1
|
Lazar V, Raymond E, Magidi S, Bresson C, Wunder F, Berindan-Neagoe I, Tijeras-Rabaland A, Raynaud J, Onn A, Ducreux M, Batist G, Lassen U, Cilius Nielsen F, Schilsky RL, Rubin E, Kurzrock R. Identification of a central network hub of key prognostic genes based on correlation between transcriptomics and survival in patients with metastatic solid tumors. Ther Adv Med Oncol 2024; 16:17588359241289200. [PMID: 39429467 PMCID: PMC11487509 DOI: 10.1177/17588359241289200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
Background Dysregulated pathways in cancer may be hub addicted. Identifying these dysregulated networks for targeting might lead to novel therapeutic options. Objective Considering the hypothesis that central hubs are associated with increased lethality, identifying key hub targets within central networks could lead to the development of novel drugs with improved efficacy in advanced metastatic solid tumors. Design Exploring transcriptomic data (22,000 gene products) from the WINTHER trial (N = 101 patients with various metastatic cancers), in which both tumor and normal organ-matched tissue were available. Methods A retrospective in silico analysis of all genes in the transcriptome was conducted to identify genes different in expression between tumor and normal tissues (paired t-test) and to determine their association with survival outcomes using survival analysis (Cox proportional hazard regression algorithm). Based on the biological relevance of the identified genes, hub targets of interest within central networks were then pinpointed. Patients were grouped based on the expression level of these genes (K-mean clustering), and the association of these groups with survival was examined (Cox proportional hazard regression algorithm, Forest plot, and Kaplan-Meier plot). Results We identified four key central hub genes-PLOD3, ARHGAP11A, RNF216, and CDCA8, for which high expression in tumor tissue compared to analogous normal tissue had the most significant correlation with worse outcomes. The correlation was independent of tumor or treatment type. The combination of the four genes showed the highest significance and correlation with the poorer outcome: overall survival (hazard ratio (95% confidence interval (CI)) = 10.5 (3.43-31.9) p = 9.12E-07 log-rank test in a Cox proportional hazard regression model). Findings were validated in independent cohorts. Conclusion The expression of PLOD3, ARHGAP11A, RNF216, and CDCA8 constitute, when combined, a prognostic tool, agnostic of tumor type and previous treatments. These genes represent potential targets for intercepting central hub networks in various cancers, offering avenues for novel therapeutic interventions.
Collapse
Affiliation(s)
- Vladimir Lazar
- Worldwide Innovative Network Association—WIN Consortium, Villejuif, France
| | - Eric Raymond
- Groupe Hospitalier Saint Joseph, Oncology Department Paris, France
| | - Shai Magidi
- Worldwide Innovative Network Association—WIN Consortium, 24, rue Albert Thuret, Chevilly-Larue 94850, France
| | - Catherine Bresson
- Worldwide Innovative Network Association—WIN Consortium, Villejuif, France
| | - Fanny Wunder
- Worldwide Innovative Network Association—WIN Consortium, Villejuif, France
| | - Ioana Berindan-Neagoe
- The Oncology Institute “Prof. Dr. Ion Chiricuta,” Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | | - Jacques Raynaud
- Worldwide Innovative Network Association—WIN Consortium, Villejuif, France
| | - Amir Onn
- Sheba Medical Center, Institute of Pulmonology, Tel HaShomer, Ramat-Gan, Israel
| | - Michel Ducreux
- Gustave Roussy, Department of Medical Oncology, Villejuif, France
- University Paris-Saclay, Department of Medical Oncology, Orsay, France
| | - Gerald Batist
- Segal Cancer Centre, Department of Oncology, Jewish General Hospital, McGill University, Montréal, QC, Canada
| | | | | | | | - Eitan Rubin
- Ben-Gurion University of the Negev, The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Be’er-Sheva, Israel
| | | |
Collapse
|
2
|
Wang S, Lee D. Community cohesion looseness in gene networks reveals individualized drug targets and resistance. Brief Bioinform 2024; 25:bbae175. [PMID: 38622359 PMCID: PMC11018546 DOI: 10.1093/bib/bbae175] [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: 02/02/2024] [Revised: 03/19/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
Community cohesion plays a critical role in the determination of an individual's health in social science. Intriguingly, a community structure of gene networks indicates that the concept of community cohesion could be applied between the genes as well to overcome the limitations of single gene-based biomarkers for precision oncology. Here, we develop community cohesion scores which precisely quantify the community ability to retain the interactions between the genes and their cellular functions in each individualized gene network. Using breast cancer as a proof-of-concept study, we measure the community cohesion score profiles of 950 case samples and predict the individualized therapeutic targets in 2-fold. First, we prioritize them by finding druggable genes present in the community with the most and relatively decreased scores in each individual. Then, we pinpoint more individualized therapeutic targets by discovering the genes which greatly contribute to the community cohesion looseness in each individualized gene network. Compared with the previous approaches, the community cohesion scores show at least four times higher performance in predicting effective individualized chemotherapy targets based on drug sensitivity data. Furthermore, the community cohesion scores successfully discover the known breast cancer subtypes and we suggest new targeted therapy targets for triple negative breast cancer (e.g. KIT and GABRP). Lastly, we demonstrate that the community cohesion scores can predict tamoxifen responses in ER+ breast cancer and suggest potential combination therapies (e.g. NAMPT and RXRA inhibitors) to reduce endocrine therapy resistance based on individualized characteristics. Our method opens new perspectives for the biomarker development in precision oncology.
Collapse
Affiliation(s)
- Seunghyun Wang
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Doheon Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
3
|
Verma G, Rebholz-Schuhmann D, Madden MG. Enabling personalised disease diagnosis by combining a patient's time-specific gene expression profile with a biomedical knowledge base. BMC Bioinformatics 2024; 25:62. [PMID: 38326757 PMCID: PMC10848462 DOI: 10.1186/s12859-024-05674-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 01/25/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Recent developments in the domain of biomedical knowledge bases (KBs) open up new ways to exploit biomedical knowledge that is available in the form of KBs. Significant work has been done in the direction of biomedical KB creation and KB completion, specifically, those having gene-disease associations and other related entities. However, the use of such biomedical KBs in combination with patients' temporal clinical data still largely remains unexplored, but has the potential to immensely benefit medical diagnostic decision support systems. RESULTS We propose two new algorithms, LOADDx and SCADDx, to combine a patient's gene expression data with gene-disease association and other related information available in the form of a KB, to assist personalized disease diagnosis. We have tested both of the algorithms on two KBs and on four real-world gene expression datasets of respiratory viral infection caused by Influenza-like viruses of 19 subtypes. We also compare the performance of proposed algorithms with that of five existing state-of-the-art machine learning algorithms (k-NN, Random Forest, XGBoost, Linear SVM, and SVM with RBF Kernel) using two validation approaches: LOOCV and a single internal validation set. Both SCADDx and LOADDx outperform the existing algorithms when evaluated with both validation approaches. SCADDx is able to detect infections with up to 100% accuracy in the cases of Datasets 2 and 3. Overall, SCADDx and LOADDx are able to detect an infection within 72 h of infection with 91.38% and 92.66% average accuracy respectively considering all four datasets, whereas XGBoost, which performed best among the existing machine learning algorithms, can detect the infection with only 86.43% accuracy on an average. CONCLUSIONS We demonstrate how our novel idea of using the most and least differentially expressed genes in combination with a KB can enable identification of the diseases that a patient is most likely to have at a particular time, from a KB with thousands of diseases. Moreover, the proposed algorithms can provide a short ranked list of the most likely diseases for each patient along with their most affected genes, and other entities linked with them in the KB, which can support health care professionals in their decision-making.
Collapse
Affiliation(s)
- Ghanshyam Verma
- Insight Centre for Data Analytics, School of Computer Science, University of Galway, Galway, Ireland.
- School of Computer Science, University of Galway, Galway, Ireland.
| | | | - Michael G Madden
- Insight Centre for Data Analytics, School of Computer Science, University of Galway, Galway, Ireland
- School of Computer Science, University of Galway, Galway, Ireland
| |
Collapse
|
4
|
Nithya C, Kiran M, Nagarajaram HA. Hubs and Bottlenecks in Protein-Protein Interaction Networks. Methods Mol Biol 2024; 2719:227-248. [PMID: 37803121 DOI: 10.1007/978-1-0716-3461-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2023]
Abstract
Protein-protein interaction networks (PPINs) represent the physical interactions among proteins in a cell. These interactions are critical in all cellular processes, including signal transduction, metabolic regulation, and gene expression. In PPINs, centrality measures are widely used to identify the most critical nodes. The two most commonly used centrality measures in networks are degree and betweenness centralities. Degree centrality is the number of connections a node has in the network, and betweenness centrality is the measure of the extent to which a node lies on the shortest paths between pairs of other nodes in the network. In PPINs, proteins with high degree and betweenness centrality are referred to as hubs and bottlenecks respectively. Hubs and bottlenecks are topologically and functionally essential proteins that play crucial roles in maintaining the network's structure and function. This article comprehensively reviews essential literature on hubs and bottlenecks, including their properties and functions.
Collapse
Affiliation(s)
- Chandramohan Nithya
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Manjari Kiran
- Department of Systems and Computational Biology, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | | |
Collapse
|
5
|
Fernando PC, Mabee PM, Zeng E. Protein-protein interaction network module changes associated with the vertebrate fin-to-limb transition. Sci Rep 2023; 13:22594. [PMID: 38114646 PMCID: PMC10730527 DOI: 10.1038/s41598-023-50050-2] [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: 06/01/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023] Open
Abstract
Evolutionary phenotypic transitions, such as the fin-to-limb transition in vertebrates, result from modifications in related proteins and their interactions, often in response to changing environment. Identifying these alterations in protein networks is crucial for a more comprehensive understanding of these transitions. However, previous research has not attempted to compare protein-protein interaction (PPI) networks associated with evolutionary transitions, and most experimental studies concentrate on a limited set of proteins. Therefore, the goal of this work was to develop a network-based platform for investigating the fin-to-limb transition using PPI networks. Quality-enhanced protein networks, constructed by integrating PPI networks with anatomy ontology data, were leveraged to compare protein modules for paired fins (pectoral fin and pelvic fin) of fishes (zebrafish) to those of the paired limbs (forelimb and hindlimb) of mammals (mouse). This also included prediction of novel protein candidates and their validation by enrichment and homology analyses. Hub proteins such as shh and bmp4, which are crucial for module stability, were identified, and their changing roles throughout the transition were examined. Proteins with preserved roles during the fin-to-limb transition were more likely to be hub proteins. This study also addressed hypotheses regarding the role of non-preserved proteins associated with the transition.
Collapse
Affiliation(s)
- Pasan C Fernando
- Department of Plant Sciences, University of Colombo, Colombo, Sri Lanka.
| | - Paula M Mabee
- Department of Biology, University of South Dakota, Vermillion, SD, USA
- National Ecological Observatory Network, Battelle, 1625 38th St. #100, Boulder, CO, 80301, USA
| | - Erliang Zeng
- Departments of Preventive & Community Dentistry, College of Dentistry, University of Iowa, Iowa City, IA, USA.
- Division of Biostatistics and Computational Biology, College of Dentistry, University of Iowa, Iowa City, IA, USA.
