1
|
Emwas AH, Zacharias HU, Alborghetti MR, Gowda GAN, Raftery D, McKay RT, Chang CK, Saccenti E, Gronwald W, Schuchardt S, Leiminger R, Merzaban J, Madhoun NY, Iqbal M, Alsiary RA, Shivapurkar R, Pain A, Shanmugam D, Ryan D, Roy R, Schirra HJ, Morris V, Zeri AC, Alahmari F, Kaddurah-Daouk R, Salek RM, LeVatte M, Berjanskii M, Lee B, Wishart DS. Recommendations for sample selection, collection and preparation for NMR-based metabolomics studies of blood. Metabolomics 2025; 21:66. [PMID: 40348843 PMCID: PMC12065766 DOI: 10.1007/s11306-025-02259-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/04/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Metabolic profiling of blood metabolites, particularly in plasma and serum, is vital for studying human diseases, human conditions, drug interventions and toxicology. The clinical significance of blood arises from its close ties to all human cells and facile accessibility. However, patient-specific variables such as age, sex, diet, lifestyle and health status, along with pre-analytical conditions (sample handling, storage, etc.), can significantly affect metabolomic measurements in whole blood, plasma, or serum studies. These factors, referred to as confounders, must be mitigated to reveal genuine metabolic changes due to illness or intervention onset. REVIEW OBJECTIVE This review aims to aid metabolomics researchers in collecting reliable, standardized datasets for NMR-based blood (whole/serum/plasma) metabolomics. The goal is to reduce the impact of confounding factors and enhance inter-laboratory comparability, enabling more meaningful outcomes in metabolomics studies. KEY CONCEPTS This review outlines the main factors affecting blood metabolite levels and offers practical suggestions for what to measure and expect, how to mitigate confounding factors, how to properly prepare, handle and store blood, plasma and serum biosamples and how to report data in targeted NMR-based metabolomics studies of blood, plasma and serum.
Collapse
Affiliation(s)
- Abdul-Hamid Emwas
- King Abdullah University of Science and Technology (KAUST), Core Labs, Thuwal, 23955-6900, Kingdom of Saudi Arabia.
| | - Helena U Zacharias
- Peter L. Reichertz Institute for Medical Informatics of TU Braunschweig and Hannover Medical School, Hannover Medical School, 30625, Hannover, Germany
| | - Marcos Rodrigo Alborghetti
- Brazilian Biosciences National Laboratory and Brazilian Center for Research in Energy and Materials, Campinas, 13083-100, Brazil
| | - G A Nagana Gowda
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican St., Seattle, WA, 98109, USA
| | - Daniel Raftery
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican St., Seattle, WA, 98109, USA
| | - Ryan T McKay
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Chung-Ke Chang
- Taiwan Biobank, Biomedical Translation Research Center, Academia Sinica, Taipei City, Taiwan
| | - Edoardo Saccenti
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Wolfram Gronwald
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Sven Schuchardt
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Nikolai-Fuchs-Str. 1, 30625, Hannover, Germany
| | - Roland Leiminger
- Bruker BioSpin GmbH & Co., Rudolf-Plank-Straße 23, 76275, Ettlingen, Germany
| | - Jasmeen Merzaban
- Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Nour Y Madhoun
- Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Mazhar Iqbal
- Drug Discovery and Structural Biology, Health Biotechnology Division, National Institute for Biotechnology & Genetic Engineering (NIBGE), Faisalabad, 38000, Pakistan
| | - Rawiah A Alsiary
- King Abdullah International Medical Research Center (KAIMRC), Saudi Arabia/King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Kingdom of Saudi Arabia
| | - Rupali Shivapurkar
- Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Arnab Pain
- Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Dhanasekaran Shanmugam
- Biochemical Sciences Division, National Chemical Laboratory, Dr. Homi Bhabha Road, 411008, Pune, India
| | - Danielle Ryan
- School of Agricultural, Environmental and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia
| | - Raja Roy
- Centre of Biomedical Research, formerly, Centre of Biomedical Magnetic Resonance, Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Rae Bareli Road, Lucknow, 226014, India
| | - Horst Joachim Schirra
- School of Environment and Sciences, Griffith University, Nathan, QLD, 4111, Australia
- Institute for Biomedicine and Glycomics, Griffith University, Don Young Road, Nathan, QLD, 4111, Australia
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Vanessa Morris
- School of Biological Sciences and Biomolecular Interaction Centre, University of Canterbury, 8140, Christchurch, New Zealand
| | - Ana Carolina Zeri
- Ilum School of Science, Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Zip Code 13083-970, Brazil
| | - Fatimah Alahmari
- Department of NanoMedicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, 31441, Dammam, Saudi Arabia
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioural Sciences, Duke University, Durham, NC, USA
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Reza M Salek
- School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0SP, UK
| | - Marcia LeVatte
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Mark Berjanskii
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Brian Lee
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - David S Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
2
|
Guo W, Hong E, Ma H, Wang J, Wang Q. Effect of the gut microbiome, skin microbiome, plasma metabolome, white blood cells subtype, immune cells, inflammatory proteins, and inflammatory cytokines on asthma: a two-sample Mendelian randomized study and mediation analysis. Front Immunol 2025; 16:1436888. [PMID: 40191192 PMCID: PMC11968350 DOI: 10.3389/fimmu.2025.1436888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Background Asthma is a chronic inflammatory disorder arising from incompletely understood heterogenic gene-environment interactions. This study aims to investigate causal relationships among gut microbiota, skin microbiota, plasma metabolomics, white blood cells subtype, immune cells, inflammatory proteins, inflammatory cytokines, and asthma. Methods First, two-sample Mendelian randomization analysis was used to identify causal relationships. The summary statistics of 412 gut microbiota traits (N = 7 738), 150 skin microbiota traits (N = 579), 1 400 plasma metabolite traits (N = 8 299), white blood cells subtype counts (N = 746 667), 731 immune cell traits (N = 3 669), 91 circulating inflammatory proteins (N = 14 744), 41 inflammatory cytokine traits (N = 8 293), and asthma traits (N = 244 562) were obtained from publicly available genome-wide association studies. Inverse-variance weighted regression was used as the primary Mendelian randomization method. A series of sensitivity analyses was performed to test the robustness of causal estimates. Subsequently, mediation analysis was performed to identify the pathway from gut or skin microbiota to asthma mediated by plasma metabolites, immune cells, and inflammatory proteins. Results Mendelian randomization revealed the causal effects of 31 gut bacterial features (abundances of 19 bacterial pathways and 12 microbiota), 10 skin bacterial features, 108 plasma metabolites (81 metabolites and 27 ratios), 81 immune cells, five circulating inflammatory proteins, and three inflammatory cytokines and asthma. Moreover, the mediation analysis results supported the mediating effects of one plasma metabolite, five immunophenotypes, and one inflammatory protein on the gut or skin microbiota in asthma pathogenesis. Conclusion The findings of this study support a causal relationship among gut microbiota, skin microbiota, plasma metabolites, immune cells, inflammatory proteins, inflammatory cytokines, and asthma. Mediating pathways through which the above factors may affect asthma were proposed. The biomarkers and mediation pathways identified in this work provide new insights into the mechanism of asthma and contribute to its prevention and treatment.
Collapse
Affiliation(s)
- Wenqian Guo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Er Hong
- Department of Respiratory Medicine, Ningbo Hospital of Traditional Chinese Medicine, Zhejiang University of Chinese Medicine, Ningbo, China
| | - Han Ma
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
- The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Ji Wang
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
3
|
Luu QQ, Kim T, Cao TBT, Choi I, Yang SY, An BS, Hwang DY, Choi Y, Park HS. Therapeutic Potential of Arginine-Loaded Red Blood Cell Nanovesicles Targeting Obese Asthma. Mediators Inflamm 2025; 2025:8248722. [PMID: 40134943 PMCID: PMC11936518 DOI: 10.1155/mi/8248722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/12/2025] [Indexed: 03/27/2025] Open
Abstract
Purpose: The role of the gut microbiomes has been emphasized in the pathogenesis of obese asthma (OA). However, the molecular mechanism of airway dysfunction underlying OA has not yet been fully elucidated. The effects of microbiomes on arginine metabolism in relation to lung functions and a novel method for delivering arginine to lung tissue based on arginine-loaded red blood cell (RBC)-derived nanovesicles (NVs) (NVArg) will be investigated. Materials and Methods: Inflammatory status, amino acid profiles, and microbial diversity were evaluated in 20 adult patients with OA compared to 30 adult patients with non-OA (NOA) and 10 healthy control (HC) groups. Changes in gut or lung microbial composition that altered arginine metabolism in relation to airway inflammation were investigated in an OA mouse model in vivo. Additionally, this study evaluated the delivery of arginine to lung tissue utilizing NVArg in vivo and in vitro. Results: Significantly increased Bacteroides abundance but decreased serum arginine concentration with lower forced exhaled volume at 1 s (FEV1) (%) was noted in the OA group compared to the NOA and HC groups. In mouse experiments, when OA mice were given living bacteria from normal control (NC) mice, lung arginine concentration and airway resistance were restored. However, the administration of arginine or its metabolite (citrulline) did not increase the arginine levels in the lung tissues. We therefore created NVArg, which successfully delivered arginine into the cytoplasm of the airway epithelial cell line in vitro. Oral administration of NVArg for OA mice significantly induced the AMP-activated protein kinase (AMPK) and endothelial nitric oxide synthase (eNOS) pathways in airway epithelial cells, which reduced airway resistance and inflammation. Conclusion: These findings suggest that microbiomes contribute to airway dysfunction by regulating arginine metabolism, whereas NVArg treatment may be a potential option for managing OA.
Collapse
Affiliation(s)
- Quoc Quang Luu
- Department of Oral and Maxillofacial Surgery, Loma Linda University School of Dentistry, Loma Linda, California, USA
| | - Taejune Kim
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Thi Bich Tra Cao
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Injung Choi
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Seung Yun Yang
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Beum-Soo An
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Dae Youn Hwang
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Youngwoo Choi
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
4
|
Albers GJ, Michalaki C, Ogger PP, Lloyd AF, Causton B, Walker SA, Caldwell A, Halket JM, Sinclair LV, Forde SH, McCarthy C, Hinks TSC, Lloyd CM, Byrne AJ. Airway macrophage glycolysis controls lung homeostasis and responses to aeroallergen. Mucosal Immunol 2025; 18:121-134. [PMID: 39426627 DOI: 10.1016/j.mucimm.2024.10.002] [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: 04/17/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/21/2024]
Abstract
The lungs represent a dynamic microenvironment where airway macrophages (AMs) are the major lung-resident macrophages. AMs dictate the balance between tissue homeostasis and immune activation and thus have contradictory functions by maintaining tolerance and tissue homeostasis, as well as initiating strong inflammatory responses. Emerging evidence has highlighted the connection between macrophage function and cellular metabolism. However, the functional importance of these processes in tissue-resident specialized macrophage populations such as those found in the airways, remain poorly elucidated. Here, we reveal that glycolysis is a fundamental pathway in AMs which regulates both lung homeostasis and responses to inhaled allergen. Using macrophage specific targeting in vivo, and multi-omics approaches, we determined that glycolytic activity in AMs is necessary to restrain type 2 (T2) immunity during homeostasis. Exposure to a range of common aeroallergens, including house dust mite (HDM), drove AM-glycolysis and furthermore, AM-specific inhibition of glycolysis altered inflammation in the airways and HDM-driven airway metabolic adaptations in vivo. Additionally, allergen sensitised asthmatics had profound metabolic changes in the airways, compared to non-sensitised asthmatic controls. Finally, we found that allergen driven AM-glycolysis in mice was TLR2 dependent. Thus, our findings demonstrate a direct relationship between glycolysis in AMs, AM-mediated homeostatic processes, and T2 immune responses in the lungs. These data suggest that glycolysis is essential for the plasticity of AMs. Depending on the immunological context, AM-glycolysis is required to exert homeostatic activity but once activated by allergen, AM-glycolysis influences inflammatory responses. Thus, precise modulation of glycolytic activity in AMs is essential for preserving lung homeostasis and regulating airway inflammation.
Collapse
Affiliation(s)
- Gesa J Albers
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Patricia P Ogger
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Amy F Lloyd
- Cell Signalling and Immunology, University of Dundee, Dundee, UK
| | - Benjamin Causton
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Simone A Walker
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Anna Caldwell
- Dept. of Nutritional Sciences, School of Life Course & Population Health Sciences, King's College London, London, UK; Department of Nutritional Sciences, KIng's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - John M Halket
- Department of Nutritional Sciences, KIng's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Linda V Sinclair
- Cell Signalling and Immunology, University of Dundee, Dundee, UK
| | - Sarah H Forde
- Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
| | - Cormac McCarthy
- Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
| | - Timothy S C Hinks
- Respiratory Medicine Unit, Nuffield Department of Medicine and National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, and the NIHR Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, UK
| | - Clare M Lloyd
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Adam J Byrne
- National Heart and Lung Institute, Imperial College London, London, UK; Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland.
| |
Collapse
|
5
|
Lin L, Wu Z, Zhong A, Luo H, Xu W, Luo W. Causal effects of genetically determined blood metabolites on asthma: a bidirectional Mendelian randomization study. J Asthma 2024; 61:1727-1737. [PMID: 39087774 DOI: 10.1080/02770903.2024.2380515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024]
Abstract
OBJECTIVE The observational association between blood metabolites and asthma has been extensively studied. However, it is still unclear whether this association is causal. In this study, we aimed to investigate the causal relationship between blood metabolites and asthma using a bidirectional Mendelian randomization (MR) analysis. Additionally, we aimed to explore the potential mechanisms underlying this relationship. METHODS The study design involved the use of genetic instruments as instrumental variables (IVs) to fulfill the assumptions of MR analysis. The data on 1,091 metabolites and 309 metabolite ratios were obtained from the Canadian Longitudinal Study on Aging (CLSA), while the data on asthma were obtained from the Integrative Epidemiology Unit (IEU) Open GWAS Project. Utilizing the inverse variance-weighted (IVW) method as the primary MR analysis approach, sensitivity tests were conducted to assess the reliability of the findings, which involved employing Cochran's Q and the MR-Egger intercept. Furthermore, Bayesian weighted MR was used to further test the robustness of the results. Additionally, pathway analysis was conducted to explore the metabolic explanations underlying asthma. RESULT In our study, a comprehensive MR Analysis identified 10 metabolites and 6 metabolite ratios significantly associated with the development of asthma (FDR < 0.05). The metabolites included glycerophosphocholines(GPCs), glycerophosphoethanolamines(GPEs), and an unknown metabolite. Of these, 1-arachidonoyl-GPC, 1-myristoyl-2-arachidonoyl-GPC, 1-palmitoyl-2-arachidonoyl-GPC, and 1-(1-enyl-palmitoyl)-2-arachidonoyl-GPC were associated with an increased risk of asthma, whereas 1,2-dilinoleoyl-GPC, 1-palmitoyl-2-linoleoyl-GPC, 1,2-dilinoleoyl-GPE, 1 - oleoyl - 2 - linoleoyl - GPE, 1-palmitoyl-2-linoleoyl-GPE, and X-21470 were found to have a protective effect. No heterogeneity and pleiotropy were observed in the significant metabolites (p > 0.05), and each metabolite exhibited a consistent effect direction across all five methods. BWMR analysis results confirmed the significance and direction of effects across exposures, except for Cholesterol to linoleoyl-arachidonoyl-glycerol ratio(p = 0.673). Pathway analysis suggests that glycerophospholipid metabolism may potentially be a mechanism underlying the development of asthma. CONCLUSION Our MR findings suggest that the identified metabolites and pathways can serve as biomarkers for clinical asthma screening and prevention, while also providing new insights for future mechanistic exploration and drug target selection.
