1
|
Ryu MH, Hur SA, Afshar T, Kolmert J, Zurita J, Wheelock CE, Carlsten C. Impact of Short-Term Diesel Exhaust Exposure on Prothrombotic Markers in Chronic Obstructive Pulmonary Disease: A Randomized, Double-Blind, Crossover Study. Ann Am Thorac Soc 2024; 21:1715-1722. [PMID: 39167788 DOI: 10.1513/annalsats.202311-955oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 08/21/2024] [Indexed: 08/23/2024] Open
Abstract
Rationale: Growing evidence suggests that air pollution exposure is a major risk factor in chronic obstructive pulmonary disease (COPD) that is associated with an increased prothrombotic state and adverse cardiovascular outcomes. However, much of this work is based on observational data or human exposure studies involving younger participants. The biological causality and mechanism of air pollution-induced prothrombotic response in patients with COPD remain to be explored. Objectives: The main aim of this work was to investigate the impact of short-term diesel exhaust (DE) exposure on circulating prothrombotic markers-fibrinogen and plasminogen activator inhibitor-1 (PAI-1)-and urinary eicosanoids in patients with COPD. Methods: Twenty-nine research participants were recruited in this randomized, double-blind, crossover, controlled human exposure study to DE. Participants included former smokers with and without mild or moderate COPD (ex-smokers [ES] and COPD group) and healthy never-smokers without COPD (nonsmoker [NS] group). Each participant was exposed to DE (300 μg/m3 of particulate matter with an aerodynamic diameter ≤2.5 μm) and filtered air for 2 hours on different occasions, in randomized order, separated by a 4-week washout. Blood and urine samples were collected before and 24 hours after each exposure. Plasma fibrinogen and serum PAI-1 concentrations were quantified using enzyme-linked immunosorbent assays. Urinary eicosanoid concentrations were quantified using ultraperformance liquid chromatography coupled to tandem mass spectrometry. Linear mixed-effects models were used for statistical comparisons. Results: Participants with COPD showed an increase in plasma fibrinogen (effect estimate, 1.27 [1.06-1.53]; P = 0.01) after DE relative to filtered air, but no significant DE-associated change in serum PAI-1 (0.95 [0.87-1.04]; P = 0.26). In never-smokers and ex-smokers without COPD, fibrinogen (NS group, 1.10 [0.99-1.23]; P = 0.08; ES group, 0.86 [0.68-1.09]; P = 0.08] and PAI-1 (NS group, 1.12 [0.96-1.32]; P = 0.15; ES group, 0.90 [0.79-1.03]; P = 0.13) were not changed after DE exposure. Participants with COPD showed a DE-attributable increase in urinary thromboxane B2 (TXB2) metabolite concentrations as follows: 11-dehydro-TXB2 (1.45 [1.02-2.08]; P = 0.04) and 2,3-dinor-TXB2 (1.45 [1.05-2.00]; P = 0.03). Conclusions: Participants with COPD had increased plasma fibrinogen and urinary TXB2 metabolites after short-term DE exposure, suggesting they may be more susceptible to a pollution-attributable prothrombotic response than healthy control subjects or ex-smokers without COPD. Clinical trial registered with www.clinicaltrials.gov (NCT02236039).
Collapse
Affiliation(s)
- Min Hyung Ryu
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Seo Am Hur
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tina Afshar
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Johan Kolmert
- Unit of Integrative Metabolomics, Karolinska Institutet, Institute of Environmental Medicine, Stockholm, Sweden; and
| | - Javier Zurita
- Unit of Integrative Metabolomics, Karolinska Institutet, Institute of Environmental Medicine, Stockholm, Sweden; and
| | - Craig E Wheelock
- Unit of Integrative Metabolomics, Karolinska Institutet, Institute of Environmental Medicine, Stockholm, Sweden; and
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Christopher Carlsten
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
2
|
Nogueira MS, Sanchez SC, Milne CE, Amin W, Thomas SJ, Milne GL. Resolvins D5 and D1 undergo phase II metabolism by uridine 5'-diphospho-glucuronosyltransferases. Prostaglandins Other Lipid Mediat 2024; 174:106870. [PMID: 39038698 DOI: 10.1016/j.prostaglandins.2024.106870] [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: 05/01/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/24/2024]
Abstract
Specialized pro-resolving mediators (SPMs) are oxidized lipid mediators that have been shown to resolve inflammation in cellular and animal models as well as humans. SPMs and their biological precursors are even commercially available as dietary supplements. It has been understood for more than forty years that pro-inflammatory oxidized lipid mediators, including prostaglandins and leukotrienes, are rapidly inactivated via metabolism. Studies on the metabolism of SPMs are, however, limited. Herein, we report that resolvin D5 (RvD5) and resolvin D1 (RvD1), well-studied SPMs, are readily metabolized by human liver microsomes (HLM) to glucuronide conjugated metabolites. We further show that this transformation is catalyzed by specific uridine 5'-diphospho-glucuronosyltransferase (UGT) isoforms. Additionally, we demonstrate that RvD5 and RvD1 metabolism by HLM is influenced by non-steroidal anti-inflammatory drugs (NSAIDs), which can act as UGT inhibitors through cyclooxygenase-independent mechanisms. The results from these studies highlight the importance of considering metabolism, as well as factors that influence metabolic enzymes, when seeking to quantify SPMs in vivo.
Collapse
Affiliation(s)
- Marina S Nogueira
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| | - Stephanie C Sanchez
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| | | | - Warda Amin
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| | - Sarah J Thomas
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| | - Ginger L Milne
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA.
| |
Collapse
|
3
|
Chen L, Brustad N, Luo Y, Wang T, Ali M, Ebrahimi P, Schoos AMM, Vahman N, Lovrić M, Rasmussen MA, Kolmert J, Wheelock CE, Lasky-Su JA, Stokholm J, Bønnelykke K, Chawes B. Prenatal Fish Oil Supplementation, Maternal COX1 Genotype, and Childhood Atopic Dermatitis: A Secondary Analysis of a Randomized Clinical Trial. JAMA Dermatol 2024; 160:1082-1090. [PMID: 39196551 PMCID: PMC11359109 DOI: 10.1001/jamadermatol.2024.2849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 06/25/2024] [Indexed: 08/29/2024]
Abstract
Importance Eicosanoids have a pathophysiological role in atopic dermatitis (AD), but it is unknown whether this is affected by prenatal ω-3 long-chain polyunsaturated fatty acid (n-3 LCPUFA; ie, fish oil) supplementation and genetic variations in the cyclooxygenase-1 (COX1) pathway. Objective To explore the association of n-3 LCPUFA supplementation during pregnancy with risk of childhood AD overall and by maternal COX1 genotype. Design, Setting, and Participants This prespecified secondary analysis of a randomized clinical trial included mother-child pairs from the Danish Copenhagen Prospective Studies on Asthma in Childhood 2010 birth cohort, with prospective follow-up until children were aged 10 years. In the trial, maternal and child COX1 genotypes were determined, and urinary eicosanoids were quantified when the child was 1 year of age. The present study was conducted from January 2019 to December 2021, and data were analyzed from January to September 2023. Intervention A total of 736 pregnant women at 24 weeks' gestation were randomized 1:1 to 2.4 g of n-3 LCPUFA (fish oil) or placebo (olive oil) per day until 1 week post partum. Main Outcomes and Measures Risk of childhood AD until age 10 years overall and by maternal COX1 genotype. Results At age 10 years, 635 children (91%; 363 [57%] female) completed the clinical follow-up, and these mother-child pairs were included in this study; 321 (51%) were in the intervention group and 314 (49%) in the control group. Pregnancy n-3 LCPUFA supplementation was associated with lower urinary thromboxane A2 metabolites at age 1 year (β, -0.46; 95% CI, -0.80 to -0.13; P = .006), which was also associated with COX1 rs1330344 genotype (β per C allele, 0.47; 95% CI, 0.20-0.73; P = .001). Although neither n-3 LCPUFA supplementation (hazard ratio [HR], 1.00; 95% CI, 0.76-1.33; P = .97) nor maternal COX1 genotype (HR, 0.94; 95% CI, 0.74-1.19; P = .60) was associated with risk of childhood AD until age 10 years, there was evidence of an interaction between these variables (P < .001 for interaction). Among mothers with the TT genotype, risk of AD was reduced in the n-3 LCPUFA group compared with the placebo group (390 mother-child pairs [61%]; HR, 0.70; 95% CI, 0.50-0.98; P = .04); there was no association for mothers with the CT genotype (209 [33%]; HR, 1.29; 95% CI, 0.79-2.10; P = .31), and risk was increased among offspring of mothers with the CC genotype (37 [6%]; HR, 5.77; 95% CI, 1.63-20.47; P = .007). There was a significant interaction between n-3 LCPUFA supplementation and child COX1 genotype and development of AD (P = .002 for interaction). Conclusions and Relevance In this secondary analysis of a randomized clinical trial, the association of prenatal n-3 LCPUFA supplementation with risk of childhood AD varied by maternal COX1 genotype. The findings could be used to inform a personalized prevention strategy of providing supplementation only to pregnant individuals with the TT genotype. Trial Registration ClinicalTrials.gov: NCT00798226.
