1
|
Su E, Song X, Wei L, Xue J, Cheng X, Xie S, Jiang H, Liu M. Endothelial GSDMD underlies LPS-induced systemic vascular injury and lethality. JCI Insight 2025; 10:e182398. [PMID: 39927458 PMCID: PMC11948583 DOI: 10.1172/jci.insight.182398] [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: 04/25/2024] [Accepted: 12/17/2024] [Indexed: 02/11/2025] Open
Abstract
Endothelial injury destroys endothelial barrier integrity, triggering organ dysfunction and ultimately resulting in sepsis-related death. Considerable attention has been focused on identifying effective targets for inhibiting damage to endothelial cells to treat endotoxemia-induced septic shock. Global gasdermin D (Gsdmd) deletion reportedly prevents death caused by endotoxemia. However, the role of endothelial GSDMD in endothelial injury and lethality in lipopolysaccharide-induced (LPS-induced) endotoxemia and the underlying regulatory mechanisms are unknown. Here, we show that LPS increases endothelial GSDMD level in aortas and lung microvessels. We demonstrated that endothelial Gsdmd deficiency, but not myeloid cell Gsdmd deletion, protects against endothelial injury and death in mice with endotoxemia or sepsis. In vivo experiments suggested that hepatocyte GSDMD mediated the release of high-mobility group box 1, which subsequently binds to the receptor for advanced glycation end products in endothelial cells to cause systemic vascular injury, ultimately resulting in acute lung injury and lethality in shock driven by endotoxemia or sepsis. Additionally, inhibiting endothelial GSDMD activation via a polypeptide inhibitor alleviated endothelial damage and improved survival in a mouse model of endotoxemia or sepsis. These data suggest that endothelial GSDMD is a viable pharmaceutical target for treating endotoxemia and endotoxemia-induced sepsis.
Collapse
Affiliation(s)
- Enyong Su
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyue Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Lili Wei
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cardiology, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai, China
| | - Junqiang Xue
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xuelin Cheng
- Department of Health Management Center, Zhongshan Hospital, and
- Department of General Practice, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shiyao Xie
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hong Jiang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, National Clinical Research Center for Interventional Medicine, Shanghai, China
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Ming Liu
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Health Management Center, Zhongshan Hospital, and
- Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Di Carlo C, Cimini C, Belda-Perez R, Valbonetti L, Bernabò N, Barboni B. Navigating the Intersection of Glycemic Control and Fertility: A Network Perspective. Int J Mol Sci 2024; 25:9967. [PMID: 39337455 PMCID: PMC11432572 DOI: 10.3390/ijms25189967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/10/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
The rising incidence of metabolic diseases is linked to elevated blood glucose levels, contributing to conditions such as diabetes and promoting the accumulation of advanced glycation end products (AGEs). AGEs, formed by non-enzymatic reactions between sugars and proteins, build up in tissues and are implicated in various diseases. This article explores the relationship between glycemic control and AGE accumulation, focusing on fertility implications. A computational model using network theory was developed, featuring a molecular database and a network with 145 nodes and 262 links, categorized as a Barabasi-Albert scale-free network. Three main subsets of nodes emerged, centered on glycemic control, fertility, and immunity, with AGEs playing a critical role. The transient receptor potential vanilloid 1 (TRPV1), a receptor expressed in several tissues including sperm, was identified as a key hub, suggesting that the modulation of TRPV1 in sperm by AGEs may influence fertility. Additionally, a novel link between glycemic control and immunity was found, indicating that immune cells may play a role in endocytosing specific AGEs. This discovery underscores the complex interplay between glycemic control and immune function, with significant implications for metabolic, immune health, and fertility.
Collapse
Affiliation(s)
- Carlo Di Carlo
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Costanza Cimini
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Ramses Belda-Perez
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Physiology, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, 30100 Murcia, Spain
| | - Luca Valbonetti
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Institute of Biochemistry and Cell Biology (CNR-IBBC/EMMA/Infrafrontier/IMPC), National Research Council, Monterotondo Scalo, 00015 Rome, Italy
| | - Nicola Bernabò
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Institute of Biochemistry and Cell Biology (CNR-IBBC/EMMA/Infrafrontier/IMPC), National Research Council, Monterotondo Scalo, 00015 Rome, Italy
| | - Barbara Barboni
- Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
3
|
Werder RB, Zhou X, Cho MH, Wilson AA. Breathing new life into the study of COPD with genes identified from genome-wide association studies. Eur Respir Rev 2024; 33:240019. [PMID: 38811034 PMCID: PMC11134200 DOI: 10.1183/16000617.0019-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 02/23/2024] [Indexed: 05/31/2024] Open
Abstract
COPD is a major cause of morbidity and mortality globally. While the significance of environmental exposures in disease pathogenesis is well established, the functional contribution of genetic factors has only in recent years drawn attention. Notably, many genes associated with COPD risk are also linked with lung function. Because reduced lung function precedes COPD onset, this association is consistent with the possibility that derangements leading to COPD could arise during lung development. In this review, we summarise the role of leading genes (HHIP, FAM13A, DSP, AGER and TGFB2) identified by genome-wide association studies in lung development and COPD. Because many COPD genome-wide association study genes are enriched in lung epithelial cells, we focus on the role of these genes in the lung epithelium in development, homeostasis and injury.
Collapse
Affiliation(s)
- Rhiannon B Werder
- Murdoch Children's Research Institute, Melbourne, Australia
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew A Wilson
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
4
|
de Souza Xavier Costa N, da Costa Sigrist G, Schalch AS, Belotti L, Dolhnikoff M, da Silva LFF. Lung tissue expression of epithelial injury markers is associated with acute lung injury severity but does not discriminate sepsis from ARDS. Respir Res 2024; 25:129. [PMID: 38500106 PMCID: PMC10949726 DOI: 10.1186/s12931-024-02761-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: 09/12/2023] [Accepted: 03/08/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a common cause of respiratory failure in critically ill patients, and diffuse alveolar damage (DAD) is considered its histological hallmark. Sepsis is one of the most common aetiology of ARDS with the highest case-fatality rate. Identifying ARDS patients and differentiate them from other causes of acute respiratory failure remains a challenge. To address this, many studies have focused on identifying biomarkers that can help assess lung epithelial injury. However, there is scarce information available regarding the tissue expression of these markers. Evaluating the expression of elafin, RAGE, and SP-D in lung tissue offers a potential bridge between serological markers and the underlying histopathological changes. Therefore, we hypothesize that the expression of epithelial injury markers varies between sepsis and ARDS as well as according to its severity. METHODS We compared the post-mortem lung tissue expression of the epithelial injury markers RAGE, SP-D, and elafin of patients that died of sepsis, ARDS, and controls that died from non-pulmonary causes. Lung tissue was collected during routine autopsy and protein expression was assessed by immunohistochemistry. We also assessed the lung injury by a semi-quantitative analysis. RESULTS We observed that all features of DAD were milder in septic group compared to ARDS group. Elafin tissue expression was increased and SP-D was decreased in the sepsis and ARDS groups. Severe ARDS expressed higher levels of elafin and RAGE, and they were negatively correlated with PaO2/FiO2 ratio, and positively correlated with bronchopneumonia percentage and hyaline membrane score. RAGE tissue expression was negatively correlated with mechanical ventilation duration in both ARDS and septic groups. In septic patients, elafin was positively correlated with ICU admission length, SP-D was positively correlated with serum lactate and RAGE was correlated with C-reactive protein. CONCLUSIONS Lung tissue expression of elafin and RAGE, but not SP-D, is associated with ARDS severity, but does not discriminate sepsis patients from ARDS patients.
Collapse
Affiliation(s)
| | - Giovana da Costa Sigrist
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, LIM-05, Brazil
| | - Alexandre Santos Schalch
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, LIM-05, Brazil
| | - Luciano Belotti
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, LIM-05, Brazil
| | - Marisa Dolhnikoff
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, LIM-05, Brazil
| | - Luiz Fernando Ferraz da Silva
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, LIM-05, Brazil
- Serviço de Verificação de Óbitos da Capital, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
5
|
Carrión-Barberà I, Triginer L, Tío L, Pérez-García C, Ribes A, Abad V, Pros A, Monfort J, Salman-Monte TC. Serum Advanced Glycation End Products and Their Soluble Receptor as New Biomarkers in Systemic Lupus Erythematosus. Biomedicines 2024; 12:610. [PMID: 38540223 PMCID: PMC10968350 DOI: 10.3390/biomedicines12030610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 11/11/2024] Open
Abstract
It has been postulated that advanced glycation end products (AGEs) and their soluble receptor (sRAGE) may play a relevant role as inducers in the chronic inflammatory pathway in various conditions, among them, in immune-mediated diseases such as systemic lupus erythematosus (SLE). However, previous studies show conflicting results about their association with SLE characteristics and their usefulness as disease biomarkers. We aimed to study the association of specific serum AGEs (pentosidine, Nξ-(carboxymethyl)lysine (CML), Nξ-(carboxyethyl)lysine (CEL)), sRAGE levels and AGEs (specific serum AGEs and skin AGEs) to sRAGE ratios with various disease parameters, in order to clarify their potential as new biomarkers in SLE and to study their relationship with cardiovascular disease (CVD). To this aim, serum pentosidine, CML, CEL and sRAGE were measured via ELISA, and skin AGEs levels were measured by skin autofluorescence. Correlations of pentosidine levels with demographic and clinical data, indexes of activity, accrual damage and patient-reported outcomes were analyzed through multiple linear regression models, while correlations of the rest of the AGEs, sRAGE and AGE to sRAGE ratios (non-normal) were analyzed using both an OLS regression model and a GML. All of the analyses were adjusted for confounders. A total of 119 SLE patients were recruited. Serum AGEs and sRAGEs were significantly associated with SLE activity indexes and/or demographic or disease characteristics: pentosidine with pulmonary manifestations; CML with anti-dsDNA antibodies, IL-6, disease duration and non-Caucasian ethnicities; CEL with anti-dsDNA antibodies, IL-6 and accumulated number of manifestations; and sRAGE with male gender, photosensitivity and being on specific immunosuppressants. These results suggest that the AGE-sRAGE axis may serve as a novel biomarker for managing and prognosticating this disease. Its correlation with certain antibodies, demographics and disease presentations may indicate a distinct clinical phenotype associated with varying levels of AGEs and/or sRAGE. The significance of specific AGE/sRAGE ratios, introduced in this study for the first time, warrants additional investigation in forthcoming research. Our study did not confirm the link between serum AGEs and CVD, which merits further exploration through studies designed for this specific purpose.
Collapse
Affiliation(s)
- Irene Carrión-Barberà
- Rheumatology Department, Hospital del Mar, 08003 Barcelona, Spain; (I.C.-B.)
- Medicine Department, Medicine Faculty, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Hospital del Mar Research Institute, 08003 Barcelona, Spain
- Clinical Expertise Unit (UEC) in Systemic Autoimmune Diseases and Vasculitis, 08003 Barcelona, Spain
| | - Laura Triginer
- Hospital del Mar Research Institute, 08003 Barcelona, Spain
| | - Laura Tío
- Hospital del Mar Research Institute, 08003 Barcelona, Spain
| | - Carolina Pérez-García
- Rheumatology Department, Hospital del Mar, 08003 Barcelona, Spain; (I.C.-B.)
- Hospital del Mar Research Institute, 08003 Barcelona, Spain
- Clinical Expertise Unit (UEC) in Systemic Autoimmune Diseases and Vasculitis, 08003 Barcelona, Spain
| | - Anna Ribes
- Hospital del Mar Research Institute, 08003 Barcelona, Spain
| | - Victoria Abad
- Rheumatology Department, Hospital del Mar, 08003 Barcelona, Spain; (I.C.-B.)
- Clinical Expertise Unit (UEC) in Systemic Autoimmune Diseases and Vasculitis, 08003 Barcelona, Spain
| | - Ana Pros
- Rheumatology Department, Hospital del Mar, 08003 Barcelona, Spain; (I.C.-B.)
- Clinical Expertise Unit (UEC) in Systemic Autoimmune Diseases and Vasculitis, 08003 Barcelona, Spain
| | - Jordi Monfort
- Rheumatology Department, Hospital del Mar, 08003 Barcelona, Spain; (I.C.-B.)
- Hospital del Mar Research Institute, 08003 Barcelona, Spain
- Clinical Expertise Unit (UEC) in Systemic Autoimmune Diseases and Vasculitis, 08003 Barcelona, Spain
| | - Tarek Carlos Salman-Monte
- Rheumatology Department, Hospital del Mar, 08003 Barcelona, Spain; (I.C.-B.)
