1
|
Halder A, Jadhav PA, Maitra A, Banerjee A, Hole A, Epari S, Shetty P, Moiyadi A, Chilkapati MK, Srivastava S. Serum Metabolomics Profiling Coupled with Machine Learning Identifies Potential Diagnostic and Prognostic Candidate Markers in Meningioma Using Raman Spectroscopy, ATR-FTIR, and LC-MS/MS. J Proteome Res 2025; 24:1180-1196. [PMID: 40000599 DOI: 10.1021/acs.jproteome.4c00806] [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: 02/27/2025]
Abstract
Meningioma, the most prevalent brain tumor, poses significant challenges due to its unclear transition from low-grade to aggressive forms, with limited knowledge about grade-specific markers. We have utilized vibrational spectroscopic techniques such as ATR-FTIR and Raman spectroscopy, alongside LC-MS/MS-based mass spectrometry to understand the systemic cues and evaluate them for clinical practice. The acquired Raman and ATR-FTIR spectra of 46 meningioma patients (27 low-grade and 19 high-grade) and 8 healthy individuals revealed 98.15% and 83.33% accuracy based on PC-LDA. The grade classification revealed an accuracy of around 70%, implying the presence of subtypes and transition phases. The observed alterations corresponded to lipids, nucleic acids, and proteins. Further, the LC-MS/MS-based study identified different derivatives of cholines, indoles, lipids, sphingosine, tryptophan, and their respective metabolic pathways as contributors in tumorigenesis and progression. Further, PRM-based targeted validation and feature selection was carried out on 43 meningioma patients and 17 healthy controls. Glycochenodeoxycholic acid, indole-3-acetic acid, trans-3-indoleacrylic acid, glycodeoxycholic acid, 5α-dihydrotestosteroneglucornide, and glycocholic acid segregated meningioma samples with an accuracy of around 90% while features like indole-3-acetic acid, stercobilin, sphingosine-1-phosphate, deoxycholic acid, and citric acid could classify grades with around 70% accuracy. These findings suggest that further validation across larger cohorts could enhance its usage in clinical settings.
Collapse
Affiliation(s)
- Ankit Halder
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Priyanka A Jadhav
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Sector-22, Kharghar, Navi Mumbai 410210, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Archisman Maitra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Arghya Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Arti Hole
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Sector-22, Kharghar, Navi Mumbai 410210, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Sridhar Epari
- Department of Pathology, Tata Memorial Centre, Mumbai 400012, India
| | - Prakash Shetty
- Department of Neurosurgery, Tata Memorial Centre, Mumbai 400012, India
| | - Aliasgar Moiyadi
- Department of Pathology, Tata Memorial Centre, Mumbai 400012, India
| | - Murali Krishna Chilkapati
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Sector-22, Kharghar, Navi Mumbai 410210, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
2
|
Bate N, Lane D, Evans SE, Salim F, Allcock NS, Haigh R, Sale JE, Jones DJL, Brindle NPJ. Engineered Receptor Capture Combined with Mass Spectrometry Enables High-Throughput Detection and Quantitation of SARS-CoV-2 Spike Protein. JACS AU 2025; 5:747-755. [PMID: 40017752 PMCID: PMC11862925 DOI: 10.1021/jacsau.4c00980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 03/01/2025]
Abstract
Mass spectrometry (MS) is a potentially powerful approach for the diagnostic detection of SARS-CoV-2 and other viruses. However, MS detection is compromised when viral antigens are present at low concentrations, especially in complex biological media. We hypothesized that viral receptors could be used for viral target capture to enable detection by MS under such conditions. This was tested using the extracellular domain of the SARS-CoV-2 receptor ACE2. To maximize recovery of the target protein, directed protein evolution was first used to increase the affinity of ACE2 for spike protein. This generated an evolved ACE2 with increased binding affinity for the spike protein receptor-binding domain (RBD). However, as with other affinity-enhanced evolved forms of ACE2, binding was sensitive to mutations in variant RBDs. As an alternative strategy to maximize capture, the native ACE2 extracellular domain was engineered for increased binding by the addition of an oligomerization scaffold to create pentameric ACE2. This bound extremely tightly to SARS-CoV-2 RBD, with an increase in apparent affinity of several thousand-fold over monomeric ACE2, and RBD retention of more than 8 h. Immobilization of multimeric ACE2 enabled quantitative enrichment of viral spike protein from saliva and increased the sensitivity of detection by MS. These data show that capture by engineered receptors combined with MS can be an effective, rapid method for detection and quantitation of target protein. A similar approach could be used for attachment proteins of other viruses or any target protein for which there are suitable receptors.
Collapse
Affiliation(s)
- Neil Bate
- Department
of Cardiovascular Sciences, University of
Leicester, University Road, Leicester, Leicester LE1 7RH, U.K.
| | - Dan Lane
- Department
of Cardiovascular Sciences, University of
Leicester, University Road, Leicester, Leicester LE1 7RH, U.K.
- van
Geest MS-OMICS Facility, University of Leicester, University Road, Leicester, Leicester LE1 7RH, U.K.
| | - Sian E. Evans
- Leicester
Drug Discovery & Diagnostics, University
of Leicester, University Road, Leicester, Leicester LE1 7RH, U.K.
| | - Farah Salim
- Department
of Cardiovascular Sciences, University of
Leicester, University Road, Leicester, Leicester LE1 7RH, U.K.
- van
Geest MS-OMICS Facility, University of Leicester, University Road, Leicester, Leicester LE1 7RH, U.K.
| | - Natalie S. Allcock
- Electron
Microscopy Facility, Core Biotechnology Services, University of Leicester, University Road, Leicester, Leicester LE1 7RH, U.K.
| | - Richard Haigh
- Leicester
Drug Discovery & Diagnostics, University
of Leicester, University Road, Leicester, Leicester LE1 7RH, U.K.
| | - Julian E. Sale
- MRC
Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, U.K.
| | - Donald J. L. Jones
- van
Geest MS-OMICS Facility, University of Leicester, University Road, Leicester, Leicester LE1 7RH, U.K.
- Department
of Genetics, Genomics & Cancer Sciences, University of Leicester, University Road, Leicester, Leicester LE1 7RH, U.K.
| | - Nicholas P. J. Brindle
- Department
of Cardiovascular Sciences, University of
Leicester, University Road, Leicester, Leicester LE1 7RH, U.K.
- Department
of Molecular & Cell Biology, University
of Leicester, University Road, Leicester, Leicester LE1 7RH, U.K.
- Leicester
Institute for Structural & Chemical Biology, University of Leicester, University Road, Leicester, Leicester LE1 7RH, U.K.
| |
Collapse
|
3
|
Coyle E, Leclercq M, Gotti C, Roux-Dalvai F, Droit A. ProPickML: Advancing Clinical Diagnostics with Automated Peak Picking in Label-Free Targeted Proteomics. J Proteome Res 2025; 24:244-255. [PMID: 39644253 PMCID: PMC11705220 DOI: 10.1021/acs.jproteome.4c00689] [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: 08/12/2024] [Revised: 11/15/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
In targeted proteomics utilizing Selected Reaction Monitoring (SRM), the precise detection of specific peptides within complex mixtures remains a significant challenge, particularly due to noise and interference in chromatograms. Existing methodologies, such as isotopic labeling and scoring algorithms, offer partial solutions but are constrained by high run times and elevated false discovery rates. To address these limitations, we have developed ProPickML a machine learning-based tool designed to accurately identify peptide peaks across diverse data sets, independent of the assumed presence of the peptide. This model was trained on a manually labeled data set and subsequently validated to assess its predictive accuracy. The results demonstrate that the model reliably identifies peptide peaks in the presence of noise, achieving a Matthews correlation coefficient (MCC) of 0.81 on an independent test data set, surpassing mProphet's MCC of 0.71. Implemented in R as ProPickML, this tool offers a competitive, cost-effective alternative to existing techniques, significantly reducing reliance on isotopic labeling and enhancing the accuracy of peptide identification in SRM workflows.
