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
|
García-Moguel I, Martínez-Mesa Á, Andújar-Espinosa R, Díaz-Campos R, Velasco-Garrido JL, Sanchez-Trincado JL, Luzon E, Nuevo J, Alconada C, Gutiérrez MÁ, Niza G, Padilla-Galo A. The impact of blood eosinophil count and FeNO on benralizumab effectiveness in clinical practice: An ORBE II subanalysis. Respir Med 2025; 237:107940. [PMID: 39814271 DOI: 10.1016/j.rmed.2025.107940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/28/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND The ORBE II study showed the real-world effectiveness of benralizumab in severe eosinophilic asthma (SEA). This subgroup analysis aimed to characterize patients and outcomes based on baseline blood eosinophil count (BEC) and/or fractional exhaled nitric oxide (FeNO) levels. METHODS In this analysis of the ORBE II retrospective study, SEA patients receiving benralizumab were categorized into subgroups based on individual or combined BEC/FeNO levels, according to the following thresholds: high BEC (hiBEC): ≥300 cells/μL; low BEC (loBEC): <300 cells/μL; high FeNO (hiFeNO): ≥50 ppb; low FeNO (loFeNO): <50 ppb. Baseline and up to 1 year of follow-up data are described. RESULTS Most patients with available data were classified as hiBEC (72.6 %) and 38.3 % as hiFeNO. The distribution according to combined baseline BEC and FeNO levels revealed a heterogeneous patient population. Although common SEA features were shared among subgroups, some distinct characteristics were observed, including elevated allergic asthma prevalence in hiBEC/loFeNO patients, high obesity prevalence and fewer non-smokers in loBEC/loFeNO patients, remarkable severe exacerbation rates in loBEC/hiFeNO patients [5.5 SD (6.0)], and more severe symptoms in the hiBEC/loBEC subgroup. All subgroups showed benefits following benralizumab treatment, with super-responder rates ranging from 68.2 % to 83.3 % and clinical remission rates reaching 70.0 %. Particularly good responses were noted in hiBEC/hiFeNO patients. CONCLUSIONS This study shows the variability of T2 biomarkers in ORBE II SEA patients, emphasizing the prevalence of high BEC values. While benralizumab benefits were important regardless of BEC, high BEC predicted good outcomes and FeNO had less influence on treatment effectiveness.
Collapse
Affiliation(s)
- Ismael García-Moguel
- Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Madrid, Spain
| | | | | | - Rocío Díaz-Campos
- Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Madrid, Spain
| | | | | | - Elisa Luzon
- Departamento Médico, AstraZeneca Farmacéutica S.A., Madrid, Spain
| | - Javier Nuevo
- Departamento Médico, AstraZeneca Farmacéutica S.A., Madrid, Spain
| | - Carlos Alconada
- Departamento Médico, AstraZeneca Farmacéutica S.A., Madrid, Spain
| | | | - Gabriel Niza
- Departamento Médico, AstraZeneca Farmacéutica S.A., Madrid, Spain
| | | |
Collapse
|
3
|
Poojary G, Vasishta S, Thomas RH, Satyamoorthy K, Padmakumar R, Joshi MB, Babu AS. Exercise improves systemic metabolism in a monocrotaline model of pulmonary hypertension. SPORTS MEDICINE AND HEALTH SCIENCE 2025; 7:37-47. [PMID: 39649790 PMCID: PMC11624410 DOI: 10.1016/j.smhs.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/04/2024] [Accepted: 03/04/2024] [Indexed: 12/11/2024] Open
Abstract
Exercise training in pulmonary arterial hypertension (PAH) has been gaining popularity with guidelines now recommending it as an important adjunct to medical therapy. Despite improvements in function and quality of life, an understanding of metabolic changes and their mechanisms remain unexplored. The objective of this study was therefore to understand the metabolic basis of exercise in a monocrotaline model of PAH. 24 male Wistar rats (age: 8-12 weeks and mean body weight: [262.16 ± 24.49] gms) were assigned to one of the four groups (i.e., Control, PAH, Exercise and PAH + Exercise). The exercise groups participated in treadmill running at 13.3 m/min, five days a week for five weeks. Demographic and clinical characteristics were monitored regularly. Following the intervention, LC-MS based metabolomics were performed on blood samples from all groups at the end of five weeks. Metabolite profiling, peak identification, alignment and isotope annotation were also performed. Statistical inference was carried out using dimensionality reducing techniques and analysis of variance. Partial-least-squares discrimination analysis and variable importance in the projection scores showed that the model was reliable, and not over lifting. The analysis demonstrated significant perturbations to lipid and amino acid metabolism, arginine and homocysteine pathways, sphingolipid (p < 0.05), glycerophospholipid (p < 0.05) and nucleotide metabolism in PAH. Exercise, however, was seen to restore arginine (p < 0.05) and homocysteine(p < 0.000 1) levels which were independent effects, irrespective of PAH. Dysregulated arginine and homocysteine pathways are seen in PAH. Exercise restores these dysregulated pathways and could potentially impact severity and outcome in PAH.
Collapse
Affiliation(s)
- Ganesha Poojary
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sampara Vasishta
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - R. Huban Thomas
- Department of Anatomy, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kapaettu Satyamoorthy
- SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara (SDM) University, Manjushree Nagar, Sattur, Dharwad, Karnataka, 580009, India
| | - Ramachandran Padmakumar
- Department of Cardiology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Manjunath B. Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Abraham Samuel Babu
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
4
|
Högman M, Pham-Ngoc H, Nguyen-Duy B, Ellingsen J, Hua-Huy T, Van Nguyen D, Dinh-Xuan AT. Measuring exhaled nitric oxide in COPD: from theoretical consideration to practical views. Expert Rev Respir Med 2024; 18:1013-1024. [PMID: 39587387 DOI: 10.1080/17476348.2024.2433537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
INTRODUCTION Chronic obstructive pulmonary disease (COPD) is traditionally perceived as Th1-inflammation, but some patients have Th2-inflammation. A high fraction of exhaled nitric oxide (FENO) is seen in asthma with Th2-inflammation, justifying FENO as a point-of-care biomarker. The use of FENO in COPD is much less frequent. We aimed to review the evidence in favor of FENO measurement in COPD and discuss its potential usefulness in clinical settings. AREAS COVERED This review covers nitric oxide production in the airways and FENO measurements in COPD patients during stable conditions and acute exacerbation. It discusses why COPD patients may have both low and high FENO levels and the potential clinical utility of FENO. EXPERT OPINION There is good evidence that FENO increases with an exacerbation irrespective of the initial low or high baseline value. However, there is insufficient evidence to establish a fixed cutoff value for elevated FENO in COPD today. Instead, a personal baseline FENO level should be established when the patient is in a stable phase of the disease, which will subsequently set high and low FENO levels in a personalized manner. In the future, home monitoring of FENO could help identify exacerbations early, allowing proper action to be taken.
Collapse
Affiliation(s)
- Marieann Högman
- Department of Medical Sciences, Lung- Allergy- and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Hà Pham-Ngoc
- Department of Respiratory and Sleep Medicine, Cochin Hospital, University Paris Cite, Paris, France
| | - Bô Nguyen-Duy
- Division of Respiratory, Allergy and Clinical Immunology, Vinmec International Hospital, Hanoi, Vietnam
| | - Jens Ellingsen
- Department of Medical Sciences, Lung- Allergy- and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Thông Hua-Huy
- Department of Respiratory and Sleep Medicine, Cochin Hospital, University Paris Cite, Paris, France
| | - Dinh Van Nguyen
- Division of Respiratory, Allergy and Clinical Immunology, Vinmec International Hospital, Hanoi, Vietnam
| | - Anh Tuan Dinh-Xuan
- Department of Respiratory and Sleep Medicine, Cochin Hospital, University Paris Cite, Paris, France
| |
Collapse
|
5
|
Delacoste FBC, Goulart CDL, Guidoti AB, Türck P, Eibel B, Irigoyen MC, de Araujo CLP, Dal Lago P. Evaluating the impact of short-term nitrate-rich dietary supplementation on endothelial function in COPD: A randomized crossover study. Respir Med 2024; 232:107745. [PMID: 39053520 DOI: 10.1016/j.rmed.2024.107745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/24/2024] [Accepted: 07/23/2024] [Indexed: 07/27/2024]
Abstract
AIM This study aimed to investigate the acute effects of dietary nitrate ingestion through l-arginine supplementation or dehydrated beet consumption on endothelial function in chronic obstructive pulmonary disease (COPD) patients. The secondary outcome was to analyze arterial stiffness, plasma nitrate, and nitrate/protein concentration. METHODS In this randomized crossover study, subjects with COPD underwent three series of supplementation: (1) l-arginine, (2) dehydrated beetroot, and (3) a placebo that appeared like the other supplements. Each intervention lasted 14 days, with a 7-day washout period between series. Participants underwent endothelial function assessment using flow-mediated dilatation (FMD), and plasma nitrate levels were measured at the end of each supplementation series. RESULTS Seventeen subjects (twelve male) completed the study protocol. Only five subjects presented endothelial dysfunction (RHI ≤0.51) at baseline. The mean baseline characteristics included age 66.5 ± 9.4 years, BMI 27.5 ± 4.5 kg/m2, FEV1, 0.79 (0.67-1.06) L. There were no differences (p > 0.05) between the groups or from pre-to post-interventions for RHI and arterial stiffness index (AIx) values, as well as parameters of endothelium-dependent vasodilation, such as blood flow velocity (BFV), shear stress, shear rate, FMD (mm), and FMD%. There was also no differences (p > 0.05) between the groups or from pre-to post-interventions plasma nitrate levels. CONCLUSIONS Acute dietary supplementation with nitrates, at the doses provided, did not show a significant improvement in endothelial function assessed by FMD, EndoPAT, or plasma nitrate levels in COPD. These findings suggest that a higher dose or prolonged supplementation might be required to achieve a therapeutic effect.
Collapse
Affiliation(s)
- Fernanda Beatriz Costa Delacoste
- Graduation Program in Rehabilitation Sciences, Experimental Laboratory of Physiology Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Cassia da Luz Goulart
- Cardiopulmonary Physiotherapy Laboratory, Department of Physical Therapy, Universidade Federal de Sao Carlos (UFSCar), São Carlos, SP, Brazil
| | - Augusto Baumhardt Guidoti
- Graduation Program in Rehabilitation Sciences, Experimental Laboratory of Physiology Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Patrick Türck
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species from the Universidade Federal do Rio Grande do Sul (UFRGS), Brazil
| | - Bruna Eibel
- Clinical Investigation Laboratory (LIC), Instituto de Cardiologia do Rio Grande do Sul (IC-FUC), Porto Alegre, Brazil
| | - Maria Claudia Irigoyen
- Experimental Laboratory of Hypertension, Heart Institute (InCor), University of São Paulo (USP), São Paulo, Brazil
| | - Cintia Laura Pereira de Araujo
- Graduation Program in Rehabilitation Sciences, Experimental Laboratory of Physiology Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Pedro Dal Lago
- Graduation Program in Rehabilitation Sciences, Experimental Laboratory of Physiology Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil; Graduation Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil.
| |
Collapse
|
6
|
Miller HA, Suliman S, Frieboes HB. Pulmonary Fibrosis Diagnosis and Disease Progression Detected Via Hair Metabolome Analysis. Lung 2024; 202:581-593. [PMID: 38861171 DOI: 10.1007/s00408-024-00712-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/30/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Fibrotic interstitial lung disease is often identified late due to non-specific symptoms, inadequate access to specialist care, and clinical unawareness precluding proper and timely treatment. Biopsy histological analysis is definitive but rarely performed due to its invasiveness. Diagnosis typically relies on high-resolution computed tomography, while disease progression is evaluated via frequent pulmonary function testing. This study tested the hypothesis that pulmonary fibrosis diagnosis and progression could be non-invasively and accurately evaluated from the hair metabolome, with the longer-term goal to minimize patient discomfort. METHODS Hair specimens collected from pulmonary fibrosis patients (n = 56) and healthy subjects (n = 14) were processed for metabolite extraction using 2DLC/MS-MS, and data were analyzed via machine learning. Metabolomic data were used to train machine learning classification models tuned via a rigorous combination of cross validation, feature selection, and testing with a hold-out dataset to evaluate classifications of diseased vs. healthy subjects and stable vs. progressed disease. RESULTS Prediction of pulmonary fibrosis vs. healthy achieved AUROCTRAIN = 0.888 (0.794-0.982) and AUROCTEST = 0.908, while prediction of stable vs. progressed disease achieved AUROCTRAIN = 0.833 (0.784 - 0.882) and AUROCTEST = 0. 799. Top metabolites for diagnosis included ornithine, 4-(methylnitrosamino)-1-3-pyridyl-N-oxide-1-butanol, Thr-Phe, desthiobiotin, and proline. Top metabolites for progression included azelaic acid, Thr-Phe, Ala-Tyr, indoleacetyl glutamic acid, and cytidine. CONCLUSION This study provides novel evidence that pulmonary fibrosis diagnosis and progression may in principle be evaluated from the hair metabolome. Longer term, this approach may facilitate non-invasive and accurate detection and monitoring of fibrotic lung diseases.
Collapse
Affiliation(s)
- Hunter A Miller
- Department of Bioengineering, University of Louisville, Lutz Hall 419, Louisville, KY, 40292, USA
| | - Sally Suliman
- Division of Pulmonary Medicine, University of Louisville, Louisville, KY, USA
- University of Arizona Medical Center Phoenix, Phoenix, AZ, USA
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville, Lutz Hall 419, Louisville, KY, 40292, USA.
- UofL Health - Brown Cancer Center, University of Louisville, Louisville, KY, USA.
