1
|
Emwas AH, Zacharias HU, Alborghetti MR, Gowda GAN, Raftery D, McKay RT, Chang CK, Saccenti E, Gronwald W, Schuchardt S, Leiminger R, Merzaban J, Madhoun NY, Iqbal M, Alsiary RA, Shivapurkar R, Pain A, Shanmugam D, Ryan D, Roy R, Schirra HJ, Morris V, Zeri AC, Alahmari F, Kaddurah-Daouk R, Salek RM, LeVatte M, Berjanskii M, Lee B, Wishart DS. Recommendations for sample selection, collection and preparation for NMR-based metabolomics studies of blood. Metabolomics 2025; 21:66. [PMID: 40348843 PMCID: PMC12065766 DOI: 10.1007/s11306-025-02259-7] [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: 02/12/2025] [Accepted: 04/04/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Metabolic profiling of blood metabolites, particularly in plasma and serum, is vital for studying human diseases, human conditions, drug interventions and toxicology. The clinical significance of blood arises from its close ties to all human cells and facile accessibility. However, patient-specific variables such as age, sex, diet, lifestyle and health status, along with pre-analytical conditions (sample handling, storage, etc.), can significantly affect metabolomic measurements in whole blood, plasma, or serum studies. These factors, referred to as confounders, must be mitigated to reveal genuine metabolic changes due to illness or intervention onset. REVIEW OBJECTIVE This review aims to aid metabolomics researchers in collecting reliable, standardized datasets for NMR-based blood (whole/serum/plasma) metabolomics. The goal is to reduce the impact of confounding factors and enhance inter-laboratory comparability, enabling more meaningful outcomes in metabolomics studies. KEY CONCEPTS This review outlines the main factors affecting blood metabolite levels and offers practical suggestions for what to measure and expect, how to mitigate confounding factors, how to properly prepare, handle and store blood, plasma and serum biosamples and how to report data in targeted NMR-based metabolomics studies of blood, plasma and serum.
Collapse
Affiliation(s)
- Abdul-Hamid Emwas
- King Abdullah University of Science and Technology (KAUST), Core Labs, Thuwal, 23955-6900, Kingdom of Saudi Arabia.
| | - Helena U Zacharias
- Peter L. Reichertz Institute for Medical Informatics of TU Braunschweig and Hannover Medical School, Hannover Medical School, 30625, Hannover, Germany
| | - Marcos Rodrigo Alborghetti
- Brazilian Biosciences National Laboratory and Brazilian Center for Research in Energy and Materials, Campinas, 13083-100, Brazil
| | - G A Nagana Gowda
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican St., Seattle, WA, 98109, USA
| | - Daniel Raftery
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican St., Seattle, WA, 98109, USA
| | - Ryan T McKay
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Chung-Ke Chang
- Taiwan Biobank, Biomedical Translation Research Center, Academia Sinica, Taipei City, Taiwan
| | - Edoardo Saccenti
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Wolfram Gronwald
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Sven Schuchardt
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Nikolai-Fuchs-Str. 1, 30625, Hannover, Germany
| | - Roland Leiminger
- Bruker BioSpin GmbH & Co., Rudolf-Plank-Straße 23, 76275, Ettlingen, Germany
| | - Jasmeen Merzaban
- Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Nour Y Madhoun
- Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Mazhar Iqbal
- Drug Discovery and Structural Biology, Health Biotechnology Division, National Institute for Biotechnology & Genetic Engineering (NIBGE), Faisalabad, 38000, Pakistan
| | - Rawiah A Alsiary
- King Abdullah International Medical Research Center (KAIMRC), Saudi Arabia/King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Kingdom of Saudi Arabia
| | - Rupali Shivapurkar
- Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Arnab Pain
- Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Dhanasekaran Shanmugam
- Biochemical Sciences Division, National Chemical Laboratory, Dr. Homi Bhabha Road, 411008, Pune, India
| | - Danielle Ryan
- School of Agricultural, Environmental and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia
| | - Raja Roy
- Centre of Biomedical Research, formerly, Centre of Biomedical Magnetic Resonance, Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Rae Bareli Road, Lucknow, 226014, India
| | - Horst Joachim Schirra
- School of Environment and Sciences, Griffith University, Nathan, QLD, 4111, Australia
- Institute for Biomedicine and Glycomics, Griffith University, Don Young Road, Nathan, QLD, 4111, Australia
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Vanessa Morris
- School of Biological Sciences and Biomolecular Interaction Centre, University of Canterbury, 8140, Christchurch, New Zealand
| | - Ana Carolina Zeri
- Ilum School of Science, Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Zip Code 13083-970, Brazil
| | - Fatimah Alahmari
- Department of NanoMedicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, 31441, Dammam, Saudi Arabia
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioural Sciences, Duke University, Durham, NC, USA
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Reza M Salek
- School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0SP, UK
| | - Marcia LeVatte
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Mark Berjanskii
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Brian Lee
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - David S Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Xiao X, Wang Q, Chai X, Zhang X, Jiang B, Liu M. Using neural networks to obtain NMR spectra of both small and macromolecules from blood samples in a single experiment. Commun Chem 2024; 7:167. [PMID: 39079950 PMCID: PMC11289489 DOI: 10.1038/s42004-024-01251-x] [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: 02/07/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
Metabolomics plays a crucial role in understanding metabolic processes within biological systems. Using specific pulse sequences, NMR-based metabolomics detects small and macromolecular metabolites that are altered in blood samples. Here we proposed a method called spectral editing neural network, which can effectively edit and separate the spectral signals of small and macromolecules in 1H NMR spectra of serum and plasma based on the linewidth of the peaks. We applied the model to process the 1H NMR spectra of plasma and serum. The extracted small and macromolecular spectra were then compared with experimentally obtained relaxation-edited and diffusion-edited spectra. Correlation analysis demonstrated the quantitative capability of the model in the extracted small molecule signals from 1H NMR spectra. The principal component analysis showed that the spectra extracted by the model and those obtained by NMR spectral editing methods reveal similar group information, demonstrating the effectiveness of the model in signal extraction.
Collapse
Affiliation(s)
- Xiongjie Xiao
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Qianqian Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Xin Chai
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Xu Zhang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
- Optics Valley Laboratory, Wuhan, China
| | - Bin Jiang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Optics Valley Laboratory, Wuhan, China.
| | - Maili Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Optics Valley Laboratory, Wuhan, China.
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Yang CH, Ho YH, Tang HY, Lo CJ. NMR-Based Analysis of Plasma Lipoprotein Subclass and Lipid Composition Demonstrate the Different Dietary Effects in ApoE-Deficient Mice. Molecules 2024; 29:988. [PMID: 38474500 DOI: 10.3390/molecules29050988] [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: 02/09/2024] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Plasma lipid levels are commonly measured using traditional methods such as triglycerides (TG), high-density lipoprotein (HDL), low-density lipoprotein (LDL), and cholesterol (CH). However, the use of newer technologies, such as nuclear magnetic resonance (NMR) with post-analysis platforms, has made it easier to assess lipoprotein profiles in research. In this study involving ApoE-deficient mice that were fed high-fat diets, significant changes were observed in TG, CH, free cholesterol (FC), and phospholipid (PL) levels within the LDL fraction. The varied proportions of TG in wild-type mice and CH, FC, and PL in ApoE-/- mice were strikingly different in very low-density lipoproteins (VLDL), LDL, intermediate-density lipoprotein (IDL), and HDL. This comprehensive analysis expands our understanding of lipoprotein subfractions and the impacts of the APOE protein and high-fat diet in mouse models. The new testing method allows for a complete assessment of plasma lipids and their correlation with genetic background and diet in mice.
Collapse
Affiliation(s)
- Cheng-Hung Yang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Yu-Hsuan Ho
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Hsiang-Yu Tang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Chi-Jen Lo
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan City 33302, Taiwan
- Clinical Metabolomics Core Laboratory, Chang Gung Memorial Hospital, Taoyuan City 33302, Taiwan
| |
Collapse
|
8
|
Chen X, Bertho G, Caradeuc C, Giraud N, Lucas-Torres C. Present and future of pure shift NMR in metabolomics. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2023; 61:654-673. [PMID: 37157858 DOI: 10.1002/mrc.5356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/30/2023] [Accepted: 05/04/2023] [Indexed: 05/10/2023]
Abstract
NMR is one of the most powerful techniques for the analysis of biological samples in the field of metabolomics. However, the high complexity of fluids, tissues, or other biological materials taken from living organisms is still a challenge for state-of-the-art pulse sequences, thereby limiting the detection, the identification, and the quantification of metabolites. In this context, the resolution enhancement provided by broadband homonuclear decoupling methods, which allows for simplifying 1 H multiplet patterns into singlets, has placed this so-called pure shift technique as a promising approach to perform metabolic profiling with unparalleled level of detail. In recent years, the many advances achieved in the design of pure shift experiments has paved the way to the analysis of a wide range of biological samples with ultra-high resolution. This review leads the reader from the early days of the main pure shift methods that have been successfully developed over the last decades to address complex samples, to the most recent and promising applications of pure shift NMR to the field of NMR-based metabolomics.
Collapse
Affiliation(s)
- Xi Chen
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Paris, France
| | - Gildas Bertho
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Paris, France
| | - Cédric Caradeuc
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Paris, France
| | - Nicolas Giraud
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Paris, France
| | - Covadonga Lucas-Torres
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Paris, France
| |
Collapse
|
9
|
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
|
10
|
Montagnani M, Bottalico L, Potenza MA, Charitos IA, Topi S, Colella M, Santacroce L. The Crosstalk between Gut Microbiota and Nervous System: A Bidirectional Interaction between Microorganisms and Metabolome. Int J Mol Sci 2023; 24:10322. [PMID: 37373470 DOI: 10.3390/ijms241210322] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Several studies have shown that the gut microbiota influences behavior and, in turn, changes in the immune system associated with symptoms of depression or anxiety disorder may be mirrored by corresponding changes in the gut microbiota. Although the composition/function of the intestinal microbiota appears to affect the central nervous system (CNS) activities through multiple mechanisms, accurate epidemiological evidence that clearly explains the connection between the CNS pathology and the intestinal dysbiosis is not yet available. The enteric nervous system (ENS) is a separate branch of the autonomic nervous system (ANS) and the largest part of the peripheral nervous system (PNS). It is composed of a vast and complex network of neurons which communicate via several neuromodulators and neurotransmitters, like those found in the CNS. Interestingly, despite its tight connections to both the PNS and ANS, the ENS is also capable of some independent activities. This concept, together with the suggested role played by intestinal microorganisms and the metabolome in the onset and progression of CNS neurological (neurodegenerative, autoimmune) and psychopathological (depression, anxiety disorders, autism) diseases, explains the large number of investigations exploring the functional role and the physiopathological implications of the gut microbiota/brain axis.
