1
|
Contreras M, Sobrino I, de la Fuente J. Paratransgenic quantum vaccinology. Trends Parasitol 2024:S1471-4922(24)00295-2. [PMID: 39462754 DOI: 10.1016/j.pt.2024.10.006] [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: 09/26/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/29/2024]
Abstract
Tick vaccines are an environmentally friendly intervention for the prevention and control of tick-borne diseases affecting humans and animals worldwide. From our perspective, the challenges in tick vaccinology have encouraged the implementation of new interventions. In this opinion article we propose paratransgenic quantum vaccinology as a new approach that integrates platform trends in biotechnology, such as omics datasets combined with big data analytics, machine learning, and paratransgenesis with a systems biology perspective. This innovative approach allows the identification of protective epitopes in tick- and/or pathogen-derived proteins for the design of chimeric vaccine candidate antigens which can be produced by commensal/symbiotic microorganisms eliciting a protective response in the host.
Collapse
Affiliation(s)
- Marinela Contreras
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain.
| | - Isidro Sobrino
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - José de la Fuente
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain; Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA.
| |
Collapse
|
2
|
Mei B, Jiang Y, Sun Y. Unveiling Commonalities and Differences in Genetic Regulations via Two-Way Fusion. J Comput Biol 2024; 31:834-870. [PMID: 39133672 DOI: 10.1089/cmb.2023.0437] [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] [Indexed: 09/10/2024] Open
Abstract
Understanding the genetic regulation, for example, gene expressions (GEs) by copy number variations and methylations, is crucial to uncover the development and progression of complex diseases. Advancing from early studies that are mostly focused on homogeneous groups of patients, some recent studies have shifted their focus toward different patient groups, explored their commonalities and differences, and led to insightful findings. However, the analysis can be very challenging with one GE possibly regulated by multiple regulators and one regulator potentially regulating the expressions of multiple genes, leading to two distinct types of commonalities/differences in the patterns of genetic regulation. In addition, the high dimensionality of both sides of regulation poses challenges to computation. In this study, we develop a two-way fusion integrative analysis approach, which innovatively applies two fusion penalties to simultaneously identify commonalities/differences in the regulated pattern of GEs and regulating pattern of regulators, and adopt a Huber loss function to accommodate the possible data contamination. Moreover, a simple yet efficient iterative optimization algorithm is developed, which does not need to introduce any auxiliary variables and extra tuning parameters and is guaranteed to converge to a globally optimal solution. The advantages of the proposed approach are demonstrated in extensive simulations. The analysis of The Cancer Genome Atlas data on melanoma and lung cancer leads to interesting findings and satisfactory prediction performance.
Collapse
Affiliation(s)
- Biao Mei
- Center for Applied Statistics, School of Statistics, Renmin University of China, Beijing, China
| | - Yu Jiang
- School of Public Health, University of Memphis, Memphis, Tennessee, USA
| | - Yifan Sun
- Center for Applied Statistics, School of Statistics, Renmin University of China, Beijing, China
- Beijing Advanced Innovation Center for Future Blockchain and Privacy Computing, Beijing, China
| |
Collapse
|
3
|
Nguyen H, Pham VD, Nguyen H, Tran B, Petereit J, Nguyen T. CCPA: cloud-based, self-learning modules for consensus pathway analysis using GO, KEGG and Reactome. Brief Bioinform 2024; 25:bbae222. [PMID: 39041916 PMCID: PMC11264295 DOI: 10.1093/bib/bbae222] [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: 11/22/2023] [Revised: 03/15/2024] [Accepted: 04/25/2024] [Indexed: 07/24/2024] Open
Abstract
This manuscript describes the development of a resource module that is part of a learning platform named 'NIGMS Sandbox for Cloud-based Learning' (https://github.com/NIGMS/NIGMS-Sandbox). The module delivers learning materials on Cloud-based Consensus Pathway Analysis in an interactive format that uses appropriate cloud resources for data access and analyses. Pathway analysis is important because it allows us to gain insights into biological mechanisms underlying conditions. But the availability of many pathway analysis methods, the requirement of coding skills, and the focus of current tools on only a few species all make it very difficult for biomedical researchers to self-learn and perform pathway analysis efficiently. Furthermore, there is a lack of tools that allow researchers to compare analysis results obtained from different experiments and different analysis methods to find consensus results. To address these challenges, we have designed a cloud-based, self-learning module that provides consensus results among established, state-of-the-art pathway analysis techniques to provide students and researchers with necessary training and example materials. The training module consists of five Jupyter Notebooks that provide complete tutorials for the following tasks: (i) process expression data, (ii) perform differential analysis, visualize and compare the results obtained from four differential analysis methods (limma, t-test, edgeR, DESeq2), (iii) process three pathway databases (GO, KEGG and Reactome), (iv) perform pathway analysis using eight methods (ORA, CAMERA, KS test, Wilcoxon test, FGSEA, GSA, SAFE and PADOG) and (v) combine results of multiple analyses. We also provide examples, source code, explanations and instructional videos for trainees to complete each Jupyter Notebook. The module supports the analysis for many model (e.g. human, mouse, fruit fly, zebra fish) and non-model species. The module is publicly available at https://github.com/NIGMS/Consensus-Pathway-Analysis-in-the-Cloud. This manuscript describes the development of a resource module that is part of a learning platform named ``NIGMS Sandbox for Cloud-based Learning'' https://github.com/NIGMS/NIGMS-Sandbox. The overall genesis of the Sandbox is described in the editorial NIGMS Sandbox [1] at the beginning of this Supplement. This module delivers learning materials on the analysis of bulk and single-cell ATAC-seq data in an interactive format that uses appropriate cloud resources for data access and analyses.
Collapse
Affiliation(s)
- Ha Nguyen
- Department of Computer Science and Software Engineering, Auburn University, AL 36849, USA
| | - Van-Dung Pham
- Department of Computer Science and Software Engineering, Auburn University, AL 36849, USA
| | - Hung Nguyen
- Department of Computer Science and Software Engineering, Auburn University, AL 36849, USA
| | - Bang Tran
- Department of Computer Science, California State University, Sacramento, CA 95819, USA
| | - Juli Petereit
- Nevada Bioinformatics Center, University of Nevada, Reno, NV 89557, USA
| | - Tin Nguyen
- Department of Computer Science and Software Engineering, Auburn University, AL 36849, USA
| |
Collapse
|
4
|
Ahmed F, Samantasinghar A, Manzoor Soomro A, Kim S, Hyun Choi K. A systematic review of computational approaches to understand cancer biology for informed drug repurposing. J Biomed Inform 2023; 142:104373. [PMID: 37120047 DOI: 10.1016/j.jbi.2023.104373] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/25/2023] [Accepted: 04/23/2023] [Indexed: 05/01/2023]
Abstract
Cancer is the second leading cause of death globally, trailing only heart disease. In the United States alone, 1.9 million new cancer cases and 609,360 deaths were recorded for 2022. Unfortunately, the success rate for new cancer drug development remains less than 10%, making the disease particularly challenging. This low success rate is largely attributed to the complex and poorly understood nature of cancer etiology. Therefore, it is critical to find alternative approaches to understanding cancer biology and developing effective treatments. One such approach is drug repurposing, which offers a shorter drug development timeline and lower costs while increasing the likelihood of success. In this review, we provide a comprehensive analysis of computational approaches for understanding cancer biology, including systems biology, multi-omics, and pathway analysis. Additionally, we examine the use of these methods for drug repurposing in cancer, including the databases and tools that are used for cancer research. Finally, we present case studies of drug repurposing, discussing their limitations and offering recommendations for future research in this area.
Collapse
Affiliation(s)
- Faheem Ahmed
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea
| | | | | | - Sejong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| | - Kyung Hyun Choi
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea.
| |
Collapse
|
5
|
Dorraki M, Muratovic D, Fouladzadeh A, Verjans JW, Allison A, Findlay DM, Abbott D. Hip osteoarthritis: A novel network analysis of subchondral trabecular bone structures. PNAS NEXUS 2022; 1:pgac258. [PMID: 36712355 PMCID: PMC9802325 DOI: 10.1093/pnasnexus/pgac258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/26/2022] [Accepted: 11/09/2022] [Indexed: 11/23/2022]
Abstract
Hip osteoarthritis (HOA) is a degenerative joint disease that leads to the progressive destruction of subchondral bone and cartilage at the hip joint. Development of effective treatments for HOA remains an open problem, primarily due to the lack of knowledge of its pathogenesis and a typically late-stage diagnosis. We describe a novel network analysis methodology for microcomputed tomography (micro-CT) images of human trabecular bone. We explored differences between the trabecular bone microstructure of femoral heads with and without HOA. Large-scale automated extraction of the network formed by trabecular bone revealed significant network properties not previously reported for bone. Profound differences were discovered, particularly in the proximal third of the femoral head, where HOA networks demonstrated elevated numbers of edges, vertices, and graph components. When further differentiating healthy joint and HOA networks, the latter showed fewer small-world network properties, due to decreased clustering coefficient and increased characteristic path length. Furthermore, we found that HOA networks had reduced length of edges, indicating the formation of compressed trabecular structures. In order to assess our network approach, we developed a deep learning model for classifying HOA and control cases, and we fed it with two separate inputs: (i) micro-CT images of the trabecular bone, and (ii) the network extracted from them. The model with plain micro-CT images achieves 74.6% overall accuracy while the trained model with extracted networks attains 96.5% accuracy. We anticipate our findings to be a starting point for a novel description of bone microstructure in HOA, by considering the phenomenon from a graph theory viewpoint.
