1
|
Sui J, Chen J, Chen Y, Iwamori N, Sun J. GASIDN: identification of sub-Golgi proteins with multi-scale feature fusion. BMC Genomics 2024; 25:1019. [PMID: 39478465 PMCID: PMC11526662 DOI: 10.1186/s12864-024-10954-3] [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: 03/03/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024] Open
Abstract
The Golgi apparatus is a crucial component of the inner membrane system in eukaryotic cells, playing a central role in protein biosynthesis. Dysfunction of the Golgi apparatus has been linked to neurodegenerative diseases. Accurate identification of sub-Golgi protein types is therefore essential for developing effective treatments for such diseases. Due to the expensive and time-consuming nature of experimental methods for identifying sub-Golgi protein types, various computational methods have been developed as identification tools. However, the majority of these methods rely solely on neighboring features in the protein sequence and neglect the crucial spatial structure information of the protein.To discover alternative methods for accurately identifying sub-Golgi proteins, we have developed a model called GASIDN. The GASIDN model extracts multi-dimension features by utilizing a 1D convolution module on protein sequences and a graph learning module on contact maps constructed from AlphaFold2.The model utilizes the deep representation learning model SeqVec to initialize protein sequences. GASIDN achieved accuracy values of 98.4% and 96.4% in independent testing and ten-fold cross-validation, respectively, outperforming the majority of previous predictors. To the best of our knowledge, this is the first method that utilizes multi-scale feature fusion to identify and locate sub-Golgi proteins. In order to assess the generalizability and scalability of our model, we conducted experiments to apply it in the identification of proteins from other organelles, including plant vacuoles and peroxisomes. The results obtained from these experiments demonstrated promising outcomes, indicating the effectiveness and versatility of our model. The source code and datasets can be accessed at https://github.com/SJNNNN/GASIDN .
Collapse
Affiliation(s)
- Jianan Sui
- School of Information Science and Engineering, University of Jinan, Jinan, China
| | - Jiazi Chen
- Laboratory of Zoology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka-shi, Fukuoka, Japan
| | - Yuehui Chen
- School of Artificial Intelligence Institute and Information Science and Engineering, University of Jinan, Jinan, China.
| | - Naoki Iwamori
- Laboratory of Zoology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka-shi, Fukuoka, Japan
| | - Jin Sun
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China
| |
Collapse
|
2
|
Bao W, Gu Y, Chen B, Yu H. Golgi_DF: Golgi proteins classification with deep forest. Front Neurosci 2023; 17:1197824. [PMID: 37250391 PMCID: PMC10213405 DOI: 10.3389/fnins.2023.1197824] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/19/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction Golgi is one of the components of the inner membrane system in eukaryotic cells. Its main function is to send the proteins involved in the synthesis of endoplasmic reticulum to specific parts of cells or secrete them outside cells. It can be seen that Golgi is an important organelle for eukaryotic cells to synthesize proteins. Golgi disorders can cause various neurodegenerative and genetic diseases, and the accurate classification of Golgi proteins is helpful to develop corresponding therapeutic drugs. Methods This paper proposed a novel Golgi proteins classification method, which is Golgi_DF with the deep forest algorithm. Firstly, the classified proteins method can be converted the vector features containing various information. Secondly, the synthetic minority oversampling technique (SMOTE) is utilized to deal with the classified samples. Next, the Light GBM method is utilized to feature reduction. Meanwhile, the features can be utilized in the penultimate dense layer. Therefore, the reconstructed features can be classified with the deep forest algorithm. Results In Golgi_DF, this method can be utilized to select the important features and identify Golgi proteins. Experiments show that the well-performance than the other art-of-the state methods. Golgi_DF as a standalone tools, all its source codes publicly available at https://github.com/baowz12345/golgiDF. Discussion Golgi_DF employed reconstructed feature to classify the Golgi proteins. Such method may achieve more available features among the UniRep features.