Collapse
Affiliation(s)
- Monica Montagnani
- Department of Precision and Regenerative Medicine and Ionian Area-Section of Pharmacology, School of Medicine, University of Bari "Aldo Moro", Policlinico University Hospital of Bari, Piazza G. Cesare 11, 70124 Bari, Italy
| | - Lucrezia Bottalico
- School of Technical Medical Sciences, "Alexander Xhuvani" University of Elbasan, 3001-3006 Elbasan, Albania
| | - Maria Assunta Potenza
- Department of Precision and Regenerative Medicine and Ionian Area-Section of Pharmacology, School of Medicine, University of Bari "Aldo Moro", Policlinico University Hospital of Bari, Piazza G. Cesare 11, 70124 Bari, Italy
| | - Ioannis Alexandros Charitos
- Pneumology and Respiratory Rehabilitation Division, Maugeri Clinical Scientific Research Institutes (IRCCS), 70124 Bari, Italy
| | - Skender Topi
- School of Technical Medical Sciences, "Alexander Xhuvani" University of Elbasan, 3001-3006 Elbasan, Albania
| | - Marica Colella
- Interdisciplinary Department of Medicine, Microbiology and Virology Unit, School of Medicine, University of Bari "Aldo Moro", Piazza G. Cesare, 11, 70124 Bari, Italy
| | - Luigi Santacroce
- Interdisciplinary Department of Medicine, Microbiology and Virology Unit, School of Medicine, University of Bari "Aldo Moro", Piazza G. Cesare, 11, 70124 Bari, Italy
| |
Collapse
|
11
|
Goshina A, Matyushenko V, Mezhenskaya D, Rak A, Katelnikova A, Gusev D, Rudenko L, Isakova-Sivak I. RDE Treatment Prevents Non-Specific Detection of SARS-CoV-2- and Influenza-Specific IgG Antibodies in Heat-Inactivated Serum Samples. Antibodies (Basel) 2023; 12:39. [PMID: 37366655 PMCID: PMC10295076 DOI: 10.3390/antib12020039] [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: 05/01/2023] [Revised: 05/28/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
Assessing the levels of serum IgG antibodies is widely used to measure immunity to influenza and the new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) after natural infection or vaccination with specific vaccines, as well as to study immune responses to these viruses in animal models. For safety reasons, sometimes serum specimens collected from infected individuals are subjected to heat inactivation at 56 °C to reduce the risk of infecting personnel during serological studies. However, this procedure may affect the level of virus-specific antibodies, making the results of antibody immunoassays uninterpretable. Here, we evaluated the effect of the heat inactivation of human, ferret and hamster serum samples on the binding of IgG antibodies to the influenza and SARS-CoV-2 antigens. For this, serum samples of naive and immune hosts were analyzed in three variants: (i) untreated sera, (ii) heated at 56 °C for 1 h, and (iii) treated with receptor-destroying enzyme (RDE). The samples were studied through an in-house enzyme-linked immunosorbent assay (ELISA) using whole influenza virus or recombinant proteins corresponding to nucleocapsid (N) protein and the receptor-binding domain of SARS-CoV-2 Spike (RBD) as antigens. We demonstrated that the heat inactivation of the naive serum samples of various hosts can lead to false-positive results, while RDE treatment abolished the effect of the non-specific binding of IgG antibodies to the viral antigens. Furthermore, RDE also significantly decreased the level of virus-specific IgG antibodies in SARS-CoV-2 and influenza-immune sera of humans and animals, although it is unknown whether it actually removes true virus-specific IgG antibodies or only non-specifically binding artifacts. Nevertheless, we suggest that the RDE treatment of human and animal sera may be useful in preventing false-positive results in various immunoassays, while also neutralizing infectious virus, since the standard protocol for the use of RDE also includes heating the sample at 56 °C.
Collapse
Affiliation(s)
- Arina Goshina
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (A.G.); (V.M.); (D.M.); (A.R.)
| | - Victoria Matyushenko
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (A.G.); (V.M.); (D.M.); (A.R.)
| | - Daria Mezhenskaya
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (A.G.); (V.M.); (D.M.); (A.R.)
| | - Alexandra Rak
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (A.G.); (V.M.); (D.M.); (A.R.)
| | - Anastasia Katelnikova
- Department of Toxicology and Microbiology, Institute of Preclinical Research Ltd., 188663 Saint Petersburg, Russia;
| | - Denis Gusev
- Botkin Infectious Diseases Hospital, Piskarovskiy Ave 49, 195067 Saint Petersburg, Russia
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (A.G.); (V.M.); (D.M.); (A.R.)
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (A.G.); (V.M.); (D.M.); (A.R.)
| |
Collapse
|
12
|
Del Coco L, Greco M, Inguscio A, Munir A, Danieli A, Cossa L, Musarò D, Coscia MR, Fanizzi FP, Maffia M. Blood Metabolite Profiling of Antarctic Expedition Members: An 1H NMR Spectroscopy-Based Study. Int J Mol Sci 2023; 24:ijms24098459. [PMID: 37176166 PMCID: PMC10179003 DOI: 10.3390/ijms24098459] [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: 03/28/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Serum samples from eight participants during the XV winter-over at Concordia base (Antarctic expedition) collected at defined time points, including predeparture, constituted the key substrates for a specific metabolomics study. To ascertain acute changes and chronic adaptation to hypoxia, the metabolic profiles of the serum samples were analyzed using NMR spectroscopy, with principal components analysis (PCA) followed by partial least squares and orthogonal partial least squares discriminant analyses (PLS-DA and OPLS-DA) used as supervised classification methods. Multivariate data analyses clearly highlighted an adaptation period characterized by an increase in the levels of circulating glutamine and lipids, mobilized to supply the body energy needs. At the same time, a reduction in the circulating levels of glutamate and N-acetyl glycoproteins, stress condition indicators, and proinflammatory markers were also found in the NMR data investigation. Subsequent pathway analysis showed possible perturbations in metabolic processes, potentially related to the physiological adaptation, predominantly found by comparing the baseline (at sea level, before mission onset), the base arrival, and the mission ending collected values.
Collapse
Affiliation(s)
- Laura Del Coco
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| | - Marco Greco
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| | - Alessandra Inguscio
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| | - Anas Munir
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
- Department of Mathematics and Physics "E. De Giorgi", University of Salento, Via Lecce-Arnesano, 73100 Lecce, Italy
| | - Antonio Danieli
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| | - Luca Cossa
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| | - Debora Musarò
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| | - Maria Rosaria Coscia
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Via P. Castellino 111, 80131 Naples, Italy
| | - Francesco Paolo Fanizzi
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| | - Michele Maffia
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| |
Collapse
|
13
|
Kalbitzer T, Lobenhofer K, Martin S, Beck Erlach M, Kremer W, Kalbitzer HR. NMR derived changes of lipoprotein particle concentrations related to impaired fasting glucose, impaired glucose tolerance, or manifest type 2 diabetes mellitus. Lipids Health Dis 2023; 22:42. [PMID: 36964528 PMCID: PMC10037821 DOI: 10.1186/s12944-023-01801-7] [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: 12/02/2022] [Accepted: 03/06/2023] [Indexed: 03/26/2023] Open
Abstract
Background Type 2 diabetes mellitus (T2D) and corresponding borderline states, impaired fasting glucose (IFG) and/or glucose tolerance (IGT), are associated with dyslipoproteinemia. It is important to distinguish between factors that cause T2D and that are the direct result of T2D. Methods The lipoprotein subclass patterns of blood donors with IFG, IGT, with IFG combined with IGT, and T2D are analyzed by nuclear magnetic resonance (NMR) spectroscopy. The development of lipoprotein patterns with time is investigated by using samples retained for an average period of 6 years. In total 595 blood donors are classified by oral glucose tolerance test (oGTT) and their glycosylated hemoglobin (HbA1c) concentrations. Concentrations of lipoprotein particles of 15 different subclasses are analyzed in the 10,921 NMR spectra recorded under fasting and non-fasting conditions. The subjects are assumed healthy according to the strict regulations for blood donors before performing the oGTT. Results Under fasting conditions manifest T2D exhibits a significant concentration increase of the smallest HDL particles (HDL A) combined with a decrease in all other HDL subclasses. In contrast to other studies reviewed in this paper, a general concentration decrease of all LDL particles is observed that is most prominent for the smallest LDL particles (LDL A). Under normal nutritional conditions a large, significant increase of the concentrations of VLDL and chylomicrons is observed for all groups with IFG and/or IGT and most prominently for manifest T2D. As we show it is possible to obtain an estimate of the concentrations of the apolipoproteins Apo-A1, Apo-B100, and Apo-B48 from the NMR data. In the actual study cohort, under fasting conditions the concentrations of the lipoproteins are not increased significantly in T2D, under non-fasting conditions only Apo-B48 increases significantly. Conclusion In contrast to other studies, in our cohort of “healthy” blood donors the T2D associated dyslipoproteinemia does not change the total concentrations of the lipoprotein particles produced in the liver under fasting and non-fasting conditions significantly but only their subclass distributions. Compared to the control group, under non-fasting conditions participants with IGT and IFG or T2D show a substantial increase of plasma concentrations of those lipoproteins that are produced in the intestinal tract. The intestinal insulin resistance becomes strongly observable.
