1
|
Guo L, Han L, Zhang J, Shen M, Li J, Zhang K, Chen R, Liu H. HMGB1 mediates epithelial-mesenchymal transition and fibrosis in silicosis via RAGE/β-catenin signaling. Chem Biol Interact 2025; 408:111385. [PMID: 39800143 DOI: 10.1016/j.cbi.2025.111385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/15/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Epithelial-mesenchymal transition (EMT) is implicated in the pathogenesis of silicosis. High mobility group box 1 (HMGB1) has been found to induce EMT in fibrotic diseases. Previous studies have revealed a critical role of HMGB1 in silicosis, whereas the detail mechanisms still obscure. Here, we observed that HMGB1 protein was increased in the serum of silicosis patients and in the lung tissues of silicotic mice. The levels of HMGB1, receptor for advanced glycation end products (RAGE) and β-catenin protein were increased in the alveolar EMT cell model established by the treatment of transforming growth factor β1 (TGF-β1) and conditioned mediums derived from silica-stimulated macrophages. The activation of HMGB1, RAGE, β-catenin, EMT process, as well as cell migration triggered by TGF-β1 in RLE-6TN cells could be enhanced by treatment with recombinant HMGB1 protein (rHMGB1) and decreased by HMGB1 chemical inhibitor glycyrrhizin or RAGE inhibitor FPS-ZM1. And RAGE suppression could alleviate HMGB1-mediated the aggravation of β-catenin signaling, cell migration and EMT process induced by TGF-β1. Furthermore, both HMGB1 inhibition and RAGE knockout effectively alleviated the lung function impairment, EMT process, pulmonary inflammation and fibrosis in silicotic mice. These findings suggested that HMGB1 might promote EMT through RAGE/β-catenin axis in silicosis. And HMGB1 might constitute a therapeutic target for ameliorating the fibrosis of silicosis.
Collapse
Affiliation(s)
- Lingli Guo
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China; Hebei Key Laboratory of Organ Fibrosis, School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Lu Han
- Hebei Key Laboratory of Organ Fibrosis, School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Jing Zhang
- LinYi Center for Disease Control and Prevention, LinYi, Shangdong, 276000, China
| | - Mengyao Shen
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China
| | - Jiacheng Li
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China
| | - Kuijie Zhang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China
| | - Ruru Chen
- Hebei Key Laboratory of Organ Fibrosis, School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Heliang Liu
- Hebei Key Laboratory of Organ Fibrosis, School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China.
| |
Collapse
|
2
|
Wang J, Li Y, Chen H, Wang J. Vascular volume changes in radiological patterns of usual interstitial pneumonia in patients with type 2 diabetes. Diabetol Metab Syndr 2024; 16:298. [PMID: 39696634 DOI: 10.1186/s13098-024-01551-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVE This research primarily focuses on exploring the changes in intrapulmonary vascular volume (IPVV) in radiological patterns of usual interstitial pneumonia (UIP) associated with Type 2 Diabetes Mellitus (T2DM), thereby inferring the possible mechanisms of the co-occurrence of diabetes and UIP patterns. METHODS Thin-layer data were post-processed on the basis of high-resolution computed tomography (HRCT) and quantitatively assessed for IPVV. Changes in IPVV were compared between T2DM combined with UIP modality and T2DM non-UIP modality. Correlations between UIP patterns and various markers and confounders, including IPVV, were determined via logistic regression analysis. In this study, the potential of IPVV as a predictor for UIP presence was analysed through the application of subject operating characteristic curve analysis. RESULTS In patients with T2DM, the IPVV demonstrated smaller size in those with combined UIP patterns compared to T2DM patients without UIP patterns (164.4 ± 68.7 vs 202.9 ± 76.3 mL, P = 0.005). We detected a positive correlation between IPVV levels and several variables, including fasting plasma glucose (FPG) (r = 0.404, P < 0.0001), glycated hemoglobin (HbA1c) (r = 0.225, P = 0.022), serum uric acid (SUA) (r = 0.332, P = 0.0007) and HRCT scores (r = 0.288, P = 0.024). Conversely, negative correlations were noted with total cholesterol (TC) (r = -0.220, P = 0.028) and cystatin-C (Cys-C) (r = -0.215, P = 0.038). Multivariate logistic regression analysis identified independent associations between the presence of UIP and several factors: IPVV, age, smoking history, and FPG. In assessing the combined UIP pattern among T2DM patients, IPVV levels exhibited a sensitivity of 70.5% and a specificity of 58.5%, generating an AUC of 0.645. CONCLUSION In individuals diagnosed with T2DM alongside UIP, a substantial decline in IPVV was documented. This diminution correlates with the presence of UIP, suggesting that IPVV may serve as a potent biomarker for detecting UIP patterns in individuals with T2DM. This may suggest that the mechanism behind the co-occurrence of T2DM with UIP patterns is attributed to alterations in the pulmonary microvasculature, potentially representing one of the vascular complications associated with diabetes.
Collapse
Affiliation(s)
- Jiarong Wang
- Department of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Department of Radiology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yuanchao Li
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Hao Chen
- Department of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jianbo Wang
- Department of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
3
|
Cross K, Vetter SW, Alam Y, Hasan MZ, Nath AD, Leclerc E. Role of the Receptor for Advanced Glycation End Products (RAGE) and Its Ligands in Inflammatory Responses. Biomolecules 2024; 14:1550. [PMID: 39766257 PMCID: PMC11673996 DOI: 10.3390/biom14121550] [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: 10/21/2024] [Revised: 11/30/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
Since its discovery in 1992, the receptor for advanced glycation end products (RAGE) has emerged as a key receptor in many pathological conditions, especially in inflammatory conditions. RAGE is expressed by most, if not all, immune cells and can be activated by many ligands. One characteristic of RAGE is that its ligands are structurally very diverse and belong to different classes of molecules, making RAGE a promiscuous receptor. Many of RAGE ligands are damaged associated molecular patterns (DAMPs) that are released by cells under inflammatory conditions. Although RAGE has been at the center of a lot of research in the past three decades, a clear understanding of the mechanisms of RAGE activation by its ligands is still missing. In this review, we summarize the current knowledge of the role of RAGE and its ligands in inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | - Estelle Leclerc
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA; (K.C.); (S.W.V.); (Y.A.); (M.Z.H.); (A.D.N.)
| |
Collapse
|
4
|
Kitadai E, Yamaguchi K, Ohshimo S, Iwamoto H, Sakamoto S, Horimasu Y, Masuda T, Nakashima T, Hamada H, Bonella F, Guzman J, Costabel U, Hattori N. Serum soluble isoform of receptor for advanced glycation end product is a predictive biomarker for acute exacerbation of idiopathic pulmonary fibrosis: a German and Japanese cohort study. Respir Res 2024; 25:405. [PMID: 39529063 PMCID: PMC11552171 DOI: 10.1186/s12931-024-03014-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The receptor for advanced glycation end product (RAGE) is a transmembrane receptor accelerating a pro-inflammatory signal. RAGE signalling is promoted by decreased soluble isoform of RAGE (sRAGE), which is a decoy receptor for RAGE ligands, and RAGE SNP rs2070600 minor allele. In Caucasian and Japanese cohorts, low circulatory sRAGE levels and presence of the minor allele are associated with poor survival of idiopathic pulmonary fibrosis (IPF) and increased disease susceptibility to interstitial lung disease, respectively. However, whether sRAGE and RAGE SNP rs2070600 are associated with acute exacerbation of IPF (AE-IPF) is unclear. METHODS This retrospective cohort study evaluated the association between the onset of AE-IPF and serum sRAGE levels in 69 German and 102 Japanese patients with IPF. The association of AE-IPF with RAGE SNP rs2070600 in 51 German and 84 Japanese patients, whose DNA samples were stored, was also investigated. RESULTS In each cohort, the incidence of AE-IPF was significantly and reproducibly higher in the patients with sRAGE < 467.1 pg/mL. In a pooled exploratory analysis, the incidence of AE-IPF was lowest in the patients with higher sRAGE levels and rs2070600 minor allele, although no significant difference in the incidence was observed between the patients with and without the rs2070600 minor allele. CONCLUSIONS Low sRAGE levels were associated with increased incidence of AE-IPF in two independent cohorts of different ethnicities. The combination of rs2070600 and sRAGE levels may stratify patients with IPF for the risk of AE.
