1
|
Keresztes D, Kerestély M, Szarka L, Kovács BM, Schulc K, Veres DV, Csermely P. Cancer drug resistance as learning of signaling networks. Biomed Pharmacother 2025; 183:117880. [PMID: 39884030 DOI: 10.1016/j.biopha.2025.117880] [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: 09/27/2024] [Revised: 01/08/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025] Open
Abstract
Drug resistance is a major cause of tumor mortality. Signaling networks became useful tools for driving pharmacological interventions against cancer drug resistance. Signaling datasets now cover the entire human cell. Recently, network adaptation became understood as a learning process. We review rapidly increasing evidence showing that the development of cancer drug resistance can be described as learning of signaling networks. During drug adaptation, the network forgets drug-affected pathways by desensitization and relearns by strengthening alternative pathways. Thus, resistant cancer cells develop a drug resistance memory. We show that all key players of cellular learning (i.e., IDPs, protein translocation, microRNAs/lncRNAs, scaffolding proteins and epigenetic/chromatin memory) have important roles in the development of cancer drug resistance. Moreover, all of them are central components of the epithelial-mesenchymal transition leading to metastases and resistance. Phenotypic plasticity was recently listed as a hallmark of cancer. We review how network plasticity induces rare, pre-existent drug-resistant cells in the absence of drug treatment. Key network methods assessing the development of drug resistance and network pharmacological interventions against drug resistance are summarized. Finally, we highlight the class of cellular memory drugs affecting cellular learning and forgetting, and we summarize current challenges to prevent or break drug resistance using network models.
Collapse
Affiliation(s)
- Dávid Keresztes
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Márk Kerestély
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Levente Szarka
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Borbála M Kovács
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Klára Schulc
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary; Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Dániel V Veres
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary; Turbine Simulated Cell Technologies, Budapest, Hungary
| | - Peter Csermely
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
2
|
Ray AK, Priya A, Malik MZ, Thanaraj TA, Singh AK, Mago P, Ghosh C, Shalimar, Tandon R, Chaturvedi R. A bioinformatics approach to elucidate conserved genes and pathways in C. elegans as an animal model for cardiovascular research. Sci Rep 2024; 14:7471. [PMID: 38553458 PMCID: PMC10980734 DOI: 10.1038/s41598-024-56562-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/07/2024] [Indexed: 04/02/2024] Open
Abstract
Cardiovascular disease (CVD) is a collective term for disorders of the heart and blood vessels. The molecular events and biochemical pathways associated with CVD are difficult to study in clinical settings on patients and in vitro conditions. Animal models play a pivotal and indispensable role in CVD research. Caenorhabditis elegans, a nematode species, has emerged as a prominent experimental organism widely utilized in various biomedical research fields. However, the specific number of CVD-related genes and pathways within the C. elegans genome remains undisclosed to date, limiting its in-depth utilization for investigations. In the present study, we conducted a comprehensive analysis of genes and pathways related to CVD within the genomes of humans and C. elegans through a systematic bioinformatic approach. A total of 1113 genes in C. elegans orthologous to the most significant CVD-related genes in humans were identified, and the GO terms and pathways were compared to study the pathways that are conserved between the two species. In order to infer the functions of CVD-related orthologous genes in C. elegans, a PPI network was constructed. Orthologous gene PPI network analysis results reveal the hubs and important KRs: pmk-1, daf-21, gpb-1, crh-1, enpl-1, eef-1G, acdh-8, hif-1, pmk-2, and aha-1 in C. elegans. Modules were identified for determining the role of the orthologous genes at various levels in the created network. We also identified 9 commonly enriched pathways between humans and C. elegans linked with CVDs that include autophagy (animal), the ErbB signaling pathway, the FoxO signaling pathway, the MAPK signaling pathway, ABC transporters, the biosynthesis of unsaturated fatty acids, fatty acid metabolism, glutathione metabolism, and metabolic pathways. This study provides the first systematic genomic approach to explore the CVD-associated genes and pathways that are present in C. elegans, supporting the use of C. elegans as a prominent animal model organism for cardiovascular diseases.
