1
|
Guo W, Luo J, Zhao S, Li L, Xing W, Gao R. The critical role of RAGE in severe influenza infection: A target for control of inflammatory response in the disease. Clin Immunol 2024; 262:110178. [PMID: 38460892 DOI: 10.1016/j.clim.2024.110178] [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: 12/24/2023] [Revised: 01/30/2024] [Accepted: 02/21/2024] [Indexed: 03/11/2024]
Abstract
Controlling the excessive inflammatory response is one of the key ways to reduce the severity and mortality of severe influenza virus infections. RAGE is involved in inflammatory responses and acute lung injuries. Here, we investigated the role of RAGE and its potential application as a target for severe influenza treatment through serological correlation analysis for influenza patients, and treatment with the RAGE inhibitor FPS-ZM1 on A549 cells or mice with influenza A (H1N1) infection. The results showed high levels of RAGE were correlated with immunopathological injury and severity of influenza, and FPS-ZM1 treatment increased the viability of A549 cells with influenza A infection and decreased morbidity and mortality of influenza A virus infection in mice. The RAGE/NF-κb inflammatory signaling pathway is a major targeting pathway for FPS-ZM1 treatment in severe influenza. These findings provide further insights into the immune injury of severe influenza and a potential targeting candidate for the disease treatment.
Collapse
Affiliation(s)
- Wenhui Guo
- NHC Key Laboratory of Biosafety, NHC Key Laboratory of Medical Virology and Viral Diseases, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Junhao Luo
- NHC Key Laboratory of Biosafety, NHC Key Laboratory of Medical Virology and Viral Diseases, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Song Zhao
- NHC Key Laboratory of Biosafety, NHC Key Laboratory of Medical Virology and Viral Diseases, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Li Li
- NHC Key Laboratory of Biosafety, NHC Key Laboratory of Medical Virology and Viral Diseases, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Wenge Xing
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| | - Rongbao Gao
- NHC Key Laboratory of Biosafety, NHC Key Laboratory of Medical Virology and Viral Diseases, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| |
Collapse
|
2
|
Delrue C, Speeckaert R, Delanghe JR, Speeckaert MM. Breath of fresh air: Investigating the link between AGEs, sRAGE, and lung diseases. VITAMINS AND HORMONES 2024; 125:311-365. [PMID: 38997169 DOI: 10.1016/bs.vh.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Advanced glycation end products (AGEs) are compounds formed via non-enzymatic reactions between reducing sugars and amino acids or proteins. AGEs can accumulate in various tissues and organs and have been implicated in the development and progression of various diseases, including lung diseases. The receptor of advanced glycation end products (RAGE) is a receptor that can bind to advanced AGEs and induce several cellular processes such as inflammation and oxidative stress. Several studies have shown that both AGEs and RAGE play a role in the pathogenesis of lung diseases, such as chronic obstructive pulmonary disease, asthma, idiopathic pulmonary fibrosis, cystic fibrosis, and acute lung injury. Moreover, the soluble form of the receptor for advanced glycation end products (sRAGE) has demonstrated its ability to function as a decoy receptor, possessing beneficial characteristics such as anti-inflammatory, antioxidant, and anti-fibrotic properties. These qualities make it an encouraging focus for therapeutic intervention in managing pulmonary disorders. This review highlights the current understanding of the roles of AGEs and (s)RAGE in pulmonary diseases and their potential as biomarkers and therapeutic targets for preventing and treating these pathologies.
Collapse
Affiliation(s)
- Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium
| | | | - Joris R Delanghe
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Marijn M Speeckaert
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium; Research Foundation-Flanders (FWO), Brussels, Belgium.
| |
Collapse
|
3
|
Dong H, Sun Y, Nie L, Cui A, Zhao P, Leung WK, Wang Q. Metabolic memory: mechanisms and diseases. Signal Transduct Target Ther 2024; 9:38. [PMID: 38413567 PMCID: PMC10899265 DOI: 10.1038/s41392-024-01755-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/29/2024] Open
Abstract
Metabolic diseases and their complications impose health and economic burdens worldwide. Evidence from past experimental studies and clinical trials suggests our body may have the ability to remember the past metabolic environment, such as hyperglycemia or hyperlipidemia, thus leading to chronic inflammatory disorders and other diseases even after the elimination of these metabolic environments. The long-term effects of that aberrant metabolism on the body have been summarized as metabolic memory and are found to assume a crucial role in states of health and disease. Multiple molecular mechanisms collectively participate in metabolic memory management, resulting in different cellular alterations as well as tissue and organ dysfunctions, culminating in disease progression and even affecting offspring. The elucidation and expansion of the concept of metabolic memory provides more comprehensive insight into pathogenic mechanisms underlying metabolic diseases and complications and promises to be a new target in disease detection and management. Here, we retrace the history of relevant research on metabolic memory and summarize its salient characteristics. We provide a detailed discussion of the mechanisms by which metabolic memory may be involved in disease development at molecular, cellular, and organ levels, with emphasis on the impact of epigenetic modulations. Finally, we present some of the pivotal findings arguing in favor of targeting metabolic memory to develop therapeutic strategies for metabolic diseases and provide the latest reflections on the consequences of metabolic memory as well as their implications for human health and diseases.
Collapse
Affiliation(s)
- Hao Dong
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuezhang Sun
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lulingxiao Nie
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Aimin Cui
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Pengfei Zhao
- Periodontology and Implant Dentistry Division, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Wai Keung Leung
- Periodontology and Implant Dentistry Division, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Qi Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
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
|
5
|
Salgar S, Bolívar BE, Flanagan JM, Anum SJ, Bouchier-Hayes L. The NLRP3 inflammasome fires up heme-induced inflammation in hemolytic conditions. Transl Res 2023; 252:34-44. [PMID: 36041706 DOI: 10.1016/j.trsl.2022.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/29/2022] [Accepted: 08/21/2022] [Indexed: 01/14/2023]
Abstract
Overactive inflammatory responses are central to the pathophysiology of many hemolytic conditions including sickle cell disease. Excessive hemolysis leads to elevated serum levels of heme due to saturation of heme scavenging mechanisms. Extracellular heme has been shown to activate the NLRP3 inflammasome, leading to activation of caspase-1 and release of pro-inflammatory cytokines IL-1β and IL-18. Heme also activates the non-canonical inflammasome pathway, which may contribute to NLRP3 inflammasome formation and leads to pyroptosis, a type of inflammatory cell death. Some clinical studies indicate there is a benefit to blocking the NLRP3 inflammasome pathway in patients with sickle cell disease and other hemolytic conditions. However, a thorough understanding of the mechanisms of heme-induced inflammasome activation is needed to fully leverage this pathway for clinical benefit. This review will explore the mechanisms of heme-induced NLRP3 inflammasome activation and the role of this pathway in hemolytic conditions including sickle cell disease.
Collapse
Affiliation(s)
- Suruchi Salgar
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas
| | - Beatriz E Bolívar
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jonathan M Flanagan
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas
| | - Shaniqua J Anum
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas
| | - Lisa Bouchier-Hayes
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
6
|
Liu YD, Tang F, Li XL, Liu YF, Zhang P, Yang CL, Du T, Li H, Wang CC, Liu Y, Yang B, Duan RS. Type 2 diabetes mellitus as a possible risk factor for myasthenia gravis: a case-control study. Front Neurol 2023; 14:1125842. [PMID: 37139075 PMCID: PMC10149973 DOI: 10.3389/fneur.2023.1125842] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/21/2023] [Indexed: 05/05/2023] Open
Abstract
Background A certain number of myasthenia gravis (MG) patients clinically had type 2 diabetes mellitus (T2DM) prior to MG onset, which suggests that the onset of MG may correlate with the history of T2DM. This study aimed to examine the correlation between MG and T2DM. Methods In a single-center, retrospective, 1:5 matched case-control study, all 118 hospitalized patients with a diagnosis of MG from 8 August 2014 to 22 January 2019 were enrolled. In total, four datasets with different sources of the control group were retrieved from the electronic medical records (EMRs). Data were collected at the individual level. A conditional logistic regression analysis was used to test the risk of MG associated with T2DM. Findings The risk of MG was significantly associated with T2DM, and there were notable differences by sex and age. Whether compared to the general population, general hospitalized patients without autoimmune diseases (AIDs), or patients with other AIDs except MG, women aged over 50 years with T2DM had an increased risk of MG. The mean onset age of diabetic MG patients was more than that of the non-diabetic MG patients. Interpretation This study demonstrates that T2DM is strongly associated with the subsequent risk of MG and varies significantly by sex and age. It reveals that diabetic MG may be a unique subtype that is different from the conventional MG subgroup classification. More clinical and immunological features of diabetic MG patients need to be explored in further studies.
Collapse
Affiliation(s)
- Yu-Dong Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Fang Tang
- Center for Big Data Research in Health and Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Neuroimmunology, Jinan, China
| | - Xiao-Li Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Neuroimmunology, Jinan, China
| | - Ya-Fei Liu
- Center for Big Data Research in Health and Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Peng Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Neuroimmunology, Jinan, China
| | - Chun-Lin Yang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Tong Du
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Heng Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Neuroimmunology, Jinan, China
| | - Cong-Cong Wang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Neuroimmunology, Jinan, China
| | - Ying Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Bing Yang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Neuroimmunology, Jinan, China
| | - Rui-Sheng Duan
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Neuroimmunology, Jinan, China
- *Correspondence: Rui-Sheng Duan
| |
Collapse
|
7
|
Johnson LL, Tekabe Y, Zelonina T, Ma X, Zhang G, Goldklang M, D’Armiento J. Blocking RAGE expression after injury reduces inflammation in mouse model of acute lung injury. Respir Res 2023; 24:21. [PMID: 36670409 PMCID: PMC9852798 DOI: 10.1186/s12931-023-02324-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/11/2023] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Receptor for Advanced Glycated Endproducts (RAGE) plays a major role in the inflammatory response to infectious and toxin induced acute lung injury. We tested the hypothesis that a RAGE blocking antibody when administered after the onset of injury can reduce lung inflammation compared to control antibody. METHODS Male and female C57BL/6 (WT) mice were used. Forty-six received lipopolysaccharide (LPS) and 26 PBS by nasal instillation on day one, repeated on day three. On day 2, 36 mice receiving LPS were divided into two groups of 18, one treated with 200 μg of non-immune isotype control IgG and the second group treated with 200 μg of anti-RAGE Ab, each dose divided between IV and IP. Ten of the 46 were not treated. On day 4, before euthanasia, mice were injected with fluorescein isothiocyanate (FITC) labelled albumen. BALF and serum samples were collected as well as lung tissue for immunohistochemistry (IHC). BALF was analyzed for cell (leukocyte) counts, for FITC BALF/serum ratios indicating pulmonary vascular leak, and for cytokines/chemokines using bead based multiplex assays. Quantitative IHC was performed for MPO and RAGE. RESULTS Ten LPS mice showed minimal inflammation by all measures indicating poor delivery of LPS and were excluded from analysis leaving n = 11 in the LPS + IgG group and n = 12 in the LPS + anti-RAGE group. BALF cell counts were low in the PBS administered mice (4.9 ± 2.1 × 105/ml) and high in the LPS injured untreated mice (109 ± 34) and in the LPS + IgG mice (91 ± 54) while in comparison, LPS + anti-RAGE ab mice counts were significantly lower (51.3 ± 18 vs. LPS + IgG, P = 0.03). The BALF/serum FITC ratios were lower for the LPS + anti-RAGE mice than for the LPS + IgG mice indicating less capillary leakiness. Quantitative IHC RAGE staining was lower in the LPS + anti-RAGE ab mice than in the LPS + IgG treated mice (P = 0.02). CONCLUSIONS These results describe a four-day LPS protocol to sustain lung injury and allow for treatment and suggests that treatment aimed at blocking RAGE when given after onset of injury can reduce lung inflammation.
