1
|
He Y, Liang H, Yang X, Hao F, Huang K, Wang Q. Dynamic changes of lung sRAGE in mice with chronic obstructive pulmonary disease induced by cigarette smoke exposure. PLoS One 2024; 19:e0313872. [PMID: 39591423 PMCID: PMC11594483 DOI: 10.1371/journal.pone.0313872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024] Open
Abstract
OBJECTIVE To study the changes of lung function, pathophysiology, inflammatory cytokines and related inflammatory responses in COPD mouse model, and to analyze the role of sRAGE in the pathogenesis of COPD induced by cigarette smoke (CS) exposure in mice. METHODS 24 healthy male C57BL/6J mice aged 6 to 8 weeks were randomly divided into Smoke-Exposed (SE) group and Control group. The mice in SE group were exposed to 7 time points at 3, 7, 15, 30, 60, 90 and 120 days, while mice in control group were exposed to fresh room air, with 3 mice in each group. Lung function of mice was detected at different exposure time points, and the lung tissue sections were stained with HE to observe the lung histopathological changes of mice in each group, and the lung tissue morphological quantitative analysis was performed to evaluate the degree of emphysema. The content of inflammatory cytokines including IL-1β, IL-6 and TNF-α in the supernatant of BALF was detected by ELISA to evaluate the pulmonary inflammation of mice. The expression of sRAGE in BALF supernatant was detected by ELISA. BALF cell precipitates were classified and counted under light microscope. RESULTS After 90 days of exposure to cigarette smoke, the lung function of mice was significantly reduced, emphysema appeared significantly, and the expression of inflammatory cells and inflammatory cytokines in BALF was significantly increased (all P<0.05). sRAGE increased significantly in the early stage of CS exposure (7-15 days) compared with the control group, and the number of macrophages and levels of inflammatory cytokines in BALF also increased temporarily (P<0.05). With the gradual exposure of CS, sRAGE expression gradually decreased, and was significantly reduced after COPD formation compared with the control group. CONCLUSION In the process of the occurrence and development of chronic obstructive pulmonary disease induced by cigarette smoke exposure, the level of sRAGE in bronchoalveolar lavage fluid showed a dynamic change of first increase and then decrease. The expression of sRAGE increased in the early stage of smoke exposure and played a transient pro-inflammatory role. With long-term exposure to cigarette smoke, the inflammatory response is gradually aggravated in lung, and the expression of sRAGE is significantly decreased, and its reduction degree is closely related to the degree of reduced lung function and inflammation.
Collapse
Affiliation(s)
- Yue He
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongyu Liang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaohui Yang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fengyun Hao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kai Huang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qiang Wang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Delrue C, Speeckaert R, Delanghe JR, Speeckaert MM. Breath of fresh air: Investigating the link between AGEs, sRAGE, and lung diseases. VITAMINS AND HORMONES 2024; 125:311-365. [PMID: 38997169 DOI: 10.1016/bs.vh.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Advanced glycation end products (AGEs) are compounds formed via non-enzymatic reactions between reducing sugars and amino acids or proteins. AGEs can accumulate in various tissues and organs and have been implicated in the development and progression of various diseases, including lung diseases. The receptor of advanced glycation end products (RAGE) is a receptor that can bind to advanced AGEs and induce several cellular processes such as inflammation and oxidative stress. Several studies have shown that both AGEs and RAGE play a role in the pathogenesis of lung diseases, such as chronic obstructive pulmonary disease, asthma, idiopathic pulmonary fibrosis, cystic fibrosis, and acute lung injury. Moreover, the soluble form of the receptor for advanced glycation end products (sRAGE) has demonstrated its ability to function as a decoy receptor, possessing beneficial characteristics such as anti-inflammatory, antioxidant, and anti-fibrotic properties. These qualities make it an encouraging focus for therapeutic intervention in managing pulmonary disorders. This review highlights the current understanding of the roles of AGEs and (s)RAGE in pulmonary diseases and their potential as biomarkers and therapeutic targets for preventing and treating these pathologies.
Collapse
Affiliation(s)
- Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium
| | | | - Joris R Delanghe
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Marijn M Speeckaert
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium; Research Foundation-Flanders (FWO), Brussels, Belgium.
| |
Collapse
|
3
|
Reynaert NL, Vanfleteren LEGW, Perkins TN. The AGE-RAGE Axis and the Pathophysiology of Multimorbidity in COPD. J Clin Med 2023; 12:jcm12103366. [PMID: 37240472 DOI: 10.3390/jcm12103366] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/24/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease of the airways and lungs due to an enhanced inflammatory response, commonly caused by cigarette smoking. Patients with COPD are often multimorbid, as they commonly suffer from multiple chronic (inflammatory) conditions. This intensifies the burden of individual diseases, negatively affects quality of life, and complicates disease management. COPD and comorbidities share genetic and lifestyle-related risk factors and pathobiological mechanisms, including chronic inflammation and oxidative stress. The receptor for advanced glycation end products (RAGE) is an important driver of chronic inflammation. Advanced glycation end products (AGEs) are RAGE ligands that accumulate due to aging, inflammation, oxidative stress, and carbohydrate metabolism. AGEs cause further inflammation and oxidative stress through RAGE, but also through RAGE-independent mechanisms. This review describes the complexity of RAGE signaling and the causes of AGE accumulation, followed by a comprehensive overview of alterations reported on AGEs and RAGE in COPD and in important co-morbidities. Furthermore, it describes the mechanisms by which AGEs and RAGE contribute to the pathophysiology of individual disease conditions and how they execute crosstalk between organ systems. A section on therapeutic strategies that target AGEs and RAGE and could alleviate patients from multimorbid conditions using single therapeutics concludes this review.
Collapse
Affiliation(s)
- Niki L Reynaert
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, 6229 ER Maastricht, The Netherlands
| | - Lowie E G W Vanfleteren
- COPD Center, Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Timothy N Perkins
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
4
|
Abstract
Smoking is a well-established risk factor for chronic obstructive pulmonary disease (COPD). Chronic lung inflammation continues even after smoking cessation and leads to COPD progression. To date, anti-inflammatory therapies are ineffective in improving pulmonary function and COPD symptoms, and new molecular targets are urgently needed to deal with this challenge. The receptor for advanced glycation end-products (RAGE) was shown to be relevant in COPD pathogenesis, since it is both a genetic determinant of low lung function and a determinant of COPD susceptibility. Moreover, RAGE is involved in the physiological response to cigarette smoke exposure. Since innate and acquired immunity plays an essential role in the development of chronic inflammation and emphysema in COPD, here we summarized the roles of RAGE and its ligand HMGB1 in COPD immunity.
Collapse
Affiliation(s)
- Lin Chen
- Department of Respiratory and Critical Care Medicine, Liuzhou People's Hospital, LiuZhou, Guangxi, China
| | - Xuejiao Sun
- Department of Respiratory and Critical Care Medicine, Liuzhou People's Hospital, LiuZhou, Guangxi, China
| | - Xiaoning Zhong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
5
|
Östling J, Van Geest M, Olsson HK, Dahlen SE, Viklund E, Gustafsson PM, Mirgorodskaya E, Olin AC. A novel non-invasive method allowing for discovery of pathologically relevant proteins from small airways. Clin Proteomics 2022; 19:20. [PMID: 35668386 PMCID: PMC9167914 DOI: 10.1186/s12014-022-09348-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/04/2022] [Indexed: 01/01/2023] Open
Abstract
Background There is a lack of early and precise biomarkers for personalized respiratory medicine. Breath contains an aerosol of droplet particles, which are formed from the epithelial lining fluid when the small airways close and re-open during inhalation succeeding a full expiration. These particles can be collected by impaction using the PExA method (Particles in Exhaled Air), and are derived from an area of high clinical interest previously difficult to access, making them a potential source of biomarkers reflecting pathological processes in the small airways. Research question Our aim was to investigate if PExA method is useful for discovery of biomarkers that reflect pathology of small airways. Methods and analysis Ten healthy controls and 20 subjects with asthma, of whom 10 with small airway involvement as indicated by a high lung clearance index (LCI ≥ 2.9 z-score), were examined in a cross-sectional design, using the PExA instrument. The samples were analysed with the SOMAscan proteomics platform (SomaLogic Inc.). Results Two hundred-seven proteins were detected in up to 80% of the samples. Nine proteins showed differential abundance in subjects with asthma and high LCI as compared to healthy controls. Two of these were less abundant (ALDOA4, C4), and seven more abundant (FIGF, SERPINA1, CD93, CCL18, F10, IgM, IL1RAP). sRAGE levels were lower in ex-smokers (n = 14) than in never smokers (n = 16). Gene Ontology (GO) annotation database analyses revealed that the PEx proteome is enriched in extracellular proteins associated with extracellular exosome-vesicles and innate immunity. Conclusion The applied analytical method was reproducible and allowed identification of pathologically interesting proteins in PEx samples from asthmatic subjects with high LCI. The results suggest that PEx based proteomics is a novel and promising approach to study respiratory diseases with small airway involvement. Supplementary Information The online version contains supplementary material available at 10.1186/s12014-022-09348-y. Key question Can the PExA method identify individual protein profiles that reflect pathology of small airways, using the SOMAscan platform? What is the bottom line? Two hundred-seven proteins were detected in up to 80% of the PEx samples, with a strong overrepresentation of proteins related to innate immune responses, including nine proteins that that discriminated subjects with asthma and high LCI as compared to healthy controls. Why read on The results support that PEx based proteomics is a novel and promising approach to study respiratory diseases with small airway involvement. Supplementary Information The online version contains supplementary material available at 10.1186/s12014-022-09348-y.
