1
|
Sunildutt N, Ahmed F, Salih ARC, Kim HC, Choi KH. Unraveling new avenues in pancreatic cancer treatment: A comprehensive exploration of drug repurposing using transcriptomic data. Comput Biol Med 2025; 185:109481. [PMID: 39644581 DOI: 10.1016/j.compbiomed.2024.109481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/28/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
Pancreatic cancer, a malignancy notorious for its late-stage diagnosis and low patient survival rates, remains a formidable global health challenge. The currently available FDA-approved treatments for pancreatic cancer, notably chemotherapeutic agents, exhibit suboptimal efficacy, often accompanied by concerns regarding toxicity. Given the intricate nature of pancreatic cancer pathogenesis and the time-intensive nature of in silico drug discovery approaches, drug repurposing emerges as a compelling strategy to expedite the development of novel therapeutic interventions. In our study, we harnessed transcriptomic data from an exhaustive exploration of four diverse databases, ensuring a rigorous and unbiased analysis of differentially expressed genes, with a particular focus on upregulated genes associated with pancreatic cancer. Leveraging these pancreatic cancer-associated host protein targets, we employed a battery of cutting-edge bioinformatics tools, including Cytoscape STRING, GeneMANIA, Connectivity Map, and NetworkAnalyst, to identify potential small molecule drug candidates and elucidate their interactions. Subsequently, we conducted meticulous docking and redocking simulations for the selected drug-protein target pairs. This rigorous computational approach culminated in the identification of two promising broad-spectrum drug candidates against four pivotal host genes implicated in pancreatic cancer. Our findings strongly advocate for further investigation and preclinical validation of these candidates. Specifically, we propose prioritizing Dasatinib for evaluation against MMP3, MMP9, and EGFR due to their remarkable binding affinities, as well as Pioglitazone against MMP3, MMP2 and MMP9. These discoveries hold great promise in advancing the therapeutic landscape for pancreatic cancer, offering new avenues for improving patient outcomes.
Collapse
Affiliation(s)
- Naina Sunildutt
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea
| | - Faheem Ahmed
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea
| | - Abdul Rahim Chethikkattuveli Salih
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea; Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, US; BioSpero, Inc, Jeju, Republic of Korea
| | - Hyung Chul Kim
- Department of Future Science and Technology Business, Korea University, Seoul, 02841, Republic of Korea.
| | - Kyung Hyun Choi
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea.
| |
Collapse
|
2
|
Orfanoudaki G, Psatha K, Aivaliotis M. Insight into Mantle Cell Lymphoma Pathobiology, Diagnosis, and Treatment Using Network-Based and Drug-Repurposing Approaches. Int J Mol Sci 2024; 25:7298. [PMID: 39000404 PMCID: PMC11242097 DOI: 10.3390/ijms25137298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Mantle cell lymphoma (MCL) is a rare, incurable, and aggressive B-cell non-Hodgkin lymphoma (NHL). Early MCL diagnosis and treatment is critical and puzzling due to inter/intra-tumoral heterogeneity and limited understanding of the underlying molecular mechanisms. We developed and applied a multifaceted analysis of selected publicly available transcriptomic data of well-defined MCL stages, integrating network-based methods for pathway enrichment analysis, co-expression module alignment, drug repurposing, and prediction of effective drug combinations. We demonstrate the "butterfly effect" emerging from a small set of initially differentially expressed genes, rapidly expanding into numerous deregulated cellular processes, signaling pathways, and core machineries as MCL becomes aggressive. We explore pathogenicity-related signaling circuits by detecting common co-expression modules in MCL stages, pointing out, among others, the role of VEGFA and SPARC proteins in MCL progression and recommend further study of precise drug combinations. Our findings highlight the benefit that can be leveraged by such an approach for better understanding pathobiology and identifying high-priority novel diagnostic and prognostic biomarkers, drug targets, and efficacious combination therapies against MCL that should be further validated for their clinical impact.
Collapse
Affiliation(s)
- Georgia Orfanoudaki
- Functional Proteomics and Systems Biology (FunPATh), Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center, GR-54124 Thessaloniki, Greece
- Institute of Molecular Biology and Biotechnology Foundation for Research and Technology-Hellas, GR-70013 Heraklion, Greece
| | - Konstantina Psatha
- Functional Proteomics and Systems Biology (FunPATh), Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center, GR-54124 Thessaloniki, Greece
- Institute of Molecular Biology and Biotechnology Foundation for Research and Technology-Hellas, GR-70013 Heraklion, Greece
- Laboratory of Medical Biology-Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Michalis Aivaliotis
- Functional Proteomics and Systems Biology (FunPATh), Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center, GR-54124 Thessaloniki, Greece
- Institute of Molecular Biology and Biotechnology Foundation for Research and Technology-Hellas, GR-70013 Heraklion, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
- Laboratory of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| |
Collapse
|
3
|
Sakai N, Kamimura K, Terai S. Repurposable Drugs for Immunotherapy and Strategies to Find Candidate Drugs. Pharmaceutics 2023; 15:2190. [PMID: 37765160 PMCID: PMC10536625 DOI: 10.3390/pharmaceutics15092190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Conventional drug discovery involves significant steps, time, and expenses; therefore, novel methods for drug discovery remain unmet, particularly for patients with intractable diseases. For this purpose, the drug repurposing method has been recently used to search for new therapeutic agents. Repurposed drugs are mostly previously approved drugs, which were carefully tested for their efficacy for other diseases and had their safety for the human body confirmed following careful pre-clinical trials, clinical trials, and post-marketing surveillance. Therefore, using these approved drugs for other diseases that cannot be treated using conventional therapeutic methods could save time and economic costs for testing their clinical applicability. In this review, we have summarized the methods for identifying repurposable drugs focusing on immunotherapy.