Collapse
Affiliation(s)
- Liyu Lin
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zilun Wu
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Anqi Zhong
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haocheng Luo
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenjie Xu
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wen Luo
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
6
|
Feng M, Qiao L, Yu Q, Liu M, Zhang J, Wen S, Li X, Teng V, Yan L, Zhang C, Li S, Guo Y, Lu P. Blood chromium and lung function among Chinese young adults: A comprehensive analysis based on epidemiology and metabolomics. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 281:116594. [PMID: 38941662 DOI: 10.1016/j.ecoenv.2024.116594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/30/2024]
Abstract
Chromium (Cr) exposure is associated with various respiratory system diseases, but there are limited studies investigating its impact on lung function in young adults. The Cr exposure-related metabolomic changes are not well elucidated. This study recruited 608 students from a university in Shandong Province, China in 2019. We used cohort design fitted with linear mixed-effects models to assess the association between blood Cr concentration and lung function. In addition, we performed metabolomic and lipidomic analyses of baseline serum samples (N = 582) using liquid chromatography-mass spectrometry. Two-step statistical analysis (analysis of variance and mixed-linear effect model) was used to evaluate the effect of blood Cr exposure on metabolites. We found that blood Cr was associated with decreased lung function in young adults. Each 2-fold increase in blood Cr concentrations was significantly associated with decreased FEV1 and FVC by 35.26 mL (95 % CI: -60.75, -9.78) and 38.56 mL (95 % CI: -66.60, -10.51), respectively. In the metabolomics analysis, blood Cr exposure was significantly associated with 14 key metabolites. The changed metabolites were mainly enriched in six pathways including lipid metabolism, amino acid metabolism, and cofactor vitamin metabolism. Blood Cr may affect lung function through oxidative stress and inflammation related pathways.
Collapse
Affiliation(s)
- Mingyu Feng
- Binzhou Medical University, Yantai, Shandong, China
| | - Lingyan Qiao
- Binzhou Medical University, Yantai, Shandong, China
| | - Qingxia Yu
- Binzhou Medical University, Yantai, Shandong, China
| | - Meiling Liu
- YanTaiShan Hospital, YanTai, Shandong, China
| | - Jia Zhang
- Binzhou Medical University, Yantai, Shandong, China
| | - Shuo Wen
- Binzhou Medical University, Yantai, Shandong, China
| | - Xinyuan Li
- Binzhou Medical University, Yantai, Shandong, China
| | - Victor Teng
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Victoria, Melbourne, Australia
| | - Lailai Yan
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, China
| | | | - Shanshan Li
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Victoria, Melbourne, Australia
| | - Yuming Guo
- Binzhou Medical University, Yantai, Shandong, China; Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Victoria, Melbourne, Australia.
| | - Peng Lu
- Binzhou Medical University, Yantai, Shandong, China.
| |
Collapse
|
7
|
Delgado Dolset MI, Pablo-Torres C, Contreras N, Couto-Rodríguez A, Escolar-Peña A, Graña-Castro O, Izquierdo E, López-Rodríguez JC, Macías-Camero A, Pérez-Gordo M, Villaseñor A, Zubeldia-Varela E, Barber D, Escribese MM. Severe Allergy as a Chronic Inflammatory Condition From a Systems Biology Perspective. Clin Exp Allergy 2024; 54:550-584. [PMID: 38938054 DOI: 10.1111/cea.14517] [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: 01/17/2024] [Revised: 05/14/2024] [Accepted: 05/26/2024] [Indexed: 06/29/2024]
Abstract
Persistent and unresolved inflammation is a common underlying factor observed in several and seemingly unrelated human diseases, including cardiovascular and neurodegenerative diseases. Particularly, in atopic conditions, acute inflammatory responses such as those triggered by insect venom, food or drug allergies possess also a life-threatening potential. However, respiratory allergies predominantly exhibit late immune responses associated with chronic inflammation, that can eventually progress into a severe phenotype displaying similar features as those observed in other chronic inflammatory diseases, as is the case of uncontrolled severe asthma. This review aims to explore the different facets and systems involved in chronic allergic inflammation, including processes such as tissue remodelling and immune cell dysregulation, as well as genetic, metabolic and microbiota alterations, which are common to other inflammatory conditions. Our goal here was to deepen on the understanding of an entangled disease as is chronic allergic inflammation and expose potential avenues for the development of better diagnostic and intervention strategies.
Collapse
Affiliation(s)
- M I Delgado Dolset
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - C Pablo-Torres
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - N Contreras
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Couto-Rodríguez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Escolar-Peña
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - O Graña-Castro
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - E Izquierdo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - J C López-Rodríguez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Macías-Camero
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - M Pérez-Gordo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Villaseñor
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - E Zubeldia-Varela
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - D Barber
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - M M Escribese
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| |
Collapse
|
8
|
Wang L, Lv Z. Causal associations among gut microbiota, 1400 plasma metabolites, and asthma: a two-sample Mendelian randomization study. Front Mol Biosci 2024; 11:1370919. [PMID: 39104371 PMCID: PMC11298384 DOI: 10.3389/fmolb.2024.1370919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/21/2024] [Indexed: 08/07/2024] Open
Abstract
Background Emerging evidence indicates a correlation between imbalances in intestinal microbiota and changes in plasma metabolites in the progression of asthma. However, the causal link between these factors remains unclear. Methods A two-sample Mendelian randomization (MR) study was employed to evaluate the potential causal connection between gut microbiota, plasma metabolites, and asthma susceptibility. Gut microbiota data from expansive genome-wide genotype studies and 16S fecal microbiome datasets were examined by the MiBioGen Alliance. Asthma data were procured from the FinnGen biobank analysis, while comprehensive Genome-Wide Association Studies (GWAS) summary statistics for plasma metabolites were derived from the NHGRI-EBI GWAS Catalog. Fluctuations in intestinal flora and plasma metabolites in asthma patients were evaluated using the weighted mode method. Additionally, pleiotropic and heterogeneity analyses were performed to ascertain the reliability of the findings. Results Upon examining the gut microbiota through MR with the IVW method, alongside tests for heterogeneity and pleiotropy, findings reveal a negative association between the abundance of the Christensenellaceae R.7 group and asthma risk. In contrast, the Bifidobacterium and Prevotella 7 genera exhibit a positive association with asthma risk, indicating they may be potential risk factors (p < 0.05). Furthermore, MR analysis of 1,400 metabolites employing Weighted median, IVW, and Weighted mode methods resulted in p-values below 0.05. Subsequent tests for pleiotropy and heterogeneity showed that the levels of 3,5-dichloro-2,6-dihydroxybenzoic acid have a negative correlation with asthma, whereas the phenylalanine to phosphate ratio has a positive correlation, suggesting their potential as risk factors for asthma (p < 0.05). Conclusion The current Mendelian randomization study provides evidence supporting a potential causal link between specific gut microbiota taxa, plasma metabolites, and asthma. These findings offer novel perspectives for future research and the development of treatment and prevention strategies for asthma.
Collapse
Affiliation(s)
- Lizhu Wang
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zhe Lv
- Air Force Medical University Tangdu Hospital, Xi’an, China
| |
Collapse
|
9
|
Sasaki M, Suaini NHA, Afghani J, Heye KN, O'Mahony L, Venter C, Lauener R, Frei R, Roduit C. Systematic review of the association between short-chain fatty acids and allergic diseases. Allergy 2024; 79:1789-1811. [PMID: 38391245 DOI: 10.1111/all.16065] [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: 09/26/2023] [Revised: 01/23/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024]
Abstract
We performed a systematic review to investigate the current evidence on the association between allergic diseases and short chain fatty acids (SCFAs), which are microbially produced and suggested as one mechanism on how gut microbiome affects the risk of allergic diseases. Medline, Embase and Web of Science were searched from data inception until September 2022. We identified 37 papers, of which 17 investigated prenatal or early childhood SCFAs and the development of allergic diseases in childhood, and 20 assessed SCFAs in patients with pre-existing allergic diseases. Study design, study populations, outcome definition, analysis method and reporting of the results varied between papers. Overall, there was some evidence showing that the three main SCFAs (acetate, propionate and butyrate) in the first few years of life had a protective effect against allergic diseases, especially for atopic dermatitis, wheeze or asthma and IgE-mediated food allergy in childhood. The association between each SCFA and allergic disease appeared to be different by disease and the age of assessment. Further research that can determine the potentially timing specific effect of each SCFA will be useful to investigate how SCFAs can be used in treatment or in prevention against allergic diseases.
Collapse
Affiliation(s)
- Mari Sasaki
- University Children's Hospital Zürich, Zürich, Switzerland
- Division of Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, University of Bern, Bern, Switzerland
| | - Noor H A Suaini
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jamie Afghani
- Environmental Medicine Faculty of Medicine, University of Augsburg, Augsburg, Germany
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
- Institute of Environmental Medicine, Environmental Health Centre, Helmholtz Munich - German Research Centre for Environmental Health (GmbH), Neuherberg, Germany
| | - Kristina N Heye
- Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
| | - Liam O'Mahony
- Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland
- APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, Ireland
- School of Microbiology, University College Cork, National University of Ireland, Cork, Ireland
| | - Carina Venter
- Pediatric Allergy and Immunology, University of Colorado/Childrens Hospital Colorado, Aurora, Colorado, USA
| | - Roger Lauener
- Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
- Christine Kühne Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Remo Frei
- Division of Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, University of Bern, Bern, Switzerland
- Christine Kühne Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Caroline Roduit
- University Children's Hospital Zürich, Zürich, Switzerland
- Division of Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, University of Bern, Bern, Switzerland
- Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
- Christine Kühne Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| |
Collapse
|
10
|
Shi J, Lin Y, Jiang Y, Qiu G, Jian F, Lin W, Zhang S. Dietary choline intake and its association with asthma: A study based on the National Health and Nutrition Examination Survey database. Clin Transl Allergy 2024; 14:e12359. [PMID: 38860615 PMCID: PMC11165556 DOI: 10.1002/clt2.12359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/11/2024] [Accepted: 04/25/2024] [Indexed: 06/12/2024] Open
Abstract
OBJECTIVE This work endeavored to examine the correlation between dietary choline intake and the odds of asthma, utilizing data from the National Health and Nutrition Examination Survey (NHANES). METHODS Aggregated data from seven cycles (2005-2018) in the NHANES database were utilized. The independent variable was dietary choline intake, and the dependent variable was asthma. The weighted logistic regression method was used to construct a model reflecting the relationship between these two factors. This work employed stratified analysis without adjusting for confounding factors and subgroup analysis with adjusted confounding factors to mine the association between dietary choline intake and asthma. Additionally, restricted cubic spline analysis examined nonlinear associations of the two in age subgroups. RESULTS Forty five thousand and seven hundreds ninety seven samples were included here. The model indicating the relationship between dietary choline intake and asthma was constructed (OR: 0.86, 95% CI: 0.79-0.93, p < 0.001). Stratified analysis indicated that the interaction terms of age (p < 0.001) and body mass index (BMI) (p = 0.002) with dietary choline intake significantly influenced the relationship model. In the adjusted models, accounting for demographic characteristics, poverty impact ratio, BMI, exposure to environmental tobacco smoke, and total energy intake, an increase in dietary choline intake significantly reduced the odds of asthma (OR: 0.79, 95% CI: 0.72-0.88, p < 0.001). Subgroup analyses based on age and BMI revealed a significant negative correlation between dietary choline intake and the odds of asthma in the adult population (OR: 0.76, 95% CI: 0.67-0.86, p < 0.001), as well as in individuals with a BMI between 25 and 30 kg/m2 (OR: 0.79, 95% CI: 0.63-0.99, p = 0.042), and those with a BMI >30 kg/m2 (OR: 0.73, 95% CI: 0.60-0.89, p = 0.002). CONCLUSION Dietary choline intake was significantly inversely correlated with asthma prevalence, especially in adults and overweight/obese individuals, suggesting that increasing choline intake may reduce asthma risk. Further research is needed to explore this relationship and provide tailored dietary recommendations for different age and BMI groups to enhance asthma prevention and management.
Collapse
Affiliation(s)
- Jiaqiang Shi
- Department of PediatricsLongyan First Hospital of Fujian Medical UniversityLongyanFujian ProvinceChina
| | - Yuming Lin
- Department of PediatricsLongyan First Hospital of Fujian Medical UniversityLongyanFujian ProvinceChina
| | - Yingxiu Jiang
- Minxi Vocational College (Fujian)LongyanFujian ProvinceChina
| | - Guoguo Qiu
- Department of PediatricsLongyan First Hospital of Fujian Medical UniversityLongyanFujian ProvinceChina
| | - Fanghua Jian
- Department of PediatricsLongyan First Hospital of Fujian Medical UniversityLongyanFujian ProvinceChina
| | - Wei Lin
- Department of PediatricsLongyan First Hospital of Fujian Medical UniversityLongyanFujian ProvinceChina
| | - Shihao Zhang
- Department of Respiratory and Critical Care MedicineGanzhou People's HospitalZhangzhouJiangxi ProvinceChina
| |
Collapse
|
11
|
Zhu T, Ma Y, Wang J, Xiong W, Mao R, Cui B, Min Z, Song Y, Chen Z. Serum Metabolomics Reveals Metabolomic Profile and Potential Biomarkers in Asthma. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:235-252. [PMID: 38910282 PMCID: PMC11199150 DOI: 10.4168/aair.2024.16.3.235] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 11/05/2023] [Accepted: 01/27/2024] [Indexed: 06/25/2024]
Abstract
PURPOSE Asthma is a highly heterogeneous disease. Metabolomics plays a pivotal role in the pathogenesis and development of asthma. The main aims of our study were to explore the underlying mechanism of asthma and to identify novel biomarkers through metabolomics approach. METHODS Serum samples from 102 asthmatic patients and 18 healthy controls were collected and analyzed using liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) system. Multivariate analysis and weighted gene co-expression network analysis (WGCNA) were performed to explore asthma-associated metabolomics profile and metabolites. The Kyoto Encyclopedia of Genes and Genomes (KEGG) was used for pathway enrichment analysis. Subsequently, 2 selected serum hub metabolites, myristoleic acid and dodecanoylcarnitine, were replicated in a validation cohort using ultra-high performance LC-MS/MS system (UHPLC-MS/MS). RESULTS Distinct metabolomics profile of asthma was revealed by multivariate analysis. Then, 116 overlapped asthma-associated metabolites between multivariate analysis and WGCNA, including 12 hub metabolites, were identified. Clinical features-associated hub metabolites were also identified by WGCNA. Among 116 asthma-associated metabolites, Sphingolipid metabolism and valine, leucine and isoleucine biosynthesis were revealed by KEGG analysis. Furthermore, serum myristoleic acid and dodecanoylcarnitine were significantly higher in asthmatic patients than in healthy controls in validation cohort. Additionally, serum myristoleic acid and dodecanoylcarnitine demonstrated high sensitivities and specificities in predicting asthma. CONCLUSIONS Collectively, asthmatic patients showed a unique serum metabolome. Sphingolipid metabolism and valine, leucine and isoleucine biosynthesis were involved in the pathogenesis of asthma. Furthermore, our results suggest the promising values of serum myristoleic acid and dodecanoylcarnitine for asthma diagnosis in adults.