Collapse
Affiliation(s)
- Liang Chen
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Nicklas Brustad
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Yang Luo
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Tingting Wang
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Mina Ali
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Parvaneh Ebrahimi
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Food Microbiology, Gut Health, and Fermentation, Department of Food Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Ann-Marie M. Schoos
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
| | - Nilo Vahman
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Mario Lovrić
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Centre for Applied Bioanthropology, Institute for Anthropological Research, Zagreb, Croatia
- Faculty of Electrical Engineering, Computer Science and Information Technology Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Morten A. Rasmussen
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Food Microbiology, Gut Health, and Fermentation, Department of Food Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Johan Kolmert
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Craig E. Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Jessica A. Lasky-Su
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Food Microbiology, Gut Health, and Fermentation, Department of Food Science, University of Copenhagen, Frederiksberg C, Denmark
- Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bo Chawes
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Chen L, Brustad N, Kim M, Luo Y, Wang T, Ali M, Prince N, Chen Y, Chu S, Begum S, Mendez K, Kelly RS, Schoos AM, Rasmussen MA, Zurita J, Kolmert J, Stokholm J, Litonjua A, Weiss ST, Bønnelykke K, Wheelock CE, Lasky-Su J, Chawes B. Urinary eicosanoid levels in early life and risk of atopic disease in childhood. J Allergy Clin Immunol 2024; 154:670-678. [PMID: 38825025 DOI: 10.1016/j.jaci.2024.05.022] [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: 11/17/2023] [Revised: 05/23/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Eicosanoids are lipid mediators including thromboxanes (TXs), prostaglandins (PGs), and leukotrienes with a pathophysiological role in established atopic disease. However, their role in the inception of disease is unclear. This study aimed to investigate the association between urinary eicosanoids in early life and development of atopic disease. METHODS This study quantified the levels of 21 eicosanoids in urine from children from the COPSAC2010 (Copenhagen Prospective Studies on Asthma in Childhood 2010) (age 1 year, n = 450) and VDAART (Vitamin D Antenatal Asthma Reduction Trial) (age 3 years, n = 575) mother-child cohorts and analyzed the associations with development of wheeze/asthma, atopic dermatitis, and biomarkers of type-2 inflammation, applying false discovery rate of 5% (FDR5%) multiple testing correction. RESULTS In both cohorts, analyses adjusted for environmental determinants showed that higher TXA2 eicosanoids in early life were associated with increased risk of developing atopic dermatitis (P < FDR5%) and type-2 inflammation (P < .05). In VDAART, lower PGE2 and PGI2 eicosanoids and higher isoprostanes were also associated with increased risk of atopic dermatitis (P < FDR5%). For wheeze/asthma, analyses in COPSAC2010 showed that lower isoprostanes and PGF2 eicosanoids and higher PGD2 eicosanoids at age 1 year associated with an increased risk at age 1-10 years (P < .05), whereas analyses in VDAART showed that lower PGE2 and higher TXA2 eicosanoids at age 3 years associated with an increased risk at 6 years (P < FDR5%). CONCLUSIONS This study suggests that early life perturbations in the eicosanoid metabolism are present before the onset of atopic disease in childhood, which provides pathophysiological insight in the inception of atopic diseases.
Collapse
Affiliation(s)
- Liang Chen
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Nicklas Brustad
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Min Kim
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Yang Luo
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Tingting Wang
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Mina Ali
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Nicole Prince
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Yulu Chen
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Su Chu
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Sofina Begum
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Kevin Mendez
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Ann-Marie Schoos
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
| | - Morten A Rasmussen
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Slagelse, Denmark
| | - Javier Zurita
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Kolmert
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark; Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Slagelse, Denmark
| | - Augusto Litonjua
- Division of Pediatric Pulmonary Medicine, Golisano Children's Hospital, University of Rochester Medical Center, Rochester, NY
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Craig E Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Bo Chawes
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
5
|
Djukanović R, Brinkman P, Kolmert J, Gomez C, Schofield J, Brandsma J, Shapanis A, Skipp PJS, Postle A, Wheelock C, Dahlen SE, Sterk PJ, Brown T, Jackson DJ, Mansur A, Pavord I, Patel M, Brightling C, Siddiqui S, Bradding P, Sabroe I, Saralaya D, Chishimba L, Porter J, Robinson D, Fowler S, Howarth PH, Little L, Oliver T, Hill K, Stanton L, Allen A, Ellis D, Griffiths G, Harrison T, Akenroye A, Lasky-Su J, Heaney L, Chaudhuri R, Kurukulaaratchy R. Biomarker Predictors of Clinical Efficacy of the Anti-IgE Biologic Omalizumab in Severe Asthma in Adults: Results of the SoMOSA Study. Am J Respir Crit Care Med 2024; 210:288-297. [PMID: 38635834 PMCID: PMC11348961 DOI: 10.1164/rccm.202310-1730oc] [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: 10/03/2023] [Accepted: 04/18/2024] [Indexed: 04/20/2024] Open
Abstract
Background: The anti-IgE monoclonal antibody omalizumab is widely used for severe asthma. This study aimed to identify biomarkers that predict clinical improvement during 1 year of omalizumab treatment. Methods: One-year open-label Study of Mechanisms of action of Omalizumab in Severe Asthma (SoMOSA) involving 216 patients with severe (Global Initiative for Asthma step 4/5) uncontrolled atopic asthma (at least two severe exacerbations in the previous year) taking high-dose inhaled corticosteroids and long-acting β-agonists with or without maintenance oral corticosteroids. It had two phases: 0-16 weeks, to assess early clinical improvement by Global Evaluation of Therapeutic Effectiveness (GETE); and 16-52 weeks, to assess late responses based on ⩾50% reduction in exacerbations or mOCS dose. All participants provided samples (exhaled breath, blood, sputum, urine) before and after 16 weeks of omalizumab treatment. Measurements and Main Results: A total of 191 patients completed phase 1; 63% had early improvement. Of 173 who completed phase 2, 69% had reduced exacerbations by ⩾50% and 57% (37 of 65) taking mOCSs had reduced their dose by ⩾50%. The primary outcomes 2,3-dinor-11-β-PGF2α, GETE score, and standard clinical biomarkers (blood and sputum eosinophils, exhaled nitric oxide, serum IgE) did not predict either clinical response. Five volatile organic compounds and five plasma lipid biomarkers strongly predicted the ⩾50% reduction in exacerbations (receiver operating characteristic areas under the curve of 0.780 and 0.922, respectively) and early responses (areas under the curve of 0.835 and 0.949, respectively). In an independent cohort, gas chromatography/mass spectrometry biomarkers differentiated between severe and mild asthma. Conclusions: This is the first discovery of omics biomarkers that predict improvement in asthma with biologic agent treatment. Prospective validation and development for clinical use is justified.
