1
|
Brito Rodrigues P, de Rezende Rodovalho V, Sencio V, Benech N, Creskey M, Silva Angulo F, Delval L, Robil C, Gosset P, Machelart A, Haas J, Descat A, Goosens JF, Beury D, Maurier F, Hot D, Wolowczuk I, Sokol H, Zhang X, Ramirez Vinolo MA, Trottein F. Integrative metagenomics and metabolomics reveal age-associated gut microbiota and metabolite alterations in a hamster model of COVID-19. Gut Microbes 2025; 17:2486511. [PMID: 40172215 PMCID: PMC11970752 DOI: 10.1080/19490976.2025.2486511] [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: 11/12/2024] [Revised: 03/08/2025] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
Aging is a key contributor of morbidity and mortality during acute viral pneumonia. The potential role of age-associated dysbiosis on disease outcomes is still elusive. In the current study, we used high-resolution shotgun metagenomics and targeted metabolomics to characterize SARS-CoV-2-associated changes in the gut microbiota from young (2-month-old) and aged (22-month-old) hamsters, a valuable model of COVID-19. We show that age-related dysfunctions in the gut microbiota are linked to disease severity and long-term sequelae in older hamsters. Our data also reveal age-specific changes in the composition and metabolic activity of the gut microbiota during both the acute phase (day 7 post-infection, D7) and the recovery phase (D22) of infection. Aged hamsters exhibited the most notable shifts in gut microbiota composition and plasma metabolic profiles. Through an integrative analysis of metagenomics, metabolomics, and clinical data, we identified significant associations between bacterial taxa, metabolites and disease markers in the aged group. On D7 (high viral load and lung epithelial damage) and D22 (body weight loss and fibrosis), numerous amino acids, amino acid-related molecules, and indole derivatives were found to correlate with disease markers. In particular, a persistent decrease in phenylalanine, tryptophan, glutamic acid, and indoleacetic acid in aged animals positively correlated with poor recovery of body weight and/or lung fibrosis by D22. In younger hamsters, several bacterial taxa (Eubacterium, Oscillospiraceae, Lawsonibacter) and plasma metabolites (carnosine and cis-aconitic acid) were associated with mild disease outcomes. These findings support the need for age-specific microbiome-targeting strategies to more effectively manage acute viral pneumonia and long-term disease outcomes.
Collapse
Affiliation(s)
- Patrícia Brito Rodrigues
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | | | - Valentin Sencio
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Nicolas Benech
- Gastroenterology Department, Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, France
- Hospices Civils de Lyon, Lyon GEM Microbiota Study Group, Lyon, France
| | - Marybeth Creskey
- Regulatory Research Division, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, University of Ottawa, Ottawa, Canada
| | - Fabiola Silva Angulo
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Lou Delval
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Cyril Robil
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Philippe Gosset
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Arnaud Machelart
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Joel Haas
- U1011-EGID, University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Amandine Descat
- EA 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées, University of Lille, CHU Lille, Lille, France
| | - Jean François Goosens
- EA 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées, University of Lille, CHU Lille, Lille, France
| | - Delphine Beury
- US 41 - UAR 2014 - PLBS, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Florence Maurier
- US 41 - UAR 2014 - PLBS, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - David Hot
- US 41 - UAR 2014 - PLBS, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Isabelle Wolowczuk
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Harry Sokol
- Gastroenterology Department, Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, France
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Xu Zhang
- Regulatory Research Division, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, University of Ottawa, Ottawa, Canada
- School of Pharmaceutical Sciences, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | | | - François Trottein
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
2
|
Agrawal R, Pal VK, K S S, Menon GJ, Singh IR, Malhotra N, C S N, Ganesh K, Rajmani RS, Narain Seshasayee AS, Chandra N, Joshi MB, Singh A. Hydrogen sulfide (H2S) coordinates redox balance, carbon metabolism, and mitochondrial bioenergetics to suppress SARS-CoV-2 infection. PLoS Pathog 2025; 21:e1013164. [PMID: 40388397 DOI: 10.1371/journal.ppat.1013164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/28/2025] [Indexed: 05/21/2025] Open
Abstract
Viruses modulate various aspects of host physiology, including carbon metabolism, redox balance, and mitochondrial bioenergetics to acquire the building blocks for replication and regulation of the immune response. Understanding how SARS-CoV-2 alters the host metabolism may lead to treatments for COVID-19. We report that a ubiquitous gaseous molecule, hydrogen sulfide (H2S), regulates redox, metabolism, and mitochondrial bioenergetics to control SARS-CoV-2. Virus replication is associated with down-regulation of the H2S-producing enzymes cystathionine-β-synthase (CBS), cystathionine-γ-lyase (CTH), and 3-mercaptopyruvate sulfurtransferase (3-MST) in multiple cell lines and nasopharyngeal swabs of symptomatic COVID-19 patients. Consequently, SARS-CoV-2-infected cells showed diminished endogenous H2S levels and a protein modification (S-sulfhydration) caused by H2S. Genetic silencing or chemical inhibition of CTH resulted in SARS-CoV-2 proliferation. Chemical supplementation of H2S using a slow-releasing H2S donor, GYY4137, diminished virus replication. Using a redox biosensor, metabolomics, transcriptomics, and XF-flux analyzer, we showed that GYY4137 blocked SARS-CoV-2 replication by inducing the Nrf2/Keap1 pathway, restoring redox balance and carbon metabolites and potentiating mitochondrial oxidative phosphorylation. Treatment of SARS-CoV-2-infected mice or hamsters with GYY4137 suppressed viral replication and ameliorated lung pathology. GYY4137 treatment reduced the expression of inflammatory cytokines and re-established the expression of Nrf2-dependent antioxidant genes in the lungs of SARS-CoV-2-infected mice. Notably, non-invasive measurement of respiratory functions using unrestrained whole-body plethysmography (uWBP) of SARS-CoV-2-infected mice showed improved pulmonary function variables, including pulmonary obstruction (Penh), end-expiratory pause (EEP), and relaxation time (RT) upon GYY4137 treatment. Together, our findings significantly extend our understanding of H2S-mediated regulation of viral infections and open new avenues for investigating the pathogenic mechanisms and therapeutic opportunities for coronavirus-associated disorders.
Collapse
Affiliation(s)
- Ragini Agrawal
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka, India
- Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, Karnataka, India
- Department of Aging Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Udupi, Karnataka, India
| | - Virender Kumar Pal
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka, India
- Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Suhas K S
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka, India
- Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Gopika Jayan Menon
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka, India
- Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Inder Raj Singh
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, Karnataka, India
| | - Nitish Malhotra
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, Karnataka, India
| | - Naren C S
- Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Kailash Ganesh
- Department of Aging Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Udupi, Karnataka, India
| | - Raju S Rajmani
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Aswin Sai Narain Seshasayee
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, Karnataka, India
| | - Nagasuma Chandra
- Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Manjunath B Joshi
- Department of Aging Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Udupi, Karnataka, India
| | - Amit Singh
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka, India
- Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, Karnataka, India
| |
Collapse
|
3
|
Li L, Haijun W, Tianyu C, Wenjing W, Zi W, Yibing C. Metabolomics and machine learning identify urine metabolic characteristics and potential biomarkers for severe Mycoplasma pneumoniae pneumonia. Sci Rep 2025; 15:17090. [PMID: 40379752 PMCID: PMC12084370 DOI: 10.1038/s41598-025-01895-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 05/08/2025] [Indexed: 05/19/2025] Open
Abstract
To study the differences in the urine metabolome between pediatric patients with severe Mycoplasma pneumoniae pneumonia (SMPP) and those with general Mycoplasma pneumoniae pneumonia (GMPP) via non-targeted metabolomics method, and potential biomarkers were explored through machine learning (ML) algorithms. The urine metabonomics data of 48 children with SMPP and 85 children with GMPP were collected via high performance liquid chromatography‒mass spectrometry (HPLC-MS/MS). The differential metabolites between the two groups were obtained via principal component analysis (PCA) and partial least squares-discriminant analysis (PLS-DA), and the significant metabolic pathways were screened via enrichment analysis. Potential biomarkers were identified using the random forest algorithm, and their relationships with clinical indicators were subsequently analyzed. A total of 136 significantly differential metabolites were identified in the urine samples from SMPP and GMPP. Of these, 68 metabolites were upregulated, and 68 were downregulated, predominantly belonging to the amino acid group. A total of 6 differential metabolic pathways were enriched, including Galactose metabolism, Pantothenate and CoA biosynthesis, Cysteine and methionine metabolism, Biotin metabolism, Glycine, serine and threonine metabolism, Arginine biosynthesis. Three significant potential biomarkers were identified through machine learning: 3-Hydroxyanthranilic acid (3-HAA), L-Kynurenine, and 16(R)-HETE. The area under the receiver operating characteristic curve (AUC) for this three-metabolite panel was 0.9142. There are great differences in the urine metabolome between SMPP and GMPP children, with multiple metabolic pathways being abnormally expressed. Three metabolites have been identified as potential biomarkers for the early detection of SMPP.
Collapse
Affiliation(s)
- Lin Li
- Department of Emergency, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Wang Haijun
- Department of Emergency, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Chen Tianyu
- JIUHE Diagnostics Co., Ltd, Zhengzhou, 450016, China
| | - Wang Wenjing
- JIUHE Diagnostics Co., Ltd, Zhengzhou, 450016, China
| | - Wang Zi
- Department of Emergency, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Cheng Yibing
- Department of Emergency, Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China.
| |
Collapse
|
4
|
Loveday EK, Welhaven H, Erdogan AE, Hain KS, Domanico LF, Chang CB, June RK, Taylor MP. Starve a cold or feed a fever? Identifying cellular metabolic changes following infection and exposure to SARS-CoV-2. PLoS One 2025; 20:e0305065. [PMID: 39937842 PMCID: PMC11819565 DOI: 10.1371/journal.pone.0305065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 12/03/2024] [Indexed: 02/14/2025] Open
Abstract
Viral infections induce major shifts in cellular metabolism elicited by active viral replication and antiviral responses. For the virus, harnessing cellular metabolism and evading changes that limit replication are essential for productive viral replication. In contrast, the cellular response to infection disrupts metabolic pathways to prevent viral replication and promote an antiviral state in the host cell and neighboring bystander cells. This competition between the virus and cell results in measurable shifts in cellular metabolism that differ depending on the virus, cell type, and extracellular environment. The resulting metabolic shifts can be observed and analyzed using global metabolic profiling techniques to identify pathways that are critical for either viral replication or cellular defense. SARS-CoV-2 is a respiratory virus that can exhibit broad tissue tropism and diverse, yet inconsistent, symptomatology. While the factors that determine the presentation and severity of SARS-CoV-2 infection remain unclear, metabolic syndromes are associated with more severe manifestations of SARS-CoV-2 disease. Despite these observations a critical knowledge gap remains between cellular metabolic responses and SARS-CoV-2 infection. Using a well-established untargeted metabolomics analysis workflow, we compared SARS-CoV-2 infection of human lung carcinoma cells. We identified significant changes in metabolic pathways that correlate with either productive or non-productive viral infection. This information is critical for characterizing the factors that contribute to SARS-CoV-2 replication that could be targeted for therapeutic interventions to limit viral disease.
Collapse
Affiliation(s)
- Emma K. Loveday
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, United States of America
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, Montana, United States of America
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, United States of America
| | - Hope Welhaven
- Chemistry and Biochemistry, Montana State University, Bozeman, Montana, United States of America
| | - Ayten Ebru Erdogan
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, Montana, United States of America
| | - Kyle S. Hain
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, United States of America
| | - Luke F. Domanico
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, United States of America
| | - Connie B. Chang
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, United States of America
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, Montana, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ronald K. June
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, Montana, United States of America
| | - Matthew P. Taylor
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, United States of America
| |
Collapse
|
5
|
Li X, Edén A, Malwade S, Cunningham JL, Bergquist J, Weidenfors JA, Sellgren CM, Engberg G, Piehl F, Gisslen M, Kumlien E, Virhammar J, Orhan F, Rostami E, Schwieler L, Erhardt S. Central and peripheral kynurenine pathway metabolites in COVID-19: Implications for neurological and immunological responses. Brain Behav Immun 2025; 124:163-176. [PMID: 39615604 DOI: 10.1016/j.bbi.2024.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/31/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024] Open
Abstract
Long-term symptoms such as pain, fatigue, and cognitive impairments are commonly observed in individuals affected by coronavirus disease 2019 (COVID-19). Metabolites of the kynurenine pathway have been proposed to account for cognitive impairment in COVID-19 patients. Here, cerebrospinal fluid (CSF) and plasma levels of kynurenine pathway metabolites in 53 COVID-19 patients and 12 non-inflammatory neurological disease controls in Sweden were measured with an ultra-performance liquid chromatography-tandem mass spectrometry system (UPLC-MS/MS) and correlated with immunological markers and neurological markers. Single cell transcriptomic data from a previous study of 130 COVID-19 patients was used to investigate the expression of key genes in the kynurenine pathway. The present study reveals that the neuroactive kynurenine pathway metabolites quinolinic acid (QUIN) and kynurenic acid (KYNA) are increased in CSF in patients with acute COVID-19. In addition, CSF levels of kynurenine, ratio of kynurenine/tryptophan (rKT) and QUIN correlate with neurodegenerative markers. Furthermore, tryptophan is significantly decreased in plasma but not in the CSF. In addition, the kynurenine pathway is strongly activated in the plasma and correlates with the peripheral immunological marker neopterin. Single-cell transcriptomics revealed upregulated gene expressions of the rate-limiting enzyme indoleamine 2,3- dioxygenase1 (IDO1) in CD14+ and CD16+ monocytes that correlated with type II-interferon response exclusively in COVID-19 patients. In summary, our study confirms significant activation of the peripheral kynurenine pathway in patients with acute COVID-19 and, notably, this is the first study to identify elevated levels of kynurenine metabolites in the central nervous system associated with the disease. Our findings suggest that peripheral inflammation, potentially linked to overexpression of IDO1 in monocytes, activates the kynurenine pathway. Increased plasma kynurenine, crossing the blood-brain barrier, serves as a source for elevated brain KYNA and neurotoxic QUIN. We conclude that blocking peripheral-to-central kynurenine transport could be a promising strategy to protect against neurotoxic effects of QUIN in COVID-19 patients.
Collapse
Affiliation(s)
- Xueqi Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Arvid Edén
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41685, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Infectious Disease, Gothenburg, 41685, Sweden
| | - Susmita Malwade
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Janet L Cunningham
- Department of Medical Science, Psychiatry, Uppsala University, Uppsala 75185, Sweden; Department of Neuroscience, Karolinska Institute, Stockholm 17177, Sweden
| | - Jonas Bergquist
- Analytical Chemistry and Neurochemistry, Department of Chemistry─BMC, Uppsala University, Box 599, 751 24 Uppsala, Sweden; The ME/CFS Collaborative Research Centre at Uppsala University, 751 24 Uppsala, Sweden
| | | | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Göran Engberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden; Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - Fredrik Piehl
- Unit of Neuroimmunology, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm 17177, Sweden; Division of Neurology, Karolinska University Hospital, Stockholm 17176, Sweden
| | - Magnus Gisslen
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41685, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Infectious Disease, Gothenburg, 41685, Sweden; Public Health Agency of Sweden, Solna, Sweden
| | - Eva Kumlien
- Department of Medical Sciences, Neurology, Uppsala University, Uppsala 75185, Sweden
| | - Johan Virhammar
- Department of Medical Sciences, Neurology, Uppsala University, Uppsala 75185, Sweden
| | - Funda Orhan
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Elham Rostami
- Department of Neuroscience, Karolinska Institute, Stockholm 17177, Sweden; Department of Medical Sciences, Neurology, Uppsala University, Uppsala 75185, Sweden
| | - Lilly Schwieler
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Sophie Erhardt
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden.
| |
Collapse
|
6
|
Zárate-Segura PB, Martínez-Castillo M, Garduño-Gutiérrez AP, Hernández-Hernández JM, Cano-Martínez LJ, García-Mena J, Coral-Vázquez RM, Bastida-González F. Changes in miRNA Pattern Expression Associated With COVID-19 Severity. In Vivo 2025; 39:482-490. [PMID: 39740904 PMCID: PMC11705121 DOI: 10.21873/invivo.13852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND/AIM Coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2 infection, manifests a wide range of clinical symptoms ranging from mild to moderate and severe. Host-related factors influence the course of SARS-CoV-2 infection; for instance, the expression of host microRNAs (miRNAs) could influence the progression and complications of COVID-19. This study aimed to determine the expression pattern of endogenous miRNAs in 80 severe COVID-19 patients compared to a group of healthy individuals. MATERIALS AND METHODS The miRNA screening expression analysis was performed using TaqMan Low-Density Array, and the expression changes of miR-490-3p, miR-195-5p, miR-454-3p, and miR-431-5p were validated using RT-qPCR. In silico analysis was used to identify new targets and predict the pathways, biological processes, and interactions of the selected miRNAs. RESULTS The miR-490-3p, miR-195-5p, miR-454-3p, and miR-431-5p, were over-expressed in the total population of severe COVID-19 patients compared to the control group. miR-490-3p was found to be over-expressed in both female and male COVID-19 patients. CONCLUSION Specific miRNAs might be a potential biomarker for predicting the clinical course of COVID-19.
