1
|
Sontam T, Deutz NEP, Cruthirds CL, Mbilinyi R, Ruebush LE, Ten Have GA, Thaden JJ, Engelen MPKJ. Prolonged disturbances in citrulline metabolism following resistance exercise in COPD. Clin Nutr 2025; 49:21-32. [PMID: 40233541 DOI: 10.1016/j.clnu.2025.03.021] [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: 01/29/2025] [Revised: 03/21/2025] [Accepted: 03/28/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND & AIMS Disturbances in arginine (ARG) and protein metabolism, as well as in gut function have been observed in response to an endurance exercise session in patients with Chronic Obstructive Pulmonary Disease (COPD). We studied whether resistance exercise also affects the acute response in arginine (role in nitric oxide synthesis), citrulline (CIT, marker of gut health), and (muscle) protein metabolism differently in COPD as compared to healthy older adults. METHODS Patients with stable moderate to severe COPD (n = 24) and healthy controls (n = 25) completed a high-intensity resistance exercise session in the postabsorptive state. We administered a pulse of multiple stable isotopes of amino acids before, and 1 h and 24 h post-resistance exercise to assess the whole body production (WBP) and intracellular productions by compartmental analysis of ARG and CIT, and of tau-methylhistidine (TauMETHIS), phenylalanine (PHE), tyrosine (TYR), and PHE > TYR conversion as markers of muscle (myofibrillar) protein breakdown and whole body (net) protein breakdown, respectively. Muscle fatigue was determined by assessing the decay in peak leg extension torque post-resistance exercise. RESULTS COPD patients overall exhibited lower WBP ARG (p < 0.0001), CIT (p < 0.0001), PHE (p = 0.0001), TYR (p < 0.0001), and tau-METHIS (p = 0.0004) compared to controls. Resistance exercise did not change WBP of PHE, tau-METHIS, or PHE > TYR conversion, despite prolonged muscle fatigue in COPD. WBP CIT was increased at 1- and 24-h post-exercise in both groups (p < 0.003). Plasma CIT concentrations were reduced in both groups (p < 0.006) and remained lower at 24 h post-exercise in COPD only (p < 0.05) despite a third less work performed. CONCLUSIONS Both COPD and healthy participants exhibited upregulated whole-body citrulline production following resistance exercise. However, in COPD, this increase was insufficient to counteract the sustained reduction in plasma citrulline concentration, despite performing significantly less work during the exercise session. This prolonged disturbance in citrulline metabolism in COPD points to a potential exercise-induced enterocyte dysfunction, highlighting a novel area for understanding the impact of resistance exercise on gut health in this population. CLINICAL TRIAL REGISTRY Trial registration ClinicalTrials.gov: NCT02780219.
Collapse
Affiliation(s)
- Tarun Sontam
- Center for Translational Research in Aging & Longevity, Dept of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA; Department of Medical Education, Texas A&M School of Medicine, College Station, TX, USA
| | - Nicolaas E P Deutz
- Center for Translational Research in Aging & Longevity, Dept of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA; Department of Primary Care & Rural Medicine, Texas A&M School of Medicine, College Station, TX, USA
| | - Clayton L Cruthirds
- Center for Translational Research in Aging & Longevity, Dept of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA
| | - Robert Mbilinyi
- Center for Translational Research in Aging & Longevity, Dept of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA; Department of Medical Education, Texas A&M School of Medicine, College Station, TX, USA
| | - Laura E Ruebush
- Center for Translational Research in Aging & Longevity, Dept of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA
| | - Gabriella Am Ten Have
- Center for Translational Research in Aging & Longevity, Dept of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA
| | - John J Thaden
- Center for Translational Research in Aging & Longevity, Dept of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA
| | - Mariёlle P K J Engelen
- Center for Translational Research in Aging & Longevity, Dept of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA; Department of Primary Care & Rural Medicine, Texas A&M School of Medicine, College Station, TX, USA.
| |
Collapse
|
2
|
Abdel-Aziz MI, Hashimoto S, Neerincx AH, Haarman EG, Cecil A, Lintelmann J, Witting M, Hauck SM, Kerssemakers N, Verster JC, Bang C, Franke A, Dierdorp BS, Dekker T, Metwally NKA, Duitman JW, Lutter R, Gorenjak M, Toncheva AA, Kheiroddin P, Harner S, Brandstetter S, Wolff C, Corcuera-Elosegui P, López-Fernández L, Perez-Garcia J, Martin-Almeida M, Sardón-Prado O, Pino-Yanes M, Potočnik U, Kabesch M, Vijverberg SJH, Kraneveld AD, Maitland-van der Zee AH. Metabotypes are linked to uncontrolled childhood asthma, gut microbiota, and systemic inflammation. J Allergy Clin Immunol 2025:S0091-6749(25)00457-9. [PMID: 40280190 DOI: 10.1016/j.jaci.2025.04.017] [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: 12/20/2024] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Childhood asthma has been linked to distinct metabolomic profiles. OBJECTIVE We sought to identify phenotypes (metabotypes) in children with moderate to severe asthma through integrative fecal and serum metabolome analysis. METHODS Children from the Systems Pharmacology Approach to Uncontrolled Pediatric Asthma cohort with Global Initiative for Asthma treatment step 3 or higher were recruited. Asthma control was defined by the Asthma Control Test and annual exacerbation history. Targeted metabolomic profiling of feces and serum was performed using liquid chromatography and flow injection electrospray ionization-triple quadrupole mass spectrometry. Similarity network fusion integrated fecal and serum metabolome profiles, followed by spectral clustering. Clusters were analyzed for differences in asthma characteristics, food diaries, fecal microbiota composition, and levels of serum inflammatory markers and blood cells. RESULTS Integrative fecal and serum metabolome analysis of 92 children with moderate to severe asthma (median age, 11.5 years, 34% female) revealed 3 metabotypes. Metabotype 1 had the lowest percentage of allergic rhinitis, with elevated serum ceramides and triglycerides. Metabotype 2 had higher odds of asthma control, the highest percentage of children with 4 or more months of breast-feeding, reduced sugar intake, lowest levels of blood neutrophils and serum inflammatory markers, and elevated serum acylcarnitines and ω-3 fatty acids. Metabotype 3 included the highest percentage of uncontrolled asthma patients, with decreased serum cholesteryl esters, phosphatidylcholines, and sphingomyelins, elevated fecal amino acids, and reduced fecal microbiota diversity. CONCLUSIONS Metabotypes in children with moderate to severe asthma are linked to asthma control, distinct fecal microbiota, and systemic inflammatory patterns. The findings suggest that metabotyping can be valuable in precision medicine approaches for asthma.
Collapse
Affiliation(s)
- Mahmoud I Abdel-Aziz
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands; Amsterdam Public Health, Personalized Medicine, Amsterdam, The Netherlands; Department of Clinical Pharmacy, Faculty of Pharmacy, Assiut University, Assiut, Egypt.
| | - Simone Hashimoto
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands; Amsterdam Public Health, Personalized Medicine, Amsterdam, The Netherlands; Department of Pediatric Pulmonology and Allergy, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
| | - Anne H Neerincx
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Eric G Haarman
- Department of Pediatric Pulmonology and Allergy, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
| | - Alexander Cecil
- Metabolomic and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Jutta Lintelmann
- Metabolomic and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Michael Witting
- Metabolomic and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; Chair of Analytical Food Chemistry, TUM School of Life Sciences, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Stefanie M Hauck
- Metabolomic and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Nikki Kerssemakers
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Joris C Verster
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands; Centre for Mental Health and Brain sciences, Swinburne University, Melbourne, Australia; Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Barbara S Dierdorp
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands; Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Tamara Dekker
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands; Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nariman K A Metwally
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan Willem Duitman
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands; Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - René Lutter
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands; Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Mario Gorenjak
- Center for Human Molecular Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Antoaneta A Toncheva
- Department of Pediatric Pneumology and Allergy, University Children's Hospital Regensburg (KUNO), Regensburg, Germany
| | - Parastoo Kheiroddin
- Department of Pediatric Pneumology and Allergy, University Children's Hospital Regensburg (KUNO), Regensburg, Germany
| | - Susanne Harner
- Department of Pediatric Pneumology and Allergy, University Children's Hospital Regensburg (KUNO), Regensburg, Germany
| | | | - Christine Wolff
- University Children's Hospital, University of Regensburg, Regensburg, Germany
| | - Paula Corcuera-Elosegui
- Division of Pediatric Respiratory Medicine, Donostia University Hospital, San Sebastián, Spain
| | - Leyre López-Fernández
- Division of Pediatric Respiratory Medicine, Donostia University Hospital, San Sebastián, Spain
| | - Javier Perez-Garcia
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain
| | - Mario Martin-Almeida
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain
| | - Olaia Sardón-Prado
- Division of Pediatric Respiratory Medicine, Donostia University Hospital, San Sebastián, Spain; Department of Pediatrics, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), San Sebastián, Spain
| | - Maria Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain; Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain
| | - Uroš Potočnik
- Center for Human Molecular Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Maribor, Slovenia; Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova Ulica 17, Maribor, Slovenia; Department for Science and Research, University Medical Centre Maribor, Ljubljanska Ulica 5, Maribor, Slovenia
| | - Michael Kabesch
- Department of Pediatric Pneumology and Allergy, University Children's Hospital Regensburg (KUNO), Regensburg, Germany; University Children's Hospital, University of Regensburg, Regensburg, Germany
| | - Susanne J H Vijverberg
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands; Amsterdam Public Health, Personalized Medicine, Amsterdam, The Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Anke H Maitland-van der Zee
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands; Amsterdam Public Health, Personalized Medicine, Amsterdam, The Netherlands; Department of Pediatric Pulmonology and Allergy, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Grunwell JR, Fitzpatrick AM. Asthma Phenotypes and Biomarkers. Respir Care 2025. [PMID: 40013975 DOI: 10.1089/respcare.12352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Asthma experienced by both adults and children is a phenotypically heterogeneous condition. Severe asthma, characterized by ongoing symptoms and airway inflammation despite high doses of inhaled and/or systemic corticosteroids, is the focus of research efforts to understand this underlying heterogeneity. Clinical phenotypes in both adult and pediatric asthma have been determined using supervised definition-driven classification and unsupervised data-driven clustering methods. Efforts to understand the underlying inflammatory patterns of severe asthma have led to the seminal discovery of type 2-high versus type 2-low phenotypes and to the development of biologics targeted at type 2-high inflammation to reduce the rates of severe asthma exacerbations. Type 2-high asthma is characterized by upregulation of T helper 2 immune pathways including interleukin (IL)-4, IL-5, and IL-13 along with eosinophilic airway inflammation, sometimes allergic sensitization, and responsiveness to treatment with corticosteroids. Type 2-low asthma is poorly responsive to corticosteroids and is not as well characterized as type 2-high asthma. Type 2-low asthma is limited by being defined as the absence of type 2-high inflammatory markers. Choosing a biologic for the treatment of severe asthma involves the evaluation of a panel of biomarkers such as blood eosinophils, total and specific immunoglobulin E/allergic sensitization, and fractional exhaled nitric oxide. In this review, we focus on the underlying pathobiology of adult and pediatric asthma, discuss the different phenotype-based treatment options for adult and pediatric type 2-high with or without allergic asthma and type 2-low asthma, and describe a clinical phenotyping approach to patients to guide out-patient therapy. Finally, we end with a discussion of whether pediatric asthma exacerbations necessitating admission to an ICU constitute their own high-risk phenotype and/or whether it is a part of other previously defined high-risk subgroups such as difficult-to-control asthma, exacerbation-prone asthma, and severe treatment-resistant asthma.
Collapse
Affiliation(s)
- Jocelyn R Grunwell
- Dr. Grunwell is affiliated with Division of Critical Care Medicine, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia
| | - Anne M Fitzpatrick
- Dr. Fitzpatrick is affiliated with Division of Pulmonary, Allergy/Immunology, Cystic Fibrosis, and Sleep Medicine, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
4
|
Xu Z, Wu Y, Zhao X, Zhou H. Integrating nontargeted metabolomics and RNA sequencing of dexamethasone-treated and untreated asthmatic mice reveals changes of amino acids and aminoacyl-tRNA in group 2 innate lymphoid cells. Int J Biol Macromol 2024; 283:137630. [PMID: 39547613 DOI: 10.1016/j.ijbiomac.2024.137630] [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: 08/21/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Bronchial asthma is the most common multifactorial and heterogeneous disease in childhood. The glucocorticoid dexamethasone is a classic treatment for asthma. Research indicates that group 2 innate lymphoid cells (ILC2s) are crucial to the pathogenesis of asthma. However, few studies have focused on ILC2s metabolism and transcription. This study aims to establish an ovalbumin (OVA)-induced asthma model and a dexamethasone-treated asthma model to explore the regulation of lung ILC2s at the genetic and metabolic levels during the progression and remission of asthma, utilizing single-cell metabolomics and transcriptomics approaches. The results showed that ILC2s regulated the metabolic pathways and transcriptional levels of amino acids (such as arginine, proline, and histidine) and linoleic acid, as well as the metabolic biomarkers of arginine, urocanic acid, and linoleic acid in asthma. Additionally, the cytokine pathways and NF-γB pathways have been altered at the genetic level. At the same time, we revealed that dexamethasone regulates ILC2s amino acid and aminoacyl tRNA metabolism, as well as related genes, thereby alleviating asthma symptoms. Furthermore, we identified the genes Eno3 and Tap1, which are significantly associated with asthma. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to verify the accuracy of the RNA sequencing results. This study, for the first time, revealed the mechanistic changes of ILC2s in the development and treatment of asthma using multiomics techniques, laying a foundation for targeted therapies in asthma.