- Center for Predictive Medicine, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
7
|
Lee JU, Song KS, Hong J, Shin H, Park E, Baek J, Park S, Baek AR, Lee J, Jang AS, Kim DJ, Chin SS, Kim UJ, Jeong SH, Park SW. Role of lung ornithine aminotransferase in idiopathic pulmonary fibrosis: regulation of mitochondrial ROS generation and TGF-β1 activity. Exp Mol Med 2024; 56:478-490. [PMID: 38413821 PMCID: PMC10907606 DOI: 10.1038/s12276-024-01170-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/12/2023] [Accepted: 11/23/2023] [Indexed: 02/29/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by aberrant lung remodeling and the excessive accumulation of extracellular matrix (ECM) proteins. In a previous study, we found that the levels of ornithine aminotransferase (OAT), a principal enzyme in the proline metabolism pathway, were increased in the lungs of patients with IPF. However, the precise role played by OAT in the pathogenesis of IPF is not yet clear. The mechanism by which OAT affects fibrogenesis was assessed in vitro using OAT-overexpressing and OAT-knockdown lung fibroblasts. The therapeutic effects of OAT inhibition were assessed in the lungs of bleomycin-treated mice. OAT expression was increased in fibrotic areas, principally in interstitial fibroblasts, of lungs affected by IPF. OAT levels in the bronchoalveolar lavage fluid of IPF patients were inversely correlated with lung function. The survival rate was significantly lower in the group with an OAT level >75.659 ng/mL than in the group with an OAT level ≤75.659 ng/mL (HR, 29.53; p = 0.0008). OAT overexpression and knockdown increased and decreased ECM component production by lung fibroblasts, respectively. OAT knockdown also inhibited transforming growth factor-β1 (TGF)-β1 activity and TGF-β1 pathway signaling. OAT overexpression increased the generation of mitochondrial reactive oxygen species (ROS) by activating proline dehydrogenase. The OAT inhibitor L-canaline significantly attenuated bleomycin-induced lung injury and fibrosis. In conclusion, increased OAT levels in lungs affected by IPF contribute to the progression of fibrosis by promoting excessive mitochondrial ROS production, which in turn activates TGF-β1 signaling. OAT may be a useful target for treating patients with fibrotic lung diseases, including IPF.
Collapse
Affiliation(s)
- Jong-Uk Lee
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Gyeonggi-Do, South Korea
| | - Ki Sung Song
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Gyeonggi-Do, South Korea
| | - Jisu Hong
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Gyeonggi-Do, South Korea
| | - Hyesun Shin
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Gyeonggi-Do, South Korea
| | - Eunji Park
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Gyeonggi-Do, South Korea
| | - Junyeong Baek
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Gyeonggi-Do, South Korea
| | - Shinhee Park
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Gyeonggi-Do, South Korea
| | - Ae-Rin Baek
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Gyeonggi-Do, South Korea
| | - Junehyuk Lee
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Gyeonggi-Do, South Korea
| | - An Soo Jang
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Gyeonggi-Do, South Korea
| | - Do Jin Kim
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Gyeonggi-Do, South Korea
| | - Su Sie Chin
- Department of Pathology, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Gyeonggi-Do, South Korea
| | - U-Jin Kim
- Department of Internal Medicine, Environmental Health Center Kangwon National University, Gangwondaehakgil, Chuncheon-si, Gangwon-do, South Korea
| | - Sung Hwan Jeong
- Department of Allergy, Pulmonary and Critical Care Medicine, Gachon University, Gil Medical Center, Incheon, South Korea
| | - Sung-Woo Park
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Gyeonggi-Do, South Korea.
| |
Collapse
|
8
|
Xu W, Cerna-Vargas JP, Tajuelo A, Lozano-Montoya A, Kivoloka M, Krink N, Monteagudo-Cascales E, Matilla MA, Krell T, Sourjik V. Systematic mapping of chemoreceptor specificities for Pseudomonas aeruginosa. mBio 2023; 14:e0209923. [PMID: 37791891 PMCID: PMC10653921 DOI: 10.1128/mbio.02099-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 08/16/2023] [Indexed: 10/05/2023] Open
Abstract
IMPORTANCE Chemotaxis of motile bacteria has multiple physiological functions. It enables bacteria to locate optimal ecological niches, mediates collective behaviors, and can play an important role in infection. These multiple functions largely depend on ligand specificities of chemoreceptors, and the number and identities of chemoreceptors show high diversity between organisms. Similar diversity is observed for the spectra of chemoeffectors, which include not only chemicals of high metabolic value but also bacterial, plant, and animal signaling molecules. However, the systematic identification of chemoeffectors and their mapping to specific chemoreceptors remains a challenge. Here, we combined several in vivo and in vitro approaches to establish a systematic screening strategy for the identification of receptor ligands and we applied it to identify a number of new physiologically relevant chemoeffectors for the important opportunistic human pathogen P. aeruginosa. This strategy can be equally applicable to map specificities of sensory domains from a wide variety of receptor types and bacteria.
Collapse
Affiliation(s)
- Wenhao Xu
- Max Planck Institute for Terrestrial Microbiology & Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
| | - Jean Paul Cerna-Vargas
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Granada, Spain
- Centro de Biotecnología y Genómica de Plantas CBGP, Universidad Politécnica de Madrid-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria/CSIC, Parque Científico y Tecnológico de la UPM, Pozuelo de Alarcón, Madrid, Spain
| | - Ana Tajuelo
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Andrea Lozano-Montoya
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Melissa Kivoloka
- Max Planck Institute for Terrestrial Microbiology & Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
| | - Nicolas Krink
- Max Planck Institute for Terrestrial Microbiology & Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
| | - Elizabet Monteagudo-Cascales
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Miguel A. Matilla
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Tino Krell
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Victor Sourjik
- Max Planck Institute for Terrestrial Microbiology & Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
| |
Collapse
|
9
|
Esposito A, Cotta CK, Lacchini R. Beyond eNOS: Genetic influence in NO pathway affecting drug response. Genet Mol Biol 2022; 45:e20220157. [PMID: 36264109 PMCID: PMC9583294 DOI: 10.1590/1678-4685-gmb-2022-0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/08/2022] [Indexed: 11/04/2022] Open
Abstract
Nitric Oxide (NO) has important biological functions, and its production may be
influenced by genetic polymorphisms. Since NO mediates the drug response, the
same genetic polymorphism that alter NO levels may also impact drug therapy. The
vast majority of studies in the literature that assess the genetic influence on
NO-related drug response focus on NOS3 (which encodes
endothelial nitric oxide synthase), however several other proteins are
interconnected in the same pathway and may also impact NO availability and drug
response. The aim of this study was to review the literature regarding genetic
polymorphisms that influence NO in response to pharmacological agents located in
genes other than NOS3. Articles were obtained from Pubmed and
consisted of 17 manuscripts that assessed polymorphisms of the following
targets: Arginases 1 and 2 (ARG1 and ARG2),
dimethylarginine dimethylaminohydrolases 1 and 2 (DDAH1 and
DDAH2), and vascular endothelial growth factor
(VEGF). Here we analyze the main results of these articles,
which show promising evidences that may suggest that the NO-driven
pharmacological response is affected by more than the eNOS gene. The search for
genetic markers may result in better understanding of the variability of drug
response and turn pharmacotherapy involving NO safer and more effective.
Collapse
Affiliation(s)
- Aline Esposito
- Universidade de São Paulo, Departamento de Farmacologia, Ribeirão
Preto, São Paulo, SP, Brazil
| | - Cezar Kayzuka Cotta
- Universidade de São Paulo, Departamento de Farmacologia, Ribeirão
Preto, São Paulo, SP, Brazil
| | - Riccardo Lacchini
- Universidade de São Paulo, Departamento de Enfermagem Psiquiátrica e
Ciências Humanas, Ribeirão Preto, São Paulo, SP, Brazil
| |
Collapse
|
10
|
The Arginine Catabolism-Derived Amino Acid L-ornithine Is a Chemoattractant for Pseudomonas aeruginosa. Microorganisms 2022; 10:microorganisms10020264. [PMID: 35208720 PMCID: PMC8875649 DOI: 10.3390/microorganisms10020264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/05/2022] [Accepted: 01/21/2022] [Indexed: 02/01/2023] Open
Abstract
Pseudomonas aeruginosa is a common, opportunistic bacterial pathogen among patients with cystic fibrosis, asthma, and chronic obstructive pulmonary disease. During the course of these diseases, l-ornithine, a non-proteinogenic amino acid, becomes more abundant. P. aeruginosa is chemotactic towards other proteinogenic amino acids. Here, we evaluated the chemotaxis response of P. aeruginosa towards l-ornithine. Our results show that l-ornithine serves as a chemoattractant for several strains of P. aeruginosa, including clinical isolates, and that the chemoreceptors involved in P. aeruginosa PAO1 are PctA and PctB. It seems likely that P. aeruginosa’s chemotactic response to l-ornithine might be a common feature and thus could potentially contribute to pathogenesis processes during colonization and infection scenarios.
Collapse
|
11
|
Ramasubramanian R, Kalhan R, Jacobs DR, Washko GR, Hou L, Gross MD, Guan W, Thyagarajan B. Gene expression of oxidative stress markers and lung function: A CARDIA lung study. Mol Genet Genomic Med 2021; 9:e1832. [PMID: 34800009 PMCID: PMC8683624 DOI: 10.1002/mgg3.1832] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/14/2021] [Accepted: 09/14/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Circulating markers of oxidative stress have been associated with lower lung function. Our objective was to study the association of gene expression levels of oxidative stress pathway genes (ALOX12, ALOX15, ARG2, GSTT1, LPO, MPO, NDUFB3, PLA2G7, and SOD3) and lung function forced expiratory volume in one second (FEV1 ), forced vital capacity (FVC) in Coronary Artery Risk Development in Young Adults study. METHODS Lung function was measured using spirometry and the Nanostring platform was used to estimate gene expression levels. Linear regression models were used to study association of lung function measured at year 30, 10-year decline in lung function and gene expression after adjustment for center, smoking, and BMI, measured at year 25. RESULTS The 10-year decline of FEV1 was faster in highest NDUFB3 quartile compared to the lowest (difference = -2.09%; p = 0.001) after adjustment for multiple comparisons. The 10-year decline in FEV1 and FVC was nominally slower in highest versus lowest quartile of PLA2G7 (difference = 1.14%; p = 0.02, and difference = 1.06%; p = 0.005, respectively). The other genes in the study were not associated with FEV1 or FVC. CONCLUSION Higher gene expression levels in oxidative stress pathway genes are associated with faster 10-year FEV1 decline.
Collapse
Affiliation(s)
- Ramya Ramasubramanian
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, Minnesota, USA
| | - Ravi Kalhan
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - David R Jacobs
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, Minnesota, USA
| | - George R Washko
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Myron D Gross
- Department of Pathology and Laboratory Medicine, University of Minnesota School of Medicine, Minneapolis, Minnesota, USA
| | - Weihua Guan
- Department of Biostatistics, University of Minnesota School of Public Health, Minneapolis, Minnesota, USA
| | - Bharat Thyagarajan
- Department of Pathology and Laboratory Medicine, University of Minnesota School of Medicine, Minneapolis, Minnesota, USA
| |
Collapse
|
12
|
Hu B, Xu G, Jin X, Chen D, Qian X, Li W, Xu L, Zhu J, Tang J, Jin X, Hou J. Novel Prognostic Predictor for Primary Pulmonary Hypertension: Focus on Blood Urea Nitrogen. Front Cardiovasc Med 2021; 8:724179. [PMID: 34760940 PMCID: PMC8572959 DOI: 10.3389/fcvm.2021.724179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/29/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Primary pulmonary hypertension (PPH) is a life-threatening disease associated with increased mortality. The urea cycle pathway plays a major role in PPH severity and treatment response. Little is known about the association of the blood urea nitrogen (BUN) and PPH prognosis. Methods: Clinical data were extracted from the Medical Information Mart for Intensive Care III (MIMIC-III) database. Adult patients (≥18 years) patients with primary pulmonary hypertension (PPH) in the database were enrolled. Spearman correlation was used to analyze the association of BUN with length of hospital and intensive care unit (ICU) stays. The chi-square test was used to analyze the association of BUN with mortality rate. Survival curves were estimated using the Kaplan-Meier method and compared by the log-rank test. Multivariable logistic regression was used to identify the BUN as an independent prognostic factor of mortality. Receiver operating characteristic (ROC) curves and the area under the curve (AUC) were used to analyze the sensitivity and specificity for mortality. Results: In total, 263 patients who met the selection criteria were enrolled. BUN was significantly positively associated with length of hospital stay and ICU stay (hospital stay: ρ = 0.282, ICU stay: ρ = 0.276; all P < 0.001). Higher hospital, 90-day and 4-year mortality rates were observed in the higher BUN quartile of PPH patients (hospital: P = 0.002; 90-day: P = 0.025; 4-year: P < 0.001). The Kaplan-Meier survival curves showed that patients in higher BUN quartile tended to have lower 4-year survival (Q1:7.65%, Q2: 10.71%; Q3: 14.80%, Q4: 16.84%; P < 0.0001). Logistic regression analyses found a significant association of BUN and mortality (hospital: OR = 1.05, 95% CI = 1.02–1.08, P = 0.001; 90-day: OR = 1.02, 95% CI = 1.00–1.05, P = 0.027; 4-year: OR = 1.05, 95% CI = 1.02–1.08, P = 0.001). Results of ROC and AUC showed that the diagnostic performance of BUN for mortality was moderately good. Conclusion: BUN was positively correlated with the length of hospital stay and ICU stay of PPH patients. Higher BUN was associated with higher hospital, 90-day and 4-year mortality and lower 4-year survival of PPH patients. These findings indicate that BUN can be a novel potential prognostic predictor for PPH.
