Pub Date : 2025-08-31DOI: 10.1007/s12031-025-02402-y
Sanya Kapoor, Valentina L. Kouznetsova, Santosh Kesari, Igor F. Tsigelny
Glioblastoma (GBM) represents one of the most aggressive brain tumors with a poor prognosis despite decades of research. Epigenetic regulation has emerged as a promising strategy for managing aggressive cancers, such as GBM, by modulating pro-tumorigenic gene expression. The role of pro-tumorigenic genes, such as oligodendrocyte transcription factor 2 (OLIG2), has been heavily associated with cancer progression and treatment resistance and is a potential target for GBM. The objective of this study is to analyze the effectiveness of various epigenetic regulators, including histone modifiers, DNA methylases, chromatin remodelers, and miRNAs, on OLIG2 expression, including the effectiveness of individual epigenetic regulators and their combinations. The effects of epigenetic regulators in GBM that are found in the literature were reviewed for their survival and co-expression with OLIG2. We found that KDM6B, BRG1, DNMT1, and HDAC2 were associated with significant co-expression with OLIG2 and decreased survival in GBM patients, reinforcing their suitability as targets. Additionally, miR-17-3p miRNAs associated with silencing OLIG2 as gene expression was downregulated in GBM. Additionally, this paper highlights the potential of combination therapies targeting multiple epigenetic pathways simultaneously. A kinase inhibitor (alisertib), together with JQ1, reduced the tumor growth of GBM cells in vivo more than either treatment alone, making combination therapies a promising solution.
{"title":"Epigenetic Regulation of OLIG2 in Glioblastoma: Mechanisms and Therapeutic Targets to Combat Treatment Resistance","authors":"Sanya Kapoor, Valentina L. Kouznetsova, Santosh Kesari, Igor F. Tsigelny","doi":"10.1007/s12031-025-02402-y","DOIUrl":"10.1007/s12031-025-02402-y","url":null,"abstract":"<div><p>Glioblastoma (GBM) represents one of the most aggressive brain tumors with a poor prognosis despite decades of research. Epigenetic regulation has emerged as a promising strategy for managing aggressive cancers, such as GBM, by modulating pro-tumorigenic gene expression. The role of pro-tumorigenic genes, such as oligodendrocyte transcription factor 2 (OLIG2), has been heavily associated with cancer progression and treatment resistance and is a potential target for GBM. The objective of this study is to analyze the effectiveness of various epigenetic regulators, including histone modifiers, DNA methylases, chromatin remodelers, and miRNAs, on OLIG2 expression, including the effectiveness of individual epigenetic regulators and their combinations. The effects of epigenetic regulators in GBM that are found in the literature were reviewed for their survival and co-expression with OLIG2. We found that KDM6B, BRG1, DNMT1, and HDAC2 were associated with significant co-expression with OLIG2 and decreased survival in GBM patients, reinforcing their suitability as targets. Additionally, miR-17-3p miRNAs associated with silencing OLIG2 as gene expression was downregulated in GBM. Additionally, this paper highlights the potential of combination therapies targeting multiple epigenetic pathways simultaneously. A kinase inhibitor (alisertib), together with JQ1, reduced the tumor growth of GBM cells in vivo more than either treatment alone, making combination therapies a promising solution.</p></div>","PeriodicalId":652,"journal":{"name":"Journal of Molecular Neuroscience","volume":"75 3","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144920497","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-08-30DOI: 10.1007/s12031-025-02314-x
Weina Wang, Chenglu Li, Long Chen, Yuquan Zhang