Collapse
Affiliation(s)
| | | | - Anahita Fouladzadeh
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Johan W Verjans
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia,Australian Institute for Machine Learning (AIML), The University of Adelaide, Adelaide, SA 5000, Australia,Royal Adelaide Hospital, Adelaide, SA 5000, Australia,Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Andrew Allison
- School of Electrical and Electronic Engineering, The University of Adelaide, Adelaide, SA 5000, Australia,Centre for Biomedical Engineering (CBME), The University of Adelaide, Adelaide, SA 5000, Australia
| | - David M Findlay
- Centre for Orthopaedic and Trauma Research, Discipline of Orthopaedics and Trauma, The University of Adelaide, Adelaide, SA 5000, Australia,Centre for Biomedical Engineering (CBME), The University of Adelaide, Adelaide, SA 5000, Australia
| | - Derek Abbott
- School of Electrical and Electronic Engineering, The University of Adelaide, Adelaide, SA 5000, Australia,Centre for Biomedical Engineering (CBME), The University of Adelaide, Adelaide, SA 5000, Australia
| |
Collapse
|
6
|
Identification of miRNA-mRNA-TFs regulatory network and crucial pathways involved in asthma through advanced systems biology approaches. PLoS One 2022; 17:e0271262. [PMID: 36264868 PMCID: PMC9584516 DOI: 10.1371/journal.pone.0271262] [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: 04/06/2022] [Accepted: 06/28/2022] [Indexed: 12/07/2022] Open
Abstract
Asthma is a life-threatening and chronic inflammatory lung disease that is posing a true global health challenge. The genetic basis of the disease is fairly well examined. However, the molecular crosstalk between microRNAs (miRNAs), target genes, and transcription factors (TFs) networks and their contribution to disease pathogenesis and progression is not well explored. Therefore, this study was aimed at dissecting the molecular network between mRNAs, miRNAs, and TFs using robust computational biology approaches. The transcriptomic data of bronchial epithelial cells of severe asthma patients and healthy controls was studied by different systems biology approaches like differentially expressed gene detection, functional enrichment, miRNA-target gene pairing, and mRNA-miRNA-TF molecular networking. We detected the differential expression of 1703 (673 up-and 1030 down-regulated) genes and 71 (41 up-and 30 down-regulated) miRNAs in the bronchial epithelial cells of asthma patients. The DEGs were found to be enriched in key pathways like IL-17 signaling (KEGG: 04657), Th1 and Th2 cell differentiation (KEGG: 04658), and the Th17 cell differentiation (KEGG: 04659) (p-values = 0.001). The results from miRNAs-target gene pairs-transcription factors (TFs) have detected the key roles of 3 miRs (miR-181a-2-3p; miR-203a-3p; miR-335-5p), 6 TFs (TFAM, FOXO1, GFI1, IRF2, SOX9, and HLF) and 32 miRNA target genes in eliciting autoimmune reactions in bronchial epithelial cells of the respiratory tract. Through systemic implementation of comprehensive system biology tools, this study has identified key miRNAs, TFs, and miRNA target gene pairs as potential tissue-based asthma biomarkers.
Collapse
|
7
|
Abstract
The growth of solid tumours relies on an ever-increasing supply of oxygen and nutrients that are delivered via vascular networks. Tumour vasculature includes endothelial cell lined angiogenesis and the less common cancer cell lined vasculogenic mimicry (VM). To study and compare the development of vascular networks formed during angiogenesis and VM (represented here by breast cancer and pancreatic cancer cell lines) a number of in vitro assays were utilised. From live cell imaging, we performed a large-scale automated extraction of network parameters and identified properties not previously reported. We show that for both angiogenesis and VM, the characteristic network path length reduces over time; however, only endothelial cells increase network clustering coefficients thus maintaining small-world network properties as they develop. When compared to angiogenesis, the VM network efficiency is improved by decreasing the number of edges and vertices, and also by increasing edge length. Furthermore, our results demonstrate that angiogenic and VM networks appear to display similar properties to road traffic networks and are also subject to the well-known Braess paradox. This quantitative measurement framework opens up new avenues to potentially evaluate the impact of anti-cancer drugs and anti-vascular therapies. Fouladzadeh, Dorraki and colleagues investigate the development of angiogenic networks for in vitro cancer cell lines. They demonstrate that during the growth stages of vasculogenic mimicry, the number of edges and vertices decreases but the edge length increases resulting in improved network efficiency.
Collapse
|
8
|
Zhang S, Hu X, Luo Z, Jiang Y, Sun Y, Ma S. Biomarker-guided heterogeneity analysis of genetic regulations via multivariate sparse fusion. Stat Med 2021; 40:3915-3936. [PMID: 33906263 PMCID: PMC8277716 DOI: 10.1002/sim.9006] [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: 04/20/2020] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 11/06/2022]
Abstract
Heterogeneity is a hallmark of many complex diseases. There are multiple ways of defining heterogeneity, among which the heterogeneity in genetic regulations, for example, gene expressions (GEs) by copy number variations (CNVs), and methylation, has been suggested but little investigated. Heterogeneity in genetic regulations can be linked with disease severity, progression, and other traits and is biologically important. However, the analysis can be very challenging with the high dimensionality of both sides of regulation as well as sparse and weak signals. In this article, we consider the scenario where subjects form unknown subgroups, and each subgroup has unique genetic regulation relationships. Further, such heterogeneity is "guided" by a known biomarker. We develop a multivariate sparse fusion (MSF) approach, which innovatively applies the penalized fusion technique to simultaneously determine the number and structure of subgroups and regulation relationships within each subgroup. An effective computational algorithm is developed, and extensive simulations are conducted. The analysis of heterogeneity in the GE-CNV regulations in melanoma and GE-methylation regulations in stomach cancer using the TCGA data leads to interesting findings.
Collapse
Affiliation(s)
- Sanguo Zhang
- School of Mathematical Sciences, and Key Laboratory of Big Data Mining and Knowledge Management, Chinese Academy of Science, Beijing, China
| | - Xiaonan Hu
- School of Mathematical Sciences, and Key Laboratory of Big Data Mining and Knowledge Management, Chinese Academy of Science, Beijing, China
| | - Ziye Luo
- Center for Applied Statistics, School of Statistics, Renmin University of China, Beijing, China
| | - Yu Jiang
- School of Public Health, University of Memphis, Tennessee, USA
| | - Yifan Sun
- Center for Applied Statistics, School of Statistics, Renmin University of China, Beijing, China
| | - Shuangge Ma
- Center for Applied Statistics, School of Statistics, Renmin University of China, Beijing, China
- Department of Biostatistics, Yale University, Connecticut, USA
| |
Collapse
|
9
|
Li YK, Hsu HM, Lin MC, Chang CW, Chu CM, Chang YJ, Yu JC, Chen CT, Jian CE, Sun CA, Chen KH, Kuo MH, Cheng CS, Chang YT, Wu YS, Wu HY, Yang YT, Lin C, Lin HC, Hu JM, Chang YT. Genetic co-expression networks contribute to creating predictive model and exploring novel biomarkers for the prognosis of breast cancer. Sci Rep 2021; 11:7268. [PMID: 33790307 PMCID: PMC8012617 DOI: 10.1038/s41598-021-84995-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Genetic co-expression network (GCN) analysis augments the understanding of breast cancer (BC). We aimed to propose GCN-based modeling for BC relapse-free survival (RFS) prediction and to discover novel biomarkers. We used GCN and Cox proportional hazard regression to create various prediction models using mRNA microarray of 920 tumors and conduct external validation using independent data of 1056 tumors. GCNs of 34 identified candidate genes were plotted in various sizes. Compared to the reference model, the genetic predictors selected from bigger GCNs composed better prediction models. The prediction accuracy and AUC of 3 ~ 15-year RFS are 71.0-81.4% and 74.6-78% respectively (rfm, ACC 63.2-65.5%, AUC 61.9-74.9%). The hazard ratios of risk scores of developing relapse ranged from 1.89 ~ 3.32 (p < 10-8) over all models under the control of the node status. External validation showed the consistent finding. We found top 12 co-expressed genes are relative new or novel biomarkers that have not been explored in BC prognosis or other cancers until this decade. GCN-based modeling creates better prediction models and facilitates novel genes exploration on BC prognosis.
Collapse
Affiliation(s)
- Yuan-Kuei Li
- Division of Colorectal Surgery, Department of Surgery, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan.,Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Huan-Ming Hsu
- Division of General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Surgery, Songshan Branch of Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Meng-Chiung Lin
- Division of Gastroenterology, Department of Medicine, Taichung Armed Forces General Hospital, Taichung, Taiwan
| | - Chi-Wen Chang
- School of Nursing, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Nursing, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Chi-Ming Chu
- Division of Medical Informatics, Department of Epidemiology, School of Public Health, National Defense Medical Center, Taipei, Taiwan.,Big Data Research Center, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan.,Department of Public Health, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan.,Department of Public Health, China Medical University, Taichung City, Taiwan.,Department of Healthcare Administration and Medical Informatics College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Jia Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Jyh-Cherng Yu
- Division of General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Ting Chen
- Division of Medical Informatics, Department of Epidemiology, School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Chen-En Jian
- Division of Medical Informatics, Department of Epidemiology, School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Chien-An Sun
- Big Data Research Center, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Kang-Hua Chen
- School of Nursing, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Nursing, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Ming-Hao Kuo
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Shiang Cheng
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ya-Ting Chang
- Division of Medical Informatics, Department of Epidemiology, School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Syuan Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Hao-Yi Wu
- Division of Medical Informatics, Department of Epidemiology, School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Ya-Ting Yang
- Division of Medical Informatics, Department of Epidemiology, School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Chen Lin
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan.,Center for Biotechnology and Biomedical Engineering, National Central University, Taoyuan, Taiwan
| | - Hung-Che Lin
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Hualien Armed Forces General Hospital, Xincheng, Hualien, 97144, Taiwan
| | - Je-Ming Hu
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan.,School of Medicine, National Defense Medical Center, Taipei City, Taiwan
| | - Yu-Tien Chang
- Division of Medical Informatics, Department of Epidemiology, School of Public Health, National Defense Medical Center, Taipei, Taiwan. .,Big Data Research Center, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
10
|
Suresh NT, E R V, U K. Multi-scale top-down approach for modelling epileptic protein-protein interaction network analysis to identify driver nodes and pathways. Comput Biol Chem 2020; 88:107323. [PMID: 32653778 DOI: 10.1016/j.compbiolchem.2020.107323] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/04/2020] [Accepted: 06/23/2020] [Indexed: 12/23/2022]
Abstract
Protein - Protein Interaction Network (PPIN) analysis unveils molecular level mechanisms involved in disease condition. To explore the complex regulatory mechanisms behind epilepsy and to address the clinical and biological issues of epilepsy, in silico techniques are feasible in a cost- effective manner. In this work, a hierarchical procedure to identify influential genes and regulatory pathways in epilepsy prognosis is proposed. To obtain key genes and pathways causing epilepsy, integration of two benchmarked datasets which are exclusively devoted for complex disorders is done as an initial step. Using STRING database, PPIN is constructed for modelling protein-protein interactions. Further, key interactions are obtained from the established PPIN using network centrality measures followed by network propagation algorithm -Random Walk with Restart (RWR). The outcome of the method reveals some influential genes behind epilepsy prognosis, along with their associated pathways like PI3 kinase, VEGF signaling, Ras, Wnt signaling etc. In comparison with similar works, our results have shown improvement in identifying unique molecular functions, biological processes, gene co-occurrences etc. Also, CORUM provides an annotation for approximately 60% of similarity in human protein complexes with the obtained result. We believe that the formulated strategy can put-up the vast consideration of indigenous drugs towards meticulous identification of genes encoded by protein against several combinatorial disorders.
