1
|
Sun M, Wang J, Wan S. Accurate identification of medulloblastoma subtypes from diverse data sources with severe batch effects by RaMBat. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.640010. [PMID: 40060540 PMCID: PMC11888263 DOI: 10.1101/2025.02.24.640010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
As the most common pediatric brain malignancy, medulloblastoma (MB) includes multiple distinct molecular subtypes characterized by clinical heterogeneity and genetic alterations. Accurate identification of MB subtypes is essential for downstream risk stratification and tailored therapeutic design. Existing MB subtyping approaches perform poorly due to limited cohorts and severe batch effects when integrating various MB data sources. To address these concerns, we propose a novel approach called RaMBat for accurate MB subtyping from diverse data sources with severe batch effects. Benchmarking tests based on 13 datasets with severe batch effects suggested that RaMBat achieved a median accuracy of 99%, significantly outperforming state-of-the-art MB subtyping approaches and conventional machine learning classifiers. RaMBat could efficiently deal with the batch effects and clearly separate subtypes of MB samples from diverse data sources. We believe RaMBat will bring direct positive impacts on downstream MB risk stratification and tailored treatment design.
Collapse
|
2
|
Wu C, Xie X, Yang X, Du M, Lin H, Huang J. Applications of gene pair methods in clinical research: advancing precision medicine. MOLECULAR BIOMEDICINE 2025; 6:22. [PMID: 40202606 PMCID: PMC11982013 DOI: 10.1186/s43556-025-00263-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
The rapid evolution of high-throughput sequencing technologies has revolutionized biomedical research, producing vast amounts of gene expression data that hold immense potential for biological discovery and clinical applications. Effectively mining these large-scale, high-dimensional data is crucial for facilitating disease detection, subtype differentiation, and understanding the molecular mechanisms underlying disease progression. However, the conventional paradigm of single-gene profiling, measuring absolute expression levels of individual genes, faces critical limitations in clinical implementation. These include vulnerability to batch effects and platform-dependent normalization requirements. In contrast, emerging approaches analyzing relative expression relationships between gene pairs demonstrate unique advantages. By focusing on binary comparisons of two genes' expression magnitudes, these methods inherently normalize experimental variations while capturing biologically stable interaction patterns. In this review, we systematically evaluate gene pair-based analytical frameworks. We classify eleven computational approaches into two fundamental categories: expression value-based methods quantifying differential expression patterns, and rank-based methods exploiting transcriptional ordering relationships. To bridge methodological development with practical implementation, we establish a reproducible analytical pipeline incorporating feature selection, classifier construction, and model evaluation modules using real-world benchmark datasets from pulmonary tuberculosis studies. These findings position gene pair analysis as a transformative paradigm for mining high-dimensional omics data, with direct implications for precision biomarker discovery and mechanistic studies of disease progression.
Collapse
Affiliation(s)
- Changchun Wu
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xueqin Xie
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xin Yang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Mengze Du
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China
| | - Hao Lin
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Jian Huang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| |
Collapse
|
3
|
Leng L, Wang H, Hu Y, Hu L. LINC02363: a potential biomarker for early diagnosis and treatment of sepsis. BMC Immunol 2025; 26:23. [PMID: 40089725 PMCID: PMC11909972 DOI: 10.1186/s12865-025-00702-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Sepsis remains a leading cause of global morbidity and mortality, yet early diagnosis is hindered by the limited specificity and sensitivity of current biomarkers. AIM The aim of this study was to identify lncRNAs that play a key role in sepsis and provide potential biomarkers for the diagnosis and treatment of sepsis. METHODS Transcriptomic data from sepsis patients were retrieved from the Chinese National Genebank (CNGBdb). Differential expression analysis identified 2,348 LncRNAs and 5,125 mRNAs (|FC|≥2, FDR < 0.05). Weighted gene co-expression network analysis (WGCNA) and meta-analysis were applied to screen core genes. Gene set enrichment analysis (GSEA) explored functional pathways, while single-cell sequencing and qPCR validated cellular localization and expression patterns. RESULTS WGCNA identified three key genes: LINC02363 (LncRNA), DYNLT1, and FCGR1B. Survival and meta-analyses revealed strong correlations between these genes and sepsis outcomes. GSEA highlighted LINC02363's involvement in "herpes simplex virus type 1 infection," "tuberculosis," and ribosome pathways. Single-cell sequencing showed FCGR1B's broad distribution across immune cells, while DYNLT1 localized predominantly in macrophages. qPCR confirmed significant upregulation of LINC02363 (p < 0.01), FCGR1B (p < 0.05), and DYNLT1 (p < 0.05) in sepsis patients compared to controls. CONCLUSION LINC02363 may serve as a new biomarker for the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Linghan Leng
- Department of Intensive Care Unit, Chengdu Fifth People's Hospital, Chengdu, People's Republic of China
| | - Hao Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, People's Republic of China
| | - Yingchun Hu
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China.
| | - Li Hu
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China.
| |
Collapse
|
4
|
Gu D, Wu Y, Ding Z, Dai Y. Biliary HMGB1 levels and biochemical indices in the assessment of acute obstructive septic cholangitis combined with septic shock. Clinics (Sao Paulo) 2025; 80:100611. [PMID: 40054422 PMCID: PMC11928834 DOI: 10.1016/j.clinsp.2025.100611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/23/2025] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Acute Obstructive Septic Cholangitis (AOSC) is a serious infectious disease of the biliary system. It is prone to develop into septic shock without prompt management. METHODS Bile was collected from 71 AOSC patients (45 AOSC without septic shock, 26 AOSC with septic shock) during biliary drainage and on days 1 and 3 postoperatively. The levels of High Mobility Group Protein 1 (HMGB1), Interleukin (IL)-1, IL-6, and Tumor Necrosis Factor alpha (TNF-α) were measured. The differences in the levels of biliary factors and their correlation with clinical biochemical indicators were assessed in the two groups. RESULTS HMGB1 gradually decreased in both groups in the postoperative period. Intraoperative levels of biliary HMGB1 were significantly higher in patients with AOSC with septic shock. TNF-α and HMGB1 decreased slowly in patients with AOSC with septic shock on postoperative days 1 and 3, and the levels of the factors decreased less. Biliary HMGB1 levels were negatively correlated with white blood cell count and positively correlated with blood urea nitrogen, blood creatinine, procalcitonin, and C-reactive protein. A bile HMGB1 level of 1108.3 pg/mL was the cut-off value to differentiate patients with AOSC with or without septic shock. CONCLUSION Biliary HMGB1 levels are elevated in patients with AOSC with septic shock and decrease slowly in the postoperative period. This suggests that HMGB1 is of considerable importance as a potential therapeutic target in the pathogenesis of septic shock in AOSC patients.
Collapse
Affiliation(s)
- DanYang Gu
- Department of General Surgery, Postgraduate Union Training Base of Xiangyang No 1 People's Hospital, School of Medicine, Wuhan University of Science and Technology, Xiangyang City, Hubei Province, China
| | - YuHao Wu
- Department of General Surgery, Xiangyang No 1 People's Hospital, Hubei University of Medicine, Xiangyang City, Hubei Province, China
| | - ZhengHua Ding
- Department of General Surgery, Xiangyang No 1 People's Hospital, Hubei University of Medicine, Xiangyang City, Hubei Province, China
| | - Yang Dai
- Department of General Surgery, Xiangyang No 1 People's Hospital, Hubei University of Medicine, Xiangyang City, Hubei Province, China.
| |
Collapse
|
5
|
Zheng X, Jin N, Wu Q, Zhang N, Wu H, Wang Y, Luo R, Liu T, Ding W, Geng Q, Cheng L. Less is more: relative rank is more informative than absolute abundance for compositional NGS data. Brief Funct Genomics 2025; 24:elae045. [PMID: 39568388 PMCID: PMC11735744 DOI: 10.1093/bfgp/elae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/24/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024] Open
Abstract
High-throughput gene expression data have been extensively generated and utilized in biological mechanism investigations, biomarker detection, disease diagnosis and prognosis. These applications encompass not only bulk transcriptome, but also single cell RNA-seq data. However, extracting reliable biological information from transcriptome data remains challenging due to the constrains of Compositional Data Analysis. Current data preprocessing methods, including dataset normalization and batch effect correction, are insufficient to address these issues and improve data quality for downstream analysis. Alternatively, qualification methods focusing on the relative order of gene expression (ROGER) are more informative than the quantification methods that rely on gene expression abundance. The Pairwise Analysis of Gene expression method is an enhancement of ROGER, designed for data integration in either sample space or feature space. In this review, we summarize the methods applied to transcriptome data analysis and discuss their potentials in predicting clinical outcomes.