Collapse
Affiliation(s)
- Tina Kalbitzer
- grid.7727.50000 0001 2190 5763Institute of Biophysics and Physical Biochemistry and Centre of Magnetic Resonance in Chemistry and Biomedicine, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| | - Kristina Lobenhofer
- grid.7727.50000 0001 2190 5763Institute of Biophysics and Physical Biochemistry and Centre of Magnetic Resonance in Chemistry and Biomedicine, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| | - Silke Martin
- Blutspendedienst des Bayerischen Roten Kreuzes Gemeinnützige GmbH, Herzog-Heinrich-Straße 2, 80336 Munich, Germany
| | - Markus Beck Erlach
- grid.7727.50000 0001 2190 5763Institute of Biophysics and Physical Biochemistry and Centre of Magnetic Resonance in Chemistry and Biomedicine, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| | - Werner Kremer
- grid.7727.50000 0001 2190 5763Institute of Biophysics and Physical Biochemistry and Centre of Magnetic Resonance in Chemistry and Biomedicine, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| | - Hans Robert Kalbitzer
- grid.7727.50000 0001 2190 5763Institute of Biophysics and Physical Biochemistry and Centre of Magnetic Resonance in Chemistry and Biomedicine, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| |
Collapse
|
14
|
Tian M, Liu H, Chen S, Yang Z, Tao W, Peng S, Che H, Jin L. 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
|
15
|
Ruffieux H, Hanson AL, Lodge S, Lawler NG, Whiley L, Gray N, Nolan TH, Bergamaschi L, Mescia F, Turner L, de Sa A, Pelly VS, Kotagiri P, Kingston N, Bradley JR, Holmes E, Wist J, Nicholson JK, Lyons PA, Smith KGC, Richardson S, Bantug GR, Hess C. A patient-centric modeling framework captures recovery from SARS-CoV-2 infection. Nat Immunol 2023; 24:349-358. [PMID: 36717723 PMCID: PMC9892000 DOI: 10.1038/s41590-022-01380-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 11/03/2022] [Indexed: 02/01/2023]
Abstract
The biology driving individual patient responses to severe acute respiratory syndrome coronavirus 2 infection remains ill understood. Here, we developed a patient-centric framework leveraging detailed longitudinal phenotyping data and covering a year after disease onset, from 215 infected individuals with differing disease severities. Our analyses revealed distinct 'systemic recovery' profiles, with specific progression and resolution of the inflammatory, immune cell, metabolic and clinical responses. In particular, we found a strong inter-patient and intra-patient temporal covariation of innate immune cell numbers, kynurenine metabolites and lipid metabolites, which highlighted candidate immunologic and metabolic pathways influencing the restoration of homeostasis, the risk of death and that of long COVID. Based on these data, we identified a composite signature predictive of systemic recovery, using a joint model on cellular and molecular parameters measured soon after disease onset. New predictions can be generated using the online tool http://shiny.mrc-bsu.cam.ac.uk/apps/covid-19-systemic-recovery-prediction-app , designed to test our findings prospectively.
Collapse
Affiliation(s)
- Hélène Ruffieux
- MRC Biostatistics Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
| | - Aimee L Hanson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Samantha Lodge
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Nathan G Lawler
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Luke Whiley
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| | - Nicola Gray
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Tui H Nolan
- MRC Biostatistics Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Laura Bergamaschi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Federica Mescia
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Lorinda Turner
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Aloka de Sa
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Victoria S Pelly
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Prasanti Kotagiri
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Nathalie Kingston
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - John R Bradley
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Elaine Holmes
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Julien Wist
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Chemistry Department, Universidad del Valle, Cali, Colombia
| | - Jeremy K Nicholson
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Institute of Global Health Innovation, Imperial College London, London, UK
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Sylvia Richardson
- MRC Biostatistics Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Glenn R Bantug
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Botnar Research Centre for Child Health (BRCCH) University Basel & ETH Zurich, Basel, Switzerland
| | - Christoph Hess
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland.
- Botnar Research Centre for Child Health (BRCCH) University Basel & ETH Zurich, Basel, Switzerland.
| |
Collapse
|
16
|
Chang KH, Cheng ML, Lo CJ, Fan CM, Wu YR, Chen CM. Alternations of Lipoprotein Profiles in the Plasma as Biomarkers of Huntington's Disease. Cells 2023; 12:cells12030385. [PMID: 36766727 PMCID: PMC9913722 DOI: 10.3390/cells12030385] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Alterations in lipid composition and disturbed lipoprotein metabolism are involved in the pathomechanism of Huntington's disease (HD). Here, we measured 112 lipoprotein subfractions and components in the plasma of 20 normal controls, 24 symptomatic (sympHD) and 9 presymptomatic (preHD) HD patients. Significant changes were found in 30 lipoprotein subfractions and components in all HD patients. Plasma levels of total cholesterol (CH), apolipoprotein (Apo)B, ApoB-particle number (PN), and components of low-density lipoprotein (LDL) were lower in preHD and sympHD patients. Components of LDL4, LDL5, LDL6 and high-density lipoprotein (HDL)4 demonstrated lower levels in preHD and sympHD patients compared with controls. Components in LDL3 displayed lower levels in sympHD compared with the controls, whereas components in very low-density lipoprotein (VLDL)5 were higher in sympHD patients compared to the controls. The levels of components in HDL4 and VLDL5 demonstrated correlation with the scores of motor assessment, independence scale or functional capacity of Unified Huntington's Disease Rating Scale. These findings indicate the potential of components of VLDL5, LDL3, LDL4, LDL5 and HDL4 to serve as the biomarkers for HD diagnosis and disease progression, and demonstrate substantial evidence of the involvement of lipids and apolipoproteins in HD pathogenesis.
Collapse
Affiliation(s)
- Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan 333, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Mei-Ling Cheng
- Department of Biomedical Sciences, Chang Gung University, Taoyuan 333, Taiwan
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan
- Clinical Metabolomics Core Laboratory, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Chi-Jen Lo
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan
| | - Chun-Ming Fan
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan 333, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan 333, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Correspondence: ; Tel.: +886-3-3281200 (ext. 8729); Fax: +886-3-3288849
| |
Collapse
|
17
|
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
|
18
|
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: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
19
|
Debik J, Isaksen SH, Strømmen M, Spraul M, Schäfer H, Bathen TF, Giskeødegård GF. Effect of Delayed Centrifugation on the Levels of NMR-Measured Lipoproteins and Metabolites in Plasma and Serum Samples. Anal Chem 2022; 94:17003-17010. [PMID: 36454175 DOI: 10.1021/acs.analchem.2c02167] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Metabolic profiling is widely used for large-scale association studies, based on biobank material. The main obstacle to the translation of metabolomic findings into clinical application is the lack of standardization, making validation in independent cohorts challenging. One reason for this is sensitivity of metabolites to preanalytical conditions. We present a systematic investigation of the effect of delayed centrifugation on the levels of NMR-measured metabolites and lipoproteins in serum and plasma samples. Blood was collected from 20 anonymous donors, of which 10 were recruited from an obesity clinic. Samples were stored at room temperature until centrifugation after 30 min, 1, 2, 4, or 8 h, which is within a realistic time scenario in clinical practice. The effect of delaying centrifugation on plasma and serum metabolic concentrations, and on concentrations of lipoprotein subfractions, was investigated. Our results show that lipoproteins are only minimally affected by a delay in centrifugation while metabolite levels are more sensitive to a delay. Metabolites significantly increased or decreased in concentration depending on delay duration. Further, we describe differences in the stability of serum and plasma, showing that plasma is more stable for metabolites, while lipoprotein subfractions are equally stable for both types of matrices.
Collapse
Affiliation(s)
- Julia Debik
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim 7491, Norway.,K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Sylvia Hetlelid Isaksen
- Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Magnus Strømmen
- Centre for Obesity Research, Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim 7030, Norway.,The Clinical Research Ward, Department for Research and Development, St. Olavs Hospital, Trondheim University Hospital, Trondheim 7030, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Manfred Spraul
- Bruker BioSpin AIC Division, Ettlingen, Rheinstetten 76287, Germany
| | - Hartmut Schäfer
- Bruker BioSpin AIC Division, Ettlingen, Rheinstetten 76287, Germany
| | - Tone F Bathen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim 7030, Norway
| | - Guro F Giskeødegård
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim 7491, Norway.,Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim 7030, Norway
| |
Collapse
|
20
|
Gong L, Miao Z, Zhang L, Shi B, Xiao Z, Qiu P, Liu M, Zou W. Pharmacokinetics and pharmacodynamics of bioactive compounds in Penyanqing preparation in THP-1 inflammatory cells induced by Lipopolysaccharide. BMC Complement Med Ther 2022; 22:323. [PMID: 36474249 PMCID: PMC9727977 DOI: 10.1186/s12906-022-03784-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Penyanqing (PYQ), a traditional Chinese medicine (TCM), has a good clinical efficacy for the treatment of pelvic inflammatory disease (PID). Previously, researches on its anti-inflammatory effect and mechanism in vitro, in silico, and in vivo have been reported by our team. However, the interrelationship between the anti-inflammatory activity and the active compounds in PYQ are not clear. Here, the pharmacokinetics-pharmacodynamics (PK-PD) study was carried out for more proper clinical use. METHODS The plasma concentrations of salvianolic acid B (SAB), protocatechualdehyde (PRO), paeoniflorin (PE), astilbin (AST), ferulic acid (FE), and chlorogenic acid (CH) in SD rats after PYQ administration were determined by a selective and rapid HPLC-MS/MS method. In addition, the PK-PD on cell model was used to explore the relationship between the plasma concentration and inflammatory biomarkers (TNF-α, IL-1β). RESULTS The results of this study showed that the six components could reach the peak blood concentration within 0.29 h, indicating the rapid absorption of it. The eliminations of AST, CH, FE, PE, and PRO were relatively fast due to their mean residence times (MRTs) within 3 h, while the elimination of SAB was slower (MRT 5.67 ± 0.66 h). Combined with a THP-1 cell model, there was a significant correlation between inflammatory factors and component plasma concentrations with correlation coefficients in the range of -0.9--0.746. Correspondingly, the drug-containing plasma obtained at 0.25 h point exhibited the best inhibition effect on production of IL-1β and TNF-α in LPS-induced THP-1 cells. CONCLUSION The six main components in PYQ could be quickly absorbed, and there was a potential good correlation between their pharmacokinetics and the pharmacodynamics of PYQ.