Collapse
Affiliation(s)
- Norihiro Sakai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Aasahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan; (N.S.); (S.T.)
| | - Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Aasahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan; (N.S.); (S.T.)
- Department of General Medicine, Niigata University School of Medicine, 1-757, Aasahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Aasahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan; (N.S.); (S.T.)
| |
Collapse
|
4
|
Firoozbakht F, Rezaeian I, Rueda L, Ngom A. Computationally repurposing drugs for breast cancer subtypes using a network-based approach. BMC Bioinformatics 2022; 23:143. [PMID: 35443626 PMCID: PMC9020161 DOI: 10.1186/s12859-022-04662-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 03/30/2022] [Indexed: 11/22/2022] Open
Abstract
‘De novo’ drug discovery is costly, slow, and with high risk. Repurposing known drugs for treatment of other diseases offers a fast, low-cost/risk and highly-efficient method toward development of efficacious treatments. The emergence of large-scale heterogeneous biomolecular networks, molecular, chemical and bioactivity data, and genomic and phenotypic data of pharmacological compounds is enabling the development of new area of drug repurposing called ‘in silico’ drug repurposing, i.e., computational drug repurposing (CDR). The aim of CDR is to discover new indications for an existing drug (drug-centric) or to identify effective drugs for a disease (disease-centric). Both drug-centric and disease-centric approaches have the common challenge of either assessing the similarity or connections between drugs and diseases. However, traditional CDR is fraught with many challenges due to the underlying complex pharmacology and biology of diseases, genes, and drugs, as well as the complexity of their associations. As such, capturing highly non-linear associations among drugs, genes, diseases by most existing CDR methods has been challenging. We propose a network-based integration approach that can best capture knowledge (and complex relationships) contained within and between drugs, genes and disease data. A network-based machine learning approach is applied thereafter by using the extracted knowledge and relationships in order to identify single and pair of approved or experimental drugs with potential therapeutic effects on different breast cancer subtypes. Indeed, further clinical analysis is needed to confirm the therapeutic effects of identified drugs on each breast cancer subtype.
Collapse
Affiliation(s)
- Forough Firoozbakht
- School of Computer Science, University of Windsor, 401 Sunset Ave., Windsor, ON, Canada
| | - Iman Rezaeian
- School of Computer Science, University of Windsor, 401 Sunset Ave., Windsor, ON, Canada.,Rocket Innovation Studio, 156 Chatham St W, Windsor, ON, Canada
| | - Luis Rueda
- School of Computer Science, University of Windsor, 401 Sunset Ave., Windsor, ON, Canada.
| | - Alioune Ngom
- School of Computer Science, University of Windsor, 401 Sunset Ave., Windsor, ON, Canada
| |
Collapse
|
5
|
Pham TH, Qiu Y, Liu J, Zimmer S, O’Neill E, Xie L, Zhang P. Chemical-induced gene expression ranking and its application to pancreatic cancer drug repurposing. PATTERNS 2022; 3:100441. [PMID: 35465231 PMCID: PMC9023899 DOI: 10.1016/j.patter.2022.100441] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/13/2021] [Accepted: 01/12/2022] [Indexed: 12/18/2022]
Abstract
Chemical-induced gene expression profiles provide critical information of chemicals in a biological system, thus offering new opportunities for drug discovery. Despite their success, large-scale analysis leveraging gene expressions is limited by time and cost. Although several methods for predicting gene expressions were proposed, they only focused on imputation and classification settings, which have limited applications to real-world scenarios of drug discovery. Therefore, a chemical-induced gene expression ranking (CIGER) framework is proposed to target a more realistic but more challenging setting in which overall rankings in gene expression profiles induced by de novo chemicals are predicted. The experimental results show that CIGER significantly outperforms existing methods in both ranking and classification metrics. Furthermore, a drug screening pipeline based on CIGER is proposed to identify potential treatments of drug-resistant pancreatic cancer. Our predictions have been validated by experiments, thereby showing the effectiveness of CIGER for phenotypic compound screening of precision medicine. A new deep-learning method (CIGER) for chemical-induced gene expression ranking CIGER can predict gene expression for de novo chemicals from chemical structures We discovered drugs for the treatment of drug-resistant pancreatic cancer
In recent years, a phenotype-based drug discovery approach using chemical-induced gene expressions has shown to be effective in drug discovery and precision medicine. However, it is not feasible to experimentally determine chemical-induced gene expressions for all available chemicals of interest, thereby hindering the application of gene expression-based compound screening on a large scale. Thus, it is crucial to design a computational approach that can generate gene expression information for any chemicals. We proposed a new, deep-learning framework named chemical-induced gene expression ranking (CIGER) to predict a landmark gene expression profile (i.e., gene ranking) induced by de novo chemicals based on their chemical structures. Leveraging CIGER, we predicted and experimentally validated that several existing drugs can increase the therapeutic response on drug-resistant pancreatic cancer. Our results demonstrated the effectiveness of CIGER for precision drug discovery in practice.