Collapse
Affiliation(s)
- Nikhila T Suresh
- Department of Computer Science and IT, Amrita School of Arts and Sciences, Amrita Vishwa Vidyapeetham, Kochi Campus, India
| | - Vimina E R
- Department of Computer Science and IT, Amrita School of Arts and Sciences, Amrita Vishwa Vidyapeetham, Kochi Campus, India.
| | - Krishnakumar U
- Department of Computer Science and IT, Amrita School of Arts and Sciences, Amrita Vishwa Vidyapeetham, Kochi Campus, India
| |
Collapse
|
11
|
Ahmed F. Integrated Network Analysis Reveals FOXM1 and MYBL2 as Key Regulators of Cell Proliferation in Non-small Cell Lung Cancer. Front Oncol 2019; 9:1011. [PMID: 31681566 PMCID: PMC6804573 DOI: 10.3389/fonc.2019.01011] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/20/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Loss of control on cell division is an important factor for the development of non-small cell lung cancer (NSCLC), however, its molecular mechanism and gene regulatory network are not clearly understood. This study utilized the systems bioinformatics approach to reveal the “driver-network” involve in tumorigenic processes in NSCLC. Methods: A meta-analysis of gene expression data of NSCLC was integrated with protein-protein interaction (PPI) data to construct an NSCLC network. MCODE and iRegulone were used to identify the local clusters and its upstream transcription regulators involve in NSCLC. Pair-wise gene expression correlation was performed using GEPIA. The survival analysis was performed by the Kaplan-Meier plot. Results: This study identified a local “driver-network” with highest MCODE score having 26 up-regulated genes involved in the process of cell proliferation in NSCLC. Interestingly, the “driver-network” is under the regulation of TFs FOXM1 and MYBL2 as well as miRNAs. Furthermore, the overexpression of member genes in “driver-network” and the TFs are associated with poor overall survival (OS) in NSCLC patients. Conclusion: This study identified a local “driver-network” and its upstream regulators responsible for the cell proliferation in NSCLC, which could be promising biomarkers and therapeutic targets for NSCLC treatment.
Collapse
Affiliation(s)
- Firoz Ahmed
- Department of Biochemistry, University of Jeddah, Jeddah, Saudi Arabia.,University of Jeddah Center for Scientific and Medical Research, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
12
|
Stefania DD, Vergara D. The Many-Faced Program of Epithelial-Mesenchymal Transition: A System Biology-Based View. Front Oncol 2017; 7:274. [PMID: 29181337 PMCID: PMC5694026 DOI: 10.3389/fonc.2017.00274] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/31/2017] [Indexed: 12/16/2022] Open
Abstract
System biology uses a range of experimental and statistical methods to dissect complex processes that results from alterations in biological models. Given the complexity of the epithelial–mesenchymal transition (EMT) program, system biology represents a promising approach to understanding its fine molecular regulation by the interpretation of high-throughput datasets. Herein, we review recent contributions of system biology applied to the field of EMT physiology and illustrate the importance of these approaches to model biological networks that are perturbed during the transition. Together, these results allowed the definition of an EMT signature across different tumor types, the identification of dysregulated processes and new modules of regulation, making possible to reveal the EMT molecular visage underneath.
Collapse
Affiliation(s)
- De Domenico Stefania
- Biotecgen, Department of Biological and Environmental Sciences and Technologies, Lecce, Italy.,Institute of Sciences of Food Production, National Research Council, Lecce, Italy
| | - Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| |
Collapse
|
13
|
Wang H, Luo J, Liu C, Niu H, Wang J, Liu Q, Zhao Z, Xu H, Ding Y, Sun J, Zhang Q. Investigating MicroRNA and transcription factor co-regulatory networks in colorectal cancer. BMC Bioinformatics 2017; 18:388. [PMID: 28865443 PMCID: PMC5581471 DOI: 10.1186/s12859-017-1796-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 08/21/2017] [Indexed: 02/06/2023] Open
Abstract
Background Colorectal cancer (CRC) is one of the most common malignancies worldwide with poor prognosis. Studies have showed that abnormal microRNA (miRNA) expression can affect CRC pathogenesis and development through targeting critical genes in cellular system. However, it is unclear about which miRNAs play central roles in CRC’s pathogenesis and how they interact with transcription factors (TFs) to regulate the cancer-related genes. Results To address this issue, we systematically explored the major regulation motifs, namely feed-forward loops (FFLs), that consist of miRNAs, TFs and CRC-related genes through the construction of a miRNA-TF regulatory network in CRC. First, we compiled CRC-related miRNAs, CRC-related genes, and human TFs from multiple data sources. Second, we identified 13,123 3-node FFLs including 25 miRNA-FFLs, 13,005 TF-FFLs and 93 composite-FFLs, and merged the 3-node FFLs to construct a CRC-related regulatory network. The network consists of three types of regulatory subnetworks (SNWs): miRNA-SNW, TF-SNW, and composite-SNW. To enhance the accuracy of the network, the results were filtered by using The Cancer Genome Atlas (TCGA) expression data in CRC, whereby we generated a core regulatory network consisting of 58 significant FFLs. We then applied a hub identification strategy to the significant FFLs and found 5 significant components, including two miRNAs (hsa-miR-25 and hsa-miR-31), two genes (ADAMTSL3 and AXIN1) and one TF (BRCA1). The follow up prognosis analysis indicated all of the 5 significant components having good prediction of overall survival of CRC patients. Conclusions In summary, we generated a CRC-specific miRNA-TF regulatory network, which is helpful to understand the complex CRC regulatory mechanisms and guide clinical treatment. The discovered 5 regulators might have critical roles in CRC pathogenesis and warrant future investigation. Electronic supplementary material The online version of this article (10.1186/s12859-017-1796-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hao Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Department of Pathology, College of Basic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jiamao Luo
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Department of Pathology, College of Basic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Chun Liu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Department of Pathology, College of Basic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Huilin Niu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Department of Pathology, College of Basic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jing Wang
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Zhongming Zhao
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Hua Xu
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yanqing Ding
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Department of Pathology, College of Basic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jingchun Sun
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Qingling Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China. .,Department of Pathology, College of Basic Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
14
|
Shrestha R, Hodzic E, Sauerwald T, Dao P, Wang K, Yeung J, Anderson S, Vandin F, Haffari G, Collins CC, Sahinalp SC. HIT'nDRIVE: patient-specific multidriver gene prioritization for precision oncology. Genome Res 2017; 27:1573-1588. [PMID: 28768687 PMCID: PMC5580716 DOI: 10.1101/gr.221218.117] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/06/2017] [Indexed: 12/12/2022]
Abstract
Prioritizing molecular alterations that act as drivers of cancer remains a crucial bottleneck in therapeutic development. Here we introduce HIT'nDRIVE, a computational method that integrates genomic and transcriptomic data to identify a set of patient-specific, sequence-altered genes, with sufficient collective influence over dysregulated transcripts. HIT'nDRIVE aims to solve the "random walk facility location" (RWFL) problem in a gene (or protein) interaction network, which differs from the standard facility location problem by its use of an alternative distance measure: "multihitting time," the expected length of the shortest random walk from any one of the set of sequence-altered genes to an expression-altered target gene. When applied to 2200 tumors from four major cancer types, HIT'nDRIVE revealed many potentially clinically actionable driver genes. We also demonstrated that it is possible to perform accurate phenotype prediction for tumor samples by only using HIT'nDRIVE-seeded driver gene modules from gene interaction networks. In addition, we identified a number of breast cancer subtype-specific driver modules that are associated with patients' survival outcome. Furthermore, HIT'nDRIVE, when applied to a large panel of pan-cancer cell lines, accurately predicted drug efficacy using the driver genes and their seeded gene modules. Overall, HIT'nDRIVE may help clinicians contextualize massive multiomics data in therapeutic decision making, enabling widespread implementation of precision oncology.
Collapse
Affiliation(s)
- Raunak Shrestha
- Bioinformatics Training Program, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z4.,Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, British Columbia, Canada V6H 3Z6
| | - Ermin Hodzic
- School of Computing Science, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| | - Thomas Sauerwald
- Computer Laboratory, University of Cambridge, Cambridge CB3 0FD, United Kingdom
| | - Phuong Dao
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20894, USA
| | - Kendric Wang
- Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, British Columbia, Canada V6H 3Z6
| | - Jake Yeung
- Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, British Columbia, Canada V6H 3Z6
| | - Shawn Anderson
- Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, British Columbia, Canada V6H 3Z6
| | - Fabio Vandin
- Department of Information Engineering, University of Padova, 35131 Padova, Italy
| | - Gholamreza Haffari
- Faculty of Information Technology, Monash University, Melbourne 3800, Australia
| | - Colin C Collins
- Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, British Columbia, Canada V6H 3Z6.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada V5Z 1M9
| | - S Cenk Sahinalp
- Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, British Columbia, Canada V6H 3Z6.,School of Computing Science, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6.,School of Informatics and Computing, Indiana University, Bloomington, Indiana 47408, USA
| |
Collapse
|
15
|
Ren W, Li W, Wang D, Hu S, Suo J, Ying X. Combining multi-dimensional data to identify key genes and pathways in gastric cancer. PeerJ 2017; 5:e3385. [PMID: 28603669 PMCID: PMC5463969 DOI: 10.7717/peerj.3385] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 05/06/2017] [Indexed: 12/22/2022] Open
Abstract
Gastric cancer is an aggressive cancer that is often diagnosed late. Early detection and treatment require a better understanding of the molecular pathology of the disease. The present study combined data on gene expression and regulatory levels (microRNA, methylation, copy number) with the aim of identifying key genes and pathways for gastric cancer. Data used in this study was retrieved from The Cancer Genomic Atlas. Differential analyses between gastric cancer and normal tissues were carried out using Limma. Copy number alterations were identified for tumor samples. Bimodal filtering of differentially expressed genes (DEGs) based on regulatory changes was performed to identify candidate genes. Protein–protein interaction networks for candidate genes were generated by Cytoscape software. Gene ontology and pathway analyses were performed, and disease-associated network was constructed using the Agilent literature search plugin on Cytoscape. In total, we identified 3602 DEGs, 251 differentially expressed microRNAs, 604 differential methylation-sites, and 52 copy number altered regions. Three groups of candidate genes controlled by different regulatory mechanisms were screened out. Interaction networks for candidate genes were constructed consisting of 415, 228, and 233 genes, respectively, all of which were enriched in cell cycle, P53 signaling, DNA replication, viral carcinogenesis, HTLV-1 infection, and progesterone mediated oocyte maturation pathways. Nine hub genes (SRC, KAT2B, NR3C1, CDK6, MCM2, PRKDC, BLM, CCNE1, PARK2) were identified that were presumed to be key regulators of the networks; seven of these were shown to be implicated in gastric cancer through disease-associated network construction. The genes and pathways identified in our study may play pivotal roles in gastric carcinogenesis and have clinical significance.