Collapse
Affiliation(s)
- Jörgen Östling
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden.,PExA AB, Gothenburg, Sweden
| | - Marleen Van Geest
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden.,Hansa Biopharma AB, Lund, Sweden
| | - Henric K Olsson
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Sven-Erik Dahlen
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Emilia Viklund
- Occupational and Environmental Medicine, Department of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Anna-Carin Olin
- Occupational and Environmental Medicine, Department of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
6
|
Ferraro M, Di Vincenzo S, Sangiorgi C, Leto Barone S, Gangemi S, Lanata L, Pace E. Carbocysteine Modifies Circulating miR-21, IL-8, sRAGE, and fAGEs Levels in Mild Acute Exacerbated COPD Patients: A Pilot Study. Pharmaceuticals (Basel) 2022; 15:ph15020218. [PMID: 35215330 PMCID: PMC8880736 DOI: 10.3390/ph15020218] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
Patients with Chronic Obstructive Pulmonary Disease (COPD) periodically experience acute exacerbation (AECOPD). Carbocysteine represents a valid add on therapy in COPD by exerting antioxidant and anti-inflammatory activities. The in vivo effects of carbocysteine on inflammatory markers are not yet fully understood. The aims of this study were to assess: (i) miR-21, IL-8, soluble Receptor for Advanced Glycation End Products (sRAGE), and fluorescent Advanced Glycation End Products (fAGEs) in control subjects (n = 9), stable (n = 9), and AECOPD patients (n = 24); and (ii) whether carbocysteine modifies these markers and the functional parameters in mild AECOPD patients. Mild AECOPD patients received or not carbocysteine along with background inhalation therapy for 20 days. At the onset and at the end of the observation period, the following parameters were evaluated: FEV1, FEF25–75%, CAT questionnaire; miR-21 by Real Time PCR; IL-8 and sRAGE by ELISA; and fAGEs by spectro-fluorescence method. COPD patients showed higher levels of miR-21, IL-8, fAGEs and lower levels of sRAGE compared to that of controls. miR-21 inversely correlated with FEV1. IL-8 and fAGEs were significantly different in stable and exacerbated COPD patients. Carbocysteine improved symptoms, FEV1 and FEF25–75%, increased sRAGE, and reduced miR-21, IL-8, and fAGEs in mild AECOPD patients. The present study provides compelling evidence that carbocysteine may help to manage mild AECOPD by downregulating some parameters of systemic inflammation.
Collapse
Affiliation(s)
- Maria Ferraro
- Institute for Biomedical Research and Innovation (IRIB)—Consiglio Nazionale delle Ricerche, 90146 Palermo, Italy; (M.F.); (S.D.V.); (C.S.)
- Institute of Translational Pharmacology (IFT)—National Research Council, 90146 Palermo, Italy
| | - Serena Di Vincenzo
- Institute for Biomedical Research and Innovation (IRIB)—Consiglio Nazionale delle Ricerche, 90146 Palermo, Italy; (M.F.); (S.D.V.); (C.S.)
- Institute of Translational Pharmacology (IFT)—National Research Council, 90146 Palermo, Italy
| | - Claudia Sangiorgi
- Institute for Biomedical Research and Innovation (IRIB)—Consiglio Nazionale delle Ricerche, 90146 Palermo, Italy; (M.F.); (S.D.V.); (C.S.)
| | | | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy;
| | | | - Elisabetta Pace
- Institute for Biomedical Research and Innovation (IRIB)—Consiglio Nazionale delle Ricerche, 90146 Palermo, Italy; (M.F.); (S.D.V.); (C.S.)
- Institute of Translational Pharmacology (IFT)—National Research Council, 90146 Palermo, Italy
- Correspondence: ; Tel.: +39-091-680-9148; Fax: +39-091-680-9122
| |
Collapse
|
7
|
Keefe J, Yao C, Hwang SJ, Courchesne P, Lee GY, Dupuis J, Mizgerd JP, O’Connor G, Washko GR, Cho MH, Silverman EK, Levy D. An Integrative Genomic Strategy Identifies sRAGE as a Causal and Protective Biomarker of Lung Function. Chest 2022; 161:76-84. [PMID: 34237330 PMCID: PMC8783029 DOI: 10.1016/j.chest.2021.06.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND There are few clinically useful circulating biomarkers of lung function and lung disease. We hypothesized that genome-wide association studies (GWAS) of circulating proteins in conjunction with GWAS of pulmonary traits represents a clinically relevant approach to identifying causal proteins and therapeutically useful insights into mechanisms related to lung function and disease. STUDY QUESTION Can an integrative genomic strategy using GWAS of plasma soluble receptor for advanced glycation end-products (sRAGE) levels in conjunction with GWAS of lung function traits identify putatively causal relations of sRAGE to lung function? STUDY DESIGN AND METHODS Plasma sRAGE levels were measured in 6,861 Framingham Heart Study participants and GWAS of sRAGE was conducted to identify protein quantitative trait loci (pQTL), including cis-pQTL variants at the sRAGE protein-coding gene locus (AGER). We integrated sRAGE pQTL variants with variants from GWAS of lung traits. Colocalization of sRAGE pQTL variants with lung trait GWAS variants was conducted, and Mendelian randomization was performed using sRAGE cis-pQTL variants to infer causality of sRAGE for pulmonary traits. Cross-sectional and longitudinal protein-trait association analyses were conducted for sRAGE in relation to lung traits. RESULTS Colocalization identified shared genetic signals for sRAGE with lung traits. Mendelian randomization analyses suggested protective causal relations of sRAGE to several pulmonary traits. Protein-trait association analyses demonstrated higher sRAGE levels to be cross-sectionally and longitudinally associated with preserved lung function. INTERPRETATION sRAGE is produced by type I alveolar cells, and it acts as a decoy receptor to block the inflammatory cascade. Our integrative genomics approach provides evidence for sRAGE as a causal and protective biomarker of lung function, and the pattern of associations is suggestive of a protective role of sRAGE against restrictive lung physiology. We speculate that targeting the AGER/sRAGE axis may be therapeutically beneficial for the treatment and prevention of inflammation-related lung disease.
Collapse
Affiliation(s)
- Joshua Keefe
- Framingham Heart Study, Framingham, MA,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Chen Yao
- Framingham Heart Study, Framingham, MA,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Shih-Jen Hwang
- Framingham Heart Study, Framingham, MA,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Paul Courchesne
- Framingham Heart Study, Framingham, MA,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Gha Young Lee
- Framingham Heart Study, Framingham, MA,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Joseph P. Mizgerd
- Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA
| | - George O’Connor
- Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA
| | - George R. Washko
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Michael H. Cho
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Edwin K. Silverman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD,CORRESPONDENCE TO: Daniel Levy, MD
| |
Collapse
|
8
|
Drosatos IA, Tsoporis JN, Izhar S, Gupta S, Tsirebolos G, Sakadakis E, Triantafyllis AS, Rigopoulos A, Rigopoulos D, Rallidis LS, Rizos I, Parker TG. Differential Regulation of Circulating Soluble Receptor for Advanced Glycation End Products (sRAGEs) and Its Ligands S100A8/A9 Four Weeks Post an Exercise Intervention in a Cohort of Young Army Recruits. Biomolecules 2021; 11:1354. [PMID: 34572568 PMCID: PMC8469473 DOI: 10.3390/biom11091354] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/17/2022] Open
Abstract
Apart from its beneficial effects on cardiovascular risk factors, an anti-inflammatory effect of exercise is strongly implicated. Yet, data regarding the effect of an exercise intervention on healthy individuals are limited and contradictory. The present study aimed to investigate the effects of a physical activity intervention on the soluble form of the receptor for advanced glycation end products (sRAGEs) and its ligands S100A8/A9. A total of 332 young army recruits volunteered and 169 completed the study. The participants underwent the standard basic training of Greek army recruits. IL-6, IL-1β, S100A8/A9, and sRAGEs were measured at the beginning and at the end of the training period. Primary rodent adult aortic smooth muscle cells (ASMCs) were analyzed for responsiveness to direct stimulation with S100A8/A9 alone or in combination with sRAGEs. At the end of the training period, we observed a statistically significant reduction in S100A8/A9 (630.98 vs. 472.12 ng/mL, p = 0.001), IL-1β (9.39 [3.8, 44.14] vs. 5.03 [2.44, 27.3] vs. pg/mL, p = 0.001), and sRAGEs (398.38 vs. 220.1 pg/mL, p = 0.001). IL-6 values did not change significantly after exercise. S100A8/A9 reduction was positively correlated with body weight (r = 0.236 [0.095, 0.370], p = 0.002) and BMI (r = 0.221 [0.092, 0.346], p = 0.004). Direct stimulation of ASMCs with S100A8/A9 increased the expression of IL-6, IL-1β, and TNF-α and, in the presence of sRAGEs, demonstrated a dose-dependent inhibition. A 4-week military training resulted in significant reduction in the pro-inflammatory cytokines IL-1β and S100A8/A9 complex. The observed reduction in sRAGEs may possibly reflect diminished RAGE axis activation. Altogether, our findings support the anti-inflammatory properties of physical activity.
Collapse
Affiliation(s)
- Ioannis-Alexandros Drosatos
- Second Department of Cardiology, Attikon University Hospital, 12462 Athens, Greece; (I.-A.D.); (G.T.); (E.S.); (A.S.T.); (A.R.); (L.S.R.); (I.R.)
- Department of Preventive Medicine, 414 Military Hospital, 15236 Athens, Greece;
| | - James N. Tsoporis
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Unity Health Toronto, University of Toronto, Toronto, ON M5B 1W8, Canada; (S.I.); (S.G.); (T.G.P.)
| | - Shehla Izhar
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Unity Health Toronto, University of Toronto, Toronto, ON M5B 1W8, Canada; (S.I.); (S.G.); (T.G.P.)
| | - Sahil Gupta
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Unity Health Toronto, University of Toronto, Toronto, ON M5B 1W8, Canada; (S.I.); (S.G.); (T.G.P.)
| | - George Tsirebolos
- Second Department of Cardiology, Attikon University Hospital, 12462 Athens, Greece; (I.-A.D.); (G.T.); (E.S.); (A.S.T.); (A.R.); (L.S.R.); (I.R.)
