1
|
De Filippis GM, Amalfitano D, Russo C, Tommasino C, Rinaldi AM. A systematic mapping study of semantic technologies in multi-omics data integration. J Biomed Inform 2025; 165:104809. [PMID: 40154721 DOI: 10.1016/j.jbi.2025.104809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/03/2025] [Accepted: 03/07/2025] [Indexed: 04/01/2025]
Abstract
OBJECTIVE The integration of multi-omics data is essential for understanding complex biological systems, providing insights beyond single-omics approaches. However, challenges related to data heterogeneity, standardization, and computational scalability persist. This study explores the interdisciplinary application of semantic technologies to enhance data integration, standardization, and analysis in multi-omics research. METHODS We performed a systematic mapping study assessing literature from 2014 to 2024, focusing on the utilization of ontologies, knowledge graphs, and graph-based methods for multi-omics integration. RESULTS Our findings indicate a growing number of publications in this field, predominantly appearing in high-impact journals. The deployment of semantic technologies has notably improved data visualization, querying, and management, thus enhancing gene and pathway discovery, and providing deeper disease insights and more accurate predictive modeling. CONCLUSION The study underscores the significance of semantic technologies in overcoming multi-omics integration challenges. Future research should focus on integrating diverse data types, developing advanced computational tools, and incorporating AI and machine learning to foster personalized medicine applications.
Collapse
Affiliation(s)
- Giovanni Maria De Filippis
- Department of Electrical Engineering and Information Technology DIETI, University of Naples Federico II, Via Claudio, 21, Naples, 80125, Italy.
| | - Domenico Amalfitano
- Department of Electrical Engineering and Information Technology DIETI, University of Naples Federico II, Via Claudio, 21, Naples, 80125, Italy.
| | - Cristiano Russo
- Department of Electrical Engineering and Information Technology DIETI, University of Naples Federico II, Via Claudio, 21, Naples, 80125, Italy.
| | - Cristian Tommasino
- Department of Electrical Engineering and Information Technology DIETI, University of Naples Federico II, Via Claudio, 21, Naples, 80125, Italy.
| | - Antonio Maria Rinaldi
- Department of Electrical Engineering and Information Technology DIETI, University of Naples Federico II, Via Claudio, 21, Naples, 80125, Italy.
| |
Collapse
|
2
|
Serra A, Fratello M, Federico A, Greco D. An update on knowledge graphs and their current and potential applications in drug discovery. Expert Opin Drug Discov 2025:1-21. [PMID: 40223439 DOI: 10.1080/17460441.2025.2490253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 04/03/2025] [Indexed: 04/15/2025]
Abstract
INTRODUCTION Knowledge graphs are becoming prominent tools in computational drug discovery. They effectively integrate heterogeneous biomedical data and generate new hypotheses and knowledge. AREAS COVERED This article is based on a literature review using Google Scholar and PubMed to retrieve articles on existing knowledge graphs relevant to the drug discovery field. The authors compare the types of entities, relationships, and data sources they encompass. Additionally, the authors provide examples of their use in the drug discovery field and discuss potential strategies for advancing this research area. EXPERT OPINION Knowledge graphs are crucial in drug discovery, but their construction leads to challenges in data integration and consistency. Future research should prioritize the standardization of data sources and data modeling. More efforts are needed for the integration in knowledge graphs of diverse data types, such as chemical structures and epigenetic data, to enhance their effectiveness. Additionally, advancements in large language models should be pursued to aid the development of knowledge graphs, provide intuitive querying capabilities for non-expert users, and explain knowledge graphs -derived predictions, thereby making these tools more accessible and their insights more interpretable for a wider audience.
Collapse
Affiliation(s)
- Angela Serra
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
| | - Michele Fratello
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Antonio Federico
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
| | - Dario Greco
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
3
|
He L, Luo Y, He M, Sun T, Yan Y, Ye W, Lin Y. Shale wastewater treatment policies recommended by integrated knowledge graph and probability-based algorithms. ENVIRONMENTAL MONITORING AND ASSESSMENT 2025; 197:520. [PMID: 40198401 DOI: 10.1007/s10661-025-13964-0] [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: 11/15/2024] [Accepted: 03/26/2025] [Indexed: 04/10/2025]
Abstract
Shale wastewater (SWW) has received much attention recently due to its non-ignorable threat to the environment and human health. Selection of an appropriate SWW treatment technology or a set of technology combinations could be a challenge particularly when no substantial professional-knowledge (or prior experience) is available. This paper develops a modeling framework integrating knowledge graph (KG) and probability-based recommendation algorithms to produce SWW treatment policies without depending on conventional physically based mathematical models and their associated quantitative data. The model is applied to the shale regions in China, including Chongqing city, the provinces of Sichuan, Yunnan, Guizhou, Shaanxi, and Inner Mongolia Autonomous Region, and utilizes the Monte-Carlo (MC) technique to assess the stability of the output policies. Results show that chemical precipitation, ultrafiltration, membrane bioreactor, and electrodialysis in Knowledge Graph Convolutional Networks (KGCN), as well as ultrafiltration, electrocoagulation, reverse osmosis, and electrocatalytic oxidation in RippleNet, are all preferentially recommended in one scenario, with the MC-based probabilities all higher than 0.86, indicating the stability of recommendation results and robustness of the model. Future studies would focus on improving the current KG and algorithms and offering explanatory mechanisms behind policy recommendations.
Collapse
Affiliation(s)
- Li He
- State Key Laboratory of Hydraulic Engineering Intelligent Construction and Operation, Tianjin University, Tianjin, 300350, China.
| | - Yugeng Luo
- State Key Laboratory of Hydraulic Engineering Intelligent Construction and Operation, Tianjin University, Tianjin, 300350, China
| | - Mengxi He
- State Key Laboratory of Hydraulic Engineering Intelligent Construction and Operation, Tianjin University, Tianjin, 300350, China
| | - Tong Sun
- State Key Laboratory of Hydraulic Engineering Intelligent Construction and Operation, Tianjin University, Tianjin, 300350, China
| | - Yiming Yan
- Key Laboratory of Water Cycle and Related Land Surface Processes, Institute of Geographic Sciences and Natural Resources Research, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wei Ye
- Yalong River Hydropower Development Company, Ltd, Chengdu, 610051, China
| | - Yupeng Lin
- Shenzhen Dazheng Construction Consulting Company, Ltd, Shenzhen, 518000, China
| |
Collapse
|
4
|
Chytas A, Gavriilides G, Kapetanakis A, de Langlais A, Jaulent MC, Natsiavas P. OpenPVSignal Knowledge Graph: Pharmacovigilance Signal Reports in a Computationally Exploitable FAIR Representation. Drug Saf 2025; 48:425-436. [PMID: 39921707 PMCID: PMC11903642 DOI: 10.1007/s40264-024-01503-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2024] [Indexed: 02/10/2025]
Abstract
INTRODUCTION Pharmacovigilance signal report (PVSR) documents contain valuable condensed information published by drug monitoring organizations, typically in a free-text format. They provide initial insights into potential links between drugs and harmful effects. Still, their unstructured format prevents this valuable information from being integrated into data-processing pipelines (e.g., to support either the investigation of drug safety signals or decision-making in the clinical context). OBJECTIVE OpenPVSignal is a data model designed specifically to publish PVSRs via a computationally exploitable format, compliant with the FAIR (Findable, Accessible, Interoperable, Reusable) principles to promote ease of access and reusability of these valuable data. METHODS This paper outlines the procedure for converting pharmacovigilance signals published by the World Health Organization Uppsala Monitoring Centre (WHO-UMC) into the OpenPVSignal data model, resulting in a Knowledge Graph (KG). It details each step of the process, including the technical validation by KG engineers and the qualitative verification by medical and pharmacovigilance experts, leading to the finalized KG. RESULTS A total of 101 PVSRs from 2011 to 2019 were incorporated into the openly available KG. CONCLUSION The presented KG could be useful in various data-processing pipelines, including systems that support drug safety activities.
Collapse
Affiliation(s)
- Achilleas Chytas
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - George Gavriilides
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Anargyros Kapetanakis
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Alix de Langlais
- School of Engineering in Electrotechnics and Electronics, ESIEE Paris, Noisy-le-Grand, France
| | | | - Pantelis Natsiavas
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece.
| |
Collapse
|
5
|
Hansen CB, Møller MEE, Pérez-Alós L, Israelsen SB, Drici L, Ottenheijm ME, Nielsen AB, Wewer Albrechtsen NJ, Benfield T, Garred P. Differences in biomarker levels and proteomic survival prediction across two COVID-19 cohorts with distinct treatments. iScience 2025; 28:112046. [PMID: 40124495 PMCID: PMC11927729 DOI: 10.1016/j.isci.2025.112046] [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: 09/09/2024] [Revised: 12/07/2024] [Accepted: 02/13/2025] [Indexed: 03/25/2025] Open
Abstract
Prognostic biomarkers have been widely studied in COVID-19, but their levels may be influenced by treatment strategies. This study examined plasma biomarkers and proteomic survival prediction in two unvaccinated hospitalized COVID-19 cohorts receiving different treatments. In a derivation cohort (n = 126) from early 2020, we performed plasma proteomic profiling and evaluated innate and complement system immune markers. A proteomic model based on differentially expressed proteins predicted 30-day mortality with an area under the curve (AUC) of 0.81. The model was tested in a validation cohort (n = 80) from late 2020, where patients received remdesivir and dexamethasone, and performed with an AUC of 0.75. Biomarker levels varied considerably between cohorts, sometimes in opposite directions, highlighting the impact of treatment regimens on biomarker expression. These findings underscore the need to account for treatment effects when developing prognostic models, as treatment differences may limit their generalizability across populations.
Collapse
Affiliation(s)
- Cecilie Bo Hansen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | | | - Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Simone Bastrup Israelsen
- Department of Infectious Diseases, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
| | - Lylia Drici
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg Hospital, Copenhagen, Denmark
| | - Maud Eline Ottenheijm
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg Hospital, Copenhagen, Denmark
| | - Annelaura Bach Nielsen
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg Hospital, Copenhagen, Denmark
| | - Nicolai J. Wewer Albrechtsen
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Benfield
- Department of Infectious Diseases, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Kumari M, Chauhan R, Garg P. MedKG: enabling drug discovery through a unified biomedical knowledge graph. Mol Divers 2025:10.1007/s11030-025-11164-z. [PMID: 40085402 DOI: 10.1007/s11030-025-11164-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Biomedical knowledge graphs have emerged as powerful tools for drug discovery, but existing platforms often suffer from outdated information, limited accessibility, and insufficient integration of complex data. This study presents MedKG, a comprehensive and continuously updated knowledge graph designed to address these challenges in precision medicine and drug discovery. MedKG integrates data from 35 authoritative sources, encompassing 34 node types and 79 relationships. A Continuous Integration/Continuous Update pipeline ensures MedKG remains current, addressing a critical limitation of static knowledge bases. The integration of molecular embeddings enhances semantic analysis capabilities, bridging the gap between chemical structures and biological entities. To demonstrate MedKG's utility, a novel hybrid Relational Graph Convolutional Network for disease-drug link prediction, MedLINK was developed and used in case studies on clinical trial data for disease drug link prediction. Furthermore, a web-based application with user-friendly APIs and visualization tools was built, making MedKG accessible to both technical and non-technical users, which is freely available at http://pitools.niper.ac.in/medkg/.
Collapse
Affiliation(s)
- Madhavi Kumari
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Rohit Chauhan
- Department of Computer Science, National Institute of Technology (NIT), Durgapur, MG Road, Durgapur, West Bengal, 713209, India
| | - Prabha Garg
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, S.A.S. Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
7
|
Gnanaolivu R, Oliver G, Jenkinson G, Blake E, Chen W, Chia N, Klee EW, Wang C. A clinical knowledge graph-based framework to prioritize candidate genes for facilitating diagnosis of Mendelian diseases and rare genetic conditions. BMC Bioinformatics 2025; 26:82. [PMID: 40087567 PMCID: PMC11908102 DOI: 10.1186/s12859-025-06096-2] [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: 10/31/2024] [Accepted: 02/25/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Diagnosing Mendelian and rare genetic conditions requires identifying phenotype-associated genetic findings and prioritizing likely disease-causing genes. This task is labor-intensive for molecular and clinical geneticists, who must review extensive literature and databases to link patient phenotypes with causal genotypes. The challenge is further complicated by the large number of genetic variants detected through next-generation sequencing, which impacts both diagnosis timelines and patient care strategies. To address this, in silico methods that prioritize causal genes based on patient-derived phenotypes offer an effective solution, reducing the time involved in diagnostic case reviews and enhancing the efficiency of clinical diagnosis. RESULTS We developed the phenotype prioritization and analysis for rare diseases (PPAR) to rank genes based on human phenotype ontology (HPO) terms, with the specific goal of aiding the interpretation of genetic testing for Mendelian and rare diseases. PPAR leverages embeddings from a knowledge graph and incorporates knowledge from connections between genes, HPO terms, and gene ontology annotations. When applied on a clinical rare disease cohort and the publicly available deciphering developmental disorders (DDD) dataset. PPAR ranked the causal gene in the top 10 for 27% of cases in the clinical cohort and for 85% of cases in the DDD dataset, outperforming other established HPO-based methods. CONCLUSION Our findings demonstrate that PPAR, a method developed from the clinical knowledge graph, effectively ranks causal genes based on patient-derived HPO terms in rare and Mendelian disease contexts. PPAR has shown superior performance compared to other well-established HPO-only methods and provides an efficient, accessible solution for clinical geneticists. The Python-based tool is publicly available at https://github.com/dimi-lab/PPAR , offering a user-friendly platform for gene prioritization.
Collapse
Affiliation(s)
- Rohan Gnanaolivu
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Gavin Oliver
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Garrett Jenkinson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Emily Blake
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Wenan Chen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Nicholas Chia
- Department of Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Eric W Klee
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Chen Wang
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
8
|
Eriksen M, Hansen AM, Nielsen AB, Mundt F, Mann M, Lassen U, Ahlborn LB, Højgaard M, Spanggaard I, Qvortrup C, Yde CW, Rohrberg KS. Multiomics Identifies Potential Molecular Profiles Associated With Outcomes After BRAF-Targeted Therapy in Patients With BRAF V600E-Mutated Advanced Solid Tumors. JCO Precis Oncol 2025; 9:e2400266. [PMID: 40080754 PMCID: PMC11922189 DOI: 10.1200/po.24.00266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 11/18/2024] [Accepted: 01/21/2025] [Indexed: 03/15/2025] Open
Abstract
PURPOSE It is a clinical challenge to select patients for BRAF-targeted therapy because of the lack of predictive biomarkers besides the BRAF V600E mutation. By analyzing the genome, transcriptome, and proteome, this study investigated the association between baseline molecular alterations and outcomes of BRAF-targeted therapy. PATIENTS AND METHODS Fresh tumor tissue from patients enrolled in the Copenhagen Prospective Personalized Oncology study was collected and underwent comprehensive molecular profiling. RESULTS TP53 comutations were most frequently detected. Patients with a TP53 wild-type tumor had a significantly longer median progression-free survival than those with TP53 comutations (hazard ratio, 2.8 [95% CI, 1.13 to 7.08]; P = .02). RNAseq revealed a distinct gene expression signature for patients with long-term disease control (LDC), including hallmarks of cell cycle arrest and proliferation in the p53 pathway. The protein analysis demonstrated that ubiquitin-conjugating enzyme EK2 was significantly downregulated in patients with LDC. CONCLUSION Using a multiomic approach, we identified molecular alterations associated with treatment outcomes. The potential of analyzing multiomic data is promising and should be prioritized in translational cancer research to uncover the full potential within precision oncology.