Collapse
Affiliation(s)
- Wu Ren
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, China.,Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Wei Li
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, China
| | - Daguang Wang
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, China
| | - Shuofeng Hu
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jian Suo
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xiaomin Ying
- Beijing Institute of Basic Medical Sciences, Beijing, China
| |
Collapse
|
16
|
Zhang J, Zheng H, Li Y, Li H, Liu X, Qin H, Dong L, Wang D. Coexpression network analysis of the genes regulated by two types of resistance responses to powdery mildew in wheat. Sci Rep 2016; 6:23805. [PMID: 27033636 PMCID: PMC4817125 DOI: 10.1038/srep23805] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/15/2016] [Indexed: 02/06/2023] Open
Abstract
Powdery mildew disease caused by Blumeria graminis f. sp. tritici (Bgt) inflicts severe economic losses in wheat crops. A systematic understanding of the molecular mechanisms involved in wheat resistance to Bgt is essential for effectively controlling the disease. Here, using the diploid wheat Triticum urartu as a host, the genes regulated by immune (IM) and hypersensitive reaction (HR) resistance responses to Bgt were investigated through transcriptome sequencing. Four gene coexpression networks (GCNs) were developed using transcriptomic data generated for 20 T. urartu accessions showing IM, HR or susceptible responses. The powdery mildew resistance regulated (PMRR) genes whose expression was significantly correlated with Bgt resistance were identified, and they tended to be hubs and enriched in six major modules. A wide occurrence of negative regulation of PMRR genes was observed. Three new candidate immune receptor genes (TRIUR3_13045, TRIUR3_01037 and TRIUR3_06195) positively associated with Bgt resistance were discovered. Finally, the involvement of TRIUR3_01037 in Bgt resistance was tentatively verified through cosegregation analysis in a F2 population and functional expression assay in Bgt susceptible leaf cells. This research provides insights into the global network properties of PMRR genes. Potential molecular differences between IM and HR resistance responses to Bgt are discussed.
Collapse
Affiliation(s)
- Juncheng Zhang
- The State Key Laboratory of Plant Cell and chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongyuan Zheng
- The Collaborative Innovation Center for Grain Crops, Henan Agricultural University, Zhengzhou 450002, China
| | - Yiwen Li
- The State Key Laboratory of Plant Cell and chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongjie Li
- The National Key Facility for Crop Gene Resources and Genetic Improvement, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xin Liu
- The State Key Laboratory of Plant Cell and chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Huanju Qin
- The State Key Laboratory of Plant Cell and chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lingli Dong
- The State Key Laboratory of Plant Cell and chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Daowen Wang
- The State Key Laboratory of Plant Cell and chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- The Collaborative Innovation Center for Grain Crops, Henan Agricultural University, Zhengzhou 450002, China
| |
Collapse
|
17
|
Identifying Cancer Subtypes from miRNA-TF-mRNA Regulatory Networks and Expression Data. PLoS One 2016; 11:e0152792. [PMID: 27035433 PMCID: PMC4818025 DOI: 10.1371/journal.pone.0152792] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 03/18/2016] [Indexed: 12/18/2022] Open
Abstract
Background Identifying cancer subtypes is an important component of the personalised medicine framework. An increasing number of computational methods have been developed to identify cancer subtypes. However, existing methods rarely use information from gene regulatory networks to facilitate the subtype identification. It is widely accepted that gene regulatory networks play crucial roles in understanding the mechanisms of diseases. Different cancer subtypes are likely caused by different regulatory mechanisms. Therefore, there are great opportunities for developing methods that can utilise network information in identifying cancer subtypes. Results In this paper, we propose a method, weighted similarity network fusion (WSNF), to utilise the information in the complex miRNA-TF-mRNA regulatory network in identifying cancer subtypes. We firstly build the regulatory network where the nodes represent the features, i.e. the microRNAs (miRNAs), transcription factors (TFs) and messenger RNAs (mRNAs) and the edges indicate the interactions between the features. The interactions are retrieved from various interatomic databases. We then use the network information and the expression data of the miRNAs, TFs and mRNAs to calculate the weight of the features, representing the level of importance of the features. The feature weight is then integrated into a network fusion approach to cluster the samples (patients) and thus to identify cancer subtypes. We applied our method to the TCGA breast invasive carcinoma (BRCA) and glioblastoma multiforme (GBM) datasets. The experimental results show that WSNF performs better than the other commonly used computational methods, and the information from miRNA-TF-mRNA regulatory network contributes to the performance improvement. The WSNF method successfully identified five breast cancer subtypes and three GBM subtypes which show significantly different survival patterns. We observed that the expression patterns of the features in some miRNA-TF-mRNA sub-networks vary across different identified subtypes. In addition, pathway enrichment analyses show that the top pathways involving the most differentially expressed genes in each of the identified subtypes are different. The results would provide valuable information for understanding the mechanisms characterising different cancer subtypes and assist the design of treatment therapies. All datasets and the R scripts to reproduce the results are available online at the website: http://nugget.unisa.edu.au/Thuc/cancersubtypes/.
Collapse
|
18
|
Kumar P, Aggarwal R. An overview of triple-negative breast cancer. Arch Gynecol Obstet 2015; 293:247-69. [PMID: 26341644 DOI: 10.1007/s00404-015-3859-y] [Citation(s) in RCA: 433] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 08/18/2015] [Indexed: 12/22/2022]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is a heterogeneous group of tumors comprising various breast cancers simply defined by the absence of estrogen receptor, progesterone receptor and overexpression of human epidermal growth factor receptor 2 gene. In this review, we discuss the epidemiology, risk factors, clinical characteristics and prognostic variables of TNBC, and present the summary of recommended treatment strategies and all other available treatment options. METHODS We performed a systematic literature search using Medline and selected those articles which seemed relevant for this review. In addition, the ClinicalTrials.gov was also scanned for ongoing trials. RESULTS TNBC accounts for 10-20 % of all invasive breast cancers and has been found to be associated with African-American race, younger age, higher grade and mitotic index, and more advanced stage at diagnosis. Locoregional treatment is similar to other invasive breast cancer subtypes and involves surgery-mastectomy with or without adjuvant radiotherapy or breast conservation followed by adjuvant radiotherapy. Due to lack of drug-targetable receptors, chemotherapy is the only recommended systemic treatment to improve disease outcome. TNBC is sensitive to chemotherapy as demonstrated by high pathological complete response rates achieved after neoadjuvant chemotherapy, and this approach also allows for breast-conserving surgery. The peak risk of relapse is at 3 years after surgery, thereafter recurrence risk rapidly decreases. Survival after metastatic relapse is shorter as compared to other breast cancer subtypes, treatment options are few and response rates are poor and lack durability. Important molecular characteristics have now been identified that can subdivide this group of breast cancers further and can provide alternative systemic therapies. CONCLUSIONS To improve therapeutic outcome of TNBC, reliable predictive biomarkers and newer drugs against the known molecular pathways are required.
Collapse
Affiliation(s)
- Pankaj Kumar
- Department of Radiation Oncology, Max Super Speciality Hospital, Phase-6, Mohali, 160055, Punjab, India.
| | - Rupali Aggarwal
- Department of Radiation Oncology, Indus Super Speciality Hospital, Phase-1, Mohali, 160055, Punjab, India
| |
Collapse
|
19
|
Verbeke LPC, Van den Eynden J, Fierro AC, Demeester P, Fostier J, Marchal K. Pathway Relevance Ranking for Tumor Samples through Network-Based Data Integration. PLoS One 2015. [PMID: 26217958 PMCID: PMC4517887 DOI: 10.1371/journal.pone.0133503] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The study of cancer, a highly heterogeneous disease with different causes and clinical outcomes, requires a multi-angle approach and the collection of large multi-omics datasets that, ideally, should be analyzed simultaneously. We present a new pathway relevance ranking method that is able to prioritize pathways according to the information contained in any combination of tumor related omics datasets. Key to the method is the conversion of all available data into a single comprehensive network representation containing not only genes but also individual patient samples. Additionally, all data are linked through a network of previously identified molecular interactions. We demonstrate the performance of the new method by applying it to breast and ovarian cancer datasets from The Cancer Genome Atlas. By integrating gene expression, copy number, mutation and methylation data, the method's potential to identify key pathways involved in breast cancer development shared by different molecular subtypes is illustrated. Interestingly, certain pathways were ranked equally important for different subtypes, even when the underlying (epi)-genetic disturbances were diverse. Next to prioritizing universally high-scoring pathways, the pathway ranking method was able to identify subtype-specific pathways. Often the score of a pathway could not be motivated by a single mutation, copy number or methylation alteration, but rather by a combination of genetic and epi-genetic disturbances, stressing the need for a network-based data integration approach. The analysis of ovarian tumors, as a function of survival-based subtypes, demonstrated the method's ability to correctly identify key pathways, irrespective of tumor subtype. A differential analysis of survival-based subtypes revealed several pathways with higher importance for the bad-outcome patient group than for the good-outcome patient group. Many of the pathways exhibiting higher importance for the bad-outcome patient group could be related to ovarian tumor proliferation and survival.
