1
|
Valerio M, Inno A, Zambelli A, Cortesi L, Lorusso D, Viassolo V, Verzè M, Nicolis F, Gori S. Deep Neural Network Integrated into Network-Based Stratification (D3NS): A Method to Uncover Cancer Subtypes from Somatic Mutations. Cancers (Basel) 2024; 16:2845. [PMID: 39199616 PMCID: PMC11352240 DOI: 10.3390/cancers16162845] [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: 07/19/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 09/01/2024] Open
Abstract
(1) Background: The identification of tumor subtypes is fundamental in precision medicine for accurate diagnoses and personalized therapies. Cancer development is often driven by the accumulation of somatic mutations that can cause alterations in tissue functions and morphologies. In this work, a method based on a deep neural network integrated into a network-based stratification framework (D3NS) is proposed to stratify tumors according to somatic mutations. (2) Methods: This approach leverages the power of deep neural networks to detect hidden information in the data by combining the knowledge contained in a network of gene interactions, as typical of network-based stratification methods. D3NS was applied using real-world data from The Cancer Genome Atlas for bladder, ovarian, and kidney cancers. (3) Results: This technique allows for the identification of tumor subtypes characterized by different survival rates and significant associations with several clinical outcomes (tumor stage, grade or response to therapy). (4) Conclusion: D3NS can provide a base model in cancer research and could be considered as a useful tool for tumor stratification, offering potential support in clinical settings.
Collapse
Affiliation(s)
- Matteo Valerio
- Medical Oncology, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Verona, Italy
| | - Alessandro Inno
- Medical Oncology, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Verona, Italy
| | - Alberto Zambelli
- Medical Oncology Unit, IRCCS Istituto Clinico Humanitas and Department of Biomedical Sciences, Humanitas University, 20089 Rozzano, Milan, Italy;
| | - Laura Cortesi
- Oncology, Hematology, and Respiratory Diseases, Azienda Ospedaliera-Universitaria, Policlinico di Modena, 41124 Modena, Italy
| | - Domenica Lorusso
- Gynecologic Oncology Unit, Humanitas San Pio X, Milan and Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Valeria Viassolo
- Medical Genetics, Medical Direction, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Verona, Italy;
| | - Matteo Verzè
- Medical Direction, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Verona, Italy; (M.V.)
| | - Fabrizio Nicolis
- Medical Direction, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Verona, Italy; (M.V.)
| | - Stefania Gori
- Medical Oncology, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Verona, Italy
| |
Collapse
|
2
|
Zou M, Li H, Su D, Xiong Y, Wei H, Wang S, Sun H, Wang T, Xi Q, Zuo Y, Yang L. Integrating somatic mutation profiles with structural deep clustering network for metabolic stratification in pancreatic cancer: a comprehensive analysis of prognostic and genomic landscapes. Brief Bioinform 2023; 25:bbad430. [PMID: 38040491 PMCID: PMC10783866 DOI: 10.1093/bib/bbad430] [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: 08/16/2023] [Revised: 09/29/2023] [Accepted: 11/05/2023] [Indexed: 12/03/2023] Open
Abstract
Pancreatic cancer is a globally recognized highly aggressive malignancy, posing a significant threat to human health and characterized by pronounced heterogeneity. In recent years, researchers have uncovered that the development and progression of cancer are often attributed to the accumulation of somatic mutations within cells. However, cancer somatic mutation data exhibit characteristics such as high dimensionality and sparsity, which pose new challenges in utilizing these data effectively. In this study, we propagated the discrete somatic mutation data of pancreatic cancer through a network propagation model based on protein-protein interaction networks. This resulted in smoothed somatic mutation profile data that incorporate protein network information. Based on this smoothed mutation profile data, we obtained the activity levels of different metabolic pathways in pancreatic cancer patients. Subsequently, using the activity levels of various metabolic pathways in cancer patients, we employed a deep clustering algorithm to establish biologically and clinically relevant metabolic subtypes of pancreatic cancer. Our study holds scientific significance in classifying pancreatic cancer based on somatic mutation data and may provide a crucial theoretical basis for the diagnosis and immunotherapy of pancreatic cancer patients.
Collapse
Affiliation(s)
- Min Zou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Honghao Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Dongqing Su
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yuqiang Xiong
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Haodong Wei
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Shiyuan Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Hongmei Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Tao Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Qilemuge Xi
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Yongchun Zuo
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
- Digital College, Inner Mongolia Intelligent Union Big Data Academy, Inner Mongolia Wesure Date Technology Co., Ltd. Hohhot 010010, China
- Inner Mongolia International Mongolian Hospital, Hohhot 010065, China
| | - Lei Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
3
|
Petti M, Farina L. Network medicine for patients' stratification: From single-layer to multi-omics. WIREs Mech Dis 2023; 15:e1623. [PMID: 37323106 DOI: 10.1002/wsbm.1623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 03/08/2023] [Accepted: 05/30/2023] [Indexed: 06/17/2023]
Abstract
Precision medicine research increasingly relies on the integrated analysis of multiple types of omics. In the era of big data, the large availability of different health-related information represents a great, but at the same time untapped, chance with a potentially fundamental role in the prevention, diagnosis and prognosis of diseases. Computational methods are needed to combine this data to create a comprehensive view of a given disease. Network science can model biomedical data in terms of relationships among molecular players of different nature and has been successfully proposed as a new paradigm for studying human diseases. Patient stratification is an open challenge aimed at identifying subtypes with different disease manifestations, severity, and expected survival time. Several stratification approaches based on high-throughput gene expression measurements have been successfully applied. However, few attempts have been proposed to exploit the integration of various genotypic and phenotypic data to discover novel sub-types or improve the detection of known groupings. This article is categorized under: Cancer > Biomedical Engineering Cancer > Computational Models Cancer > Genetics/Genomics/Epigenetics.
