1
|
Feng F, Liu T, Hou X, Lin X, Zhou S, Tian Y, Qi X. Targeting the FSH/FSHR axis in ovarian cancer: advanced treatment using nanotechnology and immunotherapy. Front Endocrinol (Lausanne) 2024; 15:1489767. [PMID: 39741875 PMCID: PMC11685086 DOI: 10.3389/fendo.2024.1489767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
Ovarian cancer (OC) is the gynecological malignancy with the poorest prognosis. Surgery and chemotherapy are the primary therapies for OC; however, patients often experience recurrence. Given the intimate interaction between OC cells and the tumor microenvironment (TME), it is imperative to devise treatments that target both tumor cells and TME components. Recently, follicle-stimulating hormone (FSH) levels in the blood have been shown to correlate with poorer prognosis in individuals with OC. Ovarian carcinoma cells express FSH receptors (FSHRs). Thus, FSH is an important target in the development of novel therapeutic agents. Here, we review the effects of FSH on normal physiology, including the reproductive, skeletal, cardiac, and fat metabolic systems. Importantly, this review outlines the role and mechanism of the FSH/FSHR axis in the proliferation, survival, and metastasis of OC, providing theoretical support for the targeted FSHR treatment of OC. Current progress in targeting FSHR for OC, including the recent application of nanotechnology and immunotherapy, is presented. Finally, we discuss prospects and future directions of targeted FSHR therapy in OC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaoyi Qi
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
2
|
Luchetti N, Smith KM, Matarrese MAG, Loppini A, Filippi S, Chiodo L. A statistical mechanics investigation of unfolded protein response across organisms. Sci Rep 2024; 14:27658. [PMID: 39532983 PMCID: PMC11557608 DOI: 10.1038/s41598-024-79086-8] [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: 08/13/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
Living systems rely on coordinated molecular interactions, especially those related to gene expression and protein activity. The Unfolded Protein Response is a crucial mechanism in eukaryotic cells, activated when unfolded proteins exceed a critical threshold. It maintains cell homeostasis by enhancing protein folding, initiating quality control, and activating degradation pathways when damage is irreversible. This response functions as a dynamic signaling network, with proteins as nodes and their interactions as edges. We analyze these protein-protein networks across different organisms to understand their intricate intra-cellular interactions and behaviors. In this work, analyzing twelve organisms, we assess how fundamental measures in network theory can individuate seed proteins and specific pathways across organisms. We employ network robustness to evaluate and compare the strength of the investigated protein-protein interaction networks, and the structural controllability of complex networks to find and compare the sets of driver nodes necessary to control the overall networks. We find that network measures are related to phylogenetics, and advanced network methods can identify main pathways of significance in the complete Unfolded Protein Response mechanism.
Collapse
Affiliation(s)
- Nicole Luchetti
- Department of Engineering, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo 21, Rome, 00128, Italy.
- Center for Life Nano- & Neuro-Science, Italian Institute of Technology, Viale Regina Elena 291, Rome, 00161, Italy.
| | - Keith M Smith
- Computer and Information Sciences, University of Strathclyde, 26 Richmond Street, Glasgow, G1 1XH, United Kingdom
| | - Margherita A G Matarrese
- Department of Engineering, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo 21, Rome, 00128, Italy
| | - Alessandro Loppini
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo 21, Rome, 00128, Italy
| | - Simonetta Filippi
- Department of Engineering, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo 21, Rome, 00128, Italy.
- National Institute of Optics, National Research Council, Largo Enrico Fermi 6, Florence, 50125, Italy.
- International Center for Relativistic Astrophysics Network, Piazza della Repubblica 10, Pescara, 65122, Italy.
| | - Letizia Chiodo
- Department of Engineering, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo 21, Rome, 00128, Italy
| |
Collapse
|
3
|
Azmal M, Paul JK, Prima FS, Talukder OF, Ghosh A. An in silico molecular docking and simulation study to identify potential anticancer phytochemicals targeting the RAS signaling pathway. PLoS One 2024; 19:e0310637. [PMID: 39298437 PMCID: PMC11412525 DOI: 10.1371/journal.pone.0310637] [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: 06/09/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
The dysregulation of the rat sarcoma (RAS) signaling pathway, particularly the MAPK/ERK cascade, is a hallmark of many cancers, leading to uncontrolled cellular proliferation and resistance to apoptosis-inducing treatments. Dysregulation of the MAPK/ERK pathway is common in various cancers including pancreatic, lung, and colon cancers, making it a critical target for therapeutic intervention. Natural compounds, especially phytochemicals, offer a promising avenue for developing new anticancer therapies due to their potential to interfere with these signaling pathways. This study investigates the potential of anticancer phytochemicals to inhibit the MAPK/ERK pathway through molecular docking and simulation techniques. A total of 26 phytochemicals were screened from an initial set of 340 phytochemicals which were retrieved from Dr. Duke's database using in silico methods for their binding affinity and stability. Molecular docking was performed to identify key interactions with ERK2, followed by molecular dynamics (MD) simulations to evaluate the stability of these interactions. The study identified several phytochemicals, including luteolin, hispidulin, and isorhamnetin with a binding score of -10.1±0 Kcal/mol, -9.86±0.15 Kcal/mol, -9.76±0.025 Kcal/mol, respectively as promising inhibitors of the ERK2 protein. These compounds demonstrated significant binding affinities and stable interactions with ERK2 in MD simulation studies up to 200ns, particularly at the active site. The radius of gyration analysis confirmed the stability of these phytochemical-protein complexes' compactness, indicating their potential to inhibit ERK activity. The stability and binding affinity of these compounds suggest that they can effectively inhibit ERK2 activity, potentially leading to more effective and less toxic cancer treatments. The findings underscore the therapeutic promise of these phytochemicals, which could serve as a basis for developing new cancer therapies.
Collapse
Affiliation(s)
- Mahir Azmal
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Jibon Kumar Paul
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Fatema Sultana Prima
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Omar Faruk Talukder
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| |
Collapse
|
4
|
Kim D, Nita-Lazar A. Progress in mass spectrometry approaches to profiling protein-protein interactions in the studies of the innate immune system. JOURNAL OF PROTEINS AND PROTEOMICS 2024; 15:545-559. [PMID: 39380887 PMCID: PMC11460538 DOI: 10.1007/s42485-024-00156-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/04/2024] [Accepted: 06/24/2024] [Indexed: 10/10/2024]
Abstract
Understanding protein-protein interactions (PPIs) is pivotal for deciphering the intricacies of biological processes. Dysregulation of PPIs underlies a spectrum of diseases, including cancer, neurodegenerative disorders, and autoimmune conditions, highlighting the imperative of investigating these interactions for therapeutic advancements. This review delves into the realm of mass spectrometry-based techniques for elucidating PPIs and their profound implications in biological research. Mass spectrometry in the PPI research field not only facilitates the evaluation of protein-protein interaction modulators but also discovers unclear molecular mechanisms and sheds light on both on- and off-target effects, thus aiding in drug development. Our discussion navigates through six pivotal techniques: affinity purification mass spectrometry (AP-MS), proximity labeling mass spectrometry (PL-MS), cross-linking mass spectrometry (XL-MS), size exclusion chromatography coupled with mass spectrometry (SEC-MS), limited proteolysis-coupled mass spectrometry (LiP-MS), and thermal proteome profiling (TPP).
Collapse
Affiliation(s)
- Doeun Kim
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1892, USA
| | - Aleksandra Nita-Lazar
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1892, USA
| |
Collapse
|
5
|
Arora G, Banerjee M, Langthasa J, Bhat R, Chatterjee S. Targeting metabolic fluxes reverts metastatic transitions in ovarian cancer. iScience 2023; 26:108081. [PMID: 37876796 PMCID: PMC10590820 DOI: 10.1016/j.isci.2023.108081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/05/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023] Open
Abstract
The formation of spheroids during epithelial ovarian cancer progression is correlated with peritoneal metastasis, disease recurrence, and poor prognosis. Although metastasis has been demonstrated to be driven by metabolic changes in transformed cells, mechanistic associations between metabolism and phenotypic transitions remain ill-explored. We performed quantitative proteomics to identify protein signatures associated with three distinct phenotypic morphologies (2D monolayers and two geometrically distinct three-dimensional spheroidal states) of the high-grade serous ovarian cancer line OVCAR-3. We obtained disease-driving phenotype-specific metabolic reaction modules and elucidated gene knockout strategies to reduce metabolic alterations that could drive phenotypic transitions. Exploring the DrugBank database, we identified and evaluated drugs that could impair such transitions and, hence, cancer progression. Finally, we experimentally validated our predictions by confirming the ability of one of our predicted drugs, the neuraminidase inhibitor oseltamivir, to inhibit spheroidogenesis in three ovarian cancer cell lines without any cytotoxic effects on untransformed stromal mesothelia.
