1
|
Kramaric T, Thein OS, Parekh D, Scott A, Vangberg A, Beckmann M, Phillips H, Thickett D, Mur LAJ. SARS-CoV2 variants differentially impact on the plasma metabolome. Metabolomics 2025; 21:50. [PMID: 40186806 PMCID: PMC11972186 DOI: 10.1007/s11306-025-02238-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/11/2025] [Indexed: 04/07/2025]
Abstract
INTRODUCTION Infection with severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) leads to COVID19 disease and caused a worldwide pandemic in 2019. Since the first wave of infections, there has been significant antigenic shifts, leading to the emergence of new variants. Today, infections have shifted away from the severe, fatal infection seen in 2019. OBJECTIVE This study aimed to assess how the plasma metabolomes from patients varied with infection with different strains and could reflect disease severity. METHODS Patients with COVID19 not requiring intensive care were recruited between January 2021 and May 2022 from the Queen Elizabeth Hospital Birmingham; 33 patients with alpha, 13 delta and 14 omicron variants. These were compared to 26 age matched contemporaneously recruited controls. Plasma samples were extracted into chloroform/methanol/water (1:2.5/1 v/v) and assessed by flow injection electrospray mass spectrometry (FIE-MS) using an Exactive Orbitrap mass spectrometer. Derived data were assessed using the R based MetaboAnalyst platform. RESULTS Plasma metabolomes from COVID19 patients were clearly different from controls. Metabolite variation could be related to infection with different SARS-CoV2 variants. Variant showed different levels of some phospholipids, ganglioside GD1a and a dihydroxyvitamin D3 derivative. Correlations of the plasma metabolomes indicated negative correlations between selected phospholipids and the levels of C-reactive protein, creatinine, neutrophil and D-dimer. CONCLUSION The plasma metabolomes of COVID19 patients show changes, particularly in phospholipids, which could reflect disease severity and SARS-CoV2 variant infection.
Collapse
Affiliation(s)
- Tina Kramaric
- Department of Life Sciences, Aberystwyth University, Penglais Campus, Aberystwyth, SY23 3DA, UK
| | - Onn Shaun Thein
- Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
- Birmingham Biomedical Research Centre, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR), Birmingham, UK
| | - Dhruv Parekh
- Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
- Birmingham Biomedical Research Centre, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR), Birmingham, UK
| | - Aaron Scott
- Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
- Birmingham Biomedical Research Centre, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR), Birmingham, UK
| | - Andrine Vangberg
- Department of Life Sciences, Aberystwyth University, Penglais Campus, Aberystwyth, SY23 3DA, UK
| | - Manfred Beckmann
- Department of Life Sciences, Aberystwyth University, Penglais Campus, Aberystwyth, SY23 3DA, UK
| | - Helen Phillips
- Department of Life Sciences, Aberystwyth University, Penglais Campus, Aberystwyth, SY23 3DA, UK
| | - David Thickett
- Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
- Birmingham Biomedical Research Centre, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR), Birmingham, UK
| | - Luis A J Mur
- Department of Life Sciences, Aberystwyth University, Penglais Campus, Aberystwyth, SY23 3DA, UK.
| |
Collapse
|
2
|
Lang J, Bernal A, Wist J, Egan S, Bong SH, Millet O, Ryan M, Lee AC, Hall D, Nitschke P, Masuda R, Imrie A, Holmes E, Nicholson J, Loo RL. Longitudinal study on immunologic, lipoproteomic, and inflammatory responses indicates the safety of sequential COVID-19 vaccination. J Mol Med (Berl) 2025; 103:421-433. [PMID: 40074874 PMCID: PMC12003606 DOI: 10.1007/s00109-025-02527-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025]
Abstract
COVID-19 vaccines are crucial in reducing SARS-CoV-2 transmission and severe health outcomes. Despite widespread administration, their long-term systemic effects on human metabolism remain inadequately understood. This longitudinal study aims to evaluate IgG responses, 34 cytokines, 112 lipoproteins, and 21 low-molecular-weight metabolites in 33 individuals receiving two to four COVID-19 vaccine doses. Changes in metabolic profiles for the first 16 days post each dose of vaccine, and up to 480 days post-initial dose, were compared to baseline (before vaccination). Additionally, metabolic profiles of vaccinated participants were compared to a reference cohort of unvaccinated individuals without prior exposure to SARS-CoV-2 infection (controls) and SARS-CoV-2 cases. Positive IgG responses were observed in 78.8% (N = 26) of participants after the first dose, reaching 100% with subsequent doses. The most common side effects were localized pain at the injection site and "flu-like" symptoms, reported by > 50% of participants. Systemic side effects, e.g., sore lymph nodes, fatigue, and brain fog, were reported but showed no significant correlations to IgG responses. Transient temporal changes were observed for cytokine IP10 (CXCL10) and glutamic acid around the third vaccine dose. Compared to the reference cohort, 497 vaccinated samples (95.0%) had profiles similar to the controls, while the remaining 26 samples with prior infection exposures were similar to mild cases of SARS-CooV-2 infection. In conclusion, COVID-19 vaccination did not induce lasting changes in inflammatory and metabolic responses, nor did it induce changes similar to mild cases of SARS-CoV-2 infection. This supports the metabolic safety of the vaccine and contributes to increased vaccine confidence. KEY MESSAGES: Minimal changes in inflammatory/metabolic markers up to 480 days post-vaccination. Transient increase in IP10 (CXCL10) and glutamic acid around the third dose. Post-vaccination IgG response did not alter metabolic profiles like SARS-CoV-2 cases. Our findings provide insights into the safety of repeated COVID-19 vaccinations.
Collapse
Affiliation(s)
- Jurissa Lang
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Andres Bernal
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Julien Wist
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
- Chemistry Department, Universidad del Valle, Cali, 76001, Colombia
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London, SW7 2AZ, UK
| | - Siobhon Egan
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Sze How Bong
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Oscar Millet
- Centro de Investigación Cooperativa en Biociencias -CIC bioGUNE, Precision Medicine and Metabolism Laboratory, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, 48160, Derio, Spain
| | - Monique Ryan
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Aude-Claire Lee
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Drew Hall
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Philipp Nitschke
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Reika Masuda
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
| | - Allison Imrie
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA, 6009, Australia
| | - Elaine Holmes
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia
- Nutrition Research, Department of Metabolism, Nutrition and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, London, SW7 2AZ, UK
| | - Jeremy Nicholson
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia.
- Nutrition Research, Department of Metabolism, Nutrition and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, London, SW7 2AZ, UK.
- Institute of Global Health and Innovation, Imperial College London, Faculty Building South Kensington Campus, London, SW7 2AZ, UK.
| | - Ruey Leng Loo
- Australian National Phenome Centre and Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA, 6150, Australia.
| |
Collapse
|
3
|
Ghini V, Pecchioli V, Celli T, Boccia N, Bertini L, Veneziani F, Vannucchi V, Turano P. Metabolomic and lipoproteomic differences and similarities between COVID-19 and other types of pneumonia. Sci Rep 2025; 15:7507. [PMID: 40032933 PMCID: PMC11876652 DOI: 10.1038/s41598-025-91965-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 02/24/2025] [Indexed: 03/05/2025] Open
Abstract
COVID-19 infection has revealed significant effects on the human blood metabolome and lipoproteome, which have been coherently observed in different cohorts worldwide and across the various waves of SARS-CoV-2 pandemic. As one of the main clinical manifestations of COVID-19 is a severe acute respiratory illness, it is pertinent to explore whether this metabolic/lipoproteomic disturbance is associated with the respiratory symptoms. To this purpose we are here reporting comparative1H NMR analyses of the plasma of 252 COVID-19 patients and of patients with non-COVID-19 interstitial (24 individuals) or lobar (21 individuals) pneumonia, all matched by age, gender and disease severity. The analysis is based on 24 metabolites and 114 lipoprotein parameters. Several common traits are observed among the three groups, albeit with some peculiar features characteristic of each group. The main differences were observed between the lobar cases and all the others.
Collapse
Affiliation(s)
- Veronica Ghini
- Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, FI, Italy
| | - Valentina Pecchioli
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), Sesto Fiorentino, FI, Italy
| | - Tommaso Celli
- Internal Medicine, USL Toscana Centro, Santa Maria Nuova Hospital, Florence, Italy
| | - Nunzia Boccia
- Internal Medicine, USL Toscana Centro, Santa Maria Nuova Hospital, Florence, Italy
| | - Laura Bertini
- Internal Medicine, USL Toscana Centro, Santa Maria Nuova Hospital, Florence, Italy
| | - Francesca Veneziani
- Laboratory of Clinical Pathology, USL Toscana Centro, Santa Maria Nuova Hospital, Florence, Italy
| | - Vieri Vannucchi
- Internal Medicine, USL Toscana Centro, Santa Maria Nuova Hospital, Florence, Italy
| | - Paola Turano
- Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, FI, Italy.
- CERM, University of Florence, Sesto Fiorentino, FI, Italy.
| |
Collapse
|
4
|
Loveday EK, Welhaven H, Erdogan AE, Hain KS, Domanico LF, Chang CB, June RK, Taylor MP. Starve a cold or feed a fever? Identifying cellular metabolic changes following infection and exposure to SARS-CoV-2. PLoS One 2025; 20:e0305065. [PMID: 39937842 PMCID: PMC11819565 DOI: 10.1371/journal.pone.0305065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 12/03/2024] [Indexed: 02/14/2025] Open
Abstract
Viral infections induce major shifts in cellular metabolism elicited by active viral replication and antiviral responses. For the virus, harnessing cellular metabolism and evading changes that limit replication are essential for productive viral replication. In contrast, the cellular response to infection disrupts metabolic pathways to prevent viral replication and promote an antiviral state in the host cell and neighboring bystander cells. This competition between the virus and cell results in measurable shifts in cellular metabolism that differ depending on the virus, cell type, and extracellular environment. The resulting metabolic shifts can be observed and analyzed using global metabolic profiling techniques to identify pathways that are critical for either viral replication or cellular defense. SARS-CoV-2 is a respiratory virus that can exhibit broad tissue tropism and diverse, yet inconsistent, symptomatology. While the factors that determine the presentation and severity of SARS-CoV-2 infection remain unclear, metabolic syndromes are associated with more severe manifestations of SARS-CoV-2 disease. Despite these observations a critical knowledge gap remains between cellular metabolic responses and SARS-CoV-2 infection. Using a well-established untargeted metabolomics analysis workflow, we compared SARS-CoV-2 infection of human lung carcinoma cells. We identified significant changes in metabolic pathways that correlate with either productive or non-productive viral infection. This information is critical for characterizing the factors that contribute to SARS-CoV-2 replication that could be targeted for therapeutic interventions to limit viral disease.
Collapse
Affiliation(s)
- Emma K. Loveday
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, United States of America
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, Montana, United States of America
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, United States of America
| | - Hope Welhaven
- Chemistry and Biochemistry, Montana State University, Bozeman, Montana, United States of America
| | - Ayten Ebru Erdogan
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, Montana, United States of America
| | - Kyle S. Hain
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, United States of America
| | - Luke F. Domanico
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, United States of America
| | - Connie B. Chang
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, United States of America
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, Montana, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ronald K. June
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, Montana, United States of America
| | - Matthew P. Taylor
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, United States of America
| |
Collapse
|
5
|
Trautwein C. Quantitative Blood Serum IVDr NMR Spectroscopy in Clinical Metabolomics of Cancer, Neurodegeneration, and Internal Medicine. Methods Mol Biol 2025; 2855:427-443. [PMID: 39354321 DOI: 10.1007/978-1-0716-4116-3_24] [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: 10/03/2024]
Abstract
Despite more than two decades of metabolomics having joined the "omics" scenery, to date only a few novel blood metabolite biomarkers have found their way into the clinic. This is changing now by massive large-scale population metabolic phenotyping for both healthy and disease cohorts. Here, nuclear magnetic resonance (NMR) spectroscopy is a method of choice, as typical blood serum markers can be easily quantified and by knowledge of precise reference concentrations, more and more NMR-amenable biomarkers are established, moving NMR from research to clinical application. Besides customized approaches, to date two major commercial platforms have evolved based on either 600 MHz (14.1 Tesla) or 500 MHz (11.7 Tesla) high-field NMR systems. This chapter provides an introduction into the field of quantitative in vitro diagnostics research (IVDr) NMR at 600 MHz and its application within clinical research of cancer, neurodegeneration, and internal medicine.
Collapse
|
6
|
Iftimie S, Amigó N, Martínez-Micaelo N, López-Azcona AF, Martínez-Navidad C, Castañé H, Jiménez-Franco A, Ribalta J, Parra S, Castro A, Camps J, Joven J. Differential analysis of lipoprotein and glycoprotein profiles in bacterial infections and COVID-19 using proton nuclear magnetic resonance and machine learning. Heliyon 2024; 10:e37115. [PMID: 39286163 PMCID: PMC11402779 DOI: 10.1016/j.heliyon.2024.e37115] [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: 02/07/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Background We scrutinized variations in the proton nuclear magnetic resonance (1H NMR) lipoprotein and glycoprotein profiles among hospitalized individuals with infectious diseases. Methods We obtained sera from 124 patients with COVID-19, 50 patients with catheter-related bacterial infections, and 50 healthy volunteers. Results were interpreted using machine learning. Results COVID-19 patients had bigger and more abundant VLDL particles than the control group and higher VLDL-cholesterol and VLDL-triglyceride concentrations. Patients with bacterial infections showed similar trends, but differences often did not reach statistical significance. Both types of patients showed lower LDL-cholesterol concentrations than the controls. LDL were larger, and the number of particles was lower than that of the healthy individuals. HDL particles had decreased cholesterol and increased triglycerides. Small particles were reduced. Glycoproteins were increased in both groups of patients. All these alterations were more pronounced in COVID-19 patients than those with bacterial infections. The diagnostic accuracy of these profiles exceeded 90 % when distinguishing between healthy individuals and patients, and 85 % when differentiating between the two patient groups. Conclusion Our findings highlight the potential of 1H NMR analysis for lipoproteins and glycoproteins as infection biomarkers. Additionally, they reveal differences between viral and bacterial infections, shedding light on an area with promising clinical significance.
Collapse
Affiliation(s)
- Simona Iftimie
- Department of Internal Medicine, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
- Department of Medicine and Surgery, Universitat Rovira i Virgili, Reus, Spain
| | - Núria Amigó
- Department of Medicine and Surgery, Universitat Rovira i Virgili, Reus, Spain
- CIBER of Diabetes and Associated Metabolic Disease (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Biosfer Teslab, Reus, Spain
| | - Neus Martínez-Micaelo
- Department of Medicine and Surgery, Universitat Rovira i Virgili, Reus, Spain
- CIBER of Diabetes and Associated Metabolic Disease (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Biosfer Teslab, Reus, Spain
| | - Ana F López-Azcona
- Department of Internal Medicine, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
- Department of Medicine and Surgery, Universitat Rovira i Virgili, Reus, Spain
| | - Cristian Martínez-Navidad
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Helena Castañé
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Andrea Jiménez-Franco
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Josep Ribalta
- Department of Medicine and Surgery, Universitat Rovira i Virgili, Reus, Spain
- CIBER of Diabetes and Associated Metabolic Disease (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Unitat de Recerca de Lípids i Arteriosclerosi, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Sandra Parra
- Department of Internal Medicine, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
- Department of Medicine and Surgery, Universitat Rovira i Virgili, Reus, Spain
| | - Antoni Castro
- Department of Internal Medicine, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
- Department of Medicine and Surgery, Universitat Rovira i Virgili, Reus, Spain
| | - Jordi Camps
- Department of Medicine and Surgery, Universitat Rovira i Virgili, Reus, Spain
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Jorge Joven
- Department of Medicine and Surgery, Universitat Rovira i Virgili, Reus, Spain
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
7
|
Chao AS, Lin CY, Chiang MH, Lu KY, Tsai CK, Chen KJ, Chien CW, Wu TS, Chang YL, Chao A, Lin G, Chiu CY. Metabolomic profiling of maternal plasma identifies inverse associations of acetate and urea with anti-SARS-CoV-2 antibody titers following COVID-19 vaccination during pregnancy. J Mol Med (Berl) 2024; 102:819-830. [PMID: 38568327 DOI: 10.1007/s00109-024-02438-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 02/19/2024] [Accepted: 03/12/2024] [Indexed: 05/21/2024]
Abstract
We conducted a comprehensive metabolomic analysis of plasma samples obtained from pregnant women who displayed varying post-vaccination antibody titers after receiving mRNA-1273-SARS-CoV-2 vaccines. The study involved 62 pregnant women, all of whom had been vaccinated after reaching 24 weeks of gestation. To quantify post-vaccination plasma antibody titers, we employed binding antibody units (BAU) in accordance with the World Health Organization International Standard. Subsequently, we classified the study participants into three distinct BAU/mL categories: those with high titers (above 2000), medium titers (ranging from 1000 to 2000), and low titers (below 1000). Plasma metabolomic profiling was conducted using 1H nuclear magnetic resonance spectroscopy, and the obtained data were correlated with the categorized antibody titers. Notably, in pregnant women exhibiting elevated anti-SARS-CoV-2 antibody titers, reduced plasma concentrations of acetate and urea were observed. A significant negative correlation between these compounds and antibody titers was also evident. An analysis of metabolomics pathways revealed significant inverse associations between antibody titers and four distinct amino acid metabolic pathways: (1) biosynthesis of phenylalanine, tyrosine, and tryptophan; (2) biosynthesis of valine, leucine, and isoleucine; (3) phenylalanine metabolism; and (4) degradation of valine, leucine, and isoleucine. Additionally, an association between the synthesis and degradation pathways of ketone bodies was evident. In conclusion, we identified different metabolic pathways that underlie the diverse humoral responses triggered by COVID-19 mRNA vaccines during pregnancy. Our data hold significant implications for refining COVID-19 vaccination approaches in expectant mothers. KEY MESSAGES : Anti-SARS-CoV-2 antibody titers decline as the number of days since COVID-19 vaccination increases. Anti-SARS-CoV-2 antibody titers are inversely associated with acetate, a microbial-derived metabolite, and urea. Amino acid metabolism is significantly associated with SARS-CoV-2 antibody titers.
