51
|
Dehghan A, Abbasi K, Razzaghi P, Banadkuki H, Gharaghani S. CCL-DTI: contributing the contrastive loss in drug-target interaction prediction. BMC Bioinformatics 2024; 25:48. [PMID: 38291364 PMCID: PMC11264960 DOI: 10.1186/s12859-024-05671-3] [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: 09/13/2023] [Accepted: 01/22/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND The Drug-Target Interaction (DTI) prediction uses a drug molecule and a protein sequence as inputs to predict the binding affinity value. In recent years, deep learning-based models have gotten more attention. These methods have two modules: the feature extraction module and the task prediction module. In most deep learning-based approaches, a simple task prediction loss (i.e., categorical cross entropy for the classification task and mean squared error for the regression task) is used to learn the model. In machine learning, contrastive-based loss functions are developed to learn more discriminative feature space. In a deep learning-based model, extracting more discriminative feature space leads to performance improvement for the task prediction module. RESULTS In this paper, we have used multimodal knowledge as input and proposed an attention-based fusion technique to combine this knowledge. Also, we investigate how utilizing contrastive loss function along the task prediction loss could help the approach to learn a more powerful model. Four contrastive loss functions are considered: (1) max-margin contrastive loss function, (2) triplet loss function, (3) Multi-class N-pair Loss Objective, and (4) NT-Xent loss function. The proposed model is evaluated using four well-known datasets: Wang et al. dataset, Luo's dataset, Davis, and KIBA datasets. CONCLUSIONS Accordingly, after reviewing the state-of-the-art methods, we developed a multimodal feature extraction network by combining protein sequences and drug molecules, along with protein-protein interaction networks and drug-drug interaction networks. The results show it performs significantly better than the comparable state-of-the-art approaches.
Collapse
Affiliation(s)
- Alireza Dehghan
- Department of Bioinformatics, Kish International Campus, University of Tehran, Kish, 1417614411, Iran
| | - Karim Abbasi
- Laboratory of System Biology, Bioinformatics and Artificial Intelligence in Medicine (LBB&AI), Faculty of Mathematics and Computer Science, Kharazmi University, Tehran, 1417614411, Iran
| | - Parvin Razzaghi
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 4513766731, Iran.
| | - Hossein Banadkuki
- Laboratory of Bioinformatics and Drug Design (LBD), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, 1417614411, Iran
| | - Sajjad Gharaghani
- Laboratory of Bioinformatics and Drug Design (LBD), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, 1417614411, Iran.
| |
Collapse
|
52
|
Duman ET, Tuna G, Ak E, Avsar G, Pir P. Optimized network based natural language processing approach to reveal disease comorbidities in COVID-19. Sci Rep 2024; 14:2325. [PMID: 38282038 PMCID: PMC10822845 DOI: 10.1038/s41598-024-52819-5] [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: 03/23/2023] [Accepted: 01/24/2024] [Indexed: 01/30/2024] Open
Abstract
A novel virus emerged from Wuhan, China, at the end of 2019 and quickly evolved into a pandemic, significantly impacting various industries, especially healthcare. One critical lesson from COVID-19 is the importance of understanding and predicting underlying comorbidities to better prioritize care and pharmacological therapies. Factors like age, race, and comorbidity history are crucial in determining disease mortality. While clinical data from hospitals and cohorts have led to the identification of these comorbidities, traditional approaches often lack a mechanistic understanding of the connections between them. In response, we utilized a deep learning approach to integrate COVID-19 data with data from other diseases, aiming to detect comorbidities with mechanistic insights. Our modified algorithm in the mpDisNet package, based on word-embedding deep learning techniques, incorporates miRNA expression profiles from SARS-CoV-2 infected cell lines and their target transcription factors. This approach is aligned with the emerging field of network medicine, which seeks to define diseases based on distinct pathomechanisms rather than just phenotypes. The main aim is discovery of possible unknown comorbidities by connecting the diseases by their miRNA mediated regulatory interactions. The algorithm can predict the majority of COVID-19's known comorbidities, as well as several diseases that have yet to be discovered to be comorbid with COVID-19. These potentially comorbid diseases should be investigated further to raise awareness and prevention, as well as informing the comorbidity research for the next possible outbreak.
Collapse
Affiliation(s)
- Emre Taylan Duman
- Department of Bioengineering, Gebze Technical University, Kocaeli, Turkey.
- NGS-Core Unit for Integrative Genomics, Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany.
| | - Gizem Tuna
- Department of Molecular Biology, Gebze Technical University, Kocaeli, Turkey
| | - Enes Ak
- Department of Bioengineering, Gebze Technical University, Kocaeli, Turkey
| | - Gülben Avsar
- Department of Bioengineering, Gebze Technical University, Kocaeli, Turkey
| | - Pinar Pir
- Department of Bioengineering, Gebze Technical University, Kocaeli, Turkey
| |
Collapse
|
53
|
Kong X, Diao L, Jiang P, Nie S, Guo S, Li D. DDK-Linker: a network-based strategy identifies disease signals by linking high-throughput omics datasets to disease knowledge. Brief Bioinform 2024; 25:bbae111. [PMID: 38517698 PMCID: PMC10959161 DOI: 10.1093/bib/bbae111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/24/2024] Open
Abstract
The high-throughput genomic and proteomic scanning approaches allow investigators to measure the quantification of genome-wide genes (or gene products) for certain disease conditions, which plays an essential role in promoting the discovery of disease mechanisms. The high-throughput approaches often generate a large gene list of interest (GOIs), such as differentially expressed genes/proteins. However, researchers have to perform manual triage and validation to explore the most promising, biologically plausible linkages between the known disease genes and GOIs (disease signals) for further study. Here, to address this challenge, we proposed a network-based strategy DDK-Linker to facilitate the exploration of disease signals hidden in omics data by linking GOIs to disease knowns genes. Specifically, it reconstructed gene distances in the protein-protein interaction (PPI) network through six network methods (random walk with restart, Deepwalk, Node2Vec, LINE, HOPE, Laplacian) to discover disease signals in omics data that have shorter distances to disease genes. Furthermore, benefiting from the establishment of knowledge base we established, the abundant bioinformatics annotations were provided for each candidate disease signal. To assist in omics data interpretation and facilitate the usage, we have developed this strategy into an application that users can access through a website or download the R package. We believe DDK-Linker will accelerate the exploring of disease genes and drug targets in a variety of omics data, such as genomics, transcriptomics and proteomics data, and provide clues for complex disease mechanism and pharmacological research. DDK-Linker is freely accessible at http://ddklinker.ncpsb.org.cn/.
Collapse
Affiliation(s)
- Xiangren Kong
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Lihong Diao
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Peng Jiang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Shiyan Nie
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Shuzhen Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Dong Li
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| |
Collapse
|
54
|
Hu C, Mi W, Li F, Zhu L, Ou Q, Li M, Li T, Ma Y, Zhang Y, Xu Y. Optimizing drug combination and mechanism analysis based on risk pathway crosstalk in pan cancer. Sci Data 2024; 11:74. [PMID: 38228620 PMCID: PMC10791624 DOI: 10.1038/s41597-024-02915-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 01/03/2024] [Indexed: 01/18/2024] Open
Abstract
Combination therapy can greatly improve the efficacy of cancer treatment, so identifying the most effective drug combination and interaction can accelerate the development of combination therapy. Here we developed a computational network biological approach to identify the effective drug which inhibition risk pathway crosstalk of cancer, and then filtrated and optimized the drug combination for cancer treatment. We integrated high-throughput data concerning pan-cancer and drugs to construct miRNA-mediated crosstalk networks among cancer pathways and further construct networks for therapeutic drug. Screening by drug combination method, we obtained 687 optimized drug combinations of 83 first-line anticancer drugs in pan-cancer. Next, we analyzed drug combination mechanism, and confirmed that the targets of cancer-specific crosstalk network in drug combination were closely related to cancer prognosis by survival analysis. Finally, we save all the results to a webpage for query ( http://bio-bigdata.hrbmu.edu.cn/oDrugCP/ ). In conclusion, our study provided an effective method for screening precise drug combinations for various cancer treatments, which may have important scientific significance and clinical application value for tumor treatment.
Collapse
Affiliation(s)
- Congxue Hu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Wanqi Mi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Feng Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Lun Zhu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Qi Ou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Maohao Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Tengyue Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yuheng Ma
- Department of Pharmacy, Inner Mongolia Medical University, Jinshan Development Zone, Hohhot, 010100, China
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yingqi Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
- Department of Pharmacy, Inner Mongolia Medical University, Jinshan Development Zone, Hohhot, 010100, China.
| |
Collapse
|
55
|
Elkin R, Oh JH, Dela Cruz F, Norton L, Deasy JO, Kung AL, Tannenbaum AR. Dynamic network curvature analysis of gene expression reveals novel potential therapeutic targets in sarcoma. Sci Rep 2024; 14:488. [PMID: 38177639 PMCID: PMC10766622 DOI: 10.1038/s41598-023-49930-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/13/2023] [Indexed: 01/06/2024] Open
Abstract
Network properties account for the complex relationship between genes, making it easier to identify complex patterns in their interactions. In this work, we leveraged these network properties for dual purposes. First, we clustered pediatric sarcoma tumors using network information flow as a similarity metric, computed by the Wasserstein distance. We demonstrate that this approach yields the best concordance with histological subtypes, validated against three state-of-the-art methods. Second, to identify molecular targets that would be missed by more conventional methods of analysis, we applied a novel unsupervised method to cluster gene interactomes represented as networks in pediatric sarcoma. RNA-Seq data were mapped to protein-level interactomes to construct weighted networks that were then subjected to a non-Euclidean, multi-scale geometric approach centered on a discrete notion of curvature. This provides a measure of the functional association among genes in the context of their connectivity. In confirmation of the validity of this method, hierarchical clustering revealed the characteristic EWSR1-FLI1 fusion in Ewing sarcoma. Furthermore, assessing the effects of in silico edge perturbations and simulated gene knockouts as quantified by changes in curvature, we found non-trivial gene associations not previously identified.
Collapse
Affiliation(s)
- Rena Elkin
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, 10065, USA.
| | - Jung Hun Oh
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, 10065, USA
| | - Filemon Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, 10065, USA
| | - Larry Norton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, 10065, USA
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, 10065, USA
| | - Andrew L Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, 10065, USA
| | - Allen R Tannenbaum
- Departments of Computer Science and Applied Mathematics and Statistics, Stony Brook University, Stony Brook, 11794, USA
| |
Collapse
|
56
|
Nithya C, Kiran M, Nagarajaram HA. Hubs and Bottlenecks in Protein-Protein Interaction Networks. Methods Mol Biol 2024; 2719:227-248. [PMID: 37803121 DOI: 10.1007/978-1-0716-3461-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2023]
Abstract
Protein-protein interaction networks (PPINs) represent the physical interactions among proteins in a cell. These interactions are critical in all cellular processes, including signal transduction, metabolic regulation, and gene expression. In PPINs, centrality measures are widely used to identify the most critical nodes. The two most commonly used centrality measures in networks are degree and betweenness centralities. Degree centrality is the number of connections a node has in the network, and betweenness centrality is the measure of the extent to which a node lies on the shortest paths between pairs of other nodes in the network. In PPINs, proteins with high degree and betweenness centrality are referred to as hubs and bottlenecks respectively. Hubs and bottlenecks are topologically and functionally essential proteins that play crucial roles in maintaining the network's structure and function. This article comprehensively reviews essential literature on hubs and bottlenecks, including their properties and functions.
Collapse
Affiliation(s)
- Chandramohan Nithya
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Manjari Kiran
- Department of Systems and Computational Biology, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | | |
Collapse
|
57
|
Liu H, Xu Q, Wufuer H, Li Z, Sun R, Jiang Z, Dou X, Fu Q, Campisi J, Sun Y. Rutin is a potent senomorphic agent to target senescent cells and can improve chemotherapeutic efficacy. Aging Cell 2024; 23:e13921. [PMID: 37475161 PMCID: PMC10776113 DOI: 10.1111/acel.13921] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/24/2023] [Accepted: 06/20/2023] [Indexed: 07/22/2023] Open
Abstract
Aging is a major risk factor for most chronic disorders, for which cellular senescence is one of the central hallmarks. Senescent cells develop the pro-inflammatory senescence-associated secretory phenotype (SASP), which significantly contributes to organismal aging and age-related disorders. Development of senotherapeutics, an emerging class of therapeutic agents to target senescent cells, allows to effectively delay aging and alleviate chronic pathologies. Here we report preliminary outputs from screening of a natural medicinal agent (NMA) library for senotherapeutic candidates and validated several agents with prominent potential as senomorphics. Rutin, a phytochemical constituent found in a number of plants, showed remarkable capacity in targeting senescent cells by dampening expression of the full spectrum SASP. Further analysis indicated that rutin restrains the acute stress-associated phenotype (ASAP) by specifically interfering with the interactions of ATM with HIF1α, a master regulator of cellular and systemic homeostasis activated during senescence, and of ATM with TRAF6, part of a key signaling axis supporting the ASAP development toward the SASP. Conditioned media produced by senescent stromal cells enhanced the malignant phenotypes of prostate cancer cells, including in vitro proliferation, migration, invasion, and more importantly, chemoresistance, while rutin remarkably downregulated these gain-of-functions. Although classic chemotherapy reduced tumor progression, the treatment outcome was substantially improved upon combination of a chemotherapeutic agent with rutin. Our study provides a proof of concept for rutin as an emerging natural senomorphic agent, and presents an effective therapeutic avenue for alleviating age-related pathologies including cancer.