Collapse
Affiliation(s)
- Thai-Hoang Pham
- Department of Computer Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Yue Qiu
- Ph.D. Program in Biology, The Graduate Center, The City University of New York, New York, NY 10016, USA
| | - Jiahui Liu
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Eric O’Neill
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
- EpiCombi.AI Therapeutics, Oxford OX7 3SB, UK
| | - Lei Xie
- Ph.D. Program in Biology, The Graduate Center, The City University of New York, New York, NY 10016, USA
- Department of Computer Science, Hunter College, The City University of New York, New York, NY 10065, USA
- Ph.D. Program in Computer Science and Biochemistry, The Graduate Center, The City University of New York, New York, NY 10016, USA
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Ping Zhang
- Department of Computer Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
- Translational Data Analytics Institute, The Ohio State University, Columbus, OH 43210, USA
- Corresponding author
| |
Collapse
|
6
|
Al-Taie Z, Hannink M, Mitchem J, Papageorgiou C, Shyu CR. Drug Repositioning and Subgroup Discovery for Precision Medicine Implementation in Triple Negative Breast Cancer. Cancers (Basel) 2021; 13:6278. [PMID: 34944904 PMCID: PMC8699385 DOI: 10.3390/cancers13246278] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 12/29/2022] Open
Abstract
Breast cancer (BC) is the leading cause of death among female patients with cancer. Patients with triple-negative breast cancer (TNBC) have the lowest survival rate. TNBC has substantial heterogeneity within the BC population. This study utilized our novel patient stratification and drug repositioning method to find subgroups of BC patients that share common genetic profiles and that may respond similarly to the recommended drugs. After further examination of the discovered patient subgroups, we identified five homogeneous druggable TNBC subgroups. A drug repositioning algorithm was then applied to find the drugs with a high potential for each subgroup. Most of the top drugs for these subgroups were chemotherapy used for various types of cancer, including BC. After analyzing the biological mechanisms targeted by these drugs, ferroptosis was the common cell death mechanism induced by the top drugs in the subgroups with neoplasm subdivision and race as clinical variables. In contrast, the antioxidative effect on cancer cells was the common targeted mechanism in the subgroup of patients with an age less than 50. Literature reviews were used to validate our findings, which could provide invaluable insights to streamline the drug repositioning process and could be further studied in a wet lab setting and in clinical trials.
Collapse
Affiliation(s)
- Zainab Al-Taie
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA; (Z.A.-T.); (J.M.)
- Department of Computer Science, College of Science for Women, University of Baghdad, Baghdad 10070, Iraq
| | - Mark Hannink
- Department of Biochemistry, University of Missouri, Columbia, Missouri, MO 65211, USA;
- Department of Animal Sciences, Bond Life Sciences Center, University of Missouri, 1201 Rollins Street, Columbia, MO 65211, USA
| | - Jonathan Mitchem
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA; (Z.A.-T.); (J.M.)
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65212, USA
- Department of Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Christos Papageorgiou
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Chi-Ren Shyu
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA; (Z.A.-T.); (J.M.)
- Electrical Engineering and Computer Science Department, University of Missouri, Columbia, MO 65211, USA
- Department of Medicine, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
7
|
Wu S, Chen D, Snyder MP. Network biology bridges the gaps between quantitative genetics and multi-omics to map complex diseases. Curr Opin Chem Biol 2021; 66:102101. [PMID: 34861483 DOI: 10.1016/j.cbpa.2021.102101] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/17/2021] [Accepted: 10/27/2021] [Indexed: 12/27/2022]
Abstract
With advances in high-throughput sequencing technologies, quantitative genetics approaches have provided insights into genetic basis of many complex diseases. Emerging in-depth multi-omics profiling technologies have created exciting opportunities for systematically investigating intricate interaction networks with different layers of biological molecules underlying disease etiology. Herein, we summarized two main categories of biological networks: evidence-based and statistically inferred. These different types of molecular networks complement each other at both bulk and single-cell levels. We also review three main strategies to incorporate quantitative genetics results with multi-omics data by network analysis: (a) network propagation, (b) functional module-based methods, (c) comparative/dynamic networks. These strategies not only aid in elucidating molecular mechanisms of complex diseases but can guide the search for therapeutic targets.