Collapse
Affiliation(s)
- Linna Gong
- grid.284723.80000 0000 8877 7471Biopharmaceutics, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Zhishuo Miao
- grid.284723.80000 0000 8877 7471Biopharmaceutics, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Li Zhang
- Changsha Research and Development Center On Obstetric and Gynecologic Traditional Chinese Medicine Preparation, NHC Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008 Hunan China
| | - Birui Shi
- grid.284723.80000 0000 8877 7471Biopharmaceutics, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Zuoqi Xiao
- Changsha Research and Development Center On Obstetric and Gynecologic Traditional Chinese Medicine Preparation, NHC Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008 Hunan China
| | - Panzi Qiu
- Changsha Research and Development Center On Obstetric and Gynecologic Traditional Chinese Medicine Preparation, NHC Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008 Hunan China
| | - Menghua Liu
- grid.284723.80000 0000 8877 7471Biopharmaceutics, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Wei Zou
- Changsha Research and Development Center On Obstetric and Gynecologic Traditional Chinese Medicine Preparation, NHC Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008 Hunan China
| |
Collapse
|
21
|
An NMR-Based Model to Investigate the Metabolic Phenoreversion of COVID-19 Patients throughout a Longitudinal Study. Metabolites 2022; 12:metabo12121206. [PMID: 36557244 PMCID: PMC9788519 DOI: 10.3390/metabo12121206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/19/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
After SARS-CoV-2 infection, the molecular phenoreversion of the immunological response and its associated metabolic dysregulation are required for a full recovery of the patient. This process is patient-dependent due to the manifold possibilities induced by virus severity, its phylogenic evolution and the vaccination status of the population. We have here investigated the natural history of COVID-19 disease at the molecular level, characterizing the metabolic and immunological phenoreversion over time in large cohorts of hospitalized severe patients (n = 886) and non-hospitalized recovered patients that self-reported having passed the disease (n = 513). Non-hospitalized recovered patients do not show any metabolic fingerprint associated with the disease or immune alterations. Acute patients are characterized by the metabolic and lipidomic dysregulation that accompanies the exacerbated immunological response, resulting in a slow recovery time with a maximum probability of around 62 days. As a manifestation of the heterogeneity in the metabolic phenoreversion, age and severity become factors that modulate their normalization time which, in turn, correlates with changes in the atherogenesis-associated chemokine MCP-1. Our results are consistent with a model where the slow metabolic normalization in acute patients results in enhanced atherosclerotic risk, in line with the recent observation of an elevated number of cardiovascular episodes found in post-COVID-19 cohorts.
Collapse
|
22
|
Assante G, Tourna A, Carpani R, Ferrari F, Prati D, Peyvandi F, Blasi F, Bandera A, Le Guennec A, Chokshi S, Patel VC, Cox IJ, Valenti L, Youngson NA. Reduced circulating FABP2 in patients with moderate to severe COVID-19 may indicate enterocyte functional change rather than cell death. Sci Rep 2022; 12:18792. [PMID: 36335131 PMCID: PMC9637119 DOI: 10.1038/s41598-022-23282-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 10/25/2022] [Indexed: 11/08/2022] Open
Abstract
The gut is of importance in the pathology of COVID-19 both as a route of infection, and gut dysfunction influencing the severity of disease. Systemic changes caused by SARS-CoV-2 gut infection include alterations in circulating levels of metabolites, nutrients and microbial products which alter immune and inflammatory responses. Circulating plasma markers for gut inflammation and damage such as zonulin, lipopolysaccharide and β-glycan increase in plasma along with severity of disease. However, Intestinal Fatty Acid Binding Protein / Fatty Acid Binding Protein 2 (I-FABP/FABP2), a widely used biomarker for gut cell death, has paradoxically been shown to be reduced in moderate to severe COVID-19. We also found this pattern in a pilot cohort of mild (n = 18) and moderately severe (n = 19) COVID-19 patients in Milan from March to June 2020. These patients were part of the first phase of COVID-19 in Europe and were therefore all unvaccinated. After exclusion of outliers, patients with more severe vs milder disease showed reduced FABP2 levels (median [IQR]) (124 [368] vs. 274 [558] pg/mL, P < 0.01). A reduction in NMR measured plasma relative lipid-CH3 levels approached significance (median [IQR]) (0.081 [0.011] vs. 0.073 [0.024], P = 0.06). Changes in circulating lipid levels are another feature commonly observed in severe COVID-19 and a weak positive correlation was observed in the more severe group between reduced FABP2 and reduced relative lipid-CH3 and lipid-CH2 levels. FABP2 is a key regulator of enterocyte lipid import, a process which is inhibited by gut SARS-CoV-2 infection. We propose that the reduced circulating FABP2 in moderate to severe COVID-19 is a marker of infected enterocyte functional change rather than gut damage, which could also contribute to the development of hypolipidemia in patients with more severe disease.
Collapse
Affiliation(s)
- G Assante
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
| | - A Tourna
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
| | - R Carpani
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - F Ferrari
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - D Prati
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - F Peyvandi
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| | - F Blasi
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| | - A Bandera
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| | - A Le Guennec
- Randall Centre for Cell & Molecular Biophysics, King's College, London, UK
| | - S Chokshi
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
| | - V C Patel
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
- Institute of Liver Studies, King's College Hospital, London, UK
| | - I J Cox
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK.
- Faculty of Life Sciences & Medicine, King's College, London, UK.
| | - L Valenti
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy.
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy.
| | - N A Youngson
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK.
- Faculty of Life Sciences & Medicine, King's College, London, UK.
| |
Collapse
|
23
|
Stability of Metabolomic Content during Sample Preparation: Blood and Brain Tissues. Metabolites 2022; 12:metabo12090811. [PMID: 36144215 PMCID: PMC9505456 DOI: 10.3390/metabo12090811] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 08/26/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Thermal and enzymatic reactions can significantly change the tissue metabolomic content during the sample preparation. In this work, we evaluated the stability of metabolites in human whole blood, serum, and rat brain, as well as in metabolomic extracts from these tissues. We measured the concentrations of 63 metabolites in brain and 52 metabolites in blood. We have shown that metabolites in the extracts from biological tissues are stable within 24 h at 4 °C. Serum and whole blood metabolomes are also rather stable, changes in metabolomic content of the whole blood homogenate become apparent only after 1–2 h of incubation at 4 °C, and become strong after 24 h. The most significant changes correspond to energy metabolites: the concentrations of ATP and ADP decrease fivefold, and the concentrations of NAD, NADH, and NADPH decrease below the detectable level. A statistically significant increase was observed for AMP, IMP, hypoxanthine, and nicotinamide. The brain tissue is much more metabolically active than human blood, and significant metabolomic changes occur already within the first several minutes during the brain harvest and sample homogenization. At a longer timescale (hours), noticeable changes were observed for all classes of compounds, including amino acids, organic acids, alcohols, amines, sugars, nitrogenous bases, nucleotides, and nucleosides.
Collapse
|
24
|
Spick M, Lewis HM, Frampas CF, Longman K, Costa C, Stewart A, Dunn-Walters D, Greener D, Evetts G, Wilde MJ, Sinclair E, Barran PE, Skene DJ, Bailey MJ. An integrated analysis and comparison of serum, saliva and sebum for COVID-19 metabolomics. Sci Rep 2022; 12:11867. [PMID: 35831456 PMCID: PMC9278322 DOI: 10.1038/s41598-022-16123-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 07/05/2022] [Indexed: 12/15/2022] Open
Abstract
The majority of metabolomics studies to date have utilised blood serum or plasma, biofluids that do not necessarily address the full range of patient pathologies. Here, correlations between serum metabolites, salivary metabolites and sebum lipids are studied for the first time. 83 COVID-19 positive and negative hospitalised participants provided blood serum alongside saliva and sebum samples for analysis by liquid chromatography mass spectrometry. Widespread alterations to serum-sebum lipid relationships were observed in COVID-19 positive participants versus negative controls. There was also a marked correlation between sebum lipids and the immunostimulatory hormone dehydroepiandrosterone sulphate in the COVID-19 positive cohort. The biofluids analysed herein were also compared in terms of their ability to differentiate COVID-19 positive participants from controls; serum performed best by multivariate analysis (sensitivity and specificity of 0.97), with the dominant changes in triglyceride and bile acid levels, concordant with other studies identifying dyslipidemia as a hallmark of COVID-19 infection. Sebum performed well (sensitivity 0.92; specificity 0.84), with saliva performing worst (sensitivity 0.78; specificity 0.83). These findings show that alterations to skin lipid profiles coincide with dyslipidaemia in serum. The work also signposts the potential for integrated biofluid analyses to provide insight into the whole-body atlas of pathophysiological conditions.
Collapse
Affiliation(s)
- Matt Spick
- Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Holly-May Lewis
- Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Cecile F Frampas
- Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, GU2 7XH, UK
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Katie Longman
- Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Catia Costa
- Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, GU2 7XH, UK
- Surrey Ion Beam Centre, University of Surrey, Guildford, GU2 7XH, UK
| | - Alexander Stewart
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Deborah Dunn-Walters
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Danni Greener
- Frimley Park Hospital, Frimley Health NHS Trust, Frimley, GU16 7UJ, UK
| | - George Evetts
- Frimley Park Hospital, Frimley Health NHS Trust, Frimley, GU16 7UJ, UK
| | - Michael J Wilde
- School of Geography, Earth and Environmental Sciences, University of Plymouth, Plymouth, PL4 8AA, UK
| | - Eleanor Sinclair
- Manchester Institute of Biotechnology, University of Manchester, Manchester, M1 7DN, UK
| | - Perdita E Barran
- Manchester Institute of Biotechnology, University of Manchester, Manchester, M1 7DN, UK
| | - Debra J Skene
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Melanie J Bailey
- Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, GU2 7XH, UK.