Collapse
Affiliation(s)
- Bo Hu
- Department of Pathology and Municipal Key-Innovative Discipline of Molecular Diagnostics, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing University, Jiaxing, China
| | - Guangtao Xu
- Forensic and Pathology Laboratory, Department of Pathology, Institute of Forensic Science, Jiaxing University, Jiaxing, China
| | - Xin Jin
- Forensic and Pathology Laboratory, Department of Pathology, Institute of Forensic Science, Jiaxing University, Jiaxing, China
| | - Deqing Chen
- Forensic and Pathology Laboratory, Department of Pathology, Institute of Forensic Science, Jiaxing University, Jiaxing, China
| | - Xiaolan Qian
- Department of Pathology and Municipal Key-Innovative Discipline of Molecular Diagnostics, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing University, Jiaxing, China
| | - Wanlu Li
- Forensic and Pathology Laboratory, Department of Pathology, Institute of Forensic Science, Jiaxing University, Jiaxing, China
| | - Long Xu
- Forensic and Pathology Laboratory, Department of Pathology, Institute of Forensic Science, Jiaxing University, Jiaxing, China
| | - Jia Zhu
- Forensic and Pathology Laboratory, Department of Pathology, Institute of Forensic Science, Jiaxing University, Jiaxing, China
| | - Jie Tang
- Department of Pathology and Municipal Key-Innovative Discipline of Molecular Diagnostics, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing University, Jiaxing, China
| | - Xiuhui Jin
- Department of Immunology and Human Biology, University of Toronto, Toronto, ON, Canada
| | - Jian Hou
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
13
|
OZTAN O, TÜRKSOY VA, DENİZ S, COŞKUN BEYAN A, İRİTAŞ SB, ERCAN M, TUTKUN E. Silicosis and methylated arginines/L-arginines: case-control adapted a cross-sectional design. JOURNAL OF HEALTH SCIENCES AND MEDICINE 2021. [DOI: 10.32322/jhsm.982776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
14
|
Rodríguez-Guzmán MJ, Peces-Barba Romero G, Pérez Rial S, Serrano Del Castillo C, Palomero Rodríguez MÁ, Mahillo-Fernandez I, Villar-Álvarez F. Elevated levels of arginase activity are related to inflammation in patients with COPD exacerbation. BMC Pulm Med 2021; 21:271. [PMID: 34418988 PMCID: PMC8379850 DOI: 10.1186/s12890-021-01629-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 08/05/2021] [Indexed: 11/10/2022] Open
Abstract
Introduction Within the pathogenesis of the chronic obstructive pulmonary disease (COPD) there are interactions between different inflammatory mediators that are enhanced during an exacerbation. Arginase is present in bronchial epithelial cells, endothelial, fibroblasts and alveolar macrophages, which make it a probable key enzyme in the regulation of inflammation and remodelling. We aimed to find a potential relationship between arginase activity, inflammatory mediators in COPD patients in stable phase and during exacerbations. Methods We performed a prospective, observational study of cases and controls, with 4 study groups (healthy controls, stable COPD, COPD during an exacerbation and COPD 3 months after exacerbation). We measured arginase, inflammation markers (IL-6, IL-8, TNF-∝, IFN-γ and C reactive protein), and mediators of immunity: neutrophils, monocytes, total TCD3 + lymphocytes (CD3ζ), CD4 + T cells, CD8 + T cells, NK cells. Results A total of 49 subjects were recruited, average age of 69.73 years (59.18% male). Arginase activity is elevated during an exacerbation of COPD, and this rise is related to an increase in IL-6 production. The levels of IL-6 and IL-8 remained elevated in patients with COPD at 3 months after hospital exacerbation. We did not find a clear relationship between arginase activity, immunity or with the degree of obstruction in COPD patients. Conclusions Arginase activity is elevated during an exacerbation of COPD, and it could be related to an increase in the production of IL-6. Levels of IL-6, IL-8, and arginase activity remain elevated in patients with COPD at 3 months after hospital exacerbation. Arginase activity could contribute to the development of COPD.
Collapse
Affiliation(s)
| | - Germán Peces-Barba Romero
- Pulmonology Department, IIS Jiménez Díaz Foundation, CIBERES, Ave Reyes Catolicos #2, 28040, Madrid, Spain
| | - Sandra Pérez Rial
- Pulmonology Department, IIS Jiménez Díaz Foundation, CIBERES, Ave Reyes Catolicos #2, 28040, Madrid, Spain
| | | | | | | | - Felipe Villar-Álvarez
- Pulmonology Department, IIS Jiménez Díaz Foundation, CIBERES, Ave Reyes Catolicos #2, 28040, Madrid, Spain
| |
Collapse
|
15
|
Bayrak BB, Tunali S, Bal-Demirci T, Ulkuseven B, Yanardag R. Glycoprotein levels and oxidative lung injury in experimental diabetes: effect of oxovanadium(IV) complex based on thiosemicarbazone. Toxicol Mech Methods 2021; 31:581-588. [PMID: 34240667 DOI: 10.1080/15376516.2021.1941462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Diabetes mellitus (DM) is chronic and metabolic disorder, which is mainly attributed by hyperglycemia. Vanadium salts and their oxo-complexes have been shown to possess insulin-mimetic and anti-diabetic activities in animal models and diabetic patients. The main goal of this study was to investigate the protective effect of oxovanadium(IV) complex based on thiosemicarbazone (VOL) [L: (N(1)-2,4-dihydroxybenzylidene-N-(4)-2-hydroxybenzylidene-S-methyl-isothiosemicarbazidato-oxovanadium(IV)] on glycoprotein components levels and oxidative lung injury of streptozotocin (STZ)-induced diabetic rats. Male Swiss albino rats were separated into four groups. Group I (n = 5): Control (normal) animals, Group II (n = 5): Control animals administered with VOL, Group III (n = 6): STZ-induced diabetic animals, and Group IV (n = 5): STZ-induced diabetic rats treated with VOL. VOL was given to the experimental animals by gavage at a dose of 0.2 mM/kg body weight every day for 12 days. Diabetes was induced by single intraperitoneal injection of STZ (65 mg/kg body weight). On the 12th day, lung tissue samples were taken. Glycoprotein components, advanced oxidation protein products, protein carbonyl, hydroxyproline levels, and prolidase, arginase, xanthine oxidase, catalase, superoxide dismutase, glutathione peroxidase, glutathione reductase, glutathione-S-transferase and adenosine deaminase activities significantly increased whereas aryl esterase, paraoxonase-1, carbonic anhydrase, Na+/K+-ATPase activities remarkably decreased in lung tissue of diabetic rats. Treatment with VOL reversed these effects showing a beneficial effect. The present study shows that VOL has a protective effect against diabetes-induced lung damage as well as on abnormal glycoprotein component levels.
Collapse
Affiliation(s)
- Bertan Boran Bayrak
- Department of Chemistry, Division of Biochemistry, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Sevim Tunali
- Department of Chemistry, Division of Biochemistry, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Tulay Bal-Demirci
- Department of Chemistry, Division of Inorganic Chemistry, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Bahri Ulkuseven
- Department of Chemistry, Division of Inorganic Chemistry, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Refiye Yanardag
- Department of Chemistry, Division of Biochemistry, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| |
Collapse
|
16
|
Hautbergue T, Antigny F, Boët A, Haddad F, Masson B, Lambert M, Delaporte A, Menager JB, Savale L, Pavec JL, Fadel E, Humbert M, Junot C, Fenaille F, Colsch B, Mercier O. Right Ventricle Remodeling Metabolic Signature in Experimental Pulmonary Hypertension Models of Chronic Hypoxia and Monocrotaline Exposure. Cells 2021; 10:1559. [PMID: 34205639 PMCID: PMC8235667 DOI: 10.3390/cells10061559] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/12/2021] [Accepted: 06/16/2021] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Over time and despite optimal medical management of patients with pulmonary hypertension (PH), the right ventricle (RV) function deteriorates from an adaptive to maladaptive phenotype, leading to RV failure (RVF). Although RV function is well recognized as a prognostic factor of PH, no predictive factor of RVF episodes has been elucidated so far. We hypothesized that determining RV metabolic alterations could help to understand the mechanism link to the deterioration of RV function as well as help to identify new biomarkers of RV failure. METHODS In the current study, we aimed to characterize the metabolic reprogramming associated with the RV remodeling phenotype during experimental PH induced by chronic-hypoxia-(CH) exposure or monocrotaline-(MCT) exposure in rats. Three weeks after PH initiation, we hemodynamically characterized PH (echocardiography and RV catheterization), and then we used an untargeted metabolomics approach based on liquid chromatography coupled to high-resolution mass spectrometry to analyze RV and LV tissues in addition to plasma samples from MCT-PH and CH-PH rat models. RESULTS CH exposure induced adaptive RV phenotype as opposed to MCT exposure which induced maladaptive RV phenotype. We found that predominant alterations of arginine, pyrimidine, purine, and tryptophan metabolic pathways were detected on the heart (LV+RV) and plasma samples regardless of the PH model. Acetylspermidine, putrescine, guanidinoacetate RV biopsy levels, and cytosine, deoxycytidine, deoxyuridine, and plasmatic thymidine levels were correlated to RV function in the CH-PH model. It was less likely correlated in the MCT model. These pathways are well described to regulate cell proliferation, cell hypertrophy, and cardioprotection. These findings open novel research perspectives to find biomarkers for early detection of RV failure in PH.
Collapse
Affiliation(s)
- Thaïs Hautbergue
- Département Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, INRAE, SPI, MetaboHUB, 91191 Gif-sur-Yvette, France; (T.H.); (C.J.); (F.F.); (B.C.)
| | - Fabrice Antigny
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - Angèle Boët
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Réanimation des Cardiopathies Congénitales, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| | - François Haddad
- Cardiovascular Medicine, Stanford Hospital, Stanford University, Stanford, CA 94305, USA;
| | - Bastien Masson
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - Mélanie Lambert
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - Amélie Delaporte
- Service d’Anesthésie, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France;
| | - Jean-Baptiste Menager
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| | - Laurent Savale
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Jérôme Le Pavec
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| | - Elie Fadel
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| | - Marc Humbert
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Christophe Junot
- Département Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, INRAE, SPI, MetaboHUB, 91191 Gif-sur-Yvette, France; (T.H.); (C.J.); (F.F.); (B.C.)
| | - François Fenaille
- Département Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, INRAE, SPI, MetaboHUB, 91191 Gif-sur-Yvette, France; (T.H.); (C.J.); (F.F.); (B.C.)
| | - Benoit Colsch
- Département Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, INRAE, SPI, MetaboHUB, 91191 Gif-sur-Yvette, France; (T.H.); (C.J.); (F.F.); (B.C.)
| | - Olaf Mercier
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| |
Collapse
|
17
|
Grunwell JR, Rad MG, Stephenson ST, Mohammad AF, Opolka C, Fitzpatrick AM, Kamaleswaran R. Machine Learning-Based Discovery of a Gene Expression Signature in Pediatric Acute Respiratory Distress Syndrome. Crit Care Explor 2021; 3:e0431. [PMID: 34151274 PMCID: PMC8208445 DOI: 10.1097/cce.0000000000000431] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVES To identify differentially expressed genes and networks from the airway cells within 72 hours of intubation of children with and without pediatric acute respiratory distress syndrome. To test the use of a neutrophil transcription reporter assay to identify immunogenic responses to airway fluid from children with and without pediatric acute respiratory distress syndrome. DESIGN Prospective cohort study. SETTING Thirty-six bed academic PICU. PATIENTS Fifty-four immunocompetent children, 28 with pediatric acute respiratory distress syndrome, who were between 2 days to 18 years old within 72 hours of intubation for acute hypoxemic respiratory failure. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS We applied machine learning methods to a Nanostring transcriptomics on primary airway cells and a neutrophil reporter assay to discover gene networks differentiating pediatric acute respiratory distress syndrome from no pediatric acute respiratory distress syndrome. An analysis of moderate or severe pediatric acute respiratory distress syndrome versus no or mild pediatric acute respiratory distress syndrome was performed. Pathway network visualization was used to map pathways from 62 genes selected by ElasticNet associated with pediatric acute respiratory distress syndrome. The Janus kinase/signal transducer and activator of transcription pathway emerged. Support vector machine performed best for the primary airway cells and the neutrophil reporter assay using a leave-one-out cross-validation with an area under the operating curve and 95% CI of 0.75 (0.63-0.87) and 0.80 (0.70-1.0), respectively. CONCLUSIONS We identified gene networks important to the pediatric acute respiratory distress syndrome airway immune response using semitargeted transcriptomics from primary airway cells and a neutrophil reporter assay. These pathways will drive mechanistic investigations into pediatric acute respiratory distress syndrome. Further studies are needed to validate our findings and to test our models.
Collapse
Affiliation(s)
- Jocelyn R Grunwell
- Children's Healthcare of Atlanta, Egleston Hospital, Atlanta, GA
- Emory University School of Medicine, Department of Pediatrics, Division of Critical Care Medicine, Atlanta, GA
| | - Milad G Rad
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Susan T Stephenson
- Emory University School of Medicine, Department of Pediatrics, Division of Critical Care Medicine, Atlanta, GA
| | - Ahmad F Mohammad
- Emory University School of Medicine, Department of Pediatrics, Division of Critical Care Medicine, Atlanta, GA
| | - Cydney Opolka
- Children's Healthcare of Atlanta, Egleston Hospital, Atlanta, GA
| | - Anne M Fitzpatrick
- Emory University School of Medicine, Department of Pediatrics, Division of Critical Care Medicine, Atlanta, GA
| | - Rishikesan Kamaleswaran
- Emory University School of Medicine, Department of Pediatrics, Division of Critical Care Medicine, Atlanta, GA
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA
| |
Collapse
|
18
|
Koroglu-Aydın P, Bayrak BB, Bugan I, Karabulut-Bulan O, Yanardag R. Histological and biochemical investigation of the renoprotective effects of metformin in diabetic and prostate cancer model. Toxicol Mech Methods 2021; 31:489-500. [PMID: 34039237 DOI: 10.1080/15376516.2021.1919810] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Diabetes and cancer have common physiological and biochemical mechanisms. Metformin is the preferred drug of choice for the treatment of diabetes. Prostate cancer can be modeled in by injection of MAT-Lylu cells. A model of diabetes in rats is induced by streptozotocin injectıon. In the current study, we explored the mechanisms by which diabetes accelerates cancer, and evaluated the effects of metformin to know whether it has any impact against the damage caused by cancer and diabetic + cancer via histopathological and biochemical parameters of kidney tissue. METHODS The experiment was carried out in rats. Groups 1-Control, 2- Diabetic, 3-Cancer, 4-Diabetic + cancer, 5-Diabetic + cancer + metformin, 6-Cancer + metformin. Metformin treatment was applied by gavage every day. The research ended on the 14th day. The collected kidney tissue sections were stained with Hematoxylin-Eosin. RESULTS Histological evaluation showed moderate to severe damage to the kidney tissue following diabetic and cancer processess. In diabetic, cancer and diabetic + cancer groups, reduced glutathione levels, total antioxidant status, sodium/potassium-ATPase and paraoxonase1 activities were found to be significantly abated. While advanced oxidized protein products, lipid peroxidation, nitric oxide, tumor necrosis factor-alpha, reactive oxygen species levels, total oxidant status, catalase, superoxide dismutase, glutathione-related antioxidant enzymes, myeloperoxidase, and arginase activities were significantly raised. The administration of metformin reversed these defects. The outcome of the reveals that histopathological and biochemical damage in cancer and diabetes + cancer groups decreased in the groups that received metformin. CONCLUSION In conclusion, metformin treatment can be considered an adjuvant candidate for kidney tissue in diabetes, prostate cancer and cancer therapy related damage.