Collapse
Affiliation(s)
- Xubin Zheng
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, Luohu District, Shenzhen 518020, China
- Health Data Science Center, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Luohu District, Shenzhen 518020, China
- School of Computing and Information Technology, Great Bay University, Dongguan 523000, Guangdong, China
| | - Nana Jin
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, Luohu District, Shenzhen 518020, China
- Health Data Science Center, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Luohu District, Shenzhen 518020, China
| | - Qiong Wu
- School of Basic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Ning Zhang
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, Luohu District, Shenzhen 518020, China
- Health Data Science Center, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Luohu District, Shenzhen 518020, China
| | - Haonan Wu
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, Luohu District, Shenzhen 518020, China
- Health Data Science Center, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Luohu District, Shenzhen 518020, China
| | - Yuanhao Wang
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, Luohu District, Shenzhen 518020, China
- Health Data Science Center, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Luohu District, Shenzhen 518020, China
| | - Rui Luo
- Department of Systems Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Tao Liu
- International Digital Economy Academy (IDEA), Futian District, Shenzhen 518020, China
| | - Wanfu Ding
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, Luohu District, Shenzhen 518020, China
| | - Qingshan Geng
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, Luohu District, Shenzhen 518020, China
| | - Lixin Cheng
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, Luohu District, Shenzhen 518020, China
- Health Data Science Center, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Luohu District, Shenzhen 518020, China
| |
Collapse
|
6
|
Meng D, Feng Y, Yuan K, Yu Z, Cao Q, Cheng L, Zheng X. scMMAE: masked cross-attention network for single-cell multimodal omics fusion to enhance unimodal omics. Brief Bioinform 2024; 26:bbaf010. [PMID: 39851073 PMCID: PMC11757910 DOI: 10.1093/bib/bbaf010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/18/2024] [Accepted: 01/08/2025] [Indexed: 01/25/2025] Open
Abstract
Multimodal omics provide deeper insight into the biological processes and cellular functions, especially transcriptomics and proteomics. Computational methods have been proposed for the integration of single-cell multimodal omics of transcriptomics and proteomics. However, existing methods primarily concentrate on the alignment of different omics, overlooking the unique information inherent in each omics type. Moreover, as the majority of single-cell cohorts only encompass one omics, it becomes critical to transfer the knowledge learnt from multimodal omics to enhance unimodal omics analysis. Therefore, we proposed a novel framework that leverages masked autoencoder with cross-attention mechanism, called scMMAE (single-cell multimodal masked autoencoder), to fuse multimodal omics and enhance unimodal omics analysis. scMMAE simultaneously captures both the shared features and the distinctive information of two single-cell omics modalities and transfers the knowledge to enhance single-cell transcriptome data. Comparative evaluations against benchmarking methods across various cohorts revealed a notable improvement, with an increase of up to 21% in the adjusted Rand index and up to 12% in normalized mutual information in the context of multimodal fusion. In the realm of unimodal omics, scMMAE demonstrated an overall enhancement of approximately 20% in the adjusted Rand index and nearly 10% in normalized mutual information. Other nine metrics, including the Fowlkes-Mallows index and silhouette coefficient, further underscored the high performance of scMMAE. Significantly, scMMAE exhibits an elevated level of proficiency in distinguishing between different cell types, particularly on CD4 and CD8 T cells. Availability and implementation: scMMAE source code at https://github.com/DM0815/scMMAE/.
Collapse
Affiliation(s)
- Dian Meng
- Guangdong Provincial Key Laboratory of Mathematical and Neural Dynamical Systems, Great Bay University, No. 16 Daxue Rd, Songshanhu District, Dongguan, Guangdong, 523000, China
- School of Computing and Information Technology, Great Bay University, No. 16 Daxue Rd, Songshanhu District, Dongguan, Guangdong, 523000, China
| | - Yu Feng
- BGI-Research, Shenzhen, 518083, China
| | - Kaishen Yuan
- Guangdong Provincial Key Laboratory of Mathematical and Neural Dynamical Systems, Great Bay University, No. 16 Daxue Rd, Songshanhu District, Dongguan, Guangdong, 523000, China
| | - Zitong Yu
- Guangdong Provincial Key Laboratory of Mathematical and Neural Dynamical Systems, Great Bay University, No. 16 Daxue Rd, Songshanhu District, Dongguan, Guangdong, 523000, China
- School of Computing and Information Technology, Great Bay University, No. 16 Daxue Rd, Songshanhu District, Dongguan, Guangdong, 523000, China
| | - Qin Cao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, 518057, China
| | - Lixin Cheng
- Shenzhen People’s Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, China
| | - Xubin Zheng
- Guangdong Provincial Key Laboratory of Mathematical and Neural Dynamical Systems, Great Bay University, No. 16 Daxue Rd, Songshanhu District, Dongguan, Guangdong, 523000, China
- School of Computing and Information Technology, Great Bay University, No. 16 Daxue Rd, Songshanhu District, Dongguan, Guangdong, 523000, China
| |
Collapse
|
7
|
Jin N, Nan C, Li W, Lin P, Xin Y, Wang J, Chen Y, Wang Y, Yu K, Wang C, Chen C, Geng Q, Cheng L. PAGE-based transfer learning from single-cell to bulk sequencing enhances model generalization for sepsis diagnosis. Brief Bioinform 2024; 26:bbae661. [PMID: 39710434 PMCID: PMC11962595 DOI: 10.1093/bib/bbae661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/04/2024] [Accepted: 12/12/2024] [Indexed: 12/24/2024] Open
Abstract
Sepsis, caused by infections, sparks a dangerous bodily response. The transcriptional expression patterns of host responses aid in the diagnosis of sepsis, but the challenge lies in their limited generalization capabilities. To facilitate sepsis diagnosis, we present an updated version of single-cell Pair-wise Analysis of Gene Expression (scPAGE) using transfer learning method, scPAGE2, dedicated to data fusion between single-cell and bulk transcriptome. Compared to scPAGE, the upgrade to scPAGE2 featured ameliorated Differentially Expressed Gene Pairs (DEPs) for pretraining a model in single-cell transcriptome and retrained it using bulk transcriptome data to construct a sepsis diagnostic model, which effectively transferred cell-layer information from single-cell to bulk transcriptome. Seven datasets across three transcriptome platforms and fluorescence-activated cell sorting (FACS) were used for performance validation. The model involved four DEPs, showing robust performance across next-generation sequencing and microarray platforms, surpassing state-of-the-art models with an average AUROC of 0.947 and an average AUPRC of 0.987. Analysis of scRNA-seq data reveals higher cell proportions with JAM3-PIK3AP1 expression in sepsis monocytes, decreased ARG1-CCR7 in B and T cells. Elevated IRF6-HP in sepsis monocytes confirmed by both scRNA-seq and an independent cohort using FACS. Both the superior performance of the model and the in vitro validation of IRF6-HP in monocytes emphasize that scPAGE2 is effective and robust in the construction of sepsis diagnostic model. We additionally applied scPAGE2 to acute myeloid leukemia and demonstrated its superior classification performance. Overall, we provided a strategy to improve the generalizability of classification model that can be adapted to a broad range of clinical prediction scenarios.
Collapse
Affiliation(s)
- Nana Jin
- Guangdong Provincial Clinical Research Center for Geriatrics; Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, 1017 Dongmen Rd N, Luohu District, Shenzhen 518020, China
- Post-doctoral Scientific Research Station of Basic Medicine, Jinan University, 601 Huangpu Blvd W, Tianhe District, Guangzhou 510632, China
| | - Chuanchuan Nan
- Department of Critical Care Medicine, Shenzhen People’s Hospital, the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, 1017 Dongmen Rd N, Luohu District, Shenzhen 518020, China
| | - Wanyang Li
- Guangdong Provincial Clinical Research Center for Geriatrics; Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, 1017 Dongmen Rd N, Luohu District, Shenzhen 518020, China
| | - Peijing Lin
- Guangdong Provincial Clinical Research Center for Geriatrics; Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, 1017 Dongmen Rd N, Luohu District, Shenzhen 518020, China
| | - Yu Xin
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang 150001, China
| | - Jun Wang
- Bioinformatics Centre, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, 2200 København, Denmark
| | - Yuelong Chen
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China
| | - Yuanhao Wang
- Guangdong Provincial Clinical Research Center for Geriatrics; Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, 1017 Dongmen Rd N, Luohu District, Shenzhen 518020, China
| | - Kaijiang Yu
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang 150001, China
| | - Changsong Wang
- Department of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang 150001, China
| | - Chunbo Chen
- Department of Critical Care Medicine, Shenzhen People’s Hospital, the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, 1017 Dongmen Rd N, Luohu District, Shenzhen 518020, China
| | - Qingshan Geng
- Guangdong Provincial Clinical Research Center for Geriatrics; Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, 1017 Dongmen Rd N, Luohu District, Shenzhen 518020, China
| | - Lixin Cheng
- Guangdong Provincial Clinical Research Center for Geriatrics; Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, 1017 Dongmen Rd N, Luohu District, Shenzhen 518020, China
- Department of Critical Care Medicine, Shenzhen People’s Hospital, the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, 1017 Dongmen Rd N, Luohu District, Shenzhen 518020, China
- Health Data Science Center, Shenzhen People's Hospital, 1017 Dongmen Rd N, Luohu District, Shenzhen 518020, China
| |
Collapse
|
8
|
Li N, Hu Z, Zhang N, Liang Y, Feng Y, Ding W, Cheng L, Zheng Y. Pairwise analysis of gene expression for oral squamous cell carcinoma via a large-scale transcriptome integration. J Cell Mol Med 2024; 28:e70153. [PMID: 39470584 PMCID: PMC11520439 DOI: 10.1111/jcmm.70153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/09/2024] [Accepted: 10/01/2024] [Indexed: 10/30/2024] Open
Abstract
Among all cancers occurring in the head and neck region, oral squamous cell carcinoma (OSCC) is the most common oral malignant tumours characterized by its aggressiveness and metastasis. The development of transcriptomics technology has greatly facilitated the diagnosis of various cancers. However, identifying genetic biomarkers is limited by data from a single batch of OSCC samples, and integrating analysis across different platforms remains a great challenge. In this study, we integrated five OSCC transcriptome datasets using an innovative strategy capable of mitigating batch effect, and extracting information from different datasets based on changes in the relative expression of gene pairs. By leveraging a machine learning method, we developed a prediction model including 27 differential gene pairs (DGPs) to discriminate OSCC from control samples, achieving an area under the receiver operating characteristic curve (AUC) of 0.8987 for the training set. Moreover, the model demonstrated commendable performance in four external validation sets, with AUCs of 0.9926, 0.9688, 0.8052 and 0.8565, respectively. Subsequently, a prognostic model was constructed based on six key gene pairs through univariate and multivariate Cox regression analysis. The AUCs of the model at 1-year and 3-year overall survival time prediction were 0.717 and 0.779 in an independent dataset. Our result demonstrates the effectiveness of this new method of integrating data and identifying DGPs. Using DGPs can significantly improve the performance of both diagnostic and prognostic models.