Collapse
Affiliation(s)
- An-Shine Chao
- Department of Obstetrics and Gynecology, New Taipei Municipal Tu Cheng Hospital, New Taipei City, Taiwan.
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Chiao-Yun Lin
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
- Gynecologic Cancer Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Meng-Han Chiang
- Clinical Metabolomics Core Lab, Chang , Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Kuan-Ying Lu
- Clinical Metabolomics Core Lab, Chang , Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Cheng-Kun Tsai
- Clinical Metabolomics Core Lab, Chang , Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Kuan-Ju Chen
- Department of Obstetrics and Gynecology, New Taipei Municipal Tu Cheng Hospital, New Taipei City, Taiwan
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chih-Wei Chien
- Department of Obstetrics and Gynecology, New Taipei Municipal Tu Cheng Hospital, New Taipei City, Taiwan
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ting-Shu Wu
- Department of Infectious Control, Chang , Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yao-Lung Chang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Angel Chao
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
- Gynecologic Cancer Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Gigin Lin
- Clinical Metabolomics Core Lab, Chang , Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Medical Imaging and Intervention and Institute for Radiological Research, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Chih-Yung Chiu
- Clinical Metabolomics Core Lab, Chang , Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
- Division of Pediatric Pulmonology, Department of Pediatrics, Chang , Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
8
|
Elgedawy GA, Samir M, Elabd NS, Elsaid HH, Enar M, Salem RH, Montaser BA, AboShabaan HS, Seddik RM, El-Askaeri SM, Omar MM, Helal ML. Metabolic profiling during COVID-19 infection in humans: Identification of potential biomarkers for occurrence, severity and outcomes using machine learning. PLoS One 2024; 19:e0302977. [PMID: 38814977 PMCID: PMC11139268 DOI: 10.1371/journal.pone.0302977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/15/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND After its emergence in China, the coronavirus SARS-CoV-2 has swept the world, leading to global health crises with millions of deaths. COVID-19 clinical manifestations differ in severity, ranging from mild symptoms to severe disease. Although perturbation of metabolism has been reported as a part of the host response to COVID-19 infection, scarce data exist that describe stage-specific changes in host metabolites during the infection and how this could stratify patients based on severity. METHODS Given this knowledge gap, we performed targeted metabolomics profiling and then used machine learning models and biostatistics to characterize the alteration patterns of 50 metabolites and 17 blood parameters measured in a cohort of 295 human subjects. They were categorized into healthy controls, non-severe, severe and critical groups with their outcomes. Subject's demographic and clinical data were also used in the analyses to provide more robust predictive models. RESULTS The non-severe and severe COVID-19 patients experienced the strongest changes in metabolite repertoire, whereas less intense changes occur during the critical phase. Panels of 15, 14, 2 and 2 key metabolites were identified as predictors for non-severe, severe, critical and dead patients, respectively. Specifically, arginine and malonyl methylmalonyl succinylcarnitine were significant biomarkers for the onset of COVID-19 infection and tauroursodeoxycholic acid were potential biomarkers for disease progression. Measuring blood parameters enhanced the predictive power of metabolic signatures during critical illness. CONCLUSIONS Metabolomic signatures are distinctive for each stage of COVID-19 infection. This has great translation potential as it opens new therapeutic and diagnostic prospective based on key metabolites.
Collapse
Affiliation(s)
- Gamalat A. Elgedawy
- Department of Clinical Biochemistry and Molecular Diagnostics, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Mohamed Samir
- Faculty of Veterinary Medicine, Department of Zoonoses, Zagazig University, Zagazig, Egypt
| | - Naglaa S. Elabd
- Faculty of Medicine, Department of Tropical Medicine, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Hala H. Elsaid
- Department of Clinical Biochemistry and Molecular Diagnostics, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Mohamed Enar
- Al Mahala Elkobra Fever Hospital, Al Mahala Elkobra, Egypt
| | - Radwa H. Salem
- Department of Clinical Microbiology and Immunology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Belal A. Montaser
- Faculty of Medicine, Department of Clinical Pathology, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Hind S. AboShabaan
- Ph.D. of Biochemistry, National Liver Institute Hospital, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Randa M. Seddik
- Faculty of Medicine, Department of Tropical Medicine, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Shimaa M. El-Askaeri
- Department of Clinical Microbiology and Immunology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Marwa M. Omar
- Faculty of Medicine, Department of Clinical Pathology, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Marwa L. Helal
- Department of Clinical Biochemistry and Molecular Diagnostics, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| |
Collapse
|
9
|
Loveday EK, Welhaven H, Erdogan AE, Hain K, Chang CB, June RK, Taylor MP. Starve a cold or feed a fever? Identifying cellular metabolic changes following infection and exposure to SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595410. [PMID: 38826440 PMCID: PMC11142155 DOI: 10.1101/2024.05.22.595410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Viral infections induce major shifts in cellular metabolism elicited by active viral replication and antiviral responses. For the virus, harnessing cellular metabolism and evading changes that limit replication are essential for productive viral replication. In contrast, the cellular response to infection disrupts metabolic pathways to prevent viral replication and promote an antiviral state in the host cell and neighboring bystander cells. This competition between the virus and cell results in measurable shifts in cellular metabolism that differ depending on the virus, cell type, and extracellular environment. The resulting metabolic shifts can be observed and analyzed using global metabolic profiling techniques to identify pathways that are critical for either viral replication or cellular defense. SARS-CoV-2 is a respiratory virus that can exhibit broad tissue tropism and diverse, yet inconsistent, symptomatology. While the factors that determine the presentation and severity of SARS-CoV-2 infection remain unclear, metabolic syndromes are associated with more severe manifestations of SARS-CoV-2 disease. Despite these observations a critical knowledge gap remains between cellular metabolic responses and SARS-CoV-2 infection. Using a well-established untargeted metabolomics analysis workflow, we compared SARS-CoV-2 infection of human lung carcinoma cells. We identified significant changes in metabolic pathways that correlate with either productive or non-productive viral infection. This information is critical for characterizing the factors that contribute to SARS-CoV-2 replication that could be targeted for therapeutic interventions to limit viral disease.
Collapse
Affiliation(s)
- Emma Kate Loveday
- Center for Biofilm Engineering, Montana State University, Bozeman MT 59717
- Department of Chemical and Biological Engineering, Montana State University, Bozeman MT 59717
| | - Hope Welhaven
- Chemistry and Biochemistry, Montana State University, Bozeman MT 59717
| | - Ayten Ebru Erdogan
- Department of Chemical and Biological Engineering, Montana State University, Bozeman MT 59717
| | - Kyle Hain
- Microbiology and Cell Biology, Montana State University, Bozeman MT 59717
| | - Connie B. Chang
- Center for Biofilm Engineering, Montana State University, Bozeman MT 59717
- Department of Chemical and Biological Engineering, Montana State University, Bozeman MT 59717
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905
| | - Ronald K. June
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman MT 59717
| | - Matthew P. Taylor
- Microbiology and Cell Biology, Montana State University, Bozeman MT 59717
| |
Collapse
|
10
|
Lodge S, Litton E, Gray N, Ryan M, Millet O, Fear M, Raby E, Currie A, Wood F, Holmes E, Wist J, Nicholson JK. Stratification of Sepsis Patients on Admission into the Intensive Care Unit According to Differential Plasma Metabolic Phenotypes. J Proteome Res 2024; 23:1328-1340. [PMID: 38513133 PMCID: PMC11002934 DOI: 10.1021/acs.jproteome.3c00803] [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: 11/20/2023] [Revised: 02/15/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024]
Abstract
Delayed diagnosis of patients with sepsis or septic shock is associated with increased mortality and morbidity. UPLC-MS and NMR spectroscopy were used to measure panels of lipoproteins, lipids, biogenic amines, amino acids, and tryptophan pathway metabolites in blood plasma samples collected from 152 patients within 48 h of admission into the Intensive Care Unit (ICU) where 62 patients had no sepsis, 71 patients had sepsis, and 19 patients had septic shock. Patients with sepsis or septic shock had higher concentrations of neopterin and lower levels of HDL cholesterol and phospholipid particles in comparison to nonsepsis patients. Septic shock could be differentiated from sepsis patients based on different concentrations of 10 lipids, including significantly lower concentrations of five phosphatidylcholine species, three cholesterol esters, one dihydroceramide, and one phosphatidylethanolamine. The Supramolecular Phospholipid Composite (SPC) was reduced in all ICU patients, while the composite markers of acute phase glycoproteins were increased in the sepsis and septic shock patients within 48 h admission into ICU. We show that the plasma metabolic phenotype obtained within 48 h of ICU admission is diagnostic for the presence of sepsis and that septic shock can be differentiated from sepsis based on the lipid profile.
Collapse
Affiliation(s)
- Samantha Lodge
- Australian
National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Edward Litton
- Intensive
Care Unit, Fiona Stanley Hospital, Murdoch, WA 6150, Australia
- Intensive
Care Unit, St John of God Hospital, Subiaco, WA 6009, Australia
- School
of Medicine, University of Western Australia, Crawley, WA 6009, Australia
| | - Nicola Gray
- Australian
National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Monique Ryan
- Australian
National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Oscar Millet
- Precision
Medicine and Metabolism Laboratory, CIC
bioGUNE, Parque Tecnológico
de Bizkaia, Bld. 800, Derio 48160, Spain
| | - Mark Fear
- Burn
Injury Research Unit, School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
- Fiona
Wood Foundation, Perth, WA 6150, Australia
| | - Edward Raby
- Department
of Infectious Diseases, Fiona Stanley Hospital, Murdoch, WA 6150, Australia
| | - Andrew Currie
- School
of Medical, Molecular & Forensic Sciences, Murdoch University, Perth, WA 6150, Australia
- Centre
for Molecular Medicine & Innovative Therapeutics, Murdoch University, Perth, WA 6150, Australia
- Wesfarmers
Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia
| | - Fiona Wood
- Burn
Injury Research Unit, School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
- Fiona
Wood Foundation, Perth, WA 6150, Australia
- Burns
service of Western Australia, WA Department
of Health, Murdoch, WA 6150, Australia
| | - Elaine Holmes
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Institute
of Global Health Innovation, Faculty of Medicine, Imperial College London, Level 1, Faculty Building, South Kensington Campus, London SW7 2NA, U.K.
| | - Julien Wist
- Australian
National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Chemistry
Department, Universidad del Valle, Cali 76001, Colombia
- Department of Metabolism, Digestion and
Reproduction, Faculty of Medicine, Imperial
College London, Sir Alexander
Fleming Building, South Kensington, London SW7 2AZ, U.K.
| | - Jeremy K. Nicholson
- Australian
National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
- Department of Metabolism, Digestion and
Reproduction, Faculty of Medicine, Imperial
College London, Sir Alexander
Fleming Building, South Kensington, London SW7 2AZ, U.K.
| |
Collapse
|
11
|
Lonati C, Berezhnoy G, Lawler N, Masuda R, Kulkarni A, Sala S, Nitschke P, Zizmare L, Bucci D, Cannet C, Schäfer H, Singh Y, Gray N, Lodge S, Nicholson J, Merle U, Wist J, Trautwein C. Urinary phenotyping of SARS-CoV-2 infection connects clinical diagnostics with metabolomics and uncovers impaired NAD + pathway and SIRT1 activation. Clin Chem Lab Med 2024; 62:770-788. [PMID: 37955280 DOI: 10.1515/cclm-2023-1017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/22/2023] [Indexed: 11/14/2023]
Abstract
OBJECTIVES The stratification of individuals suffering from acute and post-acute SARS-CoV-2 infection remains a critical challenge. Notably, biomarkers able to specifically monitor viral progression, providing details about patient clinical status, are still not available. Herein, quantitative metabolomics is progressively recognized as a useful tool to describe the consequences of virus-host interactions considering also clinical metadata. METHODS The present study characterized the urinary metabolic profile of 243 infected individuals by quantitative nuclear magnetic resonance (NMR) spectroscopy and liquid chromatography mass spectrometry (LC-MS). Results were compared with a historical cohort of noninfected subjects. Moreover, we assessed the concentration of recently identified antiviral nucleosides and their association with other metabolites and clinical data. RESULTS Urinary metabolomics can stratify patients into classes of disease severity, with a discrimination ability comparable to that of clinical biomarkers. Kynurenines showed the highest fold change in clinically-deteriorated patients and higher-risk subjects. Unique metabolite clusters were also generated based on age, sex, and body mass index (BMI). Changes in the concentration of antiviral nucleosides were associated with either other metabolites or clinical variables. Increased kynurenines and reduced trigonelline excretion indicated a disrupted nicotinamide adenine nucleotide (NAD+) and sirtuin 1 (SIRT1) pathway. CONCLUSIONS Our results confirm the potential of urinary metabolomics for noninvasive diagnostic/prognostic screening and show that the antiviral nucleosides could represent novel biomarkers linking viral load, immune response, and metabolism. Moreover, we established for the first time a casual link between kynurenine accumulation and deranged NAD+/SIRT1, offering a novel mechanism through which SARS-CoV-2 manipulates host physiology.
Collapse
Affiliation(s)
- Caterina Lonati
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany
| | - Georgy Berezhnoy
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany
| | - Nathan Lawler
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Reika Masuda
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Aditi Kulkarni
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany
| | - Samuele Sala
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Philipp Nitschke
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Laimdota Zizmare
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany
| | - Daniele Bucci
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany
| | - Claire Cannet
- Bruker BioSpin GmbH, AIC Division, Ettlingen, Germany
| | | | - Yogesh Singh
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Nicola Gray
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Samantha Lodge
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Jeremy Nicholson
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Uta Merle
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Julien Wist
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Christoph Trautwein
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
12
|
Roberts J, Whiley L, Gray N, Gay M, Nitschke P, Masuda R, Holmes E, Nicholson JK, Wist J, Lawler NG. Rapid and Self-Administrable Capillary Blood Microsampling Demonstrates Statistical Equivalence with Standard Venous Collections in NMR-Based Lipoprotein Analysis. Anal Chem 2024; 96:4505-4513. [PMID: 38372289 PMCID: PMC10955515 DOI: 10.1021/acs.analchem.3c05152] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/17/2024] [Accepted: 01/29/2024] [Indexed: 02/20/2024]
Abstract
We investigated plasma and serum blood derivatives from capillary blood microsamples (500 μL, MiniCollect tubes) and corresponding venous blood (10 mL vacutainers). Samples from 20 healthy participants were analyzed by 1H NMR, and 112 lipoprotein subfraction parameters; 3 supramolecular phospholipid composite (SPC) parameters from SPC1, SPC2, and SPC3 subfractions; 2 N-acetyl signals from α-1-acid glycoprotein (Glyc), GlycA, and GlycB; and 3 calculated parameters, SPC (total), SPC3/SPC2, and Glyc (total) were assessed. Using linear regression between capillary and venous collection sites, we explained that agreement (Adj. R2 ≥ 0.8, p < 0.001) was witnessed for 86% of plasma parameters (103/120) and 88% of serum parameters (106/120), indicating that capillary lipoprotein, SPC, and Glyc concentrations follow changes in venous concentrations. These results indicate that capillary blood microsamples are suitable for sampling in remote areas and for high-frequency longitudinal sampling of the majority of lipoproteins, SPCs, and Glycs.
