1
|
Jang J, Lee J, Park J, Cha S, Lee SB, Park SM, Hong SH, Kim WJ, Lee M, Yang SR. Recombinant RAGE antagonist peptide promotes alveolar epithelial cell regeneration via the RAGE/MAPKs/MMP2 pathway in emphysema. Biochem Pharmacol 2025; 231:116668. [PMID: 39608502 DOI: 10.1016/j.bcp.2024.116668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/25/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
The progression of chronic obstructive pulmonary disease (COPD) results in irreversible pulmonary damage and sustained inflammatory responses. While alternative approaches have been explored, the specific role of alveolar epithelial cells in the pathogenesis of COPD remains unclear. Additionally, the association between emphysema and DAMP-RAGE signaling in COPD patients are not understood. Therefore, this study demonstrates to determine the therapeutic effect of a RAGE antagonist peptide (RAP), which we previously identified on the pathogenesis of COPD. We assessed the expression of RAGE ligands and RAGE binding signaling in COPD patients using GEO data. PPE-induced emphysema mouse model and AGER-/- mouse were employed, along treated with RAP. The association between RAGE and the development of emphysema was examined in H&E staining and western blot analysis in mouse lung tissue and BALF. We next analyzed the damage caused by oxidative stress and inflammation through CSE and RAP in human alveolar epithelial cell line A549. Our results show that inhibiting of RAGE alleviates emphysema by suppressing inflammation and MMP activity. Inhibition of RAGE in alveolar epithelial cells significantly induced the mitigation of lung injury, independent of macrophage infiltration. Furthermore, it was confirmed that RAP ameliorated CSE-induced oxidative stress, inflammation, and cell cycle arrest in human alveolar epithelial cells. These findings demonstrate that inhibiting RAGE in alveolar epithelial cells suppress lung injury and emphysema by inhibiting oxidative stress-induced inflammation and MMPs, while promoting alveolar epithelial cell proliferation. Furthermore, blocking of the DAMP-RAGE interaction through RAP offers a promising therapeutic approach for mitigating emphysema.
Collapse
Affiliation(s)
- Jimin Jang
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Jooyeon Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Jaehyun Park
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Sangryul Cha
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Se Bi Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Sung-Min Park
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Woo Jin Kim
- Department of Internal Medicine, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea; Institute of Medical Science, School of Medicine, Kangwon National University, Chuncheon, Gangwon State, South Korea.
| |
Collapse
|
2
|
Han H, Peng X, Huang M, Zhao W, Yang S, Lan Z, Cai S, Zhao H. PM2.5 Exposure Aggravates Inflammatory Response and Mucus Production in 16HBE Cells through Inducing Oxidative Stress and RAGE Expression. Cell Biochem Biophys 2024:10.1007/s12013-024-01526-z. [PMID: 39294419 DOI: 10.1007/s12013-024-01526-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2024] [Indexed: 09/20/2024]
Abstract
Particulate matter 2.5 (PM2.5)-induced oxidative stress has been extensively proposed as a pivotal event in lung diseases. Receptor for advanced glycation end-products (RAGE) is a receptor of pro-inflammatory ligands that has been supported to be implied in the progression of multiple lung diseases. This study attempts to delineate the specific effects of PM2.5 on human bronchial epithelial 16HBE cells in vitro and figure out whether PM2.5 functions via mediating oxidative stress and RAGE. In PM2.5-challenged 16HBE cells, MTT assay detected cell viability. ELISA estimated inflammatory levels. Flow cytometry analysis measured ROS activity and related assay kits examined oxidative stress levels. Western blot tested nuclear factor E2-related factor 2 (Nrf2), RAGE, β-catenin, and mucin 5AC (MUC5AC) expression. Immunofluorescence staining evaluated nuclear translocation of β-catenin. It was noticed that PM2.5 exposure exacerbated inflammatory response, oxidative stress, and mucus production. Additionally, PM2.5 elevated RAGE expression while declined Nrf2 expression as well as stimulated the nuclear translocation of β-catenin. Furthermore, RAGE inhibition or nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor VAS2870 mitigated inflammatory response, oxidative stress, and mucus generation in PM2.5-exposed 16HBE cells. In addition, RAGE inhibition or VAS2870 raised Nrf2 expression, reduced RAGE expression, and hampered β-catenin nuclear translocation. Briefly, PM2.5 might act as a leading driver of inflammatory response and mucus production in lung injury, the mechanism of which might be related to the activation of oxidative stress and the up-regulation of RAGE.
Collapse
Affiliation(s)
- Huishan Han
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of General Practice, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Xianru Peng
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Minyu Huang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenqu Zhao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuluan Yang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zihan Lan
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Haijin Zhao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
3
|
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
|
4
|
Meshanni JA, Lee JM, Vayas KN, Sun R, Jiang C, Guo GL, Gow AJ, Laskin JD, Laskin DL. Suppression of Lung Oxidative Stress, Inflammation, and Fibrosis following Nitrogen Mustard Exposure by the Selective Farnesoid X Receptor Agonist Obeticholic Acid. J Pharmacol Exp Ther 2024; 388:586-595. [PMID: 37188530 PMCID: PMC10801770 DOI: 10.1124/jpet.123.001557] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/26/2023] [Accepted: 04/22/2023] [Indexed: 05/17/2023] Open
Abstract
Nitrogen mustard (NM) is a cytotoxic vesicant known to cause pulmonary injury that can progress to fibrosis. NM toxicity is associated with an influx of inflammatory macrophages in the lung. Farnesoid X receptor (FXR) is a nuclear receptor involved in bile acid and lipid homeostasis that has anti-inflammatory activity. In these studies, we analyzed the effects of FXR activation on lung injury, oxidative stress, and fibrosis induced by NM. Male Wistar rats were exposed to phosphate-buffered saline (vehicle control) or NM (0.125 mg/kg) by intratracheal Penncentury-MicroSprayer aerosolization; this was followed by treatment with the FXR synthetic agonist, obeticholic acid (OCA, 15 mg/kg), or vehicle control (0.13-0.18 g peanut butter) 2 hours later and then once per day, 5 days per week thereafter for 28 days. NM caused histopathological changes in the lung, including epithelial thickening, alveolar circularization, and pulmonary edema. Picrosirius red staining and lung hydroxyproline content were increased, indicative of fibrosis; foamy lipid-laden macrophages were also identified in the lung. This was associated with aberrations in pulmonary function, including increases in resistance and hysteresis. Following NM exposure, lung expression of HO-1 and iNOS, and the ratio of nitrates/nitrites in bronchoalveolar lavage fluid (BAL), markers of oxidative stress increased, along with BAL levels of inflammatory proteins, fibrinogen, and sRAGE. Administration of OCA attenuated NM-induced histopathology, oxidative stress, inflammation, and altered lung function. These findings demonstrate that FXR plays a role in limiting NM-induced lung injury and chronic disease, suggesting that activating FXR may represent an effective approach to limiting NM-induced toxicity. SIGNIFICANCE STATEMENT: In this study, the role of farnesoid-X-receptor (FXR) in mustard vesicant-induced pulmonary toxicity was analyzed using nitrogen mustard (NM) as a model. This study's findings that administration of obeticholic acid, an FXR agonist, to rats reduces NM-induced pulmonary injury, oxidative stress, and fibrosis provide novel mechanistic insights into vesicant toxicity, which may be useful in the development of efficacious therapeutics.
Collapse
Affiliation(s)
- Jaclynn A Meshanni
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Jordan M Lee
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Kinal N Vayas
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Rachel Sun
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Chenghui Jiang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Grace L Guo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Jeffrey D Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| |
Collapse
|
5
|
Gao J, Zhang Z, Yan JY, Ge YX, Gao Y. Inflammation and coagulation abnormalities via the activation of the HMGB1‑RAGE/NF‑κB and F2/Rho pathways in lung injury induced by acute hypoxia. Int J Mol Med 2023; 52:67. [PMID: 37350396 PMCID: PMC10555482 DOI: 10.3892/ijmm.2023.5270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/15/2023] [Indexed: 06/24/2023] Open
Abstract
High‑altitude acute hypoxia is commonly associated with respiratory cardiovascular diseases. The inability to adapt to acute hypoxia may lead to cardiovascular dysfunction, lung injury and even death. Therefore, understanding the molecular basis of the adaptation to high‑altitude acute hypoxia may reveal novel therapeutic approaches with which to counteract the detrimental consequences of hypoxia. In the present study, a high‑altitude environment was simulated in a rat model in order to investigate the role of the high mobility group protein‑1 (HMGB1)/receptor for advanced glycation end products (RAGE)/NF‑κB and F2/Rho signaling pathways in lung injury induced by acute hypoxia. It was found that acute hypoxia caused inflammation through the HMGB1/RAGE/NF‑κB pathway and coagulation dysfunction through the F2/Rho pathway, both of which may be key processes in acute hypoxia‑induced lung injury. The present study provides new insight into the molecular basis of lung injury induced by acute hypoxia. The simultaneous activation of the HMGB1/RAGE/NF‑κB and F2/Rho signaling pathways plays a critical role in hypoxia‑induced inflammatory responses and coagulation abnormalities, and provides a theoretical basis for the development of potential therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Jia-Yi Yan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Yun-Xuan Ge
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| |
Collapse
|
6
|
Cha SR, Jang J, Park SM, Ryu SM, Cho SJ, Yang SR. Cigarette Smoke-Induced Respiratory Response: Insights into Cellular Processes and Biomarkers. Antioxidants (Basel) 2023; 12:1210. [PMID: 37371940 DOI: 10.3390/antiox12061210] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Cigarette smoke (CS) poses a significant risk factor for respiratory, vascular, and organ diseases owing to its high content of harmful chemicals and reactive oxygen species (ROS). These substances are known to induce oxidative stress, inflammation, apoptosis, and senescence due to their exposure to environmental pollutants and the presence of oxidative enzymes. The lung is particularly susceptible to oxidative stress. Persistent oxidative stress caused by chronic exposure to CS can lead to respiratory diseases such as chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), and lung cancer. Avoiding exposure to environmental pollutants, like cigarette smoke and air pollution, can help mitigate oxidative stress. A comprehensive understanding of oxidative stress and its impact on the lungs requires future research. This includes identifying strategies for preventing and treating lung diseases as well as investigating the underlying mechanisms behind oxidative stress. Thus, this review aims to investigate the cellular processes induced by CS, specifically inflammation, apoptosis, senescence, and their associated biomarkers. Furthermore, this review will delve into the alveolar response provoked by CS, emphasizing the roles of potential therapeutic target markers and strategies in inflammation and oxidative stress.