Collapse
Affiliation(s)
- Pınar Koroglu-Aydın
- Department of Histology and Embryology, Faculty of Medicine, Halic University, Istanbul, Turkey
| | - Bertan Boran Bayrak
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Ilknur Bugan
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Vezneciler, Turkey
| | - Omur Karabulut-Bulan
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Vezneciler, Turkey
| | - Refiye Yanardag
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| |
Collapse
|
19
|
Zhang L, Qu S, Wang L, Wang C, Yu Q, Zhang Z, Diao Y, Zhang B, Li Y, Shi Y, Wang P. Tianlongkechuanling Inhibits Pulmonary Fibrosis Through Down-Regulation of Arginase-Ornithine Pathway. Front Pharmacol 2021; 12:661129. [PMID: 33995084 PMCID: PMC8114272 DOI: 10.3389/fphar.2021.661129] [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: 01/30/2021] [Accepted: 03/30/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Pulmonary Fibrosis (PF) is an interstitial lung disease characterized by excessive accumulation of extracellular matrix in the lungs, which disrupts the structure and gas exchange of the alveoli. There are only two approved therapies for PF, nintedanib (Nib) and pirfenidone. Therefore, the use of Chinese medicine for PF is attracting attention. Tianlongkechuanling (TL) is an effective Chinese formula that has been applied clinically to alleviate PF, which can enhance lung function and quality of life. Purpose: The potential effects and specific mechanisms of TL have not been fully explored, yet. In the present study, proteomics was performed to explore the therapeutic protein targets of TL on Bleomycin (BLM)-induced Pulmonary Fibrosis. Method: BLM-induced PF mice models were established. Hematoxylineosin staining and Masson staining were used to analyze histopathological changes and collagen deposition. To screen the differential proteins expression between the Control, BLM, BLM + TL and BLM + Nib (BLM + nintedanib) groups, quantitative proteomics was performed using tandem mass tag (TMT) labeling with nanoLC-MS/MS [nano liquid chromatographymass spectrometry]). Changes in the profiles of the expressed proteins were analyzed using the bioinformatics tools Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG). The protein–protein interactions (PPI) were established by STRING. Expressions of α-smooth muscle actin (α-SMA), Collagen I (Col1a1), Fibronectin (Fn1) and enzymes in arginase-ornithine pathway were detected by Western blot or RT-PCR. Result: TL treatments significantly ameliorated BLM-induced collagen deposition in lung tissues. Moreover, TL can inhibit the protein expressions of α-SMA and the mRNA expressions of Col1a1 and Fn1. Using TMT technology, we observed 253 differentially expressed proteins related to PPI networks and involved different KEGG pathways. Arginase-ornithine pathway is highly significant. The expression of arginase1 (Arg1), carbamoyltransferase (OTC), carbamoy-phosphate synthase (CPS1), argininosuccinate synthase (ASS1), ornithine aminotransferase (OAT) argininosuccinate lyase (ASL) and inducible nitric oxide synthase (iNOS) was significantly decreased after TL treatments. Conclusion: Administration of TL in BLM-induced mice resulted in decreasing pulmonary fibrosis. Our findings propose that the down regulation of arginase-ornithine pathway expression with the reduction of arginase biosynthesis is a central mechanism and potential treatment for pulmonary fibrosis with the prevention of TL.
Collapse
Affiliation(s)
- Lili Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Sihao Qu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Lu Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Chunguo Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qinghe Yu
- Traditional Chinese Medicine, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhimin Zhang
- Traditional Chinese Medicine, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yirui Diao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Binbin Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yadong Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.,Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China
| | - Peng Wang
- Traditional Chinese Medicine, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
20
|
Falck-Jones S, Vangeti S, Yu M, Falck-Jones R, Cagigi A, Badolati I, Österberg B, Lautenbach MJ, Åhlberg E, Lin A, Lepzien R, Szurgot I, Lenart K, Hellgren F, Maecker H, Sälde J, Albert J, Johansson N, Bell M, Loré K, Färnert A, Smed-Sörensen A. Functional monocytic myeloid-derived suppressor cells increase in blood but not airways and predict COVID-19 severity. J Clin Invest 2021; 131:144734. [PMID: 33492309 DOI: 10.1172/jci144734] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
The immunopathology of coronavirus disease 2019 (COVID-19) remains enigmatic, causing immunodysregulation and T cell lymphopenia. Monocytic myeloid-derived suppressor cells (M-MDSCs) are T cell suppressors that expand in inflammatory conditions, but their role in acute respiratory infections remains unclear. We studied the blood and airways of patients with COVID-19 across disease severities at multiple time points. M-MDSC frequencies were elevated in blood but not in nasopharyngeal or endotracheal aspirates of patients with COVID-19 compared with healthy controls. M-MDSCs isolated from patients with COVID-19 suppressed T cell proliferation and IFN-γ production partly via an arginase 1-dependent (Arg-1-dependent) mechanism. Furthermore, patients showed increased Arg-1 and IL-6 plasma levels. Patients with COVID-19 had fewer T cells and downregulated expression of the CD3ζ chain. Ordinal regression showed that early M-MDSC frequency predicted subsequent disease severity. In conclusion, M-MDSCs expanded in the blood of patients with COVID-19, suppressed T cells, and were strongly associated with disease severity, indicating a role for M-MDSCs in the dysregulated COVID-19 immune response.
Collapse
Affiliation(s)
- Sara Falck-Jones
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sindhu Vangeti
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Meng Yu
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ryan Falck-Jones
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Alberto Cagigi
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Isabella Badolati
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Björn Österberg
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Maximilian Julius Lautenbach
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Eric Åhlberg
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ang Lin
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Stemirna Therapeutics Inc., Shanghai, China
| | - Rico Lepzien
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Inga Szurgot
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Klara Lenart
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Fredrika Hellgren
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Holden Maecker
- Stanford University Medical Center, Stanford, California, USA
| | - Jörgen Sälde
- Health Care Services Stockholm County (SLSO), Stockholm, Sweden
| | - Jan Albert
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Microbiology and
| | - Niclas Johansson
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Max Bell
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Karin Loré
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Färnert
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
21
|
Xue C, Wu N, Fan Y, Ma J, Ye Q. Distinct metabolic features in the plasma of patients with silicosis and dust-exposed workers in China: a case-control study. BMC Pulm Med 2021; 21:91. [PMID: 33731064 PMCID: PMC7971960 DOI: 10.1186/s12890-021-01462-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/09/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Silicosis is a progressive pneumoconiosis characterized by interstitial fibrosis following exposure to silica dust. The role of metabolic dysregulation in the pathogenesis of silicosis has not been investigated in detail. This study aimed to identify different metabolic features in the plasma of patients with silicosis and dust-exposed workers without silicosis in metabolomics studies. METHODS Patients with silicosis, dust-exposed workers (DEWs) without silicosis and age-matched healthy controls were recruited in a case-control study. The metabolomics analyses by ultra-high performance liquid chromatography-mass spectrometry were conducted. Distinct metabolic features (DMFs) were identified in the pilot study and were validated in the validation study. The enriched signalling pathways of these DMFs were determined. The ability of DMFs to discriminate among the groups was analysed through receiver operating characteristic (ROC) curves. The correlations between DMFs and clinical features were also explored. RESULTS Twenty-nine DMFs and 9 DMFs were detected and had the same trend in the pilot study and the validation study in the plasma of the DEW and silicosis groups, respectively. Sphingolipid metabolism was the major metabolic pathway in the DEWs, and arginine and proline metabolism was associated with silicosis. Twenty DMFs in the DEWs and 3 DMFs in the patients with silicosis showed a discriminatory ability with ROC curve analysis. The abundance of kynurenine was higher in Stage III silicosis than in Stage I or Stage II silicosis. L-arginine and kynurenine were both negatively correlated with the percentage of forced vital capacity predicted in silicosis. CONCLUSIONS Distinct metabolic features in the plasma of DEWs and the patients with silicosis were found to be different. Sphingolipid metabolism and arginine and proline metabolism were identified as the major metabolic pathway in the DEW and silicosis groups, respectively. L-arginine and kynurenine were correlated with the severity of silicosis.
Collapse
Affiliation(s)
- Changjiang Xue
- Department of Occupational Medicine and Toxicology, Clinical Centre for Interstitial Lung Diseases, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chao-Yang District, Beijing, 100020, China
| | - Na Wu
- Department of Occupational Medicine and Toxicology, Clinical Centre for Interstitial Lung Diseases, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chao-Yang District, Beijing, 100020, China
| | - Yali Fan
- Department of Occupational Medicine and Toxicology, Clinical Centre for Interstitial Lung Diseases, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chao-Yang District, Beijing, 100020, China
| | - Jing Ma
- Department of Occupational Medicine and Toxicology, Clinical Centre for Interstitial Lung Diseases, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chao-Yang District, Beijing, 100020, China
| | - Qiao Ye
- Department of Occupational Medicine and Toxicology, Clinical Centre for Interstitial Lung Diseases, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chao-Yang District, Beijing, 100020, China.
| |
Collapse
|
22
|
Li JM, Yang DC, Oldham J, Linderholm A, Zhang J, Liu J, Kenyon NJ, Chen CH. Therapeutic targeting of argininosuccinate synthase 1 (ASS1)-deficient pulmonary fibrosis. Mol Ther 2021; 29:1487-1500. [PMID: 33508432 DOI: 10.1016/j.ymthe.2021.01.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 12/10/2020] [Accepted: 01/19/2021] [Indexed: 12/30/2022] Open
Abstract
Argininosuccinate synthase 1 (ASS1) serves as a critical enzyme in arginine biosynthesis; however, its role in interstitial lung diseases, particularly idiopathic pulmonary fibrosis (IPF), remains largely unknown. This study aims at characterization and targeting of ASS1 deficiency in pulmonary fibrosis. We find that ASS1 was significantly decreased and inversely correlated with fibrotic status. Transcriptional downregulation of ASS1 was noted in fibroblastic foci of primary lung fibroblasts isolated from IPF patients. Genetic manipulations of ASS1 studies confirm that ASS1 expression inhibited fibroblast cell proliferation, migration, and invasion. We further show that the hepatocyte growth factor receptor (Met) receptor was activated and acted upstream of the Src-STAT3 axis signaling in ASS1-knockdown fibroblasts. Interestingly, both arginine-free conditions and arginine deiminase treatment were demonstrated to kill fibrotic fibroblasts, attenuated bleomycin-induced pulmonary fibrosis in mice, as well as synergistically increased nintedanib efficacy. Our data suggest ASS1 deficiency as a druggable target and also provide a unique therapeutic strategy against pulmonary fibrosis.
Collapse
Affiliation(s)
- Ji-Min Li
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, USA; Division of Nephrology, Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
| | - David C Yang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, USA; Division of Nephrology, Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Justin Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, USA
| | - Angela Linderholm
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, USA
| | - Jun Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, USA; Division of Nephrology, Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Jun Liu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, USA; Division of Nephrology, Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Nicholas J Kenyon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, USA
| | - Ching-Hsien Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, USA; Division of Nephrology, Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
23
|
Mamazhakypov A, Weiß A, Zukunft S, Sydykov A, Kojonazarov B, Wilhelm J, Vroom C, Petrovic A, Kosanovic D, Weissmann N, Seeger W, Fleming I, Iglarz M, Grimminger F, Ghofrani HA, Pullamsetti SS, Schermuly RT. Effects of macitentan and tadalafil monotherapy or their combination on the right ventricle and plasma metabolites in pulmonary hypertensive rats. Pulm Circ 2020; 10:2045894020947283. [PMID: 33240483 PMCID: PMC7672745 DOI: 10.1177/2045894020947283] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 07/10/2020] [Indexed: 12/19/2022] Open
Abstract
Pulmonary arterial hypertension is a severe respiratory disease characterized by pulmonary artery remodeling. RV dysfunction and dysregulated circulating metabolomics are associated with adverse outcomes in pulmonary arterial hypertension. We investigated effects of tadalafil and macitentan alone or in combination on the RV and plasma metabolomics in SuHx and PAB models. For SuHx model, rats were injected with SU5416 and exposed to hypoxia for three weeks and then were returned to normoxia and treated with either tadalafil (10 mg/kg in chow) or macitentan (10 mg/kg in chow) or their combination (both 10 mg/kg in chow) for two weeks. For PAB model, rats were subjected to either sham or PAB surgery for three weeks and treated with above-mentioned drugs from week 1 to week 3. Following terminal echocardiographic and hemodynamic measurements, tissue samples were collected for metabolomic, histological and gene expression analysis. Both SuHx and PAB rats developed RV remodeling/dysfunction with severe and mild plasma metabolomic alterations, respectively. In SuHx rats, tadalafil and macitentan alone or in combination improved RV remodeling/function with the effects of macitentan and combination therapy being superior to tadalafil. All therapies similarly attenuated SuHx-induced changes in plasma metabolomics. In PAB rats, only macitentan improved RV remodeling/function, while only tadalafil attenuated PAB-induced changes in plasma metabolomics.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Astrid Weiß
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Sven Zukunft
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany & German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany
| | - Akylbek Sydykov
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Baktybek Kojonazarov
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Jochen Wilhelm
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Christina Vroom
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Aleksandar Petrovic
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Djuro Kosanovic
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany.,Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Norbert Weissmann
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Werner Seeger
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany.,Department of Lung Development and Remodelling, Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany & German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany
| | - Marc Iglarz
- Actelion Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Friedrich Grimminger
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Hossein A Ghofrani
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Soni S Pullamsetti
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany.,Department of Lung Development and Remodelling, Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ralph T Schermuly
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center, Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
24
|
Arginine and Arginine/ADMA Ratio Predict 90-Day Mortality in Patients with Out-of-Hospital Cardiac Arrest-Results from the Prospective, Observational COMMUNICATE Trial. J Clin Med 2020; 9:jcm9123815. [PMID: 33255752 PMCID: PMC7760544 DOI: 10.3390/jcm9123815] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/08/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
(1) Background: In patients with shock, the L-arginine nitric oxide pathway is activated, causing an elevation of nitric oxide, asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA) levels. Whether these metabolites provide prognostic information in patients after out-of-hospital cardiac arrest (OHCA) remains unclear. (2) Methods: We prospectively included OHCA patients, recorded clinical parameters and measured plasma ADMA, SDMA and Arginine levels by liquid chromatography tandem mass spectrometry (LC-MS). The primary endpoint was 90-day mortality. (3) Results: Of 263 patients, 130 (49.4%) died within 90 days after OHCA. Compared to survivors, non-survivors had significantly higher levels of ADMA and lower Arginine and Arginine/ADMA ratios in univariable regression analyses. Arginine levels and Arginine/ADMA ratio were significantly associated with 90-day mortality (OR 0.51 (95%CI 0.34 to 0.76), p < 0.01 and OR 0.40 (95%CI 0.26 to 0.61), p < 0.001, respectively). These associations remained significant in several multivariable models. Arginine/ADMA ratio had the highest predictive value with an area under the curve (AUC) of 0.67 for 90-day mortality. Results for secondary outcomes were similar with significant associations with in-hospital mortality and neurological outcome. (4) Conclusion: Arginine and Arginine/ADMA ratio were independently associated with 90-day mortality and other adverse outcomes in patients after OHCA. Whether therapeutic modification of the L-arginine-nitric oxide pathway has the potential to improve outcome should be evaluated.