Collapse
Affiliation(s)
- Ashwini Kumar Ray
- Department of Environmental Studies, University of Delhi, New Delhi, India.
| | - Anjali Priya
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Md Zubbair Malik
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Kuwait City, Kuwait.
| | | | - Alok Kumar Singh
- Department of Zoology, Ramjas College, University of Delhi, New Delhi, India
| | - Payal Mago
- Shaheed Rajguru College of Applied Science for Women, University of Delhi, New Delhi, India
- Campus of Open Learning, University of Delhi, New Delhi, India
| | - Chirashree Ghosh
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Shalimar
- Department of Gastroenterology, All India Institute of Medical Science, New Delhi, India
| | - Ravi Tandon
- Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
3
|
Ray AK, Priya A, Malik MZ, Thanaraj TA, Singh AK, Mago P, Ghosh C, Shalimar, Tandon R, Chaturvedi R. Conserved Cardiovascular Network: Bioinformatics Insights into Genes and Pathways for Establishing Caenorhabditis elegans as an Animal Model for Cardiovascular Diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.24.573256. [PMID: 38234826 PMCID: PMC10793405 DOI: 10.1101/2023.12.24.573256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Cardiovascular disease (CVD) is a collective term for disorders of the heart and blood vessels. The molecular events and biochemical pathways associated with CVD are difficult to study in clinical settings on patients and in vitro conditions. Animal models play a pivotal and indispensable role in cardiovascular disease (CVD) research. Caenorhabditis elegans , a nematode species, has emerged as a prominent experimental organism widely utilised in various biomedical research fields. However, the specific number of CVD-related genes and pathways within the C. elegans genome remains undisclosed to date, limiting its in-depth utilisation for investigations. In the present study, we conducted a comprehensive analysis of genes and pathways related to CVD within the genomes of humans and C. elegans through a systematic bioinformatic approach. A total of 1113 genes in C. elegans orthologous to the most significant CVD-related genes in humans were identified, and the GO terms and pathways were compared to study the pathways that are conserved between the two species. In order to infer the functions of CVD-related orthologous genes in C. elegans, a PPI network was constructed. Orthologous gene PPI network analysis results reveal the hubs and important KRs: pmk-1, daf-21, gpb-1, crh-1, enpl-1, eef-1G, acdh-8, hif-1, pmk-2, and aha-1 in C. elegans. Modules were identified for determining the role of the orthologous genes at various levels in the created network. We also identified 9 commonly enriched pathways between humans and C. elegans linked with CVDs that include autophagy (animal), the ErbB signalling pathway, the FoxO signalling pathway, the MAPK signalling pathway, ABC transporters, the biosynthesis of unsaturated fatty acids, fatty acid metabolism, glutathione metabolism, and metabolic pathways. This study provides the first systematic genomic approach to explore the CVD-associated genes and pathways that are present in C. elegans, supporting the use of C. elegans as a prominent animal model organism for cardiovascular diseases.
Collapse
|
4
|
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
|
5
|
Brooks-Warburton J, Modos D, Sudhakar P, Madgwick M, Thomas JP, Bohar B, Fazekas D, Zoufir A, Kapuy O, Szalay-Beko M, Verstockt B, Hall LJ, Watson A, Tremelling M, Parkes M, Vermeire S, Bender A, Carding SR, Korcsmaros T. A systems genomics approach to uncover patient-specific pathogenic pathways and proteins in ulcerative colitis. Nat Commun 2022; 13:2299. [PMID: 35484353 PMCID: PMC9051123 DOI: 10.1038/s41467-022-29998-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 04/06/2022] [Indexed: 12/11/2022] Open
Abstract
We describe a precision medicine workflow, the integrated single nucleotide polymorphism network platform (iSNP), designed to determine the mechanisms by which SNPs affect cellular regulatory networks, and how SNP co-occurrences contribute to disease pathogenesis in ulcerative colitis (UC). Using SNP profiles of 378 UC patients we map the regulatory effects of the SNPs to a human signalling network containing protein-protein, miRNA-mRNA and transcription factor binding interactions. With unsupervised clustering algorithms we group these patient-specific networks into four distinct clusters driven by PRKCB, HLA, SNAI1/CEBPB/PTPN1 and VEGFA/XPO5/POLH hubs. The pathway analysis identifies calcium homeostasis, wound healing and cell motility as key processes in UC pathogenesis. Using transcriptomic data from an independent patient cohort, with three complementary validation approaches focusing on the SNP-affected genes, the patient specific modules and affected functions, we confirm the regulatory impact of non-coding SNPs. iSNP identified regulatory effects for disease-associated non-coding SNPs, and by predicting the patient-specific pathogenic processes, we propose a systems-level way to stratify patients.
