1
|
Madrid-Gambin F, Haro N, Mason NL, Mallaroni P, Theunissen EL, Toennes SW, Pozo OJ, Ramaekers JG. Metabolomic profiling of cannabis use and cannabis intoxication in humans. Neuropsychopharmacology 2025; 50:920-927. [PMID: 40074870 PMCID: PMC12032370 DOI: 10.1038/s41386-025-02082-7] [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: 12/10/2024] [Revised: 02/14/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025]
Abstract
Acute intoxication from Δ9-tetrahydrocannabinol (THC, the primary active ingredient of cannabis) can lead to neurocognitive impairment and interference with day-to-day operations, such as driving. Present evaluations of THC-induced impairment in legal settings rely on biological drug tests that solely establish cannabis use, rather than cannabis impairment. The current study evaluated the metabolome in blood collected from occasional and chronic cannabis users (N = 35) at baseline and following treatments with cannabis (300 μg/kg THC) and placebo, with the aim to identify unique metabolic alterations that are associated with acute cannabis intoxication and cannabis use frequency. Blood samples were collected at baseline and repeatedly during 70 min after treatment. Sustained attention performance and ratings of subjective high were taken twice within 40 min after treatment. Metabolomic fingerprints of occasional and chronic cannabis users were distinctly different at baseline, when both groups were not intoxicated. A total of 14 metabolites, mainly related to endocannabinoid and amino acid metabolism, were identified that distinguished chronic from occasional cannabis users and that yielded a discriminant analysis model with an 80% classification rate (95% CI: 61-91%). Distinct metabolomic fingerprints were found for occasional cannabis users who, in contrast to chronic cannabis users, showed attentional impairment and elevated ratings of subjective high during cannabis intoxication. These included increments in organic acids, β-hydroxybutyrate and second messenger ceramides. The current study demonstrates the feasibility of the metabolomics approach to identify metabolic changes that are specific to the neurocognitive state of cannabis intoxication and to the history of cannabis use.
Collapse
Affiliation(s)
- Francisco Madrid-Gambin
- Applied Metabolomics Research Group, Hospital del Mar Research Institute, 08003, Barcelona, Spain
| | - Noemí Haro
- Applied Metabolomics Research Group, Hospital del Mar Research Institute, 08003, Barcelona, Spain
| | - Natasha L Mason
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, 6200 MD, Maastricht, the Netherlands
| | - Pablo Mallaroni
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, 6200 MD, Maastricht, the Netherlands
| | - Eef L Theunissen
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, 6200 MD, Maastricht, the Netherlands
| | - Stefan W Toennes
- Goethe University Frankfurt, University Hospital, Institute of Legal Medicine, Frankfurt, Germany
| | - Oscar J Pozo
- Applied Metabolomics Research Group, Hospital del Mar Research Institute, 08003, Barcelona, Spain
| | - Johannes G Ramaekers
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, 6200 MD, Maastricht, the Netherlands.
| |
Collapse
|
2
|
Khymenets O, Vilarroya O, Benet G, Feixas G, Arranz Betegon A, McLeod MD, Pozo OJ. Profile of steroid metabolites in human breast milk in different stages of lactation. Food Funct 2025. [PMID: 40277187 PMCID: PMC12023736 DOI: 10.1039/d4fo05713a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/02/2025] [Indexed: 04/26/2025]
Abstract
Breast milk (BM), as an optimal food, provides the newborn with a variety of minor compounds relevant for health and wellbeing. Endogenous steroids, also minor constituents, are mainly secreted in BM as conjugated metabolites. Recent research has revealed the relevance of steroid conjugates in many physiological processes. Thus, their presence in BM appears to be very intriguing, especially in relation to breastfeeding. The objective of our study was to profile conjugated steroid metabolites present in BM in relation to the lactation stage, and to promote further evaluation of their importance in breastfeeding. For this purpose, we developed and used a direct UHPLC-MS/MS metabolomics approach capable to detect more than 60 conjugated metabolites (mono-sulfated, mono-glucuronylated, bis-sulfated and sulfate-glucuronylated) from all steroid families. We compared the occurrence of these metabolites in samples collected from breastfeeding mothers and stratified by lactation stages: colostrum, transitional and mature milk. Our results showed that many biologically relevant conjugated steroids are secreted in BM. Their concentrations were highest in colostrum, decreased remarkably in transitional and were much lower in mature milk, with some exceptions. The profile of metabolites also differed considerably between lactation stages. The approximate daily secretion in BM indicated that infants are exposed to significant oral doses of steroid conjugates during the first week of lactation. The supply of these metabolites in BM declined and became constant after the second week postpartum. Overall, our data provide a foundation for further investigation on the physiological relevance of BM secreted steroid metabolites in relation to both mother and child.
Collapse
Affiliation(s)
- Olha Khymenets
- Applied Metabolomics Research Group, Neurosciences Research Programme, Hospital del Mar Research Institute, Barcelona, Spain.
| | - Oscar Vilarroya
- Department of Psychiatry and Legal Medicine, Autonomous University of Barcelona, Barcelona, Spain
- Neuroimaging Research Group, Neurosciences Programme, Hospital del Mar Research Institute, Barcelona, Spain
| | - Georgina Benet
- Neuroimaging Research Group, Neurosciences Programme, Hospital del Mar Research Institute, Barcelona, Spain
| | - Georgina Feixas
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Angela Arranz Betegon
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, Institut Clinic de Ginecologia, Obstetricia i Neonatologia, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Malcolm D McLeod
- Research School of Chemistry, Australian National University, Canberra, Australia
| | - Oscar J Pozo
- Applied Metabolomics Research Group, Neurosciences Research Programme, Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
3
|
Machado-Vieira R, Krause TM, Jones G, Teixeira AL, Shahani LR, Lane SD, Soares JC, Truong CN. Protective effects of psychiatric medications against COVID-19 mortality before vaccines. PLoS One 2025; 20:e0310438. [PMID: 39992948 PMCID: PMC11849848 DOI: 10.1371/journal.pone.0310438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/14/2025] [Indexed: 02/26/2025] Open
Abstract
The coronavirus disease pandemic caused by the coronavirus SARS-CoV-2, which emerged in the United States in late 2019 to early 2020 and quickly escalated into a national public health crisis. Research has identified psychiatric conditions as possible risk factors associated with COVID-19 infection and symptom severity. This study aims to determine whether specific classes of psychiatric medications could reduce the likelihood of infection and alleviate the severity of the disease. The objective of this study is to investigate the relationship between neuropsychiatric medication usage and COVID-19 outcomes before the widespread utilization of COVID-19 vaccines. This cross-sectional study used Optum's de-identified Clinformatics Data Mart Database to identify patients diagnosed with COVID-19 in 2020 and their psychiatric medication prescriptions in the United States. Ordered logistic regression was used to predict the likelihood of a higher COVID-19 severity level for long-term and new users. Results were adjusted for demographic characteristics and medical and psychiatric comorbidities. Most users were classified into the long-term user analysis group. Long-term users were 9% less likely to have a higher severity score (CI: 0.89-0.93, p-value < 0.001) than non-users. SSRI antidepressant users, both long-term (OR: 1.09; CI: 1.06-1.12) and short-term (OR: 1.17; CI: 1.07-1.27) were significantly more likely to have a lower severity score. However, the results varied across long-term and short-term users for all medication classes. Results of the current study suggest that psychopharmacological agents are associated with reduced COVID-19 severity levels and that antidepressant medications may have a protective role against COVID-19.
Collapse
Affiliation(s)
- Rodrigo Machado-Vieira
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Trudy M. Krause
- School of Public Health Center for Health Care Data, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Gregory Jones
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Antonio L. Teixeira
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Lokesh R. Shahani
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Scott D. Lane
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Jair C. Soares
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Chau N. Truong
- School of Public Health Center for Health Care Data, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| |
Collapse
|
4
|
Pérez-Reche FJ, Cheetham NJ, Bowyer RCE, Thompson EJ, Tettamanzi F, Menni C, Steves CJ. ESPClust: unsupervised identification of modifiers for the effect size profile in omics association studies. Bioinformatics 2025; 41:btaf065. [PMID: 39913362 PMCID: PMC11879214 DOI: 10.1093/bioinformatics/btaf065] [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: 09/03/2024] [Revised: 01/08/2025] [Accepted: 02/04/2025] [Indexed: 03/06/2025] Open
Abstract
MOTIVATION High-throughput omics technologies have revolutionized the identification of associations between individual traits and underlying biological characteristics, but still use 'one effect-size fits all' approaches. While covariates are often used, their potential as effect modifiers often remains unexplored. RESULTS We propose ESPClust, a novel unsupervised method designed to identify covariates that modify the effect size of associations between sets of omics variables and outcomes. By extending the concept of moderators to encompass multiple exposures, ESPClust analyses the effect size profile (ESP) to identify regions in covariate space with different ESP, enabling the discovery of subpopulations with distinct associations. Applying ESPClust to synthetic data, insulin resistance and COVID-19 symptom manifestation, we demonstrate its versatility and ability to uncover nuanced effect size modifications that traditional analyses may overlook. By integrating information from multiple exposures, ESPClust identifies effect size modifiers in datasets that are too small for traditional univariate stratified analyses. This method provides a robust framework for understanding complex omics data and holds promise for personalised medicine. AVAILABILITY AND IMPLEMENTATION The source code ESPClust is available at https://github.com/fjpreche/ESPClust.git. It can be installed via Python package repositories as 'pip install ESPClust==1.1.0'.
Collapse
Affiliation(s)
- Francisco J Pérez-Reche
- School of Natural and Computing Sciences, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
- Department of Twin Research and Genetic Epidemiology, School of Life Course & Population Sciences, King’s College London, London SE1 7EH, United Kingdom
| | - Nathan J Cheetham
- Department of Twin Research and Genetic Epidemiology, School of Life Course & Population Sciences, King’s College London, London SE1 7EH, United Kingdom
| | - Ruth C E Bowyer
- Department of Twin Research and Genetic Epidemiology, School of Life Course & Population Sciences, King’s College London, London SE1 7EH, United Kingdom
- The Alan Turing Institute, British Library, London NW1 2DB, United Kingdom
| | - Ellen J Thompson
- School of Psychology, Faculty of Science, Engineering and Medicine, University of Sussex, Brighton BN1 9QH, United Kingdom
| | - Francesca Tettamanzi
- Department of Twin Research and Genetic Epidemiology, School of Life Course & Population Sciences, King’s College London, London SE1 7EH, United Kingdom
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, School of Life Course & Population Sciences, King’s College London, London SE1 7EH, United Kingdom
| | - Claire J Steves
- Department of Twin Research and Genetic Epidemiology, School of Life Course & Population Sciences, King’s College London, London SE1 7EH, United Kingdom
| |
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
|
Sanduzzi Zamparelli S, Sanduzzi Zamparelli A, Bocchino M. Immune-Boosting and Antiviral Effects of Antioxidants in COVID-19 Pneumonia: A Therapeutic Perspective. Life (Basel) 2025; 15:113. [PMID: 39860053 PMCID: PMC11766556 DOI: 10.3390/life15010113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/05/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
The COVID-19 pandemic caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has profoundly impacted global health, with pneumonia emerging as a major complication in severe cases. The pathogenesis of COVID-19 is marked by the overproduction of reactive oxygen species (ROS) and an excessive inflammatory response, resulting in oxidative stress and significant tissue damage, particularly in the respiratory system. Antioxidants have garnered considerable attention for their potential role in managing COVID-19 pneumonia by mitigating oxidative stress and modulating immune responses. This review provides a comprehensive overview of the literature on the use of antioxidants in hospitalized patients with mild-to-moderate COVID-19. Studies exploring antioxidants, including vitamins, trace elements, nitric oxide (NO), ozone (O3), glutathione (GSH), L-carnitine, melatonin, bromelain, N-acetylcysteine (NAC), and numerous polyphenols, have yielded promising outcomes. Through their ROS-scavenging properties, these molecules support endothelial function, reduce the thrombosis risk, and may help mitigate the effects of the cytokine storm, a key contributor to COVID-19 morbidity and mortality. Clinical evidence suggests that antioxidant supplementation may improve patient outcomes by decreasing inflammation, supporting immune cell function, and potentially shortening recovery times. Furthermore, these molecules may mitigate the symptoms of COVID-19 by exerting direct antiviral effects that inhibit the infection process and genomic replication of SARS-CoV-2 in host cells. Moreover, antioxidants may work synergistically with standard antiviral treatments to reduce viral-induced oxidative damage. By integrating findings from the literature with real-world data from our clinical experience, we gain a more profound understanding of the role of antioxidants in managing COVID-19 pneumonia. Further research combining comprehensive literature reviews with real-world data analysis is crucial to validate the efficacy of antioxidants and establish evidence-based guidelines for their use in clinical practice.
Collapse
Affiliation(s)
| | - Alessandro Sanduzzi Zamparelli
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (A.S.Z.); (M.B.)
- UNESCO Chair for Health Education and Sustainable Development, University of Naples “Federico II”, 80131 Naples, Italy
- ERN Lung, 60596 Frankfurt am Main, Germany
| | - Marialuisa Bocchino
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (A.S.Z.); (M.B.)
| |
Collapse
|
7
|
Pimentel E, Banoei MM, Kaur J, Lee CH, Winston BW. Metabolomic Insights into COVID-19 Severity: A Scoping Review. Metabolites 2024; 14:617. [PMID: 39590853 PMCID: PMC11596841 DOI: 10.3390/metabo14110617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/29/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND In 2019, SARS-CoV-2, the novel coronavirus, entered the world scene, presenting a global health crisis with a broad spectrum of clinical manifestations. Recognizing the significance of metabolomics as the omics closest to symptomatology, it has become a useful tool for predicting clinical outcomes. Several metabolomic studies have indicated variations in the metabolome corresponding to different disease severities, highlighting the potential of metabolomics to unravel crucial insights into the pathophysiology of SARS-CoV-2 infection. METHODS The PRISMA guidelines were followed for this scoping review. Three major scientific databases were searched: PubMed, the Directory of Open Access Journals (DOAJ), and BioMed Central, from 2020 to 2024. Initially, 2938 articles were identified and vetted with specific inclusion and exclusion criteria. Of these, 42 articles were retrieved for analysis and summary. RESULTS Metabolites were identified that were repeatedly noted to change with COVID-19 and its severity. Phenylalanine, glucose, and glutamic acid increased with severity, while tryptophan, proline, and glutamine decreased, highlighting their association with COVID-19 severity. Additionally, pathway analysis revealed that phenylalanine, tyrosine and tryptophan biosynthesis, and arginine biosynthesis were the most significantly impacted pathways in COVID-19 severity. CONCLUSIONS COVID-19 severity is intricately linked to significant metabolic alterations that span amino acid metabolism, energy production, immune response modulation, and redox balance.
Collapse
Affiliation(s)
- Eric Pimentel
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (E.P.); (M.M.B.); (J.K.); (C.H.L.)
| | - Mohammad Mehdi Banoei
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (E.P.); (M.M.B.); (J.K.); (C.H.L.)
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Jasnoor Kaur
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (E.P.); (M.M.B.); (J.K.); (C.H.L.)
| | - Chel Hee Lee
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (E.P.); (M.M.B.); (J.K.); (C.H.L.)
- Department of Mathematics and Statistics, Faculty of Science, University of Calgary, Calgary, AB T2N 5A1, Canada
| | - Brent W. Winston
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (E.P.); (M.M.B.); (J.K.); (C.H.L.)