Collapse
Affiliation(s)
- Martina Eriksen
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Anne M Hansen
- Department of Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Annelaura B Nielsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Filip Mundt
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Matthias Mann
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Lassen
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lise B Ahlborn
- Department of Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Martin Højgaard
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Iben Spanggaard
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Camilla Qvortrup
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christina W Yde
- Department of Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kristoffer S Rohrberg
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
9
|
Bolliger M, Wasinger D, Brunmair J, Hagn G, Wolf M, Preindl K, Reiter B, Bileck A, Gerner C, Fitzal F, Meier-Menches SM. Mass spectrometry-based analysis of eccrine sweat supports predictive, preventive and personalised medicine in a cohort of breast cancer patients in Austria. EPMA J 2025; 16:165-182. [PMID: 39991101 PMCID: PMC11842658 DOI: 10.1007/s13167-025-00396-6] [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: 10/09/2024] [Accepted: 01/07/2025] [Indexed: 02/25/2025]
Abstract
Objective Metabolomics measurements of eccrine sweat may provide novel and relevant biomedical information to support predictive, preventive and personalised medicine (3PM). However, only limited data is available regarding metabolic alterations accompanying chemotherapy of breast cancer patients related to residual cancer burden (RCB) or therapy response. Here, we have applied Metabo-Tip, a non-invasive metabolomics assay based on the analysis of eccrine sweat from the fingertips, to investigate the feasibility of such an approach, especially with respect to drug monitoring, assessing lifestyle parameters and stratification of breast cancer patients. Methods Eccrine sweat samples were collected from breast cancer patients (n = 9) during the first cycle of neoadjuvant chemotherapy at four time points in this proof-of-concept study at a Tertiary University Hospital. Metabolites in eccrine sweat were analysed using mass spectrometry. Blood plasma samples from the same timepoints were also collected and analysed using a validated targeted metabolomics kit, in addition to proteomics and fatty acids/oxylipin analysis. Results A total of 247 exogenous small molecules and endogenous metabolites were identified in eccrine sweat of the breast cancer patients. Cyclophosphamide and ondansetron were successfully detected and monitored in eccrine sweat of individual patients and accurately reflected the administration schedule. The non-essential amino acids asparagine, serine and proline, as well as ornithine were significantly regulated in eccrine sweat and blood plasma over the therapy cycle. However, their distinct time-dependent profiles indicated compartment-specific distributions. Indeed, the metabolite composition of eccrine sweat seems to largely resemble the composition of the interstitial fluid. Plasma proteins and fatty acids/oxylipins were not affected by the first treatment cycle. Individual smoking habit was revealed by the simultaneous detection of nicotine and its primary metabolite cotinine in eccrine sweat. Stratification according to RCB revealed pronounced differences in the metabolic composition of eccrine sweat in these patients at baseline, e.g., essential amino acids, possibly due to the systemic contribution of breast cancer and its impact on metabolic turnover. Conclusion Mass spectrometry-based analysis of metabolites from eccrine sweat of breast cancer patients successfully qualified lifestyle parameters for risk assessment and allowed us to monitor drug treatment and systemic response to therapy. Moreover, eccrine sweat revealed a potentially predictive metabolic pattern stratifying patients by the extent of the metabolic activity of breast cancer tissue at baseline. Eccrine sweat is derived from the otherwise hardly accessible interstitial fluid and, thus, opens up a new dimension for biomonitoring of breast cancer in secondary and tertiary care. The simple sample collection without the need for trained personnel could also enable decentralised long-term biomonitoring to assess stable disease or disease progression. Eccrine sweat analysis may indeed significantly advance 3PM for the benefit of breast cancer patients. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-025-00396-6.
Collapse
Affiliation(s)
- Michael Bolliger
- Department of General Surgery (Division of Visceral Surgery), Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Department of Surgery, St. Francis Hospital, Nikolsdorfergasse 32, 1050 Vienna, Austria
| | - Daniel Wasinger
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Vienna Doctoral School in Chemistry, University of Vienna, Waehringer Str. 38-42, 1090 Vienna, Austria
| | - Julia Brunmair
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| | - Gerhard Hagn
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Vienna Doctoral School in Chemistry, University of Vienna, Waehringer Str. 38-42, 1090 Vienna, Austria
| | - Michael Wolf
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Vienna Doctoral School in Chemistry, University of Vienna, Waehringer Str. 38-42, 1090 Vienna, Austria
| | - Karin Preindl
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18–20, Vienna, 1090 Austria
- Joint Metabolome Facility, University of Vienna and Medical University Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| | - Birgit Reiter
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18–20, Vienna, 1090 Austria
- Joint Metabolome Facility, University of Vienna and Medical University Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| | - Andrea Bileck
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| | - Christopher Gerner
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| | - Florian Fitzal
- Department of Surgery and Vascular Surgery, Hanusch Hospital, Heinrich-Collin-Str. 30, 1140 Vienna, Austria
| | - Samuel M. Meier-Menches
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Faculty of Chemistry, Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| |
Collapse
|
10
|
Guo T, Steen JA, Mann M. Mass-spectrometry-based proteomics: from single cells to clinical applications. Nature 2025; 638:901-911. [PMID: 40011722 DOI: 10.1038/s41586-025-08584-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 01/02/2025] [Indexed: 02/28/2025]
Abstract
Mass-spectrometry (MS)-based proteomics has evolved into a powerful tool for comprehensively analysing biological systems. Recent technological advances have markedly increased sensitivity, enabling single-cell proteomics and spatial profiling of tissues. Simultaneously, improvements in throughput and robustness are facilitating clinical applications. In this Review, we present the latest developments in proteomics technology, including novel sample-preparation methods, advanced instrumentation and innovative data-acquisition strategies. We explore how these advances drive progress in key areas such as protein-protein interactions, post-translational modifications and structural proteomics. Integrating artificial intelligence into the proteomics workflow accelerates data analysis and biological interpretation. We discuss the application of proteomics to single-cell analysis and spatial profiling, which can provide unprecedented insights into cellular heterogeneity and tissue architecture. Finally, we examine the transition of proteomics from basic research to clinical practice, including biomarker discovery in body fluids and the promise and challenges of implementing proteomics-based diagnostics. This Review provides a broad and high-level overview of the current state of proteomics and its potential to revolutionize our understanding of biology and transform medical practice.
Collapse
Affiliation(s)
- Tiannan Guo
- State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, China.
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China.
| | - Judith A Steen
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
11
|
Perco P, Ley M, Kęska-Izworska K, Wojenska D, Bono E, Walter SM, Fillinger L, Kratochwill K. Computational Drug Repositioning in Cardiorenal Disease: Opportunities, Challenges, and Approaches. Proteomics 2025:e202400109. [PMID: 39888210 DOI: 10.1002/pmic.202400109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 02/01/2025]
Affiliation(s)
- Paul Perco
- Delta4 GmbH, Vienna, Austria
- Department of Internal Medicine IV, Medical University of Innsbruck, Innsbruck, Austria
| | - Matthias Ley
- Delta4 GmbH, Vienna, Austria
- Comprehensive Center for Pediatrics, Department of, Pediatrics and Adolescent Medicine, Division of Pediatric Nephrology and Gastroenterology, Medical University of Vienna, Vienna, Austria
| | | | | | - Enrico Bono
- Delta4 GmbH, Vienna, Austria
- Comprehensive Center for Pediatrics, Department of, Pediatrics and Adolescent Medicine, Division of Pediatric Nephrology and Gastroenterology, Medical University of Vienna, Vienna, Austria
| | | | | | - Klaus Kratochwill
- Delta4 GmbH, Vienna, Austria
- Comprehensive Center for Pediatrics, Department of, Pediatrics and Adolescent Medicine, Division of Pediatric Nephrology and Gastroenterology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Frau F, Loustalot P, Törnqvist M, Temam N, Cupe J, Montmerle M, Augé F. Connecting electronic health records to a biomedical knowledge graph to link clinical phenotypes and molecular endotypes in atopic dermatitis. Sci Rep 2025; 15:3082. [PMID: 39856093 PMCID: PMC11760951 DOI: 10.1038/s41598-024-78794-5] [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: 04/17/2024] [Accepted: 11/04/2024] [Indexed: 01/27/2025] Open
Abstract
Precision medicine is defined by the U.S. Food & Drug Administration as "an innovative approach to tailoring disease prevention and treatment that considers differences in people's genes, environments, and lifestyles". To succeed in providing personalized medicine to patients, it will be necessary to integrate medical, biological and molecular data in order to identify all complex disease subtypes and understand their pathobiological mechanism. Since biomedical knowledge graphs (BKGs) are limited to the integration of prior knowledge data and do not integrate real-world data (RWD) that would allow for the incorporation of patient level information, we propose a first step towards using RWD, BKGs and graph machine learning (ML) to enable a fully integrated precision medicine strategy. In this study, we established a link between RWD and a BKG. Our methodology introduced a novel patient representation using graph ML applied to the BKG. This approach facilitated the interpretation and extension of ML findings, particularly in disease subtype identification with molecular data contained in the BKG. We applied our innovative methodology to deepen our understanding of atopic dermatitis, a condition with a complex underlying pathophysiological mechanism. Through our analysis, we identified seven subgroups of patients each characterized by clinical and genomic characteristics.
Collapse
Affiliation(s)
- Francesca Frau
- Sanofi R&D, Development Real World Evidence, 65926, Frankfurt am Main, Germany
| | | | | | - Nina Temam
- Quinten Health, 8 rue Vernier, 75017, Paris, France
| | - Jean Cupe
- Quinten Health, 8 rue Vernier, 75017, Paris, France
| | | | - Franck Augé
- Sanofi R&D - Translational Medicine & Early Development - Translational Precision Medicine, 13 Quai Jules Guesde, 94400, Vitry-sur-Seine, France.
| |
Collapse
|
13
|
Zhou S, Ye L, Huang Y, Valsecchi C, Liu Y, Shao L, Liu J, He T, Liu L, Fan M. Rapid diagnosis and recurrence prediction of choledocholithiasis disease using raw bile with machine learning assisted SERS. Talanta 2025; 282:126979. [PMID: 39383718 DOI: 10.1016/j.talanta.2024.126979] [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: 05/09/2024] [Revised: 09/06/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Surface-enhanced Raman spectroscopy (SERS) analysis based on body fluids has been widely applied in disease diagnose. Choledocholithiasis is a widespread and often recurrent digestive system disease, with limited data on factors predicting its formation and reappearance. Bile contains many components that could provide valuable diagnostic information; however, the current diagnosis of biliary disease by SERS focuses on detecting specific component in the bile, overlooking the complex interplay and correlations among multiple factors that could be crucial for accurate diagnosis. This work directly obtained multi-component SERS spectral information of raw bile from 46 patients. Characteristic information was extracted from bile SERS spectra using Principal Component Analysis (PCA), revealing variations in the content of characteristic components associated with different choledocholithiasis types and their recurrence frequency. Pearson correlation analysis was also introduced to reveal the interactions of primary active substances pertinent to choledocholithiasis diagnosis. The efficacy of PCA and Support Vector Machine (SVM) models in classifying stone types, presented an accuracy of 99.2 %. Furthermore, the interaction patterns among SERS characteristic components in choledocholithiasis recurrence frequency were revealed, and with the support of SVM, the prediction for different recurrence rates reached an accuracy of 95.2 %. Overall, this work demonstrates that integrating SERS with machine learning can support disease diagnosis and the interpretation of correlations among multiple components, facilitating elucidating the disease mechanisms.
Collapse
Affiliation(s)
- Shana Zhou
- School of Environmental Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Liansong Ye
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yuting Huang
- Department of Clinical Laboratory Medicine, First Affiliated Hospital, Army Medical University, Chongqing, 400038, China
| | - Chiara Valsecchi
- Federal University of Pampa, Campus Alegrete, 97542-160, Alegrete, RS, Brazil
| | - Yingying Liu
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Limei Shao
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Jiao Liu
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Tian He
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Ling Liu
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, West China Hospital, Sichuan University, Chengdu 610064, China.
| | - Meikun Fan
- School of Environmental Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China.
| |
Collapse
|
14
|
Blears D, Lou J, Fong N, Mitter R, Sheridan RM, He D, Dirac-Svejstrup AB, Bentley D, Svejstrup JQ. Redundant pathways for removal of defective RNA polymerase II complexes at a promoter-proximal pause checkpoint. Mol Cell 2024; 84:4790-4807.e11. [PMID: 39504960 DOI: 10.1016/j.molcel.2024.10.012] [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: 01/26/2024] [Revised: 07/09/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024]
Abstract
The biological purpose of Integrator and RNA polymerase II (RNAPII) promoter-proximal pausing remains uncertain. Here, we show that loss of INTS6 in human cells results in increased interaction of RNAPII with proteins that can mediate its dissociation from the DNA template, including the CRL3ARMC5 E3 ligase, which ubiquitylates CTD serine5-phosphorylated RPB1 for degradation. ARMC5-dependent RNAPII ubiquitylation is activated by defects in factors acting at the promoter-proximal pause, including Integrator, DSIF, and capping enzyme. This ARMC5 checkpoint normally curtails a sizeable fraction of RNAPII transcription, and ARMC5 knockout cells produce more uncapped transcripts. When both the Integrator and CRL3ARMC5 turnover mechanisms are compromised, cell growth ceases and RNAPII with high pausing propensity disperses from the promoter-proximal pause site into the gene body. These data support a model in which CRL3ARMC5 functions alongside Integrator in a checkpoint mechanism that removes faulty RNAPII complexes at promoter-proximal pause sites to safeguard transcription integrity.