Collapse
|
25
|
Patel BS, Ravix J, Pabelick C, Prakash YS. Class C GPCRs in the airway. Curr Opin Pharmacol 2020; 51:19-28. [PMID: 32375079 DOI: 10.1016/j.coph.2020.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/17/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023]
Abstract
Understanding and targeting of GPCRs remain a critical aspect of airway pharmacology and therapeutics for diseases such as asthma or COPD. Most attention has been on the large Class A GPCRs towards improved bronchodilation and blunting of remodeling. Better known in the central or peripheral nervous system, there is increasing evidence that Class C GPCRs which include metabotropic glutamate and GABA receptors, the calcium sensing receptor, sweet/umami taste receptors and a number of orphan receptors, can contribute to airway structure and function. In this review, we will summarize current state of knowledge regarding the pharmacology of Class C GPCRs, their expression and potential functions in the airways, and the application of pharmacological agents targeting this group in the context of airway diseases.
Collapse
Affiliation(s)
- Brijeshkumar S Patel
- Department of Anesthesiologyand Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jovanka Ravix
- Department of Anesthesiologyand Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Christina Pabelick
- Department of Anesthesiologyand Perioperative Medicine, Mayo Clinic, Rochester, MN, United States; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Y S Prakash
- Department of Anesthesiologyand Perioperative Medicine, Mayo Clinic, Rochester, MN, United States; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
26
|
Song J, Eghan K, Lee S, Park JS, Yoon S, Pimtong W, Kim WK. A Phenotypic and Genotypic Evaluation of Developmental Toxicity of Polyhexamethylene Guanidine Phosphate Using Zebrafish Embryo/Larvae. TOXICS 2020; 8:E33. [PMID: 32370250 PMCID: PMC7355787 DOI: 10.3390/toxics8020033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/28/2020] [Accepted: 04/28/2020] [Indexed: 11/16/2022]
Abstract
Polyhexamethylene guanidine-phosphate (PHMG-P), a guanidine-based cationic antimicrobial polymer, is an effective antimicrobial biocide, potent even at low concentrations. Due to its resilient bactericidal properties, it has been used extensively in consumer products. It was safely used until its use in humidifiers led to a catastrophic event in South Korea. Epidemiological studies have linked the use of PHMG-P as a humidifier disinfectant to pulmonary fibrosis. However, little is known about its harmful impacts other than pulmonary fibrosis. Thus, we applied a zebrafish embryo/larvae model to evaluate developmental and cardiotoxic effects and transcriptome changes using RNA-sequencing. Zebrafish embryos were exposed to 0.1, 0.2, 0.3, 0.4, 0.5, 1, and 2 mg/L of PHMG-P from 3 h to 96 h post fertilization. 2 mg/L of PHMG-P resulted in total mortality and an LC50 value at 96 h was determined at 1.18 mg/L. Significant developmental changes were not observed but the heart rate of zebrafish larvae was significantly altered. In transcriptome analysis, immune and inflammatory responses were significantly affected similarly to those in epidemiological studies. Our qPCR analysis (Itgb1b, TNC, Arg1, Arg2, IL-1β, Serpine-1, and Ptgs2b) also confirmed this following a 96 h exposure to 0.4 mg/L of PHMG-P. Based on our results, PHMG-P might induce lethal and cardiotoxic effects in zebrafish, and crucial transcriptome changes were linked to immune and inflammatory response.
Collapse
Affiliation(s)
- Jeongah Song
- Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, Jeongeup 56212, Korea;
| | - Kojo Eghan
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Korea; (K.E.); (S.L.); (J.-S.P.); (S.Y.)
- Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Korea
| | - Sangwoo Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Korea; (K.E.); (S.L.); (J.-S.P.); (S.Y.)
| | - Jong-Su Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Korea; (K.E.); (S.L.); (J.-S.P.); (S.Y.)
| | - Seokjoo Yoon
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Korea; (K.E.); (S.L.); (J.-S.P.); (S.Y.)
- Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Korea
| | - Wittaya Pimtong
- Nano Environmental and Health Safety Research Team, National Nanotechnology Center, National Science and Technology Development Agency, Pathum Thani 12120, Thailand;
| | - Woo-Keun Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Korea; (K.E.); (S.L.); (J.-S.P.); (S.Y.)
- Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Korea
| |
Collapse
|
27
|
Otoupalova E, Smith S, Cheng G, Thannickal VJ. Oxidative Stress in Pulmonary Fibrosis. Compr Physiol 2020; 10:509-547. [PMID: 32163196 DOI: 10.1002/cphy.c190017] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Oxidative stress has been linked to various disease states as well as physiological aging. The lungs are uniquely exposed to a highly oxidizing environment and have evolved several mechanisms to attenuate oxidative stress. Idiopathic pulmonary fibrosis (IPF) is a progressive age-related disorder that leads to architectural remodeling, impaired gas exchange, respiratory failure, and death. In this article, we discuss cellular sources of oxidant production, and antioxidant defenses, both enzymatic and nonenzymatic. We outline the current understanding of the pathogenesis of IPF and how oxidative stress contributes to fibrosis. Further, we link oxidative stress to the biology of aging that involves DNA damage responses, loss of proteostasis, and mitochondrial dysfunction. We discuss the recent findings on the role of reactive oxygen species (ROS) in specific fibrotic processes such as macrophage polarization and immunosenescence, alveolar epithelial cell apoptosis and senescence, myofibroblast differentiation and senescence, and alterations in the acellular extracellular matrix. Finally, we provide an overview of the current preclinical studies and clinical trials targeting oxidative stress in fibrosis and potential new strategies for future therapeutic interventions. © 2020 American Physiological Society. Compr Physiol 10:509-547, 2020.
Collapse
Affiliation(s)
- Eva Otoupalova
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sam Smith
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Guangjie Cheng
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
28
|
Kuo WK, Liu YC, Chu CM, Hua CC, Huang CY, Liu MH, Wang CH. Amino Acid-Based Metabolic Indexes Identify Patients With Chronic Obstructive Pulmonary Disease And Further Discriminates Patients In Advanced BODE Stages. Int J Chron Obstruct Pulmon Dis 2019; 14:2257-2266. [PMID: 31631995 PMCID: PMC6778323 DOI: 10.2147/copd.s220557] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/12/2019] [Indexed: 01/13/2023] Open
Abstract
Background The BODE index is a multidimensional grading system for predicting the prognoses of patients with chronic obstructive pulmonary disease (COPD). This study investigated whether an amino acids-based metabolic profile developed for heart failure patients (including histidine, ornithine, phenylalanine, and leucine) could identify COPD patients and further discriminates COPD patients in advanced BODE stages. Methods Ultra-performance liquid chromatography was performed on 119 participants, including 75 COPD patients at different BODE stages and 44 normal controls. Albumin, pre-albumin, transferrin, high sensitivity C-reactive protein, and hand grip strength were also measured. Receiver operating characteristic curves and area under curves were used for estimation. Results The BODE points in our patients were 3.29 [95% confidence interval (CI) = 2.74-3.85]. Compared to normal controls, COPD patients had lower leucine but higher ornithine levels. A COPD score, developed based on leucine and ornithine, significantly discriminated COPD from normal controls [odds ratio (OR) = 2.71, 95% CI = 1.83-4.04, p <0.001]. A COPD score of ≥ 3.00 had an OR of 15.58 (95% CI = 5.96-40.73, p <0.001). In COPD patients from BODE 1 to BODE 4, the levels of histidine, ornithine and phenylalanine increased significantly. In multivariable analysis, histidine and phenylalanine were independently able to distinguish BODE stages 3 and 4 from BODE 1 and were adopted to develop a metabolic score. Metabolic scores identified patients at BODE 3 and 4 (OR = 2.74, 95% CI =1.41-5.29, p = 0.003) better than hand grip strength, high sensitive C-reactive protein, albumin, pre-albumin, and transferrin value. A metabolic score of ≥9.53 significantly discriminated BODE 3 and 4 from BODE 1 and 2 (OR = 8.56, 95% CI = 2.77-26.39, p <0.001). Conclusion Amino acid-based COPD score and metabolic score discriminate COPD patients from normal controls, and identify patients in advanced stages of COPD.
Collapse
Affiliation(s)
- Wei-Ke Kuo
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Yu-Chih Liu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
- Chang Gung University, College of Medicine, Taoyuan, Taiwan
| | - Chien-Ming Chu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chung-Ching Hua
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
- Chang Gung University, College of Medicine, Taoyuan, Taiwan
| | - Chih-Yu Huang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Min-Hui Liu
- Chang Gung University, College of Medicine, Taoyuan, Taiwan
- Heart Failure Research Center, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chao-Hung Wang
- Chang Gung University, College of Medicine, Taoyuan, Taiwan
- Heart Failure Research Center, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| |
Collapse
|
29
|
Haydar D, Cory TJ, Birket SE, Murphy BS, Pennypacker KR, Sinai AP, Feola DJ. Azithromycin Polarizes Macrophages to an M2 Phenotype via Inhibition of the STAT1 and NF-κB Signaling Pathways. THE JOURNAL OF IMMUNOLOGY 2019; 203:1021-1030. [PMID: 31263039 DOI: 10.4049/jimmunol.1801228] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 06/14/2019] [Indexed: 12/26/2022]
Abstract
Azithromycin is effective at controlling exaggerated inflammation and slowing the long-term decline of lung function in patients with cystic fibrosis. We previously demonstrated that the drug shifts macrophage polarization toward an alternative, anti-inflammatory phenotype. In this study we investigated the immunomodulatory mechanism of azithromycin through its alteration of signaling via the NF-κB and STAT1 pathways. J774 murine macrophages were plated, polarized (with IFN-γ, IL-4/-13, or with azithromycin plus IFN-γ) and stimulated with LPS. The effect of azithromycin on NF-κB and STAT1 signaling mediators was assessed by Western blot, homogeneous time-resolved fluorescence assay, nuclear translocation assay, and immunofluorescence. The drug's effect on gene and protein expression of arginase was evaluated as a marker of alternative macrophage activation. Azithromycin blocked NF-κB activation by decreasing p65 nuclear translocation, although blunting the degradation of IκBα was due, at least in part, to a decrease in IKKβ kinase activity. A direct correlation was observed between increasing azithromycin concentrations and increased IKKβ protein expression. Moreover, incubation with the IKKβ inhibitor IKK16 decreased arginase expression and activity in azithromycin-treated cells but not in cells treated with IL-4 and IL-13. Importantly, azithromycin treatment also decreased STAT1 phosphorylation in a concentration-dependent manner, an effect that was reversed with IKK16 treatment. We conclude that azithromycin anti-inflammatory mechanisms involve inhibition of the STAT1 and NF-κB signaling pathways through the drug's effect on p65 nuclear translocation and IKKβ.
Collapse
Affiliation(s)
- Dalia Haydar
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, KY 40536
| | - Theodore J Cory
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Susan E Birket
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama-Birmingham, Birmingham, AL 35294
| | | | - Keith R Pennypacker
- Department of Neurology, University of Kentucky College of Medicine, Lexington, KY 40536.,Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536; and
| | - Anthony P Sinai
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY 40536
| | - David J Feola
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, KY 40536;
| |
Collapse
|
30
|
Cho A, Eskandari R, Granlund KL, Keshari KR. Hyperpolarized [6- 13C, 15N 3]-Arginine as a Probe for in Vivo Arginase Activity. ACS Chem Biol 2019; 14:665-673. [PMID: 30893552 DOI: 10.1021/acschembio.8b01044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Alterations in arginase enzyme expression are linked with various diseases and have been shown to support disease progression, thus motivating the development of an imaging probe for this enzymatic target. 13C-enriched arginine can be used as a hyperpolarized (HP) magnetic resonance (MR) probe for arginase flux since the arginine carbon-6 resonance (157 ppm) is converted to urea (163 ppm) following arginase-catalyzed hydrolysis. However, scalar relaxation from adjacent 14N-nuclei shortens cabon-6 T 1 and T 2 times, yielding poor spectral properties. To address these limitations, we report the synthesis of [6-13C,15N3]-arginine and demonstrate that 15N-enrichment increases carbon-6 relaxation times, thereby improving signal-to-noise ratio and spectral resolution. By overcoming these limitations with this novel isotope-labeling scheme, we were able to perform in vitro and in vivo arginase activity measurements with HP MR. We present HP [6-13C,15N3]-arginine as a noninvasive arginase imaging agent for preclinical studies, with the potential for future clinical diagnostic use.
Collapse
Affiliation(s)
- Andrew Cho
- Department of Biochemistry and Structural Biology, Weill Cornell Graduate School, New York, New York 10065, United States
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, New York 10065, United States
| | - Roozbeh Eskandari
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Kristin L. Granlund
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Kayvan R. Keshari
- Department of Biochemistry and Structural Biology, Weill Cornell Graduate School, New York, New York 10065, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| |
Collapse
|
31
|
Krystofova J, Pathipati P, Russ J, Sheldon A, Ferriero D. The Arginase Pathway in Neonatal Brain Hypoxia-Ischemia. Dev Neurosci 2019; 40:437-450. [PMID: 30995639 PMCID: PMC6784534 DOI: 10.1159/000496467] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022] Open
Abstract
Brain damage after hypoxia-ischemia (HI) occurs in an age-dependent manner. Neuroprotective strategies assumed to be effective in adults might have deleterious effects in the immature brain. In order to create effective therapies, the complex pathophysiology of HI in the developing brain requires exploring new mechanisms. Critical determinants of neuronal survival after HI are the extent of vascular dysfunction, inflammation, and oxidative stress, followed later by tissue repair. The key enzyme of these processes in the human body is arginase (ARG) that acts via the bioavailability of nitric oxide, and the synthesis of polyamines and proline. ARG is expressed throughout the brain in different cells. However, little is known about the effect of ARG in pathophysiological states of the brain, especially hypoxia-ischemia. Here, we summarize the role of ARG during neurodevelopment as well as in various brain pathologies.