Collapse
Affiliation(s)
- Sang-Ryul Cha
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon 24341, Republic of Korea
| | - Jimin Jang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon 24341, Republic of Korea
| | - Sung-Min Park
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon 24341, Republic of Korea
| | - Se Min Ryu
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon 24341, Republic of Korea
| | - Seong-Joon Cho
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon 24341, Republic of Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon 24341, Republic of Korea
| |
Collapse
|
7
|
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: 13.0] [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
|
8
|
Malaviya R, Gardner CR, Rancourt RC, Smith LC, Abramova EV, Vayas KN, Gow AJ, Laskin JD, Laskin DL. Lung injury and oxidative stress induced by inhaled chlorine in mice is associated with proinflammatory activation of macrophages and altered bioenergetics. Toxicol Appl Pharmacol 2023; 461:116388. [PMID: 36690086 PMCID: PMC9960611 DOI: 10.1016/j.taap.2023.116388] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
Abstract
Chlorine (Cl2) gas is a highly toxic and oxidizing irritant that causes life-threatening lung injuries. Herein, we investigated the impact of Cl2-induced injury and oxidative stress on lung macrophage phenotype and function. Spontaneously breathing male C57BL/6J mice were exposed to air or Cl2 (300 ppm, 25 min) in a whole-body exposure chamber. Bronchoalveolar lavage (BAL) fluid and cells, and lung tissue were collected 24 h later and analyzed for markers of injury, oxidative stress and macrophage activation. Exposure of mice to Cl2 resulted in increases in numbers of BAL cells and levels of IgM, total protein, and fibrinogen, indicating alveolar epithelial barrier dysfunction and inflammation. BAL levels of inflammatory proteins including surfactant protein (SP)-D, soluble receptor for glycation end product (sRAGE) and matrix metalloproteinase (MMP)-9 were also increased. Cl2 inhalation resulted in upregulation of phospho-histone H2A.X, a marker of double-strand DNA breaks in the bronchiolar epithelium and alveolar cells; oxidative stress proteins, heme oxygenase (HO)-1 and catalase were also upregulated. Flow cytometric analysis of BAL cells revealed increases in proinflammatory macrophages following Cl2 exposure, whereas numbers of resident and antiinflammatory macrophages were not altered. This was associated with increases in numbers of macrophages expressing cyclooxygenase (COX)-2 and inducible nitric oxide synthase (iNOS), markers of proinflammatory activation, with no effect on mannose receptor (MR) or Ym-1 expression, markers of antiinflammatory activation. Metabolic analysis of lung cells showed increases in glycolytic activity following Cl2 exposure in line with proinflammatory macrophage activation. Mechanistic understanding of Cl2-induced injury will be useful in the identification of efficacious countermeasures for mitigating morbidity and mortality of this highly toxic gas.
Collapse
Affiliation(s)
- Rama Malaviya
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, USA
| | - Carol R Gardner
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, USA
| | - Raymond C Rancourt
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, USA
| | - Ley Cody Smith
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, USA
| | - Elena V Abramova
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, USA
| | - Kinal N Vayas
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, USA
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, USA
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health and Justice, School of Public Health, Rutgers University, Piscataway, NJ 08854, USA
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, USA.
| |
Collapse
|
9
|
Kwon OS, Decker ST, Zhao J, Hoidal JR, Heuckstadt T, Sanders KA, Richardson RS, Layec G. The receptor for advanced glycation end products (RAGE) is involved in mitochondrial function and cigarette smoke-induced oxidative stress. Free Radic Biol Med 2023; 195:261-269. [PMID: 36586455 DOI: 10.1016/j.freeradbiomed.2022.12.089] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/14/2022] [Accepted: 12/24/2022] [Indexed: 12/30/2022]
Abstract
The mechanisms underlying muscle dysfunction with Chronic Obstructive Pulmonary Disease (COPD) are poorly understood. Indirect evidence has recently suggested a role of Advanced Glycation End Products (AGEs) and their receptor (RAGE) in the pathophysiology of COPD. Accordingly, this study aimed to examine the redox balance and mitochondrial alterations in the skeletal muscle of a mouse model deficient in the receptor for AGE (RAGE-KO) and wild-type C57BL/6 exposed to cigarette smoke for 8-months using immunoblotting, spectrophotometry, and high-resolution respirometry. Cigarette smoke exposure increased by two-fold 4-HNE levels (P < 0.001), a marker of oxidative stress, and markedly downregulated contractile proteins, mitochondrial respiratory complexes, and uncoupling proteins levels (P < 0.001). Functional alterations with cigarette smoke exposure included a greater reliance on complex-I supported respiration (P < 0.01) and lower relative respiratory capacity for fatty acid (P < 0.05). RAGE knockout resulted in 47% lower 4-HNE protein levels than the corresponding WT control mice exposed to cigarette smoke (P < 0.05), which was partly attributed to increased Complex III protein levels. Independent of cigarette smoke exposure, RAGE KO decreased mitochondrial specific maximal respiration (P < 0.05), resulting in a compensatory increase in mitochondrial content measured by citrate synthase activity (P < 0.001) such that muscle respiratory capacity remained unaltered. Together, these findings suggest that knockout of RAGE protected the skeletal muscle against oxidative damage induced by 8 months of cigarette smoke exposure. In addition, this study supports a role for RAGE in regulating mitochondrial content and function and can thus serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Oh Sung Kwon
- Department of Kinesiology, University of Connecticut, Storrs, CT, USA; UConn Center on Aging and Department of Orthopaedic Surgery, University of Connecticut, School of Medicine, Farmington, CT, USA; Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, UT, USA; Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA
| | - Stephen T Decker
- Department of Kinesiology, University of Massachusetts Amherst, USA
| | - Jia Zhao
- Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, UT, USA; Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA
| | - John R Hoidal
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, USA; Department of Internal Medicine, Pulmonary Division, University of Utah, Salt Lake City, UT, USA; Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT, USA
| | - Thomas Heuckstadt
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, USA; Department of Internal Medicine, Pulmonary Division, University of Utah, Salt Lake City, UT, USA; Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT, USA
| | - Karl A Sanders
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, USA; Department of Internal Medicine, Pulmonary Division, University of Utah, Salt Lake City, UT, USA; Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT, USA
| | - Russell S Richardson
- Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, UT, USA; Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA; Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Gwenael Layec
- Department of Kinesiology, University of Massachusetts Amherst, USA; Institute of Applied Life Science, University of Massachusetts Amherst, USA.
| |
Collapse
|
10
|
Wismans LV, Lopuhaä B, de Koning W, Moeniralam H, van Oosterhout M, Ambarus C, Hofman FN, Kuiken T, Endeman H, Mustafa DAM, von der Thüsen JH. Increase of mast cells in COVID-19 pneumonia may contribute to pulmonary fibrosis and thrombosis. Histopathology 2023; 82:407-419. [PMID: 36366933 PMCID: PMC9877713 DOI: 10.1111/his.14838] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/10/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022]
Abstract
AIMS Lung tissue from COVID-19 patients shares similar histomorphological features with chronic lung allograft disease, also suggesting activation of autoimmune-related pathways in COVID-19. To more clearly understand the underlying spectrum of pathophysiology in COVID-19 pneumonia, we analysed mRNA expression of autoimmune-related genes in post-mortem lung tissue from COVID-19 patients. METHODS AND RESULTS Formalin-fixed, paraffin-embedded lung tissue samples of 18 COVID-19 patients and eight influenza patients were used for targeted gene expression profiling using NanoString technology. Multiplex immunofluorescence for tryptase and chymase was applied for validation. Genes related to mast cells were significantly increased in COVID-19. This finding was strengthened by multiplex immunofluorescence also showing a significant increase of tryptase- and chymase-positive cells in COVID-19. Furthermore, receptors for advanced glycation end-products (RAGE) and pro-platelet basic protein (PPBP) were up-regulated in COVID-19 compared to influenza. Genes associated with Type I interferon signalling showed a significant correlation to detected SARS-CoV2 pathway-related genes. The comparison of lung tissue samples from both groups based on the presence of histomorphological features indicative of acute respiratory distress syndrome did not result in finding any specific gene or pathways. CONCLUSION Two separate means of measuring show a significant increase of mast cells in SARS-CoV-2-infected lung tissue compared to influenza. Additionally, several genes involved in fibrosis and thrombosis, among which are RAGE and PPBP, are up-regulated in COVID-19. As mast cells are able to induce thrombosis and fibrosis, they may play an important role in the pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Leonoor V Wismans
- Present address:
Department of SurgeryErasmus Medical CenterRotterdamthe Netherlands,The Tumor Immuno‐Pathology Laboratory, Department of PathologyJosephine Nefkens Institute, Erasmus Medical CenterRotterdamthe Netherlands
| | - Boaz Lopuhaä
- Present address:
Department of SurgeryErasmus Medical CenterRotterdamthe Netherlands,Department of PathologyJosephine Nefkens Institute, Erasmus Medical CenterRotterdamthe Netherlands
| | - Willem de Koning
- The Tumor Immuno‐Pathology Laboratory, Department of PathologyJosephine Nefkens Institute, Erasmus Medical CenterRotterdamthe Netherlands,Clinical Bioinformatics Unit, Department of PathologyErasmus Medical CenterRotterdamthe Netherlands
| | - Hazra Moeniralam
- Department of Internal Medicine and Intensive CareSt. Antonius HospitalNieuwegeinthe Netherlands
| | | | - Carmen Ambarus
- Department of Pathology DNASt. Antonius HospitalNieuwegeinthe Netherlands
| | - Frederik N Hofman
- Department of Cardiothoracic SurgerySt. Antonius HospitalNieuwegeinthe Netherlands
| | - Thijs Kuiken
- Department of ViroscienceErasmus Medical CenterRotterdamthe Netherlands
| | - Henrik Endeman
- Department of Adult Intensive CareErasmus Medical CenterRotterdamthe Netherlands
| | - Dana A M Mustafa
- The Tumor Immuno‐Pathology Laboratory, Department of PathologyJosephine Nefkens Institute, Erasmus Medical CenterRotterdamthe Netherlands,Department of PathologyJosephine Nefkens Institute, Erasmus Medical CenterRotterdamthe Netherlands
| | - Jan H von der Thüsen
- Department of PathologyJosephine Nefkens Institute, Erasmus Medical CenterRotterdamthe Netherlands
| |
Collapse
|
11
|
Lin H, Li H. How does cigarette smoking affect airway remodeling in asthmatics? Tob Induc Dis 2023; 21:13. [PMID: 36741543 PMCID: PMC9881586 DOI: 10.18332/tid/156047] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 03/07/2022] [Accepted: 10/25/2022] [Indexed: 01/30/2023] Open
Abstract
Asthma is a prevalent chronic airway inflammatory disease involving multiple cells, and the prolonged course of the disease can cause airway remodeling, resulting in irreversible or partial irreversible airflow limitation and persistent airway hyperresponsiveness (AHR) in asthmatics. Therefore, we must ascertain the factors that affect the occurrence and development of airway remodeling in asthmatics. Smokers are not uncommon in asthmatics. However, there is no systematic description of how smoking promotes airway remodeling in asthmatics. This narrative review summarizes the effects of smoking on airway remodeling in asthmatics, and the progress of the methods for evaluating airway remodeling.
Collapse
Affiliation(s)
- Huihui Lin
- Department of Respiratory Diseases, The First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Zhejiang, China
| | - Hequan Li
- Department of Respiratory Diseases, The First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Zhejiang, China
| |
Collapse
|
12
|
Magee N, Ahamed F, Eppler N, Jones E, Ghosh P, He L, Zhang Y. Hepatic transcriptome profiling reveals early signatures associated with disease transition from non-alcoholic steatosis to steatohepatitis. LIVER RESEARCH 2022; 6:238-250. [PMID: 36864891 PMCID: PMC9977163 DOI: 10.1016/j.livres.2022.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background and aim Non-alcoholic fatty liver disease (NAFLD) is becoming a leading cause of chronic liver disease worldwide. The molecular events that influence disease progression from non-alcoholic fatty liver (NAFL) to aggressive non-alcoholic steatohepatitis (NASH) remain incompletely understood, leading to lack of mechanism-based targeted treatment options for NASH. This study aims to identify early signatures associated with disease progression from NAFL to NASH in mice and humans. Materials and methods Male C57BL/6J mice were fed a high-fat, -cholesterol, and - fructose (HFCF) diet for up to 9 months. The extent of steatosis, inflammation, and fibrosis was evaluated in liver tissues. Total RNA sequencing (RNA-seq) was conducted to determine liver transcriptomic changes. Results After being fed the HFCF diet, mice sequentially developed steatosis, early steatohepatitis, steatohepatitis with fibrosis, and eventually spontaneous liver tumor. Hepatic RNA-seq revealed that the key signatures during steatosis progression to early steatohepatitis were pathways related to extracellular matrix organization and immune responses such as T cell migration, arginine biosynthesis, C-type lectin receptor signaling, and cytokine-cytokine receptor interaction. Genes regulated by transcription factors forkhead box M1 (FOXM1) and negative elongation factor complex member E (NELFE) were significantly altered during disease progression. This phenomenon was also observed in patients with NASH. Conclusions In summary, we identified early signatures associated with disease progression from NAFL to early NASH in a mouse model that recapitulated key metabolic, histologic, and transcriptomic changes seen in humans. The findings from our study may shed light on the development of novel preventative, diagnostic, and therapeutic strategies for NASH.