Collapse
Affiliation(s)
- Johanne Brooks-Warburton
- Earlham Institute, Norwich Research Park, Norwich, UK
- Gut Microbes and Health Programme, The Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Department of Clinical, Pharmaceutical and Biological Sciences, University of Hertfordshire, Hertford, UK
- Gastroenterology Department, Lister Hospital, Stevenage, UK
| | - Dezso Modos
- Earlham Institute, Norwich Research Park, Norwich, UK
- Gut Microbes and Health Programme, The Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Centre for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Padhmanand Sudhakar
- Earlham Institute, Norwich Research Park, Norwich, UK
- Gut Microbes and Health Programme, The Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- KU Leuven, Department of Chronic diseases, Metabolism and Ageing, Leuven, Belgium
| | - Matthew Madgwick
- Earlham Institute, Norwich Research Park, Norwich, UK
- Gut Microbes and Health Programme, The Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - John P Thomas
- Earlham Institute, Norwich Research Park, Norwich, UK
- Gut Microbes and Health Programme, The Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Department of Gastroenterology, Norfolk and Norwich University Hospitals, Norwich, UK
| | - Balazs Bohar
- Earlham Institute, Norwich Research Park, Norwich, UK
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - David Fazekas
- Earlham Institute, Norwich Research Park, Norwich, UK
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Azedine Zoufir
- Centre for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Orsolya Kapuy
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | | | - Bram Verstockt
- KU Leuven, Department of Chronic diseases, Metabolism and Ageing, Leuven, Belgium
- University Hospitals Leuven, Department of Gastroenterology and Hepatology, KU Leuven, Leuven, Belgium
| | - Lindsay J Hall
- Gut Microbes and Health Programme, The Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
- School of Life Sciences, ZIEL - Institute for Food & Health, Technical University of Munich, 80333, Freising, Germany
| | - Alastair Watson
- Department of Gastroenterology, Norfolk and Norwich University Hospitals, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Mark Tremelling
- Department of Gastroenterology, Norfolk and Norwich University Hospitals, Norwich, UK
| | - Miles Parkes
- Inflammatory Bowel Disease Research Group, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Severine Vermeire
- KU Leuven, Department of Chronic diseases, Metabolism and Ageing, Leuven, Belgium
- University Hospitals Leuven, Department of Gastroenterology and Hepatology, KU Leuven, Leuven, Belgium
| | - Andreas Bender
- Centre for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Simon R Carding
- Gut Microbes and Health Programme, The Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
- Norwich Medical School, University of East Anglia, Norwich, UK.
| | - Tamas Korcsmaros
- Earlham Institute, Norwich Research Park, Norwich, UK.
- Gut Microbes and Health Programme, The Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
| |
Collapse
|
6
|
Badkas A, De Landtsheer S, Sauter T. Construction and contextualization approaches for protein-protein interaction networks. Comput Struct Biotechnol J 2022; 20:3280-3290. [PMID: 35832626 PMCID: PMC9251778 DOI: 10.1016/j.csbj.2022.06.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 11/17/2022] Open
Abstract
Protein-protein interaction network (PPIN) analysis is a widely used method to study the contextual role of proteins of interest, to predict novel disease genes, disease or functional modules, and to identify novel drug targets. PPIN-based analysis uses both generic and context-specific networks. Multiple contextualization methodologies have been described, such as shortest-path algorithms, neighborhood-based methods, and diffusion/propagation algorithms. This review discusses these methods, provides intuitive representations of PPIN contextualization, and also examines how the quality of such context-specific networks could be improved by considering additional sources of evidence. As a heuristic, we observe that tasks such as identifying disease genes, drug targets, and protein complexes should consider local neighborhoods, while uncovering disease mechanisms and discovering disease-pathways would gain from diffusion-based construction.
Collapse
|
7
|
Thomas JP, Modos D, Korcsmaros T, Brooks-Warburton J. Network Biology Approaches to Achieve Precision Medicine in Inflammatory Bowel Disease. Front Genet 2021; 12:760501. [PMID: 34745229 PMCID: PMC8566351 DOI: 10.3389/fgene.2021.760501] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/08/2021] [Indexed: 12/22/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic immune-mediated condition arising due to complex interactions between multiple genetic and environmental factors. Despite recent advances, the pathogenesis of the condition is not fully understood and patients still experience suboptimal clinical outcomes. Over the past few years, investigators are increasingly capturing multi-omics data from patient cohorts to better characterise the disease. However, reaching clinically translatable endpoints from these complex multi-omics datasets is an arduous task. Network biology, a branch of systems biology that utilises mathematical graph theory to represent, integrate and analyse biological data through networks, will be key to addressing this challenge. In this narrative review, we provide an overview of various types of network biology approaches that have been utilised in IBD including protein-protein interaction networks, metabolic networks, gene regulatory networks and gene co-expression networks. We also include examples of multi-layered networks that have combined various network types to gain deeper insights into IBD pathogenesis. Finally, we discuss the need to incorporate other data sources including metabolomic, histopathological, and high-quality clinical meta-data. Together with more robust network data integration and analysis frameworks, such efforts have the potential to realise the key goal of precision medicine in IBD.