Collapse
Affiliation(s)
- Tao Zhu
- Department of Respiratory Medicine and Critical Care Medicine, and Preclinical Research Center, Suining Central Hospital, Suining, China
| | - Yuan Ma
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai Institute of Respiratory Disease, Shanghai, China
| | - Jiajia Wang
- Rheumatology Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Xiong
- Department of Respiratory Medicine and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ruolin Mao
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai Institute of Respiratory Disease, Shanghai, China
| | - Bo Cui
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai Institute of Respiratory Disease, Shanghai, China
| | - Zhihui Min
- Research Center of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuanlin Song
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai Institute of Respiratory Disease, Shanghai, China.
| | - Zhihong Chen
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai Institute of Respiratory Disease, Shanghai, China.
| |
Collapse
|
12
|
Roth‐Walter F, Adcock IM, Benito‐Villalvilla C, Bianchini R, Bjermer L, Caramori G, Cari L, Chung KF, Diamant Z, Eguiluz‐Gracia I, Knol EF, Jesenak M, Levi‐Schaffer F, Nocentini G, O'Mahony L, Palomares O, Redegeld F, Sokolowska M, Van Esch BCAM, Stellato C. Metabolic pathways in immune senescence and inflammaging: Novel therapeutic strategy for chronic inflammatory lung diseases. An EAACI position paper from the Task Force for Immunopharmacology. Allergy 2024; 79:1089-1122. [PMID: 38108546 PMCID: PMC11497319 DOI: 10.1111/all.15977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
The accumulation of senescent cells drives inflammaging and increases morbidity of chronic inflammatory lung diseases. Immune responses are built upon dynamic changes in cell metabolism that supply energy and substrates for cell proliferation, differentiation, and activation. Metabolic changes imposed by environmental stress and inflammation on immune cells and tissue microenvironment are thus chiefly involved in the pathophysiology of allergic and other immune-driven diseases. Altered cell metabolism is also a hallmark of cell senescence, a condition characterized by loss of proliferative activity in cells that remain metabolically active. Accelerated senescence can be triggered by acute or chronic stress and inflammatory responses. In contrast, replicative senescence occurs as part of the physiological aging process and has protective roles in cancer surveillance and wound healing. Importantly, cell senescence can also change or hamper response to diverse therapeutic treatments. Understanding the metabolic pathways of senescence in immune and structural cells is therefore critical to detect, prevent, or revert detrimental aspects of senescence-related immunopathology, by developing specific diagnostics and targeted therapies. In this paper, we review the main changes and metabolic alterations occurring in senescent immune cells (macrophages, B cells, T cells). Subsequently, we present the metabolic footprints described in translational studies in patients with chronic asthma and chronic obstructive pulmonary disease (COPD), and review the ongoing preclinical studies and clinical trials of therapeutic approaches aiming at targeting metabolic pathways to antagonize pathological senescence. Because this is a recently emerging field in allergy and clinical immunology, a better understanding of the metabolic profile of the complex landscape of cell senescence is needed. The progress achieved so far is already providing opportunities for new therapies, as well as for strategies aimed at disease prevention and supporting healthy aging.
Collapse
Affiliation(s)
- F. Roth‐Walter
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine ViennaMedical University Vienna and University ViennaViennaAustria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - I. M. Adcock
- Molecular Cell Biology Group, National Heart & Lung InstituteImperial College LondonLondonUK
| | - C. Benito‐Villalvilla
- Department of Biochemistry and Molecular Biology, School of ChemistryComplutense University of MadridMadridSpain
| | - R. Bianchini
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine ViennaMedical University Vienna and University ViennaViennaAustria
| | - L. Bjermer
- Department of Respiratory Medicine and Allergology, Lung and Allergy research, Allergy, Asthma and COPD Competence CenterLund UniversityLundSweden
| | - G. Caramori
- Department of Medicine and SurgeryUniversity of ParmaPneumologiaItaly
| | - L. Cari
- Department of Medicine, Section of PharmacologyUniversity of PerugiaPerugiaItaly
| | - K. F. Chung
- Experimental Studies Medicine at National Heart & Lung InstituteImperial College London & Royal Brompton & Harefield HospitalLondonUK
| | - Z. Diamant
- Department of Respiratory Medicine and Allergology, Institute for Clinical ScienceSkane University HospitalLundSweden
- Department of Respiratory Medicine, First Faculty of MedicineCharles University and Thomayer HospitalPragueCzech Republic
- Department of Clinical Pharmacy & PharmacologyUniversity Groningen, University Medical Center Groningen and QPS‐NLGroningenThe Netherlands
| | - I. Eguiluz‐Gracia
- Allergy UnitHospital Regional Universitario de Málaga‐Instituto de Investigación Biomédica de Málaga (IBIMA)‐ARADyALMálagaSpain
| | - E. F. Knol
- Departments of Center of Translational Immunology and Dermatology/AllergologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - M. Jesenak
- Department of Paediatrics, Department of Pulmonology and Phthisiology, Comenius University in Bratislava, Jessenius Faculty of Medicine in MartinUniversity Teaching HospitalMartinSlovakia
| | - F. Levi‐Schaffer
- Institute for Drug Research, Pharmacology Unit, Faculty of MedicineThe Hebrew University of JerusalemJerusalemIsrael
| | - G. Nocentini
- Department of Medicine, Section of PharmacologyUniversity of PerugiaPerugiaItaly
| | - L. O'Mahony
- APC Microbiome IrelandUniversity College CorkCorkIreland
- Department of MedicineUniversity College CorkCorkIreland
- School of MicrobiologyUniversity College CorkCorkIreland
| | - O. Palomares
- Department of Biochemistry and Molecular Biology, School of ChemistryComplutense University of MadridMadridSpain
| | - F. Redegeld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of ScienceUtrecht UniversityUtrechtThe Netherlands
| | - M. Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZürichDavosSwitzerland
- Christine Kühne – Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| | - B. C. A. M. Van Esch
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of ScienceUtrecht UniversityUtrechtThe Netherlands
| | - C. Stellato
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”University of SalernoSalernoItaly
| |
Collapse
|
13
|
Barman M, Gio-Batta M, Andrieux L, Stråvik M, Saalman R, Fristedt R, Rabe H, Sandin A, Wold AE, Sandberg AS. Short-chain fatty acids (SCFA) in infants' plasma and corresponding mother's milk and plasma in relation to subsequent sensitisation and atopic disease. EBioMedicine 2024; 101:104999. [PMID: 38340558 PMCID: PMC10869761 DOI: 10.1016/j.ebiom.2024.104999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Short-chain fatty acids (SCFAs) in intestinal contents may influence immune function, while less is known about SCFAs in blood plasma. The aims were to investigate the relation between infants' and maternal plasma SCFAs, as well as SCFAs in mother's milk, and relate SCFA concentrations in infant plasma to subsequent sensitisation and atopic disease. METHODS Infant plasma (N = 148) and corresponding mother's milk and plasma were collected four months postpartum. Nine SCFA (formic, acetic, propionic, isobutyric, butyric, succinic, valeric, isovaleric, and caproic acid) were analysed by UPLC-MS. At 12 months of age, atopic disease was diagnosed by a pediatric allergologist, and sensitisation was measured by skin prick test. All families participated in the Swedish birth cohort NICE (Nutritional impact on Immunological maturation during Childhood in relation to the Environment). FINDINGS Infants with sensitisation, atopic eczema, or food allergy had significantly lower concentrations of five, three, and two SCFAs, respectively, in plasma at four months. Logistic regressions models showed significant negative associations between formic, succinic, and caproic acid and sensitisation [ORadj (95% CI) per SD: 0.41 (0.19-0.91); 0.19 (0.05-0.75); 0.25 (0.09-0.66)], and between acetic acid and atopic eczema [0.42 (0.18-0.95)], after adjusting for maternal allergy. Infants' and maternal plasma SCFA concentrations correlated strongly, while milk SCFA concentrations were unrelated to both. Butyric and caproic acid concentrations were enriched around 100-fold, and iso-butyric and valeric acid around 3-5-fold in mother's milk, while other SCFAs were less prevalent in milk than in plasma. INTERPRETATION Butyric and caproic acid might be actively transported into breast milk to meet the needs of the infant, although mechanistic studies are needed to confirm this. The negative associations between certain SCFAs on sensitisation and atopic disease adds to prior evidence regarding their immunoregulatory potential. FUNDING Swedish Research Council (Nr. 2013-3145, 2019-0137 and 2023-02217 to A-S.S.), Swedish Research Council for Health, Working Life and Welfare FORTE, Nr 2018-00485 to A.W.), The Swedish Asthma and Allergy Association's Research Fund (2020-0020 to A.S.).
Collapse
Affiliation(s)
- Malin Barman
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden.
| | - Monica Gio-Batta
- Institute of Biomedicine, Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Léna Andrieux
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden; Département de Biologie, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, 69342 Lyon Cedex 07, France
| | - Mia Stråvik
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Robert Saalman
- Institute of Clinical Sciences, Department of Pediatrics, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Rikard Fristedt
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Hardis Rabe
- Institute of Biomedicine, Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Anna Sandin
- Department of Clinical Science, Pediatrics, Sunderby Research Unit, Umeå University, Umeå 901 87, Sweden
| | - Agnes E Wold
- Institute of Biomedicine, Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Ann-Sofie Sandberg
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden
| |
Collapse
|
14
|
Lejeune S, Kaushik A, Parsons ES, Chinthrajah S, Snyder M, Desai M, Manohar M, Prunicki M, Contrepois K, Gosset P, Deschildre A, Nadeau K. Untargeted metabolomic profiling in children identifies novel pathways in asthma and atopy. J Allergy Clin Immunol 2024; 153:418-434. [PMID: 38344970 DOI: 10.1016/j.jaci.2023.09.040] [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: 05/06/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 02/15/2024]
Abstract
BACKGROUND Asthma and other atopic disorders can present with varying clinical phenotypes marked by differential metabolomic manifestations and enriched biological pathways. OBJECTIVE We sought to identify these unique metabolomic profiles in atopy and asthma. METHODS We analyzed baseline nonfasted plasma samples from a large multisite pediatric population of 470 children aged <13 years from 3 different sites in the United States and France. Atopy positivity (At+) was defined as skin prick test result of ≥3 mm and/or specific IgE ≥ 0.35 IU/mL and/or total IgE ≥ 173 IU/mL. Asthma positivity (As+) was based on physician diagnosis. The cohort was divided into 4 groups of varying combinations of asthma and atopy, and 6 pairwise analyses were conducted to best assess the differential metabolomic profiles between groups. RESULTS Two hundred ten children were classified as At-As-, 42 as At+As-, 74 as At-As+, and 144 as At+As+. Untargeted global metabolomic profiles were generated through ultra-high-performance liquid chromatography-tandem mass spectroscopy. We applied 2 independent machine learning classifiers and short-listed 362 metabolites as discriminant features. Our analysis showed the most diverse metabolomic profile in the At+As+/At-As- comparison, followed by the At-As+/At-As- comparison, indicating that asthma is the most discriminant condition associated with metabolomic changes. At+As+ metabolomic profiles were characterized by higher levels of bile acids, sphingolipids, and phospholipids, and lower levels of polyamine, tryptophan, and gamma-glutamyl amino acids. CONCLUSION The At+As+ phenotype displays a distinct metabolomic profile suggesting underlying mechanisms such as modulation of host-pathogen and gut microbiota interactions, epigenetic changes in T-cell differentiation, and lower antioxidant properties of the airway epithelium.
Collapse
Affiliation(s)
- Stéphanie Lejeune
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; University of Lille, Pediatric Pulmonology and Allergy Department, Hôpital Jeanne de Flandre, CHU Lille, Lille, France; University of Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France.
| | - Abhinav Kaushik
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Department of Environmental Health, T. H. Chan School of Public Health, Harvard University, Boston, Mass
| | - Ella S Parsons
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Sharon Chinthrajah
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Michael Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, Calif
| | - Manisha Desai
- Quantitative Science Unit, Department of Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Monali Manohar
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Mary Prunicki
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Department of Environmental Health, T. H. Chan School of Public Health, Harvard University, Boston, Mass
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, Calif
| | - Philippe Gosset
- University of Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
| | - Antoine Deschildre
- University of Lille, Pediatric Pulmonology and Allergy Department, Hôpital Jeanne de Flandre, CHU Lille, Lille, France; University of Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
| | - Kari Nadeau
- Department of Environmental Health, T. H. Chan School of Public Health, Harvard University, Boston, Mass
| |
Collapse
|
15
|
Barosova R, Baranovicova E, Hanusrichterova J, Mokra D. Metabolomics in Animal Models of Bronchial Asthma and Its Translational Importance for Clinics. Int J Mol Sci 2023; 25:459. [PMID: 38203630 PMCID: PMC10779398 DOI: 10.3390/ijms25010459] [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: 11/15/2023] [Revised: 12/17/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Bronchial asthma is an extremely heterogenous chronic respiratory disorder with several distinct endotypes and phenotypes. These subtypes differ not only in the pathophysiological changes and/or clinical features but also in their response to the treatment. Therefore, precise diagnostics represent a fundamental condition for effective therapy. In the diagnostic process, metabolomic approaches have been increasingly used, providing detailed information on the metabolic alterations associated with human asthma. Further information is brought by metabolomic analysis of samples obtained from animal models. This article summarizes the current knowledge on metabolomic changes in human and animal studies of asthma and reveals that alterations in lipid metabolism, amino acid metabolism, purine metabolism, glycolysis and the tricarboxylic acid cycle found in the animal studies resemble, to a large extent, the changes found in human patients with asthma. The findings indicate that, despite the limitations of animal modeling in asthma, pre-clinical testing and metabolomic analysis of animal samples may, together with metabolomic analysis of human samples, contribute to a novel way of personalized treatment of asthma patients.
Collapse
Affiliation(s)
- Romana Barosova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (R.B.); (J.H.)
| | - Eva Baranovicova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Juliana Hanusrichterova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (R.B.); (J.H.)
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Daniela Mokra
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (R.B.); (J.H.)
| |
Collapse
|
16
|
Barosova R, Baranovicova E, Adamcakova J, Prso K, Hanusrichterova J, Mokra D. Sex differences in plasma metabolites in a guinea pig model of allergic asthma. Physiol Res 2023; 72:S499-S508. [PMID: 38165754 PMCID: PMC10861256 DOI: 10.33549/physiolres.935218] [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: 03/28/2023] [Accepted: 09/11/2023] [Indexed: 02/01/2024] Open
Abstract
Sex seems to be a contributing factor in the pathogenesis of bronchial asthma. This study aimed to find sex-related differences in metabolome measured by hydrogen-1 nuclear magnetic resonance ((1)H NMR) spectroscopy in healthy and ovalbumin (OVA)-sensitized guinea pigs. Adult male and female animals were divided into controls and OVA-sensitized groups. OVA-sensitization was performed by OVA systemic and inhalational administration within 14 days; on day 15, animals were killed by anesthetic overdose followed by exsanguination. Blood was taken and differential white blood cell count was measured. Left lung was saline-lavaged and differential cell count in the bronchoalveolar lavage fluid (BALF) was measured. After blood centrifugation, plasma was processed for (1)H NMR analysis. Metabolomic data was evaluated by principal component analysis (PCA). Eosinophil counts elevated in the BALF confirming eosinophil-mediated inflammation in OVA-sensitized animals of both sexes. Sex differences for lactate, glucose, and citrate were found in controls, where these parameters were lower in males than in females. In OVA-sensitized males higher glucose and lower pyruvate were found compared to controls. OVA-sensitized females showed lower lactate, glucose, alanine, 3-hydroxy-butyrate, creatine, pyruvate, and succinate concentrations compared to controls. In OVA-sensitized animals, lactate concentration was lower in males. Data from females (healthy and OVA-sensitized) were generally more heterogeneous. Significant sex differences in plasma concentrations of metabolites were found in both healthy and OVA-sensitized animals suggesting that sex may influence the metabolism and may thereby contribute to different clinical picture of asthma in males and females.
Collapse
Affiliation(s)
- R Barosova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic.
| | | | | | | | | | | |
Collapse
|
17
|
Xie QM, Chen N, Song SM, Zhao CC, Ruan Y, Sha JF, Liu Q, Jiang XQ, Fei GH, Wu HM. Itaconate Suppresses the Activation of Mitochondrial NLRP3 Inflammasome and Oxidative Stress in Allergic Airway Inflammation. Antioxidants (Basel) 2023; 12:489. [PMID: 36830047 PMCID: PMC9951851 DOI: 10.3390/antiox12020489] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Itaconate has emerged as a novel anti-inflammatory and antioxidative endogenous metabolite, yet its role in allergic airway inflammation (AAI) and the underlying mechanism remains elusive. Here, the itaconate level in the lung was assessed by High Performance Liquid Chromatography (HPLC), and the effects of the Irg1/itaconate pathway on AAI and alveolar macrophage (AM) immune responses were evaluated using an ovalbumin (OVA)-induced AAI model established by wild type (WT) and Irg1-/- mice, while the mechanism of this process was investigated by metabolomics analysis, mitochondrial/cytosolic protein fractionation and transmission electron microscopy in the lung tissues. The results demonstrated that the Irg1 mRNA/protein expression and itaconate production in the lung were significantly induced by OVA. Itaconate ameliorated while Irg1 deficiency augmented AAI, and this may be attributed to the fact that itaconate suppressed mitochondrial events such as NLRP3 inflammasome activation, oxidative stress and metabolic dysfunction. Furthermore, we identified that the Irg1/itaconate pathway impacted the NLRP3 inflammasome activation and oxidative stress in AMs. Collectively, our findings provide evidence for the first time, supporting the conclusion that in the allergic lung, the itaconate level is markedly increased, which directly regulates AMs' immune responses. We therefore propose that the Irg1/itaconate pathway in AMs is a potential anti-inflammatory and anti-oxidative therapeutic target for AAI.