- Surrey Ion Beam Centre, University of Surrey, Guildford, GU2 7XH, UK.
| |
Collapse
|
25
|
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: 17] [Impact Index Per Article: 5.7] [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
|
26
|
Masuda R, Lodge S, Whiley L, Gray N, Lawler N, Nitschke P, Bong SH, Kimhofer T, Loo RL, Boughton B, Zeng AX, Hall D, Schaefer H, Spraul M, Dwivedi G, Yeap BB, Diercks T, Bernardo-Seisdedos G, Mato JM, Lindon JC, Holmes E, Millet O, Wist J, Nicholson JK. Exploration of Human Serum Lipoprotein Supramolecular Phospholipids Using Statistical Heterospectroscopy in n-Dimensions (SHY- n): Identification of Potential Cardiovascular Risk Biomarkers Related to SARS-CoV-2 Infection. Anal Chem 2022; 94:4426-4436. [PMID: 35230805 DOI: 10.1021/acs.analchem.1c05389] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SARS-CoV-2 infection causes a significant reduction in lipoprotein-bound serum phospholipids give rise to supramolecular phospholipid composite (SPC) signals observed in diffusion and relaxation edited 1H NMR spectra. To characterize the chemical structural components and compartmental location of SPC and to understand further its possible diagnostic properties, we applied a Statistical HeterospectroscopY in n-dimensions (SHY-n) approach. This involved statistically linking a series of orthogonal measurements made on the same samples, using independent analytical techniques and instruments, to identify the major individual phospholipid components giving rise to the SPC signals. Thus, an integrated model for SARS-CoV-2 positive and control adults is presented that relates three identified diagnostic subregions of the SPC signal envelope (SPC1, SPC2, and SPC3) generated using diffusion and relaxation edited (DIRE) NMR spectroscopy to lipoprotein and lipid measurements obtained by in vitro diagnostic NMR spectroscopy and ultrahigh-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). The SPC signals were then correlated sequentially with (a) total phospholipids in lipoprotein subfractions; (b) apolipoproteins B100, A1, and A2 in different lipoproteins and subcompartments; and (c) MS-measured total serum phosphatidylcholines present in the NMR detection range (i.e., PCs: 16.0,18.2; 18.0,18.1; 18.2,18.2; 16.0,18.1; 16.0,20.4; 18.0,18.2; 18.1,18.2), lysophosphatidylcholines (LPCs: 16.0 and 18.2), and sphingomyelin (SM 22.1). The SPC3/SPC2 ratio correlated strongly (r = 0.86) with the apolipoprotein B100/A1 ratio, a well-established marker of cardiovascular disease risk that is markedly elevated during acute SARS-CoV-2 infection. These data indicate the considerable potential of using a serum SPC measurement as a metric of cardiovascular risk based on a single NMR experiment. This is of specific interest in relation to understanding the potential for increased cardiovascular risk in COVID-19 patients and risk persistence in post-acute COVID-19 syndrome (PACS).
Collapse
Affiliation(s)
- Reika Masuda
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia
| | - Samantha Lodge
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia
| | - Luke Whiley
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia
| | - Nicola Gray
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia
| | - Nathan Lawler
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia
| | - Philipp Nitschke
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia
| | - Sze-How Bong
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia
| | - Torben Kimhofer
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia
| | - Ruey Leng Loo
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia
| | - Berin Boughton
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia
| | - Annie X Zeng
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia
| | - Drew Hall
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia
| | | | - Manfred Spraul
- Bruker Biospin GmbH, Silberstreifen, Ettlingen 76275, Germany
| | - Girish Dwivedi
- Department of Cardiology, Fiona Stanley Hospital, Medical School, University of Western Australia, Perth 6150, Western Australia, Australia
| | - Bu B Yeap
- Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Medical School, University of Western Australia, Perth 6150, Western Australia, Australia
| | - Tammo Diercks
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160 Derio, Spain
| | - Ganeko Bernardo-Seisdedos
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160 Derio, Spain
| | - José M Mato
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160 Derio, Spain
| | - 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
| | - Elaine Holmes
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia.,Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, U.K
| | - Oscar Millet
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Parque Tecnológico de Bizkaia, Bld. 800, 48160 Derio, Spain
| | - Julien Wist
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia.,Chemistry Department, Universidad del Valle, 76001 Cali, Colombia
| | - Jeremy K Nicholson
- Australian National Phenome Center, and Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth 6150, Western Australia, Australia.,Department of Cardiology, Fiona Stanley Hospital, Medical School, University of Western Australia, Perth 6150, Western Australia, Australia.,Institute of Global Health Innovation, Faculty of Medicine, Imperial College London, Level 1, Faculty Building, South Kensington Campus, London SW7 2NA, U.K
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
Molecular Phenomic Approaches to Deconvolving the Systemic Effects of SARS-CoV-2 Infection and Post-acute COVID-19 Syndrome. PHENOMICS 2021; 1:143-150. [PMID: 35233558 PMCID: PMC8295979 DOI: 10.1007/s43657-021-00020-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/25/2021] [Accepted: 06/17/2021] [Indexed: 11/09/2022]
Abstract
SARS COV-2 infection causes acute and frequently severe respiratory disease with associated multi-organ damage and systemic disturbances in many biochemical pathways. Metabolic phenotyping provides deep insights into the complex immunopathological problems that drive the resulting COVID-19 disease and is also a source of novel metrics for assessing patient recovery. A multiplatform metabolic phenotyping approach to studying the pathology and systemic metabolic sequelae of COVID-19 is considered here, together with a framework for assessing post-acute COVID-19 Syndrome (PACS) that is a major long-term health consequence for many patients. The sudden emergence of the disease presents a biological discovery challenge as we try to understand the pathological mechanisms of the disease and develop effective mitigation strategies. This requires technologies to measure objectively the extent and sub-phenotypes of the disease at the molecular level. Spectroscopic methods can reveal metabolic sub-phenotypes and new biomarkers that can be monitored during the acute disease phase and beyond. This approach is scalable and translatable to other pathologies and provides as an exemplar strategy for the investigation of other emergent zoonotic diseases with complex immunological drivers, multi-system involvements and diverse persistent symptoms.
Collapse
|
30
|
Masuda R, Lodge S, Nitschke P, Spraul M, Schaefer H, Bong SH, Kimhofer T, Hall D, Loo RL, Bizkarguenaga M, Bruzzone C, Gil-Redondo R, Embade N, Mato JM, Holmes E, Wist J, Millet O, Nicholson JK. Integrative Modeling of Plasma Metabolic and Lipoprotein Biomarkers of SARS-CoV-2 Infection in Spanish and Australian COVID-19 Patient Cohorts. J Proteome Res 2021; 20:4139-4152. [PMID: 34251833 DOI: 10.1021/acs.jproteome.1c00458] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Quantitative plasma lipoprotein and metabolite profiles were measured on an autonomous community of the Basque Country (Spain) cohort consisting of hospitalized COVID-19 patients (n = 72) and a matched control group (n = 75) and a Western Australian (WA) cohort consisting of (n = 17) SARS-CoV-2 positives and (n = 20) healthy controls using 600 MHz 1H nuclear magnetic resonance (NMR) spectroscopy. Spanish samples were measured in two laboratories using one-dimensional (1D) solvent-suppressed and T2-filtered methods with in vitro diagnostic quantification of lipoproteins and metabolites. SARS-CoV-2 positive patients and healthy controls from both populations were modeled and cross-projected to estimate the biological similarities and validate biomarkers. Using the top 15 most discriminatory variables enabled construction of a cross-predictive model with 100% sensitivity and specificity (within populations) and 100% sensitivity and 82% specificity (between populations). Minor differences were observed between the control metabolic variables in the two cohorts, but the lipoproteins were virtually indistinguishable. We observed highly significant infection-related reductions in high-density lipoprotein (HDL) subfraction 4 phospholipids, apolipoproteins A1 and A2,that have previously been associated with negative regulation of blood coagulation and fibrinolysis. The Spanish and Australian diagnostic SARS-CoV-2 biomarkers were mathematically and biologically equivalent, demonstrating that NMR-based technologies are suitable for the study of the comparative pathology of COVID-19 via plasma phenotyping.
Collapse
Affiliation(s)
- Reika Masuda
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Samantha Lodge
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Philipp Nitschke
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Manfred Spraul
- Bruker Biospin GmbH, Silberstreifen, Ettlingen 76275, Germany
| | | | - Sze-How Bong
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Torben Kimhofer
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Drew Hall
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Ruey Leng Loo
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Maider Bizkarguenaga
- CIC bioGUNE, Asociación Centro de Investigación Cooperativa en Biociencias, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
| | - Chiara Bruzzone
- CIC bioGUNE, Asociación Centro de Investigación Cooperativa en Biociencias, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
| | - Rubén Gil-Redondo
- CIC bioGUNE, Asociación Centro de Investigación Cooperativa en Biociencias, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
| | - Nieves Embade
- CIC bioGUNE, Asociación Centro de Investigación Cooperativa en Biociencias, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
| | - José M Mato
- CIC bioGUNE, Asociación Centro de Investigación Cooperativa en Biociencias, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
| | - Elaine Holmes
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Section for Nutrition Research, Department of Metabolism, Nutrition and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, London SW7 2AZ, U.K
| | - Julien Wist
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Chemistry Department, Universidad del Valle, 76001 Cali, Colombia
| | - Oscar Millet
- CIC bioGUNE, Asociación Centro de Investigación Cooperativa en Biociencias, Bizkaia Science and Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
| | - Jeremy K Nicholson
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia.,Institute of Global Health Innovation, Imperial College London, Level 1, Faculty Building, South Kensington Campus, London SW7 2NA, U.K
| |
Collapse
|
31
|
Holmes E, Wist J, Masuda R, Lodge S, Nitschke P, Kimhofer T, Loo RL, Begum S, Boughton B, Yang R, Morillon AC, Chin ST, Hall D, Ryan M, Bong SH, Gay M, Edgar DW, Lindon JC, Richards T, Yeap BB, Pettersson S, Spraul M, Schaefer H, Lawler NG, Gray N, Whiley L, Nicholson JK. Incomplete Systemic Recovery and Metabolic Phenoreversion in Post-Acute-Phase Nonhospitalized COVID-19 Patients: Implications for Assessment of Post-Acute COVID-19 Syndrome. J Proteome Res 2021; 20:3315-3329. [PMID: 34009992 PMCID: PMC8147448 DOI: 10.1021/acs.jproteome.1c00224] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Indexed: 12/15/2022]
Abstract
We present a multivariate metabotyping approach to assess the functional recovery of nonhospitalized COVID-19 patients and the possible biochemical sequelae of "Post-Acute COVID-19 Syndrome", colloquially known as long-COVID. Blood samples were taken from patients ca. 3 months after acute COVID-19 infection with further assessment of symptoms at 6 months. Some 57% of the patients had one or more persistent symptoms including respiratory-related symptoms like cough, dyspnea, and rhinorrhea or other nonrespiratory symptoms including chronic fatigue, anosmia, myalgia, or joint pain. Plasma samples were quantitatively analyzed for lipoproteins, glycoproteins, amino acids, biogenic amines, and tryptophan pathway intermediates using Nuclear Magnetic Resonance (NMR) spectroscopy and mass spectrometry. Metabolic data for the follow-up patients (n = 27) were compared with controls (n = 41) and hospitalized severe acute respiratory syndrome SARS-CoV-2 positive patients (n = 18, with multiple time-points). Univariate and multivariate statistics revealed variable patterns of functional recovery with many patients exhibiting residual COVID-19 biomarker signatures. Several parameters were persistently perturbed, e.g., elevated taurine (p = 3.6 × 10-3 versus controls) and reduced glutamine/glutamate ratio (p = 6.95 × 10-8 versus controls), indicative of possible liver and muscle damage and a high energy demand linked to more generalized tissue repair or immune function. Some parameters showed near-complete normalization, e.g., the plasma apolipoprotein B100/A1 ratio was similar to that of healthy controls but significantly lower (p = 4.2 × 10-3) than post-acute COVID-19 patients, reflecting partial reversion of the metabolic phenotype (phenoreversion) toward the healthy metabolic state. Plasma neopterin was normalized in all follow-up patients, indicative of a reduction in the adaptive immune activity that has been previously detected in active SARS-CoV-2 infection. Other systemic inflammatory biomarkers such as GlycA and the kynurenine/tryptophan ratio remained elevated in some, but not all, patients. Correlation analysis, principal component analysis (PCA), and orthogonal-partial least-squares discriminant analysis (O-PLS-DA) showed that the follow-up patients were, as a group, metabolically distinct from controls and partially comapped with the acute-phase patients. Significant systematic metabolic differences between asymptomatic and symptomatic follow-up patients were also observed for multiple metabolites. The overall metabolic variance of the symptomatic patients was significantly greater than that of nonsymptomatic patients for multiple parameters (χ2p = 0.014). Thus, asymptomatic follow-up patients including those with post-acute COVID-19 Syndrome displayed a spectrum of multiple persistent biochemical pathophysiology, suggesting that the metabolic phenotyping approach may be deployed for multisystem functional assessment of individual post-acute COVID-19 patients.