Collapse
Affiliation(s)
- Garhima Arora
- Complex Analysis Group, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Mallar Banerjee
- Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Jimpi Langthasa
- Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Ramray Bhat
- Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
- BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Samrat Chatterjee
- Complex Analysis Group, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| |
Collapse
|
6
|
Zhang X, Pan C, Wei X, Yu M, Liu S, An J, Yang J, Wei B, Hao W, Yao Y, Zhu Y, Zhang W. Cancer-keeper genes as therapeutic targets. iScience 2023; 26:107296. [PMID: 37520717 PMCID: PMC10382876 DOI: 10.1016/j.isci.2023.107296] [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: 01/17/2023] [Revised: 05/18/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
Finding cancer-driver genes has been a central theme of cancer research. We took a different perspective; instead of considering normal cells, we focused on cancerous cells and genes that maintained abnormal cell growth, which we named cancer-keeper genes (CKGs). Intervening CKGs may rectify aberrant cell growth, making them potential cancer therapeutic targets. We introduced control-hub genes and developed an efficient algorithm by extending network controllability theory. Control hub are essential for maintaining cancerous states and thus can be taken as CKGs. We applied our CKG-based approach to bladder cancer (BLCA). All genes on the cell-cycle and p53 pathways in BLCA were identified as CKGs, showing their importance in cancer. We discovered that sensitive CKGs - genes easily altered by structural perturbation - were particularly suitable therapeutic targets. Experiments on cell lines and a mouse model confirmed that six sensitive CKGs effectively suppressed cancer cell growth, demonstrating the immense therapeutic potential of CKGs.
Collapse
Affiliation(s)
- Xizhe Zhang
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Chunyu Pan
- School of Computer Science and Engineering, Northeastern University, Shenyang, China
| | - Xinru Wei
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Meng Yu
- Department of Laboratory Animal Science, China Medical University, Shenyang, China
- Key Laboratory of Transgenetic Animal Research, China Medical University, Shenyang, China
| | - Shuangjie Liu
- Department of Urology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jun An
- Department of Urology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jieping Yang
- Department of Urology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Baojun Wei
- Department of Urology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Wenjun Hao
- Department of Urology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yang Yao
- Department of Physiology, Shenyang Medical College, Shenyang, China
| | - Yuyan Zhu
- Department of Urology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Weixiong Zhang
- Department of Health Technology and Informatics, Department of Computing, The Hong Kong Polytechnic University, Hong Kong, China
- Department of Computer Science and Engineering, Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
7
|
Wei X, Pan C, Zhang X, Zhang W. Total network controllability analysis discovers explainable drugs for Covid-19 treatment. RESEARCH SQUARE 2023:rs.3.rs-3147521. [PMID: 37503262 PMCID: PMC10371104 DOI: 10.21203/rs.3.rs-3147521/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Background The active pursuit of network medicine for drug repurposing, particularly for combating Covid-19, has stimulated interest in the concept of structural control capability in cellular networks. We sought to extend this theory, focusing on the defense rather than control of the cell against viral infections. Accordingly, we extended structural controllability to total structural controllability and introduced the concept of control hubs. Perturbing any control hub may render the cell uncontrollable by exogenous stimuli like viral infections, so control hubs are ideal drug targets. Results We developed an efficient algorithm to identify all control hubs, applying it to the largest homogeneous network of human protein interactions, including interactions between human and SARS-CoV-2 proteins. Our method recognized 65 druggable control hubs with enriched antiviral functions. Utilizing these hubs, we categorized potential drugs into four groups: antiviral and anti-inflammatory agents, drugs acting on the central nervous system, dietary supplements, and compounds enhancing immunity. An exemplification of our approach's effectiveness, Fostamatinib, a drug initially developed for chronic immune thrombocytopenia, is now in clinical trials for treating Covid-19. Preclinical trial data demonstrated that Fostamatinib could reduce mortality rates, ICU stay length, and disease severity in Covid-19 patients. Conclusions Our findings confirm the efficacy of our novel strategy that leverages control hubs as drug targets. This approach provides insights into the molecular mechanisms of potential therapeutics for Covid-19, making it a valuable tool for interpretable drug discovery.
Collapse
Affiliation(s)
- Xinru Wei
- The Affiliated Brain Hospital of Nanjing Medical University
| | | | - Xizhe Zhang
- The Affiliated Brain Hospital of Nanjing Medical University
| | | |
Collapse
|
8
|
Wang Q, Nute M, Treangen TJ. Bakdrive: identifying a minimum set of bacterial species driving interactions across multiple microbial communities. Bioinformatics 2023; 39:i47-i56. [PMID: 37387148 DOI: 10.1093/bioinformatics/btad236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023] Open
Abstract
MOTIVATION Interactions among microbes within microbial communities have been shown to play crucial roles in human health. In spite of recent progress, low-level knowledge of bacteria driving microbial interactions within microbiomes remains unknown, limiting our ability to fully decipher and control microbial communities. RESULTS We present a novel approach for identifying species driving interactions within microbiomes. Bakdrive infers ecological networks of given metagenomic sequencing samples and identifies minimum sets of driver species (MDS) using control theory. Bakdrive has three key innovations in this space: (i) it leverages inherent information from metagenomic sequencing samples to identify driver species, (ii) it explicitly takes host-specific variation into consideration, and (iii) it does not require a known ecological network. In extensive simulated data, we demonstrate identifying driver species identified from healthy donor samples and introducing them to the disease samples, we can restore the gut microbiome in recurrent Clostridioides difficile (rCDI) infection patients to a healthy state. We also applied Bakdrive to two real datasets, rCDI and Crohn's disease patients, uncovering driver species consistent with previous work. Bakdrive represents a novel approach for capturing microbial interactions. AVAILABILITY AND IMPLEMENTATION Bakdrive is open-source and available at: https://gitlab.com/treangenlab/bakdrive.
Collapse
Affiliation(s)
- Qi Wang
- Systems, Synthetic, and Physical Biology (SSPB) Graduate Program, Rice University, Houston, TX 77005, United States
| | - Michael Nute
- Department of Computer Science, Rice University, Houston, TX 77005, United States
| | - Todd J Treangen
- Department of Computer Science, Rice University, Houston, TX 77005, United States
| |
Collapse
|
9
|
Rodina A, Xu C, Digwal CS, Joshi S, Patel Y, Santhaseela AR, Bay S, Merugu S, Alam A, Yan P, Yang C, Roychowdhury T, Panchal P, Shrestha L, Kang Y, Sharma S, Almodovar J, Corben A, Alpaugh ML, Modi S, Guzman ML, Fei T, Taldone T, Ginsberg SD, Erdjument-Bromage H, Neubert TA, Manova-Todorova K, Tsou MFB, Young JC, Wang T, Chiosis G. Systems-level analyses of protein-protein interaction network dysfunctions via epichaperomics identify cancer-specific mechanisms of stress adaptation. Nat Commun 2023; 14:3742. [PMID: 37353488 PMCID: PMC10290137 DOI: 10.1038/s41467-023-39241-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 06/05/2023] [Indexed: 06/25/2023] Open
Abstract
Systems-level assessments of protein-protein interaction (PPI) network dysfunctions are currently out-of-reach because approaches enabling proteome-wide identification, analysis, and modulation of context-specific PPI changes in native (unengineered) cells and tissues are lacking. Herein, we take advantage of chemical binders of maladaptive scaffolding structures termed epichaperomes and develop an epichaperome-based 'omics platform, epichaperomics, to identify PPI alterations in disease. We provide multiple lines of evidence, at both biochemical and functional levels, demonstrating the importance of these probes to identify and study PPI network dysfunctions and provide mechanistically and therapeutically relevant proteome-wide insights. As proof-of-principle, we derive systems-level insight into PPI dysfunctions of cancer cells which enabled the discovery of a context-dependent mechanism by which cancer cells enhance the fitness of mitotic protein networks. Importantly, our systems levels analyses support the use of epichaperome chemical binders as therapeutic strategies aimed at normalizing PPI networks.