Collapse
Affiliation(s)
- Lieven P. C. Verbeke
- Department of Information Technology, Ghent University—iMinds, Ghent, Belgium
- * E-mail: (LPCV); (JF); (KM)
| | - Jimmy Van den Eynden
- Department of Information Technology, Ghent University—iMinds, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Ana Carolina Fierro
- Department of Information Technology, Ghent University—iMinds, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Piet Demeester
- Department of Information Technology, Ghent University—iMinds, Ghent, Belgium
| | - Jan Fostier
- Department of Information Technology, Ghent University—iMinds, Ghent, Belgium
- * E-mail: (LPCV); (JF); (KM)
| | - Kathleen Marchal
- Department of Information Technology, Ghent University—iMinds, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- * E-mail: (LPCV); (JF); (KM)
| |
Collapse
|
20
|
Empirical inference of circuitry and plasticity in a kinase signaling network. Proc Natl Acad Sci U S A 2015; 112:7719-24. [PMID: 26060313 DOI: 10.1073/pnas.1423344112] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Our understanding of physiology and disease is hampered by the difficulty of measuring the circuitry and plasticity of signaling networks that regulate cell biology, and how these relate to phenotypes. Here, using mass spectrometry-based phosphoproteomics, we systematically characterized the topology of a network comprising the PI3K/Akt/mTOR and MEK/ERK signaling axes and confirmed its biological relevance by assessing its dynamics upon EGF and IGF1 stimulation. Measuring the activity of this network in models of acquired drug resistance revealed that cells chronically treated with PI3K or mTORC1/2 inhibitors differed in the way their networks were remodeled. Unexpectedly, we also observed a degree of heterogeneity in the network state between cells resistant to the same inhibitor, indicating that even identical and carefully controlled experimental conditions can give rise to the evolution of distinct kinase network statuses. These data suggest that the initial conditions of the system do not necessarily determine the mechanism by which cancer cells become resistant to PI3K/mTOR targeted therapies. The patterns of signaling network activity observed in the resistant cells mirrored the patterns of response to several drug combination treatments, suggesting that the activity of the defined signaling network truly reflected the evolved phenotypic diversity.
Collapse
|
21
|
Caiazza F, McGowan PM, Mullooly M, Murray A, Synnott N, O'Donovan N, Flanagan L, Tape CJ, Murphy G, Crown J, Duffy MJ. Targeting ADAM-17 with an inhibitory monoclonal antibody has antitumour effects in triple-negative breast cancer cells. Br J Cancer 2015; 112:1895-903. [PMID: 26010411 PMCID: PMC4580380 DOI: 10.1038/bjc.2015.163] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/30/2015] [Accepted: 04/08/2015] [Indexed: 12/31/2022] Open
Abstract
Background: Identification and validation of a targeted therapy for triple-negative breast cancer (TNBC), that is, breast cancers negative for oestrogen receptors, progesterone receptors and HER2 amplification, is currently one of the most urgent problems in breast cancer treatment. EGFR is one of the best-validated driver genes for TNBC. EGFR is normally activated following the release of ligands such as TGFα, mediated by the two MMP-like proteases ADAM (a disintegrin and metalloproteinase)-10 and ADAM-17. The aim of this study was to investigate the antitumour effects of a monoclonal antibody against ADAM-17 on an in vitro model of TNBC. Methods: We investigated an inhibitory cross-domain humanised monoclonal antibody targeting both the catalytic domain and the cysteine-rich domain of ADAM17-D1(A12) in the HCC1937 and HCC1143 cell lines. Results: D1(A12) was found to significantly inhibit the release of TGFα, and to decrease downstream EGFR-dependent cell signalling. D1(A12) treatment reduced proliferation in two-dimensional clonogenic assays, as well as growth in three-dimensional culture. Furthermore, D1(A12) reduced invasion of HCC1937 cells and decreased migration of HCC1143 cells. Finally, D1(A12) enhanced cell death in HCC1143 cells. Conclusion: Our in vitro findings suggest that targeting ADAM-17 with D1(A12) may have anticancer activity in TNBC cells.
Collapse
Affiliation(s)
- F Caiazza
- UCD School of Medicine and Medical Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - P M McGowan
- UCD School of Medicine and Medical Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - M Mullooly
- UCD School of Medicine and Medical Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - A Murray
- UCD School of Medicine and Medical Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - N Synnott
- UCD School of Medicine and Medical Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - N O'Donovan
- National Institute for Cellular Biotechnology (NICB), Dublin City University, Dublin, Ireland
| | - L Flanagan
- UCD School of Medicine and Medical Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - C J Tape
- 1] Department of Oncology, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK [2] Cell Communication Team, Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - G Murphy
- Department of Oncology, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - J Crown
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - M J Duffy
- 1] UCD School of Medicine and Medical Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland [2] UCD Clinical Research Centre, St Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
22
|
Identification of Novel Breast Cancer Subtype-Specific Biomarkers by Integrating Genomics Analysis of DNA Copy Number Aberrations and miRNA-mRNA Dual Expression Profiling. BIOMED RESEARCH INTERNATIONAL 2015; 2015:746970. [PMID: 25961039 PMCID: PMC4413257 DOI: 10.1155/2015/746970] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 09/15/2014] [Accepted: 09/22/2014] [Indexed: 12/11/2022]
Abstract
Breast cancer is a heterogeneous disease with well-defined molecular subtypes. Currently, comparative genomic hybridization arrays (aCGH) techniques have been developed rapidly, and recent evidences in studies of breast cancer suggest that tumors within gene expression subtypes share similar DNA copy number aberrations (CNA) which can be used to further subdivide subtypes. Moreover, subtype-specific miRNA expression profiles are also proposed as novel signatures for breast cancer classification. The identification of mRNA or miRNA expression-based breast cancer subtypes is considered an instructive means of prognosis. Here, we conducted an integrated analysis based on copy number aberrations data and miRNA-mRNA dual expression profiling data to identify breast cancer subtype-specific biomarkers. Interestingly, we found a group of genes residing in subtype-specific CNA regions that also display the corresponding changes in mRNAs levels and their target miRNAs' expression. Among them, the predicted direct correlation of BRCA1-miR-143-miR-145 pairs was selected for experimental validation. The study results indicated that BRCA1 positively regulates miR-143-miR-145 expression and miR-143-miR-145 can serve as promising novel biomarkers for breast cancer subtyping. In our integrated genomics analysis and experimental validation, a new frame to predict candidate biomarkers of breast cancer subtype is provided and offers assistance in order to understand the potential disease etiology of the breast cancer subtypes.
Collapse
|
23
|
Subramanian N, Torabi-Parizi P, Gottschalk RA, Germain RN, Dutta B. Network representations of immune system complexity. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2015; 7:13-38. [PMID: 25625853 PMCID: PMC4339634 DOI: 10.1002/wsbm.1288] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 12/09/2014] [Accepted: 12/11/2014] [Indexed: 12/25/2022]
Abstract
The mammalian immune system is a dynamic multiscale system composed of a hierarchically organized set of molecular, cellular, and organismal networks that act in concert to promote effective host defense. These networks range from those involving gene regulatory and protein–protein interactions underlying intracellular signaling pathways and single‐cell responses to increasingly complex networks of in vivo cellular interaction, positioning, and migration that determine the overall immune response of an organism. Immunity is thus not the product of simple signaling events but rather nonlinear behaviors arising from dynamic, feedback‐regulated interactions among many components. One of the major goals of systems immunology is to quantitatively measure these complex multiscale spatial and temporal interactions, permitting development of computational models that can be used to predict responses to perturbation. Recent technological advances permit collection of comprehensive datasets at multiple molecular and cellular levels, while advances in network biology support representation of the relationships of components at each level as physical or functional interaction networks. The latter facilitate effective visualization of patterns and recognition of emergent properties arising from the many interactions of genes, molecules, and cells of the immune system. We illustrate the power of integrating ‘omics’ and network modeling approaches for unbiased reconstruction of signaling and transcriptional networks with a focus on applications involving the innate immune system. We further discuss future possibilities for reconstruction of increasingly complex cellular‐ and organism‐level networks and development of sophisticated computational tools for prediction of emergent immune behavior arising from the concerted action of these networks. WIREs Syst Biol Med 2015, 7:13–38. doi: 10.1002/wsbm.1288 This article is categorized under:
Analytical and Computational Methods > Computational Methods Laboratory Methods and Technologies > Macromolecular Interactions, Methods
Collapse
Affiliation(s)
- Naeha Subramanian
- Institute for Systems Biology, Seattle, WA, USA; Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
24
|
Mitra R, Zhao Z. The oncogenic and prognostic potential of eight microRNAs identified by a synergetic regulatory network approach in lung cancer. ACTA ACUST UNITED AC 2014; 7:384-93. [PMID: 25539849 DOI: 10.1504/ijcbdd.2014.066572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Transcription factors (TFs) and microRNAs (miRNAs), the two main gene regulators in the biological system, control the gene expression at the transcriptional and post-transcriptional level, respectively. However, little is known regarding whether the miRNATF co-regulatory mechanisms, predicted by several studies, truly reflect the molecular interactions in cellular systems. To tackle this important issue, we developed an integrative framework by utilising four independent miRNA and matched mRNA expression profiling datasets to identify reproducible regulations, and demonstrated this approach in non-small cell lung cancer (NSCLC). Our analyses pinpointed several reproducible miRNA-TF co-regulatory networks in NSCLC from which we systematically prioritised eight hub miRNAs that may have strong oncogenic characteristics. Here, we discussed the major findings of our study and explored the oncogenic and prognostic potential of eight prioritised miRNAs through literature-mining based analysis and patient survival analysis. The findings provide additional insights into the miRNA-TF co-regulation in lung cancer.
Collapse
Affiliation(s)
- Ramkrishna Mitra
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Zhongming Zhao
- Departments of Biomedical Informatics, Psychiatry, and Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| |
Collapse
|
25
|
Fisher CP, Kierzek AM, Plant NJ, Moore JB. Systems biology approaches for studying the pathogenesis of non-alcoholic fatty liver disease. World J Gastroenterol 2014; 20:15070-15078. [PMID: 25386055 PMCID: PMC4223240 DOI: 10.3748/wjg.v20.i41.15070] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 03/13/2014] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a progressive disease of increasing public health concern. In western populations the disease has an estimated prevalence of 20%-40%, rising to 70%-90% in obese and type II diabetic individuals. Simplistically, NAFLD is the macroscopic accumulation of lipid in the liver, and is viewed as the hepatic manifestation of the metabolic syndrome. However, the molecular mechanisms mediating both the initial development of steatosis and its progression through non-alcoholic steatohepatitis to debilitating and potentially fatal fibrosis and cirrhosis are only partially understood. Despite increased research in this field, the development of non-invasive clinical diagnostic tools and the discovery of novel therapeutic targets has been frustratingly slow. We note that, to date, NAFLD research has been dominated by in vivo experiments in animal models and human clinical studies. Systems biology tools and novel computational simulation techniques allow the study of large-scale metabolic networks and the impact of their dysregulation on health. Here we review current systems biology tools and discuss the benefits to their application to the study of NAFLD. We propose that a systems approach utilising novel in silico modelling and simulation techniques is key to a more comprehensive, better targeted NAFLD research strategy. Such an approach will accelerate the progress of research and vital translation into clinic.