Collapse
Affiliation(s)
- Jana Krystofova
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA,
| | - Praneeti Pathipati
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey Russ
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Ann Sheldon
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Donna Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
32
|
Dos Santos TM, Righetti RF, Camargo LDN, Saraiva-Romanholo BM, Aristoteles LRCRB, de Souza FCR, Fukuzaki S, Alonso-Vale MIC, Cruz MM, Prado CM, Leick EA, Martins MA, Tibério IFLC. Effect of Anti-IL17 Antibody Treatment Alone and in Combination With Rho-Kinase Inhibitor in a Murine Model of Asthma. Front Physiol 2018; 9:1183. [PMID: 30233389 PMCID: PMC6134017 DOI: 10.3389/fphys.2018.01183] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/06/2018] [Indexed: 12/12/2022] Open
Abstract
Background: Interleukin-17 (IL-17) and Rho-kinase (ROCK) play an important role in regulating the expression of inflammatory mediators, immune cell recruitment, hyper-responsiveness, tissue remodeling, and oxidative stress. Modulation of IL-17 and ROCK proteins may represent a promising approach for the treatment of this disease. Objective: To study the effects of an anti-IL17 neutralizing antibody and ROCK inhibitor treatments, separately and in combination, in a murine model of chronic allergy-induced lung inflammation. Methods: Sixty-four BALBc mice, were divided into eight groups (n = 8): SAL (saline-instilled); OVA (exposed-ovalbumin); SAL-RHOi (saline and ROCK inhibitor), OVA-RHOi (exposed-ovalbumin and ROCK inhibitor); SAL-anti-IL17 (saline and anti-IL17); OVA-anti-IL17 (exposed-ovalbumin and anti-IL17); SAL-RHOi-anti-IL17 (saline, ROCK inhibitor and anti-IL17); and OVA-RHOi-anti-IL17 (exposed-ovalbumin, anti-IL17, and ROCK inhibitor). A 28-day protocol of albumin treatment was used for sensitization and induction of pulmonary inflammation. The anti-IL17A neutralizing antibody (7.5 μg per treatment) was administered by intraperitoneal injection and ROCK inhibitor (Y-27632) intranasally (10 mg/kg), 1 h prior to each ovalbumin challenge (days 22, 24, 26, and 28). Results: Treatment with the anti-IL17 neutralizing antibody and ROCK inhibitor attenuated the percentage of maximal increase of respiratory system resistance and respiratory system elastance after challenge with methacholine and the inflammatory response markers evaluated (CD4+, CD8+, ROCK1, ROCK2, IL-4, IL-5, IL-6, IL-10 IL-13, IL-17, TNF-α, TGF-β, NF-κB, dendritic cells, iNOS, MMP-9, MMP-12, TIMP-1, FOXP3, isoprostane, biglycan, decorin, fibronectin, collagen fibers content and gene expression of IL-17, VAChT, and arginase) compared to the OVA group (p < 0.05). Treatment with anti-IL17 and the ROCK inhibitor together resulted in potentiation in decreasing the percentage of resistance increase after challenge with methacholine, decreased the number of IL-5 positive cells in the airway, and reduced, IL-5, TGF-β, FOXP3, ROCK1 and ROCK2 positive cells in the alveolar septa compared to the OVA-RHOi and OVA-anti-IL17 groups (p < 0.05). Conclusion: Anti-IL17 treatment alone or in conjunction with the ROCK inhibitor, modulates airway responsiveness, inflammation, tissue remodeling, and oxidative stress in mice with chronic allergic lung inflammation.
Collapse
Affiliation(s)
- Tabata M Dos Santos
- Department of Medicine, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil
| | - Renato F Righetti
- Department of Medicine, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil
| | - Leandro do N Camargo
- Department of Medicine, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil
| | - Beatriz M Saraiva-Romanholo
- Department of Medicine, Laboratory of Experimental Therapeutics, LIM-20, School of Medicine, University of São Paulo, São Paulo, Brazil.,Department of Medicine, University City of São Paulo (UNICID), São Paulo, Brazil
| | | | - Flávia C R de Souza
- Department of Medicine, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil
| | - Silvia Fukuzaki
- Department of Medicine, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil
| | | | - Maysa M Cruz
- Department of Biological Sciences, Federal University of São Paulo, Diadema, Brazil
| | - Carla M Prado
- Department of Biological Sciences, Federal University of São Paulo, Diadema, Brazil.,Department of Biosciences, Federal University of São Paulo (UNIFESP), Santos, Brazil
| | - Edna A Leick
- Department of Medicine, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil
| | - Milton A Martins
- Department of Medicine, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil
| | - Iolanda F L C Tibério
- Department of Medicine, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
33
|
Systemic concentrations of asymmetric dimethylarginine (ADMA) in chronic obstructive pulmonary disease (COPD): state of the art. Amino Acids 2018; 50:1169-1176. [PMID: 29951704 DOI: 10.1007/s00726-018-2606-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/20/2018] [Indexed: 01/08/2023]
Abstract
Experimental evidence suggests that oxidative stress (OS) may increase the activity of arginine methylating enzymes that produce the endogenous nitric oxide synthase inhibitor asymmetric dimethylarginine (ADMA). In addition, it is well documented that OS can significantly decrease the synthesis and/or activity of ADMA degrading enzymes, thus causing ADMA accumulation in biological fluids. Recent reports have focused on circulating methylated arginine concentrations in chronic obstructive pulmonary disease, a disease characterized by a significant increase in OS. This review discusses the results of these studies and the opportunities for further research in this area.
Collapse
|
34
|
López V, Moraga FA, Llanos AJ, Ebensperger G, Taborda MI, Uribe E. Plasmatic Concentrations of ADMA and Homocystein in Llama ( Lama glama) and Regulation of Arginase Type II: An Animal Resistent to the Development of Pulmonary Hypertension Induced by Hypoxia. Front Physiol 2018; 9:606. [PMID: 29896110 PMCID: PMC5986928 DOI: 10.3389/fphys.2018.00606] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 05/04/2018] [Indexed: 12/21/2022] Open
Abstract
There are animal species that have adapted to life at high altitude and hypobaric hypoxia conditions in the Andean highlands. One such species is the llama (Lama glama), which seem to have developed efficient protective mechanisms to avoid maladaptation resulting from chronic hypoxia, such as a resistance to the development of hypoxia -induced pulmonary hypertension. On the other hand, it is widely known that different models of hypertension can arise as a result of changes in endothelial function. The respect, one of the common causes of deregulation in endothelial vasodilator function have been associated with down-regulation of the NO synthesis and an increase in plasma levels of asymmetric dimethylarginine (ADMA) and homocysteine. Additionally, it is also known that NO production can be regulated by plasma levels of L-arginine as a result of the competition between nitric oxide synthase (NOS) and arginase. The objective of this study, was to determine the baseline concentrations of ADMA and homocysteine in llama, and to evaluate their effect on the arginase pathway and their involvement in the resistance to the development of altitude-induced pulmonary hypertension. METHOD Lowland and highland newborn sheep and llama were investigated near sea level and at high altitude. Blood determinations of arterial blood gases, ADMA and homocysteíne are made and the effect of these on the arginase activity was evaluated. RESULTS The basal concentrations of ADMA and homocysteine were determined in llama, and they were found to be significantly lower than those found in other species and in addition, the exposure to hypoxia is unable to increase its concentration. On the other hand, it was observed that the llama exhibited 10 times less arginase II activity as compared to sheep, and the expression was not induced by hypoxia. Finally, ADMA y Hcy, has no effect on the type II arginase pathway. CONCLUSION Based on our results, we propose that low concentrations of ADMA and homocysteine found in llamas, the low expression of arginase type II, DDAH-2 and CBS, as well as its insensitivity to activation by homocysteine could constitute an adaptation mechanism of these animals to the hypoxia.
Collapse
Affiliation(s)
- Vasthi López
- Laboratorio de Metabolismo de Aminoácidos e Hipoxia, Departamento de Ciencias Biomédicas, Universidad Católica del Norte, Coquimbo, Chile
| | - Fernando A Moraga
- Laboratorio de Fisiología, Hipoxia y Función Vascular, Departamento de Ciencias Biomedicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Anibal J Llanos
- Laboratorio de Fisiología y Fisiopatología del Desarrollo, Departamento de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - German Ebensperger
- Laboratorio de Fisiología y Fisiopatología del Desarrollo, Departamento de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - María I Taborda
- Laboratorio de Metabolismo de Aminoácidos e Hipoxia, Departamento de Ciencias Biomédicas, Universidad Católica del Norte, Coquimbo, Chile
| | - Elena Uribe
- Laboratorio de Enzimología, Departamento de Bioquímica y Biología Molecular, Universidad of Concepción, Concepción, Chile
| |
Collapse
|
35
|
Zheng HK, Zhao JH, Yan Y, Lian TY, Ye J, Wang XJ, Wang Z, Jing ZC, He YY, Yang P. Metabolic reprogramming of the urea cycle pathway in experimental pulmonary arterial hypertension rats induced by monocrotaline. Respir Res 2018; 19:94. [PMID: 29751839 PMCID: PMC5948901 DOI: 10.1186/s12931-018-0800-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/01/2018] [Indexed: 12/27/2022] Open
Abstract
Background Pulmonary arterial hypertension (PAH) is a rare systemic disorder associated with considerable metabolic dysfunction. Although enormous metabolomic studies on PAH have been emerging, research remains lacking on metabolic reprogramming in experimental PAH models. We aim to evaluate the metabolic changes in PAH and provide new insight into endogenous metabolic disorders of PAH. Method A single subcutaneous injection of monocrotaline (MCT) (60 mg kg− 1) was used for rats to establish PAH model. Hemodynamics and right ventricular hypertrophy were adopted to evaluate the successful establishment of PAH model. Plasma samples were assessed through targeted metabolomic profiling platform to quantify 126 endogenous metabolites. Orthogonal partial least squares discriminant analysis (OPLS-DA) was used to discriminate between MCT-treated model and control groups. Metabolite Set Enrichment Analysis was adapted to exploit the most disturbed metabolic pathways. Results Endogenous metabolites of MCT treated PAH model and control group were well profiled using this platform. A total of 13 plasma metabolites were significantly altered between the two groups. Metabolite Set Enrichment Analysis highlighted that a disruption in the urea cycle pathway may contribute to PAH onset. Moreover, five novel potential biomarkers in the urea cycle, adenosine monophosphate, urea, 4-hydroxy-proline, ornithine, N-acetylornithine, and two candidate biomarkers, namely, O-acetylcarnitine and betaine, were found to be highly correlated with PAH. Conclusion The present study suggests a new role of urea cycle disruption in the pathogenesis of PAH. We also found five urea cycle related biomarkers and another two candidate biomarkers to facilitate early diagnosis of PAH in metabolomic profile.
Collapse
Affiliation(s)
- Hai-Kuo Zheng
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jun-Han Zhao
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, and Key Laboratory of Pulmonary Vascular Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yi Yan
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, and Key Laboratory of Pulmonary Vascular Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Tian-Yu Lian
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, and Key Laboratory of Pulmonary Vascular Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jue Ye
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, and Key Laboratory of Pulmonary Vascular Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao-Jian Wang
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, and Key Laboratory of Pulmonary Vascular Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhe Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhi-Cheng Jing
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, and Key Laboratory of Pulmonary Vascular Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yang-Yang He
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, and Key Laboratory of Pulmonary Vascular Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
36
|
Zurfluh S, Baumgartner T, Meier MA, Ottiger M, Voegeli A, Bernasconi L, Neyer P, Mueller B, Schuetz P. The role of metabolomic markers for patients with infectious diseases: implications for risk stratification and therapeutic modulation. Expert Rev Anti Infect Ther 2018; 16:133-142. [PMID: 29316826 DOI: 10.1080/14787210.2018.1426460] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Metabolomics is a rapidly growing area of research. Metabolomic markers can provide information about the interaction of different organ systems, and thereby improve the understanding of physio-pathological processes, disease risk, prognosis and therapy responsiveness in a variety of diseases. Areas covered: In this narrative review of recent clinical studies investigating metabolomic markers in adult patients presenting with acute infectious disease, we mainly focused on patients with sepsis and lower respiratory tract infections. Currently, there is a growing body of literature showing that single metabolites from distinct metabolic pathways, as well as more complex metabolomic signatures are associated with disease severity and outcome in patients with systemic infections. These pathways include, among others, metabolomic markers of oxidative stress, steroid hormone and amino acid pathways, and nutritional markers. Expert commentary: Metabolic profiling has great potential to optimize patient management, to provide new targets for individual therapy and thereby improve survival of patients. At this stage, research mainly focused on the identification of new predictive signatures and less on metabolic determinants to predict treatment response. The transition from observational studies to implementation of novel markers into clinical practice is the next crucial step to prove the usefulness of metabolomic markers in patient care.
Collapse
Affiliation(s)
- Seline Zurfluh
- a University Department of Medicine, Kantonsspital Aarau and Faculty of Medicine , University of Basel , Aarau , Switzerland
| | - Thomas Baumgartner
- a University Department of Medicine, Kantonsspital Aarau and Faculty of Medicine , University of Basel , Aarau , Switzerland
| | - Marc A Meier
- a University Department of Medicine, Kantonsspital Aarau and Faculty of Medicine , University of Basel , Aarau , Switzerland
| | - Manuel Ottiger
- a University Department of Medicine, Kantonsspital Aarau and Faculty of Medicine , University of Basel , Aarau , Switzerland
| | - Alaadin Voegeli
- a University Department of Medicine, Kantonsspital Aarau and Faculty of Medicine , University of Basel , Aarau , Switzerland
| | - Luca Bernasconi
- b Department of Laboratory Medicine, University Department of Medicine , Kantonsspital Aarau , Aarau , Switzerland
| | - Peter Neyer
- b Department of Laboratory Medicine, University Department of Medicine , Kantonsspital Aarau , Aarau , Switzerland
| | - Beat Mueller
- a University Department of Medicine, Kantonsspital Aarau and Faculty of Medicine , University of Basel , Aarau , Switzerland
| | - Philipp Schuetz
- a University Department of Medicine, Kantonsspital Aarau and Faculty of Medicine , University of Basel , Aarau , Switzerland
| |
Collapse
|
37
|
Dubey A, Izakelian L, Ayaub EA, Ho L, Stephenson K, Wong S, Kwofie K, Austin RC, Botelho F, Ask K, Richards CD. Separate roles of IL-6 and oncostatin M in mouse macrophage polarization in vitro and in vivo. Immunol Cell Biol 2017; 96:257-272. [PMID: 29363180 DOI: 10.1111/imcb.1035] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 11/08/2017] [Accepted: 11/20/2017] [Indexed: 12/21/2022]
Abstract
Arginase-1 (Arg-1)-expressing M2-like macrophages are associated with Th2-skewed immune responses, allergic airway pathology, ectopic B16 melanoma cancer growth in murine models, and can be induced by Oncostatin M (OSM) transient overexpression in vivo. Here, we compare OSM to the gp130-cytokine IL-6 in mediating macrophage polarization, and find that IL-6 overexpression alone (Ad vector, AdIL-6) did not induce Arg-1 protein in mouse lungs at day 7, nor ectopic melanoma tumor growth at day 14, in contrast to overexpression of OSM (AdOSM). AdOSM elevated levels of IL-4, IL-5 and IL-13 in bronchoalveolar lavage fluid, whereas AdIL-6 did not. Bone marrow-derived macrophages respond with Arg-1 enzymatic activity to M2 stimuli (IL-4/IL-13), which was further elevated in combination with IL-6 stimulation; however, OSM or LIF had no detectable activity in vitro. Arg-1 mRNA expression induced by AdOSM was attenuated in IL-6-/- and STAT6-/- mice, suggesting requirements for both IL-6 and IL-4/IL-13 signaling in vivo. Ectopic B16 tumor burden was also reduced in IL-6-/- mice. Thus, OSM induces Arg-1+ macrophage accumulation indirectly through elevation of Th2 cytokines and IL-6 in vivo, whereas IL-6 acts directly on macrophages but requires a Th2 microenvironment, demonstrating distinct roles for OSM and IL-6 in M2 macrophage polarization.