Collapse
Affiliation(s)
- Daniel Blears
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; Mechanisms of Transcription Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Jiangman Lou
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Nova Fong
- RNA Bioscience Initiative, Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, PO Box 6511, Aurora, CO 80045, USA
| | - Richard Mitter
- Bioinformatics and Biostatistics, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ryan M Sheridan
- RNA Bioscience Initiative, Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, PO Box 6511, Aurora, CO 80045, USA
| | - Dandan He
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - A Barbara Dirac-Svejstrup
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - David Bentley
- RNA Bioscience Initiative, Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, PO Box 6511, Aurora, CO 80045, USA
| | - Jesper Q Svejstrup
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark.
| |
Collapse
|
15
|
Johnson R, Gottlieb U, Shaham G, Eisen L, Waxman J, Devons-Sberro S, Ginder CR, Hong P, Sayeed R, Reis BY, Balicer RD, Dagan N, Zitnik M. Unified Clinical Vocabulary Embeddings for Advancing Precision Medicine. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.03.24318322. [PMID: 39677476 PMCID: PMC11643188 DOI: 10.1101/2024.12.03.24318322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Integrating clinical knowledge into AI remains challenging despite numerous medical guidelines and vocabularies. Medical codes, central to healthcare systems, often reflect operational patterns shaped by geographic factors, national policies, insurance frameworks, and physician practices rather than the precise representation of clinical knowledge. This disconnect hampers AI in representing clinical relationships, raising concerns about bias, transparency, and generalizability. Here, we developed a resource of 67,124 clinical vocabulary embeddings derived from a clinical knowledge graph tailored to electronic health record vocabularies, spanning over 1.3 million edges. Using graph transformer neural networks, we generated clinical vocabulary embeddings that provide a new representation of clinical knowledge by unifying seven medical vocabularies. These embeddings were validated through a phenotype risk score analysis involving 4.57 million patients from Clalit Healthcare Services, effectively stratifying individuals based on survival outcomes. Inter-institutional panels of clinicians evaluated the embeddings for alignment with clinical knowledge across 90 diseases and 3,000 clinical codes, confirming their robustness and transferability. This resource addresses gaps in integrating clinical vocabularies into AI models and training datasets, paving the way for knowledge-grounded population and patient-level models.
Collapse
Affiliation(s)
- Ruth Johnson
- The Ivan and Francesca Berkowitz Family Living Laboratory Collaboration at Harvard Medical School and Clalit Research Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Uri Gottlieb
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat-Gan, Israel
| | - Galit Shaham
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat-Gan, Israel
| | - Lihi Eisen
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat-Gan, Israel
| | - Jacob Waxman
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat-Gan, Israel
| | - Stav Devons-Sberro
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat-Gan, Israel
| | - Curtis R. Ginder
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter Hong
- Division of General Pediatrics, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Information Technology, Enterprise Data Analytics and Reporting, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Raheel Sayeed
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Ben Y. Reis
- The Ivan and Francesca Berkowitz Family Living Laboratory Collaboration at Harvard Medical School and Clalit Research Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Predictive Medicine Group, Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA, USA
- Harvard Data Science Initiative, Cambridge, MA, USA
| | - Ran D. Balicer
- The Ivan and Francesca Berkowitz Family Living Laboratory Collaboration at Harvard Medical School and Clalit Research Institute, Boston, MA, USA
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat-Gan, Israel
- Faculty of Health Sciences, School of Public Health, Ben Gurion University of the Negev, Be’er Sheva, Israel
| | - Noa Dagan
- The Ivan and Francesca Berkowitz Family Living Laboratory Collaboration at Harvard Medical School and Clalit Research Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Clalit Research Institute, Innovation Division, Clalit Health Services, Ramat-Gan, Israel
- Software and Information Systems Engineering, Ben Gurion University, Be’er Sheva, Israel
| | - Marinka Zitnik
- The Ivan and Francesca Berkowitz Family Living Laboratory Collaboration at Harvard Medical School and Clalit Research Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Harvard Data Science Initiative, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Allston, MA, USA
| |
Collapse
|
16
|
Chen D, Yang Y, Shi D, Zhang Z, Wang M, Pan Q, Su J, Wang Z. The use of 4D data-independent acquisition-based proteomic analysis and machine learning to reveal potential biomarkers for stress levels. J Bioinform Comput Biol 2024; 22:2450025. [PMID: 39545813 DOI: 10.1142/s0219720024500252] [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: 11/17/2024]
Abstract
Research suggests that individuals who experience prolonged exposure to stress may be at higher risk for developing psychological stress disorders. Currently, psychological stress is primarily evaluated by professional physicians using rating scales, which may be prone to subjective biases and limitations of the scales. Therefore, it is imperative to explore more objective, accurate, and efficient biomarkers for evaluating the level of psychological stress in an individual. In this study, we utilized 4D data-independent acquisition (4D-DIA) proteomics for quantitative protein analysis, and then employed support vector machine (SVM) combined with SHAP interpretation algorithm to identify potential biomarkers for psychological stress levels. Biomarkers validation was subsequently achieved through machine learning classification and a substantial amount of a priori knowledge derived from the knowledge graph. We performed cross-validation of the biomarkers using two batches of data, and the results showed that the combination of Glyceraldehyde-3-phosphate dehydrogenase and Fibronectin yielded an average area under the curve (AUC) of 92%, an average accuracy of 86%, an average F1 score of 79%, and an average sensitivity of 83%. Therefore, this combination may represent a potential approach for detecting stress levels to prevent psychological stress disorders.
Collapse
Affiliation(s)
- Dehua Chen
- School of Computer Science and Technology, DongHua University, ShangHai, P. R. China
| | - Yongsheng Yang
- School of Computer Science and Technology, DongHua University, ShangHai, P. R. China
| | - Dongdong Shi
- ShangHai Mental Health Center, Shanghai JiaoTong University, School of Medicine, P. R. China
| | - Zhenhua Zhang
- School of Computer Science and Technology, DongHua University, ShangHai, P. R. China
| | - Mei Wang
- School of Computer Science and Technology, DongHua University, ShangHai, P. R. China
| | - Qiao Pan
- School of Computer Science and Technology, DongHua University, ShangHai, P. R. China
| | - Jianwen Su
- University of California, Santa Barbara, USA
| | - Zhen Wang
- ShangHai Mental Health Center, Shanghai JiaoTong University, School of Medicine, P. R. China
| |
Collapse
|
17
|
Chen H, Lu D, Xiao Z, Li S, Zhang W, Luan X, Zhang W, Zheng G. Comprehensive applications of the artificial intelligence technology in new drug research and development. Health Inf Sci Syst 2024; 12:41. [PMID: 39130617 PMCID: PMC11310389 DOI: 10.1007/s13755-024-00300-y] [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: 08/31/2023] [Accepted: 07/27/2024] [Indexed: 08/13/2024] Open
Abstract
Purpose Target-based strategy is a prevalent means of drug research and development (R&D), since targets provide effector molecules of drug action and offer the foundation of pharmacological investigation. Recently, the artificial intelligence (AI) technology has been utilized in various stages of drug R&D, where AI-assisted experimental methods show higher efficiency than sole experimental ones. It is a critical need to give a comprehensive review of AI applications in drug R &D for biopharmaceutical field. Methods Relevant literatures about AI-assisted drug R&D were collected from the public databases (Including Google Scholar, Web of Science, PubMed, IEEE Xplore Digital Library, Springer, and ScienceDirect) through a keyword searching strategy with the following terms [("Artificial Intelligence" OR "Knowledge Graph" OR "Machine Learning") AND ("Drug Target Identification" OR "New Drug Development")]. Results In this review, we first introduced common strategies and novel trends of drug R&D, followed by characteristic description of AI algorithms widely used in drug R&D. Subsequently, we depicted detailed applications of AI algorithms in target identification, lead compound identification and optimization, drug repurposing, and drug analytical platform construction. Finally, we discussed the challenges and prospects of AI-assisted methods for drug discovery. Conclusion Collectively, this review provides comprehensive overview of AI applications in drug R&D and presents future perspectives for biopharmaceutical field, which may promote the development of drug industry.
Collapse
Affiliation(s)
- Hongyu Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dong Lu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ziyi Xiao
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - Shensuo Li
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weidong Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guangyong Zheng
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
18
|
Man J, Shi Y, Hu Z, Yang R, Huang Z, Zhou Y. KSDKG: construction and application of knowledge graph for kidney stone disease based on biomedical literature and public databases. Health Inf Sci Syst 2024; 12:54. [PMID: 39554224 PMCID: PMC11564440 DOI: 10.1007/s13755-024-00309-3] [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: 04/01/2024] [Accepted: 09/22/2024] [Indexed: 11/19/2024] Open
Abstract
Purpose Kidney stone disease (KSD) is a common urological disorder with an increasing incidence worldwide. The extensive knowledge about KSD is dispersed across multiple databases, challenging the visualization and representation of its hierarchy and connections. This paper aims at constructing a disease-specific knowledge graph for KSD to enhance the effective utilization of knowledge by medical professionals and promote clinical research and discovery. Methods Text parsing and semantic analysis were conducted on literature related to KSD from PubMed, with concept annotation based on biomedical ontology being utilized to generate semantic data in RDF format. Moreover, public databases were integrated to construct a large-scale knowledge graph for KSD. Additionally, case studies were carried out to demonstrate the practical utility of the developed knowledge graph. Results We proposed and implemented a Kidney Stone Disease Knowledge Graph (KSDKG), covering more than 90 million triples. This graph comprised semantic data extracted from 29,174 articles, integrating available data from UMLS, SNOMED CT, MeSH, DrugBank and Microbe-Disease Knowledge Graph. Through the application of three cases, we retrieved and discovered information on microbes, drugs and diseases associated with KSD. The results illustrated that the KSDKG can integrate diverse medical knowledge and provide new clinical insights for identifying the underlying mechanisms of KSD. Conclusion The KSDKG efficiently utilizes knowledge graph to reveal hidden knowledge associations, facilitating semantic search and response. As a blueprint for developing disease-specific knowledge graphs, it offers valuable contributions to medical research.
Collapse
Affiliation(s)
- Jianping Man
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Yufei Shi
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Zhensheng Hu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Rui Yang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Zhisheng Huang
- Knowledge Representation and Reasoning (KR &R) Group, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yi Zhou
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080 China
| |
Collapse
|
19
|
Cankar N, Beschorner N, Tsopanidou A, Qvist FL, Colaço AR, Andersen M, Kjaerby C, Delle C, Lambert M, Mundt F, Weikop P, Jucker M, Mann M, Skotte NH, Nedergaard M. Sleep deprivation leads to non-adaptive alterations in sleep microarchitecture and amyloid-β accumulation in a murine Alzheimer model. Cell Rep 2024; 43:114977. [PMID: 39541211 DOI: 10.1016/j.celrep.2024.114977] [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: 03/06/2024] [Revised: 09/09/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Impaired sleep is a common aspect of aging and often precedes the onset of Alzheimer's disease. Here, we compare the effects of sleep deprivation in young wild-type mice and their APP/PS1 littermates, a murine model of Alzheimer's disease. After 7 h of sleep deprivation, both genotypes exhibit an increase in EEG slow-wave activity. However, only the wild-type mice demonstrate an increase in the power of infraslow norepinephrine oscillations, which are characteristic of healthy non-rapid eye movement sleep. Notably, the APP/PS1 mice fail to enhance norepinephrine oscillations 24 h after sleep deprivation, coinciding with an accumulation of cerebral amyloid-β protein. Proteome analysis of cerebrospinal fluid and extracellular fluid further supports these findings by showing altered protein clearance in APP/PS1 mice. We propose that the suppression of infraslow norepinephrine oscillations following sleep deprivation contributes to increased vulnerability to sleep loss and heightens the risk of developing amyloid pathology in early stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Neža Cankar
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Natalie Beschorner
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Anastasia Tsopanidou
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Filippa L Qvist
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ana R Colaço
- Proteomics Research Infrastructure, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Mie Andersen
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Celia Kjaerby
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Christine Delle
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Marius Lambert
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Filip Mundt
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pia Weikop
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Matthias Mann
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department for Proteomics and Signal Transduction, Max-Planck Institute for Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Niels Henning Skotte
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark; Center for Translational Neuromedicine, University of Rochester Medical School, Elmwood Avenue 601, Rochester, NY 14642, USA.
| |
Collapse
|
20
|
Yang Y, Zheng Z, Xu Y, Wei H, Yan W. BioGSF: a graph-driven semantic feature integration framework for biomedical relation extraction. Brief Bioinform 2024; 26:bbaf025. [PMID: 39853110 PMCID: PMC11759886 DOI: 10.1093/bib/bbaf025] [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: 09/04/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/26/2025] Open
Abstract
The automatic and accurate extraction of diverse biomedical relations from literature constitutes the core elements of medical knowledge graphs, which are indispensable for healthcare artificial intelligence. Currently, fine-tuning through stacking various neural networks on pre-trained language models (PLMs) represents a common framework for end-to-end resolution of the biomedical relation extraction (RE) problem. Nevertheless, sequence-based PLMs, to a certain extent, fail to fully exploit the connections between semantics and the topological features formed by these connections. In this study, we presented a graph-driven framework named BioGSF for RE from the literature by integrating shortest dependency paths (SDP) with entity-pair graph through the employment of the graph neural network model. Initially, we leveraged dependency relationships to obtain the SDP between entities and incorporated this information into the entity-pair graph. Subsequently, the graph attention network was utilized to acquire the topological information of the entity-pair graph. Ultimately, the obtained topological information was combined with the semantic features of the contextual information for relation classification. Our method was evaluated on two distinct datasets, namely S4 and BioRED. The outcomes reveal that BioGSF not only attains the superior performance among previous models with a micro-F1 score of 96.68% (S4) and 96.03% (BioRED), but also demands the shortest running times. BioGSF emerges as an efficient framework for biomedical RE.