Collapse
Affiliation(s)
- Zhiwei Xu
- Department of Pediatrics, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Yaling Wu
- Department of Pediatrics, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Xiaoman Zhao
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui 230088, China
| | - Haoquan Zhou
- Department of Pediatrics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| |
Collapse
|
5
|
Stumpf F, Wunderle C, Ritz J, Bernasconi L, Neyer P, Tribolet P, Stanga Z, Mueller B, Bischoff SC, Schuetz P. Prognostic implications of the arginine metabolism in patients at nutritional risk: A secondary analysis of the randomized EFFORT trial. Clin Nutr 2024; 43:660-673. [PMID: 38309228 DOI: 10.1016/j.clnu.2024.01.012] [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: 10/16/2023] [Revised: 12/26/2023] [Accepted: 01/14/2024] [Indexed: 02/05/2024]
Abstract
BACKGROUND Arginine, a conditionally essential amino acid, is key component in metabolic pathways including immune regulation and protein synthesis. Depletion of arginine contributes to worse outcomes in severely ill and surgical patient populations. We assessed prognostic implications of arginine levels and its metabolites and ratios in polymorbid medical inpatients at nutritional risk regarding clinical outcomes and treatment response. METHODS Within this secondary analysis of the randomized controlled Effect of early nutritional support on Frailty, Functional Outcomes, and Recovery of malnourished medical inpatients Trial (EFFORT), we investigated the association of arginine, its metabolites and ratios (i.e., ADMA and SDMA, ratios of arginine/ADMA, arginine/ornithine, and global arginine bioavailability ratio) measured on hospital admission with short-term and long-term mortality by means of regression analysis. RESULTS Among the 231 patients with available measurements, low arginine levels ≤90.05 μmol/l (n = 86; 37 %) were associated with higher all-cause mortality at 30 days (primary endpoint, adjusted HR 3.27, 95 % CI 1.86 to 5.75, p < 0.001) and at 5 years (adjusted HR 1.50, 95 % CI 1.07 to 2.12, p = 0.020). Arginine metabolites and ratios were also associated with adverse outcome, but had lower prognostic value. There was, however, no evidence that treatment response was influenced by admission arginine levels. CONCLUSION This secondary analysis focusing on medical inpatients at nutritional risk confirms a strong association of low plasma arginine levels and worse clinical courses. The potential effects of arginine-enriched nutritional supplements should be investigated in this population of patients. CLINICAL TRIAL REGISTRATION clinicaltrials.gov as NCT02517476 (registered 7 August 2015).
Collapse
Affiliation(s)
- Franziska Stumpf
- Medical University Department, Division of General Internal and Emergency Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; Institute of Nutritional Medicine, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Carla Wunderle
- Medical University Department, Division of General Internal and Emergency Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland
| | - Jacqueline Ritz
- Medical University Department, Division of General Internal and Emergency Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland
| | - Peter Neyer
- Institute of Laboratory Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland
| | - Pascal Tribolet
- Medical University Department, Division of General Internal and Emergency Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; Department of Health Professions, Bern University of Applied Sciences, Falkenplatz 24, 3012 Bern, Switzerland; Faculty of Life Sciences University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Zeno Stanga
- Division of Diabetes, Endocrinology, Nutritional Medicine, and Metabolism, Bern University Hospital and University of Bern, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Beat Mueller
- Medical University Department, Division of General Internal and Emergency Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; Medical Faculty of the University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Philipp Schuetz
- Medical University Department, Division of General Internal and Emergency Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; Medical Faculty of the University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland.
| |
Collapse
|
6
|
Wang Z, Zhang J, Shi S, Ma H, Wang D, Zuo C, Zhang Q, Lian C. Predicting lung adenocarcinoma prognosis, immune escape, and pharmacomic profile from arginine and proline-related genes. Sci Rep 2023; 13:15198. [PMID: 37709932 PMCID: PMC10502151 DOI: 10.1038/s41598-023-42541-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/12/2023] [Indexed: 09/16/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is a highly heterogeneous disease that ranks first in morbidity and mortality. Abnormal arginine metabolism is associated with inflammatory lung disease and may influence alterations in the tumor immune microenvironment. However, the potential role of arginine and proline metabolic patterns and immune molecular markers in LUAD is unclear. Gene expression, somatic mutations, and clinicopathological information of LUAD were downloaded from The Cancer Genome Atlas (TCGA) database. Univariate Cox regression analysis was performed to identify metabolic genes associated with overall survival (OS). Unsupervised clustering divided the sample into two subtypes with different metabolic and immunological profiles. Gene set enrichment analysis (GESA) and gene set variation analysis (GSVA) were used to analyze the underlying biological processes of the two subtypes. Drug sensitivity between subtypes was also predicted; then prognostic features were developed by multivariate Cox regression analysis. In addition, validation was obtained in the GSE68465, and GSE50081 dataset. Then, gene expression, and clinical characterization of hub genes CPS1 and SMS were performed; finally, in vitro validation experiments for knockdown of SMS were performed in LUAD cell lines. In this study, we first identified 12 arginine and proline-related genes (APRGs) significantly associated with OS and characterized the clinicopathological features and tumor microenvironmental landscape of two different subtypes. Then, we established an arginine and proline metabolism-related scoring system and identified two hub genes highly associated with prognosis, namely CPS1, and SMS. In addition, we performed CCK8, transwell, and other functional experiments on SMS to obtain consistent results. Our comprehensive analysis revealed the potential molecular features and clinical applications of APRGs in LUAD. A model based on 2 APRGs can accurately predict survival outcomes in LUAD, improve our understanding of APRGs in LUAD, and pave a new pathway to guide risk stratification and treatment strategy development for LUAD patients.
Collapse
Affiliation(s)
- Ziqiang Wang
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu, 233030, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu, 233030, China
| | - Shuhua Shi
- Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu, 233030, China
| | - Hongyu Ma
- Department of Clinical Medicine, Bengbu Medical College, Bengbu, 233030, China
| | - Dongqin Wang
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu, 233030, China
| | - Chao Zuo
- Department of Clinical Laboratory, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Qiang Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China.
| | - Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu, 233030, China.
| |
Collapse
|
7
|
Cottrill KA, Chandler JD, Kobara S, Stephenson ST, Mohammad AF, Tidwell M, Mason C, Van Dresser M, Patrignani J, Kamaleswaran R, Fitzpatrick AM, Grunwell JR. Metabolomics identifies disturbances in arginine, phenylalanine, and glycine metabolism as differentiating features of exacerbating atopic asthma in children. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:100115. [PMID: 37609569 PMCID: PMC10443927 DOI: 10.1016/j.jacig.2023.100115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Background Asthma exacerbations are highly prevalent in children, but only a few studies have examined the biologic mechanisms underlying exacerbations in this population. Objective High-resolution metabolomics analyses were performed to understand the differences in metabolites in children with exacerbating asthma who were hospitalized in a pediatric intensive care unit for status asthmaticus. We hypothesized that compared with a similar population of stable outpatients with asthma, children with exacerbating asthma would have differing metabolite abundance patterns with distinct clustering profiles. Methods A total of 98 children aged 6 through 17 years with exacerbating asthma (n = 69) and stable asthma (n = 29) underwent clinical characterization procedures and submitted plasma samples for metabolomic analyses. High-confidence metabolites were retained and utilized for pathway enrichment analyses to identify the most relevant metabolic pathways that discriminated between groups. Results In all, 118 and 131 high-confidence metabolites were identified in positive and negative ionization mode, respectively. A total of 103 unique metabolites differed significantly between children with exacerbating asthma and children with stable asthma. In all, 8 significantly enriched pathways that were largely associated with alterations in arginine, phenylalanine, and glycine metabolism were identified. However, other metabolites and pathways of interest were also identified. Conclusion Metabolomic analyses identified multiple perturbed metabolites and pathways that discriminated children with exacerbating asthma who were hospitalized for status asthmaticus. These results highlight the complex biology of inflammation in children with exacerbating asthma and argue for additional studies of the metabolic determinants of asthma exacerbations in children because many of the identified metabolites of interest may be amenable to targeted interventions.
Collapse
Affiliation(s)
| | - Joshua D. Chandler
- Department of Pediatrics, Emory University, Atlanta
- Children’s Healthcare of Atlanta
| | - Seibi Kobara
- Department of Biomedical Informatics, Emory University, Atlanta
| | | | | | | | | | | | | | - Rishikesan Kamaleswaran
- Department of Pediatrics, Emory University, Atlanta
- Department of Biomedical Informatics, Emory University, Atlanta
| | - Anne M. Fitzpatrick
- Department of Pediatrics, Emory University, Atlanta
- Children’s Healthcare of Atlanta
| | - Jocelyn R. Grunwell
- Department of Pediatrics, Emory University, Atlanta
- Children’s Healthcare of Atlanta
| |
Collapse
|
8
|
Packi K, Matysiak J, Plewa S, Klupczyńska-Gabryszak A, Matuszewska E, Rzetecka N, Bręborowicz A, Matysiak J. Amino Acid Profiling Identifies Disease-Specific Signatures in IgE-Mediated and Non-IgE-Mediated Food Allergy in Pediatric Patients with Atopic Dermatitis. Biomedicines 2023; 11:1919. [PMID: 37509558 PMCID: PMC10377369 DOI: 10.3390/biomedicines11071919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/02/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
An IgE-mediated food allergy (FA) in atopic dermatitis (AD) children should be easily differentiated from other immune-mediated adverse effects related to food. Specific IgEs for particular protein components has provided additional diagnostic value. However, component-resolved diagnostics (CRD) has not solved all diagnostic problems either. We analysed the serum profile of 42 amino acids (AAs) in 76 AD children aged 2-60 months with an IgE-mediated FA (n = 36), with a non-IgE-mediated FA (n = 15) and without an FA (n = 25) using high-performance liquid chromatography coupled with mass spectrometry (LC-MS/MS) and an aTRAQ kit. We identified homocitrulline (Hcit), sarcosine (Sar) and L-tyrosine (Tyr) as features that differentiated the studied groups (one-way ANOVA with least significant difference post hoc test). The Hcit concentrations in the non-IgE-mediated FA group were significantly decreased compared with the IgE-mediated FA group (p = 0.018) and the control group (p = 0.008). In AD children with a non-IgE-mediated FA, the Tyr levels were also significantly reduced compared with the controls (p = 0.009). The mean concentration of Sar was the highest in the non-IgE-mediated FA group and the lowest in the IgE-mediated FA group (p = 0.047). Future studies should elucidate the involvement of these AAs in the molecular pathway of IgE- and non-IgE-mediated allergic responses.
Collapse
Affiliation(s)
- Kacper Packi
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-806 Poznan, Poland
- AllerGen, Center of Personalized Medicine, 97-300 Piotrkow Trybunalski, Poland
| | - Joanna Matysiak
- Faculty of Health Sciences, Calisia University-Kalisz, 62-800 Kalisz, Poland
| | - Szymon Plewa
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | | | - Eliza Matuszewska
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Natalia Rzetecka
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Anna Bręborowicz
- Department of Pulmonology, Pediatric Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Jan Matysiak
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| |
Collapse
|
9
|
Althoff MD, Peterson R, McGrath M, Jin Y, Grasemann H, Sharma S, Federman A, Wisnivesky JP, Holguin F. Phenotypic characteristics of asthma and morbidity are associated with distinct longitudinal changes in L-arginine metabolism. BMJ Open Respir Res 2023; 10:e001683. [PMID: 37270184 PMCID: PMC10254613 DOI: 10.1136/bmjresp-2023-001683] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/12/2023] [Indexed: 06/05/2023] Open
Abstract
BACKGROUND The L-arginine metabolome is dysregulated in asthma, though it is not understood how longitudinal changes in L-arginine metabolism differ among asthma phenotypes and relate to disease outcomes. OBJECTIVES To determine the longitudinal associations between phenotypic characteristics with L-arginine metabolites and their relationships with asthma morbidity. METHODS This is a prospective cohort study of 321 patients with asthma followed semiannually for over 18 months with assessments of plasma L-arginine metabolites, asthma control, spirometry, quality of life and exacerbations. Metabolite concentrations and ratios were transformed using the natural logarithm. RESULTS There were many differences in L-arginine metabolism among asthma phenotypes in the adjusted models. Increasing body mass index was associated with increased asymmetric dimethylarginine (ADMA) and depleted L-citrulline. Latinx was associated with increased metabolism via arginase, with higher L-ornithine, proline and L-ornithine/L-citrulline levels, and was found to have higher L-arginine availability compared with white race. With respect to asthma outcomes, increasing L-citrulline was associated with improved asthma control and increasing L-arginine and L-arginine/ADMA were associated with improved quality of life. Increased variability in L-arginine, L-arginine/ADMA, L-arginine/L-ornithine and L-arginine availability index over 12 months were associated with increased exacerbations, OR 4.70 (95% CI 1.35 to 16.37), OR 8.69 (95% CI 1.98 to 38.08), OR 4.17 (95% CI 1.40 to 12.41) and OR 4.95 (95% CI 1.42 to 17.16), respectively. CONCLUSIONS Our findings suggest that L-arginine metabolism is associated with multiple measures of asthma control and may explain, in part, the relationship between age, race/ethnicity and obesity with asthma outcomes.
Collapse
Affiliation(s)
- Meghan Dolan Althoff
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ryan Peterson
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Max McGrath
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Ying Jin
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Hartmut Grasemann
- Respiratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sunita Sharma
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Alex Federman
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Juan Pablo Wisnivesky
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fernando Holguin
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
10
|
Starikova EA, Rubinstein AA, Mammedova JT, Isakov DV, Kudryavtsev IV. Regulated Arginine Metabolism in Immunopathogenesis of a Wide Range of Diseases: Is There a Way to Pass between Scylla and Charybdis? Curr Issues Mol Biol 2023; 45:3525-3551. [PMID: 37185755 PMCID: PMC10137093 DOI: 10.3390/cimb45040231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
More than a century has passed since arginine was discovered, but the metabolism of the amino acid never ceases to amaze researchers. Being a conditionally essential amino acid, arginine performs many important homeostatic functions in the body; it is involved in the regulation of the cardiovascular system and regeneration processes. In recent years, more and more facts have been accumulating that demonstrate a close relationship between arginine metabolic pathways and immune responses. This opens new opportunities for the development of original ways to treat diseases associated with suppressed or increased activity of the immune system. In this review, we analyze the literature describing the role of arginine metabolism in the immunopathogenesis of a wide range of diseases, and discuss arginine-dependent processes as a possible target for therapeutic approaches.
Collapse
Affiliation(s)
- Eleonora A Starikova
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L'va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Artem A Rubinstein
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
| | - Jennet T Mammedova
- Laboratory of General Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
| | - Dmitry V Isakov
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L'va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Igor V Kudryavtsev
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
- School of Biomedicine, Far Eastern Federal University, FEFU Campus, 10 Ajax Bay, Russky Island, 690922 Vladivostok, Russia
| |
Collapse
|
11
|
Weber R, Streckenbach B, Welti L, Inci D, Kohler M, Perkins N, Zenobi R, Micic S, Moeller A. Online breath analysis with SESI/HRMS for metabolic signatures in children with allergic asthma. Front Mol Biosci 2023; 10:1154536. [PMID: 37065443 PMCID: PMC10102578 DOI: 10.3389/fmolb.2023.1154536] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/16/2023] [Indexed: 04/03/2023] Open
Abstract
Introduction: There is a need to improve the diagnosis and management of pediatric asthma. Breath analysis aims to address this by non-invasively assessing altered metabolism and disease-associated processes. Our goal was to identify exhaled metabolic signatures that distinguish children with allergic asthma from healthy controls using secondary electrospray ionization high-resolution mass spectrometry (SESI/HRMS) in a cross-sectional observational study. Methods: Breath analysis was performed with SESI/HRMS. Significant differentially expressed mass-to-charge features in breath were extracted using the empirical Bayes moderated t-statistics test. Corresponding molecules were putatively annotated by tandem mass spectrometry database matching and pathway analysis. Results: 48 allergic asthmatics and 56 healthy controls were included in the study. Among 375 significant mass-to-charge features, 134 were putatively identified. Many of these could be grouped to metabolites of common pathways or chemical families. We found several pathways that are well-represented by the significant metabolites, for example, lysine degradation elevated and two arginine pathways downregulated in the asthmatic group. Assessing the ability of breath profiles to classify samples as asthmatic or healthy with supervised machine learning in a 10 times repeated 10-fold cross-validation revealed an area under the receiver operating characteristic curve of 0.83. Discussion: For the first time, a large number of breath-derived metabolites that discriminate children with allergic asthma from healthy controls were identified by online breath analysis. Many are linked to well-described metabolic pathways and chemical families involved in pathophysiological processes of asthma. Furthermore, a subset of these volatile organic compounds showed high potential for clinical diagnostic applications.