- Hospital del Mar Research Institute, 08003 Barcelona, Spain
- Clinical Expertise Unit (UEC) in Systemic Autoimmune Diseases and Vasculitis, 08003 Barcelona, Spain
| |
Collapse
|
6
|
Xia J, Wang J, Ying L, Huang R, Zhang K, Zhang R, Tang W, Xu Q, Lai D, Zhang Y, Hu Y, Zhang X, Zang R, Fan J, Shu Q, Xu J. RAGE Is a Receptor for SARS-CoV-2 N Protein and Mediates N Protein-induced Acute Lung Injury. Am J Respir Cell Mol Biol 2023; 69:508-520. [PMID: 37478333 PMCID: PMC10633841 DOI: 10.1165/rcmb.2022-0351oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 07/19/2023] [Indexed: 07/23/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleocapsid protein (N-protein) increases early in body fluids during infection and has recently been identified as a direct inducer for lung injury. However, the signal mechanism of N-protein in the lung inflammatory response remains poorly understood. The goal of this study was to determine whether RAGE (receptor for advanced glycation endproducts) participated in N-protein-induced acute lung injury. The binding between N-protein and RAGE was examined via assays for protein-protein interaction. To determine the signaling mechanism in vitro, cells were treated with recombinant N-protein and assayed for the activation of the RAGE/MAPK (mitogen-activated protein kinase)/NF-ĸB pathway. RAGE deficiency mice and antagonist were used to study N-protein-induced acute lung injury in vivo. Binding between N-protein and RAGE was confirmed via flow cytometry-based binding assay, surface plasmon resonance, and ELISA. Pull-down and coimmunoprecipitation assays revealed that N-protein bound RAGE via both N-terminal and C-terminal domains. In vitro, N-protein activated the RAGE-ERK1/2-NF-ĸB signaling pathway and induced a proinflammatory response. RAGE deficiency subdued N-protein-induced proinflammatory signaling and response. In vivo, RAGE was upregulated in the BAL and lung tissue after recombinant N-protein insult. RAGE deficiency and small molecule antagonist partially protected mice from N-protein-induced acute lung injury. Our study demonstrated that RAGE is a receptor for N-protein. RAGE is partially responsible for N-protein-induced acute lung injury and has the potential to become a therapeutic target for treating coronavirus disease.
Collapse
Affiliation(s)
- Jie Xia
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Jiangmei Wang
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Liyang Ying
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Ruoqiong Huang
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Kai Zhang
- The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; and
| | - Ruoyang Zhang
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Wenqi Tang
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Qi Xu
- Hangzhou Medical College, Hangzhou, China
| | - Dengming Lai
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Yan Zhang
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Yaoqin Hu
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiaodie Zhang
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Ruoxi Zang
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Jiajie Fan
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Qiang Shu
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Jianguo Xu
- The Children’s Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
7
|
Katsoulis O, Guo R, Singanayagam A. When It Comes to COVID-19, Inflammation Is All the RAGE. Am J Respir Cell Mol Biol 2023; 69:489-490. [PMID: 37489933 PMCID: PMC10633835 DOI: 10.1165/rcmb.2023-0227ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023] Open
Affiliation(s)
- Orestis Katsoulis
- Department of Infectious Disease Imperial College London London, United Kingdom
| | - Rui Guo
- Department of Critical Care Medicine The First Affiliated Hospital of Chongqing Medical University Chongqing, China
| | - Aran Singanayagam
- Department of Infectious Disease Imperial College London London, United Kingdom
| |
Collapse
|
8
|
Reynaert NL, Vanfleteren LEGW, Perkins TN. The AGE-RAGE Axis and the Pathophysiology of Multimorbidity in COPD. J Clin Med 2023; 12:jcm12103366. [PMID: 37240472 DOI: 10.3390/jcm12103366] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/24/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease of the airways and lungs due to an enhanced inflammatory response, commonly caused by cigarette smoking. Patients with COPD are often multimorbid, as they commonly suffer from multiple chronic (inflammatory) conditions. This intensifies the burden of individual diseases, negatively affects quality of life, and complicates disease management. COPD and comorbidities share genetic and lifestyle-related risk factors and pathobiological mechanisms, including chronic inflammation and oxidative stress. The receptor for advanced glycation end products (RAGE) is an important driver of chronic inflammation. Advanced glycation end products (AGEs) are RAGE ligands that accumulate due to aging, inflammation, oxidative stress, and carbohydrate metabolism. AGEs cause further inflammation and oxidative stress through RAGE, but also through RAGE-independent mechanisms. This review describes the complexity of RAGE signaling and the causes of AGE accumulation, followed by a comprehensive overview of alterations reported on AGEs and RAGE in COPD and in important co-morbidities. Furthermore, it describes the mechanisms by which AGEs and RAGE contribute to the pathophysiology of individual disease conditions and how they execute crosstalk between organ systems. A section on therapeutic strategies that target AGEs and RAGE and could alleviate patients from multimorbid conditions using single therapeutics concludes this review.
Collapse
Affiliation(s)
- Niki L Reynaert
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, 6229 ER Maastricht, The Netherlands
| | - Lowie E G W Vanfleteren
- COPD Center, Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Timothy N Perkins
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
9
|
Bajwa S, Luebbe A, Vo NDN, Piskor EM, Kosan C, Wolf G, Loeffler I. RAGE is a critical factor of sex-based differences in age-induced kidney damage. Front Physiol 2023; 14:1154551. [PMID: 37064891 PMCID: PMC10090518 DOI: 10.3389/fphys.2023.1154551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
Introduction: Advanced glycation end products (AGEs) are a heterogeneous group of molecules with potential pathophysiological effects on the kidneys. Fibrosis together with the accumulation of AGEs has been investigated for its contribution to age-related decline in renal function. AGEs mediate their effects in large parts through their interactions with the receptor for AGEs (RAGE). RAGE is a transmembrane protein that belongs to the immunoglobulin superfamily and has the ability to interact with multiple pro-inflammatory/pro-oxidative ligands. The role of RAGE in aging kidneys has not been fully characterized, especially for sex-based differences. Methods: Therefore, we analyzed constitutive RAGE knockout (KO) mice in an age- and sex-dependent manner. Paraffin-embedded kidney sections were used for histological analysis and protein expression of fibrosis and damage markers. RNA expression analysis from the kidney cortex was done by qPCR for AGE receptors, kidney damage, and early inflammation/fibrosis factors. FACS analysis was used for immune cell profiling of the kidneys. Results: Histological analysis revealed enhanced infiltration of immune cells (positive for B220) in aged (>70 weeks old) KO mice in both sexes. FACS analysis revealed a similar pattern of enhanced B-1a cells in aged KO mice. There was an age-based increase in pro-fibrotic and pro-inflammatory markers (IL-6, TNF, TGF-β1, and SNAIL1) in KO male mice that presumably contributed to renal fibrosis and renal damage (glomerular and tubular). In fact, in KO mice, there was an age-dependent increase in renal damage (assessed by NGAL and KIM1) that was accompanied by increased fibrosis (assessed by CTGF). This effect was more pronounced in male KO mice than in the female KO mice. In contrast to the KO animals, no significant increase in damage markers was detectable in wild-type animals at the age examined (>70 weeks old). Moreover, there is an age-based increase in AGEs and scavenger receptor MSR-A2 in the kidneys. Discussion: Our data suggest that the loss of the clearance receptor RAGE in male animals further accelerates age-dependent renal damage; this could be in part due to an increase in AGEs load during aging and the absence of protective female hormones. By contrast, in females, RAGE expression seems to play only a minor role when compared to tissue pathology.
Collapse
Affiliation(s)
- Seerat Bajwa
- Department of Internal Medicine III, Jena University Hospital, Jena, Germany
| | - Alexander Luebbe
- Department of Internal Medicine III, Jena University Hospital, Jena, Germany
| | - Ngoc Dong Nhi Vo
- Department of Internal Medicine III, Jena University Hospital, Jena, Germany
| | - Eva-Maria Piskor
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University, Jena, Germany
| | - Christian Kosan
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University, Jena, Germany
| | - Gunter Wolf
- Department of Internal Medicine III, Jena University Hospital, Jena, Germany
| | - Ivonne Loeffler
- Department of Internal Medicine III, Jena University Hospital, Jena, Germany
| |
Collapse
|
10
|
Yan S, Li M, Liu B, Ma Z, Yang Q. Neutrophil extracellular traps and pulmonary fibrosis: an update. J Inflamm (Lond) 2023; 20:2. [PMID: 36658568 PMCID: PMC9851107 DOI: 10.1186/s12950-023-00329-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Pulmonary fibrosis (PF) is a serious and often fatal illness that occurs in various clinical settings and represents a significant unmet medical need. Increasing evidence indicates that neutrophil extracellular traps (NETs) contribute significantly to the progression of PF. Therefore, understanding the pathways by which NETs contribute to the disease is crucial for developing effective treatments. This review focuses on the formation of NETs and the common mechanisms of NETs in PF.
Collapse
Affiliation(s)
- Suyan Yan
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| | - Meiqi Li
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| | - Baocheng Liu
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| | - Zhenzhen Ma
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| | - Qingrui Yang
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| |
Collapse
|
11
|
Receptor for advanced glycation end-products (RAGE) mediates phagocytosis in nonprofessional phagocytes. Commun Biol 2022; 5:824. [PMID: 35974093 PMCID: PMC9381800 DOI: 10.1038/s42003-022-03791-1] [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: 12/30/2021] [Accepted: 08/03/2022] [Indexed: 11/28/2022] Open
Abstract
In mammals, both professional phagocytes and nonprofessional phagocytes (NPPs) can perform phagocytosis. However, limited targets are phagocytosed by NPPs, and thus, the mechanism remains unclear. We find that spores of the yeast Saccharomyces cerevisiae are internalized efficiently by NPPs. Analyses of this phenomenon reveals that RNA fragments derived from cytosolic RNA species are attached to the spore wall, and these fragments serve as ligands to induce spore internalization. Furthermore, we show that a multiligand receptor, RAGE (receptor for advanced glycation end-products), mediates phagocytosis in NPPs. RAGE-mediated phagocytosis is not uniquely induced by spores but is an intrinsic mechanism by which NPPs internalize macromolecules containing RAGE ligands. In fact, artificial particles labeled with polynucleotides, HMGB1, or histone (but not bovine serum albumin) are internalized in NPPs. Our findings provide insight into the molecular basis of phagocytosis by NPPs, a process by which a variety of macromolecules are targeted for internalization. The multiligand receptor RAGE (receptor for advanced glycation end-products) mediates phagocytosis in non-professional phagocytes (NPPs), for example through the use of RNA fragments as ligands for internalization.
Collapse
|
12
|
The HMGB1-RAGE axis induces apoptosis in acute respiratory distress syndrome through PERK/eIF2α/ATF4-mediated endoplasmic reticulum stress. Inflamm Res 2022; 71:1245-1260. [DOI: 10.1007/s00011-022-01613-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/06/2022] [Accepted: 07/13/2022] [Indexed: 11/05/2022] Open
|
13
|
Li L, Beeraka NM, Xie L, Dong L, Liu J, Wang L. Co-expression of High-mobility group box 1 protein (HMGB1) and receptor for advanced glycation end products (RAGE) in the prognosis of esophageal squamous cell carcinoma. Discov Oncol 2022; 13:64. [PMID: 35829833 PMCID: PMC9279518 DOI: 10.1007/s12672-022-00527-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/15/2022] [Indexed: 12/24/2022] Open
Abstract
Esophageal cancer is a malignant type of cancer with a high mortality rate. The aim of this study is to determine co-expression patterns of High-mobility group box 1 protein (HMGB1) and receptor for advanced glycation end products (RAGE) in ESCC (esophageal squamous cell carcinoma) conditions and their prognostic role in cancer progression. The expression of HMGB1 and RAGE in ESCC tissues has been analyzed using qRT-PCR and Western blotting. Co-localized expression patterns of HMGB1 and RAGE in ESCC tissues were determined using immunohistochemistry and analyzed for clinical-pathological parameters. Overall survival was performed based on co-expression of HMGB1 and RAGE proteins. A higher expression pattern of HMGB1, and RAGE was observed at mRNA and protein level in the ESCC group compared to the adjacent tissue group. Expression of HMGB1 was significantly correlated with lymph node, metastasis, lymphatic invasion, and venous invasion (p < 0.05). RAGE expression exhibited a significant correlation with venous invasion. Overall survival was significantly shorter (P < 0.05) in the patients with co-expression of HMGB1 and RAGE compared to the patients without co-expression. A significant difference in the overall survival was evident between the patients with co-expression of HMGB1 and RAGE and the patients without coexpression. HMGB1 and RAGE expression patterns were associated with aggressive metastatic characteristics of ESCC. The co-expression of HMGB1 and RAGE was correlated with shorter survival times. Results concluded the co-expression patterns of HMGB1 and RAGE exhibited a prognostic relevance in ESCC conditions.