Collapse
Affiliation(s)
- Yang Yang
- Computing Science and Artificial Intelligence College, Suzhou City University, No. 1188 Wuzhong Avenue, Wuzhong District Suzhou, Suzhou 215004, China
- Suzhou Key Lab of Multi-modal Data Fusion and Intelligent Healthcare, No. 1188 Wuzhong Avenue, Wuzhong District Suzhou, Suzhou 215004, China
- School of Computer Science & Technology, Soochow University, No. 1 Shizi Street, Suzhou 215000, China
| | - Zixuan Zheng
- School of Computer Science & Technology, Soochow University, No. 1 Shizi Street, Suzhou 215000, China
| | - Yuyang Xu
- School of Computer Science & Technology, Soochow University, No. 1 Shizi Street, Suzhou 215000, China
| | - Huifang Wei
- School of Basic Medical Sciences, Suzhou Medical College of Soochow University, No. 199 Renai Road, SIP, Suzhou 215123, China
| | - Wenying Yan
- Suzhou Key Lab of Multi-modal Data Fusion and Intelligent Healthcare, No. 1188 Wuzhong Avenue, Wuzhong District Suzhou, Suzhou 215004, China
- School of Basic Medical Sciences, Suzhou Medical College of Soochow University, No. 199 Renai Road, SIP, Suzhou 215123, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, No. 199 Renai Road, SIP, Suzhou 215123, China
| |
Collapse
|
21
|
Ma L, Gao W, Hu X, Zhou D, Wang C, Yu J, Tang K. An improved cancer diagnosis algorithm for protein mass spectrometry based on PCA and a one-dimensional neural network combining ResNet and SENet. Analyst 2024; 149:5675-5683. [PMID: 39492792 DOI: 10.1039/d4an00784k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Cancer is one of the most serious health problems worldwide. Because cancer has no specific symptoms in its early stages, it is often not diagnosed until it is in advanced stages, reducing the likelihood of successful treatment. Therefore, early diagnosis of cancer is a formidable challenge. Mass spectrometry-based proteomics offers a robust technical foundation for cancer diagnosis. However, mass spectrometry data are characterized by high dimensionality, large data volume, and noise interference, which can lead to diagnostic errors in clinical applications. To address this challenge, an improved algorithm combining principal component analysis (PCA) with a convolutional neural network (CNN) algorithm (denoted as PCA-1DSE-ResCNN) was proposed to assist in analyzing high-dimensional mass spectral data. The algorithm initially reduced the dimensionality of the data through the PCA technique. Subsequently, the convolutional neural network algorithm (1DSE-ResCNN) integrating residual blocks and squeeze-and-excitation blocks was used as a classifier. This approach can not only alleviate the issues of overfitting and gradient vanishing caused by deep network layers but also reduce redundant information, enabling the algorithm to effectively learn high-dimensional data features and deal with nonlinear relationships. To validate the effectiveness of the algorithm, the high-dimensional ovarian cancer mass spectrometry dataset was selected as an example to examine its application performance in early diagnosis of ovarian cancer. The experimental results demonstrated that the PCA-1DSE-ResCNN algorithm outperforms other methods in terms of accuracy, specificity, and sensitivity on three high-dimensional ovarian cancer datasets. This study will contribute to the rapid diagnosis and early detection of cancer.
Collapse
Affiliation(s)
- Liang Ma
- Faculty of Electrical Engineering and Computer Science, Ningbo University, Ningbo, P. R. China.
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, P. R. China.
| | - Wenqing Gao
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, P. R. China.
- Ningbo Zhenhai Institute of Mass Spectrometry, Ningbo, P.R. China
| | - Xiangyang Hu
- Faculty of Electrical Engineering and Computer Science, Ningbo University, Ningbo, P. R. China.
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, P. R. China.
| | - Dongdong Zhou
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, P. R. China.
- Ningbo Zhenhai Institute of Mass Spectrometry, Ningbo, P.R. China
| | - Chenlu Wang
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, P. R. China.
- Ningbo Zhenhai Institute of Mass Spectrometry, Ningbo, P.R. China
| | - Jiancheng Yu
- Faculty of Electrical Engineering and Computer Science, Ningbo University, Ningbo, P. R. China.
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, P. R. China.
- Ningbo Zhenhai Institute of Mass Spectrometry, Ningbo, P.R. China
| | - Keqi Tang
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, P. R. China.
- Ningbo Zhenhai Institute of Mass Spectrometry, Ningbo, P.R. China
| |
Collapse
|
22
|
Qin G, Narsinh K, Wei Q, Roach JC, Joshi A, Goetz SL, Moxon ST, Brush MH, Xu C, Yao Y, Glen AK, Morris ED, Ralevski A, Roper R, Belhu B, Zhang Y, Shmulevich I, Hadlock J, Glusman G. Generating Biomedical Knowledge Graphs from Knowledge Bases, Registries, and Multiomic Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.14.623648. [PMID: 39605475 PMCID: PMC11601480 DOI: 10.1101/2024.11.14.623648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
As large clinical and multiomics datasets and knowledge resources accumulate, they need to be transformed into computable and actionable information to support automated reasoning. These datasets range from laboratory experiment results to electronic health records (EHRs). Barriers to accessibility and sharing of such datasets include diversity of content, size and privacy. Effective transformation of data into information requires harmonization of stakeholder goals, implementation, enforcement of standards regarding quality and completeness, and availability of resources for maintenance and updates. Systems such as the Biomedical Data Translator leverage knowledge graphs (KGs), structured and machine learning readable knowledge representation, to encode knowledge extracted through inference. We focus here on the transformation of data from multiomics datasets and EHRs into compact knowledge, represented in a KG data structure. We demonstrate this data transformation in the context of the Translator ecosystem, including clinical trials, drug approvals, cancer, wellness, and EHR data. These transformations preserve individual privacy. We provide access to the five resulting KGs through the Translator framework. We show examples of biomedical research questions supported by our KGs, and discuss issues arising from extracting biomedical knowledge from multiomics data.
Collapse
Affiliation(s)
- Guangrong Qin
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Kamileh Narsinh
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Qi Wei
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Jared C. Roach
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Arpita Joshi
- The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Skye L. Goetz
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Sierra T. Moxon
- Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Matthew H. Brush
- UNC Chapel Hill, Department of Genetics, 120 Mason Farm Rd, Chapel Hill, NC 27599, USA
| | - Colleen Xu
- The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Yao Yao
- Oregon State University, 1500 SW Jefferson Way, Corvallis, OR 97331
| | - Amy K. Glen
- Oregon State University, 1500 SW Jefferson Way, Corvallis, OR 97331
| | - Evan D. Morris
- Renaissance Computing Institute, 100 Europa Dr, Ste 540, Chapel Hill, NC 27517, USA
| | | | - Ryan Roper
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Basazin Belhu
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Yue Zhang
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Ilya Shmulevich
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Jennifer Hadlock
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Gwênlyn Glusman
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| |
Collapse
|
23
|
Li M, Li X, Pan K, Geva A, Yang D, Sweet SM, Bonzel CL, Ayakulangara Panickan V, Xiong X, Mandl K, Cai T. Multisource representation learning for pediatric knowledge extraction from electronic health records. NPJ Digit Med 2024; 7:319. [PMID: 39533050 PMCID: PMC11558010 DOI: 10.1038/s41746-024-01320-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Electronic Health Record (EHR) systems are particularly valuable in pediatrics due to high barriers in clinical studies, but pediatric EHR data often suffer from low content density. Existing EHR code embeddings tailored for the general patient population fail to address the unique needs of pediatric patients. To bridge this gap, we introduce a transfer learning approach, MUltisource Graph Synthesis (MUGS), aimed at accurate knowledge extraction and relation detection in pediatric contexts. MUGS integrates graphical data from both pediatric and general EHR systems, along with hierarchical medical ontologies, to create embeddings that adaptively capture both the homogeneity and heterogeneity between hospital systems. These embeddings enable refined EHR feature engineering and nuanced patient profiling, proving particularly effective in identifying pediatric patients similar to specific profiles, with a focus on pulmonary hypertension (PH). MUGS embeddings, resistant to negative transfer, outperform other benchmark methods in multiple applications, advancing evidence-based pediatric research.
Collapse
Affiliation(s)
- Mengyan Li
- Department of Mathematical Sciences, Bentley University, Waltham, MA, USA
| | - Xiaoou Li
- School of Statistics, University of Minnesota, Minneapolis, MN, USA
| | - Kevin Pan
- Mission San Jose High School, Fremont, CA, USA
| | - Alon Geva
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| | - Doris Yang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Sara Morini Sweet
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Clara-Lea Bonzel
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | | | - Xin Xiong
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kenneth Mandl
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Tianxi Cai
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
24
|
Brbić M, Yasunaga M, Agarwal P, Leskovec J. Predicting drug outcome of population via clinical knowledge graph. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.06.24303800. [PMID: 38496488 PMCID: PMC10942490 DOI: 10.1101/2024.03.06.24303800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Optimal treatments depend on numerous factors such as drug chemical properties, disease biology, and patient characteristics to which the treatment is applied. To realize the promise of AI in healthcare, there is a need for designing systems that can capture patient heterogeneity and relevant biomedical knowledge. Here we present PlaNet, a geometric deep learning framework that reasons over population variability, disease biology, and drug chemistry by representing knowledge in the form of a massive clinical knowledge graph that can be enhanced by language models. Our framework is applicable to any sub-population, any drug as well drug combinations, any disease, and a wide range of pharmacological tasks. We apply the PlaNet framework to reason about outcomes of clinical trials: PlaNet predicts drug efficacy and adverse events, even for experimental drugs and their combinations that have never been seen by the model. Furthermore, PlaNet can estimate the effect of changing population on trial outcomes with direct implications for patient stratification in clinical trials. PlaNet takes fundamental steps towards AI-guided clinical trials design, offering valuable guidance for realizing the vision of precision medicine using AI.
Collapse
Affiliation(s)
- Maria Brbić
- School of Computer and Communication Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Michihiro Yasunaga
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| | - Prabhat Agarwal
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| | - Jure Leskovec
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
25
|
Malusare A, Aggarwal V. Improving Molecule Generation and Drug Discovery with a Knowledge-enhanced Generative Model. ARXIV 2024:arXiv:2402.08790v2. [PMID: 38410649 PMCID: PMC10896363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Recent advancements in generative models have established state-of-the-art benchmarks in the generation of molecules and novel drug candidates. Despite these successes, a significant gap persists between generative models and the utilization of extensive biomedical knowledge, often systematized within knowledge graphs, whose potential to inform and enhance generative processes has not been realized. In this paper, we present a novel approach that bridges this divide by developing a framework for knowledge-enhanced generative models called KARL. We develop a scalable methodology to extend the functionality of knowledge graphs while preserving semantic integrity, and incorporate this contextual information into a generative framework to guide a diffusion-based model. The integration of knowledge graph embeddings with our generative model furnishes a robust mechanism for producing novel drug candidates possessing specific characteristics while ensuring validity and synthesizability. KARL outperforms state-of-the-art generative models on both unconditional and targeted generation tasks.
Collapse
Affiliation(s)
- Aditya Malusare
- Edwardson School of Industrial Engineering and the Institute of Cancer Research, Purdue University
| | - Vaneet Aggarwal
- Edwardson School of Industrial Engineering and the Institute of Cancer Research, Purdue University
| |
Collapse
|
26
|
Laitila J, Seaborne RAE, Ranu N, Kolb JS, Wallgren-Pettersson C, Witting N, Vissing J, Vilchez JJ, Zanoteli E, Palmio J, Huovinen S, Granzier H, Ochala J. Myosin ATPase inhibition fails to rescue the metabolically dysregulated proteome of nebulin-deficient muscle. J Physiol 2024; 602:5229-5245. [PMID: 39216086 DOI: 10.1113/jp286870] [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: 05/13/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Nemaline myopathy (NM) is a genetic muscle disease, primarily caused by mutations in the NEB gene (NEB-NM) and with muscle myosin dysfunction as a major molecular pathogenic mechanism. Recently, we have observed that the myosin biochemical super-relaxed state was significantly impaired in NEB-NM, inducing an aberrant increase in ATP consumption and remodelling of the energy proteome in diseased muscle fibres. Because the small-molecule Mavacamten is known to promote the myosin super-relaxed state and reduce the ATP demand, we tested its potency in the context of NEB-NM. We first conducted in vitro experiments in isolated single myofibres from patients and found that Mavacamten successfully reversed the myosin ATP overconsumption. Following this, we assessed its short-term in vivo effects using the conditional nebulin knockout (cNeb KO) mouse model and subsequently performing global proteomics profiling in dissected soleus myofibres. After a 4 week treatment period, we observed a remodelling of a large number of proteins in both cNeb KO mice and their wild-type siblings. Nevertheless, these changes were not related to the energy proteome, indicating that short-term Mavacamten treatment is not sufficient to properly counterbalance the metabolically dysregulated proteome of cNeb KO mice. Taken together, our findings emphasize Mavacamten potency in vitro but challenge its short-term efficacy in vivo. KEY POINTS: No cure exists for nemaline myopathy, a type of genetic skeletal muscle disease mainly derived from mutations in genes encoding myofilament proteins. Applying Mavacamten, a small molecule directly targeting the myofilaments, to isolated membrane-permeabilized muscle fibres from human patients restored myosin energetic disturbances. Treating a mouse model of nemaline myopathy in vivo with Mavacamten for 4 weeks, remodelled the skeletal muscle fibre proteome without any noticeable effects on energetic proteins. Short-term Mavacamten treatment may not be sufficient to reverse the muscle phenotype in nemaline myopathy.
Collapse
Affiliation(s)
- Jenni Laitila
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Robert A E Seaborne
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Centre of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Natasha Ranu
- Centre of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Justin S Kolb
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, MO, USA
| | - Carina Wallgren-Pettersson
- The Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland and Department of Medical and Clinical Genetics, Medicum, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Nanna Witting
- Copenhagen Neuromuscular Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - John Vissing
- Copenhagen Neuromuscular Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Juan Jesus Vilchez
- Neuromuscular and Ataxias Research Group, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Spain, Valencia, Spain
| | - Edmar Zanoteli
- Department of Neurology, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil
| | - Johanna Palmio
- Neuromuscular Research Center, Department of Neurology, Tampere University and University Hospital, Tampere, Finland
| | - Sanna Huovinen
- Department of Pathology, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Henk Granzier
- The Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland and Department of Medical and Clinical Genetics, Medicum, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Julien Ochala
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Andersen JV, Marian OC, Qvist FL, Westi EW, Aldana BI, Schousboe A, Don AS, Skotte NH, Wellendorph P. Deficient brain GABA metabolism leads to widespread impairments of astrocyte and oligodendrocyte function. Glia 2024; 72:1821-1839. [PMID: 38899762 DOI: 10.1002/glia.24585] [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: 04/03/2024] [Revised: 06/03/2024] [Accepted: 06/09/2024] [Indexed: 06/21/2024]
Abstract
The neurometabolic disorder succinic semialdehyde dehydrogenase (SSADH) deficiency leads to great neurochemical imbalances and severe neurological manifestations. The cause of the disease is loss of function of the enzyme SSADH, leading to impaired metabolism of the principal inhibitory neurotransmitter GABA. Despite the known identity of the enzymatic deficit, the underlying pathology of SSADH deficiency remains unclear. To uncover new mechanisms of the disease, we performed an untargeted integrative analysis of cerebral protein expression, functional metabolism, and lipid composition in a genetic mouse model of SSADH deficiency (ALDH5A1 knockout mice). Our proteomic analysis revealed a clear regional vulnerability, as protein alterations primarily manifested in the hippocampus and cerebral cortex of the ALDH5A1 knockout mice. These regions displayed aberrant expression of proteins linked to amino acid homeostasis, mitochondria, glial function, and myelination. Stable isotope tracing in acutely isolated brain slices demonstrated an overall maintained oxidative metabolism of glucose, but a selective decrease in astrocyte metabolic activity in the cerebral cortex of ALDH5A1 knockout mice. In contrast, an elevated capacity of oxidative glutamine metabolism was observed in the ALDH5A1 knockout brain, which may serve as a neuronal compensation of impaired astrocyte glutamine provision. In addition to reduced expression of critical oligodendrocyte proteins, a severe depletion of myelin-enriched sphingolipids was found in the brains of ALDH5A1 knockout mice, suggesting degeneration of myelin. Altogether, our study highlights that impaired astrocyte and oligodendrocyte function is intimately linked to SSADH deficiency pathology, suggesting that selective targeting of glial cells may hold therapeutic potential in this disease.