Collapse
Affiliation(s)
- Nancy Magee
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Forkan Ahamed
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Natalie Eppler
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Elizabeth Jones
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Priyanka Ghosh
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Lily He
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
- Liver Center, University of Kansas, Kansas City, KS, USA
| |
Collapse
|
13
|
Mumby S, Adcock IM. Recent evidence from omic analysis for redox signalling and mitochondrial oxidative stress in COPD. J Inflamm (Lond) 2022; 19:10. [PMID: 35820851 PMCID: PMC9277949 DOI: 10.1186/s12950-022-00308-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/27/2022] [Indexed: 11/30/2022] Open
Abstract
COPD is driven by exogenous and endogenous oxidative stress derived from inhaled cigarette smoke, air pollution and reactive oxygen species from dysregulated mitochondria in activated inflammatory cells within the airway and lung. This is compounded by the loss in antioxidant defences including FOXO and NRF2 and other antioxidant transcription factors together with various key enzymes that attenuate oxidant effects. Oxidative stress enhances inflammation; airway remodelling including fibrosis and emphysema; post-translational protein modifications leading to autoantibody generation; DNA damage and cellular senescence. Recent studies using various omics technologies in the airways, lungs and blood of COPD patients has emphasised the importance of oxidative stress, particularly that derived from dysfunctional mitochondria in COPD and its role in immunity, inflammation, mucosal barrier function and infection. Therapeutic interventions targeting oxidative stress should overcome the deleterious pathologic effects of COPD if targeted to the lung. We require novel, more efficacious antioxidant COPD treatments among which mitochondria-targeted antioxidants and Nrf2 activators are promising.
Collapse
|
14
|
Lin L, Li J, Song Q, Cheng W, Chen P. The role of HMGB1/RAGE/TLR4 signaling pathways in cigarette smoke-induced inflammation in chronic obstructive pulmonary disease. Immun Inflamm Dis 2022; 10:e711. [PMID: 36301039 PMCID: PMC9552978 DOI: 10.1002/iid3.711] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/10/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common chronic respiratory disease with irreversible and continuous progression. It has become the fifth most burdensome disease and the third most deadly disease globally. Therefore, the prevention and treatment of COPD are urgent, and it is also important to clarify the pathogenesis of it. Smoking is the main and most common risk factor for COPD. Cigarette smoke (CS) can cause lung inflammation and other pathological mechanisms in the airways and lung tissue. Airway inflammation is one of the important mechanisms leading to the pathogenesis of COPD. Recent studies have shown that high mobility group box 1 (HMGB1) is involved in the occurrence and development of respiratory diseases, including COPD. HMGB1 is a typical damage-associated molecular pattern (DAMP) protein, which mainly exerts its activity by binding to the receptor for advanced glycation end products (RAGE) and toll-like receptor 4 (TLR4) and further participate in the process of airway inflammation. Studies have shown that the abnormal expression of HMGB1, RAGE, and TLR4 are related to inflammation in COPD. Herein, we discuss the roles of HMGB1, RAGE, and TLR4 in CS/cigarette smoke extract-induced inflammation in COPD, providing a new target for the diagnosis, treatment and prevention of COPD.
Collapse
Affiliation(s)
- Ling Lin
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Jing Li
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Qing Song
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Wei Cheng
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Ping Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| |
Collapse
|
15
|
Palzer EF, Wendt CH, Bowler RP, Hersh CP, Safo SE, Lock EF. sJIVE: Supervised Joint and Individual Variation Explained. Comput Stat Data Anal 2022; 175:107547. [PMID: 36119152 PMCID: PMC9481062 DOI: 10.1016/j.csda.2022.107547] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Analyzing multi-source data, which are multiple views of data on the same subjects, has become increasingly common in molecular biomedical research. Recent methods have sought to uncover underlying structure and relationships within and/or between the data sources, and other methods have sought to build a predictive model for an outcome using all sources. However, existing methods that do both are presently limited because they either (1) only consider data structure shared by all datasets while ignoring structures unique to each source, or (2) they extract underlying structures first without consideration to the outcome. The proposed method, supervised joint and individual variation explained (sJIVE), can simultaneously (1) identify shared (joint) and source-specific (individual) underlying structure and (2) build a linear prediction model for an outcome using these structures. These two components are weighted to compromise between explaining variation in the multi-source data and in the outcome. Simulations show sJIVE to outperform existing methods when large amounts of noise are present in the multi-source data. An application to data from the COPDGene study explores gene expression and proteomic patterns associated with lung function.
Collapse
Affiliation(s)
- Elise F. Palzer
- Division of Biostatistics, University of Minnesota, Minneapolis, 55455, USA
| | - Christine H. Wendt
- Division of Pulmonary, Allergy and Critical Care, University of Minnesota, Minneapolis, 55455, USA
| | - Russell P. Bowler
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Craig P. Hersh
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sandra E. Safo
- Division of Biostatistics, University of Minnesota, Minneapolis, 55455, USA
| | - Eric F. Lock
- Division of Biostatistics, University of Minnesota, Minneapolis, 55455, USA
| |
Collapse
|
16
|
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
|
17
|
Buscetta M, Cristaldi M, Cimino M, La Mensa A, Dino P, Bucchieri F, Rappa F, Amato S, Aronica TS, Pace E, Bertani A, Cipollina C. Cigarette smoke promotes inflammasome-independent activation of caspase-1 and -4 leading to gasdermin D cleavage in human macrophages. FASEB J 2022; 36:e22525. [PMID: 36004615 DOI: 10.1096/fj.202200837r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/02/2022] [Accepted: 08/17/2022] [Indexed: 11/11/2022]
Abstract
Mechanisms and consequences of gasdermin D (GSDMD) activation in cigarette smoke (CS)-associated inflammation and lung disease are unknown. GSDMD is a downstream effector of caspase-1, -8, and -4. Upon cleavage, GSDMD generates pores into cell membranes. Different degrees of GSDMD activation are associated with a range of physiological outputs ranging from cell hyperactivation to pyroptosis. We have previously reported that in human monocyte-derived macrophages CS extract (CSE) inhibits the NLRP3 inflammasome and shifts the response to lipopolysaccharide (LPS) towards the TLR4-TRIF axis leading to activation of caspase-8, which, in turn, activates caspase-1. In the present work, we investigated whether other ASC-dependent inflammasomes could be involved in caspase activation by CSE and whether caspase activation led to GSDMD cleavage and other downstream effects. Presented results demonstrate that CSE promoted ASC-independent activation of caspase-1 leading to GSDMD cleavage and increased cell permeability, in the absence of cell death. GSDMD cleavage was strongly enhanced upon stimulation with LPS+CSE, suggesting a synergistic effect between the two stimuli. Noteworthy, CSE promoted LPS internalization leading to caspase-4 activation, thus contributing to increased GSDMD cleavage. Caspase-dependent GSDMD cleavage was associated with mitochondrial superoxide generation. Increased cleaved GSDMD was found in lung macrophages of smokers compared to ex-smokers and non-smoking controls. Our findings revealed that ASC-independent activation of caspase-1, -4, and -8 and GSDMD cleavage upon exposure to CS may contribute to macrophage dysfunction and feed the chronic inflammation observed in the smokers' lung.
Collapse
Affiliation(s)
| | | | | | - Agnese La Mensa
- Fondazione RiMED, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Paola Dino
- Fondazione RiMED, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Fabio Bucchieri
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Francesca Rappa
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Santina Amato
- Azienda di Rilievo Nazionale ed Alta Specializzazione Ospedali (A.R.N.A.S) "Civico Di Cristina Benfratelli", Palermo, Italy
| | - Tommaso Silvano Aronica
- Azienda di Rilievo Nazionale ed Alta Specializzazione Ospedali (A.R.N.A.S) "Civico Di Cristina Benfratelli", Palermo, Italy
| | - Elisabetta Pace
- Istituto di Farmacologia Traslazionale (IFT)-CNR, Palermo, Italy
| | | | - Chiara Cipollina
- Fondazione RiMED, Palermo, Italy.,Istituto di Farmacologia Traslazionale (IFT)-CNR, Palermo, Italy
| |
Collapse
|
18
|
D’Erminio DN, Krishnamoorthy D, Lai A, Hoy RC, Natelson DM, Poeran J, Torres A, Laudier DM, Nasser P, Vashishth D, Illien-Jünger S, Iatridis JC. High fat diet causes inferior vertebral structure and function without disc degeneration in RAGE-KO mice. J Orthop Res 2022; 40:1672-1686. [PMID: 34676612 PMCID: PMC9021327 DOI: 10.1002/jor.25191] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 08/12/2021] [Accepted: 09/30/2021] [Indexed: 02/04/2023]
Abstract
Back pain and spinal pathologies are associated with obesity in juveniles and adults, yet studies identifying causal relationships are lacking and none investigate sex differences. This study determined if high fat (HF) diet causes structural and functional changes to vertebrae and intervertebral discs (IVDs); if these changes are modulated in mice with systematic ablation for the receptor for advanced glycation endproducts (RAGE-KO); and if these changes are sex-dependent. Wild-type (WT) and RAGE-KO mice were fed a low fat (LF) or HF diet for 12 weeks starting at 6 weeks, representing the juvenile population. HF diet led to weight/fat gain, glucose intolerance, and increased cytokine levels (IL-5, MIG, and RANTES); with less fat gain in RAGE-KO females. Most importantly, HF diet reduced vertebral trabecular bone volume fraction and compressive and shear moduli, without a modifying effect of RAGE-KO, but with a more pronounced effect in females. HF diet caused reduced cortical area fraction only in WT males. Neither HF diet nor RAGE-KO affected IVD degeneration grade. Biomechanical properties of coccygeal motion segments were affected by RAGE-KO but not diet, with some interactions identified. In conclusion, HF diet resulted in inferior vertebral structure and function with some sex differences, no IVD degeneration, and few modifying effects of RAGE-KO. These structural and functional deficiencies with HF diet provide further evidence that diet can affect spinal structures and may increase the risk for spinal injury and degeneration with aging and additional stressors. Back pain and spinal pathologies are associated with obesity in juveniles and adults, yet studies identifying causal relationships are lacking and none investigate sex differences.
