetal diseases. background: Traditional diagnostic methods in ultrasound are known for being time-consuming and laborious, prompting the need for more efficient approaches. objective: The objective of this research is to develop an end-to-end automatic diagnosis system using convolutional neural networks with ensemble learning to enhance robustness and accuracy in classifying ultrasound images. method: The study involves constructing and implementing the automatic diagnosis system, training it on a diverse dataset encompassing six categories: abdomen, brain, femur, thorax, maternal cervix, and other planes. result: Experimental results demonstrate that the proposed end-to-end system significantly improves the detection accuracy of the standard plane in ultrasound images. conclusion: The application of artificial intelligence through an ensemble learning-based automatic diagnosis system shows promise in advancing ultrasound-based medical diagnosis, particularly in fetal development assessment. other: This research contributes to the ongoing efforts in leveraging technology for more efficient and accurate medical diagnostic processes.
目的本研究旨在利用人工智能提高医疗诊断水平,重点关注胎儿发育的超声评估和胎儿疾病的检测。 背景:众所周知,传统的超声诊断方法费时费力,因此需要更高效的方法:众所周知,传统的超声诊断方法费时费力,因此需要更高效的方法:本研究的目的是利用卷积神经网络和集合学习开发端到端自动诊断系统,以提高超声图像分类的鲁棒性和准确性:研究包括构建和实施自动诊断系统,并在包括腹部、脑部、股骨、胸部、产妇宫颈和其他平面等六个类别的不同数据集上对其进行训练:实验结果表明,所提出的端到端系统显著提高了超声图像中标准平面的检测准确率。 结论:通过基于集合学习的自动识别系统应用人工智能,有望推动基于超声的医学诊断,尤其是胎儿发育评估:这项研究有助于利用技术提高医疗诊断过程的效率和准确性。
{"title":"Automatic Detection of Standard Planes in Fetal Ultrasound Images based on Convolutional Neural Networks and Ensemble Learning","authors":"Baoping Zhu, Fan Yang, Hongliang Duan, Zhipeng Gao","doi":"10.2174/0115748936295679240620094626","DOIUrl":"https://doi.org/10.2174/0115748936295679240620094626","url":null,"abstract":"aims: This study aims to leverage artificial intelligence for enhancing medical diagnosis, focusing on ultrasound evaluation of fetal development and detection of fetal diseases. background: Traditional diagnostic methods in ultrasound are known for being time-consuming and laborious, prompting the need for more efficient approaches. objective: The objective of this research is to develop an end-to-end automatic diagnosis system using convolutional neural networks with ensemble learning to enhance robustness and accuracy in classifying ultrasound images. method: The study involves constructing and implementing the automatic diagnosis system, training it on a diverse dataset encompassing six categories: abdomen, brain, femur, thorax, maternal cervix, and other planes. result: Experimental results demonstrate that the proposed end-to-end system significantly improves the detection accuracy of the standard plane in ultrasound images. conclusion: The application of artificial intelligence through an ensemble learning-based automatic diagnosis system shows promise in advancing ultrasound-based medical diagnosis, particularly in fetal development assessment. other: This research contributes to the ongoing efforts in leveraging technology for more efficient and accurate medical diagnostic processes.","PeriodicalId":10801,"journal":{"name":"Current Bioinformatics","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141586136","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
Rank Matrix Approach for Endometriosis: Integrating Data and Constructing Diagnostic Models 子宫内膜异位症的等级矩阵法:整合数据并构建诊断模型
IF 4 3区 生物学 Q3 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-10 DOI: 10.2174/0115748936296151240605053713
Ranze Xie, Deqing Hong, Jiaqi Yuan, Peng Xu, Wenbin Liu, Zheng Ye
Background: Endometriosis is a debilitating gynecological disorder characterized by chronic pain, infertility, and the growth of endometrial tissue outside the uterus. Accurate and early detection of this condition is crucial for effective management and treatment. Methods: We developed a gene rank matrix-based model to integrate endometriosis cohorts across multiple platforms. After removing batch effects, we identified 83 genes associated with endometriosis and further refined a diagnostic model using 11 of these genes. The model was trained on two platforms and validated on two others using SVM, Random Forest, Logistic Regression, and gradient-boosting machine learning algorithms. Results: The integration via the gene rank matrix effectively mitigated batch effects. Utilizing a gradient boosting classifier with a subset of 11 genes, the model demonstrated commendable diagnostic efficacy, achieving an Area Under the Curve (AUC) of 0.77, an accuracy of 0.72, and an F1 score of 0.72 for the training dataset. When subjected to validation, the model maintained its performance, yielding an AUC of 0.769, an accuracy of 0.719, and an F1 score of 0.732. These 11 genes were found to be associated with immunosuppression. Conclusion: Our approach to integrating gene rank matrices effectively consolidates endometriosis data across diverse platforms. The diagnostic model, harnessing the predictive power of 11 specific genes, surpasses alternative models, thereby offering promising prospects for aiding clinical diagnosis of endometriosis. Further validation is imperative to elucidate the functional significance of these 11 genes. Our study underscores the potential of data integration coupled with machine learning techniques in advancing the diagnosis of intricate diseases, such as endometriosis.