Collapse
Affiliation(s)
- Nan Li
- Department of StomatologyShenzhen People's Hospital (Second Clinical Medical School of Jinan University; First Affiliated Hospital of Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Zunkai Hu
- Department of Critical Care MedicineShenzhen People's Hospital (Second Clinical Medical School of Jinan University; First Affiliated Hospital of Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Ning Zhang
- Department of Critical Care MedicineShenzhen People's Hospital (Second Clinical Medical School of Jinan University; First Affiliated Hospital of Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Yining Liang
- School of MedicineSouthern University of Science and TechnologyShenzhenGuangdongChina
| | - Yating Feng
- School of MedicineSouthern University of Science and TechnologyShenzhenGuangdongChina
| | - Wanfu Ding
- Department of Information and TechnologyShenzhen People's HospitalShenzhenGuangdongChina
| | - Lixin Cheng
- Department of Critical Care MedicineShenzhen People's Hospital (Second Clinical Medical School of Jinan University; First Affiliated Hospital of Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Yuyan Zheng
- Department of StomatologyShenzhen People's Hospital (Second Clinical Medical School of Jinan University; First Affiliated Hospital of Southern University of Science and Technology)ShenzhenGuangdongChina
| |
Collapse
|
9
|
Zheng X, Meng D, Chen D, Wong WK, To KH, Zhu L, Wu J, Liang Y, Leung KS, Wong MH, Cheng L. scCaT: An explainable capsulating architecture for sepsis diagnosis transferring from single-cell RNA sequencing. PLoS Comput Biol 2024; 20:e1012083. [PMID: 39432561 PMCID: PMC11527285 DOI: 10.1371/journal.pcbi.1012083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 10/31/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
Sepsis is a life-threatening condition characterized by an exaggerated immune response to pathogens, leading to organ damage and high mortality rates in the intensive care unit. Although deep learning has achieved impressive performance on prediction and classification tasks in medicine, it requires large amounts of data and lacks explainability, which hinder its application to sepsis diagnosis. We introduce a deep learning framework, called scCaT, which blends the capsulating architecture with Transformer to develop a sepsis diagnostic model using single-cell RNA sequencing data and transfers it to bulk RNA data. The capsulating architecture effectively groups genes into capsules based on biological functions, which provides explainability in encoding gene expressions. The Transformer serves as a decoder to classify sepsis patients and controls. Our model achieves high accuracy with an AUROC of 0.93 on the single-cell test set and an average AUROC of 0.98 on seven bulk RNA cohorts. Additionally, the capsules can recognize different cell types and distinguish sepsis from control samples based on their biological pathways. This study presents a novel approach for learning gene modules and transferring the model to other data types, offering potential benefits in diagnosing rare diseases with limited subjects.
Collapse
Affiliation(s)
- Xubin Zheng
- School of Computing and Information Technology, Great Bay University, Guangdong, China
- Department of Critical Care Medicine, Shenzhen People’s Hospital, the First Affiliated Hospital of Southern University of Science and Technology, the Second Clinical Medicine College of Jinan University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Mathematical and Neural Dynamical Systems, Guangzhou, China
| | - Dian Meng
- School of Computing and Information Technology, Great Bay University, Guangdong, China
| | - Duo Chen
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Department of Nosocomial Infection Prevention and Control, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Wan-Ki Wong
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ka-Ho To
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Lei Zhu
- Department of Computer Science, City University of Hong Kong, Kowloon, Hong Kong
| | | | - Yining Liang
- Southern University of Science and Technology, Guangdong, China
| | - Kwong-Sak Leung
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong
- Department of Applied Data Science, Shue Yan University, North Point, Hong Kong
| | - Man-Hon Wong
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Lixin Cheng
- Department of Critical Care Medicine, Shenzhen People’s Hospital, the First Affiliated Hospital of Southern University of Science and Technology, the Second Clinical Medicine College of Jinan University, Shenzhen, China
| |
Collapse
|
10
|
Wei Y, Bai C, Xu S, Cui M, Wang R, Wu M. Diagnostic and Predictive Value of LncRNA MCM3AP-AS1 in Sepsis and Its Regulatory Role in Sepsis-Induced Myocardial Dysfunction. Cardiovasc Toxicol 2024; 24:1125-1138. [PMID: 39085530 DOI: 10.1007/s12012-024-09903-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
The present study focused on exploring the clinical value and molecular mechanism of LncRNA MCM3AP antisense RNA 1 (MCM3AP-AS1) in sepsis and sepsis-induced myocardial dysfunction (SIMD). 122 sepsis patients and 90 healthy were included. Sepsis patients were categorized into SIMD and non-MD. The expression levels of MCM3AP-AS1 and miRNA were examined using RT-qPCR. Diagnostic value of MCM3AP-AS1 in sepsis assessed by ROC curves. Logistic regression to explore risk factors influencing the occurrence of SIMD. Cardiomyocytes were induced by LPS to construct cell models in vitro. CCK-8, flow cytometry, and ELISA to analyze cell viability, apoptosis, and inflammation levels. Serum MCM3AP-AS1 was upregulated in patients with sepsis. The sensitivity and specificity of MCM3AP-AS1 were 75.41% and 93.33%, for recognizing sepsis from healthy controls. Additionally, elevated MCM3AP-AS1 is a risk factor for SIMD and can predict SIMD development. Compared with the LPS-induced cardiomyocytes, inhibition of MCM3AP-AS1 significantly attenuated LPS-induced apoptosis and inflammation; however, this attenuation was partially reversed by lowered miR-28-5p, but this reversal was partially eliminated by CASP2. MCM3AP-AS1 may be a novel diagnostic biomarker for sepsis and can predict the development of SIMD. MCM3AP-AS1 probably participated in SIMD progression by regulating cardiomyocyte inflammation and apoptosis through the target miR-28-5p/CASP2 axis.
Collapse
Affiliation(s)
- Yunwei Wei
- Department of Anesthesiology, Women's Health Center of Shanxi, Children's Hospital of Shanxi, Taiyuan, Shanxi, China
| | - Cui Bai
- Department of Critical Care Medicine, Chongqing Yubei District People's Hospital, Chongqing, 401120, China
| | - Shuying Xu
- Department of Emergency, Binzhou Medical University Hospital, 661 Huanghe 2nd Road, Bincheng District, Binzhou, 256600, Shandong, China
| | - Mingli Cui
- Department of Cardiovascular Medicine, Binzhou Medical University Hospital, Binzhou, 256600, Shandong, China
| | - Ruixia Wang
- Department of Emergency, Binzhou Medical University Hospital, 661 Huanghe 2nd Road, Bincheng District, Binzhou, 256600, Shandong, China.
| | - Meizhen Wu
- Department of Intensive Care Unit, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, 3 Xincun Road, Xinghualing District, Taiyuan, 030013, Shanxi, China.
| |
Collapse
|
11
|
Elkahwagy DMAS, Kiriacos CJ, Mansour M. Logistic regression and other statistical tools in diagnostic biomarker studies. Clin Transl Oncol 2024; 26:2172-2180. [PMID: 38530558 PMCID: PMC11333519 DOI: 10.1007/s12094-024-03413-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/16/2024] [Indexed: 03/28/2024]
Abstract
A biomarker is a measured indicator of a variety of processes, and is often used as a clinical tool for the diagnosis of diseases. While the developmental process of biomarkers from lab to clinic is complex, initial exploratory stages often focus on characterizing the potential of biomarkers through utilizing various statistical methods that can be used to assess their discriminatory performance, establish an appropriate cut-off that transforms continuous data to apt binary responses of confirming or excluding a diagnosis, or establish a robust association when tested against confounders. This review aims to provide a gentle introduction to the most common tools found in diagnostic biomarker studies used to assess the performance of biomarkers with an emphasis on logistic regression.
Collapse
Affiliation(s)
| | - Caroline Joseph Kiriacos
- Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, 11835, Egypt
| | - Manar Mansour
- Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, 11835, Egypt
| |
Collapse
|
12
|
Xie J, Zheng X, Yan J, Li Q, Jin N, Wang S, Zhao P, Li S, Ding W, Cheng L, Geng Q. Deep learning model to discriminate diverse infection types based on pairwise analysis of host gene expression. iScience 2024; 27:109908. [PMID: 38827397 PMCID: PMC11141160 DOI: 10.1016/j.isci.2024.109908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 03/01/2024] [Accepted: 05/03/2024] [Indexed: 06/04/2024] Open
Abstract
Accurate detection of pathogens, particularly distinguishing between Gram-positive and Gram-negative bacteria, could improve disease treatment. Host gene expression can capture the immune system's response to infections caused by various pathogens. Here, we present a deep neural network model, bvnGPS2, which incorporates the attention mechanism based on a large-scale integrated host transcriptome dataset to precisely identify Gram-positive and Gram-negative bacterial infections as well as viral infections. We performed analysis of 4,949 blood samples across 40 cohorts from 10 countries using our previously designed omics data integration method, iPAGE, to select discriminant gene pairs and train the bvnGPS2. The performance of the model was evaluated on six independent cohorts comprising 374 samples. Overall, our deep neural network model shows robust capability to accurately identify specific infections, paving the way for precise medicine strategies in infection treatment and potentially also for identifying subtypes of other diseases.