- Department of Cardiology, 401 General Military Hospital of Athens, 11525 Athens, Greece
| | - Eleftherios Sakadakis
- Second Department of Cardiology, Attikon University Hospital, 12462 Athens, Greece; (I.-A.D.); (G.T.); (E.S.); (A.S.T.); (A.R.); (L.S.R.); (I.R.)
| | - Andreas S. Triantafyllis
- Second Department of Cardiology, Attikon University Hospital, 12462 Athens, Greece; (I.-A.D.); (G.T.); (E.S.); (A.S.T.); (A.R.); (L.S.R.); (I.R.)
| | - Angelos Rigopoulos
- Second Department of Cardiology, Attikon University Hospital, 12462 Athens, Greece; (I.-A.D.); (G.T.); (E.S.); (A.S.T.); (A.R.); (L.S.R.); (I.R.)
| | | | - Loukianos S. Rallidis
- Second Department of Cardiology, Attikon University Hospital, 12462 Athens, Greece; (I.-A.D.); (G.T.); (E.S.); (A.S.T.); (A.R.); (L.S.R.); (I.R.)
| | - Ioannis Rizos
- Second Department of Cardiology, Attikon University Hospital, 12462 Athens, Greece; (I.-A.D.); (G.T.); (E.S.); (A.S.T.); (A.R.); (L.S.R.); (I.R.)
| | - Thomas G. Parker
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Unity Health Toronto, University of Toronto, Toronto, ON M5B 1W8, Canada; (S.I.); (S.G.); (T.G.P.)
| |
Collapse
|
9
|
Nucera F, Lo Bello F, Shen SS, Ruggeri P, Coppolino I, Di Stefano A, Stellato C, Casolaro V, Hansbro PM, Adcock IM, Caramori G. Role of Atypical Chemokines and Chemokine Receptors Pathways in the Pathogenesis of COPD. Curr Med Chem 2021; 28:2577-2653. [PMID: 32819230 DOI: 10.2174/0929867327999200819145327] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/11/2020] [Accepted: 06/18/2020] [Indexed: 11/22/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) represents a heightened inflammatory response in the lung generally resulting from tobacco smoking-induced recruitment and activation of inflammatory cells and/or activation of lower airway structural cells. Several mediators can modulate activation and recruitment of these cells, particularly those belonging to the chemokines (conventional and atypical) family. There is emerging evidence for complex roles of atypical chemokines and their receptors (such as high mobility group box 1 (HMGB1), antimicrobial peptides, receptor for advanced glycosylation end products (RAGE) or toll-like receptors (TLRs)) in the pathogenesis of COPD, both in the stable disease and during exacerbations. Modulators of these pathways represent potential novel therapies for COPD and many are now in preclinical development. Inhibition of only a single atypical chemokine or receptor may not block inflammatory processes because there is redundancy in this network. However, there are many animal studies that encourage studies for modulating the atypical chemokine network in COPD. Thus, few pharmaceutical companies maintain a significant interest in developing agents that target these molecules as potential antiinflammatory drugs. Antibody-based (biological) and small molecule drug (SMD)-based therapies targeting atypical chemokines and/or their receptors are mostly at the preclinical stage and their progression to clinical trials is eagerly awaited. These agents will most likely enhance our knowledge about the role of atypical chemokines in COPD pathophysiology and thereby improve COPD management.
Collapse
Affiliation(s)
- Francesco Nucera
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Federica Lo Bello
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Sj S Shen
- Faculty of Science, Centre for Inflammation, Centenary Institute, University of Technology, Ultimo, Sydney, Australia
| | - Paolo Ruggeri
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Irene Coppolino
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Antonino Di Stefano
- Division of Pneumology, Cyto- Immunopathology Laboratory of the Cardio-Respiratory System, Clinical Scientific Institutes Maugeri IRCCS, Veruno, Italy
| | - Cristiana Stellato
- Department of Medicine, Surgery and Dentistry, Salerno Medical School, University of Salerno, Salerno, Italy
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry, Salerno Medical School, University of Salerno, Salerno, Italy
| | - Phil M Hansbro
- Faculty of Science, Centre for Inflammation, Centenary Institute, University of Technology, Ultimo, Sydney, Australia
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Gaetano Caramori
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| |
Collapse
|
10
|
Aida Y, Kamide T, Ishii H, Kitao Y, Uchiyama N, Nakada M, Hori O. Soluble receptor for advanced glycation end products as a biomarker of symptomatic vasospasm in subarachnoid hemorrhage. J Neurosurg 2021; 134:122-130. [PMID: 31675694 DOI: 10.3171/2019.8.jns191269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/16/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The receptor for advanced glycation end products (RAGE) is a membrane protein associated with the induction of oxidative stress and inflammation in several pathological conditions. Previous studies have demonstrated that soluble RAGE (sRAGE) acts as a decoy for RAGE and protects cells against RAGE-mediated injury. The authors and other groups have reported that the expression of RAGE increases after brain ischemia and subarachnoid hemorrhage (SAH), and deletion of RAGE or overexpression of sRAGE improves neuronal survival. It has also been demonstrated that the plasma sRAGE level could be a predictor of the outcome after ischemic stroke. This study aimed to evaluate plasma sRAGE as a biomarker for symptomatic vasospasm (SVS) in SAH patients, as well as a rat model. METHODS The authors measured and compared plasma sRAGE levels in 27 SAH patients (7 with SVS and 20 without SVS) from day 5 to day 14 post-SAH. They also examined plasma sRAGE levels and expression of RAGE and heme oxygenase-1 (HO-1) in a rat SAH model. RESULTS The relative plasma sRAGE levels were significantly lower in the SVS group than in the non-SVS group of patients. A cut-off value of 0.84 for predicting SVS was considered to be appropriate for the relative plasma sRAGE levels on day 7 versus day 5. In the rat SAH model, plasma sRAGE levels were significantly lower than those in sham-treated rats, and the expressions of RAGE and HO-1 were enhanced in the SAH group compared with the non-SAH group. CONCLUSIONS Plasma sRAGE levels can be used as a potential biomarker for predicting SVS after SAH.
Collapse
Affiliation(s)
| | | | - Hiroshi Ishii
- 2Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Yasuko Kitao
- 2Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | | | | | - Osamu Hori
- 2Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| |
Collapse
|
11
|
Serum Soluble Receptor for AGE (sRAGE) Levels Are Associated With Unhealthy Lifestyle and Nonalcoholic Fatty Liver Disease. Clin Transl Gastroenterol 2020; 10:1-10. [PMID: 31082855 PMCID: PMC6602762 DOI: 10.14309/ctg.0000000000000040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has been demonstrated to be positively associated with serum advanced glycation end products (AGEs) and negatively with soluble receptor for AGE (sRAGE) in a few small studies. We aimed to test the association between lifestyle and sRAGE levels and the association between sRAGE levels or AGEs intake and NAFLD, insulin resistance (IR), and elevated alanine aminotransferase (ALT).
Collapse
|
12
|
Yamaguchi K, Iwamoto H, Mazur W, Miura S, Sakamoto S, Horimasu Y, Masuda T, Miyamoto S, Nakashima T, Ohshimo S, Fujitaka K, Hamada H, Hattori N. Reduced endogenous secretory RAGE in blood and bronchoalveolar lavage fluid is associated with poor prognosis in idiopathic pulmonary fibrosis. Respir Res 2020; 21:145. [PMID: 32527263 PMCID: PMC7291663 DOI: 10.1186/s12931-020-01410-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 05/31/2020] [Indexed: 12/16/2022] Open
Abstract
Background The endogenous secretory receptor for advanced glycation end products (esRAGE) is a soluble isoform produced by alternative splicing of the RAGE gene. The isoform has anti-inflammatory properties due to its inhibition of the RAGE/ligand interaction and is reduced in the lung tissue of patients with idiopathic pulmonary fibrosis (IPF). This study aimed to investigate the association of esRAGE serum and bronchoalveolar lavage fluid (BALF) levels with progression of IPF. Methods This study included 79 IPF patients and 90 healthy controls. IPF and control serum esRAGE levels were compared, and the correlation between serum and BALF esRAGE levels was analyzed in 57 IPF patient samples. We also investigated the relationship of esRAGE serum and BALF levels with prognoses and lung function parameters in patients with IPF. Results Serum esRAGE levels in IPF patients were significantly lower than those in healthy controls (162.0 ± 102.4 ng/ml and 200.7 ± 107.3 ng/ml, p = 0.009), although the baseline characteristics of age and smoking history were not matched. Serum levels of esRAGE were correlated with BALF esRAGE levels (rs = 0.317). The BALF esRAGE levels were also correlated with diffusion capacity for carbon monoxide (rs = 0.406). A Kaplan-Meier curve analysis and univariate/multivariate Cox hazard proportion analysis revealed that lower levels of esRAGE in blood and BALF were significantly associated with poorer prognoses in patients with IPF. Conclusions Decreased esRAGE levels in BALF and blood were associated with poor prognoses in patients with IPF. These results suggest that esRAGE could be related to the pathophysiology of IPF and serum esRAGE could be a potential prognostic marker of IPF.
Collapse
Affiliation(s)
- Kakuhiro Yamaguchi
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hiroshi Iwamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Witold Mazur
- Heart and Lung Centre, Division of Pulmonary Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Shinichiro Miura
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shinjiro Sakamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yasushi Horimasu
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Takeshi Masuda
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shintaro Miyamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Taku Nakashima
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shinichiro Ohshimo
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazunori Fujitaka
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hironobu Hamada
- Department of Physical Analysis and Therapeutic Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
13
|
Decreased Soluble Receptor of Advanced Glycation End Product Levels Correlated with Inflammation in Silicosis. Mediators Inflamm 2020; 2020:2683753. [PMID: 32351319 PMCID: PMC7178542 DOI: 10.1155/2020/2683753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/07/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
Silicosis is a devastating disease caused by inhalation of silica dust that leads to inflammatory cascade and then scarring of the lung tissue. Increasing evidences indicate that soluble receptor for advanced glycation end products (sRAGE) is involved in inflammatory diseases. However, no data on the possible relationship between sRAGE and inflammation of silicosis are available. In this study, serum from subjects with silicosis (n = 59) or from healthy controls (HC, n = 14) was analyzed for the secretion of sRAGE, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), transforming growth factor-β1 (TGF-β1), and oxidized low-density lipoprotein (ox-LDL). The associations between sRAGE and cytokines and ox-LDL and lung function were assessed by Pearson's correlation analyses. Mean levels of serum sRAGE were lower in silicosis than those in controls (p < 0.05). The subjects who had a longer term of occupational exposure had higher levels of sRAGE (p < 0.05). The secretion of TNF-α, IL-1β, IL-6, TGF-β1, and ox-LDL was significantly higher in the silicosis group than that in the HC group (p < 0.05). Furthermore, the levels of sRAGE were negatively correlated with TNF-α, IL-6, IL-1β, and ox-LDL. There is no correlation between sRAGE and TGF-β1 and lung function. The optimal point of sRAGE for differentiating silicosis from healthy controls was 14250.02 pg/ml by ROC curve analysis. A decrease in serum sRAGE and its association with inflammatory response might suggest a role for sRAGE in the pathogenesis of silicosis.