Collapse
Affiliation(s)
- Hanxin Liu
- Department of PharmacologyInstitute of Aging Medicine, Binzhou Medical UniversityYantaiChina
| | - Qixia Xu
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and Health, Chinese Academy of SciencesShanghaiChina
| | - Halidan Wufuer
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and Health, Chinese Academy of SciencesShanghaiChina
| | - Zi Li
- Shanghai Institute of Nutrition and Health, Chinese Academy of SciencesShanghaiChina
| | - Rong Sun
- Department of Discovery BiologyBioduro‐Sundia, Zhangjiang Hi‐Tech ParkShanghaiChina
| | - Zhirui Jiang
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and Health, Chinese Academy of SciencesShanghaiChina
| | - Xuefeng Dou
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and Health, Chinese Academy of SciencesShanghaiChina
| | - Qiang Fu
- Department of PharmacologyInstitute of Aging Medicine, Binzhou Medical UniversityYantaiChina
| | - Judith Campisi
- Buck Institute for Research on AgingNovatoCaliforniaUSA
- Lawrence Berkeley National LaboratoryUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Yu Sun
- Department of PharmacologyInstitute of Aging Medicine, Binzhou Medical UniversityYantaiChina
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and Health, Chinese Academy of SciencesShanghaiChina
- Department of Medicine and VAPSHCSUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
58
|
Csabai L, Bohár B, Türei D, Prabhu S, Földvári-Nagy L, Madgwick M, Fazekas D, Módos D, Ölbei M, Halka T, Poletti M, Kornilova P, Kadlecsik T, Demeter A, Szalay-Bekő M, Kapuy O, Lenti K, Vellai T, Gul L, Korcsmáros T. AutophagyNet: high-resolution data source for the analysis of autophagy and its regulation. Autophagy 2024; 20:188-201. [PMID: 37589496 PMCID: PMC10761021 DOI: 10.1080/15548627.2023.2247737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/31/2023] [Accepted: 08/06/2023] [Indexed: 08/18/2023] Open
Abstract
Macroautophagy/autophagy is a highly-conserved catabolic procss eliminating dysfunctional cellular components and invading pathogens. Autophagy malfunction contributes to disorders such as cancer, neurodegenerative and inflammatory diseases. Understanding autophagy regulation in health and disease has been the focus of the last decades. We previously provided an integrated database for autophagy research, the Autophagy Regulatory Network (ARN). For the last eight years, this resource has been used by thousands of users. Here, we present a new and upgraded resource, AutophagyNet. It builds on the previous database but contains major improvements to address user feedback and novel needs due to the advancement in omics data availability. AutophagyNet contains updated interaction curation and integration of over 280,000 experimentally verified interactions between core autophagy proteins and their protein, transcriptional and post-transcriptional regulators as well as their potential upstream pathway connections. AutophagyNet provides annotations for each core protein about their role: 1) in different types of autophagy (mitophagy, xenophagy, etc.); 2) in distinct stages of autophagy (initiation, expansion, termination, etc.); 3) with subcellular and tissue-specific localization. These annotations can be used to filter the dataset, providing customizable download options tailored to the user's needs. The resource is available in various file formats (e.g. CSV, BioPAX and PSI-MI), and data can be analyzed and visualized directly in Cytoscape. The multi-layered regulation of autophagy can be analyzed by combining AutophagyNet with tissue- or cell type-specific (multi-)omics datasets (e.g. transcriptomic or proteomic data). The resource is publicly accessible at http://autophagynet.org.Abbreviations: ARN: Autophagy Regulatory Network; ATG: autophagy related; BCR: B cell receptor pathway; BECN1: beclin 1; GABARAP: GABA type A receptor-associated protein; IIP: innate immune pathway; LIR: LC3-interacting region; lncRNA: long non-coding RNA; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; miRNA: microRNA; NHR: nuclear hormone receptor; PTM: post-translational modification; RTK: receptor tyrosine kinase; TCR: T cell receptor; TLR: toll like receptor.
Collapse
Affiliation(s)
- Luca Csabai
- Earlham Institute, Norwich, UK
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Balázs Bohár
- Earlham Institute, Norwich, UK
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Dénes Türei
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg, Germany
| | | | - László Földvári-Nagy
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Matthew Madgwick
- Earlham Institute, Norwich, UK
- Quadram Institute, Norwich Research Park, Norwich, UK
| | - Dávid Fazekas
- Earlham Institute, Norwich, UK
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Dezső Módos
- Earlham Institute, Norwich, UK
- Quadram Institute, Norwich Research Park, Norwich, UK
| | - Márton Ölbei
- Earlham Institute, Norwich, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Themis Halka
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Martina Poletti
- Earlham Institute, Norwich, UK
- Quadram Institute, Norwich Research Park, Norwich, UK
| | | | - Tamás Kadlecsik
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
| | | | | | - Orsolya Kapuy
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Katalin Lenti
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Tibor Vellai
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
- ELKH/MTA-ELTE Genetics Research Group, Budapest, Hungary
| | - Lejla Gul
- Earlham Institute, Norwich, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Tamás Korcsmáros
- Earlham Institute, Norwich, UK
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
- Quadram Institute, Norwich Research Park, Norwich, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| |
Collapse
|
59
|
Chen J, Li J, Zhong C, Ling Y, Liu D, Li X, Xu J, Liu Q, Guo Y, Wang L. Nanobody-loaded nanobubbles targeting the G250 antigen with ultrasound/photoacoustic/fluorescence multimodal imaging capabilities for specifically enhanced imaging of RCC. NANOSCALE 2023; 16:343-359. [PMID: 38062769 DOI: 10.1039/d3nr04097f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Clinicians have attempted to discover a noninvasive, easy-to-perform, and accurate method to distinguish benign and malignant renal masses. The targeted nanobubbles (NBs) we constructed that target the specific membrane antigen of renal cell carcinoma (RCC), G250, and contain indocyanine green (ICG) provide multimodal enhanced imaging capability in ultrasound/photoacoustic/fluorescence for RCC which may possibly solve this problem. In this study, we encapsulated ICG in the lipid shell of the NBs by mechanical oscillation, then anti-G250 nanobodies (AGN) were coupled to the surfaces by the biotin-streptavidin bridge method, and the nanobubble named AGN/ICG-NB was completely constructed. The average particle diameter of the prepared AGN/ICG-NBs was (427.2 ± 4.50) nm, and the zeta potential was (-13.33 ± 1.01) mV. Immunofluorescence and flow cytometry confirmed the specific binding capability of AGN/ICG-NBs to G250-positive cells. In vitro imaging experiments confirmed the multimodal imaging capability of AGN/ICG-NBs, and the in vivo imaging experiments demonstrated the specifically enhanced ability of AGN/ICG-NBs for ultrasound/photoacoustic/fluorescence imaging of human-derived RCC tumors. The biosafety of AGN/ICG-NB was verified by CCK-8 assay, organ H&E staining and blood biochemical indices. In conclusion, the targeted nanobubbles we prepared with ultrasound/photoacoustic/fluorescence multimodal imaging capabilities provide a potentially feasible approach to address the need for early diagnosis and differential diagnosis of renal masses.
Collapse
Affiliation(s)
- Jiajiu Chen
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China.
| | - Jingyi Li
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China.
| | - Chengjie Zhong
- The Second Clinical Medical College, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yi Ling
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing 400038, P.R. China.
| | - Deng Liu
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing 400038, P.R. China.
| | - Xin Li
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing 400038, P.R. China.
| | - Jing Xu
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China.
| | - Qiuli Liu
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China.
| | - Yanli Guo
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing 400038, P.R. China.
| | - Luofu Wang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China.
| |
Collapse
|
60
|
Wang Y, Zhou B, Ru J, Meng X, Wang Y, Liu W. Advances in computational methods for identifying cancer driver genes. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:21643-21669. [PMID: 38124614 DOI: 10.3934/mbe.2023958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Cancer driver genes (CDGs) are crucial in cancer prevention, diagnosis and treatment. This study employed computational methods for identifying CDGs, categorizing them into four groups. The major frameworks for each of these four categories were summarized. Additionally, we systematically gathered data from public databases and biological networks, and we elaborated on computational methods for identifying CDGs using the aforementioned databases. Further, we summarized the algorithms, mainly involving statistics and machine learning, used for identifying CDGs. Notably, the performances of nine typical identification methods for eight types of cancer were compared to analyze the applicability areas of these methods. Finally, we discussed the challenges and prospects associated with methods for identifying CDGs. The present study revealed that the network-based algorithms and machine learning-based methods demonstrated superior performance.
Collapse
Affiliation(s)
- Ying Wang
- School of Computer Science and Engineering, Changshu Institute of Technology, Changshu 215500, China
| | - Bohao Zhou
- School of Computer Science and Engineering, Changshu Institute of Technology, Changshu 215500, China
| | - Jidong Ru
- School of Textile Garment and Design, Changshu Institute of Technology, Changshu 215500, China
| | - Xianglian Meng
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou 213032, China
| | - Yundong Wang
- School of Computer Science and Engineering, Changshu Institute of Technology, Changshu 215500, China
| | - Wenjie Liu
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou 213032, China
| |
Collapse
|
61
|
Guo J, Zhang Y, Gao Y, Li S, Xu G, Tian Z, Xu Q, Li X, Li Y, Zhang Y. Systematical analyses of large-scale transcriptome reveal viral infection-related genes and disease comorbidities. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2023; 51:453-465. [PMID: 37651591 DOI: 10.1080/21691401.2023.2252477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 09/02/2023]
Abstract
Perturbation of transcriptome in viral infection patients is a recurrent theme impacting symptoms and mortality, yet a detailed understanding of pertinent transcriptome and identification of robust biomarkers is not complete. In this study, we manually collected 23 datasets related to 6,197 blood transcriptomes across 16 types of respiratory virus infections. We applied a comprehensive systems biology approach starting with whole-blood transcriptomes combined with multilevel bioinformatics analyses to characterize the expression, functional pathways, and protein-protein interaction (PPI) networks to identify robust biomarkers and disease comorbidities. Robust gene markers of infection with different viruses were identified, which can accurately classify the normal and infected patients in train and validation cohorts. The biological processes (BP) of different viruses showed great similarity and enriched in infection and immune response pathways. Network-based analyses revealed that a variety of viral infections were associated with nervous system diseases, neoplasms and metabolic diseases, and significantly correlated with brain tissues. In summary, our manually collected transcriptomes and comprehensive analyses reveal key molecular markers and disease comorbidities in the process of viral infection, which could provide a valuable theoretical basis for the prevention of subsequent public health events for respiratory virus infections.
Collapse
Affiliation(s)
- Jing Guo
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Ya Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Yueying Gao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Si Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Gang Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Zhanyu Tian
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Qi Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Xia Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongsheng Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
62
|
Li R, Zhao H, Huang X, Zhang J, Bai R, Zhuang L, Wen S, Wu S, Zhou Q, Li M, Zeng L, Zhang S, Deng S, Su J, Zuo Z, Chen R, Lin D, Zheng J. Super-enhancer RNA m 6A promotes local chromatin accessibility and oncogene transcription in pancreatic ductal adenocarcinoma. Nat Genet 2023; 55:2224-2234. [PMID: 37957340 DOI: 10.1038/s41588-023-01568-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 10/12/2023] [Indexed: 11/15/2023]
Abstract
The biological functions of noncoding RNA N6-methyladenosine (m6A) modification remain poorly understood. In the present study, we depict the landscape of super-enhancer RNA (seRNA) m6A modification in pancreatic ductal adenocarcinoma (PDAC) and reveal a regulatory axis of m6A seRNA, H3K4me3 modification, chromatin accessibility and oncogene transcription. We demonstrate the cofilin family protein CFL1, overexpressed in PDAC, as a METTL3 cofactor that helps seRNA m6A methylation formation. The increased seRNA m6As are recognized by the reader YTHDC2, which recruits H3K4 methyltransferase MLL1 to promote H3K4me3 modification cotranscriptionally. Super-enhancers with a high level of H3K4me3 augment chromatin accessibility and facilitate oncogene transcription. Collectively, these results shed light on a CFL1-METTL3-seRNA m6A-YTHDC2/MLL1 axis that plays a role in the epigenetic regulation of local chromatin state and gene expression, which strengthens our knowledge about the functions of super-enhancers and their transcripts.
Collapse
Affiliation(s)
- Rui Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Hongzhe Zhao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Xudong Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Jialiang Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Ruihong Bai
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Lisha Zhuang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shujuan Wen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shaojia Wu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Quanbo Zhou
- Department of Pancreaticobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mei Li
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lingxing Zeng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shaoping Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shuang Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Jiachun Su
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Zhixiang Zuo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Rufu Chen
- Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Dongxin Lin
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China.