Collapse
Affiliation(s)
- Manuela Petti
- Department of Computer, Control and Management Engineering, Sapienza University of Rome, Rome, Italy
| | - Lorenzo Farina
- Department of Computer, Control and Management Engineering, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
4
|
Shetty KS, Jose A, Bani M, Vinod PK. Network diffusion-based approach for survival prediction and identification of biomarkers using multi-omics data of papillary renal cell carcinoma. Mol Genet Genomics 2023; 298:871-882. [PMID: 37093328 DOI: 10.1007/s00438-023-02022-4] [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: 09/08/2022] [Accepted: 04/12/2023] [Indexed: 04/25/2023]
Abstract
Identification of cancer subtypes based on molecular knowledge is crucial for improving the patient diagnosis, prognosis, and treatment. In this work, we integrated copy number variations (CNVs) and transcriptomic data of Kidney Papillary Renal Cell Carcinoma (KIRP) using a network diffusion strategy to stratify cancers into clinically and biologically relevant subtypes. We constructed GeneNet, a KIRP specific gene expression network from RNA-seq data. The copy number variation data was projected onto GeneNet and propagated on the network for clustering. We identified robust subtypes that are biologically informative and significantly associated with patient survival, tumor stage and clinical subtypes of KIRP. We performed a Singular Value Decomposition (SVD) analysis of KIRP subtypes, which revealed the genes/silent players related to poor survival. A differential gene expression analysis between subtypes showed that genes related to immune, extracellular matrix organization, and genomic instability are upregulated in the poor survival group. Overall, the network-based approach revealed the molecular subtypes of KIRP and captured the relationship between gene expression and CNVs. This framework can be further expanded to integrate other omics data.
Collapse
Affiliation(s)
- Keerthi S Shetty
- Center for Computational Natural Sciences and Bioinformatics, IIIT Hyderabad, Hyderabad, 500032, India
| | - Aswin Jose
- Center for Computational Natural Sciences and Bioinformatics, IIIT Hyderabad, Hyderabad, 500032, India
| | - Mihir Bani
- Center for Computational Natural Sciences and Bioinformatics, IIIT Hyderabad, Hyderabad, 500032, India
| | - P K Vinod
- Center for Computational Natural Sciences and Bioinformatics, IIIT Hyderabad, Hyderabad, 500032, India.
| |
Collapse
|
5
|
Network Approaches for Precision Oncology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1361:199-213. [DOI: 10.1007/978-3-030-91836-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
6
|
Sun Y, Wang Q, Zhang Y, Geng M, Wei Y, Liu Y, Liu S, Petersen RB, Yue J, Huang K, Zheng L. Multigenerational maternal obesity increases the incidence of HCC in offspring via miR-27a-3p. J Hepatol 2020; 73:603-615. [PMID: 32593682 DOI: 10.1016/j.jhep.2020.03.050] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Obesity is an independent risk factor for malignancies, including hepatocellular carcinoma (HCC). However, it remains unknown whether maternal obesity affects the incidence of HCC in offspring. Thus, we aimed to investigate this association and its underlying mechanisms. METHODS Diethylnitrosamine (DEN) was used to induce HCC in a high-fat diet (HFD)-induced multigenerational obesity model. RNA-sequencing was performed to identify the genes and microRNAs (miRNAs) that were altered over generations. The role of the miR-27a-3p-Acsl1/Aldh2 axis in HCC was evaluated in cell lines and HCC-bearing nude mice, and its intergenerational impact was studied in pregnant mice and their offspring. RESULTS Under HFD stress, maternal obesity caused susceptibility of offspring to DEN-induced HCC, and such susceptibility was cumulative over generations. We identified that Acsl1 and Aldh2, direct targets of miR-27a-3p, were gradually changed over generations. Under hyperlipidemic conditions, downregulation of Acsl1 and Aldh2 increased cell proliferation (in vitro) or tumor growth (in vivo) in synergy. Intratumor injection of an miR-27a-3p agomir exacerbated tumor growth by downregulating Acsl1 and Aldh2; while intratumor injection of an miR-27a-3p antagomir had the opposite effect. Moreover, serum miR-27a-3p levels gradually increased in the HFD-fed maternal lineage over generations. Injecting pregnant mice with an miR-27a-3p agomir not only upregulated hepatic miR-27a-3p and downregulated Acsl1/Aldh2 in offspring (fetus, young and adult stages), but also exacerbated HCC development in DEN-treated offspring. In human HCC, upregulated miR-27a-3p and downregulated Acsl1/Aldh2 were negatively correlated with survival on TCGA analysis; while, hepatic miR-27a-3p was negatively correlated with Acsl1/Aldh2 expression in tumor/non-tumor tissues from fatty/non-fatty livers. CONCLUSIONS Maternal obesity plays a role in regulating cumulative susceptibility to HCC development in offspring over multiple generations through the miR-27a-3p-Acsl1/Aldh2 axis. LAY SUMMARY It is not currently known how maternal obesity affects the incidence of liver cancer in offspring. In this study, we identified a microRNA (miR-27a-3p) that was upregulated in obese mothers and could be passed on to their offspring. This microRNA enhanced the risk of liver cancer in offspring by regulating 2 genes (Acsl1 and Aldh2). This mechanism could be a future therapeutic target.
Collapse
Affiliation(s)
- Yu Sun
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China, 430072
| | - Qing Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China, 430072
| | - Yu Zhang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, 430030
| | - Mengyuan Geng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China, 430072
| | - Yujuan Wei
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, 430030
| | - Yanrui Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China, 430072
| | - Shanshan Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China, 430072
| | - Robert B Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant, MI, USA, 48858
| | - Junqiu Yue
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, 430030
| | - Kun Huang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, 430030.
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China, 430072; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China, 430072.
| |
Collapse
|
7
|
Human Microbe-Disease Association Prediction by a Novel Double-Ended Random Walk with Restart. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3978702. [PMID: 32851068 PMCID: PMC7439206 DOI: 10.1155/2020/3978702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/07/2020] [Accepted: 07/20/2020] [Indexed: 11/17/2022]
Abstract
Microorganisms in the human body play a vital role in metabolism, immune defense, nutrient absorption, cancer control, and prevention of pathogen colonization. More and more biological and clinical studies have shown that the imbalance of microbial communities is closely related to the occurrence and development of various complex human diseases. Finding potential microbial-disease associations is critical for understanding the pathology of a few diseases and thus further improving disease diagnosis and prognosis. In this study, we proposed a novel computational model to predict disease-associated microbes. Specifically, we first constructed a heterogeneous interconnection network based on known microbe-disease associations deposited in a few databases, the similarity between diseases, and the similarity between microorganisms. We then predicted novel microbe-disease associations by a new method called the double-ended restart random walk model (DRWHMDA) implemented on the interconnection network. In addition, we performed case studies of colon cancer and asthma for further evaluation. The results indicate that 10 and 9 of the top 10 microorganisms predicted to be associated with colorectal cancer and asthma were validated by relevant literatures, respectively. Our method is expected to be effective in identifying disease-related microorganisms and will help to reveal the relationship between microorganisms and complex human diseases.