Collapse
Affiliation(s)
- Danielle N D’Erminio
- Leni & Peter W. May Dept. of Orthopaedics, Mount Sinai Health System, New York, NY
- Dept. of Biomedical Engineering, The City College of New York at CUNY, NY, NY
| | - Divya Krishnamoorthy
- Leni & Peter W. May Dept. of Orthopaedics, Mount Sinai Health System, New York, NY
- 3DBio Therapeutics, New York, NY
| | - Alon Lai
- Leni & Peter W. May Dept. of Orthopaedics, Mount Sinai Health System, New York, NY
| | - Robert C Hoy
- Leni & Peter W. May Dept. of Orthopaedics, Mount Sinai Health System, New York, NY
| | - Devorah M Natelson
- Leni & Peter W. May Dept. of Orthopaedics, Mount Sinai Health System, New York, NY
| | - Jashvant Poeran
- Dept. of Population Health Science & Policy, and Medicine, Mount Sinai Health System, New York, NY
| | - Andrew Torres
- Leni & Peter W. May Dept. of Orthopaedics, Mount Sinai Health System, New York, NY
| | - Damien M Laudier
- Leni & Peter W. May Dept. of Orthopaedics, Mount Sinai Health System, New York, NY
| | - Philip Nasser
- Leni & Peter W. May Dept. of Orthopaedics, Mount Sinai Health System, New York, NY
| | - Deepak Vashishth
- Ctr. for Biotechnology & Interdisciplinary Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY
| | - Svenja Illien-Jünger
- Leni & Peter W. May Dept. of Orthopaedics, Mount Sinai Health System, New York, NY
- Emory University School of Medicine, Department of Orthopaedics, Atlanta, GA
| | - James C Iatridis
- Leni & Peter W. May Dept. of Orthopaedics, Mount Sinai Health System, New York, NY
| |
Collapse
|
19
|
Röhl A, Baek SH, Kachroo P, Morrow JD, Tantisira K, Silverman EK, Weiss ST, Sharma A, Glass K, DeMeo DL. Protein interaction networks provide insight into fetal origins of chronic obstructive pulmonary disease. Respir Res 2022; 23:69. [PMID: 35331221 PMCID: PMC8944072 DOI: 10.1186/s12931-022-01963-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 02/08/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a leading cause of death in adults that may have origins in early lung development. It is a complex disease, influenced by multiple factors including genetic variants and environmental factors. Maternal smoking during pregnancy may influence the risk for diseases during adulthood, potentially through epigenetic modifications including methylation. METHODS In this work, we explore the fetal origins of COPD by utilizing lung DNA methylation marks associated with in utero smoke (IUS) exposure, and evaluate the network relationships between methylomic and transcriptomic signatures associated with adult lung tissue from former smokers with and without COPD. To identify potential pathobiological mechanisms that may link fetal lung, smoke exposure and adult lung disease, we study the interactions (physical and functional) of identified genes using protein-protein interaction networks. RESULTS We build IUS-exposure and COPD modules, which identify connected subnetworks linking fetal lung smoke exposure to adult COPD. Studying the relationships and connectivity among the different modules for fetal smoke exposure and adult COPD, we identify enriched pathways, including the AGE-RAGE and focal adhesion pathways. CONCLUSIONS The modules identified in our analysis add new and potentially important insights to understanding the early life molecular perturbations related to the pathogenesis of COPD. We identify AGE-RAGE and focal adhesion as two biologically plausible pathways that may reveal lung developmental contributions to COPD. We were not only able to identify meaningful modules but were also able to study interconnections between smoke exposure and lung disease, augmenting our knowledge about the fetal origins of COPD.
Collapse
Affiliation(s)
- Annika Röhl
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Seung Han Baek
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Priyadarshini Kachroo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jarrett D Morrow
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kelan Tantisira
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Division of Pediatric Respiratory Medicine, University of California San Diego, San Diego, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Amitabh Sharma
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Center for Complex Network Research, Northeastern University, Boston, MA, USA
| | - Kimberly Glass
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
20
|
Gupta A, Burgess JK, Borghuis T, de Vries MP, Kuipers J, Permentier HP, Bischoff R, Slebos DJ, Pouwels SD. Identification of damage associated molecular patterns and extracellular matrix proteins as major constituents of the surface proteome of lung implantable silicone/nitinol devices. Acta Biomater 2022; 141:209-218. [PMID: 35038586 DOI: 10.1016/j.actbio.2022.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/23/2021] [Accepted: 01/07/2022] [Indexed: 11/15/2022]
Abstract
Lung implantable devices have been widely adopted as mechanical interventions for a wide variety of pulmonary pathologies. Despite successful initial treatment, long-term efficacy can often be impacted by fibrotic or granulation tissue formation at the implant sites. This study aimed to explore the lung-device interface by identifying the adhered proteome on lung devices explanted from patients with severe emphysema. In this study, scanning electron microscopy is used to visualize the adhesion of cells and proteins to silicone and nitinol surfaces of explanted endobronchial valves. By applying high-resolution mass-spectrometry, the surface proteome of eight explanted valves is characterized, identifying 263 unique protein species to be mutually adsorbed on the valves. This subset is subjected to gene enrichment analysis, matched with known databases and further validated using immunohistochemistry. Enrichment analyses reveal dominant clusters of functionally-related ontology terms associated with coagulation, pattern recognition receptor signaling, immune responses, cytoskeleton organization, cell adhesion and migration. Matching results show that extracellular matrix proteins and damage-associated molecular patterns are cardinal in the formation of the surface proteome. This is the first study investigating the composition of the adhered proteome on explanted lung devices, setting the groundwork for hypothesis generation and further exploration. STATEMENT OF SIGNIFICANCE: This is the first study investigating the composition of the adhered proteome on explanted lung devices. Lung implantable devices have been widely adopted as mechanical interventions for pulmonary pathologies. Despite successful initial treatment, long-term efficacy can often be impacted by fibrotic or granulation tissue formation around the implant sites. We identified the adhered proteome on explanted lung devices using several techniques. We identified 263 unique protein species to be mutually adsorbed on explanted lung devices. Pathway analyses revealed that these proteins are associated with coagulation, pattern recognition receptor signaling, immune responses, cytoskeleton organization, cell adhesion and migration. Furthermore, we identified that especially extracellular matrix proteins and damage-associated molecular patterns were cardinal in the formation of the surface proteome.
Collapse
Affiliation(s)
- Akash Gupta
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Pathology & Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology & Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands
| | - Theo Borghuis
- University of Groningen, University Medical Center Groningen, Department of Pathology & Medical Biology, Groningen, the Netherlands
| | - Marcel P de Vries
- Department of Analytical Biochemistry and Interfaculty Mass Spectrometry Center, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Jeroen Kuipers
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Hjalmar P Permentier
- Department of Analytical Biochemistry and Interfaculty Mass Spectrometry Center, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Rainer Bischoff
- Department of Analytical Biochemistry and Interfaculty Mass Spectrometry Center, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Dirk-Jan Slebos
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands
| | - Simon D Pouwels
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Pathology & Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands.
| |
Collapse
|
21
|
Sin S, Lim MN, Kim J, Bak SH, Kim WJ. Association between plasma sRAGE and emphysema according to the genotypes of AGER gene. BMC Pulm Med 2022; 22:58. [PMID: 35144588 PMCID: PMC8832795 DOI: 10.1186/s12890-022-01848-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 01/31/2022] [Indexed: 11/22/2022] Open
Abstract
Background Higher soluble receptor for advanced glycation end product (sRAGE) levels are considered to be associated with severe emphysema. However, the relationship remains uncertain when the advanced glycation end-product specific receptor (AGER) gene is involved. We aimed to analyse the association between sRAGE levels and emphysema according to the genotypes of rs2070600 in the AGER gene. Methods We genotyped rs2070600 and measured the plasma concentration of sRAGE in each participant. Emphysema was quantified based on the chest computed tomography findings. We compared sRAGE levels based on the presence or absence and severity of emphysema in each genotype. Multiple logistic and linear regression models were used for the analyses. Results A total of 436 participants were included in the study. Among them, 64.2% had chronic obstructive pulmonary disease and 34.2% had emphysema. Among the CC-genotyped participants, the sRAGE level was significantly higher in participants without emphysema than in those with emphysema (P < 0.001). In addition, sRAGE levels were negatively correlated with emphysema severity in CC-genotyped patients (r = − 0.268 P < 0.001). Multiple regression analysis revealed that sRAGE was an independent protective factor for the presence of emphysema (adjusted odds ratio, 0.24; 95% confidence interval (CI) 0.11–0.51) and severity of emphysema (β = − 3.28, 95% CI − 4.86 to − 1.70) in CC-genotyped participants. Conclusion Plasma sRAGE might be a biomarker with a protective effect on emphysema among CC-genotyped patients of rs2070600 on the AGER gene. This is important in determining the target group for the future prediction and treatment of emphysema. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-022-01848-9.
Collapse
Affiliation(s)
- Sooim Sin
- Department of Internal Medicine, School of Medicine, Kangwon National University Hospital, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Myung-Nam Lim
- Department of Internal Medicine and Environmental Health Center, School of Medicine, Kangwon National University Hospital, Kangwon National University, Chuncheon, Republic of Korea
| | - Jeeyoung Kim
- Department of Internal Medicine and Environmental Health Center, School of Medicine, Kangwon National University Hospital, Kangwon National University, Chuncheon, Republic of Korea
| | - So Hyeon Bak
- Department of Radiology, , School of Medicine, Kangwon National University Hospital, Kangwon National University, Chuncheon, Republic of Korea
| | - Woo Jin Kim
- Department of Internal Medicine, School of Medicine, Kangwon National University Hospital, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
22
|
Medeiros ML, Oliveira AL, de Oliveira MG, Mónica FZ, Antunes E. Methylglyoxal Exacerbates Lipopolysaccharide-Induced Acute Lung Injury via RAGE-Induced ROS Generation: Protective Effects of Metformin. J Inflamm Res 2021; 14:6477-6489. [PMID: 34880648 PMCID: PMC8648108 DOI: 10.2147/jir.s337115] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/15/2021] [Indexed: 01/11/2023] Open
Abstract
Purpose Methylglyoxal (MGO) is a highly reactive dicarbonyl species implicated in diabetic-associated diseases. Acute lung injury (ALI) symptoms and prognosis are worsened by diabetes and obesity. Here, we hypothesized that elevated MGO levels aggravate ALI, which can be prevented by metformin. Therefore, this study evaluated the lung inflammation in lipopolysaccharide (LPS)-exposed mice pretreated with MGO. Methods C57Bl/6 male mice treated or not with MGO for 12 weeks were intranasally instilled with LPS (30 µg) to induce ALI, and metformin (300 mg/kg) was given as gavage in the last two weeks of treatment. After 6 h, bronchoalveolar lavage fluid (BALF) and lung tissues were collected to quantify the cell infiltration, cytokine levels, reactive-oxygen species (ROS) production, and RAGE expression. Results LPS exposure markedly increased the neutrophil infiltration in BALF and lung tissue, which was accompanied by higher levels of IFN-γ, TNF-α and IL-1β compared with untreated group. MGO treatment significantly increased the airways neutrophil infiltration and mRNA expressions of TNF-α and IL-1β, whereas COX-2 expression remained unchanged. In lung tissues of LPS-exposed mice, MGO treatment significantly increased the immunostaining and mRNA expression of RAGE, and the ROS levels. Serum MGO concentration achieved after 12-week intake was 9.2-fold higher than control mice, which was normalized by metformin treatment. Metformin also reduced the inflammatory markers in response to MGO. Conclusion MGO intake potentiates the LPS-induced ALI, increases RAGE expression and ROS generation, which is normalized by metformin. MGO scavengers may be a good adjuvant therapy to reduce ALI in patients with cardiometabolic diseases.