- Departments of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA.
- Departments of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
6
|
Liu X, Yang B, Huang X, Yan W, Zhang Y, Hu G. Identifying Lymph Node Metastasis-Related Factors in Breast Cancer Using Differential Modular and Mutational Structural Analysis. Interdiscip Sci 2023; 15:525-541. [PMID: 37115388 DOI: 10.1007/s12539-023-00568-w] [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: 01/25/2023] [Revised: 04/13/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023]
Abstract
Complex diseases are generally caused by disorders of biological networks and/or mutations in multiple genes. Comparisons of network topologies between different disease states can highlight key factors in their dynamic processes. Here, we propose a differential modular analysis approach that integrates protein-protein interactions with gene expression profiles for modular analysis, and introduces inter-modular edges and date hubs to identify the "core network module" that quantifies the significant phenotypic variation. Then, based on this core network module, key factors, including functional protein-protein interactions, pathways, and driver mutations, are predicted by the topological-functional connection score and structural modeling. We applied this approach to analyze the lymph node metastasis (LNM) process in breast cancer. The functional enrichment analysis showed that both inter-modular edges and date hubs play important roles in cancer metastasis and invasion, and in metastasis hallmarks. The structural mutation analysis suggested that the LNM of breast cancer may be the outcome of the dysfunction of rearranged during transfection (RET) proto-oncogene-related interactions and the non-canonical calcium signaling pathway via an allosteric mutation of RET. We believe that the proposed method can provide new insights into disease progression such as cancer metastasis.
Collapse
Affiliation(s)
- Xingyi Liu
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Bin Yang
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Xinpeng Huang
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Wenying Yan
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China.
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Suzhou, 215123, Jiangsu, China.
| | - Yujuan Zhang
- Experimental Center of Suzhou Medical College, Soochow University, Suzhou, 215123, Jiangsu, China.
| | - Guang Hu
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China.
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
7
|
Gaiteri C, Connell DR, Sultan FA, Iatrou A, Ng B, Szymanski BK, Zhang A, Tasaki S. Robust, scalable, and informative clustering for diverse biological networks. Genome Biol 2023; 24:228. [PMID: 37828545 PMCID: PMC10571258 DOI: 10.1186/s13059-023-03062-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/19/2023] [Indexed: 10/14/2023] Open
Abstract
Clustering molecular data into informative groups is a primary step in extracting robust conclusions from big data. However, due to foundational issues in how they are defined and detected, such clusters are not always reliable, leading to unstable conclusions. We compare popular clustering algorithms across thousands of synthetic and real biological datasets, including a new consensus clustering algorithm-SpeakEasy2: Champagne. These tests identify trends in performance, show no single method is universally optimal, and allow us to examine factors behind variation in performance. Multiple metrics indicate SpeakEasy2 generally provides robust, scalable, and informative clusters for a range of applications.
Collapse
Affiliation(s)
- Chris Gaiteri
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA.
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA.
| | - David R Connell
- Rush University Graduate College, Rush University Medical Center, Chicago, IL, USA
| | - Faraz A Sultan
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Artemis Iatrou
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Harvard University, Belmont, MA, USA
| | - Bernard Ng
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Boleslaw K Szymanski
- Department of Computer Science, Rensselaer Polytechnic Institute, Troy, NY, USA
- Network Science and Technology Center, Rensselaer Polytechnic Institute, Troy, NY, USA
- Academy of Social Sciences, Łódź, Poland
| | - Ada Zhang
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
8
|
Gao M, Zhao L, Zhang Z, Wang J, Wang C. Using a stacked ensemble learning framework to predict modulators of protein-protein interactions. Comput Biol Med 2023; 161:107032. [PMID: 37230018 DOI: 10.1016/j.compbiomed.2023.107032] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/13/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023]
Abstract
Identifying small molecule protein-protein interaction modulators (PPIMs) is a highly promising and meaningful research direction for drug discovery, cancer treatment, and other fields. In this study, we developed a stacking ensemble computational framework, SELPPI, based on a genetic algorithm and tree-based machine learning method for effectively predicting new modulators targeting protein-protein interactions. More specifically, extremely randomized trees (ExtraTrees), adaptive boosting (AdaBoost), random forest (RF), cascade forest, light gradient boosting machine (LightGBM), and extreme gradient boosting (XGBoost) were used as basic learners. Seven types of chemical descriptors were taken as the input characteristic parameters. Primary predictions were obtained with each basic learner-descriptor pair. Then, the 6 methods mentioned above were used as meta learners and trained on the primary prediction in turn. The most efficient method was utilized as the meta learner. Finally, the genetic algorithm was used to select the optimal primary prediction output as the input of the meta learner for secondary prediction to obtain the final result. We systematically evaluated our model on the pdCSM-PPI datasets. To our knowledge, our model outperformed all existing models, which demonstrates its great power.
Collapse
Affiliation(s)
- Mengyao Gao
- Faculty of Computing, Harbin Institute of Technology, Harbin, China.
| | - Lingling Zhao
- Faculty of Computing, Harbin Institute of Technology, Harbin, China.
| | - Zitong Zhang
- Faculty of Computing, Harbin Institute of Technology, Harbin, China.
| | - Junjie Wang
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China.
| | - Chunyu Wang
- Faculty of Computing, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
9
|
Nithya C, Kiran M, Nagarajaram HA. Dissection of hubs and bottlenecks in a protein-protein interaction network. Comput Biol Chem 2023; 102:107802. [PMID: 36603332 DOI: 10.1016/j.compbiolchem.2022.107802] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/20/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022]
Abstract
Analysis of degree centrality in conjunction with betweenness centrality of proteins in a human protein-protein interaction network revealed three categories of centrally important proteins: a) proteins with high degree and betweenness (hub-bottlenecks denoted as MX), b) proteins with high betweenness and low degree (non-hub-bottlenecks/pure bottlenecks denoted as PB) and c) proteins with high degree and low betweenness (hub-non-bottlenecks/pure hubs denoted as PH). When subjected to a detailed statistical analysis of their molecular-level properties, the proteins belonging to each of these categories were found to be associated with distinct canonical molecular properties, i.e., "molecular markers". The MX proteins are a) conformationally versatile, mainly comprising of essential proteins, b) the targets for interactions by the proteins of viral and bacterial pathogens, c) evolutionally constrained, involved in multiple pathways, enriched with disease genes and d) involved in the functions such as protein stabilization, phosphorylation, and mRNA slicing processes. PB proteins are a) enriched with extracellular and cancer-related proteins, b) enriched with the approved drug targets and c) involved in cell-cell signaling processes. Finally, PH are a) structurally versatile, b) enriched with essential proteins primarily involved in housekeeping processes (transcription and replication). The fact that the proteins belonging to these three categories form three distinct sets in terms of their molecular properties reveals the existence of trichotomy among hubs and bottlenecks, and this knowledge is of paramount importance while prioritizing protein targets for further studies such as drug design and disease association studies based on their network centrality values.
Collapse
Affiliation(s)
- Chandramohan Nithya
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana 500046, India
| | - Manjari Kiran
- Department of Systems and Computational Biology, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana 500046, India
| | | |
Collapse
|
10
|
Discovering Entities Similarities in Biological Networks Using a Hybrid Immune Algorithm. INFORMATICS 2023. [DOI: 10.3390/informatics10010018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Disease phenotypes are generally caused by the failure of gene modules which often have similar biological roles. Through the study of biological networks, it is possible to identify the intrinsic structure of molecular interactions in order to identify the so-called “disease modules”. Community detection is an interesting and valuable approach to discovering the structure of the community in a complex network, revealing the internal organization of the nodes, and has become a leading research topic in the analysis of complex networks. This work investigates the link between biological modules and network communities in test-case biological networks that are commonly used as a reference point and which include Protein–Protein Interaction Networks, Metabolic Networks and Transcriptional Regulation Networks. In order to identify small and structurally well-defined communities in the biological context, a hybrid immune metaheuristic algorithm Hybrid-IA is proposed and compared with several metaheuristics, hyper-heuristics, and the well-known greedy algorithm Louvain, with respect to modularity maximization. Considering the limitation of modularity optimization, which can fail to identify smaller communities, the reliability of Hybrid-IA was also analyzed with respect to three well-known sensitivity analysis measures (NMI, ARI and NVI) that assess how similar the detected communities are to real ones. By inspecting all outcomes and the performed comparisons, we will see that on one hand Hybrid-IA finds slightly lower modularity values than Louvain, but outperforms all other metaheuristics, while on the other hand, it can detect communities more similar to the real ones when compared to those detected by Louvain.
Collapse
|
11
|
Petrosius V, Schoof EM. Recent advances in the field of single-cell proteomics. Transl Oncol 2023; 27:101556. [PMID: 36270102 PMCID: PMC9587008 DOI: 10.1016/j.tranon.2022.101556] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/06/2022] Open
Abstract
The field of single-cell omics is rapidly progressing. Although DNA and RNA sequencing-based methods have dominated the field to date, global proteome profiling has also entered the main stage. Single-cell proteomics was facilitated by advancements in different aspects of mass spectrometry (MS)-based proteomics, such as instrument design, sample preparation, chromatography and ion mobility. Single-cell proteomics by mass spectrometry (scp-MS) has moved beyond being a mere technical development, and is now able to deliver actual biological application and has been successfully applied to characterize different cell states. Here, we review some key developments of scp-MS, provide a background to the field, discuss the various available methods and foresee possible future directions.
Collapse
Affiliation(s)
- Valdemaras Petrosius
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Erwin M Schoof
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark.
| |
Collapse
|
12
|
Kato M, Kori H. Partial synchronization and community switching in phase-oscillator networks and its analysis based on a bidirectional, weighted chain of three oscillators. Phys Rev E 2023; 107:014210. [PMID: 36797893 DOI: 10.1103/physreve.107.014210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/22/2022] [Indexed: 01/22/2023]
Abstract
Complex networks often possess communities defined based on network connectivity. When dynamics undergo in a network, one can also consider dynamical communities, i.e., a group of nodes displaying a similar dynamical process. We have investigated both analytically and numerically the development of a dynamical community structure, where the community is referred to as a group of nodes synchronized in frequency, in networks of phase oscillators. We first demonstrate that using a few example networks, the community structure changes when network connectivity or interaction strength is varied. In particular, we found that community switching, i.e., a portion of oscillators change the group to which they synchronize, occurs for a range of parameters. We then propose a three-oscillator model: a bidirectional, weighted chain of three Kuramoto phase oscillators, as a theoretical framework for understanding the community formation and its variation. Our analysis demonstrates that the model shows a variety of partially synchronized patterns: oscillators with similar natural frequencies tend to synchronize for weak coupling, while tightly connected oscillators tend to synchronize for strong coupling. We obtain approximate expressions for the critical coupling strengths by employing a perturbative approach in a weak coupling regime and a geometric approach in strong coupling regimes. Moreover, we elucidate the bifurcation types of transitions between different patterns. Our theory might be useful for understanding the development of partially synchronized patterns in a wider class of complex networks than community structured networks.