Collapse
Affiliation(s)
- Erika Kitadai
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Kakuhiro Yamaguchi
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan.
| | - Shinichiro Ohshimo
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroshi Iwamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Shinjiro Sakamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Yasushi Horimasu
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Takeshi Masuda
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Taku Nakashima
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Hironobu Hamada
- Department of Physical Analysis and Therapeutic Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Francesco Bonella
- Center for Interstitial and Rare Lung Disease, Department of Pneumology, Ruhrlandklinik University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Josune Guzman
- General and Experimental Pathology, Ruhr-University, Bochum, Germany
| | - Ulrich Costabel
- Center for Interstitial and Rare Lung Disease, Department of Pneumology, Ruhrlandklinik University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
5
|
Zhang H, Hua H, Liu J, Wang C, Zhu C, Xia Q, Jiang W, Cheng X, Hu X, Zhang Y. Integrative analysis of the efficacy and pharmacological mechanism of Xuefu Zhuyu decoction in idiopathic pulmonary fibrosis via evidence-based medicine, bioinformatics, and experimental verification. Heliyon 2024; 10:e38122. [PMID: 39416822 PMCID: PMC11481653 DOI: 10.1016/j.heliyon.2024.e38122] [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: 02/11/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Objective We used evidence-based medicine, bioinformatics and experimental verification to comprehensively analyze the efficacy and pharmacological mechanism of Xuefu Zhuyu decoction (XFZYD) in the treatment of idiopathic pulmonary fibrosis (IPF). Methods Major databases were retrieved for randomized controlled trials (RCTs) of XFZYD treating IPF to perform meta-analysis. Active ingredients and target genes of XFZYD were identified from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). IPF-related differentially expressed genes (DEGs) were identified from the Gene Expression Omnibus (GEO) database. The RGUI software was utilized for Gene Ontology (GO) functional enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. The ingredient-target and protein-protein interaction (PPI) networks were achieved through Cytoscape software and the STRING database to identify the key compounds and target proteins. Molecular docking was performed using AutoDockTool and AutoDock Vina software. The effect between key compounds and target proteins was verified in animal experiments. Results Six RCTs were included for meta-analysis, which uncovered that the total effective rate of clinical efficacy was higher in the experimental group than control group. Then, 156 active ingredients and 254 target genes of XFZYD, and 1,566 IPF-related DEGs were identified. The intersection analysis identified 48 target genes correlating with 130 active ingredients of XFZYD treating IPF. GO functional enrichment, KEGG pathway enrichment, ingredient-target network and PPI network were achieved. Following the identification of key compounds and target proteins, we performed molecular docking. Ultimately, our research focused on the key compound quercetin for experimental validation to assess its interactions with two key target proteins, JUN and PTGS2. Conclusion The effectiveness of XFZYD on IPF has been substantiated through evidence-based medicine. The pharmacological mechanism of XFZYD for IPF treatment involves a complex interplay of various compounds and targets, with quercetin exerting pronounced impacts on JUN and PTGS2 proteins.
Collapse
Affiliation(s)
- Huizhe Zhang
- Department of Respiratory Medicine, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng TCM Hospital, Yancheng, Jiangsu, 224005, China
| | - Haibing Hua
- Department of Gastroenterology, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu, 214400, China
| | - Jian Liu
- Department of Respiratory Medicine, Xuejia People's Hospital of Xinbei District, Changzhou, Jiangsu, 213003, China
| | - Cong Wang
- Department of Pulmonary and Critical Care Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu, 214400, China
- Research Institute of Respiratory Diseases, Jiangsu Province Clinical Academy of Traditional Chinese Medicine (Jiangyin Branch), Jiangyin, Jiangsu, 214400, China
| | - Chenjing Zhu
- Department of Pulmonary and Critical Care Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu, 214400, China
- Research Institute of Respiratory Diseases, Jiangsu Province Clinical Academy of Traditional Chinese Medicine (Jiangyin Branch), Jiangyin, Jiangsu, 214400, China
| | - Qingqing Xia
- Department of Pulmonary and Critical Care Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu, 214400, China
- Research Institute of Respiratory Diseases, Jiangsu Province Clinical Academy of Traditional Chinese Medicine (Jiangyin Branch), Jiangyin, Jiangsu, 214400, China
| | - Weilong Jiang
- Department of Pulmonary and Critical Care Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu, 214400, China
- Research Institute of Respiratory Diseases, Jiangsu Province Clinical Academy of Traditional Chinese Medicine (Jiangyin Branch), Jiangyin, Jiangsu, 214400, China
| | - Xiangjin Cheng
- Department of Critical Care Medicine, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng TCM Hospital, Yancheng, Jiangsu, 224005, China
| | - Xiaodong Hu
- Department of Pulmonary and Critical Care Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu, 214400, China
- Research Institute of Respiratory Diseases, Jiangsu Province Clinical Academy of Traditional Chinese Medicine (Jiangyin Branch), Jiangyin, Jiangsu, 214400, China
| | - Yufeng Zhang
- Department of Pulmonary and Critical Care Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu, 214400, China
- Research Institute of Respiratory Diseases, Jiangsu Province Clinical Academy of Traditional Chinese Medicine (Jiangyin Branch), Jiangyin, Jiangsu, 214400, China
| |
Collapse
|
6
|
Burgy O, Mayr CH, Schenesse D, Fousekis Papakonstantinou E, Ballester B, Sengupta A, She Y, Hu Q, Melo-Narvaéz MC, Jain E, Pestoni JC, Mozurak M, Estrada-Bernal A, Onwuka U, Coughlan C, Parimon T, Chen P, Heimerl T, Bange G, Schmeck BT, Lindner M, Hilgendorff A, Ruppert C, Güenther A, Mann M, Yildirim AÖ, Eickelberg O, Jung AL, Schiller HB, Lehmann M, Burgstaller G, Königshoff M. Fibroblast-derived extracellular vesicles contain SFRP1 and mediate pulmonary fibrosis. JCI Insight 2024; 9:e168889. [PMID: 39315549 PMCID: PMC11457858 DOI: 10.1172/jci.insight.168889] [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: 02/17/2023] [Accepted: 08/07/2024] [Indexed: 09/25/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal chronic lung disease characterized by aberrant intercellular communication, extracellular matrix deposition, and destruction of functional lung tissue. While extracellular vesicles (EVs) accumulate in the IPF lung, their cargo and biological effects remain unclear. We interrogated the proteome of EV and non-EV fractions during pulmonary fibrosis and characterized their contribution to fibrosis. EVs accumulated 14 days after bleomycin challenge, correlating with decreased lung function and initiated fibrogenesis in healthy precision-cut lung slices. Label-free proteomics of bronchoalveolar lavage fluid EVs (BALF-EVs) collected from mice challenged with bleomycin or control identified 107 proteins enriched in fibrotic vesicles. Multiomic analysis revealed fibroblasts as a major cellular source of BALF-EV cargo, which was enriched in secreted frizzled related protein 1 (SFRP1). Sfrp1 deficiency inhibited the activity of fibroblast-derived EVs to potentiate lung fibrosis in vivo. SFRP1 led to increased transitional cell markers, such as keratin 8, and WNT/β-catenin signaling in primary alveolar type 2 cells. SFRP1 was expressed within the IPF lung and localized at the surface of EVs from patient-derived fibroblasts and BALF. Our work reveals altered EV protein cargo in fibrotic EVs promoting fibrogenesis and identifies fibroblast-derived vesicular SFRP1 as a fibrotic mediator and potential therapeutic target for IPF.