Collapse
Affiliation(s)
- John P Thomas
- Earlham Institute, Norwich, United Kingdom
- Quadram Institute Bioscience, Norwich, United Kingdom
- Department of Gastroenterology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Dezso Modos
- Earlham Institute, Norwich, United Kingdom
- Quadram Institute Bioscience, Norwich, United Kingdom
| | - Tamas Korcsmaros
- Earlham Institute, Norwich, United Kingdom
- Quadram Institute Bioscience, Norwich, United Kingdom
| | - Johanne Brooks-Warburton
- Department of Gastroenterology, Lister Hospital, Stevenage, United Kingdom
- Department of Clinical, Pharmaceutical and Biological Sciences, University of Hertfordshire, Hatfield, United Kingdom
| |
Collapse
|
8
|
Iskandar A, Zulkifli NW, Ahmad MK, Theva Das K, Zulkifle N. OTUB1 expression and interaction network analyses in MCF-7 breast cancer cells. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
9
|
Al-Karawi D, Al-Assam H, Du H, Sayasneh A, Landolfo C, Timmerman D, Bourne T, Jassim S. An Evaluation of the Effectiveness of Image-based Texture Features Extracted from Static B-mode Ultrasound Images in Distinguishing between Benign and Malignant Ovarian Masses. ULTRASONIC IMAGING 2021; 43:124-138. [PMID: 33629652 DOI: 10.1177/0161734621998091] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Significant successes in machine learning approaches to image analysis for various applications have energized strong interest in automated diagnostic support systems for medical images. The evolving in-depth understanding of the way carcinogenesis changes the texture of cellular networks of a mass/tumor has been informing such diagnostics systems with use of more suitable image texture features and their extraction methods. Several texture features have been recently applied in discriminating malignant and benign ovarian masses by analysing B-mode images from ultrasound scan of the ovary with different levels of performance. However, comparative performance evaluation of these reported features using common sets of clinically approved images is lacking. This paper presents an empirical evaluation of seven commonly used texture features (histograms, moments of histogram, local binary patterns [256-bin and 59-bin], histograms of oriented gradients, fractal dimensions, and Gabor filter), using a collection of 242 ultrasound scan images of ovarian masses of various pathological characteristics. The evaluation examines not only the effectiveness of classification schemes based on the individual texture features but also the effectiveness of various combinations of these schemes using the simple majority-rule decision level fusion. Trained support vector machine classifiers on the individual texture features without any specific pre-processing, achieve levels of accuracy between 75% and 85% where the seven moments and the 256-bin LBP are at the lower end while the Gabor filter is at the upper end. Combining the classification results of the top k (k = 3, 5, 7) best performing features further improve the overall accuracy to a level between 86% and 90%. These evaluation results demonstrate that each of the investigated image-based texture features provides informative support in distinguishing benign or malignant ovarian masses.
Collapse
Affiliation(s)
| | - Hisham Al-Assam
- School of Computing, University of Buckingham, Buckingham, UK
| | - Hongbo Du
- School of Computing, University of Buckingham, Buckingham, UK
| | - Ahmad Sayasneh
- Faculty of Life Sciences and Medicine, St Thomas Hospital, King's College London, London, UK
| | - Chiara Landolfo
- Department of Development and Regeneration; Obstetrics and Gynaecology, University Hospitals KU Leuven, Leuven, Belgium
- Queen Charlotte's and Chelsea Hospital, Imperial College, London, UK
- Dipartimento Scienze della Salute della Donna, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Dirk Timmerman
- Department of Development and Regeneration; Obstetrics and Gynaecology, University Hospitals KU Leuven, Leuven, Belgium
| | - Tom Bourne
- Department of Development and Regeneration; Obstetrics and Gynaecology, University Hospitals KU Leuven, Leuven, Belgium
- Queen Charlotte's and Chelsea Hospital, Imperial College, London, UK
| | - Sabah Jassim
- School of Computing, University of Buckingham, Buckingham, UK
| |
Collapse
|
10
|
Yin J, Li X, Li F, Lu Y, Zeng S, Zhu F. Identification of the key target profiles underlying the drugs of narrow therapeutic index for treating cancer and cardiovascular disease. Comput Struct Biotechnol J 2021; 19:2318-2328. [PMID: 33995923 PMCID: PMC8105181 DOI: 10.1016/j.csbj.2021.04.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/09/2021] [Accepted: 04/15/2021] [Indexed: 12/14/2022] Open
Abstract
An appropriate therapeutic index is crucial for drug discovery and development since narrow therapeutic index (NTI) drugs with slight dosage variation may induce severe adverse drug reactions or potential treatment failure. To date, the shared characteristics underlying the targets of NTI drugs have been explored by several studies, which have been applied to identify potential drug targets. However, the association between the drug therapeutic index and the related disease has not been dissected, which is important for revealing the NTI drug mechanism and optimizing drug design. Therefore, in this study, two classes of disease (cancers and cardiovascular disorders) with the largest number of NTI drugs were selected, and the target property of the corresponding NTI drugs was analyzed. By calculating the biological system profiles and human protein–protein interaction (PPI) network properties of drug targets and adopting an AI-based algorithm, differentiated features between two diseases were discovered to reveal the distinct underlying mechanisms of NTI drugs in different diseases. Consequently, ten shared features and four unique features were identified for both diseases to distinguish NTI from NNTI drug targets. These computational discoveries, as well as the newly found features, suggest that in the clinical study of avoiding narrow therapeutic index in those diseases, the ability of target to be a hub and the efficiency of target signaling in the human PPI network should be considered, and it could thus provide novel guidance in the drug discovery and clinical research process and help to estimate the drug safety of cancer and cardiovascular disease.