背景:子宫内膜异位症是一种使人衰弱的妇科疾病,其特点是慢性疼痛、不孕和子宫内膜组织在子宫腔外生长。准确、及早地发现这种疾病对于有效管理和治疗至关重要。方法我们开发了一个基于基因排序矩阵的模型来整合多个平台的子宫内膜异位症队列。去除批次效应后,我们确定了 83 个与子宫内膜异位症相关的基因,并利用其中 11 个基因进一步完善了诊断模型。该模型在两个平台上进行了训练,并使用 SVM、随机森林、逻辑回归和梯度提升机器学习算法在另外两个平台上进行了验证。结果:通过基因等级矩阵进行整合有效地减轻了批次效应。利用梯度提升分类器的 11 个基因子集,该模型展示了值得称赞的诊断效果,训练数据集的曲线下面积(AUC)为 0.77,准确率为 0.72,F1 得分为 0.72。在进行验证时,该模型的性能保持不变,AUC 为 0.769,准确率为 0.719,F1 得分为 0.732。发现这 11 个基因与免疫抑制有关。结论我们整合基因排序矩阵的方法有效地整合了不同平台上的子宫内膜异位症数据。该诊断模型利用 11 个特定基因的预测能力,超越了其他模型,从而为子宫内膜异位症的临床诊断提供了广阔的前景。要阐明这 11 个基因的功能意义,进一步的验证势在必行。我们的研究强调了数据整合与机器学习技术在推进子宫内膜异位症等复杂疾病诊断方面的潜力。
{"title":"Rank Matrix Approach for Endometriosis: Integrating Data and Constructing Diagnostic Models","authors":"Ranze Xie, Deqing Hong, Jiaqi Yuan, Peng Xu, Wenbin Liu, Zheng Ye","doi":"10.2174/0115748936296151240605053713","DOIUrl":"https://doi.org/10.2174/0115748936296151240605053713","url":null,"abstract":"Background: Endometriosis is a debilitating gynecological disorder characterized by chronic pain, infertility, and the growth of endometrial tissue outside the uterus. Accurate and early detection of this condition is crucial for effective management and treatment. Methods: We developed a gene rank matrix-based model to integrate endometriosis cohorts across multiple platforms. After removing batch effects, we identified 83 genes associated with endometriosis and further refined a diagnostic model using 11 of these genes. The model was trained on two platforms and validated on two others using SVM, Random Forest, Logistic Regression, and gradient-boosting machine learning algorithms. Results: The integration via the gene rank matrix effectively mitigated batch effects. Utilizing a gradient boosting classifier with a subset of 11 genes, the model demonstrated commendable diagnostic efficacy, achieving an Area Under the Curve (AUC) of 0.77, an accuracy of 0.72, and an F1 score of 0.72 for the training dataset. When subjected to validation, the model maintained its performance, yielding an AUC of 0.769, an accuracy of 0.719, and an F1 score of 0.732. These 11 genes were found to be associated with immunosuppression. Conclusion: Our approach to integrating gene rank matrices effectively consolidates endometriosis data across diverse platforms. The diagnostic model, harnessing the predictive power of 11 specific genes, surpasses alternative models, thereby offering promising prospects for aiding clinical diagnosis of endometriosis. Further validation is imperative to elucidate the functional significance of these 11 genes. Our study underscores the potential of data integration coupled with machine learning techniques in advancing the diagnosis of intricate diseases, such as endometriosis.","PeriodicalId":10801,"journal":{"name":"Current Bioinformatics","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141586217","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
GenRepAI: Utilizing Artificial Intelligence to Identify Repeats in Genomic Suffix Trees GenRepAI:利用人工智能识别基因组后缀树中的重复序列
IF 4 3区 生物学 Q3 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-10 DOI: 10.2174/0115748936303435240702112205
Freeson Kaniwa
Background: The human genome is densely populated with repetitive DNA sequences that play crucial roles in genomic functions and structures but are also implicated in over 40 human diseases. The computational challenge of identifying and characterizing these repeats is significant due to the complexity and size of the genome, which are overwhelming traditional algorithms. Methods: To address these challenges, we propose GenRepAI, a deep learning framework to navigate and analyze genomic suffix trees. GenRepAI employs supervised machine learning classifiers trained on labeled datasets of repeat annotations and unsupervised anomaly detection to identify novel repeat sequences. The models are trained using convolutional neural networks (CNNs), long short-term memory networks (LSTMs), and vision transformers to classify and annotate repeats within the human genome. Results: GenRepAI is designed to comprehensively profile repeats that underlie various neurological diseases, allowing researchers to identify pathogenic expansions. The framework will integrate into existing genomic analysis pipelines, with the capability to screen patient genomes and highlight potential causal variants for further validation. Conclusion: GenRepAI is set to become a foundational tool in genomics, leveraging artificial intelligence to enhance the characterization of repetitive sequences. It promises significant advancements in the molecular diagnosis of repeat expansion disorders and contributes to a deeper understanding of genomic structure and function, with broad applications in personalized medicine.