Collapse
Affiliation(s)
- Qiu-Meng Xie
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei 230022, China
| | - Ning Chen
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei 230022, China
| | - Si-Ming Song
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei 230022, China
| | - Cui-Cui Zhao
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei 230022, China
| | - Ya Ruan
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei 230022, China
| | - Jia-Feng Sha
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei 230022, China
| | - Qian Liu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Department of Respiratory Medicine, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230001, China
| | - Xu-Qin Jiang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Department of Respiratory Medicine, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230001, China
| | - Guang-He Fei
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei 230022, China
| | - Hui-Mei Wu
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road 218, Hefei 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei 230022, China
| |
Collapse
|
18
|
Grueso-Navarro E, Navarro P, Laserna-Mendieta EJ, Lucendo AJ, Arias-González L. Blood-Based Biomarkers for Eosinophilic Esophagitis and Concomitant Atopic Diseases: A Look into the Potential of Extracellular Vesicles. Int J Mol Sci 2023; 24:ijms24043669. [PMID: 36835081 PMCID: PMC9967575 DOI: 10.3390/ijms24043669] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/17/2023] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic, Th2-inflammatory disease of the esophagus that can severely affect food intake. Currently, diagnosis and assessing response to treatment of EoE is highly invasive and requires endoscopy with esophageal biopsies. Finding non-invasive and accurate biomarkers is important for improving patient well-being. Unfortunately, EoE is usually accompanied by other atopies, which make it difficult to identify specific biomarkers. Providing an update of circulating EoE biomarkers and concomitant atopies is therefore timely. This review summarizes the current knowledge in EoE blood biomarkers and two of its most common comorbidities, bronchial asthma (BA) and atopic dermatitis (AD), focusing on dysregulated proteins, metabolites, and RNAs. It also revises the current knowledge on extracellular vesicles (EVs) as non-invasive biomarkers for BA and AD, and concludes with the potential use of EVs as biomarkers in EoE.
Collapse
Affiliation(s)
- Elena Grueso-Navarro
- Department of Gastroenterology, Hospital General de Tomelloso, Tomelloso, 13700 Ciudad Real, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45004 Toledo, Spain
- Correspondence: (E.G.-N.); (A.J.L.)
| | - Pilar Navarro
- Department of Gastroenterology, Hospital General de Tomelloso, Tomelloso, 13700 Ciudad Real, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45004 Toledo, Spain
| | - Emilio J. Laserna-Mendieta
- Department of Gastroenterology, Hospital General de Tomelloso, Tomelloso, 13700 Ciudad Real, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45004 Toledo, Spain
- Laboratory Medicine Department, Hospital Universitario de La Princesa, 28006 Madrid, Spain
- Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006 Madrid, Spain
| | - Alfredo J. Lucendo
- Department of Gastroenterology, Hospital General de Tomelloso, Tomelloso, 13700 Ciudad Real, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45004 Toledo, Spain
- Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28006 Madrid, Spain
- Correspondence: (E.G.-N.); (A.J.L.)
| | - Laura Arias-González
- Department of Gastroenterology, Hospital General de Tomelloso, Tomelloso, 13700 Ciudad Real, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45004 Toledo, Spain
- Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28006 Madrid, Spain
| |
Collapse
|
19
|
Wang CJ, Noble PB, Elliot JG, James AL, Wang KCW. From Beneath the Skin to the Airway Wall: Understanding the Pathological Role of Adipose Tissue in Comorbid Asthma-Obesity. Compr Physiol 2023; 13:4321-4353. [PMID: 36715283 DOI: 10.1002/cphy.c220011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This article provides a contemporary report on the role of adipose tissue in respiratory dysfunction. Adipose tissue is distributed throughout the body, accumulating beneath the skin (subcutaneous), around organs (visceral), and importantly in the context of respiratory disease, has recently been shown to accumulate within the airway wall: "airway-associated adipose tissue." Excessive adipose tissue deposition compromises respiratory function and increases the severity of diseases such as asthma. The mechanisms of respiratory impairment are inflammatory, structural, and mechanical in nature, vary depending on the anatomical site of deposition and adipose tissue subtype, and likely contribute to different phenotypes of comorbid asthma-obesity. An understanding of adipose tissue-driven pathophysiology provides an opportunity for diagnostic advancement and patient-specific treatment. As an exemplar, the potential impact of airway-associated adipose tissue is highlighted, and how this may change the management of a patient with asthma who is also obese. © 2023 American Physiological Society. Compr Physiol 13:4321-4353, 2023.
Collapse
Affiliation(s)
- Carolyn J Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Peter B Noble
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - John G Elliot
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Kimberley C W Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
20
|
Goretzki A, Zimmermann J, Rainer H, Lin YJ, Schülke S. Immune Metabolism in TH2 Responses: New Opportunities to Improve Allergy Treatment - Disease-Specific Findings (Part 1). Curr Allergy Asthma Rep 2023; 23:29-40. [PMID: 36441389 PMCID: PMC9832111 DOI: 10.1007/s11882-022-01057-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE OF REVIEW Recent high-level publications have shown an intricate connection between immune effector function and the metabolic state of the respective cells. In the last years, studies have begun analyzing the metabolic changes associated with allergies. As the first part of a two-article series, this review will briefly summarize the basics of immune metabolism and then focus on the recently published studies on metabolic changes observed in allergic patients. RECENT FINDINGS In the last 3 years, immune-metabolic research in allergology had a clear focus on asthma with some studies also reporting findings in food allergy and atopic dermatitis. Current results suggest asthma to be associated with a shift in cellular metabolism towards increased aerobic glycolysis (Warburg metabolism), while also displaying substantial changes in fatty acid- and amino acid metabolism (depending on investigated patient collective, asthma phenotype, and disease severity). Understanding immune-metabolic changes in allergies will allow us to (I) better understand allergic disease pathology and (II) modulate immune-metabolic pathways to improve allergy treatment.
Collapse
Affiliation(s)
- A. Goretzki
- Vice President’s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, 63225 Langen, Germany
| | - J. Zimmermann
- Vice President’s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, 63225 Langen, Germany
| | - H. Rainer
- Vice President’s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, 63225 Langen, Germany
| | - Y.-J. Lin
- Vice President’s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, 63225 Langen, Germany
| | - Stefan Schülke
- Vice President's Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, 63225, Langen, Germany.
| |
Collapse
|
21
|
Frank BS, Khailova L, Dekermanjian J, Mitchell MB, Morgan GJ, Twite M, Christians U, DiMaria MV, Klawitter J, Davidson JA. Interstage Single Ventricle Heart Disease Infants Show Dysregulation in Multiple Metabolic Pathways: Targeted Metabolomics Analysis. JACC. ADVANCES 2023; 2:100169. [PMID: 36875009 PMCID: PMC9979841 DOI: 10.1016/j.jacadv.2022.100169] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/03/2022] [Accepted: 11/01/2022] [Indexed: 01/29/2023]
Abstract
BACKGROUND Infants with SVHD experience morbidity related to pulmonary vascular inadequacy. Metabolomic analysis involves a systems biology approach to identifying novel biomarkers and pathways in complex diseases. The metabolome of infants with SVHD is not well understood and no prior study has evaluated the relationship between serum metabolite patterns and pulmonary vascular readiness for staged SVHD palliation. OBJECTIVES The purpose of this study was to evaluate the circulating metabolome of interstage infants with single ventricle heart disease (SVHD) and determine whether metabolite levels were associated with pulmonary vascular inadequacy. METHODS This was a prospective cohort study of 52 infants with SVHD undergoing Stage 2 palliation and 48 healthy infants. Targeted metabolomic phenotyping (175 metabolites) was performed by tandem mass spectrometry on SVHD pre-Stage 2, post-Stage 2, and control serum samples. Clinical variables were extracted from the medical record. RESULTS Random forest analysis readily distinguished between cases and controls and preoperative and postoperative samples. Seventy-four of 175 metabolites differed between SVHD and controls. Twenty-seven of 39 metabolic pathways were altered including pentose phosphate and arginine metabolism. Seventy-one metabolites differed in SVHD patients between timepoints. Thirty-three of 39 pathways were altered postoperatively including arginine and tryptophan metabolism. We found trends toward increased preoperative methionine metabolites in patients with higher pulmonary vascular resistance and higher postoperative tryptophan metabolites in patients with greater postoperative hypoxemia. CONCLUSIONS The circulating metabolome of interstage SVHD infants differs significantly from controls and is further disrupted after Stage 2. Several metabolites showed trends toward association with adverse outcomes. Metabolic dysregulation may be an important factor in early SVHD pathobiology.
Collapse
Affiliation(s)
- Benjamin S. Frank
- Section of Cardiology, Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
| | - Ludmila Khailova
- Section of Cardiology, Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
| | - Jonathan Dekermanjian
- Center for Innovative Design and Analysis, University of Colorado, Aurora, Colorado, USA
| | - Max B. Mitchell
- Department of Surgery, University of Colorado, Aurora, Colorado, USA
| | - Gareth J. Morgan
- Section of Cardiology, Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
| | - Mark Twite
- Department of Anesthesiology, University of Colorado, Aurora, Colorado, USA
| | - Uwe Christians
- Department of Anesthesiology, University of Colorado, Aurora, Colorado, USA
| | - Michael V. DiMaria
- Section of Cardiology, Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
| | - Jelena Klawitter
- Department of Anesthesiology, University of Colorado, Aurora, Colorado, USA
| | - Jesse A. Davidson
- Section of Cardiology, Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
22
|
Huang F, Yu J, Lai T, Luo L, Zhang W. The Combination of Bioinformatics Analysis and Untargeted Metabolomics Reveals Potential Biomarkers and Key Metabolic Pathways in Asthma. Metabolites 2022; 13:25. [PMID: 36676950 PMCID: PMC9860906 DOI: 10.3390/metabo13010025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Asthma is a complex chronic airway inflammatory disease that seriously impacts patients' quality of life. As a novel approach to exploring the pathogenesis of diseases, metabolomics provides the potential to identify biomarkers of asthma host susceptibility and elucidate biological pathways. The aim of this study was to screen potential biomarkers and biological pathways so as to provide possible pharmacological therapeutic targets for asthma. In the present study, we merged the differentially expressed genes (DEGs) of asthma in the GEO database with the metabolic genes obtained by Genecard for bioinformatics analysis and successfully screened out the metabolism-related hub genes (HIF1A, OCRL, NNMT, and PER1). Then, untargeted metabolic techniques were utilized to reveal HDM-induced metabolite alterations in 16HBE cells. A total of 45 significant differential metabolites and 5 differential metabolic pathways between the control group and HDM group were identified based on the OPLS-DA model. Finally, three key metabolic pathways, including glycerophospholipid metabolism, galactose metabolism, and alanine, aspartate, and glutamate metabolism, were screened through the integrated analysis of bioinformatics data and untargeted metabolomics data. Taken together, these findings provide valuable insights into the pathophysiology and targeted therapy of asthma and lay a foundation for further research.
Collapse
Affiliation(s)
- Fangfang Huang
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Graduate School, Guangdong Medical University, Zhanjiang 524023, China
| | - Jinjin Yu
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, China
| | - Tianwen Lai
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang 524023, China
| | - Weizhen Zhang
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
23
|
Delgado Dolset MI, Obeso D, Rodriguez-Coira J, Villaseñor A, González Cuervo H, Arjona A, Barbas C, Barber D, Carrillo T, Escribese MM. Contribution of allergy in the acquisition of uncontrolled severe asthma. Front Med (Lausanne) 2022; 9:1009324. [PMID: 36213665 PMCID: PMC9532527 DOI: 10.3389/fmed.2022.1009324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/22/2022] [Indexed: 12/02/2022] Open
Abstract
Asthma is a multifactorial, heterogeneous disease that has a challenging management. It can be divided in non-allergic and allergic (usually associated with house dust mites (HDM) sensitization). There are several treatments options for asthma (corticosteroids, bronchodilators, antileukotrienes, anticholinergics,…); however, there is a subset of patients that do not respond to any of the treatments, who can display either a T2 or a non-T2 phenotype. A deeper understanding of the differential mechanisms underlying each phenotype will help to decipher the contribution of allergy to the acquisition of this uncontrolled severe phenotype. Here, we aim to elucidate the biological pathways associated to allergy in the uncontrolled severe asthmatic phenotype. To do so, twenty-three severe uncontrolled asthmatic patients both with and without HDM-allergy were recruited from Hospital Universitario de Gran Canaria Dr. Negrin. A metabolomic fingerprint was obtained through liquid chromatography coupled to mass spectrometry, and identified metabolites were associated with their pathways. 9/23 patients had uncontrolled HDM-allergic asthma (UCA), whereas 14 had uncontrolled, non-allergic asthma (UCNA). 7/14 (50%) of the UCNA patients had Aspirin Exacerbated Respiratory Disease. There were no significant differences regarding gender or body mass index; but there were significant differences in age and onset age, which were higher in UCNA patients; and in total IgE, which was higher in UCA. The metabolic fingerprint revealed that 103 features were significantly different between UCNA and UCA (p < 0.05), with 97 being increased in UCA and 6 being decreased. We identified lysophosphocholines (LPC) 18:2, 18:3 and 20:4 (increased in UCA patients); and deoxycholic acid and palmitoleoylcarnitine (decreased in UCA). These metabolites were related with a higher activation of phospholipase A2 (PLA2) and other phospholipid metabolism pathways. Our results show that allergy induces the activation of specific inflammatory pathways, such as the PLA2 pathway, which supports its role in the development of an uncontrolled asthma phenotype. There are also clinical differences, such as higher levels of IgE and earlier onset ages for the allergic asthmatic group, as expected. These results provide evidences to better understand the contribution of allergy to the establishment of a severe uncontrolled phenotype.