Collapse
Affiliation(s)
- Si Wu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Dijun Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
8
|
Khanjani F, Jafari L, Azadiyan S, Roozbehi S, Moradian C, Zahiri J, Hasannia S, Sajedi RH. Drug repositioning based on gene expression data for human HER2-positive breast cancer. Arch Biochem Biophys 2021; 712:109043. [PMID: 34597657 DOI: 10.1016/j.abb.2021.109043] [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: 05/26/2021] [Revised: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 10/20/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2)-positive breast cancer represents approximately 15-30% of all invasive breast cancers. Despite the recent advances in therapeutic practices of HER2 subtype, drug resistance and tumor recurrence still have remained as major problems. Drug discovery is a long and difficult process, so the aim of this study is to find potential new application for existing therapeutic agents. Gene expression data for breast invasive carcinoma were retrieved from The Cancer Genome Atlas (TCGA) database. The normal and tumor samples were analyzed using Linear Models for Microarray Data (LIMMA) R package in order to find the differentially expressed genes (DEGs). These genes were used as entry for the library of integrated network-based cellular signatures (LINCS) L1000CDS2 software and suggested 24 repurposed drugs. According to the obtained results, some of these drugs including vorinostat, mocetinostat, alvocidib, CGP-60474, BMS-387032, AT-7519, and curcumin have significant functional similarity and structural correlation with FDA-approved breast cancer drugs. Based on the drug-target network, which consisted of the repurposed drugs and their target genes, the aforementioned drugs had the highest degrees. Moreover, the experimental approach verified curcumin as an effective therapeutic agent for HER2 positive breast cancer. Hence, our work suggested that some repurposed drugs based on gene expression data can be noticed as potential drugs for the treatment of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Farkhondeh Khanjani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Leila Jafari
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences, Zanjan, Iran
| | - Somayeh Azadiyan
- Bioinformatics and Computational Omics Lab (BioCOOL), Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sahar Roozbehi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Cobra Moradian
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Javad Zahiri
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
| | - Sadegh Hasannia
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza H Sajedi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
9
|
Human microRNA similarity in breast cancer. Biosci Rep 2021; 41:229885. [PMID: 34612484 PMCID: PMC8529337 DOI: 10.1042/bsr20211123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/28/2021] [Accepted: 10/04/2021] [Indexed: 11/25/2022] Open
Abstract
MicroRNAs (miRNAs) play important roles in a variety of human diseases, including breast cancer. A number of miRNAs are up- and down-regulated in breast cancer. However, little is known about miRNA similarity and similarity network in breast cancer. Here, a collection of 272 breast cancer-associated miRNA precursors (pre-miRNAs) were utilized to calculate similarities of sequences, target genes, pathways and functions and construct a combined similarity network. Well-characterized miRNAs and their similarity network were highlighted. Interestingly, miRNA sequence-dependent similarity networks were not identified in spite of sequence–target gene association. Similarity networks with minimum and maximum number of miRNAs originate from pathway and mature sequence, respectively. The breast cancer-associated miRNAs were divided into seven functional classes (classes I–VII) followed by disease enrichment analysis and novel miRNA-based disease similarities were found. The finding would provide insight into miRNA similarity, similarity network and disease heterogeneity in breast cancer.
Collapse
|
10
|
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
|
11
|
Apoptotic effects of valproic acid on miR-34a, miR-520h and HDAC1 gene in breast cancer. Life Sci 2021; 269:119027. [PMID: 33453248 DOI: 10.1016/j.lfs.2021.119027] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 12/17/2022]
Abstract
Identifying miRNAs involved in cancer and devising strategies to control their expression is a new therapeutic approach. Valproic acid (VPA) has attracted a lot of interest in cancer research. We evaluated the impact of VPA on the expression of miR-34a, miR-520h, and their target gene histone deacetylase 1 (HDAC1), as well as their relationship with apoptosis in breast cancer. First, through bioinformatics analyses, the possible target genes of miR-34a and miR-520h and their roles in apoptosis regulation were investigated. Then, miR-34a, miR-520h, and HDAC1 gene expression in tissues of breast cancer patients were determined using the qRT-PCR method. The anticancer impact of VPA on apoptosis and the expression levels of miR-34a, miR-520h, and HDAC1 gene were measured in MCF-7 and MDA-MB-231 cell lines. The bioinformatics analyses indicated that miR-34a and miR-520h might make a unique contribution in regulating the apoptosis pathway. The relative expression of miR-34a and miR-520h significantly decreased in cancer tissues, while the relative expression of HDAC1 increased. In the in vitro study, VPA led to apoptosis induction and increased lipid peroxidation products in breast cancer cells. Moreover, VPA increased the expression of miR- 34a and miR-520h and decreased HDAC1 expression in MCF-7 cells. In MDA-MB-231 cells, VPA decreased the expression of these miRNAs and increased the expression of HDAC1. It can be concluded that miR-34a and miR-520h are implicated in the apoptosis pathways, and thus, VPA can recruit as a possible option in breast cancer research due to its interference with epigenetic processes.
Collapse
|
12
|
Blatti C, Emad A, Berry MJ, Gatzke L, Epstein M, Lanier D, Rizal P, Ge J, Liao X, Sobh O, Lambert M, Post CS, Xiao J, Groves P, Epstein AT, Chen X, Srinivasan S, Lehnert E, Kalari KR, Wang L, Weinshilboum RM, Song JS, Jongeneel CV, Han J, Ravaioli U, Sobh N, Bushell CB, Sinha S. Knowledge-guided analysis of "omics" data using the KnowEnG cloud platform. PLoS Biol 2020; 18:e3000583. [PMID: 31971940 PMCID: PMC6977717 DOI: 10.1371/journal.pbio.3000583] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 12/19/2019] [Indexed: 12/19/2022] Open
Abstract
We present Knowledge Engine for Genomics (KnowEnG), a free-to-use computational system for analysis of genomics data sets, designed to accelerate biomedical discovery. It includes tools for popular bioinformatics tasks such as gene prioritization, sample clustering, gene set analysis, and expression signature analysis. The system specializes in "knowledge-guided" data mining and machine learning algorithms, in which user-provided data are analyzed in light of prior information about genes, aggregated from numerous knowledge bases and encoded in a massive "Knowledge Network." KnowEnG adheres to "FAIR" principles (findable, accessible, interoperable, and reuseable): its tools are easily portable to diverse computing environments, run on the cloud for scalable and cost-effective execution, and are interoperable with other computing platforms. The analysis tools are made available through multiple access modes, including a web portal with specialized visualization modules. We demonstrate the KnowEnG system's potential value in democratization of advanced tools for the modern genomics era through several case studies that use its tools to recreate and expand upon the published analysis of cancer data sets.