Collapse
Affiliation(s)
- Jize Xie
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University), Shenzhen 518020, China
- John Hopcroft Center for Computer Science, Shanghai Jiao Tong University, Shanghai, China
| | - Xubin Zheng
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University), Shenzhen 518020, China
- Great Bay University, Dongguan, China
| | - Jianlong Yan
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University), Shenzhen 518020, China
| | - Qizhi Li
- John Hopcroft Center for Computer Science, Shanghai Jiao Tong University, Shanghai, China
| | - Nana Jin
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University), Shenzhen 518020, China
- Health Data Science Center, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Shuojia Wang
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University), Shenzhen 518020, China
| | - Pengfei Zhao
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University), Shenzhen 518020, China
| | - Shuai Li
- John Hopcroft Center for Computer Science, Shanghai Jiao Tong University, Shanghai, China
| | - Wanfu Ding
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University), Shenzhen 518020, China
| | - Lixin Cheng
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University), Shenzhen 518020, China
- Health Data Science Center, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Qingshan Geng
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University), Shenzhen 518020, China
| |
Collapse
|
13
|
Weng S, Fu H, Xu S, Li J. Validating core therapeutic targets for osteoporosis treatment based on integrating network pharmacology and informatics. SLAS Technol 2024; 29:100122. [PMID: 38364892 DOI: 10.1016/j.slast.2024.100122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 01/24/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024]
Abstract
OBJECTIVE Our goal was to find metabolism-related lncRNAs that were associated with osteoporosis (OP) and construct a model for predicting OP progression using these lncRNAs. METHODS The GEO database was employed to obtain gene expression profiles. The WGCNA technique and differential expression analysis were used to identify hypoxia-related lncRNAs. A Lasso regression model was applied to select 25 hypoxia-related genes, from which a classification model was created. Its robust classification performance was confirmed with an area under the ROC curve close to 1, as verified on the validation set. Concurrently, we constructed a ceRNA network based on these genes to unveil potential regulatory processes. Biologically active compounds of STZYD were identified using the Traditional Chinese Medicine System Pharmacology Database and Analysis Platform (TCMSP) database. BATMAN was used to identify its targets, and we obtained OP-related genes from Malacards and DisGeNET, followed by identifying intersection genes with metabolism-related genes. A pharmacological network was then constructed based on the intersecting genes. The pharmacological network was further integrated with the ceRNA network, resulting in the creation of a comprehensive network that encompasses herb-active components, pathways, lncRNAs, miRNAs, and targets. Expression levels of hypoxia-related lncRNAs in mononuclear cells isolated from peripheral blood of OP and normal patients were subsequently validated using quantitative real-time PCR (qRT-PCR). Protein levels of RUNX2 were determined through a western blot assay. RESULTS CBFB, GLO1, NFKB2 and PIK3CA were identified as central therapeutic targets, and ADD3-AS1, DTX2P1-UPK3BP1-PMS2P11, TTTY1B, ZNNT1 and LINC00623 were identified as core lncRNAs. CONCLUSIONS Our work uncovers a possible therapeutic mechanism for STZYD, providing a potential therapeutic target for OP. In addition, a prediction model of metabolism-related lncRNAs of OP progression was constructed to provide a reference for the diagnosis of OP patients.
Collapse
Affiliation(s)
- Shiyang Weng
- Department of Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Huichao Fu
- Department of Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Shengxiang Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang 310009, China.
| | - Jieruo Li
- Department of Sport Medicine, Institute of Orthopedics Diseases and Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
14
|
Zhao P, Meng D, Hu Z, Liang Y, Feng Y, Sun T, Cheng L, Zheng X, Li H. Intra-sample reversed pairs based on differentially ranked genes reveal biosignature for ovarian cancer. Comput Biol Med 2024; 172:108208. [PMID: 38484696 DOI: 10.1016/j.compbiomed.2024.108208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/08/2024] [Accepted: 02/25/2024] [Indexed: 03/26/2024]
Abstract
Ovarian cancer, a major gynecological malignancy, often remains undetected until advanced stages, necessitating more effective early screening methods. Existing biomarker based on differential genes often suffers from variations in clinical practice. To overcome the limitations of absolute gene expression values including batch effects and biological heterogeneity, we introduced a pairwise biosignature leveraging intra-sample differentially ranked genes (DRGs) and machine learning for ovarian cancer detection across diverse cohorts. We analyzed ten cohorts comprising 872 samples with 796 ovarian cancer and 76 normal. Our method, DRGpair, involves three stages: intra-sample ranking differential analysis, reversed gene pair analysis, and iterative LASSO regression. We identified four DRG pairs, demonstrating superior diagnostic performance compared to current state-of-the-art biomarkers and differentially expressed genes in seven independent cohorts. This rank-based approach not only reduced computational complexity but also enhanced the specificity and effectiveness of biomarkers, revealing DRGs as promising candidates for ovarian cancer detection and offering a scalable model adaptable to varying cohort characteristics.
Collapse
Affiliation(s)
- Pengfei Zhao
- School of Medicine, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China
| | - Dian Meng
- School of Computing and Information Technology, Great Bay University, Guangdong, China
| | - Zunkai Hu
- School of Medicine, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China
| | - Yining Liang
- School of Medicine, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China
| | - Yating Feng
- School of Medicine, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China
| | - Tongjie Sun
- School of Medicine, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China
| | - Lixin Cheng
- School of Medicine, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China
| | - Xubin Zheng
- School of Computing and Information Technology, Great Bay University, Guangdong, China; Great Bay Institute for Advanced Study, Guangdong, China
| | - Haili Li
- School of Medicine, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China.
| |
Collapse
|
15
|
Zhang N, Yang F, Zhao P, Jin N, Wu H, Liu T, Geng Q, Yang X, Cheng L. MrGPS: an m6A-related gene pair signature to predict the prognosis and immunological impact of glioma patients. Brief Bioinform 2023; 25:bbad498. [PMID: 38171932 PMCID: PMC10782913 DOI: 10.1093/bib/bbad498] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 11/17/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024] Open
Abstract
N6-methyladenosine (m6A) RNA methylation is the predominant epigenetic modification for mRNAs that regulates various cancer-related pathways. However, the prognostic significance of m6A modification regulators remains unclear in glioma. By integrating the TCGA lower-grade glioma (LGG) and glioblastoma multiforme (GBM) gene expression data, we demonstrated that both the m6A regulators and m6A-target genes were associated with glioma prognosis and activated various cancer-related pathways. Then, we paired m6A regulators and their target genes as m6A-related gene pairs (MGPs) using the iPAGE algorithm, among which 122 MGPs were significantly reversed in expression between LGG and GBM. Subsequently, we employed LASSO Cox regression analysis to construct an MGP signature (MrGPS) to evaluate glioma prognosis. MrGPS was independently validated in CGGA and GEO glioma cohorts with high accuracy in predicting overall survival. The average area under the receiver operating characteristic curve (AUC) at 1-, 3- and 5-year intervals were 0.752, 0.853 and 0.831, respectively. Combining clinical factors of age and radiotherapy, the AUC of MrGPS was much improved to around 0.90. Furthermore, CIBERSORT and TIDE algorithms revealed that MrGPS is indicative for the immune infiltration level and the response to immune checkpoint inhibitor therapy in glioma patients. In conclusion, our study demonstrated that m6A methylation is a prognostic factor for glioma and the developed prognostic model MrGPS holds potential as a valuable tool for enhancing patient management and facilitating accurate prognosis assessment in cases of glioma.
Collapse
Affiliation(s)
- Ning Zhang
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
- The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University
- Neuroscience Center, Shantou University Medical College, Shantou, China
| | - Fengxia Yang
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
- The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University
- Neuroscience Center, Shantou University Medical College, Shantou, China
| | - Pengfei Zhao
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
- The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University
| | - Nana Jin
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
- The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University
| | - Haonan Wu
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
- The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University
| | - Tao Liu
- International Digital Economy Academy, Shenzhen, China
| | - Qingshan Geng
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
- The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University
| | - Xiaojun Yang
- Neuroscience Center, Shantou University Medical College, Shantou, China
| | - Lixin Cheng
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
- The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University
| |
Collapse
|
16
|
Liu X, Hong C, Jiang Y, Li W, Chen Y, Ma Y, Zhao P, Li T, Chen H, Liu X, Cheng L. Co-expression module analysis reveals high expression homogeneity for both coding and non-coding genes in sepsis. BMC Genomics 2023; 24:418. [PMID: 37488493 PMCID: PMC10364430 DOI: 10.1186/s12864-023-09460-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/16/2023] [Indexed: 07/26/2023] Open
Abstract
Sepsis is a life-threatening condition characterized by a harmful host response to infection with organ dysfunction. Annually about 20 million people are dead owing to sepsis and its mortality rates is as high as 20%. However, no studies have been carried out to investigate sepsis from the system biology point of view, as previous research predominantly focused on individual genes without considering their interactions and associations. Here, we conducted a comprehensive exploration of genome-wide expression alterations in both mRNAs and long non-coding RNAs (lncRNAs) in sepsis, using six microarray datasets. Co-expression networks were conducted to identify mRNA and lncRNA modules, respectively. Comparing these sepsis modules with normal modules, we observed a homogeneous expression pattern within the mRNA/lncRNA members, with the majority of them displaying consistent expression direction. Moreover, we identified consistent modules across diverse datasets, consisting of 20 common mRNA members and two lncRNAs, namely CHRM3-AS2 and PRKCQ-AS1, which are potential regulators of sepsis. Our results reveal that the up-regulated common mRNAs are mainly involved in the processes of neutrophil mediated immunity, while the down-regulated mRNAs and lncRNAs are significantly overrepresented in T-cell mediated immunity functions. This study sheds light on the co-expression patterns of mRNAs and lncRNAs in sepsis, providing a novel perspective and insight into the sepsis transcriptome, which may facilitate the exploration of candidate therapeutic targets and molecular biomarkers for sepsis.