Collapse
Affiliation(s)
- Lynne L. Johnson
- grid.21729.3f0000000419368729Departments of Medicine, Anesthesiology, and Pathology, Columbia University, 622 West 168th St, PH 10-203, New York, NY 10032 USA
| | - Yared Tekabe
- grid.21729.3f0000000419368729Departments of Medicine, Anesthesiology, and Pathology, Columbia University, 622 West 168th St, PH 10-203, New York, NY 10032 USA
| | - Tina Zelonina
- grid.21729.3f0000000419368729Departments of Medicine, Anesthesiology, and Pathology, Columbia University, 622 West 168th St, PH 10-203, New York, NY 10032 USA
| | - Xinran Ma
- grid.21729.3f0000000419368729Departments of Medicine, Anesthesiology, and Pathology, Columbia University, 622 West 168th St, PH 10-203, New York, NY 10032 USA
| | - Geping Zhang
- grid.21729.3f0000000419368729Departments of Medicine, Anesthesiology, and Pathology, Columbia University, 622 West 168th St, PH 10-203, New York, NY 10032 USA
| | - Monica Goldklang
- grid.21729.3f0000000419368729Departments of Medicine, Anesthesiology, and Pathology, Columbia University, 622 West 168th St, PH 10-203, New York, NY 10032 USA
| | - Jeanine D’Armiento
- grid.21729.3f0000000419368729Departments of Medicine, Anesthesiology, and Pathology, Columbia University, 622 West 168th St, PH 10-203, New York, NY 10032 USA
| |
Collapse
|
8
|
Liu C, Li P, Zheng J, Wang Y, Wu W, Liu X. Role of necroptosis in airflow limitation in chronic obstructive pulmonary disease: focus on small-airway disease and emphysema. Cell Death Dis 2022; 8:363. [PMID: 35973987 PMCID: PMC9381515 DOI: 10.1038/s41420-022-01154-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/31/2022] [Accepted: 08/01/2022] [Indexed: 11/09/2022]
Abstract
Airflow limitation with intractable progressive mechanisms is the main disease feature of chronic obstructive pulmonary disease (COPD). The pathological process of airflow limitation in COPD involves necroptosis, a form of programmed necrotic cell death with pro-inflammatory properties. In this paper, the correlations of small-airway disease and emphysema with airflow limitation in COPD were firstly reviewed; then, based on this, the effects of necroptosis on small-airway disease and emphysema were analysed, and the possible mechanisms of necroptosis causing airflow limitation in COPD were explored. The results showed that airflow limitation is caused by a combination of small-airway disease and emphysema. In addition, toxic particulate matter stimulates epithelial cells to trigger necroptosis, and necroptosis promotes the expulsion of cell contents, the abnormal hyperplasia of pro-inflammatory mediators and the insufficient clearance of dead cells by macrophages; these processes, coupled with the interaction of necroptosis and oxidative stress, collectively result in small-airway disease and emphysema in COPD.
Collapse
Affiliation(s)
- Chanjing Liu
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Peijun Li
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Jiejiao Zheng
- Department of Rehabilitation Medicine, Huadong Hospital, Shanghai, People's Republic of China
| | - Yingqi Wang
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Weibing Wu
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, People's Republic of China.
| | - Xiaodan Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
9
|
Xu W, Wu CJ, Jiao YM, Mei XL, Huang L, Qin EQ, Tu B, Zhao P, Wang LF, Chen WW. Soluble Receptor for Advanced Glycation End Product Is Involved in the Inflammatory Response of Human Adenovirus-Infected Patients. Front Microbiol 2022; 13:923215. [PMID: 35875560 PMCID: PMC9301492 DOI: 10.3389/fmicb.2022.923215] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Human adenovirus (HAdV) infection causes excessive inflammation associated with severe tissue injury, such as pneumonia. The molecules involved in the underlying inflammatory mechanisms remain to be elucidated. Receptor for advanced glycation end product (RAGE) is mainly expressed on immune cells and lung tissues, and it is a key factor in the initiation and development of inflammation. RAGE can be cleaved by metalloprotease 9 (MMP9) to release the extracellular segment, which is named soluble RAGE (sRAGE), into the intercellular space, where it can bind to RAGE ligands and block RAGE activation and subsequent inflammation. In our study, we enrolled HAdV-infected patients and their contacts to examine the relationship between sRAGE and inflammation induced by HAdV infection. The results showed that HAdV infection stimulated inflammatory cytokine secretion, increased such as high mobility group box 1 (HMGB1) levels, and suppressed sRAGE expression. sRAGE levels were significantly different between patients with or without pneumonia. We also found that MMP9 was significantly lower in patients with pneumonia, and it was positively correlated with sRAGE levels over 7 days after disease onset. The mitogen-activated protein kinase (MAPK) pathway is an important immune activation signaling pathway that is regulated by RAGE. We observed the activation of the MAPK pathway in the peripheral blood mononuclear cells (PBMCs) of patients. Negative correlations between sRAGE and phosphorylated JNK and p38 were observed. These results suggest that sRAGE is involved in HAdV-induced inflammatory responses, and might be a potential therapeutic target to alleviate the HAdV-induced excessive inflammation.
Collapse
Affiliation(s)
- Wen Xu
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Cheng-Jun Wu
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- IC Technology Key Lab of Liaoning, School of Biomedical Engineering, Dalian University of Technology, Dalian, China
| | - Yan-Mei Jiao
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Xiao-Le Mei
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Lei Huang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - En-Qiang Qin
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Bo Tu
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Peng Zhao
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Li-Feng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Wei-Wei Chen
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- *Correspondence: Wei-Wei Chen,
| |
Collapse
|
10
|
He CP, Chen C, Jiang XC, Li H, Zhu LX, Wang PX, Xiao T. The role of AGEs in pathogenesis of cartilage destruction in osteoarthritis. Bone Joint Res 2022; 11:292-300. [PMID: 35549515 PMCID: PMC9130677 DOI: 10.1302/2046-3758.115.bjr-2021-0334.r1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative disease resulting from progressive joint destruction caused by many factors. Its pathogenesis is complex and has not been elucidated to date. Advanced glycation end products (AGEs) are a series of irreversible and stable macromolecular complexes formed by reducing sugar with protein, lipid, and nucleic acid through a non-enzymatic glycosylation reaction (Maillard reaction). They are an important indicator of the degree of ageing. Currently, it is considered that AGEs accumulation in vivo is a molecular basis of age-induced OA, and AGEs production and accumulation in vivo is one of the important reasons for the induction and acceleration of the pathological changes of OA. In recent years, it has been found that AGEs are involved in a variety of pathological processes of OA, including extracellular matrix degradation, chondrocyte apoptosis, and autophagy. Clearly, AGEs play an important role in regulating the expression of OA-related genes and maintaining the chondrocyte phenotype and the stability of the intra-articular environment. This article reviews the latest research results of AGEs in a variety of pathological processes of OA, to provide a new direction for the study of OA pathogenesis and a new target for prevention and treatment. Cite this article: Bone Joint Res 2022;11(5):292–300.
Collapse
Affiliation(s)
- Chao-Peng He
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Cheng Chen
- Department of Orthopedics, Second Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Xin-Chen Jiang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.,Hunan Provincial Key Laboratory of Neurorestoratology, Second Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Hui Li
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Li-Xin Zhu
- Department of Orthopedics, Second Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Ping-Xiao Wang
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Tao Xiao
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
11
|
Wang W, Zheng F, Zhang A. Arsenic-induced lung inflammation and fibrosis in a rat model: Contribution of the HMGB1/RAGE, PI3K/AKT, and TGF-β1/SMAD pathways. Toxicol Appl Pharmacol 2021; 432:115757. [PMID: 34673086 DOI: 10.1016/j.taap.2021.115757] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/10/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023]
Abstract
An increasing number of studies have shown that arsenic exposure increases the risk of lung cancer as well as a variety of non-malignant respiratory diseases, including bronchitis and tracheobronchitis. HMGB1 is widely expressed in a variety of tissues and cells and is involved in the pathological processes of many lung diseases through binding to the corresponding receptors and activating the downstream signaling pathways. However, the exact role of HMGB1/RAGE in arsenic-induced lung injury remains unknown. The aim of this study was to investigate whether HMGB1/RAGE and its activated downstream pathways are involved in the process of arsenic exposure-induced lung injury in rats. In this study, an animal model of oral exposure to arsenic was induced using 2.5, 5 and 10 mg/kg NaAsO2. The results showed that capillary permeability (LDH, TP, ACP, and AKP) was increased in the arsenic exposure groups, resulting in cell damage; this was accompanied by acute inflammation marked by significant neutrophil infiltration. Meanwhile, obvious histopathological damage, including thickening of the lung epithelium, increased infiltration of inflammatory cells, rupture of the alveolar wall, swelling of the mitochondria, and chromatin agglutination was observed by H&E staining and transmission electron microscopy. Furthermore, the results confirmed that the expressions of HMGB1 and RAGE in lung tissue were enhanced, and protein expression of PI3K, p-AKT, IL-1β, IL-18, and MMP-9 was increased in lung homogenates from the arsenic-exposed groups compared to the control group. Finally, Masson's staining results revealed arsenic-induced fibrosis and collagen deposition. Moreover, a significant increase in key fibrosis factors, including TGF-β1, p-SMAD2, p-SMAD3, and SMAD4 was observed in the lung homogenates in arsenic-exposed groups. In conclusion, the current study demonstrates that sub-chronic arsenic exposure triggers the inflammatory response and collagen fiber deposition in rat lung tissue. The potential mechanism may be closely related to activation of the pro-inflammatory-related HMGB1/RAGE pathway and initiation of the PI3K/AKT and TGF-β1/SMAD pathways.
Collapse
Affiliation(s)
- Wenjuan Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Fanyan Zheng
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China.
| |
Collapse
|
12
|
Higher-Dose DHA Supplementation Modulates Immune Responses in Pregnancy and Is Associated with Decreased Preterm Birth. Nutrients 2021; 13:nu13124248. [PMID: 34959801 PMCID: PMC8703393 DOI: 10.3390/nu13124248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
Pregnancy and parturition involve extensive changes in the maternal immune system. In our randomized, multi-site, double-blind superiority trial using a Bayesian adaptive design, we demonstrated that 1000 mg/day of docosahexaenoic acid (DHA) was superior to 200 mg/day in preventing both early preterm birth (less than 34 weeks' gestation) and preterm birth (less than 37 weeks' gestation). The goal of this secondary study is to compare the effects of 1000 mg/day versus 200 mg/day on maternal inflammation, a possible mechanism by which DHA may prevent preterm birth. Maternal blood samples were collected at enrollment (12-20 weeks' gestation) and at delivery. Red blood cell DHA levels were measured by gas chromatography, and plasma concentrations of sRAGE, IL-6, IL-1β, TNFα, and INFγ were measured by ELISA. Data were analyzed for associations with the DHA dose, gestational age at birth, and preterm birth (<37 weeks). Higher baseline and lower delivery levels of maternal sRAGE were associated with a greater probability of longer gestation and delivery at term gestation. Higher-dose DHA supplementation increased the probability of a smaller decrease in delivery sRAGE levels. Higher IL-6 concentrations at delivery were associated with the probability of delivering after 37 weeks, and higher-dose DHA supplementation increased the probability of greater increases in IL-6 concentrations between enrollment and delivery. These data provide a proposed mechanistic explanation of how a higher dose of DHA during pregnancy provides immunomodulatory regulation in the initiation of parturition by influencing sRAGE and IL-6 levels, which may explain its ability to reduce the risk of preterm birth.
Collapse
|
13
|
Kunimoto M, Yokoyama M, Shimada K, Matsubara T, Aikawa T, Ouchi S, Fukao K, Miyazaki T, Fujiwara K, Abulimiti A, Honzawa A, Shimada A, Yamamoto T, Amano A, Saitoh M, Morisawa T, Takahashi T, Daida H, Minamino T. Relationship between skin autofluorescence levels and clinical events in patients with heart failure undergoing cardiac rehabilitation. Cardiovasc Diabetol 2021; 20:208. [PMID: 34656131 PMCID: PMC8520614 DOI: 10.1186/s12933-021-01398-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 10/06/2021] [Indexed: 12/11/2022] Open
Abstract
Background Advanced glycation end-products, indicated by skin autofluorescence (SAF) levels, could be prognostic predictors of all-cause and cardiovascular mortality in patients with diabetes mellitus (DM) and renal disease. However, the clinical usefulness of SAF levels in patients with heart failure (HF) who underwent cardiac rehabilitation (CR) remains unclear. This study aimed to investigate the associations between SAF and MACE risk in patients with HF who underwent CR. Methods This study enrolled 204 consecutive patients with HF who had undergone CR at our university hospital between November 2015 and October 2017. Clinical characteristics and anthropometric data were collected at the beginning of CR. SAF levels were noninvasively measured with an autofluorescence reader. Major adverse cardiovascular event (MACE) was a composite of all-cause mortality and unplanned hospitalization for HF. Follow-up data concerning primary endpoints were collected until November 2017. Results Patients’ mean age was 68.1 years, and 61% were male. Patients were divided into two groups according to the median SAF levels (High and Low SAF groups). Patients in the High SAF group were significantly older, had a higher prevalence of chronic kidney disease, and more frequently had history of coronary artery bypass surgery; however, there were no significant between-group differences in sex, prevalence of DM, left ventricular ejection fraction, and physical function. During a mean follow-up period of 590 days, 18 patients had all-cause mortality and 36 were hospitalized for HF. Kaplan–Meier analysis showed that patients in the high SAF group had a higher incidence of MACE (log-rank P < 0.05). After adjusting for confounding factors, Cox regression multivariate analysis revealed that SAF levels were independently associated with the incidence of MACE (odds ratio, 1.86; 95% confidence interval, 1.08–3.12; P = 0.03). Conclusion SAF levels were significantly associated with the incidence of MACE in patients with HF and may be useful for risk stratification in patients with HF who underwent CR. Supplementary Information The online version contains supplementary material available at 10.1186/s12933-021-01398-0.