Collapse
Affiliation(s)
- Lingzhao Li
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007 Henan People’s Republic of China
| | - Narasimha M. Beeraka
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People’s Republic of China
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Street, Moscow, 119991 Russian Federation
| | - Linsen Xie
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007 Henan People’s Republic of China
| | - Li Dong
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007 Henan People’s Republic of China
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People’s Republic of China
| | - Lei Wang
- Department of Radiation Oncology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, 195# Tongbai Road, Zhengzhou, 450052 Henan People’s Republic of China
| |
Collapse
|
14
|
Singh H, Agrawal DK. Therapeutic potential of targeting the receptor for advanced glycation end products (RAGE) by small molecule inhibitors. Drug Dev Res 2022; 83:1257-1269. [PMID: 35781678 PMCID: PMC9474610 DOI: 10.1002/ddr.21971] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/06/2022] [Accepted: 06/12/2022] [Indexed: 01/05/2023]
Abstract
Receptor for advanced glycation end products (RAGE) is a 45 kDa transmembrane receptor of immunoglobulin family that can bind to various endogenous and exogenous ligands and initiate the inflammatory downstream signaling pathways. RAGE is involved in various disorders including cardiovascular and neurodegenerative diseases, cancer, and diabetes. This review summarizes the structural features of RAGE and its various isoforms along with their pathological effects. Mainly, the article emphasized on the translational significance of antagonizing the interactions of RAGE with its ligands using small molecules reported in the last 5 years and discusses future approaches that could be employed to block the interactions in the treatment of chronic inflammatory ailments. The RAGE inhibitors described in this article could prove as a powerful approach in the management of immune‐inflammatory diseases. A critical review of the literature suggests that there is a dire need to dive deeper into the molecular mechanism of action to resolve critical issues that must be addressed to understand RAGE‐targeting therapy and long‐term blockade of RAGE in human diseases.
Collapse
Affiliation(s)
- Harbinder Singh
- Department of Translational Research, College of Osteopathic Medicine of the Pacific Western University of Health Sciences, Pomona, California, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
15
|
Age, obesity and hyperglycaemia: Activation of innate immunity initiates a series of molecular interactions involving anionic surfaces leading to COVID-19 morbidity and mortality. Med Hypotheses 2021; 155:110646. [PMID: 34392108 PMCID: PMC8330138 DOI: 10.1016/j.mehy.2021.110646] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/28/2021] [Accepted: 07/25/2021] [Indexed: 12/20/2022]
Abstract
Obesity and type 2 diabetes are major factors in COVID-19 causing a progression to excessive morbidity and mortality. An important characteristic of these conditions is poor glycaemic control leading to inappropriate chemical reactions and the production of glycated proteins in which positively charged lysine and arginine residues are neutralised. We propose that this protein glycation primes the inflammatory system as the presence of aspartate and glutamate residues in any glycated zwitterionic protein will thus increase its anionic characteristics. As a result, these macromolecules will be recognised by the innate immune system and identified as originating from infection or cell damage (sterile inflammation). Many proteins in the body exist to non-specifically target these anionic macromolecules and rely heavily on positively charged (cationic) binding-sites to produce a relatively non-specific interaction as the first step in the body’s response. Proteins involved in this innate immunity are collectively referred to as damage associated molecular pattern molecules or pathogen associated molecular pattern molecules. A crucial player in this process is RAGE (Receptor for Advanced Glycation End products). RAGE plays a central role in the inflammatory response and on ligand binding stimulates many aspects of inflammation including the production of the key inflammatory mediator NF-κB, and the subsequent production of inflammatory cytokines. This process has the potential to show a positive feedback loop resulting in a dramatic response within the tissue. We propose that protein glycation primes the inflammatory system by generating negatively charged surfaces so that when a SARS-Cov-2 infection occurs within the lung the further release of negatively-charged macromolecules due to cell damage results in a potentially catastrophic inflammatory response resulting in the cytokine storm associated with COVID-19 morbidity and mortality. That part of the population who do not suffer from inflammatory priming (Phase 1), such as the young and the non-obese, should not be subjected to the catastrophic inflammatory response seen in others (Phase 2). This hypothesis further highlights the need for improved dietary intake to minimise the inflammatory priming resulting from poor glycaemic control.
Collapse
|
16
|
Viurcos-Sanabria R, Escobedo G. Immunometabolic bases of type 2 diabetes in the severity of COVID-19. World J Diabetes 2021; 12:1026-1041. [PMID: 34326952 PMCID: PMC8311488 DOI: 10.4239/wjd.v12.i7.1026] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/16/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) is caused by the novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). COVID-19 and type 2 diabetes (T2D) have now merged into an ongoing global syndemic that is threatening the lives of millions of people around the globe. For this reason, there is a deep need to understand the immunometabolic bases of the main etiological factors of T2D that affect the severity of COVID-19. Here, we discuss how hyperglycemia contributes to the cytokine storm commonly associated with COVID-19 by stimulating monocytes and macrophages to produce interleukin IL-1β, IL-6, and TNF-α in the airway epithelium. The main mechanisms through which hyperglycemia promotes reactive oxygen species release, inhibition of T cell activation, and neutrophil extracellular traps in the lungs of patients with severe SARS-CoV-2 infection are also studied. We further examine the molecular mechanisms by which proinflammatory cytokines induce insulin resistance, and their deleterious effects on pancreatic β-cell exhaustion in T2D patients critically ill with COVID-19. We address the effect of excess glucose on advanced glycation end product (AGE) formation and the role of AGEs in perpetuating pneumonia and acute respiratory distress syndrome. Finally, we discuss the contribution of preexisting endothelial dysfunction secondary to diabetes in the development of neutrophil trafficking, vascular leaking, and thrombotic events in patients with severe SARS-CoV-2 infection. As we outline here, T2D acts in synergy with SARS-CoV-2 infection to increase the progression, severity, and mortality of COVID-19. We think a better understanding of the T2D-related immunometabolic factors that contribute to exacerbate the severity of COVID-19 will improve our ability to identify patients with high mortality risk and prevent adverse outcomes.
Collapse
Affiliation(s)
| | - Galileo Escobedo
- Laboratorio de Proteómica, Dirección de Investigación, Hospital General de Mexico “Dr. Eduardo Liceaga”, Mexico City 06720, Mexico
| |
Collapse
|
17
|
Madhavan BK, Han Z, Singh B, Bordt N, Kaymak S, Bandapalli OR, Kihm L, Shahzad K, Isermann B, Herzig S, Nawroth P, Kumar V. Elevated Expression of the RAGE Variant- V in SCLC Mitigates the Effect of Chemotherapeutic Drugs. Cancers (Basel) 2021; 13:cancers13112843. [PMID: 34200336 PMCID: PMC8201239 DOI: 10.3390/cancers13112843] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Radiomimetic drugs induce extensive genotoxic insults to their target cells. Irreparable DNA damage leaves cells with the choice between a program leading to cell death or senescence, but not DNA repair. Among the challenges of an advanced stage of small cell lung carcinoma (SCLC), the resistance to radiomimetic drugs is the most prominent one. In SCLC, the initial chemotherapeutic treatment primes cell to modify their DNA repair and cell cycle regulatory systems, using alternative but highly efficient forms of DNA repair and auxiliary factors. This modulated system now bypasses several regulatory controls. Thus, at this stage, cells become resistant to any beneficial effects of chemotherapeutic drugs. In the present study, we observed that variant-V of the receptor for advanced glycation end-products (RAGE) is abundantly expressed in advancing and metastasizing SCLC. Therefore, it may serve as a potential target for specific therapeutic interventions directed to SCLC. Abstract Small cell lung carcinoma (SCLC) is a highly aggressive malignancy with a very high mortality rate. A prominent part of this is because these carcinomas are refractory to chemotherapies, such as etoposide or cisplatin, making effective treatment almost impossible. Here, we report that elevated expression of the RAGE variant-V in SCLC promotes homology-directed DNA DSBs repair when challenged with anti-cancer drugs. This variant exclusively localizes to the nucleus, interacts with members of the double-strand break (DSB) repair machinery and thus promotes the recruitment of DSBs repair factors at the site of damage. Increased expression of this variant thus, promotes timely DNA repair. Congruently, the tumor cells expressing high levels of variant-V can tolerate chemotherapeutic drug treatment better than the RAGE depleted cells. Our findings reveal a yet undisclosed role of the RAGE variant-V in the homology-directed DNA repair. This variant thus can be a potential target to be considered for future therapeutic approaches in advanced SSLC.
Collapse
Affiliation(s)
- Bindhu K. Madhavan
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
| | - Zhe Han
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
| | - Bishal Singh
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
| | - Nico Bordt
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
| | - Serap Kaymak
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
| | - Obul Reddy Bandapalli
- Hopp Children’s Cancer Center (KiTZ), 69120 Heidelberg, Germany;
- Medical Faculty, Heidelberg University, 69117 Heidelberg, Germany
| | - Lars Kihm
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
| | - Khurrum Shahzad
- Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany; (K.S.); (B.I.)
| | - Berend Isermann
- Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany; (K.S.); (B.I.)
| | - Stephan Herzig
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- Molecular Metabolic Control, Technical University Munich, 80333 Munich, Germany
- Helmholtz Center Munich, Institute for Diabetes and Cancer, D-85764 Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Programm, Helmholtz-Zentrum, 69120 Heidelberg, Germany
| | - Peter Nawroth
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- Joint Heidelberg-IDC Translational Diabetes Programm, Helmholtz-Zentrum, 69120 Heidelberg, Germany
| | - Varun Kumar
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- European Molecular Biology Laboratory, Advanced Light Microscopy Facility, 69117 Heidelberg, Germany
- Correspondence: ; Tel.: +49-6221-56-6960
| |
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: 14] [Impact Index Per Article: 3.5] [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
|
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: 6.8] [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
|
20
|
Lim A, Radujkovic A, Weigand MA, Merle U. Soluble receptor for advanced glycation end products (sRAGE) as a biomarker of COVID-19 disease severity and indicator of the need for mechanical ventilation, ARDS and mortality. Ann Intensive Care 2021; 11:50. [PMID: 33751264 PMCID: PMC7983090 DOI: 10.1186/s13613-021-00836-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/08/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND COVID-19 pneumonia and subsequent respiratory failure is causing an immense strain on intensive care units globally. Early prediction of severe disease enables clinicians to avoid acute respiratory distress syndrome (ARDS) development and improve management of critically ill patients. The soluble receptor of advanced glycation endproducts (sRAGE) is a biomarker shown to predict ARDS. Although sRAGE level varies depending on the type of disease, there is limited information available on changes in sRAGE levels in COVID-19. Therefore, sRAGE was measured in COVID-19 patients to determine sRAGE level variation in COVID-19 severity and to examine its ability to predict the need for mechanical ventilation (MV) and mortality in COVID-19. METHODS In this single-centre observational cohort study in Germany, serum sRAGE during acute COVID-19, 20 weeks after the start of COVID-19 symptoms, as well as in control groups of non-COVID-19 pneumonia patients and healthy controls were measured using ELISA. The primary endpoint was severe disease (high-flow nasal oxygen therapy (HFNO)/MV and need of organ support). The secondary endpoints were respiratory failure with need of MV and 30-day mortality. The area under the curve (AUC), cut-off based on Youden's index and odds ratio with 95% CI for sRAGE were calculated with regard to prediction of MV need and mortality. RESULTS Serum sRAGE in 164 COVID-19 patients, 101 matched COVID-19 convalescent patients, 23 non-COVID-19 pneumonia patients and 15 healthy volunteers were measured. sRAGE level increased with COVID-19 severity, need for oxygen therapy, HFNO/MV, ARDS severity, need of dialysis and catecholamine support, 30-day mortality, sequential organ failure assessment (SOFA) and quick SOFA (qSOFA) score. sRAGE was found to be a good predictor of MV need in COVID-19 inpatients and mortality with an AUC of 0.871 (0.770-0.973) and 0.903 (0.817-0.990), respectively. When adjusted for male gender, age, comorbidity and SOFA score ≥ 3, sRAGE was independently associated with risk of need for HFNO/MV. When adjusted for SOFA score ≥ 3, sRAGE was independently associated with risk of need for MV. CONCLUSIONS Serum sRAGE concentrations are elevated in COVID-19 patients as disease severity increases. sRAGE should be considered as a biomarker for predicting the need for MV and mortality in COVID-19.