背景:人类基因组中存在大量重复的 DNA 序列,它们在基因组功能和结构中发挥着至关重要的作用,同时也与 40 多种人类疾病有关。由于基因组的复杂性和规模,识别和表征这些重复序列的计算难度很大,传统算法难以承受。方法:为了应对这些挑战,我们提出了 GenRepAI,这是一个导航和分析基因组后缀树的深度学习框架。GenRepAI 采用在重复注释的标记数据集上训练的监督机器学习分类器和无监督异常检测来识别新的重复序列。模型使用卷积神经网络(CNN)、长短期记忆网络(LSTM)和视觉转换器进行训练,以对人类基因组中的重复序列进行分类和注释。结果GenRepAI 旨在全面剖析导致各种神经系统疾病的重复序列,使研究人员能够识别致病性扩展。该框架将集成到现有的基因组分析管道中,能够筛选患者基因组并突出潜在的因果变异,以便进一步验证。结论GenRepAI 将成为基因组学的基础工具,利用人工智能加强重复序列的特征描述。它有望在重复扩增疾病的分子诊断方面取得重大进展,并有助于加深对基因组结构和功能的理解,在个性化医疗方面有着广泛的应用。
{"title":"GenRepAI: Utilizing Artificial Intelligence to Identify Repeats in Genomic Suffix Trees","authors":"Freeson Kaniwa","doi":"10.2174/0115748936303435240702112205","DOIUrl":"https://doi.org/10.2174/0115748936303435240702112205","url":null,"abstract":"Background: The human genome is densely populated with repetitive DNA sequences that play crucial roles in genomic functions and structures but are also implicated in over 40 human diseases. The computational challenge of identifying and characterizing these repeats is significant due to the complexity and size of the genome, which are overwhelming traditional algorithms. Methods: To address these challenges, we propose GenRepAI, a deep learning framework to navigate and analyze genomic suffix trees. GenRepAI employs supervised machine learning classifiers trained on labeled datasets of repeat annotations and unsupervised anomaly detection to identify novel repeat sequences. The models are trained using convolutional neural networks (CNNs), long short-term memory networks (LSTMs), and vision transformers to classify and annotate repeats within the human genome. Results: GenRepAI is designed to comprehensively profile repeats that underlie various neurological diseases, allowing researchers to identify pathogenic expansions. The framework will integrate into existing genomic analysis pipelines, with the capability to screen patient genomes and highlight potential causal variants for further validation. Conclusion: GenRepAI is set to become a foundational tool in genomics, leveraging artificial intelligence to enhance the characterization of repetitive sequences. It promises significant advancements in the molecular diagnosis of repeat expansion disorders and contributes to a deeper understanding of genomic structure and function, with broad applications in personalized medicine.","PeriodicalId":10801,"journal":{"name":"Current Bioinformatics","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141586135","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
Screening Analysis of Predictive Markers for Cytokine Release Syndrome Risk in CAR-T Cell Therapy CAR-T 细胞疗法中细胞因子释放综合征风险预测标记物的筛选分析
IF 4 3区 生物学 Q3 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-07-03 DOI: 10.2174/0115748936295986240619162816
Jiayu Xu, Chengkui Zhao, Zhenyu Wei, Weixin Xie, Qi Cheng, Min Zhang, Shuangze Han, Liqing Kang, Nan Xu, Lei Yu, Weixing Feng
Background: Chimeric Antigen Receptor (CAR)-T cell therapy has emerged as a highly effective treatment for hematological tumors. However, the associated adverse reaction, Cytokine Release Syndrome (CRS), poses a significant challenge. While numerous studies have investigated CRS biomarkers during CAR-T cell therapy, the ability to predict CRS risk prior to treatment initiation remains a crucial yet underexplored aspect. Objective: The primary purpose of this study was to address the issue of limited data, explore an alternative approach using public data to identify predictive markers for CRS risk assessment from RNA-Seq in pre-treatment patients data, and comprehend the inducible mechanisms underlying CRS. Methods: We integrated information from two public databases, the FDA Adverse Event Reporting System (FAERS) for adverse reaction reports of CAR-T cell therapy and the Cancer Genome Atlas (TCGA) for RNA-Seq data on corresponding hematological tumors. Candidate genes were screened by correlation analysis between Reported Odds Ratio (ROR) values and RNA-Seq gene expression levels, and then core factors were identified through stepwise analysis of pathway enrichment, cluster analysis, and protein interactions. Results: Our analysis highlighted the correlation between CRS risk and pre-treatment T cell activation/ proliferation, identifying key genes (IFN-γ, IL1β, IL2, IL6, and IL10) as significant CRS indicators. Conclusion: This study offers a unique perspective on predicting CRS risk before CAR-T cell therapy, circumventing the challenges of scarce clinical data by leveraging analysis of public databases. It elucidates the crucial role of T cell activation/proliferation dynamics in CRS. The analytical methods and identified markers provide a reference for the research and clinical application of CAR-T cell therapy.