Collapse
Affiliation(s)
- Anna Rodina
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Chao Xu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Chander S Digwal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Suhasini Joshi
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yogita Patel
- Department of Biochemistry, Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Anand R Santhaseela
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Sadik Bay
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Swathi Merugu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Aftab Alam
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Pengrong Yan
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Chenghua Yang
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Tanaya Roychowdhury
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Palak Panchal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Liza Shrestha
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yanlong Kang
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Sahil Sharma
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Justina Almodovar
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Adriana Corben
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Maimonides Medical Center, Brooklyn, NY, USA
| | - Mary L Alpaugh
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Rowan University, Glassboro, NJ, USA
| | - Shanu Modi
- Department of Medicine, Division of Solid Tumors, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Monica L Guzman
- Department of Medicine, Division of Hematology Oncology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Tony Taldone
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Stephen D Ginsberg
- Departments of Psychiatry, Neuroscience & Physiology & the NYU Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
| | - Hediye Erdjument-Bromage
- Department of Neuroscience and Physiology and Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Thomas A Neubert
- Department of Neuroscience and Physiology and Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Katia Manova-Todorova
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Meng-Fu Bryan Tsou
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jason C Young
- Department of Biochemistry, Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Tai Wang
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Department of Medicine, Division of Solid Tumors, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
10
|
Ginsberg SD, Sharma S, Norton L, Chiosis G. Targeting stressor-induced dysfunctions in protein-protein interaction networks via epichaperomes. Trends Pharmacol Sci 2023; 44:20-33. [PMID: 36414432 PMCID: PMC9789192 DOI: 10.1016/j.tips.2022.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/31/2022] [Accepted: 10/31/2022] [Indexed: 11/21/2022]
Abstract
Diseases are manifestations of complex changes in protein-protein interaction (PPI) networks whereby stressors, genetic, environmental, and combinations thereof, alter molecular interactions and perturb the individual from the level of cells and tissues to the entire organism. Targeting stressor-induced dysfunctions in PPI networks has therefore become a promising but technically challenging frontier in therapeutics discovery. This opinion provides a new framework based upon disrupting epichaperomes - pathological entities that enable dysfunctional rewiring of PPI networks - as a mechanism to revert context-specific PPI network dysfunction to a normative state. We speculate on the implications of recent research in this area for a precision medicine approach to detecting and treating complex diseases, including cancer and neurodegenerative disorders.
Collapse
Affiliation(s)
- Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY 10962, USA; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY 10016, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sahil Sharma
- Program in Chemical Biology, Sloan Kettering Institute, New York, NY 10065, USA
| | - Larry Norton
- Breast Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Gabriela Chiosis
- Program in Chemical Biology, Sloan Kettering Institute, New York, NY 10065, USA; Breast Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
11
|
Halder S, Ghosh S, Chattopadhyay J, Chatterjee S. Understanding noise in cell signalling in the prospect of drug-targets. J Theor Biol 2022; 555:111298. [PMID: 36202233 DOI: 10.1016/j.jtbi.2022.111298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/04/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022]
Abstract
The introduction of noise to signals can alter central regulatory switches of cellular processes leading to diseases. Noise is inherently present in the cellular signalling system and plays a decisive role in the input-output (I/O) relation. The current study aims to understand the noise tolerance of motif structures in the cell signalling processes. The vulnerability of a node to noise could be a significant factor in causing signalling error and need to be controlled. We developed stochastic differential equation (SDE) based mathematical models for different network motifs with two nodes and studied the association between motif structure and signal-noise relation. A two-dimensional parameter space analysis on motif sensitivity with noise and input signal variation was performed to classify and rank the motifs. Identifying sensitive motifs and their high druggability infers their significance in screening potential drug-target candidates. Finally, we proposed a theoretical framework to identify nodes from a network as potential drug targets. We applied this mathematical formalism to three cancer networks to identify drug-targets and validated them with existing databases.
Collapse
Affiliation(s)
- Suvankar Halder
- Complex Analysis Group, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, India
| | - Sumana Ghosh
- Complex Analysis Group, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, India
| | - Joydev Chattopadhyay
- Agricultural and Ecological Research Unit, Indian Statistical Institute, 203 B.T. Road, Kolkata 700108, India
| | - Samrat Chatterjee
- Complex Analysis Group, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, India.
| |
Collapse
|
12
|
Sales de Queiroz A, Sales Santa Cruz G, Jean-Marie A, Mazauric D, Roux J, Cazals F. Gene prioritization based on random walks with restarts and absorbing states, to define gene sets regulating drug pharmacodynamics from single-cell analyses. PLoS One 2022; 17:e0268956. [PMID: 36342924 PMCID: PMC9639845 DOI: 10.1371/journal.pone.0268956] [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: 08/27/2021] [Accepted: 05/12/2022] [Indexed: 11/09/2022] Open
Abstract
Prioritizing genes for their role in drug sensitivity, is an important step in understanding drugs mechanisms of action and discovering new molecular targets for co-treatment. To formalize this problem, we consider two sets of genes X and P respectively composing the gene signature of cell sensitivity at the drug IC50 and the genes involved in its mechanism of action, as well as a protein interaction network (PPIN) containing the products of X and P as nodes. We introduce Genetrank, a method to prioritize the genes in X for their likelihood to regulate the genes in P. Genetrank uses asymmetric random walks with restarts, absorbing states, and a suitable renormalization scheme. Using novel so-called saturation indices, we show that the conjunction of absorbing states and renormalization yields an exploration of the PPIN which is much more progressive than that afforded by random walks with restarts only. Using MINT as underlying network, we apply Genetrank to a predictive gene signature of cancer cells sensitivity to tumor-necrosis-factor-related apoptosis-inducing ligand (TRAIL), performed in single-cells. Our ranking provides biological insights on drug sensitivity and a gene set considerably enriched in genes regulating TRAIL pharmacodynamics when compared to the most significant differentially expressed genes obtained from a statistical analysis framework alone. We also introduce gene expression radars, a visualization tool embedded in MA plots to assess all pairwise interactions at a glance on graphical representations of transcriptomics data. Genetrank is made available in the Structural Bioinformatics Library (https://sbl.inria.fr/doc/Genetrank-user-manual.html). It should prove useful for mining gene sets in conjunction with a signaling pathway, whenever other approaches yield relatively large sets of genes.
Collapse
Affiliation(s)
| | | | | | | | - Jérémie Roux
- CNRS UMR 7284, Inserm U 1081, Institut de Recherche sur le Cancer et le Vieillissement de Nice, Centre Antoine Lacassagne, Universite Côte d’Azur, Nice, France
- * E-mail: (FC); (JR)
| | - Frédéric Cazals
- Inria, Université Côte d’Azur, Nice, France
- * E-mail: (FC); (JR)
| |
Collapse
|
13
|
An integrated network representation of multiple cancer-specific data for graph-based machine learning. NPJ Syst Biol Appl 2022; 8:14. [PMID: 35487924 PMCID: PMC9054771 DOI: 10.1038/s41540-022-00226-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 04/04/2022] [Indexed: 12/20/2022] Open
Abstract
Genomic profiles of cancer cells provide valuable information on genetic alterations in cancer. Several recent studies employed these data to predict the response of cancer cell lines to drug treatment. Nonetheless, due to the multifactorial phenotypes and intricate mechanisms of cancer, the accurate prediction of the effect of pharmacotherapy on a specific cell line based on the genetic information alone is problematic. Emphasizing on the system-level complexity of cancer, we devised a procedure to integrate multiple heterogeneous data, including biological networks, genomics, inhibitor profiling, and gene-disease associations, into a unified graph structure. In order to construct compact, yet information-rich cancer-specific networks, we developed a novel graph reduction algorithm. Driven by not only the topological information, but also the biological knowledge, the graph reduction increases the feature-only entropy while preserving the valuable graph-feature information. Subsequent comparative benchmarking simulations employing a tissue level cross-validation protocol demonstrate that the accuracy of a graph-based predictor of the drug efficacy is 0.68, which is notably higher than those measured for more traditional, matrix-based techniques on the same data. Overall, the non-Euclidean representation of the cancer-specific data improves the performance of machine learning to predict the response of cancer to pharmacotherapy. The generated data are freely available to the academic community at https://osf.io/dzx7b/.
Collapse
|
14
|
Popescu VB, Kanhaiya K, Năstac DI, Czeizler E, Petre I. Network controllability solutions for computational drug repurposing using genetic algorithms. Sci Rep 2022; 12:1437. [PMID: 35082323 PMCID: PMC8791995 DOI: 10.1038/s41598-022-05335-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 12/29/2021] [Indexed: 12/22/2022] Open
Abstract
Control theory has seen recently impactful applications in network science, especially in connections with applications in network medicine. A key topic of research is that of finding minimal external interventions that offer control over the dynamics of a given network, a problem known as network controllability. We propose in this article a new solution for this problem based on genetic algorithms. We tailor our solution for applications in computational drug repurposing, seeking to maximize its use of FDA-approved drug targets in a given disease-specific protein-protein interaction network. We demonstrate our algorithm on several cancer networks and on several random networks with their edges distributed according to the Erdős-Rényi, the Scale-Free, and the Small World properties. Overall, we show that our new algorithm is more efficient in identifying relevant drug targets in a disease network, advancing the computational solutions needed for new therapeutic and drug repurposing approaches.