Collapse
Affiliation(s)
- Adeline Lim
- Department of Internal Medicine IV, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
| | - Aleksandar Radujkovic
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Markus A Weigand
- Department of Anesthesiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Uta Merle
- Department of Internal Medicine IV, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| |
Collapse
|
21
|
Bobkova NV. The Balance between Two Branches of RAS Can Protect from Severe COVID-19 Course. BIOCHEMISTRY (MOSCOW) SUPPLEMENT. SERIES A, MEMBRANE AND CELL BIOLOGY 2021; 15:36-51. [PMID: 33643542 PMCID: PMC7897458 DOI: 10.1134/s1990747821010037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/09/2020] [Accepted: 09/22/2020] [Indexed: 12/23/2022]
Abstract
The COVID-19 pandemic has swept the world and required the mobilization of scientists and clinicians around the world to combat this serious disease. Along with SARS-CoV-2 virology research, understanding of the fundamental physiological processes, molecular and cellular mechanisms and intracellular signaling pathways underlying the clinical manifestations of COVID-19 is important for effective therapy of this disease. The review describes in detail the interaction of the components of the renin-angiotensin system (RAS) and receptors of end-glycosylated products (RAGE), which plays a special role in normal lung physiology and in pathological conditions in COVID-19, including the development of acute respiratory distress syndrome and "cytokine storm". A separate section is devoted to the latest developments aimed at correcting the dysfunction of the RAS caused by the binding of the virus to angiotensin converting enzyme 2 (ACE2)- the central element of this system. Analysis of published theoretical, clinical, and experimental data indicates the need for a complex treatment to prevent a severe course of COVID-19 using MasR agonists, blockers of the AT1R and NF-κB signaling pathway, as well as compounds with neuroprotective and neuroregenerative effects.
Collapse
Affiliation(s)
- N. V. Bobkova
- Institute of Cell Biophysics, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Moscow oblast Russia
| |
Collapse
|
22
|
Roy D, Ramasamy R, Schmidt AM. Journey to a Receptor for Advanced Glycation End Products Connection in Severe Acute Respiratory Syndrome Coronavirus 2 Infection: With Stops Along the Way in the Lung, Heart, Blood Vessels, and Adipose Tissue. Arterioscler Thromb Vasc Biol 2021; 41:614-627. [PMID: 33327744 PMCID: PMC7837689 DOI: 10.1161/atvbaha.120.315527] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 11/30/2020] [Indexed: 01/08/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has affected millions of people worldwide and the pandemic has yet to wane. Despite its associated significant morbidity and mortality, there are no definitive cures and no fully preventative measures to combat SARS-CoV-2. Hence, the urgency to identify the pathobiological mechanisms underlying increased risk for and the severity of SARS-CoV-2 infection is mounting. One contributing factor, the accumulation of damage-associated molecular pattern molecules, is a leading trigger for the activation of nuclear factor-kB and the IRF (interferon regulatory factors), such as IRF7. Activation of these pathways, particularly in the lung and other organs, such as the heart, contributes to a burst of cytokine release, which predisposes to significant tissue damage, loss of function, and mortality. The receptor for advanced glycation end products (RAGE) binds damage-associated molecular patterns is expressed in the lung and heart, and in priming organs, such as the blood vessels (in diabetes) and adipose tissue (in obesity), and transduces the pathological signals emitted by damage-associated molecular patterns. It is proposed that damage-associated molecular pattern-RAGE enrichment in these priming tissues, and in the lungs and heart during active infection, contributes to the widespread tissue damage induced by SARS-CoV-2. Accordingly, the RAGE axis might play seminal roles in and be a target for therapeutic intervention in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Divya Roy
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine (D.R., R.R., A.M.S.)
- New York Institute of Technology College of Osteopathic Medicine, Glen Head (D.R.)
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine (D.R., R.R., A.M.S.)
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine (D.R., R.R., A.M.S.)
| |
Collapse
|
23
|
Mukherjee TK, Malik P, Hoidal JR. Receptor for Advanced Glycation End Products (RAGE) and Its Polymorphic Variants as Predictive Diagnostic and Prognostic Markers of NSCLCs: a Perspective. Curr Oncol Rep 2021; 23:12. [PMID: 33399986 DOI: 10.1007/s11912-020-00992-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Non-small cell lung cancers (NSCLCs) account for ~ 85% of all lung cancers, and 5-year survival in Europe and the USA is ~ 13-17%. In this review, we focus on the significance of Receptor for Advanced Glycation End products (RAGE) as a diagnostic or post-therapeutic prognostic marker for various forms of NSCLCs. RECENT FINDINGS The lungs have the highest levels of basal RAGE expression in mammals. The physiologic RAGE in lungs may be involved in adhesion and spreading of AT-1 cells and maintenance of pulmonary homeostasis. However, high level expression of RAGE complicates various diseases including acute lung injury. In NSCLCs, while a number of studies report decreased RAGE expression, inferring a protective role, others suggest that RAGE expression may contribute to NSCLC pathogenesis. Genetic polymorphisms of RAGE are reportedly associated with NSCLC development and complications. RAGE and its polymorphic variants may be useful diagnostic or post-therapeutic prognostic markers of NSCLCs.
Collapse
Affiliation(s)
- Tapan K Mukherjee
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT, USA. .,Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA. .,George E. Wahlen Department of Veterans Affairs Medical Center, 500, Foothil Drive, Building#45, Salt Lake City, UT, 84148, USA.
| | - Parth Malik
- School of Chemical Sciences, Central University of Gujarat (Gandhinagar), Gandhinagar, India.,School of Nano Sciences, Central University of Gujarat (Gandhinagar), Gandhinagar, India
| | - John R Hoidal
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT, USA.,Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA.,George E. Wahlen Department of Veterans Affairs Medical Center, 500, Foothil Drive, Building#45, Salt Lake City, UT, 84148, USA
| |
Collapse
|
24
|
Haider SH, Veerappan A, Crowley G, Caraher EJ, Ostrofsky D, Mikhail M, Lam R, Wang Y, Sunseri M, Kwon S, Prezant DJ, Liu M, Schmidt AM, Nolan A. Multiomics of World Trade Center Particulate Matter-induced Persistent Airway Hyperreactivity. Role of Receptor for Advanced Glycation End Products. Am J Respir Cell Mol Biol 2020; 63:219-233. [PMID: 32315541 DOI: 10.1165/rcmb.2019-0064oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pulmonary disease after World Trade Center particulate matter (WTC-PM) exposure is associated with dyslipidemia and the receptor for advanced glycation end products (RAGE); however, the mechanisms are not well understood. We used a murine model and a multiomics assessment to understand the role of RAGE in the pulmonary long-term effects of a single high-intensity exposure to WTC-PM. After 1 month, WTC-PM-exposed wild-type (WT) mice had airway hyperreactivity, whereas RAGE-deficient (Ager-/-) mice were protected. PM-exposed WT mice also had histologic evidence of airspace disease, whereas Ager-/- mice remained unchanged. Inflammatory mediators such as G-CSF (granulocyte colony-stimulating factor), IP-10 (IFN-γ-induced protein 10), and KC (keratinocyte chemoattractant) were differentially expressed after WTC-PM exposure. WTC-PM induced α-SMA, DIAPH1 (protein diaphanous homolog 1), RAGE, and significant lung collagen deposition in WT compared with Ager-/- mice. Compared with WT mice with PM exposure, relative expression of phosphorylated to total CREB (cAMP response element-binding protein) and JNK (c-Jun N-terminal kinase) was significantly increased in the lung of PM-exposed Ager-/- mice, whereas Akt (protein kinase B) was decreased. Random forests of the refined lung metabolomic profile classified subjects with 92% accuracy; principal component analysis captured 86.7% of the variance in three components and demonstrated prominent subpathway involvement, including known mediators of lung disease such as vitamin B6 metabolites, sphingolipids, fatty acids, and phosphatidylcholines. Treatment with a partial RAGE antagonist, pioglitazone, yielded similar fold-change expression of metabolites (N6-carboxymethyllysine, 1-methylnicotinamide, N1+N8-acetylspermidine, and succinylcarnitine [C4-DC]) between WT and Ager-/- mice exposed to WTC-PM. RAGE can mediate WTC-PM-induced airway hyperreactivity and warrants further investigation.
Collapse
Affiliation(s)
- Syed H Haider
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Arul Veerappan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - George Crowley
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Erin J Caraher
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Dean Ostrofsky
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Mena Mikhail
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Rachel Lam
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Yuyan Wang
- Division of Biostatistics, Department of Population Health
| | - Maria Sunseri
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Sophia Kwon
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - David J Prezant
- Bureau of Health Services and Office of Medical Affairs, Fire Department of New York, Brooklyn, New York; and.,Division of Pulmonary Medicine, Department of Medicine, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York
| | - Mengling Liu
- Division of Biostatistics, Department of Population Health.,Department of Environmental Medicine, and
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Anna Nolan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine.,Department of Environmental Medicine, and.,Bureau of Health Services and Office of Medical Affairs, Fire Department of New York, Brooklyn, New York; and
| |
Collapse
|
25
|
Cicco S, Cicco G, Racanelli V, Vacca A. Neutrophil Extracellular Traps (NETs) and Damage-Associated Molecular Patterns (DAMPs): Two Potential Targets for COVID-19 Treatment. Mediators Inflamm 2020; 2020:7527953. [PMID: 32724296 PMCID: PMC7366221 DOI: 10.1155/2020/7527953] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/11/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022] Open
Abstract
COVID-19 is a pandemic disease caused by the new coronavirus SARS-CoV-2 that mostly affects the respiratory system. The consequent inflammation is not able to clear viruses. The persistent excessive inflammatory response can build up a clinical picture that is very difficult to manage and potentially fatal. Modulating the immune response plays a key role in fighting the disease. One of the main defence systems is the activation of neutrophils that release neutrophil extracellular traps (NETs) under the stimulus of autophagy. Various molecules can induce NETosis and autophagy; some potent activators are damage-associated molecular patterns (DAMPs) and, in particular, the high-mobility group box 1 (HMGB1). This molecule is released by damaged lung cells and can induce a robust innate immunity response. The increase in HMGB1 and NETosis could lead to sustained inflammation due to SARS-CoV-2 infection. Therefore, blocking these molecules might be useful in COVID-19 treatment and should be further studied in the context of targeted therapy.
Collapse
Affiliation(s)
- Sebastiano Cicco
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro Medical School, Piazza G. Cesare 11, I-70124 Bari, Italy
| | - Gerolamo Cicco
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro Medical School, Piazza G. Cesare 11, I-70124 Bari, Italy
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro Medical School, Piazza G. Cesare 11, I-70124 Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro Medical School, Piazza G. Cesare 11, I-70124 Bari, Italy
| |
Collapse
|
26
|
Degani G, Altomare A, Digiovanni S, Arosio B, Fritz G, Raucci A, Aldini G, Popolo L. Prothrombin is a binding partner of the human receptor of advanced glycation end products. J Biol Chem 2020; 295:12498-12511. [PMID: 32665403 DOI: 10.1074/jbc.ra120.013692] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/11/2020] [Indexed: 01/02/2023] Open
Abstract
The receptor for advanced glycation end products (RAGE) plays a key role in mammal physiology and in the etiology and progression of inflammatory and oxidative stress-based diseases. In adults, RAGE expression is normally high only in the lung where the protein concentrates in the basal membrane of alveolar Type I epithelial cells. In diseases, RAGE levels increase in the affected tissues and sustain chronic inflammation. RAGE exists as a membrane glycoprotein with an ectodomain, a transmembrane helix, and a short carboxyl-terminal tail, or as a soluble ectodomain that acts as a decoy receptor (sRAGE). VC1 domain is responsible for binding to the majority of RAGE ligands including advanced glycation end products (AGEs), S100 proteins, and HMGB1. To ascertain whether other ligands exist, we analyzed by MS the material pulled down by VC1 from human plasma. Twenty of 295 identified proteins were selected and associated to coagulation and complement processes and to extracellular matrix. Four of them contained a γ-carboxyl glutamic acid (Gla) domain, a calcium-binding module, and prothrombin (PT) was the most abundant. Using MicroScale thermophoresis, we quantified the interaction of PT with VC1 and sRAGE in the absence or presence of calcium that acted as a competitor. PT devoid of the Gla domain (PT des-Gla) did not bind to sRAGE, providing further evidence that the Gla domain is critical for the interaction. Finally, the presence of VC1 delayed plasma clotting in a dose-dependent manner. We propose that RAGE is involved in modulating blood coagulation presumably in conditions of lung injury.