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Jian Zheng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
63
|
Li Y, Fan Z, Rao J, Chen Z, Chu Q, Zheng M, Li X. An overview of recent advances and challenges in predicting compound-protein interaction (CPI). MEDICAL REVIEW (2021) 2023; 3:465-486. [PMID: 38282802 PMCID: PMC10808869 DOI: 10.1515/mr-2023-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/30/2023] [Indexed: 01/30/2024]
Abstract
Compound-protein interactions (CPIs) are critical in drug discovery for identifying therapeutic targets, drug side effects, and repurposing existing drugs. Machine learning (ML) algorithms have emerged as powerful tools for CPI prediction, offering notable advantages in cost-effectiveness and efficiency. This review provides an overview of recent advances in both structure-based and non-structure-based CPI prediction ML models, highlighting their performance and achievements. It also offers insights into CPI prediction-related datasets and evaluation benchmarks. Lastly, the article presents a comprehensive assessment of the current landscape of CPI prediction, elucidating the challenges faced and outlining emerging trends to advance the field.
Collapse
Affiliation(s)
- Yanbei Li
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, Zhejiang Province, China
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhehuan Fan
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jingxin Rao
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhiyi Chen
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, Zhejiang Province, China
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qinyu Chu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, Zhejiang Province, China
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mingyue Zheng
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, Zhejiang Province, China
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xutong Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
64
|
Esmaili F, Pourmirzaei M, Ramazi S, Shojaeilangari S, Yavari E. A Review of Machine Learning and Algorithmic Methods for Protein Phosphorylation Site Prediction. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:1266-1285. [PMID: 37863385 PMCID: PMC11082408 DOI: 10.1016/j.gpb.2023.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 01/16/2023] [Accepted: 03/23/2023] [Indexed: 10/22/2023]
Abstract
Post-translational modifications (PTMs) have key roles in extending the functional diversity of proteins and, as a result, regulating diverse cellular processes in prokaryotic and eukaryotic organisms. Phosphorylation modification is a vital PTM that occurs in most proteins and plays a significant role in many biological processes. Disorders in the phosphorylation process lead to multiple diseases, including neurological disorders and cancers. The purpose of this review is to organize this body of knowledge associated with phosphorylation site (p-site) prediction to facilitate future research in this field. At first, we comprehensively review all related databases and introduce all steps regarding dataset creation, data preprocessing, and method evaluation in p-site prediction. Next, we investigate p-site prediction methods, which are divided into two computational groups: algorithmic and machine learning (ML). Additionally, it is shown that there are basically two main approaches for p-site prediction by ML: conventional and end-to-end deep learning methods, both of which are given an overview. Moreover, this review introduces the most important feature extraction techniques, which have mostly been used in p-site prediction. Finally, we create three test sets from new proteins related to the released version of the database of protein post-translational modifications (dbPTM) in 2022 based on general and human species. Evaluating online p-site prediction tools on newly added proteins introduced in the dbPTM 2022 release, distinct from those in the dbPTM 2019 release, reveals their limitations. In other words, the actual performance of these online p-site prediction tools on unseen proteins is notably lower than the results reported in their respective research papers.
Collapse
Affiliation(s)
- Farzaneh Esmaili
- Department of Information Technology, Tarbiat Modares University, Tehran 14115-111, Iran
| | - Mahdi Pourmirzaei
- Department of Information Technology, Tarbiat Modares University, Tehran 14115-111, Iran
| | - Shahin Ramazi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-111, Iran.
| | - Seyedehsamaneh Shojaeilangari
- Biomedical Engineering Group, Department of Electrical Engineering and Information Technology, Iranian Research Organization for Science and Technology (IROST), Tehran 33535-111, Iran
| | - Elham Yavari
- Department of Information Technology, Tarbiat Modares University, Tehran 14115-111, Iran
| |
Collapse
|
65
|
Zhao Y, Yin J, Zhang L, Zhang Y, Chen X. Drug-drug interaction prediction: databases, web servers and computational models. Brief Bioinform 2023; 25:bbad445. [PMID: 38113076 PMCID: PMC10782925 DOI: 10.1093/bib/bbad445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/26/2023] [Accepted: 11/14/2023] [Indexed: 12/21/2023] Open
Abstract
In clinical treatment, two or more drugs (i.e. drug combination) are simultaneously or successively used for therapy with the purpose of primarily enhancing the therapeutic efficacy or reducing drug side effects. However, inappropriate drug combination may not only fail to improve efficacy, but even lead to adverse reactions. Therefore, according to the basic principle of improving the efficacy and/or reducing adverse reactions, we should study drug-drug interactions (DDIs) comprehensively and thoroughly so as to reasonably use drug combination. In this review, we first introduced the basic conception and classification of DDIs. Further, some important publicly available databases and web servers about experimentally verified or predicted DDIs were briefly described. As an effective auxiliary tool, computational models for predicting DDIs can not only save the cost of biological experiments, but also provide relevant guidance for combination therapy to some extent. Therefore, we summarized three types of prediction models (including traditional machine learning-based models, deep learning-based models and score function-based models) proposed during recent years and discussed the advantages as well as limitations of them. Besides, we pointed out the problems that need to be solved in the future research of DDIs prediction and provided corresponding suggestions.
Collapse
Affiliation(s)
- Yan Zhao
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou 221116, China
| | - Jun Yin
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou 221116, China
| | - Li Zhang
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou 221116, China
| | - Yong Zhang
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou 221116, China
| | - Xing Chen
- School of Science, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
66
|
Varshney N, Murmu S, Baral B, Kashyap D, Singh S, Kandpal M, Bhandari V, Chaurasia A, Kumar S, Jha HC. Unraveling the Aurora kinase A and Epstein-Barr nuclear antigen 1 axis in Epstein Barr virus associated gastric cancer. Virology 2023; 588:109901. [PMID: 37839162 DOI: 10.1016/j.virol.2023.109901] [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: 08/04/2023] [Revised: 09/18/2023] [Accepted: 09/29/2023] [Indexed: 10/17/2023]
Abstract
Aurora kinase A (AURKA) is one of the crucial cell cycle regulators associated with gastric cancer. Here, we explored Epstein Barr Virus-induced gastric cancer progression through EBV protein EBNA1 with AURKA. We found that EBV infection enhanced cell proliferation and migration of AGS cells and upregulation of AURKA levels. AURKA knockdown markedly reduced the proliferation and migration of the AGS cells even with EBV infection. Moreover, MD-simulation data deciphered the probable connection between EBNA1 and AURKA. The in-vitro analysis through the transcript and protein expression showed that AURKA knockdown reduces the expression of EBNA1. Moreover, EBNA1 alone can enhance AURKA protein expression in AGS cells. Co-immunoprecipitation and NMR analysis between AURKA and EBNA1 depicts the interaction between two proteins. In addition, AURKA knockdown promotes apoptosis in EBV-infected AGS cells through cleavage of Caspase-3, -9, and PARP1. This study demonstrates that EBV oncogenic modulators EBNA1 possibly modulate AURKA in EBV-mediated gastric cancer progression.
Collapse
Affiliation(s)
- Nidhi Varshney
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, India
| | - Sneha Murmu
- Division of Agricultural Bioinformatics (DABin), ICAR-Indian Agricultural Statistics Research Institute (IASRI), India
| | - Budhadev Baral
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, India
| | - Dharmendra Kashyap
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, India
| | - Siddharth Singh
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, India
| | - Meenakshi Kandpal
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, India
| | - Vasundhra Bhandari
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | | | - Sunil Kumar
- Division of Agricultural Bioinformatics (DABin), ICAR-Indian Agricultural Statistics Research Institute (IASRI), India.
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, India.
| |
Collapse
|
67
|
Somers J, Fenner M, Kong G, Thirumalaisamy D, Yashar WM, Thapa K, Kinali M, Nikolova O, Babur Ö, Demir E. A framework for considering prior information in network-based approaches to omics data analysis. Proteomics 2023; 23:e2200402. [PMID: 37986684 DOI: 10.1002/pmic.202200402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 11/22/2023]
Abstract
For decades, molecular biologists have been uncovering the mechanics of biological systems. Efforts to bring their findings together have led to the development of multiple databases and information systems that capture and present pathway information in a computable network format. Concurrently, the advent of modern omics technologies has empowered researchers to systematically profile cellular processes across different modalities. Numerous algorithms, methodologies, and tools have been developed to use prior knowledge networks (PKNs) in the analysis of omics datasets. Interestingly, it has been repeatedly demonstrated that the source of prior knowledge can greatly impact the results of a given analysis. For these methods to be successful it is paramount that their selection of PKNs is amenable to the data type and the computational task they aim to accomplish. Here we present a five-level framework that broadly describes network models in terms of their scope, level of detail, and ability to inform causal predictions. To contextualize this framework, we review a handful of network-based omics analysis methods at each level, while also describing the computational tasks they aim to accomplish.
Collapse
Affiliation(s)
- Julia Somers
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - Madeleine Fenner
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - Garth Kong
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
- Division of Oncological Sciences, Oregon Health and Science University, Portland, Oregon, USA
| | - Dharani Thirumalaisamy
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - William M Yashar
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
- Division of Oncological Sciences, Oregon Health and Science University, Portland, Oregon, USA
| | - Kisan Thapa
- Computer Science Department, University of Massachusetts Boston, College of Science and Mathematics, Boston, Massachusetts, USA
| | - Meric Kinali
- Computer Science Department, University of Massachusetts Boston, College of Science and Mathematics, Boston, Massachusetts, USA
| | - Olga Nikolova
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
- Division of Oncological Sciences, Oregon Health and Science University, Portland, Oregon, USA
| | - Özgün Babur
- Computer Science Department, University of Massachusetts Boston, College of Science and Mathematics, Boston, Massachusetts, USA
| | - Emek Demir
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
68
|
Pfeifer B, Chereda H, Martin R, Saranti A, Clemens S, Hauschild AC, Beißbarth T, Holzinger A, Heider D. Ensemble-GNN: federated ensemble learning with graph neural networks for disease module discovery and classification. Bioinformatics 2023; 39:btad703. [PMID: 37988152 PMCID: PMC10684359 DOI: 10.1093/bioinformatics/btad703] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/06/2023] [Accepted: 11/20/2023] [Indexed: 11/22/2023] Open
Abstract
SUMMARY Federated learning enables collaboration in medicine, where data is scattered across multiple centers without the need to aggregate the data in a central cloud. While, in general, machine learning models can be applied to a wide range of data types, graph neural networks (GNNs) are particularly developed for graphs, which are very common in the biomedical domain. For instance, a patient can be represented by a protein-protein interaction (PPI) network where the nodes contain the patient-specific omics features. Here, we present our Ensemble-GNN software package, which can be used to deploy federated, ensemble-based GNNs in Python. Ensemble-GNN allows to quickly build predictive models utilizing PPI networks consisting of various node features such as gene expression and/or DNA methylation. We exemplary show the results from a public dataset of 981 patients and 8469 genes from the Cancer Genome Atlas (TCGA). AVAILABILITY AND IMPLEMENTATION The source code is available at https://github.com/pievos101/Ensemble-GNN, and the data at Zenodo (DOI: 10.5281/zenodo.8305122).
Collapse
Affiliation(s)
- Bastian Pfeifer
- Institute for Medical Informatics, Statistics and Documentation, Medical University Graz, Graz 8036, Austria
| | - Hryhorii Chereda
- Medical Bioinformatics, University Medical Center Göttingen, Göttingen 37077, Germany
| | - Roman Martin
- Data Science in Biomedicine, Department of Mathematics and Computer Science, University of Marburg, Marburg 35043, Germany
| | - Anna Saranti
- Institute for Medical Informatics, Statistics and Documentation, Medical University Graz, Graz 8036, Austria
- Human-Centered AI Lab, University of Natural Resources and Life Sciences, Vienna 1190, Austria
| | - Sandra Clemens
- Data Science in Biomedicine, Department of Mathematics and Computer Science, University of Marburg, Marburg 35043, Germany
| | - Anne-Christin Hauschild
- Institute for Medical Informatics, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Tim Beißbarth
- Medical Bioinformatics, University Medical Center Göttingen, Göttingen 37077, Germany
| | - Andreas Holzinger
- Institute for Medical Informatics, Statistics and Documentation, Medical University Graz, Graz 8036, Austria
- Human-Centered AI Lab, University of Natural Resources and Life Sciences, Vienna 1190, Austria
| | - Dominik Heider
- Data Science in Biomedicine, Department of Mathematics and Computer Science, University of Marburg, Marburg 35043, Germany
| |
Collapse
|
69
|
Pollin G, De Assuncao T, Doria Jorge S, Chi YI, Charlesworth M, Madden B, Iovanna J, Zimmermann M, Urrutia R, Lomberk G. Writers and readers of H3K9me2 form distinct protein networks during the cell cycle that include candidates for H3K9 mimicry. Biosci Rep 2023; 43:BSR20231093. [PMID: 37782747 PMCID: PMC10611923 DOI: 10.1042/bsr20231093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/15/2023] [Accepted: 10/02/2023] [Indexed: 10/04/2023] Open
Abstract
Histone H3 lysine 9 methylation (H3K9me), which is written by the Euchromatic Histone Lysine Methyltransferases EHMT1 and EHMT2 and read by the heterochromatin protein 1 (HP1) chromobox (CBX) protein family, is dysregulated in many types of cancers. Approaches to inhibit regulators of this pathway are currently being evaluated for therapeutic purposes. Thus, knowledge of the complexes supporting the function of these writers and readers during the process of cell proliferation is critical for our understanding of their role in carcinogenesis. Here, we immunopurified each of these proteins and used mass spectrometry to define their associated non-histone proteins, individually and at two different phases of the cell cycle, namely G1/S and G2/M. Our findings identify novel binding proteins for these writers and readers, as well as corroborate known interactors, to show the formation of distinct protein complex networks in a cell cycle phase-specific manner. Furthermore, there is an organizational switch between cell cycle phases for interactions among specific writer-reader pairs. Through a multi-tiered bioinformatics-based approach, we reveal that many interacting proteins exhibit histone mimicry, based on an H3K9-like linear motif. Gene ontology analyses, pathway enrichment, and network reconstruction inferred that these comprehensive EHMT and CBX-associated interacting protein networks participate in various functions, including transcription, DNA repair, splicing, and membrane disassembly. Combined, our data reveals novel complexes that provide insight into key functions of cell cycle-associated epigenomic processes that are highly relevant for better understanding these chromatin-modifying proteins during cell cycle and carcinogenesis.