- Departments of Medicine, Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| |
Collapse
|
8
|
Ember KJI, Ksantini N, Dallaire F, Sheehy G, Tran T, Dehaes M, Durand M, Trudel D, Leblond F. Liquid saliva-based Raman spectroscopy device with on-board machine learning detects COVID-19 infection in real-time. Analyst 2024; 149:5535-5545. [PMID: 39435472 DOI: 10.1039/d4an00729h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
With greater population density, the likelihood of viral outbreaks achieving pandemic status is increasing. However, current viral screening techniques use specific reagents, and as viruses mutate, test accuracy decreases. Here, we present the first real-time, reagent-free, portable analysis platform for viral detection in liquid saliva, using COVID-19 as a proof-of-concept. We show that vibrational molecular spectroscopy and machine learning (ML) detect biomolecular changes consistent with the presence of viral infection. Saliva samples were collected from 470 individuals, including 65 that were infected with COVID-19 (28 from hospitalized patients and 37 from a walk-in testing clinic) and 251 that had a negative polymerase chain reaction (PCR) test. A further 154 were collected from healthy volunteers. Saliva measurements were achieved in 6 minutes or less and led to machine learning models predicting COVID-19 infection with sensitivity and specificity reaching 90%, depending on volunteer symptoms and disease severity. Machine learning models were based on linear support vector machines (SVM). This platform could be deployed to manage future pandemics using the same hardware but using a tunable machine learning model that could be rapidly updated as new viral strains emerge.
Collapse
Affiliation(s)
- Katherine J I Ember
- Department of Engineering Physics, Polytechnique Montréal, Montreal, Quebec, Canada.
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Nassim Ksantini
- Department of Engineering Physics, Polytechnique Montréal, Montreal, Quebec, Canada.
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Frédérick Dallaire
- Department of Engineering Physics, Polytechnique Montréal, Montreal, Quebec, Canada.
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Guillaume Sheehy
- Department of Engineering Physics, Polytechnique Montréal, Montreal, Quebec, Canada.
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Trang Tran
- Department of Engineering Physics, Polytechnique Montréal, Montreal, Quebec, Canada.
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Mathieu Dehaes
- Department of Radiology, Radio-oncology and Nuclear Medicine, Université de Montréal, Montreal, Canada
- Institute of Biomedical Engineering, Université de Montréal, Montreal, Canada
- Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CRCHUSJ), Montreal, Canada
| | - Madeleine Durand
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
- Internal Medicine service, Centre Hospitalier de l'Univsersité de Montréal (CHUM), Montreal, Quebec, Canada
| | - Dominique Trudel
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Frédéric Leblond
- Department of Engineering Physics, Polytechnique Montréal, Montreal, Quebec, Canada.
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| |
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
|
Pilger BI, Castro A, Vasconcellos FF, Moura KF, Signini ÉDF, Marqueze LFB, Fiorenza-Neto EA, Rocha MT, Pedroso GS, Cavaglieri CR, Ferreira AG, Figueiredo C, Minuzzi LG, Gatti da Silva GH, Castro GS, Lira FS, Seelaender M, Pinho RA. Obesity-dependent molecular alterations in fatal COVID-19: A retrospective postmortem study of metabolomic profile of adipose tissue. J Cell Biochem 2024; 125:e30566. [PMID: 38591648 DOI: 10.1002/jcb.30566] [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: 01/26/2024] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/10/2024]
Abstract
We investigated the effects of obesity on metabolic, inflammatory, and oxidative stress parameters in the adipose tissue of patients with fatal COVID-19. Postmortem biopsies of subcutaneous adipose tissue were obtained from 25 unvaccinated inpatients who passed from COVID-19, stratified as nonobese (N-OB; body mass index [BMI], 26.5 ± 2.3 kg m-2) or obese (OB BMI 34.2 ± 5.1 kg m-2). Univariate and multivariate analyses revealed that body composition was responsible for most of the variations detected in the metabolome, with greater dispersion observed in the OB group. Fifteen metabolites were major segregation factors. Results from the OB group showed higher levels of creatinine, myo-inositol, O-acetylcholine, and succinate, and lower levels of sarcosine. The N-OB group showed lower levels of glutathione peroxidase activity, as well as higher content of IL-6 and adiponectin. We revealed significant changes in the metabolomic profile of the adipose tissue in fatal COVID-19 cases, with high adiposity playing a key role in these observed variations. These findings highlight the potential involvement of metabolic and inflammatory pathways, possibly dependent on hypoxia, shedding light on the impact of obesity on disease pathogenesis and suggesting avenues for further research and possible therapeutic targets.
Collapse
Affiliation(s)
- Bruna I Pilger
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Alex Castro
- Laboratory of Nuclear Magnetic Resonance, Department of Chemistry, Universidade Federal de São Carlos, São Carlos, Brazil
- Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Franciane F Vasconcellos
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Karen F Moura
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Étore De Favari Signini
- Cardiovascular Physical Therapy Laboratory, Department of Physical Therapy, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Luis Felipe B Marqueze
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Edson A Fiorenza-Neto
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Mateus T Rocha
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Giulia S Pedroso
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Claudia R Cavaglieri
- Exercise Physiology Laboratory, Faculty of Physical Education, University of Campinas, Campinas, Brazil
| | - Antonio G Ferreira
- Laboratory of Nuclear Magnetic Resonance, Department of Chemistry, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Caique Figueiredo
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Luciele G Minuzzi
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Guilherme H Gatti da Silva
- Cancer Metabolism Research Group, Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | - Gabriela S Castro
- Cancer Metabolism Research Group, Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | - Fábio S Lira
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Marilia Seelaender
- Cancer Metabolism Research Group, Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | - Ricardo A Pinho
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| |
Collapse
|
11
|
Camelo ALM, Zamora Obando HR, Rocha I, Dias AC, Mesquita ADS, Simionato AVC. COVID-19 and Comorbidities: What Has Been Unveiled by Metabolomics? Metabolites 2024; 14:195. [PMID: 38668323 PMCID: PMC11051775 DOI: 10.3390/metabo14040195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/14/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
The COVID-19 pandemic has brought about diverse impacts on the global population. Individuals with comorbidities were more susceptible to the severe symptoms caused by the virus. Within the crisis scenario, metabolomics represents a potential area of science capable of providing relevant information for understanding the metabolic pathways associated with the intricate interaction between the viral disease and previous comorbidities. This work aims to provide a comprehensive description of the scientific production pertaining to metabolomics within the specific context of COVID-19 and comorbidities, while highlighting promising areas for exploration by those interested in the subject. In this review, we highlighted the studies of metabolomics that indicated a variety of metabolites associated with comorbidities and COVID-19. Furthermore, we observed that the understanding of the metabolic processes involved between comorbidities and COVID-19 is limited due to the urgent need to report disease outcomes in individuals with comorbidities. The overlap of two or more comorbidities associated with the severity of COVID-19 hinders the comprehension of the significance of each condition. Most identified studies are observational, with a restricted number of patients, due to challenges in sample collection amidst the emergent situation.
Collapse
Affiliation(s)
- André Luiz Melo Camelo
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Hans Rolando Zamora Obando
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Isabela Rocha
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Aline Cristina Dias
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Alessandra de Sousa Mesquita
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Ana Valéria Colnaghi Simionato
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
- National Institute of Science and Technology for Bioanalytics—INCTBio, Institute of Chemistry, Universidade Estadual de (UNICAMP), Campinas 13083-970, São Paulo, Brazil
| |
Collapse
|
12
|
Lv Y, Yang X, Song Y, Yang D, Zheng K, Zhou S, Xie H, Guo R, Tang S. The Correlation Between Essential Amino Acid Tryptophan, Lysine, Phenylalanine and Chemotherapy of Breast Cancer. Technol Cancer Res Treat 2024; 23:15330338241286872. [PMID: 39435510 PMCID: PMC11497521 DOI: 10.1177/15330338241286872] [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: 02/27/2024] [Revised: 07/28/2024] [Accepted: 08/09/2024] [Indexed: 10/23/2024] Open
Abstract
To investigate the differences in serum tryptophan, lysine, and phenylalanine levels in breast cancer patients, the correlation between the three amino acids with the chemotherapy regimen, and their significance in the clinical diagnosis and treatment of breast cancer.Clinical data were collected from the Department of Breast Surgery at Yunnan Cancer Hospital, encompassing 216 cases from July to December 2020, including 91 healthy individuals, 38 with benign tumors, and 87 with cancer. Amino acid levels were measured using liquid chromatography-tandem mass spectrometry. Statistical analyses, such as the Kruskal-Wallis H-test and Wilcoxon test, were conducted to compare the levels of these amino acids across the healthy group, benign tumor group, and breast cancer group. The χ2 test and Fisher's exact probability method were employed to assess the relationship between amino acid levels and breast cancer stage, grade, and chemotherapy regimen.The results indicated that there were significant differences in serum lysine (H = 36.13, P < .001) and phenylalanine (H = 34.03, P < .001) levels among the three groups. However, tryptophan levels did not show statistically significant variances. Specifically, lysine and phenylalanine levels were significantly different when comparing the healthy group with the breast cancer group and the benign tumor group with the breast cancer group. These differences were not significant when comparing the healthy group with the benign tumor group. Furthermore, there were no statistically significant distinctions observed in lysine (F = 0.836, P > .05) and phenylalanine (F = 1.466, P > .05) levels across different conventional chemotherapy regimens among the breast cancer cases studied.Serum lysine and phenylalanine levels might serve as potential biomarkers for breast cancer, and the choice of chemotherapy regimen is unlikely to impact significant changes in these amino acid levels.
Collapse
Affiliation(s)
- Yafeng Lv
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Xuan Yang
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Ying Song
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Dechun Yang
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Kai Zheng
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Shaoqiang Zhou
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Hanhui Xie
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Rong Guo
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| | - Shicong Tang
- Department of Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, China
| |
Collapse
|
13
|
Cesar-Silva D, Pereira-Dutra FS, Giannini ALM, Maya-Monteiro CM, de Almeida CJG. Lipid compartments and lipid metabolism as therapeutic targets against coronavirus. Front Immunol 2023; 14:1268854. [PMID: 38106410 PMCID: PMC10722172 DOI: 10.3389/fimmu.2023.1268854] [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: 07/28/2023] [Accepted: 10/24/2023] [Indexed: 12/19/2023] Open
Abstract
Lipids perform a series of cellular functions, establishing cell and organelles' boundaries, organizing signaling platforms, and creating compartments where specific reactions occur. Moreover, lipids store energy and act as secondary messengers whose distribution is tightly regulated. Disruption of lipid metabolism is associated with many diseases, including those caused by viruses. In this scenario, lipids can favor virus replication and are not solely used as pathogens' energy source. In contrast, cells can counteract viruses using lipids as weapons. In this review, we discuss the available data on how coronaviruses profit from cellular lipid compartments and why targeting lipid metabolism may be a powerful strategy to fight these cellular parasites. We also provide a formidable collection of data on the pharmacological approaches targeting lipid metabolism to impair and treat coronavirus infection.
Collapse
Affiliation(s)
- Daniella Cesar-Silva
- Laboratory of Immunopharmacology, Department of Genetics, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Filipe S. Pereira-Dutra
- Laboratory of Immunopharmacology, Department of Genetics, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Ana Lucia Moraes Giannini
- Laboratory of Functional Genomics and Signal Transduction, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarissa M. Maya-Monteiro
- Laboratory of Immunopharmacology, Department of Genetics, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- Laboratory of Endocrinology and Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Cecília Jacques G. de Almeida
- Laboratory of Immunopharmacology, Department of Genetics, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
14
|
Chenchula S, Sharma S, Tripathi M, Chavan M, Misra AK, Rangari G. Prevalence of overweight and obesity and their effect on COVID-19 severity and hospitalization among younger than 50 years versus older than 50 years population: A systematic review and meta-analysis. Obes Rev 2023; 24:e13616. [PMID: 37574901 DOI: 10.1111/obr.13616] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/14/2023] [Indexed: 08/15/2023]
Abstract
Cohort studies have shown that both overweight and obesity have their impact by increasing hospitalization with COVID-19. We conducted a systematic literature search in PubMed, Google Scholar, and MedRxiv databases following the PRISMA guidelines. Statistical analyses were performed using STATA software version 16 MP (Stata Corp, College Station, TX, USA) and Med Calc software version 22.009(Med Calc software Ltd, Ostend, Belgium). The primary outcome was to measure the prevalence of overweight and obesity and their impact on the risk of hospitalization among COVID-19 patients under and above 50 years of age. In total, 184 studies involving 2,365,377 patients were included. The prevalence of overweight was highest among those younger than 50 years of age over those older than 50 years of age, (26.33% vs. 30.46%), but there was no difference in obesity (36.30% vs. 36.02%). Overall, the pooled prevalence of overweight and obesity among hospitalized COVID-19 patients was 31.0% and 36.26%, respectively. Compared with normal weight, the odds of hospitalization with overweight (odds ratio [OR] 2.186, 95% confidence interval [CI] [1.19, 3.99], p < 0.01) and obesity (OR 3.069, 95% CI [1.67, 5.61], p < 0.001) in those younger than 50 years and obesity (OR 3.977, 95% CI [2.75, 5.73], p < 0.001) in the older than 50 years age group were significantly high. The increased prevalence of overweight and obesity among the under 50 years age group and obesity among the older than 50 years age group significantly increased the rate of COVID-19 infections, severity and hospitalization.
Collapse
Affiliation(s)
- Santenna Chenchula
- Department of Pharmacology, All India Institute of Medical Sciences, Mangalagiri, India
| | - Sushil Sharma
- Department of Pharmacology, All India Institute of Medical Sciences, Mangalagiri, India
| | - Mukesh Tripathi
- Department of Anaesthesia and Critical care Medicine, All India Institute of Medical Sciences, Mangalagiri, India
| | - Madhavrao Chavan
- Department of Pharmacology, All India Institute of Medical Sciences, Mangalagiri, India
| | - Arup Kumar Misra
- Department of Pharmacology, All India Institute of Medical Sciences, Mangalagiri, India
| | - Gaurav Rangari
- Department of Pharmacology, All India Institute of Medical Sciences, Mangalagiri, India
| |
Collapse
|
15
|
Sánchez-Rico M, Edán-Sánchez A, Olfson M, Alvarado JM, Airagnes G, Rezaei K, Delcuze A, Peyre H, Limosin F, Hoertel N. Antipsychotic use and 28-day mortality in patients hospitalized with COVID-19: A multicenter observational retrospective study. Eur Neuropsychopharmacol 2023; 75:93-104. [PMID: 37713738 PMCID: PMC10272945 DOI: 10.1016/j.euroneuro.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 09/17/2023]
Abstract
Prior research has yielded conflicting results about the potential influence of antipsychotics in patients with COVID-19. In this multicenter retrospective study, we examined the association of antipsychotic use at admission with 28-day all-cause mortality in a sample of 59,021 adult patients hospitalized with COVID-19 from January 2020 to November 2021. In a 1:1 ratio matched analytic sample (N=1,454) accounting for age, sex, hospital, hospitalization period, the Elixhauser Comorbidity Index, other psychotropic medications, medications prescribed according to compassionate use or as part of a clinical trial, current diagnoses of psychiatric disorders, and clinical and biological markers of COVID-19 severity, antipsychotic use was not associated with 28-day mortality [23.5% (N=727) versus 18.6% (N=727); OR=1.16; 95%CI=0.89-1.51; p=0.280]. This association remained non-significant in exploratory analyses across all classes of antipsychotics and individual molecules, except for typical antipsychotics and loxapine, which were significantly linked to increased 28-day mortality, associations likely due to residual indication bias. Contrariwise, antipsychotics prescribed at daily doses higher than 200 mg of chlorpromazine-equivalents might be associated with reduced 28-day mortality when compared to patients not taking antipsychotics in the matched analytic sample [10.4% (N=154) versus 18.6% (N=727); AOR=0.56; 95%CI=0.31-0.96; p=0.040]. These results suggest that antipsychotic use, when prescribed at usual doses, are not be associated with 28-day mortality in patients hospitalized with COVID-19.