Collapse
Affiliation(s)
- Genny Degani
- Department of Biosciences, University of Milan, Milan, Italy
| | | | | | - Beatrice Arosio
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico and University of Milan, Via Pace 9, Milan, Italy
| | - Guenter Fritz
- Institute of Microbiology, University of Hohenheim, Stuttgart, Germany
| | - Angela Raucci
- Experimental Cardio-oncology and Cardiovascular Aging Unit, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea, 4, Milan, Italy
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Laura Popolo
- Department of Biosciences, University of Milan, Milan, Italy
| |
Collapse
|
27
|
Rojas A, Gonzalez I, Morales MA. SARS-CoV-2-mediated inflammatory response in lungs: should we look at RAGE? Inflamm Res 2020; 69:641-643. [PMID: 32372149 PMCID: PMC7200049 DOI: 10.1007/s00011-020-01353-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 04/21/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Affiliation(s)
- Armando Rojas
- Biomedical Research Laboratories, Medicine Faculty, Catholic University of Maule, Talca, Chile.
| | - Ileana Gonzalez
- Biomedical Research Laboratories, Medicine Faculty, Catholic University of Maule, Talca, Chile
| | - Miguel A Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| |
Collapse
|
28
|
Andersson U, Ottestad W, Tracey KJ. Extracellular HMGB1: a therapeutic target in severe pulmonary inflammation including COVID-19? Mol Med 2020; 26:42. [PMID: 32380958 PMCID: PMC7203545 DOI: 10.1186/s10020-020-00172-4] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 04/23/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The 2019 novel coronavirus disease (COVID-19) causes for unresolved reasons acute respiratory distress syndrome in vulnerable individuals. There is a need to identify key pathogenic molecules in COVID-19-associated inflammation attainable to target with existing therapeutic compounds. The endogenous damage-associated molecular pattern (DAMP) molecule HMGB1 initiates inflammation via two separate pathways. Disulfide-HMGB1 triggers TLR4 receptors generating pro-inflammatory cytokine release. Extracellular HMGB1, released from dying cells or secreted by activated innate immunity cells, forms complexes with extracellular DNA, RNA and other DAMP or pathogen-associated molecular (DAMP) molecules released after lytic cell death. These complexes are endocytosed via RAGE, constitutively expressed at high levels in the lungs only, and transported to the endolysosomal system, which is disrupted by HMGB1 at high concentrations. Danger molecules thus get access to cytosolic proinflammatory receptors instigating inflammasome activation. It is conceivable that extracellular SARS-CoV-2 RNA may reach the cellular cytosol via HMGB1-assisted transfer combined with lysosome leakage. Extracellular HMGB1 generally exists in vivo bound to other molecules, including PAMPs and DAMPs. It is plausible that these complexes are specifically removed in the lungs revealed by a 40% reduction of HMGB1 plasma levels in arterial versus venous blood. Abundant pulmonary RAGE expression enables endocytosis of danger molecules to be destroyed in the lysosomes at physiological HMGB1 levels, but causing detrimental inflammasome activation at high levels. Stress induces apoptosis in pulmonary endothelial cells from females but necrosis in cells from males. CONCLUSION Based on these observations we propose extracellular HMGB1 to be considered as a therapeutic target for COVID-19.
Collapse
Affiliation(s)
- Ulf Andersson
- Department of Women’s and Children’s Health, Karolinska Institutet at Karolinska University Hospital, Tomtebodavägen 18A, 171 77 Stockholm, Sweden
| | - William Ottestad
- Air Ambulance department, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kevin J. Tracey
- Center for Biomedical Science and Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030 USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra University, Hempstead, New York, 11030 USA
- Department of Surgery, North Shore University Hospital, Northwell Health, 300 Community Drive, Manhasset, NY 11030 USA
| |
Collapse
|
29
|
Kida Y, Ohshimo S, Kyo M, Hosokawa K, Amatya VJ, Takeshima Y, Shime N. Retrospective immunohistological study of autopsied lungs in patients with acute exacerbation of interstitial pneumonia managed with extracorporeal membrane oxygenation. J Thorac Dis 2019; 11:4436-4443. [PMID: 31903231 DOI: 10.21037/jtd.2019.11.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Acute exacerbation of interstitial pneumonia (AE-IP) is a life-threatening pulmonary condition that involves various pathogeneses. In patients with AE-IP who need mechanical ventilation with high driving pressure and oxygen concentration, veno-venous extracorporeal membrane oxygenation (V-V ECMO) may diminish alveolar epithelial damage by decreasing ventilator settings. The pathophysiological benefit of this therapeutic option is not well investigated. Methods We retrospectively collected 15 autopsied patients with AE-IP who were treated with mechanical ventilation in the intensive care unit (ICU) at Hiroshima University Hospital (Hiroshima, Japan) between 2010 and 2016. The patients were grouped by whether they were managed with mechanical ventilation only (the ventilator group, n=6) or with mechanical ventilation and V-V ECMO (the ECMO group, n=9). Results The median age of the ventilator and ECMO group patients were similar (65 and 64 years, respectively). The severity score APACHE II in the ECMO group (35.0) is significantly higher than that of ventilator group (14.5) (P=0.006). Ventilator days were significantly shorter in the ventilator group (17.5 days) than in the ECMO group (30.0 days) (P=0.04). Compared with the ECMO group, the ventilator group had a stronger Masson-trichrome stain grade (4 vs. 6, P=0.04) and higher immunoreactivity grades for Krebs von den Lungen-6 (4 vs. 6, P=0.04) and IL-8 (3 vs. 6, P=0.02). Between the ventilator and ECMO groups, the immunoreactivity grades of angiopoietin 2 (4 vs. 1, P=0.08) and receptor for advanced glycation end products (2 vs. 1, P=0.52) did not differ. Conclusions The lungs of mechanically ventilated AE-IP patients treated with V-V ECMO had decreased fibrosis, endothelial injury, and inflammation. This finding suggests the lung-protective efficacy of adjunctive V-V ECMO therapy.
Collapse
Affiliation(s)
- Yoshiko Kida
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shinichiro Ohshimo
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Michihito Kyo
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Koji Hosokawa
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Vishwa Jeet Amatya
- Department of Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukio Takeshima
- Department of Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nobuaki Shime
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
30
|
Haider SH, Oskuei A, Crowley G, Kwon S, Lam R, Riggs J, Mikhail M, Talusan A, Veerappan A, Kim JS, Caraher EJ, Nolan A. Receptor for advanced glycation end-products and environmental exposure related obstructive airways disease: a systematic review. Eur Respir Rev 2019; 28:28/151/180096. [PMID: 30918021 PMCID: PMC7006869 DOI: 10.1183/16000617.0096-2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 02/09/2019] [Indexed: 12/11/2022] Open
Abstract
Background Our group has identified the receptor for advanced glycation end-products (RAGE) as a predictor of World Trade Center particulate matter associated lung injury. The aim of this systematic review is to assess the relationship between RAGE and obstructive airways disease secondary to environmental exposure. Methods A comprehensive search using PubMed and Embase was performed on January 5, 2018 utilising keywords focusing on environmental exposure, obstructive airways disease and RAGE and was registered with PROSPERO (CRD42018093834). We included original human research studies in English, focusing on pulmonary end-points associated with RAGE and environmental exposure. Results A total of 213 studies were identified by the initial search. After removing the duplicates and applying inclusion and exclusion criteria, we screened the titles and abstracts of 61 studies. Finally, 19 full-text articles were included. The exposures discussed in these articles include particulate matter (n=2) and cigarette smoke (n=17). Conclusion RAGE is a mediator of inflammation associated end-organ dysfunction such as obstructive airways disease. Soluble RAGE, a decoy receptor, may have a protective effect in some pulmonary processes. Overall, RAGE is biologically relevant in environmental exposure associated lung disease. Future investigations should focus on further understanding the role and therapeutic potential of RAGE in particulate matter exposure associated lung disease. RAGE is biologically relevant in environmental exposure associated lung disease. Future investigations should focus on further understanding the role and therapeutic potential of RAGE in particulate matter exposure associated lung diseasehttp://ow.ly/gfZz30o7otU
Collapse
Affiliation(s)
- Syed H Haider
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA.,Bureau of Health Services and Office of Medical Affairs, Fire Department of New York, New York, NY, USA
| | - Assad Oskuei
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - George Crowley
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - Sophia Kwon
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - Rachel Lam
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - Jessica Riggs
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - Mena Mikhail
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - Angela Talusan
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - Arul Veerappan
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - James S Kim
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - Erin J Caraher
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA
| | - Anna Nolan
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, NY, USA .,Bureau of Health Services and Office of Medical Affairs, Fire Department of New York, New York, NY, USA.,Dept of Environmental Medicine, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
31
|
Machahua C, Montes-Worboys A, Planas-Cerezales L, Buendia-Flores R, Molina-Molina M, Vicens-Zygmunt V. Serum AGE/RAGEs as potential biomarker in idiopathic pulmonary fibrosis. Respir Res 2018; 19:215. [PMID: 30409203 PMCID: PMC6225674 DOI: 10.1186/s12931-018-0924-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/28/2018] [Indexed: 12/16/2022] Open
Abstract
Background The soluble receptor for advanced glycation end-products (sRAGE) has been suggested that it acts as a decoy for capturing advanced glycation end-products (AGEs) and inhibits the activation of the oxidative stress and apoptotic pathways. Lung AGEs/sRAGE is increased in idiopathic pulmonary fibrosis (IPF). The objective of the study was to evaluate the AGEs and sRAGE levels in serum as a potential biomarker in IPF. Methods Serum samples were collected from adult patients: 62 IPF, 22 chronic hypersensitivity pneumonitis (cHP), 20 fibrotic non-specific interstitial pneumonia (fNSIP); and 12 healthy controls. In addition, 23 IPF patients were re-evaluated after 3-year follow-up period. Epidemiological and clinical features were recorded: age, sex, smoking habits, and lung function. AGEs and sRAGE were evaluated by ELISA, and the results were correlated with pulmonary functional test values. Results IPF and cHP groups presented a significant increase of AGE/sRAGE serum concentration compared with fNSIP patients. Moreover, an inverse correlation between AGEs and sRAGE levels were found in IPF, and serum sRAGE at diagnosis correlated with FVC and DLCO values. Additionally, changes in serum AGEs and sRAGE correlated with % change of FVC, DLCO and TLC during the follow-up. sRAGE levels below 428.25 pg/ml evolved poor survival rates. Conclusions These findings demonstrate that the increase of AGE/sRAGE ratio is higher in IPF, although the levels were close to cHP. AGE/sRAGE increase correlates with respiratory functional progression. Furthermore, the concentration of sRAGE in blood stream at diagnosis and follow-up could be considered as a potential prognostic biomarker. Electronic supplementary material The online version of this article (10.1186/s12931-018-0924-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Carlos Machahua
- Pneumology Research Group, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.,Biomedical Research Network Centers in Respiratory Diseases (CIBERES), Barcelona, Spain
| | - Ana Montes-Worboys
- Pneumology Research Group, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.,Biomedical Research Network Centers in Respiratory Diseases (CIBERES), Barcelona, Spain.,Unit of Interstitial Lung Diseases, Department of Pneumology, University Hospital of Bellvitge, C. Feixa Llarga sn., 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Lurdes Planas-Cerezales
- Pneumology Research Group, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.,Unit of Interstitial Lung Diseases, Department of Pneumology, University Hospital of Bellvitge, C. Feixa Llarga sn., 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Maria Molina-Molina
- Pneumology Research Group, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain. .,Biomedical Research Network Centers in Respiratory Diseases (CIBERES), Barcelona, Spain. .,Unit of Interstitial Lung Diseases, Department of Pneumology, University Hospital of Bellvitge, C. Feixa Llarga sn., 08907 L'Hospitalet de Llobregat, Barcelona, Spain.
| | - Vanesa Vicens-Zygmunt
- Pneumology Research Group, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.,Unit of Interstitial Lung Diseases, Department of Pneumology, University Hospital of Bellvitge, C. Feixa Llarga sn., 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
32
|
Sutton K, Costa T, Alber A, Bryson K, Borowska D, Balic A, Kaiser P, Stevens M, Vervelde L. Visualisation and characterisation of mononuclear phagocytes in the chicken respiratory tract using CSF1R-transgenic chickens. Vet Res 2018; 49:104. [PMID: 30305141 PMCID: PMC6389226 DOI: 10.1186/s13567-018-0598-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
The respiratory tract is a key organ for many avian pathogens as well as a major route for vaccination in the poultry industry. To improve immune responses after vaccination of chickens through increased uptake of vaccines and targeting to antigen presenting cells, a better understanding of the avian respiratory immune system is required. Transgenic MacReporter birds were used expressing a reporter gene (eGFP or mApple) under the control of the CSF1R promoter and enhancer in cells of the mononuclear phagocyte (MNP) lineage to visualize the ontogeny of the lymphoid tissue, macrophages and dendritic cells, in the trachea, lung and air sac of birds from embryonic day 18-63 weeks of age. Small aggregates of CSF1R-transgene+ cells start to form at the openings of the secondary bronchi at 1 week of age, indicative of the early development of the organised bronchus-associated lymphoid tissue. Immunohistochemical staining revealed subpopulations of MNPs in the lung, based on expression of CSF1R-transgene, CD11, TIM4, LAMP1, and MHC II. Specialised epithelial cells or M cells covering the bronchus-associated lymphoid tissue expressed CSF1R-transgene and type II pneumocytes expressed LAMP1 suggesting that these epithelial cells are phagocytic and transcytose antigen. Highly organised lymphoid tissue was seen in trachea from 4 weeks onwards. Throughout the air sacs at all ages, CSF1R-transgene+ cells were scattered and at later stages, CSF1R-transgene+ cells lined capillaries. These results will serve as a base for further functional characterization of macrophages and dendritic cells and their role in respiratory diseases and vaccine responses.