Collapse
Affiliation(s)
- María Isabel Delgado Dolset
- Institute for Applied Molecular Medicine Nemesio Díez, School of Medicine, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - David Obeso
- Institute for Applied Molecular Medicine Nemesio Díez, School of Medicine, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centre of Metabolomics and Bioanalysis (CEMBIO), School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - Juan Rodriguez-Coira
- Institute for Applied Molecular Medicine Nemesio Díez, School of Medicine, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centre of Metabolomics and Bioanalysis (CEMBIO), School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - Alma Villaseñor
- Institute for Applied Molecular Medicine Nemesio Díez, School of Medicine, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centre of Metabolomics and Bioanalysis (CEMBIO), School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - Heleia González Cuervo
- Allergy Service, Hospital Universitario de Gran Canaria Doctor Negrin, Las Palmas de Gran Canaria, Spain
| | - Ana Arjona
- Allergy Service, Hospital Universitario de Gran Canaria Doctor Negrin, Las Palmas de Gran Canaria, Spain
| | - Coral Barbas
- Centre of Metabolomics and Bioanalysis (CEMBIO), School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - Domingo Barber
- Institute for Applied Molecular Medicine Nemesio Díez, School of Medicine, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - Teresa Carrillo
- Allergy Service, Hospital Universitario de Gran Canaria Doctor Negrin, Las Palmas de Gran Canaria, Spain
- Department of Medical and Surgical Sciences, School of Health Sciences, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - María M. Escribese
- Institute for Applied Molecular Medicine Nemesio Díez, School of Medicine, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Department of Basic Medical Sciences, School of Medicine, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- *Correspondence: María M. Escribese
| |
Collapse
|
24
|
Tang X, Rönnberg E, Säfholm J, Thulasingam M, Trauelsen M, Schwartz TW, Wheelock CE, Dahlén S, Nilsson G, Haeggström JZ. Activation of succinate receptor 1 boosts human mast cell reactivity and allergic bronchoconstriction. Allergy 2022; 77:2677-2687. [PMID: 35122266 PMCID: PMC9545225 DOI: 10.1111/all.15245] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/31/2021] [Accepted: 01/23/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND SUCNR1 is a sensor of extracellular succinate, a Krebs cycle intermediate generated in excess during oxidative stress and has been linked to metabolic regulation and inflammation. While mast cells express SUCNR1, its role in mast cell reactivity and allergic conditions such as asthma remains to be elucidated. METHODS Cord blood-derived mast cells and human mast cell line LAD-2 challenged by SUCNR1 ligands were analyzed for the activation and mediator release. Effects on mast cell-dependent bronchoconstriction were assessed in guinea pig trachea and isolated human small bronchi challenged with antigen and anti-IgE, respectively. RESULTS SUCNR1 is abundantly expressed on human mast cells. Challenge with succinate, or the synthetic non-metabolite agonist cis-epoxysuccinate, renders mast cells hypersensitive to IgE-dependent activation, resulting in augmented degranulation and histamine release, de novo biosynthesis of eicosanoids and cytokine secretion. The succinate-potentiated mast cell reactivity was attenuated by SUCNR1 knockdown and selective SUCNR1 antagonists and could be tuned by pharmacologically targeting protein kinase C and extracellular signal-regulated kinase. Both succinate and cis-epoxysuccinate dose-dependently potentiated antigen-induced contraction in a mast cell-dependent guinea pig airway model, associated with increased generation of cysteinyl-leukotrienes and histamine in trachea. Similarly, cis-epoxysuccinate aggravated IgE-receptor-induced contraction of human bronchi, which was blocked by SUCNR1 antagonism. CONCLUSION SUCNR1 amplifies IgE-receptor-induced mast cell activation and allergic bronchoconstriction, suggesting a role for this pathway in aggravation of allergic asthma, thus linking metabolic perturbations to mast cell-dependent inflammation.
Collapse
Affiliation(s)
- Xiao Tang
- Division of Physiological Chemistry IIDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Elin Rönnberg
- Division of Immunology and AllergyDepartment of Medicine SolnaKarolinska Institutet, and Karolinska University HospitalSolnaSweden
| | - Jesper Säfholm
- Unit of Experimental Asthma and Allergy ResearchInstitute of Environmental MedicineKarolinska InstitutetStockholmSweden
| | - Madhuranayaki Thulasingam
- Division of Physiological Chemistry IIDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Mette Trauelsen
- Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Thue W. Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Craig E. Wheelock
- Division of Physiological Chemistry IIDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Sven‐Erik Dahlén
- Unit of Experimental Asthma and Allergy ResearchInstitute of Environmental MedicineKarolinska InstitutetStockholmSweden,Department of Respiratory MedicineKarolinska University Hospital HuddingeStockholmSweden
| | - Gunnar Nilsson
- Division of Immunology and AllergyDepartment of Medicine SolnaKarolinska Institutet, and Karolinska University HospitalSolnaSweden,Department of Medical SciencesUppsala UniversityUppsalaSweden
| | - Jesper Z. Haeggström
- Division of Physiological Chemistry IIDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| |
Collapse
|
25
|
Machine-learning algorithms for asthma, COPD, and lung cancer risk assessment using circulating microbial extracellular vesicle data and their application to assess dietary effects. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1586-1595. [PMID: 36180580 PMCID: PMC9534896 DOI: 10.1038/s12276-022-00846-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/17/2022] [Accepted: 07/12/2022] [Indexed: 11/08/2022]
Abstract
Although mounting evidence suggests that the microbiome has a tremendous influence on intractable disease, the relationship between circulating microbial extracellular vesicles (EVs) and respiratory disease remains unexplored. Here, we developed predictive diagnostic models for COPD, asthma, and lung cancer by applying machine learning to microbial EV metagenomes isolated from patient serum and coded by their accumulated taxonomic hierarchy. All models demonstrated high predictive strength with mean AUC values ranging from 0.93 to 0.99 with various important features at the genus and phylum levels. Application of the clinical models in mice showed that various foods reduced high-fat diet-associated asthma and lung cancer risk, while COPD was minimally affected. In conclusion, this study offers a novel methodology for respiratory disease prediction and highlights the utility of serum microbial EVs as data-rich features for noninvasive diagnosis. Artificial intelligence (AI) has enabled researchers to intercept microbial messages bearing clinically useful information about of a variety of respiratory disorders. The organisms that comprise our microbiome communicate via the release of tiny, biomolecule-laden membrane bubbles called ‘extracellular vesicles’ (EVs) into the bloodstream. EVs are also influenced by human disease. South Korean researchers led by Yoon-Keun Kim of MD Healthcare, Seoul, and Young-Koo Jee of Dankook University College of Medicine, Cheonan, have used an AI algorithm to assemble EV-based profiles that can discriminate between healthy people and those with conditions like asthma or lung cancer. Their analysis of 1727 patient serum samples revealed microbial signatures that enabled accurate diagnosis of several respiratory disorders. Preliminary experiments in mice suggest that certain dietary changes could help shift the microbiome of high-risk individuals towards a healthier profile.
Collapse
|
26
|
Daley-Yates P, Keppler B, Brealey N, Shabbir S, Singh D, Barnes N. Inhaled glucocorticoid-induced metabolome changes in asthma. Eur J Endocrinol 2022; 187:413-427. [PMID: 35900313 PMCID: PMC9346266 DOI: 10.1530/eje-21-0912] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 07/04/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The aim of this study was toidentify dose-related systemic effects of inhaled glucocorticoids (GCs) on the global metabolome. DESIGN AND METHODS Metabolomics/lipidomic analysis from plasma was obtained from 54 subjects receiving weekly escalating doses (µg/day) of fluticasone furoate (FF; 25, 100, 200, 400 and 800), fluticasone propionate (FP; 50, 200, 500, 1000 and 2000), budesonide (BUD; 100, 400, 800, 1600 and 3200) or placebo. Samples (pre- and post-dose) were analysed using ultrahigh-performance liquid chromatography-tandem mass spectroscopy and liquid chromatography-mass spectrometry. Ions were matched to library standards for identification and quantification. Statistical analysis involved repeated measures ANOVA, cross-over model, random forest and principal component analysis using log-transformed data. RESULTS Quantifiable metabolites (1971) had few significant changes (% increases/decreases; P < 0.05) vs placebo: FF 1.34 (0.42/0.92), FP 1.95 (0.41/1.54) and BUD 2.05 (0.60/1.45). Therapeutic doses had fewer changes: FF 0.96 (0.36/0.61), FP 1.66 (0.44/1.22) and BUD 1.45 (0.56/0.90). At highest/supratherapeutic doses, changes were qualitatively similar: reduced adrenal steroids, particularly glucuronide metabolites of cortisol and cortisone and pregnenolone metabolite DHEA-S; increased amino acids and glycolytic intermediates; decreased fatty acid β-oxidation and branched-chain amino acids. Notable qualitative differences were lowered dopamine metabolites (BUD) and secondary bile acid profiles (BUD/FF), suggesting CNS and gut microbiome effects. CONCLUSIONS Dose-dependent metabolomic changes occurred with inhaled GCs but were seen predominately at highest/supratherapeutic doses, supporting the safety of low and mid therapeutic doses. At comparable therapeutic doses (FF 100, FP 500 and BUD 800 µg/day), FF had the least effect on the most sensitive markers (adrenal steroids) vs BUD and FP.
Collapse
Affiliation(s)
- Peter Daley-Yates
- Clinical Pharmacology and Experimental Medicine, GSK, Uxbridge, UK
- Correspondence should be addressed to P Daley-Yates;
| | - Brian Keppler
- Discovery and Translational Sciences, Metabolon Inc., Morrisville, North Carolina, USA
| | | | | | - Dave Singh
- Medicines Evaluation Unit, University of Manchester, Manchester University NHS Foundation Trust, Manchester, UK
| | - Neil Barnes
- Global Medical Franchise, GSK, Brentford, UK
- William Harvey Institute, Bart’s and the London School of Medicine and Dentistry, London, UK
| |
Collapse
|
27
|
Wang M, Deng R. Effects of carbon black nanoparticles and high humidity on the lung metabolome in Balb/c mice with established allergic asthma. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:65100-65111. [PMID: 35484453 DOI: 10.1007/s11356-022-20349-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/15/2022] [Indexed: 06/14/2023]
Abstract
In respiratory diseases, the induction of allergic asthma has gradually aroused public concerns. Co-exposures of environmental risk factors such as nanoparticles and high humidity could play important roles in the development of allergic asthma. However, the relevant researches are still lacking and the involved mechanisms, especially metabolic changes, remain unclear. We took the lead in studying the combined induction effect and underlying mechanisms of carbon black nanoparticles (CB NPs) and high humidity on allergic asthma. In this work, murine models of allergic asthma were established with ovalbumin under the single and combined exposures of 15 μg/kg CB NPs and 90% relative humidity. The two risk factors, particularly their co-exposure, exhibited adjuvant effect on airway hyperresponsiveness, remodeling, and inflammation in Balb/c mice. Untargeted metabolomics identified the potential biomarkers in lung for asthma occurrence and for asthma exacerbation caused by CB NPs and high humidity. The significantly dysregulated metabolic pathways in asthmatic mice were proposed, and the disturbed metabolic pathways under the exposures of CB NPs and/or high humidity were mainly implicated in asthma symptoms. This work sheds light on the understanding for health risks of NP pollutions and high environmental humidity and contributes to useful biomarker identification and asthma control.
Collapse
Affiliation(s)
- Mingpu Wang
- School of Civil Engineering, Chongqing University, Chongqing, 400045, China
| | - Rui Deng
- School of Civil Engineering, Chongqing University, Chongqing, 400045, China.
| |
Collapse
|
28
|
Tang X, Hou Y, Schwartz TW, Haeggström JZ. Metabolite G-protein coupled receptor signaling: Potential regulation of eicosanoids. Biochem Pharmacol 2022; 204:115208. [PMID: 35963340 DOI: 10.1016/j.bcp.2022.115208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/05/2022] [Accepted: 08/05/2022] [Indexed: 11/19/2022]
Abstract
Eicosanoids are a family of bioactive compounds derived from arachidonic acid (AA) that play pivotal roles in physiology and disease, including inflammatory conditions of multiple organ systems. The biosynthesis of eicosanoids requires a series of catalytic steps that are controlled by designated enzymes, which can be regulated by inflammatory and stress signals via transcriptional and translational mechanisms. In the past decades, evidence have emerged indicating that G-protein coupled receptors (GPCRs) can sense extracellular metabolites, and regulate inflammatory responses including eicosanoid production. This review focuses on the recent advances of metabolite GPCRs research, their role in regulation of eicosanoid biosynthesis, and the link to pathophysiological conditions.
Collapse
Affiliation(s)
- Xiao Tang
- Division of Physiological Chemistry II, Biomedicum 9A, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden.
| | - Yaolin Hou
- Division of Physiological Chemistry II, Biomedicum 9A, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Thue W Schwartz
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; Laboratory for Molecular Pharmacology, Department for Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jesper Z Haeggström
- Division of Physiological Chemistry II, Biomedicum 9A, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden.
| |
Collapse
|
29
|
Liang L, Hu M, Chen Y, Liu L, Wu L, Hang C, Luo X, Xu X. Metabolomics of bronchoalveolar lavage in children with persistent wheezing. Respir Res 2022; 23:161. [PMID: 35718784 PMCID: PMC9208141 DOI: 10.1186/s12931-022-02087-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/11/2022] [Indexed: 11/28/2022] Open
Abstract
Background Recent studies have demonstrated the important role of metabolomics in the pathogenesis of asthma. However, the role of lung metabolomics in childhood persistent wheezing (PW) or wheezing recurrence remains poorly understood. Methods In this prospective observational study, we performed a liquid chromatography/mass spectrometry-based metabolomic survey on bronchoalveolar lavage samples collected from 30 children with PW and 30 age-matched infants (control group). A 2-year follow-up study on these PW children was conducted. Results Children with PW showed a distinct characterization of respiratory metabolome compared with control group. Children with PW had higher abundances of choline, oleamide, nepetalactam, butyrylcarnitine, l-palmitoylcarnitine, palmitoylethanolamide, and various phosphatidylcholines. The glycerophospholipid metabolism pathway was the most relevant pathway involving in PW pathophysiologic process. Additionally, different gender, prematurity, and systemic corticoids use demonstrated a greater impact in airway metabolite compositions. Furthermore, for PW children with recurrence during the follow-up period, children who were born prematurely had an increased abundance of butyrylcarnitine relative to those who were carried to term. Conclusions This study suggests that the alterations of lung metabolites could be associated with the development of wheezing, and this early alteration could also be correlated with wheezing recurrence later in life. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02087-6.
Collapse
Affiliation(s)
- Lingfang Liang
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China
| | - Minfei Hu
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China
| | - Yuanling Chen
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China
| | - Lingke Liu
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China
| | - Lei Wu
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China
| | - Chengcheng Hang
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China
| | - Xiaofei Luo
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China
| | - Xuefeng Xu
- Department of Rheumatology Immunology and Allergy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310003, People's Republic of China.
| |
Collapse
|
30
|
A Metabolomics Approach to Sulforaphane Efficacy in Secondhand Smoking-Induced Pulmonary Damage in Mice. Metabolites 2022; 12:metabo12060518. [PMID: 35736451 PMCID: PMC9227370 DOI: 10.3390/metabo12060518] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 02/01/2023] Open
Abstract
Sulforaphane is an isocyanate abundantly present in cruciferous vegetables. In the present study, we aimed to investigate the effects of sulforaphane on secondhand smoking (SHS)-induced pulmonary damage in mice. Additionally, a metabolomic study was performed to identify biomarkers associated with pulmonary disease using proton nuclear magnetic resonance (1H-NMR) analysis. Male C57BL6J mice were divided into a control group, an SHS exposure group (positive control group, PC), and a sulforaphane treatment group exposed to secondhand smoke (SS) (n = 5 per group). The PC and SS groups were exposed to secondhand smoke in a chamber twice daily for four weeks. Mice in the SS group were orally administered sulforaphane (50 mg/kg) for four weeks during secondhand smoke exposure. Histopathological examination of the lungs revealed pulmonary damage in PC mice, including loss of bronchial epithelial cells, bronchial wall thickening, and infiltration of macrophages. In contrast, mice in the SS group showed little or no epithelial thickening, thereby exhibiting reduced lung damage. Mouse serum and lung tissues were collected and analyzed to determine changes in endogenous metabolites using 1H-NMR. After target profiling, we identified metabolites showing the same tendency in the serum and lung as biomarkers for SHS-induced pulmonary damage, including taurine, glycerol, creatine, arginine, and leucine. As a result of histopathological examination, sulforaphane might inhibit SHS-induced lung damage, and metabolite analysis results suggest potential biomarkers for SHS-induced pulmonary damage in mice.
Collapse
|
31
|
Xu S, Panettieri RA, Jude J. Metabolomics in asthma: A platform for discovery. Mol Aspects Med 2022; 85:100990. [PMID: 34281719 PMCID: PMC9088882 DOI: 10.1016/j.mam.2021.100990] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 06/21/2021] [Accepted: 07/06/2021] [Indexed: 12/28/2022]
Abstract
Asthma, characterized by airway hyperresponsiveness, inflammation and remodeling, is a chronic airway disease with complex etiology. Severe asthma is characterized by frequent exacerbations and poor therapeutic response to conventional asthma therapy. A clear understanding of cellular and molecular mechanisms of asthma is critical for the discovery of novel targets for optimal therapeutic control of asthma. Metabolomics is emerging as a powerful tool to elucidate novel disease mechanisms in a variety of diseases. In this review, we summarize the current status of knowledge in asthma metabolomics at systemic and cellular levels. The findings demonstrate that various metabolic pathways, related to energy metabolism, macromolecular biosynthesis and redox signaling, are differentially modulated in asthma. Airway smooth muscle cell plays pivotal roles in asthma by contributing to airway hyperreactivity, inflammatory mediator release and remodeling. We posit that metabolomic profiling of airway structural cells, including airway smooth muscle cells, will shed light on molecular mechanisms of asthma and airway hyperresponsiveness and help identify novel therapeutic targets.