Collapse
|
8
|
Giang TT, Nguyen TP, Tran DH. Stratifying patients using fast multiple kernel learning framework: case studies of Alzheimer's disease and cancers. BMC Med Inform Decis Mak 2020; 20:108. [PMID: 32546157 PMCID: PMC7296686 DOI: 10.1186/s12911-020-01140-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 05/28/2020] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Predictive patient stratification is greatly emerging, because it allows us to prospectively identify which patients will benefit from what interventions before their condition worsens. In the biomedical research, a number of stratification methods have been successfully applied and have assisted treatment process. Because of heterogeneity and complexity of medical data, it is very challenging to integrate them and make use of them in practical clinic. There are two major challenges of data integration. Firstly, since the biomedical data has a high number of dimensions, combining multiple data leads to the hard problem of vast dimensional space handling. The computation is enormously complex and time-consuming. Secondly, the disparity of different data types causes another critical problem in machine learning for biomedical data. It has a great need to develop an efficient machine learning framework to handle the challenges. METHODS In this paper, we propose a fast-multiple kernel learning framework, referred to as fMKL-DR, that optimise equations to calculate matrix chain multiplication and reduce dimensions in data space. We applied our framework to two case studies, Alzheimer's disease (AD) patient stratification and cancer patient stratification. We performed several comparative evaluations on various biomedical datasets. RESULTS In the case study of AD patients, we enhanced significantly the multiple-ROIs approach based on MRI image data. The method could successfully classify not only AD patients and non-AD patients but also different phases of AD patients with AUC close to 1. In the case study of cancer patients, the framework was applied to six types of cancers, i.e., glioblastoma multiforme cancer, ovarian cancer, lung cancer, breast cancer, kidney cancer, and liver cancer. We efficiently integrated gene expression, miRNA expression, and DNA methylation. The results showed that the classification model basing on integrated datasets was much more accurate than classification model basing on the single data type. CONCLUSIONS The results demonstrated that the fMKL-DR remarkably improves computational cost and accuracy for both AD patient and cancer patient stratification. We optimised the data integration, dimension reduction, and kernel fusion. Our framework has great potential for mining large-scale cohort data and aiding personalised prevention.
Collapse
Affiliation(s)
- Thanh-Trung Giang
- VNU University of Engineering and Technology, Hanoi, Vietnam.,TayBac University, Son La, Vietnam
| | - Thanh-Phuong Nguyen
- Life Sciences Research Unit, Belval, University of Luxembourg, Luxembourg City, Luxembourg. .,Megeno S.A., Belval, Esch-sur-Alzette, Luxembourg.
| | | |
Collapse
|
9
|
Di Nanni N, Bersanelli M, Milanesi L, Mosca E. Network Diffusion Promotes the Integrative Analysis of Multiple Omics. Front Genet 2020; 11:106. [PMID: 32180795 PMCID: PMC7057719 DOI: 10.3389/fgene.2020.00106] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/29/2020] [Indexed: 02/01/2023] Open
Abstract
The development of integrative methods is one of the main challenges in bioinformatics. Network-based methods for the analysis of multiple gene-centered datasets take into account known and/or inferred relations between genes. In the last decades, the mathematical machinery of network diffusion—also referred to as network propagation—has been exploited in several network-based pipelines, thanks to its ability of amplifying association between genes that lie in network proximity. Indeed, network diffusion provides a quantitative estimation of network proximity between genes associated with one or more different data types, from simple binary vectors to real vectors. Therefore, this powerful data transformation method has also been increasingly used in integrative analyses of multiple collections of biological scores and/or one or more interaction networks. We present an overview of the state of the art of bioinformatics pipelines that use network diffusion processes for the integrative analysis of omics data. We discuss the fundamental ways in which network diffusion is exploited, open issues and potential developments in the field. Current trends suggest that network diffusion is a tool of broad utility in omics data analysis. It is reasonable to think that it will continue to be used and further refined as new data types arise (e.g. single cell datasets) and the identification of system-level patterns will be considered more and more important in omics data analysis.
Collapse
Affiliation(s)
- Noemi Di Nanni
- Institute of Biomedical Technologies, National Research Council, Milan, Italy.,Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy
| | - Matteo Bersanelli
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy.,National Institute of Nuclear Physics (INFN), Bologna, Italy
| | - Luciano Milanesi
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Ettore Mosca
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| |
Collapse
|
10
|
Li GXH, Munro D, Fermin D, Vogel C, Choi H. A protein-centric approach for exome variant aggregation enables sensitive association analysis with clinical outcomes. Hum Mutat 2020; 41:934-945. [PMID: 31930623 DOI: 10.1002/humu.23979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 12/14/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023]
Abstract
Somatic mutations are early drivers of tumorigenesis and tumor progression. However, the mutations typically occur at variable positions across different individuals, resulting in the data being too sparse to test meaningful associations between variants and phenotypes. To overcome this challenge, we devised a novel approach called Gene-to-Protein-to-Disease (GPD) which accumulates variants into new sequence units as the degree of genetic assault on structural or functional units of each protein. The variant frequencies in the sequence units were highly reproducible between two large cancer cohorts. Survival analysis identified 232 sequence units in which somatic mutations had deleterious effects on overall survival, including consensus driver mutations obtained from multiple calling algorithms. By contrast, around 76% of the survival predictive units had been undetected by conventional gene-level analysis. We demonstrate the ability of these signatures to separate patient groups according to overall survival, therefore, providing novel prognostic tools for various cancers. GPD also identified sequence units with somatic mutations whose impact on survival was modified by the occupancy of germline variants in the surrounding regions. The findings indicate that a patient's genetic predisposition interacts with the effect of somatic mutations on survival outcomes in some cancers.