Collapse
Affiliation(s)
- Charles Blatti
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Amin Emad
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Electrical and Computer Engineering, McGill University, Montreal, Canada
| | - Matthew J. Berry
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Lisa Gatzke
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Milt Epstein
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Daniel Lanier
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Pramod Rizal
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Jing Ge
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Xiaoxia Liao
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Omar Sobh
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Mike Lambert
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Corey S. Post
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Jinfeng Xiao
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Peter Groves
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Aidan T. Epstein
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Xi Chen
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Subhashini Srinivasan
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Erik Lehnert
- Seven Bridges Genomics, Charlestown, Massachusetts, United States of America
| | - Krishna R. Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Richard M. Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jun S. Song
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - C. Victor Jongeneel
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Jiawei Han
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Umberto Ravaioli
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Nahil Sobh
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Colleen B. Bushell
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Saurabh Sinha
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
13
|
Lin CY, Ruan P, Li R, Yang JM, See S, Song J, Akutsu T. Deep learning with evolutionary and genomic profiles for identifying cancer subtypes. J Bioinform Comput Biol 2019; 17:1940005. [DOI: 10.1142/s0219720019400055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer subtype identification is an unmet need in precision diagnosis. Recently, evolutionary conservation has been indicated to contain informative signatures for functional significance in cancers. However, the importance of evolutionary conservation in distinguishing cancer subtypes remains largely unclear. Here, we identified the evolutionarily conserved genes (i.e. core genes) and observed that they are primarily involved in cellular pathways relevant to cell growth and metabolisms. By using these core genes, we developed two novel strategies, namely a feature-based strategy (FES) and an image-based strategy (IMS) by integrating their evolutionary and genomic profiles with the deep learning algorithm. In comparison with the FES using the random set and the strategy using the PAM50 classifier, the core gene set-based FES achieved a higher accuracy for identifying breast cancer subtypes. The IMS and FES using the core gene set yielded better performances than the other strategies, in terms of classifying both breast cancer subtypes and multiple cancer types. Moreover, the IMS is reproducible even using different gene expression data (i.e. RNA-seq and microarray). Comprehensive analysis of eight cancer types demonstrates that our evolutionary conservation-based models represent a valid and helpful approach for identifying cancer subtypes and the core gene set offers distinguishable clues of cancer subtypes.
Collapse
Affiliation(s)
- Chun-Yu Lin
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji, Kyoto 6110011, Japan
| | - Peiying Ruan
- NVIDIA AI Technology Center, NVIDIA Corporation Japan, Tokyo 1070052, Japan
| | - Ruiming Li
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji, Kyoto 6110011, Japan
| | - Jinn-Moon Yang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Simon See
- NVIDIA AI Technology Center, NVIDIA Corporation Singapore, Singapore 138522, Singapore
| | - Jiangning Song
- Monash Biomedicine Discovery Institute and Department of Biochemistry & Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Tatsuya Akutsu
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji, Kyoto 6110011, Japan
| |
Collapse
|
14
|
Oulas A, Minadakis G, Zachariou M, Sokratous K, Bourdakou MM, Spyrou GM. Systems Bioinformatics: increasing precision of computational diagnostics and therapeutics through network-based approaches. Brief Bioinform 2019; 20:806-824. [PMID: 29186305 PMCID: PMC6585387 DOI: 10.1093/bib/bbx151] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/17/2017] [Indexed: 02/01/2023] Open
Abstract
Systems Bioinformatics is a relatively new approach, which lies in the intersection of systems biology and classical bioinformatics. It focuses on integrating information across different levels using a bottom-up approach as in systems biology with a data-driven top-down approach as in bioinformatics. The advent of omics technologies has provided the stepping-stone for the emergence of Systems Bioinformatics. These technologies provide a spectrum of information ranging from genomics, transcriptomics and proteomics to epigenomics, pharmacogenomics, metagenomics and metabolomics. Systems Bioinformatics is the framework in which systems approaches are applied to such data, setting the level of resolution as well as the boundary of the system of interest and studying the emerging properties of the system as a whole rather than the sum of the properties derived from the system's individual components. A key approach in Systems Bioinformatics is the construction of multiple networks representing each level of the omics spectrum and their integration in a layered network that exchanges information within and between layers. Here, we provide evidence on how Systems Bioinformatics enhances computational therapeutics and diagnostics, hence paving the way to precision medicine. The aim of this review is to familiarize the reader with the emerging field of Systems Bioinformatics and to provide a comprehensive overview of its current state-of-the-art methods and technologies. Moreover, we provide examples of success stories and case studies that utilize such methods and tools to significantly advance research in the fields of systems biology and systems medicine.