Collapse
|
14
|
Zaigham S, Persson M, Jujic A, Frantz S, Borné Y, Malinovschi A, Wollmer P, Engström G. Measures of lung function and their relationship with advanced glycation end-products. ERJ Open Res 2020; 6:00356-2019. [PMID: 32523964 PMCID: PMC7261968 DOI: 10.1183/23120541.00356-2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/11/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Advanced glycation end-products (AGEs) have been implicated in the pathophysiology of chronic obstructive pulmonary disease (COPD). However, the association between AGE accumulation in the skin measured by skin autofluorescence (SAF) and lung function in healthy subjects has not been explored in detail. We use a population-based study of 50-64-year-olds to assess spirometry, diffusing capacity of the lung for carbon monoxide (D LCO) and impulse oscillometry (IOS) in relation to SAF. METHODS Participants with information on SAF, lung function and potential confounding variables were included from the Swedish Cardiopulmonary Bioimage Study (SCAPIS) cohort (spirometry, n=4111; D LCO, n=3889; IOS, n=3970). Linear regression was used to assess changes in lung function (as measured by spirometry (forced expiratory volume in 1 s (FEV1), forced vital capacity (FVC) and FEV1/FVC), D LCO and IOS (resistance measured at 5 (R 5) and 20 Hz (R 20), R 5-R 20, area of reactance, reactance measured at 5 Hz (X- 5), and resonant frequency)) by a 1-sd increase in SAF. RESULTS FEV1, FVC and D LCO were significantly and inversely associated with SAF after adjustment for potential confounding factors (per 1-sd increase in SAF: FEV1 -0.03 L (95% CI -0.04- -0.02 L), p<0.001; FVC -0.03 L (95% CI -0.05- -0.02 L), p<0.001; D LCO -0.07 mmol·min-1·kPa-1 (95% CI -0.11- -0.03 mmol·min-1·kPa-1), p<0.001). This association was also found in nonsmokers and in non-COPD subjects. Pulmonary reactance (X 5) but not pulmonary resistance (R 5, R 20 and R 5-R 20) was significantly associated with SAF (per 1-sd increase in SAF: X 5 -0.001 kPa·L-1·s (95% CI -0.003-0.00 kPa·L-1·s), p=0.042), which was mirrored in non-COPD patients but not in current nonsmokers. CONCLUSIONS AGE accumulation, as measured by SAF, is significantly associated with lung function decrements indicative of changes in the lung parenchyma.
Collapse
Affiliation(s)
- Suneela Zaigham
- Dept of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | | | - Amra Jujic
- Dept of Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Dept of Cardiology, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Sophia Frantz
- Dept of Translational Medicine, Lund University, Malmö, Sweden
| | - Yan Borné
- Dept of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Andrei Malinovschi
- Dept of Medical Sciences, Clinical Physiology, Uppsala University, Sweden
| | - Per Wollmer
- Dept of Translational Medicine, Lund University, Malmö, Sweden
| | - Gunnar Engström
- Dept of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| |
Collapse
|
15
|
Mastej E, Gillenwater L, Zhuang Y, Pratte KA, Bowler RP, Kechris K. Identifying Protein-metabolite Networks Associated with COPD Phenotypes. Metabolites 2020; 10:metabo10040124. [PMID: 32218378 PMCID: PMC7241079 DOI: 10.3390/metabo10040124] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/06/2020] [Accepted: 03/23/2020] [Indexed: 02/02/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease in which airflow obstruction in the lung makes it difficult for patients to breathe. Although COPD occurs predominantly in smokers, there are still deficits in our understanding of the additional risk factors in smokers. To gain a deeper understanding of the COPD molecular signatures, we used Sparse Multiple Canonical Correlation Network (SmCCNet), a recently developed tool that uses sparse multiple canonical correlation analysis, to integrate proteomic and metabolomic data from the blood of 1008 participants of the COPDGene study to identify novel protein-metabolite networks associated with lung function and emphysema. Our aim was to integrate -omic data through SmCCNet to build interpretable networks that could assist in the discovery of novel biomarkers that may have been overlooked in alternative biomarker discovery methods. We found a protein-metabolite network consisting of 13 proteins and 7 metabolites which had a -0.34 correlation (p-value = 2.5 × 10-28) to lung function. We also found a network of 13 proteins and 10 metabolites that had a -0.27 correlation (p-value = 2.6 × 10-17) to percent emphysema. Protein-metabolite networks can provide additional information on the progression of COPD that complements single biomarker or single -omic analyses.
Collapse
Affiliation(s)
- Emily Mastej
- Computational Bioscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Correspondence:
| | | | - Yonghua Zhuang
- Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | | | - Russell P. Bowler
- National Jewish Health, Denver, CO 80206, USA (K.A.P.)
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Katerina Kechris
- Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
16
|
Xia S, Zhou C, Kalionis B, Shuang X, Ge H, Gao W. Combined Antioxidant, Anti-inflammaging and Mesenchymal Stem Cell Treatment: A Possible Therapeutic Direction in Elderly Patients with Chronic Obstructive Pulmonary Disease. Aging Dis 2020; 11:129-140. [PMID: 32010487 PMCID: PMC6961773 DOI: 10.14336/ad.2019.0508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/08/2019] [Indexed: 12/19/2022] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a worldwide health problem associated with high morbidity and mortality, especially in elderly patients. Aging functions include mitochondrial dysfunction, cell-to-cell information exchange, protein homeostasis and extracellular matrix dysregulation, which are closely related to chronic inflammatory response and oxidation-antioxidant imbalance in the pathogenesis of COPD. COPD displays distinct inflammaging features, including increased cellular senescence and oxidative stress, stem cell exhaustion, alterations in the extracellular matrix, reduced levels of endogenous anti-inflammaging molecules, and reduced autophagy. Given that COPD and inflammaging share similar general features, it is very important to identify the specific mechanisms of inflammaging, which involve oxidative stress, inflammation and lung mesenchymal stem cell function in the development of COPD, especially in elderly COPD patients. In this review, we highlight the studies relevant to COPD progression, and focus on mechanisms associated with inflammaging.
Collapse
Affiliation(s)
- Shijin Xia
- 1Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai, China
| | - Changxi Zhou
- 2Department of Respiratory Medicine, The Second Medical Center of PLA General Hospital, Beijing, China
| | - Bill Kalionis
- 3Department of Maternal-Fetal Medicine Pregnancy Research Centre and University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Xiaoping Shuang
- 4Department of Cardiovascular Diseases, Xiangyang Hospital of Traditional Chinese Medicine, Xiangyang, Hubei, China
| | - Haiyan Ge
- 5Department of Pulmonary Diseases, Huadong Hospital, Fudan University, Shanghai, China
| | - Wen Gao
- 6Department of Thoracic Surgery, Huadong Hospital, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Singh KP, Lawyer G, Muthumalage T, Maremanda KP, Khan NA, McDonough SR, Ye D, McIntosh S, Rahman I. Systemic biomarkers in electronic cigarette users: implications for noninvasive assessment of vaping-associated pulmonary injuries. ERJ Open Res 2019; 5:00182-2019. [PMID: 31886159 PMCID: PMC6926365 DOI: 10.1183/23120541.00182-2019] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 10/03/2019] [Indexed: 12/20/2022] Open
Abstract
Background Electronic cigarettes (e-cigs) were introduced as electronic nicotine delivery systems, and have become very popular in the USA and globally. There is a paucity of data on systemic injury biomarkers of vaping in e-cig users that can be used as a noninvasive assessment of vaping-associated lung injuries. We hypothesised that characterisation of systemic biomarkers of inflammation, anti-inflammatory, oxidative stress, vascular and lipid mediators, growth factors, and extracellular matrix breakdown may provide information regarding the toxicity of vaping in e-cig users. Methods We collected various biological fluids, i.e. plasma, urine, saliva and exhaled breath condensate (EBC), measured pulmonary function and vaping characteristics, and assessed various biomarkers in e-cig users and nonusers. Results The plasma samples of e-cig users showed a significant increase in biomarkers of inflammation (interleukin (IL)-1β, IL-6, IL-8, IL-13, interferon (IFN)-γ, matrix metalloproteinase-9, intercellular cell adhesion molecule-1) and extracellular matrix breakdown (desmosine), and decreased pro-resolving lipid mediators (resolvin D1 and resolvin D2). There was a significant increase in growth factor (endothelial growth factor, vascular endothelial growth factor, β-nerve growth factor, platelet-derived growth factor-AA, stem cell factor, hepatocyte growth factor and placental growth factor) levels in plasma of e-cig users versus nonusers. E-cig users showed a significant increase in levels of inflammatory biomarker IFN-γ, oxidative stress biomarker 8-isoprostane and oxidative DNA damage biomarker 8-oxo-dG in urine samples, and of inflammatory biomarker IL-1β in saliva samples. EBC showed a slight increase in levels of triglycerides and 8-isoprostane in e-cig users compared with normal nonusers. Conclusion E-cig users have increased levels of biomarkers of inflammation and oxidative stress, reduced pro-resolving anti-inflammatory mediators, and endothelial dysfunction, which may act as risk factors for increasing susceptibility to systemic diseases. The identified noninvasive biomarkers can be used for determining e-cig vaping-associated lung injuries, and for regulatory and diagnostic aspects of vaping in humans. E-cig use adversely affects oxidative stress and inflammatory responses, and induces tissue remodelling. The identified biomarkers can be used for assessment of vaping-associated lung injuries, and for regulatory and diagnostic aspects of vaping in humans.http://bit.ly/2nxZQ8R
Collapse
Affiliation(s)
- Kameshwar P Singh
- Dept of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.,These authors contributed equally to this work
| | - Gina Lawyer
- Dept of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.,These authors contributed equally to this work
| | - Thivanka Muthumalage
- Dept of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Krishna P Maremanda
- Dept of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Naushad Ahmad Khan
- Dept of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Samantha R McDonough
- Dept of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Dongxia Ye
- Dept of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Scott McIntosh