Collapse
Affiliation(s)
- Jiayi Yin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaoxu Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yinjing Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Su Zeng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China.,Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
11
|
Treveil A, Sudhakar P, Matthews ZJ, Wrzesiński T, Jones EJ, Brooks J, Ölbei M, Hautefort I, Hall LJ, Carding SR, Mayer U, Powell PP, Wileman T, Di Palma F, Haerty W, Korcsmáros T. Regulatory network analysis of Paneth cell and goblet cell enriched gut organoids using transcriptomics approaches. Mol Omics 2021; 16:39-58. [PMID: 31819932 DOI: 10.1039/c9mo00130a] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The epithelial lining of the small intestine consists of multiple cell types, including Paneth cells and goblet cells, that work in cohort to maintain gut health. 3D in vitro cultures of human primary epithelial cells, called organoids, have become a key model to study the functions of Paneth cells and goblet cells in normal and diseased conditions. Advances in these models include the ability to skew differentiation to particular lineages, providing a useful tool to study cell type specific function/dysfunction in the context of the epithelium. Here, we use comprehensive profiling of mRNA, microRNA and long non-coding RNA expression to confirm that Paneth cell and goblet cell enrichment of murine small intestinal organoids (enteroids) establishes a physiologically accurate model. We employ network analysis to infer the regulatory landscape altered by skewing differentiation, and using knowledge of cell type specific markers, we predict key regulators of cell type specific functions: Cebpa, Jun, Nr1d1 and Rxra specific to Paneth cells, Gfi1b and Myc specific for goblet cells and Ets1, Nr3c1 and Vdr shared between them. Links identified between these regulators and cellular phenotypes of inflammatory bowel disease (IBD) suggest that global regulatory rewiring during or after differentiation of Paneth cells and goblet cells could contribute to IBD aetiology. Future application of cell type enriched enteroids combined with the presented computational workflow can be used to disentangle multifactorial mechanisms of these cell types and propose regulators whose pharmacological targeting could be advantageous in treating IBD patients with Crohn's disease or ulcerative colitis.
Collapse
Affiliation(s)
- A Treveil
- Earlham Institute, Norwich Research Park, Norwich, Norfolk NR4 7UZ, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Cortés-Ciriano I, Škuta C, Bender A, Svozil D. QSAR-derived affinity fingerprints (part 2): modeling performance for potency prediction. J Cheminform 2020; 12:41. [PMID: 33431016 PMCID: PMC7339533 DOI: 10.1186/s13321-020-00444-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 05/16/2020] [Indexed: 01/22/2023] Open
Abstract
Affinity fingerprints report the activity of small molecules across a set of assays, and thus permit to gather information about the bioactivities of structurally dissimilar compounds, where models based on chemical structure alone are often limited, and model complex biological endpoints, such as human toxicity and in vitro cancer cell line sensitivity. Here, we propose to model in vitro compound activity using computationally predicted bioactivity profiles as compound descriptors. To this aim, we apply and validate a framework for the calculation of QSAR-derived affinity fingerprints (QAFFP) using a set of 1360 QSAR models generated using Ki, Kd, IC50 and EC50 data from ChEMBL database. QAFFP thus represent a method to encode and relate compounds on the basis of their similarity in bioactivity space. To benchmark the predictive power of QAFFP we assembled IC50 data from ChEMBL database for 18 diverse cancer cell lines widely used in preclinical drug discovery, and 25 diverse protein target data sets. This study complements part 1 where the performance of QAFFP in similarity searching, scaffold hopping, and bioactivity classification is evaluated. Despite being inherently noisy, we show that using QAFFP as descriptors leads to errors in prediction on the test set in the ~ 0.65-0.95 pIC50 units range, which are comparable to the estimated uncertainty of bioactivity data in ChEMBL (0.76-1.00 pIC50 units). We find that the predictive power of QAFFP is slightly worse than that of Morgan2 fingerprints and 1D and 2D physicochemical descriptors, with an effect size in the 0.02-0.08 pIC50 units range. Including QSAR models with low predictive power in the generation of QAFFP does not lead to improved predictive power. Given that the QSAR models we used to compute the QAFFP were selected on the basis of data availability alone, we anticipate better modeling results for QAFFP generated using more diverse and biologically meaningful targets. Data sets and Python code are publicly available at https://github.com/isidroc/QAFFP_regression .