Collapse
|
26
|
Mitra R, Edmonds MD, Sun J, Zhao M, Yu H, Eischen CM, Zhao Z. Reproducible combinatorial regulatory networks elucidate novel oncogenic microRNAs in non-small cell lung cancer. RNA (NEW YORK, N.Y.) 2014; 20:1356-68. [PMID: 25024357 PMCID: PMC4138319 DOI: 10.1261/rna.042754.113] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 05/01/2014] [Indexed: 06/03/2023]
Abstract
While previous studies reported aberrant expression of microRNAs (miRNAs) in non-small cell lung cancer (NSCLC), little is known about which miRNAs play central roles in NSCLC's pathogenesis and its regulatory mechanisms. To address this issue, we presented a robust computational framework that integrated matched miRNA and mRNA expression profiles in NSCLC using feed-forward loops. The network consists of miRNAs, transcription factors (TFs), and their common predicted target genes. To discern the biological meaning of their associations, we introduced the direction of regulation. A network edge validation strategy using three independent NSCLC expression profiling data sets pinpointed reproducible biological regulations. Reproducible regulation, which may reflect the true molecular interaction, has not been applied to miRNA-TF co-regulatory network analyses in cancer or other diseases yet. We revealed eight hub miRNAs that connected to a higher proportion of targets validated by independent data sets. Network analyses showed that these miRNAs might have strong oncogenic characteristics. Furthermore, we identified a novel miRNA-TF co-regulatory module that potentially suppresses the tumor suppressor activity of the TGF-β pathway by targeting a core pathway molecule (TGFBR2). Follow-up experiments showed two miRNAs (miR-9-5p and miR-130b-3p) in this module had increased expression while their target gene TGFBR2 had decreased expression in a cohort of human NSCLC. Moreover, we demonstrated these two miRNAs directly bind to the 3' untranslated region of TGFBR2. This study enhanced our understanding of miRNA-TF co-regulatory mechanisms in NSCLC. The combined bioinformatics and validation approach we described can be applied to study other types of diseases.
Collapse
Affiliation(s)
- Ramkrishna Mitra
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Mick D Edmonds
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Jingchun Sun
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Min Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Hui Yu
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Christine M Eischen
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Zhongming Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, Tennessee 37212, USA Center for Quantitative Sciences, Vanderbilt University, Nashville, Tennessee 37232, USA
| |
Collapse
|
27
|
Zawadzka AM, Schilling B, Cusack MP, Sahu AK, Drake P, Fisher SJ, Benz CC, Gibson BW. Phosphoprotein secretome of tumor cells as a source of candidates for breast cancer biomarkers in plasma. Mol Cell Proteomics 2014; 13:1034-49. [PMID: 24505115 PMCID: PMC3977182 DOI: 10.1074/mcp.m113.035485] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Breast cancer is a heterogeneous disease whose molecular diversity is not well reflected in clinical and pathological markers used for prognosis and treatment selection. As tumor cells secrete proteins into the extracellular environment, some of these proteins reach circulation and could become suitable biomarkers for improving diagnosis or monitoring response to treatment. As many signaling pathways and interaction networks are altered in cancerous tissues by protein phosphorylation, changes in the secretory phosphoproteome of cancer tissues could reflect both disease progression and subtype. To test this hypothesis, we compared the phosphopeptide-enriched fractions obtained from proteins secreted into conditioned media (CM) derived from five luminal and five basal type breast cancer cell lines using label-free quantitative mass spectrometry. Altogether over 5000 phosphosites derived from 1756 phosphoproteins were identified, several of which have the potential to qualify as phosphopeptide plasma biomarker candidates for the more aggressive basal and also the luminal-type breast cancers. The analysis of phosphopeptides from breast cancer patient plasma and controls allowed us to construct a discovery list of phosphosites under rigorous collection conditions, and second to qualify discovery candidates generated from the CM studies. Indeed, a set of basal-specific phosphorylation CM site candidates derived from IBP3, CD44, OPN, FSTL3, LAMB1, and STC2, and luminal-specific candidates derived from CYTC and IBP5 were selected and, based on their presence in plasma, quantified across all cell line CM samples using Skyline MS1 intensity data. Together, this approach allowed us to assemble a set of novel cancer subtype specific phosphopeptide candidates for subsequent biomarker verification and clinical validation.
Collapse
Affiliation(s)
- Anna M Zawadzka
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, California 94945
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Hortobágyi G, Conte P. Interpreting cancer biology: refining our therapeutic algorithm in breast cancer. Oncologist 2013; 18:e8-10. [PMID: 23633451 DOI: 10.1634/theoncologist.2013-0117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Gabriel Hortobágyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Texas 77230-1439, USA.
| | | |
Collapse
|
29
|
Chen Y, Hao J, Jiang W, He T, Zhang X, Jiang T, Jiang R. Identifying potential cancer driver genes by genomic data integration. Sci Rep 2013; 3:3538. [PMID: 24346768 PMCID: PMC3866686 DOI: 10.1038/srep03538] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 12/02/2013] [Indexed: 12/17/2022] Open
Abstract
Cancer is a genomic disease associated with a plethora of gene mutations resulting in a loss of control over vital cellular functions. Among these mutated genes, driver genes are defined as being causally linked to oncogenesis, while passenger genes are thought to be irrelevant for cancer development. With increasing numbers of large-scale genomic datasets available, integrating these genomic data to identify driver genes from aberration regions of cancer genomes becomes an important goal of cancer genome analysis and investigations into mechanisms responsible for cancer development. A computational method, MAXDRIVER, is proposed here to identify potential driver genes on the basis of copy number aberration (CNA) regions of cancer genomes, by integrating publicly available human genomic data. MAXDRIVER employs several optimization strategies to construct a heterogeneous network, by means of combining a fused gene functional similarity network, gene-disease associations and a disease phenotypic similarity network. MAXDRIVER was validated to effectively recall known associations among genes and cancers. Previously identified as well as novel driver genes were detected by scanning CNAs of breast cancer, melanoma and liver carcinoma. Three predicted driver genes (CDKN2A, AKT1, RNF139) were found common in these three cancers by comparative analysis.
Collapse
Affiliation(s)
- Yong Chen
- 1] National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China [2] MOE Key Laboratory of Bioinformatics and Bioinformatics Division, TNLIST/Department of Automation, Tsinghua University, Beijing 100084, China
| | - Jingjing Hao
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, TNLIST/Department of Automation, Tsinghua University, Beijing 100084, China
| | - Wei Jiang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, TNLIST/Department of Automation, Tsinghua University, Beijing 100084, China
| | - Tong He
- School of Applied Mathematics, Central University of Finance and Economics, Beijing 102206, China
| | - Xuegong Zhang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, TNLIST/Department of Automation, Tsinghua University, Beijing 100084, China
| | - Tao Jiang
- 1] MOE Key Laboratory of Bioinformatics and Bioinformatics Division, TNLIST/Department of Automation, Tsinghua University, Beijing 100084, China [2] Department of Computer Science and Engineering, University of California, Riverside, CA 92521, USA
| | - Rui Jiang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, TNLIST/Department of Automation, Tsinghua University, Beijing 100084, China
| |
Collapse
|
30
|
Udyavar AR, Hoeksema MD, Clark JE, Zou Y, Tang Z, Li Z, Li M, Chen H, Statnikov A, Shyr Y, Liebler DC, Field J, Eisenberg R, Estrada L, Massion PP, Quaranta V. Co-expression network analysis identifies Spleen Tyrosine Kinase (SYK) as a candidate oncogenic driver in a subset of small-cell lung cancer. BMC SYSTEMS BIOLOGY 2013; 7 Suppl 5:S1. [PMID: 24564859 PMCID: PMC4029366 DOI: 10.1186/1752-0509-7-s5-s1] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Oncogenic mechanisms in small-cell lung cancer remain poorly understood leaving this tumor with the worst prognosis among all lung cancers. Unlike other cancer types, sequencing genomic approaches have been of limited success in small-cell lung cancer, i.e., no mutated oncogenes with potential driver characteristics have emerged, as it is the case for activating mutations of epidermal growth factor receptor in non-small-cell lung cancer. Differential gene expression analysis has also produced SCLC signatures with limited application, since they are generally not robust across datasets. Nonetheless, additional genomic approaches are warranted, due to the increasing availability of suitable small-cell lung cancer datasets. Gene co-expression network approaches are a recent and promising avenue, since they have been successful in identifying gene modules that drive phenotypic traits in several biological systems, including other cancer types. Results We derived an SCLC-specific classifier from weighted gene co-expression network analysis (WGCNA) of a lung cancer dataset. The classifier, termed SCLC-specific hub network (SSHN), robustly separates SCLC from other lung cancer types across multiple datasets and multiple platforms, including RNA-seq and shotgun proteomics. The classifier was also conserved in SCLC cell lines. SSHN is enriched for co-expressed signaling network hubs strongly associated with the SCLC phenotype. Twenty of these hubs are actionable kinases with oncogenic potential, among which spleen tyrosine kinase (SYK) exhibits one of the highest overall statistical associations to SCLC. In patient tissue microarrays and cell lines, SCLC can be separated into SYK-positive and -negative. SYK siRNA decreases proliferation rate and increases cell death of SYK-positive SCLC cell lines, suggesting a role for SYK as an oncogenic driver in a subset of SCLC. Conclusions SCLC treatment has thus far been limited to chemotherapy and radiation. Our WGCNA analysis identifies SYK both as a candidate biomarker to stratify SCLC patients and as a potential therapeutic target. In summary, WGCNA represents an alternative strategy to large scale sequencing for the identification of potential oncogenic drivers, based on a systems view of signaling networks. This strategy is especially useful in cancer types where no actionable mutations have emerged.