Collapse
Affiliation(s)
- Jens V Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oana C Marian
- Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
- School of Medical Sciences, The University of Sydney, Camperdown, New South Wales, Australia
| | - Filippa L Qvist
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emil W Westi
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anthony S Don
- Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
- School of Medical Sciences, The University of Sydney, Camperdown, New South Wales, Australia
| | - Niels H Skotte
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Petrine Wellendorph
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Wang L, Zheng G, Wang P, Jia X. Unlocking the secrets of NPSLE: the role of dendritic cell-secreted CCL2 in blood-brain barrier disruption. Front Immunol 2024; 15:1343805. [PMID: 39403387 PMCID: PMC11472714 DOI: 10.3389/fimmu.2024.1343805] [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: 11/24/2023] [Accepted: 05/27/2024] [Indexed: 11/02/2024] Open
Abstract
Background This study employed RNA-seq technology and meta-analysis to unveil the molecular mechanisms of neuropsychiatric systemic lupus erythematosus (NPSLE) within the central nervous system. Methods Downloaded transcriptomic data on systemic lupus erythematosus (SLE) from the Gene Expression Omnibus (GEO) and analyzed differential genes in peripheral blood samples of NPSLE patients and healthy individuals. Employed WGCNA to identify key genes related to cognitive impairment and validated findings via RNA-seq. Conducted GO, KEGG, and GSEA analyses, and integrated PPI networks to explore gene regulatory mechanisms. Assessed gene impacts on dendritic cells and blood-brain barrier using RT-qPCR, ELISA, and in vitro models. Results Public databases and RNA-seq data have revealed a significant upregulation of CCL2 (C-C motif chemokine ligand 2) in the peripheral blood of both SLE and NPSLE patients, primarily secreted by mature dendritic cells. Furthermore, the secretion of CCL2 by mature dendritic cells may act through the RSAD2-ISG15 axis and is associated with the activation of the NLRs (Nod Like Receptor Signaling Pathway) signaling pathway in vascular endothelial cells. Subsequent in vitro cell experiments confirmed the high expression of CCL2 in peripheral blood dendritic cells of NPSLE patients, with its secretion being regulated by the RSAD2-ISG15 axis and inducing vascular endothelial cell pyroptosis through the activation of the NLRs signaling pathway. Clinical trial results ultimately confirmed that NPSLE patients exhibiting elevated CCL2 expression also experienced cognitive decline. Conclusions The secretion of CCL2 by dendritic cells induces pyroptosis in vascular endothelial cells, thereby promoting blood-brain barrier damage and triggering cognitive impairment in patients with systemic lupus erythematosus.
Collapse
Affiliation(s)
- Lei Wang
- Department of Medical Imaging, Hebei General Hospital, Shijiazhuang, China
| | - Guimin Zheng
- Department of Rheumatology and Immunology, Hebei General Hospital, Shijiazhuang, China
| | - Peiwen Wang
- 3 Major Classes of Clinical Medicine Department, Grade 2021, Hebei Medical University, Shijiazhuang, China
| | - Xiuchuan Jia
- Department of Medical Imaging, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
29
|
Mazein I, Rougny A, Mazein A, Henkel R, Gütebier L, Michaelis L, Ostaszewski M, Schneider R, Satagopam V, Jensen LJ, Waltemath D, Wodke JAH, Balaur I. Graph databases in systems biology: a systematic review. Brief Bioinform 2024; 25:bbae561. [PMID: 39565895 PMCID: PMC11578065 DOI: 10.1093/bib/bbae561] [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: 04/18/2024] [Revised: 09/28/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
Graph databases are becoming increasingly popular across scientific disciplines, being highly suitable for storing and connecting complex heterogeneous data. In systems biology, they are used as a backend solution for biological data repositories, ontologies, networks, pathways, and knowledge graph databases. In this review, we analyse all publications using or mentioning graph databases retrieved from PubMed and PubMed Central full-text search, focusing on the top 16 available graph databases, Publications are categorized according to their domain and application, focusing on pathway and network biology and relevant ontologies and tools. We detail different approaches and highlight the advantages of outstanding resources, such as UniProtKB, Disease Ontology, and Reactome, which provide graph-based solutions. We discuss ongoing efforts of the systems biology community to standardize and harmonize knowledge graph creation and the maintenance of integrated resources. Outlining prospects, including the use of graph databases as a way of communication between biological data repositories, we conclude that efficient design, querying, and maintenance of graph databases will be key for knowledge generation in systems biology and other research fields with heterogeneous data.
Collapse
Affiliation(s)
- Ilya Mazein
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Adrien Rougny
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Alexander Mazein
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Ron Henkel
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Lea Gütebier
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Lea Michaelis
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Venkata Satagopam
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| | - Lars Juhl Jensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Grønnegårdsvej 15, 1870 Frederiksberg C, Denmark
| | - Dagmar Waltemath
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Judith A H Wodke
- Medical Informatics Laboratory, University Medicine Greifswald, Walther-Rathenau-Straße 48, Greifswald 17475, Germany
| | - Irina Balaur
- Luxembourg Centre for Systems Biology, University of Luxembourg, 6 Avenue du Swing, Belvaux L-4367, Luxembourg
| |
Collapse
|
30
|
Perdomo-Quinteiro P, Belmonte-Hernández A. Knowledge Graphs for drug repurposing: a review of databases and methods. Brief Bioinform 2024; 25:bbae461. [PMID: 39325460 PMCID: PMC11426166 DOI: 10.1093/bib/bbae461] [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: 05/22/2024] [Revised: 08/07/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024] Open
Abstract
Drug repurposing has emerged as a effective and efficient strategy to identify new treatments for a variety of diseases. One of the most effective approaches for discovering potential new drug candidates involves the utilization of Knowledge Graphs (KGs). This review comprehensively explores some of the most prominent KGs, detailing their structure, data sources, and how they facilitate the repurposing of drugs. In addition to KGs, this paper delves into various artificial intelligence techniques that enhance the process of drug repurposing. These methods not only accelerate the identification of viable drug candidates but also improve the precision of predictions by leveraging complex datasets and advanced algorithms. Furthermore, the importance of explainability in drug repurposing is emphasized. Explainability methods are crucial as they provide insights into the reasoning behind AI-generated predictions, thereby increasing the trustworthiness and transparency of the repurposing process. We will discuss several techniques that can be employed to validate these predictions, ensuring that they are both reliable and understandable.
Collapse
Affiliation(s)
- Pablo Perdomo-Quinteiro
- Grupo de Aplicación de Telecomunicaciones Visuales, Escuela Técnica Superior de Ingenieros de Telecomunicación, Universidad Politécnica de Madrid, Avenida Complutense 30, 28040 Madrid, Spain
| | - Alberto Belmonte-Hernández
- Grupo de Aplicación de Telecomunicaciones Visuales, Escuela Técnica Superior de Ingenieros de Telecomunicación, Universidad Politécnica de Madrid, Avenida Complutense 30, 28040 Madrid, Spain
| |
Collapse
|
31
|
Ran P, Wang Y, Li K, He S, Tan S, Lv J, Zhu J, Tang S, Feng J, Qin Z, Li Y, Huang L, Yin Y, Zhu L, Yang W, Ding C. STAVER: a standardized benchmark dataset-based algorithm for effective variation reduction in large-scale DIA-MS data. Brief Bioinform 2024; 25:bbae553. [PMID: 39504480 PMCID: PMC11540132 DOI: 10.1093/bib/bbae553] [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: 07/05/2024] [Revised: 09/12/2024] [Accepted: 10/19/2024] [Indexed: 11/08/2024] Open
Abstract
Mass spectrometry (MS)-based proteomics has become instrumental in comprehensively investigating complex biological systems. Data-independent acquisition (DIA)-MS, utilizing hybrid spectral library search strategies, allows for the simultaneous quantification of thousands of proteins, showing promise in enhancing protein identification and quantification precision. However, low-quality profiles can considerably undermine quantitative precision, resulting in inaccurate protein quantification. To tackle this challenge, we introduced STAVER, a novel algorithm that leverages standardized benchmark datasets to reduce non-biological variation in large-scale DIA-MS analyses. By eliminating unwanted noise in MS signals, STAVER significantly improved protein quantification precision, especially in hybrid spectral library searches. Moreover, we validated STAVER's robustness and applicability across multiple large-scale DIA datasets, demonstrating significantly enhanced precision and reproducibility of protein quantification. STAVER offers an innovative and effective approach for enhancing the quality of large-scale DIA proteomic data, facilitating cross-platform and cross-laboratory comparative analyses. This advancement significantly enhances the consistency and reliability of findings in clinical research. The complete package is available at https://github.com/Ran485/STAVER.
Collapse
Affiliation(s)
- Peng Ran
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Yunzhi Wang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Kai Li
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Shiman He
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Subei Tan
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Jiacheng Lv
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Jiajun Zhu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Shaoshuai Tang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Jinwen Feng
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Zhaoyu Qin
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Yan Li
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Lin Huang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Yanan Yin
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Lingli Zhu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
| | - Wenjun Yang
- Department of Pediatric Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Road, Yangpu District, Shanghai 200092, China
| | - Chen Ding
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, E301, School of Life Sciences, No. 2005, Songhu Road, Yangpu District, Shanghai 200438, P.R. China
- Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xinjiang Medical University Xinjiang Key Laboratory of Translational Biomedical Engineering, Urumqi 830000, P. R. China
| |
Collapse
|
32
|
Oliinyk D, Will A, Schneidmadel FR, Böhme M, Rinke J, Hochhaus A, Ernst T, Hahn N, Geis C, Lubeck M, Raether O, Humphrey SJ, Meier F. µPhos: a scalable and sensitive platform for high-dimensional phosphoproteomics. Mol Syst Biol 2024; 20:972-995. [PMID: 38907068 PMCID: PMC11297287 DOI: 10.1038/s44320-024-00050-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/23/2024] Open
Abstract
Mass spectrometry has revolutionized cell signaling research by vastly simplifying the analysis of many thousands of phosphorylation sites in the human proteome. Defining the cellular response to perturbations is crucial for further illuminating the functionality of the phosphoproteome. Here we describe µPhos ('microPhos'), an accessible phosphoproteomics platform that permits phosphopeptide enrichment from 96-well cell culture and small tissue amounts in <8 h total processing time. By greatly minimizing transfer steps and liquid volumes, we demonstrate increased sensitivity, >90% selectivity, and excellent quantitative reproducibility. Employing highly sensitive trapped ion mobility mass spectrometry, we quantify ~17,000 Class I phosphosites in a human cancer cell line using 20 µg starting material, and confidently localize ~6200 phosphosites from 1 µg. This depth covers key signaling pathways, rendering sample-limited applications and perturbation experiments with hundreds of samples viable. We employ µPhos to study drug- and time-dependent response signatures in a leukemia cell line, and by quantifying 30,000 Class I phosphosites in the mouse brain we reveal distinct spatial kinase activities in subregions of the hippocampal formation.
Collapse
Affiliation(s)
- Denys Oliinyk
- Functional Proteomics, Jena University Hospital, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
| | - Andreas Will
- Functional Proteomics, Jena University Hospital, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
| | - Felix R Schneidmadel
- Functional Proteomics, Jena University Hospital, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
| | - Maximilian Böhme
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
- Klinik für Innere Medizin II, Jena University Hospital, 07747, Jena, Germany
| | - Jenny Rinke
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
- Klinik für Innere Medizin II, Jena University Hospital, 07747, Jena, Germany
| | - Andreas Hochhaus
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
- Klinik für Innere Medizin II, Jena University Hospital, 07747, Jena, Germany
| | - Thomas Ernst
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
- Klinik für Innere Medizin II, Jena University Hospital, 07747, Jena, Germany
| | - Nina Hahn
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, 07747, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
| | - Christian Geis
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, 07747, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
| | - Markus Lubeck
- Bruker Daltonics GmbH & Co. KG, 28359, Bremen, Germany
| | | | - Sean J Humphrey
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, 3052, Victoria, Australia.
| | - Florian Meier
- Functional Proteomics, Jena University Hospital, 07747, Jena, Germany.
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany.
| |
Collapse
|
33
|
London A, Richter MM, Sjøberg KA, Wewer Albrechtsen NJ, Považan M, Drici L, Schaufuss A, Madsen L, Øyen J, Madsbad S, Holst JJ, van Hall G, Siebner HR, Richter EA, Kiens B, Lundsgaard A, Bojsen-Møller KN. The impact of short-term eucaloric low- and high-carbohydrate diets on liver triacylglycerol content in males with overweight and obesity: a randomized crossover study. Am J Clin Nutr 2024; 120:283-293. [PMID: 38914224 DOI: 10.1016/j.ajcnut.2024.06.006] [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: 01/25/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Intrahepatic triacylglycerol (liver TG) content is associated with hepatic insulin resistance and dyslipidemia. Liver TG content can be modulated within days under hypocaloric conditions. OBJECTIVES We hypothesized that 4 d of eucaloric low-carbohydrate/high-fat (LC) intake would decrease liver TG content, whereas a high-carbohydrate/low-fat (HC) intake would increase liver TG content, and further that alterations in liver TG would be linked to dynamic changes in hepatic glucose and lipid metabolism. METHODS A randomized crossover trial in males with 4 d + 4 d of LC and HC, respectively, with ≥2 wk of washout. 1H-magnetic resonance spectroscopy (1H-MRS) was used to measure liver TG content, with metabolic testing before and after intake of an LC diet (11E% carbohydrate corresponding to 102 ± 12 {mean ± standard deviation [SD]) g/d, 70E% fat} and an HC diet (65E% carbohydrate corresponding to 537 ± 56 g/d, 16E% fat). Stable [6,6-2H2]-glucose and [1,1,2,3,3-D5]-glycerol tracer infusions combined with hyperinsulinemic-euglycemic clamps and indirect calorimetry were used to measure rates of hepatic glucose production and lipolysis, whole-body insulin sensitivity and substrate oxidation. RESULTS Eleven normoglycemic males with overweight or obesity (BMI 31.6 ± 3.7 kg/m2) completed both diets. The LC diet reduced liver TG content by 35.3% (95% confidence interval: -46.6, -24.1) from 4.9% [2.4-11.0] (median interquartile range) to 2.9% [1.4-6.9], whereas there was no change after the HC diet. After the LC diet, fasting whole-body fat oxidation and plasma beta-hydroxybutyrate concentration increased, whereas markers of de novo lipogenesis (DNL) diminished. Fasting plasma TG and insulin concentrations were lowered and the hepatic insulin sensitivity index increased after LC. Peripheral glucose disposal was unchanged. CONCLUSIONS Reduced carbohydrate and increased fat intake for 4 d induced a marked reduction in liver TG content and increased hepatic insulin sensitivity. Increased rates of fat oxidation and ketogenesis combined with lower rates of DNL are suggested to be responsible for lowering liver TG. This trial was registered at clinicaltrials.gov as NCT04581421.