Collapse
Affiliation(s)
- Olivier Burgy
- INSERM U1231 Center for Translational and Molecular Medicine (CTM), Faculty of Health Sciences, Université de Bourgogne, Dijon, France
- Reference Center for Rare Pulmonary Diseases, University Hospital Dijon-Bourgogne, Dijon, France
| | - Christoph H. Mayr
- Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Déborah Schenesse
- INSERM U1231 Center for Translational and Molecular Medicine (CTM), Faculty of Health Sciences, Université de Bourgogne, Dijon, France
- Reference Center for Rare Pulmonary Diseases, University Hospital Dijon-Bourgogne, Dijon, France
- Department of Pulmonary Medicine and Intensive Care Unit, University Hospital Dijon-Bourgogne, Dijon, France
| | | | - Beatriz Ballester
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
- Faculty of Health Sciences, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| | - Arunima Sengupta
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Yixin She
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Qianjiang Hu
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Maria Camila Melo-Narvaéz
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Eshita Jain
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Jeanine C. Pestoni
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Molly Mozurak
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adriana Estrada-Bernal
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ugochi Onwuka
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christina Coughlan
- Division of Neurology, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Tanyalak Parimon
- Women’s Guild Lung Institute, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Peter Chen
- Women’s Guild Lung Institute, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | | | - Gert Bange
- Center for Synthetic Microbiology (SYNMIKRO) and
| | - Bernd T. Schmeck
- Department of Pulmonary Medicine and Intensive Care Unit, University Hospital Dijon-Bourgogne, Dijon, France
- Center for Synthetic Microbiology (SYNMIKRO) and
- Core Facility Flow Cytometry – Bacterial Vesicles, Philipps-University Marburg, Marburg, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC) Giessen Biobank, Justus-Liebig-University Giessen, DZL, Giessen, Germany
| | - Michael Lindner
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
- Paracelsus Medical Private University, Salzburg, Austria
| | - Anne Hilgendorff
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Clemens Ruppert
- Universities of Giessen and Marburg Lung Center (UGMLC) Giessen Biobank, Justus-Liebig-University Giessen, DZL, Giessen, Germany
| | - Andreas Güenther
- Universities of Giessen and Marburg Lung Center (UGMLC) Giessen Biobank, Justus-Liebig-University Giessen, DZL, Giessen, Germany
- European IPF Registry (eurIPFreg), Center for Interstitial and Rare Lung Diseases, UGMLC, Justus-Liebig University Giessen, DZL, Giessen, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Oliver Eickelberg
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anna Lena Jung
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
- Core Facility Flow Cytometry – Bacterial Vesicles, Philipps-University Marburg, Marburg, Germany
| | - Herbert B. Schiller
- Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians University, Munich, Germany
- Research Unit for Precision Regenerative Medicine, Helmholtz Munich, Munich, Germany
| | - Mareike Lehmann
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Gerald Burgstaller
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Melanie Königshoff
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Geriatric Research Education and Clinical Center (GRECC) at the VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
Ancer-Rodríguez J, Gopar-Cuevas Y, García-Aguilar K, Chávez-Briones MDL, Miranda-Maldonado I, Ancer-Arellano A, Ortega-Martínez M, Jaramillo-Rangel G. Cell Proliferation and Apoptosis-Key Players in the Lung Aging Process. Int J Mol Sci 2024; 25:7867. [PMID: 39063108 PMCID: PMC11276691 DOI: 10.3390/ijms25147867] [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: 06/18/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Currently, the global lifespan has increased, resulting in a higher proportion of the population over 65 years. Changes that occur in the lung during aging increase the risk of developing acute and chronic lung diseases, such as acute respiratory distress syndrome, chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and lung cancer. During normal tissue homeostasis, cell proliferation and apoptosis create a dynamic balance that constitutes the physiological cell turnover. In basal conditions, the lungs have a low rate of cell turnover compared to other organs. During aging, changes in the rate of cell turnover in the lung are observed. In this work, we review the literature that evaluates the role of molecules involved in cell proliferation and apoptosis in lung aging and in the development of age-related lung diseases. The list of molecules that regulate cell proliferation, apoptosis, or both processes in lung aging includes TNC, FOXM1, DNA-PKcs, MicroRNAs, BCL-W, BCL-XL, TCF21, p16, NOX4, NRF2, MDM4, RPIA, DHEA, and MMP28. However, despite the studies carried out to date, the complete signaling pathways that regulate cell turnover in lung aging are still unknown. More research is needed to understand the changes that lead to the development of age-related lung diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Gilberto Jaramillo-Rangel
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico; (J.A.-R.); (Y.G.-C.); (M.-d.-L.C.-B.); (I.M.-M.); (A.A.-A.); (M.O.-M.)
| |
Collapse
|
8
|
Wang Y, Wang X, Du C, Wang Z, Wang J, Zhou N, Wang B, Tan K, Fan Y, Cao P. Glycolysis and beyond in glucose metabolism: exploring pulmonary fibrosis at the metabolic crossroads. Front Endocrinol (Lausanne) 2024; 15:1379521. [PMID: 38854692 PMCID: PMC11157045 DOI: 10.3389/fendo.2024.1379521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/07/2024] [Indexed: 06/11/2024] Open
Abstract
At present, pulmonary fibrosis (PF) is a prevalent and irreversible lung disease with limited treatment options, and idiopathic pulmonary fibrosis (IPF) is one of its most common forms. Recent research has highlighted PF as a metabolic-related disease, including dysregulated iron, mitochondria, lipid, and glucose homeostasis. Systematic reports on the regulatory roles of glucose metabolism in PF are rare. This study explores the intricate relationships and signaling pathways between glucose metabolic processes and PF, delving into how key factors involved in glucose metabolism regulate PF progression, and the interplay between them. Specifically, we examined various enzymes, such as hexokinase (HK), 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), pyruvate kinase (PK), and lactate dehydrogenase (LDH), illustrating their regulatory roles in PF. It highlights the significance of lactate, alongside the role of pyruvate dehydrogenase kinase (PDK) and glucose transporters (GLUTs) in modulating pulmonary fibrosis and glucose metabolism. Additionally, critical regulatory factors such as transforming growth factor-beta (TGF-β), interleukin-1 beta (IL-1β), and hypoxia-inducible factor 1 subunit alpha (HIF-1α) were discussed, demonstrating their impact on both PF and glucose metabolic pathways. It underscores the pivotal role of AMP-activated protein kinase (AMPK) in this interplay, drawing connections between diabetes mellitus, insulin, insulin-like growth factors, and peroxisome proliferator-activated receptor gamma (PPARγ) with PF. This study emphasizes the role of key enzymes, regulators, and glucose transporters in fibrogenesis, suggesting the potential of targeting glucose metabolism for the clinical diagnosis and treatment of PF, and proposing new promising avenues for future research and therapeutic development.