Collapse
Affiliation(s)
- Ronja Weber
- Department of Respiratory Medicine, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Bettina Streckenbach
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Lara Welti
- Department of Respiratory Medicine, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Demet Inci
- Department of Respiratory Medicine, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Malcolm Kohler
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
| | - Nathan Perkins
- Division of Clinical Chemistry and Biochemistry, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Renato Zenobi
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Srdjan Micic
- Department of Respiratory Medicine, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Alexander Moeller
- Department of Respiratory Medicine, University Children’s Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Cottrill KA, Stephenson ST, Mohammad AF, Kim SO, McCarty NA, Kamaleswaran R, Fitzpatrick AM, Chandler JD. Exacerbation-prone pediatric asthma is associated with arginine, lysine, and methionine pathway alterations. J Allergy Clin Immunol 2023; 151:118-127.e10. [PMID: 36096204 PMCID: PMC9825634 DOI: 10.1016/j.jaci.2022.07.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND The asthma of some children remains poorly controlled, with recurrent exacerbations despite treatment with inhaled corticosteroids. Aside from prior exacerbations, there are currently no reliable predictors of exacerbation-prone asthma in these children and only a limited understanding of the potential underlying mechanisms. OBJECTIVE We sought to quantify small molecules in the plasma of children with exacerbation-prone asthma through mass spectrometry-based metabolomics. We hypothesized that the plasma metabolome of these children would differ from that of children with non-exacerbation-prone asthma. METHODS Plasma metabolites were extracted from 4 pediatric asthma cohorts (215 total subjects, with 41 having exacerbation-prone asthma) and detected with a mass spectrometer. High-confidence annotations were retained for univariate analysis and were confirmed by a sensitivity analysis in subjects receiving high-dose inhaled corticosteroids. Metabolites that varied by cohort were excluded. MetaboAnalyst software was used to identify pathways of interest. Concentrations were calculated by reference standardization. RESULTS We identified 32 unique, cohort-independent metabolites that differed in children with exacerbation-prone asthma compared to children with non-exacerbation-prone asthma. Comparison of metabolite concentrations to literature-reported values for healthy children revealed that most metabolites were decreased in both asthma groups, but more so in exacerbation-prone asthma. Pathway analysis identified arginine, lysine, and methionine pathways as most impacted. CONCLUSIONS Several plasma metabolites are perturbed in children with exacerbation-prone asthma and are largely related to arginine, lysine, and methionine pathways. While validation is needed, plasma metabolites may be potential biomarkers for exacerbation-prone asthma in children.
Collapse
Affiliation(s)
| | | | | | - Susan O Kim
- Department of Pediatrics, Emory University, Atlanta, Ga
| | | | - Rishikesan Kamaleswaran
- Department of Pediatrics, Emory University, Atlanta, Ga; Department of Biomedical Informatics, Emory University, Atlanta, Ga
| | - Anne M Fitzpatrick
- Department of Pediatrics, Emory University, Atlanta, Ga; Children's Healthcare of Atlanta, Atlanta, Ga
| | - Joshua D Chandler
- Department of Pediatrics, Emory University, Atlanta, Ga; Children's Healthcare of Atlanta, Atlanta, Ga.
| |
Collapse
|
13
|
Momma TY, Ottaviani JI. There is no direct competition between arginase and nitric oxide synthase for the common substrate l-arginine. Nitric Oxide 2022; 129:16-24. [PMID: 36126859 DOI: 10.1016/j.niox.2022.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/08/2022] [Accepted: 09/10/2022] [Indexed: 11/19/2022]
Abstract
AIMS Extrahepatic arginases are postulated to be involved in cardiovascular-related pathologies by competing with nitric oxide synthase (NOS) for the common substrate l-arginine, subsequently decreasing nitric oxide production. However, previous models used to study arginase and NOS competition did not account for steady state level of l-arginine pool, which is dependent on conditions of l-arginine supply and utilization pathways. This work aimed at revisiting the concept of NOS and arginase competition while considering different conditions of l-arginine supply and l-arginine utilization pathways. METHODS AND RESULTS Mouse macrophage-like RAW cells and human vascular endothelial cells co-expressing NOS and arginase were used to reevaluate the concept of substrate competition between arginase and NOS under conditions of l-arginine supply that mimicked either a continuous (similar to in vivo conditions) or a limited supply (similar to previous in vitro models). Enzyme kinetics simulation models were used to gain mechanistic insight and to evaluate the tenability of a substrate competition between the two enzymes. In addition to arginase and NOS, other l-arginine pathways such as transporters and utilization towards protein synthesis were considered to understand the intricacies of l-arginine metabolism. Our results indicate that when there is a continuous supply of l-arginine, as is the case for most cells in vivo, arginase does not affect NOS activity by a substrate competition. Furthermore, we demonstrate that l-arginine pathways such as transporters and protein synthesis are more likely to affect NOS activity than arginase. CONCLUSIONS Arginase does not outcompete NOS for the common substrate l-arginine. Findings from this study should be considered to better understand the role of arginase in certain pathologies and for the interpretation of in vivo studies with arginase inhibitors.
Collapse
Affiliation(s)
- Tony Y Momma
- College of Agricultural and Environmental Sciences, University of California, Davis, CA, 95616, USA.
| | - Javier I Ottaviani
- College of Agricultural and Environmental Sciences, University of California, Davis, CA, 95616, USA; Mars Inc., McLean, VA, 22101, USA
| |
Collapse
|
14
|
Matysiak J, Packi K, Klimczak S, Bukowska P, Matuszewska E, Klupczyńska-Gabryszak A, Bręborowicz A, Matysiak J. Cytokine profile in childhood asthma. JOURNAL OF MEDICAL SCIENCE 2022. [DOI: 10.20883/medical.e725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Childhood asthma is a chronic airway disease, which pathogenesis is markedly heterogeneous–with multiple phenotypes defining visible characteristics and endotypes defining molecular mechanisms. Cytokines and chemokines released during inflammatory responses are key immune mediators. The cytokine response can largely determine the susceptibility to childhood asthma and its severity. The purpose of this study was to characterize the immune profile of childhood asthma. The study involved 26 children (3–18 years old), who were divided into 2 groups: study–with childhood asthma; control–without asthma. The innovative Bio-Plex method was used to determine the serum concentration of 37 inflammatory proteins in one experiment. The results were analyzed using univariate statistical tests. In the study group, the level of the 10 tested markers increased, while the level of the remaining 9 decreased compared to the control; a statistically significant reduction in concentration was obtained only for the MMP-1(p<0.05). According to the ROC curve, MMP-1 can be considered an effective discriminator of childhood asthma (p<0.05; AUC=0.752). Cytokines/chemokines may be useful in the diagnosis of childhood asthma and may also become a prognostic target in determining the phenotype/endotype of this condition. This study should be a prelude to and an incentive for more complex proteomic analyzes.
Collapse
|
15
|
Characterization of the L-Arginine/Nitric Oxide Pathway and Oxidative Stress in Pediatric Patients with Atopic Diseases. Int J Mol Sci 2022; 23:ijms23042136. [PMID: 35216250 PMCID: PMC8878534 DOI: 10.3390/ijms23042136] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 01/27/2023] Open
Abstract
Introduction: L-Arginine (Arg) is a semi-essential amino acid. Constitutive and inducible nitric oxide synthase (NOS) isoforms convert Arg to nitric oxide (NO), a potent vaso- and bronchodilator with multiple biological functions. Atopic dermatitis (AD) and bronchial asthma (BA) are atopic diseases affecting many children globally. Several studies analyzed NO in airways, yet the systemic synthesis of NO in AD and BA in children with BA, AD or both is elusive. Methods: In a multicenter study, blood and urine were obtained from 130 of 302 participating children for the measurement of metabolites of the Arg/NO pathway (BA 31.5%; AD 5.4%; AD + BA 36.1%; attention deficit hyperactivity disorder (ADHD) 12.3%). In plasma and urine amino acids Arg and homoarginine (hArg), both substrates of NOS, asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA), both inhibitors of NOS, dimethylamine (DMA), and nitrite and nitrate, were measured by gas chromatography–mass spectrometry. Malondialdehyde (MDA) was measured in plasma and urine samples to evaluate possible effects of oxidative stress. Results: There were no differences in the Arg/NO pathway between the groups of children with different atopic diseases. In comparison to children with ADHD, children with AD, BA or AD and BA had higher plasma nitrite (p < 0.001) and nitrate (p < 0.001) concentrations, suggesting higher systemic NO synthesis in AD and BA. Urinary excretion of DMA was also higher (p = 0.028) in AD and BA compared to patients with ADHD, suggesting elevated ADMA metabolization. Discussion/Conclusion: The Arg/NO pathway is activated in atopic diseases independent of severity. Systemic NO synthesis is increased in children with an atopic disease. Plasma and urinary MDA levels did not differ between the groups, suggesting no effect of oxidative stress on the Arg/NO pathway in atopic diseases.
Collapse
|
16
|
Morris CR, Kuypers FA, Hagar R, Larkin S, Lavrisha L, Saulys A, Vichinsky EP, Suh JH. Implications for the Metabolic Fate of Oral Glutamine Supplementation within Plasma and Erythrocytes of Patients with Sickle Cell Disease: A Pharmacokinetics Study. Complement Ther Med 2022; 64:102803. [PMID: 35032556 DOI: 10.1016/j.ctim.2022.102803] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 11/13/2021] [Accepted: 01/10/2022] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVES L-Glutamine is FDA-approved for sickle cell disease (SCD), yet the mechanism(s)-of-action are poorly understood. We performed a pharmacokinetics (pK) study to determine the metabolic fate of glutamine supplementation on plasma and erythrocyte amino acids in patients with SCD. DESIGN A pK study was performed where patients with SCD fasting for >8hours received oral L-glutamine (10 grams). Blood was analyzed at baseline, 30/60/90minutes/2/3/4/8 hrs. A standardized diet was administered to all participants at 3 established time-points (after 2/5/7hrs). A subset of patients also had pK studies performed without glutamine supplementation to follow normal diurnal fluctuations in amino acids. SETTING Comprehensive SCD Center in Oakland, California RESULTS: Five patients with SCD were included, three of whom performed pK studies both with and without glutamine supplementation. Average age was 50.6 ± 5.6 years, 60% were female, 40% SS, 60% SC. Plasma glutamine levels increased significantly after oral glutamine supplementation, compared to minimal fluctuations with diet. Plasma glutamine concentration peaked within 30-minutes of ingestion (p=0.01) before decreasing to a plateau by 2-hours that remained higher than baseline by 8hours. Oral glutamine also increased plasma arginine concentration, which peaked by 4-hrs (p=0.03) and remained elevated through 8-hrs. Erythrocyte glutamine levels began to increase by 8-hours, while erythrocyte arginine concentration peaked at 4-hours. CONCLUSIONS Oral glutamine supplementation acutely improved glutamine and arginine bioavailability in both plasma and erythrocytes. This is the first study to demonstrate that glutamine therapy increases arginine bioavailability and may provide insight into shared mechanisms-of-action between these conditionally-essential amino acids.
Collapse
Affiliation(s)
- Claudia R Morris
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA; Children's Healthcare of Atlanta, Atlanta, Georgia, USA.
| | - Frans A Kuypers
- Department of Hematology/Oncology, UCSF-Benioff Children's Hospital Oakland, Oakland, CA, USA
| | - Robert Hagar
- Department of Hematology/Oncology, UCSF-Benioff Children's Hospital Oakland, Oakland, CA, USA
| | - Sandra Larkin
- Department of Hematology/Oncology, UCSF-Benioff Children's Hospital Oakland, Oakland, CA, USA
| | - Lisa Lavrisha
- Department of Hematology/Oncology, UCSF-Benioff Children's Hospital Oakland, Oakland, CA, USA
| | - Augusta Saulys
- Department of Emergency Medicine, UCSF-Benioff Children's Hospital Oakland, Oakland, CA, USA
| | - Elliott P Vichinsky
- Department of Hematology/Oncology, UCSF-Benioff Children's Hospital Oakland, Oakland, CA, USA
| | - Jung H Suh
- Department of Hematology/Oncology, UCSF-Benioff Children's Hospital Oakland, Oakland, CA, USA
| |
Collapse
|
17
|
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
|
18
|
Wang C, Jiang S, Zhang S, Ouyang Z, Wang G, Wang F. Research Progress of Metabolomics in Asthma. Metabolites 2021; 11:567. [PMID: 34564383 PMCID: PMC8466166 DOI: 10.3390/metabo11090567] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/10/2021] [Accepted: 08/19/2021] [Indexed: 12/25/2022] Open
Abstract
Asthma is a highly heterogeneous disease, but the pathogenesis of asthma is still unclear. It is well known that the airway inflammatory immune response is the pathological basis of asthma. Metabolomics is a systems biology method to analyze the difference of low molecular weight metabolites (<1.5 kDa) and explore the relationship between metabolic small molecules and pathophysiological changes of the organisms. The functional interdependence between immune response and metabolic regulation is one of the cores of the body's steady-state regulation, and its dysfunction will lead to a series of metabolic disorders. The signal transduction effect of specific metabolites may affect the occurrence of the airway inflammatory immune response, which may be closely related to the pathogenesis of asthma. Emerging metabolomic analysis may provide insights into the pathogenesis and diagnosis of asthma. The review aims to analyze the changes of metabolites in blood/serum/plasma, urine, lung tissue, and exhaled breath condensate (EBC) samples, and further reveals the potential pathogenesis of asthma according to the disordered metabolic pathways.