背景:嵌合抗原受体(CAR)-T 细胞疗法已成为治疗血液肿瘤的高效疗法。然而,与之相关的不良反应--细胞因子释放综合征(CRS)--带来了巨大挑战。虽然已有许多研究调查了 CAR-T 细胞治疗过程中的 CRS 生物标志物,但在治疗开始前预测 CRS 风险的能力仍然是一个至关重要但尚未得到充分探索的方面。研究目的本研究的主要目的是解决数据有限的问题,探索一种使用公共数据的替代方法,从治疗前患者数据中的 RNA-Seq 中确定 CRS 风险评估的预测标记物,并了解 CRS 的诱导机制。方法:我们整合了两个公共数据库的信息,一个是FDA不良事件报告系统(FAERS)的CAR-T细胞疗法不良反应报告,另一个是癌症基因组图谱(TCGA)的相应血液肿瘤的RNA-Seq数据。通过报告比值(ROR)和RNA-Seq基因表达水平之间的相关性分析筛选候选基因,然后通过路径富集、聚类分析和蛋白质相互作用的逐步分析确定核心因子。结果我们的分析强调了CRS风险与治疗前T细胞活化/增殖之间的相关性,确定了关键基因(IFN-γ、IL1β、IL2、IL6和IL10)作为重要的CRS指标。结论这项研究为预测 CAR-T 细胞治疗前的 CRS 风险提供了一个独特的视角,通过对公共数据库的分析规避了临床数据稀缺所带来的挑战。它阐明了 T 细胞活化/增殖动态在 CRS 中的关键作用。分析方法和确定的标记物为 CAR-T 细胞疗法的研究和临床应用提供了参考。
{"title":"Screening Analysis of Predictive Markers for Cytokine Release Syndrome Risk in CAR-T Cell Therapy","authors":"Jiayu Xu, Chengkui Zhao, Zhenyu Wei, Weixin Xie, Qi Cheng, Min Zhang, Shuangze Han, Liqing Kang, Nan Xu, Lei Yu, Weixing Feng","doi":"10.2174/0115748936295986240619162816","DOIUrl":"https://doi.org/10.2174/0115748936295986240619162816","url":null,"abstract":"Background: Chimeric Antigen Receptor (CAR)-T cell therapy has emerged as a highly effective treatment for hematological tumors. However, the associated adverse reaction, Cytokine Release Syndrome (CRS), poses a significant challenge. While numerous studies have investigated CRS biomarkers during CAR-T cell therapy, the ability to predict CRS risk prior to treatment initiation remains a crucial yet underexplored aspect. Objective: The primary purpose of this study was to address the issue of limited data, explore an alternative approach using public data to identify predictive markers for CRS risk assessment from RNA-Seq in pre-treatment patients data, and comprehend the inducible mechanisms underlying CRS. Methods: We integrated information from two public databases, the FDA Adverse Event Reporting System (FAERS) for adverse reaction reports of CAR-T cell therapy and the Cancer Genome Atlas (TCGA) for RNA-Seq data on corresponding hematological tumors. Candidate genes were screened by correlation analysis between Reported Odds Ratio (ROR) values and RNA-Seq gene expression levels, and then core factors were identified through stepwise analysis of pathway enrichment, cluster analysis, and protein interactions. Results: Our analysis highlighted the correlation between CRS risk and pre-treatment T cell activation/ proliferation, identifying key genes (IFN-γ, IL1β, IL2, IL6, and IL10) as significant CRS indicators. Conclusion: This study offers a unique perspective on predicting CRS risk before CAR-T cell therapy, circumventing the challenges of scarce clinical data by leveraging analysis of public databases. It elucidates the crucial role of T cell activation/proliferation dynamics in CRS. The analytical methods and identified markers provide a reference for the research and clinical application of CAR-T cell therapy.","PeriodicalId":10801,"journal":{"name":"Current Bioinformatics","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141550364","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
Insights into Co-Expression Network Analysis of MicroProteins and their Target Transcription Factors in Plant Embryo Development 植物胚胎发育过程中微蛋白及其目标转录因子共表达网络分析的启示
IF 4 3区 生物学 Q3 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-06-26 DOI: 10.2174/0115748936304167240530091051
Khadijeh Shokri, Naser Farrokhi, Asadollah Ahmadikhah, Mahdi Safaeizade, Amir Mousavi
Background: Gene expression is regulated in a spatiotemporal manner, and the roles of microProteins (MiPs) in this concept have started to become clear in plants. Methods: Here, a microarray data analysis was carried out to decipher the spatiotemporal role of MiPs in embryo development. The guilt-by-association method was used to determine the corresponding regulatory factors. Results: Module network analyses and protein-protein interaction (PPI) assays suggested 13 modules for embryo development in the Arabidopsis model plant. Various biological processes such as metabolite biosynthesis, hormone transition and regulation, fatty acid and