Collapse
Affiliation(s)
| | | | - Dumitru Iulian Năstac
- POLITEHNICA University of Bucharest, Faculty of Electronics, Telecommunications and Information Technology, 061071, Bucharest, Romania
| | - Eugen Czeizler
- Computer Science, Åbo Akademi University, 20500, Turku, Finland
- National Institute for Research and Development in Biological Sciences, 060031, Bucharest, Romania
| | - Ion Petre
- Department of Mathematics and Statistics, University of Turku, 20014, Turku, Finland.
- National Institute for Research and Development in Biological Sciences, 060031, Bucharest, Romania.
| |
Collapse
|
15
|
Joshi S, Gomes ED, Wang T, Corben A, Taldone T, Gandu S, Xu C, Sharma S, Buddaseth S, Yan P, Chan LYL, Gokce A, Rajasekhar VK, Shrestha L, Panchal P, Almodovar J, Digwal CS, Rodina A, Merugu S, Pillarsetty N, Miclea V, Peter RI, Wang W, Ginsberg SD, Tang L, Mattar M, de Stanchina E, Yu KH, Lowery M, Grbovic-Huezo O, O'Reilly EM, Janjigian Y, Healey JH, Jarnagin WR, Allen PJ, Sander C, Erdjument-Bromage H, Neubert TA, Leach SD, Chiosis G. Pharmacologically controlling protein-protein interactions through epichaperomes for therapeutic vulnerability in cancer. Commun Biol 2021; 4:1333. [PMID: 34824367 PMCID: PMC8617294 DOI: 10.1038/s42003-021-02842-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 11/03/2021] [Indexed: 12/03/2022] Open
Abstract
Cancer cell plasticity due to the dynamic architecture of interactome networks provides a vexing outlet for therapy evasion. Here, through chemical biology approaches for systems level exploration of protein connectivity changes applied to pancreatic cancer cell lines, patient biospecimens, and cell- and patient-derived xenografts in mice, we demonstrate interactomes can be re-engineered for vulnerability. By manipulating epichaperomes pharmacologically, we control and anticipate how thousands of proteins interact in real-time within tumours. Further, we can essentially force tumours into interactome hyperconnectivity and maximal protein-protein interaction capacity, a state whereby no rebound pathways can be deployed and where alternative signalling is supressed. This approach therefore primes interactomes to enhance vulnerability and improve treatment efficacy, enabling therapeutics with traditionally poor performance to become highly efficacious. These findings provide proof-of-principle for a paradigm to overcome drug resistance through pharmacologic manipulation of proteome-wide protein-protein interaction networks.
Collapse
Affiliation(s)
- Suhasini Joshi
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Erica DaGama Gomes
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Tai Wang
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Adriana Corben
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Tony Taldone
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Srinivasa Gandu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Chao Xu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Sahil Sharma
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Salma Buddaseth
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Pengrong Yan
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Lon Yin L Chan
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Askan Gokce
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Vinagolu K Rajasekhar
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Lisa Shrestha
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Palak Panchal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Justina Almodovar
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Chander S Digwal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Anna Rodina
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Swathi Merugu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | - Vlad Miclea
- Faculty of Automation and Computer Science, Technical University of Cluj-Napoca, Cluj-Napoca, CJ, 400114, Romania
| | - Radu I Peter
- Faculty of Automation and Computer Science, Technical University of Cluj-Napoca, Cluj-Napoca, CJ, 400114, Romania
| | - Wanyan Wang
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
- Departments of Psychiatry, Neuroscience & Physiology, and the NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Laura Tang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Marissa Mattar
- Antitumour Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Elisa de Stanchina
- Antitumour Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Kenneth H Yu
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Maeve Lowery
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Olivera Grbovic-Huezo
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Eileen M O'Reilly
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yelena Janjigian
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, 10065, USA
| | - John H Healey
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - William R Jarnagin
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Peter J Allen
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Chris Sander
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Hediye Erdjument-Bromage
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, NYU School of Medicine, New York, NY, 10016, USA
| | - Thomas A Neubert
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, NYU School of Medicine, New York, NY, 10016, USA
| | - Steven D Leach
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Dartmouth Geisel School of Medicine and Norris Cotton Cancer Center, Lebanon, NH, 03766, USA
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
16
|
Guo WF, Zhang SW, Zeng T, Akutsu T, Chen L. Network control principles for identifying personalized driver genes in cancer. Brief Bioinform 2021; 21:1641-1662. [PMID: 31711128 DOI: 10.1093/bib/bbz089] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 02/02/2023] Open
Abstract
To understand tumor heterogeneity in cancer, personalized driver genes (PDGs) need to be identified for unraveling the genotype-phenotype associations corresponding to particular patients. However, most of the existing driver-focus methods mainly pay attention on the cohort information rather than on individual information. Recent developing computational approaches based on network control principles are opening a new way to discover driver genes in cancer, particularly at an individual level. To provide comprehensive perspectives of network control methods on this timely topic, we first considered the cancer progression as a network control problem, in which the expected PDGs are altered genes by oncogene activation signals that can change the individual molecular network from one health state to the other disease state. Then, we reviewed the network reconstruction methods on single samples and introduced novel network control methods on single-sample networks to identify PDGs in cancer. Particularly, we gave a performance assessment of the network structure control-based PDGs identification methods on multiple cancer datasets from TCGA, for which the data and evaluation package also are publicly available. Finally, we discussed future directions for the application of network control methods to identify PDGs in cancer and diverse biological processes.
Collapse
Affiliation(s)
- Wei-Feng Guo
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| | - Tao Zeng
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tatsuya Akutsu
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji, 611-0011, Japan
| | - Luonan Chen
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China.,Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, 201210 Shanghai, China.,Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| |
Collapse
|
17
|
Popescu VB, Sanchez-Martinez JA, Schacherer D, Safadoust S, Majidi N, Andronescu A, Nedea A, Ion D, Mititelu E, Czeizler E, Petre I. NetControl4BioMed: A web-based platform for controllability analysis of protein-protein interaction networks. Bioinformatics 2021; 37:3976-3978. [PMID: 34352070 PMCID: PMC8570810 DOI: 10.1093/bioinformatics/btab570] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/12/2021] [Accepted: 08/03/2021] [Indexed: 12/23/2022] Open
Abstract
MOTIVATION There is an increasing amount of data coming from genome-wide studies identifying disease-specific survivability-essential proteins and host factors critical to a cell becoming infected. Targeting such proteins has a strong potential for targeted, precision therapies. Typically however, too few of them are drug targetable. An alternative approach is to influence them through drug targetable proteins upstream of them. Structural target network controllability is a suitable solution to this problem. It aims to discover suitable source nodes (e.g., drug targetable proteins) in a directed interaction network that can control (through a suitable set of input functions) a desired set of targets. RESULTS We introduce NetControl4BioMed, a free open-source web-based application that allows users to generate or upload directed protein-protein interaction networks and to perform target structural network controllability analyses on them. The analyses can be customized to focus the search on drug targetable source nodes, thus providing drug therapeutic suggestions. The application integrates protein data from HGNC, Ensemble, UniProt, NCBI, and InnateDB, directed interaction data from InnateDB, Omnipath, and SIGNOR, cell-line data from COLT and DepMap, and drug-target data from DrugBank. AVAILABILITY The application and data are available online at https://netcontrol.combio.org/. The source code is available at https://github.com/Vilksar/NetControl4BioMed under an MIT license.