Collapse
Affiliation(s)
- Jayden
Lee Roberts
- Australian
National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute,
Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
| | - Luke Whiley
- Australian
National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute,
Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
| | - Nicola Gray
- Australian
National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute,
Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
| | - Melvin Gay
- Bruker
Pty Ltd., Preston, VIC 3072, Australia
| | - Philipp Nitschke
- Australian
National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute,
Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
| | - Reika Masuda
- Australian
National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute,
Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
| | - Elaine Holmes
- Australian
National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute,
Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Department
of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, U.K.
| | - Jeremy K. Nicholson
- Australian
National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute,
Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Department
of Cardiology, Fiona Stanley Hospital, Medical School, University of Western Australia, Murdoch, WA 6150, Australia
- Institute
of Global Health Innovation, Faculty of Medicine, Imperial College London, Level 1, Faculty Building, South Kensington, London SW7 2NA, U.K.
| | - Julien Wist
- Australian
National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute,
Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Faculty
of Medicine, Department of Metabolism, Digestion and Reproduction,
Division of Digestive Diseases, Imperial
College, London SW7 2AZ, United Kingdom
- Chemistry
Department, Universidad del Valle, Melendez 76001, Cali, Colombia
| | - Nathan G. Lawler
- Australian
National Phenome Centre, Health Futures Institute, Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute,
Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Murdoch, WA 6150, Australia
| |
Collapse
|
13
|
Sala S, Nitschke P, Masuda R, Gray N, Lawler NG, Wood JM, Buckler JN, Berezhnoy G, Bolaños J, Boughton BA, Lonati C, Rössler T, Singh Y, Wilson ID, Lodge S, Morillon AC, Loo RL, Hall D, Whiley L, Evans GB, Grove TL, Almo SC, Harris LD, Holmes E, Merle U, Trautwein C, Nicholson JK, Wist J. Integrative Molecular Structure Elucidation and Construction of an Extended Metabolic Pathway Associated with an Ancient Innate Immune Response in COVID-19 Patients. J Proteome Res 2024; 23:956-970. [PMID: 38310443 PMCID: PMC10913068 DOI: 10.1021/acs.jproteome.3c00654] [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: 10/06/2023] [Revised: 12/01/2023] [Accepted: 12/29/2023] [Indexed: 02/05/2024]
Abstract
We present compelling evidence for the existence of an extended innate viperin-dependent pathway, which provides crucial evidence for an adaptive response to viral agents, such as SARS-CoV-2. We show the in vivo biosynthesis of a family of novel endogenous cytosine metabolites with potential antiviral activities. Two-dimensional nuclear magnetic resonance (NMR) spectroscopy revealed a characteristic spin-system motif, indicating the presence of an extended panel of urinary metabolites during the acute viral replication phase. Mass spectrometry additionally enabled the characterization and quantification of the most abundant serum metabolites, showing the potential diagnostic value of the compounds for viral infections. In total, we unveiled ten nucleoside (cytosine- and uracil-based) analogue structures, eight of which were previously unknown in humans allowing us to propose a new extended viperin pathway for the innate production of antiviral compounds. The molecular structures of the nucleoside analogues and their correlation with an array of serum cytokines, including IFN-α2, IFN-γ, and IL-10, suggest an association with the viperin enzyme contributing to an ancient endogenous innate immune defense mechanism against viral infection.
Collapse
Affiliation(s)
- Samuele Sala
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Philipp Nitschke
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Reika Masuda
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Nicola Gray
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Nathan G. Lawler
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - James M. Wood
- Ferrier
Research Institute, Victoria University
of Wellington, Wellington 6012, New Zealand
- The
Maurice Wilkins Centre for Molecular Biodiscovef Wellington, Welry, The University of Auckland, Auckland 1010, New Zealand
| | - Joshua N. Buckler
- Ferrier
Research Institute, Victoria University
of Wellington, Wellington 6012, New Zealand
| | - Georgy Berezhnoy
- Department
of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, 72074 Tübingen, Germany
| | - Jose Bolaños
- Chemistry
Department, Universidad del Valle, Cali 76001, Colombia
| | - Berin A. Boughton
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Caterina Lonati
- Center
for Preclinical Research, Fondazione IRCCS
Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Titus Rössler
- Department
of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, 72074 Tübingen, Germany
| | - Yogesh Singh
- Institute
of Medical Genetics and Applied Genomics, University Hospital Tübingen, 72074 Tübingen, Germany
| | - Ian D. Wilson
- Division
of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College, Burlington Danes Building, Du Cane Road, London W12 0NN, U.K.
| | - Samantha Lodge
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Aude-Claire Morillon
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Ruey Leng Loo
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Drew Hall
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Luke Whiley
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Gary B. Evans
- Ferrier
Research Institute, Victoria University
of Wellington, Wellington 6012, New Zealand
- The
Maurice Wilkins Centre for Molecular Biodiscovef Wellington, Welry, The University of Auckland, Auckland 1010, New Zealand
| | - Tyler L. Grove
- Department
of Biochemistry, Albert Einstein College
of Medicine, Bronx, New York 10461, United States
| | - Steven C. Almo
- Department
of Biochemistry, Albert Einstein College
of Medicine, Bronx, New York 10461, United States
| | - Lawrence D. Harris
- Ferrier
Research Institute, Victoria University
of Wellington, Wellington 6012, New Zealand
- The
Maurice Wilkins Centre for Molecular Biodiscovef Wellington, Welry, The University of Auckland, Auckland 1010, New Zealand
| | - Elaine Holmes
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
- Division
of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College, Burlington Danes Building, Du Cane Road, London W12 0NN, U.K.
| | - Uta Merle
- Department
of Internal Medicine IV, University Hospital
Heidelberg, 69120 Heidelberg, Germany
| | - Christoph Trautwein
- Department
of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, 72074 Tübingen, Germany
| | - Jeremy K. Nicholson
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
- Institute
of Global Health Innovation, Faculty of
Medicine, Imperial College London, Level 1, Faculty Building, South Kensington Campus, London SW7 2NA, U.K.
| | - Julien Wist
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
- Chemistry
Department, Universidad del Valle, Cali 76001, Colombia
- Faculty of Medicine, Department of Metabolism,
Digestion and Reproduction,
Division of Digestive Diseases at Imperial College, London SW7 2AZ, U.K.
| |
Collapse
|
14
|
Ahmad MS, Minaee N, Serrano-Contreras JI, Kaluarachchi M, Shen EYL, Boulange C, Ahmad S, Phetcharaburanin J, Holmes E, Wist J, Albaloshi AH, Alaama T, Damanhouri ZA, Lodge S. Exploring the Interactions between Obesity and Diabetes: Implications for Understanding Metabolic Dysregulation in a Saudi Arabian Adult Population. J Proteome Res 2024; 23:809-821. [PMID: 38230637 PMCID: PMC10846529 DOI: 10.1021/acs.jproteome.3c00717] [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: 10/31/2023] [Revised: 12/07/2023] [Accepted: 12/14/2023] [Indexed: 01/18/2024]
Abstract
The rising prevalence of obesity in Saudi Arabia is a major contributor to the nation's high levels of cardiometabolic diseases such as type 2 diabetes. To assess the impact of obesity on the diabetic metabolic phenotype presented in young Saudi Arabian adults, participants (n = 289, aged 18-40 years) were recruited and stratified into four groups: healthy weight (BMI 18.5-24.99 kg/m2) with (n = 57) and without diabetes (n = 58) or overweight/obese (BMI > 24.99 kg/m2) with (n = 102) and without diabetes (n = 72). Distinct plasma metabolic phenotypes associated with high BMI and diabetes were identified using nuclear magnetic resonance spectroscopy and ultraperformance liquid chromatography mass spectrometry. Increased plasma glucose and dysregulated lipoproteins were characteristics of obesity in individuals with and without diabetes, but the obesity-associated lipoprotein phenotype was partially masked in individuals with diabetes. Although there was little difference between diabetics and nondiabetics in the global plasma LDL cholesterol and phospholipid concentration, the distribution of lipoprotein particles was altered in diabetics with a shift toward denser and more atherogenic LDL5 and LDL6 particles, which was amplified in the presence of obesity. Further investigation is warranted in larger Middle Eastern populations to explore the dysregulation of metabolism driven by interactions between obesity and diabetes in young adults.
Collapse
Affiliation(s)
- Muhammad Saeed Ahmad
- Department
of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, U.K.
- Drug
Metabolism Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Novia Minaee
- Health
Futures Institute, Murdoch University, Perth, WA 6150, Australia
| | | | - Manuja Kaluarachchi
- Department
of Metabolism, Digestion and Reproduction, Imperial College, London SW7 2AZ, U.K.
| | - Eric Yi-Liang Shen
- Department
of Metabolism, Digestion and Reproduction, Imperial College, London SW7 2AZ, U.K.
- Department
of Radiation Oncology, Chang Gung Memorial
Hospital and Chang Gung University, Taoyuan 333, Taiwan
| | - Claire Boulange
- Department
of Metabolism, Digestion and Reproduction, Imperial College, London SW7 2AZ, U.K.
| | - Sultan Ahmad
- Drug
Metabolism Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Jutarop Phetcharaburanin
- Department
of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Elaine Holmes
- Health
Futures Institute, Murdoch University, Perth, WA 6150, Australia
- Department
of Metabolism, Digestion and Reproduction, Imperial College, London SW7 2AZ, U.K.
| | - Julien Wist
- Health
Futures Institute, Murdoch University, Perth, WA 6150, Australia
- Department
of Metabolism, Digestion and Reproduction, Imperial College, London SW7 2AZ, U.K.
- Chemistry
Department, Universidad del Valle, Cali 76001, Colombia
| | - Ahmed Hakem Albaloshi
- King
Abdulaziz Hospital and Endocrine and Diabetic Center, Jeddah 23436, Saudi Arabia
| | - Tareef Alaama
- Department
of Medicine, Faculty of Medicine, King Abdulaziz
University, Jeddah 21589, Saudi Arabia
| | - Zoheir Abdullah Damanhouri
- Drug
Metabolism Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department
of Pharmacology, Faculty of Medicine, King
Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Samantha Lodge
- Health
Futures Institute, Murdoch University, Perth, WA 6150, Australia
| |
Collapse
|
15
|
Ahmad AF, Caparrós-Martin JA, Gray N, Lodge S, Wist J, Lee S, O'Gara F, Dwivedi G, Ward NC. Gut microbiota and metabolomics profiles in patients with chronic stable angina and acute coronary syndrome. Physiol Genomics 2024; 56:48-64. [PMID: 37811721 DOI: 10.1152/physiolgenomics.00072.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/05/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. The gut microbiota and its associated metabolites may be involved in the development and progression of CVD, although the mechanisms and impact on clinical outcomes are not fully understood. This study investigated the gut microbiome profile and associated metabolites in patients with chronic stable angina (CSA) and acute coronary syndrome (ACS) compared with healthy controls. Bacterial alpha diversity in stool from patients with ACS or CSA was comparable to healthy controls at both baseline and follow-up visits. Differential abundance analysis identified operational taxonomic units (OTUs) assigned to commensal taxa differentiating patients with ACS from healthy controls at both baseline and follow-up. Patients with CSA and ACS had significantly higher levels of trimethylamine N-oxide compared with healthy controls (CSA: 0.032 ± 0.023 mmol/L, P < 0.01 vs. healthy, and ACS: 0.032 ± 0.023 mmol/L, P = 0.02 vs. healthy, respectively). Patients with ACS had reduced levels of propionate and butyrate (119 ± 4 vs. 139 ± 5.1 µM, P = 0.001, and 14 ± 4.3 vs. 23.5 ± 8.1 µM, P < 0.001, respectively), as well as elevated serum sCD14 (2245 ± 75.1 vs. 1834 ± 45.8 ng/mL, P < 0.0001) and sCD163 levels (457.3 ± 31.8 vs. 326.8 ± 20.7 ng/mL, P = 0.001), compared with healthy controls at baseline. Furthermore, a modified small molecule metabolomic and lipidomic signature was observed in patients with CSA and ACS compared with healthy controls. These findings provide evidence of a link between gut microbiome composition and gut bacterial metabolites with CVD. Future time course studies in patients to observe temporal changes and subsequent associations with gut microbiome composition are required to provide insight into how these are affected by transient changes following an acute coronary event.NEW & NOTEWORTHY The study found discriminative microorganisms differentiating patients with acute coronary syndrome (ACS) from healthy controls. In addition, reduced levels of certain bacterial metabolites and elevated sCD14 and sCD163 were observed in patients with ACS compared with healthy controls. Furthermore, modified small molecule metabolomic and lipidomic signatures were found in both patient groups. Although it is not known whether these differences in profiles are associated with disease development and/or progression, the findings provide exciting options for potential new disease-related mechanism(s) and associated therapeutic target(s).
Collapse
Affiliation(s)
- Adilah F Ahmad
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
| | - Jose A Caparrós-Martin
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia, Australia
| | - Nicola Gray
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Samantha Lodge
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Julien Wist
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Silvia Lee
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
- Department of Microbiology, PathWest Laboratory Medicine, Perth, Western Australia, Australia
| | - Fergal O'Gara
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia, Australia
- BIOMERIT Research Centre, School of Microbiology, University College Cork, Cork, Ireland
| | - Girish Dwivedi
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
- Department of Cardiology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Natalie C Ward
- Dobney Hypertension Centre, Medical School, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
16
|
Ghini V, Meoni G, Vignoli A, Di Cesare F, Tenori L, Turano P, Luchinat C. Fingerprinting and profiling in metabolomics of biosamples. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2023; 138-139:105-135. [PMID: 38065666 DOI: 10.1016/j.pnmrs.2023.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/13/2023] [Accepted: 10/15/2023] [Indexed: 12/18/2023]
Abstract
This review focuses on metabolomics from an NMR point of view. It attempts to cover the broad scope of metabolomics and describes the NMR experiments that are most suitable for each sample type. It is addressed not only to NMR specialists, but to all researchers who wish to approach metabolomics with a clear idea of what they wish to achieve but not necessarily with a deep knowledge of NMR. For this reason, some technical parts may seem a bit naïve to the experts. The review starts by describing standard metabolomics procedures, which imply the use of a dedicated 600 MHz instrument and of four properly standardized 1D experiments. Standardization is a must if one wants to directly compare NMR results obtained in different labs. A brief mention is also made of standardized pre-analytical procedures, which are even more essential. Attention is paid to the distinction between fingerprinting and profiling, and the advantages and disadvantages of fingerprinting are clarified. This aspect is often not fully appreciated. Then profiling, and the associated problems of signal assignment and quantitation, are discussed. We also describe less conventional approaches, such as the use of different magnetic fields, the use of signal enhancement techniques to increase sensitivity, and the potential of field-shuttling NMR. A few examples of biomedical applications are also given, again with the focus on NMR techniques that are most suitable to achieve each particular goal, including a description of the most common heteronuclear experiments. Finally, the growing applications of metabolomics to foodstuffs are described.
Collapse
Affiliation(s)
- Veronica Ghini
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy; Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Gaia Meoni
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy; Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Alessia Vignoli
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy; Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Francesca Di Cesare
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy; Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Leonardo Tenori
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy; Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy; Consorzio Interuniversitario Risonanze Magnetiche Metallo Proteine (CIRMMP), Sesto Fiorentino, Italy
| | - Paola Turano
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy; Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy; Consorzio Interuniversitario Risonanze Magnetiche Metallo Proteine (CIRMMP), Sesto Fiorentino, Italy.
| | - Claudio Luchinat
- Consorzio Interuniversitario Risonanze Magnetiche Metallo Proteine (CIRMMP), Sesto Fiorentino, Italy; Giotto Biotech S.r.l., Sesto Fiorentino, Italy.
| |
Collapse
|
17
|
Ghini V, Vieri W, Celli T, Pecchioli V, Boccia N, Alonso-Vásquez T, Pelagatti L, Fondi M, Luchinat C, Bertini L, Vannucchi V, Landini G, Turano P. COVID-19: A complex disease with a unique metabolic signature. PLoS Pathog 2023; 19:e1011787. [PMID: 37943960 PMCID: PMC10662774 DOI: 10.1371/journal.ppat.1011787] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 11/21/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
Plasma of COVID-19 patients contains a strong metabolomic/lipoproteomic signature, revealed by the NMR analysis of a cohort of >500 patients sampled during various waves of COVID-19 infection, corresponding to the spread of different variants, and having different vaccination status. This composite signature highlights common traits of the SARS-CoV-2 infection. The most dysregulated molecules display concentration trends that scale with disease severity and might serve as prognostic markers for fatal events. Metabolomics evidence is then used as input data for a sex-specific multi-organ metabolic model. This reconstruction provides a comprehensive view of the impact of COVID-19 on the entire human metabolism. The human (male and female) metabolic network is strongly impacted by the disease to an extent dictated by its severity. A marked metabolic reprogramming at the level of many organs indicates an increase in the generic energetic demand of the organism following infection. Sex-specific modulation of immune response is also suggested.