Collapse
Affiliation(s)
- Ginny X H Li
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Dan Munro
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, New York
| | - Damian Fermin
- Department of Pediatric Nephrology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Christine Vogel
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, New York
| | - Hyungwon Choi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research, Singapore, Singapore
| |
Collapse
|
11
|
Dincer C, Kaya T, Keskin O, Gursoy A, Tuncbag N. 3D spatial organization and network-guided comparison of mutation profiles in Glioblastoma reveals similarities across patients. PLoS Comput Biol 2019; 15:e1006789. [PMID: 31527881 PMCID: PMC6782092 DOI: 10.1371/journal.pcbi.1006789] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 10/08/2019] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of brain tumor. Molecular heterogeneity is a hallmark of GBM tumors that is a barrier in developing treatment strategies. In this study, we used the nonsynonymous mutations of GBM tumors deposited in The Cancer Genome Atlas (TCGA) and applied a systems level approach based on biophysical characteristics of mutations and their organization in patient-specific subnetworks to reduce inter-patient heterogeneity and to gain potential clinically relevant insights. Approximately 10% of the mutations are located in "patches" which are defined as the set of residues spatially in close proximity that are mutated across multiple patients. Grouping mutations as 3D patches reduces the heterogeneity across patients. There are multiple patches that are relatively small in oncogenes, whereas there are a small number of very large patches in tumor suppressors. Additionally, different patches in the same protein are often located at different domains that can mediate different functions. We stratified the patients into five groups based on their potentially affected pathways that are revealed from the patient-specific subnetworks. These subnetworks were constructed by integrating mutation profiles of the patients with the interactome data. Network-guided clustering showed significant association between the groups and patient survival (P-value = 0.0408). Also, each group carries a set of signature 3D mutation patches that affect predominant pathways. We integrated drug sensitivity data of GBM cell lines with the mutation patches and the patient groups to analyze the possible therapeutic outcome of these patches. We found that Pazopanib might be effective in Group 3 by targeting CSF1R. Additionally, inhibiting ATM that is a mediator of PTEN phosphorylation may be ineffective in Group 2. We believe that from mutations to networks and eventually to clinical and therapeutic data, this study provides a novel perspective in the network-guided precision medicine.
Collapse
Affiliation(s)
- Cansu Dincer
- Department of Health Informatics, Graduate School of Informatics, METU, Ankara, Turkey
| | - Tugba Kaya
- Department of Health Informatics, Graduate School of Informatics, METU, Ankara, Turkey
| | - Ozlem Keskin
- Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey
- Research Center for Translational Medicine (KUTTAM), Koc University, Istanbul, Turkey
| | - Attila Gursoy
- Research Center for Translational Medicine (KUTTAM), Koc University, Istanbul, Turkey
- Department of Computer Engineering, Koc University, Istanbul, Turkey
| | - Nurcan Tuncbag
- Department of Health Informatics, Graduate School of Informatics, METU, Ankara, Turkey
- Cancer Systems Biology Laboratory (CanSyL-METU), Ankara, Turkey
- * E-mail:
| |
Collapse
|
12
|
Timilsina M, Yang H, Sahay R, Rebholz-Schuhmann D. Predicting links between tumor samples and genes using 2-Layered graph based diffusion approach. BMC Bioinformatics 2019; 20:462. [PMID: 31500564 PMCID: PMC6734347 DOI: 10.1186/s12859-019-3056-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 08/26/2019] [Indexed: 12/21/2022] Open
Abstract
Background Determining the association between tumor sample and the gene is demanding because it requires a high cost for conducting genetic experiments. Thus, the discovered association between tumor sample and gene further requires clinical verification and validation. This entire mechanism is time-consuming and expensive. Due to this issue, predicting the association between tumor samples and genes remain a challenge in biomedicine. Results Here we present, a computational model based on a heat diffusion algorithm which can predict the association between tumor samples and genes. We proposed a 2-layered graph. In the first layer, we constructed a graph of tumor samples and genes where these two types of nodes are connected by “hasGene” relationship. In the second layer, the gene nodes are connected by “interaction” relationship. We applied the heat diffusion algorithms in nine different variants of genetic interaction networks extracted from STRING and BioGRID database. The heat diffusion algorithm predicted the links between tumor samples and genes with mean AUC-ROC score of 0.84. This score is obtained by using weighted genetic interactions of fusion or co-occurrence channels from the STRING database. For the unweighted genetic interaction from the BioGRID database, the algorithms predict the links with an AUC-ROC score of 0.74. Conclusions We demonstrate that the gene-gene interaction scores could improve the predictive power of the heat diffusion model to predict the links between tumor samples and genes. We showed the efficient runtime of the heat diffusion algorithm in various genetic interaction network. We statistically validated our prediction quality of the links between tumor samples and genes. Electronic supplementary material The online version of this article (10.1186/s12859-019-3056-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mohan Timilsina
- Insight Centre for Data Analytics, National University of Ireland Galway, Galway, Ireland.