Collapse
Affiliation(s)
- Chao Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (C.W.); (S.J.); (S.Z.)
| | - Shengyu Jiang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (C.W.); (S.J.); (S.Z.)
| | - Siyu Zhang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (C.W.); (S.J.); (S.Z.)
| | - Zhuoer Ouyang
- Department of Cellular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China;
| | - Guoqiang Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (C.W.); (S.J.); (S.Z.)
| | - Fang Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (C.W.); (S.J.); (S.Z.)
| |
Collapse
|
19
|
Zou XL, Wu JJ, Ye HX, Feng DY, Meng P, Yang HL, Wu WB, Li HT, He Z, Zhang TT. Associations Between Gut Microbiota and Asthma Endotypes: A Cross-Sectional Study in South China Based on Patients with Newly Diagnosed Asthma. J Asthma Allergy 2021; 14:981-992. [PMID: 34408443 PMCID: PMC8367087 DOI: 10.2147/jaa.s320088] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/26/2021] [Indexed: 12/16/2022] Open
Abstract
Objective This study aimed to investigate the gut microbiome profile in different inflammatory phenotypes of treatment-naive newly diagnosed asthmatic adults, to gain insight on the associations between intestinal microbiota and phenotypic features that characterize asthma heterogeneity to develop new treatments for asthma. Methods Fresh stool samples were obtained from 20 healthy subjects and 47 newly diagnosed asthmatic patients prior to any interventions. The asthmatics were divided into allergic and non-allergic cohorts. Intestinal microbiota was analyzed by 16S rRNA next-generation sequencing. Demographic and clinical parameters were collected. Alpha and beta diversity analysis were calculated to detect differences within sample phylotype richness and evenness between controls and asthmatic patients. Statistically significant differences between samples were analyzed for all used metrics, and features of gut bacterial community structure were evaluated in relation to extensive clinical characteristics of asthmatic patients. Results Gut microbial compositions were significantly different between asthmatic and healthy groups. Alpha-diversity of the gut microbiome was significantly lower in asthmatics than in controls. The microbiome between allergic and non-allergic asthmatic patients were also different, and 28 differential species were identified. PPAR signaling pathway, carotenoid biosynthesis, and flavonoid biosynthesis were significantly positively correlated with allergy-associated clinical index, including FENO value, blood eosinophil counts, and serum IgE and IL-4 levels. A combination of Ruminococcus bromii, Brevundimonas vesicularis, and Clostridium disporicum showed an AUC of 0.743 in the specific allergic/non-allergic cohort. When integrating C. disporicum, flavone, flavonol biosynthesis, and serum IL-4 values, the AUC achieved 0.929 to classify asthmatics. At the same time, C. colinum and its associated functional pathway exhibited an AUC of 0.78 to distinguish allergic asthmatics from those without allergies. Conclusion We demonstrated a distinct taxonomic composition of gut microbiota in different asthmatic phenotypes, highlighting their significant relationships. Our study may support considerations of intestinal microbial signatures in delineating asthma phenotypes.
Collapse
Affiliation(s)
- Xiao-Ling Zou
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jin-Jie Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hui-Xia Ye
- Department of Gynecology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ding-Yun Feng
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ping Meng
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hai-Ling Yang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wen-Bin Wu
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hong-Tao Li
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhen He
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Tian-Tuo Zhang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
20
|
Ritz T, Salsman ML, Young DA, Lippert AR, Khan DA, Ginty AT. Boosting nitric oxide in stress and respiratory infection: Potential relevance for asthma and COVID-19. Brain Behav Immun Health 2021; 14:100255. [PMID: 33842899 PMCID: PMC8019595 DOI: 10.1016/j.bbih.2021.100255] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 01/12/2023] Open
Abstract
Nitric oxide (NO) is a ubiquitous signaling molecule that is critical for supporting a plethora of processes in biological organisms. Among these, its role in the innate immune system as a first line of defense against pathogens has received less attention. In asthma, levels of exhaled NO have been utilized as a window into airway inflammation caused by allergic processes. However, respiratory infections count among the most important triggers of disease exacerbations. Among the multitude of factors that affect NO levels are psychological processes. In particular, longer lasting states of psychological stress and depression have been shown to attenuate NO production. The novel SARS-CoV-2 virus, which has caused a pandemic, and with that, sustained levels of psychological stress globally, also adversely affects NO signaling. We review evidence on the role of NO in respiratory infection, including COVID-19, and stress, and argue that boosting NO bioavailability may be beneficial in protection from infections, thus benefitting individuals who suffer from stress in asthma or SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Thomas Ritz
- Department of Psychology, Southern Methodist University, 6116 N. Central Expressway, Suite 1160, Dallas, TX, USA
| | - Margot L Salsman
- Department of Psychology, Southern Methodist University, 6116 N. Central Expressway, Suite 1160, Dallas, TX, USA
| | - Danielle A Young
- Department of Psychology and Neuroscience, Baylor University, One Bear Place, 97334, Baylor Sciences Building, Suite B.309, Waco, TX, USA
| | - Alexander R Lippert
- Department of Chemistry, Southern Methodist University, Fondren Science Building 303, P.O. Box, 750314, Dallas, TX, USA
| | - Dave A Khan
- Department of Internal Medicine, Allergy and Immunology, The University of Texas Southwestern Medical Center, 5323, Harry Hines Blvd., Dallas, TX, USA
| | - Annie T Ginty
- Department of Psychology and Neuroscience, Baylor University, One Bear Place, 97334, Baylor Sciences Building, Suite B.309, Waco, TX, USA
| |
Collapse
|
21
|
Abstract
Low plasma arginine bioavailability has been implicated in endothelial dysfunction and immune dysregulation. The role of arginine in COVID-19 is unknown, but could contribute to cellular damage if low. Our objective was to determine arginine bioavailability in adults and children with COVID-19 vs. healthy controls. We hypothesized that arginine bioavailability would be low in patients with COVID-19 and multisystem inflammatory syndrome in children (MIS-C). We conducted a prospective observational study of three patient cohorts; arginine bioavailability was determined in asymptomatic healthy controls, adults hospitalized with COVID-19, and hospitalized children/adolescents <21 y old with COVID-19, MIS-C, or asymptomatic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection identified on admission screen. Mean patient plasma amino acids were compared to controls using the Student’s t test. Arginine-to-ornithine ratio, a biomarker of arginase activity, and global arginine bioavailability ratio (GABR, arginine/[ornithine+citrulline]) were assessed in all three groups. A total of 80 patients were included (28 controls, 32 adults with COVID-19, and 20 pediatric patients with COVID-19/MIS-C). Mean plasma arginine and arginine bioavailability ratios were lower among adult and pediatric patients with COVID-19/MIS-C compared to controls. There was no difference between arginine bioavailability in children with COVID-19 vs. MIS-C. Adults and children with COVID-19 and MIS-C in our cohort had low arginine bioavailability compared to healthy adult controls. This may contribute to immune dysregulation and endothelial dysfunction in COVID-19. Low arginine-to-ornithine ratio in patients with COVID-19 or MIS-C suggests an elevation of arginase activity. Further study is merited to explore the role of arginine dysregulation in COVID-19.
Collapse
|
22
|
Marei ES, El-Maghraby DF, Ashry K. Correlation of vitamin D and asymmetric dimethylarginine in children with bronchial asthma. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2021. [DOI: 10.1080/16878507.2021.1923898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Elham S. Marei
- Health Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Dina F. El-Maghraby
- Health Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Khaled Ashry
- Health Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
23
|
Fitzpatrick AM, Chipps BE, Holguin F, Woodruff PG. T2-"Low" Asthma: Overview and Management Strategies. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 8:452-463. [PMID: 32037109 DOI: 10.1016/j.jaip.2019.11.006] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/11/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023]
Abstract
Although the term "asthma" has been applied to all patients with airway lability and variable chest symptoms for centuries, phenotypes of asthma with distinct clinical and molecular features that may warrant different treatment approaches are well recognized. Patients with type 2 (T2)-"high" asthma are characterized by upregulation of T2 immune pathways (ie, IL-4 and IL-13 gene sets) and eosinophilic airway inflammation, whereas these features are absent in patients with T2-"low" asthma and may contribute to poor responsiveness to corticosteroid treatment. This review details definitions and clinical features of T2-"low" asthma, potential mechanisms and metabolic aspects, pediatric considerations, and potential treatment approaches. Priority research questions for T2-"low" asthma are also discussed.
Collapse
Affiliation(s)
| | - Bradley E Chipps
- Capital Allergy and Respiratory Disease Center, Sacramento, Calif
| | - Fernando Holguin
- University of Colorado, Pulmonary Sciences and Critical Care Medicine, Denver, Colo
| | - Prescott G Woodruff
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, and the Cardiovascular Research Institute, University of California, San Francisco, Calif
| |
Collapse
|
24
|
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
|
25
|
Scott JA, Maarsingh H, Holguin F, Grasemann H. Arginine Therapy for Lung Diseases. Front Pharmacol 2021; 12:627503. [PMID: 33833679 PMCID: PMC8022134 DOI: 10.3389/fphar.2021.627503] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/04/2021] [Indexed: 12/15/2022] Open
Abstract
Nitric oxide (NO) is produced by a family of isoenzymes, nitric oxide synthases (NOSs), which all utilize L-arginine as substrate. The production of NO in the lung and airways can play a number of roles during lung development, regulates airway and vascular smooth muscle tone, and is involved in inflammatory processes and host defense. Altered L-arginine/NO homeostasis, due to the accumulation of endogenous NOS inhibitors and competition for substrate with the arginase enzymes, has been found to play a role in various conditions affecting the lung and in pulmonary diseases, such as asthma, chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), pulmonary hypertension, and bronchopulmonary dysplasia. Different therapeutic strategies to increase L-arginine levels or bioavailability are currently being explored in pre-clinical and clinical studies. These include supplementation of L-arginine or L-citrulline and inhibition of arginase.
Collapse
Affiliation(s)
- Jeremy A Scott
- Occupational and Environmental Health, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Harm Maarsingh
- Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, FL, United States
| | - Fernando Holguin
- Division of Pulmonary Sciences and Critical Care, University of Colorado, Aurora, CO, United States
| | - Hartmut Grasemann
- Division of Respiratory Medicine, Department of Paediatrics and Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
26
|
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
|
27
|
Piyadasa H, Lloyd D, Lee AHY, Altieri A, Hemshekhar M, Osawa N, Basu S, Blimkie T, Falsafi R, Halayko AJ, Hancock REW, Mookherjee N. Characterization of immune responses and the lung transcriptome in a murine model of IL-33 challenge. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165950. [PMID: 32841733 DOI: 10.1016/j.bbadis.2020.165950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/26/2020] [Accepted: 08/20/2020] [Indexed: 12/25/2022]
Abstract
IL-33 induces airway inflammation and hyper-responsiveness in respiratory diseases. Although defined as a therapeutic target, there are limited studies that have comprehensively investigated IL-33-mediated responses in the lungs in vivo. In this study, we characterized immunological and physiological responses induced by intranasal IL-33 challenge, in a mouse model. We identified specific cytokines, IL-4, IL-5, IL-6, IL-10, IP-10 and MIP1-α, that are increased in bronchoalveolar lavage and lung tissues by IL-33. Using transcriptomics (RNA-Seq) we demonstrated that 2279 transcripts were up-regulated and 1378 downregulated (≥ 2-fold, p < 0.01) in lung tissues, in response to IL-33. Bioinformatic interrogation of the RNA-Seq data was used to predict biological pathways and upstream regulators involved in IL-33-mediated responses. We showed that the mRNA and protein of STAT4, a predicted upstream regulator of IL-33-induced transcripts, was significantly enhanced in the lungs following IL-33 challenge. Overall, this study provides specific IL-33-induced molecular targets and endpoints that can be used as a resource for in vivo studies, e.g. in preclinical murine models examining novel interventions to target downstream effects of IL-33.
Collapse
Affiliation(s)
- Hadeesha Piyadasa
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Canada; Department of Immunology, University of Manitoba, Winnipeg, Canada
| | - Dylan Lloyd
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Amy H Y Lee
- Centre for Microbial Disease and Immunity Research, University of British Columbia, Vancouver, Canada; Department of Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Anthony Altieri
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Canada; Department of Immunology, University of Manitoba, Winnipeg, Canada
| | - Mahadevappa Hemshekhar
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Natasha Osawa
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Sujata Basu
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada; Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Travis Blimkie
- Centre for Microbial Disease and Immunity Research, University of British Columbia, Vancouver, Canada
| | - Reza Falsafi
- Centre for Microbial Disease and Immunity Research, University of British Columbia, Vancouver, Canada
| | - Andrew J Halayko
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada; Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Robert E W Hancock
- Centre for Microbial Disease and Immunity Research, University of British Columbia, Vancouver, Canada
| | - Neeloffer Mookherjee
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Canada; Department of Immunology, University of Manitoba, Winnipeg, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg, Canada.
| |
Collapse
|
28
|
Fernando Y, Wickramasinghe P, De Silva U, Alahakoon M, Anuradha KWDA, Handunnetti S. Differences in serum markers of oxidative stress in well controlled and poorly controlled asthma in Sri Lankan children: a pilot study. Allergy Asthma Clin Immunol 2020; 16:66. [PMID: 32944026 PMCID: PMC7491346 DOI: 10.1186/s13223-020-00463-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/10/2020] [Indexed: 12/22/2022] Open
Abstract
Background Asthma is a disease characterised by hyper responsiveness and bronchoconstriction of airways, and is a major health burden globally. A dysfunction of the oxidant-antioxidant balance, termed oxidative stress, has been implicated in the pathophysiology of asthma. The present study aims to assess the changes in oxidative stress markers, namely nitric oxide metabolites and antioxidant capacity, in children with poorly controlled and well controlled asthma, in comparison to healthy controls. Methods The present study enrolled 72 children (ages 5-15 years) classified into three groups: (1) poorly controlled asthma (n = 20), (2) well controlled asthma (n = 24) and (3) healthy controls (n = 27). An interviewer-administered questionnaire was used to record socio-demographic data of the participants. The serum concentrations of the oxidant markers (nitrite, nitrate and total nitric oxide metabolites [NOx]) were determined using the Griess test, and the total antioxidant capacity (TAOC) was determined using the ABTS decolorisation method. The concentrations of these markers were compared across the three groups. Results The three study groups were similar in terms of socio-demographic data. The differences across the three groups were statistically significant for serum concentrations of nitrate and NOx (but not nitrite) and serum TAOC. Further analyses showed that the disparity for nitrate and NOx concentrations was greatest between poorly controlled asthma and healthy controls (p = 0.001 and p < 0.001) compared to the well-controlled asthmatics and healthy controls (p = 0.036 and p = 0.049). A significant difference in serum nitrate and NOx concentrations was not observed between the two asthma groups (p = 0.311 and 0.203). The TAOC were significantly lower in poorly controlled asthmatics as compared to well-controlled asthmatics (p = 0.003) and healthy controls (p < 0.001). However, there was no significant difference in the serum TAOC between healthy controls and well-controlled asthmatics (p = 0.496). These findings may indicate that it is perhaps the higher TAOC that contributes to the well controlled state of asthma. Conclusions The present study indicated that an imbalance of oxidants and antioxidants in the serum may have an underlying role in asthma pathophysiology, and how these markers may be effective in asthma management.