Collapse
Affiliation(s)
- Ratko Djukanović
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton and National Institute for Health and Care Research Southampton Biomedical Research Centre, Southampton, United Kingdom
| | - Paul Brinkman
- Department of Respiratory Medicine, Amsterdam University Medical Center, University of Amsterdam, the Netherlands
| | - Johan Kolmert
- Institute of Environmental Medicine, Karolinska Institutet, and the Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Cristina Gomez
- Institute of Environmental Medicine, Karolinska Institutet, and the Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - James Schofield
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton and National Institute for Health and Care Research Southampton Biomedical Research Centre, Southampton, United Kingdom
- Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Joost Brandsma
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton and National Institute for Health and Care Research Southampton Biomedical Research Centre, Southampton, United Kingdom
| | - Andy Shapanis
- Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Paul J. S. Skipp
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton and National Institute for Health and Care Research Southampton Biomedical Research Centre, Southampton, United Kingdom
- Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Anthony Postle
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton and National Institute for Health and Care Research Southampton Biomedical Research Centre, Southampton, United Kingdom
| | - Craig Wheelock
- Institute of Environmental Medicine, Karolinska Institutet, and the Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Sven-Erik Dahlen
- Institute of Environmental Medicine, Karolinska Institutet, and the Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Peter J. Sterk
- Department of Respiratory Medicine, Amsterdam University Medical Center, University of Amsterdam, the Netherlands
| | - Thomas Brown
- Portsmouth Hospitals University National Health Service Trust, Queen Alexandra Hospital, Portsmouth, United Kingdom
| | - David J. Jackson
- Guy’s Severe Asthma Centre, School of Immunology & Microbial Sciences, King’s College London, London, United Kingdom
| | - Adel Mansur
- University of Birmingham and Heartlands Hospital, University Hospitals Birmingham National Health Service Foundation Trust, Birmingham, United Kingdom
| | - Ian Pavord
- Oxford Respiratory National Institute for Health and Care Research Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Mitesh Patel
- Respiratory Medicine and R&D, University Hospitals Plymouth National Health Service Trust, Plymouth, United Kingdom
| | - Christopher Brightling
- Institute for Lung Health and Leicester National Institute for Health and Care Research Biomedical Research Centre, University of Leicester, Leicester, United Kingdom
| | - Salman Siddiqui
- Institute for Lung Health and Leicester National Institute for Health and Care Research Biomedical Research Centre, University of Leicester, Leicester, United Kingdom
| | - Peter Bradding
- Institute for Lung Health and Leicester National Institute for Health and Care Research Biomedical Research Centre, University of Leicester, Leicester, United Kingdom
| | - Ian Sabroe
- Clinical Research Facility, Sheffield Teaching Hospitals National Health Service Foundation Trust, Sheffield, United Kingdom
| | - Dinesh Saralaya
- Bradford Institute for Health Research and the National Patient Recruitment Centre, Bradford, United Kingdom
| | - Livingstone Chishimba
- Clinical Sciences, Liverpool University Hospitals National Health Service Foundation Trust, Liverpool, United Kingdom
| | - Joanna Porter
- University College London Respiratory and National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Douglas Robinson
- University College London Respiratory and National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Stephen Fowler
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
- Manchester Academic Health Science Centre and National Institute for Health and Care Research Manchester Biomedical Research Centre, Manchester University Hospitals National Health Service Foundation Trust, Manchester, United Kingdom
| | - Peter H. Howarth
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton and National Institute for Health and Care Research Southampton Biomedical Research Centre, Southampton, United Kingdom
| | - Louisa Little
- Southampton Clinical Trials Unit, University of Southampton, and University Hospital Southampton National Health Service Foundation Trust, Southampton, United Kingdom
| | - Thomas Oliver
- Southampton Clinical Trials Unit, University of Southampton, and University Hospital Southampton National Health Service Foundation Trust, Southampton, United Kingdom
| | - Kayleigh Hill
- Southampton Clinical Trials Unit, University of Southampton, and University Hospital Southampton National Health Service Foundation Trust, Southampton, United Kingdom
| | - Louise Stanton
- Southampton Clinical Trials Unit, University of Southampton, and University Hospital Southampton National Health Service Foundation Trust, Southampton, United Kingdom
| | - Alexander Allen
- Southampton Clinical Trials Unit, University of Southampton, and University Hospital Southampton National Health Service Foundation Trust, Southampton, United Kingdom
| | - Deborah Ellis
- Southampton Clinical Trials Unit, University of Southampton, and University Hospital Southampton National Health Service Foundation Trust, Southampton, United Kingdom
| | - Gareth Griffiths
- Southampton Clinical Trials Unit, University of Southampton, and University Hospital Southampton National Health Service Foundation Trust, Southampton, United Kingdom
| | - Tim Harrison
- Nottingham Respiratory National Institute for Health and Care Research Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom
| | - Ayobami Akenroye
- Division of Allergy and Clinical Immunology and
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Liam Heaney
- Wellcome-Wolfson Institute for Experimental Medicine, Belfast, Northern Ireland; and
| | - Rekha Chaudhuri
- Gartnavel General Hospital and School of Infection & Immunity, University of Glasgow, Glasgow, United Kingdom
| | - Ramesh Kurukulaaratchy
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton and National Institute for Health and Care Research Southampton Biomedical Research Centre, Southampton, United Kingdom
| |
Collapse
|
6
|
Fernandez Requena B, Gonzalez-Riano C, Barbas C. Addressing the untargeted lipidomics challenge in urine samples: Comparative study of extraction methods by UHPLC-ESI-QTOF-MS. Anal Chim Acta 2024; 1299:342433. [PMID: 38499427 DOI: 10.1016/j.aca.2024.342433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/20/2024]
Abstract
Urine analysis has remained a fundamental and widely used method in clinical diagnostics for over a century. With its minimal invasive nature and comprehensive range of analytes, urine has established itself as a clinical diagnostic tool for various disorders, including renal, urological, metabolic, and endocrine diseases. Furthermore, urine's unique attributes make it an attractive matrix for biomarker discovery, as well as in assessing the metabolic and physiological states of patients and healthy individuals alike. However, limitations in our knowledge of average values and sources of urinary lipids decrease the wider clinical application of urinary lipidomics. In this context, untargeted lipidomics analysis relies heavily on the extraction and analysis of lipids in biological samples. Nevertheless, this type of analysis presents challenges in lipid identification due to the diverse nature of lipids. Therefore, proper sample treatment before analysis is crucial to obtain robust and reproducible lipidomic profiles. To address this gap, we conducted a comparative study of a urine pool sample collected from twenty healthy volunteers using four different lipid extraction methods: one biphasic and three monophasic protocols. The extracted lipids were then analyzed using UHPLC-MS and MS/MS, and the semi-quantification of all the accurately annotated lipid species was performed for each extraction method.
Collapse
Affiliation(s)
- Belen Fernandez Requena
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Madrid, España
| | - Carolina Gonzalez-Riano
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Madrid, España
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Madrid, España.
| |
Collapse
|
7
|
Milne GL, Nogueira MS, Gao B, Sanchez SC, Amin W, Thomas S, Oger C, Galano JM, Murff HJ, Yang G, Durand T. Identification of novel F 2-isoprostane metabolites by specific UDP-glucuronosyltransferases. Redox Biol 2024; 70:103020. [PMID: 38211441 PMCID: PMC10821610 DOI: 10.1016/j.redox.2023.103020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/19/2023] [Accepted: 12/27/2023] [Indexed: 01/13/2024] Open
Abstract
UDP-glucuronosyltransferases (UGTs) catalyze the conjugation of glucuronic acid with endogenous and exogenous lipophilic small molecules to facilitate their inactivation and excretion from the body. This represents approximately 35 % of all phase II metabolic transformations. Fatty acids and their oxidized eicosanoid derivatives can be metabolized by UGTs. F2-isoprostanes (F2-IsoPs) are eicosanoids formed from the free radical oxidation of arachidonic acid. These molecules are potent vasoconstrictors and are widely used as biomarkers of endogenous oxidative damage. An increasing body of evidence demonstrates the efficacy of measuring the β-oxidation metabolites of F2-IsoPs rather than the unmetabolized F2-IsoPs to quantify oxidative damage in certain settings. Yet, the metabolism of F2-IsoPs is incompletely understood. This study sought to identify and characterize novel phase II metabolites of 15-F2t-IsoP and 5-epi-5-F2t-IsoP, two abundantly produced F2-IsoPs, in human liver microsomes (HLM). Utilizing liquid chromatography-mass spectrometry, we demonstrated that glucuronide conjugates are the major metabolites of these F2-IsoPs in HLM. Further, we showed that these molecules are metabolized by specific UGT isoforms. 15-F2t-IsoP is metabolized by UGT1A3, 1A9, and 2B7, while 5-epi-5-F2t-IsoP is metabolized by UGT1A7, 1A9, and 2B7. We identified, for the first time, the formation of intact glucuronide F2-IsoPs in human urine and showed that F2-IsoP glucuronidation is reduced in people supplemented with eicosapentaenoic and docosahexaenoic acids for 12 weeks. These studies demonstrate that endogenous F2-IsoP levels can be modified by factors other than redox mechanisms.
Collapse
Affiliation(s)
- Ginger L Milne
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232-6602, USA.
| | - Marina S Nogueira
- Division of Epidemiology, Department of Medicine, Vanderbilt Univiersity Medical Center, Nashville, TN, 37232, USA
| | - Benlian Gao
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232-6602, USA
| | - Stephanie C Sanchez
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232-6602, USA
| | - Warda Amin
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232-6602, USA
| | - Sarah Thomas
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232-6602, USA
| | - Camille Oger
- Institut des Biomolécules Max Mousseron, IBMM, UMR 5247, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM. Montpellier, France
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron, IBMM, UMR 5247, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM. Montpellier, France
| | - Harvey J Murff
- Division of Geriatric Medicine, Department of Medicine, Vanderbilt Univiersity Medical Center, Nashville, TN, 37232, USA
| | - Gong Yang
- Division of Epidemiology, Department of Medicine, Vanderbilt Univiersity Medical Center, Nashville, TN, 37232, USA
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron, IBMM, UMR 5247, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM. Montpellier, France
| |
Collapse
|
8
|
Petrucci G, Hatem D, Langley R, Cleary S, Gentry-Maharaj A, Pitocco D, Rizzi A, Ranalli P, Zaccardi F, Habib A, Rocca B. Effect of very long-term storage and multiple freeze and thaw cycles on 11-dehydro-thromboxane-B 2 and 8-iso-prostaglandin F 2α, levels in human urine samples by validated enzyme immunoassays. Sci Rep 2024; 14:5546. [PMID: 38448541 PMCID: PMC10917770 DOI: 10.1038/s41598-024-55720-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 02/27/2024] [Indexed: 03/08/2024] Open
Abstract
Biological samples are often frozen and stored for years and/or thawed multiple times, thus assessing their stability on long-term storage and repeated freeze-thaw cycles is crucial. The study aims were to assess:-the long-term stability of two major enzymatic and non-enzymatic metabolites of arachidonic acid, i.e. urinary 11-dehydro-thromboxane-(Tx) B2, 8-iso-prostaglandin (PG)F2α, and creatinine in frozen urine samples;-the effect of multiple freeze-thaw cycles. Seven-hundred and three urine samples measured in previously-published studies, stored at -40 °C, and measured for a second time for 11-dehydro-TxB2 (n = 677) and/or 8-iso-PGF2α (n = 114) and/or creatinine (n = 610) were stable over 10 years and the 2 measurements were highly correlated (all rho = 0.99, P < 0.0001). Urine samples underwent 10 sequential freeze-thaw cycles, with and without the antioxidant 4-hydroxy-2,2,6,6-tetramethylpiperidin-1-oxyl (10 mM); urinary 11-dehydro-TxB2 and creatinine were stable across all cycles (11-dehydro-TxB2: 100.4 ± 21%; creatinine: 101 ± 7% of baseline at cycle ten; n = 17), while 8-iso-PGF2α significantly increased by cycle 6 (151 ± 22% of baseline at cycle ten, n = 17, P < 0.05) together with hydrogen peroxide only in the absence of antioxidant. Arachidonic acid metabolites and creatinine appear stable in human urines stored at -40 °C over 10 years. Multiple freeze-thaw cycles increase urinary 8-iso-PGF2α in urine samples without antioxidants. These data are relevant for studies using urine samples stored over long-term and/or undergoing multiple freezing-thawing.