Collapse
Affiliation(s)
- Xiaojun Liu
- Department of Critical Care, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, 518020, China
| | - Chengying Hong
- Department of Critical Care, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, 518020, China
| | - Yichun Jiang
- Department of Critical Care, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, 518020, China
| | - Wei Li
- Department of Critical Care, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, 518020, China
| | - Youlian Chen
- Department of Critical Care, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, 518020, China
| | - Yonghui Ma
- Department of Critical Care, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, 518020, China
| | - Pengfei Zhao
- Department of Critical Care, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, 518020, China
| | - Tiyuan Li
- Department of Critical Care, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, 518020, China
| | - Huaisheng Chen
- Department of Critical Care, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, 518020, China.
| | - Xueyan Liu
- Department of Critical Care, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, 518020, China.
| | - Lixin Cheng
- Department of Critical Care, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, 518020, China.
| |
Collapse
|
17
|
Pelaia TM, Shojaei M, McLean AS. The Role of Transcriptomics in Redefining Critical Illness. Crit Care 2023; 27:89. [PMID: 36941625 PMCID: PMC10027592 DOI: 10.1186/s13054-023-04364-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
This article is one of ten reviews selected from the Annual Update in Intensive Care and Emergency Medicine 2023. Other selected articles can be found online at https://www.biomedcentral.com/collections/annualupdate2023 . Further information about the Annual Update in Intensive Care and Emergency Medicine is available from https://link.springer.com/bookseries/8901 .
Collapse
Affiliation(s)
- Tiana M Pelaia
- Department of Intensive Care Medicine, Nepean Hospital, Kingswood, NSW, Australia.
| | - Maryam Shojaei
- Department of Intensive Care Medicine, Nepean Hospital, Kingswood, NSW, Australia
- Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Anthony S McLean
- Department of Intensive Care Medicine, Nepean Hospital, Kingswood, NSW, Australia
| |
Collapse
|
18
|
Li Q, Zheng X, Xie J, Wang R, Li M, Wong MH, Leung KS, Li S, Geng Q, Cheng L. bvnGPS: a generalizable diagnostic model for acute bacterial and viral infection using integrative host transcriptomics and pretrained neural networks. Bioinformatics 2023; 39:7066914. [PMID: 36857587 PMCID: PMC9997702 DOI: 10.1093/bioinformatics/btad109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/05/2023] [Accepted: 02/28/2023] [Indexed: 03/03/2023] Open
Abstract
MOTIVATION The confusion of acute inflammation infected by virus and bacteria or noninfectious inflammation will lead to missing the best therapy occasion resulting in poor prognoses. The diagnostic model based on host gene expression has been widely used to diagnose acute infections, but the clinical usage was hindered by the capability across different samples and cohorts due to the small sample size for signature training and discovery. RESULTS Here, we construct a large-scale dataset integrating multiple host transcriptomic data and analyze it using a sophisticated strategy which removes batch effect and extracts the common information from different cohorts based on the relative expression alteration of gene pairs. We assemble 2680 samples across 16 cohorts and separately build gene pair signature (GPS) for bacterial, viral, and noninfected patients. The three GPSs are further assembled into an antibiotic decision model (bacterial-viral-noninfected GPS, bvnGPS) using multiclass neural networks, which is able to determine whether a patient is bacterial infected, viral infected, or noninfected. bvnGPS can distinguish bacterial infection with area under the receiver operating characteristic curve (AUC) of 0.953 (95% confidence interval, 0.948-0.958) and viral infection with AUC of 0.956 (0.951-0.961) in the test set (N = 760). In the validation set (N = 147), bvnGPS also shows strong performance by attaining an AUC of 0.988 (0.978-0.998) on bacterial-versus-other and an AUC of 0.994 (0.984-1.000) on viral-versus-other. bvnGPS has the potential to be used in clinical practice and the proposed procedure provides insight into data integration, feature selection and multiclass classification for host transcriptomics data. AVAILABILITY AND IMPLEMENTATION The codes implementing bvnGPS are available at https://github.com/Ritchiegit/bvnGPS. The construction of iPAGE algorithm and the training of neural network was conducted on Python 3.7 with Scikit-learn 0.24.1 and PyTorch 1.7. The visualization of the results was implemented on R 4.2, Python 3.7, and Matplotlib 3.3.4.
Collapse
Affiliation(s)
- Qizhi Li
- Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen 518020, China.,John Hopcroft Center for Computer Science, Shanghai Jiao Tong University, Shanghai, China
| | - Xubin Zheng
- Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen 518020, China.,Department of Computer Science and Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.,Great Bay University, Dongguan, China
| | - Jize Xie
- Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen 518020, China.,John Hopcroft Center for Computer Science, Shanghai Jiao Tong University, Shanghai, China
| | - Ran Wang
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Mengyao Li
- Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen 518020, China
| | - Man-Hon Wong
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Kwong-Sak Leung
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.,Department of Applied Data Science, Hong Kong Shue Yan University, North Point, Hong Kong
| | - Shuai Li
- John Hopcroft Center for Computer Science, Shanghai Jiao Tong University, Shanghai, China
| | - Qingshan Geng
- Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen 518020, China
| | - Lixin Cheng
- Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen 518020, China
| |
Collapse
|
19
|
Cheng L, Wu H, Zheng X, Zhang N, Zhao P, Wang R, Wu Q, Liu T, Yang X, Geng Q. GPGPS: a robust prognostic gene pair signature of glioma ensembling IDH mutation and 1p/19q co-deletion. Bioinformatics 2023; 39:6986965. [PMID: 36637205 PMCID: PMC9843586 DOI: 10.1093/bioinformatics/btac850] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/14/2022] [Indexed: 01/14/2023] Open
Abstract
MOTIVATION Many studies have shown that IDH mutation and 1p/19q co-deletion can serve as prognostic signatures of glioma. Although these genetic variations affect the expression of one or more genes, the prognostic value of gene expression related to IDH and 1p/19q status is still unclear. RESULTS We constructed an ensemble gene pair signature for the risk evaluation and survival prediction of glioma based on the prior knowledge of the IDH and 1p/19q status. First, we separately built two gene pair signatures IDH-GPS and 1p/19q-GPS and elucidated that they were useful transcriptome markers projecting from corresponding genome variations. Then, the gene pairs in these two models were assembled to develop an integrated model named Glioma Prognostic Gene Pair Signature (GPGPS), which demonstrated high area under the curves (AUCs) to predict 1-, 3- and 5-year overall survival (0.92, 0.88 and 0.80) of glioma. GPGPS was superior to the single GPSs and other existing prognostic signatures (avg AUC = 0.70, concordance index = 0.74). In conclusion, the ensemble prognostic signature with 10 gene pairs could serve as an independent predictor for risk stratification and survival prediction in glioma. This study shed light on transferring knowledge from genetic alterations to expression changes to facilitate prognostic studies. AVAILABILITY AND IMPLEMENTATION Codes are available at https://github.com/Kimxbzheng/GPGPS.git. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Lixin Cheng
- To whom correspondence should be addressed. or
| | | | - Xubin Zheng
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen 518020, China
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Ning Zhang
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Pengfei Zhao
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen 518020, China
- Department of Geriatrics, Shenzhen Clinical Research Center for Aging, Shenzhen 518020, China
| | - Ran Wang
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen 518020, China
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Qiong Wu
- Hong Kong Genome Institute, Shatin, New Territories, Hong Kong
| | - Tao Liu
- International Digital Economy Academy, Shenzhen 518020, China
| | - Xiaojun Yang
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | | |
Collapse
|
20
|
Wei SY, Feng B, Bi M, Guo HY, Ning SW, Cui R. Construction of a ferroptosis-related signature based on seven lncRNAs for prognosis and immune landscape in clear cell renal cell carcinoma. BMC Med Genomics 2022; 15:263. [PMID: 36528763 PMCID: PMC9758795 DOI: 10.1186/s12920-022-01418-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Recent studies have demonstrated that long non-coding RNAs (lncRNAs) are involved in regulating tumor cell ferroptosis. However, prognostic signatures based on ferroptosis-related lncRNAs (FRLs) and their relationship to the immune microenvironment have not been comprehensively explored in clear cell renal cell carcinoma (ccRCC). METHODS In the present study, the expression profiles of ccRCC were acquired from The Cancer Genome Atlas (TCGA) database; 459 patient specimens and 69 adjacent normal tissues were randomly separated into training or validation cohorts at a 7:3 ratio. We identified 7 FRLs that constitute a prognostic signature according to the differential analysis, correlation analysis, univariate regression, and least absolute shrinkage and selection operator (LASSO) Cox analysis. To identify the independence of risk score as a prognostic factor, univariate and multivariate regression analyses were also performed. Furthermore, CIBERSORT was conducted to analyze the immune infiltration of patients in the high-risk and low-risk groups. Subsequently, the differential expression of immune checkpoint and m6A genes was analyzed in the two risk groups. RESULTS A 7-FRLs prognostic signature of ccRCC was developed to distinguish patients into high-risk and low-risk groups with significant survival differences. This signature has great prognostic performance, with the area under the curve (AUC) for 1, 3, and 5 years of 0.713, 0.700, 0.726 in the training set and 0.727, 0.667, and 0.736 in the testing set, respectively. Moreover, this signature was significantly associated with immune infiltration. Correlation analysis showed that risk score was positively correlated with regulatory T cells (Tregs), activated CD4 memory T cells, CD8 T cells and follicular helper T cells, whereas it was inversely correlated with monocytes and M2 macrophages. In addition, the expression of fourteen immune checkpoint genes and nine m6A-related genes varied significantly between the two risk groups. CONCLUSION We established a novel FRLs-based prognostic signature for patients with ccRCC, containing seven lncRNAs with precise predictive performance. The FRLs prognostic signature may play a significant role in antitumor immunity and provide a promising idea for individualized targeted therapy for patients with ccRCC.