storage protein biosynthesis, and photosynthesis-related processes were prevalent. Different transcription factors (TFs) at different stages of embryo development were found and reviewed. Furthermore, 106 putative MiPs were identified that might be involved in the regulation of embryo development. Candidate hub MiPs (15) at embryo developmental stages were identified by PPI network analysis and their putative regulatory roles were discussed. Previously reported MiPs, AT1G14760 (KNOX), AT5G39860 (PRE1), and AT2G46410 (CPC), were noted to be present in modules M3 and M8. Conclusion: Molecular comprehension of regulatory factors including MiPs and TFs during embryo development allows targeted breeding of the corresponding traits and genome-based engineering of value-added new varieties.
背景:基因表达是以时空方式调控的,而微蛋白(MiPs)在这一概念中的作用在植物中已开始变得清晰。方法:本文通过微阵列数据分析来揭示 MiPs 在胚胎发育中的时空作用。采用 "逐个关联 "法(guilty-by-association)确定相应的调控因子。结果模块网络分析和蛋白质-蛋白质相互作用(PPI)分析表明,拟南芥模式植物的胚胎发育有 13 个模块。各种生物过程,如代谢物生物合成、激素转换和调节、脂肪酸和贮藏蛋白生物合成以及光合作用相关过程都普遍存在。发现并综述了胚胎发育不同阶段的不同转录因子(TFs)。此外,还发现了 106 个可能参与胚胎发育调控的推定 MiPs。通过PPI网络分析确定了胚胎发育阶段的候选中枢MiPs(15个),并讨论了它们的推测调控作用。之前报道的 MiPs AT1G14760(KNOX)、AT5G39860(PRE1)和 AT2G46410(CPC)被发现存在于模块 M3 和 M8 中。结论通过对胚胎发育过程中的调控因子(包括 MiPs 和 TFs)的分子理解,可对相应性状进行有针对性的育种,并基于基因组工程技术培育出高附加值的新品种。
{"title":"Insights into Co-Expression Network Analysis of MicroProteins and their Target Transcription Factors in Plant Embryo Development","authors":"Khadijeh Shokri, Naser Farrokhi, Asadollah Ahmadikhah, Mahdi Safaeizade, Amir Mousavi","doi":"10.2174/0115748936304167240530091051","DOIUrl":"https://doi.org/10.2174/0115748936304167240530091051","url":null,"abstract":"Background: Gene expression is regulated in a spatiotemporal manner, and the roles of microProteins (MiPs) in this concept have started to become clear in plants. Methods: Here, a microarray data analysis was carried out to decipher the spatiotemporal role of MiPs in embryo development. The guilt-by-association method was used to determine the corresponding regulatory factors. Results: Module network analyses and protein-protein interaction (PPI) assays suggested 13 modules for embryo development in the Arabidopsis model plant. Various biological processes such as metabolite biosynthesis, hormone transition and regulation, fatty acid and storage protein biosynthesis, and photosynthesis-related processes were prevalent. Different transcription factors (TFs) at different stages of embryo development were found and reviewed. Furthermore, 106 putative MiPs were identified that might be involved in the regulation of embryo development. Candidate hub MiPs (15) at embryo developmental stages were identified by PPI network analysis and their putative regulatory roles were discussed. Previously reported MiPs, AT1G14760 (KNOX), AT5G39860 (PRE1), and AT2G46410 (CPC), were noted to be present in modules M3 and M8. Conclusion: Molecular comprehension of regulatory factors including MiPs and TFs during embryo development allows targeted breeding of the corresponding traits and genome-based engineering of value-added new varieties.","PeriodicalId":10801,"journal":{"name":"Current Bioinformatics","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141529313","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
Detection of DNA N6-Methyladenine Modification through SMRT-seq Features and Machine Learning Model 通过 SMRT-seq 特征和机器学习模型检测 DNA N6-甲基腺嘌呤修饰
IF 4 3区 生物学 Q3 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-06-26 DOI: 10.2174/0115748936300671240523044154
Yichu Guo, Yixuan Zhang, Xiaoqing Liu, Pingan He, Yuni Zeng, Qi Dai