Collapse
Affiliation(s)
- Marina Sánchez-Rico
- AP-HP, AP-HP Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, 92130 Issy-les-Moulineaux, France; Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, Madrid, Spain.
| | - Alejandro Edán-Sánchez
- AP-HP, AP-HP Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, 92130 Issy-les-Moulineaux, France; Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, Madrid, Spain
| | - Mark Olfson
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Jesús M Alvarado
- Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, Madrid, Spain
| | - Guillaume Airagnes
- AP-HP, AP-HP Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, 92130 Issy-les-Moulineaux, France; Université Paris Cité, Paris, France
| | - Katayoun Rezaei
- AP-HP, AP-HP Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, 92130 Issy-les-Moulineaux, France
| | - Aude Delcuze
- CLINEA, Clinique Les Orchidées, Service de Psychiatrie, Andilly, France
| | | | - Frédéric Limosin
- AP-HP, AP-HP Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, 92130 Issy-les-Moulineaux, France; Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; Université Paris Cité, Paris, France
| | - Nicolas Hoertel
- AP-HP, AP-HP Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Service de Psychiatrie et Addictologie, 92130 Issy-les-Moulineaux, France; Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; INSERM UMR_1266, Institut de Psychiatrie et Neuroscience de Paris, F-75014 Paris, France
| |
Collapse
|
16
|
Fenizia S, Gaggini M, Vassalle C. The Sphingolipid-Signaling Pathway as a Modulator of Infection by SARS-CoV-2. Curr Issues Mol Biol 2023; 45:7956-7973. [PMID: 37886946 PMCID: PMC10605018 DOI: 10.3390/cimb45100503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Ceramides and other related sphingolipids, important cellular components linked to metabolic homeostasis and cardiometabolic diseases, have been found to be involved in different steps of the SARS-CoV-2 life cycle. Hence, changes in their physiological levels are identified as predictors of COVID-19 severity and prognosis, as well as potential therapeutic targets. In this review, an overview of the SARS-CoV-2 life cycle is given, followed by a description of the sphingolipid metabolism and its role in viral infection, with a particular focus on those steps required to finalize the viral life cycle. Furthermore, the use and development of pharmaceutical strategies to target sphingolipids to prevent and treat severe and long-term symptoms of infectious diseases, particularly COVID-19, are reviewed herein. Finally, research perspectives and current challenges in this research field are highlighted. Although many aspects of sphingolipid metabolism are not fully known, this review aims to highlight how the discovery and use of molecules targeting sphingolipids with reliable and selective properties may offer new therapeutic alternatives to infectious and other diseases, including COVID-19.
Collapse
Affiliation(s)
- Simona Fenizia
- Istituto di Fisiologia Clinica, Italian National Research Council, Via Moruzzi 1, 56124 Pisa, Italy
| | - Melania Gaggini
- Fondazione CNR-Regione Toscana G. Monasterio, Via Moruzzi 1, 56124 Pisa, Italy
| | - Cristina Vassalle
- Fondazione CNR-Regione Toscana G. Monasterio, Via Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
17
|
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
|
18
|
Yan H, Zhao S, Huang HX, Xie P, Cai XH, Qu YD, Zhang W, Luo JQ, Zhang L, Li X. Systematic Mendelian randomization study of the effect of gut microbiome and plasma metabolome on severe COVID-19. Front Immunol 2023; 14:1211612. [PMID: 37662924 PMCID: PMC10468967 DOI: 10.3389/fimmu.2023.1211612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/20/2023] [Indexed: 09/05/2023] Open
Abstract
BACKGROUND COVID-19 could develop severe respiratory symptoms in certain infected patients, especially in the patients with immune disorders. Gut microbiome and plasma metabolome act important immunological modulators in the human body and could contribute to the immune responses impacting the progression of COVID-19. However, the causal relationship between specific intestinal bacteria, metabolites and severe COVID-19 remains not clear. METHODS Based on two-sample Mendelian randomization (MR) framework, the causal effects of 131 intestinal taxa and 452 plasma metabolites on severe COVID-19 were evaluated. Single nucleotide polymorphisms (SNPs) strongly associated with the abundance of intestinal taxa and the concentration of plasma metabolites had been utilized as the instrument variables to infer whether they were causal factors of severe COVID-19. In addition, mediation analysis was conducted to find the potential association between the taxon and metabolite, and further colocalization analysis had been performed to validate the causal relationships. RESULTS MR analysis identified 13 taxa and 53 metabolites, which were significantly associated with severe COVID-19 as causal factors. Mediation analysis revealed 11 mediated relationships. Myo-inositol, 2-stearoylglycerophosphocholine, and alpha-glutamyltyrosine, potentially contributed to the association of Howardella and Ruminiclostridium 6 with severe COVID-19, respectively. Butyrivibrio and Ruminococcus gnavus could mediate the association of myo-inositol and N-acetylalanine, respectively. In addition, Ruminococcus torques abundance was colocalized with severe COVID-19 (PP.H4 = 0.77) and the colon expression of permeability related protein RASIP1 (PP.H4 = 0.95). CONCLUSIONS Our study highlights the potential causal relationships between gut microbiome, plasma metabolome and severe COVID-19, which potentially serve as clinical biomarkers for risk stratification and prognostication and benefit the mechanism mechanistic investigation of severe COVID-19.
Collapse
Affiliation(s)
- Han Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Si Zhao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan, China
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Han-Xue Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan, China
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Pan Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan, China
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin-He Cai
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan, China
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yun-Dan Qu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan, China
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan, China
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian-Quan Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Longbo Zhang
- Departments of Neurosurgery, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, United States
| | - Xi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan, China
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
19
|
Al-Sulaiti H, Almaliti J, Naman CB, Al Thani AA, Yassine HM. Metabolomics Approaches for the Diagnosis, Treatment, and Better Disease Management of Viral Infections. Metabolites 2023; 13:948. [PMID: 37623891 PMCID: PMC10456346 DOI: 10.3390/metabo13080948] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 08/26/2023] Open
Abstract
Metabolomics is an analytical approach that involves profiling and comparing the metabolites present in biological samples. This scoping review article offers an overview of current metabolomics approaches and their utilization in evaluating metabolic changes in biological fluids that occur in response to viral infections. Here, we provide an overview of metabolomics methods including high-throughput analytical chemistry and multivariate data analysis to identify the specific metabolites associated with viral infections. This review also focuses on data interpretation and applications designed to improve our understanding of the pathogenesis of these viral diseases.
Collapse
Affiliation(s)
- Haya Al-Sulaiti
- QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (H.A.-S.); (A.A.A.T.)
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Jehad Almaliti
- Scripps Institution of Oceanography, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA P.O. Box 92093, USA;
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Jordan, Amman P.O. Box 11942, Jordan
| | - C. Benjamin Naman
- Department of Science and Conservation, San Diego Botanic Garden, Encinitas, CA P.O. Box 92024, USA;
| | - Asmaa A. Al Thani
- QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (H.A.-S.); (A.A.A.T.)
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
- College of Health Sciences, QU-Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Hadi M. Yassine
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
- College of Health Sciences, QU-Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
20
|
Hoertel N, Rezaei K, Sánchez-Rico M, Delgado-Álvarez A, Kornhuber J, Gulbins E, Olfson M, Ouazana-Vedrines C, Carpinteiro A, Cougoule C, Becker KA, Alvarado JM, Limosin F. Medications Modulating the Acid Sphingomyelinase/Ceramide System and 28-Day Mortality among Patients with SARS-CoV-2: An Observational Study. Pharmaceuticals (Basel) 2023; 16:1107. [PMID: 37631022 PMCID: PMC10458150 DOI: 10.3390/ph16081107] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/12/2023] [Accepted: 07/20/2023] [Indexed: 08/27/2023] Open
Abstract
Prior evidence indicates the potential central role of the acid sphingomyelinase (ASM)/ceramide system in the infection of cells with SARS-CoV-2. We conducted a multicenter retrospective observational study including 72,105 adult patients with laboratory-confirmed SARS-CoV-2 infection who were admitted to 36 AP-HP (Assistance Publique-Hôpitaux de Paris) hospitals from 2 May 2020 to 31 August 2022. We examined the association between the ongoing use of medications functionally inhibiting acid sphingomyelinase (FIASMA), which reduces the infection of cells with SARS-CoV-2 in vitro, upon hospital admission with 28-day all-cause mortality in a 1:1 ratio matched analytic sample based on clinical characteristics, disease severity and other medications (N = 9714). The univariate Cox regression model of the matched analytic sample showed that FIASMA medication use at admission was associated with significantly lower risks of 28-day mortality (HR = 0.80; 95% CI = 0.72-0.88; p < 0.001). In this multicenter observational study, the use of FIASMA medications was significantly and substantially associated with reduced 28-day mortality among adult patients hospitalized with COVID-19. These findings support the continuation of these medications during the treatment of SARS-CoV-2 infections. Randomized clinical trials (RCTs) are needed to confirm these results, starting with the molecules with the greatest effect size in the study, e.g., fluoxetine, escitalopram, and amlodipine.
Collapse
Affiliation(s)
- Nicolas Hoertel
- INSERM U1266, Université Paris Cité, F-75014 Paris, France
- Service de Psychiatrie et Addictologie de l’Adulte et du Sujet Agé, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, GHU APHP.Centre, F-92130 Issy-les-Moulineaux, France
| | - Katayoun Rezaei
- Service de Psychiatrie et Addictologie de l’Adulte et du Sujet Agé, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, GHU APHP.Centre, F-92130 Issy-les-Moulineaux, France
| | - Marina Sánchez-Rico
- Service de Psychiatrie et Addictologie de l’Adulte et du Sujet Agé, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, GHU APHP.Centre, F-92130 Issy-les-Moulineaux, France
- Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, 28223 Madrid, Spain
| | - Alfonso Delgado-Álvarez
- Service de Psychiatrie et Addictologie de l’Adulte et du Sujet Agé, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, GHU APHP.Centre, F-92130 Issy-les-Moulineaux, France
- Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, 28223 Madrid, Spain
- Department of Biological and Health Psychology, Faculty of Psychology, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Erich Gulbins
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany (K.A.B.)
| | - Mark Olfson
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University, New York, NY 10032, USA
| | - Charles Ouazana-Vedrines
- Service de Psychiatrie de l’Adulte, DMU Psychiatrie et Addictologie, Hôpital Hôtel-Dieu, AP-HP, Université Paris Cité, F-75004 Paris, France
| | - Alexander Carpinteiro
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany (K.A.B.)
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany
| | - Céline Cougoule
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Katrin Anne Becker
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany (K.A.B.)
| | - Jesús M. Alvarado
- Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, 28223 Madrid, Spain
| | - Frédéric Limosin
- INSERM U1266, Université Paris Cité, F-75014 Paris, France
- Service de Psychiatrie et Addictologie de l’Adulte et du Sujet Agé, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, GHU APHP.Centre, F-92130 Issy-les-Moulineaux, France
| | | |
Collapse
|
21
|
Toro DM, da Silva-Neto PV, de Carvalho JCS, Fuzo CA, Pérez MM, Pimentel VE, Fraga-Silva TFC, Oliveira CNS, Caruso GR, Vilela AFL, Nobre-Azevedo P, Defelippo-Felippe TV, Argolo JGM, Degiovani AM, Ostini FM, Feitosa MR, Parra RS, Vilar FC, Gaspar GG, da Rocha JJR, Feres O, Costa GP, Maruyama SRC, Russo EMS, Fernandes APM, Santos IKFM, Malheiro A, Sadikot RT, Bonato VLD, Cardoso CRB, Dias-Baruffi M, Trapé ÁA, Faccioli LH, Sorgi CA. Plasma Sphingomyelin Disturbances: Unveiling Its Dual Role as a Crucial Immunopathological Factor and a Severity Prognostic Biomarker in COVID-19. Cells 2023; 12:1938. [PMID: 37566018 PMCID: PMC10417089 DOI: 10.3390/cells12151938] [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: 06/03/2023] [Revised: 06/30/2023] [Accepted: 07/20/2023] [Indexed: 08/12/2023] Open
Abstract
SARS-CoV-2 infection triggers distinct patterns of disease development characterized by significant alterations in host regulatory responses. Severe cases exhibit profound lung inflammation and systemic repercussions. Remarkably, critically ill patients display a "lipid storm", influencing the inflammatory process and tissue damage. Sphingolipids (SLs) play pivotal roles in various cellular and tissue processes, including inflammation, metabolic disorders, and cancer. In this study, we employed high-resolution mass spectrometry to investigate SL metabolism in plasma samples obtained from control subjects (n = 55), COVID-19 patients (n = 204), and convalescent individuals (n = 77). These data were correlated with inflammatory parameters associated with the clinical severity of COVID-19. Additionally, we utilized RNAseq analysis to examine the gene expression of enzymes involved in the SL pathway. Our analysis revealed the presence of thirty-eight SL species from seven families in the plasma of study participants. The most profound alterations in the SL species profile were observed in patients with severe disease. Notably, a predominant sphingomyelin (SM d18:1) species emerged as a potential biomarker for COVID-19 severity, showing decreased levels in the plasma of convalescent individuals. Elevated SM levels were positively correlated with age, hospitalization duration, clinical score, and neutrophil count, as well as the production of IL-6 and IL-8. Intriguingly, we identified a putative protective effect against disease severity mediated by SM (d18:1/24:0), while ceramide (Cer) species (d18:1/24:1) and (d18:1/24:0)were associated with increased risk. Moreover, we observed the enhanced expression of key enzymes involved in the SL pathway in blood cells from severe COVID-19 patients, suggesting a primary flow towards Cer generation in tandem with SM synthesis. These findings underscore the potential of SM as a prognostic biomarker for COVID-19 and highlight promising pharmacological targets. By targeting sphingolipid pathways, novel therapeutic strategies may emerge to mitigate the severity of COVID-19 and improve patient outcomes.
Collapse
Affiliation(s)
- Diana Mota Toro
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Postgraduate Program in Basic and Applied Immunology–PPGIBA, Institute of Biological Sciences, Federal University of Amazonas–UFAM, Manaus 69080-900, AM, Brazil;
| | - Pedro V. da Silva-Neto
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Postgraduate Program in Basic and Applied Immunology–PPGIBA, Institute of Biological Sciences, Federal University of Amazonas–UFAM, Manaus 69080-900, AM, Brazil;
| | - Jonatan C. S. de Carvalho
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
| | - Carlos A. Fuzo
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Malena M. Pérez
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Vinícius E. Pimentel
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Thais F. C. Fraga-Silva
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Camilla N. S. Oliveira
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Glaucia R. Caruso
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Adriana F. L. Vilela
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
| | - Pedro Nobre-Azevedo
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Thiago V. Defelippo-Felippe
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
| | - Jamille G. M. Argolo
- Department of General and Specialized Nursing, School of Nursing of Ribeirão Preto–EERP, University of São Paulo–USP, Ribeirão Preto 14040-902, SP, Brazil; (J.G.M.A.); (A.P.M.F.)
| | - Augusto M. Degiovani
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto 14085-000, SP, Brazil; (A.M.D.); (F.M.O.)
| | - Fátima M. Ostini
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto 14085-000, SP, Brazil; (A.M.D.); (F.M.O.)
| | - Marley R. Feitosa
- Department of Surgery and Anatomy, Faculty of Medicine of Ribeirão Preto-FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (M.R.F.); (R.S.P.); (J.J.R.d.R.); (O.F.)
- Hospital São Paulo, Ribeirão Preto 14025-100, SP, Brazil;
| | - Rogerio S. Parra
- Department of Surgery and Anatomy, Faculty of Medicine of Ribeirão Preto-FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (M.R.F.); (R.S.P.); (J.J.R.d.R.); (O.F.)
- Hospital São Paulo, Ribeirão Preto 14025-100, SP, Brazil;
| | - Fernando C. Vilar
- Hospital São Paulo, Ribeirão Preto 14025-100, SP, Brazil;
- Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil;
| | - Gilberto G. Gaspar
- Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil;
| | - José J. R. da Rocha
- Department of Surgery and Anatomy, Faculty of Medicine of Ribeirão Preto-FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (M.R.F.); (R.S.P.); (J.J.R.d.R.); (O.F.)
| | - Omar Feres
- Department of Surgery and Anatomy, Faculty of Medicine of Ribeirão Preto-FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (M.R.F.); (R.S.P.); (J.J.R.d.R.); (O.F.)