Collapse
Affiliation(s)
- Isidro Cortés-Ciriano
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK. .,European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, CB10 1SD, UK.
| | - Ctibor Škuta
- CZ-OPENSCREEN: National Infrastructure for Chemical Biology, Institute of Molecular Genetics of the ASCR, v. v. i., Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Andreas Bender
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Daniel Svozil
- CZ-OPENSCREEN: National Infrastructure for Chemical Biology, Institute of Molecular Genetics of the ASCR, v. v. i., Vídeňská 1083, 142 20, Prague, Czech Republic.,CZ-OPENSCREEN: National Infrastructure for Chemical Biology, Department of Informatics and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology Prague, Technická 5, 166 28, Prague, Czech Republic
| |
Collapse
|
13
|
Bashiri H, Rahmani H, Bashiri V, Módos D, Bender A. EMDIP: An Entropy Measure to Discover Important Proteins in PPI networks. Comput Biol Med 2020; 120:103740. [PMID: 32421645 DOI: 10.1016/j.compbiomed.2020.103740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 12/24/2022]
Abstract
Discovering important proteins in Protein-Protein Interaction (PPI) networks has attracted a lot of attention in recent years. Most of the previous work applies different network centrality measures such as Closeness, Betweenness, PageRank and many others to discover the most influential proteins in PPI networks. Although entropy is a well-known graph-based method in computer science, according to our knowledge, it is not used in the biology domain for this purpose. In this paper, first, we annotate the human PPI network with available annotation data. Second, we introduce a new concept called annotation-context that describes each protein according to annotation data of its neighbors. Third, we apply an entropy measure to discover proteins with varied annotation-context. Empirical results indicate that our proposed method succeeded in (1) differentiating essential and non-essential proteins in PPI networks with annotation data; (2) outperforming centrality measures in the task of discovering essential nodes; (3) predicting new annotated proteins based on existing annotation data.
Collapse
Affiliation(s)
- Hamid Bashiri
- School of Computer engineering, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Hossein Rahmani
- School of Computer engineering, Iran University of Science and Technology, Tehran, 16846-13114, Iran.
| | - Vahid Bashiri
- School of Computer engineering, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Dezső Módos
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, United Kingdom
| | - Andreas Bender
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, United Kingdom
| |
Collapse
|
14
|
Di Nanni N, Gnocchi M, Moscatelli M, Milanesi L, Mosca E. Gene relevance based on multiple evidences in complex networks. Bioinformatics 2019; 36:865-871. [PMID: 31504182 PMCID: PMC9883679 DOI: 10.1093/bioinformatics/btz652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/17/2019] [Accepted: 08/19/2019] [Indexed: 02/02/2023] Open
Abstract
MOTIVATION Multi-omics approaches offer the opportunity to reconstruct a more complete picture of the molecular events associated with human diseases, but pose challenges in data analysis. Network-based methods for the analysis of multi-omics leverage the complex web of macromolecular interactions occurring within cells to extract significant patterns of molecular alterations. Existing network-based approaches typically address specific combinations of omics and are limited in terms of the number of layers that can be jointly analysed. In this study, we investigate the application of network diffusion to quantify gene relevance on the basis of multiple evidences (layers). RESULTS We introduce a gene score (mND) that quantifies the relevance of a gene in a biological process taking into account the network proximity of the gene and its first neighbours to other altered genes. We show that mND has a better performance over existing methods in finding altered genes in network proximity in one or more layers. We also report good performances in recovering known cancer genes. The pipeline described in this article is broadly applicable, because it can handle different types of inputs: in addition to multi-omics datasets, datasets that are stratified in many classes (e.g., cell clusters emerging from single cell analyses) or a combination of the two scenarios. AVAILABILITY AND IMPLEMENTATION The R package 'mND' is available at URL: https://www.itb.cnr.it/mnd. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Noemi Di Nanni
- Department of Biomedical Sciences, Institute of Biomedical Technologies, National Research Council, 20090 Segrate (MI), Italy,Department of Industrial and Information Engineering, University of Pavia, Italy
| | - Matteo Gnocchi
- Department of Biomedical Sciences, Institute of Biomedical Technologies, National Research Council, 20090 Segrate (MI), Italy
| | - Marco Moscatelli