Collapse
Affiliation(s)
- Gareth Pollin
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, U.S.A
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI Center, Medical College of Wisconsin, Milwaukee, WI, U.S.A
| | - Thiago M. De Assuncao
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, U.S.A
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI Center, Medical College of Wisconsin, Milwaukee, WI, U.S.A
| | - Salomao Doria Jorge
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, U.S.A
| | - Young-In Chi
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, U.S.A
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI Center, Medical College of Wisconsin, Milwaukee, WI, U.S.A
| | | | - Benjamin Madden
- Medical Genome Facility, Proteomics Core, Mayo Clinic, Rochester, MN, U.S.A
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Michael T. Zimmermann
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, U.S.A
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, U.S.A
- Clinical and Translational Sciences Institute, Medical College of Wisconsin, Milwaukee, WI, U.S.A
| | - Raul Urrutia
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, U.S.A
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI Center, Medical College of Wisconsin, Milwaukee, WI, U.S.A
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, U.S.A
| | - Gwen Lomberk
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, U.S.A
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI Center, Medical College of Wisconsin, Milwaukee, WI, U.S.A
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, U.S.A
| |
Collapse
|
70
|
Harkin EF, Nasrallah G, Le François B, Albert PR. Transcriptional Regulation of the Human 5-HT1A Receptor Gene by Lithium: Role of Deaf1 and GSK3β. Int J Mol Sci 2023; 24:15620. [PMID: 37958600 PMCID: PMC10647674 DOI: 10.3390/ijms242115620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/11/2023] [Accepted: 10/21/2023] [Indexed: 11/15/2023] Open
Abstract
Serotonin 1A (5-HT1A) autoreceptors located on serotonin neurons inhibit their activity, and their upregulation has been implicated in depression, suicide and resistance to antidepressant treatment. Conversely, post-synaptic 5-HT1A heteroreceptors are important for antidepressant response. The transcription factor deformed epidermal autoregulatory factor 1 (Deaf1) acts as a presynaptic repressor and postsynaptic enhancer of 5-HT1A transcription, but the mechanism is unclear. Because Deaf1 interacts with and is phosphorylated by glycogen synthase kinase 3β (GSK3β)-a constitutively active protein kinase that is inhibited by the mood stabilizer lithium at therapeutic concentrations-we investigated the role of GSK3β in Deaf1 regulation of human 5-HT1A transcription. In 5-HT1A promoter-reporter assays, human HEK293 kidney and 5-HT1A-expressing SKN-SH neuroblastoma cells, transfection of Deaf1 reduced 5-HT1A promoter activity by ~45%. To identify potential GSK3β site(s) on Deaf1, point mutations of known and predicted phosphorylation sites on Deaf1 were tested. Deaf1 repressor function was not affected by any of the mutants tested except the Y300F mutant, which augmented Deaf1 repression. Both lithium and the selective GSK3 inhibitors CHIR-99021 and AR-014418 attenuated and reversed Deaf1 repression compared to vector. This inhibition was at concentrations that maximally inhibit GSK3β activity as detected by the GSK3β-sensitive TCF/LEF reporter construct. Our results support the hypothesis that GSK3β regulates the activity of Deaf1 to repress 5-HT1A transcription and provide a potential mechanism for actions of GSK3 inhibitors on behavior.
Collapse
Affiliation(s)
| | | | | | - Paul R. Albert
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa, 451 Smyth Road, Ottawa, ON K1H-8M5, Canada (B.L.F.)
| |
Collapse
|
71
|
Niinae T, Sugiyama N, Ishihama Y. Validity of the cell-extracted proteome as a substrate pool for exploring phosphorylation motifs of kinases. Genes Cells 2023; 28:727-735. [PMID: 37658684 PMCID: PMC11447832 DOI: 10.1111/gtc.13063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 09/03/2023]
Abstract
Three representative protein kinases with different substrate preferences, ERK1 (Pro-directed), CK2 (acidophilic), and PKA (basophilic), were used to investigate phosphorylation sequence motifs in substrate pools consisting of the proteomes from three different cell lines, MCF7 (human mammary carcinoma), HeLa (human cervical carcinoma), and Jurkat (human acute T-cell leukemia). Specifically, recombinant kinases were added to the cell-extracted proteomes to phosphorylate the substrates in vitro. After trypsin digestion, the phosphopeptides were enriched and subjected to nanoLC/MS/MS analysis to identify their phosphorylation sites on a large scale. By analyzing the obtained phosphorylation sites and their surrounding sequences, phosphorylation motifs were extracted for each kinase-substrate proteome pair. We found that each kinase exhibited the same set of phosphorylation motifs, independently of the substrate pool proteome. Furthermore, the identified motifs were also consistent with those found using a completely randomized peptide library. These results indicate that cell-extracted proteomes can provide kinase phosphorylation motifs with sufficient accuracy, even though their sequences are not completely random, supporting the robustness of phosphorylation motif identification based on phosphoproteome analysis of cell extracts as a substrate pool for a kinase of interest.
Collapse
Affiliation(s)
- Tomoya Niinae
- Graduate School of Pharmaceutical SciencesKyoto UniversityKyotoJapan
| | - Naoyuki Sugiyama
- Graduate School of Pharmaceutical SciencesKyoto UniversityKyotoJapan
| | - Yasushi Ishihama
- Graduate School of Pharmaceutical SciencesKyoto UniversityKyotoJapan
- Laboratory of Clinical and Analytical ChemistryNational Institute of Biomedical Innovation, Health and NutritionIbarakiOsakaJapan
| |
Collapse
|
72
|
He C, Qu Y, Yin J, Zhao Z, Ma R, Duan L. Cross-view contrastive representation learning approach to predicting DTIs via integrating multi-source information. Methods 2023; 218:176-188. [PMID: 37586602 DOI: 10.1016/j.ymeth.2023.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/26/2023] [Accepted: 08/08/2023] [Indexed: 08/18/2023] Open
Abstract
Drug-target interaction (DTI) prediction serves as the foundation of new drug findings and drug repositioning. For drugs/targets, the sequence data contains the biological structural information, while the heterogeneous network contains the biochemical functional information. These two types of information describe different aspects of drugs and targets. Due to the complexity of DTI machinery, it is necessary to learn the representation from multiple perspectives. We hereby try to design a way to leverage information from multi-source data to the maximum extent and find a strategy to fuse them. To address the above challenges, we propose a model, named MOVE (short for integrating multi-source information for predicting DTI via cross-view contrastive learning), for learning comprehensive representations of each drug and target from multi-source data. MOVE extracts information from the sequence view and the network view, then utilizes a fusion module with auxiliary contrastive learning to facilitate the fusion of representations. Experimental results on the benchmark dataset demonstrate that MOVE is effective in DTI prediction.
Collapse
Affiliation(s)
- Chengxin He
- School of Computer Science, Sichuan University, Chengdu 610065, China; Med-X Center for Informatics, Sichuan University, Chengdu 610065, China
| | - Yuening Qu
- School of Computer Science, Sichuan University, Chengdu 610065, China
| | - Jin Yin
- The West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Zhenjiang Zhao
- School of Computer Science, Sichuan University, Chengdu 610065, China
| | - Runze Ma
- School of Computer Science, Sichuan University, Chengdu 610065, China
| | - Lei Duan
- School of Computer Science, Sichuan University, Chengdu 610065, China; Med-X Center for Informatics, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
73
|
Al-Aamri A, Kamarul Azman S, Daw Elbait G, Alsafar H, Henschel A. Critical assessment of on-premise approaches to scalable genome analysis. BMC Bioinformatics 2023; 24:354. [PMID: 37735350 PMCID: PMC10512525 DOI: 10.1186/s12859-023-05470-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 09/08/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Plummeting DNA sequencing cost in recent years has enabled genome sequencing projects to scale up by several orders of magnitude, which is transforming genomics into a highly data-intensive field of research. This development provides the much needed statistical power required for genotype-phenotype predictions in complex diseases. METHODS In order to efficiently leverage the wealth of information, we here assessed several genomic data science tools. The rationale to focus on on-premise installations is to cope with situations where data confidentiality and compliance regulations etc. rule out cloud based solutions. We established a comprehensive qualitative and quantitative comparison between BCFtools, SnpSift, Hail, GEMINI, and OpenCGA. The tools were compared in terms of data storage technology, query speed, scalability, annotation, data manipulation, visualization, data output representation, and availability. RESULTS Tools that leverage sophisticated data structures are noted as the most suitable for large-scale projects in varying degrees of scalability in comparison to flat-file manipulation (e.g., BCFtools, and SnpSift). Remarkably, for small to mid-size projects, even lightweight relational database. CONCLUSION The assessment criteria provide insights into the typical questions posed in scalable genomics and serve as guidance for the development of scalable computational infrastructure in genomics.
Collapse
Affiliation(s)
- Amira Al-Aamri
- Department of Electrical Engineering and Computer Science, College of Engineering, Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates
| | - Syafiq Kamarul Azman
- Department of Electrical Engineering and Computer Science, College of Engineering, Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates
| | - Gihan Daw Elbait
- Department of Biology, College of Arts and Sciences, Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates
- Center for Biotechnology (BTC), Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates
| | - Habiba Alsafar
- Center for Biotechnology (BTC), Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates
- Department of Biomedical Engineering, Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates
| | - Andreas Henschel
- Department of Electrical Engineering and Computer Science, College of Engineering, Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates.
- Center for Biotechnology (BTC), Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
74
|
Pan Y, Ren H, Lan L, Li Y, Huang T. Review of Predicting Synergistic Drug Combinations. Life (Basel) 2023; 13:1878. [PMID: 37763281 PMCID: PMC10533134 DOI: 10.3390/life13091878] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
The prediction of drug combinations is of great clinical significance. In many diseases, such as high blood pressure, diabetes, and stomach ulcers, the simultaneous use of two or more drugs has shown clear efficacy. It has greatly reduced the progression of drug resistance. This review presents the latest applications of methods for predicting the effects of drug combinations and the bioactivity databases commonly used in drug combination prediction. These studies have played a significant role in developing precision therapy. We first describe the concept of synergy. we study various publicly available databases for drug combination prediction tasks. Next, we introduce five algorithms applied to drug combinatorial prediction, which include traditional machine learning methods, deep learning methods, mathematical methods, systems biology methods and search algorithms. In the end, we sum up the difficulties encountered in prediction models.
Collapse
Affiliation(s)
- Yichen Pan
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; (Y.P.); (H.R.)
| | - Haotian Ren
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; (Y.P.); (H.R.)
| | - Liang Lan
- Department of Interactive Media, Hong Kong Baptist University, Hong Kong, China;
| | - Yixue Li
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; (Y.P.); (H.R.)
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Guangzhou Laboratory, Guangzhou 510005, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai 200433, China
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; (Y.P.); (H.R.)
| |
Collapse
|
75
|
Yao K, Wang X, Li W, Zhu H, Jiang Y, Li Y, Tian T, Yang Z, Liu Q, Liu Q. Semi-supervised heterogeneous graph contrastive learning for drug-target interaction prediction. Comput Biol Med 2023; 163:107199. [PMID: 37421738 DOI: 10.1016/j.compbiomed.2023.107199] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/15/2023] [Accepted: 06/19/2023] [Indexed: 07/10/2023]
Abstract
Identification of drug-target interactions (DTIs) is an important step in drug discovery and drug repositioning. In recent years, graph-based methods have attracted great attention and show advantages on predicting potential DTIs. However, these methods face the problem that the known DTIs are very limited and expensive to obtain, which decreases the generalization ability of the methods. Self-supervised contrastive learning is independent of labeled DTIs, which can mitigate the impact of the problem. Therefore, we propose a framework SHGCL-DTI for predicting DTIs, which supplements the classical semi-supervised DTI prediction task with an auxiliary graph contrastive learning module. Specifically, we generate representations for the nodes through the neighbor view and meta-path view, and define positive and negative pairs to maximize the similarity between positive pairs from different views. Subsequently, SHGCL-DTI reconstructs the original heterogeneous network to predict the potential DTIs. The experiments on the public dataset show that SHGCL-DTI has significant improvement in different scenarios, compared with existing state-of-the-art methods. We also demonstrate that the contrastive learning module improves the prediction performance and generalization ability of SHGCL-DTI through ablation study. In addition, we have found several novel predicted DTIs supported by the biological literature. The data and source code are available at: https://github.com/TOJSSE-iData/SHGCL-DTI.