Collapse
Affiliation(s)
- Shi-Yao Wei
- grid.412463.60000 0004 1762 6325Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China ,grid.410736.70000 0001 2204 9268College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081 Heilongjiang Province People’s Republic of China
| | - Bei Feng
- grid.411491.8Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, 150001 Heilongjiang Province People’s Republic of China ,grid.410736.70000 0001 2204 9268College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081 Heilongjiang Province People’s Republic of China
| | - Min Bi
- grid.412463.60000 0004 1762 6325Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Hai-Ying Guo
- grid.411491.8Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, 150001 Heilongjiang Province People’s Republic of China
| | - Shang-Wei Ning
- grid.410736.70000 0001 2204 9268College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081 Heilongjiang Province People’s Republic of China
| | - Rui Cui
- grid.411491.8Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, 150001 Heilongjiang Province People’s Republic of China
| |
Collapse
|
21
|
de la Varga-Martínez O, Martín-Fernández M, Heredia-Rodríguez M, Ceballos F, Cubero-Gallego H, Priede-Vimbela JM, Bardají-Carrillo M, Sánchez-de Prada L, López-Herrero R, Jorge-Monjas P, Tamayo E, Gómez-Sánchez E. Influence of Renal Dysfunction on the Differential Behaviour of Procalcitonin for the Diagnosis of Postoperative Infection in Cardiac Surgery. J Clin Med 2022; 11:jcm11247274. [PMID: 36555891 PMCID: PMC9781060 DOI: 10.3390/jcm11247274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/26/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Background: procalcitonin is a valuable marker in the diagnosis of bacterial infections; however, the impairment of renal function can influence its diagnostic precision. The objective of this study is to evaluate the differential behaviour of procalcitonin, as well as its usefulness in the diagnosis of postoperative pulmonary infection after cardiac surgery, depending on the presence or absence of impaired renal function. Materials and methods: A total of 805 adult patients undergoing cardiac surgery with extracorporeal circulation (CBP) were prospectively recruited, comparing the behaviour of biomarkers between the groups with and without postoperative pneumonia and according to the presence or absence of renal dysfunction. Results: Pulmonary infection was diagnosed in 42 patients (5.21%). In total, 228 patients (28.32%) presented postoperative renal dysfunction. Procalcitonin was significantly higher in infected patients, even in the presence of renal dysfunction. The optimal procalcitonin threshold differed markedly in patients with renal dysfunction compared to patients without renal dysfunction (1 vs. 0.78 ng/mL p < 0.05). The diagnostic accuracy of procalcitonin increased significantly when the procalcitonin threshold was adapted to renal function. Conclusions: Procalcitonin is an accurate marker of postoperative infection in cardiac surgery, even in the presence of renal dysfunction. Renal function is an important determinant of procalcitonin levels and, therefore, its diagnostic thresholds must be adapted in the presence of renal dysfunction.
Collapse
Affiliation(s)
- Olga de la Varga-Martínez
- Department of Anaesthesiology, Infanta Leonor University Hospital, Gran Via del Este 80, 28031 Madrid, Spain
- BioCritic, Group for Biomedical Research in Critical Care Medicine, Ramon y Cajal Ave. 7, 47005 Valladolid, Spain
- Correspondence: ; Tel.: +34-911918000
| | - Marta Martín-Fernández
- BioCritic, Group for Biomedical Research in Critical Care Medicine, Ramon y Cajal Ave. 7, 47005 Valladolid, Spain
- Center for Biomedical Research in Infectious Diseases Network (CIBERINFEC), Carlos III Health Institute, 28029 Madrid, Spain
- Department of Medicine, Faculty of Medicine, Universidad de Valladolid, 47005 Valladolid, Spain
| | - María Heredia-Rodríguez
- BioCritic, Group for Biomedical Research in Critical Care Medicine, Ramon y Cajal Ave. 7, 47005 Valladolid, Spain
- Center for Biomedical Research in Infectious Diseases Network (CIBERINFEC), Carlos III Health Institute, 28029 Madrid, Spain
- Department of Anaesthesiology, Clinical University Hospital of Salamanca, P.° de San Vicente, 58, 37007 Salamanca, Spain
| | - Francisco Ceballos
- Viral Infection and Immunity Unit, National Center for Microbiology, Carlos III Health Institute, 28029 Madrid, Spain
| | - Hector Cubero-Gallego
- Interventional Cardiology Unit, Cardiology Department, Hospital del Mar, 08003 Barcelona, Spain
| | - Juan Manuel Priede-Vimbela
- Department of Anaesthesiology, Clinic University Hospital of Valladolid, Ramon y Cajal Ave. 3, 47003 Valladolid, Spain
| | - Miguel Bardají-Carrillo
- Department of Anaesthesiology, Clinic University Hospital of Valladolid, Ramon y Cajal Ave. 3, 47003 Valladolid, Spain
| | - Laura Sánchez-de Prada
- BioCritic, Group for Biomedical Research in Critical Care Medicine, Ramon y Cajal Ave. 7, 47005 Valladolid, Spain
- Microbiology and Immunology Department, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain
| | - Rocío López-Herrero
- BioCritic, Group for Biomedical Research in Critical Care Medicine, Ramon y Cajal Ave. 7, 47005 Valladolid, Spain
- Department of Anaesthesiology, Clinic University Hospital of Valladolid, Ramon y Cajal Ave. 3, 47003 Valladolid, Spain
| | - Pablo Jorge-Monjas
- BioCritic, Group for Biomedical Research in Critical Care Medicine, Ramon y Cajal Ave. 7, 47005 Valladolid, Spain
- Center for Biomedical Research in Infectious Diseases Network (CIBERINFEC), Carlos III Health Institute, 28029 Madrid, Spain
- Department of Anaesthesiology, Clinic University Hospital of Valladolid, Ramon y Cajal Ave. 3, 47003 Valladolid, Spain
- Department of Surgery, Faculty of Medicine, Universidad de Valladolid, 47005 Valladolid, Spain
| | - Eduardo Tamayo
- BioCritic, Group for Biomedical Research in Critical Care Medicine, Ramon y Cajal Ave. 7, 47005 Valladolid, Spain
- Center for Biomedical Research in Infectious Diseases Network (CIBERINFEC), Carlos III Health Institute, 28029 Madrid, Spain
- Department of Anaesthesiology, Clinic University Hospital of Valladolid, Ramon y Cajal Ave. 3, 47003 Valladolid, Spain
- Department of Surgery, Faculty of Medicine, Universidad de Valladolid, 47005 Valladolid, Spain
| | - Esther Gómez-Sánchez
- BioCritic, Group for Biomedical Research in Critical Care Medicine, Ramon y Cajal Ave. 7, 47005 Valladolid, Spain
- Center for Biomedical Research in Infectious Diseases Network (CIBERINFEC), Carlos III Health Institute, 28029 Madrid, Spain
- Department of Anaesthesiology, Clinic University Hospital of Valladolid, Ramon y Cajal Ave. 3, 47003 Valladolid, Spain
- Department of Surgery, Faculty of Medicine, Universidad de Valladolid, 47005 Valladolid, Spain
| |
Collapse
|
22
|
Yang Y, Zhang Y, Li S, Zheng X, Wong MH, Leung KS, Cheng L. A Robust and Generalizable Immune-Related Signature for Sepsis Diagnostics. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:3246-3254. [PMID: 34437068 DOI: 10.1109/tcbb.2021.3107874] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
High-throughput sequencing can detect tens of thousands of genes in parallel, providing opportunities for improving the diagnostic accuracy of multiple diseases including sepsis, which is an aggressive inflammatory response to infection that can cause organ failure and death. Early screening of sepsis is essential in clinic, but no effective diagnostic biomarkers are available yet. Here, we present a novel method, Recurrent Logistic Regression, to identify diagnostic biomarkers for sepsis from the blood transcriptome data. A panel including five immune-related genes, LRRN3, IL2RB, FCER1A, TLR5, and S100A12, are determined as diagnostic biomarkers (LIFTS) for sepsis. LIFTS discriminates patients with sepsis from normal controls in high accuracy (AUROC = 0.9959 on average; IC = [0.9722-1.0]) on nine validation cohorts across three independent platforms, which outperforms existing markers. Our analysis determined an accurate prediction model and reproducible transcriptome biomarkers that can lay a foundation for clinical diagnostic tests and biological mechanistic studies.