Collapse
Affiliation(s)
- Veronica Ghini
- Department of Chemistry “Ugo Schiff”, University of Florence, Sesto Fiorentino Florence, Italy
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Florence, Italy
| | - Walter Vieri
- Department of Chemistry “Ugo Schiff”, University of Florence, Sesto Fiorentino Florence, Italy
- Department of Biology, University of Florence, Sesto Fiorentino, Florence, Italy
| | - Tommaso Celli
- Internal Medicine, Santa Maria Nuova Hospital, Florence, Florence, Italy
| | - Valentina Pecchioli
- Department of Chemistry “Ugo Schiff”, University of Florence, Sesto Fiorentino Florence, Italy
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Florence, Italy
| | - Nunzia Boccia
- Internal Medicine, Santa Maria Nuova Hospital, Florence, Florence, Italy
| | - Tania Alonso-Vásquez
- Department of Biology, University of Florence, Sesto Fiorentino, Florence, Italy
| | - Lorenzo Pelagatti
- Internal Medicine, Santa Maria Nuova Hospital, Florence, Florence, Italy
| | - Marco Fondi
- Department of Biology, University of Florence, Sesto Fiorentino, Florence, Italy
| | - Claudio Luchinat
- Department of Chemistry “Ugo Schiff”, University of Florence, Sesto Fiorentino Florence, Italy
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Florence, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), Sesto Fiorentino Florence, Italy
| | - Laura Bertini
- Internal Medicine, Santa Maria Nuova Hospital, Florence, Florence, Italy
| | - Vieri Vannucchi
- Internal Medicine, Santa Maria Nuova Hospital, Florence, Florence, Italy
| | - Giancarlo Landini
- Internal Medicine, Santa Maria Nuova Hospital, Florence, Florence, Italy
| | - Paola Turano
- Department of Chemistry “Ugo Schiff”, University of Florence, Sesto Fiorentino Florence, Italy
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Florence, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), Sesto Fiorentino Florence, Italy
| |
Collapse
|
18
|
Kazenwadel J, Berezhnoy G, Cannet C, Schäfer H, Geisler T, Rohlfing AK, Gawaz M, Merle U, Trautwein C. Stratification of hypertension and SARS-CoV-2 infection by quantitative NMR spectroscopy of human blood serum. COMMUNICATIONS MEDICINE 2023; 3:145. [PMID: 37845506 PMCID: PMC11081957 DOI: 10.1038/s43856-023-00365-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 09/12/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Diagnostic approaches like the nuclear magnetic resonance spectroscopy (NMR) based quantification of metabolites, lipoproteins, and inflammation markers has helped to identify typical alterations in the blood serum of COVID-19 patients. However, confounders such as sex, and comorbidities, which strongly influence the metabolome, were often not considered. Therefore, the aim of this NMR study was to consider sex, as well as arterial hypertension (AHT), when investigating COVID-19-positive serum samples in a large age-and sex matched cohort. METHODS NMR serum data from 329 COVID-19 patients were compared with 305 healthy controls. 134 COVID-19 patients were affected by AHT. These were analyzed together with NMR data from 58 hypertensives without COVID-19. In addition to metabolite, lipoprotein, and glycoprotein data from NMR, common laboratory parameters were considered. Sex was considered in detail for all comparisons. RESULTS Here, we show that several differences emerge from previous NMR COVID-19 studies when AHT is considered. Especially, the previously described triglyceride-rich lipoprotein profile is no longer observed in COVID-19 patients, nor an increase in ketone bodies. Further alterations are a decrease in glutamine, leucine, isoleucine, and lysine, citric acid, HDL-4 particles, and total cholesterol. Additionally, hypertensive COVID-19 patients show higher inflammatory NMR parameters than normotensive patients. CONCLUSIONS We present a more precise picture of COVID-19 blood serum parameters. Accordingly, considering sex and comorbidities should be included in future metabolomics studies for improved and refined patient stratification. Due to metabolic similarities with other viral infections, these results can be applied to other respiratory diseases in the future.
Collapse
Affiliation(s)
- Jasmin Kazenwadel
- Werner Siemens Imaging Center, Department for Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Georgy Berezhnoy
- Werner Siemens Imaging Center, Department for Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Claire Cannet
- Bruker BioSpin GmbH, Applied Industrial and Clinical Division, Ettlingen, Germany
| | - Hartmut Schäfer
- Bruker BioSpin GmbH, Applied Industrial and Clinical Division, Ettlingen, Germany
| | - Tobias Geisler
- Department of Internal Medicine III, Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Anne-Katrin Rohlfing
- Department of Internal Medicine III, Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Internal Medicine III, Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Uta Merle
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Christoph Trautwein
- Werner Siemens Imaging Center, Department for Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany.
| |
Collapse
|
19
|
Gama-Almeida MC, Pinto GDA, Teixeira L, Hottz ED, Ivens P, Ribeiro H, Garrett R, Torres AG, Carneiro TIA, Barbalho BDO, Ludwig C, Struchiner CJ, Assunção-Miranda I, Valente APC, Bozza FA, Bozza PT, Dos Santos GC, El-Bacha T. Integrated NMR and MS Analysis of the Plasma Metabolome Reveals Major Changes in One-Carbon, Lipid, and Amino Acid Metabolism in Severe and Fatal Cases of COVID-19. Metabolites 2023; 13:879. [PMID: 37512587 PMCID: PMC10384698 DOI: 10.3390/metabo13070879] [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: 06/12/2023] [Revised: 07/15/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Brazil has the second-highest COVID-19 death rate worldwide, and Rio de Janeiro is among the states with the highest rate in the country. Although vaccine coverage has been achieved, it is anticipated that COVID-19 will transition into an endemic disease. It is concerning that the molecular mechanisms underlying clinical evolution from mild to severe disease, as well as the mechanisms leading to long COVID-19, are not yet fully understood. NMR and MS-based metabolomics were used to identify metabolites associated with COVID-19 pathophysiology and disease outcome. Severe COVID-19 cases (n = 35) were enrolled in two reference centers in Rio de Janeiro within 72 h of ICU admission, alongside 12 non-infected control subjects. COVID-19 patients were grouped into survivors (n = 18) and non-survivors (n = 17). Choline-related metabolites, serine, glycine, and betaine, were reduced in severe COVID-19, indicating dysregulation in methyl donors. Non-survivors had higher levels of creatine/creatinine, 4-hydroxyproline, gluconic acid, and N-acetylserine, indicating liver and kidney dysfunction. Several changes were greater in women; thus, patients' sex should be considered in pandemic surveillance to achieve better disease stratification and improve outcomes. These metabolic alterations may be useful to monitor organ (dys) function and to understand the pathophysiology of acute and possibly post-acute COVID-19 syndromes.
Collapse
Affiliation(s)
- Marcos C Gama-Almeida
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Gabriela D A Pinto
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Lívia Teixeira
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-361, Brazil
| | - Eugenio D Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Federal University of Juiz de Fora, Juiz de Fora 36936-900, Brazil
| | - Paula Ivens
- LabMeta, Metabolomics Laboratory, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| | - Hygor Ribeiro
- LabMeta, Metabolomics Laboratory, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
- Lipid Biochemistry and Lipidomics Laboratory, Department of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| | - Rafael Garrett
- LabMeta, Metabolomics Laboratory, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| | - Alexandre G Torres
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Lipid Biochemistry and Lipidomics Laboratory, Department of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| | - Talita I A Carneiro
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Bianca de O Barbalho
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Christian Ludwig
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2SQ, UK
| | - Claudio J Struchiner
- School of Applied Mathematics, Fundação Getúlio Vargas, Rio de Janeiro 22231-080, Brazil
- Institute of Social Medicine, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20550-013, Brazil
| | - Iranaia Assunção-Miranda
- LaRIV, Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Ana Paula C Valente
- National Center for Nuclear Magnetic Resonance-Jiri Jonas, Institute of Medical Biochemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Fernando A Bozza
- National Institute of Infectious Disease Evandro Chagas, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- D'Or Institute for Research and Education, Rio de Janeiro 22281-100, Brazil
| | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-361, Brazil
| | - Gilson C Dos Santos
- LabMet-Laboratory of Metabolomics, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Department of Genetics, State University of Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Tatiana El-Bacha
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Lipid Biochemistry and Lipidomics Laboratory, Department of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| |
Collapse
|
20
|
Lodge S, Lawler NG, Gray N, Masuda R, Nitschke P, Whiley L, Bong SH, Yeap BB, Dwivedi G, Spraul M, Schaefer H, Gil-Redondo R, Embade N, Millet O, Holmes E, Wist J, Nicholson JK. Integrative Plasma Metabolic and Lipidomic Modelling of SARS-CoV-2 Infection in Relation to Clinical Severity and Early Mortality Prediction. Int J Mol Sci 2023; 24:11614. [PMID: 37511373 PMCID: PMC10380980 DOI: 10.3390/ijms241411614] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/10/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
An integrative multi-modal metabolic phenotyping model was developed to assess the systemic plasma sequelae of SARS-CoV-2 (rRT-PCR positive) induced COVID-19 disease in patients with different respiratory severity levels. Plasma samples from 306 unvaccinated COVID-19 patients were collected in 2020 and classified into four levels of severity ranging from mild symptoms to severe ventilated cases. These samples were investigated using a combination of quantitative Nuclear Magnetic Resonance (NMR) spectroscopy and Mass Spectrometry (MS) platforms to give broad lipoprotein, lipidomic and amino acid, tryptophan-kynurenine pathway, and biogenic amine pathway coverage. All platforms revealed highly significant differences in metabolite patterns between patients and controls (n = 89) that had been collected prior to the COVID-19 pandemic. The total number of significant metabolites increased with severity with 344 out of the 1034 quantitative variables being common to all severity classes. Metabolic signatures showed a continuum of changes across the respiratory severity levels with the most significant and extensive changes being in the most severely affected patients. Even mildly affected respiratory patients showed multiple highly significant abnormal biochemical signatures reflecting serious metabolic deficiencies of the type observed in Post-acute COVID-19 syndrome patients. The most severe respiratory patients had a high mortality (56.1%) and we found that we could predict mortality in this patient sub-group with high accuracy in some cases up to 61 days prior to death, based on a separate metabolic model, which highlighted a different set of metabolites to those defining the basic disease. Specifically, hexosylceramides (HCER 16:0, HCER 20:0, HCER 24:1, HCER 26:0, HCER 26:1) were markedly elevated in the non-surviving patient group (Cliff's delta 0.91-0.95) and two phosphoethanolamines (PE.O 18:0/18:1, Cliff's delta = -0.98 and PE.P 16:0/18:1, Cliff's delta = -0.93) were markedly lower in the non-survivors. These results indicate that patient morbidity to mortality trajectories is determined relatively soon after infection, opening the opportunity to select more intensive therapeutic interventions to these "high risk" patients in the early disease stages.
Collapse
Affiliation(s)
- Samantha Lodge
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Nathan G. Lawler
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Nicola Gray
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Reika Masuda
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
| | - Philipp Nitschke
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
| | - Luke Whiley
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Sze-How Bong
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
| | - Bu B. Yeap
- Medical School, University of Western Australia, Perth, WA 6150, Australia; (B.B.Y.); (G.D.)
- Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, WA 6150, Australia
| | - Girish Dwivedi
- Medical School, University of Western Australia, Perth, WA 6150, Australia; (B.B.Y.); (G.D.)
- Department of Cardiology, Fiona Stanley Hospital, Perth, WA 6150, Australia
| | | | | | - Rubén Gil-Redondo
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160 Derio, Spain; (R.G.-R.); (N.E.); (O.M.)
| | - Nieves Embade
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160 Derio, Spain; (R.G.-R.); (N.E.); (O.M.)
| | - Oscar Millet
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160 Derio, Spain; (R.G.-R.); (N.E.); (O.M.)
| | - Elaine Holmes
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, UK
| | - Julien Wist
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Chemistry Department, Universidad del Valle, Cali 76001, Colombia
| | - Jeremy K. Nicholson
- Australian National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia; (S.L.); (N.G.L.); (N.G.); (R.M.); (P.N.); (L.W.); (S.-H.B.); (E.H.)
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Institute of Global Health Innovation, Faculty of Medicine, Imperial College London, Faculty Building, South Kensington Campus, London SW7 2NA, UK
| |
Collapse
|
21
|
Petersen E, Chudakova D, Erdyneeva D, Zorigt D, Shabalina E, Gudkov D, Karalkin P, Reshetov I, Mynbaev OA. COVID-19-The Shift of Homeostasis into Oncopathology or Chronic Fibrosis in Terms of Female Reproductive System Involvement. Int J Mol Sci 2023; 24:ijms24108579. [PMID: 37239926 DOI: 10.3390/ijms24108579] [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: 01/17/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
The COVID-19 pandemic caused by the SARS-CoV-2 coronavirus remains a global public health concern due to the systemic nature of the infection and its long-term consequences, many of which remain to be elucidated. SARS-CoV-2 targets endothelial cells and blood vessels, altering the tissue microenvironment, its secretion, immune-cell subpopulations, the extracellular matrix, and the molecular composition and mechanical properties. The female reproductive system has high regenerative potential, but can accumulate damage, including due to SARS-CoV-2. COVID-19 is profibrotic and can change the tissue microenvironment toward an oncogenic niche. This makes COVID-19 and its consequences one of the potential regulators of a homeostasis shift toward oncopathology and fibrosis in the tissues of the female reproductive system. We are looking at SARS-CoV-2-induced changes at all levels in the female reproductive system.
Collapse
Affiliation(s)
- Elena Petersen
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Daria Chudakova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Daiana Erdyneeva
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Dulamsuren Zorigt
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | | | - Denis Gudkov
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Pavel Karalkin
- P.A. Herzen Moscow Research Institute of Oncology, 125284 Moscow, Russia
- Institute of Cluster Oncology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Igor Reshetov
- Institute of Cluster Oncology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Ospan A Mynbaev
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| |
Collapse
|
22
|
de Souza Nogueira J, Santos-Rebouças CB, Piergiorge RM, Valente AP, Gama-Almeida MC, El-Bacha T, Lopes Moreira ML, Baptista Marques BS, de Siqueira JR, de Carvalho EM, da Costa Ferreira O, Porto LC, Kelly da Silva Fidalgo T, Costa Dos Santos G. Metabolic Adaptations Correlated with Antibody Response after Immunization with Inactivated SARS-CoV-2 in Brazilian Subjects. J Proteome Res 2023. [PMID: 37167433 DOI: 10.1021/acs.jproteome.3c00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
The adsorbed vaccine SARS-CoV-2 (inactivated) produced by Sinovac (SV) was the first vaccine against COVID-19 to be used in Brazil. To understand the metabolic effects of SV in Brazilian subjects, NMR-based metabolomics was used, and the immune response was studied in Brazilian subjects. Forty adults without (group-, n = 23) and with previous COVID-19 infection (group+, n = 17) were followed-up for 90 days postcompletion of the vaccine regimen. After 90 days, our results showed that subjects had increased levels of lipoproteins, lipids, and N-acetylation of glycoproteins (NAG) as well as decreased levels of amino acids, lactate, citrate, and 3-hydroxypropionate. NAG and threonine were the highest correlated metabolites with N and S proteins, and neutralizing Ab levels. This study sheds light on the immunometabolism associated with the use of SV in Brazilian subjects from Rio de Janeiro and identifies potential metabolic markers associated with the immune status.