Collapse
Affiliation(s)
- Paola B Zárate-Segura
- Laboratorio de Medicina Traslacional, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico;
| | - Macario Martínez-Castillo
- Laboratorio de Medicina Traslacional, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | | | - Luis Javier Cano-Martínez
- Laboratorio de Medicina Traslacional, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
- Subdirección de Enseñanza e Investigación, Centro Médico Nacional "20 de Noviembre", Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Mexico City, Mexico
| | - Jaime García-Mena
- Departamento de Genética y Biología Molecular, Cinvestav, Mexico City, Mexico
| | - Ramón M Coral-Vázquez
- Laboratorio de Medicina Traslacional, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
- Subdirección de Enseñanza e Investigación, Centro Médico Nacional "20 de Noviembre", Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Mexico City, Mexico
| | - Fernando Bastida-González
- Laboratorio de Biología Molecular, Laboratorio Estatal de Salud Pública del Estado de México, Toluca de Lerdo, Mexico
| |
Collapse
|
7
|
Said M, Ferrara BT, Aprodu A, Cabreiro F, Thompson EP, Everett J. Transcriptional analysis of C. elegans fmos at different life stages and their roles in ageing. Mol Genet Genomics 2024; 299:113. [PMID: 39636438 PMCID: PMC11621177 DOI: 10.1007/s00438-024-02201-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/22/2024] [Indexed: 12/07/2024]
Abstract
Flavin-containing monooxygenases (FMOs) are present in most organisms including plants, fungi, bacteria, invertebrates and vertebrates, where they catalyse the oxidative metabolism of a range of xenobiotics and endogenous metabolites. FMOs have been associated with ageing and longevity in the mouse and in C. elegans. As all five FMOs of C. elegans share an evolutionary root with mouse and human FMO5, it was of interest to discover if effects on ageing and longevity persisted across the whole group. We therefore investigated the impact of fmo gene knockout (KO) in C. elegans. We found that fmo-1, fmo-3 and fmo-4 KO significantly extended C. elegans lifespan relative to wild type and, as previously reported, FMO-2 over-expression did likewise. The transcription levels of C. elegans fmo genes were determined throughout the life cycle (embryo, larva and adult) in wild type and in each mutant to discover if their expression was related to stages in ageing, and expression levels were compared to those in human and mouse. In wild type worms, fmo-1 and fmo-4 were the mostly highly transcribed genes (especially at the larval stage), whereas fmo-2 and fmo-3 were the least transcribed, at all stages. Notably, the knockout of fmo-4 led to a 17- to 30-fold up-regulation of fmo-2, along with significantly increased levels of the other fmos. This parallels recent findings in the long-lived C. elegans tald-1 mutant where fmo-2 was also significantly up-regulated and reinforces its importance in lifespan extension.
Collapse
Affiliation(s)
- Mohamed Said
- Faculty of Engineering and Science, University of Greenwich, Chatham Maritime, Kent, ME4 4TB, UK
- Faculty of Pharmacy, October University for Modern Sciences and Arts, 6th October City, Egypt
| | - Bill T Ferrara
- Faculty of Engineering and Science, University of Greenwich, Chatham Maritime, Kent, ME4 4TB, UK
| | - Andreea Aprodu
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, 50931, Cologne, Germany
| | - Filipe Cabreiro
- Institute of Clinical Sciences, Imperial College London, London, W12 0NN, UK
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, 50931, Cologne, Germany
| | - Elinor P Thompson
- Faculty of Engineering and Science, University of Greenwich, Chatham Maritime, Kent, ME4 4TB, UK.
| | - Jeremy Everett
- Faculty of Engineering and Science, University of Greenwich, Chatham Maritime, Kent, ME4 4TB, UK.
| |
Collapse
|
8
|
Lopes de Lima I, Ap. Rosini Silva A, Brites C, Angelo da Silva Miyaguti N, Raposo Passos Mansoldo F, Vaz Nunes S, Henrique Godoy Sanches P, Regiani Cataldi T, Pais de Carvalho C, Reis da Silva A, Ribeiro da Rosa J, Magalhães Borges M, Vilarindo Oliveira W, Canevari TC, Beatriz Vermelho A, Nogueira Eberlin M, M. Porcari A. Mass Spectrometry-Based Metabolomics Reveals a Salivary Signature for Low-Severity COVID-19. Int J Mol Sci 2024; 25:11899. [PMID: 39595969 PMCID: PMC11593410 DOI: 10.3390/ijms252211899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/29/2024] [Accepted: 09/13/2024] [Indexed: 11/28/2024] Open
Abstract
Omics approaches were extensively applied during the coronavirus disease 2019 (COVID-19) pandemic to understand the disease, identify biomarkers with diagnostic and prognostic value, and discover new molecular targets for medications. COVID-19 continues to challenge the healthcare system as the virus mutates, becoming more transmissible or adept at evading the immune system, causing resurgent epidemic waves over the last few years. In this study, we used saliva from volunteers who were negative and positive for COVID-19 when Omicron and its variants became dominant. We applied a direct solid-phase extraction approach followed by non-target metabolomics analysis to identify potential salivary signatures of hospital-recruited volunteers to establish a model for COVID-19 screening. Our model, which aimed to differentiate COVID-19-positive individuals from controls in a hospital setting, was based on 39 compounds and achieved high sensitivity (85%/100%), specificity (82%/84%), and accuracy (84%/92%) in training and validation sets, respectively. The salivary diagnostic signatures were mainly composed of amino acids and lipids and were related to a heightened innate immune antiviral response and an attenuated inflammatory profile. The higher abundance of thyrotropin-releasing hormone in the COVID-19 positive group highlighted the endocrine imbalance in low-severity disease, as first reported here, underscoring the need for further studies in this area.
Collapse
Affiliation(s)
- Iasmim Lopes de Lima
- PPGEMN, School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil; (I.L.d.L.); (C.P.d.C.); (A.R.d.S.); (M.M.B.); (T.C.C.)
- MackGraphe—Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo 01302-907, SP, Brazil
| | - Alex Ap. Rosini Silva
- MS4Life Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University—USF, Bragança Paulista 12916-900, SP, Brazil; (A.A.R.S.); (N.A.d.S.M.); (P.H.G.S.); (J.R.d.R.); (A.M.P.)
| | - Carlos Brites
- LAPI-Laboratory of Research in Infectology, University Hospital Professor Edgard Santos (HUPES), Federal University of Bahia (UFBA), Salvador 40110-060, BA, Brazil; (C.B.); (S.V.N.)
| | - Natália Angelo da Silva Miyaguti
- MS4Life Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University—USF, Bragança Paulista 12916-900, SP, Brazil; (A.A.R.S.); (N.A.d.S.M.); (P.H.G.S.); (J.R.d.R.); (A.M.P.)
| | - Felipe Raposo Passos Mansoldo
- BIOINOVAR-Biotechnology Laboratories, Biocatalysis, Bioproducts and Bioenergy, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil; (F.R.P.M.); (A.B.V.)
| | - Sara Vaz Nunes
- LAPI-Laboratory of Research in Infectology, University Hospital Professor Edgard Santos (HUPES), Federal University of Bahia (UFBA), Salvador 40110-060, BA, Brazil; (C.B.); (S.V.N.)
| | - Pedro Henrique Godoy Sanches
- MS4Life Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University—USF, Bragança Paulista 12916-900, SP, Brazil; (A.A.R.S.); (N.A.d.S.M.); (P.H.G.S.); (J.R.d.R.); (A.M.P.)
| | - Thais Regiani Cataldi
- Department of Genetics, Luiz de Queiroz College of Agriculture, University of São Paulo (USP/ESALQ), Piracicaba 13418-900, SP, Brazil;
| | - Caroline Pais de Carvalho
- PPGEMN, School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil; (I.L.d.L.); (C.P.d.C.); (A.R.d.S.); (M.M.B.); (T.C.C.)
- MackGraphe—Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo 01302-907, SP, Brazil
| | - Adriano Reis da Silva
- PPGEMN, School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil; (I.L.d.L.); (C.P.d.C.); (A.R.d.S.); (M.M.B.); (T.C.C.)
- MackGraphe—Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo 01302-907, SP, Brazil
| | - Jonas Ribeiro da Rosa
- MS4Life Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University—USF, Bragança Paulista 12916-900, SP, Brazil; (A.A.R.S.); (N.A.d.S.M.); (P.H.G.S.); (J.R.d.R.); (A.M.P.)
| | - Mariana Magalhães Borges
- PPGEMN, School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil; (I.L.d.L.); (C.P.d.C.); (A.R.d.S.); (M.M.B.); (T.C.C.)
- MackGraphe—Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo 01302-907, SP, Brazil
| | - Wellisson Vilarindo Oliveira
- PPGEMN, School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil; (I.L.d.L.); (C.P.d.C.); (A.R.d.S.); (M.M.B.); (T.C.C.)
- MackGraphe—Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo 01302-907, SP, Brazil
| | - Thiago Cruz Canevari
- PPGEMN, School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil; (I.L.d.L.); (C.P.d.C.); (A.R.d.S.); (M.M.B.); (T.C.C.)
| | - Alane Beatriz Vermelho
- BIOINOVAR-Biotechnology Laboratories, Biocatalysis, Bioproducts and Bioenergy, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil; (F.R.P.M.); (A.B.V.)
| | - Marcos Nogueira Eberlin
- PPGEMN, School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil; (I.L.d.L.); (C.P.d.C.); (A.R.d.S.); (M.M.B.); (T.C.C.)
- MackGraphe—Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian Institute, São Paulo 01302-907, SP, Brazil
| | - Andreia M. Porcari
- MS4Life Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University—USF, Bragança Paulista 12916-900, SP, Brazil; (A.A.R.S.); (N.A.d.S.M.); (P.H.G.S.); (J.R.d.R.); (A.M.P.)
| |
Collapse
|
9
|
Dehhaghi M, Heydari M, Panahi HKS, Lewin SR, Heng B, Brew BJ, Guillemin GJ. The roles of the kynurenine pathway in COVID-19 neuropathogenesis. Infection 2024; 52:2043-2059. [PMID: 38802702 PMCID: PMC11499433 DOI: 10.1007/s15010-024-02293-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the highly contagious respiratory disease Corona Virus Disease 2019 (COVID-19) that may lead to various neurological and psychological disorders that can be acute, lasting days to weeks or months and possibly longer. The latter is known as long-COVID or more recently post-acute sequelae of COVID (PASC). During acute COVID-19 infection, a strong inflammatory response, known as the cytokine storm, occurs in some patients. The levels of interferon-γ (IFN-γ), interferon-β (IFN-β), interleukin-6 (IL-6) and tumour necrosis factor-alpha (TNF-α) are particularly increased. These cytokines are known to activate the enzyme indoleamine 2,3-dioxygenase 1 (IDO-1), catalysing the first step of tryptophan (Trp) catabolism through the kynurenine pathway (KP) leading to the production of several neurotoxic and immunosuppressive metabolites. There is already data showing elevation in KP metabolites both acutely and in PASC, especially regarding cognitive impairment. Thus, it is likely that KP involvement is significant in SARS-CoV-2 pathogenesis especially neurologically.
Collapse
Affiliation(s)
- Mona Dehhaghi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Mostafa Heydari
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Hamed Kazemi Shariat Panahi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Sharon R Lewin
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Infectious Diseases, The Alfred Hospital and Monash University, Melbourne, VIC, Australia
| | - Benjamin Heng
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.
| | - Bruce J Brew
- Peter Duncan Neurosciences Unit, St. Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia.
- Faculty of Medicine and Health, School of Clinical Medicine, UNSW Sydney, NSW, Australia.
- Departments of Neurology and Immunology, St. Vincent's Hospital, Sydney, NSW, Australia.
- University of Notre Dame, Darlinghurst, Sydney, NSW, Australia.
| | - Gilles J Guillemin
- Peter Duncan Neurosciences Unit, St. Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Institut Pertanian Bogor University, Bogor, Indonesia
| |
Collapse
|
10
|
Oropeza-Valdez JJ, Padron-Manrique C, Vázquez-Jiménez A, Soberon X, Resendis-Antonio O. Exploring metabolic anomalies in COVID-19 and post-COVID-19: a machine learning approach with explainable artificial intelligence. Front Mol Biosci 2024; 11:1429281. [PMID: 39314212 PMCID: PMC11417410 DOI: 10.3389/fmolb.2024.1429281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, has led to significant challenges worldwide, including diverse clinical outcomes and prolonged post-recovery symptoms known as Long COVID or Post-COVID-19 syndrome. Emerging evidence suggests a crucial role of metabolic reprogramming in the infection's long-term consequences. This study employs a novel approach utilizing machine learning (ML) and explainable artificial intelligence (XAI) to analyze metabolic alterations in COVID-19 and Post-COVID-19 patients. Samples were taken from a cohort of 142 COVID-19, 48 Post-COVID-19, and 38 control patients, comprising 111 identified metabolites. Traditional analysis methods, like PCA and PLS-DA, were compared with ML techniques, particularly eXtreme Gradient Boosting (XGBoost) enhanced by SHAP (SHapley Additive exPlanations) values for explainability. XGBoost, combined with SHAP, outperformed traditional methods, demonstrating superior predictive performance and providing new insights into the metabolic basis of the disease's progression and aftermath. The analysis revealed metabolomic subgroups within the COVID-19 and Post-COVID-19 conditions, suggesting heterogeneous metabolic responses to the infection and its long-term impacts. Key metabolic signatures in Post-COVID-19 include taurine, glutamine, alpha-Ketoglutaric acid, and LysoPC a C16:0. This study highlights the potential of integrating ML and XAI for a fine-grained description in metabolomics research, offering a more detailed understanding of metabolic anomalies in COVID-19 and Post-COVID-19 conditions.
Collapse
Affiliation(s)
- Juan José Oropeza-Valdez
- Human Systems Biology Laboratory. Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Cristian Padron-Manrique
- Human Systems Biology Laboratory. Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Aarón Vázquez-Jiménez
- Human Systems Biology Laboratory. Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Xavier Soberon
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Colonia Chamilpa, Cuernavaca, México
| | - Osbaldo Resendis-Antonio
- Human Systems Biology Laboratory. Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Coordinación de la Investigación Científica – Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
11
|
Tsuge M, Ichihara E, Hasegawa K, Kudo K, Tanimoto Y, Nouso K, Oda N, Mitsumune S, Kimura G, Yamada H, Takata I, Mitsuhashi T, Taniguchi A, Tsukahara K, Aokage T, Hagiya H, Toyooka S, Tsukahara H, Maeda Y. Increased Oxidative Stress and Decreased Citrulline in Blood Associated with Severe Novel Coronavirus Pneumonia in Adult Patients. Int J Mol Sci 2024; 25:8370. [PMID: 39125944 PMCID: PMC11313210 DOI: 10.3390/ijms25158370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
This study investigated the correlation between oxidative stress and blood amino acids associated with nitric oxide metabolism in adult patients with coronavirus disease (COVID-19) pneumonia. Clinical data and serum samples were prospectively collected from 100 adult patients hospitalized for COVID-19 between July 2020 and August 2021. Patients with COVID-19 were categorized into three groups for analysis based on lung infiltrates, oxygen inhalation upon admission, and the initiation of oxygen therapy after admission. Blood data, oxidative stress-related biomarkers, and serum amino acid levels upon admission were compared in these groups. Patients with lung infiltrations requiring oxygen therapy upon admission or starting oxygen post-admission exhibited higher serum levels of hydroperoxides and lower levels of citrulline compared to the control group. No remarkable differences were observed in nitrite/nitrate, asymmetric dimethylarginine, and arginine levels. Serum citrulline levels correlated significantly with serum lactate dehydrogenase and C-reactive protein levels. A significant negative correlation was found between serum levels of citrulline and hydroperoxides. Levels of hydroperoxides decreased, and citrulline levels increased during the recovery period compared to admission. Patients with COVID-19 with extensive pneumonia or poor oxygenation showed increased oxidative stress and reduced citrulline levels in the blood compared to those with fewer pulmonary complications. These findings suggest that combined oxidative stress and abnormal citrulline metabolism may play a role in the pathogenesis of COVID-19 pneumonia.