- Hospital São Paulo, Ribeirão Preto 14025-100, SP, Brazil;
| | - Gabriel P. Costa
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo–USP, Ribeirão Preto 14040-900, SP, Brazil; (G.P.C.); (Á.A.T.)
| | - Sandra R. C. Maruyama
- Department of Genetics and Evolution, Center for Biological and Health Sciences, Federal University of São Carlos (UFSCar), São Carlos 13565-905, SP, Brazil;
| | - Elisa M. S. Russo
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Ana Paula M. Fernandes
- Department of General and Specialized Nursing, School of Nursing of Ribeirão Preto–EERP, University of São Paulo–USP, Ribeirão Preto 14040-902, SP, Brazil; (J.G.M.A.); (A.P.M.F.)
| | - Isabel K. F. M. Santos
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Adriana Malheiro
- Postgraduate Program in Basic and Applied Immunology–PPGIBA, Institute of Biological Sciences, Federal University of Amazonas–UFAM, Manaus 69080-900, AM, Brazil;
| | - Ruxana T. Sadikot
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Vânia L. D. Bonato
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | - Cristina R. B. Cardoso
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Marcelo Dias-Baruffi
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Átila A. Trapé
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo–USP, Ribeirão Preto 14040-900, SP, Brazil; (G.P.C.); (Á.A.T.)
| | - Lúcia H. Faccioli
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto–FCFRP, University of Sao Paulo–USP, Ribeirão Preto 14040-903, SP, Brazil; (D.M.T.); (P.V.d.S.-N.); (J.C.S.d.C.); (C.A.F.); (M.M.P.); (V.E.P.); (C.N.S.O.); (G.R.C.); (E.M.S.R.); (C.R.B.C.); (M.D.-B.); (L.H.F.)
| | - Carlos A. Sorgi
- Postgraduate Program in Basic and Applied Immunology–PPGIBA, Institute of Biological Sciences, Federal University of Amazonas–UFAM, Manaus 69080-900, AM, Brazil;
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto–FFCLRP, University of São Paulo–USP, Ribeirão Preto 14040-901, SP, Brazil; (A.F.L.V.); (P.N.-A.); (T.V.D.-F.)
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto–FMRP, University of São Paulo–USP, Ribeirão Preto 14049-900, SP, Brazil; (T.F.C.F.-S.); (I.K.F.M.S.); (V.L.D.B.)
| | | |
Collapse
|
22
|
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
|
23
|
Izadpanah A, Mudd JC, Garcia JGN, Srivastav S, Abdel-Mohsen M, Palmer C, Goldman AR, Kolls JK, Qin X, Rappaport J. SARS-CoV-2 infection dysregulates NAD metabolism. Front Immunol 2023; 14:1158455. [PMID: 37457744 PMCID: PMC10344451 DOI: 10.3389/fimmu.2023.1158455] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/19/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Severe COVID-19 results initially in pulmonary infection and inflammation. Symptoms can persist beyond the period of acute infection, and patients with Post-Acute Sequelae of COVID (PASC) often exhibit a variety of symptoms weeks or months following acute phase resolution including continued pulmonary dysfunction, fatigue, and neurocognitive abnormalities. We hypothesized that dysregulated NAD metabolism contributes to these abnormalities. Methods RNAsequencing of lungs from transgenic mice expressing human ACE2 (K18-hACE2) challenged with SARS-CoV-2 revealed upregulation of NAD biosynthetic enzymes, including NAPRT1, NMNAT1, NAMPT, and IDO1 6 days post-infection. Results Our data also demonstrate increased gene expression of NAD consuming enzymes: PARP 9,10,14 and CD38. At the same time, SIRT1, a protein deacetylase (requiring NAD as a cofactor and involved in control of inflammation) is downregulated. We confirmed our findings by mining sequencing data from lungs of patients that died from SARS-CoV-2 infection. Our validated findings demonstrating increased NAD turnover in SARS-CoV-2 infection suggested that modulating NAD pathways may alter disease progression and may offer therapeutic benefits. Specifically, we hypothesized that treating K18-hACE2 mice with nicotinamide riboside (NR), a potent NAD precursor, may mitigate lethality and improve recovery from SARS-CoV-2 infection. We also tested the therapeutic potential of an anti- monomeric NAMPT antibody using the same infection model. Treatment with high dose anti-NAMPT antibody resulted in significantly decreased body weight compared to control, which was mitigated by combining HD anti-NAMPT antibody with NR. We observed a significant increase in lipid metabolites, including eicosadienoic acid, oleic acid, and palmitoyl carnitine in the low dose antibody + NR group. We also observed significantly increased nicotinamide related metabolites in NR treated animals. Discussion Our data suggest that infection perturbs NAD pathways, identify novel mechanisms that may explain some pathophysiology of CoVID-19 and suggest novel strategies for both treatment and prevention.
Collapse
Affiliation(s)
- Amin Izadpanah
- Tulane National Primate Research Center, Covington, Louisiana, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, LA, United States
| | - Joseph C. Mudd
- Tulane National Primate Research Center, Covington, Louisiana, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, LA, United States
| | - Joe G. N. Garcia
- Department of Medicine, College of Medicine Tucson, University of Arizona, Tucson, AZ, United States
| | - Sudesh Srivastav
- Biostatistics and Data Science, Tulane University School of Public Health, New Orleans, LA, United States
| | | | - Clovis Palmer
- Tulane National Primate Research Center, Covington, Louisiana, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, LA, United States
| | - Aaron R. Goldman
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, United States
- Proteomics and Metabolomics Shared Resource, The Wistar Institute, Philadelphia, PA, United States
| | - Jay K. Kolls
- Center for Translational Research in Infection and Inflammation, Tulane School of Medicine, New Orleans, Louisiana, LA, United States
| | - Xuebin Qin
- Tulane National Primate Research Center, Covington, Louisiana, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, LA, United States
| | - Jay Rappaport
- Tulane National Primate Research Center, Covington, Louisiana, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, LA, United States
| |
Collapse
|
24
|
Petrache I, Pujadas E, Ganju A, Serban KA, Borowiec A, Babbs B, Bronova IA, Egersdorf N, Hume PS, Goel K, Janssen WJ, Berdyshev EV, Cordon-Cardo C, Kolesnick R. Marked elevations in lung and plasma ceramide in COVID-19 linked to microvascular injury. JCI Insight 2023; 8:e156104. [PMID: 37212278 PMCID: PMC10322682 DOI: 10.1172/jci.insight.156104] [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: 10/22/2021] [Accepted: 04/05/2023] [Indexed: 05/23/2023] Open
Abstract
The pathogenesis of the marked pulmonary microvasculature injury, a distinguishing feature of COVID-19 acute respiratory distress syndrome (COVID-ARDS), remains unclear. Implicated in the pathophysiology of diverse diseases characterized by endothelial damage, including ARDS and ischemic cardiovascular disease, ceramide and in particular palmitoyl ceramide (C16:0-ceramide) may be involved in the microvascular injury in COVID-19. Using deidentified plasma and lung samples from COVID-19 patients, ceramide profiling by mass spectrometry was performed. Compared with healthy individuals, a specific 3-fold C16:0-ceramide elevation in COVID-19 patient plasma was identified. Compared with age-matched controls, autopsied lungs of individuals succumbing to COVID-ARDS displayed a massive 9-fold C16:0-ceramide elevation and exhibited a previously unrecognized microvascular ceramide-staining pattern and markedly enhanced apoptosis. In COVID-19 plasma and lungs, the C16-ceramide/C24-ceramide ratios were increased and reversed, respectively, consistent with increased risk of vascular injury. Indeed, exposure of primary human lung microvascular endothelial cell monolayers to C16:0-ceramide-rich plasma lipid extracts from COVID-19, but not healthy, individuals led to a significant decrease in endothelial barrier function. This effect was phenocopied by spiking healthy plasma lipid extracts with synthetic C16:0-ceramide and was inhibited by treatment with ceramide-neutralizing monoclonal antibody or single-chain variable fragment. These results indicate that C16:0-ceramide may be implicated in the vascular injury associated with COVID-19.
Collapse
Affiliation(s)
- Irina Petrache
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - Elisabet Pujadas
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aditya Ganju
- Laboratory of Signal Transduction, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Karina A. Serban
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - Alexander Borowiec
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
| | - Beatrice Babbs
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
| | - Irina A. Bronova
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
| | - Nicholas Egersdorf
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
| | - Patrick S. Hume
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - Khushboo Goel
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - William J. Janssen
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - Evgeny V. Berdyshev
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
| | - Carlos Cordon-Cardo
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Richard Kolesnick
- Laboratory of Signal Transduction, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
25
|
Chenchula S, Vidyasagar K, Pathan S, Sharma S, Chavan MR, Bhagavathula AS, Padmavathi R, Manjula M, Chhabra M, Gupta R, Amerneni KC, Ghanta MK, Mudda S. Global prevalence and effect of comorbidities and smoking status on severity and mortality of COVID-19 in association with age and gender: a systematic review, meta-analysis and meta-regression. Sci Rep 2023; 13:6415. [PMID: 37076543 PMCID: PMC10115382 DOI: 10.1038/s41598-023-33314-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/11/2023] [Indexed: 04/21/2023] Open
Abstract
A COVID-19 patient often presents with multiple comorbidities and is associated with adverse outcomes. A comprehensive assessment of the prevalence of comorbidities in patients with COVID-19 is essential. This study aimed to assess the prevalence of comorbidities, severity and mortality with regard to geographic region, age, gender and smoking status in patients with COVID-19. A systematic review and multistage meta-analyses were reported using PRISMA guidelines. PubMed/MEDLINE, SCOPUS, Google Scholar and EMBASE were searched from January 2020 to October 2022. Cross-sectional studies, cohort studies, case series studies, and case-control studies on comorbidities reporting among the COVID-19 populations that were published in English were included. The pooled prevalence of various medical conditions in COVID-19 patients was calculated based on regional population size weights. Stratified analyses were performed to understand the variations in the medical conditions based on age, gender, and geographic region. A total of 190 studies comprising 105 million COVID-19 patients were included. Statistical analyses were performed using STATA software, version 16 MP (StataCorp, College Station, TX). Meta-analysis of proportion was performed to obtain pooled values of the prevalence of medical comorbidities: hypertension (39%, 95% CI 36-42, n = 170 studies), obesity (27%, 95% CI 25-30%, n = 169 studies), diabetes (27%, 95% CI 25-30%, n = 175), and asthma (8%, 95% CI 7-9%, n = 112). Moreover, the prevalence of hospitalization was 35% (95% CI 29-41%, n = 61), intensive care admissions 17% (95% CI 14-21, n = 106), and mortality 18% (95% CI 16-21%, n = 145). The prevalence of hypertension was highest in Europe at 44% (95% CI 39-47%, n = 68), obesity and diabetes at 30% (95% CI, 26-34, n = 79) and 27% (95%CI, 24-30, n = 80) in North America, and asthma in Europe at 9% (95% CI 8-11, n = 41). Obesity was high among the ≥ 50 years (30%, n = 112) age group, diabetes among Men (26%, n = 124) and observational studies reported higher mortality than case-control studies (19% vs. 14%). Random effects meta-regression found a significant association between age and diabetes (p < 0.001), hypertension (p < 0.001), asthma (p < 0.05), ICU admission (p < 0.05) and mortality (p < 0.001). Overall, a higher global prevalence of hypertension (39%) and a lower prevalence of asthma (8%), and 18% of mortality were found in patients with COVID-19. Hence, geographical regions with respective chronic medical comorbidities should accelerate regular booster dose vaccination, preferably to those patients with chronic comorbidities, to prevent and lower the severity and mortality of COVID-19 disease with novel SARS-CoV-2 variants of concern (VOC).
Collapse
Affiliation(s)
- Santenna Chenchula
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Mangalagiri, Andhra Pradesh, 522503, India.
| | - Kota Vidyasagar
- Department of Pharmaceutical Sciences, University College of Pharmaceutical Sciences (UCPSc), Hanmakonda, Telangana, India
| | - Saman Pathan
- Department of Pharmacology, All India Institute of Medical Sciences, Bhopal, India
| | - Sushil Sharma
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Mangalagiri, Andhra Pradesh, 522503, India
| | - Madhav Rao Chavan
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Mangalagiri, Andhra Pradesh, 522503, India
| | | | - R Padmavathi
- SVS Medical College and Hospital, Mahbubnagar, Telangana, India
| | - M Manjula
- Balaji College of Nursing, Tirupathi, Andhra Pradesh, India
| | - Manik Chhabra
- Department of Pharmacy Practice, Indo-Soviet Friendship College of Pharmacy, Moga, India
| | - Rupesh Gupta
- Department of Internal Medicine, GMC, Shahdol, Madhya Pradesh, India
| | | | | | - Sofia Mudda
- Department of AYUSH, All India Institute of Medical Sciences, Bhopal, India
| |
Collapse
|
26
|
Pauletto PJT, Delgado CP, da Rocha JBT. Acid sphingomyelinase (ASM) and COVID-19: A review of the potential use of ASM inhibitors against SARS-CoV-2. Cell Biochem Funct 2023; 41:284-295. [PMID: 36929117 DOI: 10.1002/cbf.3789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 03/18/2023]
Abstract
In the last 2 years, different pharmacological agents have been indicated as potential inhibitors of SARS-CoV-2 in vitro. Specifically, drugs termed as functional inhibitors of acid sphingomyelinase (FIASMAs) have proved to inhibit the SARS-CoV-2 replication using different types of cells. Those therapeutic agents share several chemical structure characteristics and some well-known representatives are fluoxetine, escitalopram, fluvoxamine, and others. Most of the FIASMAs are primarily used as effective therapeutic agents to treat different pathologies, therefore, they are natural drug candidates for repositioning strategy. In this review, we summarize the two main proposed mechanisms mediating acid sphingomyelinase (ASM) inhibition and how they can explain the inhibition of SARS-CoV-2 replication by FIASMAs. The first mechanism implies a disruption in the lysosomal pH fall as the endosome-lysosome moves toward the interior of the cell. In fact, changes in cholesterol levels in endosome-lysosome membranes, which are associated with ASM inhibition is thought to be mediated by lysosomal proton pump (ATP-ase) inactivation. The second mechanism involves the formation of an extracellular ceramide-rich domain, which is blocked by FIASMAs. The ceramide-rich domains are believed to facilitate the SARS-CoV-2 entrance into the host cells.
Collapse
Affiliation(s)
- Pedro José Tronco Pauletto
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| | - Cassia Pereira Delgado
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| | - João Batista Teixeira da Rocha
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| |
Collapse
|
27
|
Fu J, Zhu F, Xu CJ, Li Y. Metabolomics meets systems immunology. EMBO Rep 2023; 24:e55747. [PMID: 36916532 PMCID: PMC10074123 DOI: 10.15252/embr.202255747] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 12/24/2022] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Metabolic processes play a critical role in immune regulation. Metabolomics is the systematic analysis of small molecules (metabolites) in organisms or biological samples, providing an opportunity to comprehensively study interactions between metabolism and immunity in physiology and disease. Integrating metabolomics into systems immunology allows the exploration of the interactions of multilayered features in the biological system and the molecular regulatory mechanism of these features. Here, we provide an overview on recent technological developments of metabolomic applications in immunological research. To begin, two widely used metabolomics approaches are compared: targeted and untargeted metabolomics. Then, we provide a comprehensive overview of the analysis workflow and the computational tools available, including sample preparation, raw spectra data preprocessing, data processing, statistical analysis, and interpretation. Third, we describe how to integrate metabolomics with other omics approaches in immunological studies using available tools. Finally, we discuss new developments in metabolomics and its prospects for immunology research. This review provides guidance to researchers using metabolomics and multiomics in immunity research, thus facilitating the application of systems immunology to disease research.