Collapse
Affiliation(s)
- Yenuli Fernando
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka.,Colombo International School, Colombo, 7 Sri Lanka
| | - Pujitha Wickramasinghe
- Department of Paediatrics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Udani De Silva
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka
| | | | - K W D A Anuradha
- Department of Paediatrics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Shiroma Handunnetti
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka
| |
Collapse
|
29
|
Wieczfinska J, Sitarek P, Kowalczyk T, Pawliczak R. Leonurus sibiricus root extracts decrease airway remodeling markers expression in fibroblasts. Clin Exp Immunol 2020; 202:28-46. [PMID: 32562256 DOI: 10.1111/cei.13481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/27/2020] [Accepted: 06/12/2020] [Indexed: 12/15/2022] Open
Abstract
Bronchial asthma is believed to be provoked by the interaction between airway inflammation and remodeling. Airway remodeling is a complex and poorly understood process, and controlling it appears key for halting the progression of asthma and other obstructive lung diseases. Plants synthesize a number of valuable compounds as constitutive products and as secondary metabolites, many of which have curative properties. The aim of this study was to evaluate the anti-remodeling properties of extracts from transformed and transgenic Leonurus sibiricus roots with transformed L. sibiricus roots extract with transcriptional factor AtPAP1 overexpression (AtPAP1). Two fibroblast cell lines, Wistar Institute-38 (WI-38) and human fetal lung fibroblast (HFL1), were incubated with extracts from transformed L. sibiricus roots (TR) and roots with transcriptional factor AtPAP1 over-expression (AtPAP1 TR). Additionally, remodeling conditions were induced in the cultures with rhinovirus 16 (HRV16). The expressions of metalloproteinase 9 (MMP)-9, tissue inhibitor of metalloproteinases 1 (TIMP-1), arginase I and transforming growth factor (TGF)-β were determined by quantitative polymerase chain reaction (qPCR) and immunoblotting methods. AtPAP1 TR decreased arginase I and MMP-9 expression with no effect on TIMP-1 or TGF-β mRNA expression. This extract also inhibited HRV16-induced expression of arginase I, MMP-9 and TGF-β in both cell lines (P < 0·05) Our study shows for the first time to our knowledge, that transformed AtPAP1 TR extract from L. sibiricus root may affect the remodeling process. Its effect can be attributed an increased amount of phenolic acids such as: chlorogenic acid, caffeic acid or ferulic acid and demonstrates the value of biotechnology in medicinal research.
Collapse
Affiliation(s)
- J Wieczfinska
- Department of Immunopathology, Medical University of Lodz, Lodz, Poland
| | - P Sitarek
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Lodz, Poland
| | - T Kowalczyk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Lodz, Poland
| | - R Pawliczak
- Department of Immunopathology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
30
|
Craig A, Mels CMC, Schutte AE, Bollenbach A, Tsikas D, Schwedhelm E, Kruger R. Urinary albumin-to-creatinine ratio is inversely related to nitric oxide synthesis in young black adults: the African-PREDICT study. Hypertens Res 2020; 44:71-79. [PMID: 32681162 DOI: 10.1038/s41440-020-0514-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/21/2020] [Indexed: 01/30/2023]
Abstract
Hypertension is common in black populations and is known to be associated with low nitric oxide (NO) bioavailability. We compared plasma and urinary NO-related markers and plasma creatine kinase (CK) levels between young healthy black and white adults along with the associations of these markers with the urinary albumin-to-creatinine ratio (uACR), which is a surrogate marker of endothelial and kidney function. We included 1105 participants (20-30 years). We measured the uACR, plasma CK, plasma and urinary arginine, homoarginine, asymmetric (ADMA) and symmetric dimethylarginine (SDMA), urinary ornithine/citrulline, nitrate and nitrite, and malondialdehyde (MDA). In addition, the urinary nitrate-to-nitrite ratio (UNOxR) was calculated and used as a measure of circulating NO bioavailability. The uACR was comparable between the groups, yet the black group had lower urinary nitrate (by -15%) and UNOxR values (by -18%) (both p ≤ 0.001), higher plasma (by +9.6%) and urinary (by +5.9%) arginine (both p ≤ 0.004), higher plasma (by +13%) and urinary (by +3.7%) ADMA (both p ≤ 0.033), and higher CK (by +9.5%) and MDA (by +19%) (both p < 0.001) compared with white adults. Plasma and urinary homoarginine were similar between the groups. In the multiple regression analysis, we confirmed the inverse associations of the uACR with both plasma (adj. R2 = 0.066; β = -0.209; p = 0.005) and urinary (adj. R2 = 0.066; β = -0.149; p = 0.010) homoarginine and with the UNOxR (adj. R2 = 0.060; β = -0.122; p = 0.031) in the black group only. The overall less favorable NO profile and higher CK and MDA levels in the black cohort along with the adverse associations with the uACR may reflect the vulnerability of this cohort to the early development of hypertension.
Collapse
Affiliation(s)
- Ashleigh Craig
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
| | - Catharina M C Mels
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Aletta E Schutte
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa.,School of Public Health and Community Medicine, University of New South Wales and The George Institute for Global Health, Sydney, Australia
| | - Alexander Bollenbach
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, Hannover, Germany
| | - Dimitrios Tsikas
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, Hannover, Germany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.,Deutsches Zentrum fuer Herz-Kreislauf-Forschung E.V. (DZHK), Partner Site Hamburg/Kiel/Lüebeck, Hamburg, Germany
| | - Ruan Kruger
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa. .,MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
31
|
Matysiak J, Klupczynska A, Packi K, Mackowiak-Jakubowska A, Bręborowicz A, Pawlicka O, Olejniczak K, Kokot ZJ, Matysiak J. Alterations in Serum-Free Amino Acid Profiles in Childhood Asthma. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E4758. [PMID: 32630672 PMCID: PMC7370195 DOI: 10.3390/ijerph17134758] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 02/07/2023]
Abstract
Asthma often begins in childhood, although making an early diagnosis is difficult. Clinical manifestations, the exclusion of other causes of bronchial obstruction, and responsiveness to anti-inflammatory therapy are the main tool of diagnosis. However, novel, precise, and functional biochemical markers are needed in the differentiation of asthma phenotypes, endotypes, and creating personalized therapy. The aim of the study was to search for metabolomic-based asthma biomarkers among free amino acids (AAs). A wide panel of serum-free AAs in asthmatic children, covering both proteinogenic and non-proteinogenic AAs, were analyzed. The examination included two groups of individuals between 3 and 18 years old: asthmatic children and the control group consisted of children with neither asthma nor allergies. High-performance liquid chromatography combined with tandem mass spectrometry (LC-MS/MS technique) was used for AA measurements. The data were analyzed by applying uni- and multivariate statistical tests. The obtained results indicate the decreased serum concentration of taurine, L-valine, DL-β-aminoisobutyric acid, and increased levels of ƴ-amino-n-butyric acid and L-arginine in asthmatic children when compared to controls. The altered concentration of these AAs can testify to their role in the pathogenesis of childhood asthma. The authors' results should contribute to the future introduction of new diagnostic markers into clinical practice.
Collapse
Affiliation(s)
- Joanna Matysiak
- Faculty of Health Sciences, The President Stanisław Wojciechowski State University of Applied Sciences in Kalisz, 62-800 Kalisz, Poland;
| | - Agnieszka Klupczynska
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60 -780 Poznan, Poland; (A.K.); (K.P.); (A.M.-J.); (O.P.); (J.M.)
| | - Kacper Packi
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60 -780 Poznan, Poland; (A.K.); (K.P.); (A.M.-J.); (O.P.); (J.M.)
| | - Anna Mackowiak-Jakubowska
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60 -780 Poznan, Poland; (A.K.); (K.P.); (A.M.-J.); (O.P.); (J.M.)
| | - Anna Bręborowicz
- Department of Pulmonology, Pediatric Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznan, Poland; (A.B.); (K.O.)
| | - Olga Pawlicka
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60 -780 Poznan, Poland; (A.K.); (K.P.); (A.M.-J.); (O.P.); (J.M.)
| | - Katarzyna Olejniczak
- Department of Pulmonology, Pediatric Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznan, Poland; (A.B.); (K.O.)
| | - Zenon J. Kokot
- Faculty of Health Sciences, The President Stanisław Wojciechowski State University of Applied Sciences in Kalisz, 62-800 Kalisz, Poland;
| | - Jan Matysiak
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60 -780 Poznan, Poland; (A.K.); (K.P.); (A.M.-J.); (O.P.); (J.M.)
| |
Collapse
|
32
|
van den Berg MPM, Kurhade SH, Maarsingh H, Erceg S, Hulsbeek IR, Boekema PH, Kistemaker LEM, van Faassen M, Kema IP, Elsinga PH, Dömling A, Meurs H, Gosens R. Pharmacological Screening Identifies SHK242 and SHK277 as Novel Arginase Inhibitors with Efficacy against Allergen-Induced Airway Narrowing In Vitro and In Vivo. J Pharmacol Exp Ther 2020; 374:62-73. [PMID: 32269169 DOI: 10.1124/jpet.119.264341] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 03/31/2020] [Indexed: 02/02/2023] Open
Abstract
Arginase is a potential target for asthma treatment. However, there are currently no arginase inhibitors available for clinical use. Here, a novel class of arginase inhibitors was synthesized, and their efficacy was pharmacologically evaluated. The reference compound 2(S)-amino-6-boronohexanoic acid (ABH) and >200 novel arginase inhibitors were tested for their ability to inhibit recombinant human arginase 1 and 2 in vitro. The most promising compounds were separated as enantiomers. Enantiomer pairs SHK242 and SHK243, and SHK277 and SHK278 were tested for functional efficacy by measuring their effect on allergen-induced airway narrowing in lung slices of ovalbumin-sensitized guinea pigs ex vivo. A guinea pig model of acute allergic asthma was used to examine the effect of the most efficacious enantiopure arginase inhibitors on allergen-induced airway hyper-responsiveness (AHR), early and late asthmatic reactions (EAR and LAR), and airway inflammation in vivo. The novel compounds were efficacious in inhibiting arginase 1 and 2 in vitro. The enantiopure SHK242 and SHK277 fully inhibited arginase activity, with IC50 values of 3.4 and 10.5 μM for arginase 1 and 2.9 and 4.0 µM for arginase 2, respectively. Treatment of slices with ABH or novel compounds resulted in decreased ovalbumin-induced airway narrowing compared with control, explained by increased local nitric oxide production in the airway. In vivo, ABH, SHK242, and SHK277 protected against allergen-induced EAR and LAR but not against AHR or lung inflammation. We have identified promising novel arginase inhibitors for the potential treatment of allergic asthma that were able to protect against allergen-induced early and late asthmatic reactions. SIGNIFICANCE STATEMENT: Arginase is a potential drug target for asthma treatment, but currently there are no arginase inhibitors available for clinical use. We have identified promising novel arginase inhibitors for the potential treatment of allergic asthma that were able to protect against allergen-induced early and late asthmatic reactions. Our new inhibitors show protective effects in reducing airway narrowing in response to allergens and reductions in the early and late asthmatic response.
Collapse
Affiliation(s)
- M P M van den Berg
- Departments of Molecular Pharmacology (M.P.M.v.d.B., S.E., I.R.H., P.H.B., L.E.M.K., H.Me., R.G.) and Drug Design (S.H.K., A.D.), Groningen Research Institute of Pharmacy, University of Groningen. Department of Laboratory Medicine, University Medical Center Groningen (M.v.F., I.P.K.), University of Groningen, Groningen, The Netherlands; Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida (H.Ma.); and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (P.H.E.)
| | - S H Kurhade
- Departments of Molecular Pharmacology (M.P.M.v.d.B., S.E., I.R.H., P.H.B., L.E.M.K., H.Me., R.G.) and Drug Design (S.H.K., A.D.), Groningen Research Institute of Pharmacy, University of Groningen. Department of Laboratory Medicine, University Medical Center Groningen (M.v.F., I.P.K.), University of Groningen, Groningen, The Netherlands; Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida (H.Ma.); and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (P.H.E.)
| | - H Maarsingh
- Departments of Molecular Pharmacology (M.P.M.v.d.B., S.E., I.R.H., P.H.B., L.E.M.K., H.Me., R.G.) and Drug Design (S.H.K., A.D.), Groningen Research Institute of Pharmacy, University of Groningen. Department of Laboratory Medicine, University Medical Center Groningen (M.v.F., I.P.K.), University of Groningen, Groningen, The Netherlands; Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida (H.Ma.); and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (P.H.E.)
| | - S Erceg
- Departments of Molecular Pharmacology (M.P.M.v.d.B., S.E., I.R.H., P.H.B., L.E.M.K., H.Me., R.G.) and Drug Design (S.H.K., A.D.), Groningen Research Institute of Pharmacy, University of Groningen. Department of Laboratory Medicine, University Medical Center Groningen (M.v.F., I.P.K.), University of Groningen, Groningen, The Netherlands; Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida (H.Ma.); and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (P.H.E.)
| | - I R Hulsbeek
- Departments of Molecular Pharmacology (M.P.M.v.d.B., S.E., I.R.H., P.H.B., L.E.M.K., H.Me., R.G.) and Drug Design (S.H.K., A.D.), Groningen Research Institute of Pharmacy, University of Groningen. Department of Laboratory Medicine, University Medical Center Groningen (M.v.F., I.P.K.), University of Groningen, Groningen, The Netherlands; Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida (H.Ma.); and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (P.H.E.)
| | - P H Boekema
- Departments of Molecular Pharmacology (M.P.M.v.d.B., S.E., I.R.H., P.H.B., L.E.M.K., H.Me., R.G.) and Drug Design (S.H.K., A.D.), Groningen Research Institute of Pharmacy, University of Groningen. Department of Laboratory Medicine, University Medical Center Groningen (M.v.F., I.P.K.), University of Groningen, Groningen, The Netherlands; Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida (H.Ma.); and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (P.H.E.)
| | - L E M Kistemaker
- Departments of Molecular Pharmacology (M.P.M.v.d.B., S.E., I.R.H., P.H.B., L.E.M.K., H.Me., R.G.) and Drug Design (S.H.K., A.D.), Groningen Research Institute of Pharmacy, University of Groningen. Department of Laboratory Medicine, University Medical Center Groningen (M.v.F., I.P.K.), University of Groningen, Groningen, The Netherlands; Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida (H.Ma.); and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (P.H.E.)
| | - M van Faassen
- Departments of Molecular Pharmacology (M.P.M.v.d.B., S.E., I.R.H., P.H.B., L.E.M.K., H.Me., R.G.) and Drug Design (S.H.K., A.D.), Groningen Research Institute of Pharmacy, University of Groningen. Department of Laboratory Medicine, University Medical Center Groningen (M.v.F., I.P.K.), University of Groningen, Groningen, The Netherlands; Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida (H.Ma.); and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (P.H.E.)