- Department of Biomedical Sciences, Institute of Biomedical Technologies, National Research Council, 20090 Segrate (MI), Italy
| | - Luciano Milanesi
- Department of Biomedical Sciences, Institute of Biomedical Technologies, National Research Council, 20090 Segrate (MI), Italy
| | | |
Collapse
|
15
|
Kim TR, Jeong HH, Sohn KA. Topological integration of RPPA proteomic data with multi-omics data for survival prediction in breast cancer via pathway activity inference. BMC Med Genomics 2019; 12:94. [PMID: 31296204 PMCID: PMC6624183 DOI: 10.1186/s12920-019-0511-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The analysis of integrated multi-omics data enables the identification of disease-related biomarkers that cannot be identified from a single omics profile. Although protein-level data reflects the cellular status of cancer tissue more directly than gene-level data, past studies have mainly focused on multi-omics integration using gene-level data as opposed to protein-level data. However, the use of protein-level data (such as mass spectrometry) in multi-omics integration has some limitations. For example, the correlation between the characteristics of gene-level data (such as mRNA) and protein-level data is weak, and it is difficult to detect low-abundance signaling proteins that are used to target cancer. The reverse phase protein array (RPPA) is a highly sensitive antibody-based quantification method for signaling proteins. However, the number of protein features in RPPA data is extremely low compared to the number of gene features in gene-level data. In this study, we present a new method for integrating RPPA profiles with RNA-Seq and DNA methylation profiles for survival prediction based on the integrative directed random walk (iDRW) framework proposed in our previous study. In the iDRW framework, each omics profile is merged into a single pathway profile that reflects the topological information of the pathway. In order to address the sparsity of RPPA profiles, we employ the random walk with restart (RWR) approach on the pathway network. RESULTS Our model was validated using survival prediction analysis for a breast cancer dataset from The Cancer Genome Atlas. Our proposed model exhibited improved performance compared with other methods that utilize pathway information and also out-performed models that did not include the RPPA data utilized in our study. The risk pathways identified for breast cancer in this study were closely related to well-known breast cancer risk pathways. CONCLUSIONS Our results indicated that RPPA data is useful for survival prediction for breast cancer patients under our framework. We also observed that iDRW effectively integrates RNA-Seq, DNA methylation, and RPPA profiles, while variation in the composition of the omics data can affect both prediction performance and risk pathway identification. These results suggest that omics data composition is a critical parameter for iDRW.
Collapse
Affiliation(s)
- Tae Rim Kim
- Department of Computer Engineering, Ajou University, Suwon, 16499 South Korea
| | - Hyun-Hwan Jeong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030 USA
| | - Kyung-Ah Sohn
- Department of Computer Engineering, Ajou University, Suwon, 16499 South Korea
| |
Collapse
|
16
|
Grimes T, Potter SS, Datta S. Integrating gene regulatory pathways into differential network analysis of gene expression data. Sci Rep 2019; 9:5479. [PMID: 30940863 PMCID: PMC6445151 DOI: 10.1038/s41598-019-41918-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/12/2019] [Indexed: 12/22/2022] Open
Abstract
The advent of next-generation sequencing has introduced new opportunities in analyzing gene expression data. Research in systems biology has taken advantage of these opportunities by gleaning insights into gene regulatory networks through the analysis of gene association networks. Contrasting networks from different populations can reveal the many different roles genes fill, which can lead to new discoveries in gene function. Pathologies can also arise from aberrations in these gene-gene interactions. Exposing these network irregularities provides a new avenue for understanding and treating diseases. A general framework for integrating known gene regulatory pathways into a differential network analysis between two populations is proposed. The framework importantly allows for any gene-gene association measure to be used, and inference is carried out through permutation testing. A simulation study investigates the performance in identifying differentially connected genes when incorporating known pathways, even if the pathway knowledge is partially inaccurate. Another simulation study compares the general framework with four state-of-the-art methods. Two RNA-seq datasets are analyzed to illustrate the use of this framework in practice. In both examples, the analysis reveals genes and pathways that are known to be biologically significant along with potentially novel findings that may be used to motivate future research.