Collapse
Affiliation(s)
- Mitsuru Tsuge
- Department of Pediatrics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan;
| | - Eiki Ichihara
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama 700-8558, Japan; (E.I.); (A.T.)
| | - Kou Hasegawa
- Department of General Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (K.H.); (H.H.)
| | - Kenichiro Kudo
- Department of Respiratory Medicine, National Hospital Organization Minami-Okayama Medical Center, Okayama 701-1192, Japan; (K.K.); (S.M.)
| | - Yasushi Tanimoto
- Department of Allergy and Respiratory Medicine, National Hospital Organization Minami-Okayama Medical Center, Okayama 701-0304, Japan; (Y.T.); (G.K.)
| | - Kazuhiro Nouso
- Department of Gastroenterology, Okayama City Hospital, Okayama 700-0962, Japan;
| | - Naohiro Oda
- Department of Internal Medicine, Fukuyama City Hospital, Fukuyama 721-0971, Japan; (N.O.); (I.T.)
| | - Sho Mitsumune
- Department of Respiratory Medicine, National Hospital Organization Minami-Okayama Medical Center, Okayama 701-1192, Japan; (K.K.); (S.M.)
| | - Goro Kimura
- Department of Allergy and Respiratory Medicine, National Hospital Organization Minami-Okayama Medical Center, Okayama 701-0304, Japan; (Y.T.); (G.K.)
| | - Haruto Yamada
- Department of Infectious Disease, Okayama City Hospital, Okayama 700-0962, Japan;
| | - Ichiro Takata
- Department of Internal Medicine, Fukuyama City Hospital, Fukuyama 721-0971, Japan; (N.O.); (I.T.)
| | - Toshiharu Mitsuhashi
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama 700-8558, Japan;
| | - Akihiko Taniguchi
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama 700-8558, Japan; (E.I.); (A.T.)
| | - Kohei Tsukahara
- Department of Emergency, Critical Care and Disaster Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (K.T.); (T.A.)
| | - Toshiyuki Aokage
- Department of Emergency, Critical Care and Disaster Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (K.T.); (T.A.)
| | - Hideharu Hagiya
- Department of General Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (K.H.); (H.H.)
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrine Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan;
| | - Hirokazu Tsukahara
- Department of Pediatrics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan;
| | - Yoshinobu Maeda
- Department of Hematology, Oncology and Respiratory Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan;
| |
Collapse
|
12
|
Agamah FE, Ederveen THA, Skelton M, Martin DP, Chimusa ER, ’t Hoen PAC. Network-based integrative multi-omics approach reveals biosignatures specific to COVID-19 disease phases. Front Mol Biosci 2024; 11:1393240. [PMID: 39040605 PMCID: PMC11260748 DOI: 10.3389/fmolb.2024.1393240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/22/2024] [Indexed: 07/24/2024] Open
Abstract
Background COVID-19 disease is characterized by a spectrum of disease phases (mild, moderate, and severe). Each disease phase is marked by changes in omics profiles with corresponding changes in the expression of features (biosignatures). However, integrative analysis of multiple omics data from different experiments across studies to investigate biosignatures at various disease phases is limited. Exploring an integrative multi-omics profile analysis through a network approach could be used to determine biosignatures associated with specific disease phases and enable the examination of the relationships between the biosignatures. Aim To identify and characterize biosignatures underlying various COVID-19 disease phases in an integrative multi-omics data analysis. Method We leveraged a multi-omics network-based approach to integrate transcriptomics, metabolomics, proteomics, and lipidomics data. The World Health Organization Ordinal Scale WHO Ordinal Scale was used as a disease severity reference to harmonize COVID-19 patient metadata across two studies with independent data. A unified COVID-19 knowledge graph was constructed by assembling a disease-specific interactome from the literature and databases. Disease-state specific omics-graphs were constructed by integrating multi-omics data with the unified COVID-19 knowledge graph. We expanded on the network layers of multiXrank, a random walk with restart on multilayer network algorithm, to explore disease state omics-specific graphs and perform enrichment analysis. Results Network analysis revealed the biosignatures involved in inducing chemokines and inflammatory responses as hubs in the severe and moderate disease phases. We observed distinct biosignatures between severe and moderate disease phases as compared to mild-moderate and mild-severe disease phases. Mild COVID-19 cases were characterized by a unique biosignature comprising C-C Motif Chemokine Ligand 4 (CCL4), and Interferon Regulatory Factor 1 (IRF1). Hepatocyte Growth Factor (HGF), Matrix Metallopeptidase 12 (MMP12), Interleukin 10 (IL10), Nuclear Factor Kappa B Subunit 1 (NFKB1), and suberoylcarnitine form hubs in the omics network that characterizes the moderate disease state. The severe cases were marked by biosignatures such as Signal Transducer and Activator of Transcription 1 (STAT1), Superoxide Dismutase 2 (SOD2), HGF, taurine, lysophosphatidylcholine, diacylglycerol, triglycerides, and sphingomyelin that characterize the disease state. Conclusion This study identified both biosignatures of different omics types enriched in disease-related pathways and their associated interactions (such as protein-protein, protein-transcript, protein-metabolite, transcript-metabolite, and lipid-lipid interactions) that are unique to mild, moderate, and severe COVID-19 disease states. These biosignatures include molecular features that underlie the observed clinical heterogeneity of COVID-19 and emphasize the need for disease-phase-specific treatment strategies. The approach implemented here can be used to find associations between transcripts, proteins, lipids, and metabolites in other diseases.
Collapse
Affiliation(s)
- Francis E. Agamah
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Thomas H. A. Ederveen
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | - Michelle Skelton
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Darren P. Martin
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Emile R. Chimusa
- Department of Applied Science, Faculty of Health and Life Sciences, Northumbria University, Newcastle, United Kingdom
| | - Peter A. C. ’t Hoen
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| |
Collapse
|
13
|
Abdallah AM, Doudin A, Sulaiman TO, Jamil O, Arif R, Sada FA, Yassine HM, Elrayess MA, Elzouki AN, Emara MM, Thillaiappan NB, Cyprian FS. Metabolic predictors of COVID-19 mortality and severity: a survival analysis. Front Immunol 2024; 15:1353903. [PMID: 38799469 PMCID: PMC11127595 DOI: 10.3389/fimmu.2024.1353903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction The global healthcare burden of COVID-19 pandemic has been unprecedented with a high mortality. Metabolomics, a powerful technique, has been increasingly utilized to study the host response to infections and to understand the progression of multi-system disorders such as COVID-19. Analysis of the host metabolites in response to SARS-CoV-2 infection can provide a snapshot of the endogenous metabolic landscape of the host and its role in shaping the interaction with SARS-CoV-2. Disease severity and consequently the clinical outcomes may be associated with a metabolic imbalance related to amino acids, lipids, and energy-generating pathways. Hence, the host metabolome can help predict potential clinical risks and outcomes. Methods In this prospective study, using a targeted metabolomics approach, we studied the metabolic signature in 154 COVID-19 patients (males=138, age range 48-69 yrs) and related it to disease severity and mortality. Blood plasma concentrations of metabolites were quantified through LC-MS using MxP Quant 500 kit, which has a coverage of 630 metabolites from 26 biochemical classes including distinct classes of lipids and small organic molecules. We then employed Kaplan-Meier survival analysis to investigate the correlation between various metabolic markers, disease severity and patient outcomes. Results A comparison of survival outcomes between individuals with high levels of various metabolites (amino acids, tryptophan, kynurenine, serotonin, creatine, SDMA, ADMA, 1-MH and carnitine palmitoyltransferase 1 and 2 enzymes) and those with low levels revealed statistically significant differences in survival outcomes. We further used four key metabolic markers (tryptophan, kynurenine, asymmetric dimethylarginine, and 1-Methylhistidine) to develop a COVID-19 mortality risk model through the application of multiple machine-learning methods. Conclusions Metabolomics analysis revealed distinct metabolic signatures among different severity groups, reflecting discernible alterations in amino acid levels and perturbations in tryptophan metabolism. Notably, critical patients exhibited higher levels of short chain acylcarnitines, concomitant with higher concentrations of SDMA, ADMA, and 1-MH in severe cases and non-survivors. Conversely, levels of 3-methylhistidine were lower in this context.
Collapse
Affiliation(s)
| | - Asmma Doudin
- Biomedical Research Center (BRC), Qatar University, Doha, Qatar
| | - Theeb Osama Sulaiman
- Department of Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Omar Jamil
- Department of Radiology, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Rida Arif
- Emergency Medicine Department, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Fatima Al Sada
- Neurosurgery Department, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Hadi M. Yassine
- Biomedical Research Center (BRC), Qatar University, Doha, Qatar
| | - Mohamed A. Elrayess
- College of Medicine, Qatar University (QU) Health, Qatar University, Doha, Qatar
- Biomedical Research Center (BRC), Qatar University, Doha, Qatar
| | - Abdel-Naser Elzouki
- College of Medicine, Qatar University (QU) Health, Qatar University, Doha, Qatar
- Department of Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Mohamed M. Emara
- College of Medicine, Qatar University (QU) Health, Qatar University, Doha, Qatar
| | | | - Farhan S. Cyprian
- College of Medicine, Qatar University (QU) Health, Qatar University, Doha, Qatar
| |
Collapse
|
14
|
Lodge S, Litton E, Gray N, Ryan M, Millet O, Fear M, Raby E, Currie A, Wood F, Holmes E, Wist J, Nicholson JK. Stratification of Sepsis Patients on Admission into the Intensive Care Unit According to Differential Plasma Metabolic Phenotypes. J Proteome Res 2024; 23:1328-1340. [PMID: 38513133 PMCID: PMC11002934 DOI: 10.1021/acs.jproteome.3c00803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/15/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024]
Abstract
Delayed diagnosis of patients with sepsis or septic shock is associated with increased mortality and morbidity. UPLC-MS and NMR spectroscopy were used to measure panels of lipoproteins, lipids, biogenic amines, amino acids, and tryptophan pathway metabolites in blood plasma samples collected from 152 patients within 48 h of admission into the Intensive Care Unit (ICU) where 62 patients had no sepsis, 71 patients had sepsis, and 19 patients had septic shock. Patients with sepsis or septic shock had higher concentrations of neopterin and lower levels of HDL cholesterol and phospholipid particles in comparison to nonsepsis patients. Septic shock could be differentiated from sepsis patients based on different concentrations of 10 lipids, including significantly lower concentrations of five phosphatidylcholine species, three cholesterol esters, one dihydroceramide, and one phosphatidylethanolamine. The Supramolecular Phospholipid Composite (SPC) was reduced in all ICU patients, while the composite markers of acute phase glycoproteins were increased in the sepsis and septic shock patients within 48 h admission into ICU. We show that the plasma metabolic phenotype obtained within 48 h of ICU admission is diagnostic for the presence of sepsis and that septic shock can be differentiated from sepsis based on the lipid profile.
Collapse
Affiliation(s)
- Samantha Lodge
- Australian
National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Edward Litton
- Intensive
Care Unit, Fiona Stanley Hospital, Murdoch, WA 6150, Australia
- Intensive
Care Unit, St John of God Hospital, Subiaco, WA 6009, Australia
- School
of Medicine, University of Western Australia, Crawley, WA 6009, Australia
| | - Nicola Gray
- Australian
National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Monique Ryan
- Australian
National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
| | - Oscar Millet
- Precision
Medicine and Metabolism Laboratory, CIC
bioGUNE, Parque Tecnológico
de Bizkaia, Bld. 800, Derio 48160, Spain
| | - Mark Fear
- Burn
Injury Research Unit, School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
- Fiona
Wood Foundation, Perth, WA 6150, Australia
| | - Edward Raby
- Department
of Infectious Diseases, Fiona Stanley Hospital, Murdoch, WA 6150, Australia
| | - Andrew Currie
- School
of Medical, Molecular & Forensic Sciences, Murdoch University, Perth, WA 6150, Australia
- Centre
for Molecular Medicine & Innovative Therapeutics, Murdoch University, Perth, WA 6150, Australia
- Wesfarmers
Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia
| | - Fiona Wood
- Burn
Injury Research Unit, School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
- Fiona
Wood Foundation, Perth, WA 6150, Australia
- Burns
service of Western Australia, WA Department
of Health, Murdoch, WA 6150, Australia
| | - Elaine Holmes
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Institute
of Global Health Innovation, Faculty of Medicine, Imperial College London, Level 1, Faculty Building, South Kensington Campus, London SW7 2NA, U.K.
| | - Julien Wist
- Australian
National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
- Center
for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA 6150, Australia
- Chemistry
Department, Universidad del Valle, Cali 76001, Colombia
- Department of Metabolism, Digestion and
Reproduction, Faculty of Medicine, Imperial
College London, Sir Alexander
Fleming Building, South Kensington, London SW7 2AZ, U.K.
| | - Jeremy K. Nicholson
- Australian
National Phenome Center, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, WA6150, Australia
- Department of Metabolism, Digestion and
Reproduction, Faculty of Medicine, Imperial
College London, Sir Alexander
Fleming Building, South Kensington, London SW7 2AZ, U.K.
| |
Collapse
|
15
|
Whiley L, Lawler NG, Zeng AX, Lee A, Chin ST, Bizkarguenaga M, Bruzzone C, Embade N, Wist J, Holmes E, Millet O, Nicholson JK, Gray N. Cross-Validation of Metabolic Phenotypes in SARS-CoV-2 Infected Subpopulations Using Targeted Liquid Chromatography-Mass Spectrometry (LC-MS). J Proteome Res 2024; 23:1313-1327. [PMID: 38484742 PMCID: PMC11002931 DOI: 10.1021/acs.jproteome.3c00797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 04/06/2024]
Abstract
To ensure biological validity in metabolic phenotyping, findings must be replicated in independent sample sets. Targeted workflows have long been heralded as ideal platforms for such validation due to their robust quantitative capability. We evaluated the capability of liquid chromatography-mass spectrometry (LC-MS) assays targeting organic acids and bile acids to validate metabolic phenotypes of SARS-CoV-2 infection. Two independent sample sets were collected: (1) Australia: plasma, SARS-CoV-2 positive (n = 20), noninfected healthy controls (n = 22) and COVID-19 disease-like symptoms but negative for SARS-CoV-2 infection (n = 22). (2) Spain: serum, SARS-CoV-2 positive (n = 33) and noninfected healthy controls (n = 39). Multivariate modeling using orthogonal projections to latent structures discriminant analyses (OPLS-DA) classified healthy controls from SARS-CoV-2 positive (Australia; R2 = 0.17, ROC-AUC = 1; Spain R2 = 0.20, ROC-AUC = 1). Univariate analyses revealed 23 significantly different (p < 0.05) metabolites between healthy controls and SARS-CoV-2 positive individuals across both cohorts. Significant metabolites revealed consistent perturbations in cellular energy metabolism (pyruvic acid, and 2-oxoglutaric acid), oxidative stress (lactic acid, 2-hydroxybutyric acid), hypoxia (2-hydroxyglutaric acid, 5-aminolevulinic acid), liver activity (primary bile acids), and host-gut microbial cometabolism (hippuric acid, phenylpropionic acid, indole-3-propionic acid). These data support targeted LC-MS metabolic phenotyping workflows for biological validation in independent sample sets.