Collapse
Affiliation(s)
- Masaki Kato
- Department of Mathematical Informatics, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Kori
- Department of Mathematical Informatics, The University of Tokyo, Tokyo, Japan and Department of Complexity Sciences and Engineering, The University of Tokyo, Kashiwa, Chiba, Japan
| |
Collapse
|
13
|
Ye Q, Guo NL. Inferencing Bulk Tumor and Single-Cell Multi-Omics Regulatory Networks for Discovery of Biomarkers and Therapeutic Targets. Cells 2022; 12:101. [PMID: 36611894 PMCID: PMC9818242 DOI: 10.3390/cells12010101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 12/28/2022] Open
Abstract
There are insufficient accurate biomarkers and effective therapeutic targets in current cancer treatment. Multi-omics regulatory networks in patient bulk tumors and single cells can shed light on molecular disease mechanisms. Integration of multi-omics data with large-scale patient electronic medical records (EMRs) can lead to the discovery of biomarkers and therapeutic targets. In this review, multi-omics data harmonization methods were introduced, and common approaches to molecular network inference were summarized. Our Prediction Logic Boolean Implication Networks (PLBINs) have advantages over other methods in constructing genome-scale multi-omics networks in bulk tumors and single cells in terms of computational efficiency, scalability, and accuracy. Based on the constructed multi-modal regulatory networks, graph theory network centrality metrics can be used in the prioritization of candidates for discovering biomarkers and therapeutic targets. Our approach to integrating multi-omics profiles in a patient cohort with large-scale patient EMRs such as the SEER-Medicare cancer registry combined with extensive external validation can identify potential biomarkers applicable in large patient populations. These methodologies form a conceptually innovative framework to analyze various available information from research laboratories and healthcare systems, accelerating the discovery of biomarkers and therapeutic targets to ultimately improve cancer patient survival outcomes.
Collapse
Affiliation(s)
- Qing Ye
- West Virginia University Cancer Institute, Morgantown, WV 26506, USA
- Lane Department of Computer Science and Electrical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Nancy Lan Guo
- West Virginia University Cancer Institute, Morgantown, WV 26506, USA
- Department of Occupational and Environmental Health Sciences, School of Public Health, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
14
|
Millar-Wilson A, Ward Ó, Duffy E, Hardiman G. Multiscale modeling in the framework of biological systems and its potential for spaceflight biology studies. iScience 2022; 25:105421. [DOI: 10.1016/j.isci.2022.105421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
15
|
Kong Q, Ke M, Weng Y, Qin Y, He A, Li P, Cai Z, Tian R. Dynamic Phosphotyrosine-Dependent Signaling Profiling in Living Cells by Two-Dimensional Proximity Proteomics. J Proteome Res 2022; 21:2727-2735. [DOI: 10.1021/acs.jproteome.2c00418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Qian Kong
- Department of Chemistry, College of Science, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
- Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong SAR, China
| | - Mi Ke
- Department of Chemistry, College of Science, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
- Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
| | - Yicheng Weng
- Department of Chemistry, College of Science, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
- Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
| | - Yunqiu Qin
- Department of Chemistry, College of Science, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
- Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
| | - An He
- Department of Chemistry, College of Science, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
- Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
| | - Pengfei Li
- Department of Chemistry, College of Science, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
- Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
- Shenzhen Grubbs Institute, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong SAR, China
| | - Ruijun Tian
- Department of Chemistry, College of Science, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
- Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
- Shenzhen Grubbs Institute, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
| |
Collapse
|
16
|
Yue R, Dutta A. Computational systems biology in disease modeling and control, review and perspectives. NPJ Syst Biol Appl 2022; 8:37. [PMID: 36192551 PMCID: PMC9528884 DOI: 10.1038/s41540-022-00247-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 09/05/2022] [Indexed: 02/02/2023] Open
Abstract
Omics-based approaches have become increasingly influential in identifying disease mechanisms and drug responses. Considering that diseases and drug responses are co-expressed and regulated in the relevant omics data interactions, the traditional way of grabbing omics data from single isolated layers cannot always obtain valuable inference. Also, drugs have adverse effects that may impair patients, and launching new medicines for diseases is costly. To resolve the above difficulties, systems biology is applied to predict potential molecular interactions by integrating omics data from genomic, proteomic, transcriptional, and metabolic layers. Combined with known drug reactions, the resulting models improve medicines' therapeutical performance by re-purposing the existing drugs and combining drug molecules without off-target effects. Based on the identified computational models, drug administration control laws are designed to balance toxicity and efficacy. This review introduces biomedical applications and analyses of interactions among gene, protein and drug molecules for modeling disease mechanisms and drug responses. The therapeutical performance can be improved by combining the predictive and computational models with drug administration designed by control laws. The challenges are also discussed for its clinical uses in this work.
Collapse
Affiliation(s)
- Rongting Yue
- Department of Electrical and Computer Engineering, University of Connecticut, 371 Fairfield Way, Storrs, CT, 06269, USA.
| | - Abhishek Dutta
- Department of Electrical and Computer Engineering, University of Connecticut, 371 Fairfield Way, Storrs, CT, 06269, USA
| |
Collapse
|
17
|
Large-scale prediction of key dynamic interacting proteins in multiple cancers. Int J Biol Macromol 2022; 220:1124-1132. [PMID: 36027989 DOI: 10.1016/j.ijbiomac.2022.08.125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 11/21/2022]
Abstract
Tracking cancer dynamic protein-protein interactions (PPIs) and deciphering their pathogenesis remain a challenge. We presented a dynamic PPIs' hypothesis: permanent and transient interactions might achieve dynamic switchings from normal cells to malignancy, which could cause maintenance functions to be interrupted and transient functions to be sustained. Based on the hypothesis, we first predicted >1400 key cancer genes (KCG) by applying PPI-express we proposed to 18 cancer gene expression datasets. We then further screened out key dynamic interactions (KDI) of cancer based on KCG and transient and permanent interactions under both conditions. Two prominent functional characteristics, "Cell cycle-related" and "Immune-related", were presented for KCG, suggesting that these might be their general characteristics. We found that, compared to permanent to transient KDI pairs (P2T) in the network, transient to permanent (T2P) have significantly higher edge betweenness (EB), and P2T pairs tending to locate intra-functional modules may play roles in maintaining normal biological functions, while T2P KDI pairs tending to locate inter-modules may play roles in biological signal transduction. It was consistent with our hypothesis. Also, we analyzed network characteristics of KDI pairs and their functions. Our findings of KDI may serve to understand and explain a few hallmarks of cancer.
Collapse
|
18
|
Artime O, De Domenico M. From the origin of life to pandemics: emergent phenomena in complex systems. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2022; 380:20200410. [PMID: 35599559 PMCID: PMC9125231 DOI: 10.1098/rsta.2020.0410] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 05/31/2023]
Abstract
When a large number of similar entities interact among each other and with their environment at a low scale, unexpected outcomes at higher spatio-temporal scales might spontaneously arise. This non-trivial phenomenon, known as emergence, characterizes a broad range of distinct complex systems-from physical to biological and social-and is often related to collective behaviour. It is ubiquitous, from non-living entities such as oscillators that under specific conditions synchronize, to living ones, such as birds flocking or fish schooling. Despite the ample phenomenological evidence of the existence of systems' emergent properties, central theoretical questions to the study of emergence remain unanswered, such as the lack of a widely accepted, rigorous definition of the phenomenon or the identification of the essential physical conditions that favour emergence. We offer here a general overview of the phenomenon of emergence and sketch current and future challenges on the topic. Our short review also serves as an introduction to the theme issue Emergent phenomena in complex physical and socio-technical systems: from cells to societies, where we provide a synthesis of the contents tackled in the issue and outline how they relate to these challenges, spanning from current advances in our understanding on the origin of life to the large-scale propagation of infectious diseases. This article is part of the theme issue 'Emergent phenomena in complex physical and socio-technical systems: from cells to societies'.
Collapse
Affiliation(s)
- Oriol Artime
- Fondazione Bruno Kessler, Via Sommarive 18, Povo, TN 38123, Italy
| | - Manlio De Domenico
- Department of Physics and Astronomy ‘Galileo Galilei’, University of Padua, Padova, Veneto, Italy
| |
Collapse
|
19
|
Ma L, Shao Z, Li L, Huang J, Wang S, Lin Q, Li J, Gong M, Nandi AK. Heuristics and metaheuristics for biological network alignment: A review. Neurocomputing 2022. [DOI: 10.1016/j.neucom.2021.08.156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
You Y, Lai X, Pan Y, Zheng H, Vera J, Liu S, Deng S, Zhang L. Artificial intelligence in cancer target identification and drug discovery. Signal Transduct Target Ther 2022; 7:156. [PMID: 35538061 PMCID: PMC9090746 DOI: 10.1038/s41392-022-00994-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 03/14/2022] [Accepted: 04/05/2022] [Indexed: 02/08/2023] Open
Abstract
Artificial intelligence is an advanced method to identify novel anticancer targets and discover novel drugs from biology networks because the networks can effectively preserve and quantify the interaction between components of cell systems underlying human diseases such as cancer. Here, we review and discuss how to employ artificial intelligence approaches to identify novel anticancer targets and discover drugs. First, we describe the scope of artificial intelligence biology analysis for novel anticancer target investigations. Second, we review and discuss the basic principles and theory of commonly used network-based and machine learning-based artificial intelligence algorithms. Finally, we showcase the applications of artificial intelligence approaches in cancer target identification and drug discovery. Taken together, the artificial intelligence models have provided us with a quantitative framework to study the relationship between network characteristics and cancer, thereby leading to the identification of potential anticancer targets and the discovery of novel drug candidates.
Collapse
Affiliation(s)
- Yujie You
- College of Computer Science, Sichuan University, Chengdu, 610065, China
| | - Xin Lai
- Laboratory of Systems Tumor Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91052, Germany
| | - Yi Pan
- Faculty of Computer Science and Control Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Room D513, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, China
| | - Huiru Zheng
- School of Computing, Ulster University, Belfast, BT15 1ED, UK
| | - Julio Vera
- Laboratory of Systems Tumor Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91052, Germany
| | - Suran Liu
- College of Computer Science, Sichuan University, Chengdu, 610065, China
| | - Senyi Deng
- Institute of Thoracic Oncology, Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610065, China.
| | - Le Zhang
- College of Computer Science, Sichuan University, Chengdu, 610065, China.