Collapse
Affiliation(s)
- Jianbo Fu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany.,College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Cheng-Jian Xu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany.,Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Yang Li
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany.,Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
28
|
Wilson AD, Forse LB. Potential for Early Noninvasive COVID-19 Detection Using Electronic-Nose Technologies and Disease-Specific VOC Metabolic Biomarkers. SENSORS (BASEL, SWITZERLAND) 2023; 23:2887. [PMID: 36991597 PMCID: PMC10054641 DOI: 10.3390/s23062887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/19/2023] [Accepted: 03/03/2023] [Indexed: 06/12/2023]
Abstract
The established efficacy of electronic volatile organic compound (VOC) detection technologies as diagnostic tools for noninvasive early detection of COVID-19 and related coronaviruses has been demonstrated from multiple studies using a variety of experimental and commercial electronic devices capable of detecting precise mixtures of VOC emissions in human breath. The activities of numerous global research teams, developing novel electronic-nose (e-nose) devices and diagnostic methods, have generated empirical laboratory and clinical trial test results based on the detection of different types of host VOC-biomarker metabolites from specific chemical classes. COVID-19-specific volatile biomarkers are derived from disease-induced changes in host metabolic pathways by SARS-CoV-2 viral pathogenesis. The unique mechanisms proposed from recent researchers to explain how COVID-19 causes damage to multiple organ systems throughout the body are associated with unique symptom combinations, cytokine storms and physiological cascades that disrupt normal biochemical processes through gene dysregulation to generate disease-specific VOC metabolites targeted for e-nose detection. This paper reviewed recent methods and applications of e-nose and related VOC-detection devices for early, noninvasive diagnosis of SARS-CoV-2 infections. In addition, metabolomic (quantitative) COVID-19 disease-specific chemical biomarkers, consisting of host-derived VOCs identified from exhaled breath of patients, were summarized as possible sources of volatile metabolic biomarkers useful for confirming and supporting e-nose diagnoses.
Collapse
Affiliation(s)
- Alphus Dan Wilson
- Pathology Department, Center for Forest Health & Disturbance, Forest Genetics and Ecosystems Biology, Southern Research Station, USDA Forest Service, Stoneville, MS 38776, USA
| | - Lisa Beth Forse
- Southern Hardwoods Laboratory, Southern Research Station, USDA Forest Service, Stoneville, MS 38776, USA
| |
Collapse
|
29
|
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
|
30
|
Bourgin M, Durand S, Kroemer G. Diagnostic, Prognostic and Mechanistic Biomarkers of COVID-19 Identified by Mass Spectrometric Metabolomics. Metabolites 2023; 13:metabo13030342. [PMID: 36984782 PMCID: PMC10056171 DOI: 10.3390/metabo13030342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/14/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
A number of studies have assessed the impact of SARS-CoV-2 infection and COVID-19 severity on the metabolome of exhaled air, saliva, plasma, and urine to identify diagnostic and prognostic biomarkers. In spite of the richness of the literature, there is no consensus about the utility of metabolomic analyses for the management of COVID-19, calling for a critical assessment of the literature. We identified mass spectrometric metabolomic studies on specimens from SARS-CoV2-infected patients and subjected them to a cross-study comparison. We compared the clinical design, technical aspects, and statistical analyses of published studies with the purpose to identify the most relevant biomarkers. Several among the metabolites that are under- or overrepresented in the plasma from patients with COVID-19 may directly contribute to excessive inflammatory reactions and deficient immune control of SARS-CoV2, hence unraveling important mechanistic connections between whole-body metabolism and the course of the disease. Altogether, it appears that mass spectrometric approaches have a high potential for biomarker discovery, especially if they are subjected to methodological standardization.
Collapse
Affiliation(s)
- Mélanie Bourgin
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805 Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75005 Paris, France
- Correspondence:
| | - Sylvère Durand
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805 Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75005 Paris, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805 Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75005 Paris, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, 75610 Paris, France
| |
Collapse
|
31
|
Bruzzone C, Conde R, Embade N, Mato JM, Millet O. Metabolomics as a powerful tool for diagnostic, pronostic and drug intervention analysis in COVID-19. Front Mol Biosci 2023; 10:1111482. [PMID: 36876049 PMCID: PMC9975567 DOI: 10.3389/fmolb.2023.1111482] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
COVID-19 currently represents one of the major health challenges worldwide. Albeit its infectious character, with onset affectation mainly at the respiratory track, it is clear that the pathophysiology of COVID-19 has a systemic character, ultimately affecting many organs. This feature enables the possibility of investigating SARS-CoV-2 infection using multi-omic techniques, including metabolomic studies by chromatography coupled to mass spectrometry or by nuclear magnetic resonance (NMR) spectroscopy. Here we review the extensive literature on metabolomics in COVID-19, that unraveled many aspects of the disease including: a characteristic metabotipic signature associated to COVID-19, discrimination of patients according to severity, effect of drugs and vaccination treatments and the characterization of the natural history of the metabolic evolution associated to the disease, from the infection onset to full recovery or long-term and long sequelae of COVID.
Collapse
Affiliation(s)
- Chiara Bruzzone
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bilbao, Bizkaia, Spain
| | - Ricardo Conde
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bilbao, Bizkaia, Spain
| | - Nieves Embade
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bilbao, Bizkaia, Spain
| | - José M. Mato
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bilbao, Bizkaia, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Oscar Millet
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bilbao, Bizkaia, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
32
|
Chisanga M, Williams H, Boudreau D, Pelletier JN, Trottier S, Masson JF. Label-Free SERS for Rapid Differentiation of SARS-CoV-2-Induced Serum Metabolic Profiles in Non-Hospitalized Adults. Anal Chem 2023; 95:3638-3646. [PMID: 36763490 PMCID: PMC9940618 DOI: 10.1021/acs.analchem.2c04514] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
COVID-19 represents a multi-system infectious disease with broad-spectrum manifestations, including changes in host metabolic processes connected to the disease pathogenesis. Understanding biochemical dysregulation patterns as a consequence of COVID-19 illness promises to be crucial for tracking disease course and clinical outcomes. Surface-enhanced Raman scattering (SERS) has attracted considerable interest in biomedical diagnostics for the sensitive detection of intrinsic profiles of unique fingerprints of serum biomolecules indicative of SARS-CoV-2 infection in a label-free format. Here, we applied label-free SERS and chemometrics for rapid interrogation of temporal metabolic dynamics in longitudinal sera of mildly infected non-hospitalized patients (n = 22), at 4 and 16 weeks post PCR-positive diagnosis, and compared them with negative controls (n = 8). SERS spectral markers revealed distinct metabolic profiles in patient sera that significantly deviated from the healthy metabolic state at the two sampling time intervals. Multivariate and univariate analyses of the spectral data identified abundance dynamics in amino acids, lipids, and protein vibrations as the key spectral features underlying the metabolic differences detected in convalescent samples and perhaps associated with patient recovery progression. A validation study performed using spontaneous Raman spectroscopy yielded spectral data results that corroborated SERS spectral findings and confirmed the detected disease-specific molecular phenotypes in clinical samples. Label-free SERS promises to be a valuable analytical technique for rapid screening of the metabolic phenotype induced by SARS-CoV-2 infection to allow appropriate healthcare intervention.
Collapse
Affiliation(s)
- Malama Chisanga
- Department
of Chemistry, Québec Centre for Advanced Materials (QCAM),
Regroupement Québécois sur les Matériaux de Pointe
(RQMP), and Centre Interdisciplinaire de Recherche sur le Cerveau
et l’Apprentissage (CIRCA), Université
de Montréal, CP 6128 Succ. Centre-Ville, Montreal, Québec H3C 3J7, Canada
| | - Hannah Williams
- Department
of Chemistry, Québec Centre for Advanced Materials (QCAM),
Regroupement Québécois sur les Matériaux de Pointe
(RQMP), and Centre Interdisciplinaire de Recherche sur le Cerveau
et l’Apprentissage (CIRCA), Université
de Montréal, CP 6128 Succ. Centre-Ville, Montreal, Québec H3C 3J7, Canada
| | - Denis Boudreau
- Department
of Chemistry and Centre for Optics, Photonics and Lasers (COPL), Université Laval, 1045, av. de la Médecine, Québec, Québec G1V 0A6, Canada
| | - Joelle N. Pelletier
- Department
of Chemistry, Department of Biochemistry and PROTEO, Québec
Network for Research on Protein Function, Engineering and Applications, Université de Montréal, CP 6128 Succ. Centre-Ville, Montreal, Québec H3C 3J7, Canada
| | - Sylvie Trottier
- Centre
de Recherche du Centre Hospitalier Universitaire de Québec
and Département de Microbiologie-Infectiologie et d’Immunologie, Université Laval, 2705, boulevard Laurier, Québec, Québec G1V 4G2, Canada
| | - Jean-Francois Masson
- Department
of Chemistry, Québec Centre for Advanced Materials (QCAM),
Regroupement Québécois sur les Matériaux de Pointe
(RQMP), and Centre Interdisciplinaire de Recherche sur le Cerveau
et l’Apprentissage (CIRCA), Université
de Montréal, CP 6128 Succ. Centre-Ville, Montreal, Québec H3C 3J7, Canada,. Phone: +1-514-343-7342
| |
Collapse
|
33
|
Qin R, He L, Yang Z, Jia N, Chen R, Xie J, Fu W, Chen H, Lin X, Huang R, Luo T, Liu Y, Yao S, Jiang M, Li J. Identification of Parameters Representative of Immune Dysfunction in Patients with Severe and Fatal COVID-19 Infection: a Systematic Review and Meta-analysis. Clin Rev Allergy Immunol 2023; 64:33-65. [PMID: 35040086 PMCID: PMC8763427 DOI: 10.1007/s12016-021-08908-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2021] [Indexed: 01/26/2023]
Abstract
Abnormal immunological indicators associated with disease severity and mortality in patients with COVID-19 have been reported in several observational studies. However, there are marked heterogeneities in patient characteristics and research methodologies in these studies. We aimed to provide an updated synthesis of the association between immune-related indicators and COVID-19 prognosis. We conducted an electronic search of PubMed, Scopus, Ovid, Willey, Web of Science, Cochrane library, and CNKI for studies reporting immunological and/or immune-related parameters, including hematological, inflammatory, coagulation, and biochemical variables, tested on hospital admission of COVID-19 patients with different severities and outcomes. A total of 145 studies were included in the current meta-analysis, with 26 immunological, 11 hematological, 5 inflammatory, 4 coagulation, and 10 biochemical variables reported. Of them, levels of cytokines, including IL-1β, IL-1Ra, IL-2R, IL-4, IL-6, IL-8, IL-10, IL-18, TNF-α, IFN-γ, IgA, IgG, and CD4+ T/CD8+ T cell ratio, WBC, neutrophil, platelet, ESR, CRP, ferritin, SAA, D-dimer, FIB, and LDH were significantly increased in severely ill patients or non-survivors. Moreover, non-severely ill patients or survivors presented significantly higher counts of lymphocytes, monocytes, lymphocyte/monocyte ratio, eosinophils, CD3+ T,CD4+T and CD8+T cells, B cells, and NK cells. The currently updated meta-analysis primarily identified a hypercytokinemia profile with the severity and mortality of COVID-19 containing IL-1β, IL-1Ra, IL-2R, IL-4, IL-6, IL-8, IL-10, IL-18, TNF-α, and IFN-γ. Impaired innate and adaptive immune responses, reflected by decreased eosinophils, lymphocytes, monocytes, B cells, NK cells, T cells, and their subtype CD4+ and CD8+ T cells, and augmented inflammation, coagulation dysfunction, and nonpulmonary organ injury, were marked features of patients with poor prognosis. Therefore, parameters of immune response dysfunction combined with inflammatory, coagulated, or nonpulmonary organ injury indicators may be more sensitive to predict severe patients and those non-survivors.
Collapse
Affiliation(s)
- Rundong Qin
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Li He
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhaowei Yang
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Nan Jia
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ruchong Chen
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiaxing Xie
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wanyi Fu
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hao Chen
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xinliu Lin
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Renbin Huang
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tian Luo
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yukai Liu
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Siyang Yao
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mei Jiang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Jing Li
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
34
|
An NMR-Based Model to Investigate the Metabolic Phenoreversion of COVID-19 Patients throughout a Longitudinal Study. Metabolites 2022; 12:metabo12121206. [PMID: 36557244 PMCID: PMC9788519 DOI: 10.3390/metabo12121206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/19/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
After SARS-CoV-2 infection, the molecular phenoreversion of the immunological response and its associated metabolic dysregulation are required for a full recovery of the patient. This process is patient-dependent due to the manifold possibilities induced by virus severity, its phylogenic evolution and the vaccination status of the population. We have here investigated the natural history of COVID-19 disease at the molecular level, characterizing the metabolic and immunological phenoreversion over time in large cohorts of hospitalized severe patients (n = 886) and non-hospitalized recovered patients that self-reported having passed the disease (n = 513). Non-hospitalized recovered patients do not show any metabolic fingerprint associated with the disease or immune alterations. Acute patients are characterized by the metabolic and lipidomic dysregulation that accompanies the exacerbated immunological response, resulting in a slow recovery time with a maximum probability of around 62 days. As a manifestation of the heterogeneity in the metabolic phenoreversion, age and severity become factors that modulate their normalization time which, in turn, correlates with changes in the atherogenesis-associated chemokine MCP-1. Our results are consistent with a model where the slow metabolic normalization in acute patients results in enhanced atherosclerotic risk, in line with the recent observation of an elevated number of cardiovascular episodes found in post-COVID-19 cohorts.
Collapse
|
35
|
Asadi Anar M, Foroughi E, Sohrabi E, Peiravi S, Tavakoli Y, Kameli Khouzani M, Behshood P, Shamshiri M, Faridzadeh A, Keylani K, Langari SF, Ansari A, Khalaji A, Garousi S, Mottahedi M, Honari S, Deravi N. Selective serotonin reuptake inhibitors: New hope in the fight against COVID-19. Front Pharmacol 2022; 13:1036093. [PMID: 36532776 PMCID: PMC9748354 DOI: 10.3389/fphar.2022.1036093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
The emerging COVID-19 pandemic led to a dramatic increase in global mortality and morbidity rates. As in most infections, fatal complications of coronavirus affliction are triggered by an untrammeled host inflammatory response. Cytokine storms created by high levels of interleukin and other cytokines elucidate the pathology of severe COVID-19. In this respect, repurposing drugs that are already available and might exhibit anti-inflammatory effects have received significant attention. With the in vitro and clinical investigation of several studies on the effect of antidepressants on COVID-19 prognosis, previous data suggest that selective serotonin reuptake inhibitors (SSRIs) might be the new hope for the early treatment of severely afflicted patients. SSRIs' low cost and availability make them potentially eligible for COVID-19 repurposing. This review summarizes current achievements and literature about the connection between SSRIs administration and COVID-19 prognosis.