Collapse
Affiliation(s)
- Yuejiao Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, Hebei, China
| | - Xue Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, Hebei, China
| | - Chaoqi Du
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, Hebei, China
| | - Zeming Wang
- Department of Laboratory, Hebei Provincial People’s Hospital, Shijiazhuang, Hebei, China
| | - Jiahui Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, Hebei, China
| | - Nan Zhou
- Department of Gynecology, Xingtai People’s Hospital, Xingtai, Hebei, China
| | - Baohua Wang
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ke Tan
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, Hebei, China
| | - Yumei Fan
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, Hebei, China
| | - Pengxiu Cao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, Hebei, China
| |
Collapse
|
9
|
Lv X, Shang Y, Ning Y, Yu W, Wang J. Pharmacological targets of SGLT2 inhibitors on IgA nephropathy and membranous nephropathy: a mendelian randomization study. Front Pharmacol 2024; 15:1399881. [PMID: 38846092 PMCID: PMC11155304 DOI: 10.3389/fphar.2024.1399881] [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: 03/12/2024] [Accepted: 04/30/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Emerging research suggests that sodium-glucose cotransporter 2 (SGLT2) inhibitors may play a pivotal role in the treatment of primary glomerular diseases. This study was aimed to investigate potential pharmacological targets connecting SGLT2 inhibitors with IgA nephropathy (IgAN) and membranous nephropathy (MN). Methods A univariate Mendelian randomization (MR) analysis was conducted using publicly available genome-wide association studies (GWAS) datasets. Co-localization analysis was used to identify potential connections between target genes and IgAN and MN. Then, Comparative Toxicogenomics Database (CTD) was employed to predict diseases associated with these target genes and SGLT2 inhibitors (canagliflozin, dapagliflozin, and empagliflozin). Subsequently, phenotypic scan analyses were applied to explore the causal relationships between the predicted diseases and target genes. Finally, we analyzed the immune signaling pathways involving pharmacological target genes using the Kyoto encyclopedia of genes and genomes (KEGG). Results The results of MR analysis revealed that eight drug targets were causally linked to the occurrence of IgAN, while 14 drug targets were linked to MN. In the case of IgAN, LCN2 and AGER emerged as co-localized genes related to the pharmacological agent of dapagliflozin and the occurrence of IgAN. LCN2 was identified as a risk factor, while AGER was exhibited a protective role. KEGG analysis revealed that LCN2 is involved in the interleukin (IL)-17 immune signaling pathway, while AGER is associated with the neutrophil extracellular traps (NETs) signaling immune pathway. No positive co-localization results of the target genes were observed between two other SGLT2 inhibitors (canagliflozin and empagliflozin) and the occurrence of IgAN, nor between the three SGLT2 inhibitors and the occurrence of MN. Conclusion Our study provided evidence supporting a causal relationship between specific SGLT2 inhibitors and IgAN. Furthermore, we found that dapagliflozin may act on IgAN through the genes LCN2 and AGER.
Collapse
Affiliation(s)
- Xin Lv
- Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Shang
- Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Ning
- Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weimin Yu
- Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Wang
- Department of Nephrology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
10
|
Delrue C, Speeckaert R, Delanghe JR, Speeckaert MM. Breath of fresh air: Investigating the link between AGEs, sRAGE, and lung diseases. VITAMINS AND HORMONES 2024; 125:311-365. [PMID: 38997169 DOI: 10.1016/bs.vh.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Advanced glycation end products (AGEs) are compounds formed via non-enzymatic reactions between reducing sugars and amino acids or proteins. AGEs can accumulate in various tissues and organs and have been implicated in the development and progression of various diseases, including lung diseases. The receptor of advanced glycation end products (RAGE) is a receptor that can bind to advanced AGEs and induce several cellular processes such as inflammation and oxidative stress. Several studies have shown that both AGEs and RAGE play a role in the pathogenesis of lung diseases, such as chronic obstructive pulmonary disease, asthma, idiopathic pulmonary fibrosis, cystic fibrosis, and acute lung injury. Moreover, the soluble form of the receptor for advanced glycation end products (sRAGE) has demonstrated its ability to function as a decoy receptor, possessing beneficial characteristics such as anti-inflammatory, antioxidant, and anti-fibrotic properties. These qualities make it an encouraging focus for therapeutic intervention in managing pulmonary disorders. This review highlights the current understanding of the roles of AGEs and (s)RAGE in pulmonary diseases and their potential as biomarkers and therapeutic targets for preventing and treating these pathologies.
Collapse
Affiliation(s)
- Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium
| | | | - Joris R Delanghe
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Marijn M Speeckaert
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium; Research Foundation-Flanders (FWO), Brussels, Belgium.
| |
Collapse
|
11
|
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
|
12
|
Zhang Z, Deng X, Gu W, Jiao Y, Su C, Liu H, Ma W, Zhang H, Xiang R, Wang D, Wang Y, Chunguo W, Meng F. Jianghu decoction and its active component polydatin inhibit inflammation and fibrotic lesions in the lungs of ILD mice via the AMPK signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117003. [PMID: 37543150 DOI: 10.1016/j.jep.2023.117003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 07/29/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Recently, interstitial lung disease (ILD) morbidity and mortality have been increasing with insidious epidemiological characteristics. Jianghu decoction (JH) is an effective Chinese medicine for ILD. AIM OF THE STUDY We aimed to reveal the material basis and mechanism of action of JH in the treatment of ILD. MATERIALS AND METHODS In this study, an ILD mouse model was constructed with bleomycin. HE staining, transcriptome analysis, parallel reaction monitoring-mass spectrometry (PRM-MS), UPLC‒MS, and western blotting assays were conducted. RESULTS HE staining results showed that JH effectively reduced inflammation and fibrosis foci in the lungs of the ILD model. Furthermore, transcriptome analysis revealed that JH regulates a set of biological signaling pathways related to immune inflammation and fibrosis. PRM-MS combined with western blotting was applied to detect inflammation and fibrosis involving proteins in lung tissue. JH effectively reversed the aberrant expression of HMGB1, RAGE, SEPTIN4, ACTA2, and ITGAV proteins in the model group. AMPK was identified as the core upstream regulatory protein for JH-mediated ILD regulation. In addition, UHPLC‒MS technology was applied to determine the active ingredients of JH. A total of 80 components were identified from JH, and polydatin (PD) was identified as the active ingredient that effectively alleviated lung fibrosis and inflammatory injury in ILD mice. To illustrate the molecular regulatory network of JH and PD in alleviating lung fibrosis and inflammatory injury, we also examined inflammation and fibrosis-related molecules downstream of the AMPK pathway with RT‒qPCR and western blotting. CONCLUSIONS The results showed that both JH and its active component PD exert synergistic inhibition on pulmonary fibrosis and inflammation. Specifically, the AMPK/PGC1α/PPARγ signaling pathway was activated, and the AMPK/HMGB1/RAGE signaling pathway was inhibited in ILD lungs responding to JH or PD administration.