Collapse
Affiliation(s)
- Jeane de Souza Nogueira
- Histocompatibility and Cryopreservation Laboratory, IBRAG, Rio de Janeiro State University, CEP 20950-003 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cíntia Barros Santos-Rebouças
- Department of Genetics, IBRAG, Rio de Janeiro State University, CEP 20550-013 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael Mina Piergiorge
- Department of Genetics, IBRAG, Rio de Janeiro State University, CEP 20550-013 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula Valente
- CENABIO I, Institute of Medical Biochemistry, CNRMN, BioNMR, Federal University of Rio de Janeiro, CEP 21941-902 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos C Gama-Almeida
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, CEP 21941-902 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tatiana El-Bacha
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, CEP 21941-902 Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | - Orlando da Costa Ferreira
- Molecular Virology Laboratory, Federal University of Rio de Janeiro, CEP 21941-902 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luís Cristóvão Porto
- Histocompatibility and Cryopreservation Laboratory, IBRAG, Rio de Janeiro State University, CEP 20950-003 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tatiana Kelly da Silva Fidalgo
- Department of Preventive and Community Dentistry, Dental School, Rio de Janeiro State University, CEP 20551-030 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gilson Costa Dos Santos
- Department of Genetics, IBRAG, Rio de Janeiro State University, CEP 20550-013 Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
23
|
Berezhnoy G, Bissinger R, Liu A, Cannet C, Schäfer H, Kienzle K, Bitzer M, Häberle H, Göpel S, Trautwein C, Singh Y. Maintained imbalance of triglycerides, apolipoproteins, energy metabolites and cytokines in long-term COVID-19 syndrome patients. Front Immunol 2023; 14:1144224. [PMID: 37228606 PMCID: PMC10203989 DOI: 10.3389/fimmu.2023.1144224] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 04/17/2023] [Indexed: 05/27/2023] Open
Abstract
Background Deep metabolomic, proteomic and immunologic phenotyping of patients suffering from an infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have matched a wide diversity of clinical symptoms with potential biomarkers for coronavirus disease 2019 (COVID-19). Several studies have described the role of small as well as complex molecules such as metabolites, cytokines, chemokines and lipoproteins during infection and in recovered patients. In fact, after an acute SARS-CoV-2 viral infection almost 10-20% of patients experience persistent symptoms post 12 weeks of recovery defined as long-term COVID-19 syndrome (LTCS) or long post-acute COVID-19 syndrome (PACS). Emerging evidence revealed that a dysregulated immune system and persisting inflammation could be one of the key drivers of LTCS. However, how these biomolecules altogether govern pathophysiology is largely underexplored. Thus, a clear understanding of how these parameters within an integrated fashion could predict the disease course would help to stratify LTCS patients from acute COVID-19 or recovered patients. This could even allow to elucidation of a potential mechanistic role of these biomolecules during the disease course. Methods This study comprised subjects with acute COVID-19 (n=7; longitudinal), LTCS (n=33), Recov (n=12), and no history of positive testing (n=73). 1H-NMR-based metabolomics with IVDr standard operating procedures verified and phenotyped all blood samples by quantifying 38 metabolites and 112 lipoprotein properties. Univariate and multivariate statistics identified NMR-based and cytokine changes. Results Here, we report on an integrated analysis of serum/plasma by NMR spectroscopy and flow cytometry-based cytokines/chemokines quantification in LTCS patients. We identified that in LTCS patients lactate and pyruvate were significantly different from either healthy controls (HC) or acute COVID-19 patients. Subsequently, correlation analysis in LTCS group only among cytokines and amino acids revealed that histidine and glutamine were uniquely attributed mainly with pro-inflammatory cytokines. Of note, triglycerides and several lipoproteins (apolipoproteins Apo-A1 and A2) in LTCS patients demonstrate COVID-19-like alterations compared with HC. Interestingly, LTCS and acute COVID-19 samples were distinguished mostly by their phenylalanine, 3-hydroxybutyrate (3-HB) and glucose concentrations, illustrating an imbalanced energy metabolism. Most of the cytokines and chemokines were present at low levels in LTCS patients compared with HC except for IL-18 chemokine, which tended to be higher in LTCS patients. Conclusion The identification of these persisting plasma metabolites, lipoprotein and inflammation alterations will help to better stratify LTCS patients from other diseases and could help to predict ongoing severity of LTCS patients.
Collapse
Affiliation(s)
- Georgy Berezhnoy
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Rosi Bissinger
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | - Anna Liu
- Research Institute of Women’s Health, University of Tübingen, Tübingen, Germany
| | - Claire Cannet
- Bruker BioSpin, Applied Industrial and Clinical Division, Ettlingen, Germany
| | - Hartmut Schäfer
- Bruker BioSpin, Applied Industrial and Clinical Division, Ettlingen, Germany
| | - Katharina Kienzle
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | - Michael Bitzer
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
- Center for Personalized Medicine, University Hospital Tübingen, Tubingen, Germany
| | - Helene Häberle
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Siri Göpel
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | - Christoph Trautwein
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Yogesh Singh
- Research Institute of Women’s Health, University of Tübingen, Tübingen, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Next Generation Sequencing (NGS) Competence Center Tübingen (NCCT), University of Tübingen, Tübingen, Germany
| |
Collapse
|
24
|
Dubey R, Sinha N, Jagannathan NR. Potential of in vitro nuclear magnetic resonance of biofluids and tissues in clinical research. NMR IN BIOMEDICINE 2023; 36:e4686. [PMID: 34970810 DOI: 10.1002/nbm.4686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/18/2021] [Accepted: 12/21/2021] [Indexed: 06/14/2023]
Abstract
Body fluids, cells, and tissues contain a wide variety of metabolites that consist of a mixture of various low-molecular-weight compounds, including amino acids, peptides, lipids, nucleic acids, and organic acids, which makes comprehensive analysis more difficult. Quantitative nuclear magnetic resonance (NMR) spectroscopy is a well-established analytical technique for analyzing the metabolic profiles of body fluids, cells, and tissues. It enables fast and comprehensive detection, characterization, a high level of experimental reproducibility, minimal sample preparation, and quantification of various endogenous metabolites. In recent times, NMR-based metabolomics has been appreciably utilized in diverse branches of medicine, including microbiology, toxicology, pathophysiology, pharmacology, nutritional intervention, and disease diagnosis/prognosis. In this review, the utility of NMR-based metabolomics in clinical studies is discussed. The significance of in vitro NMR-based metabolomics as an effective tool for detecting metabolites and their variations in different diseases are discussed, together with the possibility of identifying specific biomarkers that can contribute to early detection and diagnosis of disease.
Collapse
Affiliation(s)
- Richa Dubey
- Centre of Biomedical Research, SGPGIMS Campus, Lucknow, India
| | - Neeraj Sinha
- Centre of Biomedical Research, SGPGIMS Campus, Lucknow, India
| | - Naranamangalam R Jagannathan
- Department of Radiology, Chettinad Hospital & Research Institute, Chettinad Academy of Research & Education, Kelambakkam, India
- Department of Radiology, Sri Ramachandra Institute of Higher Education & Research, Chennai, India
- Department of Electrical Engineering, Indian Institute Technology, Madras, Chennai, India
| |
Collapse
|
25
|
Bruzzone C, Conde R, Embade N, Mato JM, Millet O. Metabolomics as a powerful tool for diagnostic, pronostic and drug intervention analysis in COVID-19. Front Mol Biosci 2023; 10:1111482. [PMID: 36876049 PMCID: PMC9975567 DOI: 10.3389/fmolb.2023.1111482] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
COVID-19 currently represents one of the major health challenges worldwide. Albeit its infectious character, with onset affectation mainly at the respiratory track, it is clear that the pathophysiology of COVID-19 has a systemic character, ultimately affecting many organs. This feature enables the possibility of investigating SARS-CoV-2 infection using multi-omic techniques, including metabolomic studies by chromatography coupled to mass spectrometry or by nuclear magnetic resonance (NMR) spectroscopy. Here we review the extensive literature on metabolomics in COVID-19, that unraveled many aspects of the disease including: a characteristic metabotipic signature associated to COVID-19, discrimination of patients according to severity, effect of drugs and vaccination treatments and the characterization of the natural history of the metabolic evolution associated to the disease, from the infection onset to full recovery or long-term and long sequelae of COVID.
Collapse
Affiliation(s)
- Chiara Bruzzone
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bilbao, Bizkaia, Spain
| | - Ricardo Conde
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bilbao, Bizkaia, Spain
| | - Nieves Embade
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bilbao, Bizkaia, Spain
| | - José M. Mato
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bilbao, Bizkaia, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Oscar Millet
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bilbao, Bizkaia, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
26
|
Tian M, Liu H, Chen S, Yang Z, Tao W, Peng S, Che H, Jin L, The Secretariat of International Human Phenome Consortium. Report on the 3rd Board Meeting of the International Human Phenome Consortium. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:77-82. [PMID: 35757389 PMCID: PMC9215143 DOI: 10.1007/s43657-022-00065-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 01/31/2023]
Affiliation(s)
- Mei Tian
- Human Phenome Institute, Fudan University, Shanghai, 200438 China
- International Human Phenome Institutes (Shanghai), Shanghai, 200433 China
| | - Han Liu
- Human Phenome Institute, Fudan University, Shanghai, 200438 China
- International Human Phenome Institutes (Shanghai), Shanghai, 200433 China
| | - Shunling Chen
- International Human Phenome Institutes (Shanghai), Shanghai, 200433 China
| | - Zhong Yang
- International Human Phenome Institutes (Shanghai), Shanghai, 200433 China
- School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Weishuo Tao
- Human Phenome Institute, Fudan University, Shanghai, 200438 China
| | - Shiwen Peng
- Human Phenome Institute, Fudan University, Shanghai, 200438 China
| | - Huiting Che
- Human Phenome Institute, Fudan University, Shanghai, 200438 China
| | - Li Jin
- Human Phenome Institute, Fudan University, Shanghai, 200438 China
- International Human Phenome Institutes (Shanghai), Shanghai, 200433 China
- School of Life Sciences, Fudan University, Shanghai, 200438 China
- Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | | |
Collapse
|
27
|
Affiliation(s)
- G. A. Nagana Gowda
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington
| | - Daniel Raftery
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| |
Collapse
|
28
|
Manzi M, Zabalegui N, Monge ME. Postoperative Metabolic Phenoreversion in Clear Cell Renal Cell Carcinoma. J Proteome Res 2023; 22:1-15. [PMID: 36484409 DOI: 10.1021/acs.jproteome.2c00293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ultimate goal of surgical treatment in cancer is to remove the tumor mass for restoring a healthy state. A 16-lipid panel that discriminated healthy controls from clear cell renal cell carcinoma (ccRCC) patients in a prior study was evaluated in the present work in paired-serum samples collected from patients (n = 41) before and after nephrectomy. Changes in the lipid and metabolite fingerprints from ccRCC patients were investigated and compared with fingerprints from healthy individuals obtained by means of ultra-performance liquid chromatography-high-resolution mass spectrometry. The lipid panel differentiated phenotypes associated with metabolic restoration after surgery, representing a serum signature of phenoreversion to a healthy metabolic state. In particular, PC 16:0/0:0, PC 18:2/18:2, and linoleic acid allowed discriminating serum samples from ccRCC patients with poor prognosis from those with an improved outcome during the follow-up period. Ratios of PC 16:0/0:0 and PC 18:2/18:2 with linoleic acid levels may contribute as prognostic tools to support decision-making during the patient follow-up care. The preliminary character of these results should be validated with larger cohorts, including subjects with different ethnicities, life style, and diets. MetaboLights study references: MTBLS1839, MTBLS3838, and MTBLS4629.
Collapse
Affiliation(s)
- Malena Manzi
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2390, C1425FQD Ciudad de Buenos Aires, Argentina.,Departamento de Fisiología, Biología molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA Buenos Aires, Argentina
| | - Nicolás Zabalegui
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2390, C1425FQD Ciudad de Buenos Aires, Argentina.,Departamento de Química Inorgánica, Analítica y Química Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, C1428EGA Buenos Aires, Argentina
| | - María Eugenia Monge
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2390, C1425FQD Ciudad de Buenos Aires, Argentina
| |
Collapse
|
29
|
de Moraes Pontes JG, Dos Santos RV, Tasic L. NMR-Metabolomics in COVID-19 Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1412:197-209. [PMID: 37378768 DOI: 10.1007/978-3-031-28012-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
COVID-19 stands for Corona Virus Disease 2019, which starts as a viral infection that provokes illness with different symptoms and severity. The infected individuals can be asymptomatic or present with mild, moderate, severe, and critical illness with acute respiratory distress syndrome (ARDS), acute cardiac injury, and multiorgan failure. When the virus enters the cells, it replicates and provokes responses. Most diseased individuals resolve the problems in a short time but unfortunately, some may die, and almost 3 years after the first reported cases, COVID-19 still kills thousands per day worldwide. One of the problems in not curing the viral infection is that the virus passes by undetected in cells. This can be caused by the lack of pathogen-associated molecular patterns (PAMPs) that start an orchestrated immune response, such as activation of type 1 interferons (IFNs), inflammatory cytokines, chemokines, and antiviral defenses. Before all of these events can happen, the virus uses the infected cells and numerous small molecules as sources of energy and building blocks for newly synthesized viral nanoparticles that travel to and infect other host cells. Therefore, studying the cell metabolome and metabolomic changes in biofluids might give insights into the state of the viral infection, viral loads, and defense response. NMR-metabolomics can help in solving the real-time host interactions by monitoring concentration changes in metabolites. This chapter addresses the state of the art of COVIDomics by NMR analyses and presents exemplified biomolecules identified in different world regions and gravities of illness as potential biomarkers.
Collapse
Affiliation(s)
| | - Roney Vander Dos Santos
- Laboratory of Chemical Biology, Institute of Chemistry, University of Campinas (UNICAMP), CampinaEs, Sao Paulo, Brazil
| | - Ljubica Tasic
- Laboratory of Chemical Biology, Institute of Chemistry, University of Campinas (UNICAMP), CampinaEs, Sao Paulo, Brazil.
| |
Collapse
|
30
|
Bizkarguenaga M, Gil-Redondo R, Bruzzone C, Bernardo-Seisdedos G, Laín A, González-Valle B, Embade N, Mato JM, Millet O. Prospective Metabolomic Studies in Precision Medicine: The AKRIBEA Project. Handb Exp Pharmacol 2023; 277:275-297. [PMID: 36253553 DOI: 10.1007/164_2022_610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
For a long time, conventional medicine has analysed biomolecules to diagnose diseases. Yet, this approach has proven valid only for a limited number of metabolites and often through a bijective relationship with the disease (i.e. glucose relationship with diabetes), ultimately offering incomplete diagnostic value. Nowadays, precision medicine emerges as an option to improve the prevention and/or treatment of numerous pathologies, focusing on the molecular mechanisms, acting in a patient-specific dimension, and leveraging multiple contributing factors such as genetic, environmental, or lifestyle. Metabolomics grasps the required subcellular complexity while being sensitive to all these factors, which results in a most suitable technique for precision medicine. The aim of this chapter is to describe how NMR-based metabolomics can be integrated in the design of a precision medicine strategy, using the Precision Medicine Initiative of the Basque Country (the AKRIBEA project) as a case study. To that end, we will illustrate the procedures to be followed when conducting an NMR-based metabolomics study with a large cohort of individuals, emphasizing the critical points. The chapter will conclude with the discussion of some relevant biomedical applications.
Collapse
Affiliation(s)
- Maider Bizkarguenaga
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, Derio, Bizkaia, Spain
| | - Rubén Gil-Redondo
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, Derio, Bizkaia, Spain
| | - Chiara Bruzzone
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, Derio, Bizkaia, Spain
| | - Ganeko Bernardo-Seisdedos
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, Derio, Bizkaia, Spain
| | - Ana Laín
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, Derio, Bizkaia, Spain
| | - Beatriz González-Valle
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, Derio, Bizkaia, Spain
| | - Nieves Embade
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, Derio, Bizkaia, Spain
| | - José M Mato
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, Derio, Bizkaia, Spain
| | - Oscar Millet
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance, Bizkaia Science and Technology Park, Derio, Bizkaia, Spain.
| |
Collapse
|
31
|
Vignoli A, Meoni G, Ghini V, Di Cesare F, Tenori L, Luchinat C, Turano P. NMR-Based Metabolomics to Evaluate Individual Response to Treatments. Handb Exp Pharmacol 2023; 277:209-245. [PMID: 36318327 DOI: 10.1007/164_2022_618] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The aim of this chapter is to highlight the various aspects of metabolomics in relation to health and diseases, starting from the definition of metabolic space and of how individuals tend to maintain their own position in this space. Physio-pathological stimuli may cause individuals to lose their position and then regain it, or move irreversibly to other positions. By way of examples, mostly selected from our own work using 1H NMR on biological fluids, we describe the effects on the individual metabolomic fingerprint of mild external interventions, such as diet or probiotic administration. Then we move to pathologies (such as celiac disease, various types of cancer, viral infections, and other diseases), each characterized by a well-defined metabolomic fingerprint. We describe the effects of drugs on the disease fingerprint and on its reversal to a healthy metabolomic status. Drug toxicity can be also monitored by metabolomics. We also show how the individual metabolomic fingerprint at the onset of a disease may discriminate responders from non-responders to a given drug, or how it may be prognostic of e.g., cancer recurrence after many years. In parallel with fingerprinting, profiling (i.e., the identification and quantification of many metabolites and, in the case of selected biofluids, of the lipoprotein components that contribute to the 1H NMR spectral features) can provide hints on the metabolic pathways that are altered by a disease and assess their restoration after treatment.