Collapse
Affiliation(s)
- Kate Sutton
- Division of Infection and Immunity, The Roslin Institute and Royal (Dick), School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG UK
| | - Taiana Costa
- Division of Infection and Immunity, The Roslin Institute and Royal (Dick), School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG UK
| | - Andreas Alber
- Division of Infection and Immunity, The Roslin Institute and Royal (Dick), School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG UK
| | - Karen Bryson
- Division of Infection and Immunity, The Roslin Institute and Royal (Dick), School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG UK
| | - Dominika Borowska
- Division of Infection and Immunity, The Roslin Institute and Royal (Dick), School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG UK
| | - Adam Balic
- Division of Infection and Immunity, The Roslin Institute and Royal (Dick), School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG UK
| | - Pete Kaiser
- Division of Infection and Immunity, The Roslin Institute and Royal (Dick), School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG UK
| | - Mark Stevens
- Division of Infection and Immunity, The Roslin Institute and Royal (Dick), School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG UK
| | - Lonneke Vervelde
- Division of Infection and Immunity, The Roslin Institute and Royal (Dick), School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG UK
| |
Collapse
|
33
|
Wu L, Yang L. The function and mechanism of HMGB1 in lung cancer and its potential therapeutic implications. Oncol Lett 2018; 15:6799-6805. [PMID: 29725415 DOI: 10.3892/ol.2018.8215] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 01/22/2018] [Indexed: 02/06/2023] Open
Abstract
As a non-histone chromatin-associated protein, high-mobility group box-1 (HMGB1) performs a pivotal function in various human diseases, including autoimmune diseases, neurodegenerative diseases and cancer. Overexpression of HMGB1 has been demonstrated in numerous types of cancer, including breast cancer, colorectal cancer, lung cancer and hepatocellular carcinoma. However, the underlying mechanism of HMGB1 function in lung cancer remains to be elucidated. The present study aimed to analyze, and summarize the role and mechanism of HMGB1 in lung cancer by retrieving available literature regarding HMGB1 in association with lung cancer. It provides comprehensive information on the association of HMGB1 with the carcinogenesis and progression of lung cancer, and discusses the molecular mechanism of these processes. HMGB1 may induce tumorigenesis, metastasis and chemotherapy resistance in lung cancer. Overall, it is evident that HMGB1 serves an important role in the development and progression of lung cancer, and this review warrants further investigation into HMGB1 as a novel target for cancer therapy.
Collapse
Affiliation(s)
- Lei Wu
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, P.R. China.,National Clinical Research Center of Cancer, Tianjin Medical University, Tiyuanbei, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tiyuanbei, Tianjin 300060, P.R. China
| | - Lili Yang
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, P.R. China.,National Clinical Research Center of Cancer, Tianjin Medical University, Tiyuanbei, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tiyuanbei, Tianjin 300060, P.R. China
| |
Collapse
|
34
|
O'Reilly MA. Giving New Identities to Alveolar Epithelial Type I Cells. Am J Respir Cell Mol Biol 2018; 56:277-278. [PMID: 28248135 DOI: 10.1165/rcmb.2016-0383ed] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Michael A O'Reilly
- 1 Department of Pediatrics School of Medicine and Dentistry The University of Rochester Rochester, New York
| |
Collapse
|
35
|
Bai W, Zhou J, Zhou N, Liu Q, Cui J, Zou W, Zhang W. Hypoxia-increased RAGE expression regulates chemotaxis and pro-inflammatory cytokines release through nuclear translocation of NF-κ B and HIF1α in THP-1 cells. Biochem Biophys Res Commun 2017; 495:2282-2288. [PMID: 29258824 DOI: 10.1016/j.bbrc.2017.12.084] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 12/15/2017] [Indexed: 12/21/2022]
Abstract
The potential role of hypoxia in mediating the receptor for advanced glycation end products (RAGE) expression deserves to be confirmed. And the role of RAGE in hypoxia-induced chemotaxis and inflammation is still unclear. In present study, THP-1 cells were pretreated with siRNA to block HIF1α, NF-κ B, or RAGE, followed by exposed to hypoxia (combined with H2O2 or SNP), and then RAGE expression, nuclear translocation of HIF1α and NF-κ B, release of TNF-α and IL-1β, as well as expression of MCP-1 and CCR2 were measured. The results revealed that RAGE mRNA and protein in THP-1 cells were significantly increased after exposed into hypoxia atmosphere, especially into the solution containing SNP or H2O2. Moreover, SNP or H2O2 exposure could further amplify hypoxia-induced nuclear translocation of HIF-1α and NF-κ B. Knockdown HIF-1α or NF-κ B by siRNAs could reduce hypoxia- and oxidative stress-induced RAGE hyper-expression. And pretreatment THP-1 cells with RAGE siRNA or NF-κ B siRNA could reduce hypoxia- and oxidative stress-induced expression of MCP-1 and CCR2, and release of TNF-α and IL-1β. Thus, hypoxia not only increases RAGE expression in THP-1 cells by promoting nuclear translocation of NF-κ B and HIF1α, but also regulates chemotaxis and pro-inflammatory cytokines release, which may be partially mediated through upregulation of RAGE expression.
Collapse
Affiliation(s)
- Wei Bai
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Na Zhou
- The Department of Respiratory Medicine, Nanchang Third Hospital, Nanchang, China
| | - Qin Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jian Cui
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wei Zou
- Key Laboratory of Hunan Province for Traditional Chinese Medicine in Obstetrics & Gynecology Research, Hunan Province Maternal and Child Health Hospital, Changsha, China.
| | - Wei Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
36
|
Lecube A, Simó R, Pallayova M, Punjabi NM, López-Cano C, Turino C, Hernández C, Barbé F. Pulmonary Function and Sleep Breathing: Two New Targets for Type 2 Diabetes Care. Endocr Rev 2017; 38:550-573. [PMID: 28938479 DOI: 10.1210/er.2017-00173] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/29/2017] [Indexed: 02/07/2023]
Abstract
Population-based studies showing the negative impact of type 2 diabetes (T2D) on lung function are overviewed. Among the well-recognized pathophysiological mechanisms, the metabolic pathways related to insulin resistance (IR), low-grade chronic inflammation, leptin resistance, microvascular damage, and autonomic neuropathy are emphasized. Histopathological changes are exposed, and findings reported from experimental models are clearly differentiated from those described in humans. The accelerated decline in pulmonary function that appears in patients with cystic fibrosis (CF) with related abnormalities of glucose tolerance and diabetes is considered as an example to further investigate the relationship between T2D and the lung. Furthermore, a possible causal link between antihyperglycemic therapies and pulmonary function is examined. T2D similarly affects breathing during sleep, becoming an independent risk factor for higher rates of sleep apnea, leading to nocturnal hypoxemia and daytime sleepiness. Therefore, the impact of T2D on sleep breathing and its influence on sleep architecture is analyzed. Finally, the effect of improving some pathophysiological mechanisms, primarily IR and inflammation, as well as the optimization of blood glucose control on sleep breathing is evaluated. In summary, the lung should be considered by those providing care for people with diabetes and raise the central issue of whether the normalization of glucose levels can improve pulmonary function and ameliorate sleep-disordered breathing. Therefore, patients with T2D should be considered a vulnerable group for pulmonary dysfunction. However, further research aimed at elucidating how to screen for the lung impairment in the population with diabetes in a cost-effective manner is needed.
Collapse
Affiliation(s)
- Albert Lecube
- Endocrinology and Nutrition Department, Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomédica de Lleida, Universitat de Lleida, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Spain
| | - Rafael Simó
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Spain.,Endocrinology and Nutrition Department, Hospital Universitari Vall d'Hebron, Diabetes and Metabolism Research Unit, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Spain
| | - Maria Pallayova
- Department of Medicine, Weill Cornell Medicine.,Department of Human Physiology and Sleep Laboratory, Faculty of Medicine, Pavol Jozef Šafárik University, Slovak Republic
| | - Naresh M Punjabi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University.,Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University
| | - Carolina López-Cano
- Endocrinology and Nutrition Department, Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomédica de Lleida, Universitat de Lleida, Spain
| | - Cecilia Turino
- Respiratory Department, Hospital Universitari Arnau de Vilanova-Santa María, Institut de Recerca Biomédica de Lleida, Universitat de Lleida, Spain
| | - Cristina Hernández
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Spain.,Endocrinology and Nutrition Department, Hospital Universitari Vall d'Hebron, Diabetes and Metabolism Research Unit, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Spain
| | - Ferran Barbé
- Respiratory Department, Hospital Universitari Arnau de Vilanova-Santa María, Institut de Recerca Biomédica de Lleida, Universitat de Lleida, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Spain
| |
Collapse
|
37
|
Chakraborty D, Zenker S, Rossaint J, Hölscher A, Pohlen M, Zarbock A, Roth J, Vogl T. Alarmin S100A8 Activates Alveolar Epithelial Cells in the Context of Acute Lung Injury in a TLR4-Dependent Manner. Front Immunol 2017; 8:1493. [PMID: 29180999 PMCID: PMC5693860 DOI: 10.3389/fimmu.2017.01493] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/24/2017] [Indexed: 01/11/2023] Open
Abstract
Alveolar epithelial cells (AECs) are an essential part of the respiratory barrier in lungs for gas exchange and protection against pathogens. Damage to AECs occurs during lung injury and PAMPs/DAMPs have been shown to activate AECs. However, their interplay as well as the mechanism of AECs’ activation especially by the alarmin S100A8/A9 is unknown. Thus, our aim was to study the mechanism of activation of AECs (type I and type II) by S100A8 and/or lipopolysaccharide (LPS) and to understand the role of endogenous S100A8/A9 in neutrophil recruitment in the lung. For our studies, we modified a previous protocol for isolation and culturing of murine AECs. Next, we stimulated the cells with S100A8 and/or LPS and analyzed cytokine/chemokine release. We also analyzed the contribution of the known S100-receptors TLR4 and RAGE in AEC activation. In a murine model of lung injury, we investigated the role of S100A8/A9 in neutrophil recruitment to lungs. S100A8 activates type I and type II cells in a dose- and time-dependent manner which could be quantified by the release of IL-6, KC, and MCP-1. We here clearly demonstrate that AEC s are activated by S100A8 via a TLR4-dependent pathway. Surprisingly, RAGE, albeit mainly expressed in lung tissue, plays no role. Additionally, we show that S100A8/A9 is an essential factor for neutrophil recruitment to lungs. We, therefore, conclude that S100A8 promotes acute lung injury via Toll-like receptor 4-dependent activation of AECs.