Collapse
Affiliation(s)
- Wenzheng Bao
- School of Information Engineering, Xuzhou University of Technology, Xuzhou, China
| | - Yujian Gu
- School of Information Engineering, Xuzhou University of Technology, Xuzhou, China
| | - Baitong Chen
- Department of Stomatology, Xuzhou First People’s Hospital, Xuzhou, China
- The Affiliated Hospital of China University of Mining and Technology, Xuzhou, China
| | - Huiping Yu
- Department of Neurosurgery, The Hospital of Joint Logistic, Quanzhou, China
| |
Collapse
|
3
|
Olatunji I, Cui F. Multimodal AI for prediction of distant metastasis in carcinoma patients. FRONTIERS IN BIOINFORMATICS 2023; 3:1131021. [PMID: 37228671 PMCID: PMC10203594 DOI: 10.3389/fbinf.2023.1131021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Metastasis of cancer is directly related to death in almost all cases, however a lot is yet to be understood about this process. Despite advancements in the available radiological investigation techniques, not all cases of Distant Metastasis (DM) are diagnosed at initial clinical presentation. Also, there are currently no standard biomarkers of metastasis. Early, accurate diagnosis of DM is however crucial for clinical decision making, and planning of appropriate management strategies. Previous works have achieved little success in attempts to predict DM from either clinical, genomic, radiology, or histopathology data. In this work we attempt a multimodal approach to predict the presence of DM in cancer patients by combining gene expression data, clinical data and histopathology images. We tested a novel combination of Random Forest (RF) algorithm with an optimization technique for gene selection, and investigated if gene expression pattern in the primary tissues of three cancer types (Bladder Carcinoma, Pancreatic Adenocarcinoma, and Head and Neck Squamous Carcinoma) with DM are similar or different. Gene expression biomarkers of DM identified by our proposed method outperformed Differentially Expressed Genes (DEGs) identified by the DESeq2 software package in the task of predicting presence or absence of DM. Genes involved in DM tend to be more cancer type specific rather than general across all cancers. Our results also indicate that multimodal data is more predictive of metastasis than either of the three unimodal data tested, and genomic data provides the highest contribution by a wide margin. The results re-emphasize the importance for availability of sufficient image data when a weakly supervised training technique is used. Code is made available at: https://github.com/rit-cui-lab/Multimodal-AI-for-Prediction-of-Distant-Metastasis-in-Carcinoma-Patients.
Collapse
Affiliation(s)
| | - Feng Cui
- Thomas H. Gosnell School of Life Science, Rochester Institute of Technology, Rochester, NY, United States
| |
Collapse
|
4
|
Gao F, Cho WC, Gao X, Wang W. Editorial: Medical knowledge-assisted machine learning technologies in individualized medicine. Front Mol Biosci 2023; 10:1167730. [PMID: 37033449 PMCID: PMC10080393 DOI: 10.3389/fmolb.2023.1167730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 04/11/2023] Open
Affiliation(s)
- Feng Gao
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Shanghai Artificial Intelligence Laboratory, Shanghai, China
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR, China
| | - Xin Gao
- Computational Bioscience Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Wei Wang
- Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
5
|
Luo K, Xu S, Zhao J, Liu F. Upregulation of lncRNA PINK1-AS Predicts the Distant Metastasis of Patients with Small Cell Lung Cancer. Mol Biotechnol 2023; 65:28-33. [PMID: 35764723 DOI: 10.1007/s12033-022-00512-1] [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: 12/08/2021] [Accepted: 05/11/2022] [Indexed: 01/22/2023]
Abstract
PINK1-AS has been shown to participate in gastric cancer, while its role in other tumors is unclear. This study was carried out to explore the participation of PINK1-AS in small cell lung cancer (SCLC). In this study, the expression of PINK1-AS in SCLC and paired non-cancer tissues from 60 SCLC patients and in plasma samples from 60 SCLC patients and 60 healthy controls was analyzed with RT-qPCR. Chi-squared t test was applied to analyze the associations between plasma expression levels of PINK1-AS and the clinical factors of the patients. Patients were followed up for 5 years to explore the role of PINK1-AS in the prognosis of SCLC. ROC curve analysis was applied to explore the role of PINK1-AS in the prediction of distant metastasis. Transwell assays were performed to evaluate the role of silencing and overexpression of PINK1-AS in the invasion and migration of SCLC cells. We found that PINK1-AS was upregulated in SCLC tissues compared to that in non-cancer tissues. Plasma expression levels of PINK1-AS were increased in SCLC patients compared to that in the controls. High plasma expression levels of PINK1-AS were closely associated with worse survival. Plasma expression of PINK1-AS was only closely correlated with distant tumor metastasis, but not other factors. High plasma expression levels of PINK1-AS effectively separated patients with distant metastasis from non-metastatic patients. Moreover, PINK1-AS positively regulated the migration and invasion of SCLC cells. Therefore, the upregulation of PINK1-AS predicts the distant metastasis of patients with SCLC.