Collapse
Affiliation(s)
- Alessia Vignoli
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Gaia Meoni
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Veronica Ghini
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Francesca Di Cesare
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Leonardo Tenori
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy.,Consorzio Interuniversitario Risonanze Magnetiche MetalloProteine (CIRMMP), Sesto Fiorentino, Italy
| | - Claudio Luchinat
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy.,Consorzio Interuniversitario Risonanze Magnetiche MetalloProteine (CIRMMP), Sesto Fiorentino, Italy
| | - Paola Turano
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy. .,Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy. .,Consorzio Interuniversitario Risonanze Magnetiche MetalloProteine (CIRMMP), Sesto Fiorentino, Italy.
| |
Collapse
|
32
|
Rössler T, Berezhnoy G, Singh Y, Cannet C, Reinsperger T, Schäfer H, Spraul M, Kneilling M, Merle U, Trautwein C. Quantitative Serum NMR Spectroscopy Stratifies COVID-19 Patients and Sheds Light on Interfaces of Host Metabolism and the Immune Response with Cytokines and Clinical Parameters. Metabolites 2022; 12:metabo12121277. [PMID: 36557315 PMCID: PMC9781847 DOI: 10.3390/metabo12121277] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
The complex manifestations of COVID-19 are still not fully decoded on the molecular level. We combined quantitative the nuclear magnetic resonance (NMR) spectroscopy serum analysis of metabolites, lipoproteins and inflammation markers with clinical parameters and a targeted cytokine panel to characterize COVID-19 in a large (534 patient samples, 305 controls) outpatient cohort of recently tested PCR-positive patients. The COVID-19 cohort consisted of patients who were predominantly in the initial phase of the disease and mostly exhibited a milder disease course. Concerning the metabolic profiles of SARS-CoV-2-infected patients, we identified markers of oxidative stress and a severe dysregulation of energy metabolism. NMR markers, such as phenylalanine, inflammatory glycoproteins (Glyc) and their ratio with the previously reported supramolecular phospholipid composite (Glyc/SPC), showed a predictive power comparable to laboratory parameters such as C-reactive protein (CRP) or ferritin. We demonstrated interfaces between the metabolism and the immune system, e.g., we could trace an interleukin (IL-6)-induced transformation of a high-density lipoprotein (HDL) to a pro-inflammatory actor. Finally, we showed that metadata such as age, sex and constitution (e.g., body mass index, BMI) need to be considered when exploring new biomarkers and that adding NMR parameters to existing diagnoses expands the diagnostic toolbox for patient stratification and personalized medicine.
Collapse
Affiliation(s)
- Titus Rössler
- Werner Siemens Imaging Center, Department for Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Georgy Berezhnoy
- Werner Siemens Imaging Center, Department for Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Yogesh Singh
- Institute of Medical Genetics & Applied Genomics, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Claire Cannet
- Bruker BioSpin GmbH, Applied Industrial and Clinical Division, 76275 Ettlingen, Germany
| | - Tony Reinsperger
- Bruker BioSpin GmbH, Applied Industrial and Clinical Division, 76275 Ettlingen, Germany
| | - Hartmut Schäfer
- Bruker BioSpin GmbH, Applied Industrial and Clinical Division, 76275 Ettlingen, Germany
| | - Manfred Spraul
- Bruker BioSpin GmbH, Applied Industrial and Clinical Division, 76275 Ettlingen, Germany
| | - Manfred Kneilling
- Werner Siemens Imaging Center, Department for Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Department of Dermatology, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-guided and Functionally Instructed Tumor Therapies”, Medical Faculty, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Uta Merle
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Christoph Trautwein
- Werner Siemens Imaging Center, Department for Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Correspondence:
| |
Collapse
|
33
|
Edgar M, Kuhn S, Page G, Grootveld M. Computational simulation of 1 H NMR profiles of complex biofluid analyte mixtures at differential operating frequencies: Applications to low-field benchtop spectra. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2022; 60:1097-1112. [PMID: 34847251 DOI: 10.1002/mrc.5236] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/30/2021] [Accepted: 11/23/2021] [Indexed: 06/13/2023]
Abstract
Estimations of accurate and reliable NMR chemical shift values, coupling patterns and constants within a reasonable timeframe remain significantly challenging, and the unavailability of reliable software strategies for the prediction of low-field (e.g., 60 MHz) spectra from those acquired at higher operating frequencies hampers their direct comparison. Hence, this study explored the applications of accessible software options for predicting these parameters in the 1 H NMR profiles of analytes as a function of magnetic field strength; this was performed for individual analytes and also for complex biofluid matrices featured in metabolomics investigations. For this purpose, results from the very first successful experimental acquisition and simulation of the 1 H NMR profiles of intact human salivary supernatant samples on a 60 MHz benchtop spectrometer were evaluated. Using salivary metabolite concentrations determined at 400 MHz, it was demonstrated that simulation of the low-field spectra of five biomolecules with the most prominent 1 H resonances detectable allowed multiple component fits to be applied to experimental spectra. Hence, these salivary 1 H NMR profiles could be successfully predicted throughout the 45-600 MHz operating frequency range. With the exception of propionate resonance multiplets, which revealed more complex coupling patterns at low field and required more astute computational and fitting options, valuable quantitative metabolomics data on salivary acetate, formate, methanol and glycine could be attained from low-field spectrometres. These studies are both timely and pertinent in view of the recent advancement of low-field benchtop NMR facilities for diagnostically significant biomarker tracking in biofluids. Experiments performed with added ammonium chloride to facilitate the release of salivary metabolites from biopolymer binding sites provided evidence that a small but nevertheless significant proportion of propionate, but not lactate, was bound to such sites, an observation of much relevance to biomolecule quantification in salivary metabolomics investigations.
Collapse
Affiliation(s)
- Mark Edgar
- Department of Chemistry, University of Loughborough, Loughborough, UK
| | - Stefan Kuhn
- School of Computer Science and Informatics, De Montfort University, Leicester, UK
| | - Georgina Page
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| | - Martin Grootveld
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| |
Collapse
|
34
|
Ghorasaini M, Tsezou KI, Verhoeven A, Mohammed Y, Vlachoyiannopoulos P, Mikros E, Giera M. Congruence and Complementarity of Differential Mobility Spectrometry and NMR Spectroscopy for Plasma Lipidomics. Metabolites 2022; 12:1030. [PMID: 36355113 PMCID: PMC9699282 DOI: 10.3390/metabo12111030] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
The lipid composition of lipoprotein particles is determinative of their respective formation and function. In turn, the combination and correlation of nuclear magnetic resonance (NMR)-based lipoprotein measurements with mass spectrometry (MS)-based lipidomics is an appealing technological combination for a better understanding of lipid metabolism in health and disease. Here, we developed a combined workflow for subsequent NMR- and MS-based analysis on single sample aliquots of human plasma. We evaluated the quantitative agreement of the two platforms for lipid quantification and benchmarked our combined workflow. We investigated the congruence and complementarity between the platforms in order to facilitate a better understanding of patho-physiological lipoprotein and lipid alterations. We evaluated the correlation and agreement between the platforms. Next, we compared lipid class concentrations between healthy controls and rheumatoid arthritis patient samples to investigate the consensus among the platforms on differentiating the two groups. Finally, we performed correlation analysis between all measured lipoprotein particles and lipid species. We found excellent agreement and correlation (r > 0.8) between the platforms and their respective diagnostic performance. Additionally, we generated correlation maps detailing lipoprotein/lipid interactions and describe disease-relevant correlations.
Collapse
Affiliation(s)
- Mohan Ghorasaini
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Konstantina Ismini Tsezou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 157 71 Athens, Greece
- Pharmagnose S.A., 320 11 Inofyta, Greece
| | - Aswin Verhoeven
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Yassene Mohammed
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Genome BC Proteomics Centre, University of Victoria, Victoria, BC V8Z 5N3, Canada
| | - Panayiotis Vlachoyiannopoulos
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 157 71 Athens, Greece
- Institute for Autoimmune Systemic and Neurologic Diseases, 104 31 Athens, Greece
| | - Emmanuel Mikros
- Pharmagnose S.A., 320 11 Inofyta, Greece
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistiomiopolis, Zografou, 157 71 Athens, Greece
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
35
|
Quantitative NMR-Based Lipoprotein Analysis Identifies Elevated HDL-4 and Triglycerides in the Serum of Alzheimer’s Disease Patients. Int J Mol Sci 2022; 23:ijms232012472. [PMID: 36293327 PMCID: PMC9604278 DOI: 10.3390/ijms232012472] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/30/2022] [Accepted: 10/08/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia in the elderly and has been associated with changes in lipoprotein metabolism. We performed quantitative lipoprotein analysis in a local cohort of cognitively impaired elderly and control subjects using standardized nuclear magnetic resonance (NMR) spectroscopy. A commercially available quantitative NMR-based assay covering 112 lipoprotein main and subtype variables was used to investigate blood serum samples from a moderate cohort size of 161 persons (71 female, 90 male), including measures of quality control. Additionally, clinical metadata and cerebrospinal fluid AD biomarkers were collected and used for analysis. High-density lipoprotein (HDL) HDL-4 subfraction levels were mostly high in female individuals with mild cognitive impairment (MCI), followed by AD. Low-density lipoprotein (LDL) LDL-2 cholesterol was slightly elevated in male AD patients. HDL-2 apolipoprotein Apo-A1, HDL-2 phospholipids, and HDL-3 triglycerides were highly abundant in AD and MCI women compared to men. When considering clinical biomarkers (Aβ, tau), very low-density lipoprotein (VLDL) VLDL-1 and intermediate-density lipoprotein (IDL) triglycerides were substantially higher in AD compared to MCI. In addition, triglyceride levels correlated positively with dementia. Different lipoprotein serum patterns were identified for AD, MCI, and control subjects. Interestingly, HDL-4 and LDL-2 cholesterol parameters revealed strong gender-specific changes in the context of AD-driven dementia. As gender-based comparisons were based on smaller sub-groups with a low n-number, several statistical findings did not meet the significance threshold for multiple comparisons testing. Still, our finding suggests that serum HDL-4 parameters and various triglycerides correlate positively with AD pathology which could be a read-out of extended lipids traveling through the blood-brain barrier, supporting amyloid plaque formation processes. Thereof, we see herein a proof of concept that this quantitative NMR-based lipoprotein assay can generate important and highly interesting data for refined AD diagnosis and patient stratification, especially when larger cohorts are available.
Collapse
|
36
|
Cheung E, Xia Y, Caporini MA, Gilmore JL. Tools shaping drug discovery and development. BIOPHYSICS REVIEWS 2022; 3:031301. [PMID: 38505278 PMCID: PMC10903431 DOI: 10.1063/5.0087583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 06/21/2022] [Indexed: 03/21/2024]
Abstract
Spectroscopic, scattering, and imaging methods play an important role in advancing the study of pharmaceutical and biopharmaceutical therapies. The tools more familiar to scientists within industry and beyond, such as nuclear magnetic resonance and fluorescence spectroscopy, serve two functions: as simple high-throughput techniques for identification and purity analysis, and as potential tools for measuring dynamics and structures of complex biological systems, from proteins and nucleic acids to membranes and nanoparticle delivery systems. With the expansion of commercial small-angle x-ray scattering instruments into the laboratory setting and the accessibility of industrial researchers to small-angle neutron scattering facilities, scattering methods are now used more frequently in the industrial research setting, and probe-less time-resolved small-angle scattering experiments are now able to be conducted to truly probe the mechanism of reactions and the location of individual components in complex model or biological systems. The availability of atomic force microscopes in the past several decades enables measurements that are, in some ways, complementary to the spectroscopic techniques, and wholly orthogonal in others, such as those related to nanomechanics. As therapies have advanced from small molecules to protein biologics and now messenger RNA vaccines, the depth of biophysical knowledge must continue to serve in drug discovery and development to ensure quality of the drug, and the characterization toolbox must be opened up to adapt traditional spectroscopic methods and adopt new techniques for unraveling the complexities of the new modalities. The overview of the biophysical methods in this review is meant to showcase the uses of multiple techniques for different modalities and present recent applications for tackling particularly challenging situations in drug development that can be solved with the aid of fluorescence spectroscopy, nuclear magnetic resonance spectroscopy, atomic force microscopy, and small-angle scattering.
Collapse
Affiliation(s)
- Eugene Cheung
- Moderna, 200 Technology Square, Cambridge, Massachusetts 02139, USA
| | - Yan Xia
- Moderna, 200 Technology Square, Cambridge, Massachusetts 02139, USA
| | - Marc A. Caporini
- Moderna, 200 Technology Square, Cambridge, Massachusetts 02139, USA
| | - Jamie L. Gilmore
- Moderna, 200 Technology Square, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
37
|
Ciccarelli M, Merciai F, Carrizzo A, Sommella E, Di Pietro P, Caponigro V, Salviati E, Musella S, Sarno VD, Rusciano M, Toni AL, Iesu P, Izzo C, Schettino G, Conti V, Venturini E, Vitale C, Scarpati G, Bonadies D, Rispoli A, Polverino B, Poto S, Pagliano P, Piazza O, Licastro D, Vecchione C, Campiglia P. Untargeted lipidomics reveals specific lipid profiles in COVID-19 patients with different severity from Campania region (Italy). J Pharm Biomed Anal 2022; 217:114827. [PMID: 35569273 PMCID: PMC9085356 DOI: 10.1016/j.jpba.2022.114827] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 02/08/2023]
Abstract
COVID-19 infection evokes various systemic alterations that push patients not only towards severe acute respiratory syndrome but causes an important metabolic dysregulation with following multi-organ alteration and potentially poor outcome. To discover novel potential biomarkers able to predict disease's severity and patient's outcome, in this study we applied untargeted lipidomics, by a reversed phase ultra-high performance liquid chromatography-trapped ion mobility mass spectrometry platform (RP-UHPLC-TIMS-MS), on blood samples collected at hospital admission in an Italian cohort of COVID-19 patients (45 mild, 54 severe, 21 controls). In a subset of patients, we also collected a second blood sample in correspondence of clinical phenotype modification (longitudinal population). Plasma lipid profiles revealed several lipids significantly modified in COVID-19 patients with respect to controls and able to discern between mild and severe clinical phenotype. Severe patients were characterized by a progressive decrease in the levels of LPCs, LPC-Os, PC-Os, and, on the contrary, an increase in overall TGs, PEs, and Ceramides. A machine learning model was built by using both the entire dataset and with a restricted lipid panel dataset, delivering comparable results in predicting severity (AUC= 0.777, CI: 0.639-0.904) and outcome (AUC= 0.789, CI: 0.658-0.910). Finally, re-building the model with 25 longitudinal (t1) samples, this resulted in 21 patients correctly classified. In conclusion, this study highlights specific lipid profiles that could be used monitor the possible trajectory of COVID-19 patients at hospital admission, which could be used in targeted approaches.