Collapse
Affiliation(s)
- Amalie London
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; Department of Nutrition, Exercise and Sports, The August Krogh Section for Molecular Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Michael M Richter
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; Department of Nutrition, Exercise and Sports, The August Krogh Section for Molecular Physiology, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Biochemistry, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Kim Anker Sjøberg
- Department of Nutrition, Exercise and Sports, The August Krogh Section for Molecular Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Clinical Biochemistry, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark; Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Michal Považan
- Danish Research Center for Magnetic Resonance (DRCMR), Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Lylia Drici
- Department of Clinical Biochemistry, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark; Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Amanda Schaufuss
- Department of Nutrition, Exercise and Sports, The August Krogh Section for Molecular Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Lise Madsen
- Department of Biology, Laboratory of Genomics and Molecular Biomedicine, University of Copenhagen, Copenhagen, Denmark; Institute of Marine Research, Bergen, Norway
| | | | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Gerrit van Hall
- Department of Clinical Metabolomics, Rigshospitalet, Denmark
| | - Hartwig Roman Siebner
- Danish Research Center for Magnetic Resonance (DRCMR), Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark; Department of Neurology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Erik A Richter
- Department of Nutrition, Exercise and Sports, The August Krogh Section for Molecular Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Bente Kiens
- Department of Nutrition, Exercise and Sports, The August Krogh Section for Molecular Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Annemarie Lundsgaard
- Department of Nutrition, Exercise and Sports, The August Krogh Section for Molecular Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Kirstine Nyvold Bojsen-Møller
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
34
|
Cousins HC, Nayar G, Altman RB. Computational Approaches to Drug Repurposing: Methods, Challenges, and Opportunities. Annu Rev Biomed Data Sci 2024; 7:15-29. [PMID: 38598857 DOI: 10.1146/annurev-biodatasci-110123-025333] [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: 04/12/2024]
Abstract
Drug repurposing refers to the inference of therapeutic relationships between a clinical indication and existing compounds. As an emerging paradigm in drug development, drug repurposing enables more efficient treatment of rare diseases, stratified patient populations, and urgent threats to public health. However, prioritizing well-suited drug candidates from among a nearly infinite number of repurposing options continues to represent a significant challenge in drug development. Over the past decade, advances in genomic profiling, database curation, and machine learning techniques have enabled more accurate identification of drug repurposing candidates for subsequent clinical evaluation. This review outlines the major methodologic classes that these approaches comprise, which rely on (a) protein structure, (b) genomic signatures, (c) biological networks, and (d) real-world clinical data. We propose that realizing the full impact of drug repurposing methodologies requires a multidisciplinary understanding of each method's advantages and limitations with respect to clinical practice.
Collapse
Affiliation(s)
- Henry C Cousins
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA;
| | - Gowri Nayar
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA;
| | - Russ B Altman
- Departments of Genetics, Medicine, and Bioengineering, Stanford University, Stanford, California, USA
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA;
| |
Collapse
|
35
|
Lolansen SD, Rostgaard N, Olsen MH, Ottenheijm ME, Drici L, Capion T, Nørager NH, MacAulay N, Juhler M. Proteomic profile and predictive markers of outcome in patients with subarachnoid hemorrhage. Clin Proteomics 2024; 21:51. [PMID: 39044147 PMCID: PMC11267790 DOI: 10.1186/s12014-024-09493-6] [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: 11/28/2023] [Accepted: 05/31/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND The molecular mechanisms underlying development of posthemorrhagic hydrocephalus (PHH) following subarachnoid hemorrhage (SAH) remain incompletely understood. Consequently, treatment strategies tailored towards the individual patient remain limited. This study aimed to identify proteomic cerebrospinal fluid (CSF) biomarkers capable of predicting shunt dependency and functional outcome in patients with SAH in order to improve informed clinical decision making. METHODS Ventricular CSF samples were collected twice from 23 patients with SAH who required external ventricular drain (EVD) insertion (12 patients with successful EVD weaning, 11 patients in need of permanent CSF shunting due to development of PHH). The paired CSF samples were collected acutely after ictus and later upon EVD removal. Cisternal CSF samples were collected from 10 healthy control subjects undergoing vascular clipping of an unruptured aneurysm. All CSF samples were subjected to mass spectrometry-based proteomics analysis. Proteomic biomarkers were quantified using area under the curve (AUC) estimates from a receiver operating curve (ROC). RESULTS CSF from patients with SAH displayed a distinct proteomic profile in comparison to that of healthy control subjects. The CSF collected acutely after ictus from patients with SAH was moreover distinct from that collected weeks later but appeared similar in the weaned and shunted patient groups. Sixteen unique proteins were identified as potential predictors of shunt dependency, while three proteins were identified as potential predictors of functional outcome assessed six months after ictus with the modified Rankin Scale. CONCLUSIONS We here identified several potential proteomic biomarkers in CSF from patients with SAH capable of predicting (i) shunt dependency and thus development of PHH and (ii) the functional outcome assessed six months after ictus. These proteomic biomarkers may have the potential to aid clinical decision making by predicting shunt dependency and functional outcome following SAH.
Collapse
Affiliation(s)
- Sara Diana Lolansen
- Department of Neurosurgery, the Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Nina Rostgaard
- Department of Neurosurgery, the Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Markus Harboe Olsen
- Department of Neuroanaesthesiology, the Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Anaesthesiology, Zealand University Hospital, Køge, Denmark
| | - Maud Eline Ottenheijm
- NNF Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Lylia Drici
- NNF Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Tenna Capion
- Department of Neurosurgery, the Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Nicolas Hernandez Nørager
- Department of Neurosurgery, the Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.
| | - Marianne Juhler
- Department of Neurosurgery, the Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
36
|
Sudhahar S, Ozer B, Chang J, Chadwick W, O'Donovan D, Campbell A, Tulip E, Thompson N, Roberts I. An experimentally validated approach to automated biological evidence generation in drug discovery using knowledge graphs. Nat Commun 2024; 15:5703. [PMID: 38977662 PMCID: PMC11231212 DOI: 10.1038/s41467-024-50024-6] [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: 05/17/2023] [Accepted: 06/27/2024] [Indexed: 07/10/2024] Open
Abstract
Explaining predictions for drug repositioning with biological knowledge graphs is a challenging problem. Graph completion methods using symbolic reasoning predict drug treatments and associated rules to generate evidence representing the therapeutic basis of the drug. Yet the vast amounts of generated paths that are biologically irrelevant or not mechanistically meaningful within the context of disease biology can limit utility. We use a reinforcement learning based knowledge graph completion model combined with an automatic filtering approach that produces the most relevant rules and biological paths explaining the predicted drug's therapeutic connection to the disease. In this work we validate the approach against preclinical experimental data for Fragile X syndrome demonstrating strong correlation between automatically extracted paths and experimentally derived transcriptional changes of selected genes and pathways of drug predictions Sulindac and Ibudilast. Additionally, we show it reduces the number of generated paths in two case studies, 85% for Cystic fibrosis and 95% for Parkinson's disease.
Collapse
|
37
|
Valous NA, Popp F, Zörnig I, Jäger D, Charoentong P. Graph machine learning for integrated multi-omics analysis. Br J Cancer 2024; 131:205-211. [PMID: 38729996 PMCID: PMC11263675 DOI: 10.1038/s41416-024-02706-7] [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: 11/15/2023] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
Multi-omics experiments at bulk or single-cell resolution facilitate the discovery of hypothesis-generating biomarkers for predicting response to therapy, as well as aid in uncovering mechanistic insights into cellular and microenvironmental processes. Many methods for data integration have been developed for the identification of key elements that explain or predict disease risk or other biological outcomes. The heterogeneous graph representation of multi-omics data provides an advantage for discerning patterns suitable for predictive/exploratory analysis, thus permitting the modeling of complex relationships. Graph-based approaches-including graph neural networks-potentially offer a reliable methodological toolset that can provide a tangible alternative to scientists and clinicians that seek ideas and implementation strategies in the integrated analysis of their omics sets for biomedical research. Graph-based workflows continue to push the limits of the technological envelope, and this perspective provides a focused literature review of research articles in which graph machine learning is utilized for integrated multi-omics data analyses, with several examples that demonstrate the effectiveness of graph-based approaches.
Collapse
Affiliation(s)
- Nektarios A Valous
- Applied Tumor Immunity Clinical Cooperation Unit, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany.
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany.
| | - Ferdinand Popp
- Applied Tumor Immunity Clinical Cooperation Unit, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Inka Zörnig
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Dirk Jäger
- Applied Tumor Immunity Clinical Cooperation Unit, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Pornpimol Charoentong
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| |
Collapse
|
38
|
Ertürk A. Deep 3D histology powered by tissue clearing, omics and AI. Nat Methods 2024; 21:1153-1165. [PMID: 38997593 DOI: 10.1038/s41592-024-02327-1] [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] [Received: 11/24/2022] [Accepted: 05/28/2024] [Indexed: 07/14/2024]
Abstract
To comprehensively understand tissue and organism physiology and pathophysiology, it is essential to create complete three-dimensional (3D) cellular maps. These maps require structural data, such as the 3D configuration and positioning of tissues and cells, and molecular data on the constitution of each cell, spanning from the DNA sequence to protein expression. While single-cell transcriptomics is illuminating the cellular and molecular diversity across species and tissues, the 3D spatial context of these molecular data is often overlooked. Here, I discuss emerging 3D tissue histology techniques that add the missing third spatial dimension to biomedical research. Through innovations in tissue-clearing chemistry, labeling and volumetric imaging that enhance 3D reconstructions and their synergy with molecular techniques, these technologies will provide detailed blueprints of entire organs or organisms at the cellular level. Machine learning, especially deep learning, will be essential for extracting meaningful insights from the vast data. Further development of integrated structural, molecular and computational methods will unlock the full potential of next-generation 3D histology.
Collapse
Affiliation(s)
- Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Zentrum München, Neuherberg, Germany.
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University, Munich, Germany.
- School of Medicine, Koç University, İstanbul, Turkey.
- Deep Piction GmbH, Munich, Germany.
| |
Collapse
|
39
|
Lange E, Kranert L, Krüger J, Benndorf D, Heyer R. Microbiome modeling: a beginner's guide. Front Microbiol 2024; 15:1368377. [PMID: 38962127 PMCID: PMC11220171 DOI: 10.3389/fmicb.2024.1368377] [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: 01/10/2024] [Accepted: 05/27/2024] [Indexed: 07/05/2024] Open
Abstract
Microbiomes, comprised of diverse microbial species and viruses, play pivotal roles in human health, environmental processes, and biotechnological applications and interact with each other, their environment, and hosts via ecological interactions. Our understanding of microbiomes is still limited and hampered by their complexity. A concept improving this understanding is systems biology, which focuses on the holistic description of biological systems utilizing experimental and computational methods. An important set of such experimental methods are metaomics methods which analyze microbiomes and output lists of molecular features. These lists of data are integrated, interpreted, and compiled into computational microbiome models, to predict, optimize, and control microbiome behavior. There exists a gap in understanding between microbiologists and modelers/bioinformaticians, stemming from a lack of interdisciplinary knowledge. This knowledge gap hinders the establishment of computational models in microbiome analysis. This review aims to bridge this gap and is tailored for microbiologists, researchers new to microbiome modeling, and bioinformaticians. To achieve this goal, it provides an interdisciplinary overview of microbiome modeling, starting with fundamental knowledge of microbiomes, metaomics methods, common modeling formalisms, and how models facilitate microbiome control. It concludes with guidelines and repositories for modeling. Each section provides entry-level information, example applications, and important references, serving as a valuable resource for comprehending and navigating the complex landscape of microbiome research and modeling.
Collapse
Affiliation(s)
- Emanuel Lange
- Multidimensional Omics Data Analysis, Department for Bioanalytics, Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
- Graduate School Digital Infrastructure for the Life Sciences, Bielefeld Institute for Bioinformatics Infrastructure (BIBI), Faculty of Technology, Bielefeld University, Bielefeld, Germany
| | - Lena Kranert
- Institute for Automation Engineering, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Jacob Krüger
- Engineering of Software-Intensive Systems, Department of Mathematics and Computer Science, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Dirk Benndorf
- Applied Biosciences and Bioprocess Engineering, Anhalt University of Applied Sciences, Köthen, Germany
| | - Robert Heyer
- Multidimensional Omics Data Analysis, Department for Bioanalytics, Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
- Graduate School Digital Infrastructure for the Life Sciences, Bielefeld Institute for Bioinformatics Infrastructure (BIBI), Faculty of Technology, Bielefeld University, Bielefeld, Germany
- Multidimensional Omics Data Analysis, Faculty of Technology, Bielefeld University, Bielefeld, Germany
| |
Collapse
|
40
|
Nygaard U, Nielsen AB, Dungu KHS, Drici L, Holm M, Ottenheijm ME, Nielsen AB, Glenthøj JP, Schmidt LS, Cortes D, Jørgensen IM, Mogensen TH, Schmiegelow K, Mann M, Vissing NH, Wewer Albrechtsen NJ. Proteomic profiling reveals diagnostic signatures and pathogenic insights in multisystem inflammatory syndrome in children. Commun Biol 2024; 7:688. [PMID: 38839859 PMCID: PMC11153518 DOI: 10.1038/s42003-024-06370-8] [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: 11/12/2023] [Accepted: 05/22/2024] [Indexed: 06/07/2024] Open
Abstract
Multisystem inflammatory syndrome in children (MIS-C) is a severe disease that emerged during the COVID-19 pandemic. Although recognized as an immune-mediated condition, the pathogenesis remains unresolved. Furthermore, the absence of a diagnostic test can lead to delayed immunotherapy. Using state-of-the-art mass-spectrometry proteomics, assisted by artificial intelligence (AI), we aimed to identify a diagnostic signature for MIS-C and to gain insights into disease mechanisms. We identified a highly specific 4-protein diagnostic signature in children with MIS-C. Furthermore, we identified seven clusters that differed between MIS-C and controls, indicating an interplay between apolipoproteins, immune response proteins, coagulation factors, platelet function, and the complement cascade. These intricate protein patterns indicated MIS-C as an immunometabolic condition with global hypercoagulability. Our findings emphasize the potential of AI-assisted proteomics as a powerful and unbiased tool for assessing disease pathogenesis and suggesting avenues for future interventions and impact on pediatric disease trajectories through early diagnosis.