Premature ovarian failure (POF) accelerates ovarian aging, leading to menstrual irregularities, reduced fertility, and decreased estrogen levels. Current hormone replacement therapy (HRT) cannot reverse the aging effects, highlighting the need for more targeted treatments. Genome-wide association studies (GWAS) and protein quantitative trait loci (pQTL) analyses can identify genetic variants and protein level changes associated with POF. Mendelian randomization (MR) evaluates the causal relationships between genetic variants and POF. pQTL analysis was conducted using plasma proteomics data from 54,219 participants and baseline cohort data of 34,557 individuals of European ancestry from the UK Biobank. GWAS data comprising 542 POF cases and 218,970 controls were obtained from the FinnGen database. MR analysis utilized inverse variance weighted (IVW), MR Egger, weighted median, and weighted mode methods. Colocalization analysis was performed using the “coloc” R package, and pathway enrichment analysis was conducted using the “clusterProfiler” package. Additionally, reverse MR analysis, molecular docking predictions, and summary data-based Mendelian randomization (SMR) analysis were performed. Finally, based on the scRNA-seq data of POF, analyses such as cell type annotation, gene set scoring, and cell–cell communication were performed. MR analysis identified significant causal relationships between specific proteins (BSG, CCL23, CTSC, FAP, IGSF21, LCN15, LILRB2, MUC16, PTN, SPINK1, TNFRSF1B, TNFRSF8, TNXB, and YJU2) and POF. Colocalization analysis indicated that key proteins (BSG, CCL23, FAP, and TNXB) share causal variants with POF traits. SMR analysis confirmed TNXB as a risk factor for POF. Finally, using the scRNA-seq data of POF, the expression of key gene sets was used to evaluate the scoring of different cell populations. Cell–cell communication analysis identified multiple communication pathways between high-scoring cell populations and other cell groups. The expression trend of key proteins was further verified by western blot assay. These findings are preliminary and require significant validation before clinical application. Combining pQTL and GWAS data, MR and colocalization analyses identified key proteins and genetic variants associated with POF, providing deeper insights into POF mechanisms and potential therapeutic targets.
{"title":"Identification of Therapeutic Targets for Premature Ovarian Failure Through Mendelian Randomization and Colocalization Analysis Using Human Plasma Proteomics","authors":"Weina Wang, Chenglu Li, Long Chen, Yuquan Zhang","doi":"10.1007/s12031-025-02314-x","DOIUrl":"10.1007/s12031-025-02314-x","url":null,"abstract":"<div><p>Premature ovarian failure (POF) accelerates ovarian aging, leading to menstrual irregularities, reduced fertility, and decreased estrogen levels. Current hormone replacement therapy (HRT) cannot reverse the aging effects, highlighting the need for more targeted treatments. Genome-wide association studies (GWAS) and protein quantitative trait loci (pQTL) analyses can identify genetic variants and protein level changes associated with POF. Mendelian randomization (MR) evaluates the causal relationships between genetic variants and POF. pQTL analysis was conducted using plasma proteomics data from 54,219 participants and baseline cohort data of 34,557 individuals of European ancestry from the UK Biobank. GWAS data comprising 542 POF cases and 218,970 controls were obtained from the FinnGen database. MR analysis utilized inverse variance weighted (IVW), MR Egger, weighted median, and weighted mode methods. Colocalization analysis was performed using the “coloc” R package, and pathway enrichment analysis was conducted using the “clusterProfiler” package. Additionally, reverse MR analysis, molecular docking predictions, and summary data-based Mendelian randomization (SMR) analysis were performed. Finally, based on the scRNA-seq data of POF, analyses such as cell type annotation, gene set scoring, and cell–cell communication were performed. MR analysis identified significant causal relationships between specific proteins (BSG, CCL23, CTSC, FAP, IGSF21, LCN15, LILRB2, MUC16, PTN, SPINK1, TNFRSF1B, TNFRSF8, TNXB, and YJU2) and POF. Colocalization analysis indicated that key proteins (BSG, CCL23, FAP, and TNXB) share causal variants with POF traits. SMR analysis confirmed TNXB as a risk factor for POF. Finally, using the scRNA-seq data of POF, the expression of key gene sets was used to evaluate the scoring of different cell populations. Cell–cell communication analysis identified multiple communication pathways between high-scoring cell populations and other cell groups. The expression trend of key proteins was further verified by western blot assay. These findings are preliminary and require significant validation before clinical application. Combining pQTL and GWAS data, MR and colocalization analyses identified key proteins and genetic variants associated with POF, providing deeper insights into POF mechanisms and potential therapeutic targets.</p></div>","PeriodicalId":652,"journal":{"name":"Journal of Molecular Neuroscience","volume":"75 3","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-08-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144918658","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-08-27DOI: 10.1007/s12031-025-02393-w
Huilin Li, Musu Li, Yue Sun, Er Yu, Jiahe Pan, Yiwen Wu, Zixuan Lu, Hongmei Wo, Fang Shao, Dongfang You, Shaowen Tang, Yang Zhao, Juncheng Dai, Honggang Yi
Low-grade gliomas (LGGs) represent a complex and aggressive category of brain tumors. Despite recent advancements in molecular subtyping and characterization, the necessity to identify additional molecular subtypes and biomarkers remains. To delineate survival subtypes in LGG, we propose a deep learning (DL)-based multi-omics SurvivalNet (MOST) model. By integrating histological RNA-seq, miRNA-seq, and DNA methylation data obtained from The Cancer Genome Atlas (TCGA), we applied the MOST model to analyze data from 497 LGG patients. We employed consensus clustering to reveal heterogeneous subtypes, validated our findings using an internal validation set through a supervised classification algorithm, and further evaluated the robustness of our model in an independent external cohort. The DL-based MOST model identified two optimal patient subtypes with significant differences in survival (P = 3.07E − 16) and demonstrated a robust model fit (C = 0.92 ± 0.02). This multi-omics model was validated using external Chinese Glioma Genome Atlas (CCGA) datasets, including RNA-Seq (N = 497, C = 0.85), miRNA array (N = 89, C = 0.80), and DNA methylation (N = 89, C = 0.61). High-risk subcategories exhibited increased expression of the homeobox (HOX) family genes, regulation of cholesterol homeostasis, glycolysis, epithelial-mesenchymal transition pathway enrichment, and a high density of M2 macrophages. Our study utilized deep learning to identify multi-omics features associated with differential survival outcomes in patients with LGG. This work is anticipated to significantly enhance prognosis prediction for LGG due to its robustness within the cohorts.
低级别胶质瘤(LGGs)是一类复杂且具有侵袭性的脑肿瘤。尽管最近在分子亚型和表征方面取得了进展,但鉴定其他分子亚型和生物标志物的必要性仍然存在。为了描述LGG的生存亚型,我们提出了一个基于深度学习(DL)的多组学生存网络(MOST)模型。通过整合从癌症基因组图谱(TCGA)中获得的组织学RNA-seq、miRNA-seq和DNA甲基化数据,我们应用MOST模型分析了497例LGG患者的数据。我们采用共识聚类来揭示异质亚型,通过监督分类算法使用内部验证集验证我们的发现,并在独立的外部队列中进一步评估我们模型的稳健性。基于dl的MOST模型确定了两种生存率差异显著的最佳患者亚型(P = 3.07E−16),并证明了稳健的模型拟合(C = 0.92±0.02)。该多组学模型使用中国胶质瘤基因组图谱(CCGA)外部数据集进行验证,包括RNA-Seq (N = 497, C = 0.85)、miRNA阵列(N = 89, C = 0.80)和DNA甲基化(N = 89, C = 0.61)。高风险亚类表现出同源盒(HOX)家族基因的表达增加,胆固醇稳态调节,糖酵解,上皮-间质转化途径富集,M2巨噬细胞密度高。我们的研究利用深度学习来识别与LGG患者差异生存结果相关的多组学特征。由于其在队列中的稳健性,预计这项工作将显著提高对LGG的预后预测。