Collapse
Affiliation(s)
- Shengjie Xu
- Rutgers Institute for Translational Medicine & Science, Rutgers, The State University of New Jersey, 89 French Street, New Brunswick, NJ, 08901, USA; Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 89 French Street, New Brunswick, NJ, 08901, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine & Science, Rutgers, The State University of New Jersey, 89 French Street, New Brunswick, NJ, 08901, USA; Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 89 French Street, New Brunswick, NJ, 08901, USA; Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, 89 French Street, New Brunswick, NJ, 08901, USA
| | - Joseph Jude
- Rutgers Institute for Translational Medicine & Science, Rutgers, The State University of New Jersey, 89 French Street, New Brunswick, NJ, 08901, USA; Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 89 French Street, New Brunswick, NJ, 08901, USA; Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, 89 French Street, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
32
|
Delgado‐Dolset MI, Obeso D, Rodríguez‐Coira J, Tarin C, Tan G, Cumplido JA, Cabrera A, Angulo S, Barbas C, Sokolowska M, Barber D, Carrillo T, Villaseñor A, Escribese MM. Understanding uncontrolled severe allergic asthma by integration of omic and clinical data. Allergy 2022; 77:1772-1785. [PMID: 34839541 DOI: 10.1111/all.15192] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/04/2021] [Accepted: 11/02/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Asthma is a complex, multifactorial disease often linked with sensitization to house dust mites (HDM). There is a subset of patients that does not respond to available treatments, who present a higher number of exacerbations and a worse quality of life. To understand the mechanisms of poor asthma control and disease severity, we aim to elucidate the metabolic and immunologic routes underlying this specific phenotype and the associated clinical features. METHODS Eighty-seven patients with a clinical history of asthma were recruited and stratified in 4 groups according to their response to treatment: corticosteroid-controlled (ICS), immunotherapy-controlled (IT), biologicals-controlled (BIO) or uncontrolled (UC). Serum samples were analysed by metabolomics and proteomics; and classifiers were built using machine-learning algorithms. RESULTS Metabolomic analysis showed that ICS and UC groups cluster separately from one another and display the highest number of significantly different metabolites among all comparisons. Metabolite identification and pathway enrichment analysis highlighted increased levels of lysophospholipids related to inflammatory pathways in the UC patients. Likewise, 8 proteins were either upregulated (CCL13, ARG1, IL15 and TNFRSF12A) or downregulated (sCD4, CCL19 and IFNγ) in UC patients compared to ICS, suggesting a significant activation of T cells in these patients. Finally, the machine-learning model built including metabolomic and clinical data was able to classify the patients with an 87.5% accuracy. CONCLUSIONS UC patients display a unique fingerprint characterized by inflammatory-related metabolites and proteins, suggesting a pro-inflammatory environment. Moreover, the integration of clinical and experimental data led to a deeper understanding of the mechanisms underlying UC phenotype.
Collapse
Affiliation(s)
- María Isabel Delgado‐Dolset
- Institute of Applied Molecular Medicine (IMMA) Department of Basic Medical Sciences Facultad de Medicina Universidad San Pablo CEU CEU Universities Urbanización Montepríncipe Madrid Spain
- Centre for Metabolomics and Bioanalysis (CEMBIO) Department of Chemistry and Biochemistry Facultad de Farmacia Universidad San Pablo CEU CEU Universities Urbanización Montepríncipe Madrid Spain
| | - David Obeso
- Institute of Applied Molecular Medicine (IMMA) Department of Basic Medical Sciences Facultad de Medicina Universidad San Pablo CEU CEU Universities Urbanización Montepríncipe Madrid Spain
- Centre for Metabolomics and Bioanalysis (CEMBIO) Department of Chemistry and Biochemistry Facultad de Farmacia Universidad San Pablo CEU CEU Universities Urbanización Montepríncipe Madrid Spain
| | - Juan Rodríguez‐Coira
- Institute of Applied Molecular Medicine (IMMA) Department of Basic Medical Sciences Facultad de Medicina Universidad San Pablo CEU CEU Universities Urbanización Montepríncipe Madrid Spain
- Centre for Metabolomics and Bioanalysis (CEMBIO) Department of Chemistry and Biochemistry Facultad de Farmacia Universidad San Pablo CEU CEU Universities Urbanización Montepríncipe Madrid Spain
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Zurich Switzerland
| | - Carlos Tarin
- Institute of Applied Molecular Medicine (IMMA) Department of Basic Medical Sciences Facultad de Medicina Universidad San Pablo CEU CEU Universities Urbanización Montepríncipe Madrid Spain
| | - Ge Tan
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Zurich Switzerland
| | - José A. Cumplido
- Hospital Universitario de Gran Canaria Doctor Negrin Las Palmas de Gran Canaria Spain
| | - Ana Cabrera
- Hospital Universitario de Gran Canaria Doctor Negrin Las Palmas de Gran Canaria Spain
| | - Santiago Angulo
- Department of Applied Mathematics and Statistics Universidad San Pablo‐CEU CEU Universities Madrid Spain
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO) Department of Chemistry and Biochemistry Facultad de Farmacia Universidad San Pablo CEU CEU Universities Urbanización Montepríncipe Madrid Spain
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Zurich Switzerland
| | - Domingo Barber
- Institute of Applied Molecular Medicine (IMMA) Department of Basic Medical Sciences Facultad de Medicina Universidad San Pablo CEU CEU Universities Urbanización Montepríncipe Madrid Spain
| | - Teresa Carrillo
- Hospital Universitario de Gran Canaria Doctor Negrin Las Palmas de Gran Canaria Spain
| | - Alma Villaseñor
- Institute of Applied Molecular Medicine (IMMA) Department of Basic Medical Sciences Facultad de Medicina Universidad San Pablo CEU CEU Universities Urbanización Montepríncipe Madrid Spain
| | - María M. Escribese
- Institute of Applied Molecular Medicine (IMMA) Department of Basic Medical Sciences Facultad de Medicina Universidad San Pablo CEU CEU Universities Urbanización Montepríncipe Madrid Spain
| |
Collapse
|
33
|
Tang S, Wu X, Zhao P, Tang K, Chen Y, Fu J, Zhou S, Yang Z, Zhang Z. A near-infrared fluorescence capillary imprinted sensor for chiral recognition and sensitive detection of l-histidine. Anal Chim Acta 2022; 1206:339794. [DOI: 10.1016/j.aca.2022.339794] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/17/2022] [Accepted: 03/30/2022] [Indexed: 12/30/2022]
|
34
|
Chang YH, Yeh KW, Huang JL, Su KW, Tsai MH, Hua MC, Liao SL, Lai SH, Chen LC, Chiu CY. Metabolomics analysis reveals molecular linkages for the impact of vitamin D on childhood allergic airway diseases. Pediatr Allergy Immunol 2022; 33:e13785. [PMID: 35616893 DOI: 10.1111/pai.13785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Several studies have reported the relevance between serum vitamin D and allergic immunoglobulin E (IgE) responses and atopic diseases. However, a metabolomics-based approach to the impacts of vitamin D on allergic reactions remains unclear. METHODS A total of 111 children completed a 3-year follow-up were enrolled and classified based on longitudinal vitamin D status (≥ 30 ng/ml, n = 54; 20-29.9 ng/ml, n = 41; <20 ng/ml, n = 16). Urinary metabolomic profiling was performed using 1 H-Nuclear magnetic resonance (NMR) spectroscopy at age 3. Integrative analyses of their associations related to vitamin D levels, atopic indices, and allergies were performed, and their roles in functional metabolic pathways were also assessed. RESULTS Six and five metabolites were identified to be significantly associated with vitamin D status and atopic diseases, respectively (FDR-adjusted p-value <.05). A further correlation analysis revealed that vitamin D-associated 3-hydroxyisobutyric acid and glutamine were positively correlated with atopic disease-associated succinic acid and alanine, respectively. Furthermore, hippuric acid was negatively correlated with atopic disease-associated formic acid, which was positively correlated with vitamin D level (p < .01). Absolute eosinophil count (AEC) was positively correlated with serum D. pteronyssinus- and D. farinae-specific IgE level (p < .01) but negatively correlated with vitamin D level (p < .05). Amino acid metabolisms were significantly associated with vitamin D related to childhood allergies. CONCLUSION Integrative metabolomic analysis provides the link of vitamin D-associated metabolites with the gut microbiome and immunoallergic reactions related to childhood allergies.
Collapse
Affiliation(s)
- Yu-Ho Chang
- School of Traditional Chinese Medicine, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Kuo-Wei Yeh
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Jing-Long Huang
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Taoyuan, Taiwan.,Department of Pediatrics, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital and Chang Gung University, Keelung, Taiwan
| | - Kuan-Wen Su
- Department of Pediatrics, Chang Gung Memorial Hospital at Keelung, and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ming-Han Tsai
- Department of Pediatrics, Chang Gung Memorial Hospital at Keelung, and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Man-Chin Hua
- Department of Pediatrics, Chang Gung Memorial Hospital at Keelung, and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Sui-Ling Liao
- Department of Pediatrics, Chang Gung Memorial Hospital at Keelung, and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Shen-Hao Lai
- Division of Pediatric Pulmonology, Chang Gung Memorial Hospital at Linkou, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Li-Chen Chen
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Taoyuan, Taiwan.,Department of Pediatrics, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital and Chang Gung University, Keelung, Taiwan
| | - Chih-Yung Chiu
- Division of Pediatric Pulmonology, Chang Gung Memorial Hospital at Linkou, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
35
|
Li J, Yao SL, Zheng TF, Xu H, Li JY, Peng Y, Chen JL, Liu SJ, Wen HR. Turn-on and blue-shift fluorescence sensor toward L-histidine based on stable Cd II metal-organic framework with tetranuclear cluster units. Dalton Trans 2022; 51:5983-5988. [PMID: 35348554 DOI: 10.1039/d2dt00390b] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A novel CdII-based two-fold interpenetrated metal-organic framework (MOF), namely {[Cd2(BTDB)2(4,4-bpy)]·DMF}n (JXUST-14), (H2BTDB = 4,4'-(benzo[c][1,2,5]thiadiazole-4,7-diyl)dibenzoic acid and 4,4-bpy = 4,4-bipyridine), has been prepared and characterized. Single-crystal structure determination reveals that JXUST-14 has a tetranuclear cluster based 6-connected pcu topological network with Schlafli symbol {412·63}. When soaked in common organic solvents and aqueous solutions with diverse pH values of 2-12 for 48 h, JXUST-14 remains stable. JXUST-14 is a highly selective and sensitive luminescent sensor for L-histidine (His) with a detection limit of 11.1 ppm. JXUST-14 is the first CdII-based MOF for the detection of His via turn-on and fluorescence blue-shift effects. Experimental study and theoretical calculation suggest that the sensing process can be mainly attributed to a charge transfer and energy transfer mechanism. More interestingly, LED lamps of JXUST-14 and JXUST-14@His were successfully developed, which endowed efficient sensitivity for His detection and thus provide great potential for future applications.
Collapse
Affiliation(s)
- Jing Li
- School of Chemistry and Chemical Engineering, Jiangxi Provincial Key Laboratory of Functional Molecular Materials Chemistry, Jiangxi University of Science and Technology, Ganzhou, Jiangxi Province 341000, P.R. China.
| | - Shu-Li Yao
- School of Chemistry and Chemical Engineering, Jiangxi Provincial Key Laboratory of Functional Molecular Materials Chemistry, Jiangxi University of Science and Technology, Ganzhou, Jiangxi Province 341000, P.R. China.
| | - Teng-Fei Zheng
- School of Chemistry and Chemical Engineering, Jiangxi Provincial Key Laboratory of Functional Molecular Materials Chemistry, Jiangxi University of Science and Technology, Ganzhou, Jiangxi Province 341000, P.R. China.
| | - Hui Xu
- School of Chemistry and Chemical Engineering, Jiangxi Provincial Key Laboratory of Functional Molecular Materials Chemistry, Jiangxi University of Science and Technology, Ganzhou, Jiangxi Province 341000, P.R. China.
| | - Jin-Yan Li
- School of Chemical and Environmental Engineering, Hanshan Normal University, Chaozhou, Guangdong Province 521041, P.R. China
| | - Yan Peng
- School of Chemistry and Chemical Engineering, Jiangxi Provincial Key Laboratory of Functional Molecular Materials Chemistry, Jiangxi University of Science and Technology, Ganzhou, Jiangxi Province 341000, P.R. China.
| | - Jing-Lin Chen
- School of Chemistry and Chemical Engineering, Jiangxi Provincial Key Laboratory of Functional Molecular Materials Chemistry, Jiangxi University of Science and Technology, Ganzhou, Jiangxi Province 341000, P.R. China.
| | - Sui-Jun Liu
- School of Chemistry and Chemical Engineering, Jiangxi Provincial Key Laboratory of Functional Molecular Materials Chemistry, Jiangxi University of Science and Technology, Ganzhou, Jiangxi Province 341000, P.R. China.
| | - He-Rui Wen
- School of Chemistry and Chemical Engineering, Jiangxi Provincial Key Laboratory of Functional Molecular Materials Chemistry, Jiangxi University of Science and Technology, Ganzhou, Jiangxi Province 341000, P.R. China.
| |
Collapse
|
36
|
Dasgupta S, Ghosh N, Choudhury P, Joshi M, Chowdhury SR, Bhattacharyya P, Chaudhury K. NMR metabolomic and microarray-based transcriptomic data integration identifies unique molecular signatures of hypersensitivity pneumonitis. Mol Omics 2021; 18:101-111. [PMID: 34881764 DOI: 10.1039/d1mo00209k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hypersensitivity pneumonitis (HP) is an immune-mediated granulomatous interstitial lung disease (ILD) that results from repeated inhalation of certain antigens. Despite major advances in research, pathophysiology of the disease remains poorly understood. The present study combines metabolomic and transcriptomic data to determine alterations in HP subjects as compared with healthy controls. Metabolic signatures were identified in serum, exhaled breath condensate (EBC) and bronchoalveolar lavage fluid (BALF) of HP patients using proton nuclear magnetic resonance (NMR) metabolomics. The expression of three metabolites, i.e., lactate, pyruvate, and proline, was found to be significantly altered in all three biofluids. The potential of differential diagnosis based on these three metabolites was investigated by including a group of patients with sarcoidosis, which is another type of granulomatous ILD. In addition, differentially expressed transcriptomic fingerprints in blood samples were identified by analyzing a Gene Expression Omnibus (GEO) database. The transcriptomics analysis of these microarray-based data revealed 59 genes to be significantly dysregulated in patients with HP. Over representation analysis of the metabolites and genes of interest was performed using IMPaLA (Integrated Molecular Pathway Level Analysis) version 12. Integrated analysis of serum metabolite signatures and blood gene expression suggests dysregulation of PI3K-AKT signaling and TCA cycle pathways in these patients. This preliminary study is a step towards better understanding of the pathogenesis of HP by identification of differentially expressed metabolites and transcriptomic fingerprints. These molecular signatures may be explored as diagnostic markers for differentiating HP from other lung diseases.