Collapse
Affiliation(s)
- Zhengju Zhang
- Dongfang Hospital of Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Xinqi Deng
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, China
| | - Wen Gu
- Beijing Hospital of Traditional Chinese Medicine of Capital Medical University, Beijing, China
| | - Yang Jiao
- Dongfang Hospital of Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Canyu Su
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Liu
- Dongfang Hospital of Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Weiguo Ma
- Dongfang Hospital of Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Honghong Zhang
- Shunyi Hospital, Beijing Traditional Chinese Medicine Hospital, Beijing, China
| | - Ruiyang Xiang
- Yongledian Health Service Center of Tongzhou District, Beijing, China
| | - Dali Wang
- Yanshan Community Health Service Center, Beijing, China
| | - Yanan Wang
- Beijing Hospital of Traditional Chinese Medicine of Capital Medical University, Beijing, China.
| | - Wang Chunguo
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Fengxian Meng
- Dongfang Hospital of Beijing University of Traditional Chinese Medicine, Beijing, China.
| |
Collapse
|
13
|
Al Hamwi G, Namasivayam V, Büschbell B, Gedschold R, Golz S, Müller CE. Proinflammatory chemokine CXCL14 activates MAS-related G protein-coupled receptor MRGPRX2 and its putative mouse ortholog MRGPRB2. Commun Biol 2024; 7:52. [PMID: 38184723 PMCID: PMC10771525 DOI: 10.1038/s42003-023-05739-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/21/2023] [Indexed: 01/08/2024] Open
Abstract
Patients with idiopathic pulmonary fibrosis show a strongly upregulated expression of chemokine CXCL14, whose target is still unknown. Screening of CXCL14 in a panel of human G protein-coupled receptors (GPCRs) revealed its potent and selective activation of the orphan MAS-related GPCR X2 (MRGPRX2). This receptor is expressed on mast cells and - like CXCL14 - upregulated in bronchial inflammation. CXCL14 induces robust activation of MRGPRX2 and its putative mouse ortholog MRGPRB2 in G protein-dependent and β-arrestin recruitment assays that is blocked by a selective MRGPRX2/B2 antagonist. Truncation combined with mutagenesis and computational studies identified the pharmacophoric sequence of CXCL14 and its presumed interaction with the receptor. Intriguingly, C-terminal domain sequences of CXCL14 consisting of 4 to 11 amino acids display similar or increased potency and efficacy compared to the full CXCL14 sequence (77 amino acids). These results provide a rational basis for the future development of potential idiopathic pulmonary fibrosis therapies.
Collapse
Affiliation(s)
- Ghazl Al Hamwi
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Vigneshwaran Namasivayam
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Beatriz Büschbell
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Robin Gedschold
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Stefan Golz
- Lead Identification & Characterization, Pharma Research and Development Center, Bayer AG, Wuppertal, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany.
| |
Collapse
|
14
|
Wei J, Zhan J, Ji H, Xu Y, Xu Q, Zhu X, Liu Y. Fibroblast Upregulation of Vitamin D Receptor Represents a Self-Protective Response to Limit Fibroblast Proliferation and Activation during Pulmonary Fibrosis. Antioxidants (Basel) 2023; 12:1634. [PMID: 37627629 PMCID: PMC10451996 DOI: 10.3390/antiox12081634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Dysregulation of vitamin D receptor (VDR) is implicated in chronic obstructive pulmonary disease. However, whether VDR dysregulation contributes to the development of pulmonary fibrosis remains largely unknown. Analysis of bulk and single-cell RNA profiling datasets revealed VDR upregulation in lung fibroblasts from patients with pulmonary fibrosis or fibrotic mice, which was validated in lung fibroblasts from bleomycin-exposed mice and bleomycin-treated fibroblasts. Stable VDR knockdown promoted, whereas the VDR agonist paricalcitol suppressed lung fibroblast proliferation and activation. Gene set enrichment analysis (GSEA) showed that the JAK/STAT pathway and unfolded protein response (UPR), a process related to endoplasmic reticulum (ER) stress, were enriched in lung fibroblasts of fibrotic lungs. Stable VDR knockdown stimulated, but paricalcitol suppressed ER stress and JAK1/STAT3 activation in lung fibroblasts. The STAT3 inhibitor blocked bleomycin- or stable VDR knockdown-induced ER stress. Paricalcitol inhibited the bleomycin-induced enrichment of STAT3 to the ATF6 promoter, thereby suppressing ATF6 expression in fibroblasts. Paricalcitol or intrapulmonary VDR overexpression inactivated JAK1/STAT3 and suppressed ER stress in bleomycin-treated mice, thus resulting in the inhibition of fibroblast proliferation and activation. Collectively, this study suggests that fibroblast VDR upregulation may be a self-protective response to limit fibroblast proliferation and activation during pulmonary fibrosis by suppressing the JAK1/STAT3/ER stress pathway.
Collapse
Affiliation(s)
- Juan Wei
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
- School of Sports and Health, Nanjing Sport Institute, Nanjing 210014, China
| | - Junhui Zhan
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| | - Hui Ji
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| | - Yitong Xu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| | - Qingfeng Xu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| | - Xiaoyan Zhu
- Department of Physiology, Navy Medical University, Shanghai 200433, China
| | - Yujian Liu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| |
Collapse
|
15
|
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
|
16
|
Liu J, Jin Z, Wang X, Jakoš T, Zhu J, Yuan Y. RAGE pathways play an important role in regulation of organ fibrosis. Life Sci 2023; 323:121713. [PMID: 37088412 DOI: 10.1016/j.lfs.2023.121713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/09/2023] [Accepted: 04/18/2023] [Indexed: 04/25/2023]
Abstract
Organ fibrosis is a pathological process of fibroblast activation and excessive deposition of extracellular matrix after persistent tissue injury and therefore is a common endpoint of many organ pathologies. Multiple cellular types and soluble mediators, including chemokines, cytokines and non-peptidic factors, are implicated in fibrogenesis and the remodeling of tissue architecture. The molecular basis of the fibrotic process is complex and consists of closely intertwined signaling networks. Research has strived for a better understanding of these pathological mechanisms to potentially reveal novel therapeutic targets for fibrotic diseases. In light of new knowledge, the receptor for advanced glycation end products (RAGE) emerged as an important candidate for the regulation of a wide variety of cellular functions related to fibrosis, including inflammation, cell proliferation, apoptosis, and angiogenesis. RAGE is a pattern recognition receptor that binds a broad range of ligands such as advanced glycation end products, high mobility group box-1, S-100 calcium-binding protein and amyloid beta protein. Although the link between RAGE and fibrosis has been established, the exact mechanisms need be investigated in further studies. The aim of this review is to collect all available information about the intricate function of RAGE and its signaling cascades in the pathogenesis of fibrotic diseases within different organs. In addition, to the major ligands and signaling pathways, we discuss potential strategies for targeting RAGE in fibrosis. We emphasize the functional links between RAGE, inflammation and fibrosis that may guide further studies and the development of improved therapeutic drugs.
Collapse
Affiliation(s)
- Jing Liu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University School of Pharmacy, Shanghai 201100, China.
| | - Zhedong Jin
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University School of Pharmacy, Shanghai 201100, China.
| | - Xiaolong Wang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University School of Pharmacy, Shanghai 201100, China.
| | - Tanja Jakoš
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University School of Pharmacy, Shanghai 201100, China.
| | - Jianwei Zhu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University School of Pharmacy, Shanghai 201100, China.
| | - Yunsheng Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University School of Pharmacy, Shanghai 201100, China.