- Public Health Sciences, University of Rochester Medical Center, Rochester, NY, USA
| | - Irfan Rahman
- Dept of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
18
|
Hao W, Li M, Zhang C, Zhang Y, Du W. Increased levels of inflammatory biomarker CX3CL1 in patients with chronic obstructive pulmonary disease. Cytokine 2019; 126:154881. [PMID: 31629111 DOI: 10.1016/j.cyto.2019.154881] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/03/2019] [Accepted: 10/07/2019] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To investigate the concentration of CX3CL1 in serum of patients with chronic obstructive pulmonary disease (COPD), and to evaluate the associations between the CX3CL1 level and systemic inflammation, small airway obstruction, and COPD assessment test (CAT) scores in COPD patients. METHODS Enzyme-linked immunosorbent assay were utilized to detect the CX3CL1 protein in serum separately from 64 patients with COPD and 53 healthy controls. RESULTS Compared with healthy non-smokers, healthy smokers and COPD non-smokers, serum CX3CL1 protein levels were significantly elevated in COPD smokers (258.33 ± 56.27 pg/mL versus 177.32 ± 43.21 pg/mL, 185.64 ± 47.03 pg/mL, and 226.55 ± 51.79 pg/mL, P < 0.05). Correlation analysis indicated that serum CX3CL1 in COPD smokers was negatively correlated with FEV1/FVC (justified r = -0.319, P < 0.001), FEV1/Pre (justified r = -0.476, P < 0.001), FEV3/FVC (justified r = -0.354, P < 0.001), MMEF25-75/Pre (justified r = -0.428, P < 0.001), but positively correlated with CRP (justified r = 0.331, P < 0.001) and MMP-12 (justified r = 0.352, P < 0.001). However, our results showed no significant correlation between serum CX3CL1 of COPD smokers and the diffusing capacity of the lung for carbon monoxide (DLCO) (justified r = 0.0397, P = 0.6025), but a positive correlation with COPD assessment test (CAT) scores (justified r = 0.367, P < 0.001). Finally, through multivariate linear analysis, statistical results demonstrated age (β = -0.2694, P = 0.005), FEV1/Pred (β = -0.2653, P = 0.003), CRP (β = 0.1427, P = 0.0478) and MMP-12 (β = 0.430, P < 0.001) are independent parameters associated with CX3CL1. CONCLUSION The results demonstrated that elevated circulating CX3CL1 level is associated with the systemic inflammation, small airway obstruction, and CAT scores in COPD patients, suggesting that CX3CL1 may play crucial roles in the pathogenesis of COPD. Blocking CX3CL1 might prevent the progression of chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Wendong Hao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China; Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yan'an University, Yan'an 716099, Shaanxi Province, PR China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China.
| | - Cailian Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yan'an University, Yan'an 716099, Shaanxi Province, PR China
| | - Yunqing Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yan'an University, Yan'an 716099, Shaanxi Province, PR China
| | - Weiping Du
- Clinical Laboratory Diagnosis Department, The Affiliated Hospital of Yan'an University, Yan'an 716099, Shaanxi Province, PR China
| |
Collapse
|
19
|
Faiz A, van den Berge M, Vermeulen CJ, Ten Hacken NHT, Guryev V, Pouwels SD. AGER expression and alternative splicing in bronchial biopsies of smokers and never smokers. Respir Res 2019; 20:70. [PMID: 30971245 PMCID: PMC6456980 DOI: 10.1186/s12931-019-1038-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 03/29/2019] [Indexed: 02/07/2023] Open
Abstract
Cigarette smoking is one of the major risk factors for the development of chronic obstructive pulmonary disease (COPD). Evidence is accumulating that Receptor for Advanced Glycation-End products (RAGE)-signaling is a key pathway in the pathophysiology of COPD. To date, it is unknown how smoking affects RAGE expression. In the current study, we investigated the effect of smoking on AGER, the gene encoding RAGE, expression and on alternative splicing of AGER. To this end, we conducted RNA-Seq on bronchial biopsies for asymptomatic smokers (n = 36) and never smokers (n = 40). Total AGER gene expression was accessed using DESeq2, while alternative splicing was investigated by measuring the number of specific split reads spanning exon-exon junctions and the total split reads. One of the major isoforms of RAGE is endogenous soluble (es) RAGE, an anti-inflammatory decoy receptor, making up for approximately 10% of the total amount of soluble (s)RAGE. We found that smokers show decreased total gene expression of AGER in bronchial biopsies, while the relative abundance of the esRAGE isoform is increased. Furthermore, no difference in the serum levels of total sRAGE were observed between smokers and non-smokers. Our data indicates that smoking initiates a protective anti-inflammatory mechanism with decreased expression of the pro-inflammatory gene AGER and increased relative abundance of the anti-inflammatory isoform esRAGE.
Collapse
Affiliation(s)
- Alen Faiz
- Department of Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Univesity of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Univesity of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Cornelis J Vermeulen
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Univesity of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nick H T Ten Hacken
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Univesity of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Victor Guryev
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Univesity of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,European Research Institute for the Biology of Ageing, University of Groningen, Groningen, The Netherlands
| | - Simon D Pouwels
- Department of Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands. .,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Univesity of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| |
Collapse
|
20
|
da Silva LF, Skupien EC, Lazzari TK, Holler SR, de Almeida EGC, Zampieri LR, Coutinho SE, Andrades M, Silva DR. Advanced glycation end products (AGE) and receptor for AGE (RAGE) in patients with active tuberculosis, and their relationship between food intake and nutritional status. PLoS One 2019; 14:e0213991. [PMID: 30870511 PMCID: PMC6417785 DOI: 10.1371/journal.pone.0213991] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 03/05/2019] [Indexed: 01/15/2023] Open
Abstract
Introduction The receptor for advanced glycation end products (RAGE) is expressed in normal lungs and is upregulated during infection. AGEs and RAGE cause oxidative stress and apoptosis in lung cells. The objective of this study is to evaluate levels of AGEs and its soluble receptor (sRAGE), and to investigate their relationship with food intake and nutritional status, in a university-affiliated hospital in Brazil. Methods Case-control study, from June 2017 to June 2018. AGE (carboxymethyl lysine, CML) and sRAGE were measured from blood samples by Elisa. Nutritional assessment was performed by body mass index, triceps skin-fold thickness, mid-arm circumference, mid-arm muscle circumference, bioelectrical impedance analysis, and food frequency questionnaire. Results We included in the study 35 tuberculosis (TB) patients and 35 controls. The mean sRAGE levels were higher in TB patients than in controls (68.5 ± 28.1 vs 57.5 ± 24.0 pg/mL; p = 0.046). Among cases that were current smokers, lower sRAGE levels were associated with mortality, evaluated at the end of hospitalization (p = 0.006), and with weight loss (p = 0.034). There was no statistically significant difference in CML levels and diet CML content between cases and controls. Malnutrition was more frequent in cases, but there was no correlation between nutritional parameters and CML or sRAGE levels. Conclusions TB patients had higher sRAGE levels than controls, although it is not clear that this difference is clinically relevant. Also, sRAGE was associated with weight loss and mortality.
Collapse
Affiliation(s)
- Lívia Fontes da Silva
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Erika Cavalheiro Skupien
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tássia Kirchmann Lazzari
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Sizuane Rieger Holler
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | - Sandra Eugênia Coutinho
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Denise Rossato Silva
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- * E-mail:
| |
Collapse
|
21
|
Pouwels SD, Klont F, Kwiatkowski M, Wiersma VR, Faiz A, van den Berge M, Horvatovich P, Bischoff R, ten Hacken NHT. Cigarette Smoking Acutely Decreases Serum Levels of the Chronic Obstructive Pulmonary Disease Biomarker sRAGE. Am J Respir Crit Care Med 2018; 198:1456-1458. [DOI: 10.1164/rccm.201807-1249le] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
| | - Frank Klont
- University of GroningenGroningen, the Netherlands
| | | | | | - Alen Faiz
- University of GroningenGroningen, the Netherlands
| | | | | | | | | |
Collapse
|
22
|
Liguori I, Russo G, Curcio F, Bulli G, Aran L, Della-Morte D, Gargiulo G, Testa G, Cacciatore F, Bonaduce D, Abete P. Oxidative stress, aging, and diseases. Clin Interv Aging 2018; 13:757-772. [PMID: 29731617 PMCID: PMC5927356 DOI: 10.2147/cia.s158513] [Citation(s) in RCA: 2162] [Impact Index Per Article: 308.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Reactive oxygen and nitrogen species (RONS) are produced by several endogenous and exogenous processes, and their negative effects are neutralized by antioxidant defenses. Oxidative stress occurs from the imbalance between RONS production and these antioxidant defenses. Aging is a process characterized by the progressive loss of tissue and organ function. The oxidative stress theory of aging is based on the hypothesis that age-associated functional losses are due to the accumulation of RONS-induced damages. At the same time, oxidative stress is involved in several age-related conditions (ie, cardiovascular diseases [CVDs], chronic obstructive pulmonary disease, chronic kidney disease, neurodegenerative diseases, and cancer), including sarcopenia and frailty. Different types of oxidative stress biomarkers have been identified and may provide important information about the efficacy of the treatment, guiding the selection of the most effective drugs/dose regimens for patients and, if particularly relevant from a pathophysiological point of view, acting on a specific therapeutic target. Given the important role of oxidative stress in the pathogenesis of many clinical conditions and aging, antioxidant therapy could positively affect the natural history of several diseases, but further investigation is needed to evaluate the real efficacy of these therapeutic interventions. The purpose of this paper is to provide a review of literature on this complex topic of ever increasing interest.