Collapse
Affiliation(s)
| | | | | | | | - Negin Majidi
- University of California, Santa Cruz, United States
| | | | | | - Diana Ion
- University "Politehnica" of Bucharest, Romania
| | | | - Eugen Czeizler
- Åbo Akademi University, Turku, Finland.,National Institute for Research and Development for Biological Sciences, Bucharest, Romania
| | - Ion Petre
- University of Turku, Finland.,National Institute for Research and Development for Biological Sciences, Bucharest, Romania
| |
Collapse
|
18
|
Morgan S, Malatras A, Duguez S, Duddy W. Optimized Molecular Interaction Networks for the Study of Skeletal Muscle. J Neuromuscul Dis 2021; 8:S223-S239. [PMID: 34308911 DOI: 10.3233/jnd-210680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Molecular interaction networks (MINs) aim to capture the complex relationships between interacting molecules within a biological system. MINs can be constructed from existing knowledge of molecular functional associations, such as protein-protein binding interactions (PPI) or gene co-expression, and these different sources may be combined into a single MIN. A given MIN may be more or less optimal in its representation of the important functional relationships of molecules in a tissue. OBJECTIVE The aim of this study was to establish whether a combined MIN derived from different types of functional association could better capture muscle-relevant biology compared to its constituent single-source MINs. METHODS MINs were constructed from functional association databases for both protein-binding and gene co-expression. The networks were then compared based on the capture of muscle-relevant genes and gene ontology (GO) terms, tested in two different ways using established biological network clustering algorithms. The top performing MINs were combined to test whether an optimal MIN for skeletal muscle could be constructed. RESULTS The STRING PPI network was the best performing single-source MIN among those tested. Combining STRING with interactions from either the MyoMiner or CoXPRESSdb gene co-expression sources resulted in a combined network with improved performance relative to its constituent networks. CONCLUSION MINs constructed from multiple types of functional association can better represent the functional relationships of molecules in a given tissue. Such networks may be used to improve the analysis and interpretation of functional genomics data in the study of skeletal muscle and neuromuscular diseases. Networks and clusters described by this study, including the combinations of STRING with MyoMiner or with CoXPRESSdb, are available for download from https://www.sys-myo.com/myominer/download.php.
Collapse
Affiliation(s)
- Stephen Morgan
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry, Northern Ireland, UK
| | - Apostolos Malatras
- Department of Biological Sciences, Molecular Medicine Research Center, University of Cyprus, University Avenue, Nicosia, Cyprus
| | - Stephanie Duguez
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry, Northern Ireland, UK
| | - William Duddy
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry, Northern Ireland, UK
| |
Collapse
|
19
|
Halder S, Ghosh S, Chattopadhyay J, Chatterjee S. Bistability in cell signalling and its significance in identifying potential drug targets. Bioinformatics 2021; 37:4156-4163. [PMID: 34021761 DOI: 10.1093/bioinformatics/btab395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/09/2021] [Accepted: 05/20/2021] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION Bistability is one of the salient dynamical features in various all-or-none kinds of decision-making processes. The presence of bistability in a cell signalling network plays a key role in input-output (I/O) relation. Our study is aiming to capture and emphasise the role of motif structure influencing the I/O relation between two nodes in the context of bistability. Here, a model-based analysis is made to investigate the critical conditions responsible for the emergence of different bistable protein-protein interaction (PPI) motifs and their possible applications to find the potential drug targets. RESULTS The global sensitivity analysis is used to identify sensitive parameters and their role in maintaining the bistability. Additionally, the bistable switching through hysteresis is explored to develop an understanding of the underlying mechanisms involved in the cell signalling processes, when significant motifs exhibiting bistability have emerged. Further, we elaborate the application of the results by the implication of the emerged PPI motifs to identify potential drug-targets in three cancer networks, which is validated with existing databases. The influence of stochastic perturbations that could hinder desired functionality of any signalling networks is also described here. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Suvankar Halder
- Complex Analysis Group, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd milestone, Faridabad-Gurgaon Expressway, Faridabad-121001, India
| | - Sumana Ghosh
- Complex Analysis Group, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd milestone, Faridabad-Gurgaon Expressway, Faridabad-121001, India
| | - Joydev Chattopadhyay
- Agricultural and Ecological Research Unit, Indian Statistical Institute, 203 B.T. Road, Kolkata-700108, India
| | - Samrat Chatterjee
- Complex Analysis Group, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd milestone, Faridabad-Gurgaon Expressway, Faridabad-121001, India
| |
Collapse
|
20
|
Korie NPU, Tandoh KZ, Kwofie SK, Quaye O. Therapeutic potential of HIV-1 entry inhibitor peptidomimetics. Exp Biol Med (Maywood) 2021; 246:1060-1068. [PMID: 33596698 PMCID: PMC8113741 DOI: 10.1177/1535370221990870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human immunodeficiency virus 1 (HIV-1) infection remains a public health concern globally. Although great strides in the management of HIV-1 have been achieved, current highly active antiretroviral therapy is limited by multidrug resistance, prolonged use-related effects, and inability to purge the HIV-1 latent pool. Even though novel therapeutic options with HIV-1 broadly neutralizing antibodies (bNAbs) are being explored, the scalability of bNAbs is limited by economic cost of production and obligatory requirement for parenteral administration. However, these limitations can be addressed by antibody mimetics/peptidomimetics of HIV-1 bNAbs. In this review we discuss the limitations of HIV-1 bNAbs as HIV-1 entry inhibitors and explore the potential therapeutic use of antibody mimetics/peptidomimetics of HIV-1 entry inhibitors as an alternative for HIV-1 bNAbs. We highlight the reduced cost of production, high specificity, and oral bioavailability of peptidomimetics compared to bNAbs to demonstrate their suitability as candidates for novel HIV-1 therapy and conclude with some perspectives on future research toward HIV-1 novel drug discovery.
Collapse
Affiliation(s)
- Nneka PU Korie
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra 00233, Ghana
| | - Kwesi Z Tandoh
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra 00233, Ghana
| | - Samuel K Kwofie
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic and Applied Sciences, University of Ghana, Accra 00233, Ghana
| | - Osbourne Quaye
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra 00233, Ghana
| |
Collapse
|
21
|
Guo WF, Yu X, Shi QQ, Liang J, Zhang SW, Zeng T. Performance assessment of sample-specific network control methods for bulk and single-cell biological data analysis. PLoS Comput Biol 2021; 17:e1008962. [PMID: 33956788 PMCID: PMC8130943 DOI: 10.1371/journal.pcbi.1008962] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/18/2021] [Accepted: 04/12/2021] [Indexed: 11/29/2022] Open
Abstract
In the past few years, a wealth of sample-specific network construction methods and structural network control methods has been proposed to identify sample-specific driver nodes for supporting the Sample-Specific network Control (SSC) analysis of biological networked systems. However, there is no comprehensive evaluation for these state-of-the-art methods. Here, we conducted a performance assessment for 16 SSC analysis workflows by using the combination of 4 sample-specific network reconstruction methods and 4 representative structural control methods. This study includes simulation evaluation of representative biological networks, personalized driver genes prioritization on multiple cancer bulk expression datasets with matched patient samples from TCGA, and cell marker genes and key time point identification related to cell differentiation on single-cell RNA-seq datasets. By widely comparing analysis of existing SSC analysis workflows, we provided the following recommendations and banchmarking workflows. (i) The performance of a network control method is strongly dependent on the up-stream sample-specific network method, and Cell-Specific Network construction (CSN) method and Single-Sample Network (SSN) method are the preferred sample-specific network construction methods. (ii) After constructing the sample-specific networks, the undirected network-based control methods are more effective than the directed network-based control methods. In addition, these data and evaluation pipeline are freely available on https://github.com/WilfongGuo/Benchmark_control.
Collapse
Affiliation(s)
- Wei-Feng Guo
- School of Electrical Engineering, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xian, China
| | - Xiangtian Yu
- Clinical Research Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Qian-Qian Shi
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Jing Liang
- School of Electrical Engineering, Zhengzhou University, Zhengzhou, China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xian, China
| | - Tao Zeng
- CAS Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
22
|
Mun J, Choi G, Lim B. A guide for bioinformaticians: 'omics-based drug discovery for precision oncology. Drug Discov Today 2020; 25:S1359-6446(20)30335-4. [PMID: 32828947 DOI: 10.1016/j.drudis.2020.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/19/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
Abstract
Bioinformatics-centric drug development is inevitable in the era of precision medicine. Clinical 'omics information, including genomics, epigenomics, transcriptomics, and proteomics, provides the most comprehensive molecular landscape in which each patient's pathological history is delineated. Hence, the capability of bioinformaticians to manage integrative 'omics data is crucial to current drug development. Bioinformatics can accelerate drug development from initial time-consuming discoveries to the clinical stage by providing information-guided solutions. However, many bioinformaticians do not have opportunities to participate in drug discovery programs. As a starting point for bioinformaticians with no prior drug development experience, here we discuss bioinformatics applications during drug development with a focus on working-level omics-based methodologies.