{"title":"Integration of Multi-omics Data Based on Deep Learning for Subtyping of Low-Grade Glioma","authors":"Huilin Li, Musu Li, Yue Sun, Er Yu, Jiahe Pan, Yiwen Wu, Zixuan Lu, Hongmei Wo, Fang Shao, Dongfang You, Shaowen Tang, Yang Zhao, Juncheng Dai, Honggang Yi","doi":"10.1007/s12031-025-02393-w","DOIUrl":"10.1007/s12031-025-02393-w","url":null,"abstract":"<div><p>Low-grade gliomas (LGGs) represent a complex and aggressive category of brain tumors. Despite recent advancements in molecular subtyping and characterization, the necessity to identify additional molecular subtypes and biomarkers remains. To delineate survival subtypes in LGG, we propose a deep learning (DL)-based multi-omics SurvivalNet (MOST) model. By integrating histological RNA-seq, miRNA-seq, and DNA methylation data obtained from The Cancer Genome Atlas (TCGA), we applied the MOST model to analyze data from 497 LGG patients. We employed consensus clustering to reveal heterogeneous subtypes, validated our findings using an internal validation set through a supervised classification algorithm, and further evaluated the robustness of our model in an independent external cohort. The DL-based MOST model identified two optimal patient subtypes with significant differences in survival (<i>P</i> = 3.07E − 16) and demonstrated a robust model fit (<i>C</i> = 0.92 ± 0.02). This multi-omics model was validated using external Chinese Glioma Genome Atlas (CCGA) datasets, including RNA-Seq (<i>N</i> = 497, <i>C</i> = 0.85), miRNA array (<i>N</i> = 89, <i>C</i> = 0.80), and DNA methylation (<i>N</i> = 89, <i>C</i> = 0.61). High-risk subcategories exhibited increased expression of the homeobox (<i>HOX</i>) family genes, regulation of cholesterol homeostasis, glycolysis, epithelial-mesenchymal transition pathway enrichment, and a high density of M2 macrophages. Our study utilized deep learning to identify multi-omics features associated with differential survival outcomes in patients with LGG. This work is anticipated to significantly enhance prognosis prediction for LGG due to its robustness within the cohorts.</p></div>","PeriodicalId":652,"journal":{"name":"Journal of Molecular Neuroscience","volume":"75 3","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144909616","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-08-20DOI: 10.1007/s12031-025-02400-0
Amod Kulkarni, Dhananjay B. Alagundagi, Mangesh Bhide, Prakash Patil
Background
Neuroinvasive pathogens are capable of breaching the blood–brain barrier (BBB), and causing central nervous system infections. Although the response of human brain microvascular endothelial cells (hBMECs), the forefront cells of BBB has been extensively studied, the roles of astrocytes and pericytes in modulating BBB integrity during infection remain less defined.
Aims
The study aims for a meta-analysis of RNA-seq data to compare the transcriptional response of hBMECs alone and in co-culture with astrocytes and pericytes (BBB-spheroids) following infection with Neisseria meningitidis and Borrelia bavariensis. Subsequently, identifying the pathogen-specific gene signatures that regulates the signalling pathways associated with infection and BBB disruption.
Methods
Unique and shared differentially expressed genes (DEGs) of hBMECs and BBB-spheroids were identified and analysed for functional enrichment using DAVID. Protein–protein interaction networks were constructed and analysed in Cytoscape using MCODE and cytoHubba to identify infection-related hub genes.
Results
A large proportion of DEGs were unique to each BBB model during infection, 49% in Neisseria and 66% in Borrelia infection, whereas only 4.9% were shared. hBMECs predominantly expressed defence-related genes, whereas BBB-spheroids expressed genes linked to barrier function. Notably, IFIH1, IFIT1, IFIT3, ISG15, MX1, OAS1, and RSAD2 were identified as regulators of the BBB’s transcriptomic response to infection.
Conclusions
The meta-analysis highlights distinct yet complementary roles of endothelial cells and the supporting pericytes and astrocytes in BBB regulation to bacterial invasion. The identified hub genes may serve as key regulators of infection-driven inflammation and form potential diagnostic or prognostic targets.