Collapse
Affiliation(s)
- Matheus L Medeiros
- Department of Pharmacology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Akila L Oliveira
- Department of Pharmacology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Mariana G de Oliveira
- Department of Pharmacology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Fabíola Z Mónica
- Department of Pharmacology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Edson Antunes
- Department of Pharmacology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| |
Collapse
|
23
|
Esquer C, Echeagaray O, Firouzi F, Savko C, Shain G, Bose P, Rieder A, Rokaw S, Witon-Paulo A, Gude N, Sussman MA. Fundamentals of vaping-associated pulmonary injury leading to severe respiratory distress. Life Sci Alliance 2021; 5:5/2/e202101246. [PMID: 34810278 PMCID: PMC8616545 DOI: 10.26508/lsa.202101246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/29/2022] Open
Abstract
Vaping of flavored liquids has been touted as safe alternative to traditional cigarette smoking with decreased health risks. The popularity of vaping has dramatically increased over the last decade, particularly among teenagers who incorporate vaping into their daily life as a social activity. Despite widespread and increasing adoption of vaping among young adults, there is little information on long-term consequences of vaping and potential health risks. This study demonstrates vaping-induced pulmonary injury using commercial JUUL pens with flavored vape juice using an inhalation exposure murine model. Profound pathological changes to upper airway, lung tissue architecture, and cellular structure are evident within 9 wk of exposure. Marked histologic changes include increased parenchyma tissue density, cellular infiltrates proximal to airway passages, alveolar rarefaction, increased collagen deposition, and bronchial thickening with elastin fiber disruption. Transcriptional reprogramming includes significant changes to gene families coding for xenobiotic response, glycerolipid metabolic processes, and oxidative stress. Cardiac systemic output is moderately but significantly impaired with pulmonary side ventricular chamber enlargement. This vaping-induced pulmonary injury model demonstrates mechanistic underpinnings of vaping-related pathologic injury.
Collapse
Affiliation(s)
- Carolina Esquer
- San Diego State University Integrated Regenerative Research Institute and Biology Department, San Diego State University, San Diego, CA, USA
| | - Oscar Echeagaray
- San Diego State University Integrated Regenerative Research Institute and Biology Department, San Diego State University, San Diego, CA, USA
| | - Fareheh Firouzi
- San Diego State University Integrated Regenerative Research Institute and Biology Department, San Diego State University, San Diego, CA, USA
| | - Clarissa Savko
- San Diego State University Integrated Regenerative Research Institute and Biology Department, San Diego State University, San Diego, CA, USA
| | - Grant Shain
- San Diego State University Integrated Regenerative Research Institute and Biology Department, San Diego State University, San Diego, CA, USA
| | - Pria Bose
- San Diego State University Integrated Regenerative Research Institute and Biology Department, San Diego State University, San Diego, CA, USA
| | - Abigail Rieder
- San Diego State University Integrated Regenerative Research Institute and Biology Department, San Diego State University, San Diego, CA, USA
| | - Sophie Rokaw
- San Diego State University Integrated Regenerative Research Institute and Biology Department, San Diego State University, San Diego, CA, USA
| | - Andrea Witon-Paulo
- San Diego State University Integrated Regenerative Research Institute and Biology Department, San Diego State University, San Diego, CA, USA
| | - Natalie Gude
- San Diego State University Integrated Regenerative Research Institute and Biology Department, San Diego State University, San Diego, CA, USA
| | - Mark A Sussman
- San Diego State University Integrated Regenerative Research Institute and Biology Department, San Diego State University, San Diego, CA, USA
| |
Collapse
|
24
|
Malik P, Hoidal JR, Mukherjee TK. Implication of RAGE Polymorphic Variants in COPD Complication and Anti-COPD Therapeutic Potential of sRAGE. COPD 2021; 18:737-748. [PMID: 34615424 DOI: 10.1080/15412555.2021.1984417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a slowly progressive and poorly reversible airway obstruction disease. It is caused either alone or in combination of emphysema, chronic bronchitis (CB), and small airways disease. COPD is thought to be a multi-factorial disorder in which genetic susceptibility, environmental factors and tobacco exposure could be doubly or simultaneously implicated. Available medicines against COPD include anti-inflammatory drugs, such as β2-agonists and anticholinergics, which efficiently reduce airflow limitation but are unable to avert disease progression and mortality. Advanced glycation end products (AGE) and their receptors i.e. receptor for advanced glycation end products (RAGE) are some molecules that have been implicated in the complication of COPD. Several RAGE single nucleotide polymorphic (SNP) variants are produced by the mammalian cells. Based on the ethnicity some SNPs aggravate the COPD severity. Mammalian cells produce several alternative RAGE splice variants including a soluble RAGE (sRAGE) and an endogenous soluble RAGE (esRAGE). Both of these act as decoy receptor and thus may help to arrest the COPD complications. Several lines of evidences indicate a decreased level of sRAGE in the COPD subjects. One of the new strategies to reduce COPD complication may be sRAGE therapeutic administration to the COPD subjects. This comprehensive discussion sheds light on the role of RAGE and its polymorphic variants in the COPD complication along with sRAGE therapeutic significance in the COPD prevention.
Collapse
Affiliation(s)
- Parth Malik
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India
| | - John R Hoidal
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA.,Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.,George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Tapan Kumar Mukherjee
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA.,Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.,George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| |
Collapse
|
25
|
Pouwels SD, Hesse L, Wu X, Allam VSRR, van Oldeniel D, Bhiekharie LJ, Phipps S, Oliver BG, Gosens R, Sukkar MB, Heijink IH. LL-37 and HMGB1 induce alveolar damage and reduce lung tissue regeneration via RAGE. Am J Physiol Lung Cell Mol Physiol 2021; 321:L641-L652. [PMID: 34405719 DOI: 10.1152/ajplung.00138.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The receptor for advanced glycation end-products (RAGE) has been implicated in the pathophysiology of chronic obstructive pulmonary disease (COPD). However, it is still unknown whether RAGE directly contributes to alveolar epithelial damage and abnormal repair responses. We hypothesize that RAGE activation not only induces lung tissue damage but also hampers alveolar epithelial repair responses. The effects of the RAGE ligands LL-37 and HMGB1 were examined on airway inflammation and alveolar tissue damage in wild-type and RAGE-deficient mice and on lung damage and repair responses using murine precision cut lung slices (PCLS) and organoids. In addition, their effects were studied on the repair response of human alveolar epithelial A549 cells, using siRNA knockdown of RAGE and treatment with the RAGE inhibitor FPS-ZM1. We observed that intranasal installation of LL-37 and HMGB1 induces RAGE-dependent inflammation and severe alveolar tissue damage in mice within 6 h, with stronger effects in a mouse strain susceptible for emphysema compared with a nonsusceptible strain. In PCLS, RAGE inhibition reduced the recovery from elastase-induced alveolar tissue damage. In organoids, RAGE ligands reduced the organoid-forming efficiency and epithelial differentiation into pneumocyte-organoids. Finally, in A549 cells, we confirmed the role of RAGE in impaired repair responses upon exposure to LL-37. Together, our data indicate that activation of RAGE by its ligands LL-37 and HMGB1 induces acute lung tissue damage and that this impedes alveolar epithelial repair, illustrating the therapeutic potential of RAGE inhibitors for lung tissue repair in emphysema.
Collapse
Affiliation(s)
- Simon D Pouwels
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Laura Hesse
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Xinhui Wu
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Venkata Sita Rama Raju Allam
- Graduate School of Health, Faculty of Health, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Daan van Oldeniel
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Linsey J Bhiekharie
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Brian G Oliver
- Graduate School of Health, Faculty of Health, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Reinoud Gosens
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Maria B Sukkar
- Graduate School of Health, Faculty of Health, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Irene H Heijink
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
26
|
Effect of Advanced Glycation End-Products and Excessive Calorie Intake on Diet-Induced Chronic Low-Grade Inflammation Biomarkers in Murine Models. Nutrients 2021; 13:nu13093091. [PMID: 34578967 PMCID: PMC8468789 DOI: 10.3390/nu13093091] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 08/29/2021] [Accepted: 09/02/2021] [Indexed: 12/29/2022] Open
Abstract
Chronic Low-Grade Inflammation (CLGI) is a non-overt inflammatory state characterized by a continuous activation of inflammation mediators associated with metabolic diseases. It has been linked to the overconsumption of Advanced Glycation End-Products (AGEs), and/or macronutrients which lead to an increase in local and systemic pro-inflammatory biomarkers in humans and animal models. This review provides a summary of research into biomarkers of diet-induced CLGI in murine models, with a focus on AGEs and obesogenic diets, and presents the physiological effects described in the literature. Diet-induced CLGI is associated with metabolic endotoxemia, and/or gut microbiota remodeling in rodents. The mechanisms identified so far are centered on pro-inflammatory axes such as the interaction between AGEs and their main receptor AGEs (RAGE) or increased levels of lipopolysaccharide. The use of murine models has helped to elucidate the local and systemic expression of CLGI mediators. These models have enabled significant advances in identification of diet-induced CLGI biomarkers and resultant physiological effects. Some limitations on the translational (murine → humans) use of biomarkers may arise, but murine models have greatly facilitated the testing of specific dietary components. However, there remains a lack of information at the whole-organism level of organization, as well as a lack of consensus on the best biomarker for use in CLGI studies and recommendations as to future research conclude this review.
Collapse
|
27
|
Pulmonary injury and oxidative stress in rats induced by inhaled sulfur mustard is ameliorated by anti-tumor necrosis factor-α antibody. Toxicol Appl Pharmacol 2021; 428:115677. [PMID: 34390737 DOI: 10.1016/j.taap.2021.115677] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 11/20/2022]
Abstract
Sulfur mustard (SM) is a bifunctional alkylating agent that causes severe injury to the respiratory tract. This is accompanied by an accumulation of macrophages in the lung and the release of the proinflammatory cytokine, tumor necrosis factor (TNF)α. In these studies, we analyzed the effects of blocking TNFα on lung injury, inflammation and oxidative stress induced by inhaled SM. Rats were treated with SM vapor (0.4 mg/kg) or air control by intratracheal inhalation. This was followed 15-30 min later by anti-TNFα antibody (15mg/kg, i.v.) or PBS control. Animals were euthanized 3 days later. Anti-TNFα antibody was found to blunt SM-induced peribronchial edema, perivascular inflammation and alveolar plasma protein and inflammatory cell accumulation in the lung; this was associated with reduced expression of PCNA in histologic sections and decreases in BAL levels of fibrinogen. SM-induced increases in inflammatory proteins including soluble receptor for glycation end products, its ligand, high mobility group box-1, and matrix metalloproteinase-9 were also reduced by anti-TNFα antibody administration, along with increases in numbers of lung macrophages expressing TNFα, cyclooxygenase-2 and inducible nitric oxide synthase. This was correlated with reduced oxidative stress as measured by expression of heme oxygenase-1 and Ym-1. Together, these data suggest that inhibiting TNFα may represent an efficacious approach to mitigating acute lung injury, inflammatory macrophage activation, and oxidative stress induced by inhaled sulfur mustard.