Collapse
Affiliation(s)
- Sanjukta Dasgupta
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| | - Nilanjana Ghosh
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| | - Priyanka Choudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| | - Mamata Joshi
- National Facility for High-field NMR, Tata Institute of Fundamental Research, Mumbai, India
| | | | | | - Koel Chaudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| |
Collapse
|
37
|
Logotheti M, Agioutantis P, Katsaounou P, Loutrari H. Microbiome Research and Multi-Omics Integration for Personalized Medicine in Asthma. J Pers Med 2021; 11:jpm11121299. [PMID: 34945771 PMCID: PMC8707330 DOI: 10.3390/jpm11121299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/13/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Asthma is a multifactorial inflammatory disorder of the respiratory system characterized by high diversity in clinical manifestations, underlying pathological mechanisms and response to treatment. It is generally established that human microbiota plays an essential role in shaping a healthy immune response, while its perturbation can cause chronic inflammation related to a wide range of diseases, including asthma. Systems biology approaches encompassing microbiome analysis can offer valuable platforms towards a global understanding of asthma complexity and improving patients' classification, status monitoring and therapeutic choices. In the present review, we summarize recent studies exploring the contribution of microbiota dysbiosis to asthma pathogenesis and heterogeneity in the context of asthma phenotypes-endotypes and administered medication. We subsequently focus on emerging efforts to gain deeper insights into microbiota-host interactions driving asthma complexity by integrating microbiome and host multi-omics data. One of the most prominent achievements of these research efforts is the association of refractory neutrophilic asthma with certain microbial signatures, including predominant pathogenic bacterial taxa (such as Proteobacteria phyla, Gammaproteobacteria class, especially species from Haemophilus and Moraxella genera). Overall, despite existing challenges, large-scale multi-omics endeavors may provide promising biomarkers and therapeutic targets for future development of novel microbe-based personalized strategies for diagnosis, prevention and/or treatment of uncontrollable asthma.
Collapse
Affiliation(s)
- Marianthi Logotheti
- G.P. Livanos and M. Simou Laboratories, 1st Department of Critical Care Medicine & Pulmonary Services, Evangelismos Hospital, Medical School, National Kapodistrian University of Athens, 3 Ploutarchou Str., 10675 Athens, Greece; (M.L.); (P.A.)
- Biotechnology Laboratory, School of Chemical Engineering, National Technical University of Athens, 5 Iroon Polytechniou Str., Zografou Campus, 15780 Athens, Greece
| | - Panagiotis Agioutantis
- G.P. Livanos and M. Simou Laboratories, 1st Department of Critical Care Medicine & Pulmonary Services, Evangelismos Hospital, Medical School, National Kapodistrian University of Athens, 3 Ploutarchou Str., 10675 Athens, Greece; (M.L.); (P.A.)
| | - Paraskevi Katsaounou
- Pulmonary Dept First ICU, Evangelismos Hospital, Medical School, National Kapodistrian University of Athens, Ipsilantou 45-7, 10675 Athens, Greece;
| | - Heleni Loutrari
- G.P. Livanos and M. Simou Laboratories, 1st Department of Critical Care Medicine & Pulmonary Services, Evangelismos Hospital, Medical School, National Kapodistrian University of Athens, 3 Ploutarchou Str., 10675 Athens, Greece; (M.L.); (P.A.)
- Correspondence:
| |
Collapse
|
38
|
Integrated metabolic and microbial analysis reveals host-microbial interactions in IgE-mediated childhood asthma. Sci Rep 2021; 11:23407. [PMID: 34862469 PMCID: PMC8642522 DOI: 10.1038/s41598-021-02925-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 11/24/2021] [Indexed: 02/07/2023] Open
Abstract
A metabolomics-based approach to address the molecular mechanism of childhood asthma with immunoglobulin E (IgE) or allergen sensitization related to microbiome in the airways remains lacking. Fifty-three children with lowly sensitized non-atopic asthma (n = 15), highly sensitized atopic asthma (n = 13), and healthy controls (n = 25) were enrolled. Blood metabolomic analysis with 1H-nuclear magnetic resonance (NMR) spectroscopy and airway microbiome composition analysis by bacterial 16S rRNA sequencing were performed. An integrative analysis of their associations with allergen-specific IgE levels for lowly and highly sensitized asthma was also assessed. Four metabolites including tyrosine, isovalerate, glycine, and histidine were uniquely associated with lowly sensitized asthma, whereas one metabolite, acetic acid, was strongly associated with highly sensitized asthma. Metabolites associated with highly sensitized asthma (valine, isobutyric acid, and acetic acid) and lowly sensitized asthma (isovalerate, tyrosine, and histidine) were strongly correlated each other (P < 0.01). Highly sensitized asthma associated metabolites were mainly enriched in pyruvate and acetyl-CoA metabolisms. Metabolites associated with highly sensitized atopic asthma were mostly correlated with microbiota in the airways. Acetic acid, a short-chain fatty acid (SCFA), was negatively correlated with the genus Atopobium (P < 0.01), but positively correlated with the genus Fusobacterium (P < 0.05). In conclusion, metabolomics reveals microbes-related metabolic pathways associated with IgE responses to house dust mite allergens in childhood asthma. A strong correlation of metabolites related to highly sensitized atopic asthma with airway microbiota provides linkages between the host-microbial interactions and asthma endotypes.
Collapse
|
39
|
De Paepe E, Van Gijseghem L, De Spiegeleer M, Cox E, Vanhaecke L. A Systematic Review of Metabolic Alterations Underlying IgE-Mediated Food Allergy in Children. Mol Nutr Food Res 2021; 65:e2100536. [PMID: 34648231 DOI: 10.1002/mnfr.202100536] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/10/2021] [Indexed: 12/24/2022]
Abstract
SCOPE Immunoglobulin E-mediated food allergies (IgE-FA) are characterized by an ever-increasing prevalence, currently reaching up to 10.4% of children in the European Union. Metabolomics has the potential to provide a deeper understanding of the pathogenic mechanisms behind IgE-FA. METHODS AND RESULTS In this work, literature is systematically searched using Web of Science, PubMed, Scopus, and Embase, from January 2010 until May 2021, including human and animal metabolomic studies on multiple biofluids (urine, blood, feces). In total, 15 studies on IgE-FA are retained and a dataset of 277 potential biomarkers is compiled for in-depth pathway mapping. Decreased indoleamine 2,3-dioxygenase-1 (IDO- 1) activity is hypothesized due to altered plasma levels of tryptophan and its metabolites in IgE-FA children. In feces of children prior to IgE-FA, aberrant metabolization of sphingolipids and histidine is noted. Decreased fecal levels of (branched) short chain fatty acids ((B)SCFAs) compel a shift towards aerobic glycolysis and suggest dysbiosis, associated with an immune system shift towards T-helper 2 (Th2) responses. During animal anaphylaxis, a similar switch towards glycolysis is observed, combined with increased ketogenic pathways. Additionally, altered histidine, purine, pyrimidine, and lipid pathways are observed. CONCLUSION To conclude, this work confirms the unprecedented opportunities of metabolomics and supports the in-depth pathophysiological qualification in the quest towards improved diagnostic and prognostic biomarkers for IgE-FA.
Collapse
Affiliation(s)
- Ellen De Paepe
- Department of Translational Physiology, Infectiology and Public Health, Laboratory of Chemical Analysis, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Lynn Van Gijseghem
- Department of Translational Physiology, Infectiology and Public Health, Laboratory of Chemical Analysis, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Margot De Spiegeleer
- Department of Translational Physiology, Infectiology and Public Health, Laboratory of Chemical Analysis, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Eric Cox
- Department of Translational Physiology, Infectiology and Public Health, Laboratory of Immunology, Ghent University, Ghent, Belgium
| | - Lynn Vanhaecke
- Department of Translational Physiology, Infectiology and Public Health, Laboratory of Chemical Analysis, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.,Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Northern Ireland, Belfast, UK
| |
Collapse
|
40
|
López-Rodríguez JC, Rodríguez-Coira J, Benedé S, Barbas C, Barber D, Villalba MT, Escribese MM, Villaseñor A, Batanero E. Comparative metabolomics analysis of bronchial epithelium during barrier establishment after allergen exposure. Clin Transl Allergy 2021; 11:e12051. [PMID: 34582104 PMCID: PMC9082991 DOI: 10.1002/clt2.12051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 02/06/2023] Open
Abstract
Background Several studies have shown a correlation between an altered metabolome and respiratory allergies. The epithelial barrier hypothesis proposes that an epithelial barrier dysfunction can result in allergic diseases development. Der p 1 allergen from house dust mite is a renowned epithelial barrier disruptor and allergy initiator due to its cysteine‐protease activity. Here, we compared the metabolic profile of the bronchial epithelium exposed or not to Der p 1 during barrier establishment to understand its active role in allergy development. Methods Calu‐3 cells were cultivated in air‐liquid interface cultures and exposed to either Der p 1 or Ole e 1 allergens during barrier establishment. The comparative metabolomics analysis of apical and basolateral media were performed using liquid chromatography and capillary electrophoresis both coupled to mass spectrometry. Results We showed that epithelial barrier disruption by Der p 1 was associated with a specific metabolic profile, which was highly dependent on the state of the epithelium at the time of contact. Moreover, an apical‐basolateral distribution of the metabolites was also observed, indicating a compartmentalization of the response with differential metabolic patterns. A number of metabolites were changed by Der p 1, mainly related to amino acids metabolism, such as L‐arginine, L‐kynurenine and L‐methionine. Conclusion This work is the first report on the metabolic response in human bronchial epithelial cells associated with cysteine‐protease Der p 1 activity, which could contribute to allergy development. Moreover, it supports a reformulated epithelial barrier hypothesis that might help to explain allergies and their increasing prevalence.
Collapse
Affiliation(s)
- Juan Carlos López-Rodríguez
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | - Juan Rodríguez-Coira
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain.,Instituto de Medicina Molecular Aplicada (IMMA), Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Sara Benedé
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Domingo Barber
- Instituto de Medicina Molecular Aplicada (IMMA), Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - María Teresa Villalba
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | - María Marta Escribese
- Instituto de Medicina Molecular Aplicada (IMMA), Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Alma Villaseñor
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain.,Instituto de Medicina Molecular Aplicada (IMMA), Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Eva Batanero
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
41
|
Ghosh N, Choudhury P, Joshi M, Bhattacharyya P, Roychowdhury S, Banerjee R, Chaudhury K. Global metabolome profiling of exhaled breath condensates in male smokers with asthma COPD overlap and prediction of the disease. Sci Rep 2021; 11:16664. [PMID: 34404870 PMCID: PMC8370999 DOI: 10.1038/s41598-021-96128-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 07/30/2021] [Indexed: 02/07/2023] Open
Abstract
Asthma-chronic obstructive pulmonary disease (COPD) overlap, termed as ACO, is a complex heterogeneous disease characterised by persistent airflow limitation, which manifests features of both asthma and COPD. These patients have a worse prognosis, in terms of more frequent and severe exacerbations, more frequent symptoms, worse quality of life, increased comorbidities and a faster lung function decline. In absence of clear diagnostic or therapeutic guidelines, ACO presents as a challenge to clinicians. The present study aims to investigate whether ACO patients have a distinct exhaled breath condensate (EBC) metabolic profile in comparison to asthma and COPD. A total of 132 age and BMI matched male smokers were recruited in the exploratory phase which consisted of (i) controls = 33 (ii) asthma = 34 (iii) COPD = 30 and (iv) ACO = 35. Using nuclear magnetic resonance (NMR) metabolomics, 8 metabolites (fatty acid, propionate, isopropanol, lactate, acetone, valine, methanol and formate) were identified to be significantly dysregulated in ACO subjects when compared to both, asthma and COPD. The expression of these dysregulated metabolites were further validated in a fresh patient cohort consisting of (i) asthma = 32 (ii) COPD = 32 and (iii) ACO = 40, which exhibited a similar expression pattern. Multivariate receiver operating characteristic (ROC) curves generated using these metabolites provided a robust ACO classification model. The findings were also integrated with previously identified serum metabolites and inflammatory markers to develop a robust predictive model for differentiation of ACO. Our findings suggest that NMR metabolomics of EBC holds potential as a platform to identify robust, non-invasive biomarkers for differentiating ACO from asthma and COPD.
Collapse
Affiliation(s)
- Nilanjana Ghosh
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Priyanka Choudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Mamata Joshi
- National Facility for High-Field NMR, Tata Institute of Fundamental Research, Mumbai, India
| | | | | | - Rintu Banerjee
- Department of Agricultural and Food Engineering, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Koel Chaudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India.
| |
Collapse
|
42
|
Metabolic Phenotypes in Asthmatic Adults: Relationship with Inflammatory and Clinical Phenotypes and Prognostic Implications. Metabolites 2021; 11:metabo11080534. [PMID: 34436475 PMCID: PMC8400680 DOI: 10.3390/metabo11080534] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 12/26/2022] Open
Abstract
Bronchial asthma is a chronic disease that affects individuals of all ages. It has a high prevalence and is associated with high morbidity and considerable levels of mortality. However, asthma is not a single disease, and multiple subtypes or phenotypes (clinical, inflammatory or combinations thereof) can be detected, namely in aggregated clusters. Most studies have characterised asthma phenotypes and clusters of phenotypes using mainly clinical and inflammatory parameters. These studies are important because they may have clinical and prognostic implications and may also help to tailor personalised treatment approaches. In addition, various metabolomics studies have helped to further define the metabolic features of asthma, using electronic noses or targeted and untargeted approaches. Besides discriminating between asthma and a healthy state, metabolomics can detect the metabolic signatures associated with some asthma subtypes, namely eosinophilic and non-eosinophilic phenotypes or the obese asthma phenotype, and this may prove very useful in point-of-care application. Furthermore, metabolomics also discriminates between asthma and other “phenotypes” of chronic obstructive airway diseases, such as chronic obstructive pulmonary disease (COPD) or Asthma–COPD Overlap (ACO). However, there are still various aspects that need to be more thoroughly investigated in the context of asthma phenotypes in adequately designed, homogeneous, multicentre studies, using adequate tools and integrating metabolomics into a multiple-level approach.
Collapse
|
43
|
Perales-Chorda C, Obeso D, Twomey L, Rojas-Benedicto A, Puchades-Carrasco L, Roca M, Pineda-Lucena A, Laguna JJ, Barbas C, Esteban V, Martí-Garrido J, Ibañez-Echevarria E, López-Salgueiro R, Barber D, Villaseñor A, Hernández Fernández de Rojas D. Characterization of anaphylaxis reveals different metabolic changes depending on severity and triggers. Clin Exp Allergy 2021; 51:1295-1309. [PMID: 34310748 DOI: 10.1111/cea.13991] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/26/2021] [Accepted: 06/27/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Despite the increasing incidence of anaphylaxis, its underlying molecular mechanisms and biomarkers for appropriate diagnosis remain undetermined. The rapid onset and potentially fatal outcome in the absence of managed treatment prevent its study. Up today, there are still no known biomarkers that allow an unequivocal diagnosis. Therefore, the aim of this study was to explore metabolic changes in patients suffering anaphylactic reactions depending on the trigger (food and/or drug) and severity (moderate and severe) in a real-life set-up. METHODS Eighteen episodes of anaphylaxis, one per patient, were analysed. Sera were collected during the acute phase (T1), the recovery phase (T2) and around 2-3 months after the anaphylactic reaction (T0: basal state). Reactions were classified following an exhaustive allergological evaluation for severity and trigger. Sera samples were analysed using untargeted metabolomics combining liquid chromatography coupled to mass spectrometry (LC-MS) and proton nuclear magnetic resonance spectroscopy (1 H-NMR). RESULTS 'Food T1 vs T2' and 'moderate T1 vs T2' anaphylaxis comparisons showed clear metabolic patterns during the onset of an anaphylactic reaction, which differed from those induced by drugs, food + drug or severe anaphylaxis. Moreover, the model of food anaphylaxis was able to distinguish the well-characterized IgE (antibiotics) from non-IgE-mediated anaphylaxis (nonsteroidal anti-inflammatory drugs), suggesting a differential metabolic pathway associated with the mechanism of action. Metabolic differences between 'moderate vs severe' at the acute phase T1 and at basal state T0 were studied. Among the altered metabolites, glucose, lipids, cortisol, betaine and oleamide were observed altered. CONCLUSIONS The results of this exploratory study provide the first evidence that different anaphylactic triggers or severity induce differential metabolic changes along time or at specific time-point, respectively. Besides, the basal status T0 might identify high-risk patients, thus opening new ways to understand, diagnose and treat anaphylaxis.