Collapse
Affiliation(s)
- Jihyeob Mun
- Center for Supercomputing Applications, Division of National Supercomputing R&D, Korea Institute of Science and Technology Information (KISTI), Daejeon, Republic of Korea
| | - Gildon Choi
- Research Center for Drug Discovery Technology, Division of Drug Discovery Research, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea.
| | - Byungho Lim
- Research Center for Drug Discovery Technology, Division of Drug Discovery Research, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea.
| |
Collapse
|
23
|
Kanhaiya K, Tyagi-Tiwari D. Identification of Drug Targets in Breast Cancer Metabolic Network. J Comput Biol 2020; 27:975-986. [DOI: 10.1089/cmb.2019.0258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Krishna Kanhaiya
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, Turku, Finland
- Department of Computer Science, Åbo Akademi University, Turku, Finland
| | - Dwitiya Tyagi-Tiwari
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, Turku, Finland
- Department of Computer Science, Åbo Akademi University, Turku, Finland
| |
Collapse
|
24
|
Singha M, Pu L, Shawky A, Busch K, Wu H, Ramanujam J, Brylinski M. GraphGR: A graph neural network to predict the effect of pharmacotherapy on the cancer cell growth.. [DOI: 10.1101/2020.05.20.107458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
AbstractGenomic profiles of cancer cells provide valuable information on genetic alterations in cancer. Several recent studies employed these data to predict the response of cancer cell lines to treatment with drugs. Nonetheless, due to the multifactorial phenotypes and intricate mechanisms of cancer, the accurate prediction of the effect of pharmacotherapy on a specific cell line based on the genetic information alone is problematic. High prediction accuracies reported in the literature likely result from significant overlaps among training, validation, and testing sets, making many predictors inapplicable to new data. To address these issues, we developed GraphGR, a graph neural network with sophisticated attention propagation mechanisms to predict the therapeutic effects of kinase inhibitors across various tumors. Emphasizing on the system-level complexity of cancer, GraphGR integrates multiple heterogeneous data, such as biological networks, genomics, inhibitor profiling, and genedisease associations, into a unified graph structure. In order to construct diverse and information-rich cancer-specific networks, we devised a novel graph reduction protocol based on not only the topological information, but also the biological knowledge. The performance of GraphGR, properly cross-validated at the tissue level, is 0.83 in terms of the area under the receiver operating characteristics, which is notably higher than those measured for other approaches on the same data. Finally, several new predictions are validated against the biomedical literature demonstrating that GraphGR generalizes well to unseen data, i.e. it can predict therapeutic effects across a variety of cancer cell lines and inhibitors. GraphGR is freely available to the academic community at https://github.com/pulimeng/GraphGR.
Collapse
|
25
|
Paananen J, Fortino V. An omics perspective on drug target discovery platforms. Brief Bioinform 2019; 21:1937-1953. [PMID: 31774113 PMCID: PMC7711264 DOI: 10.1093/bib/bbz122] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/23/2019] [Accepted: 07/27/2019] [Indexed: 01/28/2023] Open
Abstract
The drug discovery process starts with identification of a disease-modifying target. This critical step traditionally begins with manual investigation of scientific literature and biomedical databases to gather evidence linking molecular target to disease, and to evaluate the efficacy, safety and commercial potential of the target. The high-throughput and affordability of current omics technologies, allowing quantitative measurements of many putative targets (e.g. DNA, RNA, protein, metabolite), has exponentially increased the volume of scientific data available for this arduous task. Therefore, computational platforms identifying and ranking disease-relevant targets from existing biomedical data sources, including omics databases, are needed. To date, more than 30 drug target discovery (DTD) platforms exist. They provide information-rich databases and graphical user interfaces to help scientists identify putative targets and pre-evaluate their therapeutic efficacy and potential side effects. Here we survey and compare a set of popular DTD platforms that utilize multiple data sources and omics-driven knowledge bases (either directly or indirectly) for identifying drug targets. We also provide a description of omics technologies and related data repositories which are important for DTD tasks.
Collapse
Affiliation(s)
- Jussi Paananen
- Institute of Biomedicine, University of Eastern Finland, Finland.,Blueprint Genetics Ltd, Finland
| | - Vittorio Fortino
- Institute of Biomedicine, University of Eastern Finland, Finland
| |
Collapse
|
26
|
Vandana, Dixit R, Tiwari R, Katyal A, Pandey KC. Metacaspases: Potential Drug Target Against Protozoan Parasites. Front Pharmacol 2019; 10:790. [PMID: 31379569 PMCID: PMC6657590 DOI: 10.3389/fphar.2019.00790] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/18/2019] [Indexed: 02/05/2023] Open
Abstract
Among the numerous strategies/targets for controlling infectious diseases, parasites-derived proteases receive prime attention due to their essential contribution to parasite growth and development. Parasites produce a broad array of proteases, which are required for parasite entry/invasion, modification/degradation of host proteins for their nourishment, and activation of inflammation that ensures their survival to maintain infection. Presently, extensive research is focused on unique proteases termed as "metacaspases" (MCAs) in relation to their versatile functions in plants and non-metazoans. Such unique MCAs proteases could be considered as a potential drug target against parasites due to their absence in the human host. MCAs are cysteine proteases, having Cys-His catalytic dyad present in fungi, protozoa, and plants. Studies so far indicated that MCAs are broadly associated with apoptosis-like cell death, growth, and stress regulation in different protozoa. The present review comprises the important research outcomes from our group and published literature, showing the variable properties and function of MCAs for therapeutic purpose against infectious diseases.
Collapse
Affiliation(s)
- Vandana
- Host-Parasite Interaction Biology Group, ICMR-National Institute of Malaria Research, New Delhi, India.,Dr Ambedkar Center for Biomedical Research, Delhi University, New Delhi, India
| | - Rajnikant Dixit
- Host-Parasite Interaction Biology Group, ICMR-National Institute of Malaria Research, New Delhi, India
| | - Rajnarayan Tiwari
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Anju Katyal
- Dr Ambedkar Center for Biomedical Research, Delhi University, New Delhi, India
| | - Kailash C Pandey
- Host-Parasite Interaction Biology Group, ICMR-National Institute of Malaria Research, New Delhi, India.,Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| |
Collapse
|
27
|
Shahare HV, Talele GS. Designing of benzothiazole derivatives as promising EGFR tyrosine kinase inhibitors: a pharmacoinformatics study. J Biomol Struct Dyn 2019; 38:1365-1374. [DOI: 10.1080/07391102.2019.1604264] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Hitesh V. Shahare
- Department of Chemistry, SNJBs Shriman Sureshdada Jain College of Pharmacy, Chandwad, Nasik, Maharashtra, India
| | - Gokul S. Talele
- Department of Chemistry, SNJBs Shriman Sureshdada Jain College of Pharmacy, Chandwad, Nasik, Maharashtra, India
- NGSPM College of Pharmacy, Brahmavalley Educational Campus, Anjaneri, Nashik, Maharashtra, India
| |
Collapse
|
28
|
Lee LYH, Loscalzo J. Network Medicine in Pathobiology. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1311-1326. [PMID: 31014954 DOI: 10.1016/j.ajpath.2019.03.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/05/2019] [Indexed: 12/11/2022]
Abstract
The past decade has witnessed exponential growth in the generation of high-throughput human data across almost all known dimensions of biological systems. The discipline of network medicine has rapidly evolved in parallel, providing an unbiased, comprehensive biological framework through which to interrogate and integrate systematically these large-scale, multi-omic data to enhance our understanding of disease mechanisms and to design drugs that reflect a deep knowledge of molecular pathobiology. In this review, we discuss the key principles of network medicine and the human disease network and explore the latest applications of network medicine in this multi-omic era. We also highlight the current conceptual and technological challenges, which serve as exciting opportunities by which to improve and expand the network-based applications beyond the artificial boundaries of the current state of human pathobiology.
Collapse
Affiliation(s)
| | - Joseph Loscalzo
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
29
|
Latysheva NS, Babu MM. Molecular Signatures of Fusion Proteins in Cancer. ACS Pharmacol Transl Sci 2019; 2:122-133. [PMID: 32219217 PMCID: PMC7088938 DOI: 10.1021/acsptsci.9b00019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Indexed: 01/07/2023]
Abstract
![]()
Although gene fusions
are recognized as driver mutations in a wide
variety of cancers, the general molecular mechanisms underlying oncogenic
fusion proteins are insufficiently understood. Here, we employ large-scale
data integration and machine learning and (1) identify three functionally
distinct subgroups of gene fusions and their molecular signatures;
(2) characterize the cellular pathways rewired by fusion events across
different cancers; and (3) analyze the relative importance of over
100 structural, functional, and regulatory features of ∼2200
gene fusions. We report subgroups of fusions that likely act as driver
mutations and find that gene fusions disproportionately affect pathways
regulating cellular shape and movement. Although fusion proteins are
similar across different cancer types, they affect cancer type-specific
pathways. Key indicators of fusion-forming proteins include high and
nontissue specific expression, numerous splice sites, and higher centrality
in protein-interaction networks. Together, these findings provide
unifying and cancer type-specific trends across diverse oncogenic
fusion proteins.