Collapse
|
31
|
Johansson I, Ringnér M, Hedenfalk I. The landscape of candidate driver genes differs between male and female breast cancer. PLoS One 2013; 8:e78299. [PMID: 24194916 PMCID: PMC3806766 DOI: 10.1371/journal.pone.0078299] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 09/17/2013] [Indexed: 01/05/2023] Open
Abstract
The rapidly growing collection of diverse genome-scale data from multiple tumor types sheds light on various aspects of the underlying tumor biology. With the objective to identify genes of importance for breast tumorigenesis in men and to enable comparisons with genes important for breast cancer development in women, we applied the computational framework COpy Number and EXpression In Cancer (CONEXIC) to detect candidate driver genes among all altered passenger genes. Unique to this approach is that each driver gene is associated with several gene modules that are believed to be altered by the driver. Thirty candidate drivers were found in the male breast cancers and 67 in the female breast cancers. We identified many known drivers of breast cancer and other types of cancer, in the female dataset (e.g. GATA3, CCNE1, GRB7, CDK4). In contrast, only three known cancer genes were found among male breast cancers; MAP2K4, LHP, and ZNF217. Many of the candidate drivers identified are known to be involved in processes associated with tumorigenesis, including proliferation, invasion and differentiation. One of the modules identified in male breast cancer was regulated by THY1, a gene involved in invasion and related to epithelial-mesenchymal transition. Furthermore, men with THY1 positive breast cancers had significantly inferior survival. THY1 may thus be a promising novel prognostic marker for male breast cancer. Another module identified among male breast cancers, regulated by SPAG5, was closely associated with proliferation. Our data indicate that male and female breast cancers display highly different landscapes of candidate driver genes, as only a few genes were found in common between the two. Consequently, the pathobiology of male breast cancer may differ from that of female breast cancer and can be associated with differences in prognosis; men diagnosed with breast cancer may consequently require different management and treatment strategies than women.
Collapse
Affiliation(s)
- Ida Johansson
- Division of Oncology, Department of Clinical Sciences, Lund and CREATE Health Strategic Center for Translational Cancer Research, Lund University, Lund, Sweden
- * E-mail:
| | - Markus Ringnér
- Division of Oncology, Department of Clinical Sciences, Lund and CREATE Health Strategic Center for Translational Cancer Research, Lund University, Lund, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology, Department of Clinical Sciences, Lund and CREATE Health Strategic Center for Translational Cancer Research, Lund University, Lund, Sweden
| |
Collapse
|
32
|
Abstract
High throughput technologies have been applied to investigate the underlying mechanisms of complex diseases, identify disease-associations and help to improve treatment. However it is challenging to derive biological insight from conventional single gene based analysis of "omics" data from high throughput experiments due to sample and patient heterogeneity. To address these challenges, many novel pathway and network based approaches were developed to integrate various "omics" data, such as gene expression, copy number alteration, Genome Wide Association Studies, and interaction data. This review will cover recent methodological developments in pathway analysis for the detection of dysregulated interactions and disease-associated subnetworks, prioritization of candidate disease genes, and disease classifications. For each application, we will also discuss the associated challenges and potential future directions.
Collapse
|
33
|
Kaur H, Mao S, Shah S, Gorski DH, Krawetz SA, Sloane BF, Mattingly RR. Next-generation sequencing: a powerful tool for the discovery of molecular markers in breast ductal carcinoma in situ. Expert Rev Mol Diagn 2013; 13:151-65. [PMID: 23477556 DOI: 10.1586/erm.13.4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mammographic screening leads to frequent biopsies and concomitant overdiagnosis of breast cancer, particularly ductal carcinoma in situ (DCIS). Some DCIS lesions rapidly progress to invasive carcinoma, whereas others remain indolent. Because we cannot yet predict which lesions will not progress, all DCIS is regarded as malignant, and many women are overtreated. Thus, there is a pressing need for a panel of molecular markers in addition to the current clinical and pathological factors to provide prognostic information. Genomic technologies such as microarrays have made major contributions to defining subtypes of breast cancer. Next-generation sequencing (NGS) modalities offer unprecedented depth of expression analysis through revealing transcriptional boundaries, mutations, rare transcripts and alternative splice variants. NGS approaches are just beginning to be applied to DCIS. Here, the authors review the applications and challenges of NGS in discovering novel potential therapeutic targets and candidate biomarkers in the premalignant progression of breast cancer.
Collapse
Affiliation(s)
- Hitchintan Kaur
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Engebraaten O, Vollan HKM, Børresen-Dale AL. Triple-negative breast cancer and the need for new therapeutic targets. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1064-1074. [PMID: 23920327 DOI: 10.1016/j.ajpath.2013.05.033] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 05/22/2013] [Accepted: 05/28/2013] [Indexed: 12/17/2022]
Abstract
Triple-negative breast cancers (TNBCs) are a diverse and heterogeneous group of tumors that by definition lack estrogen and progesterone receptors and amplification of the HER2 gene. The majority of the tumors classified as TNBCs are highly malignant, and only a subgroup responds to conventional chemotherapy with a favorable prognosis. Results from decades of research have identified important molecular characteristics that can subdivide this group of breast cancers further. High-throughput molecular analyses including sequencing, pathway analyses, and integrated analyses of alterations at the genomic and transcriptomic levels have improved our understanding of the molecular alterations involved in tumor development and progression. How this knowledge should be used for rational selection of therapy is a challenging task and the subject of numerous ongoing research programs. This review summarizes the current knowledge on the clinical characteristics and molecular alterations of TNBCs. Currently used conventional therapeutic strategies and targeted therapy studies are discussed, with references to recently published results on the molecular characterization of TNBCs.
Collapse
Affiliation(s)
- Olav Engebraaten
- Division of Cancer Medicine, Surgery and Transplantation, Department of Oncology, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.
| | - Hans Kristian Moen Vollan
- Division of Cancer Medicine, Surgery and Transplantation, Department of Oncology, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Anne-Lise Børresen-Dale
- K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| |
Collapse
|
35
|
Hua L, Zhou P, Li L, Liu H, Yang Z. Prioritizing breast cancer subtype related miRNAs using miRNA-mRNA dysregulated relationships extracted from their dual expression profiling. J Theor Biol 2013; 331:1-11. [PMID: 23619378 DOI: 10.1016/j.jtbi.2013.04.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 04/08/2013] [Accepted: 04/09/2013] [Indexed: 01/18/2023]
Abstract
Identification of miRNA expression-based breast cancer subtypes is considered a critical means of prognostication. So far, the studies on breast cancer subtypes have not been well characterized, and few studies have performed expression profiling of both miRNA and mRNA from the same breast cancer subtypes samples. In this study we analyzed dual expression profiling data of miRNA and mRNA derived from the expression profiling of 489 miRNAs in 41 luminal-A breast tumors samples and 15 basal-like samples. We defined a correlation coefficient ratio (CCR) and used it to examine the correlative dysregulated relationships between miRNAs and mRNAs. A miRNA-mRNA dysregulated network was arisen from 6222 dysregulated relationships, and from this network, miRNA-miRNA networks specialized for luminal-A and basal-like breast cancer subtypes were extracted according to the CCR values. By analyzing the networks, we found that luminal-A trend and basal-like trend miRNA-miRNA network displayed a change in hubs which connected the most miRNAs, and therefore become the potential breast cancer subtype related miRNAs. In addition, we also used other network analysis methods for miRNA expression profiling data, such as weighted correlation network analysis (WGCNA), Bayesian network analysis, and miRNA similarity (MISIM) analysis to validate the identified miRNAs or miRNA clusters. This study provides a new analyzing method to predict candidate miRNAs of breast cancer subtype from a system biology level and help understanding the relationship between miRNA and mRNA in primary breast cancer subtype.
Collapse
Affiliation(s)
- Lin Hua
- Biomedical Engineering Institute of Capital Medical University, Beijing 100069, China.
| | | | | | | | | |
Collapse
|
36
|
Siletz A, Schnabel M, Kniazeva E, Schumacher AJ, Shin S, Jeruss JS, Shea LD. Dynamic transcription factor networks in epithelial-mesenchymal transition in breast cancer models. PLoS One 2013; 8:e57180. [PMID: 23593114 PMCID: PMC3620167 DOI: 10.1371/journal.pone.0057180] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Accepted: 01/17/2013] [Indexed: 12/11/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a complex change in cell differentiation that allows breast carcinoma cells to acquire invasive properties. EMT involves a cascade of regulatory changes that destabilize the epithelial phenotype and allow mesenchymal features to manifest. As transcription factors (TFs) are upstream effectors of the genome-wide expression changes that result in phenotypic change, understanding the sequential changes in TF activity during EMT provides rich information on the mechanism of this process. Because molecular interactions will vary as cells progress from an epithelial to a mesenchymal differentiation program, dynamic networks are needed to capture the changing context of molecular processes. In this study we applied an emerging high-throughput, dynamic TF activity array to define TF activity network changes in three cell-based models of EMT in breast cancer based on HMLE Twist ER and MCF-7 mammary epithelial cells. The TF array distinguished conserved from model-specific TF activity changes in the three models. Time-dependent data was used to identify pairs of TF activities with significant positive or negative correlation, indicative of interdependent TF activity throughout the six-day study period. Dynamic TF activity patterns were clustered into groups of TFs that change along a time course of gene expression changes and acquisition of invasive capacity. Time-dependent TF activity data was combined with prior knowledge of TF interactions to construct dynamic models of TF activity networks as epithelial cells acquire invasive characteristics. These analyses show EMT from a unique and targetable vantage and may ultimately contribute to diagnosis and therapy.