| |
Collapse
|
17
|
Yamaguchi K, Iwamoto H, Sakamoto S, Horimasu Y, Masuda T, Miyamoto S, Nakashima T, Fujitaka K, Hamada H, Hattori N. Association of the RAGE/RAGE-ligand axis with interstitial lung disease and its acute exacerbation. Respir Investig 2022; 60:531-542. [PMID: 35504814 DOI: 10.1016/j.resinv.2022.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/10/2022] [Accepted: 04/12/2022] [Indexed: 06/14/2023]
Abstract
The receptor for advanced glycation end product (RAGE) is a transmembrane receptor highly expressed in type 1 pneumocytes of healthy lungs. RAGE is considered to play a homeostatic role in the lung, as RAGE knockout mice develop lung fibrosis as they age. In contrast, RAGE can bind numerous ligands, including high-mobility group box 1 (HMGB1). These interactions initiate pro-inflammatory signaling associated with the pathogenesis of lung injury and interstitial lung disease (ILD), including idiopathic pulmonary fibrosis (IPF). ILD is a broad category of diffuse parenchymal lung disease characterized by various extents of lung fibrosis and inflammation, and IPF is a common and progressive ILD of unknown cause. The prognosis of patients with IPF is poor, and acute exacerbation of IPF (AE-IPF) is one of the main causes of death. Recent reports indicate that acute exacerbations can occur in other ILDs (AE-ILD). Notably, ILD is frequently observed in patients with lung cancer, and AE-ILD after surgical procedures or the initiation of chemotherapy for concomitant lung cancer are clinically important due to their association with increased mortality. In this review, we summarize the associations of RAGE/soluble RAGE (sRAGE)/RAGE ligands with the pathogenesis and clinical course of ILD, including IPF and AE-IPF. Additionally, the potential use of sRAGE and RAGE ligands as predictive markers of AE-IPF and cancer treatment-triggered AE-ILD is also discussed.
Collapse
Affiliation(s)
- Kakuhiro Yamaguchi
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, 734-8551, Hiroshima, Japan.
| | - Hiroshi Iwamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, 734-8551, Hiroshima, Japan
| | - Shinjiro Sakamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, 734-8551, Hiroshima, Japan
| | - Yasushi Horimasu
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, 734-8551, Hiroshima, Japan
| | - Takeshi Masuda
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, 734-8551, Hiroshima, Japan
| | - Shintaro Miyamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, 734-8551, Hiroshima, Japan
| | - Taku Nakashima
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, 734-8551, Hiroshima, Japan
| | - Kazunori Fujitaka
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, 734-8551, Hiroshima, Japan
| | - Hironobu Hamada
- Department of Physical Analysis and Therapeutic Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, 734-8551, Hiroshima, Japan
| |
Collapse
|
18
|
Kinscherf NA, Pehar M. Role and Therapeutic Potential of RAGE Signaling in Neurodegeneration. Curr Drug Targets 2022; 23:1191-1209. [PMID: 35702767 PMCID: PMC9589927 DOI: 10.2174/1389450123666220610171005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 04/29/2022] [Indexed: 01/03/2023]
Abstract
Activation of the receptor for advanced glycation end products (RAGE) has been shown to play an active role in the development of multiple neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Amyotrophic Lateral Sclerosis. Although originally identified as a receptor for advanced glycation end products, RAGE is a pattern recognition receptor able to bind multiple ligands. The final outcome of RAGE signaling is defined in a context and cell type specific manner and can exert both neurotoxic and neuroprotective functions. Contributing to the complexity of the RAGE signaling network, different RAGE isoforms with distinctive signaling capabilities have been described. Moreover, multiple RAGE ligands bind other receptors and RAGE antagonism can significantly affect their signaling. Here, we discuss the outcome of celltype specific RAGE signaling in neurodegenerative pathologies. In addition, we will review the different approaches that have been developed to target RAGE signaling and their therapeutic potential. A clear understanding of the outcome of RAGE signaling in a cell type- and disease-specific manner would contribute to advancing the development of new therapies targeting RAGE. The ability to counteract RAGE neurotoxic signaling while preserving its neuroprotective effects would be critical for the success of novel therapies targeting RAGE signaling.
Collapse
Affiliation(s)
- Noah Alexander Kinscherf
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Mariana Pehar
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,Geriatric Research Education Clinical Center, Veterans Affairs Medical Center, Madison, WI, USA
| |
Collapse
|
19
|
Baek H, Jang S, Park J, Jang J, Lee J, Hong SH, Kim WJ, Park SM, Yang SR. Reduced receptor for advanced glycation end products is associated with α-SMA expression in patients with idiopathic pulmonary fibrosis and mice. Lab Anim Res 2021; 37:28. [PMID: 34600594 PMCID: PMC8487524 DOI: 10.1186/s42826-021-00105-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/15/2021] [Indexed: 11/13/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive interstitial lung disease. Despite alveolar epithelial cells is crucial role in lung, its contribution and the associated biomarker remain unknown in the pathogenesis of IPF. Recently, environmental factors including stone dust, silica and cigarette smoking were found as risk factors involved in IPF. Receptor for advanced glycation end products (RAGE) is a member of the immunoglobulin super family of cell surface receptors. It has been shown that interaction between RAGE and its ligands on immune cells mediates cellular migration and regulation of pro-inflammation. RAGE is highly expressed in the lung, in particular, alveolar epithelial cells. Therefore, we determined whether RAGE expression is associated with fibrosis-associated genes in patients with IPF and mice. Results When bleomycin (BLM) was intratracheally administered to C57BL/6 mice for 1, 2 weeks, macrophage and neutrophils were significantly increased. The fibrotic nodule formed and accumulation of collagen was determined after BLM injection in H&E- and Masson’s trichrome staining. Levels of elastin, Col1a1 and fibronectin were increased in quantitative real-time PCR and protein levels of α-SMA was increased in western blot analysis. In the lung tissues of 1 mg/kg BLM-induced mice, RAGE expression was gradually decreased in 1- and 2 weeks in immunohistochemistry and western blot analysis, and 3 mg/kg of BLM-induced mice exhibited decreased RAGE levels while α-SMA expression was increased. We next determined RAGE expression in the lungs of IPF patients using immunohistochemistry. As a result, RAGE expression was decreased, while α-SMA expression was increased compared with non-IPF subjects. Conclusions Our findings suggest that reduced RAGE was associated with increased fibrotic genes in BLM-induced mice and patients with IPF. Therefore, RAGE could be applied with a biomarker for prognosis and diagnosis in the pathogenesis of IPF.
Collapse
Affiliation(s)
- Hyosin Baek
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Soojin Jang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Jaehyun Park
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Jimin Jang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Jooyeon Lee
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Woo Jin Kim
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Sung-Min Park
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea.
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea.
| |
Collapse
|
20
|
Perkins TN, Oury TD. The perplexing role of RAGE in pulmonary fibrosis: causality or casualty? Ther Adv Respir Dis 2021; 15:17534666211016071. [PMID: 34275342 PMCID: PMC8293846 DOI: 10.1177/17534666211016071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease in which most patients die within 3 years of diagnosis. With an unknown etiology, IPF results in progressive fibrosis of the lung parenchyma, diminishing normal lung function, which results in respiratory failure, and eventually, death. While few therapies are available to reduce disease progression, patients continue to advance toward respiratory failure, leaving lung transplantation the only viable option for survival. As incidence and mortality rates steadily increase, the need for novel therapeutics is imperative. The receptor for advanced glycation endproducts (RAGE) is most highly expressed in the lungs and plays a significant role in a number of chronic lung diseases. RAGE has long been linked to IPF; however, confounding data from both human and experimental studies have left an incomplete and perplexing story. This review examines the present understanding of the role of RAGE in human and experimental models of IPF, drawing parallels to recent advances in RAGE biology. Moreover, this review discusses the role of RAGE in lung injury response, type 2 immunity, and cellular senescence, and how such mechanisms may relate to RAGE as both a biomarker of disease progression and potential therapeutic target in IPF.The reviews of this paper are available via the supplemental material section.