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, 310024, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
21
|
Wolf C, Maus C, Persicke MRO, Filarsky K, Tausch E, Schneider C, Döhner H, Stilgenbauer S, Lichter P, Höfer T, Mertens D. Modeling the B‐cell receptor signaling on single cell level reveals a stable network circuit topology between non‐malignant B cells and chronic lymphocytic leukemia cells and between untreated cells and cells treated with kinase inhibitors. Int J Cancer 2022; 151:783-796. [DOI: 10.1002/ijc.34112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/09/2022] [Accepted: 03/16/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Christine Wolf
- Mechanisms of Leukemogenesis, German Cancer Research Center (DKFZ) Heidelberg Germany
| | - Carsten Maus
- Division of Theoretical Systems Biology German Cancer Research Center (DXDKFZ) Heidelberg Germany
- Bioquant Heidelberg University Heidelberg Germany
| | - Michael RO Persicke
- Mechanisms of Leukemogenesis, German Cancer Research Center (DKFZ) Heidelberg Germany
- Department of Internal Medicine III University Hospital Ulm Ulm Germany
- Faculty of Biosciences Heidelberg University Heidelberg Germany
| | - Katharina Filarsky
- Mechanisms of Leukemogenesis, German Cancer Research Center (DKFZ) Heidelberg Germany
| | - Eugen Tausch
- Department of Internal Medicine III University Hospital Ulm Ulm Germany
| | | | - Hartmut Döhner
- Department of Internal Medicine III University Hospital Ulm Ulm Germany
| | | | - Peter Lichter
- Division of Molecular Genetics German Cancer Research Center (DKFZ) Heidelberg Germany
| | - Thomas Höfer
- Division of Theoretical Systems Biology German Cancer Research Center (DXDKFZ) Heidelberg Germany
- Bioquant Heidelberg University Heidelberg Germany
| | - Daniel Mertens
- Mechanisms of Leukemogenesis, German Cancer Research Center (DKFZ) Heidelberg Germany
- Department of Internal Medicine III University Hospital Ulm Ulm Germany
| |
Collapse
|
22
|
Ningappa M, Rahman SA, Higgs BW, Ashokkumar CS, Sahni N, Sindhi R, Das J. A network-based approach to identify expression modules underlying rejection in pediatric liver transplantation. Cell Rep Med 2022; 3:100605. [PMID: 35492246 PMCID: PMC9044102 DOI: 10.1016/j.xcrm.2022.100605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 12/19/2021] [Accepted: 03/23/2022] [Indexed: 10/27/2022]
Abstract
Selecting the right immunosuppressant to ensure rejection-free outcomes poses unique challenges in pediatric liver transplant (LT) recipients. A molecular predictor can comprehensively address these challenges. Currently, there are no well-validated blood-based biomarkers for pediatric LT recipients before or after LT. Here, we discover and validate separate pre- and post-LT transcriptomic signatures of rejection. Using an integrative machine learning approach, we combine transcriptomics data with the reference high-quality human protein interactome to identify network module signatures, which underlie rejection. Unlike gene signatures, our approach is inherently multivariate and more robust to replication and captures the structure of the underlying network, encapsulating additive effects. We also identify, in an individual-specific manner, signatures that can be targeted by current anti-rejection drugs and other drugs that can be repurposed. Our approach can enable personalized adjustment of drug regimens for the dominant targetable pathways before and after LT in children.
Collapse
Affiliation(s)
- Mylarappa Ningappa
- Department of Surgery and Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Syed A Rahman
- Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brandon W Higgs
- Department of Surgery and Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chethan S Ashokkumar
- Department of Surgery and Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nidhi Sahni
- Department of Epigenetics, The University of Texas MD Anderson Cancer Center, Smithville, TX, USA.,Department of Molecular Carcinogenesis and Bioinformatics, The University of Texas MD Anderson Cancer Center, Smithville, TX, USA.,Department of Computational Biology, The University of Texas MD Anderson Cancer Center, Smithville, TX, USA
| | - Rakesh Sindhi
- Department of Surgery and Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jishnu Das
- Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
23
|
Majhi S, Rakshit S, Ghosh D. Oscillation suppression and chimera states in time-varying networks. CHAOS (WOODBURY, N.Y.) 2022; 32:042101. [PMID: 35489845 DOI: 10.1063/5.0087291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/11/2022] [Indexed: 06/14/2023]
Abstract
Complex network theory has offered a powerful platform for the study of several natural dynamic scenarios, based on the synergy between the interaction topology and the dynamics of its constituents. With research in network theory being developed so fast, it has become extremely necessary to move from simple network topologies to more sophisticated and realistic descriptions of the connectivity patterns. In this context, there is a significant amount of recent works that have emerged with enormous evidence establishing the time-varying nature of the connections among the constituents in a large number of physical, biological, and social systems. The recent review article by Ghosh et al. [Phys. Rep. 949, 1-63 (2022)] demonstrates the significance of the analysis of collective dynamics arising in temporal networks. Specifically, the authors put forward a detailed excerpt of results on the origin and stability of synchronization in time-varying networked systems. However, among the complex collective dynamical behaviors, the study of the phenomenon of oscillation suppression and that of other diverse aspects of synchronization are also considered to be central to our perception of the dynamical processes over networks. Through this review, we discuss the principal findings from the research studies dedicated to the exploration of the two collective states, namely, oscillation suppression and chimera on top of time-varying networks of both static and mobile nodes. We delineate how temporality in interactions can suppress oscillation and induce chimeric patterns in networked dynamical systems, from effective analytical approaches to computational aspects, which is described while addressing these two phenomena. We further sketch promising directions for future research on these emerging collective behaviors in time-varying networks.
Collapse
Affiliation(s)
- Soumen Majhi
- Department of Mathematics, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Sarbendu Rakshit
- Physics and Applied Mathematics Unit, Indian Statistical Institute, 203 B. T. Road, Kolkata 700108, India
| | - Dibakar Ghosh
- Physics and Applied Mathematics Unit, Indian Statistical Institute, 203 B. T. Road, Kolkata 700108, India
| |
Collapse
|
24
|
Systematic identification of candidate genes associated with aggressive behavior: A neurogenetic approach. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
25
|
Smith J, Arashi M, Bekker A. Empowering differential networks using Bayesian analysis. PLoS One 2022; 17:e0261193. [PMID: 35077451 PMCID: PMC8789149 DOI: 10.1371/journal.pone.0261193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/24/2021] [Indexed: 11/19/2022] Open
Abstract
Differential networks (DN) are important tools for modeling the changes in conditional dependencies between multiple samples. A Bayesian approach for estimating DNs, from the classical viewpoint, is introduced with a computationally efficient threshold selection for graphical model determination. The algorithm separately estimates the precision matrices of the DN using the Bayesian adaptive graphical lasso procedure. Synthetic experiments illustrate that the Bayesian DN performs exceptionally well in numerical accuracy and graphical structure determination in comparison to state of the art methods. The proposed method is applied to South African COVID-19 data to investigate the change in DN structure between various phases of the pandemic.
Collapse
Affiliation(s)
- Jarod Smith
- Department of Statistics, University of Pretoria, Pretoria, South Africa
| | - Mohammad Arashi
- Department of Statistics, University of Pretoria, Pretoria, South Africa
- Department of Statistics, Faculty of Mathematical Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Andriëtte Bekker
- Department of Statistics, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
26
|
Trahan C, Oeffinger M. Single-Step Affinity Purification (ssAP) and Mass Spectrometry of Macromolecular Complexes in the Yeast S. cerevisiae. Methods Mol Biol 2022; 2477:195-223. [PMID: 35524119 DOI: 10.1007/978-1-0716-2257-5_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cellular functions are mostly defined by the dynamic interactions of proteins within macromolecular networks. Deciphering the composition of macromolecular complexes and their dynamic rearrangements is the key to get a comprehensive picture of cellular behavior and to understand biological systems. In the past two decades, affinity purification coupled to mass spectrometry has become a powerful tool to comprehensively study interaction networks and their assemblies. To overcome initial limitations of the approach, in particular, the effect of protein and RNA degradation, loss of transient interactors, and poor overall yield of intact complexes from cell lysates, various modifications to affinity purification protocols have been devised over the years. In this chapter, we describe a rapid single-step affinity purification method for the efficient isolation of dynamic macromolecular complexes. The technique employs cell lysis by cryo-milling, which ensures nondegraded starting material in the submicron range, and magnetic beads, which allow for dense antibody-conjugation and thus rapid complex isolation, while avoiding loss of transient interactions. The method is epitope tag-independent, and overcomes many of the previous limitations to produce large interactomes with almost no contamination. The protocol as described here has been optimized for the yeast S. cerevisiae.
Collapse
Affiliation(s)
- Christian Trahan
- RNP Biochemistry Laboratory, Center for Genetic and Neurological Diseases, Institut de recherches cliniques de Montréal, Montréal, QC, Canada
| | - Marlene Oeffinger
- RNP Biochemistry Laboratory, Center for Genetic and Neurological Diseases, Institut de recherches cliniques de Montréal, Montréal, QC, Canada.
- Département de biochimie et médicine moléculaire, Faculté de médecine, Université de Montréal, Montréal, QC, Canada.
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montréal, QC, Canada.
| |
Collapse
|
27
|
Skaro M, Hill M, Zhou Y, Quinn S, Davis MB, Sboner A, Murph M, Arnold J. Are we there yet? A machine learning architecture to predict organotropic metastases. BMC Med Genomics 2021; 14:281. [PMID: 34819069 PMCID: PMC8611885 DOI: 10.1186/s12920-021-01122-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 11/10/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND & AIMS Cancer metastasis into distant organs is an evolutionarily selective process. A better understanding of the driving forces endowing proliferative plasticity of tumor seeds in distant soils is required to develop and adapt better treatment systems for this lethal stage of the disease. To this end, we aimed to utilize transcript expression profiling features to predict the site-specific metastases of primary tumors and second, to identify the determinants of tissue specific progression. METHODS We used statistical machine learning for transcript feature selection to optimize classification and built tree-based classifiers to predict tissue specific sites of metastatic progression. RESULTS We developed a novel machine learning architecture that analyzes 33 types of RNA transcriptome profiles from The Cancer Genome Atlas (TCGA) database. Our classifier identifies the tumor type, derives synthetic instances of primary tumors metastasizing to distant organs and classifies the site-specific metastases in 16 types of cancers metastasizing to 12 locations. CONCLUSIONS We have demonstrated that site specific metastatic progression is predictable using transcriptomic profiling data from primary tumors and that the overrepresented biological processes in tumors metastasizing to congruent distant loci are highly overlapping. These results indicate site-specific progression was organotropic and core features of biological signaling pathways are identifiable that may describe proliferative plasticity in distant soils.
Collapse
Affiliation(s)
- Michael Skaro
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA.
| | - Marcus Hill
- Department of Computer Science, University of Georgia, Athens, GA, 30602, USA
| | - Yi Zhou
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
| | - Shannon Quinn
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- Department of Computer Science, University of Georgia, Athens, GA, 30602, USA
- Department of Cellular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Melissa B Davis
- Caryl and Israel Englander Institute for Precision Medicine, New York Presbyterian Hospital-Weill Cornell Medicine, New York, NY, 10065, USA
| | - Andrea Sboner
- Caryl and Israel Englander Institute for Precision Medicine, New York Presbyterian Hospital-Weill Cornell Medicine, New York, NY, 10065, USA
- Weill Cornell Medicine, HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, New York, NY, 10021, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Mandi Murph
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, 30602, USA
| | - Jonathan Arnold
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
28
|
Ma JX, Yang Y, Li G, Ma BG. Computationally Reconstructed Interactome of Bradyrhizobium diazoefficiens USDA110 Reveals Novel Functional Modules and Protein Hubs for Symbiotic Nitrogen Fixation. Int J Mol Sci 2021; 22:11907. [PMID: 34769335 PMCID: PMC8584416 DOI: 10.3390/ijms222111907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/22/2021] [Accepted: 10/28/2021] [Indexed: 11/16/2022] Open
Abstract
Symbiotic nitrogen fixation is an important part of the nitrogen biogeochemical cycles and the main nitrogen source of the biosphere. As a classical model system for symbiotic nitrogen fixation, rhizobium-legume systems have been studied elaborately for decades. Details about the molecular mechanisms of the communication and coordination between rhizobia and host plants is becoming clearer. For more systematic insights, there is an increasing demand for new studies integrating multiomics information. Here, we present a comprehensive computational framework integrating the reconstructed protein interactome of B. diazoefficiens USDA110 with its transcriptome and proteome data to study the complex protein-protein interaction (PPI) network involved in the symbiosis system. We reconstructed the interactome of B. diazoefficiens USDA110 by computational approaches. Based on the comparison of interactomes between B. diazoefficiens USDA110 and other rhizobia, we inferred that the slow growth of B. diazoefficiens USDA110 may be due to the requirement of more protein modifications, and we further identified 36 conserved functional PPI modules. Integrated with transcriptome and proteome data, interactomes representing free-living cell and symbiotic nitrogen-fixing (SNF) bacteroid were obtained. Based on the SNF interactome, a core-sub-PPI-network for symbiotic nitrogen fixation was determined and nine novel functional modules and eleven key protein hubs playing key roles in symbiosis were identified. The reconstructed interactome of B. diazoefficiens USDA110 may serve as a valuable reference for studying the mechanism underlying the SNF system of rhizobia and legumes.