Collapse
Affiliation(s)
- Mitsuhiro Kunimoto
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Miho Yokoyama
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Cardiovascular Rehabilitation and Fitness, Juntendo University Hospital, Tokyo, Japan
| | - Kazunori Shimada
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Cardiovascular Rehabilitation and Fitness, Juntendo University Hospital, Tokyo, Japan
| | - Tomomi Matsubara
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Tatsuro Aikawa
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Shohei Ouchi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Kosuke Fukao
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Tetsuro Miyazaki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Kei Fujiwara
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Abidan Abulimiti
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Akio Honzawa
- Cardiovascular Rehabilitation and Fitness, Juntendo University Hospital, Tokyo, Japan
| | - Akie Shimada
- Department of Cardiovascular Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Taira Yamamoto
- Department of Cardiovascular Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsushi Amano
- Department of Cardiovascular Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masakazu Saitoh
- Department of Physical Therapy Faculty of Health Science, Juntendo University, Tokyo, Japan
| | - Tomoyuki Morisawa
- Department of Physical Therapy Faculty of Health Science, Juntendo University, Tokyo, Japan
| | - Tetsuya Takahashi
- Department of Physical Therapy Faculty of Health Science, Juntendo University, Tokyo, Japan
| | - Hiroyuki Daida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Department of Physical Therapy Faculty of Health Science, Juntendo University, Tokyo, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
14
|
Mete E, Sabirli R, Goren T, Turkcuer I, Kurt Ö, Koseler A. Association Between S100b Levels and COVID-19 Pneumonia: A Case Control Study. In Vivo 2021; 35:2923-2928. [PMID: 34410988 DOI: 10.21873/invivo.12583] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/20/2021] [Accepted: 06/29/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND/AIM Extracellular S100b effects are mediated by the receptor for advanced glycation end products (RAGE), which is the S100b membrane receptor. RAGE belongs to the immunoglobulin superfamily of cell surface molecules and serves as a multiligand receptor and is expressed in high abundance by alveolar type I (AT-I) cells in adult pulmonary tissue. This study aimed to provide an insight into the association between the severity of COVID-19 disease and serum S100b levels during admission to the emergency department (ED). PATIENTS AND METHODS A total of 64 patients (34 mild cases; 30 severe cases) were diagnosed with COVID-19 pneumonia and 30 healthy volunteers were admitted to study. Serum S100b levels were measured by using enzymle linked immunoassay method from blood serum samples. RESULTS Serum S100b levels showed a significantly higher mean value in mild and severe disease cohorts than in healthy controls (p=0.036 and p=0.028 respectively). Receiver operating characteristic (ROC) analysis indicated greater area under the curve (AUC) for serum S100b levels of the COVID-19 patients (AUC=0.663, 95% CI=0.541-0.785; p=0.014). In addition, serum S100b concentration was measured as 151.7 ng/ml at 79.3% sensitivity and 51.7% specificity (p=0.014). Serum S100b protein levels can serve as a valuable clinical marker in establishing diagnosis of patients. Though not useful in identifying different stages of COVID-19 infection, serum S100b concentration along with other known markers can be utilized to reliably predict clinical severity along with other clinical parameters.
Collapse
Affiliation(s)
- Ergun Mete
- Department of Microbiology, Pamukkale University Faculty of Medicine, Denizli, Turkey
| | - Ramazan Sabirli
- Department of Emergency Medicine, Kafkas University Faculty of Medicine, Kars, Turkey
| | - Tarik Goren
- Department of Emergency Medicine, Pamukkale University Faculty of Medicine, Denizli, Turkey
| | - Ibrahim Turkcuer
- Department of Emergency Medicine, Pamukkale University Faculty of Medicine, Denizli, Turkey
| | - Özgür Kurt
- Department of Microbiology, Acibadem Mehmet Ali Aydinlar University School of Medicine, Istanbul, Turkey
| | - Aylin Koseler
- Department of Biophysics, Pamukkale University Faculty of Medicine, Denizli, Turkey
| |
Collapse
|
15
|
Perkins TN, Oury TD. The perplexing role of RAGE in pulmonary fibrosis: causality or casualty? Ther Adv Respir Dis 2021; 15:17534666211016071. [PMID: 34275342 PMCID: PMC8293846 DOI: 10.1177/17534666211016071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease in which most patients die within 3 years of diagnosis. With an unknown etiology, IPF results in progressive fibrosis of the lung parenchyma, diminishing normal lung function, which results in respiratory failure, and eventually, death. While few therapies are available to reduce disease progression, patients continue to advance toward respiratory failure, leaving lung transplantation the only viable option for survival. As incidence and mortality rates steadily increase, the need for novel therapeutics is imperative. The receptor for advanced glycation endproducts (RAGE) is most highly expressed in the lungs and plays a significant role in a number of chronic lung diseases. RAGE has long been linked to IPF; however, confounding data from both human and experimental studies have left an incomplete and perplexing story. This review examines the present understanding of the role of RAGE in human and experimental models of IPF, drawing parallels to recent advances in RAGE biology. Moreover, this review discusses the role of RAGE in lung injury response, type 2 immunity, and cellular senescence, and how such mechanisms may relate to RAGE as both a biomarker of disease progression and potential therapeutic target in IPF.The reviews of this paper are available via the supplemental material section.
Collapse
Affiliation(s)
- Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, 3550 Terrace Street, S-784 Scaife Hall, Pittsburgh, PA 15261, USA
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
16
|
Nouri-Keshtkar M, Taghizadeh S, Farhadi A, Ezaddoustdar A, Vesali S, Hosseini R, Totonchi M, Kouhkan A, Chen C, Zhang JS, Bellusci S, Tahamtani Y. Potential Impact of Diabetes and Obesity on Alveolar Type 2 (AT2)-Lipofibroblast (LIF) Interactions After COVID-19 Infection. Front Cell Dev Biol 2021; 9:676150. [PMID: 34307358 PMCID: PMC8295688 DOI: 10.3389/fcell.2021.676150] [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: 03/04/2021] [Accepted: 06/11/2021] [Indexed: 01/14/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a new emerging respiratory virus, caused evolving pneumonia outbreak around the world. In SARS-Cov-2 infected patients, diabetes mellitus (DM) and obesity are two metabolic diseases associated with higher severity of SARS-CoV-2 related complications, characterized by acute lung injury requiring assisted ventilation as well as fibrosis development in surviving patients. Different factors are potentially responsible for this exacerbated response to SARS-CoV-2 infection. In patients with DM, base-line increase in inflammation and oxidative stress represent preexisting risk factors for virus-induced damages. Such factors are also likely to be found in obese patients. In addition, it has been proposed that massive injury to the alveolar epithelial type 2 (AT2) cells, which express the SARS-CoV-2 receptor angiotensin-converting enzyme 2 (ACE2), leads to the activation of their stromal niches represented by the Lipofibroblasts (LIF). LIF are instrumental in maintaining the self-renewal of AT2 stem cells. LIF have been proposed to transdifferentiate into Myofibroblast (MYF) following injury to AT2 cells, thereby contributing to fibrosis. We hypothesized that LIF's activity could be impacted by DM or obesity in an age- and gender-dependent manner, rendering them more prone to transition toward the profibrotic MYF status in the context of severe COVID-19 pneumonia. Understanding the cumulative effects of DM and/or obesity in the context of SARS-CoV-2 infection at the cellular level will be crucial for efficient therapeutic solutions.
Collapse
Affiliation(s)
- Marjan Nouri-Keshtkar
- Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sara Taghizadeh
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Excellence Cluster Cardio-Pulmonary System, Justus Liebig University Giessen, Giessen, Germany
| | - Aisan Farhadi
- Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Samira Vesali
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Roya Hosseini
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mehdi Totonchi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Azam Kouhkan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Chengshui Chen
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jin-San Zhang
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Saverio Bellusci
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Excellence Cluster Cardio-Pulmonary System, Justus Liebig University Giessen, Giessen, Germany
| | - Yaser Tahamtani
- Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
17
|
Lazzari TK, Cavalheiro E, Coutinho SE, da Silva LF, Silva DR. Leptin and advanced glycation end products receptor (RAGE) in tuberculosis patients. PLoS One 2021; 16:e0254198. [PMID: 34214138 PMCID: PMC8253382 DOI: 10.1371/journal.pone.0254198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/22/2021] [Indexed: 11/18/2022] Open
Abstract
Introduction The pathogenesis of consumptive syndrome of tuberculosis (TB) is largely unknown. Leptin concentrations may be high because of the host’s inflammatory response, contributing to weight loss in patients with TB. The receptor for advanced glycation end products (RAGE) is also associated with weight loss in patients with TB and is related to enhanced mortality. The objective of this study was to evaluate the association between leptin and AGE/RAGE. Methods Case-control study. Leptin, AGE (carboxymethyl lysine, CML) and soluble RAGE (sRAGE) were measured from blood samples by ELISA. Results We included in the study 34 patients with TB and 34 controls. We found an inverse correlation between serum leptin levels and sRAGE, only in cases (r = -0.609, p < 0.0001). sRAGE levels were lower in patients with TB who died as compared with patients who survive (21.90 ± 4.24 pg/mL vs 66.14 ± 29.49 pg/mL; p = 0.045). Leptin levels were higher in patients with TB who died as compared with patients who survive (14.11 [7.48–14.11] ng/mL vs 3.08 [0.54–6.34] ng/mL; p = 0.028). Conclusions We identified lower sRAGE levels and higher leptin levels in patients with TB who died as compared with patients who survive. In addition, an inverse and significant correlation between serum leptin and sRAGE levels was demonstrated. Future studies, with a larger sample size and in different settings, including not only hospitalized patients, are needed to confirm these findings.
Collapse
Affiliation(s)
- Tássia Kirchmann Lazzari
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Erika Cavalheiro
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Sandra Eugênia Coutinho
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lívia Fontes da Silva
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Denise Rossato Silva
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- * E-mail:
| |
Collapse
|
18
|
Nucera F, Lo Bello F, Shen SS, Ruggeri P, Coppolino I, Di Stefano A, Stellato C, Casolaro V, Hansbro PM, Adcock IM, Caramori G. Role of Atypical Chemokines and Chemokine Receptors Pathways in the Pathogenesis of COPD. Curr Med Chem 2021; 28:2577-2653. [PMID: 32819230 DOI: 10.2174/0929867327999200819145327] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/11/2020] [Accepted: 06/18/2020] [Indexed: 11/22/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) represents a heightened inflammatory response in the lung generally resulting from tobacco smoking-induced recruitment and activation of inflammatory cells and/or activation of lower airway structural cells. Several mediators can modulate activation and recruitment of these cells, particularly those belonging to the chemokines (conventional and atypical) family. There is emerging evidence for complex roles of atypical chemokines and their receptors (such as high mobility group box 1 (HMGB1), antimicrobial peptides, receptor for advanced glycosylation end products (RAGE) or toll-like receptors (TLRs)) in the pathogenesis of COPD, both in the stable disease and during exacerbations. Modulators of these pathways represent potential novel therapies for COPD and many are now in preclinical development. Inhibition of only a single atypical chemokine or receptor may not block inflammatory processes because there is redundancy in this network. However, there are many animal studies that encourage studies for modulating the atypical chemokine network in COPD. Thus, few pharmaceutical companies maintain a significant interest in developing agents that target these molecules as potential antiinflammatory drugs. Antibody-based (biological) and small molecule drug (SMD)-based therapies targeting atypical chemokines and/or their receptors are mostly at the preclinical stage and their progression to clinical trials is eagerly awaited. These agents will most likely enhance our knowledge about the role of atypical chemokines in COPD pathophysiology and thereby improve COPD management.