Collapse
Affiliation(s)
- Giovanna Petrucci
- Department of Bioethics and Safety, Section of Pharmacology, Catholic University School of Medicine, Rome, Italy.
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Duaa Hatem
- Department of Bioethics and Safety, Section of Pharmacology, Catholic University School of Medicine, Rome, Italy
| | - Ruth Langley
- Medical Research Council (MRC) Clinical Trials Units at University College London (UCL), London, UK
| | - Siobhan Cleary
- Medical Research Council (MRC) Clinical Trials Units at University College London (UCL), London, UK
| | | | - Dario Pitocco
- Diabetology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alessandro Rizzi
- Diabetology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Paola Ranalli
- Department of Hematology, S. Spirito Hospital, Pescara, Italy
| | - Francesco Zaccardi
- Leicester Real World Evidence Unit, Leicester Diabetes Centre, University of Leicester, Leicester, UK
| | - Aida Habib
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Bianca Rocca
- Department of Bioethics and Safety, Section of Pharmacology, Catholic University School of Medicine, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
9
|
Liu J, Idborg H, Korotkova M, Lend K, van Vollenhoven R, Lampa J, Rudin A, Nordström D, Gudbjornsson B, Gröndal G, Uhlig T, Hørslev-Petersen K, Lund Hetland M, Østergaard M, Nurmohamed M, Jakobsson PJ. Urinary prostanoids are elevated by anti-TNF and anti-IL6 receptor disease-modifying antirheumatic drugs but are not predictive of response to treatment in early rheumatoid arthritis. Arthritis Res Ther 2024; 26:61. [PMID: 38444034 PMCID: PMC10913231 DOI: 10.1186/s13075-024-03295-9] [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: 01/23/2024] [Accepted: 02/26/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Disease-modifying antirheumatic drugs (DMARDs) are widely used for treating rheumatoid arthritis (RA). However, there are no established biomarkers to predict a patient's response to these therapies. Prostanoids, encompassing prostaglandins, prostacyclins, and thromboxanes, are potent lipid mediators implicated in RA progression. Nevertheless, the influence of DMARDs on prostanoid biosynthesis in RA patients remains poorly understood. This study aims to assess the impact of various DMARDs on urinary prostanoids levels and to explore whether urinary prostanoid profiles correlate with disease activity or response to therapy. METHODS This study included 152 Swedish female patients with early RA, all rheumatoid factor (RF) positive, enrolled in the NORD-STAR trial (registration number: NCT01491815). Participants were randomized into four therapeutic regimes: methotrexate (MTX) combined with (i) prednisolone (arm ACT), (ii) TNF-α blocker certolizumab pegol (arm CZP), (iii) CTLA-4Ig abatacept (arm ABA), or (iv) IL-6R blocker tocilizumab (arm TCZ). Urine samples, collected before start of treatment and at 24 weeks post-treatment, were analyzed for tetranor-prostaglandin E metabolite (tPGEM), tetranor-prostaglandin D metabolite (tPGDM), 2,3-dinor thromboxane B2 (TXBM), 2,3-dinor-6-keto prostaglandin F1a (PGIM), leukotriene E4 (LTE4) and 12-hydroxyeicosatetraenoic acid (12-HETE) using liquid chromatography-mass spectrometry (LC-MS). Generalized estimating equation (GEE) models were used to analyze the change in urinary eicosanoids and their correlations to clinical outcomes. RESULTS Patients receiving MTX combined with CZP or TCZ exhibited significant elevations in urinary tPGEM and TXBM levels after 24 weeks of treatment. Other eicosanoids did not show significant alterations in response to any treatment. Baseline urinary eicosanoid levels did not correlate with baseline clinical disease activity index (CDAI) levels, nor with changes in CDAI from baseline to week 24. Their levels were also similar between patients who achieved CDAI remission and those with active disease at week 24. CONCLUSIONS Treatment with anti-TNF or anti-IL6R agents in early RA patients leads to an increased systemic production of proinflammatory and prothrombotic prostanoids. However, urinary eicosanoid levels do not appear to be predictive of the response to DMARDs therapy.
Collapse
Affiliation(s)
- Jianyang Liu
- Division of Rheumatology, Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Helena Idborg
- Division of Rheumatology, Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Marina Korotkova
- Division of Rheumatology, Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Kristina Lend
- Division of Rheumatology, Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Solna, Stockholm, Sweden
- Department of Rheumatology and Amsterdam Rheumatology Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Ronald van Vollenhoven
- Division of Rheumatology, Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Solna, Stockholm, Sweden
- Department of Rheumatology and Amsterdam Rheumatology Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Jon Lampa
- Division of Rheumatology, Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Anna Rudin
- Department of Rheumatology and Inflammation Research, University of Gothenburg Sahlgrenska Academy, Gothenburg, Sweden
| | - Dan Nordström
- Department of Medicine and Rheumatology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Bjorn Gudbjornsson
- Department of Rheumatology, Landspitali University Hospital, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Gerdur Gröndal
- Department of Rheumatology, Landspitali University Hospital, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Till Uhlig
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
- University of Oslo, Oslo, Norway
| | - Kim Hørslev-Petersen
- Danish Hospital for the Rheumatic Diseases, Sønderborg, Denmark
- University of Southern Denmark, Odense, Denmark
| | - Merete Lund Hetland
- Copenhagen Center for Arthritis Research (COPECARE), Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Østergaard
- Copenhagen Center for Arthritis Research (COPECARE), Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Nurmohamed
- Department of Rheumatology and Amsterdam Rheumatology Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Rheumatology and Immunology Center, Reade, The Netherlands
| | - Per-Johan Jakobsson
- Division of Rheumatology, Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Solna, Stockholm, Sweden.
| |
Collapse
|
10
|
Sieminska J, Kolmert J, Zurita J, Benkestock K, Revol-Cavalier J, Niklinski J, Reszec J, Dahlén SE, Ciborowski M, Wheelock CE. A single extraction 96-well method for LC-MS/MS quantification of urinary eicosanoids, steroids and drugs. Prostaglandins Other Lipid Mediat 2024; 170:106789. [PMID: 37879396 DOI: 10.1016/j.prostaglandins.2023.106789] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 10/27/2023]
Abstract
Urinary eicosanoid concentrations reflect inflammatory processes in multiple diseases and have been used as biomarkers of disease as well as suggested for patient stratification in precision medicine. However, implementation of urinary eicosanoid profiling in large-scale analyses is restricted due to sample preparation limits. Here we demonstrate a single solid-phase extraction of 300 µL urine in 96-well-format for prostaglandins, thromboxanes, isoprostanes, cysteinyl-leukotriene E4 and the linoleic acid-derived dihydroxy-octadecenoic acids (9,10- and 12,13-DiHOME). A simultaneous screening protocol was also developed for cortisol/cortisone and 7 exogenous steroids as well as 3 cyclooxygenase inhibitors. Satisfactory performance for quantification of eicosanoids with an appropriate internal standard was demonstrated for intra-plate analyses (CV = 8.5-15.1%) as well as for inter-plate (n = 35) from multiple studies (CV = 22.1-34.9%). Storage stability was evaluated at - 20 °C, and polar tetranors evidenced a 50% decrease after 5 months, while the remaining eicosanoids evidenced no significant degradation. All eicosanoids were stable over 3.5-years in urine stored at - 80 °C. This method will facilitate the implementation of urinary eicosanoid quantification in large-scale screening.