Collapse
Affiliation(s)
| | | | | | - Bin-Guang Ma
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (J.-X.M.); (Y.Y.); (G.L.)
| |
Collapse
|
29
|
Strub MD, Gao L, Tan K, McCray PB. Analysis of multiple gene co-expression networks to discover interactions favoring CFTR biogenesis and ΔF508-CFTR rescue. BMC Med Genomics 2021; 14:258. [PMID: 34717611 PMCID: PMC8557508 DOI: 10.1186/s12920-021-01106-7] [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: 04/08/2021] [Accepted: 10/20/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND We previously reported that expression of a miR-138 mimic or knockdown of SIN3A in primary cultures of cystic fibrosis (CF) airway epithelia increased ΔF508-CFTR mRNA and protein levels, and partially restored CFTR-dependent chloride transport. Global mRNA transcript profiling in ΔF508-CFBE cells treated with miR-138 mimic or SIN3A siRNA identified two genes, SYVN1 and NEDD8, whose inhibition significantly increased ΔF508-CFTR trafficking, maturation, and function. Little is known regarding the dynamic changes in the CFTR gene network during such rescue events. We hypothesized that analysis of condition-specific gene networks from transcriptomic data characterizing ΔF508-CFTR rescue could help identify dynamic gene modules associated with CFTR biogenesis. METHODS We applied a computational method, termed M-module, to analyze multiple gene networks, each of which exhibited differential activity compared to a baseline condition. In doing so, we identified both unique and shared gene pathways across multiple differential networks. To construct differential networks, gene expression data from CFBE cells were divided into three groups: (1) siRNA inhibition of NEDD8 and SYVN1; (2) miR-138 mimic and SIN3A siRNA; and (3) temperature (27 °C for 24 h, 40 °C for 24 h, and 27 °C for 24 h followed by 40 °C for 24 h). RESULTS Interrogation of individual networks (e.g., NEDD8/SYVN1 network), combinations of two networks (e.g., NEDD8/SYVN1 + temperature networks), and all three networks yielded sets of 1-modules, 2-modules, and 3-modules, respectively. Gene ontology analysis revealed significant enrichment of dynamic modules in pathways including translation, protein metabolic/catabolic processes, protein complex assembly, and endocytosis. Candidate CFTR effectors identified in the analysis included CHURC1, GZF1, and RPL15, and siRNA-mediated knockdown of these genes partially restored CFTR-dependent transepithelial chloride current to ΔF508-CFBE cells. CONCLUSIONS The ability of the M-module to identify dynamic modules involved in ΔF508 rescue provides a novel approach for studying CFTR biogenesis and identifying candidate suppressors of ΔF508.
Collapse
Affiliation(s)
- Matthew D Strub
- Department of Pediatrics, University of Iowa, 6320 PBDB, 169 Newton Road, Iowa City, IA, 52242, USA.,Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, 52245, USA
| | - Long Gao
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kai Tan
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Paul B McCray
- Department of Pediatrics, University of Iowa, 6320 PBDB, 169 Newton Road, Iowa City, IA, 52242, USA. .,Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, 52245, USA.
| |
Collapse
|
30
|
Kim M, Park J, Bouhaddou M, Kim K, Rojc A, Modak M, Soucheray M, McGregor MJ, O'Leary P, Wolf D, Stevenson E, Foo TK, Mitchell D, Herrington KA, Muñoz DP, Tutuncuoglu B, Chen KH, Zheng F, Kreisberg JF, Diolaiti ME, Gordan JD, Coppé JP, Swaney DL, Xia B, van 't Veer L, Ashworth A, Ideker T, Krogan NJ. A protein interaction landscape of breast cancer. Science 2021; 374:eabf3066. [PMID: 34591612 PMCID: PMC9040556 DOI: 10.1126/science.abf3066] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Minkyu Kim
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Jisoo Park
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA
| | - Mehdi Bouhaddou
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Kyumin Kim
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Ajda Rojc
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Maya Modak
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Margaret Soucheray
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Michael J McGregor
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Patrick O'Leary
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Denise Wolf
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Erica Stevenson
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Tzeh Keong Foo
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Dominique Mitchell
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.,Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Kari A Herrington
- Department of Biochemistry and Biophysics, Center for Advanced Light Microscopy, University of California, San Francisco, CA, USA
| | - Denise P Muñoz
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Beril Tutuncuoglu
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Kuei-Ho Chen
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Fan Zheng
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA
| | - Jason F Kreisberg
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA
| | - Morgan E Diolaiti
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - John D Gordan
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.,Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Jean-Philippe Coppé
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Danielle L Swaney
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Bing Xia
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Laura van 't Veer
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Alan Ashworth
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Trey Ideker
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA.,Department of Bioengineering, University of California, San Diego, CA, USA
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| |
Collapse
|
31
|
Dou Z, Ma X. Inferring Functional Epigenetic Modules by Integrative Analysis of Multiple Heterogeneous Networks. Front Genet 2021; 12:706952. [PMID: 34504516 PMCID: PMC8421682 DOI: 10.3389/fgene.2021.706952] [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: 05/08/2021] [Accepted: 06/29/2021] [Indexed: 02/02/2023] Open
Abstract
Gene expression and methylation are critical biological processes for cells, and how to integrate these heterogeneous data has been extensively investigated, which is the foundation for revealing the underlying patterns of cancers. The vast majority of the current algorithms fuse gene methylation and expression into a network, failing to fully explore the relations and heterogeneity of them. To resolve these problems, in this study we define the epigenetic modules as a gene set whose members are co-methylated and co-expressed. To address the heterogeneity of data, we construct gene co-expression and co-methylation networks, respectively. In this case, the epigenetic module is characterized as a common module in multiple networks. Then, a non-negative matrix factorization-based algorithm that jointly clusters the co-expression and co-methylation networks is proposed for discovering the epigenetic modules (called Ep-jNMF). Ep-jNMF is more accurate than the baselines on the artificial data. Moreover, Ep-jNMF identifies more biologically meaningful modules. And the modules can predict the subtypes of cancers. These results indicate that Ep-jNMF is efficient for the integration of expression and methylation data.
Collapse
Affiliation(s)
- Zengfa Dou
- The 20-th Research Institute, China Electronics Technology Group Corporation, Xi'an, China
| | - Xiaoke Ma
- School of Computer Science and Technology, Xidian University, Xi'an, China
| |
Collapse
|
32
|
Spatiotemporal 22q11.21 Protein Network Implicates DGCR8-Dependent MicroRNA Biogenesis as a Risk for Late-Fetal Cortical Development in Psychiatric Diseases. Life (Basel) 2021; 11:life11060514. [PMID: 34073122 PMCID: PMC8227527 DOI: 10.3390/life11060514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 12/28/2022] Open
Abstract
The chromosome 22q11.21 copy number variant (CNV) is a vital risk factor that can be a genetic predisposition to neurodevelopmental disorders (NDD). As the 22q11.21 CNV affects multiple genes, causal disease genes and mechanisms affected are still poorly understood. Thus, we aimed to identify the most impactful 22q11.21 CNV genes and the potential impacted human brain regions, developmental stages and signaling pathways. We constructed the spatiotemporal dynamic networks of 22q11.21 CNV genes using the brain developmental transcriptome and physical protein–protein interactions. The affected brain regions, developmental stages, driver genes and pathways were subsequently investigated via integrated bioinformatics analysis. As a result, we first identified that 22q11.21 CNV genes affect the cortical area mainly during late fetal periods. Interestingly, we observed that connections between a driver gene, DGCR8, and its interacting partners, MECP2 and CUL3, also network hubs, only existed in the network of the late fetal period within the cortical region, suggesting their functional specificity during brain development. We also confirmed the physical interaction result between DGCR8 and CUL3 by liquid chromatography-tandem mass spectrometry. In conclusion, our results could suggest that the disruption of DGCR8-dependent microRNA biogenesis plays a vital role in NDD for late fetal cortical development.
Collapse
|
33
|
Kuru Hİ, Buyukozkan M, Tastan O. PRER: A patient representation with pairwise relative expression of proteins on biological networks. PLoS Comput Biol 2021; 17:e1008998. [PMID: 34038408 PMCID: PMC8238204 DOI: 10.1371/journal.pcbi.1008998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 06/28/2021] [Accepted: 04/23/2021] [Indexed: 11/19/2022] Open
Abstract
Changes in protein and gene expression levels are often used as features in predictive modeling such as survival prediction. A common strategy to aggregate information contained in individual proteins is to integrate the expression levels with the biological networks. In this work, we propose a novel patient representation where we integrate proteins’ expression levels with the protein-protein interaction (PPI) networks: Patient representation with PRER (Pairwise Relative Expressions with Random walks). PRER captures the dysregulation patterns of proteins based on the neighborhood of a protein in the PPI network. Specifically, PRER computes a feature vector for a patient by comparing the source protein’s expression level with other proteins’ levels that are within its neighborhood. The neighborhood of the source protein is derived by biased random-walk strategy on the network. We test PRER’s performance in survival prediction task in 10 different cancers using random forest survival models. PRER yields a statistically significant predictive performance in 9 out of 10 cancers when compared to the same model trained with features based on individual protein expressions. Furthermore, we identified the pairs of proteins that their interactions are predictive of patient survival but their individual expression levels are not. The set of identified relations provides a valuable collection of protein biomarkers with high prognostic value. PRER can be used for other complex diseases and prediction tasks that use molecular expression profiles as input. PRER is freely available at: https://github.com/hikuru/PRER. Cancer remains to be one of the most prevalent and challenging diseases to treat. Cancer is a complex disease with several disrupted molecular mechanisms at play. The protein expression level is a fundamental indicator of how the molecular mechanisms are altered in each tumor. Predicting patient survival based on the changes is essential for understanding the cancer mechanisms and arriving at patient-specific treatment plans. For this task, existing machine learning models are used, such as random survival forest, which requires a feature-based representation of each patient based on her tumors. Most of these models use the individual molecular quantities of the tumors. However, cancer is a complex disease in which molecular mechanisms are dysregulated in various ways. In this work, we present a new patient representation scheme in which we integrate each tumor’s protein expression levels with their neighboring proteins’ expression levels in a protein-protein interaction network to capture patient-specific dysregulation patterns. Our results suggest that proteins’ relative expressions are more predictive than their individual expressions. We also analyze which of the protein interactions are more predictive of patient survival. The identified set of important protein interactions can be potentially used for cancer prognosis.
