Briefings in Functional Genomics最新文献

筛选
英文 中文
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
Briefings in Functional Genomics Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elad054
David Enoma
{"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":null,"pages":null},"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
Briefings in Functional Genomics Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elae001
Monika Khandelwal, Ranjeet Kumar Rout
{"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":null,"pages":null},"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
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
Briefings in Functional Genomics 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
{"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":null,"pages":null},"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
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
Briefings in Functional Genomics Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elae002
Chaorui Yan, Yanxu Zhu, Miao Chen, Kainan Yang, Feifei Cui, Quan Zou, Zilong Zhang
{"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":null,"pages":null},"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
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
Briefings in Functional Genomics 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
{"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":null,"pages":null},"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
Briefings in Functional Genomics 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
{"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":null,"pages":null},"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
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
Briefings in Functional Genomics Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elad040
Huixiang Peng, Jing Xu, Kangchen Liu, Fang Liu, Aidi Zhang, Xiujun Zhang
{"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":null,"pages":null},"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
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
Briefings in Functional Genomics 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
{"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":null,"pages":null},"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
DeepWalk-aware graph attention networks with CNN for circRNA-drug sensitivity association identification. 深度漫步感知图注意网络与 CNN 用于 circRNA-药物敏感性关联识别。
IF 2.5 3区 生物学
Briefings in Functional Genomics Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elad053
Guanghui Li, Youjun Li, Cheng Liang, Jiawei Luo
{"title":"DeepWalk-aware graph attention networks with CNN for circRNA-drug sensitivity association identification.","authors":"Guanghui Li, Youjun Li, Cheng Liang, Jiawei Luo","doi":"10.1093/bfgp/elad053","DOIUrl":"10.1093/bfgp/elad053","url":null,"abstract":"<p><p>Circular RNAs (circRNAs) are a class of noncoding RNA molecules that are widely found in cells. Recent studies have revealed the significant role played by circRNAs in human health and disease treatment. Several restrictions are encountered because forecasting prospective circRNAs and medication sensitivity connections through biological research is not only time-consuming and expensive but also incredibly ineffective. Consequently, the development of a novel computational method that enhances both the efficiency and accuracy of predicting the associations between circRNAs and drug sensitivities is urgently needed. Here, we present DGATCCDA, a computational method based on deep learning, for circRNA-drug sensitivity association identification. In DGATCCDA, we first construct multimodal networks from the original feature information of circRNAs and drugs. After that, we adopt DeepWalk-aware graph attention networks to sufficiently extract feature information from the multimodal networks to obtain the embedding representation of nodes. Specifically, we combine DeepWalk and graph attention network to form DeepWalk-aware graph attention networks, which can effectively capture the global and local information of graph structures. The features extracted from the multimodal networks are fused by layer attention, and eventually, the inner product approach is used to construct the association matrix of circRNAs and drugs for prediction. The ultimate experimental results obtained under 5-fold cross-validation settings show that the average area under the receiver operating characteristic curve value of DGATCCDA reaches 91.18%, which is better than those of the five current state-of-the-art calculation methods. We further guide a case study, and the excellent obtained results also show that DGATCCDA is an effective computational method for exploring latent circRNA-drug sensitivity associations.</p>","PeriodicalId":55323,"journal":{"name":"Briefings in Functional Genomics","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138806429","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
Personalized differential expression analysis in triple-negative breast cancer. 三阴性乳腺癌的个性化差异表达分析
IF 2.5 3区 生物学
Briefings in Functional Genomics Pub Date : 2024-07-19 DOI: 10.1093/bfgp/elad057
Hao Cai, Liangbo Chen, Shuxin Yang, Ronghong Jiang, You Guo, Ming He, Yun Luo, Guini Hong, Hongdong Li, Kai Song
{"title":"Personalized differential expression analysis in triple-negative breast cancer.","authors":"Hao Cai, Liangbo Chen, Shuxin Yang, Ronghong Jiang, You Guo, Ming He, Yun Luo, Guini Hong, Hongdong Li, Kai Song","doi":"10.1093/bfgp/elad057","DOIUrl":"10.1093/bfgp/elad057","url":null,"abstract":"<p><p>Identification of individual-level differentially expressed genes (DEGs) is a pre-step for the analysis of disease-specific biological mechanisms and precision medicine. Previous algorithms cannot balance accuracy and sufficient statistical power. Herein, RankCompV2, designed for identifying population-level DEGs based on relative expression orderings, was adjusted to identify individual-level DEGs. Furthermore, an optimized version of individual-level RankCompV2, named as RankCompV2.1, was designed based on the assumption that the rank positions of genes and relative rank differences of gene pairs would influence the identification of individual-level DEGs. In comparison to other individualized analysis algorithms, RankCompV2.1 performed better on statistical power, computational efficiency, and acquired coequal accuracy in both simulation and real paired cancer-normal data from ten cancer types. Besides, single sample GSEA and Gene Set Variation Analysis analysis showed that pathways enriched with up-regulated and down-regulated genes presented higher and lower enrichment scores, respectively. Furthermore, we identified 16 genes that were universally deregulated in 966 triple-negative breast cancer (TNBC) samples and interacted with Food and Drug Administration (FDA)-approved drugs or antineoplastic agents, indicating notable therapeutic targets for TNBC. In addition, we also identified genes with highly variable deregulation status and used these genes to cluster TNBC samples into three subgroups with different prognoses. The subgroup with the poorest outcome was characterized by down-regulated immune-regulated pathways, signal transduction pathways, and apoptosis-related pathways. Protein-protein interaction network analysis revealed that OAS family genes may be promising drug targets to activate tumor immunity in this subgroup. In conclusion, RankCompV2.1 is capable of identifying individual-level DEGs with high accuracy and statistical power, analyzing mechanisms of carcinogenesis and exploring therapeutic strategy.</p>","PeriodicalId":55323,"journal":{"name":"Briefings in Functional Genomics","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139405397","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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信