Collapse
Affiliation(s)
- Kainan Yao
- School of Software Engineering, Tongji University, 4800 Caoan Road, Jiading District, Shanghai, 201804, China
| | - Xiaowen Wang
- School of Software Engineering, Tongji University, 4800 Caoan Road, Jiading District, Shanghai, 201804, China
| | - Wannian Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Yangpu District, Shanghai, 200092, China.
| | - Hongming Zhu
- School of Software Engineering, Tongji University, 4800 Caoan Road, Jiading District, Shanghai, 201804, China
| | - Yizhi Jiang
- School of Software Engineering, Tongji University, 4800 Caoan Road, Jiading District, Shanghai, 201804, China
| | - Yulong Li
- School of Software Engineering, Tongji University, 4800 Caoan Road, Jiading District, Shanghai, 201804, China
| | - Tongxuan Tian
- School of Software Engineering, Tongji University, 4800 Caoan Road, Jiading District, Shanghai, 201804, China
| | - Zhaoyi Yang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, No. 96, JinZhai Road Baohe District, Hefei, 230001, Anhui, China.
| | - Qi Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Yangpu District, Shanghai, 200092, China.
| | - Qin Liu
- School of Software Engineering, Tongji University, 4800 Caoan Road, Jiading District, Shanghai, 201804, China.
| |
Collapse
|
76
|
Luo X, Wang L, Hu P, Hu L. Predicting Protein-Protein Interactions Using Sequence and Network Information via Variational Graph Autoencoder. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:3182-3194. [PMID: 37155405 DOI: 10.1109/tcbb.2023.3273567] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Protein-protein interactions (PPIs) play a critical role in the proteomics study, and a variety of computational algorithms have been developed to predict PPIs. Though effective, their performance is constrained by high false-positive and false-negative rates observed in PPI data. To overcome this problem, a novel PPI prediction algorithm, namely PASNVGA, is proposed in this work by combining the sequence and network information of proteins via variational graph autoencoder. To do so, PASNVGA first applies different strategies to extract the features of proteins from their sequence and network information, and obtains a more compact form of these features using principal component analysis. In addition, PASNVGA designs a scoring function to measure the higher-order connectivity between proteins and so as to obtain a higher-order adjacency matrix. With all these features and adjacency matrices, PASNVGA trains a variational graph autoencoder model to further learn the integrated embeddings of proteins. The prediction task is then completed by using a simple feedforward neural network. Extensive experiments have been conducted on five PPI datasets collected from different species. Compared with several state-of-the-art algorithms, PASNVGA has been demonstrated as a promising PPI prediction algorithm.
Collapse
|
77
|
Chen J, Zhang L, Cheng K, Jin B, Lu X, Che C. Predicting Drug-Target Interaction Via Self-Supervised Learning. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:2781-2789. [PMID: 35230952 DOI: 10.1109/tcbb.2022.3153963] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Recent advances in graph representation learning provide new opportunities for computational drug-target interaction (DTI) prediction. However, it still suffers from deficiencies of dependence on manual labels and vulnerability to attacks. Inspired by the success of self-supervised learning (SSL) algorithms, which can leverage input data itself as supervision,we propose SupDTI, a SSL-enhanced drug-target interaction prediction framework based on a heterogeneous network (i.e., drug-protein, drug-drug, and protein-protein interaction network; drug-disease, drug-side-effect, and protein-disease association network; drug-structure and protein-sequence similarity network). Specifically, SupDTI is an end-to-end learning framework consisting of five components. First, localized and globalized graph convolutions are designed to capture the nodes' information from both local and global perspectives, respectively. Then, we develop a variational autoencoder to constrain the nodes' representation to have desired statistical characteristics. Finally, a unified self-supervised learning strategy is leveraged to enhance the nodes' representation, namely, a contrastive learning module is employed to enable the nodes' representation to fit the graph-level representation, followed by a generative learning module which further maximizes the node-level agreement across the global and local views by learning the probabilistic connectivity distribution of the original heterogeneous network. Experimental results show that our model can achieve better prediction performance than state-of-the-art methods.
Collapse
|
78
|
Wang W, Meng X, Xiang J, Shuai Y, Bedru HD, Li M. CACO: A Core-Attachment Method With Cross-Species Functional Ortholog Information to Detect Human Protein Complexes. IEEE J Biomed Health Inform 2023; 27:4569-4578. [PMID: 37399160 DOI: 10.1109/jbhi.2023.3289490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
Protein complexes play an essential role in living cells. Detecting protein complexes is crucial to understand protein functions and treat complex diseases. Due to high time and resource consumption of experiment approaches, many computational approaches have been proposed to detect protein complexes. However, most of them are only based on protein-protein interaction (PPI) networks, which heavily suffer from the noise in PPI networks. Therefore, we propose a novel core-attachment method, named CACO, to detect human protein complexes, by integrating the functional information from other species via protein ortholog relations. First, CACO constructs a cross-species ortholog relation matrix and transfers GO terms from other species as a reference to evaluate the confidence of PPIs. Then, a PPI filter strategy is adopted to clean the PPI network and thus a weighted clean PPI network is constructed. Finally, a new effective core-attachment algorithm is proposed to detect protein complexes from the weighted PPI network. Compared to other thirteen state-of-the-art methods, CACO outperforms all of them in terms of F-measure and Composite Score, showing that integrating ortholog information and the proposed core-attachment algorithm are effective in detecting protein complexes.
Collapse
|
79
|
Zhao BW, Su XR, Hu PW, Huang YA, You ZH, Hu L. iGRLDTI: an improved graph representation learning method for predicting drug-target interactions over heterogeneous biological information network. Bioinformatics 2023; 39:btad451. [PMID: 37505483 PMCID: PMC10397422 DOI: 10.1093/bioinformatics/btad451] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 06/12/2023] [Accepted: 07/27/2023] [Indexed: 07/29/2023] Open
Abstract
MOTIVATION The task of predicting drug-target interactions (DTIs) plays a significant role in facilitating the development of novel drug discovery. Compared with laboratory-based approaches, computational methods proposed for DTI prediction are preferred due to their high-efficiency and low-cost advantages. Recently, much attention has been attracted to apply different graph neural network (GNN) models to discover underlying DTIs from heterogeneous biological information network (HBIN). Although GNN-based prediction methods achieve better performance, they are prone to encounter the over-smoothing simulation when learning the latent representations of drugs and targets with their rich neighborhood information in HBIN, and thereby reduce the discriminative ability in DTI prediction. RESULTS In this work, an improved graph representation learning method, namely iGRLDTI, is proposed to address the above issue by better capturing more discriminative representations of drugs and targets in a latent feature space. Specifically, iGRLDTI first constructs an HBIN by integrating the biological knowledge of drugs and targets with their interactions. After that, it adopts a node-dependent local smoothing strategy to adaptively decide the propagation depth of each biomolecule in HBIN, thus significantly alleviating over-smoothing by enhancing the discriminative ability of feature representations of drugs and targets. Finally, a Gradient Boosting Decision Tree classifier is used by iGRLDTI to predict novel DTIs. Experimental results demonstrate that iGRLDTI yields better performance that several state-of-the-art computational methods on the benchmark dataset. Besides, our case study indicates that iGRLDTI can successfully identify novel DTIs with more distinguishable features of drugs and targets. AVAILABILITY AND IMPLEMENTATION Python codes and dataset are available at https://github.com/stevejobws/iGRLDTI/.
Collapse
Affiliation(s)
- Bo-Wei Zhao
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Xinjiang Laboratory of Minority Speech and Language Information Processing, Urumqi 830011, China
| | - Xiao-Rui Su
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Xinjiang Laboratory of Minority Speech and Language Information Processing, Urumqi 830011, China
| | - Peng-Wei Hu
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Xinjiang Laboratory of Minority Speech and Language Information Processing, Urumqi 830011, China
| | - Yu-An Huang
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710129, China
| | - Zhu-Hong You
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710129, China
| | - Lun Hu
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Xinjiang Laboratory of Minority Speech and Language Information Processing, Urumqi 830011, China
| |
Collapse
|
80
|
Athanasiadis P, Ravikumar B, Elliott RJ, Dawson JC, Carragher NO, Clemons PA, Johanssen T, Ebner D, Aittokallio T. Chemogenomic library design strategies for precision oncology, applied to phenotypic profiling of glioblastoma patient cells. iScience 2023; 26:107209. [PMID: 37485377 PMCID: PMC10359939 DOI: 10.1016/j.isci.2023.107209] [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: 01/29/2023] [Revised: 02/21/2023] [Accepted: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
Designing a targeted screening library of bioactive small molecules is a challenging task since most compounds modulate their effects through multiple protein targets with varying degrees of potency and selectivity. We implemented analytic procedures for designing anticancer compound libraries adjusted for library size, cellular activity, chemical diversity and availability, and target selectivity. The resulting compound collections cover a wide range of protein targets and biological pathways implicated in various cancers, making them widely applicable to precision oncology. We characterized the compound and target spaces of the virtual libraries, in comparison with a minimal screening library of 1,211 compounds for targeting 1,386 anticancer proteins. In a pilot screening study, we identified patient-specific vulnerabilities by imaging glioma stem cells from patients with glioblastoma (GBM), using a physical library of 789 compounds that cover 1,320 of the anticancer targets. The cell survival profiling revealed highly heterogeneous phenotypic responses across the patients and GBM subtypes.
Collapse
Affiliation(s)
- Paschalis Athanasiadis
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, 0310 Oslo, Norway
- Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, 0317 Oslo, Norway
| | - Balaguru Ravikumar
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, 20520 00290 Helsinki, Finland
| | - Richard J.R. Elliott
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XR, UK
| | - John C. Dawson
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XR, UK
| | - Neil O. Carragher
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XR, UK
| | - Paul A. Clemons
- Chemical Biology and Therapeutics Science Program, Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA 02142, United States
| | - Timothy Johanssen
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Daniel Ebner
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Tero Aittokallio
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, 0310 Oslo, Norway
- Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, 0317 Oslo, Norway
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, 20520 00290 Helsinki, Finland
| |
Collapse
|
81
|
Gutiérrez-Casares JR, Segú-Vergés C, Sabate Chueca J, Pozo-Rubio T, Coma M, Montoto C, Quintero J. In silico evaluation of the role of lisdexamfetamine on attention-deficit/hyperactivity disorder common psychiatric comorbidities: mechanistic insights on binge eating disorder and depression. Front Neurosci 2023; 17:1118253. [PMID: 37457000 PMCID: PMC10347683 DOI: 10.3389/fnins.2023.1118253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a psychiatric condition well recognized in the pediatric population that can persist into adulthood. The vast majority of patients with ADHD present psychiatric comorbidities that have been suggested to share, to some extent, the pathophysiological mechanism of ADHD. Lisdexamfetamine (LDX) is a stimulant prodrug approved for treating ADHD and, in the US, also for binge eating disorder (BED). Herein, we evaluated, through a systems biology-based in silico method, the efficacy of a virtual model of LDX (vLDX) as ADHD treatment to improve five common ADHD psychiatric comorbidities in adults and children, and we explored the molecular mechanisms behind LDX's predicted efficacy. After the molecular characterization of vLDX and the comorbidities (anxiety, BED, bipolar disorder, depression, and tics disorder), we created a protein-protein interaction human network to which we applied artificial neural networks (ANN) algorithms. We also generated virtual populations of adults and children-adolescents totaling 2,600 individuals and obtained the predicted protein activity from Therapeutic Performance Mapping System models. The latter showed that ADHD molecular description shared 53% of its protein effectors with at least one studied psychiatric comorbidity. According to the ANN analysis, proteins targeted by vLDX are predicted to have a high probability of being related to BED and depression. In BED, vLDX was modeled to act upon neurotransmission and neuroplasticity regulators, and, in depression, vLDX regulated the hypothalamic-pituitary-adrenal axis, neuroinflammation, oxidative stress, and glutamatergic excitotoxicity. In conclusion, our modeling results, despite their limitations and although requiring in vitro or in vivo validation, could supplement the design of preclinical and potentially clinical studies that investigate treatment for patients with ADHD with psychiatric comorbidities, especially from a molecular point of view.
Collapse
Affiliation(s)
- José Ramón Gutiérrez-Casares
- Unidad Ambulatoria de Psiquiatría y Salud Mental de la Infancia, Niñez y Adolescencia, Hospital Perpetuo Socorro, Badajoz, Spain
| | - Cristina Segú-Vergés
- Anaxomics Biotech, Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | | | | | | | - Carmen Montoto
- Department of Medical, Takeda Farmacéutica España, Madrid, Spain
| | - Javier Quintero
- Servicio de Psiquiatría, Hospital Universitario Infanta Leonor, Departamento de Medicina Legal, Patología y Psiquiatría, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
82
|
Veenstra BT, Veenstra TD. Proteomic applications in identifying protein-protein interactions. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 138:1-48. [PMID: 38220421 DOI: 10.1016/bs.apcsb.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
There are many things that can be used to characterize a protein. Size, isoelectric point, hydrophobicity, structure (primary to quaternary), and subcellular location are just a few parameters that are used. The most important feature of a protein, however, is its function. While there are many experiments that can indicate a protein's role, identifying the molecules it interacts with is probably the most definitive way of determining its function. Owing to technology limitations, protein interactions have historically been identified on a one molecule per experiment basis. The advent of high throughput multiplexed proteomic technologies in the 1990s, however, made identifying hundreds and thousands of proteins interactions within single experiments feasible. These proteomic technologies have dramatically increased the rate at which protein-protein interactions (PPIs) are discovered. While the improvement in mass spectrometry technology was an early driving force in the rapid pace of identifying PPIs, advances in sample preparation and chromatography have recently been propelling the field. In this chapter, we will discuss the importance of identifying PPIs and describe current state-of-the-art technologies that demonstrate what is currently possible in this important area of biological research.