Collapse
Affiliation(s)
- Michele Ciccarelli
- Department of Medicine and Surgery, University of Salerno, Baronissi, SA, Italy
| | - Fabrizio Merciai
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy,PhD Program in Drug Discovery and Development, University of Salerno, Fisciano, SA, Italy
| | - Albino Carrizzo
- Department of Medicine and Surgery, University of Salerno, Baronissi, SA, Italy,IRCCS Neuromed, Loc. Camerelle, Pozzilli, IS, Italy
| | - Eduardo Sommella
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| | - Paola Di Pietro
- Department of Medicine and Surgery, University of Salerno, Baronissi, SA, Italy
| | - Vicky Caponigro
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| | | | - Simona Musella
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| | | | | | - Anna Laura Toni
- Department of Medicine and Surgery, University of Salerno, Baronissi, SA, Italy
| | - Paola Iesu
- Department of Medicine and Surgery, University of Salerno, Baronissi, SA, Italy
| | - Carmine Izzo
- Department of Medicine and Surgery, University of Salerno, Baronissi, SA, Italy
| | | | - Valeria Conti
- Department of Medicine and Surgery, University of Salerno, Baronissi, SA, Italy
| | | | - Carolina Vitale
- San Giovanni di Dio e Ruggi D'Aragona University Hospital, Salerno, Italy
| | - Giuliana Scarpati
- San Giovanni di Dio e Ruggi D'Aragona University Hospital, Salerno, Italy
| | - Domenico Bonadies
- San Giovanni di Dio e Ruggi D'Aragona University Hospital, Salerno, Italy
| | - Antonella Rispoli
- San Giovanni di Dio e Ruggi D'Aragona University Hospital, Salerno, Italy
| | | | - Sergio Poto
- San Giovanni di Dio e Ruggi D'Aragona University Hospital, Salerno, Italy
| | - Pasquale Pagliano
- Department of Medicine and Surgery, University of Salerno, Baronissi, SA, Italy
| | - Ornella Piazza
- Department of Medicine and Surgery, University of Salerno, Baronissi, SA, Italy
| | | | - Carmine Vecchione
- Department of Medicine and Surgery, University of Salerno, Baronissi, SA, Italy,IRCCS Neuromed, Loc. Camerelle, Pozzilli, IS, Italy,Corresponding author at: Department of Medicine and Surgery, University of Salerno, Baronissi, SA, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy,Corresponding author
| |
Collapse
|
38
|
Karu N, Kindt A, van Gammeren AJ, Ermens AAM, Harms AC, Portengen L, Vermeulen RCH, Dik WA, Langerak AW, van der Velden VHJ, Hankemeier T. Severe COVID-19 Is Characterised by Perturbations in Plasma Amines Correlated with Immune Response Markers, and Linked to Inflammation and Oxidative Stress. Metabolites 2022; 12:618. [PMID: 35888742 PMCID: PMC9321395 DOI: 10.3390/metabo12070618] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 02/01/2023] Open
Abstract
The COVID-19 pandemic raised a need to characterise the biochemical response to SARS-CoV-2 infection and find biological markers to identify therapeutic targets. In support of these aims, we applied a range of LC-MS platforms to analyse over 100 plasma samples from patients with varying COVID-19 severity and with detailed clinical information on inflammatory responses (>30 immune markers). The first publication in a series reports the results of quantitative LC-MS/MS profiling of 56 amino acids and derivatives. A comparison between samples taken from ICU and ward patients revealed a notable increase in ten post-translationally modified amino acids that correlated with markers indicative of an excessive immune response: TNF-alpha, neutrophils, markers for macrophage, and leukocyte activation. Severe patients also had increased kynurenine, positively correlated with CRP and cytokines that induce its production. ICU and ward patients with high IL-6 showed decreased levels of 22 immune-supporting and anti-oxidative amino acids and derivatives (e.g., glutathione, GABA). These negatively correlated with CRP and IL-6 and positively correlated with markers indicative of adaptive immune activation. Including corresponding alterations in convalescing ward patients, the overall metabolic picture of severe COVID-19 reflected enhanced metabolic demands to maintain cell proliferation and redox balance, alongside increased inflammation and oxidative stress.
Collapse
Affiliation(s)
- Naama Karu
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (A.K.); (A.C.H.)
| | - Alida Kindt
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (A.K.); (A.C.H.)
| | - Adriaan J. van Gammeren
- Department of Clinical Chemistry and Hematology, Amphia Hospital, 4818 CK Breda, The Netherlands; (A.J.v.G.); (A.A.M.E.)
| | - Anton A. M. Ermens
- Department of Clinical Chemistry and Hematology, Amphia Hospital, 4818 CK Breda, The Netherlands; (A.J.v.G.); (A.A.M.E.)
| | - Amy C. Harms
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (A.K.); (A.C.H.)
| | - Lutzen Portengen
- Department of Population Health Sciences, Institute for Risk Assessment Sciences, University Utrecht, 3584 CK Utrecht, The Netherlands; (L.P.); (R.C.H.V.)
| | - Roel C. H. Vermeulen
- Department of Population Health Sciences, Institute for Risk Assessment Sciences, University Utrecht, 3584 CK Utrecht, The Netherlands; (L.P.); (R.C.H.V.)
| | - Willem A. Dik
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (W.A.D.); (A.W.L.); (V.H.J.v.d.V.)
| | - Anton W. Langerak
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (W.A.D.); (A.W.L.); (V.H.J.v.d.V.)
| | - Vincent H. J. van der Velden
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (W.A.D.); (A.W.L.); (V.H.J.v.d.V.)
| | - Thomas Hankemeier
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (A.K.); (A.C.H.)
| |
Collapse
|
39
|
Correia BSB, Ferreira VG, Piagge PMFD, Almeida MB, Assunção NA, Raimundo JRS, Fonseca FLA, Carrilho E, Cardoso DR. 1H qNMR-Based Metabolomics Discrimination of Covid-19 Severity. J Proteome Res 2022; 21:1640-1653. [PMID: 35674498 PMCID: PMC9212193 DOI: 10.1021/acs.jproteome.1c00977] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Indexed: 01/08/2023]
Abstract
The coronavirus disease 2019 (Covid-19), which caused respiratory problems in many patients worldwide, led to more than 5 million deaths by the end of 2021. Experienced symptoms vary from mild to severe illness. Understanding the infection severity to reach a better prognosis could be useful to the clinics, and one study area to fulfill one piece of this biological puzzle is metabolomics. The metabolite profile and/or levels being monitored can help predict phenotype properties. Therefore, this study evaluated plasma metabolomes of 110 individual samples, 57 from control patients and 53 from recent positive cases of Covid-19 (IgM 98% reagent), representing mild to severe symptoms, before any clinical intervention. Polar metabolites from plasma samples were analyzed by quantitative 1H NMR. Glycerol, 3-aminoisobutyrate, formate, and glucuronate levels showed alterations in Covid-19 patients compared to those in the control group (Tukey's HSD p-value cutoff = 0.05), affecting the lactate, phenylalanine, tyrosine, and tryptophan biosynthesis and d-glutamine, d-glutamate, and glycerolipid metabolisms. These metabolic alterations show that SARS-CoV-2 infection led to disturbance in the energetic system, supporting the viral replication and corroborating with the severe clinical conditions of patients. Six polar metabolites (glycerol, acetate, 3-aminoisobutyrate, formate, glucuronate, and lactate) were revealed by PLS-DA and predicted by ROC curves and ANOVA to be potential prognostic metabolite panels for Covid-19 and considered clinically relevant for predicting infection severity due to their straight roles in the lipid and energy metabolism. Thus, metabolomics from samples of Covid-19 patients is a powerful tool for a better understanding of the disease mechanism of action and metabolic consequences of the infection in the human body and may corroborate allowing clinicians to intervene quickly according to the needs of Covid-19 patients.
Collapse
Affiliation(s)
- Banny S. B. Correia
- Instituto de Química de São Carlos,
Universidade de São Paulo, São Carlos, SP
13566-590, Brazil
| | - Vinicius G. Ferreira
- Instituto de Química de São Carlos,
Universidade de São Paulo, São Carlos, SP
13566-590, Brazil
- Instituto Nacional de Ciência e Tecnologia de
Bioanalítica, INCTBio, Campinas, SP 13083-861,
Brazil
| | | | - Mariana B. Almeida
- Instituto de Química de São Carlos,
Universidade de São Paulo, São Carlos, SP
13566-590, Brazil
- Instituto Nacional de Ciência e Tecnologia de
Bioanalítica, INCTBio, Campinas, SP 13083-861,
Brazil
| | - Nilson A. Assunção
- Instituto de Ciências Ambientais, Químicas
e Farmacêuticas, Universidade Federal de São
Paulo, São Paulo, SP 09972-270, Brazil
| | | | - Fernando L. A. Fonseca
- Faculdade de Medicina do
ABC, Santo André, SP 09060-870, Brazil
- Departamento de Ciências Farmacêuticas,
Universidade Federal de São Paulo, Diadema, SP
09972-270, Brazil
| | - Emanuel Carrilho
- Instituto de Química de São Carlos,
Universidade de São Paulo, São Carlos, SP
13566-590, Brazil
- Instituto Nacional de Ciência e Tecnologia de
Bioanalítica, INCTBio, Campinas, SP 13083-861,
Brazil
| | - Daniel R. Cardoso
- Instituto de Química de São Carlos,
Universidade de São Paulo, São Carlos, SP
13566-590, Brazil
| |
Collapse
|
40
|
Makaremi S, Asgarzadeh A, Kianfar H, Mohammadnia A, Asghariazar V, Safarzadeh E. The role of IL-1 family of cytokines and receptors in pathogenesis of COVID-19. Inflamm Res 2022; 71:923-947. [PMID: 35751653 PMCID: PMC9243884 DOI: 10.1007/s00011-022-01596-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/29/2022] [Indexed: 12/12/2022] Open
Abstract
A global pandemic has erupted as a result of the new brand coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This pandemic has been consociated with widespread mortality worldwide. The antiviral immune response is an imperative factor in confronting the recent coronavirus disease 2019 (COVID-19) infections. Meantime, cytokines recognize as crucial components in guiding the appropriate immune pathways in the restraining and eradication of the virus. Moreover, SARS-CoV-2 can induce uncontrolled inflammatory responses characterized by hyper-inflammatory cytokine production, which causes cytokine storm and acute respiratory distress syndrome (ARDS). As excessive inflammatory responses are contributed to the severe stage of the COVID-19 disease, therefore, the pro-inflammatory cytokines are regarded as the Achilles heel during COVID-19 infection. Among these cytokines, interleukin (IL-) 1 family cytokines (IL-1, IL-18, IL-33, IL-36, IL-37, and IL-38) appear to have a strong inflammatory role in severe COVID-19. Hence, understanding the underlying inflammatory mechanism of these cytokines during infection is critical for reducing the symptoms and severity of the disease. Here, the possible mechanisms and pathways involved in inflammatory immune responses are discussed.
Collapse
Affiliation(s)
- Shima Makaremi
- School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran.,Department of Health Information Management, School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Asgarzadeh
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.,Department of Health Information Management, School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamed Kianfar
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.,Department of Health Information Management, School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Alireza Mohammadnia
- Department of Health Information Management, School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Vahid Asghariazar
- Department of Health Information Management, School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Department of Health Information Management, School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran. .,Department of Microbiology, Parasitology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
41
|
Trovato FM, Mujib S, Jerome E, Cavazza A, Morgan P, Smith J, Depante MT, O'Reilly K, Luxton J, Mare T, Napoli S, McPhail MJ. Immunometabolic analysis shows a distinct cyto-metabotype in Covid-19 compared to sepsis from other causes. Heliyon 2022; 8:e09733. [PMID: 35774516 PMCID: PMC9225950 DOI: 10.1016/j.heliyon.2022.e09733] [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: 10/22/2021] [Revised: 03/28/2022] [Accepted: 06/10/2022] [Indexed: 12/15/2022] Open
Abstract
Background In Covid-19, profound systemic inflammatory responses are accompanied by both metabolic risk factors for severity and, separately, metabolic mechanisms have been shown to underly disease progression. It is unknown whether this reflects similar situations in sepsis or is a unique characteristic of Covid-19. Aims Define the immunometabolic signature of Covid-19. Methods 65 patients with Covid-19,19 patients with sepsis and 14 healthy controls were recruited and sampled for plasma, serum and peripheral blood mononuclear cells (PBMCs) through 10 days of critical illness. Metabotyping was performed using the Biocrates p180 kit and multiplex cytokine profiling undertaken. PBMCs underwent phenotyping by flow cytometry. Immune and metabolic readouts were integrated and underwent pathway analysis. Results Phopsphatidylcholines (PC) are reduced in Covid-19 but greater than in sepsis. Compared to controls, tryptophan is reduced in Covid-19 and inversely correlated with the severity of the disease and IFN-ɣ concentrations, conversely the kyneurine and kyneurine/tryptophan ratio increased in the most severe cases. These metabolic changes were consistent through 2 pandemic waves in our centre. PD-L1 expression in CD8+ T cells, Tregs and CD14+ monocytes was increased in Covid-19 compared to controls. Conclusions In our cohort, Covid-19 is associated with monocytopenia, increased CD14+ and Treg PD-L1 expression correlating with IFN-ɣ plasma concentration and disease severity (SOFA score). The latter is also associated with metabolic derangements of Tryptophan, LPC 16:0 and PCs. Lipid metabolism, in particular phosphatidylcholines and lysophosphatidylcolines, seems strictly linked to immune response in Covid-19. Our results support the hypothesis that IFN-ɣ -PD-L1 axis might be involved in the cytokine release syndrome typical of severe Covid-19 and the phenomenon persisted through multiple pandemic waves despite use of immunomodulation.
Collapse
Affiliation(s)
- Francesca M Trovato
- Institute of Liver Studies, King's College Hospital, London, United Kingdom.,Department of Inflammation BIology, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, Kings College London, United Kingdom
| | - Salma Mujib
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Ellen Jerome
- Institute of Liver Studies, King's College Hospital, London, United Kingdom.,Department of Inflammation BIology, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, Kings College London, United Kingdom
| | - Anna Cavazza
- Institute of Liver Studies, King's College Hospital, London, United Kingdom.,Department of Inflammation BIology, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, Kings College London, United Kingdom
| | - Phillip Morgan
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - John Smith
- Anaesthetics, Critical Care, Emergency and Trauma Research Delivery Unit, Kings College Hospital, London, United Kingdom
| | - Maria Theresa Depante
- Anaesthetics, Critical Care, Emergency and Trauma Research Delivery Unit, Kings College Hospital, London, United Kingdom
| | - Kevin O'Reilly
- Anaesthetics, Critical Care, Emergency and Trauma Research Delivery Unit, Kings College Hospital, London, United Kingdom
| | - James Luxton
- Contract R&D Department (Viapath), Kings College Hospital, London, United Kingdom
| | - Tracey Mare
- Contract R&D Department (Viapath), Kings College Hospital, London, United Kingdom
| | - Salvatore Napoli
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Mark Jw McPhail
- Institute of Liver Studies, King's College Hospital, London, United Kingdom.,Department of Inflammation BIology, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, Kings College London, United Kingdom
| |
Collapse
|
42
|
Ghini V, Maggi L, Mazzoni A, Spinicci M, Zammarchi L, Bartoloni A, Annunziato F, Turano P. Serum NMR Profiling Reveals Differential Alterations in the Lipoproteome Induced by Pfizer-BioNTech Vaccine in COVID-19 Recovered Subjects and Naïve Subjects. Front Mol Biosci 2022; 9:839809. [PMID: 35480886 PMCID: PMC9037139 DOI: 10.3389/fmolb.2022.839809] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/07/2022] [Indexed: 12/16/2022] Open
Abstract
1H NMR spectra of sera have been used to define the changes induced by vaccination with Pfizer-BioNTech vaccine (2 shots, 21 days apart) in 10 COVID-19-recovered subjects and 10 COVID-19-naïve subjects at different time points, starting from before vaccination, then weekly until 7 days after second injection, and finally 1 month after the second dose. The data show that vaccination does not induce any significant variation in the metabolome, whereas it causes changes at the level of lipoproteins. The effects are different in the COVID-19-recovered subjects with respect to the naïve subjects, suggesting that a previous infection reduces the vaccine modulation of the lipoproteome composition.
Collapse
Affiliation(s)
- Veronica Ghini
- Department of Chemistry, University of Florence, Florence, Italy
- Magnetic Resonance Center (CERM), University of Florence, Florence, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Michele Spinicci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Disease Unit, Careggi University Hospital, Florence, Italy
| | - Lorenzo Zammarchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Disease Unit, Careggi University Hospital, Florence, Italy
| | - Alessandro Bartoloni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Disease Unit, Careggi University Hospital, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| | - Paola Turano
- Department of Chemistry, University of Florence, Florence, Italy
- Magnetic Resonance Center (CERM), University of Florence, Florence, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), Florence, Italy
- *Correspondence: Paola Turano,
| |
Collapse
|
43
|
Castiglione Morelli MA, Iuliano A, Schettini SCA, Ferri A, Colucci P, Viggiani L, Matera I, Ostuni A. Are the Follicular Fluid Characteristics of Recovered Coronavirus Disease 2019 Patients Different From Those of Vaccinated Women Approaching in vitro Fertilization? Front Physiol 2022; 13:840109. [PMID: 35283772 PMCID: PMC8905595 DOI: 10.3389/fphys.2022.840109] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/26/2022] [Indexed: 12/18/2022] Open
Abstract
The aim of this pilot study is to evaluate if SARS-CoV-2 infection or vaccination against SARS-CoV-2 infection induce observable metabolic effects in follicular fluid of women who are following in vitro fertilization (IVF) treatments. The possible impact of coronavirus disease 2019 (COVID-19) on fertility and IVF outcome is considered. We have selected for this study: six women vaccinated against SARS-CoV-2 infection, five recovered COVID-19 patients, and we used nine healthy women as the control group. At the time of oocytes retrieval from participants in the study, follicular fluids were collected and metabolomic analysis was performed by 1H NMR spectroscopy in combination with multivariate analysis to interpret the spectral data. The search for antibody positivity in the follicular fluid aspirates was also carried out, together with the western blotting analysis of some inflammatory proteins, interleukin-6, tumor necrosis factor α (TNFα), and the free radical scavenger superoxide dismutase 2. Higher levels of Ala and Pro together with lower levels of lipids and trimethylamine N-oxide (TMAO) were found in follicular fluids (FFs) of vaccinated women while lower levels of many metabolites were detected in FFs of recovered COVID patients. Expression level of TNF-α was significantly lower both in recovered COVID-19 patients and vaccinated women in comparison to healthy controls.