| | - Haixuan Yang
- School of Mathematics Statistics and Applied Mathematics, National University of Ireland Galway, Galway, Ireland
| | - Ratnesh Sahay
- Insight Centre for Data Analytics, National University of Ireland Galway, Galway, Ireland
| | | |
Collapse
|
13
|
Tan A, Huang H, Zhang P, Li S. Network-based cancer precision medicine: A new emerging paradigm. Cancer Lett 2019; 458:39-45. [DOI: 10.1016/j.canlet.2019.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/29/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022]
|
14
|
Capriotti E, Ozturk K, Carter H. Integrating molecular networks with genetic variant interpretation for precision medicine. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2018; 11:e1443. [PMID: 30548534 PMCID: PMC6450710 DOI: 10.1002/wsbm.1443] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/23/2018] [Accepted: 10/30/2018] [Indexed: 02/01/2023]
Abstract
More reliable and cheaper sequencing technologies have revealed the vast mutational landscapes characteristic of many phenotypes. The analysis of such genetic variants has led to successful identification of altered proteins underlying many Mendelian disorders. Nevertheless the simple one‐variant one‐phenotype model valid for many monogenic diseases does not capture the complexity of polygenic traits and disorders. Although experimental and computational approaches have improved detection of functionally deleterious variants and important interactions between gene products, the development of comprehensive models relating genotype and phenotypes remains a challenge in the field of genomic medicine. In this context, a new view of the pathologic state as significant perturbation of the network of interactions between biomolecules is crucial for the identification of biochemical pathways associated with complex phenotypes. Seminal studies in systems biology combined the analysis of genetic variation with protein–protein interaction networks to demonstrate that even as biological systems evolve to be robust to genetic variation, their topologies create disease vulnerabilities. More recent analyses model the impact of genetic variants as changes to the “wiring” of the interactome to better capture heterogeneity in genotype–phenotype relationships. These studies lay the foundation for using networks to predict variant effects at scale using machine‐learning or algorithmic approaches. A wealth of databases and resources for the annotation of genotype–phenotype relationships have been developed to support developments in this area. This overview describes how study of the molecular interactome has generated insights linking the organization of biological systems to disease mechanism, and how this information can enable precision medicine. This article is categorized under:
Translational, Genomic, and Systems Medicine > Translational Medicine Biological Mechanisms > Cell Signaling Models of Systems Properties and Processes > Mechanistic Models Analytical and Computational Methods > Computational Methods
Collapse
Affiliation(s)
- Emidio Capriotti
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Kivilcim Ozturk
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, California
| | - Hannah Carter
- Department of Medicine and Institute for Genomic Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
15
|
Ozturk K, Dow M, Carlin DE, Bejar R, Carter H. The Emerging Potential for Network Analysis to Inform Precision Cancer Medicine. J Mol Biol 2018; 430:2875-2899. [PMID: 29908887 PMCID: PMC6097914 DOI: 10.1016/j.jmb.2018.06.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/30/2018] [Accepted: 06/06/2018] [Indexed: 12/19/2022]
Abstract
Precision cancer medicine promises to tailor clinical decisions to patients using genomic information. Indeed, successes of drugs targeting genetic alterations in tumors, such as imatinib that targets BCR-ABL in chronic myelogenous leukemia, have demonstrated the power of this approach. However, biological systems are complex, and patients may differ not only by the specific genetic alterations in their tumor, but also by more subtle interactions among such alterations. Systems biology and more specifically, network analysis, provides a framework for advancing precision medicine beyond clinical actionability of individual mutations. Here we discuss applications of network analysis to study tumor biology, early methods for N-of-1 tumor genome analysis, and the path for such tools to the clinic.
Collapse
Affiliation(s)
- Kivilcim Ozturk
- Department of Medicine, Division of Medical Genetics, University of California San Diego, La Jolla, CA 92093, USA; Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Michelle Dow
- Department of Medicine, Division of Medical Genetics, University of California San Diego, La Jolla, CA 92093, USA; Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Daniel E Carlin
- Department of Medicine, Division of Medical Genetics, University of California San Diego, La Jolla, CA 92093, USA
| | - Rafael Bejar
- Moores Cancer Center, Division of Hematology and Oncology, University of California San Diego, La Jolla, CA 92093, USA
| | - Hannah Carter
- Department of Medicine, Division of Medical Genetics, University of California San Diego, La Jolla, CA 92093, USA; Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA 92093, USA; Moores Cancer Center and Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA; CIFAR, MaRS Centre, West Tower, 661 University Ave., Suite 505, Toronto, ON M5G 1M1, Canada.
| |
Collapse
|
16
|
Abstract
Network propagation is a powerful tool for genetic analysis which is widely used to identify genes and genetic modules that underlie a process of interest. Here we provide a graphical, web-based platform (http://anat.cs.tau.ac.il/WebPropagate/) in which researchers can easily apply variants of this method to data sets of interest using up-to-date networks of protein-protein interactions in several organisms.
Collapse
Affiliation(s)
- Hadas Biran
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv 69978, Israel
| | - Tovi Almozlino
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv 69978, Israel
| | - Martin Kupiec
- Department of Molecular Microbiology and Biotechnology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Roded Sharan
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
17
|
Choi M, Shi J, Zhu Y, Yang R, Cho KH. Network dynamics-based cancer panel stratification for systemic prediction of anticancer drug response. Nat Commun 2017; 8:1940. [PMID: 29208897 PMCID: PMC5717260 DOI: 10.1038/s41467-017-02160-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 11/09/2017] [Indexed: 01/04/2023] Open
Abstract
Cancer is a complex disease involving multiple genomic alterations that disrupt the dynamic response of signaling networks. The heterogeneous nature of cancer, which results in highly variable drug response, is a major obstacle to developing effective cancer therapy. Previous studies of cancer therapeutic response mostly focus on static analysis of genome-wide alterations, thus they are unable to unravel the dynamic, network-specific origin of variation. Here we present a network dynamics-based approach to integrate cancer genomics with dynamics of biological network for drug response prediction and design of drug combination. We select the p53 network as an example and analyze its cancer-specific state transition dynamics under distinct anticancer drug treatments by attractor landscape analysis. Our results not only enable stratification of cancer into distinct drug response groups, but also reveal network-specific drug targets that maximize p53 network-mediated cell death, providing a basis to design combinatorial therapeutic strategies for distinct cancer genomic subtypes. Genomic alterations underlie the variability of drug responses between cancers, but our mechanistic understanding is limited. Here the authors use the p53 network to study how rewiring of signalling networks by genomic alterations impact their dynamic response to pharmacological perturbation.
Collapse
Affiliation(s)
- Minsoo Choi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jue Shi
- Center for Quantitative Systems Biology and Department of Physics, Hong Kong Baptist University, Hong Kong, 999077, China
| | - Yanting Zhu
- Center for Quantitative Systems Biology and Department of Physics, Hong Kong Baptist University, Hong Kong, 999077, China
| | - Ruizhen Yang
- Center for Quantitative Systems Biology and Department of Physics, Hong Kong Baptist University, Hong Kong, 999077, China
| | - Kwang-Hyun Cho
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
18
|
Carlin DE, Demchak B, Pratt D, Sage E, Ideker T. Network propagation in the cytoscape cyberinfrastructure. PLoS Comput Biol 2017; 13:e1005598. [PMID: 29023449 PMCID: PMC5638226 DOI: 10.1371/journal.pcbi.1005598] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 05/29/2017] [Indexed: 01/15/2023] Open
Abstract
Network propagation is an important and widely used algorithm in systems biology, with applications in protein function prediction, disease gene prioritization, and patient stratification. However, up to this point it has required significant expertise to run. Here we extend the popular network analysis program Cytoscape to perform network propagation as an integrated function. Such integration greatly increases the access to network propagation by putting it in the hands of biologists and linking it to the many other types of network analysis and visualization available through Cytoscape. We demonstrate the power and utility of the algorithm by identifying mutations conferring resistance to Vemurafenib.