Collapse
Affiliation(s)
- Francesco Nucera
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Federica Lo Bello
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Sj S Shen
- Faculty of Science, Centre for Inflammation, Centenary Institute, University of Technology, Ultimo, Sydney, Australia
| | - Paolo Ruggeri
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Irene Coppolino
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Antonino Di Stefano
- Division of Pneumology, Cyto- Immunopathology Laboratory of the Cardio-Respiratory System, Clinical Scientific Institutes Maugeri IRCCS, Veruno, Italy
| | - Cristiana Stellato
- Department of Medicine, Surgery and Dentistry, Salerno Medical School, University of Salerno, Salerno, Italy
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry, Salerno Medical School, University of Salerno, Salerno, Italy
| | - Phil M Hansbro
- Faculty of Science, Centre for Inflammation, Centenary Institute, University of Technology, Ultimo, Sydney, Australia
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Gaetano Caramori
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| |
Collapse
|
19
|
Rojas A, Lindner C, Gonzàlez I, Morales MA. Advanced-glycation end-products axis: A contributor to the risk of severe illness from COVID-19 in diabetes patients. World J Diabetes 2021; 12:590-602. [PMID: 33995847 PMCID: PMC8107984 DOI: 10.4239/wjd.v12.i5.590] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/29/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Compelling pieces of evidence derived from both clinical and experimental research has demonstrated the crucial role of the receptor for advanced-glycation end-products (RAGE) in orchestrating a plethora of proinflammatory cellular responses leading to many of the complications and end-organ damages reported in patients with diabetes mellitus (DM). During the coronavirus disease 2019 (COVID-19) pandemic, many clinical reports have pointed out that DM increases the risk of COVID-19 complications, hospitalization requirements, as well as the overall severe acute respiratory syndrome coronavirus 2 case-fatality rate. In the present review, we intend to focus on how the basal activation state of the RAGE axis in common preexisting conditions in DM patients such as endothelial dysfunction and hyperglycemia-related prothrombotic phenotype, as well as the contribution of RAGE signaling in lung inflammation, may then lead to the increased mortality risk of COVID-19 in these patients. Additionally, the cross-talk between the RAGE axis with either another severe acute respiratory syndrome coronavirus 2 receptor molecule different of angiotensin-converting enzyme 2 or the renin-angiotensin system imbalance produced by viral infection, as well as the role of this multi-ligand receptor on the obesity-associated low-grade inflammation in the higher risk for severe illness reported in diabetes patients with COVID-19, are also discussed.
Collapse
Affiliation(s)
- Armando Rojas
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca 3460000, Chile
| | - Cristian Lindner
- Medicine Faculty, Catholic University of Maule, Talca 3460000, Chile
| | - Ileana Gonzàlez
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca 3460000, Chile
| | - Miguel Angel Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago 8320000, Chile
| |
Collapse
|
20
|
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
|
21
|
The Effect and Regulatory Mechanism of High Mobility Group Box-1 Protein on Immune Cells in Inflammatory Diseases. Cells 2021; 10:cells10051044. [PMID: 33925132 PMCID: PMC8145631 DOI: 10.3390/cells10051044] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/18/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
High mobility group box-1 protein (HMGB1), a member of the high mobility group protein superfamily, is an abundant and ubiquitously expressed nuclear protein. Intracellular HMGB1 is released by immune and necrotic cells and secreted HMGB1 activates a range of immune cells, contributing to the excessive release of inflammatory cytokines and promoting processes such as cell migration and adhesion. Moreover, HMGB1 is a typical damage-associated molecular pattern molecule that participates in various inflammatory and immune responses. In these ways, it plays a critical role in the pathophysiology of inflammatory diseases. Herein, we review the effects of HMGB1 on various immune cell types and describe the molecular mechanisms by which it contributes to the development of inflammatory disorders. Finally, we address the therapeutic potential of targeting HMGB1.
Collapse
|
22
|
Yalcin Kehribar D, Cihangiroglu M, Sehmen E, Avci B, Capraz A, Yildirim Bilgin A, Gunaydin C, Ozgen M. The receptor for advanced glycation end product (RAGE) pathway in COVID-19. Biomarkers 2021; 26:114-118. [PMID: 33284049 PMCID: PMC7814566 DOI: 10.1080/1354750x.2020.1861099] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Coronavirus disease-2019 (COVID-19) with lung involvement frequently causes morbidity and mortality. Advanced age appears to be the most important risk factor. The receptor for advanced glycation end-product (RAGE) pathway is considered to play important roles in the physiological aging and pathogenesis of lung diseases. This study aimed to investigate the possible relationship between COVID-19 and RAGE pathway. MATERIALS AND METHODS This study included 23 asymptomatic patients and 35 patients with lung involvement who were diagnosed with COVID-19 as well as 22 healthy volunteers. Lung involvement was determined using computed tomography. Serum soluble-RAGE (sRAGE) levels were determined using enzyme-linked immunosorbent assay. RESULTS The sRAGE levels were significantly higher in the asymptomatic group than in the control group. Age, fibrinogen, C-reactive protein, and ferritin levels were higher and the sRAGE level was lower in the patients with lung involvement than in the asymptomatic patients. CONCLUSIONS In this study, patients with high sRAGE levels were younger and had asymptomatic COVID-19. Patients with low sRAGE levels were elderly patients with lung involvement, which indicates that the RAGE pathway plays an important role in the aggravation of COVID-19.
Collapse
Affiliation(s)
- Demet Yalcin Kehribar
- Ondokuz Mayis University, Faculty of Medicine, Department of Internal Medicine, Samsun, Turkey
| | - Mustafa Cihangiroglu
- Amasya University, Faculty of Medicine, Department of Infection Disease, Amasya, Turkey
| | - Emine Sehmen
- Samsun Education and Reseach Hospital, Department of Infection Disease, Samsun, Turkey
| | - Bahattin Avci
- Department of Biochemistry, Faculty of Medicine, Ondokuz Mayis University, Kurupelit, Turkey
| | - Aylin Capraz
- Amasya University, Faculty of Medicine, Department of Infection Disease, Amasya, Turkey
| | - Ayse Yildirim Bilgin
- Amasya University, Faculty of Medicine, Department of Infection Disease, Amasya, Turkey
| | - Caner Gunaydin
- Faculty of Medicine Ringgold Standard Institution, Ondokuz Mayis University, Samsun, Turkey
| | - Metin Ozgen
- Faculty of Medicine Ringgold Standard Institution, Ondokuz Mayis University, Samsun, Turkey
| |
Collapse
|
23
|
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
|
24
|
Tsai CY, Chou HC, Chen CM. Perinatal nicotine exposure alters lung development and induces HMGB1-RAGE expression in neonatal mice. Birth Defects Res 2020; 113:570-578. [PMID: 33166075 DOI: 10.1002/bdr2.1840] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/17/2020] [Accepted: 10/31/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Maternal nicotine exposure during gestation and lactation adversely affect lung development of their children. High-mobility group box 1 (HMGB1) is the encoded non-histone, nuclear DNA-binding protein that regulates transcription, and is involved in organization of DNA. Receptors for advanced glycation end products (RAGE) is a receptor for HMGB1 and activates nuclear factor-κB (NF-κB) signaling. Animal and human studies have found cigarette smoke exposure upregulates RAGE expression, suggesting that the HMGB1-RAGE pathway might be involved in maternal nicotine-induced lung injury. METHODS This study evaluated prenatal and perinatal nicotine effects on lung development and HMGB1 and RAGE expression in mouse offspring. Nicotine was administered to pregnant mice by subcutaneous osmotic mini-pump at a dose of 6 mg kg-1 day-1 from gestational Day 14 to birth (prenatal) or to postnatal Day 21 (perinatal). A control group received an equal volume of saline by the same route. Three study groups were obtained: prenatal normal saline (NS), prenatal nicotine, and perinatal nicotine groups. The mice were euthanized on postnatal Day 21, and the lung tissues were collected for histological and Western blot analyses. RESULTS Mice exposed to prenatal nicotine exhibited significantly higher lung mean chord length and oxidative stress marker 8-hydroxy-2'-deoxyguanosine and NF-κB expression compared to mice exposed to NS. Perinatal nicotine exposure further enhanced these harmful effects. These perinatal nicotine effects on lung development were associated with increased HMGB1 and RAGE expression. CONCLUSIONS HMGB1-RAGE pathway may be involved in the pathogenesis of altered lung development induced by perinatal nicotine exposure.
Collapse
Affiliation(s)
- Chin-Yen Tsai
- Department of Pediatrics, Yuan's General Hospital, Kaohsiung, Taiwan
| | - Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chung-Ming Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
25
|
Yamaguchi K, Iwamoto H, Mazur W, Miura S, Sakamoto S, Horimasu Y, Masuda T, Miyamoto S, Nakashima T, Ohshimo S, Fujitaka K, Hamada H, Hattori N. Reduced endogenous secretory RAGE in blood and bronchoalveolar lavage fluid is associated with poor prognosis in idiopathic pulmonary fibrosis. Respir Res 2020; 21:145. [PMID: 32527263 PMCID: PMC7291663 DOI: 10.1186/s12931-020-01410-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 05/31/2020] [Indexed: 12/16/2022] Open
Abstract
Background The endogenous secretory receptor for advanced glycation end products (esRAGE) is a soluble isoform produced by alternative splicing of the RAGE gene. The isoform has anti-inflammatory properties due to its inhibition of the RAGE/ligand interaction and is reduced in the lung tissue of patients with idiopathic pulmonary fibrosis (IPF). This study aimed to investigate the association of esRAGE serum and bronchoalveolar lavage fluid (BALF) levels with progression of IPF. Methods This study included 79 IPF patients and 90 healthy controls. IPF and control serum esRAGE levels were compared, and the correlation between serum and BALF esRAGE levels was analyzed in 57 IPF patient samples. We also investigated the relationship of esRAGE serum and BALF levels with prognoses and lung function parameters in patients with IPF. Results Serum esRAGE levels in IPF patients were significantly lower than those in healthy controls (162.0 ± 102.4 ng/ml and 200.7 ± 107.3 ng/ml, p = 0.009), although the baseline characteristics of age and smoking history were not matched. Serum levels of esRAGE were correlated with BALF esRAGE levels (rs = 0.317). The BALF esRAGE levels were also correlated with diffusion capacity for carbon monoxide (rs = 0.406). A Kaplan-Meier curve analysis and univariate/multivariate Cox hazard proportion analysis revealed that lower levels of esRAGE in blood and BALF were significantly associated with poorer prognoses in patients with IPF. Conclusions Decreased esRAGE levels in BALF and blood were associated with poor prognoses in patients with IPF. These results suggest that esRAGE could be related to the pathophysiology of IPF and serum esRAGE could be a potential prognostic marker of IPF.
Collapse
Affiliation(s)
- Kakuhiro Yamaguchi
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hiroshi Iwamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Witold Mazur
- Heart and Lung Centre, Division of Pulmonary Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Shinichiro Miura
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shinjiro Sakamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yasushi Horimasu
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Takeshi Masuda
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shintaro Miyamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Taku Nakashima
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shinichiro Ohshimo
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazunori Fujitaka
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hironobu Hamada
- Department of Physical Analysis and Therapeutic Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
26
|
Kosutova P, Mikolka P, Balentova S, Adamkov M, Calkovska A, Mokra D. Effects of PDE3 Inhibitor Olprinone on the Respiratory Parameters, Inflammation, and Apoptosis in an Experimental Model of Acute Respiratory Distress Syndrome. Int J Mol Sci 2020; 21:E3382. [PMID: 32403267 PMCID: PMC7247002 DOI: 10.3390/ijms21093382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/03/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022] Open
Abstract
This study aimed to investigate whether a selective phosphodiesterase-3 (PDE3) inhibitor olprinone can positively influence the inflammation, apoptosis, and respiratory parameters in animals with acute respiratory distress syndrome (ARDS) model induced by repetitive saline lung lavage. Adult rabbits were divided into 3 groups: ARDS without therapy (ARDS), ARDS treated with olprinone i.v. (1 mg/kg; ARDS/PDE3), and healthy ventilated controls (Control), and were oxygen-ventilated for the following 4 h. Dynamic lung-thorax compliance (Cdyn), mean airway pressure (MAP), arterial oxygen saturation (SaO2), alveolar-arterial gradient (AAG), ratio between partial pressure of oxygen in arterial blood to a fraction of inspired oxygen (PaO2/FiO2), oxygenation index (OI), and ventilation efficiency index (VEI) were evaluated every hour. Post mortem, inflammatory and oxidative markers (interleukin (IL)-6, IL-1β, a receptor for advanced glycation end products (RAGE), IL-10, total antioxidant capacity (TAC), 3-nitrotyrosine (3NT), and malondialdehyde (MDA) and apoptosis (apoptotic index and caspase-3) were assessed in the lung tissue. Treatment with olprinone reduced the release of inflammatory mediators and markers of oxidative damage decreased apoptosis of epithelial cells and improved respiratory parameters. The results indicate a future potential of PDE3 inhibitors also in the therapy of ARDS.