Introduction: N6-methyldeoxyadenine (6mA) is the most prevalent DNA modification in both prokaryotes and eukaryotes. While single-molecule real-time sequencing (SMRT-seq) can detect 6mA events at the individual nucleotide level, its practical application is hindered by a high rate of false positives. Methods: We propose a computational model for identifying DNA 6mA that incorporates comprehensive site features from SMRT-seq and employs machine learning classifiers. Results: The results demonstrate that 99.54% and 96.55% of the identified DNA 6mA instances in C.reinhardtii correspond with motifs and peak regions identified by methylated DNA immunoprecipitation sequencing (MeDIP-seq), respectively. Compared to SMRT-seq, the proportion of predicted DNA 6mA instances within MeDIP-seq peak regions increases by 2% to 70% across the six bacterial strains Conclusion: Our proposed method effectively reduces the false-positive rate in DNA 6mA prediction.
引言N6-甲基脱氧腺嘌呤(6mA)是原核生物和真核生物中最常见的 DNA 修饰。虽然单分子实时测序(SMRT-seq)能在单个核苷酸水平上检测 6mA 事件,但其实际应用却受到高假阳性率的阻碍。方法:我们提出了一种识别DNA 6mA的计算模型,该模型结合了SMRT-seq的综合位点特征,并采用了机器学习分类器。结果结果表明,C.reinhardtii 中 99.54% 和 96.55% 已识别的 DNA 6mA 实例分别与甲基化 DNA 免疫沉淀测序(MeDIP-seq)所识别的主题和峰值区域相对应。与 SMRT-seq 相比,MeDIP-seq 峰区中预测的 DNA 6mA 实例比例在六种细菌菌株中增加了 2% 至 70%:我们提出的方法有效降低了 DNA 6mA 预测的假阳性率。
{"title":"Detection of DNA N6-Methyladenine Modification through SMRT-seq Features and Machine Learning Model","authors":"Yichu Guo, Yixuan Zhang, Xiaoqing Liu, Pingan He, Yuni Zeng, Qi Dai","doi":"10.2174/0115748936300671240523044154","DOIUrl":"https://doi.org/10.2174/0115748936300671240523044154","url":null,"abstract":"Introduction: N6-methyldeoxyadenine (6mA) is the most prevalent DNA modification in both prokaryotes and eukaryotes. While single-molecule real-time sequencing (SMRT-seq) can detect 6mA events at the individual nucleotide level, its practical application is hindered by a high rate of false positives. Methods: We propose a computational model for identifying DNA 6mA that incorporates comprehensive site features from SMRT-seq and employs machine learning classifiers. Results: The results demonstrate that 99.54% and 96.55% of the identified DNA 6mA instances in C.reinhardtii correspond with motifs and peak regions identified by methylated DNA immunoprecipitation sequencing (MeDIP-seq), respectively. Compared to SMRT-seq, the proportion of predicted DNA 6mA instances within MeDIP-seq peak regions increases by 2% to 70% across the six bacterial strains Conclusion: Our proposed method effectively reduces the false-positive rate in DNA 6mA prediction.","PeriodicalId":10801,"journal":{"name":"Current Bioinformatics","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141506056","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
Multinomial Logistic Regression with Adaptive Regularization for Cancer Subtype Classification via Multi-omics Data 利用自适应正则化的多项式逻辑回归,通过多组学数据进行癌症亚型分类
IF 4 3区 生物学 Q3 BIOCHEMICAL RESEARCH METHODS Pub Date : 2024-06-24 DOI: 10.2174/0115748936308171240605075531
Yingdi Wu, Fuzhen Cao, Juntao Li
Background: Integrating multi-omics data for cancer classification brings complementary biological insights while also facing challenges such as data integration, gene grouping, and adaptive weight construction. Objective: This paper aims to address the challenges faced by the cancer subtype classification and gene screening based on multi-omics data. Methods: Multinomial logistic regression with adaptive regularization (MLRAR) was proposed by integrating DNA methylation, gene mutation, and RNA-seq information. A data preprocessing strategy that effectively utilizes multi-omics information was presented, and the local maximum quasiclique merging (lmQCM) algorithm was implemented to group genes. Biological pathway information was utilized to evaluate the significance of gene groups, while the significance of each gene within a group was evaluated by integrating mutation information, information theory, and methylation information. Results: Compared to MRlasso, MRGL, MSGL, MROGL, AMRSOGL, and AGLRMR, the proposed method yielded improvements in subtype classification accuracy of breast cancer by 2.6%, 2.9%, 3.5%, 2.3%, 2.0%, and 1.8%, respectively. In addition, MLRAR also achieved significant improvements in ovarian cancer by 8.2%, 5.0%, 6.8%, 5.2%, 12.7%, and 6.3%, respectively. Conclusion: The proposed method can effectively deal with data integration, gene grouping, and adaptive weight construction.