Collapse
Affiliation(s)
- Kun Luo
- Department of Pulmonary and Critical Care Medicine, First Hospital of Qinhuangdao, No. 258 Wenhua Road, Haigang District, Qinhuangdao, 066000, Hebei, People's Republic of China
| | - Shufeng Xu
- Department of Pulmonary and Critical Care Medicine, First Hospital of Qinhuangdao, No. 258 Wenhua Road, Haigang District, Qinhuangdao, 066000, Hebei, People's Republic of China.
| | - Jing Zhao
- Department of Pulmonary and Critical Care Medicine, First Hospital of Qinhuangdao, No. 258 Wenhua Road, Haigang District, Qinhuangdao, 066000, Hebei, People's Republic of China
| | - Feifei Liu
- Department of Pulmonary and Critical Care Medicine, First Hospital of Qinhuangdao, No. 258 Wenhua Road, Haigang District, Qinhuangdao, 066000, Hebei, People's Republic of China
| |
Collapse
|
6
|
Qi X, Zuo J, Yan D, Hu G, Wang R, Chen J, Fu J. A NOD-Like Receptor Signaling-Based Gene Signature Identified as a
Novel Prognostic Biomarker for Predicting Overall Survival of Colorectal
Cancer Patients. Curr Bioinform 2022. [DOI: 10.2174/1574893616666211005122422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Colorectal Cancer (CRC) is the most frequently diagnosed gastrointestinal
tract malignant tumor worldwide, which is closely associated with distant metastasis and poor prognosis.
Due to high degree of heterogeneity, reliable prognostic biomarkers are urgently needed to guide the
therapeutic intervention of CRC patients.
Objective:
The present study aimed to develop a NOD-Like Receptors (NLRs) signaling-based gene
signature that can successfully predict the overall survival of CRC patients.
Methods:
Firstly, differentially expressed NLR signaling-related genes were identified between primary
and metastatic human CRC samples. Genes with prognostic value were then screened through univariate
Cox regression analysis. Next, the NLR signaling-based prognostic signature was constructed by
LASSO-penalized Cox regression analysis, and its predictive ability was further confirmed in an independent
cohort. Furthermore, functional studies including GO, GSEA, ssGSEA and chemotherapeutic
response analyses were performed to explore the role of the NLR signaling-based signature in CRC
pathogenesis and therapy.
Results:
The established prognostic signature that consisted of 7 NLR signaling-related genes can effectively
stratify the high-risk and low-risk CRC patients in both training and validation cohorts. Moreover,
the signature proved to be an independent indicator of overall survival in CRC patients. Functional annotation
and chemotherapeutic response analyses showed that the signature was closely associated with
immune status and chemotherapeutic sensitivity of CRC patients.
Conclusion:
The novel NLR signaling-based gene signature could serve as a potential tool for survival
prediction and therapeutic evaluation, thereby contributing to the personalized prognostic management
of CRC patients.