Collapse
|
28
|
Decker ST, Kwon OS, Zhao J, Hoidal JR, Heuckstadt T, Richardson RS, Sanders KA, Layec G. Skeletal muscle mitochondrial adaptations induced by long-term cigarette smoke exposure. Am J Physiol Endocrinol Metab 2021; 321:E80-E89. [PMID: 34121449 PMCID: PMC8321829 DOI: 10.1152/ajpendo.00544.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/18/2022]
Abstract
Because patients with chronic obstructive pulmonary disease (COPD) are often physically inactive, it is still unclear whether the lower respiratory capacity in the locomotor muscles of these patients is due to cigarette smoking per se or is secondary to physical deconditioning. Accordingly, the purpose of this study was to examine mitochondrial alterations in the quadriceps muscle of 10 mice exposed to 8 mo of cigarette smoke, a sedentary mouse model of emphysema, and 9 control mice, using immunoblotting, spectrophotometry, and high-resolution respirometry in permeabilized muscle fibers. Mice exposed to smoke displayed a twofold increase in the oxidative stress marker, 4-HNE, (P < 0.05) compared with control mice. This was accompanied by significant decrease in protein expression of UCP3 (65%), ANT (58%), and mitochondrial complexes II-V (∼60%-75%). In contrast, maximal ADP-stimulated respiration with complex I and II substrates (CON: 23.6 ± 6.6 and SMO: 19.2 ± 8.2 ρM·mg-1·s-1) or octanoylcarnitine (CON: 21.8 ± 9.0 and SMO: 16.5 ± 6.6 ρM·mg-1·s-1) measured in permeabilized muscle fibers, as well as citrate synthase activity, were not significantly different between groups. Collectively, our findings revealed that sedentary mice exposed to cigarette smoke for 8 mo, which is typically associated with pulmonary inflammation and emphysema, exhibited a preserved mitochondrial respiratory capacity for various substrates, including fatty acid, in the skeletal muscle. However, the mitochondrial adaptations induced by cigarette smoke favored the development of chronic oxidative stress, which can indirectly contribute to augment the susceptibility to muscle fatigue and exercise intolerance.NEW & NOTEWORTHY It is unclear whether the exercise intolerance and skeletal muscle mitochondrial dysfunction observed in patients with COPD is due to cigarette smoke exposure, per se, or if they are secondary consequences to inactivity. Herein, while long-term exposure to cigarette smoke induces oxidative stress and an altered skeletal muscle phenotype, cigarette smoke does not directly contribute to mitochondrial dysfunction. With this evidence, we demonstrate the critical role of physical inactivity in cigarette smoke-related skeletal muscle dysfunction.
Collapse
Affiliation(s)
- Stephen T Decker
- Department of Kinesiology, University of Massachusetts Amherst, Amherst, Massachusetts
| | - Oh-Sung Kwon
- Department of Kinesiology, University of Connecticut, Storrs, Connecticut
- UConn Center on Aging and Department of Orthopaedic Surgery, University of Connecticut, School of Medicine, Farmington, Connecticut
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, Utah
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Jia Zhao
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, Utah
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - John R Hoidal
- Department of Internal Medicine, Pulmonary Division, University of Utah, Salt Lake City, Utah
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah
| | - Thomas Heuckstadt
- Department of Internal Medicine, Pulmonary Division, University of Utah, Salt Lake City, Utah
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah
| | - Russell S Richardson
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, Utah
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Karl A Sanders
- Department of Internal Medicine, Pulmonary Division, University of Utah, Salt Lake City, Utah
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah
| | - Gwenael Layec
- Department of Kinesiology, University of Massachusetts Amherst, Amherst, Massachusetts
- Institute of Applied Life Science, University of Massachusetts Amherst, Amherst, Massachusetts
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, Utah
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| |
Collapse
|
29
|
Chiappalupi S, Salvadori L, Vukasinovic A, Donato R, Sorci G, Riuzzi F. Targeting RAGE to prevent SARS-CoV-2-mediated multiple organ failure: Hypotheses and perspectives. Life Sci 2021; 272:119251. [PMID: 33636175 PMCID: PMC7900755 DOI: 10.1016/j.lfs.2021.119251] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 02/12/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
A novel infectious disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was detected in December 2019 and declared as a global pandemic by the World Health. Approximately 15% of patients with COVID-19 progress to severe pneumonia and eventually develop acute respiratory distress syndrome (ARDS), septic shock and/or multiple organ failure with high morbidity and mortality. Evidence points towards a determinant pathogenic role of members of the renin-angiotensin system (RAS) in mediating the susceptibility, infection, inflammatory response and parenchymal injury in lungs and other organs of COVID-19 patients. The receptor for advanced glycation end-products (RAGE), a member of the immunoglobulin superfamily, has important roles in pulmonary pathological states, including fibrosis, pneumonia and ARDS. RAGE overexpression/hyperactivation is essential to the deleterious effects of RAS in several pathological processes, including hypertension, chronic kidney and cardiovascular diseases, and diabetes, all of which are major comorbidities of SARS-CoV-2 infection. We propose RAGE as an additional molecular target in COVID-19 patients for ameliorating the multi-organ pathology induced by the virus and improving survival, also in the perspective of future infections by other coronaviruses.
Collapse
Affiliation(s)
- Sara Chiappalupi
- Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy
| | - Laura Salvadori
- Interuniversity Institute of Myology (IIM), Perugia 06132, Italy; Department of Translational Medicine, University of Piemonte Orientale, Novara 28100, Italy
| | - Aleksandra Vukasinovic
- Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy
| | - Rosario Donato
- Interuniversity Institute of Myology (IIM), Perugia 06132, Italy
| | - Guglielmo Sorci
- Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy; Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, Perugia 06132, Italy
| | - Francesca Riuzzi
- Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy.
| |
Collapse
|
30
|
Karnati S, Seimetz M, Kleefeldt F, Sonawane A, Madhusudhan T, Bachhuka A, Kosanovic D, Weissmann N, Krüger K, Ergün S. Chronic Obstructive Pulmonary Disease and the Cardiovascular System: Vascular Repair and Regeneration as a Therapeutic Target. Front Cardiovasc Med 2021; 8:649512. [PMID: 33912600 PMCID: PMC8072123 DOI: 10.3389/fcvm.2021.649512] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity and mortality worldwide and encompasses chronic bronchitis and emphysema. It has been shown that vascular wall remodeling and pulmonary hypertension (PH) can occur not only in patients with COPD but also in smokers with normal lung function, suggesting a causal role for vascular alterations in the development of emphysema. Mechanistically, abnormalities in the vasculature, such as inflammation, endothelial dysfunction, imbalances in cellular apoptosis/proliferation, and increased oxidative/nitrosative stress promote development of PH, cor pulmonale, and most probably pulmonary emphysema. Hypoxemia in the pulmonary chamber modulates the activation of key transcription factors and signaling cascades, which propagates inflammation and infiltration of neutrophils, resulting in vascular remodeling. Endothelial progenitor cells have angiogenesis capabilities, resulting in transdifferentiation of the smooth muscle cells via aberrant activation of several cytokines, growth factors, and chemokines. The vascular endothelium influences the balance between vaso-constriction and -dilation in the heart. Targeting key players affecting the vasculature might help in the development of new treatment strategies for both PH and COPD. The present review aims to summarize current knowledge about vascular alterations and production of reactive oxygen species in COPD. The present review emphasizes on the importance of the vasculature for the usually parenchyma-focused view of the pathobiology of COPD.
Collapse
Affiliation(s)
- Srikanth Karnati
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Michael Seimetz
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Florian Kleefeldt
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Avinash Sonawane
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Thati Madhusudhan
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Akash Bachhuka
- UniSA Science, Technology, Engineering and Mathematics, University of South Australia, Mawson Lakes Campus, Adelaide, SA, Australia
| | - Djuro Kosanovic
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany.,Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, University of Giessen, Giessen, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| |
Collapse
|
31
|
Allam VSRR, Faiz A, Lam M, Rathnayake SNH, Ditz B, Pouwels SD, Brandsma C, Timens W, Hiemstra PS, Tew GW, Neighbors M, Grimbaldeston M, van den Berge M, Donnelly S, Phipps S, Bourke JE, Sukkar MB. RAGE and TLR4 differentially regulate airway hyperresponsiveness: Implications for COPD. Allergy 2021; 76:1123-1135. [PMID: 32799375 DOI: 10.1111/all.14563] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 07/08/2020] [Accepted: 07/14/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND The receptor for advanced glycation end products (RAGE) and Toll-like receptor 4 (TLR4) is implicated in COPD. Although these receptors share common ligands and signalling pathways, it is not known whether they act in concert to drive pathological processes in COPD. We examined the impact of RAGE and/or TLR4 gene deficiency in a mouse model of COPD and also determined whether expression of these receptors correlates with airway neutrophilia and airway hyperresponsiveness (AHR) in COPD patients. METHODS We measured airway inflammation and AHR in wild-type, RAGE-/- , TLR4-/- and TLR4-/- RAGE-/- mice following acute exposure to cigarette smoke (CS). We also examined the impact of smoking status on AGER (encodes RAGE) and TLR4 bronchial gene expression in patients with and without COPD. Finally, we determined whether expression of these receptors correlates with airway neutrophilia and AHR in COPD patients. RESULTS RAGE-/- mice were protected against CS-induced neutrophilia and AHR. In contrast, TLR4-/- mice were not protected against CS-induced neutrophilia and had more severe CS-induced AHR. TLR4-/- RAGE-/- mice were not protected against CS-induced neutrophilia but were partially protected against CS-induced mediator release and AHR. Current smoking was associated with significantly lower AGER and TLR4 expression irrespective of COPD status, possibly reflecting negative feedback regulation. However, consistent with preclinical findings, AGER expression correlated with higher sputum neutrophil counts and more severe AHR in COPD patients. TLR4 expression did not correlate with neutrophilic inflammation or AHR. CONCLUSIONS Inhibition of RAGE but not TLR4 signalling may protect against airway neutrophilia and AHR in COPD.
Collapse
Affiliation(s)
| | - Alen Faiz
- School of Life Sciences Faculty of Science The University of Technology Sydney Ultimo NSW Australia
- Department of Pulmonary Diseases University of Groningen University Medical Center Groningen Groningen The Netherlands
- Department of Pathology and Medical Biology University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Maggie Lam
- Biomedicine Discovery Institute and Department of Pharmacology School of Biomedical Sciences Monash University Melbourne Vic. Australia
| | - Senani N. H. Rathnayake
- School of Life Sciences Faculty of Science The University of Technology Sydney Ultimo NSW Australia
| | - Benedikt Ditz
- Department of Pulmonary Diseases University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Simon D. Pouwels
- Department of Pulmonary Diseases University of Groningen University Medical Center Groningen Groningen The Netherlands
- Department of Pathology and Medical Biology University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Corry‐Anke Brandsma
- Department of Pathology and Medical Biology University of Groningen University Medical Center Groningen Groningen The Netherlands
- Groningen Research Institute for Asthma and COPD University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology University of Groningen University Medical Center Groningen Groningen The Netherlands
- Groningen Research Institute for Asthma and COPD University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Pieter S. Hiemstra
- Department of Pulmonology Leiden University Medical Center Leiden The Netherlands
| | - Gaik W. Tew
- OMNI‐Biomarker Development, Genentech Inc South San Francisco CA USA
| | | | | | - Maarten van den Berge
- Department of Pulmonary Diseases University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Sheila Donnelly
- School of Life Sciences Faculty of Science The University of Technology Sydney Ultimo NSW Australia
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute Herston Qld Australia
| | - Jane E. Bourke
- Biomedicine Discovery Institute and Department of Pharmacology School of Biomedical Sciences Monash University Melbourne Vic. Australia
| | - Maria B. Sukkar
- Graduate School of Health Faculty of Health The University of Technology Sydney Ultimo NSW Australia
| |
Collapse
|
32
|
Chukowry PS, Spittle DA, Turner AM. Small Airways Disease, Biomarkers and COPD: Where are We? Int J Chron Obstruct Pulmon Dis 2021; 16:351-365. [PMID: 33628018 PMCID: PMC7899307 DOI: 10.2147/copd.s280157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/11/2020] [Indexed: 11/23/2022] Open
Abstract
The response to treatment and progression of Chronic Obstructive Pulmonary Disease (COPD) varies significantly. Small airways disease (SAD) is being increasingly recognized as a key pathological feature of COPD. Studies have brought forward pathological evidence of small airway damage preceding the development of emphysema and the detection of obstruction using traditional spirometry. In recent years, there has been a renewed interest in the early detection of SAD and this has brought along an increased demand for physiological tests able to identify and quantify SAD. Early detection of SAD allows early targeted therapy and this suggests the potential for altering the course of disease. The aim of this article is to review the evidence available on the physiological testing of small airways. The first half will focus on the role of lung function tests such as maximum mid-expiratory flow, impulse oscillometry and lung clearance index in detecting and quantifying SAD. The role of Computed Tomography (CT) as a radiological biomarker will be discussed as well as the potential of recent CT analysis software to differentiate normal aging of the lungs to pathology. The evidence behind SAD biomarkers sourced from blood as well as biomarkers sourced from sputum and broncho-alveolar lavage (BAL) will be reviewed. This paper focuses on CC-16, sRAGE, PAI-1, MMP-9 and MMP-12.