Collapse
Affiliation(s)
- Elloise Coyle
- Computational
Biology Laboratory, Centre de recherche du CHU de Québec, Université Laval, Québec City, Québec G1V 4G2, Canada
| | - Mickaël Leclercq
- Computational
Biology Laboratory, Centre de recherche du CHU de Québec, Université Laval, Québec City, Québec G1V 4G2, Canada
| | - Clarisse Gotti
- Proteomics
Platform, Centre de recherche du CHU de Québec, Université Laval, Québec City, Québec G1V 4G2, Canada
| | - Florence Roux-Dalvai
- Proteomics
Platform, Centre de recherche du CHU de Québec, Université Laval, Québec City, Québec G1V 4G2, Canada
| | - Arnaud Droit
- Computational
Biology Laboratory, Centre de recherche du CHU de Québec, Université Laval, Québec City, Québec G1V 4G2, Canada
- Proteomics
Platform, Centre de recherche du CHU de Québec, Université Laval, Québec City, Québec G1V 4G2, Canada
| |
Collapse
|
4
|
Sharma G, Gupta DP, Ganguly K, Anand MP, Srivastava S. Meta-Analysis and DIA-MS-Based Proteomic Investigation of COPD Patients and Asymptomatic Smokers in the Indian Population. J Proteome Res 2024; 23:4973-4987. [PMID: 39436829 DOI: 10.1021/acs.jproteome.4c00463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is India's second largest cause of death and is largely caused by smoking. Asymptomatic smokers develop COPD due to genetic, environmental, and molecular variables, making early screening crucial. Data-independent acquisition mass spectrometry (DIA-MS) based-proteomics offers an unbiased method to analyze proteomic profiles. This study is the first to use DIA-based proteomics to analyze individual serum samples from three distinct male cohorts: healthy individuals (n = 10), asymptomatic smokers (n = 10), and COPD patients (n = 10). This comprehensive approach identified 667 proteins with a 1% false discovery rate. Differentially expressed proteins included 40 in the normal versus asymptomatic comparison, 88 in the COPD versus normal comparison, and 40 in the COPD versus asymptomatic comparison. Among them, protein-associated genes such as PRDX6, ELANE, PRKCSH, PRTN3, and MNDA could help differentiate COPD from asymptomatic smokers, while ELANE, H3-3A, IGHE, SLC4A1, and SERPINA11 could differentiate COPD from healthy subjects. Pathway enrichment and protein-protein interaction analyses revealed significant alterations in hemostasis, immune system functions, fibrin clot formation, and post-translational protein modifications. Key proteins were validated using a parallel reaction monitoring assay. DIA data are available via ProteomeXchange with identifier PXD055242. Our findings reveal key protein classifiers in COPD patients, asymptomatic smokers, and healthy individuals, helping clinicians understand disease pathobiology and improve disease management and quality of life.
Collapse
Affiliation(s)
- Gautam Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| | - Debarghya Pratim Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| | - Koustav Ganguly
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Mahesh Padukudru Anand
- Department of Respiratory Medicine, JSS Medical College, JSSAHER, Mysore, Karnataka 570015, India
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| |
Collapse
|
5
|
Kreft IC, van de Geer A, Smit ER, van der Zwaan C, van Alphen FPJ, Meijer AB, Nur E, Hoogendijk AJ, Kuijpers TW, van den Biggelaar M. Plasma Profiling of Acute Myeloid Leukemia With Fever- and Infection-Related Complications During Chemotherapy-Induced Neutropenia. Cancer Rep (Hoboken) 2024; 7:e70024. [PMID: 39441646 PMCID: PMC11498059 DOI: 10.1002/cnr2.70024] [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: 10/31/2023] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a heterogenous and complex blood cancer requiring aggressive treatment. Early identification and prediction of the complications following treatment is vital for effective disease management. AIMS We explored associations between plasma protein levels and fever- and infection-related complications in 26 AML patients during chemotherapy-induced neutropenia. MATERIAL AND METHODS Longitudinal plasma profiling was conducted using data-dependent mass spectrometry analysis. RESULTS Mass spectrometry-based plasma profiling data correlated well with laboratory parameters, including C-reactive protein, and revealed a broader inflammation protein network associated with fever- and infection-related complications. DISCUSSION AND CONCLUSION These data indicate the potential of longitudinal plasma profiling in AML patients for identifying and predicting complications that may aid in improved disease monitoring and treatment.
Collapse
Affiliation(s)
- Iris C. Kreft
- Department of Molecular HematologySanquin ResearchAmsterdamThe Netherlands
| | - Annemarie van de Geer
- Department of Blood Cell Research, Division Research and Landsteiner Laboratory of Amsterdam UMCSanquin Blood SupplyAmsterdamThe Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious DiseasesEmma Children's Hospital, Amsterdam UMCAmsterdamThe Netherlands
| | - Eva R. Smit
- Department of Molecular HematologySanquin ResearchAmsterdamThe Netherlands
| | | | | | - Alexander B. Meijer
- Department of Molecular HematologySanquin ResearchAmsterdamThe Netherlands
- Department of Biomolecular Mass Spectrometry and ProteomicsUtrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht UniversityUtrechtThe Netherlands
| | - Erfan Nur
- Department of HematologyAmsterdam UMC, location AMCAmsterdamThe Netherlands
| | - Arie J. Hoogendijk
- Department of Molecular HematologySanquin ResearchAmsterdamThe Netherlands
| | - Taco W. Kuijpers
- Department of Blood Cell Research, Division Research and Landsteiner Laboratory of Amsterdam UMCSanquin Blood SupplyAmsterdamThe Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious DiseasesEmma Children's Hospital, Amsterdam UMCAmsterdamThe Netherlands
| | | |
Collapse
|
6
|
Zhang Z, Hu Y, Zheng X, Chen C, Zhao Y, Lin H, He N. Differential short-term and long-term metabolic and cytokine responses to infection of severe fever with thrombocytopenia syndrome virus. Metabolomics 2024; 20:84. [PMID: 39066899 DOI: 10.1007/s11306-024-02150-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 06/28/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease caused by the SFTS virus (SFTSV), which has a wide geographic distribution. The primary clinical manifestations of SFTS are fever and thrombocytopenia, with multiorgan failure being the leading cause of death. While most patients recover with treatment, little is known about the potential long-term metabolic effects of SFTSV infection. OBJECTIVES This study aimed to shed light on dysregulated metabolic pathways and cytokine responses following SFTSV infection, which pose significant risks to the short-term and long-term health of affected individuals. METHODS Fourteen laboratory-confirmed clinical SFTS cases and thirty-eight healthy controls including 18 SFTSV IgG-positive and 20 IgG-negative individuals were recruited from Taizhou city of Zhejiang province, Eastern China. Inclusion criteria of healthy controls included residing in the study area for at least one year, absence of fever or other symptoms in the past two weeks, and no history of SFTS diagnosis. Ultrahigh-performance liquid chromatography-mass spectrometry (UHPLC-MS) was used to obtain the relative abundance of plasma metabolites. Short-term metabolites refer to transient alterations present only during SFTSV infection, while long-term metabolites persistently deviate from normal levels even after recovery from SFTSV infection. Additionally, the concentrations of 12 cytokines were quantified through fluorescence intensity measurements. Differential metabolites were screened using orthogonal projections to latent structures discriminant analysis (OPLS-DA) and the Wilcoxon rank test. Metabolic pathway analysis was performed using MetaboAnalyst. Between-group differences of metabolites and cytokines were examined using the Wilcoxon rank test. Correlation matrices between identified metabolites and cytokines were analyzed using Spearman's method. RESULTS AND CONCLUSIONS We screened 122 long-term metabolites and 108 short-term metabolites by analytical comparisons and analyzed their correlations with 12 cytokines. Glycerophospholipid metabolism (GPL) was identified as a significant short-term metabolic pathway suggesting that the activation of GPL might be linked to the self-replication of SFTSV, whereas pentose phosphate pathway and alanine, aspartate, and glutamate metabolism were indicated as significant long-term metabolic pathways playing a role in combating long-standing oxidative stress in the patients. Furthermore, our study suggests a new perspective that α-ketoglutarate could serve as a dietary supplement to protect recovering SFTS patients.