Collapse
Affiliation(s)
- Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, 3550 Terrace Street, S-784 Scaife Hall, Pittsburgh, PA 15261, USA
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
21
|
Transforming growth factor-β2-mediated mesenchymal transition in lens epithelial cells is repressed in the absence of RAGE. Biochem J 2021; 478:2285-2296. [PMID: 34143864 DOI: 10.1042/bcj20210069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 12/27/2022]
Abstract
Transforming growth factor-β2 (TGFβ2)-mediated epithelial to mesenchymal transition (EMT) in lens epithelial cells (LECs) has been implicated in fibrosis associated with secondary cataracts. In this study, we investigated whether the receptor for advanced glycation end products (RAGE) plays a role in TGFβ2-mediated EMT in LECs. Unlike in the LECs from wild-type mice, TGFβ2 failed to elicit an EMT response in LECs from RAGE knockout mice. The lack of RAGE also diminished TGFβ2-mediated Smad signaling. In addition, treatment with TGFβ2 increased IL-6 levels in LECs from wild-type mice but not in those from RAGE knockout mice. Treatment of human LECs with the RAGE inhibitor FPS-ZM1 reduced TGFβ2-mediated Smad signaling and the EMT response. Unlike that in wild-type lenses, the removal of fiber cell tissue in RAGE knockout lenses did not result in elevated levels of α-smooth muscle actin (α-SMA), fibronectin (FN), and integrin β1 in capsule-adherent LECs. Taken together, these results suggest that TGFβ2 signaling is intricately linked to RAGE. Targeting RAGE could be explored as a therapeutic strategy against secondary cataracts.
Collapse
|
22
|
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: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 02/12/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
A novel infectious disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was detected in December 2019 and declared as a global pandemic by the World Health. Approximately 15% of patients with COVID-19 progress to severe pneumonia and eventually develop acute respiratory distress syndrome (ARDS), septic shock and/or multiple organ failure with high morbidity and mortality. Evidence points towards a determinant pathogenic role of members of the renin-angiotensin system (RAS) in mediating the susceptibility, infection, inflammatory response and parenchymal injury in lungs and other organs of COVID-19 patients. The receptor for advanced glycation end-products (RAGE), a member of the immunoglobulin superfamily, has important roles in pulmonary pathological states, including fibrosis, pneumonia and ARDS. RAGE overexpression/hyperactivation is essential to the deleterious effects of RAS in several pathological processes, including hypertension, chronic kidney and cardiovascular diseases, and diabetes, all of which are major comorbidities of SARS-CoV-2 infection. We propose RAGE as an additional molecular target in COVID-19 patients for ameliorating the multi-organ pathology induced by the virus and improving survival, also in the perspective of future infections by other coronaviruses.
Collapse
Affiliation(s)
- Sara Chiappalupi
- Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy
| | - Laura Salvadori
- Interuniversity Institute of Myology (IIM), Perugia 06132, Italy; Department of Translational Medicine, University of Piemonte Orientale, Novara 28100, Italy
| | - Aleksandra Vukasinovic
- Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy
| | - Rosario Donato
- Interuniversity Institute of Myology (IIM), Perugia 06132, Italy
| | - Guglielmo Sorci
- Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy; Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, Perugia 06132, Italy
| | - Francesca Riuzzi
- Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy.
| |
Collapse
|
23
|
Perkins TN, Donnell ML, Oury TD. The axis of the receptor for advanced glycation endproducts in asthma and allergic airway disease. Allergy 2021; 76:1350-1366. [PMID: 32976640 DOI: 10.1111/all.14600] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/31/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022]
Abstract
Asthma is a generalized term that describes a scope of distinct pathologic phenotypes of variable severity, which share a common complication of reversible airflow obstruction. Asthma is estimated to affect almost 400 million people worldwide, and nearly ten percent of asthmatics have what is considered "severe" disease. The majority of moderate to severe asthmatics present with a "type 2-high" (T2-hi) phenotypic signature, which pathologically is driven by the type 2 cytokines Interleukin-(IL)-4, IL-5, and IL-13. However, "type 2-low" (T2-lo) phenotypic signatures are often associated with more severe, steroid-refractory neutrophilic asthma. A wide range of clinical and experimental studies have found that the receptor for advanced glycation endproducts (RAGE) plays a significant role in the pathogenesis of asthma and allergic airway disease (AAD). Current experimental data indicates that RAGE is a critical mediator of the type 2 inflammatory reactions which drive the development of T2-hi AAD. However, clinical studies demonstrate that increased RAGE ligands and signaling strongly correlate with asthma severity, especially in severe neutrophilic asthma. This review presents an overview of the current understandings of RAGE in asthma pathogenesis, its role as a biomarker of disease, and future implications for mechanistic studies, and potential therapeutic intervention strategies.
Collapse
Affiliation(s)
- Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mason L Donnell
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
24
|
Spadaro S, Fogagnolo A, Campo G, Zucchetti O, Verri M, Ottaviani I, Tunstall T, Grasso S, Scaramuzzo V, Murgolo F, Marangoni E, Vieceli Dalla Sega F, Fortini F, Pavasini R, Rizzo P, Ferrari R, Papi A, Volta CA, Contoli M. Markers of endothelial and epithelial pulmonary injury in mechanically ventilated COVID-19 ICU patients. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2021; 25:74. [PMID: 33608030 PMCID: PMC7894238 DOI: 10.1186/s13054-021-03499-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 02/10/2021] [Indexed: 01/15/2023]
Abstract
Background Biomarkers can be used to detect the presence of endothelial and/or alveolar epithelial injuries in case of ARDS. Angiopoietin-2 (Ang-2), soluble intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion protein-1 (VCAM-1), P-selectin and E-selectin are biomarkers of endothelial injury, whereas the receptor for advanced glycation end-products (RAGE) reflects alveolar epithelial injury. The aims of this study were to evaluate whether the plasma concentration of the above-mentioned biomarkers was different 1) in survivors and non-survivors of COVID-19-related ARDS and 2) in COVID-19-related and classical ARDS. Methods This prospective study was performed in two COVID-19-dedicated Intensive Care Units (ICU) and one non-COVID-19 ICU at Ferrara University Hospital. A cohort of 31 mechanically ventilated patients with COVID-19 ARDS and a cohort of 11 patients with classical ARDS were enrolled. Ang-2, ICAM-1, VCAM-1, P-selectin, E-selectin and RAGE were determined with a bead-based multiplex immunoassay at three time points: inclusion in the study (T1), after 7 ± 2 days (T2) and 14 ± 2 days (T3). The primary outcome was to evaluate the plasma trend of the biomarker levels in survivors and non-survivors. The secondary outcome was to evaluate the differences in respiratory mechanics variables and gas exchanges between survivors and non-survivors. Furthermore, we compared the plasma levels of the biomarkers at T1 in patients with COVID-19-related ARDS and classical ARDS. Results In COVID-19-related ARDS, the plasma levels of Ang-2 and ICAM-1 at T1 were statistically higher in non-survivors than survivors, (p = 0.04 and p = 0.03, respectively), whereas those of P-selectin, E-selectin and RAGE did not differ. Ang-2 and ICAM-1 at T1 were predictors of mortality (AUROC 0.650 and 0.717, respectively). At T1, RAGE and P-selectin levels were higher in classical ARDS than in COVID-19-related ARDS. Ang-2, ICAM-1 and E-selectin were lower in classical ARDS than in COVID-19-related ARDS (all p < 0.001). Conclusions COVID-19 ARDS is characterized by an early pulmonary endothelial injury, as detected by Ang-2 and ICAM-1. COVID-19 ARDS and classical ARDS exhibited a different expression of biomarkers, suggesting different pathological pathways. Trial registration NCT04343053, Date of registration: April 13, 2020
Collapse
Affiliation(s)
- Savino Spadaro
- Intensive Care Unit, Department of Translational medicine and for Romagna, University of Ferrara, Azienda Ospedaliera Universitaria di Ferrara, Via Aldo Moro 8, 44124, Ferrara, Italy.