Collapse
Affiliation(s)
- Luke Whiley
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute Harry
Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| | - Nathan G. Lawler
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute Harry
Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| | - Annie Xu Zeng
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| | - Alex Lee
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| | - Sung-Tong Chin
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| | - Maider Bizkarguenaga
- Centro
de Investigación Cooperativa en Biociencias—CIC bioGUNE,
Precision Medicine and Metabolism Laboratory, Basque Research and
Technology Alliance, Bizkaia Science and
Technology Park, Building
800, 48160 Derio, Spain
| | - Chiara Bruzzone
- Centro
de Investigación Cooperativa en Biociencias—CIC bioGUNE,
Precision Medicine and Metabolism Laboratory, Basque Research and
Technology Alliance, Bizkaia Science and
Technology Park, Building
800, 48160 Derio, Spain
| | - Nieves Embade
- Centro
de Investigación Cooperativa en Biociencias—CIC bioGUNE,
Precision Medicine and Metabolism Laboratory, Basque Research and
Technology Alliance, Bizkaia Science and
Technology Park, Building
800, 48160 Derio, Spain
| | - Julien Wist
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute Harry
Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Chemistry
Department, Universidad del Valle, Cali 76001, Colombia
| | - Elaine Holmes
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute Harry
Perkins 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.
| | - Oscar Millet
- Centro
de Investigación Cooperativa en Biociencias—CIC bioGUNE,
Precision Medicine and Metabolism Laboratory, Basque Research and
Technology Alliance, Bizkaia Science and
Technology Park, Building
800, 48160 Derio, Spain
| | - Jeremy K. Nicholson
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute Harry
Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Institute
of Global Health Innovation, Faculty Building South Kensington Campus, Imperial College London, London SW7 2AZ, U.K.
| | - Nicola Gray
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute Harry
Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| |
Collapse
|
16
|
Lonati C, Berezhnoy G, Lawler N, Masuda R, Kulkarni A, Sala S, Nitschke P, Zizmare L, Bucci D, Cannet C, Schäfer H, Singh Y, Gray N, Lodge S, Nicholson J, Merle U, Wist J, Trautwein C. Urinary phenotyping of SARS-CoV-2 infection connects clinical diagnostics with metabolomics and uncovers impaired NAD + pathway and SIRT1 activation. Clin Chem Lab Med 2024; 62:770-788. [PMID: 37955280 DOI: 10.1515/cclm-2023-1017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/22/2023] [Indexed: 11/14/2023]
Abstract
OBJECTIVES The stratification of individuals suffering from acute and post-acute SARS-CoV-2 infection remains a critical challenge. Notably, biomarkers able to specifically monitor viral progression, providing details about patient clinical status, are still not available. Herein, quantitative metabolomics is progressively recognized as a useful tool to describe the consequences of virus-host interactions considering also clinical metadata. METHODS The present study characterized the urinary metabolic profile of 243 infected individuals by quantitative nuclear magnetic resonance (NMR) spectroscopy and liquid chromatography mass spectrometry (LC-MS). Results were compared with a historical cohort of noninfected subjects. Moreover, we assessed the concentration of recently identified antiviral nucleosides and their association with other metabolites and clinical data. RESULTS Urinary metabolomics can stratify patients into classes of disease severity, with a discrimination ability comparable to that of clinical biomarkers. Kynurenines showed the highest fold change in clinically-deteriorated patients and higher-risk subjects. Unique metabolite clusters were also generated based on age, sex, and body mass index (BMI). Changes in the concentration of antiviral nucleosides were associated with either other metabolites or clinical variables. Increased kynurenines and reduced trigonelline excretion indicated a disrupted nicotinamide adenine nucleotide (NAD+) and sirtuin 1 (SIRT1) pathway. CONCLUSIONS Our results confirm the potential of urinary metabolomics for noninvasive diagnostic/prognostic screening and show that the antiviral nucleosides could represent novel biomarkers linking viral load, immune response, and metabolism. Moreover, we established for the first time a casual link between kynurenine accumulation and deranged NAD+/SIRT1, offering a novel mechanism through which SARS-CoV-2 manipulates host physiology.
Collapse
Affiliation(s)
- Caterina Lonati
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany
| | - Georgy Berezhnoy
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany
| | - Nathan Lawler
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Reika Masuda
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Aditi Kulkarni
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany
| | - Samuele Sala
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Philipp Nitschke
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Laimdota Zizmare
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany
| | - Daniele Bucci
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany
| | - Claire Cannet
- Bruker BioSpin GmbH, AIC Division, Ettlingen, Germany
| | | | - Yogesh Singh
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Nicola Gray
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Samantha Lodge
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Jeremy Nicholson
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Uta Merle
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Julien Wist
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University Perth, Australia
| | - Christoph Trautwein
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
17
|
Sala S, Nitschke P, Masuda R, Gray N, Lawler NG, Wood JM, Buckler JN, Berezhnoy G, Bolaños J, Boughton BA, Lonati C, Rössler T, Singh Y, Wilson ID, Lodge S, Morillon AC, Loo RL, Hall D, Whiley L, Evans GB, Grove TL, Almo SC, Harris LD, Holmes E, Merle U, Trautwein C, Nicholson JK, Wist J. Integrative Molecular Structure Elucidation and Construction of an Extended Metabolic Pathway Associated with an Ancient Innate Immune Response in COVID-19 Patients. J Proteome Res 2024; 23:956-970. [PMID: 38310443 PMCID: PMC10913068 DOI: 10.1021/acs.jproteome.3c00654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/01/2023] [Accepted: 12/29/2023] [Indexed: 02/05/2024]
Abstract
We present compelling evidence for the existence of an extended innate viperin-dependent pathway, which provides crucial evidence for an adaptive response to viral agents, such as SARS-CoV-2. We show the in vivo biosynthesis of a family of novel endogenous cytosine metabolites with potential antiviral activities. Two-dimensional nuclear magnetic resonance (NMR) spectroscopy revealed a characteristic spin-system motif, indicating the presence of an extended panel of urinary metabolites during the acute viral replication phase. Mass spectrometry additionally enabled the characterization and quantification of the most abundant serum metabolites, showing the potential diagnostic value of the compounds for viral infections. In total, we unveiled ten nucleoside (cytosine- and uracil-based) analogue structures, eight of which were previously unknown in humans allowing us to propose a new extended viperin pathway for the innate production of antiviral compounds. The molecular structures of the nucleoside analogues and their correlation with an array of serum cytokines, including IFN-α2, IFN-γ, and IL-10, suggest an association with the viperin enzyme contributing to an ancient endogenous innate immune defense mechanism against viral infection.
Collapse
Affiliation(s)
- Samuele Sala
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Philipp Nitschke
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Reika Masuda
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Nicola Gray
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Nathan G. Lawler
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - James M. Wood
- Ferrier
Research Institute, Victoria University
of Wellington, Wellington 6012, New Zealand
- The
Maurice Wilkins Centre for Molecular Biodiscovef Wellington, Welry, The University of Auckland, Auckland 1010, New Zealand
| | - Joshua N. Buckler
- Ferrier
Research Institute, Victoria University
of Wellington, Wellington 6012, New Zealand
| | - Georgy Berezhnoy
- Department
of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, 72074 Tübingen, Germany
| | - Jose Bolaños
- Chemistry
Department, Universidad del Valle, Cali 76001, Colombia
| | - Berin A. Boughton
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Caterina Lonati
- Center
for Preclinical Research, Fondazione IRCCS
Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Titus Rössler
- Department
of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, 72074 Tübingen, Germany
| | - Yogesh Singh
- Institute
of Medical Genetics and Applied Genomics, University Hospital Tübingen, 72074 Tübingen, Germany
| | - Ian D. Wilson
- Division
of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College, Burlington Danes Building, Du Cane Road, London W12 0NN, U.K.
| | - Samantha Lodge
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Aude-Claire Morillon
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Ruey Leng Loo
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Drew Hall
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Luke Whiley
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Gary B. Evans
- Ferrier
Research Institute, Victoria University
of Wellington, Wellington 6012, New Zealand
- The
Maurice Wilkins Centre for Molecular Biodiscovef Wellington, Welry, The University of Auckland, Auckland 1010, New Zealand
| | - Tyler L. Grove
- Department
of Biochemistry, Albert Einstein College
of Medicine, Bronx, New York 10461, United States
| | - Steven C. Almo
- Department
of Biochemistry, Albert Einstein College
of Medicine, Bronx, New York 10461, United States
| | - Lawrence D. Harris
- Ferrier
Research Institute, Victoria University
of Wellington, Wellington 6012, New Zealand
- The
Maurice Wilkins Centre for Molecular Biodiscovef Wellington, Welry, The University of Auckland, Auckland 1010, New Zealand
| | - Elaine Holmes
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
- Division
of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College, Burlington Danes Building, Du Cane Road, London W12 0NN, U.K.
| | - Uta Merle
- Department
of Internal Medicine IV, University Hospital
Heidelberg, 69120 Heidelberg, Germany
| | - Christoph Trautwein
- Department
of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, 72074 Tübingen, Germany
| | - Jeremy K. Nicholson
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
- Institute
of Global Health Innovation, Faculty of
Medicine, Imperial College London, Level 1, Faculty Building, South Kensington Campus, London SW7 2NA, U.K.
| | - Julien Wist
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
- Chemistry
Department, Universidad del Valle, Cali 76001, Colombia
- Faculty of Medicine, Department of Metabolism,
Digestion and Reproduction,
Division of Digestive Diseases at Imperial College, London SW7 2AZ, U.K.
| |
Collapse
|
18
|
Wang Y, Shen M, Li Y, Shao J, Zhang F, Guo M, Zhang Z, Zheng S. COVID-19-associated liver injury: Adding fuel to the flame. Cell Biochem Funct 2023; 41:1076-1092. [PMID: 37947373 DOI: 10.1002/cbf.3883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 11/12/2023]
Abstract
COVID-19 is mainly characterized by respiratory disorders and progresses to multiple organ involvement in severe cases. With expansion of COVID-19 and SARS-CoV-2 research, correlative liver injury has been revealed. It is speculated that COVID-19 patients exhibited abnormal liver function, as previously observed in the SARS and MERS pandemics. Furthermore, patients with underlying diseases such as chronic liver disease are more susceptible to SARS-CoV-2 and indicate a poor prognosis accompanied by respiratory symptoms, systemic inflammation, or metabolic diseases. Therefore, COVID-19 has the potential to impair liver function, while individuals with preexisting liver disease suffer from much worse infected conditions. COVID-19 related liver injury may be owing to direct cytopathic effect, immune dysfunction, gut-liver axis interaction, and inappropriate medication use. However, discussions on these issues are infancy. Expanding research have revealed that angiotensin converting enzyme 2 (ACE2) expression mediated the combination of virus and target cells, iron metabolism participated in the virus life cycle and the fate of target cells, and amino acid metabolism regulated immune response in the host cells, which are all closely related to liver health. Further exploration holds great significance in elucidating the pathogenesis, facilitating drug development, and advancing clinical treatment of COVID-19-related liver injury. This article provides a review of the clinical and laboratory hepatic characteristics in COVID-19 patients, describes the etiology and impact of liver injury, and discusses potential pathophysiological mechanisms.
Collapse
Affiliation(s)
- Yingqian Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Shen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Yujia Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mei Guo
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
19
|
Cao Q, Du X, Jiang XY, Tian Y, Gao CH, Liu ZY, Xu T, Tao XX, Lei M, Wang XQ, Ye LL, Duan DD. Phenome-wide association study and precision medicine of cardiovascular diseases in the post-COVID-19 era. Acta Pharmacol Sin 2023; 44:2347-2357. [PMID: 37532784 PMCID: PMC10692238 DOI: 10.1038/s41401-023-01119-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/29/2023] [Indexed: 08/04/2023]
Abstract
SARS-CoV-2 infection causes injuries of not only the lungs but also the heart and endothelial cells in vasculature of multiple organs, and induces systemic inflammation and immune over-reactions, which makes COVID-19 a disease phenome that simultaneously affects multiple systems. Cardiovascular diseases (CVD) are intrinsic risk and causative factors for severe COVID-19 comorbidities and death. The wide-spread infection and reinfection of SARS-CoV-2 variants and the long-COVID may become a new common threat to human health and propose unprecedented impact on the risk factors, pathophysiology, and pharmacology of many diseases including CVD for a long time. COVID-19 has highlighted the urgent demand for precision medicine which needs new knowledge network to innovate disease taxonomy for more precise diagnosis, therapy, and prevention of disease. A deeper understanding of CVD in the setting of COVID-19 phenome requires a paradigm shift from the current phenotypic study that focuses on the virus or individual symptoms to phenomics of COVID-19 that addresses the inter-connectedness of clinical phenotypes, i.e., clinical phenome. Here, we summarize the CVD manifestations in the full clinical spectrum of COVID-19, and the phenome-wide association study of CVD interrelated to COVID-19. We discuss the underlying biology for CVD in the COVID-19 phenome and the concept of precision medicine with new phenomic taxonomy that addresses the overall pathophysiological responses of the body to the SARS-CoV-2 infection. We also briefly discuss the unique taxonomy of disease as Zheng-hou patterns in traditional Chinese medicine, and their potential implications in precision medicine of CVD in the post-COVID-19 era.
Collapse
Affiliation(s)
- Qian Cao
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Xin Du
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Xiao-Yan Jiang
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yuan Tian
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Chen-Hao Gao
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Zi-Yu Liu
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Ting Xu
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Xing-Xing Tao
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Ming Lei
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Xiao-Qiang Wang
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Lingyu Linda Ye
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China.
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, China.
- Key Laboratory of Autoimmune Diseases and Precision Medicie, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750001, China.
| | - Dayue Darrel Duan
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China.
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, China.
- Key Laboratory of Autoimmune Diseases and Precision Medicie, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750001, China.
- The Department of Pharmacology, University of Nevada Reno School of Medicine, Reno, NV, 89557, USA.
| |
Collapse
|
20
|
Al-Shalan HAM, Zhou L, Dong Z, Wang P, Nicholls PK, Boughton B, Stumbles PA, Greene WK, Ma B. Systemic perturbations in amino acids/amino acid derivatives and tryptophan pathway metabolites associated with murine influenza A virus infection. Virol J 2023; 20:270. [PMID: 37990229 PMCID: PMC10664681 DOI: 10.1186/s12985-023-02239-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Influenza A virus (IAV) is the only influenza virus causing flu pandemics (i.e., global epidemics of flu disease). Influenza (the flu) is a highly contagious disease that can be deadly, especially in high-risk groups. Worldwide, these annual epidemics are estimated to result in about 3 to 5 million cases of severe illness and in about 290,000 to 650,000 respiratory deaths. We intend to reveal the effect of IAV infection on the host's metabolism, immune response, and neurotoxicity by using a mouse IAV infection model. METHODS 51 metabolites of murine blood plasma (33 amino acids/amino acid derivatives (AADs) and 18 metabolites of the tryptophan pathway) were analyzed by using Ultra-High-Performance Liquid Chromatography-Mass Spectrometry with Electrospray Ionization at the acute (7 days post-infection (dpi)), resolution (14 dpi), and recovery (21 dpi) stages of the virus infection in comparison with controls. RESULTS Among the 33 biogenic amino acids/AADs, the levels of five amino acids/AADs (1-methylhistidine, 5-oxoproline, α-aminobutyric acid, glutamine, and taurine) increased by 7 dpi, whereas the levels of ten amino acids/AADs (4-hydroxyproline, alanine, arginine, asparagine, cysteine, citrulline, glycine, methionine, proline, and tyrosine) decreased. By 14 dpi, the levels of one AAD (3-methylhistidine) increased, whereas the levels of five amino acids/AADs (α-aminobutyric acid, aminoadipic acid, methionine, threonine, valine) decreased. Among the 18 metabolites from the tryptophan pathway, the levels of kynurenine, quinolinic acid, hydroxykynurenine increased by 7 dpi, whereas the levels of indole-3-acetic acid and nicotinamide riboside decreased. CONCLUSIONS Our data may facilitate understanding the molecular mechanisms of host responses to IAV infection and provide a basis for discovering potential new mechanistic, diagnostic, and prognostic biomarkers and therapeutic targets for IAV infection.