Collapse
Affiliation(s)
- Zhiyi Zhang
- School of Public Health, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Yafei Hu
- Taizhou City Center for Disease Control and Prevention, Taizhou, Zhejiang Province, China
| | - Xiang Zheng
- Taizhou City Center for Disease Control and Prevention, Taizhou, Zhejiang Province, China
| | - Cairong Chen
- Taizhou City Center for Disease Control and Prevention, Taizhou, Zhejiang Province, China
| | - Yishuang Zhao
- Taizhou City Center for Disease Control and Prevention, Taizhou, Zhejiang Province, China
| | - Haijiang Lin
- School of Public Health, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
- Taizhou City Center for Disease Control and Prevention, Taizhou, Zhejiang Province, China.
| | - Na He
- School of Public Health, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
- Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Agamah FE, Ederveen THA, Skelton M, Martin DP, Chimusa ER, ’t Hoen PAC. Network-based integrative multi-omics approach reveals biosignatures specific to COVID-19 disease phases. Front Mol Biosci 2024; 11:1393240. [PMID: 39040605 PMCID: PMC11260748 DOI: 10.3389/fmolb.2024.1393240] [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: 02/28/2024] [Accepted: 05/22/2024] [Indexed: 07/24/2024] Open
Abstract
Background COVID-19 disease is characterized by a spectrum of disease phases (mild, moderate, and severe). Each disease phase is marked by changes in omics profiles with corresponding changes in the expression of features (biosignatures). However, integrative analysis of multiple omics data from different experiments across studies to investigate biosignatures at various disease phases is limited. Exploring an integrative multi-omics profile analysis through a network approach could be used to determine biosignatures associated with specific disease phases and enable the examination of the relationships between the biosignatures. Aim To identify and characterize biosignatures underlying various COVID-19 disease phases in an integrative multi-omics data analysis. Method We leveraged a multi-omics network-based approach to integrate transcriptomics, metabolomics, proteomics, and lipidomics data. The World Health Organization Ordinal Scale WHO Ordinal Scale was used as a disease severity reference to harmonize COVID-19 patient metadata across two studies with independent data. A unified COVID-19 knowledge graph was constructed by assembling a disease-specific interactome from the literature and databases. Disease-state specific omics-graphs were constructed by integrating multi-omics data with the unified COVID-19 knowledge graph. We expanded on the network layers of multiXrank, a random walk with restart on multilayer network algorithm, to explore disease state omics-specific graphs and perform enrichment analysis. Results Network analysis revealed the biosignatures involved in inducing chemokines and inflammatory responses as hubs in the severe and moderate disease phases. We observed distinct biosignatures between severe and moderate disease phases as compared to mild-moderate and mild-severe disease phases. Mild COVID-19 cases were characterized by a unique biosignature comprising C-C Motif Chemokine Ligand 4 (CCL4), and Interferon Regulatory Factor 1 (IRF1). Hepatocyte Growth Factor (HGF), Matrix Metallopeptidase 12 (MMP12), Interleukin 10 (IL10), Nuclear Factor Kappa B Subunit 1 (NFKB1), and suberoylcarnitine form hubs in the omics network that characterizes the moderate disease state. The severe cases were marked by biosignatures such as Signal Transducer and Activator of Transcription 1 (STAT1), Superoxide Dismutase 2 (SOD2), HGF, taurine, lysophosphatidylcholine, diacylglycerol, triglycerides, and sphingomyelin that characterize the disease state. Conclusion This study identified both biosignatures of different omics types enriched in disease-related pathways and their associated interactions (such as protein-protein, protein-transcript, protein-metabolite, transcript-metabolite, and lipid-lipid interactions) that are unique to mild, moderate, and severe COVID-19 disease states. These biosignatures include molecular features that underlie the observed clinical heterogeneity of COVID-19 and emphasize the need for disease-phase-specific treatment strategies. The approach implemented here can be used to find associations between transcripts, proteins, lipids, and metabolites in other diseases.
Collapse
Affiliation(s)
- Francis E. Agamah
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Thomas H. A. Ederveen
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | - Michelle Skelton
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Darren P. Martin
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Emile R. Chimusa
- Department of Applied Science, Faculty of Health and Life Sciences, Northumbria University, Newcastle, United Kingdom
| | - Peter A. C. ’t Hoen
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| |
Collapse
|
8
|
Rosario-Rodríguez LJ, Cantres-Rosario YM, Carrasquillo-Carrión K, Rosa-Díaz A, Rodríguez-De Jesús AE, Rivera-Nieves V, Tosado-Rodríguez EL, Méndez LB, Roche-Lima A, Bertrán J, Meléndez LM. Plasma Proteins Associated with COVID-19 Severity in Puerto Rico. Int J Mol Sci 2024; 25:5426. [PMID: 38791465 PMCID: PMC11121485 DOI: 10.3390/ijms25105426] [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: 04/07/2024] [Revised: 05/10/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
Viral strains, age, and host factors are associated with variable immune responses against SARS-CoV-2 and disease severity. Puerto Ricans have a genetic mixture of races: European, African, and Native American. We hypothesized that unique host proteins/pathways are associated with COVID-19 disease severity in Puerto Rico. Following IRB approval, a total of 95 unvaccinated men and women aged 21-71 years old were recruited in Puerto Rico from 2020-2021. Plasma samples were collected from COVID-19-positive subjects (n = 39) and COVID-19-negative individuals (n = 56) during acute disease. COVID-19-positive individuals were stratified based on symptomatology as follows: mild (n = 18), moderate (n = 13), and severe (n = 8). Quantitative proteomics was performed in plasma samples using tandem mass tag (TMT) labeling. Labeled peptides were subjected to LC/MS/MS and analyzed by Proteome Discoverer (version 2.5), Limma software (version 3.41.15), and Ingenuity Pathways Analysis (IPA, version 22.0.2). Cytokines were quantified using a human cytokine array. Proteomics analyses of severely affected COVID-19-positive individuals revealed 58 differentially expressed proteins. Cadherin-13, which participates in synaptogenesis, was downregulated in severe patients and validated by ELISA. Cytokine immunoassay showed that TNF-α levels decreased with disease severity. This study uncovers potential host predictors of COVID-19 severity and new avenues for treatment in Puerto Ricans.
Collapse
Affiliation(s)
- Lester J. Rosario-Rodríguez
- Department of Microbiology and Medical Zoology, University of Puerto Rico, Medical Sciences Campus, San Juan 00935, Puerto Rico;
| | - Yadira M. Cantres-Rosario
- Translational Proteomics Center, Research Capacity Core, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan 00935, Puerto Rico; (Y.M.C.-R.); (A.E.R.-D.J.)
| | - Kelvin Carrasquillo-Carrión
- Integrated Informatics, Research Capacity Core, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan 00935, Puerto Rico; (K.C.-C.); (E.L.T.-R.); (A.R.-L.)
| | - Alexandra Rosa-Díaz
- Interdisciplinary Studies, Natural Sciences, University of Puerto Rico, Río Piedras Campus, San Juan 00925, Puerto Rico; (A.R.-D.); (V.R.-N.)
| | - Ana E. Rodríguez-De Jesús
- Translational Proteomics Center, Research Capacity Core, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan 00935, Puerto Rico; (Y.M.C.-R.); (A.E.R.-D.J.)
| | - Verónica Rivera-Nieves
- Interdisciplinary Studies, Natural Sciences, University of Puerto Rico, Río Piedras Campus, San Juan 00925, Puerto Rico; (A.R.-D.); (V.R.-N.)
| | - Eduardo L. Tosado-Rodríguez
- Integrated Informatics, Research Capacity Core, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan 00935, Puerto Rico; (K.C.-C.); (E.L.T.-R.); (A.R.-L.)
| | - Loyda B. Méndez
- Department of Science & Technology, Ana G. Mendez University, Carolina 00928, Puerto Rico;
| | - Abiel Roche-Lima
- Integrated Informatics, Research Capacity Core, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan 00935, Puerto Rico; (K.C.-C.); (E.L.T.-R.); (A.R.-L.)
| | - Jorge Bertrán
- Infectious Diseases, Auxilio Mutuo Hospital, San Juan 00919, Puerto Rico;
| | - Loyda M. Meléndez
- Department of Microbiology and Medical Zoology, University of Puerto Rico, Medical Sciences Campus, San Juan 00935, Puerto Rico;
- Translational Proteomics Center, Research Capacity Core, Center for Collaborative Research in Health Disparities, University of Puerto Rico, Medical Sciences Campus, San Juan 00935, Puerto Rico; (Y.M.C.-R.); (A.E.R.-D.J.)
| |
Collapse
|
9
|
Camelo ALM, Zamora Obando HR, Rocha I, Dias AC, Mesquita ADS, Simionato AVC. COVID-19 and Comorbidities: What Has Been Unveiled by Metabolomics? Metabolites 2024; 14:195. [PMID: 38668323 PMCID: PMC11051775 DOI: 10.3390/metabo14040195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/14/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
The COVID-19 pandemic has brought about diverse impacts on the global population. Individuals with comorbidities were more susceptible to the severe symptoms caused by the virus. Within the crisis scenario, metabolomics represents a potential area of science capable of providing relevant information for understanding the metabolic pathways associated with the intricate interaction between the viral disease and previous comorbidities. This work aims to provide a comprehensive description of the scientific production pertaining to metabolomics within the specific context of COVID-19 and comorbidities, while highlighting promising areas for exploration by those interested in the subject. In this review, we highlighted the studies of metabolomics that indicated a variety of metabolites associated with comorbidities and COVID-19. Furthermore, we observed that the understanding of the metabolic processes involved between comorbidities and COVID-19 is limited due to the urgent need to report disease outcomes in individuals with comorbidities. The overlap of two or more comorbidities associated with the severity of COVID-19 hinders the comprehension of the significance of each condition. Most identified studies are observational, with a restricted number of patients, due to challenges in sample collection amidst the emergent situation.