Collapse
Affiliation(s)
- Petra Kosutova
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Pavol Mikolka
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Sona Balentova
- Department of Histology and Embryology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (S.B.); (M.A.)
| | - Marian Adamkov
- Department of Histology and Embryology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (S.B.); (M.A.)
| | - Andrea Calkovska
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Daniela Mokra
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| |
Collapse
|
27
|
Kim DH, Gu A, Lee JS, Yang EJ, Kashif A, Hong MH, Kim G, Park BS, Lee SJ, Kim IS. Suppressive effects of S100A8 and S100A9 on neutrophil apoptosis by cytokine release of human bronchial epithelial cells in asthma. Int J Med Sci 2020; 17:498-509. [PMID: 32174780 PMCID: PMC7053304 DOI: 10.7150/ijms.37833] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 09/05/2019] [Indexed: 12/28/2022] Open
Abstract
S100A8 and S100A9 are important proteins in the pathogenesis of allergy. Asthma is an allergic lung disease, characterized by bronchial inflammation due to leukocytes, bronchoconstriction, and allergen-specific IgE. In this study, we examined the role of S100A8 and S100A9 in the interaction of cytokine release from bronchial epithelial cells, with constitutive apoptosis of neutrophils. S100A8 and S100A9 induce increased secretion of neutrophil survival cytokines such as MCP-1, IL-6 and IL-8. This secretion is suppressed by TLR4 inhibitor), LY294002, AKT inhibitor, PD98059, SB202190, SP600125, and BAY-11-7085. S100A8 and S100A9 also induce the phosphorylation of AKT, ERK, p38 MAPK and JNK, and activation of NF-κB, which were blocked after exposure to TLR4i, LY294002, AKTi, PD98059, SB202190 or SP600125. Furthermore, supernatants collected from bronchial epithelial cells after S100A8 and S100A9 stimulation suppressed the apoptosis of normal and asthmatic neutrophils. These inhibitory mechanisms are involved in suppression of caspase 9 and caspase 3 activation, and BAX expression. The degradation of MCL-1 and BCL-2 was also blocked by S100A8 and S100A9 stimulation. Essentially, neutrophil apoptosis was blocked by co-culture of normal and asthmatic neutrophils with BEAS-2B cells in the presence of S100A8 and S100A9. These findings will enable elucidation of asthma pathogenesis.
Collapse
Affiliation(s)
- Da Hye Kim
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon 34824
| | - Ayoung Gu
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon 34824
| | - Ji-Sook Lee
- Department of Clinical Laboratory Science, Wonkwang Health Science University, Iksan, 54538
| | - Eun Ju Yang
- Department of Clinical Laboratory Science, Daegu Haany University, Gyeongsan, 38610
| | - Ayesha Kashif
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon 34824
| | - Min Hwa Hong
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon 34824
| | - Geunyeong Kim
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon 34824
| | - Beom Seok Park
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon 34824.,Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam 13135
| | - Soo Jin Lee
- Department of Pediatrics, School of Medicine, Eulji University, Daejeon, 301-746
| | - In Sik Kim
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Daejeon 34824.,Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon 34824, Republic of Korea
| |
Collapse
|
28
|
Wang M, Gauthier A, Daley L, Dial K, Wu J, Woo J, Lin M, Ashby C, Mantell LL. The Role of HMGB1, a Nuclear Damage-Associated Molecular Pattern Molecule, in the Pathogenesis of Lung Diseases. Antioxid Redox Signal 2019; 31:954-993. [PMID: 31184204 PMCID: PMC6765066 DOI: 10.1089/ars.2019.7818] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/07/2019] [Indexed: 12/11/2022]
Abstract
Significance: High-mobility group protein box 1 (HMGB1), a ubiquitous nuclear protein, regulates chromatin structure and modulates the expression of many genes involved in the pathogenesis of lung cancer and many other lung diseases, including those that regulate cell cycle control, cell death, and DNA replication and repair. Extracellular HMGB1, whether passively released or actively secreted, is a danger signal that elicits proinflammatory responses, impairs macrophage phagocytosis and efferocytosis, and alters vascular remodeling. This can result in excessive pulmonary inflammation and compromised host defense against lung infections, causing a deleterious feedback cycle. Recent Advances: HMGB1 has been identified as a biomarker and mediator of the pathogenesis of numerous lung disorders. In addition, post-translational modifications of HMGB1, including acetylation, phosphorylation, and oxidation, have been postulated to affect its localization and physiological and pathophysiological effects, such as the initiation and progression of lung diseases. Critical Issues: The molecular mechanisms underlying how HMGB1 drives the pathogenesis of different lung diseases and novel therapeutic approaches targeting HMGB1 remain to be elucidated. Future Directions: Additional research is needed to identify the roles and functions of modified HMGB1 produced by different post-translational modifications and their significance in the pathogenesis of lung diseases. Such studies will provide information for novel approaches targeting HMGB1 as a treatment for lung diseases.
Collapse
Affiliation(s)
- Mao Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Alex Gauthier
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - LeeAnne Daley
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Katelyn Dial
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Jiaqi Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Joanna Woo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Mosi Lin
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Charles Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Lin L. Mantell
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
- Center for Inflammation and Immunology, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York
| |
Collapse
|
29
|
The Modern Western Diet Rich in Advanced Glycation End-Products (AGEs): An Overview of Its Impact on Obesity and Early Progression of Renal Pathology. Nutrients 2019; 11:nu11081748. [PMID: 31366015 PMCID: PMC6724323 DOI: 10.3390/nu11081748] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/15/2019] [Accepted: 07/25/2019] [Indexed: 01/12/2023] Open
Abstract
Advanced glycation end-products (AGEs) are an assorted group of molecules formed through covalent bonds between a reduced sugar and a free amino group of proteins, lipids, and nucleic acids. Glycation alters their structure and function, leading to impaired cell function. They can be originated by physiological processes, when not counterbalanced by detoxification mechanisms, or derive from exogenous sources such as food, cigarette smoke, and air pollution. Their accumulation increases inflammation and oxidative stress through the activation of various mechanisms mainly triggered by binding to their receptors (RAGE). So far, the pathogenic role of AGEs has been evidenced in inflammatory and chronic diseases such as chronic kidney disease, cardiovascular disease, and diabetic nephropathy. This review focuses on the AGE-induced kidney damage, by describing the molecular players involved and investigating its link to the excess of body weight and visceral fat, hallmarks of obesity. Research regarding interventions to reduce AGE accumulation has been of great interest and a nutraceutical approach that would help fighting chronic diseases could be a very useful tool for patients’ everyday lives.
Collapse
|
30
|
Goldklang MP, Tekabe Y, Zelonina T, Trischler J, Xiao R, Stearns K, Rodriguez K, Shields A, Romanov A, D'Armiento JM, Johnson LL. Single-photon emission computed tomography/computed tomography imaging of RAGE in smoking-induced lung injury. Respir Res 2019; 20:116. [PMID: 31182072 PMCID: PMC6558785 DOI: 10.1186/s12931-019-1064-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 04/30/2019] [Indexed: 11/10/2022] Open
Abstract
Background Expression of the Receptor for Advanced Glycation Endproducts (RAGE) initiates pro-inflammatory pathways resulting in lung destruction. We hypothesized that RAGE directed imaging demonstrates increased lung uptake in smoke-exposure. Methods After exposure to room air or to cigarette smoke for 4-weeks or 16-weeks, rabbits were injected with 99mTc-anti-RAGE F(ab’)2 and underwent Single-Photon Emission Computed Tomography/Computed Tomography (SPECT/CT) imaging. Lung radiotracer uptake was calculated as percent injected dose (%ID). Lungs were dissected for gamma well counting and histological analysis. Results 99mTc-anti-RAGE F(ab’)2 SPECT/CT imaging demonstrated increased lung expression of RAGE with smoke exposure compared to room air control at 4-weeks: Room air right (R) 0.75 ± 0.38%ID, left (L) 0.62 ± 0.32%ID vs. Smoke exposed R 0.17 ± 0.03, L 0.17 ± 0.02%ID (p = 0.02 and 0.028, respectively). By 16-weeks of smoke exposure, the uptake decreased to 0.19 ± 0.05%ID R and 0.17 ± 0.05%ID L, significantly lower than 4-week imaging (p = 0.0076 and 0.0129 respectively). Staining for RAGE confirmed SPECT results, with the RAGE ligand HMGB1 upregulated in the macrophages of 4-week smoke-exposed rabbits. Conclusions RAGE-directed imaging identified pulmonary RAGE expression acutely in vivo in an animal model of emphysema early after smoke exposure, with diminution over time. These studies document the extent and time course of RAGE expression under smoke exposure conditions and could be utilized for disease monitoring and examining response to future RAGE-targeted therapies. Electronic supplementary material The online version of this article (10.1186/s12931-019-1064-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Monica P Goldklang
- Department of Anesthesiology, Columbia University, New York, NY, USA.,Department of Medicine, Columbia University, New York, NY, USA
| | - Yared Tekabe
- Department of Medicine, Columbia University, New York, NY, USA
| | - Tina Zelonina
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Jordis Trischler
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Rui Xiao
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Kyle Stearns
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | | | - Alexander Shields
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Alexander Romanov
- Institute for Comparative Medicine, Columbia University, New York, NY, USA
| | - Jeanine M D'Armiento
- Department of Anesthesiology, Columbia University, New York, NY, USA. .,Department of Medicine, Columbia University, New York, NY, USA. .,Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA.
| | - Lynne L Johnson
- Department of Medicine, Columbia University, New York, NY, USA.
| |
Collapse
|
31
|
Cho H, Eom Y. Potential Forensic Application of Receptor for Advanced Glycation End Products (RAGE) as a Novel Biomarker for Estimating Postmortem Interval. J Forensic Sci 2019; 64:1878-1883. [DOI: 10.1111/1556-4029.14063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/04/2019] [Accepted: 04/04/2019] [Indexed: 01/16/2023]
Affiliation(s)
- Hye‐Won Cho
- Department of Biomedical Laboratory Science College of Medical Sciences Soonchunhyang University Asan 31538 Republic of Korea
| | - Yong‐Bin Eom
- Department of Biomedical Laboratory Science College of Medical Sciences Soonchunhyang University Asan 31538 Republic of Korea
| |
Collapse
|
32
|
da Silva LF, Skupien EC, Lazzari TK, Holler SR, de Almeida EGC, Zampieri LR, Coutinho SE, Andrades M, Silva DR. Advanced glycation end products (AGE) and receptor for AGE (RAGE) in patients with active tuberculosis, and their relationship between food intake and nutritional status. PLoS One 2019; 14:e0213991. [PMID: 30870511 PMCID: PMC6417785 DOI: 10.1371/journal.pone.0213991] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 03/05/2019] [Indexed: 01/15/2023] Open
Abstract
Introduction The receptor for advanced glycation end products (RAGE) is expressed in normal lungs and is upregulated during infection. AGEs and RAGE cause oxidative stress and apoptosis in lung cells. The objective of this study is to evaluate levels of AGEs and its soluble receptor (sRAGE), and to investigate their relationship with food intake and nutritional status, in a university-affiliated hospital in Brazil. Methods Case-control study, from June 2017 to June 2018. AGE (carboxymethyl lysine, CML) and sRAGE were measured from blood samples by Elisa. Nutritional assessment was performed by body mass index, triceps skin-fold thickness, mid-arm circumference, mid-arm muscle circumference, bioelectrical impedance analysis, and food frequency questionnaire. Results We included in the study 35 tuberculosis (TB) patients and 35 controls. The mean sRAGE levels were higher in TB patients than in controls (68.5 ± 28.1 vs 57.5 ± 24.0 pg/mL; p = 0.046). Among cases that were current smokers, lower sRAGE levels were associated with mortality, evaluated at the end of hospitalization (p = 0.006), and with weight loss (p = 0.034). There was no statistically significant difference in CML levels and diet CML content between cases and controls. Malnutrition was more frequent in cases, but there was no correlation between nutritional parameters and CML or sRAGE levels. Conclusions TB patients had higher sRAGE levels than controls, although it is not clear that this difference is clinically relevant. Also, sRAGE was associated with weight loss and mortality.
Collapse
Affiliation(s)
- Lívia Fontes da Silva
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Erika Cavalheiro Skupien
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tássia Kirchmann Lazzari
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Sizuane Rieger Holler
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | - Sandra Eugênia Coutinho
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Denise Rossato Silva
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- * E-mail:
| |
Collapse
|
33
|
Ahmed MAE, El Morsy EM, Ahmed AAE. Protective effects of febuxostat against paraquat-induced lung toxicity in rats: Impact on RAGE/PI3K/Akt pathway and downstream inflammatory cascades. Life Sci 2019; 221:56-64. [PMID: 30726711 DOI: 10.1016/j.lfs.2019.02.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/25/2019] [Accepted: 02/02/2019] [Indexed: 01/09/2023]
Abstract
AIMS The herbicide paraquat causes fatal lung toxicity by induction of xanthine oxidase, production of free radicals and inflammation. Febuxostat, a xanthine oxidase inhibitor and anti-gout has recently shown anti-inflammatory activity. Accordingly, this study was carried out to investigate whether febuxostat may attenuate paraquat-induced lung toxicity and to explore the possible underlying mechanisms. MAIN METHODS Rats were administered either vehicle, a single dose of paraquat (30 mg/kg, i.p.), febuxostat (15 mg/kg, oral), or both for 14 successive days. Serum LDH and sRAGE were estimated. Lung tissue xanthine oxidase activity, SOD, TAC, MDA, and RAGE, HMGB1 gene expression, PI3K/Akt and β-catenin protein expression, MMP-9, IL-8, VEGF and COX-2 gene expression were estimated. KEY FINDINGS Results showed that paraquat induced lung injury characterized by enhanced oxidative stress and inflammation, upregulated RAGE, HMGB1 gene expression, PI3K/Akt and β-catenin protein expression. Administration of febuxostat inhibited the deleterious effects of paraquat on lung through inhibition of xanthine oxidase activity and related oxidative stress, downregulation of RAGE/PI3K/Akt pathway, and suppression of β-catenin protein expression and its downstream inflammatory mediators. SIGNIFICANCE The present study showed that febuxostat may abrogate paraquat-induced lung toxicity and demonstrated a novel mechanism for its ameliorative effects.