Collapse
Affiliation(s)
- Natasha S Latysheva
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, United Kingdom
| | - M Madan Babu
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, United Kingdom
| |
Collapse
|
30
|
Liu X, Hong Z, Liu J, Lin Y, Rodríguez-Patón A, Zou Q, Zeng X. Computational methods for identifying the critical nodes in biological networks. Brief Bioinform 2019; 21:486-497. [DOI: 10.1093/bib/bbz011] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/03/2018] [Accepted: 01/11/2019] [Indexed: 12/28/2022] Open
Abstract
Abstract
A biological network is complex. A group of critical nodes determines the quality and state of such a network. Increasing studies have shown that diseases and biological networks are closely and mutually related and that certain diseases are often caused by errors occurring in certain nodes in biological networks. Thus, studying biological networks and identifying critical nodes can help determine the key targets in treating diseases. The problem is how to find the critical nodes in a network efficiently and with low cost. Existing experimental methods in identifying critical nodes generally require much time, manpower and money. Accordingly, many scientists are attempting to solve this problem by researching efficient and low-cost computing methods. To facilitate calculations, biological networks are often modeled as several common networks. In this review, we classify biological networks according to the network types used by several kinds of common computational methods and introduce the computational methods used by each type of network.
Collapse
Affiliation(s)
- Xiangrong Liu
- Department of Computer Science, Xiamen University, China
| | - Zengyan Hong
- Department of Computer Science, Xiamen University, China
| | - Juan Liu
- Department of Computer Science, Xiamen University, China
| | - Yuan Lin
- ITOP Section, DNB Bank ASA, Solheimsgaten, Bergen, Norway
| | - Alfonso Rodríguez-Patón
- Universidad Politécnica de Madrid (UPM) Campus Montegancedo s/n, Boadilla del Monte, Madrid, Spain
| | - Quan Zou
- Department of Computer Science, Xiamen University, China
- Insitute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, China
- School of Computer Science and Technology, Tianjin University, Tianjin, China
| | | |
Collapse
|
31
|
Zhang M, Zhang M, Wang J, Cai Q, Zhao R, Yu Y, Tai H, Zhang X, Xu C. Retro-inverso follicle-stimulating hormone peptide-mediated polyethylenimine complexes for targeted ovarian cancer gene therapy. Drug Deliv 2018; 25:995-1003. [PMID: 29667478 PMCID: PMC6058519 DOI: 10.1080/10717544.2018.1461956] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 04/03/2018] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND The development of nanoparticle drug delivery systems with targeted ligands has the potential to increase treatment efficiency in ovarian cancer. METHODS We developed a 21-amino acid peptide, YTRDLVYGDPARPGIQGTGTF (L-FP21) conjugated to polyethylenimine (PEI) and methoxy polyethylene glycol (mPEG) to prepare a nanoparticle drug vehicle to target follicle-stimulating hormone receptor (FSHR) in ovarian cancer. At the same time, we optimized the ligand of the nanoparticle vehicle using D-peptides, which consist of D-amino acids (D-FP21). Nanoparticle vehicles carrying the therapeutic gene plasmid growth-regulated oncogene alpha (pGRO-α) short hairpin RNA (shRNA) (FP21-PEG-PEI/pGRO-α) were prepared for further investigation. RESULTS Compared with L-FP21, D-FP21 exhibited improved biological stability and higher uptake rate for FSHR-expressing ovarian cancer cells. The cytotoxicity of the L, D-FP21-PEG-PEI/pGRO-α complexes were significantly lower than that of the PEI/pGRO-α complex. The nanoparticle drug with the targeted ligand showed higher transfection efficiencies and improved anti-proliferation effects for ovarian cancer cells than that without the targeted ligand (mPEG-PEI/pGRO-α). Furthermore, an in vivo evaluation of an antitumor assay indicated that D-FP21-PEG-PEI/pGRO-α inhibited the growth of tumor spheroids considerably more than L-FP21-PEG-PEI/pGRO-α; their tumor inhibition rates were 58.5% and 33.3%, respectively. CONCLUSIONS D-FP21-PEG-PEI/plasmid DNA is a safe and efficient gene delivery vehicle for ovarian cancer targeted therapy.
Collapse
Affiliation(s)
- Mengyu Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Department of Gynecology, 411 Military Hospital Affiliated to Changhai Hospital of Shanghai, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Mingxing Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Jing Wang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Qingqing Cai
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Ran Zhao
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yi Yu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Haiyan Tai
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xiaoyan Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Congjian Xu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| |
Collapse
|
32
|
Li M, Gao H, Wang J, Wu FX. Control principles for complex biological networks. Brief Bioinform 2018; 20:2253-2266. [DOI: 10.1093/bib/bby088] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/16/2018] [Accepted: 08/18/2018] [Indexed: 11/13/2022] Open
Abstract
Abstract
Networks have been widely used to model the structure of various biological systems. Currently, a series of approaches have been developed to construct reliable biological networks. However, the ultimate understanding of a biological system is to steer its states to the desired ones by imposing signals. The control process is dominated by the intrinsic structure and the dynamic propagation. To understand the underlying mechanisms behind the life process, the control theory can be applied to biological networks with specific target requirements. In this article, we first introduce the structural controllability of complex networks and discuss its advantages and disadvantages. Then, we review the effective control to meet the specific requirements for complex biological networks. Moreover, we summarize the existing methods for finding the unique minimum set of driver nodes via the optimal control for complex networks. Finally, we discuss the relationships between biological networks and structural controllability, effective control and optimal control. Moreover, potential applications of general control principles are pointed out.
Collapse
Affiliation(s)
- Min Li
- School of Information Science and Engineering, Central South University, Changsha, Hunan, China
| | - Hao Gao
- School of Information Science and Engineering, Central South University, Changsha, Hunan, China
| | - Jianxin Wang
- School of Information Science and Engineering, Central South University, Changsha, Hunan, China
| | - Fang-Xiang Wu
- School of Information Science and Engineering, Central South University, Changsha, Hunan, China
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, Canada
- Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
33
|
Pyatnitskiy MA, Karpov DS, Moshkovskii SA. [Searching for essential genes in cancer genomes]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2018; 64:303-314. [PMID: 30135277 DOI: 10.18097/pbmc20186404303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The concept of essential genes, whose loss of functionality leads to cell death, is one of the fundamental concepts of genetics and is important for fundamental and applied research. This field is particularly promising in relation to oncology, since the search for genetic vulnerabilities of cancer cells allows us to identify new potential targets for antitumor therapy. The modern biotechnology capacities allow carrying out large-scale projects for sequencing somatic mutations in tumors, as well as directly interfering the genetic apparatus of cancer cells. They provided accumulation of a considerable body of knowledge about genetic variants and corresponding phenotypic manifestations in tumors. In the near future this knowledge will find application in clinical practice. This review describes the main experimental and computational approaches to the search for essential genes, concentrating on the application of these methods in the field of molecular oncology.
Collapse
Affiliation(s)
- M A Pyatnitskiy
- Institute of Biomedical Chemistry, Moscow, Russia; Higher School of Economics, Moscow, Russia
| | - D S Karpov
- Institute of Biomedical Chemistry, Moscow, Russia; Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - S A Moshkovskii
- Institute of Biomedical Chemistry, Moscow, Russia; Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
34
|
Macalino SJY, Basith S, Clavio NAB, Chang H, Kang S, Choi S. Evolution of In Silico Strategies for Protein-Protein Interaction Drug Discovery. Molecules 2018; 23:E1963. [PMID: 30082644 PMCID: PMC6222862 DOI: 10.3390/molecules23081963] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/03/2018] [Accepted: 08/04/2018] [Indexed: 12/14/2022] Open
Abstract
The advent of advanced molecular modeling software, big data analytics, and high-speed processing units has led to the exponential evolution of modern drug discovery and better insights into complex biological processes and disease networks. This has progressively steered current research interests to understanding protein-protein interaction (PPI) systems that are related to a number of relevant diseases, such as cancer, neurological illnesses, metabolic disorders, etc. However, targeting PPIs are challenging due to their "undruggable" binding interfaces. In this review, we focus on the current obstacles that impede PPI drug discovery, and how recent discoveries and advances in in silico approaches can alleviate these barriers to expedite the search for potential leads, as shown in several exemplary studies. We will also discuss about currently available information on PPI compounds and systems, along with their usefulness in molecular modeling. Finally, we conclude by presenting the limits of in silico application in drug discovery and offer a perspective in the field of computer-aided PPI drug discovery.