Collapse
Affiliation(s)
- Anastasis Oulas
- Bioinformatics European Research Area Chair, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - George Minadakis
- Bioinformatics European Research Area Chair, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Margarita Zachariou
- Bioinformatics European Research Area Chair, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Kleitos Sokratous
- Bioinformatics European Research Area Chair, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Marilena M Bourdakou
- Bioinformatics European Research Area Chair, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - George M Spyrou
- Bioinformatics European Research Area Chair, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
15
|
Larmuseau M, Verbeke LPC, Marchal K. Associating expression and genomic data using co-occurrence measures. Biol Direct 2019; 14:10. [PMID: 31072345 PMCID: PMC6507230 DOI: 10.1186/s13062-019-0240-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 04/10/2019] [Indexed: 12/11/2022] Open
Abstract
Abstract Recent technological evolutions have led to an exponential increase in data in all the omics fields. It is expected that integration of these different data sources, will drastically enhance our knowledge of the biological mechanisms behind genomic diseases such as cancer. However, the integration of different omics data still remains a challenge. In this work we propose an intuitive workflow for the integrative analysis of expression, mutation and copy number data taken from the METABRIC study on breast cancer. First, we present evidence that the expression profile of many important breast cancer genes consists of two modes or ‘regimes’, which contain important clinical information. Then, we show how the co-occurrence of these expression regimes can be used as an association measure between genes and validate our findings on the TCGA-BRCA study. Finally, we demonstrate how these co-occurrence measures can also be applied to link expression regimes to genomic aberrations, providing a more complete, integrative view on breast cancer. As a case study, an integrative analysis of the identified MLPH-FOXA1 association is performed, illustrating that the obtained expression associations are intimately linked to the underlying genomic changes. Reviewers This article was reviewed by Dirk Walther, Francisco Garcia and Isabel Nepomuceno. Electronic supplementary material The online version of this article (10.1186/s13062-019-0240-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maarten Larmuseau
- Department of Information Technology, Ghent University - Imec, Technologiepark-Zwijnaarde 126, 9052, Ghent, Belgium
| | - Lieven P C Verbeke
- Department of Plant Biotechnology and Bioinformatics, Ghent University - Imec, Technologiepark-Zwijnaarde 126, 9052, Ghent, Belgium
| | - Kathleen Marchal
- Department of Plant Biotechnology and Bioinformatics, Ghent University - Imec, Technologiepark-Zwijnaarde 126, 9052, Ghent, Belgium.
| |
Collapse
|
16
|
Li C, Liu L, Dinu V. Pathways of topological rank analysis (PoTRA): a novel method to detect pathways involved in hepatocellular carcinoma. PeerJ 2018; 6:e4571. [PMID: 29666752 PMCID: PMC5896492 DOI: 10.7717/peerj.4571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/14/2018] [Indexed: 01/01/2023] Open
Abstract
Complex diseases such as cancer are usually the result of a combination of environmental factors and one or several biological pathways consisting of sets of genes. Each biological pathway exerts its function by delivering signaling through the gene network. Theoretically, a pathway is supposed to have a robust topological structure under normal physiological conditions. However, the pathway's topological structure could be altered under some pathological condition. It is well known that a normal biological network includes a small number of well-connected hub nodes and a large number of nodes that are non-hubs. In addition, it is reported that the loss of connectivity is a common topological trait of cancer networks, which is an assumption of our method. Hence, from normal to cancer, the process of the network losing connectivity might be the process of disrupting the structure of the network, namely, the number of hub genes might be altered in cancer compared to that in normal or the distribution of topological ranks of genes might be altered. Based on this, we propose a new PageRank-based method called Pathways of Topological Rank Analysis (PoTRA) to detect pathways involved in cancer. We use PageRank to measure the relative topological ranks of genes in each biological pathway, then select hub genes for each pathway, and use Fisher's exact test to test if the number of hub genes in each pathway is altered from normal to cancer. Alternatively, if the distribution of topological ranks of gene in a pathway is altered between normal and cancer, this pathway might also be involved in cancer. Hence, we use the Kolmogorov-Smirnov test to detect pathways that have an altered distribution of topological ranks of genes between two phenotypes. We apply PoTRA to study hepatocellular carcinoma (HCC) and several subtypes of HCC. Very interestingly, we discover that all significant pathways in HCC are cancer-associated generally, while several significant pathways in subtypes of HCC are HCC subtype-associated specifically. In conclusion, PoTRA is a new approach to explore and discover pathways involved in cancer. PoTRA can be used as a complement to other existing methods to broaden our understanding of the biological mechanisms behind cancer at the system-level.