Collapse
Affiliation(s)
- Ilaria Liguori
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Gennaro Russo
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Francesco Curcio
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Giulia Bulli
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Luisa Aran
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - David Della-Morte
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,San Raffaele Roma Open University, Rome, Italy
| | - Gaetano Gargiulo
- Division of Internal Medicine, AOU San Giovanni di Dio e Ruggi di Aragona, Salerno, Italy
| | - Gianluca Testa
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy.,Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Francesco Cacciatore
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy.,Azienda Ospedaliera dei Colli, Monaldi Hospital, Heart Transplantation Unit, Naples, Italy
| | - Domenico Bonaduce
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Pasquale Abete
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
23
|
Receptor for advanced glycation end-products and ARDS prediction: a multicentre observational study. Sci Rep 2018; 8:2603. [PMID: 29422518 PMCID: PMC5805783 DOI: 10.1038/s41598-018-20994-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 01/29/2018] [Indexed: 01/06/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) prediction remains challenging despite available clinical scores. To assess soluble receptor for advanced glycation end-products (sRAGE), a marker of lung epithelial injury, as a predictor of ARDS in a high-risk population, adult patients with at least one ARDS risk factor upon admission to participating intensive care units (ICUs) were enrolled in a multicentre, prospective study between June 2014 and January 2015. Plasma sRAGE and endogenous secretory RAGE (esRAGE) were measured at baseline (ICU admission) and 24 hours later (day one). Four AGER candidate single nucleotide polymorphisms (SNPs) were also assayed because of previous reports of functionality (rs1800625, rs1800624, rs3134940, and rs2070600). The primary outcome was ARDS development within seven days. Of 500 patients enrolled, 464 patients were analysed, and 59 developed ARDS by day seven. Higher baseline and day one plasma sRAGE, but not esRAGE, were independently associated with increased ARDS risk. AGER SNP rs2070600 (Ser/Ser) was associated with increased ARDS risk and higher plasma sRAGE in this cohort, although confirmatory studies are needed to assess the role of AGER SNPs in ARDS prediction. These findings suggest that among at-risk ICU patients, higher plasma sRAGE may identify those who are more likely to develop ARDS.
Collapse
|
24
|
Klont F, Pouwels SD, Hermans J, van de Merbel NC, Horvatovich P, Ten Hacken NHT, Bischoff R. A fully validated liquid chromatography-mass spectrometry method for the quantification of the soluble receptor of advanced glycation end-products (sRAGE) in serum using immunopurification in a 96-well plate format. Talanta 2018; 182:414-421. [PMID: 29501172 DOI: 10.1016/j.talanta.2018.02.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/03/2018] [Accepted: 02/05/2018] [Indexed: 12/31/2022]
Abstract
The study of proteins is central to unraveling (patho)physiological processes and has contributed greatly to our understanding of biological systems. Corresponding studies often employ procedures to enrich proteins from their biological matrix using antibodies or other affinity binders coupled to beads with a large surface area and a correspondingly high binding capacity. Striving for maximal binding capacity may, however, not always be required or desirable, for example for proteins of low abundance. Here we describe a simplified immunoprecipitation in 96-well ELISA format (IPE) approach for fast and easy enrichment of proteins. The applicability of this approach for enriching low-abundant proteins was demonstrated by an IPE-based quantitative workflow using liquid chromatography-mass spectrometry (LC-MS) for the soluble Receptor of Advanced Glycation End-products (sRAGE), a promising biomarker in chronic obstructive pulmonary disease (COPD). The method was validated according to U.S. Food and Drug Administration (FDA) and European Medicines Agency (EMA) guidelines and enabled accurate quantitation of sRAGE between 0.1 and 10 ng/mL in 50 µL serum. The assay showed substantial correlation with the two most commonly-used sRAGE immunoassays (ELISAs) (R2-values between 0.7 and 0.8). However, the LC-MS method reported 2-4 times higher sRAGE levels compared to the ELISAs, which is largely due to a suboptimal amount of capturing antibody and/or calibration strategy used by the immunoassays. In conclusion, our simplified IPE approach proved to be an efficient strategy for enriching the low-abundant protein sRAGE from serum and may provide an easy to use platform for enriching other (low-abundant) proteins from complex, biological matrices.
Collapse
Affiliation(s)
- Frank Klont
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Simon D Pouwels
- Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Jos Hermans
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Nico C van de Merbel
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; Bioanalytical Laboratory, PRA Health Sciences, Early Development Services, Amerikaweg 18, 9407 TK Assen, The Netherlands
| | - Péter Horvatovich
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Nick H T Ten Hacken
- Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Rainer Bischoff
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| |
Collapse
|
25
|
Oczypok EA, Perkins TN, Oury TD. All the "RAGE" in lung disease: The receptor for advanced glycation endproducts (RAGE) is a major mediator of pulmonary inflammatory responses. Paediatr Respir Rev 2017; 23:40-49. [PMID: 28416135 PMCID: PMC5509466 DOI: 10.1016/j.prrv.2017.03.012] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 03/10/2017] [Indexed: 02/07/2023]
Abstract
The receptor for advanced glycation endproducts (RAGE) is a pro-inflammatory pattern recognition receptor (PRR) that has been implicated in the pathogenesis of numerous inflammatory diseases. It was discovered in 1992 on endothelial cells and was named for its ability to bind advanced glycation endproducts and promote vascular inflammation in the vessels of patients with diabetes. Further studies revealed that RAGE is most highly expressed in lung tissue and spurred numerous explorations into RAGE's role in the lung. These studies have found that RAGE is an important mediator in allergic airway inflammation (AAI) and asthma, pulmonary fibrosis, lung cancer, chronic obstructive pulmonary disease (COPD), acute lung injury, pneumonia, cystic fibrosis, and bronchopulmonary dysplasia. RAGE has not yet been targeted in the lungs of paediatric or adult clinical populations, but the development of new ways to inhibit RAGE is setting the stage for the emergence of novel therapeutic agents for patients suffering from these pulmonary conditions.
Collapse
Affiliation(s)
| | | | - Tim D. Oury
- Corresponding author. Tel.: +1 412 648 9659; Fax: +1 412 648 9527
| |
Collapse
|
26
|
Increase of Soluble RAGE in Cerebrospinal Fluid following Subarachnoid Haemorrhage. BIOMED RESEARCH INTERNATIONAL 2017. [PMID: 28630869 PMCID: PMC5467298 DOI: 10.1155/2017/7931534] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Receptors for advanced glycation end-products (RAGE) mediate the inflammatory reaction that follows aneurysmal subarachnoid haemorrhage. Soluble RAGE (sRAGE) may function as a decoy receptor. The significance of this endogenous anti-inflammatory mechanism in subarachnoid haemorrhage (SAH) remains unknown. The present study aims to analyse sRAGE levels in the cerebrospinal fluid (CSF) of SAH patients. sRAGE levels were assayed by ELISA kit in 47 CSF samples collected on post-SAH days 0–3, 5–7, and 10–14 from 27 SAH patients with acute hydrocephalus. CSF levels of sRAGE were compared with a control group and correlated with other monitored parameters. In the control group, the CSF contained only a trace amount of sRAGE. By contrast, the CSF of 20 SAH patients collected on post-SAH days 0–3 was found to contain statistically significant higher levels of sRAGE (mean concentration 3.91 pg/mL, p < 0.001). The most pronounced difference in CSF sRAGE levels between good and poor outcome patients was found on days 0–3 post-SAH but did not reach the significance threshold (p = 0.234). CSF sRAGE levels did not change significantly during hospitalisation (p = 0.868) and correlated poorly with treatment outcome, systemic inflammatory markers, and other monitored parameters. Our study revealed an early and constant increase of sRAGE level in the CSF of SAH patients.
Collapse
|
27
|
Reynaert NL, Gopal P, Rutten EP, Wouters EF, Schalkwijk CG. Advanced glycation end products and their receptor in age-related, non-communicable chronic inflammatory diseases; Overview of clinical evidence and potential contributions to disease. Int J Biochem Cell Biol 2016; 81:403-418. [PMID: 27373680 DOI: 10.1016/j.biocel.2016.06.016] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 06/24/2016] [Accepted: 06/28/2016] [Indexed: 12/31/2022]
|
28
|
Machahua C, Montes-Worboys A, Llatjos R, Escobar I, Dorca J, Molina-Molina M, Vicens-Zygmunt V. Increased AGE-RAGE ratio in idiopathic pulmonary fibrosis. Respir Res 2016; 17:144. [PMID: 27816054 PMCID: PMC5097848 DOI: 10.1186/s12931-016-0460-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 10/28/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The abnormal epithelial-mesenchymal restorative capacity in idiopathic pulmonary fibrosis (IPF) has been recently associated with an accelerated aging process as a key point for the altered wound healing. The advanced glycation end-products (AGEs) are the consequence of non-enzymatic reactions between lipid and protein with several oxidants in the aging process. The receptor for AGEs (RAGEs) has been implicated in the lung fibrotic process and the alveolar homeostasis. However, this AGE-RAGE aging pathway has been under-explored in IPF. METHODS Lung samples from 16 IPF and 9 control patients were obtained through surgical lung biopsy. Differences in AGEs and RAGE expression between both groups were evaluated by RT-PCR, Western blot and immunohistochemistry. The effect of AGEs on cell viability of primary lung fibrotic fibroblasts and alveolar epithelial cells was assessed. Cell transformation of fibrotic fibroblasts cultured into glycated matrices was evaluated in different experimental conditions. RESULTS Our study demonstrates an increase of AGEs together with a decrease of RAGEs in IPF lungs, compared with control samples. Two specific AGEs involved in aging, pentosidine and Nε-Carboxymethyl lysine, were significantly increased in IPF samples. The immunohistochemistry identified higher staining of AGEs related to extracellular matrix (ECM) proteins and the apical surface of the alveolar epithelial cells (AECs) surrounding fibroblast foci in fibrotic lungs. On the other hand, RAGE location was present at the cell membrane of AECs in control lungs, while it was almost missing in pulmonary fibrotic tissue. In addition, in vitro cultures showed that the effect of AGEs on cell viability was different for AECs and fibrotic fibroblasts. AGEs decreased cell viability in AECs, even at low concentration, while fibroblast viability was less affected. Furthermore, fibroblast to myofibroblast transformation could be enhanced by ECM glycation. CONCLUSIONS All of these findings suggest a possible role of the increased ratio AGEs-RAGEs in IPF, which could be a relevant accelerating aging tissue reaction in the abnormal wound healing of the lung fibrotic process.