Collapse
Affiliation(s)
- André Luiz Melo Camelo
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Hans Rolando Zamora Obando
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Isabela Rocha
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Aline Cristina Dias
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Alessandra de Sousa Mesquita
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Ana Valéria Colnaghi Simionato
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
- National Institute of Science and Technology for Bioanalytics—INCTBio, Institute of Chemistry, Universidade Estadual de (UNICAMP), Campinas 13083-970, São Paulo, Brazil
| |
Collapse
|
10
|
Anandakrishnan N, Yi Z, Sun Z, Liu T, Haydak J, Eddy S, Jayaraman P, DeFronzo S, Saha A, Sun Q, Yang D, Mendoza A, Mosoyan G, Wen HH, Schaub JA, Fu J, Kehrer T, Menon R, Otto EA, Godfrey B, Suarez-Farinas M, Leffters S, Twumasi A, Meliambro K, Charney AW, García-Sastre A, Campbell KN, Gusella GL, He JC, Miorin L, Nadkarni GN, Wisnivesky J, Li H, Kretzler M, Coca SG, Chan L, Zhang W, Azeloglu EU. Integrated multiomics implicates dysregulation of ECM and cell adhesion pathways as drivers of severe COVID-associated kidney injury. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.18.24304401. [PMID: 38562892 PMCID: PMC10984064 DOI: 10.1101/2024.03.18.24304401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
COVID-19 has been a significant public health concern for the last four years; however, little is known about the mechanisms that lead to severe COVID-associated kidney injury. In this multicenter study, we combined quantitative deep urinary proteomics and machine learning to predict severe acute outcomes in hospitalized COVID-19 patients. Using a 10-fold cross-validated random forest algorithm, we identified a set of urinary proteins that demonstrated predictive power for both discovery and validation set with 87% and 79% accuracy, respectively. These predictive urinary biomarkers were recapitulated in non-COVID acute kidney injury revealing overlapping injury mechanisms. We further combined orthogonal multiomics datasets to understand the mechanisms that drive severe COVID-associated kidney injury. Functional overlap and network analysis of urinary proteomics, plasma proteomics and urine sediment single-cell RNA sequencing showed that extracellular matrix and autophagy-associated pathways were uniquely impacted in severe COVID-19. Differentially abundant proteins associated with these pathways exhibited high expression in cells in the juxtamedullary nephron, endothelial cells, and podocytes, indicating that these kidney cell types could be potential targets. Further, single-cell transcriptomic analysis of kidney organoids infected with SARS-CoV-2 revealed dysregulation of extracellular matrix organization in multiple nephron segments, recapitulating the clinically observed fibrotic response across multiomics datasets. Ligand-receptor interaction analysis of the podocyte and tubule organoid clusters showed significant reduction and loss of interaction between integrins and basement membrane receptors in the infected kidney organoids. Collectively, these data suggest that extracellular matrix degradation and adhesion-associated mechanisms could be a main driver of COVID-associated kidney injury and severe outcomes.
Collapse
|
11
|
English A, McDaid D, Lynch SM, McLaughlin J, Cooper E, Wingfield B, Kelly M, Bhavsar M, McGilligan V, Irwin RE, Bucholc M, Zhang SD, Shukla P, Rai TS, Bjourson AJ, Murray E, Gibson DS, Walsh C. Genomic, Proteomic, and Phenotypic Biomarkers of COVID-19 Severity: Protocol for a Retrospective Observational Study. JMIR Res Protoc 2024; 13:e50733. [PMID: 38354037 PMCID: PMC10868637 DOI: 10.2196/50733] [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: 07/11/2023] [Revised: 10/23/2023] [Accepted: 11/09/2023] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Health organizations and countries around the world have found it difficult to control the spread of COVID-19. To minimize the future impact on the UK National Health Service and improve patient care, there is a pressing need to identify individuals who are at a higher risk of being hospitalized because of severe COVID-19. Early targeted work was successful in identifying angiotensin-converting enzyme-2 receptors and type II transmembrane serine protease dependency as drivers of severe infection. Although a targeted approach highlights key pathways, a multiomics approach will provide a clearer and more comprehensive picture of severe COVID-19 etiology and progression. OBJECTIVE The COVID-19 Response Study aims to carry out an integrated multiomics analysis to identify biomarkers in blood and saliva that could contribute to host susceptibility to SARS-CoV-2 and the development of severe COVID-19. METHODS The COVID-19 Response Study aims to recruit 1000 people who recovered from SARS-CoV-2 infection in both community and hospital settings on the island of Ireland. This protocol describes the retrospective observational study component carried out in Northern Ireland (NI; Cohort A); the Republic of Ireland cohort will be described separately. For all NI participants (n=519), SARS-CoV-2 infection has been confirmed by reverse transcription-quantitative polymerase chain reaction. A prospective Cohort B of 40 patients is also being followed up at 1, 3, 6, and 12 months postinfection to assess longitudinal symptom frequency and immune response. Data will be sourced from whole blood, saliva samples, and clinical data from the electronic care records, the general health questionnaire, and a 12-item general health questionnaire mental health survey. Saliva and blood samples were processed to extract DNA and RNA before whole-genome sequencing, RNA sequencing, DNA methylation analysis, microbiome analysis, 16S ribosomal RNA gene sequencing, and proteomic analysis were performed on the plasma. Multiomics data will be combined with clinical data to produce sensitive and specific prognostic models for severity risk. RESULTS An initial demographic and clinical profile of the NI Cohort A has been completed. A total of 249 hospitalized patients and 270 nonhospitalized patients were recruited, of whom 184 (64.3%) were female, and the mean age was 45.4 (SD 13) years. High levels of comorbidity were evident in the hospitalized cohort, with cardiovascular disease and metabolic and respiratory disorders being the most significant (P<.001), grouped according to the International Classification of Diseases 10 codes. CONCLUSIONS This study will provide a comprehensive opportunity to study the mechanisms of COVID-19 severity in recontactable participants. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/50733.
Collapse
Affiliation(s)
- Andrew English
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
- National Horizons Centre, Teesside University, Middlesbrough, United Kingdom
| | - Darren McDaid
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
| | - Seodhna M Lynch
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
| | - Joseph McLaughlin
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
| | - Eamonn Cooper
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
| | - Benjamin Wingfield
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
| | - Martin Kelly
- Western Health Social Care Trust, Londonderry, United Kingdom
| | - Manav Bhavsar
- Western Health Social Care Trust, Londonderry, United Kingdom
| | - Victoria McGilligan
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
| | - Rachelle E Irwin
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
| | - Magda Bucholc
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
| | - Shu-Dong Zhang
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
| | - Priyank Shukla
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
| | - Taranjit Singh Rai
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
| | - Anthony J Bjourson
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
| | - Elaine Murray
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
| | - David S Gibson
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry/Londonderry, United Kingdom
| | - Colum Walsh
- Department of Biomedical and Clinical Sciences, Linköping University, Uppsala, Sweden
| |
Collapse
|
12
|
Ren L, Ning L, Yang Y, Yang T, Li X, Tan S, Ge P, Li S, Luo N, Tao P, Zhang Y. MetaboliteCOVID: A manually curated database of metabolite markers for COVID-19. Comput Biol Med 2023; 167:107661. [PMID: 37925911 DOI: 10.1016/j.compbiomed.2023.107661] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/07/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
In the realm of unraveling COVID-19's intricacies, numerous metabolomic investigations have been conducted to discern the unique metabolic traits exhibited within infected patients. These endeavors have yielded a substantial reservoir of potential data pertaining to metabolic biomarkers linked to the virus. Despite these strides, a comprehensive and meticulously structured database housing these crucial biomarkers remains absent. In this study, we developed MetaboliteCOVID, a manually curated database of COVID-19-related metabolite markers. The database currently comprises 665 manually selected entries of significantly altered metabolites associated with early diagnosis, disease severity, prognosis, and drug response in COVID-19, encompassing 337 metabolites. Additionally, the database offers a user-friendly interface, containing abundant information for querying, browsing, and analyzing COVID-19-related abnormal metabolites in different body fluids. In summary, we believe that this database will effectively facilitate research on the functions and mechanisms of COVID-19-related metabolic biomarkers, thereby advancing both basic and clinical research on COVID-19. MetaboliteCOVID is free available at: https://cellknowledge.com.cn/MetaboliteCOVID.