Collapse
Affiliation(s)
- Anisha Dubey
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Laura Izakelian
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ehab A Ayaub
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Lilian Ho
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Kyle Stephenson
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Steven Wong
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Karen Kwofie
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Richard C Austin
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.,St. Joseph's Healthcare, McMaster University, Hamilton, Ontario, Canada
| | - Fernando Botelho
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Kjetil Ask
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,St. Joseph's Healthcare, McMaster University, Hamilton, Ontario, Canada
| | - Carl D Richards
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
38
|
Leem JH, Lee JH. Humidifier disinfectant-associated specific diseases should be called together as "humidifier disinfectant syndrome". ENVIRONMENTAL HEALTH AND TOXICOLOGY 2017; 32:e2017017. [PMID: 29026061 PMCID: PMC5810429 DOI: 10.5620/eht.e2017017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/07/2017] [Indexed: 06/07/2023]
Abstract
Humidifier disinfectant (HD) damage was terrible chemical damage caused by household goods that happened in only South Korea, but still very little is known in HD damage. Up to now, previous research tried to focus on interstitial fibrosis on terminal bronchioles and alveoli because it is a specific finding, compared with other diseases. To figure out whole effects from HDs, much epidemiologic and toxicologic research is underway. HDs were shown to give rise to typical toxicologic effects on various target organs, such as skin, conjunctiva, naval mucosa, bronchial mucosa, alveoli and so on, which shared common toxicological responses. On a specific target, specific toxicologic effects existed. Diverse diseases along exposure pathways can occur at the same time with a common toxicologic mechanism and cause of HDs, which can be called as HD syndrome. To gain stronger scientific evidence about it, further epidemiological and toxicological studies should be applied.
Collapse
Affiliation(s)
- Jong Han Leem
- Department of Occupational and Environmental Medicine, Inha University School of Medicine, Incheon, Korea
| | - Jong-Hyeon Lee
- Environmental Health Research and Consulting, Seoul, Korea
| |
Collapse
|
39
|
Karatzas E, Bourdakou MM, Kolios G, Spyrou GM. Drug repurposing in idiopathic pulmonary fibrosis filtered by a bioinformatics-derived composite score. Sci Rep 2017; 7:12569. [PMID: 28974751 PMCID: PMC5626774 DOI: 10.1038/s41598-017-12849-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 09/14/2017] [Indexed: 12/19/2022] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a rare disease of the respiratory system in which the lungs stiffen and get scarred, resulting in breathing weakness and eventually leading to death. Drug repurposing is a process that provides evidence for existing drugs that may also be effective in different diseases. In this study, we present a computational pipeline having as input a number of gene expression datasets from early and advanced stages of IPF and as output lists of repurposed drugs ranked with a novel composite score. We have devised and used a scoring formula in order to rank the repurposed drugs, consolidating the standard repurposing score with structural, functional and side effects' scores for each drug per stage of IPF. The whole pipeline involves the selection of proper gene expression datasets, data preprocessing and statistical analysis, selection of the most important genes related to the disease, analysis of biological pathways, investigation of related molecular mechanisms, identification of fibrosis-related microRNAs, drug repurposing, structural and literature-based analysis of the repurposed drugs.
Collapse
Affiliation(s)
- E Karatzas
- Department of Informatics and Telecommunications, University of Athens, 15784, Ilissia Athens, Greece
| | - M M Bourdakou
- Center of Systems Biology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou 4, 115 27, Athens, Greece
- Bioinformatics ERA Chair, The Cyprus Institute of Neurology and Genetics, 6 International Airport Avenue, Nicosia, 2370, Cyprus
| | - G Kolios
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - G M Spyrou
- Bioinformatics ERA Chair, The Cyprus Institute of Neurology and Genetics, 6 International Airport Avenue, Nicosia, 2370, Cyprus.
| |
Collapse
|
40
|
Wei SJ, Cheng L, Liang ES, Wang Q, Zhou SN, Xu H, Hui LH, Ge ZM, Zhang MX. Poly(ADP-ribose) polymerase 1 deficiency increases nitric oxide production and attenuates aortic atherogenesis through downregulation of arginase II. Clin Exp Pharmacol Physiol 2017; 44:114-122. [PMID: 27757983 DOI: 10.1111/1440-1681.12685] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 09/22/2016] [Accepted: 10/14/2016] [Indexed: 12/21/2022]
Abstract
Poly (ADP-ribose) polymerase (PARP) plays an important role in endothelial dysfunction, leading to atherogenesis and vascular-related diseases. However, whether PARP regulates nitric oxide (NO), a key regulator of endothelial function, is unclear so far. We investigated whether inhibition of PARP-1, the most abundant PARP isoform, prevents atherogenesis by regulating NO production and tried to elucidate the possible mechanisms involved in this phenomenon. In apolipoprotein E-deficient (apoE-/- ) mice fed a high-cholesterol diet for 12 weeks, PARP-1 inhibition via treatment with 3,4-dihydro-54-(1-piperindinyl) butoxy-1(2H)-isoquinoline (DPQ) or PARP-1 gene knockout reduced aortic atherosclerotic plaque areas (49% and 46%, respectively). Both the groups showed restored NO production in mouse aortas with reduced arginase II (Arg II) expression compared to that in the controls. In mouse peritoneal macrophages and aortic endothelial cells (MAECs), PARP-1 knockout resulted in lowered Arg II expression. Moreover, phosphorylation of endothelial NO synthase (eNOS) was preserved in the aortas and MAECs when PARP-1 was inhibited. Reduced NO production in vitro due to PARP-1 deficiency could be restored by treating the MAECs with oxidized low-density lipoprotein treatment, but this effect could not be achieved with peritoneal macrophages, which was likely due to a reduction in the expression of induced NOS expression. Our findings indicate that PARP-1 inhibition may attenuate atherogenesis by restoring NO production in endothelial cells and thus by reducing Arg II expression and consequently arginase the activity.
Collapse
Affiliation(s)
- Shu-Jian Wei
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Department of Emergency, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lin Cheng
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Er-Shun Liang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qi Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Sheng-Nan Zhou
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hao Xu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Long-Hua Hui
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China.,The First Sanatorium of Jinan Military Region, Qingdao, Shandong, China
| | - Zhi-Ming Ge
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ming-Xiang Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
41
|
Asymmetric Dimethylarginine Predicts Long-Term Outcome in Patients with Acute Exacerbation of Chronic Obstructive Pulmonary Disease. Lung 2017; 195:717-727. [DOI: 10.1007/s00408-017-0047-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/21/2017] [Indexed: 12/19/2022]
|
42
|
Shilo NR, Morris CR. Pathways to pulmonary hypertension in sickle cell disease: the search for prevention and early intervention. Expert Rev Hematol 2017; 10:875-890. [PMID: 28817980 DOI: 10.1080/17474086.2017.1364989] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Pulmonary hypertension (PH) develops in a significant number of patients with sickle cell disease (SCD), resulting in increased morbidity and mortality. This review focuses on PH pathophysiology, risk stratification, and new recommendations for screening and treatment for patients with SCD. Areas covered: An extensive PubMed literature search was performed. While the pathophysiology of PH in SCD is yet to be fully deciphered, it is known that the etiology is multifactorial; hemolysis, hypercoagulability, hypoxemia, ischemic-reperfusion injury, oxidative stress, and genetic susceptibility all contribute in varying degrees to endothelial dysfunction. Hemolysis, in particular, seems to play a key role by inciting an imbalance in the regulatory axis of nitric oxide and arginine metabolism. Systematic risk stratification starting in childhood based on clinical features and biomarkers that enable early detection is necessary. Multi-faceted, targeted interventions, before irreversible vasculopathy develops, will allow for improved patient outcomes and life expectancy. Expert commentary: Despite progress in our understanding of PH in SCD, clinically proven therapies remain elusive and additional controlled clinical trials are needed. Prevention of disease starts in childhood, a critical window for intervention. Given the complex and multifactorial nature of SCD, patients will ultimately benefit from combination therapies that simultaneously targets multiple mechanisms.
Collapse
Affiliation(s)
- Natalie R Shilo
- a Department of Pediatrics, Division of Pulmonary Medicine , University of Connecticut Heath Center , Farmington , CT , USA
| | - Claudia R Morris
- b Department of Pediatrics, Division of Pediatric Emergency Medicine, Emory-Children's Center for Cystic Fibrosis and Airways Disease Research , Emory University School of Medicine , Atlanta , GA , USA
| |
Collapse
|
43
|
Fu LQ, Li YL, Fu AK, Wu YP, Wang YY, Hu SL, Li WF. Pidotimod exacerbates allergic pulmonary infection in an OVA mouse model of asthma. Mol Med Rep 2017; 16:4151-4158. [PMID: 28731127 DOI: 10.3892/mmr.2017.7046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 05/16/2017] [Indexed: 11/05/2022] Open
Abstract
Pidotimod is a synthetic dipeptide with biological and immuno‑modulatory properties. It has been widely used for treatment and prevention of recurrent respiratory infections. However, its impact on the regulation of allergic pulmonary inflammation is still not clear. In the current study, an ovalbumin (OVA)‑induced allergic asthma model was used to investigate the immune‑modulating effects of pidotimod on airway eosinophilia, mucus metaplasia and inflammatory factor expression compared with dexamethasone (positive control). The authors determined that treatment with pidotimod exacerbated pulmonary inflammation as demonstrated by significantly increased eosinophil infiltration, dramatically elevated immunoglobulin E production, and enhanced T helper 2 response. Moreover, treatment failed to attenuate mucus production in lung tissue, and did not reduce OVA‑induced high levels of FIZZ1 and Arg1 expression in asthmatic mice. In contrast, administration of dexamethasone was efficient in alleviating allergic airway inflammation in OVA‑induced asthmatic mice. These data indicated that pidotimod as an immunotherapeutic agent should be used cautiously and the effectiveness for controlling allergic asthma needs further evaluation and research.
Collapse
Affiliation(s)
- Luo-Qin Fu
- Key Laboratory of Molecular Animal Nutrition and Feed Sciences, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Ya-Li Li
- Key Laboratory of Molecular Animal Nutrition and Feed Sciences, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Ai-Kun Fu
- Key Laboratory of Molecular Animal Nutrition and Feed Sciences, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Yan-Ping Wu
- Key Laboratory of Molecular Animal Nutrition and Feed Sciences, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Yuan-Yuan Wang
- Key Laboratory of Molecular Animal Nutrition and Feed Sciences, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Sheng-Lan Hu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong 510640, P.R. China
| | - Wei-Fen Li
- Key Laboratory of Molecular Animal Nutrition and Feed Sciences, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| |
Collapse
|
44
|
Morris CR, Hamilton-Reeves J, Martindale RG, Sarav M, Ochoa Gautier JB. Acquired Amino Acid Deficiencies: A Focus on Arginine and Glutamine. Nutr Clin Pract 2017; 32:30S-47S. [PMID: 28388380 DOI: 10.1177/0884533617691250] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Nonessential amino acids are synthesized de novo and therefore not diet dependent. In contrast, essential amino acids must be obtained through nutrition since they cannot be synthesized internally. Several nonessential amino acids may become essential under conditions of stress and catabolic states when the capacity of endogenous amino acid synthesis is exceeded. Arginine and glutamine are 2 such conditionally essential amino acids and are the focus of this review. Low arginine bioavailability plays a pivotal role in the pathogenesis of a growing number of varied diseases, including sickle cell disease, thalassemia, malaria, acute asthma, cystic fibrosis, pulmonary hypertension, cardiovascular disease, certain cancers, and trauma, among others. Catabolism of arginine by arginase enzymes is the most common cause of an acquired arginine deficiency syndrome, frequently contributing to endothelial dysfunction and/or T-cell dysfunction, depending on the clinical scenario and disease state. Glutamine, an arginine precursor, is one of the most abundant amino acids in the body and, like arginine, becomes deficient in several conditions of stress, including critical illness, trauma, infection, cancer, and gastrointestinal disorders. At-risk populations are discussed together with therapeutic options that target these specific acquired amino acid deficiencies.
Collapse
Affiliation(s)
- Claudia R Morris
- 1 Department of Pediatrics, Division of Pediatric Emergency Medicine, Emory-Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jill Hamilton-Reeves
- 2 Department of Dietetics and Nutrition, University of Kansas, Kansas City, Kansas, USA
| | - Robert G Martindale
- 3 Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA
| | - Menaka Sarav
- 4 Department of Medicine, Division of Nephrology, Northshore University Health System, University of Chicago, Chicago, Illinois, USA
| | | |
Collapse
|
45
|
Vögeli A, Ottiger M, Meier MA, Steuer C, Bernasconi L, Kulkarni P, Huber A, Christ-Crain M, Henzen C, Hoess C, Thomann R, Zimmerli W, Mueller B, Schuetz P. Admission levels of asymmetric and symmetric dimethylarginine predict long-term outcome in patients with community-acquired pneumonia. Respir Res 2017; 18:25. [PMID: 28114935 PMCID: PMC5259979 DOI: 10.1186/s12931-017-0502-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 12/28/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND During infection, there is an activation of the L-arginine-nitric-oxide pathway, with a shift from nitric oxide synthesis to a degradation of L-arginine to its metabolites, asymmetric and symmetric dimethylarginine (ADMA and SDMA). However, the prognostic implications for short-term or long-term survival remains unclear. We investigated the association of L-arginine, ADMA, and SDMA with adverse clinical outcomes in a well-defined cohort of patients with community-acquired pneumonia (CAP). METHODS We measured L-arginine, ADMA, and SDMA in 268 CAP patients from a Swiss multicenter trial by mass spectrometry and used Cox regression models to investigate associations between blood marker levels and disease severity as well as mortality over a period of 6 years. RESULTS Six-year mortality was 44.8%. Admission levels of ADMA and SDMA (μmol/L) were correlated with CAP severity as assessed by the pneumonia severity index (r = 0.32, p < 0.001 and r = 0.56, p < 0.001 for ADMA and SDMA, respectively) and higher in 6-year non-survivors versus survivors (median 0.62 vs. 0.48; p < 0.001 and 1.01 vs. 0.85; p < 0.001 for ADMA and SDMA, respectively). Both ADMA and SDMA were significantly associated with long-term mortality (hazard ratios [HR] 4.44 [95% confidence intervals (CI) 1.84 to 10.74] and 2.81 [95% CI 1.45 to 5.48], respectively). The effects were no longer significant after multivariate adjustment for age and comorbidities. No association of L-arginine with severity and outcome was found. CONCLUSIONS Both ADMA and SDMA show a severity-dependent increase in patients with CAP and are strongly associated with mortality. This association is mainly explained by age and comorbidities. TRIAL REGISTRATION ISRCTN95122877 . Registered 31 July 2006.