Collapse
Affiliation(s)
| | - Assunta Iuliano
- Center for Reproductive Medicine of "San Carlo" Hospital, Potenza, Italy
| | | | - Angela Ferri
- Center for Reproductive Medicine of "San Carlo" Hospital, Potenza, Italy
| | - Paola Colucci
- Center for Reproductive Medicine of "San Carlo" Hospital, Potenza, Italy
| | - Licia Viggiani
- Department of Sciences, University of Basilicata, Potenza, Italy
| | - Ilenia Matera
- Department of Sciences, University of Basilicata, Potenza, Italy
| | - Angela Ostuni
- Department of Sciences, University of Basilicata, Potenza, Italy
| |
Collapse
|
44
|
Valdés A, Moreno LO, Rello SR, Orduña A, Bernardo D, Cifuentes A. Metabolomics study of COVID-19 patients in four different clinical stages. Sci Rep 2022; 12:1650. [PMID: 35102215 PMCID: PMC8803913 DOI: 10.1038/s41598-022-05667-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 12/13/2021] [Indexed: 12/22/2022] Open
Abstract
SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) is the coronavirus strain causing the respiratory pandemic COVID-19 (coronavirus disease 2019). To understand the pathobiology of SARS-CoV-2 in humans it is necessary to unravel the metabolic changes that are produced in the individuals once the infection has taken place. The goal of this work is to provide new information about the altered biomolecule profile and with that the altered biological pathways of patients in different clinical situations due to SARS-CoV-2 infection. This is done via metabolomics using HPLC-QTOF-MS analysis of plasma samples at COVID-diagnose from a total of 145 adult patients, divided into different clinical stages based on their subsequent clinical outcome (25 negative controls (non-COVID); 28 positive patients with asymptomatic disease not requiring hospitalization; 27 positive patients with mild disease defined by a total time in hospital lower than 10 days; 36 positive patients with severe disease defined by a total time in hospital over 20 days and/or admission at the ICU; and 29 positive patients with fatal outcome or deceased). Moreover, follow up samples between 2 and 3 months after hospital discharge were also obtained from the hospitalized patients with mild prognosis. The final goal of this work is to provide biomarkers that can help to better understand how the COVID-19 illness evolves and to predict how a patient could progress based on the metabolites profile of plasma obtained at an early stage of the infection. In the present work, several metabolites were found as potential biomarkers to distinguish between the end-stage and the early-stage (or non-COVID) disease groups. These metabolites are mainly involved in the metabolism of carnitines, ketone bodies, fatty acids, lysophosphatidylcholines/phosphatidylcholines, tryptophan, bile acids and purines, but also omeprazole. In addition, the levels of several of these metabolites decreased to "normal" values at hospital discharge, suggesting some of them as early prognosis biomarkers in COVID-19 at diagnose.
Collapse
Affiliation(s)
- Alberto Valdés
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, CSIC, Nicolás Cabrera 9, 28049, Madrid, Spain
| | - Lorena Ortega Moreno
- Dpt. Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
| | - Silvia Rojo Rello
- Servicio de Microbiología, Hospital Clínico Universitario de Valladolid, 47004, Valladolid, Spain
| | - Antonio Orduña
- Servicio de Microbiología, Hospital Clínico Universitario de Valladolid, 47004, Valladolid, Spain
- Departamento de Microbiología, Universidad de Valladolid, Valladolid, Spain
| | - David Bernardo
- Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Alejandro Cifuentes
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, CSIC, Nicolás Cabrera 9, 28049, Madrid, Spain.
| |
Collapse
|
45
|
Ganesan R, Sekaran S, Vimalraj S. Solid-state 1H NMR-based metabolomics assessment of tributylin effects in zebrafish bone. Life Sci 2022; 289:120233. [PMID: 34921865 DOI: 10.1016/j.lfs.2021.120233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/23/2021] [Accepted: 12/08/2021] [Indexed: 12/13/2022]
Abstract
Tributyltin (TBT), an endocrine disruptor is used globally in agribusiness and industries as biocides, heat stabilizers, and in chemical catalysis. It is known for its deleterious effects on bone by negatively impacting the functions of osteoblasts, osteoclasts and mesenchymal stem cells. However, the impact of TBT on the metabolomics profile in bone is not yet studied. Here, we demonstrate alterations in chemical metabolomics profiles measured by solid state 1H nuclear magnetic resonance (1H NMR) spectroscopy in zebrafish bone following tributyltin (TBT) treatment. TBT of 0, 100, 200, 300, 400 and 500 μg/L were exposed to zebrafish. From this, zebrafish bone has subjected for further metabolomics profiling. Samples were measured via one-dimensional (1D) solvent -suppressed and T2- filtered methods with in vivo zebrafish metabolites. A dose dependent alteration in the metabolomics profile was observed and results indicated a disturbed aminoacid metabolism, TCA cycle, and glycolysis. We found a significant alteration in the levels of glutamate, glutamine, glutathione, trimethylamine N-oxide (TMAO), and other metabolites. This investigation hints us the deleterious effects of TBT on zebrafish bone enabling a comprehensive understanding of metabolomics profile and is expected to play a crucial role in understanding the deleterious effects of various endocrine disruptor on bone.
Collapse
Affiliation(s)
- Raja Ganesan
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24253, Republic of Korea; Department of Biological Sciences, Pusan National University, Busan 46241, Republic of Korea.
| | - Saravanan Sekaran
- Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai 600077, India.
| | | |
Collapse
|
46
|
Nitschke P, Lodge S, Kimhofer T, Masuda R, Bong SH, Hall D, Schäfer H, Spraul M, Pompe N, Diercks T, Bernardo-Seisdedos G, Mato JM, Millet O, Susic D, Henry A, El-Omar EM, Holmes E, Lindon JC, Nicholson JK, Wist J. J-Edited DIffusional Proton Nuclear Magnetic Resonance Spectroscopic Measurement of Glycoprotein and Supramolecular Phospholipid Biomarkers of Inflammation in Human Serum. Anal Chem 2022; 94:1333-1341. [DOI: 10.1021/acs.analchem.1c04576] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Philipp Nitschke
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia 6150, Australia
| | - Samantha Lodge
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia 6150, Australia
| | - Torben Kimhofer
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia 6150, Australia
| | - Reika Masuda
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia 6150, Australia
| | - Sze-How Bong
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia 6150, Australia
| | - Drew Hall
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia 6150, Australia
| | - Hartmut Schäfer
- Bruker Biospin GmbH, Silberstreifen, 76287, Rheinstetten 76287, Germany
| | - Manfred Spraul
- Bruker Biospin GmbH, Silberstreifen, 76287, Rheinstetten 76287, Germany
| | - Niels Pompe
- Bruker Biospin GmbH, Silberstreifen, 76287, Rheinstetten 76287, Germany
| | - Tammo Diercks
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160, Derio 48160, Spain
| | - Ganeko Bernardo-Seisdedos
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160, Derio 48160, Spain
| | - José M. Mato
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160, Derio 48160, Spain
| | - Oscar Millet
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160, Derio 48160, Spain
| | - Daniella Susic
- School of Women’s and Children’s Health, University of New South Wales, Sydney, New South Wales 2052, Australia
- UNSW Microbiome Research Centre, St George Hospital, Kogarah, New South Wales 2217, Australia
| | - Amanda Henry
- School of Women’s and Children’s Health, University of New South Wales, Sydney, New South Wales 2052, Australia
- UNSW Microbiome Research Centre, St George Hospital, Kogarah, New South Wales 2217, Australia
| | - Emad M El-Omar
- Microbiome Research Centre, St George & Sutherland Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Elaine Holmes
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia 6150, Australia
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, U.K
| | - John C. Lindon
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, U.K
| | - Jeremy K. Nicholson
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia 6150, Australia
- Institute of Global Health Innovation Faculty of Medicine, Imperial College London, Level 1, Faculty Building, South Kensington Campus, London SW7 2NA, U.K
| | - Julien Wist
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia 6150, Australia
- Chemistry Department, Universidad del Valle, Cali 76001, Colombia
| |
Collapse
|
47
|
Nitschke P, Lodge S, Hall D, Schaefer H, Spraul M, Embade N, Millet O, Holmes E, Wist J, Nicholson JK. Direct low field J-edited diffusional proton NMR spectroscopic measurement of COVID-19 inflammatory biomarkers in human serum. Analyst 2022; 147:4213-4221. [DOI: 10.1039/d2an01097f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A JEDI NMR pulse experiment incorporating relaxation, diffusion and J-modulation peak editing was implemented at a low field (80 MHz) spectrometer system to quantify two recently discovered plasma markers of SARS-CoV-2 infection and general inflammation.
Collapse
Affiliation(s)
- Philipp Nitschke
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
| | - Samantha Lodge
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
| | - Drew Hall
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
| | - Hartmut Schaefer
- Bruker Biospin GmbH, Rudolf-Plank Strasse 23, 76275 Ettlingen, Germany
| | - Manfred Spraul
- Bruker Biospin GmbH, Rudolf-Plank Strasse 23, 76275 Ettlingen, Germany
| | - Nieves Embade
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160, Derio, Spain
| | - Oscar Millet
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160, Derio, Spain
| | - Elaine Holmes
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, UK
| | - Julien Wist
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
- Chemistry Department, Universidad del Valle, Cali 76001, Colombia
| | - Jeremy K. Nicholson
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
- Institute of Global Health Innovation, Faculty of Medicine, Imperial College London, Level 1, Faculty Building, South Kensington Campus, London, SW7 2NA, UK
| |
Collapse
|
48
|
Ali S, Nedvědová Š, Badshah G, Afridi MS, Abdullah, Dutra LM, Ali U, Faria SG, Soares FL, Rahman RU, Cançado FA, Aoyanagi MM, Freire LG, Santos AD, Barison A, Oliveira CA. NMR spectroscopy spotlighting immunogenicity induced by COVID-19 vaccination to mitigate future health concerns. CURRENT RESEARCH IN IMMUNOLOGY 2022; 3:199-214. [PMID: 36032416 PMCID: PMC9393187 DOI: 10.1016/j.crimmu.2022.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
In this review, the disease and immunogenicity affected by COVID-19 vaccination at the metabolic level are described considering the use of nuclear magnetic resonance (NMR) spectroscopy for the analysis of different biological samples. Consistently, we explain how different biomarkers can be examined in the saliva, blood plasma/serum, bronchoalveolar-lavage fluid (BALF), semen, feces, urine, cerebrospinal fluid (CSF) and breast milk. For example, the proposed approach for the given samples can allow one to detect molecular biomarkers that can be relevant to disease and/or vaccine interference in a system metabolome. The analysis of the given biomaterials by NMR often produces complex chemical data which can be elucidated by multivariate statistical tools, such as PCA and PLS-DA/OPLS-DA methods. Moreover, this approach may aid to improve strategies that can be helpful in disease control and treatment management in the future. NMR analysis of various bio-samples can explore disease course and vaccine interaction. Immunogenicity and reactogenicity caused by COVID-19 vaccination can be studied by NMR. Vaccine interaction alters metabolic pathway(s) at a certain stage, and this mechanism can be probed at the metabolic level.
Collapse
|
49
|
Zamora-Mendoza BN, Díaz de León-Martínez L, Rodríguez-Aguilar M, Mizaikoff B, Flores-Ramírez R. Chemometric analysis of the global pattern of volatile organic compounds in the exhaled breath of patients with COVID-19, post-COVID and healthy subjects. Proof of concept for post-COVID assessment. Talanta 2022; 236:122832. [PMID: 34635222 PMCID: PMC8411592 DOI: 10.1016/j.talanta.2021.122832] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/21/2021] [Accepted: 08/28/2021] [Indexed: 11/04/2022]
Abstract
The objective of this research was to evaluate the application of an electronic nose and chemometric analysis to discriminate volatile organic compounds between patients with COVID-19, post-COVID syndrome and controls in exhaled breath samples. A cross-sectional study was performed on 102 exhaled breath samples, 42 with COVID-19, 30 with the post-COVID syndrome and 30 control subjects. Breath-print analysis was performed by the Cyranose 320 electronic nose with 32 sensors. Group data were evaluated by Principal Component Analysis (PCA), Canonical Discriminant Analysis (CDA), and Support Vector Machine (SVM), and the test's diagnostic power was evaluated through a Receiver Operaring Characteristic curve(ROC curve). The results of the chemometric analysis indicate in the PCA a 97.6% (PC1 = 95.9%, PC2 = 1.0%, PC3 = 0.7%) of explanation of the variability between the groups by means of 3 PCs, the CDA presents a 100% of correct classification of the study groups, SVM a 99.4% of correct classification, finally the PLS-DA indicates an observable separation between the groups and the 12 sensors that were related. The sensitivity, specificity of post-COVID vs. controls value reached 97.6% (87.4%–99.9%) and 100% (88.4%–100%) respectively, according to the ROC curve. As a perspective, we consider that this technology, due to its simplicity, low cost and portability, can support strategies for the identification and follow-up of post-COVID patients. The proposed classification model provides the basis for evaluating post-COVID patients; therefore, further studies are required to enable the implementation of this technology to support clinical management and mitigation of effects.
Collapse
Affiliation(s)
- Blanca Nohemí Zamora-Mendoza
- Faculty of Medicine-Center for Applied Research on Environment and Health (CIAAS), Autonomous University of San Luis Potosí, Avenida Sierra Leona No. 550, Colonia Lomas Segunda Sección, CP, 78210, San Luis Potosí, SLP, Mexico
| | - Lorena Díaz de León-Martínez
- Faculty of Medicine-Center for Applied Research on Environment and Health (CIAAS), Autonomous University of San Luis Potosí, Avenida Sierra Leona No. 550, Colonia Lomas Segunda Sección, CP, 78210, San Luis Potosí, SLP, Mexico.
| | | | - Boris Mizaikoff
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany; Hahn-Schickard Institute for Microanalysis Systems, Sedanstrasse 14, 89077, Ulm, Germany
| | - Rogelio Flores-Ramírez
- CONACYT Research Fellow, Coordination for Innovation and Application of Science and Technology (CIACYT), Autonomous University of San Luis Potosí, Avenida Sierra Leona No. 550, CP, 78210, Colonia Lomas Segunda Sección, San Luis Potosí, SLP, Mexico.
| |
Collapse
|
50
|
Stoian AP, Kempler P, Stulnig T, Rizvi AA, Rizzo M. Diabetes and COVID-19: What 2 Years of the Pandemic Has Taught Us. Metab Syndr Relat Disord 2021; 20:137-140. [PMID: 34967689 DOI: 10.1089/met.2021.0133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
As the world enters its third year of the COVID-19 pandemic, individuals with diabetes have faced particular challenges from the virus. A deleterious bidirectional relationship exists between the two disorders, with heightened inflammatory, immunologic, and cellular mechanisms leading to a more severe illness and increased morbidity and mortality. Tight glucose control, though necessary, is hampered by physical restrictions and difficulty accessing health care. Novel glucose-lowering medications may provide unique benefits in this regard. It is imperative that multi-pronged efforts be prioritized in order to reduce adverse outcomes in patients with diabetes at risk for COVID-19.
Collapse
Affiliation(s)
- Anca Pantea Stoian
- Department of Diabetes, Nutrition and Metabolic Diseases, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,National Institute of Diabetes, Nutrition and Metabolic Diseases "Prof N.C. Paulescu," Bucharest, Romania
| | - Peter Kempler
- Department of Medicine and Oncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Thomas Stulnig
- Department of Medicine III and Karl Landsteiner institute for Metabolic Diseases and Nephrology, Clinic Hietzing, Vienna Healthcare Group, Vienna, Austria
| | - Ali A Rizvi
- Department of Medicine, University of Central Florida College of Medicine, Orlando, Florida, USA.,Division of Endocrinology, Diabetes and Metabolism, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Manfredi Rizzo
- Department of Diabetes, Nutrition and Metabolic Diseases, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,Division of Endocrinology, Diabetes and Metabolism, University of South Carolina School of Medicine, Columbia, South Carolina, USA.,Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (Promise), University of Palermo, Palermo, Italy
| |
Collapse
|