Collapse
Affiliation(s)
- Priyamvada S Chukowry
- Respiratory Research Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Daniella A Spittle
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
| | - Alice M Turner
- Institute for Applied Health Research, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
33
|
Di Stefano A, Maniscalco M, Balbi B, Ricciardolo FLM. Oxidative and Nitrosative Stress in the Pathogenesis of Obstructive Lung Diseases of Increasing Severity. Curr Med Chem 2021; 27:7149-7158. [PMID: 32496983 DOI: 10.2174/0929867327666200604165451] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/14/2020] [Accepted: 03/16/2020] [Indexed: 11/22/2022]
Abstract
The imbalance between increased oxidative agents and antioxidant defence mechanisms is central in the pathogenesis of obstructive lung diseases such as asthma and COPD. In these patients, there are increased levels of reactive oxygen species. Superoxide anions (O2 -), Hydrogen Peroxide (H2O2) and hydroxyl radicals (•OH) are critical for the formation of further cytotoxic radicals in the bronchi and lung parenchyma. Chronic inflammation, partly induced by oxidative stress, can further increase the oxidant burden through activated phagocytic cells (neutrophils, eosinophils, macrophages), particularly in severer disease states. Antioxidants and anti-inflammatory genes are, in fact, frequently downregulated in diseased patients. Nrf2, which activates the Antioxidant Response Element (ARE) leading to upregulation of GPx, thiol metabolism-associated detoxifying enzymes (GSTs) and stressresponse genes (HO-1) are all downregulated in animal models and patients with asthma and COPD. An exaggerated production of Nitric Oxide (NO) in the presence of oxidative stress can promote the formation of oxidizing reactive nitrogen species, such as peroxynitrite (ONO2 -), leading to nitration and DNA damage, inhibition of mitochondrial respiration, protein dysfunction, and cell damage in the biological systems. Protein nitration also occurs by activation of myeloperoxidase and H2O2, promoting oxidation of nitrite (NO2 -). There is increased nitrotyrosine and myeloperoxidase in the bronchi of COPD patients, particularly in severe disease. The decreased peroxynitrite inhibitory activity found in induced sputum of COPD patients correlates with pulmonary function. Markers of protein nitration - 3- nitrotyrosine, 3-bromotyrosine, and 3-chlorotyrosine - are increased in the bronchoalveolar lavage of severe asthmatics. Targeting the oxidative, nitrosative stress and associated lung inflammation through the use of either denitration mechanisms or new drug delivery strategies for antioxidant administration could improve the treatment of these chronic disabling obstructive lung diseases.
Collapse
Affiliation(s)
- Antonino Di Stefano
- Divisione di Pneumologia e Laboratorio di Immunopatologia dell'Apparato Cardio Respiratorio, Istituti Clinici Scientifici Maugeri SpA, Societa Benefit, IRCCS, Veruno, Italy
| | - Mauro Maniscalco
- Divisione di Pneumologia, Istituti Clinici Scientifici Maugeri SpA, Societa Benefit, IRCCS, Telese, Italy
| | - Bruno Balbi
- Divisione di Pneumologia e Laboratorio di Immunopatologia dell'Apparato Cardio Respiratorio, Istituti Clinici Scientifici Maugeri SpA, Societa Benefit, IRCCS, Veruno, Italy
| | - Fabio L M Ricciardolo
- Dipartimento di Scienze Cliniche e Biologiche, AOU, San Luigi, Orbassano, Universita di Torino, Torino, Italy
| |
Collapse
|
34
|
Mukherjee TK, Malik P, Hoidal JR. Receptor for Advanced Glycation End Products (RAGE) and Its Polymorphic Variants as Predictive Diagnostic and Prognostic Markers of NSCLCs: a Perspective. Curr Oncol Rep 2021; 23:12. [PMID: 33399986 DOI: 10.1007/s11912-020-00992-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Non-small cell lung cancers (NSCLCs) account for ~ 85% of all lung cancers, and 5-year survival in Europe and the USA is ~ 13-17%. In this review, we focus on the significance of Receptor for Advanced Glycation End products (RAGE) as a diagnostic or post-therapeutic prognostic marker for various forms of NSCLCs. RECENT FINDINGS The lungs have the highest levels of basal RAGE expression in mammals. The physiologic RAGE in lungs may be involved in adhesion and spreading of AT-1 cells and maintenance of pulmonary homeostasis. However, high level expression of RAGE complicates various diseases including acute lung injury. In NSCLCs, while a number of studies report decreased RAGE expression, inferring a protective role, others suggest that RAGE expression may contribute to NSCLC pathogenesis. Genetic polymorphisms of RAGE are reportedly associated with NSCLC development and complications. RAGE and its polymorphic variants may be useful diagnostic or post-therapeutic prognostic markers of NSCLCs.
Collapse
Affiliation(s)
- Tapan K Mukherjee
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT, USA. .,Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA. .,George E. Wahlen Department of Veterans Affairs Medical Center, 500, Foothil Drive, Building#45, Salt Lake City, UT, 84148, USA.
| | - Parth Malik
- School of Chemical Sciences, Central University of Gujarat (Gandhinagar), Gandhinagar, India.,School of Nano Sciences, Central University of Gujarat (Gandhinagar), Gandhinagar, India
| | - John R Hoidal
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT, USA.,Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA.,George E. Wahlen Department of Veterans Affairs Medical Center, 500, Foothil Drive, Building#45, Salt Lake City, UT, 84148, USA
| |
Collapse
|
35
|
Sharma A, Kaur S, Sarkar M, Sarin BC, Changotra H. The AGE-RAGE Axis and RAGE Genetics in Chronic Obstructive Pulmonary Disease. Clin Rev Allergy Immunol 2020; 60:244-258. [PMID: 33170477 DOI: 10.1007/s12016-020-08815-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2020] [Indexed: 12/25/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous group of lung diseases limiting the airflow due to narrowing of airways, chronic bronchitis and emphysema that leads to difficulties in breathing. Chronic inflammation is another important characteristic of COPD which leads to immune cell infiltration and helps in the alveolar destruction. Pathology of COPD is driven by various environmental and genetic factors. COPD is mainly associated with the inhalation of toxic agents mainly the cigarette smoke. Receptor for advanced glycation end products (RAGE) has emerged as a pattern recognition receptor and is a multiligand receptor expressed moderately in various cells, tissues and highly in the lungs throughout life. RAGE recognizes various ligands produced by cigarette smoke and its role has been implicated in the pathogenesis of COPD. RAGE ligands have been reported to accumulate in the lungs of patients with COPD. RAGE is a membrane receptor but its truncated form i.e. soluble RAGE (sRAGE) mainly functions as a contender of RAGE and inhibits various RAGE dependent cell signalling. Among the various ligands of RAGE, advanced glycation end products (AGEs) are majorly linked with COPD. Accumulated AGE triggers downstream RAGE-AGE axis in COPD. Moreover, RAGE genetics has long been known to play a vital role in the pathology of various airway diseases including COPD and this gene contains an associated locus. A reliable biomarker is needed for the management of this disease. sRAGE has an inverse correlation with the RAGE showed its importance as a valuable marker in COPD. This review is focused on the role of RAGE, sRAGE, RAGE axis and RAGE genetics in COPD.
Collapse
Affiliation(s)
- Ambika Sharma
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173 234, India
| | - Sargeet Kaur
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173 234, India
| | - Malay Sarkar
- Department of Pulmonary Medicine, Indira Gandhi Medical College, Shimla, Himachal Pradesh, 171 001, India
| | - B C Sarin
- Department of Chest and TB, Sri Guru Ram Das Institute of Medical Sciences and Research, Vallah, Amritsar, 143 501, India
| | - Harish Changotra
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173 234, India.
| |
Collapse
|
36
|
Prantner D, Nallar S, Vogel SN. The role of RAGE in host pathology and crosstalk between RAGE and TLR4 in innate immune signal transduction pathways. FASEB J 2020; 34:15659-15674. [PMID: 33131091 DOI: 10.1096/fj.202002136r] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022]
Abstract
Although the innate immune receptor protein, Receptor for Advanced Glycation End products (RAGE), has been extensively studied, there has been renewed interest in RAGE for its potential role in sepsis, along with a host of other inflammatory diseases of chronic, noninfectious origin. In contrast to other innate immune receptors, for example, Toll-like receptors (TLRs), that recognize ligands derived from pathogenic organisms that are collectively known as "pathogen-associated molecular patterns" (PAMPs) or host-derived "damage-associated molecular patterns" (DAMPs), RAGE has been shown to recognize a broad collection of DAMPs exclusively. Historically, these DAMPs have been shown to be pro-inflammatory in nature. Early studies indicated that the adaptor molecule, MyD88, might be important for this change. More recent studies have explored further the mechanisms underlying this inflammatory change. Overall, the newer results have shown that there is extensive crosstalk between RAGE and TLRs. The three canonical RAGE ligands, Advanced Glycation End products (AGEs), HMGB1, and S100 proteins, have all been shown to activate both TLRs and RAGE to varying degrees in order to induce inflammation in in vitro models. As with any field that delves deeply into innate signaling, obstacles of reagent purity may be a cause of some of the discrepancies in the literature, and we have found that commercial antibodies that have been widely used exhibit a high degree of nonspecificity. Nonetheless, the weight of published evidence has led us to speculate that RAGE may be physically interacting with TLRs on the cell surface to elicit inflammation via MyD88-dependent signaling.