Collapse
Affiliation(s)
- Huda A M Al-Shalan
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
- Department of Microbiology/Virology, College of Veterinary Medicine, Baghdad University, Baghdad, Iraq
| | - Lu Zhou
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhifan Dong
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Penghao Wang
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| | - Philip K Nicholls
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| | - Berin Boughton
- Australian National Phenome Centre, Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Philip A Stumbles
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
- Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, Australia
| | - Wayne K Greene
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| | - Bin Ma
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia.
| |
Collapse
|
21
|
Arnsten AFT, Ishizawa Y, Xie Z. Scientific rationale for the use of α2A-adrenoceptor agonists in treating neuroinflammatory cognitive disorders. Mol Psychiatry 2023; 28:4540-4552. [PMID: 37029295 PMCID: PMC10080530 DOI: 10.1038/s41380-023-02057-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/09/2023]
Abstract
Neuroinflammatory disorders preferentially impair the higher cognitive and executive functions of the prefrontal cortex (PFC). This includes such challenging disorders as delirium, perioperative neurocognitive disorder, and the sustained cognitive deficits from "long-COVID" or traumatic brain injury. There are no FDA-approved treatments for these symptoms; thus, understanding their etiology is important for generating therapeutic strategies. The current review describes the molecular rationale for why PFC circuits are especially vulnerable to inflammation, and how α2A-adrenoceptor (α2A-AR) actions throughout the nervous and immune systems can benefit the circuits in PFC needed for higher cognition. The layer III circuits in the dorsolateral PFC (dlPFC) that generate and sustain the mental representations needed for higher cognition have unusual neurotransmission and neuromodulation. They are wholly dependent on NMDAR neurotransmission, with little AMPAR contribution, and thus are especially vulnerable to kynurenic acid inflammatory signaling which blocks NMDAR. Layer III dlPFC spines also have unusual neuromodulation, with cAMP magnification of calcium signaling in spines, which opens nearby potassium channels to rapidly weaken connectivity and reduce neuronal firing. This process must be tightly regulated, e.g. by mGluR3 or α2A-AR on spines, to prevent loss of firing. However, the production of GCPII inflammatory signaling reduces mGluR3 actions and markedly diminishes dlPFC network firing. Both basic and clinical studies show that α2A-AR agonists such as guanfacine can restore dlPFC network firing and cognitive function, through direct actions in the dlPFC, but also by reducing the activity of stress-related circuits, e.g. in the locus coeruleus and amygdala, and by having anti-inflammatory actions in the immune system. This information is particularly timely, as guanfacine is currently the focus of large clinical trials for the treatment of delirium, and in open label studies for the treatment of cognitive deficits from long-COVID.
Collapse
Affiliation(s)
- Amy F T Arnsten
- Department Neuroscience, Yale University School of Medicine, New Haven, CT, 056510, USA.
| | - Yumiko Ishizawa
- Department Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Zhongcong Xie
- Department Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
22
|
Michaelis S, Zelzer S, Schneider C, Schnedl WJ, Baranyi A, Meinitzer A, Herrmann M, Enko D. The possible role of quinolinic acid as a predictive marker in patients with SARS-CoV-2. Clin Chim Acta 2023; 550:117583. [PMID: 37802207 DOI: 10.1016/j.cca.2023.117583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/03/2023] [Accepted: 10/03/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND AND AIMS Quinolinic acid (QA) is a metabolite of the kynurenine pathway, which is activated by inflammatory stimuli during viral infection. We investigated the role of QA in patients infected with SARS-CoV-2, particularly its prognostic value for survival. METHODS Overall, 104 unvaccinated inpatients were included, divided into a survival (N = 80) and a deceased group (N = 24). Plasma levels of tryptophan, kynurenine, QA, C-reactive protein (CRP) and procalcitonin (PCT) were measured on admission and after seven days. The QA/TRP ratio and the relative differences between the measurements for QA (QA-Diff) and QA/TRP (Diff-QA/TRP) were calculated. RESULTS Among the kynurenine pathway markers, QA-Diff showed the highest discriminatory power for the survival prognosis (Youden index 0.467, cut-off -1.3 %, AUC 0.733, p < 0.001, sensitivity 0.79, specificity 0.675). Among the inflammatory markers, CRP showed the highest discriminatory power (Youden index 0.533, cut-off 25.0 mg/L, AUC 0.794, p < 0.001, sensitivity 0.958, specificity 0.575). A significant correlation between QA and PCT was found on admission and after one week (Spearman's rho 0.455 and 0.539, all p-values < 0.001). CONCLUSIONS QA may serve as prognostic marker for survival in patients with SARS-CoV-2. The repeated measurements during the first week of the disease may enhance the prognostic power.
Collapse
Affiliation(s)
- Simon Michaelis
- Institute of Clinical Chemistry and Laboratory Medicine, General Hospital Hochsteiermark, Vordernberger Straße 42, 8700 Leoben, Austria.
| | - Sieglinde Zelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Christopher Schneider
- Institute of Clinical Chemistry and Laboratory Medicine, General Hospital Hochsteiermark, Vordernberger Straße 42, 8700 Leoben, Austria
| | - Wolfgang J Schnedl
- Practice for General Internal Medicine, Dr.-Theodor-Körner-Straße 19b, 8600 Bruck/Mur, Austria
| | - Andreas Baranyi
- Department of Psychiatry and Psychotherapeutic Medicine, Medical, University of Graz, Auenbruggerplatz 31, 8036 Graz, Austria
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Dietmar Enko
- Institute of Clinical Chemistry and Laboratory Medicine, General Hospital Hochsteiermark, Vordernberger Straße 42, 8700 Leoben, Austria; Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| |
Collapse
|
23
|
Cascant-Vilaplana MM, Lara-Cantón I, Ramos-Garcia V, Pinilla-González A, Solaz-García Á, Quintás G, Marín-Reina P, Aguar M, Torrejón-Rodríguez L, Vento M, Kuligowski J, Cernada M. Metabolic dysregulation in term infants from SARS-CoV-2-infected mothers. World J Pediatr 2023; 19:912-917. [PMID: 37474813 DOI: 10.1007/s12519-023-00735-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/25/2023] [Indexed: 07/22/2023]
Affiliation(s)
- Mari Merce Cascant-Vilaplana
- Neonatal Research Unit, Health Research Institute Hospital La Fe, University Polytechnic Hospital La Fe, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Inmaculada Lara-Cantón
- Neonatal Research Unit, Health Research Institute Hospital La Fe, University Polytechnic Hospital La Fe, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Victoria Ramos-Garcia
- Neonatal Research Unit, Health Research Institute Hospital La Fe, University Polytechnic Hospital La Fe, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Alejandro Pinilla-González
- Neonatal Research Unit, Health Research Institute Hospital La Fe, University Polytechnic Hospital La Fe, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Álvaro Solaz-García
- Neonatal Research Unit, Health Research Institute Hospital La Fe, University Polytechnic Hospital La Fe, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Guillermo Quintás
- Leitat Technological Center, Valencia, Spain
- Analytical Unit, Health Research Institute La Fe, Valencia, Spain
| | - Purificación Marín-Reina
- Neonatal Research Unit, Health Research Institute Hospital La Fe, University Polytechnic Hospital La Fe, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
- Division of Neonatology, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Marta Aguar
- Neonatal Research Unit, Health Research Institute Hospital La Fe, University Polytechnic Hospital La Fe, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
- Division of Neonatology, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Laura Torrejón-Rodríguez
- Neonatal Research Unit, Health Research Institute Hospital La Fe, University Polytechnic Hospital La Fe, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Máximo Vento
- Neonatal Research Unit, Health Research Institute Hospital La Fe, University Polytechnic Hospital La Fe, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
- Division of Neonatology, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Julia Kuligowski
- Neonatal Research Unit, Health Research Institute Hospital La Fe, University Polytechnic Hospital La Fe, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain.
| | - María Cernada
- Neonatal Research Unit, Health Research Institute Hospital La Fe, University Polytechnic Hospital La Fe, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain.
- Division of Neonatology, University and Polytechnic Hospital La Fe, Valencia, Spain.
| |
Collapse
|
24
|
Badawy AB. The kynurenine pathway of tryptophan metabolism: a neglected therapeutic target of COVID-19 pathophysiology and immunotherapy. Biosci Rep 2023; 43:BSR20230595. [PMID: 37486805 PMCID: PMC10407158 DOI: 10.1042/bsr20230595] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/29/2023] [Accepted: 07/21/2023] [Indexed: 07/26/2023] Open
Abstract
SARS-CoV-2 (COVID-19) exerts profound changes in the kynurenine (Kyn) pathway (KP) of tryptophan (Trp) metabolism that may underpin its pathophysiology. The KP is the main source of the vital cellular effector NAD+ and intermediate metabolites that modulate immune and neuronal functions. Trp metabolism is the top pathway influenced by COVID-19. Sixteen studies established virus-induced activation of the KP mediated mainly by induction of indoleamine 2,3-dioxygenase (IDO1) in most affected tissues and of IDO2 in lung by the increased release of proinflammatory cytokines but could additionally involve increased flux of plasma free Trp and induction of Trp 2,3-dioxygenase (TDO) by cortisol. The major Kyn metabolite targeted by COVID-19 is kynurenic acid (KA), the Kyn metabolite with the greatest affinity for the aryl hydrocarbon receptor (AhR), which is also activated by COVID-19. AhR activation initiates two important series of events: a vicious circle involving IDO1 induction, KA accumulation and further AhR activation, and activation of poly (ADP-ribose) polymerase (PARP) leading to NAD+ depletion and cell death. The virus further deprives the host of NAD+ by inhibiting its main biosynthetic pathway from quinolinic acid, while simultaneously acquiring NAD+ by promoting its synthesis from nicotinamide in the salvage pathway. Additionally, the protective effects of sirtuin 1 are minimised by the PARP activation. KP dysfunction may also underpin the mood and neurological disorders acutely and during 'long COVID'. More studies of potential effects of vaccination therapy on the KP are required and exploration of therapeutic strategies involving modulation of the KP changes are proposed.
Collapse
Affiliation(s)
- Abdulla Abu-Bakr Badawy
- Formerly School of Health Sciences, Cardiff Metropolitan University, Western Avenue, Cardiff CF5 2YB, Wales, U.K
| |
Collapse
|
25
|
Abstract
The emergence of the SARS-CoV-2 (COVID-19) virus has exacted a significant toll on the global population in terms of fatalities, health consequences, and economics [...]
Collapse
Affiliation(s)
- Trevor R. Norman
- Department of Psychiatry, University of Melbourne, Austin Hospital, Heidelberg, VIC 3084, Australia
| |
Collapse
|
26
|
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
|
27
|
Piater T, Gietl M, Hofer S, Gostner JM, Sahanic S, Tancevski I, Sonnweber T, Pizzini A, Egger A, Schennach H, Loeffler-Ragg J, Weiss G, Kurz K. Persistent Symptoms and IFN-γ-Mediated Pathways after COVID-19. J Pers Med 2023; 13:1055. [PMID: 37511668 PMCID: PMC10382041 DOI: 10.3390/jpm13071055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/18/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
After COVID-19, patients have reported various complaints such as fatigue, neurological symptoms, and insomnia. Immune-mediated changes in amino acid metabolism might contribute to the development of these symptoms. Patients who had had acute, PCR-confirmed COVID-19 infection about 60 days earlier were recruited within the scope of the prospective CovILD study. We determined the inflammatory parameters and alterations in tryptophan and phenylalanine metabolism in 142 patients cross-sectionally. Symptom persistence (pain, gastrointestinal symptoms, anosmia, sleep disturbance, and neurological symptoms) and patients' physical levels of functioning were recorded. Symptoms improved in many patients after acute COVID-19 (n = 73, 51.4%). Still, a high percentage of patients had complaints, and women were affected more often. In many patients, ongoing immune activation (as indicated by high neopterin and CRP concentrations) and enhanced tryptophan catabolism were found. A higher phenylalanine to tyrosine ratio (Phe/Tyr) was found in women with a lower level of functioning. Patients who reported improvements in pain had lower Phe/Tyr ratios, while patients with improved gastrointestinal symptoms presented with higher tryptophan and kynurenine values. Our results suggest that women have persistent symptoms after COVID-19 more often than men. In addition, the physical level of functioning and the improvements in certain symptoms appear to be associated with immune-mediated changes in amino acid metabolism.
Collapse
Affiliation(s)
- Talia Piater
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Mario Gietl
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Stefanie Hofer
- Institute of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innrain 80, 6020 Innsbruck, Austria
| | - Johanna M Gostner
- Institute of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innrain 80, 6020 Innsbruck, Austria
| | - Sabina Sahanic
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Thomas Sonnweber
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Alex Pizzini
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Alexander Egger
- Central Institute for Medical and Chemical Laboratory Diagnostics (ZIMCL), Tirol Kliniken GmbH, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Harald Schennach
- Central Institute for Blood Transfusion and Immunological Department, Tirol Kliniken GmbH, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Judith Loeffler-Ragg
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Guenter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Katharina Kurz
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| |
Collapse
|
28
|
den Hartog I, Karu N, Zwep LB, Voorn GP, van de Garde EM, Hankemeier T, van Hasselt JC. Differential metabolic host response to pathogens associated with community-acquired pneumonia. Metabol Open 2023; 18:100239. [PMID: 37025095 PMCID: PMC10070890 DOI: 10.1016/j.metop.2023.100239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Background Metabolic changes induced by the host immune response to pathogens found in patients with community-acquired pneumonia (CAP) may provide insight into its pathogenesis. In this study, we characterized differences in the host metabolic response to common CAP-associated pathogens. Method Targeted metabolomic profiling was performed on serum samples obtained from hospitalized CAP patients (n = 119) at admission. We quantified 347 unique metabolites across multiple biochemical classes, including amines, acylcarnitines, and signaling lipids. We evaluated if unique associations between metabolite levels and specific CAP-associated pathogens could be identified. Results Several acylcarnitines were found to be elevated in C. burnetii and herpes simplex virus and lowered in M. pneumoniae as compared to other pathogens. Phenylalanine and kynurenine were found elevated in L. pneumophila as compared to other pathogens. S-methylcysteine was elevated in patients with M. pneumoniae, and these patients also showed lowered cortisol levels in comparison to almost all other pathogens. For the herpes simplex virus, we observed a unique elevation of eicosanoids and several amines. Many lysophosphatidylcholines showed an altered profile in C. burnetii versus S. pneumoniae, L. pneumophila, and respiratory syncytial virus. Finally, phosphatidylcholines were negatively affected by the influenza virus in comparison to S. pneumoniae. Conclusions In this exploratory analysis, metabolites from different biochemical classes were found to be altered in serum samples from patients with different CAP-associated pathogens, which may be used for hypothesis generation in studies on differences in pathogen host response and pathogenesis of CAP.
Collapse
Affiliation(s)
- Ilona den Hartog
- Division of Systems Pharmacology & Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Naama Karu
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Laura B. Zwep
- Division of Systems Pharmacology & Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - G. Paul Voorn
- Department of Medical Microbiology and Immunology, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - Ewoudt M.W. van de Garde
- Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
- Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - Thomas Hankemeier
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - J.G. Coen van Hasselt
- Division of Systems Pharmacology & Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
- Corresponding author.
| |
Collapse
|
29
|
Fesharaki Zadeh A, Arnsten AFT, Wang M. Scientific Rationale for the Treatment of Cognitive Deficits from Long COVID. Neurol Int 2023; 15:725-742. [PMID: 37368329 PMCID: PMC10303664 DOI: 10.3390/neurolint15020045] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/26/2023] [Accepted: 05/11/2023] [Indexed: 06/28/2023] Open
Abstract
Sustained cognitive deficits are a common and debilitating feature of "long COVID", but currently there are no FDA-approved treatments. The cognitive functions of the dorsolateral prefrontal cortex (dlPFC) are the most consistently afflicted by long COVID, including deficits in working memory, motivation, and executive functioning. COVID-19 infection greatly increases kynurenic acid (KYNA) and glutamate carboxypeptidase II (GCPII) in brain, both of which can be particularly deleterious to PFC function. KYNA blocks both NMDA and nicotinic-alpha-7 receptors, the two receptors required for dlPFC neurotransmission, and GCPII reduces mGluR3 regulation of cAMP-calcium-potassium channel signaling, which weakens dlPFC network connectivity and reduces dlPFC neuronal firing. Two agents approved for other indications may be helpful in restoring dlPFC physiology: the antioxidant N-acetyl cysteine inhibits the production of KYNA, and the α2A-adrenoceptor agonist guanfacine regulates cAMP-calcium-potassium channel signaling in dlPFC and is also anti-inflammatory. Thus, these agents may be helpful in treating the cognitive symptoms of long COVID.