Collapse
Affiliation(s)
- Ulrikka Nygaard
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | - Annelaura Bach Nielsen
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kia Hee Schultz Dungu
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Lylia Drici
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Holm
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Maud Eline Ottenheijm
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Allan Bybeck Nielsen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Jonathan Peter Glenthøj
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital North Zealand, Hillerød, Denmark
| | - Lisbeth Samsø Schmidt
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Dina Cortes
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Inger Merete Jørgensen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital North Zealand, Hillerød, Denmark
| | | | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Matthias Mann
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Nadja Hawwa Vissing
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| |
Collapse
|
41
|
Devarakonda MV, Mohanty S, Sunkishala RR, Mallampalli N, Liu X. Clinical trial recommendations using Semantics-Based inductive inference and knowledge graph embeddings. J Biomed Inform 2024; 154:104627. [PMID: 38561170 DOI: 10.1016/j.jbi.2024.104627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/06/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVE Designing a new clinical trial entails many decisions, such as defining a cohort and setting the study objectives to name a few, and therefore can benefit from recommendations based on exhaustive mining of past clinical trial records. This study proposes an approach based on knowledge graph embeddings and semantics-driven inductive inference for generating such recommendations. METHOD The proposed recommendation methodology is based on neural embeddings trained on first-of-its-kind knowledge graph constructed from clinical trials data. The methodology includes design of a knowledge graph for clinical trial data, evaluation of various knowledge graph embedding techniques for it, application of a novel inductive inference method using these embeddings, and generation of recommendations for clinical trial design. The study uses freely available data from clinicaltrials.gov and related sources. RESULTS The proposed approach for recommendations obtained relevance scores ranging from 70% to 83%. These scores were determined by evaluating the text similarity of recommended elements to actual elements used in clinical trials that are in progress. Furthermore, the most pertinent recommendations were consistently located towards the top of the list, indicating the effectiveness of our method. CONCLUSION Our study suggests that inductive inference using node semantics is a viable approach for generating recommendations using graphs neural embeddings, and that there is a potential for improvement in training graph embeddings using node semantics.
Collapse
Affiliation(s)
| | | | | | | | - Xiong Liu
- Biomedical Research, Novartis, Cambridge, MA, USA
| |
Collapse
|
42
|
Ebstrup E, Ansbøl J, Paez-Garcia A, Culp H, Chevalier J, Clemmens P, Coll NS, Moreno-Risueno MA, Rodriguez E. NBR1-mediated selective autophagy of ARF7 modulates root branching. EMBO Rep 2024; 25:2571-2591. [PMID: 38684906 PMCID: PMC11169494 DOI: 10.1038/s44319-024-00142-5] [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: 12/08/2023] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
Auxin dictates root architecture via the Auxin Response Factor (ARF) family of transcription factors, which control lateral root (LR) formation. In Arabidopsis, ARF7 regulates the specification of prebranch sites (PBS) generating LRs through gene expression oscillations and plays a pivotal role during LR initiation. Despite the importance of ARF7 in this process, there is a surprising lack of knowledge about how ARF7 turnover is regulated and how this impacts root architecture. Here, we show that ARF7 accumulates in autophagy mutants and is degraded through NBR1-dependent selective autophagy. We demonstrate that the previously reported rhythmic changes to ARF7 abundance in roots are modulated via autophagy and might occur in other tissues. In addition, we show that the level of co-localization between ARF7 and autophagy markers oscillates and can be modulated by auxin to trigger ARF7 turnover. Furthermore, we observe that autophagy impairment prevents ARF7 oscillation and reduces both PBS establishment and LR formation. In conclusion, we report a novel role for autophagy during development, namely by enacting auxin-induced selective degradation of ARF7 to optimize periodic root branching.
Collapse
Affiliation(s)
- Elise Ebstrup
- Department of Biology, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Jeppe Ansbøl
- Department of Biology, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Ana Paez-Garcia
- Centro de Biotecnología y Genómica de Plantas (Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria-CSIC (INIA/CSIC)). Campus de Montegancedo, Pozuelo de Alarcón, 28223, Madrid, Spain
| | - Henry Culp
- Department of Biology, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Jonathan Chevalier
- Department of Biology, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Pauline Clemmens
- Department of Biology, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Núria S Coll
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Bellaterra, 08193, Spain
- Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, 08001, Spain
| | - Miguel A Moreno-Risueno
- Centro de Biotecnología y Genómica de Plantas (Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria-CSIC (INIA/CSIC)). Campus de Montegancedo, Pozuelo de Alarcón, 28223, Madrid, Spain
| | - Eleazar Rodriguez
- Department of Biology, University of Copenhagen, 2200, Copenhagen N, Denmark.
| |
Collapse
|
43
|
Lewis CTA, Melhedegaard EG, Ognjanovic MM, Olsen MS, Laitila J, Seaborne RAE, Gronset M, Zhang C, Iwamoto H, Hessel AL, Kuehn MN, Merino C, Amigo N, Frobert O, Giroud S, Staples JF, Goropashnaya AV, Fedorov VB, Barnes B, Toien O, Drew K, Sprenger RJ, Ochala J. Remodeling of skeletal muscle myosin metabolic states in hibernating mammals. eLife 2024; 13:RP94616. [PMID: 38752835 PMCID: PMC11098559 DOI: 10.7554/elife.94616] [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] [Indexed: 05/18/2024] Open
Abstract
Hibernation is a period of metabolic suppression utilized by many small and large mammal species to survive during winter periods. As the underlying cellular and molecular mechanisms remain incompletely understood, our study aimed to determine whether skeletal muscle myosin and its metabolic efficiency undergo alterations during hibernation to optimize energy utilization. We isolated muscle fibers from small hibernators, Ictidomys tridecemlineatus and Eliomys quercinus and larger hibernators, Ursus arctos and Ursus americanus. We then conducted loaded Mant-ATP chase experiments alongside X-ray diffraction to measure resting myosin dynamics and its ATP demand. In parallel, we performed multiple proteomics analyses. Our results showed a preservation of myosin structure in U. arctos and U. americanus during hibernation, whilst in I. tridecemlineatus and E. quercinus, changes in myosin metabolic states during torpor unexpectedly led to higher levels in energy expenditure of type II, fast-twitch muscle fibers at ambient lab temperatures (20 °C). Upon repeating loaded Mant-ATP chase experiments at 8 °C (near the body temperature of torpid animals), we found that myosin ATP consumption in type II muscle fibers was reduced by 77-107% during torpor compared to active periods. Additionally, we observed Myh2 hyper-phosphorylation during torpor in I. tridecemilineatus, which was predicted to stabilize the myosin molecule. This may act as a potential molecular mechanism mitigating myosin-associated increases in skeletal muscle energy expenditure during periods of torpor in response to cold exposure. Altogether, we demonstrate that resting myosin is altered in hibernating mammals, contributing to significant changes to the ATP consumption of skeletal muscle. Additionally, we observe that it is further altered in response to cold exposure and highlight myosin as a potentially contributor to skeletal muscle non-shivering thermogenesis.
Collapse
Affiliation(s)
| | | | - Marija M Ognjanovic
- Department of Biomedical Sciences, University of CopenhagenCopenhagenDenmark
| | - Mathilde S Olsen
- Department of Biomedical Sciences, University of CopenhagenCopenhagenDenmark
| | - Jenni Laitila
- Department of Biomedical Sciences, University of CopenhagenCopenhagenDenmark
| | - Robert AE Seaborne
- Department of Biomedical Sciences, University of CopenhagenCopenhagenDenmark
- Centre for Human and Applied Physiological Sciences, Faculty of Life Sciences & Medicine, King’s College LondonLondonUnited Kingdom
| | - Magnus Gronset
- Department of Cellular and Molecular Medicine, University of CopenhagenCopenhagenDenmark
| | - Changxin Zhang
- Department of Computational Medicine and Bioinformatics, University of MichiganAnn ArborUnited States
| | - Hiroyuki Iwamoto
- Spring-8, Japan Synchrotron Radiation Research InstituteHyogoJapan
| | - Anthony L Hessel
- Institute of Physiology II, University of MuensterMuensterGermany
- Accelerated Muscle Biotechnologies ConsultantsBostonUnited States
| | - Michel N Kuehn
- Institute of Physiology II, University of MuensterMuensterGermany
- Accelerated Muscle Biotechnologies ConsultantsBostonUnited States
| | | | | | - Ole Frobert
- Department of Clinical Medicine, Faculty of Health, Aarhus UniversityAarhusDenmark
- Faculty of Health, Department of Cardiology, Örebro UniversityÖrebroSweden
| | - Sylvain Giroud
- Energetics Lab, Department of Biology, Northern Michigan UniversityMarquetteUnited States
- Research Institute of Wildlife Ecology, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine ViennaViennaAustria
| | - James F Staples
- Department of Biology, University of Western OntarioLondonCanada
| | - Anna V Goropashnaya
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska FairbanksFairbanksUnited States
| | - Vadim B Fedorov
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska FairbanksFairbanksUnited States
| | - Brian Barnes
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska FairbanksFairbanksUnited States
| | - Oivind Toien
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska FairbanksFairbanksUnited States
| | - Kelly Drew
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska FairbanksFairbanksUnited States
| | - Ryan J Sprenger
- Department of Zoology, University of British ColumbiaVancouverCanada
| | - Julien Ochala
- Department of Biomedical Sciences, University of CopenhagenCopenhagenDenmark
| |
Collapse
|
44
|
Callahan TJ, Tripodi IJ, Stefanski AL, Cappelletti L, Taneja SB, Wyrwa JM, Casiraghi E, Matentzoglu NA, Reese J, Silverstein JC, Hoyt CT, Boyce RD, Malec SA, Unni DR, Joachimiak MP, Robinson PN, Mungall CJ, Cavalleri E, Fontana T, Valentini G, Mesiti M, Gillenwater LA, Santangelo B, Vasilevsky NA, Hoehndorf R, Bennett TD, Ryan PB, Hripcsak G, Kahn MG, Bada M, Baumgartner WA, Hunter LE. An open source knowledge graph ecosystem for the life sciences. Sci Data 2024; 11:363. [PMID: 38605048 PMCID: PMC11009265 DOI: 10.1038/s41597-024-03171-w] [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: 07/26/2023] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
Translational research requires data at multiple scales of biological organization. Advancements in sequencing and multi-omics technologies have increased the availability of these data, but researchers face significant integration challenges. Knowledge graphs (KGs) are used to model complex phenomena, and methods exist to construct them automatically. However, tackling complex biomedical integration problems requires flexibility in the way knowledge is modeled. Moreover, existing KG construction methods provide robust tooling at the cost of fixed or limited choices among knowledge representation models. PheKnowLator (Phenotype Knowledge Translator) is a semantic ecosystem for automating the FAIR (Findable, Accessible, Interoperable, and Reusable) construction of ontologically grounded KGs with fully customizable knowledge representation. The ecosystem includes KG construction resources (e.g., data preparation APIs), analysis tools (e.g., SPARQL endpoint resources and abstraction algorithms), and benchmarks (e.g., prebuilt KGs). We evaluated the ecosystem by systematically comparing it to existing open-source KG construction methods and by analyzing its computational performance when used to construct 12 different large-scale KGs. With flexible knowledge representation, PheKnowLator enables fully customizable KGs without compromising performance or usability.
Collapse
Affiliation(s)
- Tiffany J Callahan
- Computational Bioscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - Ignacio J Tripodi
- Computer Science Department, Interdisciplinary Quantitative Biology, University of Colorado Boulder, Boulder, CO, 80301, USA
| | - Adrianne L Stefanski
- Computational Bioscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Luca Cappelletti
- AnacletoLab, Dipartimento di Informatica, Universit`a degli Studi di Milano, Via Celoria 18, 20133, Milan, Italy
| | - Sanya B Taneja
- Intelligent Systems Program, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Jordan M Wyrwa
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Elena Casiraghi
- AnacletoLab, Dipartimento di Informatica, Universit`a degli Studi di Milano, Via Celoria 18, 20133, Milan, Italy
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | | | - Justin Reese
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Jonathan C Silverstein
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15206, USA
| | - Charles Tapley Hoyt
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Richard D Boyce
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15206, USA
| | - Scott A Malec
- Division of Translational Informatics, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Deepak R Unni
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Marcin P Joachimiak
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Peter N Robinson
- Berlin Institute of Health at Charité-Universitatsmedizin, 10117, Berlin, Germany
| | - Christopher J Mungall
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Emanuele Cavalleri
- AnacletoLab, Dipartimento di Informatica, Universit`a degli Studi di Milano, Via Celoria 18, 20133, Milan, Italy
| | - Tommaso Fontana
- AnacletoLab, Dipartimento di Informatica, Universit`a degli Studi di Milano, Via Celoria 18, 20133, Milan, Italy
| | - Giorgio Valentini
- AnacletoLab, Dipartimento di Informatica, Universit`a degli Studi di Milano, Via Celoria 18, 20133, Milan, Italy
- ELLIS, European Laboratory for Learning and Intelligent Systems, Milan Unit, Italy
| | - Marco Mesiti
- AnacletoLab, Dipartimento di Informatica, Universit`a degli Studi di Milano, Via Celoria 18, 20133, Milan, Italy
| | - Lucas A Gillenwater
- Computational Bioscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Brook Santangelo
- Computational Bioscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Nicole A Vasilevsky
- Data Collaboration Center, Critical Path Institute, 1840 E River Rd. Suite 100, Tucson, AZ, 85718, USA
| | - Robert Hoehndorf
- Computer, Electrical and Mathematical Sciences & Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Tellen D Bennett
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Patrick B Ryan
- Janssen Research and Development, Raritan, NJ, 08869, USA
| | - George Hripcsak
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Michael G Kahn
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Michael Bada
- Division of General Internal Medicine, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - William A Baumgartner
- Division of General Internal Medicine, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| | - Lawrence E Hunter
- Computational Bioscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
45
|
Zhou J, Gu J, Sun X, Ye Q, Wu X, Xi J, Han J, Liu Y. Supramolecular Chiral Binding Affinity-Achieved Efficient Synergistic Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308493. [PMID: 38380492 DOI: 10.1002/advs.202308493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/03/2024] [Indexed: 02/22/2024]
Abstract
Supramolecular chirality-mediated selective interaction among native assemblies is essential for precise disease diagnosis and treatment. Herein, to fully understand the supramolecular chiral binding affinity-achieved therapeutic efficiency, supramolecular chiral nanoparticles (WP5⊃D/L-Arg+DOX+ICG) with the chirality transfer from chiral arginine (D/L-Arg) to water-soluble pillar[5]arene (WP5) are developed through non-covalent interactions, in which an anticancer drug (DOX, doxorubicin hydrochloride) and a photothermal agent (ICG, indocyanine green) are successfully loaded. Interestingly, the WP5⊃D-Arg nanoparticles show 107 folds stronger binding capability toward phospholipid-composed liposomes compared with WP5⊃L-Arg. The enantioselective interaction further triggers the supramolecular chirality-specific drug accumulation in cancer cells. As a consequence, WP5⊃D-Arg+DOX+ICG exhibits extremely enhanced chemo-photothermal synergistic therapeutic efficacy (tumor inhibition rate of 99.4%) than that of WP5⊃L-Arg+DOX+ICG (tumor inhibition rate of 56.4%) under the same condition. This work reveals the breakthrough that supramolecular chiral assemblies can induce surprisingly large difference in cancer therapy, providing strong support for the significance of supramolecular chirality in bio-application.