Collapse
Affiliation(s)
- Liping Ren
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China; Innovative Institute of Chinese Medicine and Pharmacy, Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lin Ning
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China
| | - Yu Yang
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China
| | - Ting Yang
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China
| | - Xinyu Li
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China
| | - Shanshan Tan
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China
| | - Peixin Ge
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China
| | - Shun Li
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China
| | - Nanchao Luo
- School of Computer Science and Technology, Aba Teachers College, WenChuan, Sichuan, 623002, China.
| | - Pei Tao
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan, 611731, China.
| | - Yang Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
13
|
Zhang F, Luna A, Tan T, Chen Y, Sander C, Guo T. COVIDpro: Database for Mining Protein Dysregulation in Patients with COVID-19. J Proteome Res 2023; 22:2847-2859. [PMID: 37555633 DOI: 10.1021/acs.jproteome.3c00092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
The ongoing pandemic of the coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 still has limited treatment options. Our understanding of the molecular dysregulations that occur in response to infection remains incomplete. We developed a web application COVIDpro (https://www.guomics.com/covidPro/) that includes proteomics data obtained from 41 original studies conducted in 32 hospitals worldwide, involving 3077 patients and covering 19 types of clinical specimens, predominantly plasma and serum. The data set encompasses 53 protein expression matrices, comprising a total of 5434 samples and 14,403 unique proteins. We identified a panel of proteins that exhibit significant dysregulation, enabling the classification of COVID-19 patients into severe and non-severe disease categories. The proteomic signatures achieved promising results in distinguishing severe cases, with a mean area under the curve of 0.87 and accuracy of 0.80 across five independent test sets. COVIDpro serves as a valuable resource for testing hypotheses and exploring potential targets for novel treatments in COVID-19 patients.
Collapse
Affiliation(s)
- Fangfei Zhang
- Fudan University, 220 Handan Road, Shanghai 200433, China
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province 310024, China
- Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang 310030, China
| | - Augustin Luna
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Broad Institute of MIT and Harvard, Cambridge, Cambridge, Massachusetts 02142, United States
| | - Tingting Tan
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province 310024, China
- Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang 310030, China
| | - Yingdan Chen
- Westlake Omics (Hangzhou) Biotechnology Company Limited, Hangzhou, Zhejiang Province 310024, China
| | - Chris Sander
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Broad Institute of MIT and Harvard, Cambridge, Cambridge, Massachusetts 02142, United States
| | - Tiannan Guo
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province 310024, China
- Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang 310030, China
| |
Collapse
|
14
|
Bernardo L, Lomagno A, Mauri PL, Di Silvestre D. Integration of Omics Data and Network Models to Unveil Negative Aspects of SARS-CoV-2, from Pathogenic Mechanisms to Drug Repurposing. BIOLOGY 2023; 12:1196. [PMID: 37759595 PMCID: PMC10525644 DOI: 10.3390/biology12091196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused the COVID-19 health emergency, affecting and killing millions of people worldwide. Following SARS-CoV-2 infection, COVID-19 patients show a spectrum of symptoms ranging from asymptomatic to very severe manifestations. In particular, bronchial and pulmonary cells, involved at the initial stage, trigger a hyper-inflammation phase, damaging a wide range of organs, including the heart, brain, liver, intestine and kidney. Due to the urgent need for solutions to limit the virus' spread, most efforts were initially devoted to mapping outbreak trajectories and variant emergence, as well as to the rapid search for effective therapeutic strategies. Samples collected from hospitalized or dead COVID-19 patients from the early stages of pandemic have been analyzed over time, and to date they still represent an invaluable source of information to shed light on the molecular mechanisms underlying the organ/tissue damage, the knowledge of which could offer new opportunities for diagnostics and therapeutic designs. For these purposes, in combination with clinical data, omics profiles and network models play a key role providing a holistic view of the pathways, processes and functions most affected by viral infection. In fact, in addition to epidemiological purposes, networks are being increasingly adopted for the integration of multiomics data, and recently their use has expanded to the identification of drug targets or the repositioning of existing drugs. These topics will be covered here by exploring the landscape of SARS-CoV-2 survey-based studies using systems biology approaches derived from omics data, paying particular attention to those that have considered samples of human origin.
Collapse
Affiliation(s)
| | | | | | - Dario Di Silvestre
- Institute for Biomedical Technologies—National Research Council (ITB-CNR), 20054 Segrate, Italy; (L.B.); (A.L.); (P.L.M.)
| |
Collapse
|
15
|
Rajoria S, Halder A, Tarnekar I, Pal P, Bansal P, Srivastava S. Detection of Mutant Peptides of SARS-CoV-2 Variants by LC/MS in the DDA Approach Using an In-House Database. J Proteome Res 2023; 22:1816-1827. [PMID: 37093804 PMCID: PMC10152398 DOI: 10.1021/acs.jproteome.2c00819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Indexed: 04/25/2023]
Abstract
Equipped with a dramatically high mutation rate, which happens to be a signature of RNA viruses, SARS-CoV-2 trampled across the globe infecting individuals of all ages and ethnicities. As the variants of concern (VOC) loomed large, definitive detection of SARS-CoV-2 strains became a matter of utmost importance in epidemiological and clinical research. Besides, unveiling the disease pathogenesis at the molecular level and deciphering the therapeutic targets became key priorities since the emergence of the pandemic. Mass spectrometry has been largely used in this regard. A critical part of mass spectrometric analyses is the proteome database required for the identification of peptides. Presently, the mutational information on proteins available on SARS-CoV-2 databases cannot be used to analyze data extracted from mass spectrometers. Hence, we developed the novel Mutant Peptide Database (MPD) for the mass spectrometry (MS)-based identification of mutated peptides, which contains information from 11 proteins of SARS-CoV-2 from a total of 21,549 SARS-CoV-2 variants across different regions of India. The database was validated using clinical samples, and its applicability was also demonstrated with the mutated peptides extracted from the literature. We believe that MPD will support broad-spectrum MS-based studies like viral detection, disease pathogenesis, and therapeutics with respect to SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Sakshi Rajoria
- Department of Biosciences and Bioengineering,
Indian Institute of Technology Bombay, Mumbai 400076,
India
| | - Ankit Halder
- Department of Biosciences and Bioengineering,
Indian Institute of Technology Bombay, Mumbai 400076,
India
| | - Ishita Tarnekar
- Thadomal Shahani Engineering
College, P.G. Kher Marg T.P.S III, Bandra West, Mumbai 400050,
India
| | - Pracheta Pal
- Department of Life Sciences, Presidency
University, 86/1 College Street, Kolkata 700073, West Bengal,
India
| | - Prakhar Bansal
- Department of Electrical Engineering,
Indian Institute of Technology Bombay, Mumbai 400076,
India
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering,
Indian Institute of Technology Bombay, Mumbai 400076,
India
| |
Collapse
|
16
|
Chisanga M, Williams H, Boudreau D, Pelletier JN, Trottier S, Masson JF. Label-Free SERS for Rapid Differentiation of SARS-CoV-2-Induced Serum Metabolic Profiles in Non-Hospitalized Adults. Anal Chem 2023; 95:3638-3646. [PMID: 36763490 PMCID: PMC9940618 DOI: 10.1021/acs.analchem.2c04514] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
COVID-19 represents a multi-system infectious disease with broad-spectrum manifestations, including changes in host metabolic processes connected to the disease pathogenesis. Understanding biochemical dysregulation patterns as a consequence of COVID-19 illness promises to be crucial for tracking disease course and clinical outcomes. Surface-enhanced Raman scattering (SERS) has attracted considerable interest in biomedical diagnostics for the sensitive detection of intrinsic profiles of unique fingerprints of serum biomolecules indicative of SARS-CoV-2 infection in a label-free format. Here, we applied label-free SERS and chemometrics for rapid interrogation of temporal metabolic dynamics in longitudinal sera of mildly infected non-hospitalized patients (n = 22), at 4 and 16 weeks post PCR-positive diagnosis, and compared them with negative controls (n = 8). SERS spectral markers revealed distinct metabolic profiles in patient sera that significantly deviated from the healthy metabolic state at the two sampling time intervals. Multivariate and univariate analyses of the spectral data identified abundance dynamics in amino acids, lipids, and protein vibrations as the key spectral features underlying the metabolic differences detected in convalescent samples and perhaps associated with patient recovery progression. A validation study performed using spontaneous Raman spectroscopy yielded spectral data results that corroborated SERS spectral findings and confirmed the detected disease-specific molecular phenotypes in clinical samples. Label-free SERS promises to be a valuable analytical technique for rapid screening of the metabolic phenotype induced by SARS-CoV-2 infection to allow appropriate healthcare intervention.