Collapse
Affiliation(s)
- Elaine Holmes
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
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, U.K.
| | - Julien Wist
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
- Chemistry Department, Universidad del
Valle, 76001 Cali, Colombia
| | - Reika Masuda
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Samantha Lodge
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
| | - Philipp Nitschke
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Torben Kimhofer
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
| | - Ruey Leng Loo
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
| | - Sofina Begum
- Department of Metabolism, Digestion, and Reproduction,
Faculty of Medicine, Imperial College London, Sir Alexander
Fleming Building, South Kensington, London SW7 2AZ, U.K.
| | - Berin Boughton
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
| | - Rongchang Yang
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Aude-Claire Morillon
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Sung-Tong Chin
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Drew Hall
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Monique Ryan
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Sze-How Bong
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
| | - Melvin Gay
- Bruker Pty. Ltd., Preston,
VIC 3072, Australia
| | - Dale W. Edgar
- State Adult Burn Unit, Fiona Stanley
Hospital, Murdoch, WA 6150, Australia
- Burn Injury Research Node, The University
of Notre Dame, Fremantle, WA 6160, Australia
| | - John C. Lindon
- Department of Surgery and Cancer, Faculty of
Medicine, Imperial College London, London SW7 2AZ,
U.K.
| | - Toby Richards
- Department of Surgery, Fiona Stanley Hospital, Medical
School, University of Western Australia,Harry Perkins Building,
Murdoch, Perth, WA 6150, Australia
| | - Bu B. Yeap
- Department of Endocrinology and Diabetes, Fiona
Stanley Hospital, Medical School, University of Western
Australia, Harry Perkins Building, Murdoch, Perth, WA 6150,
Australia
| | - Sven Pettersson
- Singapore National NeuroScience
Centre, Mandalay Road, Singapore 308232,
Singapore
- Lee Kong Chian School of Medicine.
Nanyang Technological University, Mandalay Road, Singapore
308232, Singapore
- Department of Life Science Centre,
Sunway University, Kuala Lumpur 47500,
Malaysia
| | | | | | - Nathan G. Lawler
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
| | - Nicola Gray
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
| | - Luke Whiley
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Perron Institute for Neurological and
Translational Science, Nedlands, WA 6009,
Australia
| | - Jeremy K. Nicholson
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, 5 Robin
Warren Drive, Perth, WA 6150, Australia
- Center for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, 5 Robin Warren Drive,
Perth, WA 6150, Australia
- Institute of Global Health Innovation,
Imperial College London, Level 1, Faculty Building, South
Kensington Campus, London SW7 2AZ, U.K.
| |
Collapse
|
32
|
Chen X, Gu M, Li T, Sun Y. Metabolite reanalysis revealed potential biomarkers for COVID-19: a potential link with immune response. Future Microbiol 2021; 16:577-588. [PMID: 33973485 PMCID: PMC8112156 DOI: 10.2217/fmb-2021-0047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: To understand the pathological progress of COVID-19 and to
explore the potential biomarkers. Background: The COVID-19 pandemic
is ongoing. There is metabolomics research about COVID-19 indicating the
rich information of metabolomics is worthy of further data mining.
Methods: We applied bioinformatics technology to reanalyze the
published metabolomics data of COVID-19. Results: Benzoate,
β-alanine and 4-chlorobenzoic acid were first reported to be used
as potential biomarkers to distinguish COVID-19 patients from healthy
individuals; taurochenodeoxycholic acid 3-sulfate, glucuronate
and N,N,N-trimethyl-alanylproline betaine TMAP are the top classifiers in
the receiver operating characteristic curve of COVID-severe and
COVID-nonsevere patients. Conclusion: These unique metabolites
suggest an underlying immunoregulatory treatment strategy for COVID-19.
Collapse
Affiliation(s)
- Xin Chen
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.,Princeton High School, Princeton, NJ 08540, USA
| | - Mingli Gu
- Department of Laboratory Diagnosis, Changhai Hospital, Navy Military Medical University, Shanghai, 200433, China
| | - Tengda Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Yi Sun
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| |
Collapse
|
33
|
Lawler NG, Gray N, Kimhofer T, Boughton B, Gay M, Yang R, Morillon AC, Chin ST, Ryan M, Begum S, Bong SH, Coudert JD, Edgar D, Raby E, Pettersson S, Richards T, Holmes E, Whiley L, Nicholson JK. Systemic Perturbations in Amine and Kynurenine Metabolism Associated with Acute SARS-CoV-2 Infection and Inflammatory Cytokine Responses. J Proteome Res 2021; 20:2796-2811. [PMID: 33724837 PMCID: PMC7986977 DOI: 10.1021/acs.jproteome.1c00052] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Indexed: 01/06/2023]
Abstract
We performed quantitative metabolic phenotyping of blood plasma in parallel with cytokine/chemokine analysis from participants who were either SARS-CoV-2 (+) (n = 10) or SARS-CoV-2 (-) (n = 49). SARS-CoV-2 positivity was associated with a unique metabolic phenotype and demonstrated a complex systemic response to infection, including severe perturbations in amino acid and kynurenine metabolic pathways. Nine metabolites were elevated in plasma and strongly associated with infection (quinolinic acid, glutamic acid, nicotinic acid, aspartic acid, neopterin, kynurenine, phenylalanine, 3-hydroxykynurenine, and taurine; p < 0.05), while four metabolites were lower in infection (tryptophan, histidine, indole-3-acetic acid, and citrulline; p < 0.05). This signature supports a systemic metabolic phenoconversion following infection, indicating possible neurotoxicity and neurological disruption (elevations of 3-hydroxykynurenine and quinolinic acid) and liver dysfunction (reduction in Fischer's ratio and elevation of taurine). Finally, we report correlations between the key metabolite changes observed in the disease with concentrations of proinflammatory cytokines and chemokines showing strong immunometabolic disorder in response to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Nathan G. Lawler
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Nicola Gray
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Torben Kimhofer
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Berin Boughton
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Melvin Gay
- Bruker Pty Ltd., Preston,
VIC 3072, Australia
| | - Rongchang Yang
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Aude-Claire Morillon
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Sung-Tong Chin
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Monique Ryan
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Sofina Begum
- Australian National Phenome Centre, 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, U.K.
| | - Sze How Bong
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
| | - Jerome D. Coudert
- Centre for Molecular Medicine & Innovative
Therapeutics, Murdoch University, Perth, WA 6150,
Australia
| | - Dale Edgar
- State Adult Burn Unit, Fiona Stanley
Hospital, Murdoch, WA 6150, Australia
- Burn Injury Research Node, The University of
Notre Dame, Fremantle, WA 6160, Australia
- Fiona Wood Foundation,
Murdoch, WA 6150, Australia
| | - Edward Raby
- Department of Microbiology, PathWest
Laboratory Medicine, Perth, WA 6009, Australia
- Department of Infectious Diseases, Fiona
Stanley Hospital, Perth, WA 6150, Australia
| | - Sven Pettersson
- Singapore National Neuro Science
Centre, Singapore Mandalay Road, Singapore 308232,
Singapore
- Lee Kong Chian School of Medicine,
Nanyang Technological University, Mandalay Road, Singapore
308232, Singapore
- Department of Life Science Centre,
Sunway University, 55100 Kuala Lumpur,
Malaysia
| | - Toby Richards
- Medical School, Faculty of Health and Medical
Sciences, University of Western Australia, Nedlands, WA 6009,
Australia
| | - Elaine Holmes
- Australian National Phenome Centre, 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, U.K.
| | - Luke Whiley
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
- Perron Institute for Neurological and
Translational Science, Nedlands, WA 6009,
Australia
| | - Jeremy K. Nicholson
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, WA 6150, Australia
- Medical School, Faculty of Health and Medical
Sciences, University of Western Australia, Nedlands, WA 6009,
Australia
- Institute of Global Health Innovation,
Imperial College London, Level 1, Faculty Building South
Kensington Campus, London SW7 2AZ, U.K.