| | - I P Kema
- Departments of Molecular Pharmacology (M.P.M.v.d.B., S.E., I.R.H., P.H.B., L.E.M.K., H.Me., R.G.) and Drug Design (S.H.K., A.D.), Groningen Research Institute of Pharmacy, University of Groningen. Department of Laboratory Medicine, University Medical Center Groningen (M.v.F., I.P.K.), University of Groningen, Groningen, The Netherlands; Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida (H.Ma.); and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (P.H.E.)
| | - P H Elsinga
- Departments of Molecular Pharmacology (M.P.M.v.d.B., S.E., I.R.H., P.H.B., L.E.M.K., H.Me., R.G.) and Drug Design (S.H.K., A.D.), Groningen Research Institute of Pharmacy, University of Groningen. Department of Laboratory Medicine, University Medical Center Groningen (M.v.F., I.P.K.), University of Groningen, Groningen, The Netherlands; Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida (H.Ma.); and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (P.H.E.)
| | - A Dömling
- Departments of Molecular Pharmacology (M.P.M.v.d.B., S.E., I.R.H., P.H.B., L.E.M.K., H.Me., R.G.) and Drug Design (S.H.K., A.D.), Groningen Research Institute of Pharmacy, University of Groningen. Department of Laboratory Medicine, University Medical Center Groningen (M.v.F., I.P.K.), University of Groningen, Groningen, The Netherlands; Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida (H.Ma.); and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (P.H.E.)
| | - H Meurs
- Departments of Molecular Pharmacology (M.P.M.v.d.B., S.E., I.R.H., P.H.B., L.E.M.K., H.Me., R.G.) and Drug Design (S.H.K., A.D.), Groningen Research Institute of Pharmacy, University of Groningen. Department of Laboratory Medicine, University Medical Center Groningen (M.v.F., I.P.K.), University of Groningen, Groningen, The Netherlands; Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida (H.Ma.); and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (P.H.E.)
| | - R Gosens
- Departments of Molecular Pharmacology (M.P.M.v.d.B., S.E., I.R.H., P.H.B., L.E.M.K., H.Me., R.G.) and Drug Design (S.H.K., A.D.), Groningen Research Institute of Pharmacy, University of Groningen. Department of Laboratory Medicine, University Medical Center Groningen (M.v.F., I.P.K.), University of Groningen, Groningen, The Netherlands; Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida (H.Ma.); and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (P.H.E.)
| |
Collapse
|
33
|
Momma TY, Ottaviani JI. Arginase inhibitor, N ω-hydroxy-L-norarginine, spontaneously releases biologically active NO-like molecule: Limitations for research applications. Free Radic Biol Med 2020; 152:74-82. [PMID: 32131024 DOI: 10.1016/j.freeradbiomed.2020.02.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/25/2019] [Accepted: 02/28/2020] [Indexed: 12/21/2022]
Abstract
There has been a renewed interest in the enzyme arginase for its role in various physiological and pathological processes that go beyond the urea cycle. One such role ascribed to arginase has been that of regulating nitric oxide (NO) production by a substrate (l-arginine) competition between arginase and nitric oxide synthase (NOS). Several arginase inhibitors have been developed to investigate the biological roles of arginase, of which Nω-hydroxy-l-norarginine (nor-NOHA) is commercially available and is used widely from cell culture models to clinical investigations in humans. Despite the prevalence of nor-NOHA to investigate the substrate competition between arginase and NOS, little is known regarding interferences that nor-NOHA could have on common methods to assess NO production. Therefore, we investigated if nor-NOHA has unintended consequences on common NO assessment methods. We show that nor-NOHA spontaneously releases biologically active NO-like molecule in cell culture media by reacting with riboflavin. This NO-like molecule is indistinguishable from an NO donor (NOR-3) using common methods to assess NO. Besides riboflavin, nor-NOHA spontaneously reacts with H2O2 to diminish H2O2 content and produce NO-like molecule in the process. Our investigation provides detailed evidence on unintended artefacts related to nor-NOHA that can limit its use in cell culture, as well as some ex vivo and in vivo models. Future studies on arginase should take into consideration the limitations presented here when using nor-NOHA as a research tool, not only in investigations related to arginase and NOS competition, but also for investigating other biological roles of arginase.
Collapse
Affiliation(s)
- Tony Y Momma
- Department of Nutrition, University of California, Davis, CA, 95616, USA.
| | - Javier I Ottaviani
- Department of Nutrition, University of California, Davis, CA, 95616, USA; Mars, Inc., McLean, VA, 22101, USA
| |
Collapse
|
34
|
Blumberg AH, Ebelt ST, Liang D, Morris CR, Sarnat JA. Ambient air pollution and sickle cell disease-related emergency department visits in Atlanta, GA. ENVIRONMENTAL RESEARCH 2020; 184:109292. [PMID: 32179263 PMCID: PMC7847665 DOI: 10.1016/j.envres.2020.109292] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 06/01/2023]
Abstract
BACKGROUND Sickle cell disease (SCD) is an inherited, autosomal recessive blood disorder, among the most prevalent genetic diseases, globally. While the genetic and hemolytic dynamics of SCD have been well-characterized, the etiology of SCD-related pathophysiological processes is unclear. Although limited, observational evidence suggests that environmental factors, including urban air pollution, may play a role. OBJECTIVES We assessed whether daily ambient air pollution concentrations are associated with corresponding emergency department (ED) visit counts for acute SCD exacerbations in Atlanta, Georgia, during a 9-year (2005-2013) period. We also examined heterogeneity in response by age and sex. METHODS ED visit data were from 41 hospitals in the 20-county Atlanta, GA area. Associations between daily air pollution levels for 8 urban air pollutants and counts of SCD related ED visits were estimated using Poisson generalized linear models. RESULTS We observed positive associations between pollutants generally indicative of traffic emissions and corresponding SCD ED visits [e.g., rate ratio of 1.022 (95% CI: 1.002, 1.043) per interquartile range increase in carbon monoxide]. Age stratified analyses indicated stronger associations with traffic pollutants among children (0-18 years), as compared to older age strata. Associations involving other pollutants, including ozone and particulate matter and for models of individuals >18 years old, were consistent a null hypothesis of no association. DISCUSSION This analysis represents the first North American study to examine acute risk among individuals with SCD to urban air pollution and provide evidence of urban air pollution, especially from traffic sources, as a trigger for acute exacerbations. These findings are consistent with a hypothesis that biological pathways, including several centrally associated with oxidative stress, may contribute towards enhanced susceptibility in individuals with SCD.
Collapse
|
35
|
Increased Arginase Expression and Decreased Nitric Oxide in Pig Donor Lungs after Normothermic Ex Vivo Lung Perfusion. Biomolecules 2020; 10:biom10020300. [PMID: 32075026 PMCID: PMC7072555 DOI: 10.3390/biom10020300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/30/2020] [Accepted: 02/11/2020] [Indexed: 01/12/2023] Open
Abstract
An established pig lung transplantation model was used to study the effects of cold ischemia time, normothermic acellular ex vivo lung perfusion (EVLP) and reperfusion after lung transplantation on l-arginine/NO metabolism in lung tissue. Lung tissue homogenates were analyzed for NO metabolite (NOx) concentrations by chemiluminescent NO-analyzer technique, and l-arginine, l-ornithine, l-citrulline and asymmetric dimethylarginine (ADMA) quantified using liquid chromatography-mass spectrometry (LC-MS/MS). The expression of arginase and nitric oxide synthase (NOS) isoforms in lung was measured by real-time polymerase chain reaction. EVLP preservation resulted in a significant decrease in concentrations of NOx and l-citrulline, both products of NOS, at the end of EVLP and after reperfusion following transplantation, compared to control, respectively. The ratio of l-ornithine over l-citrulline, a marker of the balance between l-arginine metabolizing enzymes, was increased in the EVLP group prior to reperfusion. The expression of both arginase isoforms was increased from baseline 1 h post reperfusion in EVLP but not in the no-EVLP group. These data suggest that EVLP results in a shift of the l-arginine balance towards arginase, leading to NO deficiency in the lung. The arginase/NOS balance may, therefore, represent a therapeutic target to improve lung quality during EVLP and, subsequently, transplant outcomes.
Collapse
|
36
|
Asosingh K, Lauruschkat CD, Alemagno M, Frimel M, Wanner N, Weiss K, Kessler S, Meyers DA, Bennett C, Xu W, Erzurum S. Arginine metabolic control of airway inflammation. JCI Insight 2020; 5:127801. [PMID: 31996482 DOI: 10.1172/jci.insight.127801] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 12/18/2019] [Indexed: 01/03/2023] Open
Abstract
Inducible nitric oxide synthase (iNOS) and arginase-2 (ARG2) share a common substrate, arginine. Higher expression of iNOS and exhaled NO are linked to airway inflammation in patients. iNOS deletion in animal models suggests that eosinophilic inflammation is regulated by arginine metabolism. Moreover, ARG2 is a regulator of Th2 response, as shown by the development of severe eosinophilic inflammation in ARG2-/- mice. However, potential synergistic roles of iNOS and ARG2 in asthma have not been explored. Here, we hypothesized that arginine metabolic fate via iNOS and ARG2 may govern airway inflammation. In an asthma cohort, ARG2 variant genotypes were associated with arginase activity. ARG2 variants with lower arginase activity, combined with levels of exhaled NO, identified a severe asthma phenotype. Airway inflammation was present in WT, ARG2-/-, iNOS-/-, and ARG2-/-/iNOS-/- mice but was greatest in ARG2-/-. Eosinophilic and neutrophilic infiltration in the ARG2-/- mice was abrogated in ARG2-/-/iNOS-/- animals. Similarly, angiogenic airway remodeling was greatest in ARG2-/- mice. Cytokines driving inflammation and remodeling were highest in lungs of asthmatic ARG2-/- mice and lowest in the iNOS-/-. ARG2 metabolism of arginine suppresses inflammation, while iNOS metabolism promotes airway inflammation, supporting a central role for arginine metabolic control of inflammation.
Collapse
Affiliation(s)
- Kewal Asosingh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Chris D Lauruschkat
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mario Alemagno
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Matthew Frimel
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nicholas Wanner
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kelly Weiss
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sean Kessler
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Deborah A Meyers
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, College of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Carole Bennett
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Weiling Xu
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Serpil Erzurum
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
37
|
Holguin F, Grasemann H, Sharma S, Winnica D, Wasil K, Smith V, Cruse MH, Perez N, Coleman E, Scialla TJ, Que LG. L-Citrulline increases nitric oxide and improves control in obese asthmatics. JCI Insight 2019; 4:131733. [PMID: 31714895 DOI: 10.1172/jci.insight.131733] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/06/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUNDThe airways of obese asthmatics have been shown to be NO deficient, and this contributes to airway dysfunction and reduced response to inhaled corticosteroids. In cultured airway epithelial cells, L-citrulline, a precursor of L-arginine recycling and NO formation, has been shown to prevent asymmetric dimethyl arginine-mediated (ADMA-mediated) NO synthase (NOS2) uncoupling, restoring NO and reducing oxidative stress.METHODSIn a proof-of-concept, open-label pilot study in which participants were analyzed before and after treatment, we hypothesized that 15 g/d L-citrulline for 2 weeks would (a) increase the fractional excretion of NO (FeNO), (b) improve asthma control, and (c) improve lung function. To this end, we recruited obese (BMI >30) asthmatics on controller therapy, with a baseline FeNO of ≤30 ppb from the University of Colorado Medical Center and Duke University Health System.RESULTSA total of 41 subjects with an average FeNO of 17 ppb (95% CI, 15-19) and poorly controlled asthma (average asthma control questionnaire [ACQ] 1.5 [95% CI, 1.2-1.8]) completed the study. Compared with baseline, L-citrulline increased whereas ADMA and arginase concentration did not (values represent the mean Δ and 95% CI): plasma L-citrulline (190 μM, 84-297), plasma L-arginine (67 μM, 38-95), and plasma L-arginine/ADMA (ratio 117, 67-167). FeNO increased by 4.2 ppb (1.7-6.7 ppb); ACQ decreased by -0.46 (-0.67 to 0.27 points); the forced vital capacity and forced exhalation volume in 1 second, respectively, changed by 86 ml (10-161 ml) and 52 ml (-11 to 132 ml). In a secondary analysis, the greatest FEV1 increments occurred in those subjects with late-onset asthma (>12 years) (63 ml [95% CI, 1-137]), in females (80 ml [95% CI, 5-154]), with a greater change seen in late-onset females (100 ml, [95% CI, 2-177]). The changes in lung function or asthma control were not significantly associated with the changes before and after treatment in L-arginine/ADMA or FeNO.CONCLUSIONShort-term L-citrulline treatment improved asthma control and FeNO levels in obese asthmatics with low or normal FeNO. Larger FEV1 increments were observed in those with late-onset asthma and in females.TRIAL REGISTRATIONClinicalTrials.gov NCT01715844.FUNDINGNIH NHLBI R01 HL146542-01.
Collapse
Affiliation(s)
- Fernando Holguin
- Department of Medicine, University of Colorado Medical School, Aurora, Colorado, USA
| | - Hartmut Grasemann
- Department of Pediatrics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sunita Sharma
- Department of Medicine, University of Colorado Medical School, Aurora, Colorado, USA
| | - Daniel Winnica
- Department of Medicine, University of Colorado Medical School, Aurora, Colorado, USA
| | - Karen Wasil
- Department of Medicine, University of Colorado Medical School, Aurora, Colorado, USA
| | - Vong Smith
- Department of Medicine, University of Colorado Medical School, Aurora, Colorado, USA
| | - Margaret H Cruse
- Department of Medicine, University of Colorado Medical School, Aurora, Colorado, USA
| | - Nancy Perez
- Department of Medicine, University of Colorado Medical School, Aurora, Colorado, USA
| | - Erika Coleman
- Department of Medicine, University of Colorado Medical School, Aurora, Colorado, USA
| | - Timothy J Scialla
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Loretta G Que
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
38
|
Abstract
Arginine metabolism is linked to several important metabolic processes, and reprogramming of arginine metabolism occurs in various physiological and pathological conditions. Here we describe a method, using a LC-MS-based metabolomics and 15N4-arginine tracing approach, to quantitatively analyze arginine metabolism. This method can reliably quantify the abundance of important intermediates and fluxes of major metabolic reactions in arginine metabolism in a variety of cultured mammalian cell models.
Collapse
Affiliation(s)
- Gretchen L Seim
- Morgridge Institute for Research, Madison, WI, USA
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Emily C Britt
- Morgridge Institute for Research, Madison, WI, USA
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jing Fan
- Morgridge Institute for Research, Madison, WI, USA.