背景:整合多组学数据用于癌症分类可带来互补的生物学见解,但同时也面临着数据整合、基因分组和自适应权重构建等挑战。目的:本文旨在解决癌症亚型分类和基因筛选所面临的挑战:本文旨在解决基于多组学数据的癌症亚型分类和基因筛选所面临的挑战。研究方法通过整合 DNA 甲基化、基因突变和 RNA-seq 信息,提出了自适应正则化多叉逻辑回归(MLRAR)。提出了一种有效利用多组学信息的数据预处理策略,并采用局部最大准斜率合并(lmQCM)算法对基因进行分组。利用生物通路信息来评估基因组的重要性,同时通过整合突变信息、信息论和甲基化信息来评估组内每个基因的重要性。结果:与 MRlasso、MRGL、MSGL、MROGL、AMRSOGL 和 AGLRMR 相比,所提出的方法在乳腺癌亚型分类准确率方面分别提高了 2.6%、2.9%、3.5%、2.3%、2.0% 和 1.8%。此外,MLRAR 对卵巢癌的分类准确率也有显著提高,分别提高了 8.2%、5.0%、6.8%、5.2%、12.7% 和 6.3%。结论所提出的方法能有效处理数据整合、基因分组和自适应权重构建等问题。
{"title":"Multinomial Logistic Regression with Adaptive Regularization for Cancer Subtype Classification via Multi-omics Data","authors":"Yingdi Wu, Fuzhen Cao, Juntao Li","doi":"10.2174/0115748936308171240605075531","DOIUrl":"https://doi.org/10.2174/0115748936308171240605075531","url":null,"abstract":"Background: Integrating multi-omics data for cancer classification brings complementary biological insights while also facing challenges such as data integration, gene grouping, and adaptive weight construction. Objective: This paper aims to address the challenges faced by the cancer subtype classification and gene screening based on multi-omics data. Methods: Multinomial logistic regression with adaptive regularization (MLRAR) was proposed by integrating DNA methylation, gene mutation, and RNA-seq information. A data preprocessing strategy that effectively utilizes multi-omics information was presented, and the local maximum quasiclique merging (lmQCM) algorithm was implemented to group genes. Biological pathway information was utilized to evaluate the significance of gene groups, while the significance of each gene within a group was evaluated by integrating mutation information, information theory, and methylation information. Results: Compared to MRlasso, MRGL, MSGL, MROGL, AMRSOGL, and AGLRMR, the proposed method yielded improvements in subtype classification accuracy of breast cancer by 2.6%, 2.9%, 3.5%, 2.3%, 2.0%, and 1.8%, respectively. In addition, MLRAR also achieved significant improvements in ovarian cancer by 8.2%, 5.0%, 6.8%, 5.2%, 12.7%, and 6.3%, respectively. Conclusion: The proposed method can effectively deal with data integration, gene grouping, and adaptive weight construction.","PeriodicalId":10801,"journal":{"name":"Current Bioinformatics","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-06-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141506060","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
EPI-HAN: Identification of Enhancer Promoter Interaction Using Hierarchical Attention Network EPI-HAN:利用层次注意网络识别增强子启动子相互作用
IF 4 3区 生物学 Q1 Mathematics Pub Date : 2024-06-12 DOI: 10.2174/0115748936294743240524113731
Fatma S. Ahmed, Saleh Aly, X. Liu
Enhancer-Promoter Interaction (EPI) recognition is crucial for understandinghuman development and transcriptional regulation. EPI in the genome plays a significant role inregulating gene expression. In Genome-Wide Association Studies (GWAS), EPIs help to improvethe mechanistic understanding of disease- or trait-associated genetic variants.Experimental methods for classifying EPIs are time-consuming and expensive. Consequently,there has been a growing emphasis on research focused on developing computational approachesthat leverage deep learning and other machine learning techniques. One of the main challengesin EPI prediction is the long sequences of enhancers and promoters, which most existing computationalapproaches struggle with. This paper proposes a new deep learning model based on the HierarchicalAttention Network (HAN) for EPI detection. The proposed EPI-HAN model has twounique features: (i) a hybrid embedding strategy (ii) a hierarchical HAN structure comprising twoattention layers that operate at both the individual token and smaller sequence levels.In benchmark comparisons, the EPI-HAN model demonstrates superior performance overstate-of-the-art methods, as evidenced by AUROC and AUPR metrics for specific cell lines. Specifically,for the cell lines HeLa-S3, HUVEC, and NHEK, the AUROC values are 0.962, 0.946, and0.987, respectively, and the AUPR values are 0.842, 0.724, and 0.926, respectively.The comparative results indicate that our model surpasses other state-of-the-art modelsin three out of six cell lines. The Superior performance in recognizing EPIs is attributed to the hierarchicalstructure of the attention mechanism.