Collapse
Affiliation(s)
- Arman Fesharaki Zadeh
- Departments of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Departments of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Amy F. T. Arnsten
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Min Wang
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA;
| |
Collapse
|
30
|
Li Y, Hook JS, Ding Q, Xiao X, Chung SS, Mettlen M, Xu L, Moreland JG, Agathocleous M. Neutrophil metabolomics in severe COVID-19 reveal GAPDH as a suppressor of neutrophil extracellular trap formation. Nat Commun 2023; 14:2610. [PMID: 37147288 PMCID: PMC10162006 DOI: 10.1038/s41467-023-37567-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/20/2023] [Indexed: 05/07/2023] Open
Abstract
Severe COVID-19 is characterized by an increase in the number and changes in the function of innate immune cells including neutrophils. However, it is not known how the metabolome of immune cells changes in patients with COVID-19. To address these questions, we analyzed the metabolome of neutrophils from patients with severe or mild COVID-19 and healthy controls. We identified widespread dysregulation of neutrophil metabolism with disease progression including in amino acid, redox, and central carbon metabolism. Metabolic changes in neutrophils from patients with severe COVID-19 were consistent with reduced activity of the glycolytic enzyme GAPDH. Inhibition of GAPDH blocked glycolysis and promoted pentose phosphate pathway activity but blunted the neutrophil respiratory burst. Inhibition of GAPDH was sufficient to cause neutrophil extracellular trap (NET) formation which required neutrophil elastase activity. GAPDH inhibition increased neutrophil pH, and blocking this increase prevented cell death and NET formation. These findings indicate that neutrophils in severe COVID-19 have an aberrant metabolism which can contribute to their dysfunction. Our work also shows that NET formation, a pathogenic feature of many inflammatory diseases, is actively suppressed in neutrophils by a cell-intrinsic mechanism controlled by GAPDH.
Collapse
Affiliation(s)
- Yafeng Li
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jessica S Hook
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qing Ding
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xue Xiao
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stephen S Chung
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marcel Mettlen
- Department of Cell Biology, Quantitative Light Microscopy Core, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lin Xu
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jessica G Moreland
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michalis Agathocleous
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
31
|
Li X, Liu Y, Xu G, Xie Y, Wang X, Wu J, Chen H. Plasma metabolomic characterization of SARS-CoV-2 Omicron infection. Cell Death Dis 2023; 14:276. [PMID: 37076483 PMCID: PMC10113737 DOI: 10.1038/s41419-023-05791-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/21/2023]
Abstract
Omicron variants of SARS-CoV-2 have spread rapidly worldwide; however, most infected patients have mild or no symptoms. This study aimed to understand the host response to Omicron infections by performing metabolomic profiling of plasma. We observed that Omicron infections triggered an inflammatory response and innate immune, and adaptive immunity was suppressed, including reduced T-cell response and immunoglobulin antibody production. Similar to the original SARS-CoV-2 strain circulating in 2019, the host developed an anti-inflammatory response and accelerated energy metabolism in response to Omicron infection. However, differential regulation of macrophage polarization and reduced neutrophil function has been observed in Omicron infections. Interferon-induced antiviral immunity was not as strong in Omicron infections as in the original SARS-CoV-2 infections. The host response to Omicron infections increased antioxidant capacity and liver detoxification more than in the original strain. Hence, these findings suggest that Omicron infections cause weaker inflammatory alterations and immune responses than the original SARS-CoV-2 strain.
Collapse
Affiliation(s)
- Xue Li
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, 300350, China
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, 300350, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, 300350, China
| | - Yimeng Liu
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, 300350, China
| | - Guiying Xu
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, 300350, China
| | - Yi Xie
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, 300350, China
| | - Ximo Wang
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, 300350, China.
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ injury and ITCWM Repair, Tianjin, China.
| | - Junping Wu
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, 300350, China.
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, 300350, China.
- Department of Tuberculosis, Haihe Hospital, Tianjin University, Tianjin, 300350, China.
| | - Huaiyong Chen
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, 300350, China.
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China.
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, 300350, China.
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, 300350, China.
| |
Collapse
|
32
|
Le AT, Wu M, Khan A, Phillips N, Rajpurkar P, Garland M, Magid K, Sibai M, Huang C, Sahoo MK, Bowen R, Cowan TM, Pinsky BA, Hogan CA. Targeted plasma metabolomics combined with machine learning for the diagnosis of severe acute respiratory syndrome virus type 2. Front Microbiol 2023; 13:1059289. [PMID: 37063449 PMCID: PMC10092816 DOI: 10.3389/fmicb.2022.1059289] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/07/2022] [Indexed: 03/31/2023] Open
Abstract
IntroductionThe routine clinical diagnosis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is largely restricted to real-time reverse transcription quantitative PCR (RT-qPCR), and tests that detect SARS-CoV-2 nucleocapsid antigen. Given the diagnostic delay and suboptimal sensitivity associated with these respective methods, alternative diagnostic strategies are needed for acute infection.MethodsWe studied the use of a clinically validated liquid chromatography triple quadrupole method (LC/MS–MS) for detection of amino acids from plasma specimens. We applied machine learning models to distinguish between SARS-CoV-2-positive and negative samples and analyzed amino acid feature importance.ResultsA total of 200 samples were tested, including 70 from individuals with COVID-19, and 130 from negative controls. The top performing model overall allowed discrimination between SARS-CoV-2-positive and negative control samples with an area under the receiver operating characteristic curve (AUC) of 0.96 (95%CI 0.91, 1.00), overall sensitivity of 0.99 (95%CI 0.92, 1.00), and specificity of 0.92 (95%CI 0.85, 0.95).DiscussionThis approach holds potential as an alternative to existing methods for the rapid and accurate diagnosis of acute SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Anthony T. Le
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Manhong Wu
- Department of Anesthesiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Afraz Khan
- British Columbia Center for Disease Control Public Health Laboratory, Vancouver, BC, Canada
| | - Nicholas Phillips
- Stanford Computer Science Department, Stanford University, Stanford, CA, United States
| | - Pranav Rajpurkar
- Stanford Computer Science Department, Stanford University, Stanford, CA, United States
| | - Megan Garland
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Kayla Magid
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Mamdouh Sibai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - ChunHong Huang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Malaya K. Sahoo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Raffick Bowen
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Biochemical Genetics Laboratory, Stanford Health Care, Palo Alto, CA, United States
| | - Tina M. Cowan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
- Clinical Chemistry and Immunology Laboratory, Stanford Health Care, Palo Alto, CA, United States
| | - Benjamin A. Pinsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Clinical Virology Laboratory, Stanford Health Care, Palo Alto, CA, United States
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Catherine A. Hogan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
- British Columbia Center for Disease Control Public Health Laboratory, Vancouver, BC, Canada
- Stanford Clinical Virology Laboratory, Stanford Health Care, Palo Alto, CA, United States
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Catherine A. Hogan,
| |
Collapse
|
33
|
Berber E, Sumbria D, Kokkaya S. A metabolic blueprint of COVID-19 and long-term vaccine efficacy. Drug Metab Pers Ther 2023; 38:15-29. [PMID: 36166711 DOI: 10.1515/dmpt-2022-0148] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/24/2022] [Indexed: 06/16/2023]
Abstract
Viruses are obligatory protein-coated units and often utilize the metabolic functions of the cells they infect. Viruses hijack cellular metabolic functions and cause consequences that can range from minor to devastating, as we have all witnessed during the COVID-19 pandemic. For understanding the virus-driven pathogenesis and its implications on the host, the cellular metabolism needs to be elucidated. How SARS-CoV-2 triggers metabolic functions and rewires the metabolism remains unidentified but the implications of the metabolic patterns are under investigation by several researchers. In this review, we have described the SARS-CoV-2-mediated metabolic alterations from in vitro studies to metabolic changes reported in victims of COVID-19. We have also discussed potential therapeutic targets to diminish the viral infection and suppress the inflammatory response, with respect to evidenced studies based on COVID-19 research. Finally, we aimed to explain how we could extend vaccine-induced immunity in people by targeting the immunometabolism.
Collapse
Affiliation(s)
- Engin Berber
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - Deepak Sumbria
- College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University, Rampura Phul, Bathinda, India
| | - Serkan Kokkaya
- Faculty of Veterinary Medicine, Bozok University, Yozgat, Turkey
| |
Collapse
|
34
|
Gardinassi LG, Servian CDP, Lima GDS, dos Anjos DCC, Gomes Junior AR, Guilarde AO, Borges MASB, dos Santos GF, Moraes BGN, Silva JMM, Masson LC, de Souza FP, da Silva RR, de Araújo GL, Rodrigues MF, da Silva LC, Meira S, Fiaccadori FS, Souza M, Romão PRT, Spadafora Ferreira M, Coelho V, Chaves AR, Simas RC, Vaz BG, Fonseca SG. Integrated Metabolic and Inflammatory Signatures Associated with Severity of, Fatality of, and Recovery from COVID-19. Microbiol Spectr 2023; 11:e0219422. [PMID: 36852984 PMCID: PMC10100880 DOI: 10.1128/spectrum.02194-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 02/04/2023] [Indexed: 03/01/2023] Open
Abstract
Severe manifestations of coronavirus disease 2019 (COVID-19) and mortality have been associated with physiological alterations that provide insights into the pathogenesis of the disease. Moreover, factors that drive recovery from COVID-19 can be explored to identify correlates of protection. The cellular metabolism represents a potential target to improve survival upon severe disease, but the associations between the metabolism and the inflammatory response during COVID-19 are not well defined. We analyzed blood laboratorial parameters, cytokines, and metabolomes of 150 individuals with mild to severe disease, of which 33 progressed to a fatal outcome. A subset of 20 individuals was followed up after hospital discharge and recovery from acute disease. We used hierarchical community networks to integrate metabolomics profiles with cytokines and markers of inflammation, coagulation, and tissue damage. Infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) promotes significant alterations in the plasma metabolome, whose activity varies according to disease severity and correlates with oxygen saturation. Differential metabolism underlying death was marked by amino acids and related metabolites, such as glutamate, glutamyl-glutamate, and oxoproline, and lipids, including progesterone, phosphocholine, and lysophosphatidylcholines (lysoPCs). Individuals who recovered from severe disease displayed persistent alterations enriched for metabolism of purines and phosphatidylinositol phosphate and glycolysis. Recovery of mild disease was associated with vitamin E metabolism. Data integration shows that the metabolic response is a hub connecting other biological features during disease and recovery. Infection by SARS-CoV-2 induces concerted activity of metabolic and inflammatory responses that depend on disease severity and collectively predict clinical outcomes of COVID-19. IMPORTANCE COVID-19 is characterized by diverse clinical outcomes that include asymptomatic to mild manifestations or severe disease and death. Infection by SARS-CoV-2 activates inflammatory and metabolic responses that drive protection or pathology. How inflammation and metabolism communicate during COVID-19 is not well defined. We used high-resolution mass spectrometry to investigate small biochemical compounds (<1,500 Da) in plasma of individuals with COVID-19 and controls. Age, sex, and comorbidities have a profound effect on the plasma metabolites of individuals with COVID-19, but we identified significant activity of pathways and metabolites related to amino acids, lipids, nucleotides, and vitamins determined by disease severity, survival outcome, and recovery. Furthermore, we identified metabolites associated with acute-phase proteins and coagulation factors, which collectively identify individuals with severe disease or individuals who died of severe COVID-19. Our study suggests that manipulating specific metabolic pathways can be explored to prevent hyperinflammation, organ dysfunction, and death.
Collapse
Affiliation(s)
- Luiz Gustavo Gardinassi
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Carolina do Prado Servian
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Gesiane da Silva Lima
- Laboratório de Cromatografia e Espectrometria de Massas, Instituto de Química, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Déborah Carolina Carvalho dos Anjos
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Antonio Roberto Gomes Junior
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Adriana Oliveira Guilarde
- Departamento de Medicina Tropical e Dermatologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Moara Alves Santa Bárbara Borges
- Departamento de Medicina Tropical e Dermatologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Gabriel Franco dos Santos
- Laboratório de Cromatografia e Espectrometria de Massas, Instituto de Química, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | | | - João Marcos Maia Silva
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Letícia Carrijo Masson
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Flávia Pereira de Souza
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Rodolfo Rodrigues da Silva
- Laboratório de Cromatografia e Espectrometria de Massas, Instituto de Química, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Giovanna Lopes de Araújo
- Laboratório de Cromatografia e Espectrometria de Massas, Instituto de Química, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Marcella Ferreira Rodrigues
- Laboratório de Cromatografia e Espectrometria de Massas, Instituto de Química, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Lidya Cardozo da Silva
- Laboratório de Cromatografia e Espectrometria de Massas, Instituto de Química, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Sueli Meira
- Laboratório Prof Margarida Dobler Komma, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Fabiola Souza Fiaccadori
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Menira Souza
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Pedro Roosevelt Torres Romão
- Laboratório de Imunologia Celular e Molecular, Programa de Pós-Graduação em Ciências da Saúde, Programa de Pós-Graduação em Ciências da Reabilitação, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Verônica Coelho
- Laboratório de Imunologia, Instituto do Coração, Faculdade de Medicina, Universidade de São Paulo, São Paulo, São Paulo, Brazil
- Laboratório de Histocompatibilidade e Imunidade Celular, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, São Paulo, Brazil
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciências e Tecnologia, São Paulo, São Paulo, Brazil
| | - Andréa Rodrigues Chaves
- Laboratório de Cromatografia e Espectrometria de Massas, Instituto de Química, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Rosineide Costa Simas
- Laboratório de Cromatografia e Espectrometria de Massas, Instituto de Química, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Boniek Gontijo Vaz
- Laboratório de Cromatografia e Espectrometria de Massas, Instituto de Química, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Simone Gonçalves Fonseca
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciências e Tecnologia, São Paulo, São Paulo, Brazil
| |
Collapse
|
35
|
da Silva Fidalgo TK, Freitas-Fernandes LB, Marques BBF, de Araújo CS, da Silva BJ, Guimarães TC, Fischer RG, Tinoco EMB, Valente AP. Salivary Metabolomic Analysis Reveals Amino Acid Metabolism Shift in SARS-CoV-2 Virus Activity and Post-Infection Condition. Metabolites 2023; 13:metabo13020263. [PMID: 36837882 PMCID: PMC9962089 DOI: 10.3390/metabo13020263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/14/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
The SARS-CoV-2 virus primarily infects salivary glands suggesting a change in the saliva metabolite profile; this shift may be used as a monitoring instrument during SARS-CoV-2 infection. The present study aims to determine the salivary metabolomic profile of patients with and post-SARS-CoV-19 infection. Patients were without (PCR-), with SARS-CoV-2 (PCR+), or post-SARS-CoV-2 infection. Unstimulated whole saliva was collected, and the 1H spectra were acquired in a 500 MHz Bruker nuclear magnetic resonance spectrometer at 25 °C. They were subjected to multivariate analysis using principal component analysis (PCA) and partial least squares discriminant analysis (PLS-DA), as well as univariate analysis through t-tests (SPSS 20.0, IL, USA), with a significance level of p < 0.05. A distinction was found when comparing PCR- subjects to those with SARS-CoV-2 infection. When comparing the three groups, the PLS-DA cross-validation presented satisfactory accuracy (ACC = 0.69, R2 = 0.39, Q2 = 0.08). Seventeen metabolites were found in different proportions among the groups. The results suggested the downregulation of major amino acid levels, such as alanine, glutamine, histidine, leucine, lysine, phenylalanine, and proline in the PCR+ group compared to the PCR- ones. In addition, acetate, valerate, and capronic acid were higher in PCR- patients than in PCR+. Sucrose and butyrate were higher in post-SARS-CoV-2 infection compared to PCR-. In general, a reduction in amino acids was observed in subjects with and post-SARS-CoV-2 disease. The salivary metabolomic strategy NMR-based was able to differentiate between non-infected individuals and those with acute and post-SARS-CoV-19 infection.