Collapse
Affiliation(s)
- Mahsa Asadi Anar
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elaheh Foroughi
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elika Sohrabi
- Department of Medicine, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Samira Peiravi
- Department of Emergency Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yasaman Tavakoli
- Department of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran, Iran
| | | | - Parisa Behshood
- Department of Microbiology, Young Researchers and Elite Club, Islamic Azad University, Shahrekord, Iran
| | - Melika Shamshiri
- School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kimia Keylani
- School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Faride Langari
- Department of Ophthalmology, Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Akram Ansari
- Shantou University Medical College, Shantou, Guangdong, China
| | | | - Setareh Garousi
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehran Mottahedi
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Honari
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Jusof FF, Lim CK, Aziz FN, Soe HJ, Raju CS, Sekaran SD, Guillemin GJ. The Cytokines CXCL10 and CCL2 and the Kynurenine Metabolite Anthranilic Acid Accurately Predict Patients at Risk of Developing Dengue With Warning Signs. J Infect Dis 2022; 226:1964-1973. [PMID: 35767283 DOI: 10.1093/infdis/jiac273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/28/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The resolution or aggravation of dengue infection depends on the patient's immune response during the critical phase. Cytokines released by immune cells increase with the worsening severity of dengue infections. Cytokines activate the kynurenine pathway (KP) and the extent of KP activation then influences disease severity. METHODS KP metabolites and cytokines in plasma samples of patients with dengue infection (dengue without warning signs [DWS-], dengue with warning signs [DWS+], or severe dengue) were analyzed. Cytokines (interferon gamma [IFN-ɣ], tumor necrosis factor, interleukin 6, CXCL10/interferon-inducile protein 10 [IP-10], interleukin 18 [IL-18], CCL2/monocyte chemoattractant protein-1 [MCP-1], and CCL4/macrophage inflammatory protein-1beta [MIP-1β] were assessed by a Human Luminex Screening Assay, while KP metabolites (tryptophan, kynurenine, anthranilic acid [AA], picolinic acid, and quinolinic acid) were assessed by ultra-high-performance liquid chromatography and Gas Chromatography Mass Spectrophotometry [GCMS] assays. RESULTS Patients with DWS+ had increased activation of the KP where kynurenine-tryptophan ratio, anthranilic acid, and picolinic acid were elevated. These patients also had higher levels of the cytokines IFN-ɣ, CXCL10, CCL4, and IL-18 than those with DWS-. Further receiver operating characteristic analysis identified 3 prognostic biomarker candidates, CXCL10, CCL2, and AA, which predicted patients with higher risks of developing DWS+ with an accuracy of 97%. CONCLUSIONS The data suggest a unique biochemical signature in patients with DWS+. CXCL10 and CCL2 together with AA are potential prognostic biomarkers that discern patients with higher risk of developing DWS+ at earlier stages of infection.
Collapse
Affiliation(s)
- Felicita Fedelis Jusof
- Department of Physiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Chai K Lim
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, New South Wales, Australia
| | - Fazidatul Nadhirah Aziz
- Department of Biomedical Sciences, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Hui Jen Soe
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Chandramathi Samudi Raju
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Shamala Devi Sekaran
- Faculty of Medical and Health Sciences, UCSI University Springhill Campus, Bandar Springhill, Port Dickson, Negri Sembilan, Malaysia
| | - Gilles J Guillemin
- Neuroinflammation Group, Motor Neurone Disease Research Centre, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| |
Collapse
|
37
|
Péricat D, Leon-Icaza SA, Sanchez Rico M, Mühle C, Zoicas I, Schumacher F, Planès R, Mazars R, Gros G, Carpinteiro A, Becker KA, Izopet J, Strub-Wourgaft N, Sjö P, Neyrolles O, Kleuser B, Limosin F, Gulbins E, Kornhuber J, Meunier E, Hoertel N, Cougoule C. Antiviral and Anti-Inflammatory Activities of Fluoxetine in a SARS-CoV-2 Infection Mouse Model. Int J Mol Sci 2022; 23:13623. [PMID: 36362409 PMCID: PMC9657171 DOI: 10.3390/ijms232113623] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/11/2022] [Accepted: 10/26/2022] [Indexed: 08/27/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic continues to cause significant morbidity and mortality worldwide. Since a large portion of the world's population is currently unvaccinated or incompletely vaccinated and has limited access to approved treatments against COVID-19, there is an urgent need to continue research on treatment options, especially those at low cost and which are immediately available to patients, particularly in low- and middle-income countries. Prior in vitro and observational studies have shown that fluoxetine, possibly through its inhibitory effect on the acid sphingomyelinase/ceramide system, could be a promising antiviral and anti-inflammatory treatment against COVID-19. In this report, we evaluated the potential antiviral and anti-inflammatory activities of fluoxetine in a K18-hACE2 mouse model of SARS-CoV-2 infection, and against variants of concern in vitro, i.e., SARS-CoV-2 ancestral strain, Alpha B.1.1.7, Gamma P1, Delta B1.617 and Omicron BA.5. Fluoxetine, administrated after SARS-CoV-2 infection, significantly reduced lung tissue viral titres and expression of several inflammatory markers (i.e., IL-6, TNFα, CCL2 and CXCL10). It also inhibited the replication of all variants of concern in vitro. A modulation of the ceramide system in the lung tissues, as reflected by the increase in the ratio HexCer 16:0/Cer 16:0 in fluoxetine-treated mice, may contribute to explain these effects. Our findings demonstrate the antiviral and anti-inflammatory properties of fluoxetine in a K18-hACE2 mouse model of SARS-CoV-2 infection, and its in vitro antiviral activity against variants of concern, establishing fluoxetine as a very promising candidate for the prevention and treatment of SARS-CoV-2 infection and disease pathogenesis.
Collapse
Affiliation(s)
- David Péricat
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Stephen Adonai Leon-Icaza
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Marina Sanchez Rico
- Faculté de Santé, Université Paris Cité, 75006 Paris, France
- Département de Psychiatrie et d’Addictologie de l’Adulte et du Sujet Agé, Assistance Publique-Hôpitaux de Paris (AP-HP), DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, 92130 Issy-les-Moulineaux, France
- INSERM, Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR_S1266, 75014 Paris, France
| | - Christiane Mühle
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Iulia Zoicas
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Fabian Schumacher
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany
| | - Rémi Planès
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Raoul Mazars
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Germain Gros
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Alexander Carpinteiro
- Institute for Molecular Biology, University Medicine Essen, University of Duisburg-Essen, 47057 Essen, Germany
| | - Katrin Anne Becker
- Institute for Molecular Biology, University Medicine Essen, University of Duisburg-Essen, 47057 Essen, Germany
| | - Jacques Izopet
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), Université Toulouse, CNRS, INSERM, UPS, 31300 Toulouse, France
- Laboratoire de Virologie, CHU Toulouse, Hôpital Purpan, 31300 Toulouse, France
| | | | - Peter Sjö
- Drugs for Neglected Diseases Initiative, 1202 Geneva, Switzerland
| | - Olivier Neyrolles
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Burkhard Kleuser
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany
| | - Frédéric Limosin
- Faculté de Santé, Université Paris Cité, 75006 Paris, France
- Département de Psychiatrie et d’Addictologie de l’Adulte et du Sujet Agé, Assistance Publique-Hôpitaux de Paris (AP-HP), DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, 92130 Issy-les-Moulineaux, France
- INSERM, Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR_S1266, 75014 Paris, France
| | - Erich Gulbins
- Institute for Molecular Biology, University Medicine Essen, University of Duisburg-Essen, 47057 Essen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Etienne Meunier
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| | - Nicolas Hoertel
- Faculté de Santé, Université Paris Cité, 75006 Paris, France
- Département de Psychiatrie et d’Addictologie de l’Adulte et du Sujet Agé, Assistance Publique-Hôpitaux de Paris (AP-HP), DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, 92130 Issy-les-Moulineaux, France
- INSERM, Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR_S1266, 75014 Paris, France
| | - Céline Cougoule
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, 31000 Toulouse, France
| |
Collapse
|
38
|
Hoertel N, Sánchez-Rico M, Kornhuber J, Gulbins E, Reiersen AM, Lenze EJ, Fritz BA, Jalali F, Mills EJ, Cougoule C, Carpinteiro A, Mühle C, Becker KA, Boulware DR, Blanco C, Alvarado JM, Strub-Wourgaft N, Lemogne C, Limosin F. Antidepressant Use and Its Association with 28-Day Mortality in Inpatients with SARS-CoV-2: Support for the FIASMA Model against COVID-19. J Clin Med 2022; 11:5882. [PMID: 36233753 PMCID: PMC9572995 DOI: 10.3390/jcm11195882] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 01/29/2023] Open
Abstract
To reduce Coronavirus Disease 2019 (COVID-19)-related mortality and morbidity, widely available oral COVID-19 treatments are urgently needed. Certain antidepressants, such as fluvoxamine or fluoxetine, may be beneficial against COVID-19. We included 388,945 adult inpatients who tested positive for SARS-CoV-2 at 36 AP−HP (Assistance Publique−Hôpitaux de Paris) hospitals from 2 May 2020 to 2 November 2021. We compared the prevalence of antidepressant use at admission in a 1:1 ratio matched analytic sample with and without COVID-19 (N = 82,586), and assessed its association with 28-day all-cause mortality in a 1:1 ratio matched analytic sample of COVID-19 inpatients with and without antidepressant use at admission (N = 1482). Antidepressant use was significantly less prevalent in inpatients with COVID-19 than in a matched control group of inpatients without COVID-19 (1.9% versus 4.8%; Odds Ratio (OR) = 0.38; 95%CI = 0.35−0.41, p < 0.001). Antidepressant use was significantly associated with reduced 28-day mortality among COVID-19 inpatients (12.8% versus 21.2%; OR = 0.55; 95%CI = 0.41−0.72, p < 0.001), particularly at daily doses of at least 40 mg fluoxetine equivalents. Antidepressants with high FIASMA (Functional Inhibitors of Acid Sphingomyelinase) activity seem to drive both associations. These treatments may reduce SARS-CoV-2 infections and COVID-19-related mortality in inpatients, and may be appropriate for prophylaxis and/or COVID-19 therapy for outpatients or inpatients.
Collapse
Affiliation(s)
- Nicolas Hoertel
- Institut de Psychiatrie et Neuroscience de Paris, Université Paris Cité, INSERM U1266, F-75014 Paris, France
- AP-HP, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Issy-les-Moulineaux, F-92130 Paris, France
| | - Marina Sánchez-Rico
- AP-HP, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Issy-les-Moulineaux, F-92130 Paris, France
- Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, 28223 Pozuelo de Alarcón (Madrid), Spain
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Erich Gulbins
- Institute for Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany
| | - Angela M. Reiersen
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eric J. Lenze
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bradley A. Fritz
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Farid Jalali
- Department of Gastroenterology, Saddleback Medical Group, Laguna Hills, CA 92653, USA
| | - Edward J. Mills
- Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Céline Cougoule
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, F-31400 Toulouse, France
| | - Alexander Carpinteiro
- Institute for Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany
| | - Christiane Mühle
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Katrin Anne Becker
- Institute for Molecular Biology, University Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany
| | - David R. Boulware
- Department of Medicine, Division of Infectious Diseases and International Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Carlos Blanco
- National Institute on Drug Abuse (NIDA), National Institutes of Health, Bethesda, MD 20852, USA
| | - Jesús M. Alvarado
- Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, 28223 Pozuelo de Alarcón (Madrid), Spain
| | - Nathalie Strub-Wourgaft
- COVID-19 Response & Pandemic Preparedness, Drugs for Neglected Diseases Initiative (DNDi), 1202 Geneva, Switzerland
| | - Cédric Lemogne
- Institut de Psychiatrie et Neuroscience de Paris, Université Paris Cité, INSERM U1266, F-75014 Paris, France
- Service de Psychiatrie de l’adulte, AP-HP, Hôpital Hôtel-Dieu, DMU Psychiatrie et Addictologie, F-75004 Paris, France
| | - Frédéric Limosin
- Institut de Psychiatrie et Neuroscience de Paris, Université Paris Cité, INSERM U1266, F-75014 Paris, France
- AP-HP, DMU Psychiatrie et Addictologie, Hôpital Corentin-Celton, Issy-les-Moulineaux, F-92130 Paris, France
| | | |
Collapse
|
39
|
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
|
40
|
Barradas M, Plaza A, Colmenarejo G, Lázaro I, Costa-Machado LF, Martín-Hernández R, Micó V, López-Aceituno JL, Herranz J, Pantoja C, Tejero H, Diaz-Ruiz A, Al-Shahrour F, Daimiel L, Loria-Kohen V, de Molina AR, Efeyan A, Serrano M, Pozo OJ, Sala-Vila A, Fernandez-Marcos PJ. Fatty acids homeostasis during fasting predicts protection from chemotherapy toxicity. Nat Commun 2022; 13:5677. [PMID: 36167809 PMCID: PMC9515185 DOI: 10.1038/s41467-022-33352-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 09/09/2022] [Indexed: 12/27/2022] Open
Abstract
Fasting exerts beneficial effects in mice and humans, including protection from chemotherapy toxicity. To explore the involved mechanisms, we collect blood from humans and mice before and after 36 or 24 hours of fasting, respectively, and measure lipid composition of erythrocyte membranes, circulating micro RNAs (miRNAs), and RNA expression at peripheral blood mononuclear cells (PBMCs). Fasting coordinately affects the proportion of polyunsaturated versus saturated and monounsaturated fatty acids at the erythrocyte membrane; and reduces the expression of insulin signaling-related genes in PBMCs. When fasted for 24 hours before and 24 hours after administration of oxaliplatin or doxorubicin, mice show a strong protection from toxicity in several tissues. Erythrocyte membrane lipids and PBMC gene expression define two separate groups of individuals that accurately predict a differential protection from chemotherapy toxicity, with important clinical implications. Our results reveal a mechanism of fasting associated with lipid homeostasis, and provide biomarkers of fasting to predict fasting-mediated protection from chemotherapy toxicity. Fasting has been reported to protect from chemotherapy-associated toxicity. Here, the authors show that fatty acid profiles in erythrocyte membranes and gene expression from peripheral blood mononuclear cells are associated to the fasting-mediated benefits during cancer treatment in mice and patients.
Collapse
Affiliation(s)
- Marta Barradas
- Metabolic Syndrome Group-BIOPROMET, CEI UAM+CSIC, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain.
| | - Adrián Plaza
- Metabolic Syndrome Group-BIOPROMET, CEI UAM+CSIC, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain.
| | - Gonzalo Colmenarejo
- Biostatistics and Bioinformatics Unit, CEI UAM+CSIC, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain
| | - Iolanda Lázaro
- Cardiovascular risk and nutrition, Hospital del Mar Medical Research Institute-IMIM, Barcelona, Spain
| | - Luis Filipe Costa-Machado
- Metabolic Syndrome Group-BIOPROMET, CEI UAM+CSIC, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain
| | - Roberto Martín-Hernández
- Biostatistics and Bioinformatics Unit, CEI UAM+CSIC, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain
| | - Victor Micó
- Nutritional Genomics of Cardiovascular Disease and Obesity, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain
| | - José Luis López-Aceituno
- Metabolic Syndrome Group-BIOPROMET, CEI UAM+CSIC, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain
| | - Jesús Herranz
- Biostatistics and Bioinformatics Unit, CEI UAM+CSIC, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain
| | - Cristina Pantoja
- Metabolic Syndrome Group-BIOPROMET, CEI UAM+CSIC, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain
| | - Hector Tejero
- Bioinformatics Unit, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
| | - Alberto Diaz-Ruiz
- Nutritional Interventions Group, Precision Nutrition and Aging, CEI UAM+CSIC, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain
| | - Fatima Al-Shahrour
- Bioinformatics Unit, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
| | - Lidia Daimiel
- Nutritional Genomics of Cardiovascular Disease and Obesity, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain
| | - Viviana Loria-Kohen
- Nutrition and Clinical Trials Unit, Platform GENYAL, CEI UAM+CSIC, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain
| | - Ana Ramirez de Molina
- Nutrition and Clinical Trials Unit, Platform GENYAL, CEI UAM+CSIC, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain.,Molecular Oncology and Nutritional Genomics of Cancer Group, CEI UAM+CSIC, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain
| | - Alejo Efeyan
- Metabolism and Cell Signaling Group, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Oscar J Pozo
- Applied Metabolomics Research Group, Hospital del Mar Medical Research Institute-(IMIM), Barcelona, Spain
| | - Aleix Sala-Vila
- Cardiovascular risk and nutrition, Hospital del Mar Medical Research Institute-IMIM, Barcelona, Spain.,Fatty Acid Research Institute, Sioux Falls, SD, USA
| | - Pablo J Fernandez-Marcos
- Metabolic Syndrome Group-BIOPROMET, CEI UAM+CSIC, Madrid Institute for Advanced Studies-IMDEA Food, Madrid, Spain.