Collapse
Affiliation(s)
- Maha A E Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), 6th of October City, Giza, Egypt.
| | - Engy M El Morsy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, Egypt
| | - Amany A E Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, Egypt
| |
Collapse
|
34
|
Lee SY, Ha EJ, Cho HW, Kim HR, Lee D, Eom YB. Potential forensic application of receptor for advanced glycation end products (RAGE) and aquaporin 5 (AQP5) as novel biomarkers for diagnosis of drowning. J Forensic Leg Med 2019; 62:56-62. [PMID: 30677703 DOI: 10.1016/j.jflm.2019.01.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 10/15/2018] [Accepted: 01/11/2019] [Indexed: 02/08/2023]
Abstract
Drowning is the most common cause of unnatural death worldwide. There is no single biomarker to diagnose drowning, so the diagnosis of drowning is one of the most difficult tasks in forensic medicine. Especially, distinguishing a victim of drowning from a body disposed of in water following death remains a problem. The objective of this study was to identify specific biomarkers of drowning compared with other causes of death such as hypoxia and postmortem submersion. The present study investigated the intrapulmonary expression of receptor for advanced glycation end products (RAGE), aquaporin-5 (AQP5), surfactant protein-A (SP-A), interleukin 6 (IL-6) and interleukin 1β (IL-1β) as markers of drowning. In animal experiments, all rats (n = 45) were classified into four groups (drowning, postmortem-submersion, hypoxia and control group). The lungs of experimental animals were analyzed as mRNA expression, immunoblot expression and immunohistochemical staining. qRT-PCR demonstrated increased mRNA expression of RAGE and AQP5 in drowning group compared with that in control, hypoxia and postmortem-submersion group, but not other molecules. Western blotting also showed high expression of RAGE and AQP5 in drowning group, immunostaining of RAGE and AQP5 was highly detected in a linear pattern in type I alveolar epithelial cells, compared with control and postmortem-submersion group. These observations indicate a difference of expression in pulmonary molecular pathology compared with other causes, suggesting RAGE and AQP5 may be useful for differentiation between drowning and postmortem-submersion.
Collapse
Affiliation(s)
- So-Yeon Lee
- Department of Medical Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea.
| | - Eun-Ju Ha
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea.
| | - Hye-Won Cho
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea.
| | - Hye-Rim Kim
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea.
| | - Dongsup Lee
- Department of Clinical Laboratory Science, Hyejeon College, Hongseoung, Chungnam, 32244, Republic of Korea.
| | - Yong-Bin Eom
- Department of Medical Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea; Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea.
| |
Collapse
|
35
|
Egron C, Roszyk L, Rochette E, Jabaudon M, Sapin V, Mulliez A, Labbé A, Coste K. Serum soluble receptor for advanced glycation end-products during acute bronchiolitis in infant: Prospective study in 93 cases. Pediatr Pulmonol 2018; 53:1429-1435. [PMID: 30113140 PMCID: PMC7167909 DOI: 10.1002/ppul.24141] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/25/2018] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Acute bronchiolitis is a major cause of acute respiratory distress in infants. The soluble receptor for advanced glycation end-products (sRAGE) is a biomarker of pulmonary damage processes, with a diagnostic and a prognostic value in acute respiratory distress syndrome (ARDS). The RAGE pathway is also implicated in the pathogenesis of other respiratory diseases like asthma, but the value of sRAGE levels in acute bronchiolitis remains under-investigated. MATERIAL AND METHODS A prospective, observational, and analytical study was conducted at Clermont-Ferrand University Hospital. The main objective was to evaluate the correlation between serum sRAGE and clinical severity of bronchiolitis in hospitalized infants aged <1 year. We analyzed correlations between serum sRAGE and Wainwright score, short-term morbidity attributable to bronchiolitis, causal viruses and risk for recurrent wheezing at 1 year. RESULTS The study included 93 infants. sRAGE levels were significantly lower in acute bronchiolitis patients (mean 1101 pg/mL) than in controls (2203 pg/mL, P < 0.001) but did not correlate with clinical severity. No correlation was found between serum sRAGE and severity score, respiratory viruses, and recurrent wheezing at 1 year. Serum sRAGE levels were negatively correlated with age (r = -0.45, P < 0.001). CONCLUSION Serum sRAGE levels are decreased in acute bronchiolitis but not correlated with disease severity. sRAGE levels should be age-adjusted in infants. Serum sRAGE levels measured in the setting of acute bronchiolitis were not predictive of recurrent wheezing.
Collapse
Affiliation(s)
- Carole Egron
- Department of Pediatrics, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Laurence Roszyk
- Department of Medical Biochemistry and Molecular Biology, CHU Clermont-Ferrand and GReD, Université Clermont Auvergne, CNRS UMR 6293, INSERM U1103, Clermont-Ferrand, France
| | - Emmanuelle Rochette
- Department of Clinical Research for Children (CRECHE) at CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Matthieu Jabaudon
- Department of Perioperative Medicine, CHU Clermont-Ferrand and GReD, Université Clermont Auvergne, CNRS UMR 6293, INSERM U1103, Clermont-Ferrand, France
| | - Vincent Sapin
- Department of Medical Biochemistry and Molecular Biology, CHU Clermont-Ferrand and GReD, Université Clermont Auvergne, CNRS UMR 6293, INSERM U1103, Clermont-Ferrand, France
| | - Aurélien Mulliez
- Department of Clinical Research and Innovation at CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - André Labbé
- Department of Pediatrics, CHU Clermont-Ferrand, Clermont-Ferrand and Université Clermont Auvergne, Clermont-Ferrand, France
| | - Karen Coste
- Department of Pediatrics, CHU Clermont-Ferrand and GReD, Université Clermont Auvergne, CNRS UMR 6293, INSERM U1103, Clermont-Ferrand, France
| |
Collapse
|
36
|
Chapman S, Mick M, Hall P, Mejia C, Sue S, Abdul Wase B, Nguyen MA, Whisenant EC, Wilcox SH, Winden D, Reynolds PR, Arroyo JA. Cigarette smoke extract induces oral squamous cell carcinoma cell invasion in a receptor for advanced glycation end-products-dependent manner. Eur J Oral Sci 2018; 126:33-40. [PMID: 29226456 DOI: 10.1111/eos.12395] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2017] [Indexed: 12/11/2022]
Abstract
Oral squamous cell carcinoma (OSCC) affects approximately 30,000 people and is associated with tobacco use. Little is known about the mechanistic effects of second-hand smoke in the development of OSSC. The receptor for advanced glycation end-products (RAGE) is a surface receptor that is upregulated by second-hand smoke and inhibited by semi-synthetic glycosaminoglycan ethers (SAGEs). Our objective was to determine the role of RAGE during cigarette smoke extract-induced cellular responses and to use SAGEs as a modulating factor of Ca9-22 OSCC cell invasion. Ca9-22 cells were cultured in the presence or absence of cigarette smoke extract and SAGEs. Cell invasion was determined and cells were lysed for western blot analysis. Ras and nuclear factor of kappa light polypeptide gene enhancer in B-cells (NF-κB) activation were determined. Treatment of cells with cigarette smoke extract resulted in: (i) increased invasion of OSCC; (ii) increased RAGE expression; (iii) inhibition of cigarette smoke extract-induced OSCC cell invasion by SAGEs; (iv) increased Ras, increased AKT and NF-κB activation, and downregulation by SAGEs; and (v) increased expression of matrix metalloproteinases (MMPs) 2, 9, and 14, and downregulation by SAGEs. We conclude that cigarette smoke extract increases invasion of OSCC cells in a RAGE-dependent manner. Inhibition of RAGE decreases the levels of its signaling molecules, which results in blocking the cigarette smoke extract-induced invasion.
Collapse
Affiliation(s)
- Steven Chapman
- Lung and Placenta Research Laboratory, Brigham Young University, Physiology and Developmental Biology, Provo, UT, USA
| | - Madison Mick
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, USA
| | - Parker Hall
- Lung and Placenta Research Laboratory, Brigham Young University, Physiology and Developmental Biology, Provo, UT, USA
| | - Camilo Mejia
- Lung and Placenta Research Laboratory, Brigham Young University, Physiology and Developmental Biology, Provo, UT, USA
| | - Stephanie Sue
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, USA
| | - Bihishta Abdul Wase
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, USA
| | - Margaret A Nguyen
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, USA
| | - Evan C Whisenant
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, USA
| | - Shalene H Wilcox
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, USA
| | - Duane Winden
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, USA
| | - Paul R Reynolds
- Lung and Placenta Research Laboratory, Brigham Young University, Physiology and Developmental Biology, Provo, UT, USA
| | - Juan A Arroyo
- Lung and Placenta Research Laboratory, Brigham Young University, Physiology and Developmental Biology, Provo, UT, USA
| |
Collapse
|
37
|
Potential contribution of alveolar epithelial type I cells to pulmonary fibrosis. Biosci Rep 2017; 37:BSR20171301. [PMID: 29026006 PMCID: PMC5696455 DOI: 10.1042/bsr20171301] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/28/2017] [Accepted: 09/29/2017] [Indexed: 12/13/2022] Open
Abstract
Pulmonary fibrosis (PF) is characterized by inflammation and fibrosis of the interstitium and destruction of alveolar histoarchitecture ultimately leading to a fatal impairment of lung function. Different concepts describe either a dominant role of inflammatory pathways or a disturbed remodeling of resident cells of the lung parenchyma during fibrogenesis. Further, a combination of both the mechanisms has been postulated. The present review emphasizes the particular involvement of alveolar epithelial type I cells in all these processes, their contribution to innate immune/inflammatory functions and maintenance of proper alveolar barrier functions. Amongst the different inflammatory and repair events the purinergic receptor P2X7, an ATP-gated cationic channel that regulates not only apoptosis, necrosis, autophagy, and NLPR3 inflammosome activation, but also the turnover of diverse tight junction (TJ) and water channel proteins, seems to be essential for the stability of alveolar barrier integrity and for the interaction with protective factors during lung injury.
Collapse
|
38
|
|
39
|
Lewis JB, Mejia C, Jordan C, Monson TD, Bodine JS, Dunaway TM, Egbert KM, Lewis AL, Wright TJ, Ogden KC, Broberg DS, Hall PD, Nelson SM, Hirschi KM, Reynolds PR, Arroyo JA. Inhibition of the receptor for advanced glycation end-products (RAGE) protects from secondhand smoke (SHS)-induced intrauterine growth restriction IUGR in mice. Cell Tissue Res 2017; 370:513-521. [PMID: 28948356 DOI: 10.1007/s00441-017-2691-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 08/29/2017] [Indexed: 01/20/2023]
Abstract
Intrauterine growth restriction (IUGR) is a disease affecting 10% of all pregnancies. IUGR is associated with maternal, fetal, or placental abnormalities. Studies investigating the effects of secondhand smoke (SHS) exposure and IUGR are limited. The receptor for advanced glycation end-products (RAGE) is a pro-inflammatory transmembrane receptor increased by SHS in the placenta. We tested the hypothesis that inhibition of RAGE during SHS exposure protects from smoke-induced IUGR. C57BL/6 mice were exposed to SHS or SHS + semi-synthetic glycosaminoglycan ethers (SAGEs) known to inhibit RAGE signaling. Trophoblast cells were treated with cigarette smoke extract (CSE) with or without SAGEs in order to address the effects of RAGE inhibition during trophoblast invasion in vitro. SHS-treated mice demonstrated a significant reduction in fetal weight (7.35-fold, P ≤ 0.0001) and placental weight (1.13-fold, P ≤ 0.0001) compared with controls. Mice co-treated with SHS and SAGEs were protected from SHS-induced fetal weights decreases. SHS treatment of C57BL/6 mice activated placental extracellular signal-regulated kinase (ERK) (3.0-fold, P ≤ 0.05), JNK (2.4-fold, P ≤ 0.05) and p38 (2.1-fold, P ≤ 0.05) and the expression of inflammatory mediators including TNF-α (1.34-fold, P ≤ 0.05) and IL-1β (1.03-fold, P ≤ 0.05). SHS-mediated activation of these molecules was reduced to basal levels when SAGE was co-administered. Invasion of trophoblast cells decreased 92% (P < 0.002) when treated with CSE and CSE-mediated invasion was completely reversed by SAGEs. We conclude that RAGE inhibition protects against fetal weight loss during SHS-induced IUGR. These studies provide insight into tobacco-mediated IUGR development and clarify avenues that may be helpful in the alleviation of placental complications.