| |
Collapse
|
34
|
Lodge S, Nitschke P, Kimhofer T, Wist J, Bong SH, Loo RL, Masuda R, Begum S, Richards T, Lindon JC, Bermel W, Reinsperger T, Schaefer H, Spraul M, Holmes E, Nicholson JK. Diffusion and Relaxation Edited Proton NMR Spectroscopy of Plasma Reveals a High-Fidelity Supramolecular Biomarker Signature of SARS-CoV-2 Infection. Anal Chem 2021; 93:3976-3986. [PMID: 33577736 PMCID: PMC7908063 DOI: 10.1021/acs.analchem.0c04952] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/28/2021] [Indexed: 12/13/2022]
Abstract
We have applied nuclear magnetic resonance spectroscopy based plasma phenotyping to reveal diagnostic molecular signatures of SARS-CoV-2 infection via combined diffusional and relaxation editing (DIRE). We compared plasma from healthy age-matched controls (n = 26) with SARS-CoV-2 negative non-hospitalized respiratory patients and hospitalized respiratory patients (n = 23 and 11 respectively) with SARS-CoV-2 rRT-PCR positive respiratory patients (n = 17, with longitudinal sampling time-points). DIRE data were modelled using principal component analysis and orthogonal projections to latent structures discriminant analysis (O-PLS-DA), with statistical cross-validation indices indicating excellent model generalization for the classification of SARS-CoV-2 positivity for all comparator groups (area under the receiver operator characteristic curve = 1). DIRE spectra show biomarker signal combinations conferred by differential concentrations of metabolites with selected molecular mobility properties. These comprise the following: (a) composite N-acetyl signals from α-1-acid glycoprotein and other glycoproteins (designated GlycA and GlycB) that were elevated in SARS-CoV-2 positive patients [p = 2.52 × 10-10 (GlycA) and 1.25 × 10-9 (GlycB) vs controls], (b) two diagnostic supramolecular phospholipid composite signals that were identified (SPC-A and SPC-B) from the -+N-(CH3)3 choline headgroups of lysophosphatidylcholines carried on plasma glycoproteins and from phospholipids in high-density lipoprotein subfractions (SPC-A) together with a phospholipid component of low-density lipoprotein (SPC-B). The integrals of the summed SPC signals (SPCtotal) were reduced in SARS-CoV-2 positive patients relative to both controls (p = 1.40 × 10-7) and SARS-CoV-2 negative patients (p = 4.52 × 10-8) but were not significantly different between controls and SARS-CoV-2 negative patients. The identity of the SPC signal components was determined using one and two dimensional diffusional, relaxation, and statistical spectroscopic experiments. The SPCtotal/GlycA ratios were also significantly different for control versus SARS-CoV-2 positive patients (p = 1.23 × 10-10) and for SARS-CoV-2 negatives versus positives (p = 1.60 × 10-9). Thus, plasma SPCtotal and SPCtotal/GlycA are proposed as sensitive molecular markers for SARS-CoV-2 positivity that could effectively augment current COVID-19 diagnostics and may have value in functional assessment of the disease recovery process in patients with long-term symptoms.
Collapse
Affiliation(s)
- Samantha Lodge
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
| | - Philipp Nitschke
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
| | - Torben Kimhofer
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
| | - Julien Wist
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
- Chemistry
Department, Universidad del Valle, Cali 76001, Colombia
| | - Sze-How Bong
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
| | - Ruey Leng Loo
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
| | - Reika Masuda
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
| | - Sofina Begum
- Australian
National Phenome Center, and Center for 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, U.K.
| | - Toby Richards
- Division
of Surgery, Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Harry Perkins Building, Murdoch, Perth WA6150, Australia
| | - 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.
| | - Wolfgang Bermel
- Bruker
Biospin GmbH, Silberstreifen, Ettlingen 76275, Germany
| | | | | | - Manfred Spraul
- Bruker
Biospin GmbH, Silberstreifen, Ettlingen 76275, Germany
| | - Elaine Holmes
- Australian
National Phenome Center, and Center for 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, U.K.
| | - Jeremy K. Nicholson
- Australian
National Phenome Center, and Center for Computational and Systems
Medicine, Health Futures Institute, Murdoch
University, Harry Perkins Building, Perth WA6150, Australia
- Division
of Surgery, Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Harry Perkins Building, Murdoch, 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.
| |
Collapse
|
35
|
Spick M, Longman K, Frampas C, Lewis H, Costa C, Walters DD, Stewart A, Wilde M, Greener D, Evetts G, Trivedi D, Barran P, Pitt A, Bailey M. Changes to the sebum lipidome upon COVID-19 infection observed via rapid sampling from the skin. EClinicalMedicine 2021; 33:100786. [PMID: 33718846 PMCID: PMC7935689 DOI: 10.1016/j.eclinm.2021.100786] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The COVID-19 pandemic has led to an unprecedented demand for testing - for diagnosis and prognosis - as well as for investigation into the impact of the disease on the host metabolism. Sebum sampling has the potential to support both needs by looking at what the virus does to us, rather than looking for the virus itself. METHODS In this pilot study, sebum samples were collected from 67 hospitalised patients (30 COVID-19 positive and 37 COVID-19 negative) by gauze swab. Lipidomics analysis was carried out using liquid chromatography mass spectrometry, identifying 998 reproducible features. Univariate and multivariate statistical analyses were applied to the resulting feature set. FINDINGS Lipid levels were depressed in COVID-19 positive participants, indicative of dyslipidemia; p-values of 0·022 and 0·015 were obtained for triglycerides and ceramides respectively, with effect sizes of 0·44 and 0·57. Partial Least Squares-Discriminant Analysis showed separation of COVID-19 positive and negative participants with sensitivity of 57% and specificity of 68%, improving to 79% and 83% respectively when controlled for confounding comorbidities. INTERPRETATION COVID-19 dysregulates many areas of metabolism; in this work we show that the skin lipidome can be added to the list. Given that samples can be provided quickly and painlessly, we conclude that sebum is worthy of future consideration for clinical sampling. FUNDING The authors acknowledge funding from the EPSRC Impact Acceleration Account for sample collection and processing, as well as EPSRC Fellowship Funding EP/R031118/1, the University of Surrey and BBSRC BB/T002212/1. Mass Spectrometry was funded under EP/P001440/1.
Collapse
Affiliation(s)
- Matt Spick
- Department of Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, United Kingdom
| | - Katherine Longman
- Department of Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, United Kingdom
| | - Cecile Frampas
- Department of Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, United Kingdom
| | - Holly Lewis
- Department of Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, United Kingdom
| | - Catia Costa
- Surrey Ion Beam Centre, University of Surrey, Guildford, United Kingdom
| | - Deborah Dunn Walters
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Alex Stewart
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | | | - Danni Greener
- Frimley Park Hospital, Frimley Health NHS Trust, United Kingdom
| | - George Evetts
- Frimley Park Hospital, Frimley Health NHS Trust, United Kingdom
| | - Drupad Trivedi
- Manchester Institute of Biotechnology, University of Manchester, United Kingdom
| | - Perdita Barran
- Manchester Institute of Biotechnology, University of Manchester, United Kingdom
| | - Andy Pitt
- Manchester Institute of Biotechnology, University of Manchester, United Kingdom
- Aston University, Birmingham, United Kingdom
| | - Melanie Bailey
- Department of Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, United Kingdom
- Surrey Ion Beam Centre, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
36
|
Lodge S, Nitschke P, Kimhofer T, Coudert JD, Begum S, Bong SH, Richards T, Edgar D, Raby E, Spraul M, Schaefer H, Lindon JC, Loo RL, Holmes E, Nicholson JK. NMR Spectroscopic Windows on the Systemic Effects of SARS-CoV-2 Infection on Plasma Lipoproteins and Metabolites in Relation to Circulating Cytokines. J Proteome Res 2021; 20:1382-1396. [PMID: 33426894 PMCID: PMC7805607 DOI: 10.1021/acs.jproteome.0c00876] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Indexed: 02/08/2023]
Abstract
To investigate the systemic metabolic effects of SARS-CoV-2 infection, we analyzed 1H NMR spectroscopic data on human blood plasma and co-modeled with multiple plasma cytokines and chemokines (measured in parallel). Thus, 600 MHz 1H solvent-suppressed single-pulse, spin-echo, and 2D J-resolved spectra were collected on plasma recorded from SARS-CoV-2 rRT-PCR-positive patients (n = 15, with multiple sampling timepoints) and age-matched healthy controls (n = 34, confirmed rRT-PCR negative), together with patients with COVID-19/influenza-like clinical symptoms who tested SARS-CoV-2 negative (n = 35). We compared the single-pulse NMR spectral data with in vitro diagnostic research (IVDr) information on quantitative lipoprotein profiles (112 parameters) extracted from the raw 1D NMR data. All NMR methods gave highly significant discrimination of SARS-CoV-2 positive patients from controls and SARS-CoV-2 negative patients with individual NMR methods, giving different diagnostic information windows on disease-induced phenoconversion. Longitudinal trajectory analysis in selected patients indicated that metabolic recovery was incomplete in individuals without detectable virus in the recovery phase. We observed four plasma cytokine clusters that expressed complex differential statistical relationships with multiple lipoproteins and metabolites. These included the following: cluster 1, comprising MIP-1β, SDF-1α, IL-22, and IL-1α, which correlated with multiple increased LDL and VLDL subfractions; cluster 2, including IL-10 and IL-17A, which was only weakly linked to the lipoprotein profile; cluster 3, which included IL-8 and MCP-1 and were inversely correlated with multiple lipoproteins. IL-18, IL-6, and IFN-γ together with IP-10 and RANTES exhibited strong positive correlations with LDL1-4 subfractions and negative correlations with multiple HDL subfractions. Collectively, these data show a distinct pattern indicative of a multilevel cellular immune response to SARS CoV-2 infection interacting with the plasma lipoproteome giving a strong and characteristic immunometabolic phenotype of the disease. We observed that some patients in the respiratory recovery phase and testing virus-free were still metabolically highly abnormal, which indicates a new role for these technologies in assessing full systemic recovery.