Collapse
Affiliation(s)
- Daniel E. Carlin
- Department of Medicine, University of California-San Diego, San Diego, California, United States of America
- * E-mail:
| | - Barry Demchak
- Department of Medicine, University of California-San Diego, San Diego, California, United States of America
| | - Dexter Pratt
- Department of Medicine, University of California-San Diego, San Diego, California, United States of America
| | - Eric Sage
- Department of Medicine, University of California-San Diego, San Diego, California, United States of America
| | - Trey Ideker
- Department of Medicine, University of California-San Diego, San Diego, California, United States of America
| |
Collapse
|
19
|
Prevalence of high-risk human papillomavirus infection and cancer gene mutations in nonmalignant tonsils. Oral Oncol 2017; 73:77-82. [PMID: 28939080 DOI: 10.1016/j.oraloncology.2017.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 02/01/2023]
Abstract
OBJECTIVES To analyze the prevalence of high-risk HPV (human papillomavirus) and genetic alterations in nonmalignant tonsils. METHODS We collected benign fresh tonsillar tissue specimens from 477 patients undergoing tonsillectomy because of chronic tonsillitis or tonsillar hypertrophy in 2012 (Group A, n=237) and in 2015 (Group B, n=240). Luminex xMAP technique served to detect E6/E7 DNA from 16 different high-risk HPV types. Tonsillar DNA and peripheral blood leukocyte DNA from the infected individuals were analyzed using Nimblegen SeqCap EZ Comprehensive Cancer Design panel. The panel targets 578 different genes that are relevant in carcinogenesis. HPV negative tonsillar specimens from age- and gender matched individuals were used as controls. All specimens harboring high-risk HPV were analyzed using fluorescence in situ hybridization (FISH). RESULTS Five of 477 (1.0%) patients tested positive for the following HPV types: HPV16 (two cases), HPV52 (one case), HPV66 (one case), HPV52 and HPV68 (coinfection, one case). FISH analyses showed that the appearance of HPV in specimens infected with HPV 16 was episomal. Benign tonsils infected with high-risk HPV harbored mutations in EP300, NF1, PIK3CA, and RB1 which are considered relevant in the development of HPV-associated head and neck squamous cell carcinoma (SCC). CONCLUSIONS The prevalence of high-risk HPV in nonmalignant tonsils is low. High-risk HPV positive tonsils harbored mutations in genes that are commonly altered in HPV-associated head and neck SCC. The role of these mutations in tonsillar carcinogenesis is an interesting target for future research.
Collapse
|
20
|
He Z, Zhang J, Yuan X, Liu Z, Liu B, Tuo S, Liu Y. Network based stratification of major cancers by integrating somatic mutation and gene expression data. PLoS One 2017; 12:e0177662. [PMID: 28520777 PMCID: PMC5433734 DOI: 10.1371/journal.pone.0177662] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/01/2017] [Indexed: 11/20/2022] Open
Abstract
The stratification of cancer into subtypes that are significantly associated with clinical outcomes is beneficial for targeted prognosis and treatment. In this study, we integrated somatic mutation and gene expression data to identify clusters of patients. In contrast to previous studies, we constructed cancer-type-specific significant co-expression networks (SCNs) rather than using a fixed gene network across all cancers, such as the network-based stratification (NBS) method, which ignores cancer heterogeneity. For each type of cancer, the gene expression data were used to construct the SCN network, while the gene somatic mutation data were mapped onto the network, propagated, and used for further clustering. For the clustering, we adopted an improved network-regularized non-negative matrix factorization (netNMF) (netNMF_HC) for a more precise classification. We applied our method to various datasets, including ovarian cancer (OV), lung adenocarcinoma (LUAD) and uterine corpus endometrial carcinoma (UCEC) cohorts derived from the TCGA (The Cancer Genome Atlas) project. Based on the results, we evaluated the performance of our method to identify survival-relevant subtypes and further compared it to the NBS method, which adopts priori networks and netNMF algorithm. The proposed algorithm outperformed the NBS method in identifying informative cancer subtypes that were significantly associated with clinical outcomes in most cancer types we studied. In particular, our method identified survival-associated UCEC subtypes that were not identified by the NBS method. Our analysis indicated valid subtyping of patient could be applied by mutation data with cancer-type-specific SCNs and netNMF_HC for individual cancers because of specific cancer co-expression patterns and more precise clustering.