Collapse
|
23
|
Xu C, Li W, Li T, Yuan J, Pang X, Liu T, Liang B, Cheng L, Sun X, Dong S. Iron metabolism-related genes reveal predictive value of acute coronary syndrome. Front Pharmacol 2022; 13:1040845. [PMID: 36330096 PMCID: PMC9622999 DOI: 10.3389/fphar.2022.1040845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/04/2022] [Indexed: 11/25/2022] Open
Abstract
Iron deficiency has detrimental effects in patients with acute coronary syndrome (ACS), which is a common nutritional disorder and inflammation-related disease affects up to one-third people worldwide. However, the specific role of iron metabolism in ACS progression is opaque. In this study, we construct an iron metabolism-related genes (IMRGs) based molecular signature of ACS and to identify novel iron metabolism gene markers for early stage of ACS. The IMRGs were mainly collected from Molecular Signatures Database (mSigDB) and two relevant studies. Two blood transcriptome datasets GSE61144 and GSE60993 were used for constructing the prediction model of ACS. After differential analysis, 22 IMRGs were differentially expressed and defined as DEIGs in the training set. Then, the 22 DEIGs were trained by the Elastic Net to build the prediction model. Five genes, PADI4, HLA-DQA1, LCN2, CD7, and VNN1, were determined using multiple Elastic Net calculations and retained to obtain the optimal performance. Finally, the generated model iron metabolism-related gene signature (imSig) was assessed by the validation set GSE60993 using a series of evaluation measurements. Compared with other machine learning methods, the performance of imSig using Elastic Net was superior in the validation set. Elastic Net consistently scores the higher than Lasso and Logistic regression in the validation set in terms of ROC, PRC, Sensitivity, and Specificity. The prediction model based on iron metabolism-related genes may assist in ACS early diagnosis.
Collapse
Affiliation(s)
- Cong Xu
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Wanyang Li
- School of Mathematics, South China University of Technology, Guangzhou, China
| | - Tangzhiming Li
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Jie Yuan
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Xinli Pang
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Tao Liu
- International Digital Economy Academy, Shenzhen, China
| | - Benhui Liang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China
| | - Lixin Cheng
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
- *Correspondence: Lixin Cheng, ; Xin Sun, ; Shaohong Dong,
| | - Xin Sun
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
- *Correspondence: Lixin Cheng, ; Xin Sun, ; Shaohong Dong,
| | - Shaohong Dong
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
- *Correspondence: Lixin Cheng, ; Xin Sun, ; Shaohong Dong,
| |
Collapse
|
24
|
Bioinformatics Analysis Identifies TNFRSF1A as a Biomarker of Liver Injury in Sepsis TNFRSF1A is a Biomarker for Septic Liver Injury. Genet Res (Camb) 2022; 2022:1493744. [PMID: 36299685 PMCID: PMC9587912 DOI: 10.1155/2022/1493744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/18/2022] Open
Abstract
Sepsis is a severe disease with high mortality, and liver injury is an independent risk factor for sepsis morbidity and mortality. We analyzed co-differentially expressed genes (co-DEGs) to explore potential biomarkers and therapeutic targets for sepsis-related liver injury. Three gene expression datasets (GSE60088, GSE23767, and GSE71530) were downloaded from the Gene Expression Omnibus (GEO). DEGs were screened between sepsis and control samples using GEO2R. The association of these DEGs with infection and liver disease was analyzed by using the CTD database. GO functional analysis, KEGG pathway enrichment analysis, and protein-protein interaction (PPI) network analysis were performed to elucidate the potential molecular mechanism of DEGs. DEGs of different tissues in GSE60088 were analyzed again to obtain specific markers of septic liver injury. Mouse model of sepsis was also established by cecal ligation and puncture (CLP), and the expression of specific markers in liver, lung, and kidney tissues was analyzed using Western blot. Here, we identified 21 DEGs in three datasets with 8 hub genes, all of which showed higher inference scores in liver diseases than bacterial infections. Among them, only TNFRSF1A had a liver-specific differential expression. TNFRSF1A was also confirmed to be specifically reduced in septic liver tissues in mice. Therefore, TNFRSF1A may serve as a potential biomarker for septic liver injury.
Collapse
|
25
|
Zhang J, Deng J, Feng X, Tan Y, Li X, Liu Y, Li M, Qi H, Tang L, Meng Q, Yan H, Qi L. Hierarchical identification of a transcriptional panel for the histological diagnosis of lung neuroendocrine tumors. Front Genet 2022; 13:944167. [PMID: 36105102 PMCID: PMC9465419 DOI: 10.3389/fgene.2022.944167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Lung cancer is a complex disease composed of neuroendocrine (NE) and non-NE tumors. Accurate diagnosis of lung cancer is essential in guiding therapeutic management. Several transcriptional signatures have been reported to distinguish between adenocarcinoma (ADC) and squamous cell carcinoma (SCC) belonging to non-NE tumors. This study aims to identify a transcriptional panel that could distinguish the histological subtypes of NE tumors to complement the morphology-based classification of an individual.Methods: A public dataset with NE subtypes, including 21 small-cell lung cancer (SCLC), 56 large-cell NE carcinomas (LCNECs), and 24 carcinoids (CARCIs), and non-NE subtypes, including 85 ADC and 61 SCC, was used as a training set. In the training set, consensus clustering was first used to filter out the samples whose expression patterns disagreed with their histological subtypes. Then, a rank-based method was proposed to develop a panel of transcriptional signatures for determining the NE subtype for an individual, based on the within-sample relative gene expression orderings of gene pairs. Twenty-three public datasets with a total of 3,454 samples, which were derived from fresh-frozen, formalin-fixed paraffin-embedded, biopsies, and single cells, were used for validation. Clinical feasibility was tested in 10 SCLC biopsy specimens collected from cancer hospitals via bronchoscopy.Results: The NEsubtype-panel was composed of three signatures that could distinguish NE from non-NE, CARCI from non-CARCI, and SCLC from LCNEC step by step and ultimately determine the histological subtype for each NE sample. The three signatures achieved high average concordance rates with 97.31%, 98.11%, and 90.63%, respectively, in the 23 public validation datasets. It is worth noting that the 10 clinic-derived SCLC samples diagnosed via immunohistochemical staining were also accurately predicted by the NEsubtype-panel. Furthermore, the subtype-specific gene expression patterns and survival analyses provided evidence for the rationality of the reclassification by the NEsubtype-panel.Conclusion: The rank-based NEsubtype-panel could accurately distinguish lung NE from non-NE tumors and determine NE subtypes even in clinically challenging samples (such as biopsy). The panel together with our previously reported signature (KRT5-AGR2) for SCC and ADC would be an auxiliary test for the histological diagnosis of lung cancer.
Collapse
Affiliation(s)
- Juxuan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jiaxing Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xiao Feng
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yilong Tan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xin Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yixin Liu
- Basic Medicine College, Harbin Medical University, Harbin, China
| | - Mengyue Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Haitao Qi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lefan Tang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Qingwei Meng
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Haidan Yan
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, China
- *Correspondence: Haidan Yan, ; Lishuang Qi,
| | - Lishuang Qi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- *Correspondence: Haidan Yan, ; Lishuang Qi,
| |
Collapse
|
26
|
Vijayan A, Fatima S, Sowmya A, Vafaee F. Blood-based transcriptomic signature panel identification for cancer diagnosis: benchmarking of feature extraction methods. Brief Bioinform 2022; 23:6658855. [PMID: 35945147 DOI: 10.1093/bib/bbac315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Liquid biopsy has shown promise for cancer diagnosis due to its minimally invasive nature and the potential for novel biomarker discovery. However, the low concentration of relevant blood-based biosources and the heterogeneity of samples (i.e. the variability of relative abundance of molecules identified), pose major challenges to biomarker discovery. Moreover, the number of molecular measurements or features (e.g. transcript read counts) per sample could be in the order of several thousand, whereas the number of samples is often substantially lower, leading to the curse of dimensionality. These challenges, among others, elucidate the importance of a robust biomarker panel identification or feature extraction step wherein relevant molecular measurements are identified prior to classification for cancer detection. In this work, we performed a benchmarking study on 12 feature extraction methods using transcriptomic profiles derived from different blood-based biosources. The methods were assessed both in terms of their predictive performance and the robustness of the biomarker panels in diagnosing cancer or stratifying cancer subtypes. While performing the comparison, the feature extraction methods are categorized into feature subset selection methods and transformation methods. A transformation feature extraction method, namely partial least square discriminant analysis, was found to perform consistently superior in terms of classification performance. As part of the benchmarking study, a generic pipeline has been created and made available as an R package to ensure reproducibility of the results and allow for easy extension of this study to other datasets (https://github.com/VafaeeLab/bloodbased-pancancer-diagnosis).