Collapse
Affiliation(s)
- Carlos Machahua
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
| | - Ana Montes-Worboys
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
- Research Network in Respiratory Diseases (CIBERES), Madrid, Spain
| | - Roger Llatjos
- Department of Pathology, University Hospital of Bellvitge, Barcelona, Spain
| | - Ignacio Escobar
- Department of Thoracic Surgery, University Hospital of Bellvitge, Barcelona, Spain
| | - Jordi Dorca
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
- Research Network in Respiratory Diseases (CIBERES), Madrid, Spain
| | - Maria Molina-Molina
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
- Research Network in Respiratory Diseases (CIBERES), Madrid, Spain
| | - Vanesa Vicens-Zygmunt
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
| |
Collapse
|
29
|
Choudhury G, MacNee W. Role of Inflammation and Oxidative Stress in the Pathology of Ageing in COPD: Potential Therapeutic Interventions. COPD 2016; 14:122-135. [DOI: 10.1080/15412555.2016.1214948] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Gourab Choudhury
- MRC Centre for Inflammation Research, Queens Medical Research Institute, 47 little France Crescent Edinburgh, United Kingdom
| | - William MacNee
- MRC Centre for Inflammation Research, Queens Medical Research Institute, 47 little France Crescent Edinburgh, United Kingdom
| |
Collapse
|
30
|
John M, McKeever TM, Haddad MA, Hall IP, Sayers I, Cockcroft JR, Bolton CE. Traditional and emerging indicators of cardiovascular risk in chronic obstructive pulmonary disease. Chron Respir Dis 2016; 13:247-55. [PMID: 26965223 PMCID: PMC5720186 DOI: 10.1177/1479972316636995] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
With the increased cardiovascular (CV) morbidity and mortality in subjects with chronic obstructive pulmonary disease (COPD), there is a priority to identify those patients at increased risk of cardiovascular disease. Stable patients with COPD (n = 185) and controls with a smoking history (n = 106) underwent aortic pulse wave velocity (PWV), blood pressure (BP) and skin autofluorescence (AF) at clinical stability. Blood was sent for fasting lipids, soluble receptor for advanced glycation end products (sRAGE) and CV risk prediction scores were calculated. More patients (18%) had a self-reported history of CV disease than controls (8%), p = 0.02, whilst diabetes was similar (14% and 10%), p = 0.44. Mean (SD) skin AF was greater in patients: 3.1 (0.5) AU than controls 2.8 (0.6) AU, p < 0.001. Aortic PWV was greater in patients: 10.2 (2.3) m/s than controls: 9.6 (2.0) m/s, p = 0.02 despite similar BP. The CV risk prediction scores did not differentiate between patients and controls nor were the individual components of the scores different. The sRAGE levels were not statistically different. We present different indicators of CV risk alongside each other in well-defined subjects with and without COPD. Two non-invasive biomarkers associated with future CV burden: skin AF and aortic PWV are both significantly greater in patients with COPD compared to the controls. The traditional CV prediction scores used in the general population were not statistically different. We provide new data to suggest that alternative approaches for optimal CV risk detection should be employed in COPD management.
Collapse
Affiliation(s)
- Michelle John
- Nottingham Respiratory Research Unit and Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, UK
| | - Tricia M McKeever
- Department of Epidemiology, School of Medicine, University of Nottingham, Nottingham, UK
| | - Maath Al Haddad
- Nottingham Respiratory Research Unit and Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, UK
| | - Ian P Hall
- Nottingham Respiratory Research Unit and Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, UK
| | - Ian Sayers
- Nottingham Respiratory Research Unit and Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, UK
| | | | - Charlotte E Bolton
- Nottingham Respiratory Research Unit and Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
31
|
Hassanein EG, ElGanady AA, Baess AI, Issa YA, ElAkhtel EM. Sputum–plasma ratio of soluble receptor for advanced glycation end-products in patients with chronic obstructive pulmonary disease. EGYPTIAN JOURNAL OF CHEST DISEASES AND TUBERCULOSIS 2016. [DOI: 10.1016/j.ejcdt.2016.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
32
|
Biswas SK. What does Cigarette Smoking do to the Circulating Level of Soluble Receptor for Advanced Glycation End Products? Int J Angiol 2016; 25:137-8. [PMID: 27231433 DOI: 10.1055/s-0036-1579690] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Affiliation(s)
- Subrata Kumar Biswas
- Department of Biochemistry, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka, Bangladesh
| |
Collapse
|
33
|
Pouwels SD, Nawijn MC, Bathoorn E, Riezebos-Brilman A, van Oosterhout AJM, Kerstjens HAM, Heijink IH. Increased serum levels of LL37, HMGB1 and S100A9 during exacerbation in COPD patients. Eur Respir J 2016; 45:1482-5. [PMID: 25931489 DOI: 10.1183/09031936.00158414] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Simon D Pouwels
- Dept of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martijn C Nawijn
- Dept of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Erik Bathoorn
- Dept of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Annelies Riezebos-Brilman
- Dept of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Antoon J M van Oosterhout
- Dept of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Huib A M Kerstjens
- GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands Dept of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Irene H Heijink
- Dept of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands Dept of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
34
|
Yonchuk JG, Silverman EK, Bowler RP, Agustí A, Lomas DA, Miller BE, Tal-Singer R, Mayer RJ. Circulating soluble receptor for advanced glycation end products (sRAGE) as a biomarker of emphysema and the RAGE axis in the lung. Am J Respir Crit Care Med 2015; 192:785-92. [PMID: 26132989 DOI: 10.1164/rccm.201501-0137pp] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a complex and heterogeneous disease that has been traditionally characterized by incompletely reversible airflow limitation. Yet, the latter is poorly correlated with many other clinically relevant characteristics of the disease. Thus, the identification of biomarkers to more accurately assess this heterogeneity and disease severity may facilitate the discovery and development of new treatments and better management of patients with COPD. One molecule that has attracted attention as a potentially useful biomarker specifically for the emphysema subpopulation is the soluble receptor for advanced glycation end products (sRAGE). As the soluble isoform of a key proinflammatory signaling receptor, sRAGE acts as a "decoy" for RAGE ligands and prevents their interaction with the receptor. Multiple reports have now linked sRAGE to COPD, and more specifically to emphysema, and evidence is accumulating that this link is likely mechanistic in nature. Here we review the current state of knowledge about sRAGE biology, the mechanistic links to COPD, and the evidence for using it as a biomarker for emphysema. We also discuss sRAGE as a potential target for therapeutic intervention in COPD.
Collapse
Affiliation(s)
- John G Yonchuk
- 1 GlaxoSmithKline Research and Development, King of Prussia, Pennsylvania
| | - Edwin K Silverman
- 2 Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Alvar Agustí
- 4 Thorax Institute, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona and Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Barcelona, Spain; and
| | - David A Lomas
- 5 Wolfson Institute for Biochemical Research, Division of Medicine, University College London, London, United Kingdom
| | - Bruce E Miller
- 1 GlaxoSmithKline Research and Development, King of Prussia, Pennsylvania
| | - Ruth Tal-Singer
- 1 GlaxoSmithKline Research and Development, King of Prussia, Pennsylvania
| | - Ruth J Mayer
- 1 GlaxoSmithKline Research and Development, King of Prussia, Pennsylvania
| |
Collapse
|
35
|
Olivas-Calderón E, Recio-Vega R, Gandolfi AJ, Lantz RC, González-Cortes T, Gonzalez-De Alba C, Froines JR, Espinosa-Fematt JA. Lung inflammation biomarkers and lung function in children chronically exposed to arsenic. Toxicol Appl Pharmacol 2015; 287:161-167. [PMID: 26048584 PMCID: PMC4751871 DOI: 10.1016/j.taap.2015.06.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 05/28/2015] [Accepted: 06/01/2015] [Indexed: 01/11/2023]
Abstract
Evidence suggests that exposure to arsenic in drinking water during early childhood or in utero has been associated with an increase in respiratory symptoms or diseases in the adulthood, however only a few studies have been carried out during those sensitive windows of exposure. Recently our group demonstrated that the exposure to arsenic during early childhood or in utero in children was associated with impairment in the lung function and suggested that this adverse effect could be due to a chronic inflammation response to the metalloid. Therefore, we designed this cross-sectional study in a cohort of children associating lung inflammatory biomarkers and lung function with urinary As levels. A total of 275 healthy children were partitioned into four study groups according with their arsenic urinary levels. Inflammation biomarkers were measured in sputum by ELISA and the lung function was evaluated by spirometry. Fifty eight percent of the studied children were found to have a restrictive spirometric pattern. In the two highest exposed groups, the soluble receptor for advanced glycation end products' (sRAGE) sputum level was significantly lower and matrix metalloproteinase-9 (MMP-9) concentration was higher. When the biomarkers were correlated to the urinary arsenic species, negative associations were found between dimethylarsinic (DMA), monomethylarsonic percentage (%MMA) and dimethylarsinic percentage (%DMA) with sRAGE and positive associations between %DMA with MMP-9 and with the MMP-9/tissue inhibitor of metalloproteinase (TIMP-1) ratio. In conclusion, chronic arsenic exposure of children negatively correlates with sRAGE, and positively correlated with MMP-9 and MMP-9/TIMP-1 levels, and increases the frequency of an abnormal spirometric pattern. Arsenic-induced alterations in inflammatory biomarkers may contribute to the development of restrictive lung diseases.