Collapse
Affiliation(s)
- Alaadin Vögeli
- Kantonsspital Aarau, University Department of Internal Medicine, Tellstrasse, CH-5001 Aarau, Switzerland
| | - Manuel Ottiger
- Kantonsspital Aarau, University Department of Internal Medicine, Tellstrasse, CH-5001 Aarau, Switzerland
| | - Marc A. Meier
- Kantonsspital Aarau, University Department of Internal Medicine, Tellstrasse, CH-5001 Aarau, Switzerland
| | - Christian Steuer
- Institute of Laboratory Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | | | - Andreas Huber
- Institute of Laboratory Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Mirjam Christ-Crain
- Endocrinology, Diabetology, and Metabolism, University Hospital Basel, Basel, Switzerland
| | - Christoph Henzen
- Department of Internal Medicine, Kantonsspital Luzern, Luzern, Switzerland
| | - Claus Hoess
- Department of Internal Medicine, Kantonsspital Münsterlingen, Münsterlingen, Switzerland
| | - Robert Thomann
- Department of Internal Medicine, Bürgerspital Solothurn, Solothurn, Switzerland
| | - Werner Zimmerli
- Kantonsspital Baselland, University Department of Internal Medicine, Liestal, Switzerland
| | - Beat Mueller
- Kantonsspital Aarau, University Department of Internal Medicine, Tellstrasse, CH-5001 Aarau, Switzerland
| | - Philipp Schuetz
- Kantonsspital Aarau, University Department of Internal Medicine, Tellstrasse, CH-5001 Aarau, Switzerland
| |
Collapse
|
46
|
Stasyuk N, Gayda G, Yepremyan H, Stepien A, Gonchar M. Fluorometric enzymatic assay of l-arginine. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2017; 170:184-190. [PMID: 27450117 DOI: 10.1016/j.saa.2016.07.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 07/07/2016] [Accepted: 07/09/2016] [Indexed: 06/06/2023]
Abstract
The enzymes of l-arginine (further - Arg) metabolism are promising tools for elaboration of selective methods for quantitative Arg analysis. In our study we propose an enzymatic method for Arg assay based on fluorometric monitoring of ammonia, a final product of Arg splitting by human liver arginase I (further - arginase), isolated from the recombinant yeast strain, and commercial urease. The selective analysis of ammonia (at 415nm under excitation at 360nm) is based on reaction with o-phthalaldehyde (OPA) in the presence of sulfite in alkali medium: these conditions permit to avoid the reaction of OPA with any amino acid. A linearity range of the fluorometric arginase-urease-OPA method is from 100nM to 6μМ with a limit of detection of 34nM Arg. The method was used for the quantitative determination of Arg in the pooled sample of blood serum. The obtained results proved to be in a good correlation with the reference enzymatic method and literature data. The proposed arginase-urease-OPA method being sensitive, economical, selective and suitable for both routine and micro-volume formats, can be used in clinical diagnostics for the simultaneous determination of Arg as well as urea and ammonia in serum samples.
Collapse
Affiliation(s)
- Nataliya Stasyuk
- Institute of Cell Biology, National Academy of Science of Ukraine, Drahomanov Str. 14/16, 79005 Lviv, Ukraine
| | - Galina Gayda
- Institute of Cell Biology, National Academy of Science of Ukraine, Drahomanov Str. 14/16, 79005 Lviv, Ukraine.
| | - Hasmik Yepremyan
- Scientific and Production Center "Armbiotechnology", National Academy of Sciences of Armenia, Armenia
| | - Agnieszka Stepien
- Department of Dietetics, Institute of Nursing and Health Sciences, Medical Faculty, University of Rzeszow, Al. mjr.W.Kopisto 2 a, 35-310 Rzeszów, Poland; Centre for Innovative Research in Medical and Natural Sciences, Medical Faculty of the University of Rzeszow, Warzywna 1A, 35-310 Rzeszów, Poland
| | - Mykhailo Gonchar
- Institute of Cell Biology, National Academy of Science of Ukraine, Drahomanov Str. 14/16, 79005 Lviv, Ukraine
| |
Collapse
|
47
|
Kim MS, Jeong SW, Choi SJ, Han JY, Kim SH, Yoon S, Oh JH, Lee K. Analysis of genomic responses in a rat lung model treated with a humidifier sterilizer containing polyhexamethyleneguanidine phosphate. Toxicol Lett 2016; 268:36-43. [PMID: 27989595 DOI: 10.1016/j.toxlet.2016.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/10/2016] [Accepted: 11/13/2016] [Indexed: 12/24/2022]
Abstract
The antimicrobial biocide polyhexamethyleneguanidine (PHMG) phosphate is the main ingredient in the commercially available humidifier disinfectant. PHMG phosphate-based humidifier disinfectants can cause pulmonary fibrosis and induce inflammatory and fibrotic responses both in vivo and in vitro. However, toxicological mechanisms including genomic alterations induced by inhalation exposure to PHMG phosphate have not been elucidated. Therefore, this study evaluated the toxicological effects of the PHMG phosphate-containing humidifier disinfectant. We used DNA microarray to identify global gene expression changes in rats treated with PHMG phosphate-containing humidifier disinfectant for 4 weeks and 10 weeks. Functional significance of differentially expressed genes (DEGs) was estimated by gene ontology (GO) analysis. Four weeks post-exposure, 320 and 392 DEGs were identified in female and male rats, respectively (>2-fold, p<0.05). Ten weeks post-exposure, 1290 and 995 DEGs were identified in females and males, respectively. Of these, 119 and 556 genes overlapped between females and males at 4 weeks and 10 weeks, respectively, post-PHMG phosphate exposure. In addition, 21 genes were upregulated and 4 genes were downregulated in response to PHMG phosphate in a time-dependent manner. Thus, we predict that changes in genomic responses could be a significant molecular mechanism underlying PHMG phosphate toxicity. Further studies are required to determine the detailed mechanism of PHMG phosphate-induced pulmonary toxicity.
Collapse
Affiliation(s)
- Min-Seok Kim
- Department of Inhalation Toxicology Research, Korea Institute of Toxicology, Jeonbuk 56212, Republic of Korea
| | - Seok Won Jeong
- Department of Research and Development, Genoplan Inc., Seoul 06221, Republic of Korea
| | - Seong-Jin Choi
- Department of Inhalation Toxicology Research, Korea Institute of Toxicology, Jeonbuk 56212, Republic of Korea
| | - Jin-Young Han
- Department of Inhalation Toxicology Research, Korea Institute of Toxicology, Jeonbuk 56212, Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, 34113, Republic of Korea
| | - Sung-Hwan Kim
- Department of Inhalation Toxicology Research, Korea Institute of Toxicology, Jeonbuk 56212, Republic of Korea
| | - Seokjoo Yoon
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, 34113, Republic of Korea
| | - Jung-Hwa Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, 34113, Republic of Korea
| | - Kyuhong Lee
- Department of Inhalation Toxicology Research, Korea Institute of Toxicology, Jeonbuk 56212, Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
48
|
Saad-Hussein A, Thabet EH, Taha MM, Shahy EM, Mahdy-Abdallah H. Association of ADAM33 gene polymorphism and arginase activity with susceptibility to ventilatory impairment in wood dust-exposed workers. Hum Exp Toxicol 2016; 35:966-73. [DOI: 10.1177/0960327115611971] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
ADAM33 represents an important gene of susceptibility for lung function impairment. This work aimed to evaluate the association between genetic polymorphism of ADAM33 at four single nucleotide polymorphisms (T1, T2, S1, and Q1) and arginase activity with respiratory functions impairment in wood workers. The study was done to compare ventilatory functions and arginase activity of 82 wood workers and 81 controls. Genotyping was determined by using the polymerase chain restriction fragment length polymorphism method. Forced expiratory volume in the first second (FEV1), forced vital capacity (FVC), and peak expiratory flow rate (PEF) of the workers were significantly reduced compared with the controls. T1 single nucleotide polymorphism (SNP) was associated with obvious decline in the FEV1, FVC, and PEF in wood workers, while T2 SNP was associated with decline in FEV1 and PEF. A significant increase in arginase activity was found in T2 and S1 SNPs of the exposed workers. Increase in duration of exposure was correlated with the decline in ventilatory functions. This inverse correlation was significant for pulmonary function indices in AA and GG genotypes of T1 and T2, respectively. Moreover, significance was detected for FVC and FEV1 in AA and GA genotypes of S1 and Q1. A positive correlation between arginase activity and duration of exposure was found to be significant in GG genotype of S1 SNP. An association between ADAM33 gene polymorphism and impaired lung functions was detected in wood dust-exposed workers. Arginase activity may play an associated important role in increasing this impairment in wood workers.
Collapse
Affiliation(s)
- A Saad-Hussein
- Environmental Research Division, Department of Environmental and Occupational Medicine, National Research Centre, Dokki, Giza, Egypt
| | - EH Thabet
- Clinical Pathology, National Research Centre, Dokki, Giza, Egypt
| | - MM Taha
- Clinical Pathology, National Research Centre, Dokki, Giza, Egypt
| | - EM Shahy
- Environmental Research Division, Department of Environmental and Occupational Medicine, National Research Centre, Dokki, Giza, Egypt
| | - H Mahdy-Abdallah
- Environmental Research Division, Department of Environmental and Occupational Medicine, National Research Centre, Dokki, Giza, Egypt
| |
Collapse
|
49
|
Jonker R, Deutz NE, Erbland ML, Anderson PJ, Engelen MP. Alterations in whole-body arginine metabolism in chronic obstructive pulmonary disease. Am J Clin Nutr 2016; 103:1458-64. [PMID: 27146652 PMCID: PMC4880996 DOI: 10.3945/ajcn.115.125187] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 04/01/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a condition characterized by systemic low-grade inflammation that could increase the production of nitric oxide (NO), of which arginine is the sole precursor. Arginine is derived from the breakdown of protein and through the conversion of citrulline to arginine (de novo arginine production). OBJECTIVE Our objective was to study whole-body arginine and citrulline and related metabolism in stable COPD patients. DESIGN With the use of stable isotope methodology, we studied whole-body arginine and citrulline rates of appearance, de novo arginine (citrulline-to-arginine flux) and NO (arginine-to-citrulline flux) production, protein synthesis and breakdown rates, and plasma amino acid concentrations in a heterogeneous group of patients with moderate-to-severe COPD [n = 23, mean ± SE age: 65 ± 2 y, forced expiratory volume in 1 s (FEV1): 40% ± 2% of predicted], and a group of healthy older adults (n = 19, mean ± SE age: 64 ± 2 y, FEV1: 95% ± 4% of predicted). RESULTS Although plasma arginine and citrulline concentrations were comparable between COPD patients and controls, whole-body arginine (P = 0.015) and citrulline (P = 0.026) rates of appearance were higher in COPD patients and related to a 57% greater de novo arginine production (P < 0.0001). Despite a higher whole-body arginine clearance in COPD patients (P < 0.0001), we found no difference in NO production. CONCLUSION In stable patients with moderate-to-severe COPD, endogenous arginine production is upregulated to support a higher arginine utilization that is unrelated to whole-body NO production. This trial was registered at clinicaltrials.gov as NCT01173354 and NCT01172314.
Collapse
Affiliation(s)
- Renate Jonker
- Center for Translational Research in Aging and Longevity, Department of Health and Kinesiology, Texas A&M University, College Station, TX; and Departments of Geriatrics and
| | - Nicolaas Ep Deutz
- Center for Translational Research in Aging and Longevity, Department of Health and Kinesiology, Texas A&M University, College Station, TX; and Departments of Geriatrics and
| | - Marcia L Erbland
- Pulmonary and Critical Care Medicine, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Paula J Anderson
- Pulmonary and Critical Care Medicine, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Mariëlle Pkj Engelen
- Center for Translational Research in Aging and Longevity, Department of Health and Kinesiology, Texas A&M University, College Station, TX; and Departments of Geriatrics and
| |
Collapse
|
50
|
Long-Term Chronic Intermittent Hypobaric Hypoxia in Rats Causes an Imbalance in the Asymmetric Dimethylarginine/Nitric Oxide Pathway and ROS Activity: A Possible Synergistic Mechanism for Altitude Pulmonary Hypertension? Pulm Med 2016; 2016:6578578. [PMID: 27313889 PMCID: PMC4904121 DOI: 10.1155/2016/6578578] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 05/08/2016] [Indexed: 12/28/2022] Open
Abstract
Chronic intermittent hypoxia (CIH) and chronic hypoxia (CH) are associated with high-altitude pulmonary hypertension (HAPH). Asymmetric dimethylarginine (ADMA), a NO synthase (NOS) inhibitor, may contribute to HAPH. This study assessed changes in the ADMA/NO pathway and the underlying mechanisms in rat lungs following exposure to CIH or CH simulated in a hypobaric chamber at 428 Torr. Twenty-four adult Wistar rats were randomly assigned to three groups: CIH2x2 (2 days of hypoxia/2 days of normoxia), CH, and NX (permanent normoxia), for 30 days. All analyses were performed in whole lung tissue. L-Arginine and ADMA were analyzed using LC-MS/MS. Under both hypoxic conditions right ventricular hypertrophy was observed (p < 0.01) and endothelial NOS mRNA increased (p < 0.001), but the phosphorylated/nonphosphorylated vasodilator-stimulated phosphoprotein (VASP) ratio was unchanged. ADMA increased (p < 0.001), whereas dimethylarginine dimethylaminohydrolase (DDAH) activity decreased only under CH (p < 0.05). Although arginase activity increased (p < 0.001) and L-arginine exhibited no changes, the L-arginine/ADMA ratio decreased significantly (p < 0.001). Moreover, NOX4 expression increased only under CH (p < 0.01), but malondialdehyde (MDA) increased (up to 2-fold) equally in CIH2x2 and CH (p < 0.001). Our results suggest that ADMA and oxidative stress likely reduce NO bioavailability under altitude hypoxia, which implies greater pulmonary vascular reactivity and tone, despite the more subdued effects observed under CIH.
Collapse
|