Collapse
Affiliation(s)
| | - David Obeso
- IMMA, Instituto de Medicina Molecular Aplicada, Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe
- Boadilla del Monte, Madrid, 28660, Spain.,CEMBIO, Centre for Metabolomics and Bioanalysis, Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe
- Boadilla del Monte, Madrid, 28660, Spain
| | - Laura Twomey
- IMMA, Instituto de Medicina Molecular Aplicada, Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe
- Boadilla del Monte, Madrid, 28660, Spain.,CEMBIO, Centre for Metabolomics and Bioanalysis, Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe
- Boadilla del Monte, Madrid, 28660, Spain
| | | | | | - Marta Roca
- Analytical Unit, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Antonio Pineda-Lucena
- Drug Discovery Unit, Health Research Institute La Fe, Valencia, Spain.,Molecular Therapeutics Program, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - José Julio Laguna
- Allergy Unit, Allergo-Anaesthesia Unit, Hospital Central de la Cruz Roja, Madrid, Spain.,Faculty of Medicine and Biomedicine, Alfonso X El Sabio University, Madrid, Spain
| | - Coral Barbas
- CEMBIO, Centre for Metabolomics and Bioanalysis, Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe
- Boadilla del Monte, Madrid, 28660, Spain
| | - Vanesa Esteban
- Faculty of Medicine and Biomedicine, Alfonso X El Sabio University, Madrid, Spain.,Department of Allergy and Immunology, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - Jaume Martí-Garrido
- Allergy Department of Hospital, Universitari i Politècnic La Fe, Valencia, Spain
| | | | | | - Domingo Barber
- IMMA, Instituto de Medicina Molecular Aplicada, Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe
- Boadilla del Monte, Madrid, 28660, Spain
| | - Alma Villaseñor
- IMMA, Instituto de Medicina Molecular Aplicada, Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe
- Boadilla del Monte, Madrid, 28660, Spain
| | | |
Collapse
|
44
|
Li L, Fang Z, Lee YK, Zhao J, Zhang H, Lu W, Chen W. Prophylactic effects of oral administration of Lactobacillus casei on house dust mite-induced asthma in mice. Food Funct 2021; 11:9272-9284. [PMID: 33047743 DOI: 10.1039/d0fo01363c] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This study investigated the prophylactic effects of five Lactobacillus casei strains on house dust mite (HDM)-induced asthma in mice. BALB/c mice were orally administered with L. casei strains for one week before HDM treatment. Ketotifen and Lactobacillus rhamnosus GG were used as positive controls. All L. casei strains decreased the number of granulocytes and the levels of Th2 and Th17 inflammatory cytokines in the lungs, L. casei3 significantly decreased the airway inflammation score. Further studies showed that L. casei3, L. casei4, and L. casei5 decreased the chemokine levels, L. casei2, L. casei4, and L. casei5 promoted the secretion of secretory immunoglobulin A (sIgA), L. casei2 upregulated the interleukin (IL)-10 levels, and L. casei1 had no effect on these immune indices. L. casei1 and L. casei4 decreased the serum levels of total IgE and HDM-specific IgG1, respectively. L. casei3 and L. casei5 decreased both HDM-specific IgG1 and total IgE levels. L. casei2 did not affect the levels of these immunoglobulins. The gut microbiota analysis revealed that all five L. casei strains enhanced the richness of the gut microbiota mainly by increasing the abundance of Firmicutes, while there were differences at the genus level.Thus, the prophylactic effects of L. casei on HDM-induced mixed chronic airway inflammatory asthma exerted as they differentially affected the immune responses and gut microbiota composition. L. casei3, which exhibited the highest prophylactic effect, increased the acetate and propionate contents in a strain-dependent manner.
Collapse
Affiliation(s)
- Lingzhi Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhifeng Fang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yuan-Kun Lee
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China and Beijing Innovation Centre of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
| |
Collapse
|
45
|
Sim S, Choi Y, Park HS. Potential Metabolic Biomarkers in Adult Asthmatics. Metabolites 2021; 11:metabo11070430. [PMID: 34209139 PMCID: PMC8306564 DOI: 10.3390/metabo11070430] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/25/2021] [Accepted: 06/25/2021] [Indexed: 12/05/2022] Open
Abstract
Asthma is the most common chronic airway inflammation, with multiple phenotypes caused by complicated interactions of genetic, epigenetic, and environmental factors. To date, various determinants have been suggested for asthma pathogenesis by a new technology termed omics, including genomics, transcriptomics, proteomics, and metabolomics. In particular, the systematic analysis of all metabolites in a biological system, such as carbohydrates, amino acids, and lipids, has helped identify a novel pathway related to complex diseases. These metabolites are involved in the regulation of hypermethylation, response to hypoxia, and immune reactions in the pathogenesis of asthma. Among them, lipid metabolism has been suggested to be related to lung dysfunction in mild-to-moderate asthma. Sphingolipid metabolites are an important mediator contributing to airway inflammation in obese asthma and aspirin-exacerbated respiratory disease. Although how these molecular variants impact the disease has not been completely determined, identification of new causative factors may possibly lead to more-personalized and precise pathway-specific approaches for better diagnosis and treatment of asthma. In this review, perspectives of metabolites related to asthma and clinical implications have been highlighted according to various phenotypes.
Collapse
Affiliation(s)
| | | | - Hae-Sim Park
- Correspondence: ; Tel.: +82-31-219-5196; Fax: +82-31-219-5154
| |
Collapse
|
46
|
Hartvigsson O, Barman M, Rabe H, Sandin A, Wold AE, Brunius C, Sandberg AS. Associations of maternal and infant metabolomes with immune maturation and allergy development at 12 months in the Swedish NICE-cohort. Sci Rep 2021; 11:12706. [PMID: 34135462 PMCID: PMC8209090 DOI: 10.1038/s41598-021-92239-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/07/2021] [Indexed: 02/05/2023] Open
Abstract
Allergic diseases are the most common chronic diseases in childrenin the Western world, but little is know about what factors influence immune maturation and allergy development. We therefore aimed to associate infant and maternal metabolomes to T- and B-cell subpopulations and allergy diagnosis. We performed liquid chromatography-mass spectrometry based untargeted metabolomics on blood plasma from mothers (third trimester, n = 605; delivery, n = 558) and from the umbilical cord (n = 366). The measured metabolomes were associated to T- and B-cell subpopulations up to 4 months after delivery and to doctor´s diagnosed eczema, food allergy and asthma at one year of age using random forest analysis. Maternal and cord plasma at delivery could predict the number of CD24+CD38low memory B-cells (p = 0.033, n = 26 and p = 0.009, n = 22), but future allergy status could not be distinguished from any of the three measured metabolomes. Replication of previous literature findings showed hypoxanthine to be upregulated in the umbilical cord of children with subsequent asthma. This exploratory study suggests foetal immune programming occuring during pregnancy as the metabolomic profiles of mothers and infants at delivery related to infants' B-cell maturation.
Collapse
Affiliation(s)
- Olle Hartvigsson
- Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg, Sweden.
| | - Malin Barman
- Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg, Sweden
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hardis Rabe
- Institute of Biomedicine, Department of Infectious Diseases, University of Gothenburg, Gothenburg, Sweden
| | - Anna Sandin
- Department of Clinical Sciences, Unit of Pediatrics, Umeå University, Umeå, Sweden
| | - Agnes E Wold
- Institute of Biomedicine, Department of Infectious Diseases, University of Gothenburg, Gothenburg, Sweden
| | - Carl Brunius
- Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg, Sweden
| | - Ann-Sofie Sandberg
- Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg, Sweden
| |
Collapse
|
47
|
Application of Metabolomics in Pediatric Asthma: Prediction, Diagnosis and Personalized Treatment. Metabolites 2021; 11:metabo11040251. [PMID: 33919626 PMCID: PMC8072856 DOI: 10.3390/metabo11040251] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/07/2021] [Accepted: 04/15/2021] [Indexed: 12/16/2022] Open
Abstract
Asthma in children remains a significant public health challenge affecting 5–20% of children in Europe and is associated with increased morbidity and societal healthcare costs. The high variation in asthma incidence among countries may be attributed to differences in genetic susceptibility and environmental factors. This respiratory disorder is described as a heterogeneous syndrome of multiple clinical manifestations (phenotypes) with varying degrees of severity and airway hyper-responsiveness, which is based on patient symptoms, lung function and response to pharmacotherapy. However, an accurate diagnosis is often difficult due to diversities in clinical presentation. Therefore, identifying early diagnostic biomarkers and improving the monitoring of airway dysfunction and inflammatory through non-invasive methods are key goals in successful pediatric asthma management. Given that asthma is caused by the interaction between genes and environmental factors, an emerging approach, metabolomics—the systematic analysis of small molecules—can provide more insight into asthma pathophysiological mechanisms, enable the identification of early biomarkers and targeted personalized therapies, thus reducing disease burden and societal cost. The purpose of this review is to present evidence on the utility of metabolomics in pediatric asthma through the analysis of intermediate metabolites of biochemical pathways that involve carbohydrates, amino acids, lipids, organic acids and nucleotides and discuss their potential application in clinical practice. Also, current challenges on the integration of metabolomics in pediatric asthma management and needed next steps are critically discussed.
Collapse
|
48
|
Dong X, Yao S, Wu W, Cao J, Sun L, Li H, Ren H, Ren W. Gas explosion-induced acute blast lung injury assessment and biomarker identification by a LC-MS-based serum metabolomics analysis. Hum Exp Toxicol 2021; 40:608-621. [PMID: 32969285 DOI: 10.1177/0960327120960761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The objective of this study was to evaluate the histopathological effect of gas explosion on rats, and to explore the metabolic alterations associated with gas explosion-induced acute blast lung injury (ABLI) in real roadway environment using metabolomics analyses. All rats were exposed to the gas explosion source at different distance points (160 m and 240 m) except the control group. Respiratory function indexes were monitored and lung tissue analysis was performed to correlate histopathological effect to serum metabolomics. Their sera samples were collected to measure the metabolic alterations by ultra-performance liquid chromatography-mass spectrometry (UPLC-MS). HE staining in lung showed that the gas explosion caused obvious inflammatory pulmonary injury, which was consistent with respiratory function monitoring results and the serum metabolomics analysis results. The metabolomics identified 9 significantly metabolites different between the control- and ABLI rats. 2-aminoadipic acid, L-methionine, L-alanine, L-lysine, L-threonine, cholic acid and L-histidine were significantly increased in the exposed groups. Citric acid and aconitic acid were significantly decreased after exposure. Pathway analyses identified 8 perturbed metabolic pathways, which provided novel potential mechanisms for the gas explosion-induced ABLI. Therefore, metabolomics analysis identified both known and unknown alterations in circulating biomarkers, adding an integral mechanistic insight into the gas explosion-induced ABLI in real roadway environment.
Collapse
Affiliation(s)
- X Dong
- Department of Environmental and Occupational Health, School of Public Health, 91593Xinxiang Medical University, Xinxiang, Henan Province, China
| | - S Yao
- Department of Environmental and Occupational Health, School of Public Health, 91593Xinxiang Medical University, Xinxiang, Henan Province, China
| | - W Wu
- Department of Environmental and Occupational Health, School of Public Health, 91593Xinxiang Medical University, Xinxiang, Henan Province, China
| | - J Cao
- Institute of Toxicology, College of Preventive Medicine, 12525Third Military Medical University, Chongqing, China
| | - L Sun
- Institute of Toxicology, College of Preventive Medicine, 12525Third Military Medical University, Chongqing, China
| | - H Li
- Department of Environmental and Occupational Health, School of Public Health, 91593Xinxiang Medical University, Xinxiang, Henan Province, China
| | - H Ren
- Human Resources Department, Sanquan College, 91593Xinxiang Medical University, Xinxiang, Henan Province, China
| | - W Ren
- Institutes of Health Central Plains, 91593Xinxiang Medical University, Xinxiang, Henan Province, China
| |
Collapse
|
49
|
Wang X, Li H, Zhang Q, Shen Q, Zhu D, Li H, Tang Z, Zhao J, Liu Z. Histological chorioamnionitis is associated with an increased risk of wheezing in preterm children less than 34 gestational weeks. BMC Pediatr 2021; 21:104. [PMID: 33648480 PMCID: PMC7919088 DOI: 10.1186/s12887-021-02572-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 02/17/2021] [Indexed: 11/21/2022] Open
Abstract
Background Chorioamnionitis is associated with various neonatal short- and long-term morbidities. The effect of chorioamnionitis on premature children’s outcomes remains controversial. The aim of this study is to investigate the relationship between histological chorioamnionitis (HCA) and physiological development, wheezing, and atopic diseases in preterm children. Methods Singleton, preterm children (< 34 weeks), whose mother underwent pathological placental examinations, were retrospectively enrolled and the outcomes were assessed at 24–40 months during follow-up. Wheezing and atopic diseases including eczema, food allergies, and allergic rhinitis were screened by a questionnaire along with medical diagnosis. Anthropometric indexes and blood pressure were measured. Cognitive and behavioural developments were assessed by the Gesell Development and Diagnosis Scale. Blood IgE and routine examination were analyzed with venous blood and serum metabolomic profiling was assessed via liquid chromatography-mass spectrometry (LC-MS). A multivariate logistic regression model was used to estimate the association between HCA and the current outcomes. Results Among the 115 enrolled children, 47 were exposed to HCA. The incidence of wheezing was significantly higher in children exposed to HCA, as 38.30% of children who were exposed to HCA and 16.18% of children who were not had been diagnosed with wheezing. After adjusting for related confounders in the multivariate logistic regression model, there remained a 2.72-fold increased risk of wheezing in children with HCA (adjusted odds ratio, aOR, 2.72; 95% confidence interval, 1.02–7.23). Moreover, 163 differential metabolites, such as butanoic acid, annotemoyin 1 and charine, were identified in the HCA exposed children’s serum. Enrichment analysis revealed that these compounds participated in diverse key metabolomic pathways relating to physical and neuro- developments, including glycerophospholipid, alpha-linolenic acid and choline metabolisms. There were no significant differences in atopic diseases, serum IgE, eosinophils’ level, anthropometric indexes, blood pressure, or cognitive or behavioural developments between the two groups. Conclusion HCA exposure is associated with an increased risk of wheezing in preterm children less than 34 gestational weeks. Supplementary Information The online version contains supplementary material available at 10.1186/s12887-021-02572-9.
Collapse
Affiliation(s)
- Xiaoli Wang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 200030, China.,International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Haiyuan Li
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 200030, China.,International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Qianqian Zhang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 200030, China.,International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Qianwen Shen
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 200030, China.,International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Dan Zhu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 200030, China.,International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Hong Li
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 200030, China.,International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Zheng Tang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 200030, China.,International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Jiuru Zhao
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 200030, China. .,International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, China. .,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China.
| | - Zhiwei Liu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 200030, China. .,International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, China. .,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China.
| |
Collapse
|
50
|
Grasemann H, Holguin F. Oxidative stress and obesity-related asthma. Paediatr Respir Rev 2021; 37:18-21. [PMID: 32660723 DOI: 10.1016/j.prrv.2020.05.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022]
Abstract
Obesity is an asthma comorbidity associated with poor control, increased exacerbation risk and reduced response to inhaled and systemic corticosteroids. It affects children and adults differentially. In those with early onset asthma, it associated with increased eosinophilic inflammation, whereas in late onset, it correlates with lower nitric oxide (NO) and predominantly non-T2 inflammation. There are probably multiple pathways by which obesity impacts asthma; airway and systemic oxidative stress has been proposed as a mechanism that could potentially explain the obesity mediated increased comorbidity and poor response to treatment. More likely than not, oxidative stress is an epiphenomenon of a very diverse set of processes driven by complex changes in airway and systemic metabolism. This article provides a comprehensive overview of the clinical, metabolic, pathophysiological and therapeutic aspects of oxidative stress in patients with obesity and asthma.
Collapse
Affiliation(s)
- Hartmut Grasemann
- Hospital for Sick Children, Respiratory Medicine, University of Toronto. Toronto, Canada
| | - Fernando Holguin
- Department of Medicine, Pulmonary Sciences and Critical Care. University of Colorado. Denver, CO, United States.
| |
Collapse
|