Collapse
Affiliation(s)
- Tatiana Kelly da Silva Fidalgo
- Department of Preventive and Community Dentistry, School of Dentistry, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
- Correspondence: (T.K.d.S.F.); (A.P.V.)
| | - Liana Bastos Freitas-Fernandes
- National Center for Nuclear Magnetic Resonance, Medical Biochemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Barbara Bruno Fagundes Marques
- Department of Periodontology, School of Dentistry, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Caroline Souza de Araújo
- Department of Preventive and Community Dentistry, School of Dentistry, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Bruno Jefferson da Silva
- National Center for Nuclear Magnetic Resonance, Medical Biochemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Taísa Coelho Guimarães
- Department of Periodontology, School of Dentistry, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Ricardo Guimarães Fischer
- Department of Periodontology, School of Dentistry, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Eduardo Muniz Barretto Tinoco
- Department of Periodontology, School of Dentistry, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Ana Paula Valente
- National Center for Nuclear Magnetic Resonance, Medical Biochemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Correspondence: (T.K.d.S.F.); (A.P.V.)
| |
Collapse
|
36
|
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
|
37
|
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
|
38
|
Federica G, Giuseppina F, Veronica L, Gianpaolo Z, Massimo T, Veronica DM, Giuseppe S, Maria TA. An untargeted metabolomic approach to investigate antiviral defence mechanisms in memory leukocytes secreting anti-SARS-CoV-2 IgG in vitro. Sci Rep 2023; 13:629. [PMID: 36635345 PMCID: PMC9835734 DOI: 10.1038/s41598-022-26156-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 12/12/2022] [Indexed: 01/13/2023] Open
Abstract
Evidence shows that individuals infected by SARS-CoV-2 experience an altered metabolic state in multiple organs. Metabolic activities are directly involved in modulating immune responses against infectious diseases, yet our understanding of how host metabolism relates to inflammatory responses remains limited. To better elucidate the underlying biochemistry of the leukocyte response, we focused our analysis on possible relationships between SARS-CoV-2 post-infection stages and distinct metabolic pathways. Indeed, we observed a significant altered metabolism of tryptophan and urea cycle pathways in cultures of peripheral blood mononuclear cells obtained 60-90 days after infection and showing in vitro IgG antibody memory for spike-S1 antigen (n = 17). This work, for the first time, identifies metabolic routes in cell metabolism possibly related to later stages of immune defence against SARS-CoV-2 infection, namely, when circulating antibodies may be absent but an antibody memory is present. The results suggest reprogramming of leukocyte metabolism after viral pathogenesis through activation of specific amino acid pathways possibly related to protective immunity against SARS-CoV-2.
Collapse
Affiliation(s)
- Gevi Federica
- grid.12597.380000 0001 2298 9743Department of Ecological and Biological Sciences, University of Tuscia, 01100 Viterbo, Italy
| | - Fanelli Giuseppina
- grid.12597.380000 0001 2298 9743Department of Ecological and Biological Sciences, University of Tuscia, 01100 Viterbo, Italy
| | - Lelli Veronica
- grid.12597.380000 0001 2298 9743Department of Ecological and Biological Sciences, University of Tuscia, 01100 Viterbo, Italy
| | - Zarletti Gianpaolo
- grid.12597.380000 0001 2298 9743Department of Innovative Biology, Agro-Food and Forestry, University of Tuscia, 01100 Viterbo, Italy
| | - Tiberi Massimo
- grid.12597.380000 0001 2298 9743Department of Innovative Biology, Agro-Food and Forestry, University of Tuscia, 01100 Viterbo, Italy
| | - De Molfetta Veronica
- grid.12597.380000 0001 2298 9743Department of Innovative Biology, Agro-Food and Forestry, University of Tuscia, 01100 Viterbo, Italy
| | - Scapigliati Giuseppe
- Department of Innovative Biology, Agro-Food and Forestry, University of Tuscia, 01100, Viterbo, Italy.
| | - Timperio Anna Maria
- Department of Ecological and Biological Sciences, University of Tuscia, 01100, Viterbo, Italy.
| |
Collapse
|
39
|
Gietl M, Burkert F, Seiwald S, Böhm A, Hofer S, Gostner JM, Piater T, Geisler S, Weiss G, Loeffler-Ragg J, Sonnweber T, Tancevski I, Pizzini A, Sahanic S, Fuchs D, Bellmann-Weiler R, Kurz K. Interferon-gamma Mediated Metabolic Pathways in Hospitalized Patients During Acute and Reconvalescent COVID-19. Int J Tryptophan Res 2023; 16:11786469231154244. [PMID: 37038445 PMCID: PMC10076985 DOI: 10.1177/11786469231154244] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/15/2023] [Indexed: 02/15/2023] Open
Abstract
Background: Fatigue, sleep disturbance, and neurological symptoms during and after COVID-19 are common and might be associated with inflammation-induced changes in tryptophan (Trp) and phenylalanine (Phe) metabolism. Aim: This pilot study investigated interferon gamma inducible biochemical pathways (namely Trp catabolism, neopterin, tyrosine [Tyr], and nitrite formation) during acute COVID-19 and reconvalescence. Patients and methods: Thirty one patients with moderate to severe COVID-19 admitted to the University Hospital of Innsbruck in early 2020 (March-May) were followed up. Neurotransmitter precursors Trp, Phe, Tyr as well as kynurenine (Kyn), neopterin, nitrite, and routine laboratory parameters were analyzed during acute infection and at a follow-up (FU) 60 days thereafter. Clinical symptoms of patients (neurological symptoms, fatigue, sleep disturbance) were recorded and associations with concentrations of laboratory parameters investigated. Results and conclusion: Almost half of the patients suffered from neurological symptoms (48.4%), the majority of patients experienced sleep difficulties (56.7%) during acute COVID-19. Fatigue was present in nearly all patients. C-reactive protein (CRP), interleukin-6 (IL-6), neopterin, Kyn, Phe concentrations were significantly increased, and Trp levels depleted during acute COVID-19. Patients with sleep impairment and neurological symptoms during acute illness presented with increased CRP and IL-6 concentrations, Trp levels were lower in patients with sleep disturbance. In general, inflammatory markers declined during reconvalescence. A high percentage of patients suffered from persistent symptoms at FU (neurological symptoms: 17.2%, fatigue: 51.7%, sleeping disturbance: 34.5%) and had higher CRP concentrations. Nitrite and Phe levels were lower in patients with sleeping difficulties at FU and Kyn/Trp ratio, as indicator of IDO activity, was significantly lower in patients with neurological symptoms compared to patients without them at FU. In summary, inflammation induced alterations of amino acid metabolism might be related to acute and persisting symptoms of COVID-19.
Collapse
Affiliation(s)
- Mario Gietl
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Francesco Burkert
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Stefanie Seiwald
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Anna Böhm
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Stefanie Hofer
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Johanna M Gostner
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Talia Piater
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Simon Geisler
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Guenter Weiss
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Judith Loeffler-Ragg
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Thomas Sonnweber
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Alex Pizzini
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Sabina Sahanic
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Dietmar Fuchs
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Rosa Bellmann-Weiler
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| | - Katharina Kurz
- Department of Internal Medicine II, Medical University Innsbruck, Biocentre, Medical Biochemistry, Innsbruck, Austria
| |
Collapse
|
40
|
|
41
|
A Time-Series Metabolomic Analysis of SARS-CoV-2 Infection in a Ferret Model. Metabolites 2022; 12:metabo12111151. [DOI: 10.3390/metabo12111151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 11/23/2022] Open
Abstract
The global threat of COVID-19 has led to an increased use of metabolomics to study SARS-CoV-2 infections in animals and humans. In spite of these efforts, however, understanding the metabolome of SARS-CoV-2 during an infection remains difficult and incomplete. In this study, metabolic responses to a SAS-CoV-2 challenge experiment were studied in nasal washes collected from an asymptomatic ferret model (n = 20) at different time points before and after infection using an LC-MS-based metabolomics approach. A multivariate analysis of the nasal wash metabolome data revealed several statistically significant features. Despite no effects of sex or interaction between sex and time on the time course of SARS-CoV-2 infection, 16 metabolites were significantly different at all time points post-infection. Among these altered metabolites, the relative abundance of taurine was elevated post-infection, which could be an indication of hepatotoxicity, while the accumulation of sialic acids could indicate SARS-CoV-2 invasion. Enrichment analysis identified several pathways influenced by SARS-CoV-2 infection. Of these, sugar, glycan, and amino acid metabolisms were the key altered pathways in the upper respiratory channel during infection. These findings provide some new insights into the progression of SARS-CoV-2 infection in ferrets at the metabolic level, which could be useful for the development of early clinical diagnosis tools and new or repurposed drug therapies.
Collapse
|
42
|
Ma Q, Ma W, Song TZ, Wu Z, Liu Z, Hu Z, Han JB, Xu L, Zeng B, Wang B, Sun Y, Yu DD, Wu Q, Yao YG, Zheng YT, Wang X. Single-nucleus transcriptomic profiling of multiple organs in a rhesus macaque model of SARS-CoV-2 infection. Zool Res 2022; 43:1041-1062. [PMID: 36349357 PMCID: PMC9700497 DOI: 10.24272/j.issn.2095-8137.2022.443] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 11/09/2022] Open
Abstract
Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes diverse clinical manifestations and tissue injuries in multiple organs. However, cellular and molecular understanding of SARS-CoV-2 infection-associated pathology and immune defense features in different organs remains incomplete. Here, we profiled approximately 77 000 single-nucleus transcriptomes of the lung, liver, kidney, and cerebral cortex in rhesus macaques ( Macaca mulatta) infected with SARS-CoV-2 and healthy controls. Integrated analysis of the multi-organ dataset suggested that the liver harbored the strongest global transcriptional alterations. We observed prominent impairment in lung epithelial cells, especially in AT2 and ciliated cells, and evident signs of fibrosis in fibroblasts. These lung injury characteristics are similar to those reported in patients with coronavirus disease 2019 (COVID-19). Furthermore, we found suppressed MHC class I/II molecular activity in the lung, inflammatory response in the liver, and activation of the kynurenine pathway, which induced the development of an immunosuppressive microenvironment. Analysis of the kidney dataset highlighted tropism of tubule cells to SARS-CoV-2, and we found membranous nephropathy (an autoimmune disease) caused by podocyte dysregulation. In addition, we identified the pathological states of astrocytes and oligodendrocytes in the cerebral cortex, providing molecular insights into COVID-19-related neurological implications. Overall, our multi-organ single-nucleus transcriptomic survey of SARS-CoV-2-infected rhesus macaques broadens our understanding of disease features and antiviral immune defects caused by SARS-CoV-2 infection, which may facilitate the development of therapeutic interventions for COVID-19.
Collapse
Affiliation(s)
- Qiang Ma
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenji Ma
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Tian-Zhang Song
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Kunming National High-Level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Zhaobo Wu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zeyuan Liu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhenxiang Hu
- LivzonBio, Inc., Zhuhai, Guangdong 519045, China
| | - Jian-Bao Han
- Kunming National High-Level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Ling Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Kunming National High-Level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Bo Zeng
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Bosong Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Yinuo Sun
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Dan-Dan Yu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Kunming National High-Level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Qian Wu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Kunming National High-Level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China. E-mail:
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Kunming National High-Level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China. E-mail:
| | - Xiaoqun Wang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
- Advanced Innovation Center for Human Brain Protection, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China. E-mail:
| |
Collapse
|
43
|
Moura AV, de Oliveira DC, Silva AAR, da Rosa JR, Garcia PHD, Sanches PHG, Garza KY, Mendes FMM, Lambert M, Gutierrez JM, Granado NM, dos Santos AC, de Lima IL, Negrini LDDO, Antonio MA, Eberlin MN, Eberlin LS, Porcari AM. Urine Metabolites Enable Fast Detection of COVID-19 Using Mass Spectrometry. Metabolites 2022; 12:1056. [PMID: 36355139 PMCID: PMC9697918 DOI: 10.3390/metabo12111056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/19/2022] [Accepted: 10/27/2022] [Indexed: 08/27/2023] Open
Abstract
The COVID-19 pandemic boosted the development of diagnostic tests to meet patient needs and provide accurate, sensitive, and fast disease detection. Despite rapid advancements, limitations related to turnaround time, varying performance metrics due to different sampling sites, illness duration, co-infections, and the need for particular reagents still exist. As an alternative diagnostic test, we present urine analysis through flow-injection-tandem mass spectrometry (FIA-MS/MS) as a powerful approach for COVID-19 diagnosis, targeting the detection of amino acids and acylcarnitines. We adapted a method that is widely used for newborn screening tests on dried blood for urine samples in order to detect metabolites related to COVID-19 infection. We analyzed samples from 246 volunteers with diagnostic confirmation via PCR. Urine samples were self-collected, diluted, and analyzed with a run time of 4 min. A Lasso statistical classifier was built using 75/25% data for training/validation sets and achieved high diagnostic performances: 97/90% sensitivity, 95/100% specificity, and 95/97.2% accuracy. Additionally, we predicted on two withheld sets composed of suspected hospitalized/symptomatic COVID-19-PCR negative patients and patients out of the optimal time-frame collection for PCR diagnosis, with promising results. Altogether, we show that the benchmarked FIA-MS/MS method is promising for COVID-19 screening and diagnosis, and is also potentially useful after the peak viral load has passed.
Collapse
Affiliation(s)
- Alexandre Varao Moura
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Danilo Cardoso de Oliveira
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Alex Ap. R. Silva
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Jonas Ribeiro da Rosa
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Pedro Henrique Dias Garcia
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Pedro Henrique Godoy Sanches
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Kyana Y. Garza
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA
| | - Flavio Marcio Macedo Mendes
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Mayara Lambert
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Junier Marrero Gutierrez
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Nicole Marino Granado
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| | - Alicia Camacho dos Santos
- Department of Material Engineering and Nanotechnology, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil
| | - Iasmim Lopes de Lima
- Department of Material Engineering and Nanotechnology, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil
| | | | - Marcia Aparecida Antonio
- Integrated Unit of Pharmacology and Gastroenterology, UNIFAG, Bragança Paulista 12916-900, SP, Brazil
| | - Marcos N. Eberlin
- Department of Material Engineering and Nanotechnology, Mackenzie Presbyterian University, São Paulo 01302-907, SP, Brazil
| | - Livia S. Eberlin
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Andreia M. Porcari
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil
| |
Collapse
|
44
|
Guaraldi G, Milic J, Cesari M, Leibovici L, Mandreoli F, Missier P, Rozzini R, Cattelan AM, Motta F, Mussini C, Cossarizza A. The interplay of post-acute COVID-19 syndrome and aging: a biological, clinical and public health approach. Ageing Res Rev 2022; 81:101686. [PMID: 35820609 PMCID: PMC9270773 DOI: 10.1016/j.arr.2022.101686] [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: 04/07/2022] [Accepted: 07/07/2022] [Indexed: 01/31/2023]
Abstract
The post-acute COVID-19 syndrome (PACS) is characterized by the persistence of fluctuating symptoms over three months from the onset of the possible or confirmed COVID-19 acute phase. Current data suggests that at least 10% of people with previously documented infection may develop PACS, and up to 50-80% of prevalence is reported among survivors after hospital discharge. This viewpoint will discuss various aspects of PACS, particularly in older adults, with a specific hypothesis to describe PACS as the expression of a modified aging trajectory induced by SARS CoV-2. This hypothesis will be argued from biological, clinical and public health view, addressing three main questions: (i) does SARS-CoV-2-induced alterations in aging trajectories play a role in PACS?; (ii) do people with PACS face immuno-metabolic derangements that lead to increased susceptibility to age-related diseases?; (iii) is it possible to restore the healthy aging trajectory followed by the individual before pre-COVID?. A particular focus will be given to the well-being of people with PACS that could be assessed by the intrinsic capacity model and support the definition of the healthy aging trajectory.