Collapse
Affiliation(s)
- Julia Sieminska
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Metabolomics Laboratory, Clinical Research Center, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Johan Kolmert
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Javier Zurita
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Johanna Revol-Cavalier
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jacek Niklinski
- Department of Clinical Molecular Biology, Medical University of Bialystok, Waszyngtona 13, 15-269 Bialystok, Poland
| | - Joanna Reszec
- Department of Medical Patomorphology, Medical University of Bialystok, Waszyngtona 13, 15-269 Bialystok, Poland
| | - Sven-Erik Dahlén
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Michal Ciborowski
- Metabolomics Laboratory, Clinical Research Center, Medical University of Bialystok, 15-276 Bialystok, Poland.
| | - Craig E Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
11
|
Pawelzik SC, Arnardottir H, Sarajlic P, Mahdi A, Vigor C, Zurita J, Zhou B, Kolmert J, Galano JM, Religa D, Durand T, Wheelock CE, Bäck M. Decreased oxidative stress and altered urinary oxylipidome by intravenous omega-3 fatty acid emulsion in a randomized controlled trial of older subjects hospitalized for COVID-19. Free Radic Biol Med 2023; 194:308-315. [PMID: 36509313 PMCID: PMC9733960 DOI: 10.1016/j.freeradbiomed.2022.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Proinflammatory bioactive lipid mediators and oxidative stress are increased in coronavirus disease 2019 (COVID-19). The randomized controlled single-blind trial COVID-Omega-F showed that intravenous omega-3 polyunsaturated fatty acids (n-3 PUFA) shifted the plasma lipid signature of COVID-19 towards increased proresolving precursor levels and decreased leukotoxin diols, associated with a beneficial immunodulatory response. The present study aimed to determine the effects of n-3 PUFA on the urinary oxylipidome and oxidative stress in COVID-19. From the COVID-Omega-F trial, 20 patients hospitalized for COVID-19 had available serial urinary samples collected at baseline, after 24-48 h, and after completing 5 days treatment with one daily intravenous infusion (2 mL/kg) of either placebo (NaCl; n = 10) or a lipid emulsion containing 10 g of n-3 PUFA per 100 mL (n = 10). Urinary eicosanoids and isoprostanes were analyzed by liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS). Erythrocytes obtained at the different time-points from n = 10 patients (n = 5 placebo and n = 5 n-3 PUFA) were used for determination of reactive oxygen species. Intravenous n-3 PUFA emulsion administration altered eicosanoid metabolites towards decreased levels for mediators of inflammation and thrombosis, and increased levels of the endothelial function mediator prostacyclin. Furthermore, non-enzymatic metabolism was skewed towards n-3 PUFA-derived metabolites with potential anti-inflammatory and pro-resolving effects. The oxidative stress marker 15-F2t-isoprostane was significantly lower in patients receiving n-3 PUFA treatment, who also exhibited significantly decreased erythrocyte oxidative stress compared with placebo-treated patients. These findings point to additional beneficial effects of intravenous n-3 PUFA emulsion treatment through a beneficial oxylipin profile and decreased oxidative stress in COVID-19.
Collapse
Affiliation(s)
- Sven-Christian Pawelzik
- Department of Medicine, Karolinska Institutet, Theme Heart, Vessels, and Neuro, Karolinska University Hospital, Stockholm, Sweden
| | - Hildur Arnardottir
- Department of Medicine, Karolinska Institutet, Theme Heart, Vessels, and Neuro, Karolinska University Hospital, Stockholm, Sweden
| | - Philip Sarajlic
- Department of Medicine, Karolinska Institutet, Theme Heart, Vessels, and Neuro, Karolinska University Hospital, Stockholm, Sweden
| | - Ali Mahdi
- Department of Medicine, Karolinska Institutet, Theme Heart, Vessels, and Neuro, Karolinska University Hospital, Stockholm, Sweden
| | - Claire Vigor
- Institut des Biomolécules Max Mousseron, IBMM, UMR 5247, Université de Montpellier, CNRS, ENSCM, Pôle Recherche Chimie Balard, 34293, Cedex 5, Montpellier, France
| | - Javier Zurita
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bingqing Zhou
- Institut des Biomolécules Max Mousseron, IBMM, UMR 5247, Université de Montpellier, CNRS, ENSCM, Pôle Recherche Chimie Balard, 34293, Cedex 5, Montpellier, France
| | - Johan Kolmert
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron, IBMM, UMR 5247, Université de Montpellier, CNRS, ENSCM, Pôle Recherche Chimie Balard, 34293, Cedex 5, Montpellier, France
| | - Dorota Religa
- Department of Neurobiology, Karolinska Institutet and Theme Ageing, Karolinska University Hospital, Stockholm, Sweden
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron, IBMM, UMR 5247, Université de Montpellier, CNRS, ENSCM, Pôle Recherche Chimie Balard, 34293, Cedex 5, Montpellier, France
| | - Craig E Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Magnus Bäck
- Department of Medicine, Karolinska Institutet, Theme Heart, Vessels, and Neuro, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
12
|
Prostanoid Metabolites as Biomarkers in Human Disease. Metabolites 2022; 12:metabo12080721. [PMID: 36005592 PMCID: PMC9414732 DOI: 10.3390/metabo12080721] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Prostaglandins (PGD2, PGE2, PGF2α), prostacyclin (PGI2), and thromboxane A2 (TXA2) together form the prostanoid family of lipid mediators. As autacoids, these five primary prostanoids propagate intercellular signals and are involved in many physiological processes. Furthermore, alterations in their biosynthesis accompany a wide range of pathological conditions, which leads to substantially increased local levels during disease. Primary prostanoids are chemically instable and rapidly metabolized. Their metabolites are more stable, integrate the local production on a systemic level, and their analysis in various biological matrices yields valuable information under different pathological settings. Therefore, prostanoid metabolites may be used as diagnostic, predictive, or prognostic biomarkers in human disease. Although their potential as biomarkers is great and extensive research has identified major prostanoid metabolites that serve as target analytes in different biofluids, the number of studies that correlate prostanoid metabolite levels to disease outcome is still limited. We review the metabolism of primary prostanoids in humans, summarize the levels of prostanoid metabolites in healthy subjects, and highlight existing biomarker studies. Since analysis of prostanoid metabolites is challenging because of ongoing metabolism and limited half-lives, an emphasis of this review lies on the reliable measurement and interpretation of obtained levels.
Collapse
|
13
|
Arriaga MB, Karim F, Queiroz ATL, Araújo-Pereira M, Barreto-Duarte B, Sales C, Moosa MYS, Mazibuko M, Milne GL, Maruri F, Serezani CH, Koethe JR, Figueiredo MC, Kritski AL, Cordeiro-Santos M, Rolla VC, Sterling TR, Leslie A, Andrade BB. Effect of Dysglycemia on Urinary Lipid Mediator Profiles in Persons With Pulmonary Tuberculosis. Front Immunol 2022; 13:919802. [PMID: 35874781 PMCID: PMC9304990 DOI: 10.3389/fimmu.2022.919802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background Oxidized lipid mediators such as eicosanoids play a central role in the inflammatory response associated with tuberculosis (TB) pathogenesis. Diabetes mellitus (DM) leads to marked changes in lipid mediators in persons with TB. However, the associations between diabetes-related changes in lipid mediators and clearance of M. tuberculosis (Mtb) among persons on anti-TB treatment (ATT) are unknown. Quantification of urinary eicosanoid metabolites can provide insights into the circulating lipid mediators involved in Mtb immune responses. Methods We conducted a multi-site prospective observational study among adults with drug-sensitive pulmonary TB and controls without active TB; both groups had sub-groups with or without dysglycemia at baseline. Participants were enrolled from RePORT-Brazil (Salvador site) and RePORT-South Africa (Durban site) and stratified according to TB status and baseline glycated hemoglobin levels: a) TB-dysglycemia (n=69); b) TB-normoglycemia (n=64); c) non-TB/dysglycemia (n=31); d) non-TB/non-dysglycemia (n=29). We evaluated the following urinary eicosanoid metabolites: 11α-hydroxy-9,15-dioxo-2,3,4,5-tetranor-prostane-1,20-dioic acid (major urinary metabolite of prostaglandin E2, PGE-M), tetranor-PGE1 (metabolite of PGE2, TN-E), 9α-hydroxy-11,15-dioxo-2,3,4,5-tetranor-prostane-1,20-dioic acid (metabolite of PGD2, PGD-M), 11-dehydro-thromboxane B2 (11dTxB2), 2,3-dinor-6-keto-PGF1α (prostaglandin I metabolite, PGI-M), and leukotriene E4 (LTE4). Comparisons between the study groups were performed at three time points: before ATT and 2 and 6 months after initiating therapy. Results PGE-M and LTE4 values were consistently higher at all three time-points in the TB-dysglycemia group compared to the other groups (p<0.001). In addition, there was a significant decrease in PGI-M and LTE4 levels from baseline to month 6 in the TB-dysglycemia and TB-normoglycemia groups. Finally, TB-dysglycemia was independently associated with increased concentrations of PGD-M, PGI-M, and LTE4 at baseline in a multivariable model adjusting for age, sex, BMI, and study site. These associations were not affected by HIV status. Conclusion The urinary eicosanoid metabolite profile was associated with TB-dysglycemia before and during ATT. These observations can help identify the mechanisms involved in the pathogenesis of TB-dysglycemia, and potential biomarkers of TB treatment outcomes, including among persons with dysglycemia.