Collapse
Affiliation(s)
- Xin Qi
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, 215011 Suzhou, China
| | - Jiachen Zuo
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, 215011 Suzhou, China
| | - Donghui Yan
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, 215011 Suzhou, China
| | - Guang Hu
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, 215123 Suzhou, China
| | - Rui Wang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, 215011 Suzhou, China
| | - Jiajia Chen
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, 215011 Suzhou, China
| | - Jiaolong Fu
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, 215011 Suzhou, China
| |
Collapse
|
7
|
Yan J, Dai P, Qin X, He Y, Zhang Y. HMGA2 promotes the migration and invasion of gallbladder cancer cells and HMGA2 knockdown inhibits angiogenesis via targeting VEGFA. Mol Med Rep 2021; 25:54. [PMID: 34913073 PMCID: PMC8711027 DOI: 10.3892/mmr.2021.12570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/17/2021] [Indexed: 11/26/2022] Open
Abstract
The high mobility group AT-hook 2 (HMGA2) protein has been found to be upregulated in the majority of tumor types and is associated with a poor prognosis. Previous studies have suggested the oncogenic role of HMGA2 in gallbladder cancer (GBC). The present study aimed to investigate the effects of HMGA2 on the invasion, migration and angiogenesis of GBC cells. To achieve this aim, HMGA2 was overexpressed or silenced in the GBC cell line, EH-GB1, and then the proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) abilities of EH-GB1 cells were investigated using Cell Counting Kit-8, wound healing, Transwell and western blotting assays. In addition, the expression levels of VEGFA were determined in EH-GB1 cells using western blotting and reverse transcription-quantitative PCR following HMGA2 overexpression or silencing. Furthermore, HMGA2-silenced EH-GB1 cells were transfected with VEGFA overexpression plasmids to evaluate the tube formation ability of HUVECs using tube formation assay. The results demonstrated that HMGA2 silencing inhibited GBC cell proliferation, migration, invasion and EMT, as evidenced by the downregulated expression of Ki67, proliferating cell nuclear antigen, MMP2, MMP9, N-cadherin, snail family transcriptional repressor 2 and zinc finger E-box-binding homeobox 1, and attenuated cell migration and invasion. However, the opposite results were obtained following HMGA2 overexpression. Moreover, HMGA2 knockdown and overexpression downregulated and upregulated VEGFA expression, respectively. In addition, the tube formation ability of HUVECs and the expression levels of CD31, VEGFR1 and VEGFR2 were downregulated following HMGA2 silencing. However, these effects were partially rescued by simultaneous VEGFA overexpression. In conclusion, the findings of the present study revealed that HMGA2 may promote GBC cell migration, invasion, EMT and angiogenesis. Therefore, inhibiting HMGA2 expression could be considered as a possible therapeutic approach for GBC.
Collapse
Affiliation(s)
- Jun Yan
- Department of General Surgery, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Peng Dai
- Department of General Surgery, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Xueliang Qin
- Department of General Surgery, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Yanping He
- Department of General Surgery, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Yu Zhang
- Department of General Surgery, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| |
Collapse
|
8
|
Gutierrez A, Demond H, Brebi P, Ili CG. Novel Methylation Biomarkers for Colorectal Cancer Prognosis. Biomolecules 2021; 11:1722. [PMID: 34827720 PMCID: PMC8615818 DOI: 10.3390/biom11111722] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) comprises the third most common cancer worldwide and the second regarding number of deaths. In order to make a correct and early diagnosis to predict metastasis formation, biomarkers are an important tool. Although there are multiple signaling pathways associated with cancer progression, the most recognized are the MAPK pathway, p53 pathway, and TGF-β pathway. These pathways regulate many important functions in the cell, such as cell cycle regulation, proliferation, differentiation, and metastasis formation, among others. Changes in expression in genes belonging to these pathways are drivers of carcinogenesis. Often these expression changes are caused by mutations; however, epigenetic changes, such as DNA methylation, are increasingly acknowledged to play a role in the deregulation of oncogenic genes. This makes DNA methylation changes an interesting biomarkers in cancer. Among the newly identified biomarkers for CRC metastasis INHBB, SMOC2, BDNF, and TBRG4 are included, all of which are highly deregulated by methylation and closely associated with metastasis. The identification of such biomarkers in metastasis of CRC may allow a better treatment and early identification of cancer formation in order to perform better diagnostics and improve the life expectancy.