Collapse
Affiliation(s)
| | - Stefanie Zenker
- Institute of Immunology, University of Münster, Münster, Germany
| | - Jan Rossaint
- Department of Anaesthesiology and Intensive Care, University of Münster, Münster, Germany
| | - Anna Hölscher
- Institute of Immunology, University of Münster, Münster, Germany
| | - Michele Pohlen
- Institute of Immunology, University of Münster, Münster, Germany
| | - Alexander Zarbock
- Department of Anaesthesiology and Intensive Care, University of Münster, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| |
Collapse
|
38
|
Kumar V, Fleming T, Terjung S, Gorzelanny C, Gebhardt C, Agrawal R, Mall MA, Ranzinger J, Zeier M, Madhusudhan T, Ranjan S, Isermann B, Liesz A, Deshpande D, Häring HU, Biswas SK, Reynolds PR, Hammes HP, Peperkok R, Angel P, Herzig S, Nawroth PP. Homeostatic nuclear RAGE-ATM interaction is essential for efficient DNA repair. Nucleic Acids Res 2017; 45:10595-10613. [PMID: 28977635 PMCID: PMC5737477 DOI: 10.1093/nar/gkx705] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 08/02/2017] [Indexed: 12/12/2022] Open
Abstract
The integrity of genome is a prerequisite for healthy life. Indeed, defects in DNA repair have been associated with several human diseases, including tissue-fibrosis, neurodegeneration and cancer. Despite decades of extensive research, the spatio-mechanical processes of double-strand break (DSB)-repair, especially the auxiliary factor(s) that can stimulate accurate and timely repair, have remained elusive. Here, we report an ATM-kinase dependent, unforeseen function of the nuclear isoform of the Receptor for Advanced Glycation End-products (nRAGE) in DSB-repair. RAGE is phosphorylated at Serine376 and Serine389 by the ATM kinase and is recruited to the site of DNA-DSBs via an early DNA damage response. nRAGE preferentially co-localized with the MRE11 nuclease subunit of the MRN complex and orchestrates its nucleolytic activity to the ATR kinase signaling. This promotes efficient RPA2S4-S8 and CHK1S345 phosphorylation and thereby prevents cellular senescence, IPF and carcinoma formation. Accordingly, loss of RAGE causatively linked to perpetual DSBs signaling, cellular senescence and fibrosis. Importantly, in a mouse model of idiopathic pulmonary fibrosis (RAGE−/−), reconstitution of RAGE efficiently restored DSB-repair and reversed pathological anomalies. Collectively, this study identifies nRAGE as a master regulator of DSB-repair, the absence of which orchestrates persistent DSB signaling to senescence, tissue-fibrosis and oncogenesis.
Collapse
Affiliation(s)
- Varun Kumar
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Helmholtz-Zentrum, München, Germany
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Helmholtz-Zentrum, München, Germany
| | - Stefan Terjung
- European Molecular Biology Laboratory, Advanced Light Microscopy Facility, Heidelberg, Germany
| | - Christian Gorzelanny
- Experimental Dermatology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christoffer Gebhardt
- Division of Dermatooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Signal Transduction and Growth Control DKFZ DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Raman Agrawal
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, INF 156, Heidelberg, Germany
| | - Marcus A Mall
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, INF 156, Heidelberg, Germany
| | - Julia Ranzinger
- Department of Nephrology, University of Heidelberg, Heidelberg, INF 410, Heidelberg, Germany
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, Heidelberg, INF 410, Heidelberg, Germany
| | - Thati Madhusudhan
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke-University, Magdeburg, Germany
| | - Satish Ranjan
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke-University, Magdeburg, Germany
| | - Berend Isermann
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke-University, Magdeburg, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD) University Hospital München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Divija Deshpande
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, Heidelberg, Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD), Helmholtz-Zentrum, München, Germany.,Department of Internal Medicine, University of Tübingen, Tübingen, Germany
| | - Subrata K Biswas
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka 1000, Bangladesh
| | - Paul R Reynolds
- Department of Physiology and Developmental Biology, Brigham Young University, 3054 Life Sciences Building, Provo, UT 84602, USA
| | - Hans-Peter Hammes
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Rainer Peperkok
- European Molecular Biology Laboratory, Advanced Light Microscopy Facility, Heidelberg, Germany
| | - Peter Angel
- Division of Signal Transduction and Growth Control DKFZ DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan Herzig
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Helmholtz-Zentrum, München, Germany.,Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz-Zentrum, München, Germany
| | - Peter P Nawroth
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, Heidelberg, Germany.,German Center for Diabetes Research (DZD), Helmholtz-Zentrum, München, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz-Zentrum, München, Germany
| |
Collapse
|
39
|
Caraher EJ, Kwon S, Haider SH, Crowley G, Lee A, Ebrahim M, Zhang L, Chen LC, Gordon T, Liu M, Prezant DJ, Schmidt AM, Nolan A. Receptor for advanced glycation end-products and World Trade Center particulate induced lung function loss: A case-cohort study and murine model of acute particulate exposure. PLoS One 2017; 12:e0184331. [PMID: 28926576 PMCID: PMC5604982 DOI: 10.1371/journal.pone.0184331] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 08/22/2017] [Indexed: 12/30/2022] Open
Abstract
World Trade Center-particulate matter(WTC-PM) exposure and metabolic-risk are associated with WTC-Lung Injury(WTC-LI). The receptor for advanced glycation end-products (RAGE) is most highly expressed in the lung, mediates metabolic risk, and single-nucleotide polymorphisms at the AGER-locus predict forced expiratory volume(FEV). Our objectives were to test the hypotheses that RAGE is a biomarker of WTC-LI in the FDNY-cohort and that loss of RAGE in a murine model would protect against acute PM-induced lung disease. We know from previous work that early intense exposure at the time of the WTC collapse was most predictive of WTC-LI therefore we utilized a murine model of intense acute PM-exposure to determine if loss of RAGE is protective and to identify signaling/cytokine intermediates. This study builds on a continuing effort to identify serum biomarkers that predict the development of WTC-LI. A case-cohort design was used to analyze a focused cohort of male never-smokers with normal pre-9/11 lung function. Odds of developing WTC-LI increased by 1.2, 1.8 and 1.0 in firefighters with soluble RAGE (sRAGE)≥97pg/mL, CRP≥2.4mg/L, and MMP-9≤397ng/mL, respectively, assessed in a multivariate logistic regression model (ROCAUC of 0.72). Wild type(WT) and RAGE-deficient(Ager-/-) mice were exposed to PM or PBS-control by oropharyngeal aspiration. Lung function, airway hyperreactivity, bronchoalveolar lavage, histology, transcription factors and plasma/BAL cytokines were quantified. WT-PM mice had decreased FEV and compliance, and increased airway resistance and methacholine reactivity after 24-hours. Decreased IFN-γ and increased LPA were observed in WT-PM mice; similar findings have been reported for firefighters who eventually develop WTC-LI. In the murine model, lack of RAGE was protective from loss of lung function and airway hyperreactivity and was associated with modulation of MAP kinases. We conclude that in a multivariate adjusted model increased sRAGE is associated with WTC-LI. In our murine model, absence of RAGE mitigated acute deleterious effects of PM and may be a biologically plausible mediator of PM-related lung disease.
Collapse
Affiliation(s)
- Erin J. Caraher
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Sophia Kwon
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Syed H. Haider
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, New York, United States of America
| | - George Crowley
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Audrey Lee
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Minah Ebrahim
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Liqun Zhang
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, New York, United States of America
- Department of Respiratory Medicine, PLA, Army General Hospital, Beijing, China
| | - Lung-Chi Chen
- Department of Environmental Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Terry Gordon
- Department of Environmental Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Mengling Liu
- Department of Environmental Medicine, New York University School of Medicine, New York, New York, United States of America
- Department of Population Health, Division of Biostatistics, New York University School of Medicine, New York, New York, United States of America
| | - David J. Prezant
- Bureau of Health Services and Office of Medical Affairs, Fire Department of New York, Brooklyn, New York, United States of America
- Department of Medicine, Pulmonary Medicine Division, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Ann Marie Schmidt
- Departments of Biochemistry and Molecular Pharmacology and Pathology, Division of Endocrinology, New York University School of Medicine, New York, New York, United States of America
| | - Anna Nolan
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, New York, New York, United States of America
- Department of Environmental Medicine, New York University School of Medicine, New York, New York, United States of America
- Bureau of Health Services and Office of Medical Affairs, Fire Department of New York, Brooklyn, New York, United States of America
| |
Collapse
|
40
|
Oczypok EA, Perkins TN, Oury TD. Alveolar Epithelial Cell-Derived Mediators: Potential Direct Regulators of Large Airway and Vascular Responses. Am J Respir Cell Mol Biol 2017; 56:694-699. [PMID: 28080134 DOI: 10.1165/rcmb.2016-0151ps] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bronchial epithelial cells and pulmonary endothelial cells are thought to be the primary modulators of conducting airways and vessels, respectively. However, histological examination of both mouse and human lung tissue reveals that alveolar epithelial cells (AECs) line the adventitia of large airways and vessels and thus are also in a position to directly regulate these structures. The primary purpose of this perspective is to highlight the fact that AECs coat the adventitial surface of every vessel and airway in the lung parenchyma. This localization is ideal for transmitting signals that can contribute to physiologic and pathologic responses in vessels and airways. A few examples of mediators produced by AECs that may contribute to vascular and airway responses are provided to illustrate some of the potential effects that AECs may modulate.
Collapse
Affiliation(s)
- Elizabeth A Oczypok
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
41
|
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: 123] [Impact Index Per Article: 15.4] [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
|
42
|
Sabbione F, Keitelman IA, Iula L, Ferrero M, Giordano MN, Baldi P, Rumbo M, Jancic C, Trevani AS. Neutrophil Extracellular Traps Stimulate Proinflammatory Responses in Human Airway Epithelial Cells. J Innate Immun 2017; 9:387-402. [PMID: 28467984 DOI: 10.1159/000460293] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 02/08/2017] [Indexed: 12/13/2022] Open
Abstract
Tissue injury leads to the release of uric acid (UA). At high local concentrations, UA can form monosodium urate crystals (MSU). MSU and UA stimulate neutrophils to release extracellular traps (NET). Here, we investigated whether these NET could be involved in the development of inflammation by stimulating cytokine release by airway epithelial cells. We found that NET significantly increased the secretion of CXCL8/IL-8 and IL-6 by alveolar and bronchial epithelial cells. These effects were not observed when NETosis was inhibited by Diphenyleneiodonium, elastase inhibitor, or Cl-amidine. Similar findings were made with NET induced by cigarette smoke extract, suggesting that NET proinflammatory capacity is independent of the inducing stimulus. Furthermore, NET affected neither the viability and morphology of epithelial cells nor the barrier integrity of polarized cells. The epithelial stimulatory capacity of NET was not affected by degradation of DNA with micrococcal nuclease, treatment with heparin, or inhibition of the elastase immobilized to DNA, but it was significantly reduced by pretreatment with an anti-HMGB-1 blocking antibody. Altogether, our findings indicate that NET exert direct proinflammatory effects on airway epithelial cells that might contribute in vivo to the further recruitment of neutrophils and the perpetuation of inflammation upon lung tissue damage.
Collapse
Affiliation(s)
- Florencia Sabbione
- Laboratorio de Inmunidad Innata, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Shaver CM, Ware LB. Primary graft dysfunction: pathophysiology to guide new preventive therapies. Expert Rev Respir Med 2017; 11:119-128. [PMID: 28074663 DOI: 10.1080/17476348.2017.1280398] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Primary graft dysfunction (PGD) is a common complication of lung transplantation characterized by acute pulmonary edema associated with bilateral pulmonary infiltrates and hypoxemia in the first 3 post-operative days. Development of PGD is a predictor of poor short- and long-term outcomes after lung transplantation, but there are currently limited tools to prevent its occurrence. Areas covered: Several potentially modifiable donor, recipient, and operative risk factors for PGD have been identified. In addition, basic and translational studies in animals and ex vivo lung perfusion systems have identified several biomarkers and mechanisms of injury in PGD. In this review, we outline the clinical and genetic risk factors for PGD and summarize experimental data exploring PGD mechanisms, with a focus on strategies to reduce PGD risk and on potential novel molecular targets for PGD prevention. Expert commentary: Because of the clinical importance of PGD, development of new therapies for prevention and treatment is critically important. Improved understanding of the pathophysiology of clinical PGD provides a framework to explore novel agents to prevent or reverse PGD. Ex vivo lung perfusion provides a new opportunity for rapid development of therapeutics that target this devastating complication of lung transplantation.