Collapse
Affiliation(s)
- Zongzhen He
- School of Computer Science and Technology, Xidian University, Xi’an, PR China
| | - Junying Zhang
- School of Computer Science and Technology, Xidian University, Xi’an, PR China
- * E-mail:
| | - Xiguo Yuan
- School of Computer Science and Technology, Xidian University, Xi’an, PR China
| | - Zhaowen Liu
- School of Computer Science and Technology, Xidian University, Xi’an, PR China
| | - Baobao Liu
- School of Computer Science and Technology, Xidian University, Xi’an, PR China
| | - Shouheng Tuo
- School of Computer Science and Technology, Xidian University, Xi’an, PR China
| | - Yajun Liu
- School of Computer Science and Technology, Xidian University, Xi’an, PR China
| |
Collapse
|
21
|
Cabanillas R, Diñeiro M, Castillo D, Pruneda PC, Penas C, Cifuentes GA, de Vicente Á, Durán NS, Álvarez R, Ordóñez GR, Cadiñanos J. A novel molecular diagnostics platform for somatic and germline precision oncology. Mol Genet Genomic Med 2017; 5:336-359. [PMID: 28717660 PMCID: PMC5511795 DOI: 10.1002/mgg3.291] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 12/28/2022] Open
Abstract
Background Next‐generation sequencing (NGS) opens new options in clinical oncology, from therapy selection to genetic counseling. However, realization of this potential not only requires succeeding in the bioinformatics and interpretation of the results, but also in their integration into the clinical practice. We have developed a novel NGS diagnostic platform aimed at detecting (1) somatic genomic alterations associated with the response to approved targeted cancer therapies and (2) germline mutations predisposing to hereditary malignancies. Methods Next‐generation sequencing libraries enriched in the exons of 215 cancer genes (97 for therapy selection and 148 for predisposition, with 30 informative for both applications), as well as selected introns from 17 genes involved in drug‐related rearrangements, were prepared from 39 tumors (paraffin‐embedded tissues/cytologies), 36 germline samples (blood) and 10 cell lines using hybrid capture. Analysis of NGS results was performed with specifically developed bioinformatics pipelines. Results The platform detects single‐nucleotide variants (SNVs) and insertions/deletions (indels) with sensitivity and specificity >99.5% (allelic frequency ≥0.1), as well as copy‐number variants (CNVs) and rearrangements. Somatic testing identified tailored approved targeted drugs in 35/39 tumors (89.74%), showing a diagnostic yield comparable to that of leading commercial platforms. A somatic EGFR p.E746_S752delinsA mutation in a mediastinal metastasis from a breast cancer prompted its anatomopathologic reassessment, its definite reclassification as a lung cancer and its treatment with gefitinib (partial response sustained for 15 months). Testing of 36 germline samples identified two pathogenic mutations (in CDKN2A and BRCA2). We propose a strategy for interpretation and reporting of results adaptable to the aim of the request, the availability of tumor and/or normal samples and the scope of the informed consent. Conclusion With an adequate methodology, it is possible to translate to the clinical practice the latest advances in precision oncology, integrating under the same platform the identification of somatic and germline genomic alterations.
Collapse
Affiliation(s)
- Rubén Cabanillas
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.Avda. Richard Grandío s/n33193OviedoSpain
| | - Marta Diñeiro
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.Avda. Richard Grandío s/n33193OviedoSpain
| | - David Castillo
- Disease Research And Medicine (DREAMgenics) S. L.Vivero Empresarial de Ciencias de la SaludC/Colegio Santo Domingo de Guzmán s/n33011OviedoSpain
| | - Patricia C Pruneda
- Disease Research And Medicine (DREAMgenics) S. L.Vivero Empresarial de Ciencias de la SaludC/Colegio Santo Domingo de Guzmán s/n33011OviedoSpain
| | - Cristina Penas
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.Avda. Richard Grandío s/n33193OviedoSpain
| | - Guadalupe A Cifuentes
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.Avda. Richard Grandío s/n33193OviedoSpain
| | - Álvaro de Vicente
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.Avda. Richard Grandío s/n33193OviedoSpain
| | - Noelia S Durán
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.Avda. Richard Grandío s/n33193OviedoSpain
| | - Rebeca Álvarez
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.Avda. Richard Grandío s/n33193OviedoSpain
| | - Gonzalo R Ordóñez
- Disease Research And Medicine (DREAMgenics) S. L.Vivero Empresarial de Ciencias de la SaludC/Colegio Santo Domingo de Guzmán s/n33011OviedoSpain
| | - Juan Cadiñanos
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA) S. A.Avda. Richard Grandío s/n33193OviedoSpain
| |
Collapse
|
22
|
Yang L, Wang S, Zhou M, Chen X, Jiang W, Zuo Y, Lv Y. Molecular classification of prostate adenocarcinoma by the integrated somatic mutation profiles and molecular network. Sci Rep 2017; 7:738. [PMID: 28389666 PMCID: PMC5429686 DOI: 10.1038/s41598-017-00872-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/20/2017] [Indexed: 01/01/2023] Open
Abstract
Prostate cancer is one of the most common cancers in men and a leading cause of cancer death worldwide, displaying a broad range of heterogeneity in terms of clinical and molecular behavior. Increasing evidence suggests that classifying prostate cancers into distinct molecular subtypes is critical to exploring the potential molecular variation underlying this heterogeneity and to better treat this cancer. In this study, the somatic mutation profiles of prostate cancer were downloaded from the TCGA database and used as the source nodes of the random walk with restart algorithm (RWRA) for generating smoothed mutation profiles in the STRING network. The smoothed mutation profiles were selected as the input matrix of the Graph-regularized Nonnegative Matrix Factorization (GNMF) for classifying patients into distinct molecular subtypes. The results were associated with most of the clinical and pathological outcomes. In addition, some bioinformatics analyses were performed for the robust subtyping, and good results were obtained. These results indicated that prostate cancers can be usefully classified according to their mutation profiles, and we hope that these subtypes will help improve the treatment stratification of this cancer in the future.
Collapse
Affiliation(s)
- Lei Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| | - Shiyuan Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Meng Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xiaowen Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Wei Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yongchun Zuo
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, 010021, China.
| | - Yingli Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
23
|
Fiorito E, Sharma Y, Gilfillan S, Wang S, Singh SK, Satheesh SV, Katika MR, Urbanucci A, Thiede B, Mills IG, Hurtado A. CTCF modulates Estrogen Receptor function through specific chromatin and nuclear matrix interactions. Nucleic Acids Res 2016; 44:10588-10602. [PMID: 27638884 PMCID: PMC5159541 DOI: 10.1093/nar/gkw785] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 08/23/2016] [Accepted: 08/24/2016] [Indexed: 11/27/2022] Open
Abstract
Enhancer regions and transcription start sites of estrogen-target regulated genes are connected by means of Estrogen Receptor long-range chromatin interactions. Yet, the complete molecular mechanisms controlling the transcriptional output of engaged enhancers and subsequent activation of coding genes remain elusive. Here, we report that CTCF binding to enhancer RNAs is enriched when breast cancer cells are stimulated with estrogen. CTCF binding to enhancer regions results in modulation of estrogen-induced gene transcription by preventing Estrogen Receptor chromatin binding and by hindering the formation of additional enhancer-promoter ER looping. Furthermore, the depletion of CTCF facilitates the expression of target genes associated with cell division and increases the rate of breast cancer cell proliferation. We have also uncovered a genomic network connecting loci enriched in cell cycle regulator genes to nuclear lamina that mediates the CTCF function. The nuclear lamina and chromatin interactions are regulated by estrogen-ER. We have observed that the chromatin loops formed when cells are treated with estrogen establish contacts with the nuclear lamina. Once there, the portion of CTCF associated with the nuclear lamina interacts with enhancer regions, limiting the formation of ER loops and the induction of genes present in the loop. Collectively, our results reveal an important, unanticipated interplay between CTCF and nuclear lamina to control the transcription of ER target genes, which has great implications in the rate of growth of breast cancer cells.