Collapse
Affiliation(s)
- Giovanni Guaraldi
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy,Department of Infectious Diseases, Azienda Ospedaliero-Universitaria, Policlinico of Modena, Modena, Italy,Correspondence to: Department of Surgical, Medical, Dental and Morphological Sciences University of Modena and Reggio Emilia, Largo del Pozzo, 71, 41124 Modena, Italy
| | - Jovana Milic
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Matteo Cesari
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | - Federica Mandreoli
- Department of Physical, Computer and Mathematical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Paolo Missier
- School of Computing, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Renzo Rozzini
- Geriatric Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | | | - Federico Motta
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Mussini
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy,Department of Infectious Diseases, Azienda Ospedaliero-Universitaria, Policlinico of Modena, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
45
|
Occelli C, Guigonis JM, Lindenthal S, Cagnard A, Graslin F, Brglez V, Seitz-Polski B, Dellamonica J, Levraut J, Pourcher T. Untargeted plasma metabolomic fingerprinting highlights several biomarkers for the diagnosis and prognosis of coronavirus disease 19. Front Med (Lausanne) 2022; 9:995069. [PMID: 36250098 PMCID: PMC9556858 DOI: 10.3389/fmed.2022.995069] [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: 07/18/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectivesThe COVID-19 pandemic has been a serious worldwide public health crisis since 2020 and is still challenging healthcare systems. New tools for the prognosis and diagnosis of COVID-19 patients remain important issues.DesignHere, we studied the metabolome of plasma samples of COVID-19 patients for the identification of prognosis biomarkers.PatientsPlasma samples of eighty-six SARS-CoV-2-infected subjects and 24 healthy controls were collected during the first peak of the COVID-19 pandemic in France in 2020.Main resultsPlasma metabolome fingerprinting allowed the successful discrimination of healthy controls, mild SARS-CoV-2 subjects, and moderate and severe COVID-19 patients at hospital admission. We found a strong effect of SARS-CoV-2 infection on the plasma metabolome in mild cases. Our results revealed that plasma lipids and alterations in their saturation level are important biomarkers for the detection of the infection. We also identified deoxy-fructosyl-amino acids as new putative plasma biomarkers for SARS-CoV-2 infection and COVID-19 severity. Finally, our results highlight a key role for plasma levels of tryptophan and kynurenine in the symptoms of COVID-19 patients.ConclusionOur results showed that plasma metabolome profiling is an efficient tool for the diagnosis and prognosis of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Céline Occelli
- Transporter in Imaging and Radiotherapy in Oncology Laboratory (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Frederic Joliot, Commissariat a l’Energie Atomique et aux Energies Alternatives (CEA), Université Côte d’Azur, School of Medicine, Nice, France
- Department of Emergency, University Hospital, Nice, France
- School of Medicine, Université Côte d’Azur, Nice, France
| | - Jean-Marie Guigonis
- Transporter in Imaging and Radiotherapy in Oncology Laboratory (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Frederic Joliot, Commissariat a l’Energie Atomique et aux Energies Alternatives (CEA), Université Côte d’Azur, School of Medicine, Nice, France
| | - Sabine Lindenthal
- Transporter in Imaging and Radiotherapy in Oncology Laboratory (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Frederic Joliot, Commissariat a l’Energie Atomique et aux Energies Alternatives (CEA), Université Côte d’Azur, School of Medicine, Nice, France
| | - Alexandre Cagnard
- Transporter in Imaging and Radiotherapy in Oncology Laboratory (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Frederic Joliot, Commissariat a l’Energie Atomique et aux Energies Alternatives (CEA), Université Côte d’Azur, School of Medicine, Nice, France
| | - Fanny Graslin
- Transporter in Imaging and Radiotherapy in Oncology Laboratory (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Frederic Joliot, Commissariat a l’Energie Atomique et aux Energies Alternatives (CEA), Université Côte d’Azur, School of Medicine, Nice, France
| | - Vesna Brglez
- Unité de Recherche Clinique Côte d’Azur (UR2CA), Université Côte d’Azur, Nice, France
- Department of Immunology, University Hospital, Nice, France
| | - Barbara Seitz-Polski
- School of Medicine, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique Côte d’Azur (UR2CA), Université Côte d’Azur, Nice, France
- Department of Immunology, University Hospital, Nice, France
| | - Jean Dellamonica
- School of Medicine, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique Côte d’Azur (UR2CA), Université Côte d’Azur, Nice, France
- Medical Intensive Care Unit, University Hospital, Nice, France
| | - Jacques Levraut
- Department of Emergency, University Hospital, Nice, France
- School of Medicine, Université Côte d’Azur, Nice, France
| | - Thierry Pourcher
- Transporter in Imaging and Radiotherapy in Oncology Laboratory (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des Sciences du Vivant Frederic Joliot, Commissariat a l’Energie Atomique et aux Energies Alternatives (CEA), Université Côte d’Azur, School of Medicine, Nice, France
- *Correspondence: Thierry Pourcher,
| |
Collapse
|
46
|
Kynurenine Pathway of Tryptophan Metabolism Is Associated with Hospital Mortality in Patients with Acute Respiratory Distress Syndrome: A Prospective Cohort Study. Antioxidants (Basel) 2022; 11:antiox11101884. [PMID: 36290606 PMCID: PMC9598717 DOI: 10.3390/antiox11101884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) involves dysregulated immune-inflammatory responses, characterized by severe oxidative stress and high mortality. Metabolites modulating the inflammatory and immune responses may play a central role in the pathogenesis of ARDS. Most biogenic amines may induce the production of reactive oxygen species, oxidative stress, mitochondrial dysfunction, and programmed cell death. We conducted a prospective study on metabolic profiling specific to the amino acids and biogenic amines of 69 patients with ARDS. Overall, hospital mortality was 52.2%. Between day 1 and day 7 after ARDS onset, plasma kynurenine levels and the kynurenine/tryptophan ratio were significantly higher among non-survivors than in survivors (all p < 0.05). Urine metabolic profiling revealed a significantly higher prevalence of tryptophan degradation and higher concentrations of metabolites downstream of the kynurenine pathway among non-survivors than among survivors upon ARDS onset. Cox regression models revealed that plasma kynurenine levels and the plasma kynurenine/tryptophan ratio on day 1 were independently associated with hospital mortality. The activation of the kynurenine pathway was associated with mortality in patients with ARDS. Metabolic phenotypes and modulating metabolic perturbations of the kynurenine pathway could perhaps serve as prognostic markers or as a target for therapeutic interventions aimed at reducing oxidative stress and mortality in ARDS.
Collapse
|
47
|
Bizjak DA, Stangl M, Börner N, Bösch F, Durner J, Drunin G, Buhl JL, Abendroth D. Kynurenine serves as useful biomarker in acute, Long- and Post-COVID-19 diagnostics. Front Immunol 2022; 13:1004545. [PMID: 36211365 PMCID: PMC9537769 DOI: 10.3389/fimmu.2022.1004545] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/06/2022] [Indexed: 12/02/2022] Open
Abstract
Introduction In patients with SARS-CoV-2, innate immunity is playing a central role, depicted by hyperinflammation and longer lasting inflammatory response. Reliable inflammatory markers that cover both acute and long-lasting COVID-19 monitoring are still lacking. Thus, we investigated one specific inflammatory marker involved as one key player of the immune system, kynurenine (Kyn), and its use for diagnosis/detection of the Long-/Post-COVID syndrome in comparison to currently used markers in both serum and saliva samples. Material and methods The study compromised in total 151 inpatients with a SARS-CoV-2 infection hospitalized between 03/2020 and 09/2021. The group NC (normal controls) included blood bank donors (n=302, 144f/158m, mean age 47.1 ± 18.3 years (range 18-75)). Two further groups were generated based on Group A (n=85, 27f/58m, mean age 63.1 ± 18.3 years (range 19-90), acute admission to the hospital) and Group B (n=66, 22f/44m, mean age 66.6 ± 17.6 years (range 17-90), admitted either for weaning or for rehabilitation period due to Long-COVID symptoms/syndrome). Plasma concentrations of Kyn, C-Reactive Protein (CRP) and interleukin-6 (IL-6) were measured on admission. In Group B we determined Kyn 4 weeks after the negative PCR-test. In a subset of patients (n=11) concentrations of Kyn and CRP were measured in sera and saliva two, three and four months after dismission. We identified 12 patients with Post-COVID symptoms >20 weeks with still significant elevated Kyn-levels. Results Mean values for NC used as reference were 2.79 ± 0.61 µM, range 1.2-4.1 µM. On admission, patients showed significantly higher concentrations of Kyn compared to NC (p-values < 0.001). Kyn significantly correlated with IL-6 peak-values (r=0.411; p-values <0.001) and CRP (r=0.488, p-values<0.001). Kyn values in Group B (Long-/Post-COVID) showed still significant higher values (8.77 ± 1.72 µM, range 5.5-16.6 µM), whereas CRP values in Group B were in the normal range. Conclusion Serum and saliva Kyn are reflecting the acute and long-term pathophysiology of the SARS-CoV-2 disease concerning the innate immune response and thus may serve a useful biomarker for diagnosis and monitoring both Long- and Post-COVID syndrome and its therapy.
Collapse
Affiliation(s)
| | - Manfred Stangl
- Division of General, Visceral and Transplant Surgery, Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Nikolaus Börner
- Division of General, Visceral and Transplant Surgery, Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Florian Bösch
- Division of General, Visceral and Transplant Surgery, Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Joachim Durner
- Neurology Department, Special Medical Clinic Ichenhausen, Ichenhausen, Germany
| | - Gergana Drunin
- Neurology Department, Special Medical Clinic Ichenhausen, Ichenhausen, Germany
| | - Jasmine-Leonike Buhl
- Division of Sports and Rehabilitation Medicine, Ulm University Hospital, Ulm, Germany
| | | |
Collapse
|
48
|
Páez-Franco JC, Maravillas-Montero JL, Mejía-Domínguez NR, Torres-Ruiz J, Tamez-Torres KM, Pérez-Fragoso A, Germán-Acacio JM, Ponce-de-León A, Gómez-Martín D, Ulloa-Aguirre A. Metabolomics analysis identifies glutamic acid and cystine imbalances in COVID-19 patients without comorbid conditions. Implications on redox homeostasis and COVID-19 pathophysiology. PLoS One 2022; 17:e0274910. [PMID: 36126080 PMCID: PMC9488784 DOI: 10.1371/journal.pone.0274910] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 09/06/2022] [Indexed: 11/18/2022] Open
Abstract
It is well known that the presence of comorbidities and age-related health issues may hide biochemical and metabolic features triggered by SARS-CoV-2 infection and other diseases associated to hypoxia, as they are by themselves chronic inflammatory conditions that may potentially disturb metabolic homeostasis and thereby negatively impact on COVID-19 progression. To unveil the metabolic abnormalities inherent to hypoxemia caused by COVID-19, we here applied gas chromatography coupled to mass spectrometry to analyze the main metabolic changes exhibited by a population of male patients less than 50 years of age with mild/moderate and severe COVID-19 without pre-existing comorbidities known to predispose to life-threatening complications from this infection. Several differences in serum levels of particular metabolites between normal controls and patients with COVID-19 as well as between mild/moderate and severe COVID-19 were identified. These included increased glutamic acid and reduced glutamine, cystine, threonic acid, and proline levels. In particular, using the entire metabolomic fingerprint obtained, we observed that glutamine/glutamate metabolism was associated with disease severity as patients in the severe COVID-19 group presented the lowest and higher serum levels of these amino acids, respectively. These data highlight the hypoxia-derived metabolic alterations provoked by SARS-CoV-2 infection in the absence of pre-existing co-morbidities as well as the value of amino acid metabolism in determining reactive oxygen species recycling pathways, which when impaired may lead to increased oxidation of proteins and cell damage. They also provide insights on new supportive therapies for COVID-19 and other disorders that involve altered redox homeostasis and lower oxygen levels that may lead to better outcomes of disease severity.
Collapse
Affiliation(s)
- José C. Páez-Franco
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José L. Maravillas-Montero
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Nancy R. Mejía-Domínguez
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jiram Torres-Ruiz
- Emergency Medicine Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Karla M. Tamez-Torres
- Department of Infectology and Microbiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Alfredo Pérez-Fragoso
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Juan Manuel Germán-Acacio
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Alfredo Ponce-de-León
- Department of Infectology and Microbiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Diana Gómez-Martín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
49
|
Lesani M, Gosmanov C, Paun A, Lewis MD, McCall LI. Impact of Visceral Leishmaniasis on Local Organ Metabolism in Hamsters. Metabolites 2022; 12:metabo12090802. [PMID: 36144206 PMCID: PMC9506185 DOI: 10.3390/metabo12090802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Leishmania is an intracellular parasite with different species pathogenic to humans and causing the disease leishmaniasis. Leishmania donovani causes visceral leishmaniasis (VL) that manifests as hepatosplenomegaly, fever, pancytopenia and hypergammaglobulinemia. If left without treatment, VL can cause death, especially in immunocompromised people. Current treatments have often significant adverse effects, and resistance has been reported in some countries. Determining the metabolites perturbed during VL can lead us to find new treatments targeting disease pathogenesis. We therefore compared metabolic perturbation between L. donovani-infected and uninfected hamsters across organs (spleen, liver, and gut). Metabolites were extracted, analyzed by liquid chromatography-mass spectrometry, and processed with MZmine and molecular networking to annotate metabolites. We found few metabolites commonly impacted by infection across all three sites, including glycerophospholipids, ceramides, acylcarnitines, peptides, purines and amino acids. In accordance with VL symptoms and parasite tropism, we found a greater overlap of perturbed metabolites between spleen and liver compared to spleen and gut, or liver and gut. Targeting pathways related to these metabolite families would be the next focus that can lead us to find more effective treatments for VL.
Collapse
Affiliation(s)
- Mahbobeh Lesani
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK 73019, USA
| | - Camil Gosmanov
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK 73019, USA
| | - Andrea Paun
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Michael D. Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Laura-Isobel McCall
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK 73019, USA
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK 73019, USA
- Correspondence:
| |
Collapse
|
50
|
Bornstein SR, Cozma D, Kamel M, Hamad M, Mohammad MG, Khan NA, Saber MM, Semreen MH, Steenblock C. Long-COVID, Metabolic and Endocrine Disease. Horm Metab Res 2022; 54:562-566. [PMID: 35724687 PMCID: PMC9363148 DOI: 10.1055/a-1878-9307] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In the aftermath of the corona pandemic, long-COVID or post-acute COVID-19 syndrome still represents a great challenge, and this topic will continue to represent a significant health problem in the coming years. At present, the impact of long-COVID on our health system cannot be fully assessed but according to current studies, up to 40% of people who have been infected with SARS-CoV-2 suffer from clinically relevant symptoms of long-COVID syndrome several weeks to months after the acute phase. The main symptoms are chronic fatigue, dyspnea, and various cognitive symptoms. Initial studies have shown that people with overweight and diabetes mellitus have a higher risk of developing long-COVID associated symptoms. Furthermore, repeated treatment of acute COVID-19 and long-COVID with steroids can contribute to long-term metabolic and endocrine disorders. Therefore, a structured program with rehabilitation and physical activity as well as optimal dietary management is of utmost importance, especially for patients with metabolic diseases and/or long-COVID. Furthermore, the removal of autoantibodies and specific therapeutic apheresis procedures could lead to a significant improvement in the symptoms of long-COVID in individual patients.
Collapse
Affiliation(s)
- Stefan R. Bornstein
- Department of Medicine III, Universitätsklinikum Carl Gustav
Carus, Dresden, Germany
- School of Cardiovascular and Metabolic Medicine and Sciences, Faculty
of Life Sciences & Medicine, King’s College London, London,
UK
| | - Diana Cozma
- Department of Medicine III, Universitätsklinikum Carl Gustav
Carus, Dresden, Germany
| | - Margrit Kamel
- Center for Regenerative Therapies Dresden, Technische
Universität Dresden, Dresden, Germany
| | - Mawieh Hamad
- Sharjah Institute for Medical Research, University of Sharjah College
of Health Sciences, Sharjah, United Arab Emirates
| | - Mohammad G. Mohammad
- Department of Medical Laboratory Sciences, College of Health Sciences,
University of Sharjah, Sharjah, United Arab Emirates
| | - Naveed A. Khan
- College of Medicine, University of Sharjah, Sharjah, United Arab
Emirates
| | - Maha M. Saber
- College of Medicine, University of Sharjah, Sharjah, United Arab
Emirates
| | | | - Charlotte Steenblock
- Department of Medicine III, Universitätsklinikum Carl Gustav
Carus, Dresden, Germany
- Correspondence Dr. Charlotte
Steenblock University Hospital Carl Gustav Carus,
Technische Universität DresdenDepartment of
Medicine IIIFetscherstraße
7401307
DresdenGermany+493514586130+493514586336
| |
Collapse
|