Collapse
Affiliation(s)
- Daniel Prantner
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Shreeram Nallar
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
37
|
Malaviya R, Abramova EV, Rancourt RC, Sunil VR, Napierala M, Weinstock D, Croutch CR, Roseman J, Tuttle R, Peters E, Casillas RP, Laskin JD, Laskin DL. Progressive Lung Injury, Inflammation, and Fibrosis in Rats Following Inhalation of Sulfur Mustard. Toxicol Sci 2020; 178:358-374. [PMID: 33002157 PMCID: PMC7751178 DOI: 10.1093/toxsci/kfaa150] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Sulfur mustard (SM) inhalation causes debilitating pulmonary injury in humans which progresses to fibrosis. Herein, we developed a rat model of SM toxicity which parallels pathological changes in the respiratory tract observed in humans. SM vapor inhalation caused dose (0.2-0.6 mg/kg)-related damage to the respiratory tract within 3 days of exposure. At 0.4-0.6 mg/kg, ulceration of the proximal bronchioles, edema and inflammation were observed, along with a proteinaceous exudate containing inflammatory cells in alveolar regions. Time course studies revealed that the pathologic response was biphasic. Thus, changes observed at 3 days post-SM were reduced at 7-16 days; this was followed by more robust aberrations at 28 days, including epithelial necrosis and hyperplasia in the distal bronchioles, thickened alveolar walls, enlarged vacuolated macrophages, and interstitial fibrosis. Histopathologic changes were correlated with biphasic increases in bronchoalveolar lavage (BAL) cell and protein content and proliferating cell nuclear antigen expression. Proinflammatory proteins receptor for advanced glycation end product (RAGE), high-mobility group box protein (HMGB)-1, and matrix metalloproteinase (MMP)-9 also increased in a biphasic manner following SM inhalation, along with surfactant protein-D (SP-D). Tumor necrosis factor (TNF)-α and inducible nitric oxide synthase (iNOS), inflammatory proteins implicated in mustard lung toxicity, and the proinflammatory/profibrotic protein, galectin (Gal)-3, were upregulated in alveolar macrophages and in bronchiolar regions at 3 and 28 days post-SM. Inflammatory changes in the lung were associated with oxidative stress, as reflected by increased expression of heme oxygenase (HO)-1. These data demonstrate a similar pathologic response to inhaled SM in rats and humans suggesting that this rodent model can be used for mechanistic studies and for the identification of efficacious therapeutics for mitigating toxicity.
Collapse
Affiliation(s)
- Rama Malaviya
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854
| | - Elena V Abramova
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854
| | - Raymond C Rancourt
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854
| | - Vasanthi R Sunil
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854
| | - Marta Napierala
- Laboratory of Environmental Research, Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland
| | - Daniel Weinstock
- Janssen Boitherapeutics, Janssen Research & Development, Spring House, Pennsylvania 19477
| | - Claire R Croutch
- Medical Research Portfolio, MRIGlobal, Kansas City, Missouri 64110
| | - Julie Roseman
- Medical Research Portfolio, MRIGlobal, Kansas City, Missouri 64110
| | - Rick Tuttle
- Medical Research Portfolio, MRIGlobal, Kansas City, Missouri 64110
| | - Eric Peters
- Medical Research Portfolio, MRIGlobal, Kansas City, Missouri 64110
| | | | - Jeffrey D Laskin
- Department of Environmental and Occupational Health, School of Public Health, Rutgers University, Piscataway, New Jersey 08854
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854,To whom correspondence should be addressed at Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Road, Piscataway, NJ 08854. E-mail:
| |
Collapse
|
38
|
Al‐Aali KA, AlHelal A, Alhamoudi N, Alhenaki AM, Javed F, Abduljabbar T. Assessment of advanced glycation end products in the peri‐implant sulcular fluid among moderate cigarette‐smokers and nonsmokers with peri‐implantitis. Clin Implant Dent Relat Res 2020; 22:380-386. [DOI: 10.1111/cid.12917] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/02/2020] [Accepted: 04/12/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Khulud A. Al‐Aali
- Department of Clinical Dental Sciences College of Dentistry, Princess Nourah Bint Abdulrahman University Ruyadh Saudi Arabia
| | - Abdulaziz AlHelal
- Department of Prosthetic Dental Science College of Dentistry, King Saud University; Research Chair for Biological Research in Dental Health, College of Dentistry Riyadh Saudi Arabia
| | - Nawaf Alhamoudi
- Department of Periodontics and Community Dentistry King Saud University Riyadh Saudi Arabia
| | - Aasem M. Alhenaki
- Department of Prosthetic Dental Science College of Dentistry, King Saud University Riyadh Saudi Arabia
| | - Fawad Javed
- Department of Periodontology, Laboratory for Periodontal‐, Implant‐, Phototherapy (LA‐PIP) School of Dental Medicine, Stony Brook University Stony Brook New York United States
- Division of Orthodontics and Dentofacial Orthopedics Eastman Institute for Oral Health, University of Rochester Rochester New York United States
| | - Tariq Abduljabbar
- Department of Prosthetic Dental Science College of Dentistry, King Saud University Riyadh Saudi Arabia
| |
Collapse
|
39
|
Magee N, Zou A, Ghosh P, Ahamed F, Delker D, Zhang Y. Disruption of hepatic small heterodimer partner induces dissociation of steatosis and inflammation in experimental nonalcoholic steatohepatitis. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49910-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
40
|
Magee N, Zou A, Ghosh P, Ahamed F, Delker D, Zhang Y. Disruption of hepatic small heterodimer partner induces dissociation of steatosis and inflammation in experimental nonalcoholic steatohepatitis. J Biol Chem 2019; 295:994-1008. [PMID: 31831621 DOI: 10.1074/jbc.ra119.010233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/23/2019] [Indexed: 12/16/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a leading cause of chronic liver disease worldwide and is characterized by steatosis, inflammation, and fibrosis. The molecular mechanisms underlying NASH development remain obscure. The nuclear receptor small heterodimer partner (Shp) plays a complex role in lipid metabolism and inflammation. Here, we sought to determine SHP's role in regulating steatosis and inflammation in NASH. Shp deletion in murine hepatocytes (Shp Hep-/-) resulted in massive infiltration of macrophages and CD4+ T cells in the liver. Shp Hep-/- mice developed reduced steatosis, but surprisingly increased hepatic inflammation and fibrosis after being fed a high-fat, -cholesterol, and -fructose (HFCF) diet. RNA-Seq analysis revealed that pathways involved in inflammation and fibrosis are significantly activated in the liver of Shp Hep-/- mice fed a chow diet. After having been fed the HFCF diet, WT mice displayed up-regulated peroxisome proliferator-activated receptor γ (Pparg) signaling in the liver; however, this response was completely abolished in the Shp Hep-/- mice. In contrast, livers of Shp Hep-/- mice had consistent NF-κB activation. To further characterize the role of Shp specifically in the transition of steatosis to NASH, mice were fed the HFCF diet for 4 weeks, followed by Shp deletion. Surprisingly, Shp deletion after steatosis development exacerbated hepatic inflammation and fibrosis without affecting liver steatosis. Together, our results indicate that, depending on NASH stage, hepatic Shp plays an opposing role in steatosis and inflammation. Mechanistically, Shp deletion in hepatocytes activated NF-κB and impaired Pparg activation, leading to the dissociation of steatosis, inflammation, and fibrosis in NASH development.
Collapse
Affiliation(s)
- Nancy Magee
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - An Zou
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Priyanka Ghosh
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Forkan Ahamed
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Don Delker
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, Utah 84108
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160 .,Liver Center, University of Kansas Medical Center, Kansas City, Kansas 66160
| |
Collapse
|
41
|
Enfermedad pulmonar obstructiva crónica y síndrome metabólico: viajar en mala compañía. Rev Clin Esp 2019; 219:492-493. [DOI: 10.1016/j.rce.2019.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 07/11/2019] [Indexed: 11/23/2022]
|
42
|
López Andreu F, López Rodríguez C. Chronic obstructive pulmonary disease and metabolic syndrome: traveling with bad companies. Rev Clin Esp 2019. [DOI: 10.1016/j.rceng.2019.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
43
|
Protein Misfolding and Endoplasmic Reticulum Stress in Chronic Lung Disease: Will Cell-Specific Targeting Be the Key to the Cure? Chest 2019; 157:1207-1220. [PMID: 31778676 DOI: 10.1016/j.chest.2019.11.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/07/2019] [Accepted: 11/10/2019] [Indexed: 12/31/2022] Open
Abstract
Chronic lung disease accounts for a significant global burden with respect to death, disability, and health-care costs. Due to the heterogeneous nature and limited treatment options for these diseases, it is imperative that the cellular and molecular mechanisms underlying the disease pathophysiology are further understood. The lung is a complex organ with a diverse cell population, and each cell type will likely have different roles in disease initiation, progression, and resolution. The effectiveness of a given therapeutic agent may depend on the net effect on each of these cell types. Over the past decade, it has been established that endoplasmic reticulum stress and the unfolded protein response are involved in the development of several chronic lung diseases. These conserved cellular pathways are important for maintaining cellular proteostasis, but their aberrant activation can result in pathology. This review discusses the current understanding of endoplasmic reticulum stress and the unfolded protein response at the cellular level in the development and progression of various chronic lung diseases. We highlight the need for increased understanding of the specific cellular contributions of unfolded protein response activation to these pathologies and suggest that the development of cell-specific targeted therapies is likely required to further decrease disease progression and to promote resolution of chronic lung disease.
Collapse
|
44
|
Shumilina J, Kusnetsova A, Tsarev A, Janse van Rensburg HC, Medvedev S, Demidchik V, Van den Ende W, Frolov A. Glycation of Plant Proteins: Regulatory Roles and Interplay with Sugar Signalling? Int J Mol Sci 2019; 20:E2366. [PMID: 31086058 PMCID: PMC6539852 DOI: 10.3390/ijms20092366] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 02/06/2023] Open
Abstract
Glycation can be defined as an array of non-enzymatic post-translational modifications of proteins formed by their interaction with reducing carbohydrates and carbonyl products of their degradation. Initial steps of this process rely on reducing sugars and result in the formation of early glycation products-Amadori and Heyns compounds via Schiff base intermediates, whereas their oxidative degradation or reactions of proteins with α-dicarbonyl compounds yield a heterogeneous group of advanced glycation end products (AGEs). These compounds accompany thermal processing of protein-containing foods and are known to impact on ageing, pathogenesis of diabetes mellitus and Alzheimer's disease in mammals. Surprisingly, despite high tissue carbohydrate contents, glycation of plant proteins was addressed only recently and its physiological role in plants is still not understood. Therefore, here we summarize and critically discuss the first steps done in the field of plant protein glycation during the last decade. We consider the main features of plant glycated proteome and discuss them in the context of characteristic metabolic background. Further, we address the possible role of protein glycation in plants and consider its probable contribution to protein degradation, methylglyoxal and sugar signalling, as well as interplay with antioxidant defense.
Collapse
Affiliation(s)
- Julia Shumilina
- Department of Biochemistry, St. Petersburg State University, Saint Petersburg 199034, Russia.
| | - Alena Kusnetsova
- Department of Biochemistry, St. Petersburg State University, Saint Petersburg 199034, Russia.
- Department of Biotechnology, St. Petersburg Chemical Pharmaceutical University, Saint Petersburg 197022, Russia.
| | - Alexander Tsarev
- Department of Biochemistry, St. Petersburg State University, Saint Petersburg 199034, Russia.
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, 06120 Halle, Germany.
| | | | - Sergei Medvedev
- Department of Plant Physiology and Biochemistry, St. Petersburg State University, Saint Petersburg 199034, Russia.
| | - Vadim Demidchik
- Department of Plant Cell Biology and Bioengineering, Belarusian State University, 220030 Minsk, Belarus.
- Department of Horticulture, Foshan University, Foshan 528231, China.
| | - Wim Van den Ende
- Laboratory of Molecular Plant Biology, KU Leuven, 3001 Leuven, Belgium.
| | - Andrej Frolov
- Department of Biochemistry, St. Petersburg State University, Saint Petersburg 199034, Russia.
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, 06120 Halle, Germany.
| |
Collapse
|