Collapse
Affiliation(s)
- Elisa Fiorito
- Breast Cancer Research group, Nordic EMBL Partnership, Centre for Molecular Medicine Norway (NCMM), University of Oslo, P.O. 1137 Blindern, 0318 Oslo, Norway
| | - Yogita Sharma
- Breast Cancer Research group, Nordic EMBL Partnership, Centre for Molecular Medicine Norway (NCMM), University of Oslo, P.O. 1137 Blindern, 0318 Oslo, Norway
| | - Siv Gilfillan
- Breast Cancer Research group, Nordic EMBL Partnership, Centre for Molecular Medicine Norway (NCMM), University of Oslo, P.O. 1137 Blindern, 0318 Oslo, Norway
| | - Shixiong Wang
- Breast Cancer Research group, Nordic EMBL Partnership, Centre for Molecular Medicine Norway (NCMM), University of Oslo, P.O. 1137 Blindern, 0318 Oslo, Norway
| | - Sachin Kumar Singh
- Breast Cancer Research group, Nordic EMBL Partnership, Centre for Molecular Medicine Norway (NCMM), University of Oslo, P.O. 1137 Blindern, 0318 Oslo, Norway
| | - Somisetty V Satheesh
- Breast Cancer Research group, Nordic EMBL Partnership, Centre for Molecular Medicine Norway (NCMM), University of Oslo, P.O. 1137 Blindern, 0318 Oslo, Norway
| | - Madhumohan R Katika
- Breast Cancer Research group, Nordic EMBL Partnership, Centre for Molecular Medicine Norway (NCMM), University of Oslo, P.O. 1137 Blindern, 0318 Oslo, Norway
| | - Alfonso Urbanucci
- Prostate Cancer Research group, Nordic EMBL Partnership, Centre for Molecular Medicine Norway (NCMM), University of Oslo, P.O. 1137 Blindern, 0318 Oslo, Norway.,Department of Molecular Oncology, Institute of Cancer Research and Oslo University Hospital, Oslo, Norway
| | - Bernd Thiede
- Proteomics Group, Department of Biosciences, Faculty of Mathematics and Natural Science, University of Oslo, P.O. 1066 Blindern, 0316 Oslo, Norway
| | - Ian G Mills
- Prostate Cancer Research group, Nordic EMBL Partnership, Centre for Molecular Medicine Norway (NCMM), University of Oslo, P.O. 1137 Blindern, 0318 Oslo, Norway.,Department of Molecular Oncology, Institute of Cancer Research and Oslo University Hospital, Oslo, Norway.,PCUK Movember Centre of Excellence, CCRCB, Queen's University, Belfast, UK
| | - Antoni Hurtado
- Breast Cancer Research group, Nordic EMBL Partnership, Centre for Molecular Medicine Norway (NCMM), University of Oslo, P.O. 1137 Blindern, 0318 Oslo, Norway .,Department of Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, N-0310 Oslo, Norway
| |
Collapse
|
24
|
Algorithmic methods to infer the evolutionary trajectories in cancer progression. Proc Natl Acad Sci U S A 2016; 113:E4025-34. [PMID: 27357673 DOI: 10.1073/pnas.1520213113] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The genomic evolution inherent to cancer relates directly to a renewed focus on the voluminous next-generation sequencing data and machine learning for the inference of explanatory models of how the (epi)genomic events are choreographed in cancer initiation and development. However, despite the increasing availability of multiple additional -omics data, this quest has been frustrated by various theoretical and technical hurdles, mostly stemming from the dramatic heterogeneity of the disease. In this paper, we build on our recent work on the "selective advantage" relation among driver mutations in cancer progression and investigate its applicability to the modeling problem at the population level. Here, we introduce PiCnIc (Pipeline for Cancer Inference), a versatile, modular, and customizable pipeline to extract ensemble-level progression models from cross-sectional sequenced cancer genomes. The pipeline has many translational implications because it combines state-of-the-art techniques for sample stratification, driver selection, identification of fitness-equivalent exclusive alterations, and progression model inference. We demonstrate PiCnIc's ability to reproduce much of the current knowledge on colorectal cancer progression as well as to suggest novel experimentally verifiable hypotheses.
Collapse
|
25
|
Ruan J, Jin V, Huang Y, Xu H, Edwards JS, Chen Y, Zhao Z. Education, collaboration, and innovation: intelligent biology and medicine in the era of big data. BMC Genomics 2015; 16 Suppl 7:S1. [PMID: 26099197 PMCID: PMC4474420 DOI: 10.1186/1471-2164-16-s7-s1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Here we present a summary of the 2014 International Conference on Intelligent Biology and Medicine (ICIBM 2014) and the editorial report of the supplement to BMC Genomics and BMC Systems Biology that includes 20 research articles selected from ICIBM 2014. The conference was held on December 4-6, 2014 at San Antonio, Texas, USA, and included six scientific sessions, four tutorials, four keynote presentations, nine highlight talks, and a poster session that covered cutting-edge research in bioinformatics, systems biology, and computational medicine.
Collapse
Affiliation(s)
- Jianhua Ruan
- Department of Computer Science, The University of Texas at San Antonio, 78249 San Antonio, TX, USA
| | - Victor Jin
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, 78229 San Antonio, TX, USA
| | - Yufei Huang
- Department of Electrical and Computer Engineering, The University of Texas at San Antonio, 78249 San Antonio, TX, USA
| | - Hua Xu
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 77030 San Antonio, TX, USA
| | - Jeremy S Edwards
- Department of Molecular Genetics and Microbiology, University of New Mexico, 87131 Albuquerque, NM, USA
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, 78229 San Antonio, TX, USA
- Department of Epidemiology & Biostatistics, The University of Texas Health Science Center at San Antonio, 78229 San Antonio, TX, USA
| | - Zhongming Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, 37203 Nashville, TN, USA
- Department of Cancer Biology, Vanderbilt University School of Medicine, 37232 Nashville, TN, USA
| |
Collapse
|