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
39
|
Ismaeel A, Franco ME, Lavado R, Papoutsi E, Casale GP, Fuglestad M, Mietus CJ, Haynatzki GR, Smith RS, Bohannon WT, Sawicki I, Pipinos II, Koutakis P. Altered Metabolomic Profile in Patients with Peripheral Artery Disease. J Clin Med 2019; 8:E1463. [PMID: 31540015 PMCID: PMC6780416 DOI: 10.3390/jcm8091463] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 12/15/2022] Open
Abstract
Peripheral artery disease (PAD) is a common atherosclerotic disease characterized by narrowed or blocked arteries in the lower extremities. Circulating serum biomarkers can provide significant insight regarding the disease progression. Here, we explore the metabolomics signatures associated with different stages of PAD and investigate potential mechanisms of the disease. We compared the serum metabolites of a cohort of 26 PAD patients presenting with claudication and 26 PAD patients presenting with critical limb ischemia (CLI) to those of 26 non-PAD controls. A difference between the metabolite profiles of PAD patients from non-PAD controls was observed for several amino acids, acylcarnitines, ceramides, and cholesteryl esters. Furthermore, our data demonstrate that patients with CLI possess an altered metabolomic signature different from that of both claudicants and non-PAD controls. These findings provide new insight into the pathophysiology of PAD and may help develop future diagnostic procedures and therapies for PAD patients.
Collapse
Affiliation(s)
- Ahmed Ismaeel
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Marco E Franco
- Department of Environmental Science, Baylor University, Waco, TX 76798, USA
| | - Ramon Lavado
- Department of Environmental Science, Baylor University, Waco, TX 76798, USA
| | - Evlampia Papoutsi
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - George P Casale
- Department of Surgery, University of Nebraska at Medical Center, Omaha, NE 68198, USA
| | - Matthew Fuglestad
- Department of Surgery, University of Nebraska at Medical Center, Omaha, NE 68198, USA
| | - Constance J Mietus
- Department of Surgery, University of Nebraska at Medical Center, Omaha, NE 68198, USA
| | - Gleb R Haynatzki
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Robert S Smith
- Department of Surgery, Baylor Scott and White Hospital, Temple, TX 76508, USA
| | - William T Bohannon
- Department of Surgery, Baylor Scott and White Hospital, Temple, TX 76508, USA
| | - Ian Sawicki
- Department of Surgery, Baylor Scott and White Hospital, Temple, TX 76508, USA
| | - Iraklis I Pipinos
- Department of Surgery, University of Nebraska at Medical Center, Omaha, NE 68198, USA
| | - Panagiotis Koutakis
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA.
| |
Collapse
|
40
|
Porro B, Songia P, Myasoedova VA, Valerio V, Moschetta D, Gripari P, Fusini L, Cavallotti L, Canzano P, Turnu L, Alamanni F, Camera M, Cavalca V, Poggio P. Endothelial Dysfunction in Patients with Severe Mitral Regurgitation. J Clin Med 2019; 8:jcm8060835. [PMID: 31212807 PMCID: PMC6616454 DOI: 10.3390/jcm8060835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 01/05/2023] Open
Abstract
Mitral valve prolapse (MVP) is the most common cause of severe mitral regurgitation. It has been reported that MVP patients—candidates for mitral valve repair (MVRep)—showed an alteration in the antioxidant defense systems as well as in the L-arginine metabolic pathway. In this study, we investigate if oxidative stress and endothelial dysfunction are an MVP consequence or driving factors. Forty-five patients undergoing MVRep were evaluated before and 6 months post surgery and compared to 29 controls. Oxidized (GSSG) and reduced (GSH) forms of glutathione, and L-arginine metabolic pathway were analyzed using liquid chromatography-tandem mass spectrometry methods while osteoprotegerin (OPG) through the ELISA kit and circulating endothelial microparticles (EMP) by flow cytometry. Six-month post surgery, in MVP patients, the GSSG/GSH ratio decreased while symmetric and asymmetric dimethylarginines levels remained comparable to the baseline. Conversely, OPG levels significantly increased when compared to their baseline. Finally, pre-MVRep EMP levels were significantly higher in patients than in controls and did not change post surgery. Overall, these results highlight that MVRep completely restores the increased oxidative stress levels, as evidenced in MVP patients. Conversely, no amelioration of endothelial dysfunction was evidenced after surgery. Thus, therapies aimed to restore a proper endothelial function before and after surgical repair could benefit MVP patients.
Collapse
Affiliation(s)
- Benedetta Porro
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
| | - Paola Songia
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
| | | | - Vincenza Valerio
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, 80138 Naples, Italy.
| | | | - Paola Gripari
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
| | - Laura Fusini
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
| | | | - Paola Canzano
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
| | - Linda Turnu
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
| | | | - Marina Camera
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy.
| | - Viviana Cavalca
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
| | - Paolo Poggio
- Centro Cardiologico Monzino, I.R.C.C.S., 20138 Milan, Italy.
| |
Collapse
|
41
|
Liang D, Ladva CN, Golan R, Yu T, Walker DI, Sarnat SE, Greenwald R, Uppal K, Tran V, Jones DP, Russell AG, Sarnat JA. Perturbations of the arginine metabolome following exposures to traffic-related air pollution in a panel of commuters with and without asthma. ENVIRONMENT INTERNATIONAL 2019; 127:503-513. [PMID: 30981021 PMCID: PMC6513706 DOI: 10.1016/j.envint.2019.04.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/30/2019] [Accepted: 04/01/2019] [Indexed: 05/05/2023]
Abstract
BACKGROUND Mechanisms underlying the effects of traffic-related air pollution on people with asthma remain largely unknown, despite the abundance of observational and controlled studies reporting associations between traffic sources and asthma exacerbation and hospitalizations. OBJECTIVES To identify molecular pathways perturbed following traffic pollution exposures, we analyzed data as part of the Atlanta Commuters Exposure (ACE-2) study, a crossover panel of commuters with and without asthma. METHODS We measured 27 air pollutants and conducted high-resolution metabolomics profiling on blood samples from 45 commuters before and after each exposure session. We evaluated metabolite and metabolic pathway perturbations using an untargeted metabolome-wide association study framework with pathway analyses and chemical annotation. RESULTS Most of the measured pollutants were elevated in highway commutes (p < 0.05). From both negative and positive ionization modes, 17,586 and 9087 metabolic features were extracted from plasma, respectively. 494 and 220 unique features were associated with at least 3 of the 27 exposures, respectively (p < 0.05), after controlling confounders and false discovery rates. Pathway analysis indicated alteration of several inflammatory and oxidative stress related metabolic pathways, including leukotriene, vitamin E, cytochrome P450, and tryptophan metabolism. We identified and annotated 45 unique metabolites enriched in these pathways, including arginine, histidine, and methionine. Most of these metabolites were not only associated with multiple pollutants, but also differentially expressed between participants with and without asthma. The analysis indicated that these metabolites collectively participated in an interrelated molecular network centering on arginine metabolism, underlying the impact of traffic-related pollutants on individuals with asthma. CONCLUSIONS We detected numerous significant metabolic perturbations associated with in-vehicle exposures during commuting and validated metabolites that were closely linked to several inflammatory and redox pathways, elucidating the potential molecular mechanisms of traffic-related air pollution toxicity. These results support future studies of metabolic markers of traffic exposures and the corresponding molecular mechanisms.
Collapse
Affiliation(s)
- Donghai Liang
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, USA.
| | - Chandresh N Ladva
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, USA
| | - Rachel Golan
- Department of Public Health, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Tianwei Yu
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, USA
| | - Douglas I Walker
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Stefanie E Sarnat
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, USA
| | - Roby Greenwald
- Division of Environmental Health, Georgia State University School of Public Health, Atlanta, USA
| | - Karan Uppal
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, USA
| | - ViLinh Tran
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, USA
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, USA
| | - Armistead G Russell
- School of Civil and Environmental Engineering, Georgia Institute of Technology, Atlanta, USA
| | - Jeremy A Sarnat
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, USA
| |
Collapse
|
42
|
Donthi S, Neela VSK, Gaddam S, Mohammed HH, Ansari SS, Valluri VL, Sivasai KSR. Association of increased risk of asthma with elevated arginase & interleukin-13 levels in serum & rs2781666 G/T genotype of arginase I. Indian J Med Res 2019; 148:159-168. [PMID: 30381539 PMCID: PMC6206777 DOI: 10.4103/ijmr.ijmr_379_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Background & objectives High expression of arginase gene and its elevated level in serum and bronchial lavage reported in animal models indicated an association with the pathogenesis of asthma. This study was undertaken to assess the serum arginase activity in symptomatic asthma patients and healthy controls and to correlate it with cytokine levels [interleukin (IL)-4 and IL-13] and arginase I (ARG1) gene polymorphism. Methods Asthma was confirmed by lung function test according to the GINA guidelines in patients attending Allergy and Pulmonology Clinic, Bhagwan Mahavir Hospital and Research Centre, Hyderabad, India, a tertiary care centre, during 2013-2015. Serum arginase was analyzed using a biochemical assay, total IgE and cytokine levels by enzyme-linked immunosorbent assay and genotyping of ARG1 for single-nucleotide polymorphisms (SNPs) rs2781666 and rs60389358 using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. Results There was a significant two-fold elevation in the arginase activity in asthmatics as compared to healthy controls which correlated with disease severity. Non-atopic asthmatics showed elevated activity of arginase compared to atopics, indicating its possible role in intrinsic asthma. Levels of serum IL-13 and IL-4 were significantly high in asthma group which correlated with disease severity that was assessed by spirometry. A positive correlation was observed between arginase activity and IL-13 concentration. Genetic analysis of ARG1 SNPs revealed that rs2781666 G/T genotype, T allele and C-T haplotype (rs60389358 and rs2781666) were associated with susceptibility to asthma. Interpretation & conclusions This study indicated that high arginase activity and IL-13 concentration in the serum and ARG1 rs2781666 G/T genotype might increase the risk of asthma in susceptible population. Further studies need to be done with a large sample to confirm these findings.
Collapse
Affiliation(s)
- Suhasini Donthi
- Department of Biotechnology, Sreenidhi Institute of Science & Technology; Immunology & Molecular Biology Division, LEPRA Society Blue Peter Public Health & Research Centre, Hyderabad, India
| | - Venkata Sanjeev Kumar Neela
- Immunology & Molecular Biology Division, LEPRA Society Blue Peter Public Health & Research Centre, Hyderabad, India
| | - Sumanlatha Gaddam
- Allergy & Pulmonology Clinic, Bhagwan Mahavir Hospital & Research Centre, Hyderabad, India
| | | | - Soheb Sadath Ansari
- Allergy & Pulmonology Clinic, Bhagwan Mahavir Hospital & Research Centre, Hyderabad, India
| | - Vijaya Lakshmi Valluri
- Immunology & Molecular Biology Division, LEPRA Society Blue Peter Public Health & Research Centre, Hyderabad, India
| | - Krovvidi S R Sivasai
- Department of Biotechnology, Sreenidhi Institute of Science & Technology, Hyderabad, India
| |
Collapse
|
43
|
SHARMA V, SINGH L, VERMA N. Establishment and Escalation of an Amino Acid Stacked Repressible Release Embedded System Using Quality by Design. Turk J Pharm Sci 2019; 16:20-26. [PMID: 32454690 PMCID: PMC7227980 DOI: 10.4274/tjps.44154] [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: 08/02/2017] [Accepted: 12/07/2017] [Indexed: 12/01/2022]
Abstract
OBJECTIVES The traditional approach of developing a new delivery system is an exhaustive task and requires a number of resources like manpower, money, material, and time. To overcome this problem Quality by Design (QbD) can be utilized to obtain pharmaceutical products of desired (best) quality with minimum use of the above resources as well as determination of the impact of one factor over the desired associated process. The present research is focused on establishing a design for formulating optimized gelatin microspheres using QbD. MATERIALS AND METHODS Repressible released embedded microspheres of L-arginine were prepared by performing cross linking of gelatin using glutaraldehyde. Characterization of the formulated embedded microspheres was done based on infrared spectroscopy, scanning electron microscopy, percentage yield, microsphere size, drug entrapment efficiency, and drug release. The impact of concentrations of gelatin and ethyl cellulose was determined over dependent response like percentage yield, microsphere size, and drug entrapment efficiency. RESULTS A response surface curve was obtained using a 32 central composite design and the optimized batch was obtained with percentage yield, microsphere size, and drug entrapment efficiency of 89.98, 333.32 mm, and 82.61%, respectively. Validation of the optimized batch was done by formulating four different batches with optimized values of independent response and a comparison of the observed responses with the predicted ones was set up and all these batches were found close to the predicted values and show the validity of the optimized data. CONCLUSION The QbD approach is quite efficient to get optimized drug delivery systems of L-arginine without exhaustive study.
Collapse
Affiliation(s)
- Vijay SHARMA
- Shri Ram Murti Smarak College of Engineering, Technology and Research, Bareilly, India
| | - Lalit SINGH
- Shri Ram Murti Smarak College of Engineering, Technology and Research, Bareilly, India
| | - Navneet VERMA
- IFTM University, Faculty of Pharmacy, Moradabad, India
| |
Collapse
|
44
|
Resanovic I, Gluvic Z, Zaric B, Sudar-Milovanovic E, Jovanovic A, Milacic D, Isakovic R, Isenovic ER. Early Effects of Hyperbaric Oxygen on Inducible Nitric Oxide Synthase Activity/Expression in Lymphocytes of Type 1 Diabetes Patients: A Prospective Pilot Study. Int J Endocrinol 2019; 2019:2328505. [PMID: 30755771 PMCID: PMC6348926 DOI: 10.1155/2019/2328505] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/03/2018] [Accepted: 10/15/2018] [Indexed: 02/05/2023] Open
Abstract
This study aimed at examining the early effects of hyperbaric oxygen therapy (HBOT) on inducible nitric oxide synthase (iNOS) activity/expression in lymphocytes of type 1 diabetes mellitus (T1DM) patients. A group of 19 patients (mean age: 63 ± 2.1) with T1DM and with the peripheral arterial disease were included in this study. Patients were exposed to 10 sessions of HBOT in the duration of 1 h to 100% oxygen inhalation at 2.4 ATA. Blood samples were collected for the plasma C-reactive protein (CRP), plasma free fatty acid (FFA), serum nitrite/nitrate, and serum arginase activity measurements. Expression of iNOS and phosphorylation of p65 subunit of nuclear factor-κB (NFκB-p65), extracellular-regulated kinases 1/2 (ERK1/2), and protein kinase B (Akt) were examined in lymphocyte lysates by Western blot. After exposure to HBOT, plasma CRP and FFA were significantly decreased (p < 0.001). Protein expression of iNOS and serum nitrite/nitrate levels were decreased (p < 0.01), while serum arginase activity was increased (p < 0.05) versus before exposure to HBOT. Increased phosphorylation of NFκB-p65 at Ser536 (p < 0.05) and decreased level of NFκB-p65 protein (p < 0.001) in lymphocytes of T1DM patients were observed after HBOT. Decreased phosphorylation of ERK1/2 (p < 0.05) and Akt (p < 0.05) was detected after HBOT. Our results indicate that exposure to HBO decreased iNOS activity/expression via decreasing phosphorylation of ERK1/2 and Akt followed by decreased activity of NFκB.