| |
Collapse
|
41
|
Ceperuelo-Mallafré V, Reverté L, Peraire J, Madeira A, Maymó-Masip E, López-Dupla M, Gutierrez-Valencia A, Ruiz-Mateos E, Buzón MJ, Jorba R, Vendrell J, Auguet T, Olona M, Vidal F, Rull A, Fernández-Veledo S. Circulating pyruvate is a potent prognostic marker for critical COVID-19 outcomes. Front Immunol 2022; 13:912579. [PMID: 36189213 PMCID: PMC9515795 DOI: 10.3389/fimmu.2022.912579] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background Coronavirus-19 (COVID-19) disease is driven by an unchecked immune response to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus which alters host mitochondrial-associated mechanisms. Compromised mitochondrial health results in abnormal reprogramming of glucose metabolism, which can disrupt extracellular signalling. We hypothesized that examining mitochondrial energy-related signalling metabolites implicated in host immune response to SARS-CoV-2 infection would provide potential biomarkers for predicting the risk of severe COVID-19 illness. Methods We used a semi-targeted serum metabolomics approach in 273 patients with different severity grades of COVID-19 recruited at the acute phase of the infection to determine the relative abundance of tricarboxylic acid (Krebs) cycle-related metabolites with known extracellular signaling properties (pyruvate, lactate, succinate and α-ketoglutarate). Abundance levels of energy-related metabolites were evaluated in a validation cohort (n=398) using quantitative fluorimetric assays. Results Increased levels of four energy-related metabolites (pyruvate, lactate, a-ketoglutarate and succinate) were found in critically ill COVID-19 patients using semi-targeted and targeted approaches (p<0.05). The combined strategy proposed herein enabled us to establish that circulating pyruvate levels (p<0.001) together with body mass index (p=0.025), C-reactive protein (p=0.039), D-Dimer (p<0.001) and creatinine (p=0.043) levels, are independent predictors of critical COVID-19. Furthermore, classification and regression tree (CART) analysis provided a cut-off value of pyruvate in serum (24.54 µM; p<0.001) as an early criterion to accurately classify patients with critical outcomes. Conclusion Our findings support the link between COVID-19 pathogenesis and immunometabolic dysregulation, and show that fluorometric quantification of circulating pyruvate is a cost-effective clinical decision support tool to improve patient stratification and prognosis prediction.
Collapse
Affiliation(s)
- Victòria Ceperuelo-Mallafré
- Universitat Rovira i Virgili (URV), Tarragona, Spain
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metaboílicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Laia Reverté
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC)-Instituto de Salud Carlos III, Madrid, Spain
| | - Joaquim Peraire
- Universitat Rovira i Virgili (URV), Tarragona, Spain
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC)-Instituto de Salud Carlos III, Madrid, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
| | - Ana Madeira
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metaboílicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Elsa Maymó-Masip
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metaboílicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel López-Dupla
- Universitat Rovira i Virgili (URV), Tarragona, Spain
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC)-Instituto de Salud Carlos III, Madrid, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
| | - Alicia Gutierrez-Valencia
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, Consejo Superior de Investigaciones Científicas (CSIC), University of Seville, Seville, Spain
| | - Ezequiel Ruiz-Mateos
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, Consejo Superior de Investigaciones Científicas (CSIC), University of Seville, Seville, Spain
| | - Maria José Buzón
- Infectious Diseases Department, Vall d’Hebron Institute of Research (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, (VHIR) Task Force COVID-19, Barcelona, Spain
| | - Rosa Jorba
- Universitat Rovira i Virgili (URV), Tarragona, Spain
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
| | - Joan Vendrell
- Universitat Rovira i Virgili (URV), Tarragona, Spain
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metaboílicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
| | - Teresa Auguet
- Universitat Rovira i Virgili (URV), Tarragona, Spain
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
| | - Montserrat Olona
- Universitat Rovira i Virgili (URV), Tarragona, Spain
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC)-Instituto de Salud Carlos III, Madrid, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
| | - Francesc Vidal
- Universitat Rovira i Virgili (URV), Tarragona, Spain
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC)-Instituto de Salud Carlos III, Madrid, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
| | - Anna Rull
- Universitat Rovira i Virgili (URV), Tarragona, Spain
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC)-Instituto de Salud Carlos III, Madrid, Spain
- Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
| | - Sonia Fernández-Veledo
- Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metaboílicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
42
|
Notarbartolo V, Montante C, Ferrante G, Giuffrè M. Antioxidant Effects of Dietary Supplements on Adult COVID-19 Patients: Why Do We Not Also Use Them in Children? Antioxidants (Basel) 2022; 11:antiox11091638. [PMID: 36139712 PMCID: PMC9495518 DOI: 10.3390/antiox11091638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/09/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Respiratory tract infections (RTIs) are very common in children, especially in the first five years of life, and several viruses, such as the influenza virus, Respiratory Syncytial Virus, and Rhinovirus, are triggers for symptoms that usually affect the upper airways. It has been known that during respiratory viral infections, a condition of oxidative stress (OS) occurs, and many studies have suggested the potential use of antioxidants as complementary components in prophylaxis and/or therapy of respiratory viral infections. Preliminary data have demonstrated that antioxidants may also interfere with the new coronavirus 2’s entry and replication in human cells, and that they have a role in the downregulation of several pathogenetic mechanisms involved in disease severity. Starting from preclinical data, the aim of this narrative review is to evaluate the current evidence about the main antioxidants that are potentially useful for preventing and treating Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection in adults and to speculate on their possible use in children by exploring the most relevant issues affecting their use in clinical practice, as well as the associated evidence gaps and research limitations.
Collapse
Affiliation(s)
- Veronica Notarbartolo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialities, University of Palermo, 90128 Palermo, Italy
- Correspondence:
| | - Claudio Montante
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialities, University of Palermo, 90128 Palermo, Italy
| | - Giuliana Ferrante
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, Pediatric Division, University of Verona, 37134 Verona, Italy
| | - Mario Giuffrè
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialities, University of Palermo, 90128 Palermo, Italy
| |
Collapse
|
43
|
Association between antidepressant use and ED or hospital visits in outpatients with SARS-CoV-2. Transl Psychiatry 2022; 12:341. [PMID: 35995770 PMCID: PMC9395392 DOI: 10.1038/s41398-022-02109-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 02/08/2023] Open
Abstract
Antidepressants have previously been associated with better outcomes in patients hospitalized with COVID-19, but their effect on clinical deterioration among ambulatory patients has not been fully explored. The objective of this study was to assess whether antidepressant exposure was associated with reduced emergency department (ED) or hospital visits among ambulatory patients with SARS-CoV-2 infection. This retrospective cohort study included adult patients (N = 25 034) with a positive SARS-CoV-2 test performed in a non-hospital setting. Logistic regression analyses tested associations between home use of antidepressant medications and a composite outcome of ED visitation or hospital admission within 30 days. Secondary exposures included individual antidepressants and antidepressants with functional inhibition of acid sphingomyelinase (FIASMA) activity. Patients with antidepressant exposure were less likely to experience the primary composite outcome compared to patients without antidepressant exposure (adjusted odds ratio [aOR] 0.89, 95% CI 0.79-0.99, p = 0.04). This association was only observed with daily doses of at least 20 mg fluoxetine-equivalent (aOR 0.87, 95% CI 0.77-0.99, p = 0.04), but not with daily doses lower than 20 mg fluoxetine-equivalent (aOR 0.94, 95% CI 0.80-1.11, p = 0.48). In exploratory secondary analyses, the outcome incidence was also reduced with exposure to selective serotonin reuptake inhibitors (aOR 0.87, 95% CI 0.75-0.99, p = 0.04), bupropion (aOR 0.70, 95% CI 0.55-0.90, p = 0.005), and FIASMA antidepressant drugs (aOR 0.87, 95% CI 0.77-0.99, p = 0.03). Antidepressant exposure was associated with a reduced incidence of emergency department visitation or hospital admission among SARS-CoV-2 positive patients, in a dose-dependent manner. These data support the FIASMA model of antidepressants' effects against COVID-19.
Collapse
|
44
|
Almulla AF, Supasitthumrong T, Tunvirachaisakul C, Algon AAA, Al-Hakeim HK, Maes M. The tryptophan catabolite or kynurenine pathway in COVID-19 and critical COVID-19: a systematic review and meta-analysis. BMC Infect Dis 2022; 22:615. [PMID: 35840908 PMCID: PMC9284970 DOI: 10.1186/s12879-022-07582-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/30/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is accompanied by activated immune-inflammatory pathways and oxidative stress, which both induce indoleamine-2,3-dioxygenase (IDO), a key enzyme of the tryptophan (TRP) catabolite (TRYCAT) pathway. The aim of this study was to systematically review and meta-analyze the status of the TRYCAT pathway, including the levels of TRP and kynurenine (KYN) and the activity of IDO, as measured by the ratio of KYN/TRP. METHODS This systematic review searched PubMed, Google Scholar, and Web of Sciences and included 14 articles that compared TRP and tryptophan catabolites (TRYCATs) in COVID-19 patients versus non-COVID-19 controls, as well as severe/critical versus mild/moderate COVID-19. The analysis was done on a total of 1269 people, including 794 COVID-19 patients and 475 controls. RESULTS The results show a significant (p < 0.0001) increase in the KYN/TRP ratio (standardized mean difference, SMD = 1.099, 95% confidence interval, CI: 0.714; 1.484) and KYN (SMD = 1.123, 95% CI: 0.730; 1.516) and significantly lower TRP (SMD = - 1.002, 95%CI: - 1.738; - 0.266) in COVID-19 versus controls. The KYN/TRP ratio (SMD = 0.945, 95%CI: 0.629; 1.262) and KYN (SMD = 0.806, 95%CI: 0.462; 1.149) were also significantly (p < 0.0001) higher and TRP lower (SMD = - 0.909, 95% CI: - 1.569; - 0.249) in severe/critical versus mild/moderate COVID-19. No significant difference was detected in kynurenic acid (KA) and the KA/KYN ratio between COVID-19 patients and controls. CONCLUSIONS Our results indicate increased activity of the IDO enzyme in COVID-19 and severe/critical patients. The TRYCAT pathway is implicated in the pathophysiology and progression of COVID-19 and may signal a worsening outcome of the disease.
Collapse
Affiliation(s)
- Abbas F. Almulla
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, 31001 Iraq
| | | | | | | | | | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
- Department of Psychiatry, IMPACT Strategic Research Centre, Deakin University, Geelong, VIC Australia
| |
Collapse
|
45
|
Martínez-Gómez LE, Ibarra-González I, Fernández-Lainez C, Tusie T, Moreno-Macías H, Martinez-Armenta C, Jimenez-Gutierrez GE, Vázquez-Cárdenas P, Vidal-Vázquez P, Ramírez-Hinojosa JP, Rodríguez-Zulueta AP, Vargas-Alarcón G, Rojas-Velasco G, Sánchez-Muñoz F, Posadas-Sanchez R, Martínez-Ruiz FDJ, Zayago-Angeles DM, Moreno ML, Barajas-Galicia E, Lopez-Cisneros G, Gonzalez-Fernández NC, Ortega-Peña S, Herrera-López B, Olea-Torres J, Juárez-Arias M, Rosas-Vásquez M, Cabrera-Nieto SA, Magaña JJ, Camacho-Rea MDC, Suarez-Ahedo C, Coronado-Zarco I, Valdespino-Vázquez MY, Martínez-Nava GA, Pineda C, Vela-Amieva M, López-Reyes A. Metabolic Reprogramming in SARS-CoV-2 Infection Impacts the Outcome of COVID-19 Patients. Front Immunol 2022; 13:936106. [PMID: 36341434 PMCID: PMC9634751 DOI: 10.3389/fimmu.2022.936106] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/10/2022] [Indexed: 11/25/2023] Open
Abstract
Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) infection triggers inflammatory clinical stages that affect the outcome of patients with coronavirus disease 2019 (COVID-19). Disease severity may be associated with a metabolic imbalance related to amino acids, lipids, and energy-generating pathways. The aim of this study was to characterize the profile of amino acids and acylcarnitines in COVID-19 patients. A multicenter, cross-sectional study was carried out. A total of 453 individuals were classified by disease severity. Levels of 11 amino acids, 31 acylcarnitines, and succinylacetone in serum samples were analyzed by electrospray ionization-triple quadrupole tandem mass spectrometry. Different clusters were observed in partial least squares discriminant analysis, with phenylalanine, alanine, citrulline, proline, and succinylacetone providing the major contribution to the variability in each cluster (variable importance in the projection >1.5). In logistic models adjusted by age, sex, type 2 diabetes mellitus, hypertension, and nutritional status, phenylalanine was associated with critical outcomes (odds ratio=5.3 (95% CI 3.16-9.2) in the severe vs. critical model, with an area under the curve of 0.84 (95% CI 0.77-0.90). In conclusion the metabolic imbalance in COVID-19 patients might affect disease progression. This work shows an association of phenylalanine with critical outcomes in COVID-19 patients, highlighting phenylalanine as a potential metabolic biomarker of disease severity.