Collapse
Affiliation(s)
- Benjamin T Veenstra
- Department of Math and Sciences, Cedarville University, Cedarville, OH, United States
| | - Timothy D Veenstra
- School of Pharmacy, Cedarville University, Cedarville, OH, United States.
| |
Collapse
|
83
|
Madugula SS, Pandey S, Amalapurapu S, Bozdag S. NRPreTo: A Machine Learning-Based Nuclear Receptor and Subfamily Prediction Tool. ACS OMEGA 2023; 8:20379-20388. [PMID: 37323377 PMCID: PMC10268018 DOI: 10.1021/acsomega.3c00286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 05/09/2023] [Indexed: 06/17/2023]
Abstract
The nuclear receptor (NR) superfamily includes phylogenetically related ligand-activated proteins, which play a key role in various cellular activities. NR proteins are subdivided into seven subfamilies based on their function, mechanism, and nature of the interacting ligand. Developing robust tools to identify NR could give insights into their functional relationships and involvement in disease pathways. Existing NR prediction tools only use a few types of sequence-based features and are tested on relatively similar independent datasets; thus, they may suffer from overfitting when extended to new genera of sequences. To address this problem, we developed Nuclear Receptor Prediction Tool (NRPreTo), a two-level NR prediction tool with a unique training approach where in addition to the sequence-based features used by existing NR prediction tools, six additional feature groups depicting various physiochemical, structural, and evolutionary features of proteins were utilized. The first level of NRPreTo allows for the successful prediction of a query protein as NR or non-NR and further subclassifies the protein into one of the seven NR subfamilies in the second level. We developed Random Forest classifiers to test on benchmark datasets, as well as the entire human protein datasets from RefSeq and Human Protein Reference Database (HPRD). We observed that using additional feature groups improved the performance. We also observed that NRPreTo achieved high performance on the external datasets and predicted 59 novel NRs in the human proteome. The source code of NRPreTo is publicly available at https://github.com/bozdaglab/NRPreTo.
Collapse
Affiliation(s)
- Sita Sirisha Madugula
- Department
of Computer Science & Engineering, University
of North Texas, Denton, Texas TX 76203, United States
| | - Suman Pandey
- Department
of Computer Science & Engineering, University
of North Texas, Denton, Texas TX 76203, United States
| | - Shreya Amalapurapu
- Department
of Computer Science & Engineering, University
of North Texas, Denton, Texas TX 76203, United States
- The
Texas Academy of Mathematics and Science, University of North Texas, Denton, Texas TX 76203, United States
| | - Serdar Bozdag
- Department
of Computer Science & Engineering, University
of North Texas, Denton, Texas TX 76203, United States
- Department
of Mathematics, University of North Texas, Denton, Texas TX 76203, United
States
- BioDiscovery
Institute, University of North Texas, Denton, Texas TX 76203, United States
| |
Collapse
|
84
|
Mo J, Li Z, Chen H, Lu Z, Ding B, Yuan X, Liu Y, Zhu W. Network medicine framework identified drug-repurposing opportunities of pharmaco-active compounds of Angelica acutiloba (Siebold & Zucc.) Kitag. for skin aging. Aging (Albany NY) 2023; 15:5144-5163. [PMID: 37310405 PMCID: PMC10292898 DOI: 10.18632/aging.204789] [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: 02/06/2023] [Accepted: 05/15/2023] [Indexed: 06/14/2023]
Abstract
Increasing incidence of skin aging has highlighted the importance of identifying effective drugs with repurposed opportunities for skin aging. We aimed to identify pharmaco-active compounds with drug-repurposing opportunities for skin aging from Angelica acutiloba (Siebold & Zucc.) Kitag. (AAK). The proximity of network medicine framework (NMF) firstly identified 8 key AAK compounds with repurposed opportunities for skin aging, which may exert by regulating 29 differentially expressed genes (DGEs) of skin aging, including 13 up-regulated targets and 16 down-regulated targets. Connectivity MAP (cMAP) analysis revealed 8 key compounds were involved in regulating the process of cell proliferation and apoptosis, mitochondrial energy metabolism and oxidative stress of skin aging. Molecular docking analysis showed that 8 key compounds had a high docked ability with AR, BCHE, HPGD and PI3, which were identified as specific biomarker for the diagnosis of skin aging. Finally, the mechanisms of these key compounds were predicted to be involved in inhibiting autophagy pathway and activating Phospholipase D signaling pathway. In conclusion, this study firstly elucidated the drug-repurposing opportunities of AAK compounds for skin aging, providing a theoretical reference for identifying repurposing drugs from Chinese medicine and new insights for our future research.
Collapse
Affiliation(s)
- Jiaxin Mo
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou Province 510006, China
| | - Zunjiang Li
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou Province 510006, China
| | - Hankun Chen
- Guangzhou Qinglan Biotechnology Co. Ltd., Guangzhou Province 515000, China
| | - Zhongyu Lu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou Province 510006, China
| | - Banghan Ding
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou Province 510006, China
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou Province 510120, China
| | - Xiaohong Yuan
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou Province 510120, China
| | - Yuan Liu
- Guangzhou Huamiao Biotechnology Research Institute Co. Ltd., Guangzhou Province 510000, China
| | - Wei Zhu
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou Province 510120, China
| |
Collapse
|
85
|
Zhao L, Zhang H, Li N, Chen J, Xu H, Wang Y, Liang Q. Network pharmacology, a promising approach to reveal the pharmacology mechanism of Chinese medicine formula. JOURNAL OF ETHNOPHARMACOLOGY 2023; 309:116306. [PMID: 36858276 DOI: 10.1016/j.jep.2023.116306] [Citation(s) in RCA: 199] [Impact Index Per Article: 199.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/06/2023] [Accepted: 02/19/2023] [Indexed: 05/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Network pharmacology is a new discipline based on systems biology theory, biological system network analysis, and multi-target drug molecule design specific signal node selection. The mechanism of action of TCM formula has the characteristics of multiple targets and levels. The mechanism is similar to the integrity, systematization and comprehensiveness of network pharmacology, so network pharmacology is suitable for the study of the pharmacological mechanism of Chinese medicine compounds. AIM OF THE STUDY The paper summarizes the present application status and existing problems of network pharmacology in the field of Chinese medicine formula, and formulates the research ideas, up-to-date key technology and application method and strategy of network pharmacology. Its purpose is to provide guidance and reference for using network pharmacology to reveal the modern scientific connotation of Chinese medicine. MATERIALS AND METHODS Literatures in this review were searched in PubMed, China National Knowledge Infrastructure (CNKI), Web of Science, ScienceDirect and Google Scholar using the keywords "traditional Chinese medicine", "Chinese herb medicine" and "network pharmacology". The literature cited in this review dates from 2002 to 2022. RESULTS Using network pharmacology methods to predict the basis and mechanism of pharmacodynamic substances of traditional Chinese medicines has become a trend. CONCLUSION Network pharmacology is a promising approach to reveal the pharmacology mechanism of Chinese medicine formula.
Collapse
Affiliation(s)
- Li Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Key Laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Hong Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Key Laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Ning Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Key Laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Jinman Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Key Laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Key Laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Key Laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Key Laboratory of Ministry of Education of Theory and Therapy of Muscles and Bones, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
86
|
Zabihian A, Sayyad FZ, Hashemi SM, Shami Tanha R, Hooshmand M, Gharaghani S. DEDTI versus IEDTI: efficient and predictive models of drug-target interactions. Sci Rep 2023; 13:9238. [PMID: 37286613 DOI: 10.1038/s41598-023-36438-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/03/2023] [Indexed: 06/09/2023] Open
Abstract
Drug repurposing is an active area of research that aims to decrease the cost and time of drug development. Most of those efforts are primarily concerned with the prediction of drug-target interactions. Many evaluation models, from matrix factorization to more cutting-edge deep neural networks, have come to the scene to identify such relations. Some predictive models are devoted to the prediction's quality, and others are devoted to the efficiency of the predictive models, e.g., embedding generation. In this work, we propose new representations of drugs and targets useful for more prediction and analysis. Using these representations, we propose two inductive, deep network models of IEDTI and DEDTI for drug-target interaction prediction. Both of them use the accumulation of new representations. The IEDTI takes advantage of triplet and maps the input accumulated similarity features into meaningful embedding corresponding vectors. Then, it applies a deep predictive model to each drug-target pair to evaluate their interaction. The DEDTI directly uses the accumulated similarity feature vectors of drugs and targets and applies a predictive model on each pair to identify their interactions. We have done a comprehensive simulation on the DTINet dataset as well as gold standard datasets, and the results show that DEDTI outperforms IEDTI and the state-of-the-art models. In addition, we conduct a docking study on new predicted interactions between two drug-target pairs, and the results confirm acceptable drug-target binding affinity between both predicted pairs.
Collapse
Affiliation(s)
- Arash Zabihian
- Laboratory of Bioinformatics and Drug Design (LBD), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- Department of Bioinformatics, Kish International Campus, University of Tehran, Kish, Iran
| | - Faeze Zakaryapour Sayyad
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Seyyed Morteza Hashemi
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Reza Shami Tanha
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Mohsen Hooshmand
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran.
| | - Sajjad Gharaghani
- Laboratory of Bioinformatics and Drug Design (LBD), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
87
|
Misetic H, Keddar MR, Jeannon JP, Ciccarelli FD. Mechanistic insights into the interactions between cancer drivers and the tumour immune microenvironment. Genome Med 2023; 15:40. [PMID: 37277866 DOI: 10.1186/s13073-023-01197-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 05/25/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND The crosstalk between cancer and the tumour immune microenvironment (TIME) has attracted significant interest in the latest years because of its impact on cancer evolution and response to treatment. Despite this, cancer-specific tumour-TIME interactions and their mechanistic insights are still poorly understood. METHODS Here, we compute the significant interactions occurring between cancer-specific genetic drivers and five anti- and pro-tumour TIME features in 32 cancer types using Lasso regularised ordinal regression. Focusing on head and neck squamous cancer (HNSC), we rebuild the functional networks linking specific TIME driver alterations to the TIME state they associate with. RESULTS The 477 TIME drivers that we identify are multifunctional genes whose alterations are selected early in cancer evolution and recur across and within cancer types. Tumour suppressors and oncogenes have an opposite effect on the TIME and the overall anti-tumour TIME driver burden is predictive of response to immunotherapy. TIME driver alterations predict the immune profiles of HNSC molecular subtypes, and perturbations in keratinization, apoptosis and interferon signalling underpin specific driver-TIME interactions. CONCLUSIONS Overall, our study delivers a comprehensive resource of TIME drivers, gives mechanistic insights into their immune-regulatory role, and provides an additional framework for patient prioritisation to immunotherapy. The full list of TIME drivers and associated properties are available at http://www.network-cancer-genes.org .
Collapse
Affiliation(s)
- Hrvoje Misetic
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, London, SE11UL, UK
| | - Mohamed Reda Keddar
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, London, SE11UL, UK
| | - Jean-Pierre Jeannon
- Department of Head & Neck Surgery, Great Maze Pond, Guy's Hospital, London, SE1 9RT, UK
| | - Francesca D Ciccarelli
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK.
- School of Cancer and Pharmaceutical Sciences, King's College London, London, SE11UL, UK.
| |
Collapse
|
88
|
Bhaoighill MN, Falcón‐Pérez JM, Royo F, Tee AR, Webber JP, Dunlop EA. Tuberous Sclerosis Complex cell-derived EVs have an altered protein cargo capable of regulating their microenvironment and have potential as disease biomarkers. J Extracell Vesicles 2023; 12:e12336. [PMID: 37337371 PMCID: PMC10279809 DOI: 10.1002/jev2.12336] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 04/12/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023] Open
Abstract
Hyperactivation of mechanistic target of rapamycin complex 1 (mTORC1) is a feature of many solid tumours and is a key pathogenic driver in the inherited condition Tuberous Sclerosis Complex (TSC). Modulation of the tumour microenvironment by extracellular vesicles (EVs) is known to facilitate the development of various cancers. The role of EVs in modulating the tumour microenvironment and their impact on the development of TSC tumours, however, remains unclear. This study, therefore, focuses on the poorly defined contribution of EVs to tumour growth in TSC. We characterised EVs secreted from TSC2-deficient and TSC2-expressing cells and identified a distinct protein cargo in TSC2-deficient EVs, containing an enrichment of proteins thought to be involved in tumour-supporting signalling pathways. We show EVs from TSC2-deficient cells promote cell viability, proliferation and growth factor secretion from recipient fibroblasts within the tumour microenvironment. Rapalogs (mTORC1 inhibitors) are the current therapy for TSC tumours. Here, we demonstrate a previously unknown intercellular therapeutic effect of rapamycin in altering EV cargo and reducing capacity to promote cell proliferation in the tumour microenvironment. Furthermore, EV cargo proteins have the potential for clinical applications as TSC biomarkers, and we reveal three EV-associated proteins that are elevated in plasma from TSC patients compared to healthy donor plasma.