Collapse
Affiliation(s)
| | | | - Priscilla Brebi
- Millennium Institute on Immunology and Immunotherapy, Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (A.G.); (H.D.)
| | - Carmen Gloria Ili
- Millennium Institute on Immunology and Immunotherapy, Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (A.G.); (H.D.)
| |
Collapse
|
9
|
Gao W, Chen Y, Yang J, Zhuo C, Huang S, Zhang H, Shi Y. Clinical Perspectives on Liquid Biopsy in Metastatic Colorectal Cancer. Front Genet 2021; 12:634642. [PMID: 33584829 PMCID: PMC7876389 DOI: 10.3389/fgene.2021.634642] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/04/2021] [Indexed: 02/05/2023] Open
Abstract
Liquid biopsy, which generally refers to the analysis of biological components such as circulating nuclear acids and circulating tumor cells in body fluids, particularly in peripheral blood, has shown good capacity to overcome several limitations faced by conventional tissue biopsies. Emerging evidence in recent decades has confirmed the promising role of liquid biopsy in the clinical management of various cancers, including colorectal cancer, which is one of the most prevalent cancers and the second leading cause of cancer-related deaths worldwide. Despite the challenges and poor clinical outcomes, patients with metastatic colorectal cancer can expect potential clinical benefits with liquid biopsy. Therefore, in this review, we focus on the clinical prospects of liquid biopsy in metastatic colorectal cancer, specifically with regard to the recently discovered various biomarkers identified on liquid biopsy. These biomarkers have been shown to be potentially useful in multiple aspects of metastatic colorectal cancer, such as auxiliary diagnosis of metastasis, prognosis prediction, and monitoring of therapy response.
Collapse
Affiliation(s)
- Wei Gao
- Department of Internal Medicine-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Yigui Chen
- Department of Internal Medicine-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Jianwei Yang
- Department of Internal Medicine-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Changhua Zhuo
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Sha Huang
- Department of Internal Medicine-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Hui Zhang
- Department of Hepatopancreatobiliary Surgical Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Yi Shi
- Department of Molecular Pathology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| |
Collapse
|
10
|
Wu G, Zhang M. A novel risk score model based on eight genes and a nomogram for predicting overall survival of patients with osteosarcoma. BMC Cancer 2020; 20:456. [PMID: 32448271 PMCID: PMC7245838 DOI: 10.1186/s12885-020-06741-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/12/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND This study aims to identify a predictive model to predict survival outcomes of osteosarcoma (OS) patients. METHODS A RNA sequencing dataset (the training set) and a microarray dataset (the validation set) were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database, respectively. Differentially expressed genes (DEGs) between metastatic and non-metastatic OS samples were identified in training set. Prognosis-related DEGs were screened and optimized by support vector machine (SVM) recursive feature elimination. A SVM classifier was built to classify metastatic and non-metastatic OS samples. Independent prognosic genes were extracted by multivariate regression analysis to build a risk score model followed by performance evaluation in two datasets by Kaplan-Meier (KM) analysis. Independent clinical prognostic indicators were identified followed by nomogram analysis. Finally, functional analyses of survival-related genes were conducted. RESULT Totally, 345 DEGs and 45 prognosis-related genes were screened. A SVM classifier could distinguish metastatic and non-metastatic OS samples. An eight-gene signature was an independent prognostic marker and used for constructing a risk score model. The risk score model could separate OS samples into high and low risk groups in two datasets (training set: log-rank p < 0.01, C-index = 0.805; validation set: log-rank p < 0.01, C-index = 0.797). Tumor metastasis and RS model status were independent prognostic factors and nomogram model exhibited accurate survival prediction for OS. Additionally, functional analyses of survival-related genes indicated they were closely associated with immune responses and cytokine-cytokine receptor interaction pathway. CONCLUSION An eight-gene predictive model and nomogram were developed to predict OS prognosis.