Collapse
Affiliation(s)
- Malama Chisanga
- Department
of Chemistry, Québec Centre for Advanced Materials (QCAM),
Regroupement Québécois sur les Matériaux de Pointe
(RQMP), and Centre Interdisciplinaire de Recherche sur le Cerveau
et l’Apprentissage (CIRCA), Université
de Montréal, CP 6128 Succ. Centre-Ville, Montreal, Québec H3C 3J7, Canada
| | - Hannah Williams
- Department
of Chemistry, Québec Centre for Advanced Materials (QCAM),
Regroupement Québécois sur les Matériaux de Pointe
(RQMP), and Centre Interdisciplinaire de Recherche sur le Cerveau
et l’Apprentissage (CIRCA), Université
de Montréal, CP 6128 Succ. Centre-Ville, Montreal, Québec H3C 3J7, Canada
| | - Denis Boudreau
- Department
of Chemistry and Centre for Optics, Photonics and Lasers (COPL), Université Laval, 1045, av. de la Médecine, Québec, Québec G1V 0A6, Canada
| | - Joelle N. Pelletier
- Department
of Chemistry, Department of Biochemistry and PROTEO, Québec
Network for Research on Protein Function, Engineering and Applications, Université de Montréal, CP 6128 Succ. Centre-Ville, Montreal, Québec H3C 3J7, Canada
| | - Sylvie Trottier
- Centre
de Recherche du Centre Hospitalier Universitaire de Québec
and Département de Microbiologie-Infectiologie et d’Immunologie, Université Laval, 2705, boulevard Laurier, Québec, Québec G1V 4G2, Canada
| | - Jean-Francois Masson
- Department
of Chemistry, Québec Centre for Advanced Materials (QCAM),
Regroupement Québécois sur les Matériaux de Pointe
(RQMP), and Centre Interdisciplinaire de Recherche sur le Cerveau
et l’Apprentissage (CIRCA), Université
de Montréal, CP 6128 Succ. Centre-Ville, Montreal, Québec H3C 3J7, Canada,. Phone: +1-514-343-7342
| |
Collapse
|
17
|
Lim EHT, van Amstel RBE, de Boer VV, van Vught LA, de Bruin S, Brouwer MC, Vlaar APJ, van de Beek D. Complement activation in COVID-19 and targeted therapeutic options: A scoping review. Blood Rev 2023; 57:100995. [PMID: 35934552 PMCID: PMC9338830 DOI: 10.1016/j.blre.2022.100995] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/07/2022] [Accepted: 07/27/2022] [Indexed: 01/28/2023]
Abstract
Increasing evidence suggests that activation of the complement system plays a key role in the pathogenesis and disease severity of Coronavirus disease 2019 (COVID-19). We used a systematic approach to create an overview of complement activation in COVID-19 based on histopathological, preclinical, multiomics, observational and clinical interventional studies. A total of 1801 articles from PubMed, EMBASE and Cochrane was screened of which 157 articles were included in this scoping review. Histopathological, preclinical, multiomics and observational studies showed apparent complement activation through all three complement pathways and a correlation with disease severity and mortality. The complement system was targeted at different levels in COVID-19, of which C5 and C5a inhibition seem most promising. Adequately powered, double blind RCTs are necessary in order to further investigate the effect of targeting the complement system in COVID-19.
Collapse
Affiliation(s)
- Endry Hartono Taslim Lim
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Neurology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Rombout Benjamin Ezra van Amstel
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| | - Vieve Victoria de Boer
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands
| | - Lonneke Alette van Vught
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam, the Netherlands
| | - Sanne de Bruin
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| | - Matthijs Christian Brouwer
- Amsterdam UMC location University of Amsterdam, Department of Neurology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Alexander Petrus Johannes Vlaar
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands.
| | - Diederik van de Beek
- Amsterdam UMC location University of Amsterdam, Department of Neurology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Neuroscience, Amsterdam, the Netherlands
| |
Collapse
|
18
|
Rajoria S, Nissa MU, Suvarna K, Khatri H, Srivastava S. Multiomics Data Analysis Workflow to Assess Severity in Longitudinal Plasma Samples of COVID-19 Patients. Data Brief 2022; 46:108765. [PMCID: PMC9678393 DOI: 10.1016/j.dib.2022.108765] [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: 08/06/2022] [Revised: 10/31/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022] Open
Abstract
Elucidation of molecular markers related to the mounted immune response is crucial for the understanding of disease pathogenesis. In this article, we present the mass-spectrometry-based metabolomic and proteomic data of blood plasma of COVID-19 patients collected at two-time points, which showed a transition from non-severe to severe conditions during these time points. Metabolites were extracted and subjected to mass spectrometric analysis using the Q-Exactive mass spectrometer. For proteomic analysis, depleted plasma samples were tryptic digested and subjected to mass spectrometry analysis. The expression of a few significant targeted proteins was also validated by employing the targeted proteomic approach of multiple reaction monitoring (MRM). Integrative pathway analysis was performed with the significant proteins to obtain biological insights into disease severity. For discussion and more information on the dataset creation, please refer to the related full-length article [1].
Collapse
|
19
|
Ciccosanti F, Antonioli M, Sacchi A, Notari S, Farina A, Beccacece A, Fusto M, Vergori A, D'Offizi G, Taglietti F, Antinori A, Nicastri E, Marchioni L, Palmieri F, Ippolito G, Piacentini M, Agrati C, Fimia GM. Proteomic analysis identifies a signature of disease severity in the plasma of COVID-19 pneumonia patients associated to neutrophil, platelet and complement activation. Clin Proteomics 2022; 19:38. [PMID: 36348270 PMCID: PMC9641302 DOI: 10.1186/s12014-022-09377-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/26/2022] [Indexed: 11/10/2022] Open
Abstract
Most patients infected with SARS-CoV-2 display mild symptoms with good prognosis, while 20% of patients suffer from severe viral pneumonia and up to 5% may require intensive care unit (ICU) admission due to severe acute respiratory syndrome, which could be accompanied by multiorgan failure.Plasma proteomics provide valuable and unbiased information about disease progression and therapeutic candidates. Recent proteomic studies have identified molecular changes in plasma of COVID-19 patients that implied significant dysregulation of several aspects of the inflammatory response accompanied by a general metabolic suppression. However, which of these plasma alterations are associated with disease severity remains only partly characterized.A known limitation of proteomic studies of plasma samples is the large difference in the macromolecule abundance, with concentration spanning at least 10 orders of magnitude. To improve the coverage of plasma contents, we performed a deep proteomic analysis of plasma from 10 COVID-19 patients with severe/fatal pneumonia compared to 10 COVID-19 patients with pneumonia who did not require ICU admission (non-ICU). To this aim, plasma samples were first depleted of the most abundant proteins, trypsin digested and peptides subjected to a high pH reversed-phase peptide fractionation before LC-MS analysis.These results highlighted an increase of proteins involved in neutrophil and platelet activity and acute phase response, which is significantly higher in severe/fatal COVID-19 patients when compared to non-ICU ones. Importantly, these changes are associated with a selective induction of complement cascade factors in severe/fatal COVID-19 patients. Data are available via ProteomeXchange with identifier PXD036491. Among these alterations, we confirmed by ELISA that higher levels of the neutrophil granule proteins DEFA3 and LCN2 are present in COVID-19 patients requiring ICU admission when compared to non-ICU and healthy donors.Altogether, our study provided an in-depth view of plasma proteome changes that occur in COVID-19 patients in relation to disease severity, which can be helpful to identify therapeutic strategies to improve the disease outcome.