Collapse
Affiliation(s)
- Laura E. Martínez-Gómez
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Isabel Ibarra-González
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Cynthia Fernández-Lainez
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México, Mexico
| | - Teresa Tusie
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Instituto de Investigaciones Biomédicas UNAM, Ciudad de México, Mexico
| | - Hortensia Moreno-Macías
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Instituto de Investigaciones Biomédicas UNAM, Ciudad de México, Mexico
- Departamento de Economía. División de Ciencias Sociales y Humanidades, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México, Mexico
| | - Carlos Martinez-Armenta
- Posgrado en Biología Experimental, Dirección de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México, Mexico
| | - Guadalupe Elizabeth Jimenez-Gutierrez
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Paola Vázquez-Cárdenas
- Centro de Innovación Médica Aplicada, Subdirección de Epidemiología e Infectología, Hospital General Dr. Manuel Gea González, Secretaría de Salud, Ciudad de México, Mexico
| | - Patricia Vidal-Vázquez
- Centro de Innovación Médica Aplicada, Subdirección de Epidemiología e Infectología, Hospital General Dr. Manuel Gea González, Secretaría de Salud, Ciudad de México, Mexico
| | - Juan P. Ramírez-Hinojosa
- Centro de Innovación Médica Aplicada, Subdirección de Epidemiología e Infectología, Hospital General Dr. Manuel Gea González, Secretaría de Salud, Ciudad de México, Mexico
| | - Ana P. Rodríguez-Zulueta
- Centro de Innovación Médica Aplicada, Subdirección de Epidemiología e Infectología, Hospital General Dr. Manuel Gea González, Secretaría de Salud, Ciudad de México, Mexico
| | - Gilberto Vargas-Alarcón
- Departamentos de Biología Molecular, Inmunología, Endocrinologia y Unidad de Cuidados Intensivos, Instituto Nacional de Cardiología Ignacio Chavez, Secretaría de Salud, Ciudad de México, Mexico
| | - Gustavo Rojas-Velasco
- Departamentos de Biología Molecular, Inmunología, Endocrinologia y Unidad de Cuidados Intensivos, Instituto Nacional de Cardiología Ignacio Chavez, Secretaría de Salud, Ciudad de México, Mexico
| | - Fausto Sánchez-Muñoz
- Departamentos de Biología Molecular, Inmunología, Endocrinologia y Unidad de Cuidados Intensivos, Instituto Nacional de Cardiología Ignacio Chavez, Secretaría de Salud, Ciudad de México, Mexico
| | - Rosalinda Posadas-Sanchez
- Departamentos de Biología Molecular, Inmunología, Endocrinologia y Unidad de Cuidados Intensivos, Instituto Nacional de Cardiología Ignacio Chavez, Secretaría de Salud, Ciudad de México, Mexico
| | - Felipe de J. Martínez-Ruiz
- Nuevo Hospital General Delegación Regional Sur de la Ciudad de México, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado (ISSSTE), Ciudad de México, Mexico
| | - Dulce M. Zayago-Angeles
- Nuevo Hospital General Delegación Regional Sur de la Ciudad de México, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado (ISSSTE), Ciudad de México, Mexico
| | - Mariana L. Moreno
- Nuevo Hospital General Delegación Regional Sur de la Ciudad de México, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado (ISSSTE), Ciudad de México, Mexico
| | - Edith Barajas-Galicia
- Hospital Central Norte Petróleos Mexicanos (PEMEX), Estado de México, Mexico City, Mexico
| | - Gerardo Lopez-Cisneros
- Hospital Central Norte Petróleos Mexicanos (PEMEX), Estado de México, Mexico City, Mexico
| | | | - Silvestre Ortega-Peña
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Brígida Herrera-López
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Jessel Olea-Torres
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Manuel Juárez-Arias
- Unidad de Investigación y Desarrollo en Alimentos, Tecnológico Nacional de México/Instituto Tecnológico (IT) Veracruz, Veracruz, Mexico
| | - Maritza Rosas-Vásquez
- Unidad de Investigación y Desarrollo en Alimentos, Tecnológico Nacional de México/Instituto Tecnológico (IT) Veracruz, Veracruz, Mexico
| | - Sara Aileen Cabrera-Nieto
- Posgrado en Ciencias Médicas, Facultad de Ciencias de la Salud, Universidad Anáhuac, Ciudad de México, Mexico
| | - Jonathan J. Magaña
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - María del Carmen Camacho-Rea
- Departamento de Nutrición Animal, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Secretaría de Salud, Ciudad de México, Mexico
| | - Carlos Suarez-Ahedo
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Irma Coronado-Zarco
- Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| | | | - Gabriela Angélica Martínez-Nava
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Carlos Pineda
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | - Marcela Vela-Amieva
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México, Mexico
| | - Alberto López-Reyes
- Laboratorio de Gerociencias, Laboratorio Facilitador, Laboratorio de Medicina Genómica, Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico
| | | |
Collapse
|
46
|
Saito K, Ishikawa R, Kitamura I, Ogawa K, Arakawa N, Sun Y, Imai K, Takuya M, Saito Y, Hasegawa C. Characterization of serotonin as a candidate biomarker of severity and prognosis of COVID-19 using LC/MS analysis. J Pharmacol Sci 2022; 150:49-55. [PMID: 36055751 PMCID: PMC9339153 DOI: 10.1016/j.jphs.2022.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/21/2022] [Accepted: 06/29/2022] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has been associated with high mortality worldwide. Owing to its complicated pathophysiology, diagnostic and prognostic biomarkers for effective patient management remain scarce. We analyzed kynurenine, tryptophan, and serotonin levels in the serum of patients with COVID-19 via liquid chromatography/mass spectrometry analysis. Serum serotonin levels were decreased in patients with more severe COVID-19, along with increased kynurenine and decreased tryptophan concentrations. Patients with moderate disease who subsequently worsened showed significantly lower serotonin concentrations compared with those who did not experience severe disease. Serum serotonin levels may represent a valuable biomarker for COVID-19 severity and prognosis.
Collapse
|
47
|
Meacci E, Pierucci F, Garcia-Gil M. Skeletal Muscle and COVID-19: The Potential Involvement of Bioactive Sphingolipids. Biomedicines 2022; 10:biomedicines10051068. [PMID: 35625805 PMCID: PMC9138286 DOI: 10.3390/biomedicines10051068] [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: 04/04/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 virus infection is the cause of the coronavirus disease 2019 (COVID-19), which is still spreading over the world. The manifestation of this disease can range from mild to severe and can be limited in time (weeks) or persist for months in about 30–50% of patients. COVID-19 is considered a multiple organ dysfunction syndrome and the musculoskeletal system manifestations are beginning to be considered of absolute importance in both COVID-19 patients and in patients recovering from the SARS-CoV-2 infection. Musculoskeletal manifestations of COVID-19 and other coronavirus infections include loss of muscle mass, muscle weakness, fatigue or myalgia, and muscle injury. The molecular mechanisms by which SARS-CoV-2 can cause damage to skeletal muscle (SkM) cells are not yet well understood. Sphingolipids (SLs) represent an important class of eukaryotic lipids with structural functions as well as bioactive molecules able to modulate crucial processes, including inflammation and viral infection. In the last two decades, several reports have highlighted the role of SLs in modulating SkM cell differentiation, regeneration, aging, response to insulin, and contraction. This review summarizes the consequences of SARS-CoV-2 infection on SkM and the potential involvement of SLs in the tissue responses to virus infection. In particular, we highlight the role of sphingosine 1-phosphate signaling in order to aid the prediction of novel targets for preventing and/or treating acute and long-term musculoskeletal manifestations of virus infection in COVID-19.
Collapse
Affiliation(s)
- Elisabetta Meacci
- Unit of Biochemical Sciences and Molecular Biology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale GB Morgagni 50, 50121 Florence, Italy;
- Interuniversity Institute of Myology, University of Florence, 50121 Florence, Italy
- Correspondence: ; Tel.: +39-055-2751231
| | - Federica Pierucci
- Unit of Biochemical Sciences and Molecular Biology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale GB Morgagni 50, 50121 Florence, Italy;
| | - Mercedes Garcia-Gil
- Unit of Physiology, Department of Biology, University of Pisa, Via S. Zeno 31, 56127 Pisa, Italy;
- Interdepartmental Research Center “Nutraceuticals and Food for Health”, University of Pisa, 56127 Pisa, Italy
| |
Collapse
|
48
|
Bonnet U, Juckel G. COVID-19 Outcomes: Does the Use of Psychotropic Drugs Make a Difference? Accumulating Evidence of a Beneficial Effect of Antidepressants-A Scoping Review. J Clin Psychopharmacol 2022; 42:284-292. [PMID: 35420565 PMCID: PMC9042214 DOI: 10.1097/jcp.0000000000001543] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/22/2022] [Indexed: 12/19/2022]
Abstract
PURPOSE/BACKGROUND Studies for repurposed drugs in severe acute respiratory syndrome coronavirus type 2-infected and coronavirus disease 2019 (COVID-19) patients are ongoing. According to preclinical research, antidepressants (ADs) might be useful in the treatment of COVID-19. METHODS/PROCEDURES We conducted a scoping review including clinical studies on AD effects on SARS-CoV-2 infection and COVID-19. FINDING/RESULTS As of January 2, 2022, we found 14 clinical studies, which could be included into this review. Among them, there were 2 randomized, placebo-controlled studies and 2 prospective parallel-group studies about the efficacy/effectiveness and tolerability of fluvoxamine. The remaining studies were mainly retrospective studies considering COVID-19 hospital populations predominantly exposed to fluoxetine (N = 3), other selective serotonin reuptake inhibitors (SSRI), selective norepinephrine reuptake inhibitors (SNRI), and trazodone. The vast majority were hospital studies and assessed COVID-19 severity (morbidity) and mortality as primary endpoints. The only outpatient study (fluvoxamine) investigated the COVID-19-related hospitalization rate, and 1 psychiatric hospital study (SSRI, SNRI, trazodone) focused on the SARS-CoV-2 infection rate. IMPLICATIONS/CONCLUSIONS At present, the best evidence of an "anti-COVID-19" potential of ADs exists for fluvoxamine and, to a lesser extent, for fluoxetine. Preliminary evidence had found that patients exposed to SSRI or SNRI substance classes might have a reduced mortality risk and that trazodone might reduce SARS-CoV-2 infection rates. Three studies found no relevant influence of ADs on COVID-19 morbidity and mortality, and 1 study described increased mortality. The latter study, however, did not differentiate between psychotropic medication and ADs. Tricyclics and monoamine oxidase inhibitors are still absolute "dark zones" in COVID-19 research. Further controlled studies testing the effectiveness/efficacy and tolerability/safety (as well as the treatment timing and duration) of different AD substance classes in COVID-19 and post/long-COVID patients of various populations are warranted.
Collapse
Affiliation(s)
- Udo Bonnet
- From the Department of Psychiatry, Psychotherapy and Psychosomatic Medicine, Evangelisches Krankenhaus Castrop-Rauxel
- Department of Psychiatry and Psychotherapy, LVR-Hospital Essen, Faculty of Medicine, University of Duisburg-Essen, Essen
| | - Georg Juckel
- Department of Psychiatry, Ruhr University Bochum, LWL University Hospital, Bochum, Germany
| |
Collapse
|
49
|
Metabolomics and integrated network analysis reveal roles of endocannabinoids and large neutral amino acid balance in the ayahuasca experience. Biomed Pharmacother 2022; 149:112845. [PMID: 35339828 DOI: 10.1016/j.biopha.2022.112845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 11/22/2022] Open
Abstract
There has been a renewed interest in the potential use of psychedelics for the treatment of psychiatric conditions. Nevertheless, little is known about the mechanism of action and molecular pathways influenced by ayahuasca use in humans. Therefore, for the first time, our study aims to investigate the human metabolomics signature after consumption of a psychedelic, ayahuasca, and its connection with both the psychedelic-induced subjective effects and the plasma concentrations of ayahuasca alkaloids. Plasma samples of 23 individuals were collected both before and after ayahuasca consumption. Samples were analysed through targeted metabolomics and further integrated with subjective ratings of the ayahuasca experience (i.e., using the 5-Dimension Altered States of Consciousness Rating Scale [ASC]), and plasma ayahuasca-alkaloids using integrated network analysis. Metabolic pathways enrichment analysis using diffusion algorithms for specific KEGG modules was performed on the metabolic output. Compared to baseline, the consumption of ayahuasca increased N-acyl-ethanolamine endocannabinoids, decreased 2-acyl-glycerol endocannabinoids, and altered several large-neutral amino acids (LNAAs). Integrated network results indicated that most of the LNAAs were inversely associated with 9 out of the 11 subscales of the ASC, except for tryptophan which was positively associated. Several endocannabinoids and hexosylceramides were directly associated with the ayahuasca alkaloids. Enrichment analysis confirmed dysregulation in several pathways involved in neurotransmission such as serotonin and dopamine synthesis. In conclusion, a crosstalk between the circulating LNAAs and the subjective effects is suggested, which is independent of the alkaloid concentrations and provides insights into the specific metabolic fingerprint and mechanism of action underlying ayahuasca experiences.
Collapse
|
50
|
Hoertel N, Sánchez-Rico M, Gulbins E, Kornhuber J, Vernet R, Beeker N, Neuraz A, Blanco C, Olfson M, Airagnes G, Lemogne C, Alvarado JM, Arnaout M, Cougoule C, Meneton P, Limosin F. Association between benzodiazepine receptor agonist use and mortality in patients hospitalised for COVID-19: a multicentre observational study. Epidemiol Psychiatr Sci 2022; 31:e18. [PMID: 35352674 PMCID: PMC8967698 DOI: 10.1017/s2045796021000743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 11/24/2021] [Accepted: 12/04/2021] [Indexed: 11/18/2022] Open
Abstract
AIMS To examine the association between benzodiazepine receptor agonist (BZRA) use and mortality in patients hospitalised for coronavirus disease 2019 (COVID-19). METHODS A multicentre observational study was performed at Greater Paris University hospitals. The sample involved 14 381 patients hospitalised for COVID-19. A total of 686 (4.8%) inpatients received a BZRA at hospital admission at a mean daily diazepam-equivalent dose of 19.7 mg (standard deviation (s.d.) = 25.4). The study baseline was the date of admission, and the primary endpoint was death. We compared this endpoint between patients who received BZRAs and those who did not in time-to-event analyses adjusted for sociodemographic characteristics, medical comorbidities and other medications. The primary analysis was a Cox regression model with inverse probability weighting (IPW). RESULTS Over a mean follow-up of 14.5 days (s.d. = 18.1), the primary endpoint occurred in 186 patients (27.1%) who received BZRAs and in 1134 patients (8.3%) who did not. There was a significant association between BZRA use and increased mortality both in the crude analysis (hazard ratio (HR) = 3.20; 95% confidence interval (CI) = 2.74-3.74; p < 0.01) and in the IPW analysis (HR = 1.61; 95% CI = 1.31-1.98, p < 0.01), with a significant dose-dependent relationship (HR = 1.55; 95% CI = 1.08-2.22; p = 0.02). This association remained significant in sensitivity analyses. Exploratory analyses indicate that most BZRAs may be associated with an increased mortality among patients hospitalised for COVID-19, except for diazepam, which may be associated with a reduced mortality compared with any other BZRA treatment. CONCLUSIONS BZRA use may be associated with an increased mortality among patients hospitalised for COVID-19, suggesting the potential benefit of decreasing dose or tapering off gradually these medications when possible.
Collapse
Affiliation(s)
- N. Hoertel
- Département de Psychiatrie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Issy-les-Moulineaux, France
- Université de Paris, Paris, France
- INSERM, Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR_S1266, Paris, France
| | - M. Sánchez-Rico
- Département de Psychiatrie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Issy-les-Moulineaux, France
- Department of Psychobiology & Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Campus de Somosaguas, Pozuelo de Alarcon, Spain
| | - E. Gulbins
- Institute for Molecular Biology, University Medicine Essen, University of Duisburg-Essen, Essen, Germany
| | - J. Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - R. Vernet
- Medical Informatics, Biostatistics and Public Health Department, AP-HP, Centre-Université de Paris, Hôpital Européen Georges Pompidou, F-75015Paris, France
| | - N. Beeker
- Assistance Publique-Hôpitaux de Paris (AP-HP), Unité de Recherche Clinique, Hôpital Cochin, Paris, France
| | - A. Neuraz
- INSERM, UMR_S 1138, Cordeliers Research Center, Université de Paris, Paris, France
- Department of Medical Informatics, AP-HP, Centre-Université de Paris, Necker-Enfants Malades Hospital, Paris, France
| | - C. Blanco
- Division of Epidemiology, Services and Prevention Research, National Institute on Drug Abuse, 6001 Executive Boulevard, Bethesda, MD20852, USA
| | - M. Olfson
- Department of Psychiatry, Columbia University/New York State Psychiatric Institute, 1051 Riverside Drive, Unit 69, New York, NY10032, USA
| | - G. Airagnes
- Département de Psychiatrie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Issy-les-Moulineaux, France
- Université de Paris, Paris, France
- INSERM, Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR_S1266, Paris, France
- INSERM, UMS 011, Population-based Epidemiologic Cohorts, Villejuif, France
| | - C. Lemogne
- Université de Paris, Paris, France
- INSERM, Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR_S1266, Paris, France
- AP-HP, Hôpital Hôtel-Dieu, DMU Psychiatrie et Addictologie, Service de Psychiatrie de l'adulte, Paris, France
| | - J. M. Alvarado
- Department of Psychobiology & Behavioural Sciences Methods, Faculty of Psychology, Universidad Complutense de Madrid, Campus de Somosaguas, Pozuelo de Alarcon, Spain
| | - M. Arnaout
- Anesthesia and Intensive Care Department, Hôpitaux Universitaires Paris Île-de-France Ouest, Boulogne-Billancourt, France
| | - C. Cougoule
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, Toulouse, France
| | - P. Meneton
- INSERM U1142 LIMICS, UMRS 1142, Sorbonne Universities, UPMC University of Paris 06, University of Paris 13, Paris, France
| | - F. Limosin
- Département de Psychiatrie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Corentin-Celton, DMU Psychiatrie et Addictologie, Issy-les-Moulineaux, France
- Université de Paris, Paris, France
- INSERM, Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR_S1266, Paris, France
| |
Collapse
|