Collapse
Affiliation(s)
- Joshua B Lewis
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - Camilo Mejia
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - Clinton Jordan
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - Troy D Monson
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - Jared S Bodine
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - Todd M Dunaway
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - Kaleb M Egbert
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - Adam L Lewis
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - Tanner J Wright
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - K Connor Ogden
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - Dallin S Broberg
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - Parker D Hall
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - Shawn M Nelson
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - Kelsey M Hirschi
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - Paul R Reynolds
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA
| | - Juan A Arroyo
- Lung and Placental Research Laboratory, Department of Physiology and Developmental Biology, Brigham Young University, 3052 LSB, Provo, UT, 84602, USA.
| |
Collapse
|
40
|
Chen M, Wang T, Shen Y, Xu D, Li X, An J, Dong J, Li D, Wen F, Chen L. Knockout of RAGE ameliorates mainstream cigarette smoke-induced airway inflammation in mice. Int Immunopharmacol 2017; 50:230-235. [DOI: 10.1016/j.intimp.2017.06.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 06/17/2017] [Accepted: 06/19/2017] [Indexed: 12/23/2022]
|
41
|
Sanders NT, Dutson DJ, Durrant JW, Lewis JB, Wilcox SH, Winden DR, Arroyo JA, Bikman BT, Reynolds PR. Cigarette smoke extract (CSE) induces RAGE-mediated inflammation in the Ca9-22 gingival carcinoma epithelial cell line. Arch Oral Biol 2017; 80:95-100. [PMID: 28399471 DOI: 10.1016/j.archoralbio.2017.03.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 03/20/2017] [Accepted: 03/28/2017] [Indexed: 12/27/2022]
Abstract
OBJECTIVE The oral environment is anatomically positioned as a significant gateway for exposure to environmental toxicants. Cigarette smoke exposure compromises oral health by orchestrating inflammation. The receptor for advanced glycation end-products (RAGE) has been implicated in smoke-induced inflammatory effects; however, its role in the oral cavity is unknown. The purpose of this study was to determine RAGE expression by immortalized gingival carcinoma cells and the degree to which RAGE-mediated signaling influences inflammation. DESIGN Gingival epithelia cells (Ca9-22) were exposed to 10% cigarette smoke extract (CSE) for six hours and screened for RAGE expression and inflammatory mediators. RESULTS Quantitative PCR and immunoblotting revealed increased RAGE expression following exposure. Furthermore, exposure activated RAGE signaling intermediates including Ras and NF-κB. IL-6 and IL-1β were also elevated in cell culture medium from CSE-exposed cells when compared to controls. A family of anionic, partially lipophilic sulfated polysaccharide derivatives known as semi-synthetic glycosaminoglycan ethers (SAGEs) were used in an effort to block RAGE signaling. Co-treatment of CSE and SAGEs ameliorated inflammatory responses. CONCLUSIONS These results provide a new perspective on a mechanism of cigarette smoke induced oral inflammation. Further work may show RAGE signaling as a potential target in the treatment of diseases of the oral cavity exacerbated by tobacco smoke exposure.
Collapse
Affiliation(s)
- Nolan T Sanders
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT, USA
| | - Derek J Dutson
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT, USA
| | - Justin W Durrant
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT, USA
| | - Joshua B Lewis
- Lung and Placenta Research Laboratory, Brigham Young University, Physiology and Developmental Biology, Provo, UT, USA
| | - Shalene H Wilcox
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT, USA
| | - Duane R Winden
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT, USA
| | - Juan A Arroyo
- Lung and Placenta Research Laboratory, Brigham Young University, Physiology and Developmental Biology, Provo, UT, USA
| | - Benjamin T Bikman
- Laboratory of Obesity and Metabolism, Brigham Young University, Physiology and Developmental Biology, Provo, UT, USA
| | - Paul R Reynolds
- Lung and Placenta Research Laboratory, Brigham Young University, Physiology and Developmental Biology, Provo, UT, USA.
| |
Collapse
|
42
|
Receptor for advanced glycation endproducts (RAGE) maintains pulmonary structure and regulates the response to cigarette smoke. PLoS One 2017; 12:e0180092. [PMID: 28678851 PMCID: PMC5497997 DOI: 10.1371/journal.pone.0180092] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 06/09/2017] [Indexed: 12/15/2022] Open
Abstract
The receptor for advanced glycation endproducts (RAGE) is highly expressed in the lung but its physiological functions in this organ is still not completely understood. To determine the contribution of RAGE to physiological functions of the lung, we analyzed pulmonary mechanics and structure of wildtype and RAGE deficient (RAGE-/-) mice. RAGE deficiency spontaneously resulted in a loss of lung structure shown by an increased mean chord length, increased respiratory system compliance, decreased respiratory system elastance and increased concentrations of serum protein albumin in bronchoalveolar lavage fluids. Pulmonary expression of RAGE was mainly localized on alveolar epithelial cells and alveolar macrophages. Primary murine alveolar epithelial cells isolated from RAGE-/- mice revealed an altered differentiation and defective barrier formation under in vitro conditions. Stimulation of interferone-y (IFNy)-activated alveolar macrophages deficient for RAGE with Toll-like receptor (TLR) ligands resulted in significantly decreased release of proinflammatory cytokines and chemokines. Exposure to chronic cigarette smoke did not affect emphysema-like changes in lung parenchyma in RAGE-/- mice. Acute cigarette smoke exposure revealed a modified inflammatory response in RAGE-/- mice that was characterized by an influx of macrophages and a decreased keratinocyte-derived chemokine (KC) release. Our data suggest that RAGE regulates the differentiation of alveolar epithelial cells and impacts on the development and maintenance of pulmonary structure. In cigarette smoke-induced lung pathology, RAGE mediates inflammation that contributes to lung damage.
Collapse
|
43
|
Oczypok EA, Perkins TN, Oury TD. All the "RAGE" in lung disease: The receptor for advanced glycation endproducts (RAGE) is a major mediator of pulmonary inflammatory responses. Paediatr Respir Rev 2017; 23:40-49. [PMID: 28416135 PMCID: PMC5509466 DOI: 10.1016/j.prrv.2017.03.012] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 03/10/2017] [Indexed: 02/07/2023]
Abstract
The receptor for advanced glycation endproducts (RAGE) is a pro-inflammatory pattern recognition receptor (PRR) that has been implicated in the pathogenesis of numerous inflammatory diseases. It was discovered in 1992 on endothelial cells and was named for its ability to bind advanced glycation endproducts and promote vascular inflammation in the vessels of patients with diabetes. Further studies revealed that RAGE is most highly expressed in lung tissue and spurred numerous explorations into RAGE's role in the lung. These studies have found that RAGE is an important mediator in allergic airway inflammation (AAI) and asthma, pulmonary fibrosis, lung cancer, chronic obstructive pulmonary disease (COPD), acute lung injury, pneumonia, cystic fibrosis, and bronchopulmonary dysplasia. RAGE has not yet been targeted in the lungs of paediatric or adult clinical populations, but the development of new ways to inhibit RAGE is setting the stage for the emergence of novel therapeutic agents for patients suffering from these pulmonary conditions.
Collapse
Affiliation(s)
| | | | - Tim D. Oury
- Corresponding author. Tel.: +1 412 648 9659; Fax: +1 412 648 9527
| |
Collapse
|
44
|
Lee H, Park JR, Kim WJ, Sundar IK, Rahman I, Park SM, Yang SR. Blockade of RAGE ameliorates elastase-induced emphysema development and progression via RAGE-DAMP signaling. FASEB J 2017; 31:2076-2089. [PMID: 28148566 DOI: 10.1096/fj.201601155r] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/17/2017] [Indexed: 12/21/2022]
Abstract
The receptor for advanced glycan end products (RAGE) has been identified as a susceptibility gene for chronic obstructive pulmonary disease (COPD) in genome-wide association studies (GWASs). However, less is known about how RAGE is involved in the pathogenesis of COPD. To determine the molecular mechanism by which RAGE influences COPD in experimental COPD models, we investigated the efficacy of the RAGE-specific antagonist FPS-ZM1 administration in in vivo and in vitro COPD models. We injected elastase intratracheally and the RAGE antagonist FPS-ZM1 in mice, and the infiltrated inflammatory cells and cytokines were assessed by ELISA. Cellular expression of RAGE was determined in protein, serum, and bronchoalveolar lavage fluid of mice and lungs and serum of human donors and patients with COPD. Downstream damage-associated molecular pattern (DAMP) pathway activation in vivo and in vitro and in patients with COPD was assessed by immunofluorescence staining, Western blot analysis, and ELISA. The expression of membrane RAGE in initiating the inflammatory response and of soluble RAGE acting as a decoy were associated with up-regulation of the DAMP-related signaling pathway via Nrf2. FPS-ZM1 administration significantly reversed emphysema in the lung of mice. Moreover, FPS-ZM1 treatment significantly reduced lung inflammation in Nrf2+/+ , but not in Nrf2-/- mice. Thus, our data indicate for the first time that RAGE inhibition has an essential protective role in COPD. Our observation of RAGE inhibition provided novel insight into its potential as a therapeutic target in emphysema/COPD.-Lee, H., Park, J.-R., Kim, W. J., Sundar, I. K., Rahman, I., Park, S.-M., Yang. S.-R. Blockade of RAGE ameliorates elastase-induced emphysema development and progression via RAGE-DAMP signaling.
Collapse
Affiliation(s)
- Hanbyeol Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, South Korea
| | - Jeong-Ran Park
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, South Korea
| | - Woo Jin Kim
- Department of Internal Medicine, Kangwon National University, Chuncheon, South Korea; and
| | - Isaac K Sundar
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Sung-Min Park
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, South Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, South Korea;
| |
Collapse
|
45
|
Machahua C, Montes-Worboys A, Llatjos R, Escobar I, Dorca J, Molina-Molina M, Vicens-Zygmunt V. Increased AGE-RAGE ratio in idiopathic pulmonary fibrosis. Respir Res 2016; 17:144. [PMID: 27816054 PMCID: PMC5097848 DOI: 10.1186/s12931-016-0460-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 10/28/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The abnormal epithelial-mesenchymal restorative capacity in idiopathic pulmonary fibrosis (IPF) has been recently associated with an accelerated aging process as a key point for the altered wound healing. The advanced glycation end-products (AGEs) are the consequence of non-enzymatic reactions between lipid and protein with several oxidants in the aging process. The receptor for AGEs (RAGEs) has been implicated in the lung fibrotic process and the alveolar homeostasis. However, this AGE-RAGE aging pathway has been under-explored in IPF. METHODS Lung samples from 16 IPF and 9 control patients were obtained through surgical lung biopsy. Differences in AGEs and RAGE expression between both groups were evaluated by RT-PCR, Western blot and immunohistochemistry. The effect of AGEs on cell viability of primary lung fibrotic fibroblasts and alveolar epithelial cells was assessed. Cell transformation of fibrotic fibroblasts cultured into glycated matrices was evaluated in different experimental conditions. RESULTS Our study demonstrates an increase of AGEs together with a decrease of RAGEs in IPF lungs, compared with control samples. Two specific AGEs involved in aging, pentosidine and Nε-Carboxymethyl lysine, were significantly increased in IPF samples. The immunohistochemistry identified higher staining of AGEs related to extracellular matrix (ECM) proteins and the apical surface of the alveolar epithelial cells (AECs) surrounding fibroblast foci in fibrotic lungs. On the other hand, RAGE location was present at the cell membrane of AECs in control lungs, while it was almost missing in pulmonary fibrotic tissue. In addition, in vitro cultures showed that the effect of AGEs on cell viability was different for AECs and fibrotic fibroblasts. AGEs decreased cell viability in AECs, even at low concentration, while fibroblast viability was less affected. Furthermore, fibroblast to myofibroblast transformation could be enhanced by ECM glycation. CONCLUSIONS All of these findings suggest a possible role of the increased ratio AGEs-RAGEs in IPF, which could be a relevant accelerating aging tissue reaction in the abnormal wound healing of the lung fibrotic process.