Collapse
Affiliation(s)
- Abhishek Vijayan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.,School of Computer Science and Engineering, University of New South Wales, Sydney, NSW, Australia
| | - Shadma Fatima
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.,Ingham Institute, NSW, Australia
| | - Arcot Sowmya
- School of Computer Science and Engineering, University of New South Wales, Sydney, NSW, Australia.,UNSW Data Science Hub, University of New South Wales, Sydney, NSW, Australia
| | - Fatemeh Vafaee
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.,UNSW Data Science Hub, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
27
|
Wu Q, Zheng X, Leung KS, Wong MH, Tsui SKW, Cheng L. meGPS: a multi-omics signature for hepatocellular carcinoma detection integrating methylome and transcriptome data. Bioinformatics 2022; 38:3513-3522. [PMID: 35674358 DOI: 10.1093/bioinformatics/btac379] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 05/08/2022] [Accepted: 06/01/2022] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION Hepatocellular carcinoma (HCC) is a primary malignancy with poor prognosis. Recently, multi-omics molecular-level measurement enables HCC diagnosis and prognosis prediction, which is crucial for early intervention of personalized therapy to diminish mortality. Here, we introduce a novel strategy utilizing DNA methylation and RNA expression data to achieve a multi-omics gene pair signature (GPS) for HCC discrimination. RESULTS The immune genes with negative correlations between expression and promoter methylation are enriched in the highly connected cancer-related pathway network, which are considered as the candidates for HCC detection. After that, we separately construct a methylation GPS (mGPS) and an expression GPS (eGPS), and then assemble them as a meGPS with five gene pairs, in which the significant methylation and expression changes occur between HCC tumor and non-tumor groups. Reliable performance has been validated by independent tissue (age, gender, and etiology) and blood datasets. This study proposes a procedure for multi-omics GPS identification and develops a novel HCC signature using both methylome and transcriptome data, suggesting potential molecular targets for the detection and therapy of HCC. AVAILABILITY AND IMPLEMENTATION Models are available at https://github.com/bioinformaticStudy/meGPS.git. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Qiong Wu
- Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, 518020, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China.,Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Xubin Zheng
- Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, 518020, China.,Department of Computer Science and Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Kwong-Sak Leung
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Man-Hon Wong
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Stephen Kwok-Wing Tsui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Lixin Cheng
- Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, 518020, China
| |
Collapse
|
28
|
Wang C, Liang G, Shen J, Kong H, Wu D, Huang J, Li X. Long Non-Coding RNAs as Biomarkers and Therapeutic Targets in Sepsis. Front Immunol 2021; 12:722004. [PMID: 34630395 PMCID: PMC8492911 DOI: 10.3389/fimmu.2021.722004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/18/2021] [Indexed: 12/14/2022] Open
Abstract
Sepsis, an infection-induced systemic inflammatory disorder, is often accompanied by multiple organ dysfunction syndromes with high incidence and mortality rates, and those who survive are often left with long-term sequelae, bringing great burden to social economy. Therefore, novel approaches to solve this puzzle are urgently needed. Previous studies revealed that long non-coding RNAs (lncRNAs) have exerted significant influences on the process of sepsis. The aim of this review is to summarize our understanding of lncRNAs as potential sepsis-related diagnostic markers and therapeutic targets, and provide new insights into the diagnosis and treatment for sepsis. In this study, we also introduced the current diagnostic markers of sepsis and discussed their limitations, while review the research advances in lncRNAs as promising biomarkers for diagnosis and prognosis of sepsis. Furthermore, the roles of lncRNAs in sepsis-induced organ dysfunction were illustrated in terms of different organ systems. Nevertheless, further studies should be carried out to elucidate underlying molecular mechanisms and pathological process of sepsis.
Collapse
Affiliation(s)
- Chuqiao Wang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Guorui Liang
- Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Jieni Shen
- Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Haifan Kong
- Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Donghong Wu
- Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Jinxiang Huang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xuefeng Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Shenzhen Luohu People's Hospital, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
29
|
Oliveira RADC, Imparato DO, Fernandes VGS, Cavalcante JVF, Albanus RD, Dalmolin RJS. Reverse Engineering of the Pediatric Sepsis Regulatory Network and Identification of Master Regulators. Biomedicines 2021; 9:biomedicines9101297. [PMID: 34680414 PMCID: PMC8533457 DOI: 10.3390/biomedicines9101297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/26/2021] [Accepted: 07/26/2021] [Indexed: 01/04/2023] Open
Abstract
Sepsis remains a leading cause of death in ICUs all over the world, with pediatric sepsis accounting for a high percentage of mortality in pediatric ICUs. Its complexity makes it difficult to establish a consensus on genetic biomarkers and therapeutic targets. A promising strategy is to investigate the regulatory mechanisms involved in sepsis progression, but there are few studies regarding gene regulation in sepsis. This work aimed to reconstruct the sepsis regulatory network and identify transcription factors (TFs) driving transcriptional states, which we refer to here as master regulators. We used public gene expression datasets to infer the co-expression network associated with sepsis in a retrospective study. We identified a set of 15 TFs as potential master regulators of pediatric sepsis, which were divided into two main clusters. The first cluster corresponded to TFs with decreased activity in pediatric sepsis, and GATA3 and RORA, as well as other TFs previously implicated in the context of inflammatory response. The second cluster corresponded to TFs with increased activity in pediatric sepsis and was composed of TRIM25, RFX2, and MEF2A, genes not previously described as acting in a coordinated way in pediatric sepsis. Altogether, these results show how a subset of master regulators TF can drive pathological transcriptional states, with implications for sepsis biology and treatment.
Collapse
Affiliation(s)
- Raffael Azevedo de Carvalho Oliveira
- Bioinformatics Multidisciplinary Environment–BioME, Instituto Metrópole Digital, Universidade Federal do Rio Grande do Norte, Natal 59078-400, Brazil; (R.A.d.C.O.); (D.O.I.); (V.G.S.F.); (J.V.F.C.)
| | - Danilo Oliveira Imparato
- Bioinformatics Multidisciplinary Environment–BioME, Instituto Metrópole Digital, Universidade Federal do Rio Grande do Norte, Natal 59078-400, Brazil; (R.A.d.C.O.); (D.O.I.); (V.G.S.F.); (J.V.F.C.)
| | - Vítor Gabriel Saldanha Fernandes
- Bioinformatics Multidisciplinary Environment–BioME, Instituto Metrópole Digital, Universidade Federal do Rio Grande do Norte, Natal 59078-400, Brazil; (R.A.d.C.O.); (D.O.I.); (V.G.S.F.); (J.V.F.C.)
| | - João Vitor Ferreira Cavalcante
- Bioinformatics Multidisciplinary Environment–BioME, Instituto Metrópole Digital, Universidade Federal do Rio Grande do Norte, Natal 59078-400, Brazil; (R.A.d.C.O.); (D.O.I.); (V.G.S.F.); (J.V.F.C.)
| | - Ricardo D’Oliveira Albanus
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Rodrigo Juliani Siqueira Dalmolin
- Bioinformatics Multidisciplinary Environment–BioME, Instituto Metrópole Digital, Universidade Federal do Rio Grande do Norte, Natal 59078-400, Brazil; (R.A.d.C.O.); (D.O.I.); (V.G.S.F.); (J.V.F.C.)
- Department of Biochemistry–DBQ–CB, Federal University of Rio Grande do Norte, Natal 59064-741, Brazil
- Correspondence:
| |
Collapse
|
30
|
Sun B, Guo S. miR-486-5p Serves as a Diagnostic Biomarker for Sepsis and Its Predictive Value for Clinical Outcomes. J Inflamm Res 2021; 14:3687-3695. [PMID: 34354365 PMCID: PMC8331108 DOI: 10.2147/jir.s323433] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/08/2021] [Indexed: 12/17/2022] Open
Abstract
Background As a molecular detection method, miRNA can quickly diagnose and prevent diseases, intervene in disease as early as possible, and reduce mortality. This study was to investigate the potential clinical diagnostic and predictive significance of miR-486-5p in sepsis and its correlation with inflammation and disease severity. Methods The serum miR-486-5p in 108 sepsis, 60 pneumonia-infected, and 101 healthy controls were detected by RT-qPCR. Spearman coefficient detects the correlation between serum miRNA and disease severity indicators (APACHE II, SOFA scores), and inflammation indicators (CRP, PCT), respectively. The diagnostic significance of miR-486-5p in sepsis was analyzed by the ROC curve. Kaplan–Meier estimator and Cox regression hazards analysis of the predictive significance of serum miR-486-5p in 28-day survival from sepsis. Results Serum miR-486-5p was increased in sepsis patients compared with healthy control and pneumonia-infected patients (P < 0.001). And increased serum miR-486-5p was positively associated with disease severity (SOFA score and APACHE II score) and inflammation (CRP and PCT). Serum miR-486-5p can not only identify sepsis patients from healthy controls (AUC = 0.914) but also significantly distinguish sepsis patients from pneumonia-infected patients (AUC = 0.814), showing good potential as a diagnostic biomarker for sepsis. In addition, serum miR-486-5p was an independent predictor of 28-day survival (log-rank P = 0.012), and patients with high levels of miR-486-5p had a poorer overall 28-day survival (HR = 3.057, 95% CI = 1.385–17.817, P = 0.014). Conclusion miR-486-5p is a potential diagnostic biomarker for sepsis, and its high level is significantly correlated with the disease severity and inflammation. In addition, miR-486-5p were predictive risk factors for 28-day survival in sepsis patients.
Collapse
Affiliation(s)
- Baobin Sun
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, 100020, People's Republic of China
| | - Shubin Guo
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, 100020, People's Republic of China
| |
Collapse
|
31
|
Song Y, Zhu S, Zhang N, Cheng L. Blood Circulating miRNA Pairs as a Robust Signature for Early Detection of Esophageal Cancer. Front Oncol 2021; 11:723779. [PMID: 34368003 PMCID: PMC8343071 DOI: 10.3389/fonc.2021.723779] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 07/08/2021] [Indexed: 01/07/2023] Open
Abstract
Esophageal cancer (EC) is a common malignant tumor in the digestive system which is often diagnosed at the middle and late stages. Noninvasive diagnosis using circulating miRNA as biomarkers enables accurate detection of early-stage EC to reduce mortality. We built a diagnostic signature consisting of four miRNA pairs for the early detection of EC using individualized Pairwise Analysis of Gene Expression (iPAGE). Profiling of miRNA expression identified 496 miRNA pairs with significant relative expression change. Four miRNA pairs consistently selected from LASSO were used to construct the final diagnostic model. The performance of the signature was validated using two independent datasets, yielding both AUCs and PRCs over 0.99. Furthermore, precision, recall, and F-score were also evaluated for clinical application, when a fixed threshold is given, resulting in all the scores are larger than 0.92 in the training set, test set, and two validation sets. Our results suggested that the 4-miRNA signature is a new biomarker for the early diagnosis of patients with EC. The clinical use of this signature would have improved the detection of EC for earlier therapy and more favorite prognosis.
Collapse
Affiliation(s)
- Yang Song
- Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Suzhu Zhu
- Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Ning Zhang
- Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Lixin Cheng
- Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| |
Collapse
|