Collapse
Affiliation(s)
- Muireann Ní Bhaoighill
- Tissue Microenvironment GroupSchool of MedicineCardiff UniversityCardiffUK
- Division of Cancer and GeneticsSchool of MedicineCardiff UniversityCardiffUK
| | - Juan M. Falcón‐Pérez
- Exosomes Lab. CICbioGUNE‐BRTAParque TecnologicoDerioSpain
- Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd)MadridSpain
- IKERBASQUEBasque Foundation for ScienceBilbaoSpain
| | - Félix Royo
- Exosomes Lab. CICbioGUNE‐BRTAParque TecnologicoDerioSpain
- Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd)MadridSpain
| | - Andrew R. Tee
- Division of Cancer and GeneticsSchool of MedicineCardiff UniversityCardiffUK
| | - Jason P. Webber
- Tissue Microenvironment GroupSchool of MedicineCardiff UniversityCardiffUK
- Institute of Life ScienceSwansea University Medical SchoolSwansea UniversitySwanseaUK
| | - Elaine A. Dunlop
- Division of Cancer and GeneticsSchool of MedicineCardiff UniversityCardiffUK
| |
Collapse
|
89
|
Sarkar S, Lucchetta M, Maier A, Abdrabbou MM, Baumbach J, List M, Schaefer MH, Blumenthal DB. Online bias-aware disease module mining with ROBUST-Web. Bioinformatics 2023; 39:btad345. [PMID: 37233198 PMCID: PMC10246579 DOI: 10.1093/bioinformatics/btad345] [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: 11/14/2022] [Revised: 04/24/2023] [Accepted: 05/25/2023] [Indexed: 05/27/2023] Open
Abstract
SUMMARY We present ROBUST-Web which implements our recently presented ROBUST disease module mining algorithm in a user-friendly web application. ROBUST-Web features seamless downstream disease module exploration via integrated gene set enrichment analysis, tissue expression annotation, and visualization of drug-protein and disease-gene links. Moreover, ROBUST-Web includes bias-aware edge costs for the underlying Steiner tree model as a new algorithmic feature, which allow to correct for study bias in protein-protein interaction networks and further improves the robustness of the computed modules. AVAILABILITY AND IMPLEMENTATION Web application: https://robust-web.net. Source code of web application and Python package with new bias-aware edge costs: https://github.com/bionetslab/robust-web, https://github.com/bionetslab/robust_bias_aware.
Collapse
Affiliation(s)
- Suryadipto Sarkar
- Biomedical Network Science Lab, Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91301, Germany
| | - Marta Lucchetta
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan 20139, Italy
| | - Andreas Maier
- Institute for Computational Systems Biology, University of Hamburg, Hamburg 22607, Germany
| | - Mohamed M Abdrabbou
- Biomedical Network Science Lab, Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91301, Germany
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, Hamburg 22607, Germany
| | - Markus List
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Martin H Schaefer
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan 20139, Italy
| | - David B Blumenthal
- Biomedical Network Science Lab, Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91301, Germany
| |
Collapse
|
90
|
Chakraborty A, Mitra S, Bhattacharjee M, De D, Pal AJ. Determining human-coronavirus protein-protein interaction using machine intelligence. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2023; 18:100228. [PMID: 37056696 PMCID: PMC10077817 DOI: 10.1016/j.medntd.2023.100228] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/29/2023] [Accepted: 04/01/2023] [Indexed: 04/08/2023] Open
Abstract
The Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2) virus spread the novel CoronaVirus -19 (nCoV-19) pandemic, resulting in millions of fatalities globally. Recent research demonstrated that the Protein-Protein Interaction (PPI) between SARS-CoV-2 and human proteins is accountable for viral pathogenesis. However, many of these PPIs are poorly understood and unexplored, necessitating a more in-depth investigation to find latent yet critical interactions. This article elucidates the host-viral PPI through Machine Learning (ML) lenses and validates the biological significance of the same using web-based tools. ML classifiers are designed based on comprehensive datasets with five sequence-based features of human proteins, namely Amino Acid Composition, Pseudo Amino Acid Composition, Conjoint Triad, Dipeptide Composition, and Normalized Auto Correlation. A majority voting rule-based ensemble method composed of the Random Forest Model (RFM), AdaBoost, and Bagging technique is proposed that delivers encouraging statistical performance compared to other models employed in this work. The proposed ensemble model predicted a total of 111 possible SARS-CoV-2 human target proteins with a high likelihood factor ≥70%, validated by utilizing Gene Ontology (GO) and KEGG pathway enrichment analysis. Consequently, this research can aid in a deeper understanding of the molecular mechanisms underlying viral pathogenesis and provide clues for developing more efficient anti-COVID medications.
Collapse
Affiliation(s)
- Arijit Chakraborty
- Bachelor of Computer Application Department, The Heritage Academy, Kolkata, India
| | - Sajal Mitra
- Department of Computer Science and Engineering, Heritage Institute of Technology, Kolkata, India
| | | | - Debashis De
- Department of Computer Science and Engineering, Maulana Abul Kalam Azad University of Technology, Kolkata, India
| | | |
Collapse
|
91
|
Faessler E, Hahn U, Schäuble S. GePI: large-scale text mining, customized retrieval and flexible filtering of gene/protein interactions. Nucleic Acids Res 2023:7177881. [PMID: 37224532 DOI: 10.1093/nar/gkad445] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/01/2023] [Accepted: 05/11/2023] [Indexed: 05/26/2023] Open
Abstract
We present GePI, a novel Web server for large-scale text mining of molecular interactions from the scientific biomedical literature. GePI leverages natural language processing techniques to identify genes and related entities, interactions between those entities and biomolecular events involving them. GePI supports rapid retrieval of interactions based on powerful search options to contextualize queries targeting (lists of) genes of interest. Contextualization is enabled by full-text filters constraining the search for interactions to either sentences or paragraphs, with or without pre-defined gene lists. Our knowledge graph is updated several times a week ensuring the most recent information to be available at all times. The result page provides an overview of the outcome of a search, with accompanying interaction statistics and visualizations. A table (downloadable in Excel format) gives direct access to the retrieved interaction pairs, together with information about the molecular entities, the factual certainty of the interactions (as verbatim expressed by the authors), and a text snippet from the original document that verbalizes each interaction. In summary, our Web application offers free, easy-to-use, and up-to-date monitoring of gene and protein interaction information, in company with flexible query formulation and filtering options. GePI is available at https://gepi.coling.uni-jena.de/.
Collapse
Affiliation(s)
- Erik Faessler
- Jena University Language and Information Engineering (JULIE) Lab, Friedrich Schiller University Jena, Fürstengraben 30, 07743 Jena, Germany
| | - Udo Hahn
- Jena University Language and Information Engineering (JULIE) Lab, Friedrich Schiller University Jena, Fürstengraben 30, 07743 Jena, Germany
| | - Sascha Schäuble
- Jena University Language and Information Engineering (JULIE) Lab, Friedrich Schiller University Jena, Fürstengraben 30, 07743 Jena, Germany
- Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745 Jena, Germany
| |
Collapse
|
92
|
Yue Y, McDonald D, Hao L, Lei H, Butler MS, He S. FLONE: fully Lorentz network embedding for inferring novel drug targets. BIOINFORMATICS ADVANCES 2023; 3:vbad066. [PMID: 37275772 PMCID: PMC10235194 DOI: 10.1093/bioadv/vbad066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/11/2023] [Accepted: 05/23/2023] [Indexed: 06/07/2023]
Abstract
Motivation To predict drug targets, graph-based machine-learning methods have been widely used to capture the relationships between drug, target and disease entities in drug-disease-target (DDT) networks. However, many methods cannot explicitly consider disease types at inference time and so will predict the same target for a given drug under any disease condition. Meanwhile, DDT networks are usually organized hierarchically carrying interactive relationships between involved entities, but these methods, especially those based on Euclidean embedding cannot fully utilize such topological information, which might lead to sub-optimal results. We hypothesized that, by importing hyperbolic embedding specifically for modeling hierarchical DDT networks, graph-based algorithms could better capture relationships between aforementioned entities, which ultimately improves target prediction performance. Results We formulated the target prediction problem as a knowledge graph completion task explicitly considering disease types. We proposed FLONE, a hyperbolic embedding-based method based on capturing hierarchical topological information in DDT networks. The experimental results on two DDT networks showed that by introducing hyperbolic space, FLONE generates more accurate target predictions than its Euclidean counterparts, which supports our hypothesis. We also devised hyperbolic encoders to fuse external domain knowledge, to make FLONE enable handling samples corresponding to previously unseen drugs and targets for more practical scenarios. Availability and implementation Source code and dataset information are at: https://github.com/arantir123/DDT_triple_prediction. Supplementary information Supplementary data are available at Bioinformatics Advances online.
Collapse
Affiliation(s)
- Yang Yue
- Centre for Computational Biology, School of Computer Science, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - David McDonald
- AIA Insights Ltd., 71-75 Shelton Street, London, Greater London, WC2H 9JQ, UK
| | - Luoying Hao
- Centre for Computational Biology, School of Computer Science, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Huangshu Lei
- YaoPharma Co., Ltd., 100 Xingguang Avenue, Renhe Town, Yubei District, Chongqing, 401121, China
| | - Mark S Butler
- AIA Insights Ltd., 71-75 Shelton Street, London, Greater London, WC2H 9JQ, UK
| | - Shan He
- Centre for Computational Biology, School of Computer Science, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| |
Collapse
|
93
|
Wysocka M, Wysocki O, Zufferey M, Landers D, Freitas A. A systematic review of biologically-informed deep learning models for cancer: fundamental trends for encoding and interpreting oncology data. BMC Bioinformatics 2023; 24:198. [PMID: 37189058 PMCID: PMC10186658 DOI: 10.1186/s12859-023-05262-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 03/30/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND There is an increasing interest in the use of Deep Learning (DL) based methods as a supporting analytical framework in oncology. However, most direct applications of DL will deliver models with limited transparency and explainability, which constrain their deployment in biomedical settings. METHODS This systematic review discusses DL models used to support inference in cancer biology with a particular emphasis on multi-omics analysis. It focuses on how existing models address the need for better dialogue with prior knowledge, biological plausibility and interpretability, fundamental properties in the biomedical domain. For this, we retrieved and analyzed 42 studies focusing on emerging architectural and methodological advances, the encoding of biological domain knowledge and the integration of explainability methods. RESULTS We discuss the recent evolutionary arch of DL models in the direction of integrating prior biological relational and network knowledge to support better generalisation (e.g. pathways or Protein-Protein-Interaction networks) and interpretability. This represents a fundamental functional shift towards models which can integrate mechanistic and statistical inference aspects. We introduce a concept of bio-centric interpretability and according to its taxonomy, we discuss representational methodologies for the integration of domain prior knowledge in such models. CONCLUSIONS The paper provides a critical outlook into contemporary methods for explainability and interpretability used in DL for cancer. The analysis points in the direction of a convergence between encoding prior knowledge and improved interpretability. We introduce bio-centric interpretability which is an important step towards formalisation of biological interpretability of DL models and developing methods that are less problem- or application-specific.
Collapse
Affiliation(s)
- Magdalena Wysocka
- Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, CRUK Manchester Institute, University of Manchester, Oxford Rd, Manchester, M13 9 PL UK
- Department of Computer Science, University of Manchester, Oxford Rd, Manchester, M13 9 PL UK
| | - Oskar Wysocki
- Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, CRUK Manchester Institute, University of Manchester, Oxford Rd, Manchester, M13 9 PL UK
- Department of Computer Science, University of Manchester, Oxford Rd, Manchester, M13 9 PL UK
- Idiap Research Institute, National University of Sciences, Rue Marconi 19, CH - 1920 Martigny, Switzerland
| | - Marie Zufferey
- Idiap Research Institute, National University of Sciences, Rue Marconi 19, CH - 1920 Martigny, Switzerland
| | - Dónal Landers
- DeLondra Oncology Ltd, 38 Carlton Avenue, Wilmslow, SK9 4EP UK
| | - André Freitas
- Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, CRUK Manchester Institute, University of Manchester, Oxford Rd, Manchester, M13 9 PL UK
- Department of Computer Science, University of Manchester, Oxford Rd, Manchester, M13 9 PL UK
- Idiap Research Institute, National University of Sciences, Rue Marconi 19, CH - 1920 Martigny, Switzerland
| |
Collapse
|
94
|
Varshney N, Mishra AK. Deep Learning in Phosphoproteomics: Methods and Application in Cancer Drug Discovery. Proteomes 2023; 11:proteomes11020016. [PMID: 37218921 DOI: 10.3390/proteomes11020016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023] Open
Abstract
Protein phosphorylation is a key post-translational modification (PTM) that is a central regulatory mechanism of many cellular signaling pathways. Several protein kinases and phosphatases precisely control this biochemical process. Defects in the functions of these proteins have been implicated in many diseases, including cancer. Mass spectrometry (MS)-based analysis of biological samples provides in-depth coverage of phosphoproteome. A large amount of MS data available in public repositories has unveiled big data in the field of phosphoproteomics. To address the challenges associated with handling large data and expanding confidence in phosphorylation site prediction, the development of many computational algorithms and machine learning-based approaches have gained momentum in recent years. Together, the emergence of experimental methods with high resolution and sensitivity and data mining algorithms has provided robust analytical platforms for quantitative proteomics. In this review, we compile a comprehensive collection of bioinformatic resources used for the prediction of phosphorylation sites, and their potential therapeutic applications in the context of cancer.