Collapse
Affiliation(s)
- María B Arriaga
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Brazil.,Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Farina Karim
- Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Artur T L Queiroz
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Center of Data and Knowledge Integration for Health (CIDACS), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Mariana Araújo-Pereira
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Brazil
| | - Beatriz Barreto-Duarte
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Curso de Medicina, Universidade Salvador (UNIFACS), Salvador, Brazil
| | - Caio Sales
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Curso de Medicina, Universidade Salvador (UNIFACS), Salvador, Brazil
| | | | - Matilda Mazibuko
- Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Ginger L Milne
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Fernanda Maruri
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Carlos Henrique Serezani
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - John R Koethe
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Marina C Figueiredo
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Afrânio L Kritski
- Programa Acadêmico de Tuberculose da Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Cordeiro-Santos
- Fundação Medicina Tropical Dr Heitor Vieira Dourado, Manaus, Brazil.,Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil.,Universidade Federal do Amazonas, Manaus, Brazil
| | - Valeria C Rolla
- Laboratório de Pesquisa Clínica em Micobacteriose, Instituto Nacional de Infectologia Evandro Chagas, Fiocruz, Rio de Janeiro, Brazil
| | - Timothy R Sterling
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Alasdair Leslie
- Africa Health Research Institute, Durban, South Africa.,Division of Infection and Immunity, University College London, London, United Kingdom
| | - Bruno B Andrade
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Brazil.,Curso de Medicina, Universidade Salvador (UNIFACS), Salvador, Brazil.,Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States.,Curso de Medicina, Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Brazil
| |
Collapse
|
14
|
Kratz D, Wilken-Schmitz A, Sens A, Hahnefeld L, Scholich K, Geisslinger G, Gurke R, Thomas D. Post-mortem changes of prostanoid concentrations in tissues of mice: Impact of fast cervical dislocation and dissection delay. Prostaglandins Other Lipid Mediat 2022; 162:106660. [PMID: 35714920 DOI: 10.1016/j.prostaglandins.2022.106660] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/18/2022] [Accepted: 06/10/2022] [Indexed: 11/29/2022]
Abstract
Prostanoids are potent lipid mediators involved in a wide variety of physiological functions like blood pressure regulation or inflammation as well as cardiovascular and malign diseases. Elucidation of their modes of action is mainly carried out in pre-clinical animal models by quantifying prostanoids in tissues of interest. Unfortunately, prostanoids are prone to post-mortem artifact formation and de novo synthesis can already be caused by external stimuli during the euthanasia of animals like prolonged hypercapnia or ischemia. Therefore, this study investigates the suitability and impact of fast cervical dislocation for the determination of prostanoids (6-keto-PGF1α, TXB2, PGF2α, PGD2, PGE2) in seven tissues of mice (spinal cord, brain, sciatic nerve, kidney, liver, lung, and spleen) to minimize time-dependent effects and approximate physiological concentrations. Tissues were dissected in a standardized sequence directly or after 10 min to investigate the influence of dissection delays. The enzyme inhibitor indomethacin (10 µM) in combination with low processing temperatures was employed to preserve prostanoid concentrations during sample preparation. Quantification of prostanoids was performed via LC-MS/MS. This study shows, that prostanoids are differentially susceptible to post-mortem artifact formation which is closely connected to their physiological function and metabolic stability in the respective tissues. Prostanoids in the brain, spinal cord, and kidney that are not involved in the regulatory response post-mortem, i.e. blood flow regulation (6-keto-PGF1α, PGE2, PGF2α) showed high reproducibility even after dissection delay and could be assessed after fast cervical dislocation if prerequisites like standardized pre-analytical workflows with immediate dissection and inhibition of residual enzymatic activity are in place. However, in tissues with high metabolic activity (liver, lung) more stable prostanoid metabolites should be used. Moreover, prostanoids in the spleen were strongly affected by dissection delays and presumably the method of euthanasia itself.
Collapse
Affiliation(s)
- D Kratz
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - A Wilken-Schmitz
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - A Sens
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - L Hahnefeld
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - K Scholich
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - G Geisslinger
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - R Gurke
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.
| | - D Thomas
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
15
|
Ryu MH, Gómez C, Yuen ACY, Brook JR, Wheelock CE, Carlsten C. Urinary Eicosanoid Levels Reflect Allergen and Diesel Exhaust Coexposure and Are Linked to Impaired Lung Function. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:7107-7118. [PMID: 35044166 DOI: 10.1021/acs.est.1c07268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Eicosanoids are potent regulators of homeostasis and inflammation. Co-exposure to allergen and diesel exhaust (DE) have been shown to lead to eosinophilic inflammation, impaired airflow, and increased airway responsiveness. It is not clear whether eicosanoids mediate the mechanism by which these exposures impair lung function. We conducted a randomized, double-blinded, and four-arm crossover study. Fourteen allergen-sensitized participants were exposed to four conditions: negative control; allergen-alone exposure; DE and allergen coexposure; coexposure with particle-reducing technology applied. Quantitative metabolic profiling of urinary eicosanoids was performed using LC-MS/MS. As expected, allergen inhalation increased eicosanoids. The prostacyclin metabolite 2,3-dinor-6-keto-PGF1α (PGF1α, prostaglandin F1α) increased with coexposure, but particle depletion suppressed this pathway. Individuals with a high genetic risk score demonstrated a greater increase in isoprostane metabolites following coexposure. Causal mediation analyses showed that allergen induced airflow impairment was mediated via leukotriene E4 and tetranor-prostaglandin D metabolite. Overall, DE exposure did not augment the allergen's effect on urinary eicosanoids, except insofar as variant genotypes conferred susceptibility to the addition of DE in terms of isoprostane metabolites. These findings will add to the body of previous controlled human exposure studies and provide greater insight into how complex environmental exposures in urban air may influence individuals with sensitivity to aeroallergens.
Collapse
Affiliation(s)
- Min Hyung Ryu
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, The University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Cristina Gómez
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm SE-171 65, Sweden
- Unit of Lung and Allergy Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Agnes C Y Yuen
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, The University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Jeffrey R Brook
- Occupational and Environmental Health Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario M5T 1P8, Canada
| | - Craig E Wheelock
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm SE-171 65, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm SE-171 76, Sweden
- Gunma University Initiative for Advanced Research (GIAR), Gunma University, Maebashi, Gunma 371-8511, Japan
| | - Christopher Carlsten
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, The University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| |
Collapse
|
16
|
Gallart-Ayala H, Teav T, Ivanisevic J. Metabolomics meets lipidomics: Assessing the small molecule component of metabolism. Bioessays 2021; 42:e2000052. [PMID: 33230910 DOI: 10.1002/bies.202000052] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 09/11/2020] [Indexed: 12/16/2022]
Abstract
Metabolomics, including lipidomics, is emerging as a quantitative biology approach for the assessment of energy flow through metabolism and information flow through metabolic signaling; thus, providing novel insights into metabolism and its regulation, in health, healthy ageing and disease. In this forward-looking review we provide an overview on the origins of metabolomics, on its role in this postgenomic era of biochemistry and its application to investigate metabolite role and (bio)activity, from model systems to human population studies. We present the challenges inherent to this analytical science, and approaches and modes of analysis that are used to resolve, characterize and measure the infinite chemical diversity contained in the metabolome (including lipidome) of complex biological matrices. In the current outbreak of metabolic diseases such as cardiometabolic disorders, cancer and neurodegenerative diseases, metabolomics appears to be ideally situated for the investigation of disease pathophysiology from a metabolite perspective.
Collapse
Affiliation(s)
- Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Tony Teav
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
17
|
Abstract
Biomarkers may be diagnostic of asthma, they may predict or reflect response to therapy or they may identify patients at risk of asthma exacerbation. A biomarker is most often measured in biologic fluids that are sampled using relatively non-invasive sampling techniques such as blood, sputum, urine or exhaled breath. Biomarkers should be stable, readily quantifiable and their measurement should be reproducible and not confounded by other host factors, or the presence of comorbidities. However, asthma comprises multiple molecular endotypes and single, sensitive, specific, biomarkers reflecting these endotypes may not exist. Combining biomarkers may improve their predictive capability in asthma. The most well-established endotypes are those described as Type2 and non-Type2 asthma. Clinical trials established the fraction of exhaled nitric oxide (FeNO) and blood eosinophil counts as key biomarkers of response to corticosteroid or targeted anti-inflammatory therapy in Type2 asthma. However, these biomarkers may have limited value in the management of asthma in real-life settings or routine clinical practise. Biomarkers for Type2 asthma are not well described or validated and more research is needed. Breathomics has provided evidence to propose a number of exhaled volatile organic compounds (VOCs) as surrogate biomarkers for airway inflammatory phenotypes, disease activity and adherence to therapy. Analysis of urinary eicosanoids has identified eicosanoids related to Type2 and non-Type2 inflammation. Future clinical trials will be important in determining how exhaled VOCs or urinary eicosanoid profiles can be used to direct precision treatments. Their future clinical use will also depend on developing simplified instrumentation for biomarker analysis at the point-of-care.
Collapse
Affiliation(s)
- Janis Shute
- School of Pharmacy and Biomedical Sciences, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK -
| |
Collapse
|
18
|
Nakamura T. The roles of lipid mediators in type I hypersensitivity. J Pharmacol Sci 2021; 147:126-131. [PMID: 34294363 DOI: 10.1016/j.jphs.2021.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/22/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Type I hypersensitivity is an immediate immune reaction that involves IgE-mediated activation of mast cells. Activated mast cells release chemical mediators, such as histamine and lipid mediators, which cause allergic reactions. Recent developments in detection devices have revealed that mast cells simultaneously release a wide variety of lipid mediators. Mounting evidence has revealed that mast cell-derived mediators exert both pro- and anti-inflammatory functions and positively and negatively regulate the development of allergic inflammation. This review presents the roles of major lipid mediators released from mast cells. Author believes this review will be helpful for a better understanding of the pathogenesis of allergic diseases and provide a new strategy for the diagnosis and treatment of allergic reactions.