增强子-启动子相互作用(EPI)识别对于理解人类发育和转录调控至关重要。基因组中的 EPI 在调控基因表达方面发挥着重要作用。在全基因组关联研究(GWAS)中,EPIs 有助于提高对疾病或性状相关遗传变异的机理认识。因此,人们越来越重视利用深度学习和其他机器学习技术开发计算方法的研究。EPI 预测的主要挑战之一是增强子和启动子的长序列,而现有的大多数计算方法都难以应对这一挑战。本文提出了一种基于层次注意网络(HAN)的新深度学习模型,用于 EPI 检测。所提出的 EPI-HAN 模型有两个特点:(在基准比较中,EPI-HAN 模型表现出优于最先进方法的性能,具体表现在特定细胞系的 AUROC 和 AUPR 指标上。具体来说,对于 HeLa-S3、HUVEC 和 NHEK 细胞系,AUROC 值分别为 0.962、0.946 和 0.987,AUPR 值分别为 0.842、0.724 和 0.926。在识别 EPI 方面的优异表现归功于注意力机制的分层结构。
{"title":"EPI-HAN: Identification of Enhancer Promoter Interaction Using Hierarchical Attention Network","authors":"Fatma S. Ahmed, Saleh Aly, X. Liu","doi":"10.2174/0115748936294743240524113731","DOIUrl":"https://doi.org/10.2174/0115748936294743240524113731","url":null,"abstract":"\u0000\u0000Enhancer-Promoter Interaction (EPI) recognition is crucial for understanding\u0000human development and transcriptional regulation. EPI in the genome plays a significant role in\u0000regulating gene expression. In Genome-Wide Association Studies (GWAS), EPIs help to improve\u0000the mechanistic understanding of disease- or trait-associated genetic variants.\u0000\u0000\u0000\u0000Experimental methods for classifying EPIs are time-consuming and expensive. Consequently,\u0000there has been a growing emphasis on research focused on developing computational approaches\u0000that leverage deep learning and other machine learning techniques. One of the main challenges\u0000in EPI prediction is the long sequences of enhancers and promoters, which most existing computational\u0000approaches struggle with. This paper proposes a new deep learning model based on the Hierarchical\u0000Attention Network (HAN) for EPI detection. The proposed EPI-HAN model has two\u0000unique features: (i) a hybrid embedding strategy (ii) a hierarchical HAN structure comprising two\u0000attention layers that operate at both the individual token and smaller sequence levels.\u0000\u0000\u0000\u0000In benchmark comparisons, the EPI-HAN model demonstrates superior performance over\u0000state-of-the-art methods, as evidenced by AUROC and AUPR metrics for specific cell lines. Specifically,\u0000for the cell lines HeLa-S3, HUVEC, and NHEK, the AUROC values are 0.962, 0.946, and\u00000.987, respectively, and the AUPR values are 0.842, 0.724, and 0.926, respectively.\u0000\u0000\u0000\u0000The comparative results indicate that our model surpasses other state-of-the-art models\u0000in three out of six cell lines. The Superior performance in recognizing EPIs is attributed to the hierarchical\u0000structure of the attention mechanism.\u0000","PeriodicalId":10801,"journal":{"name":"Current Bioinformatics","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-06-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141350099","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
Current Bioinformatics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1