Collapse
Affiliation(s)
- Carlos Machahua
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
| | - Ana Montes-Worboys
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
- Research Network in Respiratory Diseases (CIBERES), Madrid, Spain
| | - Roger Llatjos
- Department of Pathology, University Hospital of Bellvitge, Barcelona, Spain
| | - Ignacio Escobar
- Department of Thoracic Surgery, University Hospital of Bellvitge, Barcelona, Spain
| | - Jordi Dorca
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
- Research Network in Respiratory Diseases (CIBERES), Madrid, Spain
| | - Maria Molina-Molina
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
- Research Network in Respiratory Diseases (CIBERES), Madrid, Spain
| | - Vanesa Vicens-Zygmunt
- Pneumology Research Group, IDIBELL, University of Barcelona, Barcelona, Spain
- Department of Pneumology, Unit of Interstitial Lung Diseases, University Hospital of Bellvitge, Barcelona, Spain
| |
Collapse
|
46
|
Waseda K, Miyahara N, Taniguchi A, Kurimoto E, Ikeda G, Koga H, Fujii U, Yamamoto Y, Gelfand EW, Yamamoto H, Tanimoto M, Kanehiro A. Emphysema requires the receptor for advanced glycation end-products triggering on structural cells. Am J Respir Cell Mol Biol 2016; 52:482-91. [PMID: 25188021 DOI: 10.1165/rcmb.2014-0027oc] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Pulmonary emphysema is characterized by persistent inflammation and progressive alveolar destruction. The receptor for advanced glycation end-products (RAGE) is a multiligand cell surface receptor reported to be involved in the process of acute alveolar epithelial cell injury. However, studies that address the role of RAGE in pulmonary emphysema are inconclusive. We investigated the role of RAGE in the development of elastase-induced pulmonary inflammation and emphysema in mice. RAGE-sufficient (RAGE(+/+)) mice and RAGE-deficient (RAGE(-/-)) mice were treated with intratracheal elastase on Day 0. Airway inflammation, static lung compliance, lung histology, and the levels of neutrophil-related chemokine and proinflammatory cytokines in bronchoalveolar lavage fluid were determined on Days 4 and 21. Neutrophilia in bronchoalveolar lavage fluid, seen in elastase-treated RAGE(+/+) mice, was reduced in elastase-treated RAGE(-/-) mice on Day 4, and was associated with decreased levels of keratinocyte chemoattractant, macrophage inflammatory protein-2, and IL-1β. Static lung compliance values and emphysematous changes in the lung tissue were decreased in RAGE(-/-) mice compared with RAGE(+/+) mice on Day 21 after elastase treatment. Experiments using irradiated, bone marrow-chimeric mice showed that the mice expressing RAGE on radioresistant structural cells, but not hematopoietic cells, developed elastase-induced neutrophilia and emphysematous change in the lung. In contrast, mice expressing RAGE on hematopoietic cells, but not radioresistant structural cells, showed reduced neutrophilia and emphysematous change in the lung. These data identify the importance of RAGE expressed on lung structural cells in the development of elastase-induced pulmonary inflammation and emphysema. Thus, RAGE represents a novel therapeutic target for preventing pulmonary emphysema.
Collapse
Affiliation(s)
- Koichi Waseda
- 1 Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Achouiti A, de Vos AF, van ‘t Veer C, Florquin S, Tanck MW, Nawroth PP, Bierhaus A, van der Poll T, van Zoelen MAD. Receptor for Advanced Glycation End Products (RAGE) Serves a Protective Role during Klebsiella pneumoniae - Induced Pneumonia. PLoS One 2016; 11:e0141000. [PMID: 26824892 PMCID: PMC4732606 DOI: 10.1371/journal.pone.0141000] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 10/02/2015] [Indexed: 01/01/2023] Open
Abstract
Klebsiella species is the second most commonly isolated gram-negative organism in sepsis and a frequent causative pathogen in pneumonia. The receptor for advanced glycation end products (RAGE) is expressed on different cell types and plays a key role in diverse inflammatory responses. We here aimed to investigate the role of RAGE in the host response to Klebsiella (K.) pneumoniae pneumonia and intransally inoculated rage gene deficient (RAGE-/-) and normal wild-type (Wt) mice with K. pneumoniae. Klebsiella pneumonia resulted in an increased pulmonary expression of RAGE. Furthermore, the high-affinity RAGE ligand high mobility group box-1 was upregulated during K. pneumoniae pneumonia. RAGE deficiency impaired host defense as reflected by a worsened survival, increased bacterial outgrowth and dissemination in RAGE-/- mice. RAGE-/- neutrophils showed a diminished phagocytosing capacity of live K. pneumoniae in vitro. Relative to Wt mice, RAGE-/- mice demonstrated similar lung inflammation, and slightly elevated—if any—cytokine and chemokine levels and unchanged hepatocellular injury. In addition, RAGE-/- mice displayed an unaltered response to intranasally instilled Klebsiella lipopolysaccharide (LPS) with respect to pulmonary cell recruitment and local release of cytokines and chemokines. These data suggest that (endogenous) RAGE protects against K. pneumoniae pneumonia. Also, they demonstrate that RAGE contributes to an effective antibacterial defense during K. pneumoniae pneumonia, at least partly via its participation in the phagocytic properties of professional granulocytes. Additionally, our results indicate that RAGE is not essential for the induction of a local and systemic inflammatory response to either intact Klebsiella or Klebsiella LPS.
Collapse
Affiliation(s)
- Ahmed Achouiti
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Alex F. de Vos
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Cornelis van ‘t Veer
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Sandrine Florquin
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Michael W. Tanck
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Peter P. Nawroth
- Department of Internal Medicine and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - Angelika Bierhaus
- Department of Internal Medicine and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - Tom van der Poll
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Marieke A. D. van Zoelen
- Laboratory of Biomedical Science, Feinstein Institute for Medical Research, North Shore Long Island University Hospital, Manhassat, New York, United States of America
- Division of Internal Medicine and Infectious Diseases, University Medical Center of Utrecht, Utrecht, the Netherlands
- Laboratory of Translational Immunology (LTI), University Medical Center of Utrecht, Utrecht, the Netherlands
- * E-mail:
| |
Collapse
|
48
|
Gangemi S, Casciaro M, Trapani G, Quartuccio S, Navarra M, Pioggia G, Imbalzano E. Association between HMGB1 and COPD: A Systematic Review. Mediators Inflamm 2015; 2015:164913. [PMID: 26798204 PMCID: PMC4698778 DOI: 10.1155/2015/164913] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/12/2015] [Accepted: 12/03/2015] [Indexed: 01/02/2023] Open
Abstract
HMGB1 is an alarmin, a protein that warns and activates inflammation. Chronic obstructive pulmonary disease (COPD) is characterised by a progressive airflow obstruction and airway inflammation. Current anti-inflammatory therapies are poorly effective in maintaining lung function and symptoms of COPD. This underlines the need for finding new molecular targets involved in disease pathogenesis in order to block pathology progression. This review aims to analyse latest advances on HMGB1 role, utilisation, and potential application in COPD. To this purpose we reviewed experimental studies that investigated this alarmin as marker as well as a potential treatment in chronic obstructive pulmonary disease. This systematic review was conducted according to PRISMA guidelines. In almost all the studies, it emerged that HMGB1 levels are augmented in smokers and in patients affected by COPD. It emerged that cigarette smoking, the most well-known causative factor of COPD, induces neutrophils death and necrosis. The necrosis of neutrophil cells leads to HMGB1 release, which recruits other neutrophils in a self-maintaining process. According to the results reported in the paper both inhibiting HMGB1 and its receptor (RAGE) and blocking neutrophils necrosis (inducted by cigarette smoking) could be the aim for further studies.
Collapse
Affiliation(s)
- Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University Hospital “G. Martino”, University of Messina, 98125 Messina, Italy
| | - Marco Casciaro
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University Hospital “G. Martino”, University of Messina, 98125 Messina, Italy
| | - Giovanni Trapani
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Sebastiano Quartuccio
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Michele Navarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, University Pole Annunziata, 98168 Messina, Italy
| | - Giovanni Pioggia
- Institute of Applied Sciences and Intelligent Systems (ISASI), Messina Unit, 98100 Messina, Italy
| | - Egidio Imbalzano
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| |
Collapse
|
49
|
Alexander KL, Mejia CA, Jordan C, Nelson MB, Howell BM, Jones CM, Reynolds PR, Arroyo JA. Differential Receptor for Advanced Glycation End Products Expression in Preeclamptic, Intrauterine Growth Restricted, and Gestational Diabetic Placentas. Am J Reprod Immunol 2015; 75:172-80. [DOI: 10.1111/aji.12462] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/18/2015] [Indexed: 01/09/2023] Open
Affiliation(s)
- Kristen L. Alexander
- Lung and Placenta Research Laboratory; Physiology and Developmental Biology; Brigham Young University; Provo UT USA
| | - Camilo A. Mejia
- Lung and Placenta Research Laboratory; Physiology and Developmental Biology; Brigham Young University; Provo UT USA
| | - Clinton Jordan
- Lung and Placenta Research Laboratory; Physiology and Developmental Biology; Brigham Young University; Provo UT USA
| | - Michael B. Nelson
- Lung and Placenta Research Laboratory; Physiology and Developmental Biology; Brigham Young University; Provo UT USA
| | - Brian M. Howell
- Lung and Placenta Research Laboratory; Physiology and Developmental Biology; Brigham Young University; Provo UT USA
| | - Cameron M. Jones
- Lung and Placenta Research Laboratory; Physiology and Developmental Biology; Brigham Young University; Provo UT USA
| | - Paul R. Reynolds
- Lung and Placenta Research Laboratory; Physiology and Developmental Biology; Brigham Young University; Provo UT USA
| | - Juan A. Arroyo
- Lung and Placenta Research Laboratory; Physiology and Developmental Biology; Brigham Young University; Provo UT USA
| |
Collapse
|
50
|
DeChristopher LR, Uribarri J, Tucker KL. Intake of high fructose corn syrup sweetened soft drinks is associated with prevalent chronic bronchitis in U.S. Adults, ages 20-55 y. Nutr J 2015; 14:107. [PMID: 26474970 PMCID: PMC4609055 DOI: 10.1186/s12937-015-0097-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 10/07/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND High fructose corn syrup (HFCS) sweetened soft drink intake has been linked with asthma in US high-schoolers. Intake of beverages with excess free fructose (EFF), including apple juice, and HFCS sweetened fruit drinks and soft drinks, has been associated with asthma in children. One hypothesis for this association is that underlying fructose malabsorption and fructose reactivity in the GI may contribute to in situ formation of enFruAGEs. EnFruAGEs may be an overlooked source of advanced glycation end-products (AGE) that contribute to lung disease. AGE/ RAGEs are elevated in COPD lungs. EFF intake has increased in recent decades, and intakes may exceed dosages associated with adult fructose malabsorption in subsets of the population. Intestinal dysfunction has been shown to be elevated in COPD patients. The objective of this study was to investigate the association between HFCS sweetened soft drink intake and chronic bronchitis (CB), a common manifestation of COPD, in adults. METHODS DESIGN In this cross sectional analysis, the outcome variable was self-reported existing chronic bronchitis or history of CB. Exposure variable was non-diet soda. Rao Scott Ҳ(2) was used for prevalence differences and logistic regression for associations, adjusted for age, sex, race-ethnicity, BMI, smoking, exposure to in-home smoking, pre-diabetes, diabetes, SES, total energy and total fruits and beverages consumption. SETTING Data are from the National Health and Nutrition Examination Survey 2003-2006. SUBJECTS 2801 adults aged 20-55 y. RESULTS There was a statistically significant correlation between intake of non-diet soft drinks and greater prevalence and odds of chronic bronchitis (p < 0.05). Independent of all covariates, intake of non-diet soda ≥5 times a week (vs. non/low non-diet soda) was associated with nearly twice the likelihood of having chronic bronchitis (OR = 1.80; p = 0.047; 95% CI 1.01-3.20). CONCLUSIONS HFCS sweetened soft drink intake is correlated with chronic bronchitis in US adults aged 20-55 y, after adjusting for covariates, including smoking. Results support the hypothesis that underlying fructose malabsorption and fructose reactivity in the GI may contribute to chronic bronchitis, perhaps through in situ formation of enFruAGEs, which may contribute to lung disease. Longitudinal and biochemical research is needed to confirm and clarify the mechanisms involved.
Collapse
Affiliation(s)
- Luanne Robalo DeChristopher
- Biochemistry, Molecular Biology, NY Medical College, Valhalla, NY, USA.
- , P.O. Box 5542, Eugene, OR, 97405, USA.
| | - Jaime Uribarri
- Department of Medicine, the Icahn School of Medicine, New York, NY, USA
| | - Katherine L Tucker
- Department of Clinical Laboratory and Nutritional Sciences, University of Massachusetts, Lowell, MA, USA
| |
Collapse
|