Collapse
Affiliation(s)
- Tatsuro Nakamura
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan.
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Finding suitable biomarkers to phenotype asthma, identify individuals at risk of worsening and guide treatment is highly prioritized in asthma research. We aimed to provide an analysis of currently used and upcoming biomarkers, focusing on developments published in the past 2 years. RECENT FINDINGS Type 2 inflammation is the most studied asthma mechanism with the most biomarkers in the pipeline. Blood eosinophils and fractional exhaled nitric oxide (FeNO) are those most used clinically. Recent developments include their ability to identify individuals at higher risk of exacerbations, faster decline in lung function and more likely to benefit from anti-IL-5 and anti-IL-4/-13 treatment. Certain patterns of urinary eicosanoid excretion also relate to type 2 inflammation. Results of recent trials investigating the use of serum periostin or dipeptidyl peptidase-4 to guide anti-IL-13 therapy were somewhat disappointing. Less is known about non-type 2 inflammation but blood neutrophils and YKL-40 may be higher in patients with evidence of non-type 2 asthma. Volatile organic compounds show promise in their ability to distinguish both eosinophilic and neutrophilic asthma. SUMMARY The ultimate panel of biomarkers for identification of activated inflammatory pathways and treatment strategies in asthma patients still lies in the future, particularly for non-type 2 asthma, but potential candidates are available.
Collapse
|
20
|
Meister I, Zhang P, Sinha A, Sköld CM, Wheelock ÅM, Izumi T, Chaleckis R, Wheelock CE. High-Precision Automated Workflow for Urinary Untargeted Metabolomic Epidemiology. Anal Chem 2021; 93:5248-5258. [PMID: 33739820 PMCID: PMC8041248 DOI: 10.1021/acs.analchem.1c00203] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/26/2021] [Indexed: 12/15/2022]
Abstract
Urine is a noninvasive biofluid that is rich in polar metabolites and well suited for metabolomic epidemiology. However, because of individual variability in health and hydration status, the physiological concentration of urine can differ >15-fold, which can pose major challenges in untargeted liquid chromatography-mass spectrometry (LC-MS) metabolomics. Although numerous urine normalization methods have been implemented (e.g., creatinine, specific gravity-SG), most are manual and, therefore, not practical for population-based studies. To address this issue, we developed a method to measure SG in 96-well-plates using a refractive index detector (RID), which exhibited accuracy within 85-115% and <3.4% precision. Bland-Altman statistics showed a mean deviation of -0.0001 SG units (limits of agreement: -0.0014 to 0.0011) relative to a hand-held refractometer. Using this RID-based SG normalization, we developed an automated LC-MS workflow for untargeted urinary metabolomics in a 96-well-plate format. The workflow uses positive and negative ionization HILIC chromatography and acquires mass spectra in data-independent acquisition (DIA) mode at three collision energies. Five technical internal standards (tISs) were used to monitor data quality in each method, all of which demonstrated raw coefficients of variation (CVs) < 10% in the quality controls (QCs) and < 20% in the samples for a small cohort (n = 87 urine samples, n = 22 QCs). Application in a large cohort (n = 842 urine samples, n = 248 QCs) demonstrated CVQC < 5% and CVsamples < 16% for 4/5 tISs after signal drift correction by cubic spline regression. The workflow identified >540 urinary metabolites including endogenous and exogenous compounds. This platform is suitable for performing urinary untargeted metabolomic epidemiology and will be useful for applications in population-based molecular phenotyping.
Collapse
Affiliation(s)
- Isabel Meister
- Gunma
University Initiative for Advanced Research (GIAR), Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
- Division
of Physiological Chemistry 2, Department of Medical Biochemistry and
Biophysics, Karolinska Institutet, Biomedicum Quartier 9A, Stockholm 171-77, Sweden
| | - Pei Zhang
- Gunma
University Initiative for Advanced Research (GIAR), Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
- Division
of Physiological Chemistry 2, Department of Medical Biochemistry and
Biophysics, Karolinska Institutet, Biomedicum Quartier 9A, Stockholm 171-77, Sweden
| | - Anirban Sinha
- Department
of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
- Department
of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
- Computational
Physiology and Biostatistics, University
Children’s Hospital, Spitalstrasse 33, Basel 4056, Switzerland
| | - C. Magnus Sköld
- Respiratory
Medicine Unit, K2 Department of Medicine Solna and Center for Molecular
Medicine, Karolinska Institutet, Stockholm 141-86, Sweden
- Department
of Respiratory Medicine and Allergy, Karolinska
University Hospital, Stockholm 141-86, Sweden
| | - Åsa M. Wheelock
- Respiratory
Medicine Unit, K2 Department of Medicine Solna and Center for Molecular
Medicine, Karolinska Institutet, Stockholm 141-86, Sweden
- Department
of Respiratory Medicine and Allergy, Karolinska
University Hospital, Stockholm 141-86, Sweden
| | - Takashi Izumi
- Gunma
University Initiative for Advanced Research (GIAR), Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
- Department
of Biochemistry, Gunma University Graduate
School of Medicine, 3-39-22
Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Romanas Chaleckis
- Gunma
University Initiative for Advanced Research (GIAR), Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
- Division
of Physiological Chemistry 2, Department of Medical Biochemistry and
Biophysics, Karolinska Institutet, Biomedicum Quartier 9A, Stockholm 171-77, Sweden
| | - Craig E. Wheelock
- Gunma
University Initiative for Advanced Research (GIAR), Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
- Division
of Physiological Chemistry 2, Department of Medical Biochemistry and
Biophysics, Karolinska Institutet, Biomedicum Quartier 9A, Stockholm 171-77, Sweden
- Department
of Respiratory Medicine and Allergy, Karolinska
University Hospital, Stockholm 141-86, Sweden
| |
Collapse
|
21
|
Kolmert J, Gómez C, Balgoma D, Sjödin M, Bood J, Konradsen JR, Ericsson M, Thörngren JO, James A, Mikus M, Sousa AR, Riley JH, Bates S, Bakke PS, Pandis I, Caruso M, Chanez P, Fowler SJ, Geiser T, Howarth P, Horváth I, Krug N, Montuschi P, Sanak M, Behndig A, Shaw DE, Knowles RG, Holweg CTJ, Wheelock ÅM, Dahlén B, Nordlund B, Alving K, Hedlin G, Chung KF, Adcock IM, Sterk PJ, Djukanovic R, Dahlén SE, Wheelock CE. Urinary Leukotriene E 4 and Prostaglandin D 2 Metabolites Increase in Adult and Childhood Severe Asthma Characterized by Type 2 Inflammation. A Clinical Observational Study. Am J Respir Crit Care Med 2021; 203:37-53. [PMID: 32667261 PMCID: PMC7781128 DOI: 10.1164/rccm.201909-1869oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Rationale: New approaches are needed to guide personalized treatment of asthma.Objectives: To test if urinary eicosanoid metabolites can direct asthma phenotyping.Methods: Urinary metabolites of prostaglandins (PGs), cysteinyl leukotrienes (CysLTs), and isoprostanes were quantified in the U-BIOPRED (Unbiased Biomarkers for the Prediction of Respiratory Diseases Outcomes) study including 86 adults with mild-to-moderate asthma (MMA), 411 with severe asthma (SA), and 100 healthy control participants. Validation was performed internally in 302 participants with SA followed up after 12-18 months and externally in 95 adolescents with asthma.Measurement and Main Results: Metabolite concentrations in healthy control participants were unrelated to age, body mass index, and sex, except for the PGE2 pathway. Eicosanoid concentrations were generally greater in participants with MMA relative to healthy control participants, with further elevations in participants with SA. However, PGE2 metabolite concentrations were either the same or lower in male nonsmokers with asthma than in healthy control participants. Metabolite concentrations were unchanged in those with asthma who adhered to oral corticosteroid treatment as documented by urinary prednisolone detection, whereas those with SA treated with omalizumab had lower concentrations of LTE4 and the PGD2 metabolite 2,3-dinor-11β-PGF2α. High concentrations of LTE4 and PGD2 metabolites were associated with lower lung function and increased amounts of exhaled nitric oxide and eosinophil markers in blood, sputum, and urine in U-BIOPRED participants and in adolescents with asthma. These type 2 (T2) asthma associations were reproduced in the follow-up visit of the U-BIOPRED study and were found to be as sensitive to detect T2 inflammation as the established biomarkers.Conclusions: Monitoring of urinary eicosanoids can identify T2 asthma and introduces a new noninvasive approach for molecular phenotyping of adult and adolescent asthma.Clinical trial registered with www.clinicaltrials.gov (NCT01976767).