Collapse
Affiliation(s)
- Chaoxing Li
- School of Life Sciences, Arizona State University, Tempe, AZ, United States of America
| | - Li Liu
- Department of Biomedical Informatics, Arizona State University, Scottsdale, AZ, United States of America
| | - Valentin Dinu
- Department of Biomedical Informatics, Arizona State University, Scottsdale, AZ, United States of America
| |
Collapse
|
17
|
Nuncia-Cantarero M, Martinez-Canales S, Andrés-Pretel F, Santpere G, Ocaña A, Galan-Moya EM. Functional transcriptomic annotation and protein-protein interaction network analysis identify NEK2, BIRC5, and TOP2A as potential targets in obese patients with luminal A breast cancer. Breast Cancer Res Treat 2018; 168:613-623. [PMID: 29330624 PMCID: PMC5842257 DOI: 10.1007/s10549-017-4652-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/29/2017] [Indexed: 12/22/2022]
Abstract
PURPOSE Although obesity is a risk factor for breast cancer, little effort has been made in the identification of druggable molecular alterations in obese-breast cancer patients. Tumors are controlled by their surrounding microenvironment, in which the adipose tissue is a main component. In this work, we intended to describe molecular alterations at a transcriptomic and protein-protein interaction (PPI) level between obese and non-obese patients. METHODS AND RESULTS Gene expression data of 269 primary breast tumors were compared between normal-weight (BMI < 25, n = 130) and obese (IMC > 30, n = 139) patients. No significant differences were found for the global breast cancer population. However, within the luminal A subtype, upregulation of 81 genes was observed in the obese group (FC ≥ 1.4). Next, we explored the association of these genes with patient outcome, observing that 39 were linked with detrimental outcome. Their PPI map formed highly compact cluster and functional annotation analyses showed that cell cycle, cell proliferation, cell differentiation, and cellular response to extracellular stimuli were the more altered functions. Combined analyses of genes within the described functions are correlated with poor outcome. PPI network analyses for each function were to search for druggable opportunities. We identified 16 potentially druggable candidates. Among them, NEK2, BIRC5, and TOP2A were also found to be amplified in breast cancer, suggesting that they could act as strategic players in the obese-deregulated transcriptome. CONCLUSION In summary, our in silico analysis describes molecular alterations of luminal A tumors and proposes a druggable PPI network in obese patients with potential for translation to the clinical practice.
Collapse
Affiliation(s)
- Miriam Nuncia-Cantarero
- Translational Oncology Laboratory, Centro Regional de Investigaciones Biomédicas (CRIB), Universidad de Castilla La Mancha (UCLM), C/Almansa 14, 02008, Albacete, Spain
| | | | | | - Gabriel Santpere
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Alberto Ocaña
- Translational Research Unit, University Hospital, Albacete, Spain
| | - Eva Maria Galan-Moya
- Translational Oncology Laboratory, Centro Regional de Investigaciones Biomédicas (CRIB), Universidad de Castilla La Mancha (UCLM), C/Almansa 14, 02008, Albacete, Spain.
| |
Collapse
|
18
|
Bourdakou MM, Spyrou GM. Informed walks: whispering hints to gene hunters inside networks' jungle. BMC SYSTEMS BIOLOGY 2017; 11:97. [PMID: 29020948 PMCID: PMC5637247 DOI: 10.1186/s12918-017-0473-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 10/03/2017] [Indexed: 12/24/2022]
Abstract
Background Systemic approaches offer a different point of view on the analysis of several types of molecular associations as well as on the identification of specific gene communities in several cancer types. However, due to lack of sufficient data needed to construct networks based on experimental evidence, statistical gene co-expression networks are widely used instead. Many efforts have been made to exploit the information hidden in these networks. However, these approaches still need to capitalize comprehensively the prior knowledge encrypted into molecular pathway associations and improve their efficiency regarding the discovery of both exclusive subnetworks as candidate biomarkers and conserved subnetworks that may uncover common origins of several cancer types. Methods In this study we present the development of the Informed Walks model based on random walks that incorporate information from molecular pathways to mine candidate genes and gene-gene links. The proposed model has been applied to TCGA (The Cancer Genome Atlas) datasets from seven different cancer types, exploring the reconstructed co-expression networks of the whole set of genes and driving to highlighted sub-networks for each cancer type. In the sequel, we elucidated the impact of each subnetwork on the indication of underlying exclusive and common molecular mechanisms as well as on the short-listing of drugs that have the potential to suppress the corresponding cancer type through a drug-repurposing pipeline. Conclusions We have developed a method of gene subnetwork highlighting based on prior knowledge, capable to give fruitful insights regarding the underlying molecular mechanisms and valuable input to drug-repurposing pipelines for a variety of cancer types. Electronic supplementary material The online version of this article (10.1186/s12918-017-0473-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marilena M Bourdakou
- Bioinformatics ERA Chair, The Cyprus Institute of Neurology and Genetics, 6 International Airport Avenue, Ayios Dometios, 2370, Nicosia, Cyprus.,Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27, Athens, Greece
| | - George M Spyrou
- Bioinformatics ERA Chair, The Cyprus Institute of Neurology and Genetics, 6 International Airport Avenue, Ayios Dometios, 2370, Nicosia, Cyprus.
| |
Collapse
|
19
|
Karatzas E, Bourdakou MM, Kolios G, Spyrou GM. Drug repurposing in idiopathic pulmonary fibrosis filtered by a bioinformatics-derived composite score. Sci Rep 2017; 7:12569. [PMID: 28974751 PMCID: PMC5626774 DOI: 10.1038/s41598-017-12849-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 09/14/2017] [Indexed: 12/19/2022] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a rare disease of the respiratory system in which the lungs stiffen and get scarred, resulting in breathing weakness and eventually leading to death. Drug repurposing is a process that provides evidence for existing drugs that may also be effective in different diseases. In this study, we present a computational pipeline having as input a number of gene expression datasets from early and advanced stages of IPF and as output lists of repurposed drugs ranked with a novel composite score. We have devised and used a scoring formula in order to rank the repurposed drugs, consolidating the standard repurposing score with structural, functional and side effects' scores for each drug per stage of IPF. The whole pipeline involves the selection of proper gene expression datasets, data preprocessing and statistical analysis, selection of the most important genes related to the disease, analysis of biological pathways, investigation of related molecular mechanisms, identification of fibrosis-related microRNAs, drug repurposing, structural and literature-based analysis of the repurposed drugs.