Collapse
Affiliation(s)
- Stephani Joy Y Macalino
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea.
| | - Shaherin Basith
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea.
| | - Nina Abigail B Clavio
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea.
| | - Hyerim Chang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea.
| | - Soosung Kang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea.
| | - Sun Choi
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea.
| |
Collapse
|
35
|
Bidkhori G, Benfeitas R, Elmas E, Kararoudi MN, Arif M, Uhlen M, Nielsen J, Mardinoglu A. Metabolic Network-Based Identification and Prioritization of Anticancer Targets Based on Expression Data in Hepatocellular Carcinoma. Front Physiol 2018; 9:916. [PMID: 30065658 PMCID: PMC6056771 DOI: 10.3389/fphys.2018.00916] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/22/2018] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a deadly form of liver cancer with high mortality worldwide. Unfortunately, the large heterogeneity of this disease makes it difficult to develop effective treatment strategies. Cellular network analyses have been employed to study heterogeneity in cancer, and to identify potential therapeutic targets. However, the existing approaches do not consider metabolic growth requirements, i.e., biological network functionality, to rank candidate targets while preventing toxicity to non-cancerous tissues. Here, we developed an algorithm to overcome these issues based on integration of gene expression data, genome-scale metabolic models, network controllability, and dispensability, as well as toxicity analysis. This method thus predicts and ranks potential anticancer non-toxic controlling metabolite and gene targets. Our algorithm encompasses both objective-driven and-independent tasks, and uses network topology to finally rank the predicted therapeutic targets. We employed this algorithm to the analysis of transcriptomic data for 50 HCC patients with both cancerous and non-cancerous samples. We identified several potential targets that would prevent cell growth, including 74 anticancer metabolites, and 3 gene targets (PRKACA, PGS1, and CRLS1). The predicted anticancer metabolites showed good agreement with existing FDA-approved cancer drugs, and the 3 genes were experimentally validated by performing experiments in HepG2 and Hep3B liver cancer cell lines. Our observations indicate that our novel approach successfully identifies therapeutic targets for effective treatment of cancer. This approach may also be applied to any cancer type that has tumor and non-tumor gene or protein expression data.
Collapse
Affiliation(s)
- Gholamreza Bidkhori
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Rui Benfeitas
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Ezgi Elmas
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | | | - Muhammad Arif
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden.,Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
36
|
Kanhaiya K, Rogojin V, Kazemi K, Czeizler E, Petre I. NetControl4BioMed: a pipeline for biomedical data acquisition and analysis of network controllability. BMC Bioinformatics 2018; 19:185. [PMID: 30066633 PMCID: PMC6069765 DOI: 10.1186/s12859-018-2177-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Network controllability focuses on discovering combinations of external interventions that can drive a biological system to a desired configuration. In practice, this approach translates into finding a combined multi-drug therapy in order to induce a desired response from a cell; this can lead to developments of novel therapeutic approaches for systemic diseases like cancer. RESULT We develop a novel bioinformatics data analysis pipeline called NetControl4BioMed based on the concept of target structural control of linear networks. Our pipeline generates novel molecular interaction networks by combining pathway data from various public databases starting from the user's query. The pipeline then identifies a set of nodes that is enough to control a given, user-defined set of disease-specific essential proteins in the network, i.e., it is able to induce a change in their configuration from any initial state to any final state. We provide both the source code of the pipeline as well as an online web-service based on this pipeline http://combio.abo.fi/nc/net_control/remote_call.php . CONCLUSION The pipeline can be used by researchers for controlling and better understanding of molecular interaction networks through combinatorial multi-drug therapies, for more efficient therapeutic approaches and personalised medicine.
Collapse
Affiliation(s)
- Krishna Kanhaiya
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Å bo Akademi University, Domkyrkotorget 3, Turku, 20500 Finland
| | - Vladimir Rogojin
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Å bo Akademi University, Domkyrkotorget 3, Turku, 20500 Finland
| | - Keivan Kazemi
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Å bo Akademi University, Domkyrkotorget 3, Turku, 20500 Finland
| | - Eugen Czeizler
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Å bo Akademi University, Domkyrkotorget 3, Turku, 20500 Finland
- National Institute for Research and Development for Biological Sciences, Splaiul Independentei 296, Bucharest, 060031 Romania
| | - Ion Petre
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Å bo Akademi University, Domkyrkotorget 3, Turku, 20500 Finland
- National Institute for Research and Development for Biological Sciences, Splaiul Independentei 296, Bucharest, 060031 Romania
| |
Collapse
|
37
|
Czeizler E, Wu KC, Gratie C, Kanhaiya K, Petre I. Structural Target Controllability of Linear Networks. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 15:1217-1228. [PMID: 29994605 DOI: 10.1109/tcbb.2018.2797271] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Computational analysis of the structure of intra-cellular molecular interaction networks can suggest novel therapeutic approaches for systemic diseases like cancer. Recent research in the area of network science has shown that network control theory can be a powerful tool in the understanding and manipulation of such bio-medical networks. In 2011, Liu et al. developed a polynomial time algorithm computing the size of the minimal set of nodes controlling a linear network. In 2014, Gao et al. generalized the problem for target control, minimizing the set of nodes controlling a target within a linear network. The authors developed a Greedy approximation algorithm while leaving open the complexity of the optimization problem. We prove here that the target controllability problem is NP-hard in all practical setups, i.e., when the control power of any individual input is bounded by some constant. We also show that the algorithm provided by Gao et al. fails to provide a valid solution in some special cases, and an additional validation step is required. We fix and improve their algorithm using several heuristics, obtaining in the end an up to 10-fold decrease in running time and also a decrease in the size of solutions.
Collapse
|
38
|
Amgalan B, Tseveendorj I, Lee H. An integrative model for the identification of key players of cancer networks. APPLIED MATHEMATICAL MODELLING 2018; 58:65-75. [DOI: 10.1016/j.apm.2017.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
39
|
Guo WF, Zhang SW, Shi QQ, Zhang CM, Zeng T, Chen L. A novel algorithm for finding optimal driver nodes to target control complex networks and its applications for drug targets identification. BMC Genomics 2018; 19:924. [PMID: 29363426 PMCID: PMC5780855 DOI: 10.1186/s12864-017-4332-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background The advances in target control of complex networks not only can offer new insights into the general control dynamics of complex systems, but also be useful for the practical application in systems biology, such as discovering new therapeutic targets for disease intervention. In many cases, e.g. drug target identification in biological networks, we usually require a target control on a subset of nodes (i.e., disease-associated genes) with minimum cost, and we further expect that more driver nodes consistent with a certain well-selected network nodes (i.e., prior-known drug-target genes). Results Therefore, motivated by this fact, we pose and address a new and practical problem called as target control problem with objectives-guided optimization (TCO): how could we control the interested variables (or targets) of a system with the optional driver nodes by minimizing the total quantity of drivers and meantime maximizing the quantity of constrained nodes among those drivers. Here, we design an efficient algorithm (TCOA) to find the optional driver nodes for controlling targets in complex networks. We apply our TCOA to several real-world networks, and the results support that our TCOA can identify more precise driver nodes than the existing control-fucus approaches. Furthermore, we have applied TCOA to two bimolecular expert-curate networks. Source code for our TCOA is freely available from http://sysbio.sibcb.ac.cn/cb/chenlab/software.htm or https://github.com/WilfongGuo/guoweifeng. Conclusions In the previous theoretical research for the full control, there exists an observation and conclusion that the driver nodes tend to be low-degree nodes. However, for target control the biological networks, we find interestingly that the driver nodes tend to be high-degree nodes, which is more consistent with the biological experimental observations. Furthermore, our results supply the novel insights into how we can efficiently target control a complex system, and especially many evidences on the practical strategic utility of TCOA to incorporate prior drug information into potential drug-target forecasts. Thus applicably, our method paves a novel and efficient way to identify the drug targets for leading the phenotype transitions of underlying biological networks. Electronic supplementary material The online version of this article (doi: 10.1186/s12864-017-4332-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei-Feng Guo
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Qian-Qian Shi
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, University of Chinese Academy of sciences, Shanghai, 200000, China
| | - Cheng-Ming Zhang
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, University of Chinese Academy of sciences, Shanghai, 200000, China
| | - Tao Zeng
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, University of Chinese Academy of sciences, Shanghai, 200000, China.
| | - Luonan Chen
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an, 710072, China. .,Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, University of Chinese Academy of sciences, Shanghai, 200000, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, 200000, China.
| |
Collapse
|