Collapse
Affiliation(s)
- Samantha Lodge
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
- Centre for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, Murdoch, Western
Australia 6150, Australia
| | - Philipp Nitschke
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
| | - Torben Kimhofer
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
- Centre for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, Murdoch, Western
Australia 6150, Australia
| | - Jerome D. Coudert
- Centre for Molecular Medicine and Innovative
Therapeutics, Murdoch University, Harry Perkins Building,
Perth, Western Australia 6150, Australia
- Perron Institute for Neurological and
Translational Science, Nedlands, Western Australia 6009,
Australia
- School of Medicine, University of Notre
Dame, Fremantle, Western Australia 6160,
Australia
| | - Sofina Begum
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
- Section of Nutrition Research , Department of Metabolism,
Nutrition and Reproduction, Faculty of Medicine, Sir Alexander Fleming Building,
Imperial College London, London SW7 2AZ,
U.K.
| | - Sze-How Bong
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
| | - Toby Richards
- Division of Surgery, Medical School, Faculty of Health
and Medical Sciences, University of Western Australia, Harry
Perkins Building, Robert Warren Drive, Murdoch, Perth, Western Australia 6150,
Australia
| | - Dale Edgar
- Faculty of Health and Medical Sciences,
University of Western Australia, Harry Perkins Building,
Robert Warren Drive, Murdoch, Perth, Western Australia 6150,
Australia
| | - Edward Raby
- Department of Clinical Microbiology,
PathWest Laboratory Medicine WA, Murdoch, Perth, Western
Australia 6150, Australia
| | | | | | - John C. Lindon
- Division of Systems Medicine, Department of
Metabolism, Nutrition and Reproduction, Faculty of Medicine, Sir Alexander Fleming
Building, Imperial College London, London SW7 2AZ,
U.K.
| | - Ruey Leng Loo
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
- Centre for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, Murdoch, Western
Australia 6150, Australia
| | - Elaine Holmes
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
- Centre for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, Murdoch, Western
Australia 6150, Australia
- Section of Nutrition Research , Department of Metabolism,
Nutrition and Reproduction, Faculty of Medicine, Sir Alexander Fleming Building,
Imperial College London, London SW7 2AZ,
U.K.
| | - Jeremy K. Nicholson
- Australian National Phenome Centre, Health Futures
Institute, Murdoch University, Harry Perkins Building, Perth,
Western Australia 6150, Australia
- Centre for Computational and Systems Medicine, Health
Futures Institute, Murdoch University, Murdoch, Western
Australia 6150, Australia
- Division of Surgery, Medical School, Faculty of Health
and Medical Sciences, University of Western Australia, Harry
Perkins Building, Robert Warren Drive, Murdoch, Perth, Western Australia 6150,
Australia
- Institute of Global Health Innovation,
Imperial College London, Level 1, Faculty Building South
Kensington Campus, London SW7 2NA, U.K.
| |
Collapse
|
37
|
Lodge S, Nitschke P, Loo RL, Kimhofer T, Bong SH, Richards T, Begum S, Spraul M, Schaefer H, Lindon JC, Holmes E, Nicholson JK. Low Volume in Vitro Diagnostic Proton NMR Spectroscopy of Human Blood Plasma for Lipoprotein and Metabolite Analysis: Application to SARS-CoV-2 Biomarkers. J Proteome Res 2021; 20:1415-1423. [PMID: 33491459 PMCID: PMC7857136 DOI: 10.1021/acs.jproteome.0c00815] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Indexed: 11/29/2022]
Abstract
The utility of low sample volume in vitro diagnostic (IVDr) proton nuclear magnetic resonance (1H NMR) spectroscopic experiments on blood plasma for information recovery from limited availability or high value samples was exemplified using plasma from patients with SARS-CoV-2 infection and normal controls. 1H NMR spectra were obtained using solvent-suppressed 1D, spin-echo (CPMG), and 2-dimensional J-resolved (JRES) spectroscopy using both 3 mm outer diameter SampleJet NMR tubes (100 μL plasma) and 5 mm SampleJet NMR tubes (300 μL plasma) under in vitro diagnostic conditions. We noted near identical diagnostic models in both standard and low volume IVDr lipoprotein analysis (measuring 112 lipoprotein parameters) with a comparison of the two tubes yielding R2 values ranging between 0.82 and 0.99 for the 40 paired lipoprotein parameters samples. Lipoprotein measurements for the 3 mm tubes were achieved without time penalty over the 5 mm tubes as defined by biomarker recovery for SARS-CoV-2. Overall, biomarker pattern recovery for the lipoproteins was extremely similar, but there were some small positive offsets in the linear equations for several variables due to small shimming artifacts, but there was minimal degradation of the biological information. For the standard untargeted 1D, CPMG, and JRES NMR experiments on the same samples, the reduced signal-to-noise was more constraining and required greater scanning times to achieve similar differential diagnostic performance (15 min per sample per experiment for 3 mm 1D and CPMG, compared to 4 min for the 5 mm tubes). We conclude that the 3 mm IVDr method is fit-for-purpose for quantitative lipoprotein measurements, allowing the preparation of smaller volumes for high value or limited volume samples that is common in clinical studies. If there are no analytical time constraints, the lower volume experiments are equally informative for untargeted profiling.
Collapse
Affiliation(s)
- Samantha Lodge
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Philipp Nitschke
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Ruey Leng Loo
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Torben Kimhofer
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Sze-How Bong
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Toby Richards
- Division
of Surgery, Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Harry Perkins Building, Robert Warren
Drive, Murdoch, Perth, WA 6150, Australia
| | - Sofina Begum
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Division
of Systems Medicine, Department of Metabolism, Nutrition and Reproduction,
Sir Alexander Fleming Building, Imperial
College London, London SW7 2AZ, U.K.
| | | | | | - John C. Lindon
- Division
of Systems Medicine, Department of Metabolism, Nutrition and Reproduction,
Sir Alexander Fleming Building, Imperial
College London, London SW7 2AZ, U.K.
| | - Elaine Holmes
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Division
of Systems Medicine, Department of Metabolism, Nutrition and Reproduction,
Sir Alexander Fleming Building, Imperial
College London, London SW7 2AZ, U.K.
| | - Jeremy K. Nicholson
- Australian
National Phenome Centre, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Institute
of Global Health Innovation, Imperial College
London, Level 1, Faculty Building South Kensington Campus, London SW7 2NA, U.K.
| |
Collapse
|
38
|
Affiliation(s)
- Suman S. Thakur
- Proteomics and Cell Signaling, Lab
W110, Centre for Cellular & Molecular
Biology, Habsiguda, Uppal
Road, Hyderabad 500 007, Telangana, India
| |
Collapse
|
39
|
Kimhofer T, Lodge S, Whiley L, Gray N, Loo RL, Lawler NG, Nitschke P, Bong SH, Morrison DL, Begum S, Richards T, Yeap BB, Smith C, Smith KGC, Holmes E, Nicholson JK. Integrative Modeling of Quantitative Plasma Lipoprotein, Metabolic, and Amino Acid Data Reveals a Multiorgan Pathological Signature of SARS-CoV-2 Infection. J Proteome Res 2020; 19:4442-4454. [PMID: 32806897 PMCID: PMC7489050 DOI: 10.1021/acs.jproteome.0c00519] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Indexed: 02/06/2023]
Abstract
The metabolic effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection on human blood plasma were characterized using multiplatform metabolic phenotyping with nuclear magnetic resonance (NMR) spectroscopy and liquid chromatography-mass spectrometry (LC-MS). Quantitative measurements of lipoprotein subfractions, α-1-acid glycoprotein, glucose, and biogenic amines were made on samples from symptomatic coronavirus disease 19 (COVID-19) patients who had tested positive for the SARS-CoV-2 virus (n = 17) and from age- and gender-matched controls (n = 25). Data were analyzed using an orthogonal-projections to latent structures (OPLS) method and used to construct an exceptionally strong (AUROC = 1) hybrid NMR-MS model that enabled detailed metabolic discrimination between the groups and their biochemical relationships. Key discriminant metabolites included markers of inflammation including elevated α-1-acid glycoprotein and an increased kynurenine/tryptophan ratio. There was also an abnormal lipoprotein, glucose, and amino acid signature consistent with diabetes and coronary artery disease (low total and HDL Apolipoprotein A1, low HDL triglycerides, high LDL and VLDL triglycerides), plus multiple highly significant amino acid markers of liver dysfunction (including the elevated glutamine/glutamate and Fischer's ratios) that present themselves as part of a distinct SARS-CoV-2 infection pattern. A multivariate training-test set model was validated using independent samples from additional SARS-CoV-2 positive patients and controls. The predictive model showed a sensitivity of 100% for SARS-CoV-2 positivity. The breadth of the disturbed pathways indicates a systemic signature of SARS-CoV-2 positivity that includes elements of liver dysfunction, dyslipidemia, diabetes, and coronary heart disease risk that are consistent with recent reports that COVID-19 is a systemic disease affecting multiple organs and systems. Metabolights study reference: MTBLS2014.
Collapse
Affiliation(s)
- Torben Kimhofer
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, Western Australia 6150, Australia
| | - Samantha Lodge
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, Western Australia 6150, Australia
| | - Luke Whiley
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, Western Australia 6150, Australia
- Perron Institute for Neurological and
Translational Science, Nedlands, Western Australia 6009,
Australia
| | - Nicola Gray
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, Western Australia 6150, Australia
| | - Ruey Leng Loo
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, Western Australia 6150, Australia
| | - Nathan G. Lawler
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, Western Australia 6150, Australia
| | - Philipp Nitschke
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, Western Australia 6150, Australia
| | - Sze-How Bong
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, Western Australia 6150, Australia
| | - David L. Morrison
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, Western Australia 6150, Australia
| | - Sofina Begum
- Section for Nutrition Research, Imperial
College London, Sir Alexander Fleming Building, South Kensington, London
SW7 2AZ, U.K.
| | - Toby Richards
- Medical School, Faculty of Health and Medical
Sciences, University of Western Australia, and Department of Endocrinology and Diabetes,
Fiona Stanley Hospital, Harry Perkins Building, Murdoch,
Perth, Western Australia 6150, Australia
| | - Bu B. Yeap
- Medical School, Faculty of Health and Medical
Sciences, University of Western Australia, and Department of Endocrinology and Diabetes,
Fiona Stanley Hospital, Harry Perkins Building, Murdoch,
Perth, Western Australia 6150, Australia
| | - Chris Smith
- The Cambridge Institute of Therapeutic Immunology and
Infectious Disease, Department of Medicine, University of Cambridge,
Addenbrooke’s Hospital, Cambridge CB2 0QQ,
U.K.
| | - Kenneth G. C. Smith
- The Cambridge Institute of Therapeutic Immunology and
Infectious Disease, Department of Medicine, University of Cambridge,
Addenbrooke’s Hospital, Cambridge CB2 0QQ,
U.K.
| | - Elaine Holmes
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, Western Australia 6150, Australia
- Section for Nutrition Research, Imperial
College London, Sir Alexander Fleming Building, South Kensington, London
SW7 2AZ, U.K.
| | - Jeremy K. Nicholson
- Australian National Phenome Centre, Computational and
Systems Medicine, Health Futures Institute, Murdoch University,
Harry Perkins Building, Perth, Western Australia 6150, Australia
- Medical School, Faculty of Health and Medical
Sciences, University of Western Australia, and Department of Endocrinology and Diabetes,
Fiona Stanley Hospital, Harry Perkins Building, Murdoch,
Perth, Western Australia 6150, Australia
- Institute of Global Health Innovation, Imperial
College London, Level 1, Faculty Building South Kensington Campus, London
SW7 2AZ, U.K.
| |
Collapse
|