Collapse
Affiliation(s)
- Jinfeng Zhou
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225002, P. R. China
| | - Jiake Gu
- School of Medicine, Yangzhou University, Yangzhou, Jiangsu, 225002, P. R. China
| | - Xiaohuan Sun
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225002, P. R. China
| | - Qianyun Ye
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225002, P. R. China
| | - Xuan Wu
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225002, P. R. China
| | - Juqun Xi
- School of Medicine, Yangzhou University, Yangzhou, Jiangsu, 225002, P. R. China
| | - Jie Han
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225002, P. R. China
| | - Yu Liu
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, P. R. China
| |
Collapse
|
46
|
Terranova N, Renard D, Shahin MH, Menon S, Cao Y, Hop CECA, Hayes S, Madrasi K, Stodtmann S, Tensfeldt T, Vaddady P, Ellinwood N, Lu J. Artificial Intelligence for Quantitative Modeling in Drug Discovery and Development: An Innovation and Quality Consortium Perspective on Use Cases and Best Practices. Clin Pharmacol Ther 2024; 115:658-672. [PMID: 37716910 DOI: 10.1002/cpt.3053] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
Recent breakthroughs in artificial intelligence (AI) and machine learning (ML) have ushered in a new era of possibilities across various scientific domains. One area where these advancements hold significant promise is model-informed drug discovery and development (MID3). To foster a wider adoption and acceptance of these advanced algorithms, the Innovation and Quality (IQ) Consortium initiated the AI/ML working group in 2021 with the aim of promoting their acceptance among the broader scientific community as well as by regulatory agencies. By drawing insights from workshops organized by the working group and attended by key stakeholders across the biopharma industry, academia, and regulatory agencies, this white paper provides a perspective from the IQ Consortium. The range of applications covered in this white paper encompass the following thematic topics: (i) AI/ML-enabled Analytics for Pharmacometrics and Quantitative Systems Pharmacology (QSP) Workflows; (ii) Explainable Artificial Intelligence and its Applications in Disease Progression Modeling; (iii) Natural Language Processing (NLP) in Quantitative Pharmacology Modeling; and (iv) AI/ML Utilization in Drug Discovery. Additionally, the paper offers a set of best practices to ensure an effective and responsible use of AI, including considering the context of use, explainability and generalizability of models, and having human-in-the-loop. We believe that embracing the transformative power of AI in quantitative modeling while adopting a set of good practices can unlock new opportunities for innovation, increase efficiency, and ultimately bring benefits to patients.
Collapse
Affiliation(s)
- Nadia Terranova
- Quantitative Pharmacology, Merck KGaA, Lausanne, Switzerland
| | - Didier Renard
- Full Development Pharmacometrics, Novartis Pharma AG, Basel, Switzerland
| | | | - Sujatha Menon
- Clinical Pharmacology, Pfizer Inc., Groton, Connecticut, USA
| | - Youfang Cao
- Clinical Pharmacology and Translational Medicine, Eisai Inc., Nutley, New Jersey, USA
| | | | - Sean Hayes
- Quantitative Pharmacology & Pharmacometrics, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Kumpal Madrasi
- Modeling & Simulation, Sanofi, Bridgewater, New Jersey, USA
| | - Sven Stodtmann
- Pharmacometrics, AbbVie Deutschland GmbH & Co. KG, Ludwigshafen, Germany
| | | | - Pavan Vaddady
- Quantitative Clinical Pharmacology, Daiichi Sankyo, Inc., Basking Ridge, New Jersey, USA
| | | | - James Lu
- Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| |
Collapse
|
47
|
Ghandikota SK, Jegga AG. Application of artificial intelligence and machine learning in drug repurposing. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 205:171-211. [PMID: 38789178 DOI: 10.1016/bs.pmbts.2024.03.030] [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: 05/26/2024]
Abstract
The purpose of drug repurposing is to leverage previously approved drugs for a particular disease indication and apply them to another disease. It can be seen as a faster and more cost-effective approach to drug discovery and a powerful tool for achieving precision medicine. In addition, drug repurposing can be used to identify therapeutic candidates for rare diseases and phenotypic conditions with limited information on disease biology. Machine learning and artificial intelligence (AI) methodologies have enabled the construction of effective, data-driven repurposing pipelines by integrating and analyzing large-scale biomedical data. Recent technological advances, especially in heterogeneous network mining and natural language processing, have opened up exciting new opportunities and analytical strategies for drug repurposing. In this review, we first introduce the challenges in repurposing approaches and highlight some success stories, including those during the COVID-19 pandemic. Next, we review some existing computational frameworks in the literature, organized on the basis of the type of biomedical input data analyzed and the computational algorithms involved. In conclusion, we outline some exciting new directions that drug repurposing research may take, as pioneered by the generative AI revolution.
Collapse
Affiliation(s)
- Sudhir K Ghandikota
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Anil G Jegga
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.
| |
Collapse
|
48
|
Gaudry A, Pagni M, Mehl F, Moretti S, Quiros-Guerrero LM, Cappelletti L, Rutz A, Kaiser M, Marcourt L, Queiroz EF, Ioset JR, Grondin A, David B, Wolfender JL, Allard PM. A Sample-Centric and Knowledge-Driven Computational Framework for Natural Products Drug Discovery. ACS CENTRAL SCIENCE 2024; 10:494-510. [PMID: 38559298 PMCID: PMC10979503 DOI: 10.1021/acscentsci.3c00800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The ENPKG framework organizes large heterogeneous metabolomics data sets as a knowledge graph, offering exciting opportunities for drug discovery and chemodiversity characterization.
Collapse
Affiliation(s)
- Arnaud Gaudry
- Institute of Pharmaceutical
Sciences of Western Switzerland, University
of Geneva, 1211 Geneva 4, Switzerland
- School of Pharmaceutical Sciences, University
of Geneva, 1211 Geneva 4, Switzerland
| | - Marco Pagni
- Vital-IT, SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Florence Mehl
- Vital-IT, SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Sébastien Moretti
- Vital-IT, SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Luis-Manuel Quiros-Guerrero
- Institute of Pharmaceutical
Sciences of Western Switzerland, University
of Geneva, 1211 Geneva 4, Switzerland
- School of Pharmaceutical Sciences, University
of Geneva, 1211 Geneva 4, Switzerland
| | - Luca Cappelletti
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Adriano Rutz
- Institute of Pharmaceutical
Sciences of Western Switzerland, University
of Geneva, 1211 Geneva 4, Switzerland
- School of Pharmaceutical Sciences, University
of Geneva, 1211 Geneva 4, Switzerland
| | - Marcel Kaiser
- Department of Medical
and Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland
- Faculty of Science, University of Basel, 4002 Basel, Switzerland
| | - Laurence Marcourt
- Institute of Pharmaceutical
Sciences of Western Switzerland, University
of Geneva, 1211 Geneva 4, Switzerland
- School of Pharmaceutical Sciences, University
of Geneva, 1211 Geneva 4, Switzerland
| | - Emerson Ferreira Queiroz
- Institute of Pharmaceutical
Sciences of Western Switzerland, University
of Geneva, 1211 Geneva 4, Switzerland
- School of Pharmaceutical Sciences, University
of Geneva, 1211 Geneva 4, Switzerland
| | - Jean-Robert Ioset
- Drugs
for Neglected Diseases Initiative (DNDi), 1202 Geneva, Switzerland
| | - Antonio Grondin
- Green Mission Pierre Fabre, Institut de Recherche Pierre Fabre, 31562 Toulouse, France
| | - Bruno David
- Green Mission Pierre Fabre, Institut de Recherche Pierre Fabre, 31562 Toulouse, France
| | - Jean-Luc Wolfender
- Institute of Pharmaceutical
Sciences of Western Switzerland, University
of Geneva, 1211 Geneva 4, Switzerland
- School of Pharmaceutical Sciences, University
of Geneva, 1211 Geneva 4, Switzerland
| | - Pierre-Marie Allard
- Institute of Pharmaceutical
Sciences of Western Switzerland, University
of Geneva, 1211 Geneva 4, Switzerland
- School of Pharmaceutical Sciences, University
of Geneva, 1211 Geneva 4, Switzerland
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| |
Collapse
|
49
|
Luige J, Armaos A, Tartaglia GG, Ørom UAV. Predicting nuclear G-quadruplex RNA-binding proteins with roles in transcription and phase separation. Nat Commun 2024; 15:2585. [PMID: 38519458 PMCID: PMC10959947 DOI: 10.1038/s41467-024-46731-9] [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: 04/06/2023] [Accepted: 03/08/2024] [Indexed: 03/25/2024] Open
Abstract
RNA-binding proteins are central for many biological processes and their characterization has demonstrated a broad range of functions as well as a wide spectrum of target structures. RNA G-quadruplexes are important regulatory elements occurring in both coding and non-coding transcripts, yet our knowledge of their structure-based interactions is at present limited. Here, using theoretical predictions and experimental approaches, we show that many chromatin-binding proteins bind to RNA G-quadruplexes, and we classify them based on their RNA G-quadruplex-binding potential. Combining experimental identification of nuclear RNA G-quadruplex-binding proteins with computational approaches, we build a prediction tool that assigns probability score for a nuclear protein to bind RNA G-quadruplexes. We show that predicted G-quadruplex RNA-binding proteins exhibit a high degree of protein disorder and hydrophilicity and suggest involvement in both transcription and phase-separation into membrane-less organelles. Finally, we present the G4-Folded/UNfolded Nuclear Interaction Explorer System (G4-FUNNIES) for estimating RNA G4-binding propensities at http://service.tartaglialab.com/new_submission/G4FUNNIES .
Collapse
Affiliation(s)
- Johanna Luige
- RNA Biology and Innovation, Institute of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Alexandros Armaos
- Centre for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), Via Enrico Melen, 83, 16152, Genova, Italy
| | - Gian Gaetano Tartaglia
- Centre for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), Via Enrico Melen, 83, 16152, Genova, Italy.
- Catalan Institution for Research and Advanced Studies ICREA Passeig Lluis Companys, 23 08010, Barcelona, Spain.
| | - Ulf Andersson Vang Ørom
- RNA Biology and Innovation, Institute of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
50
|
Stampe NK, Ottenheijm ME, Drici L, Wewer Albrechtsen NJ, Nielsen AB, Christoffersen C, Warming PE, Engstrøm T, Winkel BG, Jabbari R, Tfelt-Hansen J, Glinge C. Discovery of plasma proteins associated with ventricular fibrillation during first ST-elevation myocardial infarction via proteomics. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2024; 13:264-272. [PMID: 37811694 DOI: 10.1093/ehjacc/zuad125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/23/2023] [Accepted: 10/06/2023] [Indexed: 10/10/2023]
Abstract
AIMS The underlying biological mechanisms of ventricular fibrillation (VF) during acute myocardial infarction are largely unknown. To our knowledge, this is the first proteomic study for this trait, with the aim to identify and characterize proteins that are associated with VF during first ST-elevation myocardial infarction (STEMI). METHODS AND RESULTS We included 230 participants from a Danish ongoing case-control study on patients with first STEMI with VF (case, n = 110) and without VF (control, n = 120) before guided catheter insertion for primary percutaneous coronary intervention. The plasma proteome was investigated using mass spectrometry-based proteomics on plasma samples collected within 24 h of symptom onset, and one patient was excluded in quality control. In 229 STEMI patients {72% men, median age 62 years [interquartile range (IQR): 54-70]}, a median of 257 proteins (IQR: 244-281) were quantified per patient. A total of 26 proteins were associated with VF; these proteins were involved in several biological processes including blood coagulation, haemostasis, and immunity. After correcting for multiple testing, two up-regulated proteins remained significantly associated with VF, actin beta-like 2 [ACTBL2, fold change (FC) 2.25, P < 0.001, q = 0.023], and coagulation factor XIII-A (F13A1, FC 1.48, P < 0.001, q = 0.023). None of the proteins were correlated with anterior infarct location. CONCLUSION Ventricular fibrillation due to first STEMI was significantly associated with two up-regulated proteins (ACTBL2 and F13A1), suggesting that they may represent novel underlying molecular VF mechanisms. Further research is needed to determine whether these proteins are predictive biomarkers or acute phase response proteins to VF during acute ischaemia.
Collapse
Affiliation(s)
- Niels Kjær Stampe
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital-Rigshospitalet, Inge Lehmanns Vej 7, Copenhagen 2100, Denmark
| | - Maud Eline Ottenheijm
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital Bispebjerg Hospital, Copenhagen, Denmark
| | - Lylia Drici
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital Bispebjerg Hospital, Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital Bispebjerg Hospital, Copenhagen, Denmark
| | - Annelaura Bach Nielsen
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital Bispebjerg Hospital, Copenhagen, Denmark
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Centre of Diagnostic Investigation, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peder Emil Warming
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital-Rigshospitalet, Inge Lehmanns Vej 7, Copenhagen 2100, Denmark
| | - Thomas Engstrøm
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital-Rigshospitalet, Inge Lehmanns Vej 7, Copenhagen 2100, Denmark
| | - Bo Gregers Winkel
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital-Rigshospitalet, Inge Lehmanns Vej 7, Copenhagen 2100, Denmark
| | - Reza Jabbari
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital-Rigshospitalet, Inge Lehmanns Vej 7, Copenhagen 2100, Denmark
| | - Jacob Tfelt-Hansen
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital-Rigshospitalet, Inge Lehmanns Vej 7, Copenhagen 2100, Denmark
- Department of Forensic Medicine, Faculty of Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Glinge
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital-Rigshospitalet, Inge Lehmanns Vej 7, Copenhagen 2100, Denmark
| |
Collapse
|