Collapse
Affiliation(s)
- Johan Kolmert
- The Institute of Environmental Medicine.,Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics.,The Center for Allergy Research
| | - Cristina Gómez
- The Institute of Environmental Medicine.,Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics.,The Center for Allergy Research
| | - David Balgoma
- The Institute of Environmental Medicine.,Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics.,The Center for Allergy Research
| | - Marcus Sjödin
- The Institute of Environmental Medicine.,Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics.,The Center for Allergy Research
| | - Johan Bood
- The Institute of Environmental Medicine.,The Center for Allergy Research.,Department of Women's and Children's Health, and
| | - Jon R Konradsen
- The Center for Allergy Research.,Respiratory Medicine Unit, Department of Medicine, Solna Campus, and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Medicine and
| | - Magnus Ericsson
- Department of Clinical Pharmacology, Huddinge Campus, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - John-Olof Thörngren
- Department of Clinical Pharmacology, Huddinge Campus, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Anna James
- The Institute of Environmental Medicine.,The Center for Allergy Research
| | - Maria Mikus
- The Institute of Environmental Medicine.,The Center for Allergy Research
| | - Ana R Sousa
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - John H Riley
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Stewart Bates
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | | | - Ioannis Pandis
- Institute of Medicine, University of Bergen, Bergen, Norway
| | - Massimo Caruso
- National Heart and Lung Institute and Department of Computing & Data Science Institute, Imperial College London, London, United Kingdom.,Department of Clinical and Experimental Medicine and
| | - Pascal Chanez
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Stephen J Fowler
- Clinique des Bronches, Allergies et Sommeil, Aix Marseille Université, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Thomas Geiser
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, and Manchester Academic Health Science Centre and National Institute for Health Research Biomedical Research Centre, Manchester University Hospitals National Health Service Foundation Trust, Manchester, United Kingdom
| | - Peter Howarth
- Department of Pulmonary Medicine, University Hospital Bern, Bern, Switzerland
| | - Ildikó Horváth
- Faculty of Medicine, Southampton University, and National Institute for Health Research Southampton Respiratory Biomedical Research Center, University Hospital Southampton, Southampton, United Kingdom
| | - Norbert Krug
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Paolo Montuschi
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Marek Sanak
- Department of Pharmacology, Catholic University of the Sacred Heart, and Agostino Gemelli University Hospital Foundation, IRCCS, Rome, Italy
| | - Annelie Behndig
- Department of Internal Medicine, Medical College, Jagiellonian University, Cracow, Poland
| | - Dominick E Shaw
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Richard G Knowles
- Nottingham National Institute for Health Research Biomedical Research Centre, University of Nottingham, United Kingdom
| | - Cécile T J Holweg
- Knowles Consulting, Stevenage Bioscience Catalyst, Stevenage, United Kingdom
| | | | - Barbro Dahlén
- The Center for Allergy Research.,Department of Women's and Children's Health, and
| | - Björn Nordlund
- Respiratory Medicine Unit, Department of Medicine, Solna Campus, and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Medicine and
| | - Kjell Alving
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden; and
| | - Gunilla Hedlin
- The Center for Allergy Research.,Respiratory Medicine Unit, Department of Medicine, Solna Campus, and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Medicine and
| | - Kian Fan Chung
- Institute of Medicine, University of Bergen, Bergen, Norway
| | - Ian M Adcock
- Institute of Medicine, University of Bergen, Bergen, Norway
| | - Peter J Sterk
- Department of Respiratory Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Ratko Djukanovic
- Department of Pulmonary Medicine, University Hospital Bern, Bern, Switzerland
| | - Sven-Erik Dahlén
- The Institute of Environmental Medicine.,The Center for Allergy Research
| | - Craig E Wheelock
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics.,The Center for Allergy Research
| | | |
Collapse
|
22
|
Tolstikov V, Moser AJ, Sarangarajan R, Narain NR, Kiebish MA. Current Status of Metabolomic Biomarker Discovery: Impact of Study Design and Demographic Characteristics. Metabolites 2020; 10:metabo10060224. [PMID: 32485899 PMCID: PMC7345110 DOI: 10.3390/metabo10060224] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/21/2020] [Accepted: 05/27/2020] [Indexed: 12/16/2022] Open
Abstract
Widespread application of omic technologies is evolving our understanding of population health and holds promise in providing precise guidance for selection of therapeutic interventions based on patient biology. The opportunity to use hundreds of analytes for diagnostic assessment of human health compared to the current use of 10–20 analytes will provide greater accuracy in deconstructing the complexity of human biology in disease states. Conventional biochemical measurements like cholesterol, creatinine, and urea nitrogen are currently used to assess health status; however, metabolomics captures a comprehensive set of analytes characterizing the human phenotype and its complex metabolic processes in real-time. Unlike conventional clinical analytes, metabolomic profiles are dramatically influenced by demographic and environmental factors that affect the range of normal values and increase the risk of false biomarker discovery. This review addresses the challenges and opportunities created by the evolving field of clinical metabolomics and highlights features of study design and bioinformatics necessary to maximize the utility of metabolomics data across demographic groups.
Collapse
Affiliation(s)
- Vladimir Tolstikov
- BERG, Precision Medicine Division, Framingham, MA 01701, USA; (V.T.); (R.S.); (N.R.N.)
| | - A. James Moser
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA;
| | | | - Niven R. Narain
- BERG, Precision Medicine Division, Framingham, MA 01701, USA; (V.T.); (R.S.); (N.R.N.)
| | - Michael A. Kiebish
- BERG, Precision Medicine Division, Framingham, MA 01701, USA; (V.T.); (R.S.); (N.R.N.)
- Correspondence: ; Tel.: +1-617-588-2245
| |
Collapse
|
23
|
Long NP, Nghi TD, Kang YP, Anh NH, Kim HM, Park SK, Kwon SW. Toward a Standardized Strategy of Clinical Metabolomics for the Advancement of Precision Medicine. Metabolites 2020; 10:E51. [PMID: 32013105 PMCID: PMC7074059 DOI: 10.3390/metabo10020051] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/18/2022] Open
Abstract
Despite the tremendous success, pitfalls have been observed in every step of a clinical metabolomics workflow, which impedes the internal validity of the study. Furthermore, the demand for logistics, instrumentations, and computational resources for metabolic phenotyping studies has far exceeded our expectations. In this conceptual review, we will cover inclusive barriers of a metabolomics-based clinical study and suggest potential solutions in the hope of enhancing study robustness, usability, and transferability. The importance of quality assurance and quality control procedures is discussed, followed by a practical rule containing five phases, including two additional "pre-pre-" and "post-post-" analytical steps. Besides, we will elucidate the potential involvement of machine learning and demonstrate that the need for automated data mining algorithms to improve the quality of future research is undeniable. Consequently, we propose a comprehensive metabolomics framework, along with an appropriate checklist refined from current guidelines and our previously published assessment, in the attempt to accurately translate achievements in metabolomics into clinical and epidemiological research. Furthermore, the integration of multifaceted multi-omics approaches with metabolomics as the pillar member is in urgent need. When combining with other social or nutritional factors, we can gather complete omics profiles for a particular disease. Our discussion reflects the current obstacles and potential solutions toward the progressing trend of utilizing metabolomics in clinical research to create the next-generation healthcare system.
Collapse
Affiliation(s)
- Nguyen Phuoc Long
- College of Pharmacy, Seoul National University, Seoul 08826, Korea; (N.P.L.); (N.H.A.); (H.M.K.)
| | - Tran Diem Nghi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea; (T.D.N.); (S.K.P.)
| | - Yun Pyo Kang
- Department of Cancer Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA;
| | - Nguyen Hoang Anh
- College of Pharmacy, Seoul National University, Seoul 08826, Korea; (N.P.L.); (N.H.A.); (H.M.K.)
| | - Hyung Min Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Korea; (N.P.L.); (N.H.A.); (H.M.K.)
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea; (T.D.N.); (S.K.P.)
| | - Sung Won Kwon
- College of Pharmacy, Seoul National University, Seoul 08826, Korea; (N.P.L.); (N.H.A.); (H.M.K.)
| |
Collapse
|
24
|
Jia M, Peng Z, Yang K, Su C, Wang Y, Yan C. A high-throughput targeted metabolomics method for the quantification of 104 non-polar metabolites in cholesterol, eicosanoid, and phospholipid metabolism: application in the study of a CCl4-induced liver injury mouse model. Analyst 2020; 145:3575-3591. [PMID: 32329491 DOI: 10.1039/d0an00385a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Experimental workflow of 104 non-polar metabolites in cholesterol, eicosanoid, and phospholipid metabolisms analysis using UPLC-QqQ-MS.
Collapse
Affiliation(s)
- Mengqi Jia
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai
- China
| | - Zhangxiao Peng
- Department of Molecular Oncology
- Eastern Hepatobiliary Surgical Hospital & National Centre for Liver Cancer
- Second Military Medical University
- Shanghai 200438
- China
| | - Kaige Yang
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai
- China
| | - Changqing Su
- Department of Molecular Oncology
- Eastern Hepatobiliary Surgical Hospital & National Centre for Liver Cancer
- Second Military Medical University
- Shanghai 200438
- China
| | - Yan Wang
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai
- China
| | - Chao Yan
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai
- China
| |
Collapse
|