Collapse
Affiliation(s)
- Edgar Olivas-Calderón
- Department of Environmental Health, Biomedical Research Center, School of Medicine, University of Coahuila, Torreon, Coahuila, Mexico; School of Medicine, University Juarez of Durango, Gomez Palacio, Durango, Mexico.
| | - Rogelio Recio-Vega
- Department of Environmental Health, Biomedical Research Center, School of Medicine, University of Coahuila, Torreon, Coahuila, Mexico.
| | - A Jay Gandolfi
- Southwest Environmental Health Science Center, University of Arizona, Tucson, AZ, USA; Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| | - R Clark Lantz
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA; Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA.
| | - Tania González-Cortes
- Department of Environmental Health, Biomedical Research Center, School of Medicine, University of Coahuila, Torreon, Coahuila, Mexico.
| | - Cesar Gonzalez-De Alba
- Department of Environmental Health, Biomedical Research Center, School of Medicine, University of Coahuila, Torreon, Coahuila, Mexico.
| | - John R Froines
- Center for Environmental and Occupational Health, School of Public Health, University of California at Los Angeles, Los Angeles, CA, USA.
| | | |
Collapse
|
36
|
Prasad K, Dhar I, Caspar-Bell G. Role of Advanced Glycation End Products and Its Receptors in the Pathogenesis of Cigarette Smoke-Induced Cardiovascular Disease. Int J Angiol 2015; 24:75-80. [PMID: 26060376 DOI: 10.1055/s-0034-1396413] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
The interaction of advanced glycation end products (AGEs) with its cell-bound receptor RAGE increases gene expression and release of proinflammatory cytokines and increase generation of reactive oxygen species (ROS). Circulating receptors, soluble RAGE (sRAGE), and endosecretory RAGE (esRAGE) by binding with RAGE ligands have protective effects against AGE-RAGE interaction. Cigarette smoking is a risk factor for coronary artery disease, stroke, and peripheral vascular disease. This article reviews; if the AGE-RAGE axis is involved in the cigarette smoke-induced cardiovascular diseases. There are various sources of AGEs in smokers including, gas/tar of cigarette, activation of macrophages and polymorphonuclear leukocytes, uncoupling of endothelial isoform of nitric oxide synthase (eNOS) and xanthine oxidase. The levels of AGEs are elevated in smokers. Serum levels of sRAGE have been reported to be reduced, elevated, or unchanged in smokers. Mostly the levels are reduced. There is one article which shows an elevation of levels of sRAGE in smokers. Serum levels of esRAGE are unaltered in smokers. Mechanism of AGE-RAGE-induced atherosclerosis has been discussed. Atherosclerosis leads to the cardiovascular diseases. It has been suggested that ratio of AGE/sRAGE or AGE/esRAGE is useful in determining the deleterious effects of AGE-RAGE interaction in smokers. sRAGE alone is not a good marker for smoke-induced cardiovascular disease. In conclusion cigarette smoke induces formation of AGEs and reduces sRAGE resulting in the development of atherosclerosis and related coronary heart disease, stroke, and peripheral vascular disease. Ratio of AGEs/sRAGE is a better marker for cardiovascular disease than AGEs or sRAGE alone in smokers.
Collapse
Affiliation(s)
- Kailash Prasad
- Department of Physiology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Indu Dhar
- Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Gudrun Caspar-Bell
- Department of Medicine, Royal University Hospital, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
37
|
Gopal P, Gosker HR, Theije CCD, Eurlings IM, Sell DR, Monnier VM, Reynaert NL. Effect of chronic hypoxia on RAGE and its soluble forms in lungs and plasma of mice. Biochim Biophys Acta Mol Basis Dis 2015; 1852:992-1000. [PMID: 25703138 DOI: 10.1016/j.bbadis.2015.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 01/30/2015] [Accepted: 02/12/2015] [Indexed: 12/24/2022]
Abstract
The receptor for advanced glycation end products (RAGE) is a multi-ligand receptor. Alternative splicing and enzymatic shedding produce soluble forms that protect against damage by ligands including Advanced Glycation End products (AGEs). A link between RAGE and oxygen levels is evident from studies showing RAGE-mediated injury following hyperoxia. The effect of hypoxia on pulmonary RAGE expression and circulating sRAGE levels is however unknown. Therefore mice were exposed to chronic hypoxia for 21 d and expression of RAGE, sheddases in lungs and circulating sRAGE were determined. In addition, accumulation of AGEs in lungs and expression of the AGE detoxifying enzyme GLO1 and receptors were evaluated. In lung tissue gene expression of total RAGE, variants 1 and 3 were elevated in mice exposed to hypoxia, whereas mRAGE and sRAGE protein levels were decreased. In the hypoxic group plasma sRAGE levels were enhanced. Although the levels of pro-ADAM10 were elevated in lungs of hypoxia exposed mice, the relative amount of the active form was decreased and gelatinase activity unaffected. In the lungs, the RAGE ligand HMGB1 was decreased and of the AGEs, only LW-1 was increased by chronic hypoxia. Gene expression of AGE receptors 2 and 3 was significantly upregulated. Chronic hypoxia is associated with downregulation of pulmonary RAGE protein levels, but a relative increase in sRAGE. These alterations might be part of the adaptive and protective response mechanism to chronic hypoxia and are not associated with AGE formation except for the fluorophore LW-1 which emerges as a novel marker of tissue hypoxia.
Collapse
Affiliation(s)
- P Gopal
- Department of Respiratory Medicine, Maastricht University, Maastricht, The Netherlands
| | - H R Gosker
- Department of Respiratory Medicine, Maastricht University, Maastricht, The Netherlands
| | - C C de Theije
- Department of Respiratory Medicine, Maastricht University, Maastricht, The Netherlands
| | - I M Eurlings
- Department of Respiratory Medicine, Maastricht University, Maastricht, The Netherlands
| | - D R Sell
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - V M Monnier
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - N L Reynaert
- Department of Respiratory Medicine, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
38
|
Yoo S, Takikawa S, Geraghty P, Argmann C, Campbell J, Lin L, Huang T, Tu Z, Feronjy R, Spira A, Schadt EE, Powell CA, Zhu J. Integrative analysis of DNA methylation and gene expression data identifies EPAS1 as a key regulator of COPD. PLoS Genet 2015; 11:e1004898. [PMID: 25569234 PMCID: PMC4287352 DOI: 10.1371/journal.pgen.1004898] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 11/17/2014] [Indexed: 01/11/2023] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a complex disease. Genetic, epigenetic, and environmental factors are known to contribute to COPD risk and disease progression. Therefore we developed a systematic approach to identify key regulators of COPD that integrates genome-wide DNA methylation, gene expression, and phenotype data in lung tissue from COPD and control samples. Our integrative analysis identified 126 key regulators of COPD. We identified EPAS1 as the only key regulator whose downstream genes significantly overlapped with multiple genes sets associated with COPD disease severity. EPAS1 is distinct in comparison with other key regulators in terms of methylation profile and downstream target genes. Genes predicted to be regulated by EPAS1 were enriched for biological processes including signaling, cell communications, and system development. We confirmed that EPAS1 protein levels are lower in human COPD lung tissue compared to non-disease controls and that Epas1 gene expression is reduced in mice chronically exposed to cigarette smoke. As EPAS1 downstream genes were significantly enriched for hypoxia responsive genes in endothelial cells, we tested EPAS1 function in human endothelial cells. EPAS1 knockdown by siRNA in endothelial cells impacted genes that significantly overlapped with EPAS1 downstream genes in lung tissue including hypoxia responsive genes, and genes associated with emphysema severity. Our first integrative analysis of genome-wide DNA methylation and gene expression profiles illustrates that not only does DNA methylation play a ‘causal’ role in the molecular pathophysiology of COPD, but it can be leveraged to directly identify novel key mediators of this pathophysiology. Chronic Obstructive Pulmonary Disease (COPD) is a common lung disease. It is the fourth leading cause of death in the world and is expected to be the third by 2020. COPD is a heterogeneous and complex disease consisting of obstruction in the small airways, emphysema, and chronic bronchitis. COPD is generally caused by exposure to noxious particles or gases, most commonly from cigarette smoking. However, only 20–25% of smokers develop clinically significant airflow obstruction. Smoking is known to cause epigenetic changes in lung tissues. Thus, genetics, epigenetic, and their interaction with environmental factors play an important role in COPD pathogenesis and progression. Currently, there are no therapeutics that can reverse COPD progression. In order to identify new targets that may lead to the development of therapeutics for curing COPD, we developed a systematic approach to identify key regulators of COPD that integrates genome-wide DNA methylation, gene expression, and phenotype data in lung tissue from COPD and control samples. Our integrative analysis identified 126 key regulators of COPD. We identified EPAS1 as the only key regulator whose downstream genes significantly overlapped with multiple genes sets associated with COPD disease severity.
Collapse
Affiliation(s)
- Seungyeul Yoo
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Sachiko Takikawa
- Division of Pulmonary, Critical Care and Sleep Medicine, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Patrick Geraghty
- Department of Medicine, St. Luke's Roosevelt Medical Center, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Carmen Argmann
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Joshua Campbell
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Luan Lin
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Tao Huang
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Zhidong Tu
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Robert Feronjy
- Department of Medicine, St. Luke's Roosevelt Medical Center, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Avrum Spira
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Eric E. Schadt
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Charles A. Powell
- Division of Pulmonary, Critical Care and Sleep Medicine, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Jun Zhu
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
39
|
Age and genetic determinants of variation of circulating levels of the receptor for advanced glycation end products (RAGE) in the general human population. Mech Ageing Dev 2015; 145:18-25. [DOI: 10.1016/j.mad.2015.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 01/22/2015] [Accepted: 01/29/2015] [Indexed: 11/23/2022]
|
40
|
Dullaart RP, Al‐Daghri NM, Ashina M, Bouzas‐Mosquera A, Brunetti ND, Buechler C, Chen H, Corrales JJ, D'Archivio M, Dei Cas A, Pino GG, Gómez‐Abril SA, Győri D, Haslacher H, Herder C, Kerstens MN, Koutsilieris M, Lombardi C, Lupattelli G, Mócsai A, Msaouel P, Orfao A, Ormazabal P, Pacher R, Perkmann T, Peteiro J, Plischke M, Reynaert NL, Ricci MA, Robles NR, Rocha M, Rutten EP, Sabico S, Santamaria F, Santoro F, Schmid A, Schmidt M, Schytz HW, Shyu K, Tada H, Thorand B, Valerio G, Vesely DL, Wu T, Yamagishi M, Yeh Y. Research update for articles published in EJCI in 2012. Eur J Clin Invest 2014; 44:1010-1023. [DOI: 10.1111/eci.12319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
|