Collapse
Affiliation(s)
- Guangzhi Wu
- Departments of Hand Surgery, The Third Hospital of Jilin University, Changchun, Jilin Province China
| | - Minglei Zhang
- Departments of Orthopedics, The Third Hospital of Jilin University, Changchun, Jilin Province China
| |
Collapse
|
11
|
Li W, Deng G, Zhang J, Hu E, He Y, Lv J, Sun X, Wang K, Chen L. Identification of breast cancer risk modules via an integrated strategy. Aging (Albany NY) 2019; 11:12131-12146. [PMID: 31860871 PMCID: PMC6949069 DOI: 10.18632/aging.102546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/19/2019] [Indexed: 12/17/2022]
Abstract
Breast cancer is one of the most common malignant cancers among females worldwide. This complex disease is not caused by a single gene, but resulted from multi-gene interactions, which could be represented by biological networks. Network modules are composed of genes with significant similarities in terms of expression, function and disease association. Therefore, the identification of disease risk modules could contribute to understanding the molecular mechanisms underlying breast cancer. In this paper, an integrated disease risk module identification strategy was proposed according to a multi-objective programming model for two similarity criteria as well as significance of permutation tests in Markov random field module score, function consistency score and Pearson correlation coefficient difference score. Three breast cancer risk modules were identified from a breast cancer-related interaction network. Genes in these risk modules were confirmed to play critical roles in breast cancer by literature review. These risk modules were enriched in breast cancer-related pathways or functions and could distinguish between breast tumor and normal samples with high accuracy for not only the microarray dataset used for breast cancer risk module identification, but also another two independent datasets. Our integrated strategy could be extended to other complex diseases to identify their risk modules and reveal their pathogenesis.
Collapse
Affiliation(s)
- Wan Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Gui Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Ji Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Erqiang Hu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yuehan He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Junjie Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xilin Sun
- Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, the Fourth Hospital of Harbin Medical University, Harbin, China
| | - Kai Wang
- Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, the Fourth Hospital of Harbin Medical University, Harbin, China
| | - Lina Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
12
|
Sun X, Sun S, Yang S. An Efficient and Flexible Method for Deconvoluting Bulk RNA-Seq Data with Single-Cell RNA-Seq Data. Cells 2019; 8:E1161. [PMID: 31569701 PMCID: PMC6830085 DOI: 10.3390/cells8101161] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 09/23/2019] [Accepted: 09/26/2019] [Indexed: 12/25/2022] Open
Abstract
Estimating cell type compositions for complex diseases is an important step to investigate the cellular heterogeneity for understanding disease etiology and potentially facilitate early disease diagnosis and prevention. Here, we developed a computationally statistical method, referring to Multi-Omics Matrix Factorization (MOMF), to estimate the cell-type compositions of bulk RNA sequencing (RNA-seq) data by leveraging cell type-specific gene expression levels from single-cell RNA sequencing (scRNA-seq) data. MOMF not only directly models the count nature of gene expression data, but also effectively accounts for the uncertainty of cell type-specific mean gene expression levels. We demonstrate the benefits of MOMF through three real data applications, i.e., Glioblastomas (GBM), colorectal cancer (CRC) and type II diabetes (T2D) studies. MOMF is able to accurately estimate disease-related cell type proportions, i.e., oligodendrocyte progenitor cells and macrophage cells, which are strongly associated with the survival of GBM and CRC, respectively.
Collapse
Affiliation(s)
- Xifang Sun
- Department of Mathematics, School of Science, Xi'an Shiyou University, 710065 Xi'an, China.
| | - Shiquan Sun
- School of Computer Science, Northwestern Polytechnical University, 710072 Xi'an, China.
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Sheng Yang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, 211166 Nanjing, China.
| |
Collapse
|