Collapse
Affiliation(s)
- Ciara M Shaver
- a Department of Medicine , Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center , Nashville , TN , USA
| | - Lorraine B Ware
- a Department of Medicine , Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center , Nashville , TN , USA.,b Department of Pathology, Microbiology and Immunology , Vanderbilt University Medical Center , Nashville , TN , USA
| |
Collapse
|
44
|
Lung adenocarcinoma expressing receptor for advanced glycation end-products with primary systemic AL amyloidosis: a case report and literature review. BMC Cancer 2017; 17:22. [PMID: 28056871 PMCID: PMC5216608 DOI: 10.1186/s12885-016-3009-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 12/15/2016] [Indexed: 01/05/2023] Open
Abstract
Background Receptor for advanced glycation end-products (RAGE), a receptor for amyloids, is constitutively expressed in lungs and generally observed to be downregulated in lung cancer tissues. However, increasing levels of RAGE or serum amyloids is associated with poor outcome in lung cancer patients. We report a rare case of primary systemic amyloid light-chain (AL) amyloidosis in biopsy-proven multiple organs with early-stage non-small cell lung cancer (NSCLC) that displayed strong staining for RAGE in the tumour tissue. Interestingly, compared with randomly selected lung cancer biopsy samples, including all representative histological subtypes of NSCLC and small-cell lung cancer, only the NSCLC in the present case showed strong expression for RAGE that can bind amyloids. Case presentation A 71-year-old woman was admitted to our hospital for comprehensive investigation of nephrotic syndrome. Computed tomography showed a small nodule in the right upper lung lobe with hilar mediastinal lymph node enlargement. Pathological examination of transbronchial biopsy samples of the nodule yielded a diagnosis of lung adenocarcinoma. Furthermore, the pathological detection of amyloid deposition in biopsy samples of a subcarinal lymph node, gastric and duodenal mucosa, cardiac muscle, and bone marrow led to a diagnosis of primary systemic AL amyloidosis with nephrotic syndrome and cardiomyopathy. In addition, RAGE was detected in lung tumour tissues surrounded by normal lung tissues with amyloid deposition. Conclusion The RAGE positivity of the lung cancer cells in this case suggests an interaction between amyloid-containing tissues and RAGE-expressing cancer cells. Lung adenocarcinoma with RAGE expression may be a complication of underlying amyloidosis.
Collapse
|
45
|
Reynaert NL, Gopal P, Rutten EP, Wouters EF, Schalkwijk CG. Advanced glycation end products and their receptor in age-related, non-communicable chronic inflammatory diseases; Overview of clinical evidence and potential contributions to disease. Int J Biochem Cell Biol 2016; 81:403-418. [PMID: 27373680 DOI: 10.1016/j.biocel.2016.06.016] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 06/24/2016] [Accepted: 06/28/2016] [Indexed: 12/31/2022]
|
46
|
Miller S, Henry AP, Hodge E, Kheirallah AK, Billington CK, Rimington TL, Bhaker SK, Obeidat M, Melén E, Merid SK, Swan C, Gowland C, Nelson CP, Stewart CE, Bolton CE, Kilty I, Malarstig A, Parker SG, Moffatt MF, Wardlaw AJ, Hall IP, Sayers I. The Ser82 RAGE Variant Affects Lung Function and Serum RAGE in Smokers and sRAGE Production In Vitro. PLoS One 2016; 11:e0164041. [PMID: 27755550 PMCID: PMC5068780 DOI: 10.1371/journal.pone.0164041] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 09/19/2016] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Genome-Wide Association Studies have identified associations between lung function measures and Chronic Obstructive Pulmonary Disease (COPD) and chromosome region 6p21 containing the gene for the Advanced Glycation End Product Receptor (AGER, encoding RAGE). We aimed to (i) characterise RAGE expression in the lung, (ii) identify AGER transcripts, (iii) ascertain if SNP rs2070600 (Gly82Ser C/T) is associated with lung function and serum sRAGE levels and (iv) identify whether the Gly82Ser variant is functionally important in altering sRAGE levels in an airway epithelial cell model. METHODS Immunohistochemistry was used to identify RAGE protein expression in 26 human tissues and qPCR was used to quantify AGER mRNA in lung cells. Gene expression array data was used to identify AGER expression during lung development in 38 fetal lung samples. RNA-Seq was used to identify AGER transcripts in lung cells. sRAGE levels were assessed in cells and patient serum by ELISA. BEAS2B-R1 cells were transfected to overexpress RAGE protein with either the Gly82 or Ser82 variant and sRAGE levels identified. RESULTS Immunohistochemical assessment of 6 adult lung samples identified high RAGE expression in the alveoli of healthy adults and individuals with COPD. AGER/RAGE expression increased across developmental stages in human fetal lung at both the mRNA (38 samples) and protein levels (20 samples). Extensive AGER splicing was identified. The rs2070600T (Ser82) allele is associated with higher FEV1, FEV1/FVC and lower serum sRAGE levels in UK smokers. Using an airway epithelium model overexpressing the Gly82 or Ser82 variants we found that HMGB1 activation of the RAGE-Ser82 receptor results in lower sRAGE production. CONCLUSIONS This study provides new information regarding the expression profile and potential role of RAGE in the human lung and shows a functional role of the Gly82Ser variant. These findings advance our understanding of the potential mechanisms underlying COPD particularly for carriers of this AGER polymorphism.
Collapse
Affiliation(s)
- Suzanne Miller
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
- * E-mail:
| | - Amanda P. Henry
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Emily Hodge
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | | | | | - Tracy L. Rimington
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Sangita K. Bhaker
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Ma’en Obeidat
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Simon K. Merid
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Caroline Swan
- Department of Biology, University of York, York, United Kingdom
| | - Catherine Gowland
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Carl P. Nelson
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Ceri E. Stewart
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Charlotte E. Bolton
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Iain Kilty
- Pfizer Worldwide Research & Development, Cambridge, Massachusetts, United States of America
| | - Anders Malarstig
- Pfizer Worldwide Research & Development, Cambridge, United Kingdom
| | - Stuart G. Parker
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, United Kingdom
| | - Miriam F. Moffatt
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Andrew J. Wardlaw
- Institute for Lung Health, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Ian P. Hall
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Ian Sayers
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
47
|
Ota C, Ishizawa K, Yamada M, Tando Y, He M, Takahashi T, Yamaya M, Yamamoto Y, Yamamoto H, Kure S, Kubo H. Receptor for advanced glycation end products expressed on alveolar epithelial cells is the main target for hyperoxia-induced lung injury. Respir Investig 2015; 54:98-108. [PMID: 26879479 DOI: 10.1016/j.resinv.2015.08.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 08/24/2015] [Accepted: 08/26/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Receptor for advanced glycation end products (RAGE) is abundantly expressed on alveolar epithelial cells (AECs) and participates in innate immune responses such as apoptosis and inflammation. However, it is unclear whether RAGE-mediated apoptosis of AECs is associated with hyperoxia-induced lung injury. METHODS We used wild-type and RAGE-knockout C57BL6/J mice in this study. In addition, we developed bone marrow chimeric mouse models expressing RAGE on hematopoietic or non-hematopoietic cells, including lung parenchymal cells, and compared survival ratios and changes in the permeability of the alveolar-capillary barrier after hyperoxia exposure. Further, we prepared single cell suspensions of lung cells and evaluated the apoptosis of AECs or microvascular endothelial cells (MVECs) by using a combination of antibodies and JC-1 dye. We also examined whether RAGE inhibition decreased hyperoxia-induced apoptosis of human lung epithelial cells in vitro. RESULTS After hyperoxia exposure, mice expressing RAGE on lung cells showed lower survival rate and increased alveolar-capillary permeability than mice expressing RAGE on hematopoietic cells. RAGE-expressing AECs showed significantly higher apoptosis than RAGE-knockout AECs after in vivo hyperoxia exposure. The level of hyperoxia-induced apoptosis was not different in MVECs. However, RAGE-null lung epithelial cells showed lower apoptosis than RAGE-expressing cells in vitro. CONCLUSION These results indicated that RAGE on AECs mainly contributed to hyperoxia-induced lung injury and alveolar-capillary barrier disruption.
Collapse
Affiliation(s)
- Chiharu Ota
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Kota Ishizawa
- Department of Molecular Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Mitsuhiro Yamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Yukiko Tando
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Mei He
- Department of Respiratory Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Toru Takahashi
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Mutsuo Yamaya
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.
| | - Hiroshi Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.
| | - Shigeo Kure
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Hiroshi Kubo
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
48
|
Serban AI, Stanca L, Geicu OI, Dinischiotu A. AGEs-Induced IL-6 Synthesis Precedes RAGE Up-Regulation in HEK 293 Cells: An Alternative Inflammatory Mechanism? Int J Mol Sci 2015; 16:20100-17. [PMID: 26307981 PMCID: PMC4613191 DOI: 10.3390/ijms160920100] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/12/2015] [Accepted: 08/06/2015] [Indexed: 01/21/2023] Open
Abstract
Advanced glycation end products (AGEs) can activate the inflammatory pathways involved in diabetic nephropathy. Understanding these molecular pathways could contribute to therapeutic strategies for diabetes complications. We evaluated the modulation of inflammatory and oxidative markers, as well as the protective mechanisms employed by human embryonic kidney cells (HEK 293) upon exposure to 200 μg/mL bovine serum albumine (BSA) or AGEs–BSA for 12, 24 and 48 h. The mRNA and protein expression levels of AGEs receptor (RAGE) and heat shock proteins (HSPs) 27, 60 and 70, the activity of antioxidant enzymes and the expression levels of eight cytokines were analysed. Cell damage via oxidative mechanisms was evaluated by glutathione and malondialdehyde levels. The data revealed two different time scale responses. First, the up-regulation of interleukin-6 (IL-6), HSP 27 and high catalase activity were detected as early as 12 h after exposure to AGEs–BSA, while the second response, after 24 h, consisted of NF-κB p65, RAGE, HSP 70 and inflammatory cytokine up-regulation, glutathione depletion, malondialdehyde increase and the activation of antioxidant enzymes. IL-6 might be important in the early ignition of inflammatory responses, while the cellular redox imbalance, RAGE activation and NF-κB p65 increased expression further enhance inflammatory signals in HEK 293 cells.
Collapse
Affiliation(s)
- Andreea Iren Serban
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine Bucharest, 105 Splaiul Independentei, district 5, Bucharest 050097, Romania.
| | - Loredana Stanca
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine Bucharest, 105 Splaiul Independentei, district 5, Bucharest 050097, Romania.
| | - Ovidiu Ionut Geicu
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine Bucharest, 105 Splaiul Independentei, district 5, Bucharest 050097, Romania.
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, district 5, Bucharest 050095, Romania.
| | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, district 5, Bucharest 050095, Romania.
| |
Collapse
|
49
|
Circulating HMGB1 and RAGE as Clinical Biomarkers in Malignant and Autoimmune Diseases. Diagnostics (Basel) 2015; 5:219-53. [PMID: 26854151 PMCID: PMC4665591 DOI: 10.3390/diagnostics5020219] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/01/2015] [Accepted: 06/05/2015] [Indexed: 12/18/2022] Open
Abstract
High molecular group box 1 (HMGB1) is a highly conserved member of the HMG-box-family; abundantly expressed in almost all human cells and released in apoptosis; necrosis or by activated immune cells. Once in the extracellular space, HMGB1 can act as a danger associated molecular pattern (DAMP), thus stimulating or inhibiting certain functions of the immune system; depending on the “combinatorial cocktail” of the surrounding milieu. HMGB1 exerts its various functions through binding to a multitude of membrane-bound receptors such as TLR-2; -4 and -9; IL-1 and RAGE (receptor for advanced glycation end products); partly complex-bound with intracellular fragments like nucleosomes. Soluble RAGE in the extracellular space, however, acts as a decoy receptor by binding to HMGB1 and inhibiting its effects. This review aims to outline today’s knowledge of structure, intra- and extracellular functions including mechanisms of release and finally the clinical relevance of HMGB1 and RAGE as clinical biomarkers in therapy monitoring, prediction and prognosis of malignant and autoimmune disease.
Collapse
|
50
|
The receptor for advanced glycation end products (RAGE) contributes to the progression of emphysema in mice. PLoS One 2015; 10:e0118979. [PMID: 25781626 PMCID: PMC4364508 DOI: 10.1371/journal.pone.0118979] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 01/27/2015] [Indexed: 01/11/2023] Open
Abstract
Several recent clinical studies have implied a role for the receptor for advanced glycation end products (RAGE) and its variants in chronic obstructive pulmonary disease (COPD). In this study we have defined a role for RAGE in the pathogenesis of emphysema in mice. RAGE deficient mice (RAGE-/-) exposed to chronic cigarette smoke were significantly protected from smoke induced emphysema as determined by airspace enlargement and had no significant reduction in lung tissue elastance when compared to their air exposed controls contrary to their wild type littermates. The progression of emphysema has been largely attributed to an increased inflammatory cell-mediated elastolysis. Acute cigarette smoke exposure in RAGE-/- mice revealed an impaired early recruitment of neutrophils, approximately a 6-fold decrease compared to wild type mice. Hence, impaired neutrophil recruitment with continued cigarette smoke exposure reduces elastolysis and consequent emphysema.
Collapse
|