Collapse
Affiliation(s)
- Fabiola Ciccosanti
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Manuela Antonioli
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Alessandra Sacchi
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Stefania Notari
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Anna Farina
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Alessia Beccacece
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Marisa Fusto
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Alessandra Vergori
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Gianpiero D'Offizi
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Fabrizio Taglietti
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Andrea Antinori
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Emanuele Nicastri
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Luisa Marchioni
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Fabrizio Palmieri
- Infectious Disease-Clinical Department, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
| | - Giuseppe Ippolito
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
- General Directorate for Research and Health Innovation, Italian Ministry of Health, Rome, Italy
| | - Mauro Piacentini
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Chiara Agrati
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy.
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy.
| | - Gian Maria Fimia
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS "L. Spallanzani", Rome, Italy.
- Department of Molecular Medicine, University of Rome "Sapienza", Rome, Italy.
| |
Collapse
|
20
|
Zhang F, Luna A, Tan T, Chen Y, Sander C, Guo T. COVIDpro: Database for mining protein dysregulation in patients with COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.09.27.509819. [PMID: 36203550 PMCID: PMC9536031 DOI: 10.1101/2022.09.27.509819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Background The ongoing pandemic of the coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) still has limited treatment options partially due to our incomplete understanding of the molecular dysregulations of the COVID-19 patients. We aimed to generate a repository and data analysis tools to examine the modulated proteins underlying COVID-19 patients for the discovery of potential therapeutic targets and diagnostic biomarkers. Methods We built a web server containing proteomic expression data from COVID-19 patients with a toolset for user-friendly data analysis and visualization. The web resource covers expert-curated proteomic data from COVID-19 patients published before May 2022. The data were collected from ProteomeXchange and from select publications via PubMed searches and aggregated into a comprehensive dataset. Protein expression by disease subgroups across projects was compared by examining differentially expressed proteins. We also visualize differentially expressed pathways and proteins. Moreover, circulating proteins that differentiated severe cases were nominated as predictive biomarkers. Findings We built and maintain a web server COVIDpro ( https://www.guomics.com/covidPro/ ) containing proteomics data generated by 41 original studies from 32 hospitals worldwide, with data from 3077 patients covering 19 types of clinical specimens, the majority from plasma and sera. 53 protein expression matrices were collected, for a total of 5434 samples and 14,403 unique proteins. Our analyses showed that the lipopolysaccharide-binding protein, as identified in the majority of the studies, was highly expressed in the blood samples of patients with severe disease. A panel of significantly dysregulated proteins was identified to separate patients with severe disease from non-severe disease. Classification of severe disease based on these proteomic signatures on five test sets reached a mean AUC of 0.87 and ACC of 0.80. Interpretation COVIDpro is an online database with an integrated analysis toolkit. It is a unique and valuable resource for testing hypotheses and identifying proteins or pathways that could be targeted by new treatments of COVID-19 patients. Funding National Key R&D Program of China: Key PDPM technologies (2021YFA1301602, 2021YFA1301601, 2021YFA1301603), Zhejiang Provincial Natural Science Foundation for Distinguished Young Scholars (LR19C050001), Hangzhou Agriculture and Society Advancement Program (20190101A04), National Natural Science Foundation of China (81972492) and National Science Fund for Young Scholars (21904107), National Resource for Network Biology (NRNB) from the National Institute of General Medical Sciences (NIGMS-P41 GM103504). Research in context Evidence before this study: Although an increasing number of therapies against COVID-19 are being developed, they are still insufficient, especially with the rise of new variants of concern. This is partially due to our incomplete understanding of the disease’s mechanisms. As data have been collected worldwide, several questions are now worth addressing via meta-analyses. Most COVID-19 drugs function by targeting or affecting proteins. Effectiveness and resistance to therapeutics can be effectively assessed via protein measurements. Empowered by mass spectrometry-based proteomics, protein expression has been characterized in a variety of patient specimens, including body fluids (e.g., serum, plasma, urea) and tissue (i.e., formalin-fixed and paraffin-embedded (FFPE)). We expert-curated proteomic expression data from COVID-19 patients published before May 2022, from the largest proteomic data repository ProteomeXhange as well as from literature search engines. Using this resource, a COVID-19 proteome meta-analysis could provide useful insights into the mechanisms of the disease and identify new potential drug targets.Added value of this study: We integrated many published datasets from patients with COVID-19 from 11 nations, with over 3000 patients and more than 5434 proteome measurements. We collected these datasets in an online database, and generated a toolbox to easily explore, analyze, and visualize the data. Next, we used the database and its associated toolbox to identify new proteins of diagnostic and therapeutic value for COVID-19 treatment. In particular, we identified a set of significantly dysregulated proteins for distinguishing severe from non-severe patients using serum samples.Implications of all the available evidence: COVIDpro will support the navigation and analysis of patterns of dysregulated proteins in various COVID-19 clinical specimens for identification and verification of protein biomarkers and potential therapeutic targets.
Collapse
|
21
|
Margraf A, Perretti M. Immune Cell Plasticity in Inflammation: Insights into Description and Regulation of Immune Cell Phenotypes. Cells 2022; 11:cells11111824. [PMID: 35681519 PMCID: PMC9180515 DOI: 10.3390/cells11111824] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 02/01/2023] Open
Abstract
Inflammation is a life-saving immune reaction occurring in response to invading pathogens. Nonetheless, inflammation can also occur in an uncontrolled, unrestricted manner, leading to chronic disease and organ damage. Mechanisms triggering an inflammatory response, hindering such a response, or leading to its resolution are well-studied but so far insufficiently elucidated with regard to precise therapeutic interventions. Notably, as an immune reaction evolves, requirements and environments for immune cells change, and thus cellular phenotypes adapt and shift, leading to the appearance of distinct cellular subpopulations with new functional features. In this article, we aim to highlight properties of, and overarching regulatory factors involved in, the occurrence of immune cell phenotypes with a special focus on neutrophils, macrophages and platelets. Additionally, we point out implications for both diagnostics and therapeutics in inflammation research.
Collapse
|
22
|
Xian R, Wang C, Gong L, Hang B, Wang W, Zhang X, Du H, Wang F, Shi F. A Species-Specific Strategy for the Identification of Hemocoagulase Agkistrodon halys pallas Based on LC-MS/MS-MRM. Front Mol Biosci 2022; 9:831293. [PMID: 35712351 PMCID: PMC9196937 DOI: 10.3389/fmolb.2022.831293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Hemocoagulase Agkistrodon halys pallas is a complex mixture composed of snake venom thrombin-like enzymes (svTLEs) and small amounts of thrombokinase-like enzymes. It has been widely used as a hemostatic with rapidly growing marketing due to its advantage of localized clotting fibrinogen other than systemic coagulation. However, svTLEs from different species have various structures, functions, and hemostatic mechanisms. To ensure the efficacy and safety of Hemocoagulase Agkistrodon halys pallas, an exclusive and sensitive method has been developed to identify specific marker peptides based on liquid chromatography-tandem mass spectrometry with multiple reaction monitoring (LC-MS/MS-MRM) mode. By combining transcriptomics and proteomics, a series of species-specific peptides of Agkistrodon halys pallas were predicted and examined by LC-MS/MS. After reduction, alkylation, and tryptic digestion were performed on Hemocoagulase Agkistrodon halys pallas, a target peptide TLCAGVMEGGIDTCNR was analyzed by LC-MS/MS-MRM. It offers a new and effective approach for the quality control of Hemocoagulase Agkistrodon halys pallas products. This method is superior to the current assays in terms of sensitivity, specificity, precision, accuracy, and throughput. The strategy can also be applied in studying other important protein-based medicines.