Collapse
Affiliation(s)
- Anaar Siletz
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois, United States of America
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Michael Schnabel
- Physical Sciences – Oncology Center, Northwestern Institute on Complex Systems, Departments of Applied Mathematics and Physics, Northwestern University, Evanston, Illinois, United States of America
| | - Ekaterina Kniazeva
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois, United States of America
| | - Andrew J. Schumacher
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Seungjin Shin
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois, United States of America
| | - Jacqueline S. Jeruss
- Department of Surgery, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, United States of America
| | - Lonnie D. Shea
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois, United States of America
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, United States of America
- Institute for BioNanotechnology in Medicine (IBNAM), Northwestern University, Chicago, Illinois, United States of America
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, United States of America
- * E-mail:
| |
Collapse
|
37
|
Limame R, de Beeck KO, Van Laere S, Croes L, De Wilde A, Dirix L, Van Camp G, Peeters M, De Wever O, Lardon F, Pauwels P. Expression profiling of migrated and invaded breast cancer cells predicts early metastatic relapse and reveals Krüppel-like factor 9 as a potential suppressor of invasive growth in breast cancer. Oncoscience 2013; 1:69-81. [PMID: 25593984 PMCID: PMC4295756 DOI: 10.18632/oncoscience.10] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 01/16/2014] [Indexed: 12/17/2022] Open
Abstract
Cell motility and invasion initiate metastasis. However, only a subpopulation of cancer cells within a tumor will ultimately become invasive. Due to this stochastic and transient nature, in an experimental setting, migrating and invading cells need to be isolated from the general population in order to study the gene expression profiles linked to these processes. This report describes microarray analysis on RNA derived from migrated or invaded subpopulations of triple negative breast cancer cells in a Transwell set-up, at two different time points during motility and invasion, pre-determined as “early” and “late” in real-time kinetic assessments. Invasion- and migration-related gene expression signatures were generated through comparison with non-invasive cells, remaining at the upper side of the Transwell membranes. Late-phase signatures of both invasion and migration indicated poor prognosis in a series of breast cancer data sets. Furthermore, evaluation of the genes constituting the prognostic invasion-related gene signature revealed Krüppel-like factor 9 (KLF9) as a putative suppressor of invasive growth in breast cancer. Next to loss in invasive vs non-invasive cell lines, KLF9 also showed significantly lower expression levels in the “early” invasive cell population, in several public expression data sets and in clinical breast cancer samples when compared to normal tissue. Overexpression of EGFP-KLF9 fusion protein significantly altered morphology and blocked invasion and growth of MDA-MB-231 cells in vitro. In addition, KLF9 expression correlated inversely with mitotic activity in clinical samples, indicating anti-proliferative effects.
Collapse
Affiliation(s)
- Ridha Limame
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium ; These authors equally contributed to this work
| | - Ken Op de Beeck
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium ; Department of Medical Genetics, University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, B-2610 Antwerp, Belgium ; These authors equally contributed to this work
| | - Steven Van Laere
- Translational Cancer Research Unit (TCRU), GZA Hospitals Sint-Augustinus, Oosterveldlaan 24, B-2610 Wilrijk (Antwerp), Belgium ; Department of Oncology, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Lieselot Croes
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium ; Department of Medical Genetics, University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, B-2610 Antwerp, Belgium ; Laboratory of Pathology, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem (Antwerp), Belgium
| | - Annemieke De Wilde
- Laboratory of Pathology, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem (Antwerp), Belgium
| | - Luc Dirix
- Translational Cancer Research Unit (TCRU), GZA Hospitals Sint-Augustinus, Oosterveldlaan 24, B-2610 Wilrijk (Antwerp), Belgium
| | - Guy Van Camp
- Department of Medical Genetics, University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Marc Peeters
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium ; Department of Oncology, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem (Antwerp), Belgium
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Radiotherapy and Experimental Cancer Research, Ghent University Hospital, De Pintelaan 185, B-9000 Ghent, Belgium
| | - Filip Lardon
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Patrick Pauwels
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium ; Laboratory of Pathology, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem (Antwerp), Belgium
| |
Collapse
|
38
|
Akan P, Alexeyenko A, Costea PI, Hedberg L, Solnestam BW, Lundin S, Hällman J, Lundberg E, Uhlén M, Lundeberg J. Comprehensive analysis of the genome transcriptome and proteome landscapes of three tumor cell lines. Genome Med 2012; 4:86. [PMID: 23158748 PMCID: PMC3580420 DOI: 10.1186/gm387] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 10/25/2012] [Accepted: 11/18/2012] [Indexed: 01/16/2023] Open
Abstract
We here present a comparative genome, transcriptome and functional network analysis of three human cancer cell lines (A431, U251MG and U2OS), and investigate their relation to protein expression. Gene copy numbers significantly influenced corresponding transcript levels; their effect on protein levels was less pronounced. We focused on genes with altered mRNA and/or protein levels to identify those active in tumor maintenance. We provide comprehensive information for the three genomes and demonstrate the advantage of integrative analysis for identifying tumor-related genes amidst numerous background mutations by relating genomic variation to expression/protein abundance data and use gene networks to reveal implicated pathways.
Collapse
Affiliation(s)
- Pelin Akan
- KTH - Royal Institute of Technology, Science for Life Laboratory, School of Biotechnology, SE-171 65 Solna, Sweden
| | - Andrey Alexeyenko
- KTH - Royal Institute of Technology, Science for Life Laboratory, School of Biotechnology, SE-171 65 Solna, Sweden
| | - Paul Igor Costea
- KTH - Royal Institute of Technology, Science for Life Laboratory, School of Biotechnology, SE-171 65 Solna, Sweden
| | - Lilia Hedberg
- KTH - Royal Institute of Technology, Science for Life Laboratory, School of Biotechnology, SE-171 65 Solna, Sweden
| | - Beata Werne Solnestam
- KTH - Royal Institute of Technology, Science for Life Laboratory, School of Biotechnology, SE-171 65 Solna, Sweden
| | - Sverker Lundin
- KTH - Royal Institute of Technology, Science for Life Laboratory, School of Biotechnology, SE-171 65 Solna, Sweden
| | - Jimmie Hällman
- KTH - Royal Institute of Technology, Science for Life Laboratory, School of Biotechnology, SE-171 65 Solna, Sweden
| | - Emma Lundberg
- KTH - Royal Institute of Technology, Science for Life Laboratory, School of Biotechnology, SE-171 65 Solna, Sweden
| | - Mathias Uhlén
- KTH - Royal Institute of Technology, Science for Life Laboratory, School of Biotechnology, SE-171 65 Solna, Sweden
| | - Joakim Lundeberg
- KTH - Royal Institute of Technology, Science for Life Laboratory, School of Biotechnology, SE-171 65 Solna, Sweden
| |
Collapse
|
39
|
Charoentong P, Angelova M, Efremova M, Gallasch R, Hackl H, Galon J, Trajanoski Z. Bioinformatics for cancer immunology and immunotherapy. Cancer Immunol Immunother 2012; 61:1885-903. [PMID: 22986455 PMCID: PMC3493665 DOI: 10.1007/s00262-012-1354-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 09/04/2012] [Indexed: 01/24/2023]
Abstract
Recent mechanistic insights obtained from preclinical studies and the approval of the first immunotherapies has motivated increasing number of academic investigators and pharmaceutical/biotech companies to further elucidate the role of immunity in tumor pathogenesis and to reconsider the role of immunotherapy. Additionally, technological advances (e.g., next-generation sequencing) are providing unprecedented opportunities to draw a comprehensive picture of the tumor genomics landscape and ultimately enable individualized treatment. However, the increasing complexity of the generated data and the plethora of bioinformatics methods and tools pose considerable challenges to both tumor immunologists and clinical oncologists. In this review, we describe current concepts and future challenges for the management and analysis of data for cancer immunology and immunotherapy. We first highlight publicly available databases with specific focus on cancer immunology including databases for somatic mutations and epitope databases. We then give an overview of the bioinformatics methods for the analysis of next-generation sequencing data (whole-genome and exome sequencing), epitope prediction tools as well as methods for integrative data analysis and network modeling. Mathematical models are powerful tools that can predict and explain important patterns in the genetic and clinical progression of cancer. Therefore, a survey of mathematical models for tumor evolution and tumor-immune cell interaction is included. Finally, we discuss future challenges for individualized immunotherapy and suggest how a combined computational/experimental approaches can lead to new insights into the molecular mechanisms of cancer, improved diagnosis, and prognosis of the disease and pinpoint novel therapeutic targets.
Collapse
Affiliation(s)
- Pornpimol Charoentong
- Biocenter, Division of Bioinformatics, Innsbruck Medical University, Innrain 80, 6020 Innsbruck, Austria
| | - Mihaela Angelova
- Biocenter, Division of Bioinformatics, Innsbruck Medical University, Innrain 80, 6020 Innsbruck, Austria
| | - Mirjana Efremova
- Biocenter, Division of Bioinformatics, Innsbruck Medical University, Innrain 80, 6020 Innsbruck, Austria
| | - Ralf Gallasch
- Biocenter, Division of Bioinformatics, Innsbruck Medical University, Innrain 80, 6020 Innsbruck, Austria
| | - Hubert Hackl
- Biocenter, Division of Bioinformatics, Innsbruck Medical University, Innrain 80, 6020 Innsbruck, Austria
| | - Jerome Galon
- INSERM U872, Integrative Cancer Immunology Laboratory, Paris, France
| | - Zlatko Trajanoski
- Biocenter, Division of Bioinformatics, Innsbruck Medical University, Innrain 80, 6020 Innsbruck, Austria
| |
Collapse
|
40
|
Kubrycht J, Sigler K, Souček P. Virtual interactomics of proteins from biochemical standpoint. Mol Biol Int 2012; 2012:976385. [PMID: 22928109 PMCID: PMC3423939 DOI: 10.1155/2012/976385] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 05/18/2012] [Accepted: 05/18/2012] [Indexed: 12/24/2022] Open
Abstract
Virtual interactomics represents a rapidly developing scientific area on the boundary line of bioinformatics and interactomics. Protein-related virtual interactomics then comprises instrumental tools for prediction, simulation, and networking of the majority of interactions important for structural and individual reproduction, differentiation, recognition, signaling, regulation, and metabolic pathways of cells and organisms. Here, we describe the main areas of virtual protein interactomics, that is, structurally based comparative analysis and prediction of functionally important interacting sites, mimotope-assisted and combined epitope prediction, molecular (protein) docking studies, and investigation of protein interaction networks. Detailed information about some interesting methodological approaches and online accessible programs or databases is displayed in our tables. Considerable part of the text deals with the searches for common conserved or functionally convergent protein regions and subgraphs of conserved interaction networks, new outstanding trends and clinically interesting results. In agreement with the presented data and relationships, virtual interactomic tools improve our scientific knowledge, help us to formulate working hypotheses, and they frequently also mediate variously important in silico simulations.
Collapse
Affiliation(s)
- Jaroslav Kubrycht
- Department of Physiology, Second Medical School, Charles University, 150 00 Prague, Czech Republic
| | - Karel Sigler
- Laboratory of Cell Biology, Institute of Microbiology, Academy of Sciences of the Czech Republic, 142 20 Prague, Czech Republic
| | - Pavel Souček
- Toxicogenomics Unit, National Institute of Public Health, 100 42 Prague, Czech Republic
| |
Collapse
|