Collapse
Affiliation(s)
- Tyler Grimes
- University of Florida, Department of Biostatistics, Gainesville, 32611, USA
| | - S Steven Potter
- University of Cincinnati, Department of Pediatrics, Cincinnati, 45229, USA
| | - Somnath Datta
- University of Florida, Department of Biostatistics, Gainesville, 32611, USA.
| |
Collapse
|
17
|
G T Zañudo J, Steinway SN, Albert R. Discrete dynamic network modeling of oncogenic signaling: Mechanistic insights for personalized treatment of cancer. ACTA ACUST UNITED AC 2018; 9:1-10. [PMID: 32954058 PMCID: PMC7487767 DOI: 10.1016/j.coisb.2018.02.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Targeted drugs disrupting proteins that are dysregulated in cancer have emerged as promising treatments because of their specificity to cancer cell aberrations and thus their improved side effect profile. However, their success remains limited, largely due to existing or emergent therapy resistance. We suggest that this is due to limited understanding of the entire relevant cellular landscape. A class of mathematical models called discrete dynamic network models can be used to understand the integrated effect of an individual tumor's aberrations. We review the recent literature on discrete dynamic models of cancer and highlight their predicted therapeutic strategies. We believe dynamic network modeling can be used to drive treatment decision-making in a personalized manner to direct improved treatments in cancer. Cancer is rooted in incorrect cellular decisions caused by genetic alterations. Dynamic models of signaling networks can map the relevant repertoire of alterations. Discrete dynamic network models can predict therapeutic interventions. Progress in personalized medicine needs integration of multiple data and model types.
Collapse
Affiliation(s)
- Jorge G T Zañudo
- Department of Physics, The Pennsylvania State University, University Park, PA 16802, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute and Broad Institute of Harvard and MIT, Boston MA, USA
| | - Steven N Steinway
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Réka Albert
- Department of Physics, The Pennsylvania State University, University Park, PA 16802, USA.,Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
18
|
Ashraf MI, Ong SK, Mujawar S, Pawar S, More P, Paul S, Lahiri C. A side-effect free method for identifying cancer drug targets. Sci Rep 2018; 8:6669. [PMID: 29703908 PMCID: PMC5923273 DOI: 10.1038/s41598-018-25042-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 04/13/2018] [Indexed: 12/20/2022] Open
Abstract
Identifying effective drug targets, with little or no side effects, remains an ever challenging task. A potential pitfall of failing to uncover the correct drug targets, due to side effect of pleiotropic genes, might lead the potential drugs to be illicit and withdrawn. Simplifying disease complexity, for the investigation of the mechanistic aspects and identification of effective drug targets, have been done through several approaches of protein interactome analysis. Of these, centrality measures have always gained importance in identifying candidate drug targets. Here, we put forward an integrated method of analysing a complex network of cancer and depict the importance of k-core, functional connectivity and centrality (KFC) for identifying effective drug targets. Essentially, we have extracted the proteins involved in the pathways leading to cancer from the pathway databases which enlist real experimental datasets. The interactions between these proteins were mapped to build an interactome. Integrative analyses of the interactome enabled us to unearth plausible reasons for drugs being rendered withdrawn, thereby giving future scope to pharmaceutical industries to potentially avoid them (e.g. ESR1, HDAC2, F2, PLG, PPARA, RXRA, etc). Based upon our KFC criteria, we have shortlisted ten proteins (GRB2, FYN, PIK3R1, CBL, JAK2, LCK, LYN, SYK, JAK1 and SOCS3) as effective candidates for drug development.
Collapse
Affiliation(s)
- Md Izhar Ashraf
- The Institute of Mathematical Sciences, Chennai, 600113, India.,B.S. Abdur Rahman Crescent Institute of Science & Technology, Vandalur, Chennai, 600048, India
| | - Seng-Kai Ong
- Department of Biological Sciences, Sunway University, 47500, Petaling Jaya, Malaysia
| | - Shama Mujawar
- Department of Biological Sciences, Sunway University, 47500, Petaling Jaya, Malaysia
| | - Shrikant Pawar
- Department of Computer Science & Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
| | - Pallavi More
- Department of Bioinformatics, University of Pune, Pune, Maharashtra, 411007, India
| | - Somnath Paul
- Department of Computer Science and Engineering, Birla Institute of Technology, Mesra, India
| | - Chandrajit Lahiri
- The Institute of Mathematical Sciences, Chennai, 600113, India. .,Department of Biological Sciences, Sunway University, 47500, Petaling Jaya, Malaysia.
| |
Collapse
|