Collapse
Affiliation(s)
- Ivana Resanovic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Belgrade, Serbia
| | - Zoran Gluvic
- Clinic for Internal Medicine, Zemun Clinical Hospital, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Bozidarka Zaric
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Belgrade, Serbia
| | - Emina Sudar-Milovanovic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Belgrade, Serbia
| | - Aleksandra Jovanovic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Belgrade, Serbia
| | - Davorka Milacic
- Department of Hyperbaric Medicine, Zemun Clinical Hospital, Belgrade, Serbia
| | - Radmilo Isakovic
- Department of Hyperbaric Medicine, Zemun Clinical Hospital, Belgrade, Serbia
| | - Esma R. Isenovic
- Institute of Nuclear Sciences Vinca, University of Belgrade, Laboratory of Radiobiology and Molecular Genetics, Belgrade, Serbia
| |
Collapse
|
45
|
Abstract
Arginine is derived from dietary intake, body protein breakdown, or endogenous de novo arginine production. Arginine methylation of non-histone proteins is used in transcriptional regulation. Protein-arginine methylation is used for regulation of transcriptional and various physiological pathological processes. Protein methylation may affect protein-protein, protein-DNA, or protein-RNA interaction. Arginine has an effect on the DNA-binding activity of NF-κB, a dominant transcriptional factor in inflammation. Adduct formation results in increased secretion of messenger molecules such as cytokines and chemokines that mediate communication among cells and promote inflammation. Arginine and lysine amino acid-rich histones in nucleosomes on modification by environmental agents form histone-DNA adducts, making it immunogenic. Alteration of DNA resulting from photomodification could lead to the development of antibodies or mutations to modified DNA. Lysine and arginine-rich histones in nucleosomes on modification by environmental agents form histone-DNA adducts, making it immunogenic. Alteration of DNA resulting from photomodification could lead to the development of antibodies or mutations to modified DNA. Therefore, the DNA-arginine photoadduct and modified photoadduct could have important implications in various pathophysiological conditions such as toxicology, carcinogenesis, and autoimmune phenomena. Abbreviations: Arg: Arginine; SLE: systemic lupus erythematosus; UV: ultraviolet; Tm: thermal melting temperature; NO: nitric oxide; O2.-: superoxide anion.
Collapse
Affiliation(s)
- Haseeb Ahsan
- a Department of Biochemistry, Faculty of Dentistry , Jamia Millia Islamia , New Delhi , India
| |
Collapse
|
46
|
Morris CR, Mauger DT, Suh JH, Phipatanakul W, Sheehan WJ, Moy JN, Paul IM, Szefler SJ, Jackson DJ, Fitzpatrick AM. Glutathione and arginine levels: Predictors for acetaminophen-associated asthma exacerbation? J Allergy Clin Immunol 2018; 142:308-311.e9. [PMID: 29518418 PMCID: PMC6035773 DOI: 10.1016/j.jaci.2018.01.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 01/18/2018] [Accepted: 01/30/2018] [Indexed: 10/17/2022]
Affiliation(s)
- Claudia R Morris
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Ga
| | - David T Mauger
- Department of Public Health Sciences, College of Medicine, Penn State University, Hershey, Pa
| | - Jung H Suh
- Children's Hospital Oakland Research Institute, Oakland, Calif
| | - Wanda Phipatanakul
- Division of Allergy/Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - William J Sheehan
- Division of Allergy/Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - James N Moy
- Stroger Hospital of Cook County, Department of Pediatrics, Rush University Medical Center, Chicago, Ill
| | - Ian M Paul
- Department of Pediatrics, College of Medicine, Penn State University, Hershey, Pa
| | - Stanley J Szefler
- Children's Hospital Colorado, the Breathing Institute, and the University of Colorado School of Medicine, Aurora, Colo
| | - Daniel J Jackson
- Pediatrics, Section of Allergy, Immunology and Rheumatology, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Anne M Fitzpatrick
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Ga.
| |
Collapse
|
47
|
Chennupati R, Meens MJ, Janssen BJ, van Dijk P, Hakvoort TBM, Lamers WH, De Mey JGR, Koehler SE. Deletion of endothelial arginase 1 does not improve vasomotor function in diabetic mice. Physiol Rep 2018; 6:e13717. [PMID: 29890043 PMCID: PMC5995309 DOI: 10.14814/phy2.13717] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 01/08/2023] Open
Abstract
Endothelial arginase 1 was ablated to assess whether this prevents hyperglycemia-induced endothelial dysfunction by improving arginine availability for nitric oxide production. Endothelial Arg1-deficient mice (Arg1-KOTie2 ) were generated by crossing Arg1fl/fl (controls) with Tie2Cretg/- mice and analyzed by immunohistochemistry, measurements of hemodynamics, and wire myography. Ablation was confirmed by immunohistochemistry. Mean arterial blood pressure was similar in conscious male control and Arg1-KOTie2 mice. Depletion of circulating arginine by intravenous infusion of arginase 1 or inhibition of nitric oxide synthase activity with L-NG -nitro-arginine methyl ester increased mean arterial pressure similarly in control (9 ± 2 and 34 ± 2 mmHg, respectively) and Arg1-KOTie2 mice (11 ± 3 and 38 ± 4 mmHg, respectively). Vasomotor responses were studied in isolated saphenous arteries of 12- and 34-week-old Arg1-KOTie2 and control animals by wire myography. Diabetes was induced in 10-week-old control and Arg1-KOTie2 mice with streptozotocin, and vasomotor responses were studied 10 weeks later. Optimal arterial diameter, contractile responses to phenylephrine, and relaxing responses to acetylcholine and sodium nitroprusside were similar in normoglycemic control and Arg1-KOTie2 mice. The relaxing response to acetylcholine was dependent on the availability of extracellular l-arginine. In the diabetic mice, arterial relaxation responses to endothelium-dependent hyperpolarization and to exogenous nitric oxide were impaired. The data show that endothelial ablation of arginase 1 in mice does not markedly modify smooth muscle and endothelial functions of a resistance artery under normo- and hyperglycemic conditions.
Collapse
Affiliation(s)
- Ramesh Chennupati
- Departments of Anatomy & EmbryologyMaastricht UniversityMaastrichtthe Netherlands
- Department of Pharmacology & ToxicologyMaastricht UniversityMaastrichtthe Netherlands
- Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtthe Netherlands
- Nutrim ‐ School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtthe Netherlands
| | - Merlijn J. Meens
- Department of Pharmacology & ToxicologyMaastricht UniversityMaastrichtthe Netherlands
- Department of Pathology and ImmunologyUniversity of GenevaGenevaSwitzerland
| | - Ben J. Janssen
- Department of Pharmacology & ToxicologyMaastricht UniversityMaastrichtthe Netherlands
- Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtthe Netherlands
| | - Paul van Dijk
- Departments of Anatomy & EmbryologyMaastricht UniversityMaastrichtthe Netherlands
| | | | - Wouter H. Lamers
- Departments of Anatomy & EmbryologyMaastricht UniversityMaastrichtthe Netherlands
- Nutrim ‐ School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtthe Netherlands
| | - Jo G. R. De Mey
- Department of Pharmacology & ToxicologyMaastricht UniversityMaastrichtthe Netherlands
- Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtthe Netherlands
- Department of Cardiovascular and Renal ResearchInstitute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- Department of CardiacThoracic and Vascular SurgeryOdense University HospitalOdenseDenmark
| | - S. Eleonore Koehler
- Departments of Anatomy & EmbryologyMaastricht UniversityMaastrichtthe Netherlands
- Nutrim ‐ School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtthe Netherlands
| |
Collapse
|
48
|
Nguyen DV, Linderholm A, Haczku A, Kenyon N. Glucagon-like peptide 1: A potential anti-inflammatory pathway in obesity-related asthma. Pharmacol Ther 2017; 180:139-143. [PMID: 28648831 PMCID: PMC5677567 DOI: 10.1016/j.pharmthera.2017.06.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Alterations in arginine metabolism and accelerated formation of advanced glycation end-products (AGEs), crucial mechanisms in obesity-related asthma, can be modulated by glucagon-like peptide 1 (GLP-1). l-arginine dysregulation in obesity promotes inflammation and bronchoconstriction. Prolonged hyperglycemia, dyslipidemia, and oxidative stress leads to production of AGEs, that bind to their receptor (RAGE) further potentiating inflammation. By binding to its widely distributed receptor, GLP-1 blunts the effects of RAGE activation and arginine dysregulation. The GLP-1 pathway, while comprehensively studied in the endocrine and cardiovascular literature, is under-recognized in pulmonary research. Insights into GLP-1 and the lung may lead to novel treatments for obesity-related asthma.
Collapse
Affiliation(s)
- Dan-Vinh Nguyen
- University of California Davis, the Veterans Affairs Northern California Healthcare System, United States.
| | - Angela Linderholm
- University of California Davis, the Veterans Affairs Northern California Healthcare System, United States
| | - Angela Haczku
- University of California Davis, the Veterans Affairs Northern California Healthcare System, United States
| | - Nicholas Kenyon
- University of California Davis, the Veterans Affairs Northern California Healthcare System, United States
| |
Collapse
|
49
|
Cloots RHE, Sankaranarayanan S, Poynter ME, Terwindt E, van Dijk P, Lamers WH, Eleonore Köhler S. Arginase 1 deletion in myeloid cells affects the inflammatory response in allergic asthma, but not lung mechanics, in female mice. BMC Pulm Med 2017; 17:158. [PMID: 29183288 PMCID: PMC5706166 DOI: 10.1186/s12890-017-0490-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 11/10/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND (Over-)expression of arginase may limit local availability of arginine for nitric oxide synthesis. We investigated the significance of arginase1 (ARG1) for the development of airway hyperresponsiveness (AHR) and lung inflammation in female mice with ovalbumin (OVA)-induced allergic asthma. METHODS Arg1 was ablated in the lung by crossing Arg1 fl/fl and Tie2Cre tg/- mice. OVA sensitization and challenge were conducted, and AHR to methacholine was determined using the Flexivent system. Changes in gene expression, chemokine and cytokine secretion, plasma IgE, and lung histology were quantified using RT-qPCR, ELISA, and immunohistochemistry, respectively. RESULTS Arg1 ablation had no influence on the development of OVA-induced AHR, but attenuated OVA-induced increases in expression of Arg2 and Nos2, Slc7a1, Slc7a2, and Slc7a7 (arginine transporters), Il4, Il5 and Il13 (TH2-type cytokines), Ccl2 and Ccl11 (chemokines), Ifng (TH1-type cytokine), Clca3 and Muc5ac (goblet cell markers), and OVA-specific IgE. Pulmonary IL-10 protein content increased, but IL-4, IL-5, IL-13, TNFα and IFNγ content, and lung histopathology, were not affected. Arg1 elimination also decreased number and tightness of correlations between adaptive changes in lung function and inflammatory parameters in OVA/OVA-treated female mice. OVA/OVA-treated female mice mounted a higher OVA-IgE response than males, but the correlation between lung function and inflammation was lower. Arg1-deficient OVA/OVA-treated females differed from males in a more pronounced decline of arginine-metabolizing and -transporting genes, higher plasma arginine levels, a smaller OVA-specific IgE response, and no improvement of peripheral lung function. CONCLUSION Complete ablation of Arg1 in the lung affects mRNA abundance of arginine-transporting and -metabolizing genes, and pro-inflammatory genes, but not methacholine responsiveness or accumulation of inflammatory cells.
Collapse
Affiliation(s)
- Roy H. E. Cloots
- Department of Anatomy & Embryology and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Selvakumari Sankaranarayanan
- Department of Anatomy & Embryology and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Matthew E. Poynter
- Division of Pulmonary Disease and Critical Care, Department of Medicine, College of Medicine, University of Vermont, Burlington, VT USA
| | - Els Terwindt
- Department of Anatomy & Embryology and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Paul van Dijk
- Department of Anatomy & Embryology and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Wouter H. Lamers
- Department of Anatomy & Embryology and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - S. Eleonore Köhler
- Department of Anatomy & Embryology and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
50
|
Trittmann JK, Jin Y, Chicoine LG, Liu Y, Chen B, Nelin LD. An arginase-1 SNP that protects against the development of pulmonary hypertension in bronchopulmonary dysplasia enhances NO-mediated apoptosis in lymphocytes. Physiol Rep 2017; 4:4/22/e13041. [PMID: 27895230 PMCID: PMC5358007 DOI: 10.14814/phy2.13041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/20/2016] [Accepted: 10/26/2016] [Indexed: 11/24/2022] Open
Abstract
Arginase and nitric oxide synthase (NOS) share a common substrate, l‐arginine, and have opposing effects on vascular remodeling. Arginase is the first step in polyamine and proline synthesis necessary for cellular proliferation, while NO produced from NOS promotes apoptosis. Previously, we identified a single nucleotide polymorphism (SNP) in the arginase‐1 (ARG1) gene, rs2781666 (T‐allele) that was associated with a decreased risk for developing pulmonary hypertension (PH) in a cohort of infants with bronchopulmonary dysplasia (BPD). In this study, we utilized lymphocytes from neonates (the only readily available cells from these patients expressing the two genotypes of interest) with either the rs2781666 SNP (TT) or wild type (GG) to test the hypothesis that the protection of the ARG1 SNP against the development of PH in BPD would involve augmented NO production leading to more apoptosis. Lymphocytes were stimulated with IL‐4, IL‐13, and phorbol myristate acetate (PMA). We found that TT lymphocytes had similar levels of arginase I and arginase II expression, but there was a tendency for lower urea production (a surrogate marker of arginase activity), than in the GG lymphocytes. The TT lymphocytes also had significantly greater NO production than did GG lymphocytes despite no differences in iNOS expression between genotypes. Furthermore, the TT lymphocytes had lower numbers of viable cells, and higher levels of cleaved caspase‐3 than did GG lymphocytes. Inhibiting NOS activity using Nω‐Nitro‐l‐arginine methyl ester hydrochloride (l‐NAME) significantly decreased cleaved caspase‐3 levels in the TT lymphocytes. These data demonstrate that the TT genotype results in greater levels of NO production leading to more apoptosis, which is consistent with the concept that BPD patients with the TT genotype are protected against the development of PH by producing greater basal levels of endogenous NO.
Collapse
Affiliation(s)
- Jennifer K Trittmann
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio .,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Yi Jin
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio
| | - Louis G Chicoine
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio.,Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio
| | - Yusen Liu
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Leif D Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|