Collapse
Affiliation(s)
- Neha Varshney
- Division of Biological Sciences, Department of Cellular and Molecular Medicine, University of California, San Diego, CA 93093, USA
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - Abhinava K Mishra
- Molecular, Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
95
|
Dong W, Yang Q, Wang J, Xu L, Li X, Luo G, Gao X. Multi-modality attribute learning-based method for drug-protein interaction prediction based on deep neural network. Brief Bioinform 2023; 24:7145903. [PMID: 37114624 DOI: 10.1093/bib/bbad161] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/19/2023] [Accepted: 04/02/2023] [Indexed: 04/29/2023] Open
Abstract
Identification of active candidate compounds for target proteins, also called drug-protein interaction (DPI) prediction, is an essential but time-consuming and expensive step, which leads to fostering the development of drug discovery. In recent years, deep network-based learning methods were frequently proposed in DPIs due to their powerful capability of feature representation. However, the performance of existing DPI methods is still limited by insufficiently labeled pharmacological data and neglected intermolecular information. Therefore, overcoming these difficulties to perfect the performance of DPIs is an urgent challenge for researchers. In this article, we designed an innovative 'multi-modality attributes' learning-based framework for DPIs with molecular transformer and graph convolutional networks, termed, multi-modality attributes (MMA)-DPI. Specifically, intermolecular sub-structural information and chemical semantic representations were extracted through an augmented transformer module from biomedical data. A tri-layer graph convolutional neural network module was applied to associate the neighbor topology information and learn the condensed dimensional features by aggregating a heterogeneous network that contains multiple biological representations of drugs, proteins, diseases and side effects. Then, the learned representations were taken as the input of a fully connected neural network module to further integrate them in molecular and topological space. Finally, the attribute representations were fused with adaptive learning weights to calculate the interaction score for the DPIs tasks. MMA-DPI was evaluated in different experimental conditions and the results demonstrate that the proposed method achieved higher performance than existing state-of-the-art frameworks.
Collapse
Affiliation(s)
- Weihe Dong
- College of information and Computer Engineering, Northeast Forestry University, Hexing Road, 150040, Harbin, China
| | - Qiang Yang
- School of Computer Science and Technology, Heilongjiang University, Xuefu Road, 150080, Harbin, China
- Postdoctoral Program of Heilongjiang Hengxun Technology Co., Ltd., Xuefu Road, 150080, Harbin, China
| | - Jian Wang
- College of information and Computer Engineering, Northeast Forestry University, Hexing Road, 150040, Harbin, China
| | - Long Xu
- School of Computer Science and Technology, Heilongjiang University, Xuefu Road, 150080, Harbin, China
- Postdoctoral Program of Heilongjiang Hengxun Technology Co., Ltd., Xuefu Road, 150080, Harbin, China
| | - Xiaokun Li
- School of Computer Science and Technology, Heilongjiang University, Xuefu Road, 150080, Harbin, China
- Postdoctoral Program of Heilongjiang Hengxun Technology Co., Ltd., Xuefu Road, 150080, Harbin, China
| | - Gongning Luo
- Computer, Electrical and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal 23955, Saudi Arabia
- School of Computer Science and Technology, Harbin Institute of Technology, West Dazhi Street, 150001, Harbin, China
| | - Xin Gao
- Computer, Electrical and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal 23955, Saudi Arabia
| |
Collapse
|
96
|
Xie D, Huang Q, Zhou P. Drug Discovery Targeting Post-Translational Modifications in Response to DNA Damages Induced by Space Radiation. Int J Mol Sci 2023; 24:ijms24087656. [PMID: 37108815 PMCID: PMC10142602 DOI: 10.3390/ijms24087656] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/07/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
DNA damage in astronauts induced by cosmic radiation poses a major barrier to human space exploration. Cellular responses and repair of the most lethal DNA double-strand breaks (DSBs) are crucial for genomic integrity and cell survival. Post-translational modifications (PTMs), including phosphorylation, ubiquitylation, and SUMOylation, are among the regulatory factors modulating a delicate balance and choice between predominant DSB repair pathways, such as non-homologous end joining (NHEJ) and homologous recombination (HR). In this review, we focused on the engagement of proteins in the DNA damage response (DDR) modulated by phosphorylation and ubiquitylation, including ATM, DNA-PKcs, CtIP, MDM2, and ubiquitin ligases. The involvement and function of acetylation, methylation, PARylation, and their essential proteins were also investigated, providing a repository of candidate targets for DDR regulators. However, there is a lack of radioprotectors in spite of their consideration in the discovery of radiosensitizers. We proposed new perspectives for the research and development of future agents against space radiation by the systematic integration and utilization of evolutionary strategies, including multi-omics analyses, rational computing methods, drug repositioning, and combinations of drugs and targets, which may facilitate the use of radioprotectors in practical applications in human space exploration to combat fatal radiation hazards.
Collapse
Affiliation(s)
- Dafei Xie
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Taiping Road 27th, Haidian District, Beijing 100850, China
| | - Qi Huang
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Taiping Road 27th, Haidian District, Beijing 100850, China
- Department of Preventive Medicine, School of Public Health, University of South China, Changsheng West Road 28th, Zhengxiang District, Hengyang 421001, China
| | - Pingkun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Taiping Road 27th, Haidian District, Beijing 100850, China
- Department of Preventive Medicine, School of Public Health, University of South China, Changsheng West Road 28th, Zhengxiang District, Hengyang 421001, China
| |
Collapse
|
97
|
Eiger DS, Smith JS, Shi T, Stepniewski TM, Tsai CF, Honeycutt C, Boldizsar N, Gardner J, Nicora CD, Moghieb AM, Kawakami K, Choi I, Hicks C, Zheng K, Warman A, Alagesan P, Knape NM, Huang O, Silverman JD, Smith RD, Inoue A, Selent J, Jacobs JM, Rajagopal S. Phosphorylation barcodes direct biased chemokine signaling at CXCR3. Cell Chem Biol 2023; 30:362-382.e8. [PMID: 37030291 PMCID: PMC10147449 DOI: 10.1016/j.chembiol.2023.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 02/10/2023] [Accepted: 03/13/2023] [Indexed: 04/10/2023]
Abstract
G protein-coupled receptor (GPCR)-biased agonism, selective activation of certain signaling pathways relative to others, is thought to be directed by differential GPCR phosphorylation "barcodes." At chemokine receptors, endogenous chemokines can act as "biased agonists", which may contribute to the limited success when pharmacologically targeting these receptors. Here, mass spectrometry-based global phosphoproteomics revealed that CXCR3 chemokines generate different phosphorylation barcodes associated with differential transducer activation. Chemokine stimulation resulted in distinct changes throughout the kinome in global phosphoproteomics studies. Mutation of CXCR3 phosphosites altered β-arrestin 2 conformation in cellular assays and was consistent with conformational changes observed in molecular dynamics simulations. T cells expressing phosphorylation-deficient CXCR3 mutants resulted in agonist- and receptor-specific chemotactic profiles. Our results demonstrate that CXCR3 chemokines are non-redundant and act as biased agonists through differential encoding of phosphorylation barcodes, leading to distinct physiological processes.
Collapse
Affiliation(s)
- Dylan S Eiger
- Department of Biochemistry, Duke University, Durham, NC 27710, USA
| | - Jeffrey S Smith
- Department of Dermatology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Dermatology, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Dermatology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Dermatology Program, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Tujin Shi
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Tomasz Maciej Stepniewski
- Research Program on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences of Pompeu Fabra University (UPF), 08003 Barcelona, Spain
| | - Chia-Feng Tsai
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | | | | | - Julia Gardner
- Trinity College, Duke University, Durham, NC 27710, USA
| | - Carrie D Nicora
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | | | - Kouki Kawakami
- Department of Pharmaceutical Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Issac Choi
- Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Chloe Hicks
- Trinity College, Duke University, Durham, NC 27710, USA
| | - Kevin Zheng
- Harvard Medical School, Boston, MA 02115, USA
| | - Anmol Warman
- Trinity College, Duke University, Durham, NC 27710, USA
| | - Priya Alagesan
- Department of Biochemistry, Duke University, Durham, NC 27710, USA
| | - Nicole M Knape
- Department of Biochemistry, Duke University, Durham, NC 27710, USA
| | - Ouwen Huang
- Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Justin D Silverman
- College of Information Sciences and Technology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Asuka Inoue
- Department of Pharmaceutical Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Jana Selent
- Research Program on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences of Pompeu Fabra University (UPF), 08003 Barcelona, Spain
| | - Jon M Jacobs
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Sudarshan Rajagopal
- Department of Biochemistry, Duke University, Durham, NC 27710, USA; Department of Pharmaceutical Sciences, Tohoku University, Sendai 980-8577, Japan.
| |
Collapse
|
98
|
Conway J, Pouryahya M, Gindin Y, Pan DZ, Carrasco-Zevallos OM, Mountain V, Subramanian GM, Montalto MC, Resnick M, Beck AH, Huss RS, Myers RP, Taylor-Weiner A, Wapinski I, Chung C. Integration of deep learning-based histopathology and transcriptomics reveals key genes associated with fibrogenesis in patients with advanced NASH. Cell Rep Med 2023; 4:101016. [PMID: 37075704 PMCID: PMC10140650 DOI: 10.1016/j.xcrm.2023.101016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 12/31/2022] [Accepted: 03/21/2023] [Indexed: 04/21/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is the most common chronic liver disease globally and a leading cause for liver transplantation in the US. Its pathogenesis remains imprecisely defined. We combined two high-resolution modalities to tissue samples from NASH clinical trials, machine learning (ML)-based quantification of histological features and transcriptomics, to identify genes that are associated with disease progression and clinical events. A histopathology-driven 5-gene expression signature predicted disease progression and clinical events in patients with NASH with F3 (pre-cirrhotic) and F4 (cirrhotic) fibrosis. Notably, the Notch signaling pathway and genes implicated in liver-related diseases were enriched in this expression signature. In a validation cohort where pharmacologic intervention improved disease histology, multiple Notch signaling components were suppressed.
Collapse
|
99
|
Chen P, Zheng H. Drug-target interaction prediction based on spatial consistency constraint and graph convolutional autoencoder. BMC Bioinformatics 2023; 24:151. [PMID: 37069493 PMCID: PMC10109239 DOI: 10.1186/s12859-023-05275-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/05/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Drug-target interaction (DTI) prediction plays an important role in drug discovery and repositioning. However, most of the computational methods used for identifying relevant DTIs do not consider the invariance of the nearest neighbour relationships between drugs or targets. In other words, they do not take into account the invariance of the topological relationships between nodes during representation learning. It may limit the performance of the DTI prediction methods. RESULTS Here, we propose a novel graph convolutional autoencoder-based model, named SDGAE, to predict DTIs. As the graph convolutional network cannot handle isolated nodes in a network, a pre-processing step was applied to reduce the number of isolated nodes in the heterogeneous network and facilitate effective exploitation of the graph convolutional network. By maintaining the graph structure during representation learning, the nearest neighbour relationships between nodes in the embedding space remained as close as possible to the original space. CONCLUSIONS Overall, we demonstrated that SDGAE can automatically learn more informative and robust feature vectors of drugs and targets, thus exhibiting significantly improved predictive accuracy for DTIs.
Collapse
Affiliation(s)
- Peng Chen
- School of Computer Science and Technology, University of Science and Technology of China, Jinzhai Road 96, Hefei, 230027, People's Republic of China
- Anhui Key Laboratory of Software Engineering in Computing and Communication, University of Science and Technology of China, Jinzhai Road 96, Hefei, 230027, People's Republic of China
| | - Haoran Zheng
- School of Computer Science and Technology, University of Science and Technology of China, Jinzhai Road 96, Hefei, 230027, People's Republic of China.
- Anhui Key Laboratory of Software Engineering in Computing and Communication, University of Science and Technology of China, Jinzhai Road 96, Hefei, 230027, People's Republic of China.
| |
Collapse
|
100
|
Pandey AK, Loscalzo J. Network medicine: an approach to complex kidney disease phenotypes. Nat Rev Nephrol 2023:10.1038/s41581-023-00705-0. [PMID: 37041415 DOI: 10.1038/s41581-023-00705-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 04/13/2023]
Abstract
Scientific reductionism has been the basis of disease classification and understanding for more than a century. However, the reductionist approach of characterizing diseases from a limited set of clinical observations and laboratory evaluations has proven insufficient in the face of an exponential growth in data generated from transcriptomics, proteomics, metabolomics and deep phenotyping. A new systematic method is necessary to organize these datasets and build new definitions of what constitutes a disease that incorporates both biological and environmental factors to more precisely describe the ever-growing complexity of phenotypes and their underlying molecular determinants. Network medicine provides such a conceptual framework to bridge these vast quantities of data while providing an individualized understanding of disease. The modern application of network medicine principles is yielding new insights into the pathobiology of chronic kidney diseases and renovascular disorders by expanding the understanding of pathogenic mediators, novel biomarkers and new options for renal therapeutics. These efforts affirm network medicine as a robust paradigm for elucidating new advances in the diagnosis and treatment of kidney disorders.
Collapse
Affiliation(s)
- Arvind K Pandey
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|