Collapse
Affiliation(s)
- E Karatzas
- Department of Informatics and Telecommunications, University of Athens, 15784, Ilissia Athens, Greece
| | - M M Bourdakou
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27, Athens, Greece
- Bioinformatics ERA Chair, The Cyprus Institute of Neurology and Genetics, 6 International Airport Avenue, Nicosia, 2370, Cyprus
| | - G Kolios
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - G M Spyrou
- Bioinformatics ERA Chair, The Cyprus Institute of Neurology and Genetics, 6 International Airport Avenue, Nicosia, 2370, Cyprus.
| |
Collapse
|
20
|
Taye B, Vaz C, Tanavde V, Kuznetsov VA, Eisenhaber F, Sugrue RJ, Maurer-Stroh S. Benchmarking selected computational gene network growing tools in context of virus-host interactions. Sci Rep 2017; 7:5805. [PMID: 28724991 PMCID: PMC5517527 DOI: 10.1038/s41598-017-06020-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 06/07/2017] [Indexed: 01/04/2023] Open
Abstract
Several available online tools provide network growing functions where an algorithm utilizing different data sources suggests additional genes/proteins that should connect an input gene set into functionally meaningful networks. Using the well-studied system of influenza host interactions, we compare the network growing function of two free tools GeneMANIA and STRING and the commercial IPA for their performance of recovering known influenza A virus host factors previously identified from siRNA screens. The result showed that given small (~30 genes) or medium (~150 genes) input sets all three network growing tools detect significantly more known host factors than random human genes with STRING overall performing strongest. Extending the networks with all the three tools significantly improved the detection of GO biological processes of known host factors compared to not growing networks. Interestingly, the rate of identification of true host factors using computational network growing is equal or better to doing another experimental siRNA screening study which could also be true and applied to other biological pathways/processes.
Collapse
Affiliation(s)
- Biruhalem Taye
- Bioinformatics Institute, A*STAR, 30 Biopolis Street #07-01 Matrix, Singapore, 138671, Singapore. .,School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore. .,Aklilu Lemma Institute of Pathobiology, Addis Ababa University, P.O.BOX 1176, Addis Ababa, Ethiopia.
| | - Candida Vaz
- Bioinformatics Institute, A*STAR, 30 Biopolis Street #07-01 Matrix, Singapore, 138671, Singapore
| | - Vivek Tanavde
- Bioinformatics Institute, A*STAR, 30 Biopolis Street #07-01 Matrix, Singapore, 138671, Singapore.,Institute of Medical Biology, A*STAR, 8A Biomedical Grove, #06-06 Immunos, Singapore, 138648, Singapore
| | - Vladimir A Kuznetsov
- Bioinformatics Institute, A*STAR, 30 Biopolis Street #07-01 Matrix, Singapore, 138671, Singapore.,School of Computer Engineering, Nanyang Technological University, 50 Nanyang Drive, Singapore, 637553, Singapore
| | - Frank Eisenhaber
- Bioinformatics Institute, A*STAR, 30 Biopolis Street #07-01 Matrix, Singapore, 138671, Singapore.,Department of Biological Sciences, National University of Singapore, 8 Medical Drive, Singapore, 117597, Singapore.,School of Computer Engineering, Nanyang Technological University, 50 Nanyang Drive, Singapore, 637553, Singapore
| | - Richard J Sugrue
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Sebastian Maurer-Stroh
- Bioinformatics Institute, A*STAR, 30 Biopolis Street #07-01 Matrix, Singapore, 138671, Singapore.,Department of Biological Sciences, National University of Singapore, 8 Medical Drive, Singapore, 117597, Singapore.,National Public Health Laboratory, Ministry of Health, 3 Biopolis Drive, Synapse #05-14/16, Singapore, 138623, Singapore
| |
Collapse
|
21
|
Differential Regulatory Analysis Based on Coexpression Network in Cancer Research. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4241293. [PMID: 27597964 PMCID: PMC4997028 DOI: 10.1155/2016/4241293] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 06/09/2016] [Accepted: 06/12/2016] [Indexed: 12/15/2022]
Abstract
With rapid development of high-throughput techniques and accumulation of big transcriptomic data, plenty of computational methods and algorithms such as differential analysis and network analysis have been proposed to explore genome-wide gene expression characteristics. These efforts are aiming to transform underlying genomic information into valuable knowledges in biological and medical research fields. Recently, tremendous integrative research methods are dedicated to interpret the development and progress of neoplastic diseases, whereas differential regulatory analysis (DRA) based on gene coexpression network (GCN) increasingly plays a robust complement to regular differential expression analysis in revealing regulatory functions of cancer related genes such as evading growth suppressors and resisting cell death. Differential regulatory analysis based on GCN is prospective and shows its essential role in discovering the system properties of carcinogenesis features. Here we briefly review the paradigm of differential regulatory analysis based on GCN. We also focus on the applications of differential regulatory analysis based on GCN in cancer research and point out that DRA is necessary and extraordinary to reveal underlying molecular mechanism in large-scale carcinogenesis studies.
Collapse
|