Collapse
Affiliation(s)
| | | | - Oznur Tastan
- Faculty of Natural Sciences and Engineering, Sabanci University, Istanbul, Turkey
- * E-mail:
| |
Collapse
|
34
|
Raghu VK, Ge X, Balajiee A, Shirer DJ, Das I, Benos PV, Chrysanthis PK. A Pipeline for Integrated Theory and Data-Driven Modeling of Biomedical Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:811-822. [PMID: 32841121 PMCID: PMC8237279 DOI: 10.1109/tcbb.2020.3019237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Genome sequencing technologies have the potential to transform clinical decision making and biomedical research by enabling high-throughput measurements of the genome at a granular level. However, to truly understand mechanisms of disease and predict the effects of medical interventions, high-throughput data must be integrated with demographic, phenotypic, environmental, and behavioral data from individuals. Further, effective knowledge discovery methods must infer relationships between these data types. We recently proposed a pipeline (CausalMGM) to achieve this. CausalMGM uses probabilistic graphical models to infer the relationships between variables in the data; however, CausalMGM's graphical structure learning algorithm can only handle small datasets efficiently. We propose a new methodology (piPref-Div) that selects the most informative variables for CausalMGM, enabling it to scale. We validate the efficacy of piPref-Div against other feature selection methods and demonstrate how the use of the full pipeline improves breast cancer outcome prediction and provides biologically interpretable views of gene expression data.
Collapse
|
35
|
Bassignana G, Fransson J, Henry V, Colliot O, Zujovic V, De Vico Fallani F. Stepwise target controllability identifies dysregulations of macrophage networks in multiple sclerosis. Netw Neurosci 2021; 5:337-357. [PMID: 34189368 PMCID: PMC8233109 DOI: 10.1162/netn_a_00180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 12/14/2020] [Indexed: 12/27/2022] Open
Abstract
Identifying the nodes able to drive the state of a network is crucial to understand, and eventually control, biological systems. Despite recent advances, such identification remains difficult because of the huge number of equivalent controllable configurations, even in relatively simple networks. Based on the evidence that in many applications it is essential to test the ability of individual nodes to control a specific target subset, we develop a fast and principled method to identify controllable driver-target configurations in sparse and directed networks. We demonstrate our approach on simulated networks and experimental gene networks to characterize macrophage dysregulation in human subjects with multiple sclerosis.
Collapse
Affiliation(s)
- Giulia Bassignana
- Sorbonne University, UPMC Univ Paris 06, Inserm U-1127, CNRS UMR-7225, Institut du Cerveau et de la Moelle Epinière, Hopital Pitié-Salpêtrière, Paris, France
- Inria Paris, Aramis Project Team, Paris, France
| | - Jennifer Fransson
- Sorbonne University, UPMC Univ Paris 06, Inserm U-1127, CNRS UMR-7225, Institut du Cerveau et de la Moelle Epinière, Hopital Pitié-Salpêtrière, Paris, France
| | - Vincent Henry
- Sorbonne University, UPMC Univ Paris 06, Inserm U-1127, CNRS UMR-7225, Institut du Cerveau et de la Moelle Epinière, Hopital Pitié-Salpêtrière, Paris, France
- Inria Paris, Aramis Project Team, Paris, France
| | - Olivier Colliot
- Sorbonne University, UPMC Univ Paris 06, Inserm U-1127, CNRS UMR-7225, Institut du Cerveau et de la Moelle Epinière, Hopital Pitié-Salpêtrière, Paris, France
- Inria Paris, Aramis Project Team, Paris, France
| | - Violetta Zujovic
- Sorbonne University, UPMC Univ Paris 06, Inserm U-1127, CNRS UMR-7225, Institut du Cerveau et de la Moelle Epinière, Hopital Pitié-Salpêtrière, Paris, France
| | - Fabrizio De Vico Fallani
- Sorbonne University, UPMC Univ Paris 06, Inserm U-1127, CNRS UMR-7225, Institut du Cerveau et de la Moelle Epinière, Hopital Pitié-Salpêtrière, Paris, France
- Inria Paris, Aramis Project Team, Paris, France
| |
Collapse
|
36
|
Sahu D, Chang YL, Lin YC, Lin CC. Characterization of the Survival Influential Genes in Carcinogenesis. Int J Mol Sci 2021; 22:4384. [PMID: 33922264 PMCID: PMC8122717 DOI: 10.3390/ijms22094384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/18/2021] [Accepted: 04/20/2021] [Indexed: 11/25/2022] Open
Abstract
The genes influencing cancer patient mortality have been studied by survival analysis for many years. However, most studies utilized them only to support their findings associated with patient prognosis: their roles in carcinogenesis have not yet been revealed. Herein, we applied an in silico approach, integrating the Cox regression model with effect size estimated by the Monte Carlo algorithm, to screen survival-influential genes in more than 6000 tumor samples across 16 cancer types. We observed that the survival-influential genes had cancer-dependent properties. Moreover, the functional modules formed by the harmful genes were consistently associated with cell cycle in 12 out of the 16 cancer types and pan-cancer, showing that dysregulation of the cell cycle could harm patient prognosis in cancer. The functional modules formed by the protective genes are more diverse in cancers; the most prevalent functions are relevant for immune response, implying that patients with different cancer types might develop different mechanisms against carcinogenesis. We also identified a harmful set of 10 genes, with potential as prognostic biomarkers in pan-cancer. Briefly, our results demonstrated that the survival-influential genes could reveal underlying mechanisms in carcinogenesis and might provide clues for developing therapeutic targets for cancers.
Collapse
Affiliation(s)
| | | | | | - Chen-Ching Lin
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (D.S.); (Y.-L.C.); (Y.-C.L.)
| |
Collapse
|
37
|
Vincenzi M, Mercurio FA, Leone M. Protein Interaction Domains: Structural Features and Drug Discovery Applications (Part 2). Curr Med Chem 2021; 28:854-892. [PMID: 31942846 DOI: 10.2174/0929867327666200114114142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/28/2019] [Accepted: 11/04/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Proteins present a modular organization made up of several domains. Apart from the domains playing catalytic functions, many others are crucial to recruit interactors. The latter domains can be defined as "PIDs" (Protein Interaction Domains) and are responsible for pivotal outcomes in signal transduction and a certain array of normal physiological and disease-related pathways. Targeting such PIDs with small molecules and peptides able to modulate their interaction networks, may represent a valuable route to discover novel therapeutics. OBJECTIVE This work represents a continuation of a very recent review describing PIDs able to recognize post-translationally modified peptide segments. On the contrary, the second part concerns with PIDs that interact with simple peptide sequences provided with standard amino acids. METHODS Crucial structural information on different domain subfamilies and their interactomes was gained by a wide search in different online available databases (including the PDB (Protein Data Bank), the Pfam (Protein family), and the SMART (Simple Modular Architecture Research Tool)). Pubmed was also searched to explore the most recent literature related to the topic. RESULTS AND CONCLUSION PIDs are multifaceted: they have all diverse structural features and can recognize several consensus sequences. PIDs can be linked to different diseases onset and progression, like cancer or viral infections and find applications in the personalized medicine field. Many efforts have been centered on peptide/peptidomimetic inhibitors of PIDs mediated interactions but much more work needs to be conducted to improve drug-likeness and interaction affinities of identified compounds.
Collapse
Affiliation(s)
- Marian Vincenzi
- Institute of Biostructures and Bioimaging, National Research Council (CNR), Via Mezzocannone 16, 80134 Naples, Italy
| | - Flavia Anna Mercurio
- Institute of Biostructures and Bioimaging, National Research Council (CNR), Via Mezzocannone 16, 80134 Naples, Italy
| | - Marilisa Leone
- Institute of Biostructures and Bioimaging, National Research Council (CNR), Via Mezzocannone 16, 80134 Naples, Italy
| |
Collapse
|
38
|
Integrative genomics analysis identifies five promising genes implicated in insomnia risk based on multiple omics datasets. Biosci Rep 2021; 40:226183. [PMID: 32830860 PMCID: PMC7468094 DOI: 10.1042/bsr20201084] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 08/15/2020] [Accepted: 08/21/2020] [Indexed: 12/27/2022] Open
Abstract
In recent decades, many genome-wide association studies on insomnia have reported numerous genes harboring multiple risk variants. Nevertheless, the molecular functions of these risk variants conveying risk to insomnia are still ill-studied. In the present study, we integrated GWAS summary statistics (N=386,533) with two independent brain expression quantitative trait loci (eQTL) datasets (N=329) to determine whether expression-associated SNPs convey risk to insomnia. Furthermore, we applied numerous bioinformatics analyses to highlight promising genes associated with insomnia risk. By using Sherlock integrative analysis, we detected 449 significant insomnia-associated genes in the discovery stage. These identified genes were significantly overrepresented in six biological pathways including Huntington’s disease (P=5.58 × 10−5), Alzheimer’s disease (P=5.58 × 10−5), Parkinson’s disease (P=6.34 × 10−5), spliceosome (P=1.17 × 10−4), oxidative phosphorylation (P=1.09 × 10−4), and wnt signaling pathways (P=2.07 × 10−4). Further, five of these identified genes were replicated in an independent brain eQTL dataset. Through a PPI network analysis, we found that there existed highly functional interactions among these five identified genes. Three genes of LDHA (P=0.044), DALRD3 (P=5.0 × 10−5), and HEBP2 (P=0.032) showed significantly lower expression level in brain tissues of insomnic patients than that in controls. In addition, the expression levels of these five genes showed prominently dynamic changes across different time points between behavioral states of sleep and sleep deprivation in mice brain cortex. Together, the evidence of the present study strongly suggested that these five identified genes may represent candidate genes and contributed risk to the etiology of insomnia.
Collapse
|
39
|
Ebrahimpour Gorji A, Roudbari Z, Ebrahimpour Gorji F, Sadeghi B. Computational study of zebrafish immune-targeted microarray data for prediction of preventive drug candidates. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2021; 12:87-93. [PMID: 33953878 PMCID: PMC8094140 DOI: 10.30466/vrf.2019.94179.2270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 04/20/2019] [Indexed: 11/04/2022]
Abstract
Viral hemorrhagic septicemia virus (VHSV) is a rhabdovirus reported to cause economic loss in fish farms. Because of the lack of adequate preventative treatments, the identification of multipath genes involved in VHS infection might be an alternative to explore the possibility of using drugs for the seasonal prevention of this fish disease. We propose labeling a category of drug molecules by further classification and interpretation of the Drug Gene Interaction Database using gene ontology and Kyoto Encyclopedia of Genes and Genomes enrichment scores. The study investigated disease networks of up-and down-regulated genes to find those with high interaction as substantial genes in pathways among the different disease networks. We prioritized these genes based on their relationship to those associated with VHS infection in the context of human protein-protein interaction networks and disease pathways. Among the 29 genes as potential drug targets, nine were selected as promising druggable genes (ERBB2, FGFR3, ITGA2B, MAP2K1, NGF, NTRK1, PDGFRA, SCN2B, and SERPINC1). PDGFRA is the most important druggable up-and down-regulated gene and is considered an important gene in the IMATINIB pathway. This study findings indicate a promising approach for drug target prediction for VHS treatment, which might be useful for disease therapeutics.