Collapse
Affiliation(s)
- Ruiqing Xian
- Biological Products Inspection Division, Shandong Institute for Food and Drug Control, Jinan, China
- National Medical Product Administration (NMPA) Key Laboratory for Research & Evaluation of Genetic Drugs, Jinan, China
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Congcong Wang
- Biological Products Inspection Division, Shandong Institute for Food and Drug Control, Jinan, China
| | - Liping Gong
- Biological Products Inspection Division, Shandong Institute for Food and Drug Control, Jinan, China
| | - Baojian Hang
- Biological Products Inspection Division, Shandong Institute for Food and Drug Control, Jinan, China
| | - Weijian Wang
- Biological Products Inspection Division, Shandong Institute for Food and Drug Control, Jinan, China
- National Medical Product Administration (NMPA) Key Laboratory for Research & Evaluation of Genetic Drugs, Jinan, China
| | - Xunjie Zhang
- Biological Products Inspection Division, Shandong Institute for Food and Drug Control, Jinan, China
| | - Hongmin Du
- R&D Department, Avanc Pharmaceutical Co., Ltd., Jizhou, China
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Feng Shi
- Biological Products Inspection Division, Shandong Institute for Food and Drug Control, Jinan, China
- National Medical Product Administration (NMPA) Key Laboratory for Research & Evaluation of Genetic Drugs, Jinan, China
| |
Collapse
|
23
|
Multi-omics evaluation of SARS-CoV-2 infected mouse lungs reveals dynamics of host responses. iScience 2022; 25:103967. [PMID: 35224468 PMCID: PMC8863311 DOI: 10.1016/j.isci.2022.103967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 01/04/2022] [Accepted: 02/17/2022] [Indexed: 12/27/2022] Open
Abstract
The outbreak of Coronavirus disease 2019 (COVID-19) throughout the world has caused millions of death, while the dynamics of host responses and the underlying regulation mechanisms during SARS-CoV-2 infection are not well depicted. Lung tissues from a mouse model sensitized to SARS-CoV-2 infection were serially collected at different time points for evaluation of transcriptome, proteome, and phosphoproteome. We showed the ebb and flow of several host responses in the lung across the viral infection. The signaling pathways and kinases regulating networks were alternated at different phases of infection. This multiplex evaluation also revealed that many kinases of the CDK and MAPK family were interactive and served as functional hubs in mediating the signal transduction during SARS-CoV-2 infection. Our study not only revealed the dynamics of lung pathophysiology and their underlying molecular mechanisms during SARS-CoV-2 infection, but also highlighted some molecules and signaling pathways that might guide future investigations on COVID-19 therapies. Multi-omics analysis profiles temporal host responses in SARS-CoV-2 infected lungs Signaling pathways and kinase regulating networks are dynamically altered The CDK and MAPK family are interactive and involved in regulating host responses
Collapse
|
24
|
Rajczewski AT, Jagtap PD, Griffin TJ. An overview of technologies for MS-based proteomics-centric multi-omics. Expert Rev Proteomics 2022; 19:165-181. [PMID: 35466851 PMCID: PMC9613604 DOI: 10.1080/14789450.2022.2070476] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Mass spectrometry-based proteomics reveals dynamic molecular signatures underlying phenotypes reflecting normal and perturbed conditions in living systems. Although valuable on its own, the proteome has only one level of moleclar information, with the genome, epigenome, transcriptome, and metabolome, all providing complementary information. Multi-omic analysis integrating information from one or more of these other domains with proteomic information provides a more complete picture of molecular contributors to dynamic biological systems. AREAS COVERED Here, we discuss the improvements to mass spectrometry-based technologies, focused on peptide-based, bottom-up approaches that have enabled deep, quantitative characterization of complex proteomes. These advances are facilitating the integration of proteomics data with other 'omic information, providing a more complete picture of living systems. We also describe the current state of bioinformatics software and approaches for integrating proteomics and other 'omics data, critical for enabling new discoveries driven by multi-omics. EXPERT COMMENTARY Multi-omics, centered on the integration of proteomics information with other 'omic information, has tremendous promise for biological and biomedical studies. Continued advances in approaches for generating deep, reliable proteomic data and bioinformatics tools aimed at integrating data across 'omic domains will ensure the discoveries offered by these multi-omic studies continue to increase.
Collapse
Affiliation(s)
- Andrew T. Rajczewski
- Department of Biochemistry, Molecular and Cell Biology Building, University of Minnesota, 420 Washington Ave SE 7-129, Minneapolis, MN, 55455, USA
| | - Pratik D. Jagtap
- Department of Biochemistry, Molecular and Cell Biology Building, University of Minnesota, 420 Washington Ave SE 7-129, Minneapolis, MN, 55455, USA,Coauthor, Research Department of Biochemistry, Molecular and Cell Biology Building, University of Minnesota, 420 Washington Ave SE 7-129, Minneapolis, MN, 55455, USA
| | - Timothy J. Griffin
- Department of Biochemistry, Molecular and Cell Biology Building, University of Minnesota, 420 Washington Ave SE 7-129, Minneapolis, MN, 55455, USA,Department of Biochemistry, Molecular and Cell Biology Building, University of Minnesota, 420 Washington Ave SE 7-129, Minneapolis, MN, 55455, USA
| |
Collapse
|
25
|
Costanzo M, Caterino M, Fedele R, Cevenini A, Pontillo M, Barra L, Ruoppolo M. COVIDomics: The Proteomic and Metabolomic Signatures of COVID-19. Int J Mol Sci 2022; 23:ijms23052414. [PMID: 35269564 PMCID: PMC8910221 DOI: 10.3390/ijms23052414] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/04/2022] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Omics-based technologies have been largely adopted during this unprecedented global COVID-19 pandemic, allowing the scientific community to perform research on a large scale to understand the pathobiology of the SARS-CoV-2 infection and its replication into human cells. The application of omics techniques has been addressed to every level of application, from the detection of mutations, methods of diagnosis or monitoring, drug target discovery, and vaccine generation, to the basic definition of the pathophysiological processes and the biochemical mechanisms behind the infection and spread of SARS-CoV-2. Thus, the term COVIDomics wants to include those efforts provided by omics-scale investigations with application to the current COVID-19 research. This review summarizes the diverse pieces of knowledge acquired with the application of COVIDomics techniques, with the main focus on proteomics and metabolomics studies, in order to capture a common signature in terms of proteins, metabolites, and pathways dysregulated in COVID-19 disease. Exploring the multiomics perspective and the concurrent data integration may provide new suitable therapeutic solutions to combat the COVID-19 pandemic.
Collapse
Affiliation(s)
- Michele Costanzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (M.C.); (M.C.); (A.C.)
- CEINGE–Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (R.F.); (M.P.); (L.B.)
| | - Marianna Caterino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (M.C.); (M.C.); (A.C.)
- CEINGE–Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (R.F.); (M.P.); (L.B.)
| | - Roberta Fedele
- CEINGE–Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (R.F.); (M.P.); (L.B.)
| | - Armando Cevenini
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (M.C.); (M.C.); (A.C.)
- CEINGE–Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (R.F.); (M.P.); (L.B.)
| | - Mariarca Pontillo
- CEINGE–Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (R.F.); (M.P.); (L.B.)
| | - Lucia Barra
- CEINGE–Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (R.F.); (M.P.); (L.B.)
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (M.C.); (M.C.); (A.C.)
- CEINGE–Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (R.F.); (M.P.); (L.B.)
- Correspondence:
| |
Collapse
|
26
|
Yang J, Yan Y, Zhong W. Application of omics technology to combat the COVID-19 pandemic. MedComm (Beijing) 2021; 2:381-401. [PMID: 34766152 PMCID: PMC8554664 DOI: 10.1002/mco2.90] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 12/17/2022] Open
Abstract
As of August 27, 2021, the ongoing pandemic of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread to over 220 countries, areas, and territories. Thus far, 214,468,601 confirmed cases, including 4,470,969 deaths, have been reported to the World Health Organization. To combat the COVID-19 pandemic, multiomics-based strategies, including genomics, transcriptomics, proteomics, and metabolomics, have been used to study the diagnosis methods, pathogenesis, prognosis, and potential drug targets of COVID-19. In order to help researchers and clinicians to keep up with the knowledge of COVID-19, we summarized the most recent progresses reported in omics-based research papers. This review discusses omics-based approaches for studying COVID-19, summarizing newly emerged SARS-CoV-2 variants as well as potential diagnostic methods, risk factors, and pathological features of COVID-19. This review can help researchers and clinicians gain insight into COVID-19 features, providing direction for future drug development and guidance for clinical treatment, so that patients can receive appropriate treatment as soon as possible to reduce the risk of disease progression.
Collapse
Affiliation(s)
- Jingjing Yang
- National Engineering Research Center for the Emergency DrugBeijing Institute of Pharmacology and ToxicologyBeijingChina
- School of Pharmaceutical SciencesHainan UniversityHaikouHainanChina
| | - Yunzheng Yan
- National Engineering Research Center for the Emergency DrugBeijing Institute of Pharmacology and ToxicologyBeijingChina
| | - Wu Zhong
- National Engineering Research Center for the Emergency DrugBeijing Institute of Pharmacology and ToxicologyBeijingChina
| |
Collapse
|