Collapse
Affiliation(s)
- Abdolvahab Ebrahimpour Gorji
- Department of Fisheries, Faculty of Animal Sciences and Fisheries, Sari Agricultural and Natural Resources University, Sari, Iran
| | - Zahra Roudbari
- Department of Animal Science, Faculty of Agriculture, University of Jiroft, Jiroft, Iran
| | - Fatemeh Ebrahimpour Gorji
- Department of Cell and Molecular Biology, Faculty of Science, University of Andishesazan, Neka, Iran
| | - Balal Sadeghi
- Department of Food Hygiene and Public Health, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| |
Collapse
|
40
|
Shojaie A. Differential Network Analysis: A Statistical Perspective. WILEY INTERDISCIPLINARY REVIEWS. COMPUTATIONAL STATISTICS 2021; 13:e1508. [PMID: 37050915 PMCID: PMC10088462 DOI: 10.1002/wics.1508] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/03/2020] [Indexed: 11/06/2022]
Abstract
Networks effectively capture interactions among components of complex systems, and have thus become a mainstay in many scientific disciplines. Growing evidence, especially from biology, suggest that networks undergo changes over time, and in response to external stimuli. In biology and medicine, these changes have been found to be predictive of complex diseases. They have also been used to gain insight into mechanisms of disease initiation and progression. Primarily motivated by biological applications, this article provides a review of recent statistical machine learning methods for inferring networks and identifying changes in their structures.
Collapse
Affiliation(s)
- Ali Shojaie
- Department of Biostatistics, University of Washington, Seattle WA
| |
Collapse
|
41
|
Bar H, Bang S. A mixture model to detect edges in sparse co-expression graphs with an application for comparing breast cancer subtypes. PLoS One 2021; 16:e0246945. [PMID: 33571253 PMCID: PMC7877669 DOI: 10.1371/journal.pone.0246945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/28/2021] [Indexed: 11/19/2022] Open
Abstract
We develop a method to recover a gene network's structure from co-expression data, measured in terms of normalized Pearson's correlation coefficients between gene pairs. We treat these co-expression measurements as weights in the complete graph in which nodes correspond to genes. To decide which edges exist in the gene network, we fit a three-component mixture model such that the observed weights of 'null edges' follow a normal distribution with mean 0, and the non-null edges follow a mixture of two lognormal distributions, one for positively- and one for negatively-correlated pairs. We show that this so-called L2 N mixture model outperforms other methods in terms of power to detect edges, and it allows to control the false discovery rate. Importantly, our method makes no assumptions about the true network structure. We demonstrate our method, which is implemented in an R package called edgefinder, using a large dataset consisting of expression values of 12,750 genes obtained from 1,616 women. We infer the gene network structure by cancer subtype, and find insightful subtype characteristics. For example, we find thirteen pathways which are enriched in each of the cancer groups but not in the Normal group, with two of the pathways associated with autoimmune diseases and two other with graft rejection. We also find specific characteristics of different breast cancer subtypes. For example, the Luminal A network includes a single, highly connected cluster of genes, which is enriched in the human diseases category, and in the Her2 subtype network we find a distinct, and highly interconnected cluster which is uniquely enriched in drug metabolism pathways.
Collapse
Affiliation(s)
- Haim Bar
- Department of Statistics, University of Connecticut, Storrs, CT, United States of America
| | - Seojin Bang
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, United States of America
| |
Collapse
|
42
|
Yang K, Lu K, Wu Y, Yu J, Liu B, Zhao Y, Chen J, Zhou X. A network-based machine-learning framework to identify both functional modules and disease genes. Hum Genet 2021; 140:897-913. [PMID: 33409574 DOI: 10.1007/s00439-020-02253-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/22/2020] [Indexed: 01/20/2023]
Abstract
Disease gene identification is a critical step towards uncovering the molecular mechanisms of diseases and systematically investigating complex disease phenotypes. Despite considerable efforts to develop powerful computing methods, candidate gene identification remains a severe challenge owing to the connectivity of an incomplete interactome network, which hampers the discovery of true novel candidate genes. We developed a network-based machine-learning framework to identify both functional modules and disease candidate genes. In this framework, we designed a semi-supervised non-negative matrix factorization model to obtain the functional modules related to the diseases and genes. Of note, we proposed a disease gene-prioritizing method called MapGene that integrates the correlations from both functional modules and network closeness. Our framework identified a set of functional modules with highly functional homogeneity and close gene interactions. Experiments on a large-scale benchmark dataset showed that MapGene performs significantly better than the state-of-the-art algorithms. Further analysis demonstrates MapGene can effectively relieve the impact of the incompleteness of interactome networks and obtain highly reliable rankings of candidate genes. In addition, disease cases on Parkinson's disease and diabetes mellitus confirmed the generalization of MapGene for novel candidate gene identification. This work proposed, for the first time, an integrated computing framework to predict both functional modules and disease candidate genes. The methodology and results support that our framework has the potential to help discover underlying functional modules and reliable candidate genes in human disease.
Collapse
Affiliation(s)
- Kuo Yang
- School of Computer and Information Technology, Institute of Medical Intelligence, Beijing Jiaotong University, Beijing, 100044, China.,Institute for TCM-X, MOE Key Laboratory of Bioinformatics / Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, 10084, China
| | - Kezhi Lu
- School of Computer and Information Technology, Institute of Medical Intelligence, Beijing Jiaotong University, Beijing, 100044, China.,imec-DistriNet, KU Leuven, Leuven, 3001, Belgium
| | - Yang Wu
- Key Laboratory of Intelligent Information Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jian Yu
- Beijing Key Laboratory of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing, 100044, China
| | - Baoyan Liu
- Data Center of Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yi Zhao
- Key Laboratory of Intelligent Information Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jianxin Chen
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xuezhong Zhou
- School of Computer and Information Technology, Institute of Medical Intelligence, Beijing Jiaotong University, Beijing, 100044, China. .,Data Center of Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
43
|
Abstract
Cell-surface adhesion receptors mediate interactions with the extracellular matrix (ECM) to control many fundamental aspects of cell behavior, including cell migration, survival, and proliferation. Integrin adhesion receptors recruit structural and signaling proteins to form multimolecular adhesion complexes that link the plasma membrane to the actomyosin cytoskeleton. The assembly and turnover of adhesion complexes are tightly regulated, governed in part by the networks of physical protein interactions and functional signaling associations between components of the adhesome. Proteomic profiling of adhesion complexes has begun to reveal their molecular complexity and diversity. To interrogate the composition of cell-ECM adhesions, we detail herein an approach for the network analysis of adhesion complex proteomes. Integration of these proteomic data with adhesome databases in the context of predicted protein interactions enables the mapping of experimentally defined adhesion complex networks. Computational analysis of resultant network models can identify subnetworks of putative functionally linked adhesion protein communities. This approach provides a framework to predict functional adhesion protein relationships and generate new mechanistic hypotheses for further experimental testing.
Collapse
Affiliation(s)
- Frederic Li Mow Chee
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Adam Byron
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
44
|
Savino A, Provero P, Poli V. Differential Co-Expression Analyses Allow the Identification of Critical Signalling Pathways Altered during Tumour Transformation and Progression. Int J Mol Sci 2020; 21:E9461. [PMID: 33322692 PMCID: PMC7764314 DOI: 10.3390/ijms21249461] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/02/2020] [Accepted: 12/09/2020] [Indexed: 02/02/2023] Open
Abstract
Biological systems respond to perturbations through the rewiring of molecular interactions, organised in gene regulatory networks (GRNs). Among these, the increasingly high availability of transcriptomic data makes gene co-expression networks the most exploited ones. Differential co-expression networks are useful tools to identify changes in response to an external perturbation, such as mutations predisposing to cancer development, and leading to changes in the activity of gene expression regulators or signalling. They can help explain the robustness of cancer cells to perturbations and identify promising candidates for targeted therapy, moreover providing higher specificity with respect to standard co-expression methods. Here, we comprehensively review the literature about the methods developed to assess differential co-expression and their applications to cancer biology. Via the comparison of normal and diseased conditions and of different tumour stages, studies based on these methods led to the definition of pathways involved in gene network reorganisation upon oncogenes' mutations and tumour progression, often converging on immune system signalling. A relevant implementation still lagging behind is the integration of different data types, which would greatly improve network interpretability. Most importantly, performance and predictivity evaluation of the large variety of mathematical models proposed would urgently require experimental validations and systematic comparisons. We believe that future work on differential gene co-expression networks, complemented with additional omics data and experimentally tested, will considerably improve our insights into the biology of tumours.
Collapse
Affiliation(s)
- Aurora Savino
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Paolo Provero
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Corso Massimo D’Ázeglio 52, 10126 Turin, Italy;
- Center for Omics Sciences, Ospedale San Raffaele IRCCS, Via Olgettina 60, 20132 Milan, Italy
| | - Valeria Poli
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| |
Collapse
|
45
|
Niss K, Jakobsson ME, Westergaard D, Belling KG, Olsen JV, Brunak S. Effects of active farnesoid X receptor on GLUTag enteroendocrine L cells. Mol Cell Endocrinol 2020; 517:110923. [PMID: 32702472 DOI: 10.1016/j.mce.2020.110923] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/27/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022]
Abstract
Activated transcription factor (TF) farnesoid X receptor (FXR) represses glucagon-like peptide-1 (GLP-1) secretion in enteroendocrine L cells. This, in turn, reduces insulin secretion, which is triggered when β cells bind GLP-1. Preventing FXR activation could boost GLP-1 production and insulin secretion. Yet, FXR's broader role in L cell biology still lacks understanding. Here, we show that FXR is a multifaceted TF in L cells using proteomics and gene expression data generated on GLUTag L cells. Most striking, 252 proteins regulated upon glucose stimulation have their abundances neutralized upon FXR activation. Mitochondrial repression or glucose import block are likely mechanisms of this. Further, FXR physically targets bile acid metabolism proteins, growth factors and other TFs, regulates ChREBP, while extensive text-mining found 30 FXR-regulated proteins to be well-known in L cell biology. Taken together, this outlines FXR as a powerful TF, where GLP-1 secretion block is just one of many downstream effects.
Collapse
Affiliation(s)
- Kristoffer Niss
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Magnus E Jakobsson
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark; Department of Immunotechnology, Lund University, Medicon Village, 22100, Lund, Sweden
| | - David Westergaard
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark; Dept. of Health Technology, Technical University of Denmark, DK-2800, Lyngby, Denmark
| | - Kirstine G Belling
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark; Dept. of Health Technology, Technical University of Denmark, DK-2800, Lyngby, Denmark.
| |
Collapse
|
46
|
Lucchetta M, Pellegrini M. Finding disease modules for cancer and COVID-19 in gene co-expression networks with the Core&Peel method. Sci Rep 2020; 10:17628. [PMID: 33077837 PMCID: PMC7573595 DOI: 10.1038/s41598-020-74705-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/30/2020] [Indexed: 12/21/2022] Open
Abstract
Genes are organized in functional modules (or pathways), thus their action and their dysregulation in diseases may be better understood by the identification of the modules most affected by the disease (aka disease modules, or active subnetworks). We describe how an algorithm based on the Core&Peel method is used to detect disease modules in co-expression networks of genes. We first validate Core&Peel for the general task of functional module detection by comparison with 42 methods participating in the Disease Module Identification DREAM challenge. Next, we use four specific disease test cases (colorectal cancer, prostate cancer, asthma, and rheumatoid arthritis), four state-of-the-art algorithms (ModuleDiscoverer, Degas, KeyPathwayMiner, and ClustEx), and several pathway databases to validate the proposed algorithm. Core&Peel is the only method able to find significant associations of the predicted disease module with known validated relevant pathways for all four diseases. Moreover, for the two cancer datasets, Core&Peel detects further eight relevant pathways not discovered by the other methods used in the comparative analysis. Finally, we apply Core&Peel and other methods to explore the transcriptional response of human cells to SARS-CoV-2 infection, finding supporting evidence for drug repositioning efforts at a pre-clinical level.
Collapse
Affiliation(s)
- Marta Lucchetta
- Institute of Informatics and Telematics (IIT), CNR, Pisa, 56124, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, 53100, Italy
| | - Marco Pellegrini
- Institute of Informatics and Telematics (IIT), CNR, Pisa, 56124, Italy.
| |
Collapse
|
47
|
Dong Z, Ma Y, Zhou H, Shi L, Ye G, Yang L, Liu P, Zhou L. Integrated genomics analysis highlights important SNPs and genes implicated in moderate-to-severe asthma based on GWAS and eQTL datasets. BMC Pulm Med 2020; 20:270. [PMID: 33066754 PMCID: PMC7568423 DOI: 10.1186/s12890-020-01303-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 09/27/2020] [Indexed: 02/06/2023] Open
Abstract
Background Severe asthma is a chronic disease contributing to disproportionate disease morbidity and mortality. From the year of 2007, many genome-wide association studies (GWAS) have documented a large number of asthma-associated genetic variants and related genes. Nevertheless, the molecular mechanism of these identified variants involved in asthma or severe asthma risk remains largely unknown. Methods In the current study, we systematically integrated 3 independent expression quantitative trait loci (eQTL) data (N = 1977) and a large-scale GWAS summary data of moderate-to-severe asthma (N = 30,810) by using the Sherlock Bayesian analysis to identify whether expression-related variants contribute risk to severe asthma. Furthermore, we performed various bioinformatics analyses, including pathway enrichment analysis, PPI network enrichment analysis, in silico permutation analysis, DEG analysis and co-expression analysis, to prioritize important genes associated with severe asthma. Results In the discovery stage, we identified 1129 significant genes associated with moderate-to-severe asthma by using the Sherlock Bayesian analysis. Two hundred twenty-eight genes were prominently replicated by using MAGMA gene-based analysis. These 228 replicated genes were enriched in 17 biological pathways including antigen processing and presentation (Corrected P = 4.30 × 10− 6), type I diabetes mellitus (Corrected P = 7.09 × 10− 5), and asthma (Corrected P = 1.72 × 10− 3). With the use of a series of bioinformatics analyses, we highlighted 11 important genes such as GNGT2, TLR6, and TTC19 as authentic risk genes associated with moderate-to-severe/severe asthma. With respect to GNGT2, there were 3 eSNPs of rs17637472 (PeQTL = 2.98 × 10− 8 and PGWAS = 3.40 × 10− 8), rs11265180 (PeQTL = 6.0 × 10− 6 and PGWAS = 1.99 × 10− 3), and rs1867087 (PeQTL = 1.0 × 10− 4 and PGWAS = 1.84 × 10− 5) identified. In addition, GNGT2 is significantly expressed in severe asthma compared with mild-moderate asthma (P = 0.045), and Gngt2 shows significantly distinct expression patterns between vehicle and various glucocorticoids (Anova P = 1.55 × 10− 6). Conclusions Our current study provides multiple lines of evidence to support that these 11 identified genes as important candidates implicated in the pathogenesis of severe asthma.
Collapse
Affiliation(s)
- Zhouzhou Dong
- Critical Care Unit, Ningbo Medical Center Lihuili Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang, 315100, P.R. China
| | - Yunlong Ma
- Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.,School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Hua Zhou
- Department of Respiratory Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Linhui Shi
- Critical Care Unit, Ningbo Medical Center Lihuili Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang, 315100, P.R. China
| | - Gongjie Ye
- Critical Care Unit, Ningbo Medical Center Lihuili Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang, 315100, P.R. China
| | - Lei Yang
- Critical Care Unit, Ningbo Medical Center Lihuili Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang, 315100, P.R. China
| | - Panpan Liu
- Critical Care Unit, Ningbo Medical Center Lihuili Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang, 315100, P.R. China
| | - Li Zhou
- Department of Immunology and Rheumatology, Ningbo Medical Center Lihuili Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang, 315100, P.R. China.
| |
Collapse
|
48
|
Stolfi P, Manni L, Soligo M, Vergni D, Tieri P. Designing a Network Proximity-Based Drug Repurposing Strategy for COVID-19. Front Cell Dev Biol 2020; 8:545089. [PMID: 33123533 PMCID: PMC7573309 DOI: 10.3389/fcell.2020.545089] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
The ongoing COVID-19 pandemic still requires fast and effective efforts from all fronts, including epidemiology, clinical practice, molecular medicine, and pharmacology. A comprehensive molecular framework of the disease is needed to better understand its pathological mechanisms, and to design successful treatments able to slow down and stop the impressive pace of the outbreak and harsh clinical symptomatology, possibly via the use of readily available, off-the-shelf drugs. This work engages in providing a wider picture of the human molecular landscape of the SARS-CoV-2 infection via a network medicine approach as the ground for a drug repurposing strategy. Grounding on prior knowledge such as experimentally validated host proteins known to be viral interactors, tissue-specific gene expression data, and using network analysis techniques such as network propagation and connectivity significance, the host molecular reaction network to the viral invasion is explored and exploited to infer and prioritize candidate target genes, and finally to propose drugs to be repurposed for the treatment of COVID-19. Ranks of potential target genes have been obtained for coherent groups of tissues/organs, potential and distinct sites of interaction between the virus and the organism. The normalization and the aggregation of the different scores allowed to define a preliminary, restricted list of genes candidates as pharmacological targets for drug repurposing, with the aim of contrasting different phases of the virus infection and viral replication cycle.
Collapse
Affiliation(s)
- Paola Stolfi
- National Research Council (CNR), Institute for Applied Computing (IAC), Rome, Italy
| | - Luigi Manni
- National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| | - Marzia Soligo
- National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| | - Davide Vergni
- National Research Council (CNR), Institute for Applied Computing (IAC), Rome, Italy
| | - Paolo Tieri
- National Research Council (CNR), Institute for Applied Computing (IAC), Rome, Italy
| |
Collapse
|
49
|
Nam JH, Couch D, da Silveira WA, Yu Z, Chung D. PALMER: improving pathway annotation based on the biomedical literature mining with a constrained latent block model. BMC Bioinformatics 2020; 21:432. [PMID: 33008309 PMCID: PMC7532116 DOI: 10.1186/s12859-020-03756-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 09/16/2020] [Indexed: 11/23/2022] Open
Abstract
Background In systems biology, it is of great interest to identify previously unreported associations between genes. Recently, biomedical literature has been considered as a valuable resource for this purpose. While classical clustering algorithms have popularly been used to investigate associations among genes, they are not tuned for the literature mining data and are also based on strong assumptions, which are often violated in this type of data. For example, these approaches often assume homogeneity and independence among observations. However, these assumptions are often violated due to both redundancies in functional descriptions and biological functions shared among genes. Latent block models can be alternatives in this case but they also often show suboptimal performances, especially when signals are weak. In addition, they do not allow to utilize valuable prior biological knowledge, such as those available in existing databases. Results In order to address these limitations, here we propose PALMER, a constrained latent block model that allows to identify indirect relationships among genes based on the biomedical literature mining data. By automatically associating relevant Gene Ontology terms, PALMER facilitates biological interpretation of novel findings without laborious downstream analyses. PALMER also allows researchers to utilize prior biological knowledge about known gene-pathway relationships to guide identification of gene–gene associations. We evaluated PALMER with simulation studies and applications to studies of pathway-modulating genes relevant to cancer signaling pathways, while utilizing biological pathway annotations available in the KEGG database as prior knowledge. Conclusions We showed that PALMER outperforms traditional latent block models and it provides reliable identification of novel gene–gene associations by utilizing prior biological knowledge, especially when signals are weak in the biomedical literature mining dataset. We believe that PALMER and its relevant user-friendly software will be powerful tools that can be used to improve existing pathway annotations and identify novel pathway-modulating genes.
Collapse
Affiliation(s)
- Jin Hyun Nam
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA.,School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Daniel Couch
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | | | - Zhenning Yu
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Dongjun Chung
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
50
|
Adnan N, Lei C, Ruan J. Robust edge-based biomarker discovery improves prediction of breast cancer metastasis. BMC Bioinformatics 2020; 21:359. [PMID: 32998692 PMCID: PMC7526355 DOI: 10.1186/s12859-020-03692-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background The abundance of molecular profiling of breast cancer tissues entailed active research on molecular marker-based early diagnosis of metastasis. Recently there is a surging interest in combining gene expression with gene networks such as protein-protein interaction (PPI) network, gene co-expression (CE) network and pathway information to identify robust and accurate biomarkers for metastasis prediction, reflecting the common belief that cancer is a systems biology disease. However, controversy exists in the literature regarding whether network markers are indeed better features than genes alone for predicting as well as understanding metastasis. We believe much of the existing results may have been biased by the overly complicated prediction algorithms, unfair evaluation, and lack of rigorous statistics. In this study, we propose a simple approach to use network edges as features, based on two types of networks respectively, and compared their prediction power using three classification algorithms and rigorous statistical procedure on one of the largest datasets available. To detect biomarkers that are significant for the prediction and to compare the robustness of different feature types, we propose an unbiased and novel procedure to measure feature importance that eliminates the potential bias from factors such as different sample size, number of features, as well as class distribution. Results Experimental results reveal that edge-based feature types consistently outperformed gene-based feature type in random forest and logistic regression models under all performance evaluation metrics, while the prediction accuracy of edge-based support vector machine (SVM) model was poorer, due to the larger number of edge features compared to gene features and the lack of feature selection in SVM model. Experimental results also show that edge features are much more robust than gene features and the top biomarkers from edge feature types are statistically more significantly enriched in the biological processes that are well known to be related to breast cancer metastasis. Conclusions Overall, this study validates the utility of edge features as biomarkers but also highlights the importance of carefully designed experimental procedures in order to achieve statistically reliable comparison results.
Collapse
Affiliation(s)
- Nahim Adnan
- Department of Computer Science, The University of Texas at San Antonio, One UTSA Circle, San Antonio, 78249, TX, USA
| | - Chengwei Lei
- Department of Computer & Electrical Engineering/Computer Science, California State University, Bakersfield, 9001 Stockdale Highway, Bakersfield, 93311, CA, USA
| | - Jianhua Ruan
- Department of Computer Science, The University of Texas at San Antonio, One UTSA Circle, San Antonio, 78249, TX, USA.
| |
Collapse
|