| | - Alberto Fogagnolo
- Intensive Care Unit, Department of Translational medicine and for Romagna, University of Ferrara, Azienda Ospedaliera Universitaria di Ferrara, Via Aldo Moro 8, 44124, Ferrara, Italy
| | - Gianluca Campo
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Cona, FE, Italy.,Maria Cecilia Hospital, GVM Care and Research, Cotignola, RA, Italy
| | - Ottavio Zucchetti
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Cona, FE, Italy.,Maria Cecilia Hospital, GVM Care and Research, Cotignola, RA, Italy
| | - Marco Verri
- Intensive Care Unit, Department of Translational medicine and for Romagna, University of Ferrara, Azienda Ospedaliera Universitaria di Ferrara, Via Aldo Moro 8, 44124, Ferrara, Italy
| | - Irene Ottaviani
- Intensive Care Unit, Department of Translational medicine and for Romagna, University of Ferrara, Azienda Ospedaliera Universitaria di Ferrara, Via Aldo Moro 8, 44124, Ferrara, Italy
| | - Tanushree Tunstall
- Department of Infection Biology, School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Salvatore Grasso
- Dipartimento dell'Emergenza e Trapianti d'Organo (DETO), Sezione di Anestesiologia e Rianimazione, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Valentina Scaramuzzo
- Intensive Care Unit, Department of Translational medicine and for Romagna, University of Ferrara, Azienda Ospedaliera Universitaria di Ferrara, Via Aldo Moro 8, 44124, Ferrara, Italy
| | - Francesco Murgolo
- Dipartimento dell'Emergenza e Trapianti d'Organo (DETO), Sezione di Anestesiologia e Rianimazione, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Elisabetta Marangoni
- Intensive Care Unit, Department of Translational medicine and for Romagna, University of Ferrara, Azienda Ospedaliera Universitaria di Ferrara, Via Aldo Moro 8, 44124, Ferrara, Italy
| | - Francesco Vieceli Dalla Sega
- Dipartimento dell'Emergenza e Trapianti d'Organo (DETO), Sezione di Anestesiologia e Rianimazione, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Francesca Fortini
- Dipartimento dell'Emergenza e Trapianti d'Organo (DETO), Sezione di Anestesiologia e Rianimazione, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Rita Pavasini
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Cona, FE, Italy
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, RA, Italy.,Department of Morphology, Surgery, and Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Roberto Ferrari
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Cona, FE, Italy.,Maria Cecilia Hospital, GVM Care and Research, Cotignola, RA, Italy
| | - Alberto Papi
- Respiratory Section, Department of Morphology, Surgery, and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Carlo Alberto Volta
- Intensive Care Unit, Department of Translational medicine and for Romagna, University of Ferrara, Azienda Ospedaliera Universitaria di Ferrara, Via Aldo Moro 8, 44124, Ferrara, Italy
| | - Marco Contoli
- Respiratory Section, Department of Morphology, Surgery, and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
25
|
Kopf S, Kumar V, Kender Z, Han Z, Fleming T, Herzig S, Nawroth PP. Diabetic Pneumopathy-A New Diabetes-Associated Complication: Mechanisms, Consequences and Treatment Considerations. Front Endocrinol (Lausanne) 2021; 12:765201. [PMID: 34899603 PMCID: PMC8655305 DOI: 10.3389/fendo.2021.765201] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/22/2021] [Indexed: 01/04/2023] Open
Abstract
Patients with diabetes are over-represented among the total cases reported with "idiopathic" pulmonary fibrosis (IPF). This raises the question, whether this is an association only or whether diabetes itself can cause pulmonary fibrosis. Recent studies in mouse models of type 1 and type 2 diabetes demonstrated that diabetes causes pulmonary fibrosis. Both types of diabetes trigger a cascade, starting with increased DNA damage, an impaired DNA repair, and leading to persistent DNA damage signaling. This response, in turn, induces senescence, a senescence-associated-secretory phenotype (SASP), marked by the release of pro-inflammatory cytokines and growth factors, finally resulting in fibrosis. Restoring DNA repair drives fibrosis into remission, thus proving causality. These data can be translated clinically to patients with type 2 diabetes, characterized by long-term diabetes and albuminuria. Hence there are several arguments, to substitute the term "idiopathic" pulmonary fibrosis (IPF) in patients with diabetes (and exclusion of other causes of lung diseases) by the term "diabetes-induced pulmonary fibrosis" (DiPF). However, future studies are required to establish this term and to study whether patients with diabetes respond to the established therapies similar to non-diabetic patients.
Collapse
Affiliation(s)
- Stefan Kopf
- Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Varun Kumar
- Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
- European Molecular Biology Laboratory, Advanced Light Microscopy Facility, Heidelberg, Germany
| | - Zoltan Kender
- Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Zhe Han
- Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Stephan Herzig
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Munich-Neuherberg, Germany
- Joint Heidelberg-Institute for Diabetes and Cancer (IDC) Translational Diabetes Programme, Helmholtz-Zentrum, Munich, Germany
| | - Peter P. Nawroth
- Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
- Joint Heidelberg-Institute for Diabetes and Cancer (IDC) Translational Diabetes Programme, Helmholtz-Zentrum, Munich, Germany
- *Correspondence: Peter P. Nawroth,
| |
Collapse
|
26
|
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
|
27
|
Vlachogiannis NI, Pappa M, Ntouros PA, Nezos A, Mavragani CP, Souliotis VL, Sfikakis PP. Association Between DNA Damage Response, Fibrosis and Type I Interferon Signature in Systemic Sclerosis. Front Immunol 2020; 11:582401. [PMID: 33123169 PMCID: PMC7566292 DOI: 10.3389/fimmu.2020.582401] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 09/14/2020] [Indexed: 01/22/2023] Open
Abstract
Increased endogenous DNA damage and type I interferon pathway activation have been implicated in systemic sclerosis (SSc) pathogenesis. Because experimental evidence suggests an interplay between DNA damage response/repair (DDR/R) and immune response, we hypothesized that deregulated DDR/R is associated with a type I interferon signature and/or fibrosis extent in SSc. DNA damage levels, oxidative stress, induction of abasic sites and the efficiency of DNA double-strand break repair (DSB/R) and nucleotide excision repair (NER) were assessed in peripheral blood mononuclear cells (PBMCs) derived from 37 SSc patients and 55 healthy controls; expression of DDR/R-associated genes and type I interferon-induced genes was also quantified. Endogenous DNA damage was significantly higher in untreated diffuse or limited SSc (Olive tail moment; 14.7 ± 7.0 and 9.5 ± 4.1, respectively) as well as in patients under cytotoxic treatment (15.0 ± 5.4) but not in very early onset SSc (5.6 ± 1.2) compared with controls (4.9 ± 2.6). Moreover, patients with pulmonary fibrosis had significantly higher DNA damage levels than those without (12.6 ± 5.8 vs. 8.8 ± 4.8, respectively). SSc patients displayed increased oxidative stress and abasic sites, defective DSB/R but not NER capacity, downregulation of genes involved in DSB/R (MRE11A, PRKDC) and base excision repair (PARP1, XRCC1), and upregulation of apoptosis-related genes (BAX, BBC3). Individual levels of DNA damage in SSc PBMCs correlated significantly with the corresponding mRNA expression of type I interferon-induced genes (IFIT1, IFI44 and MX1, r=0.419-0.490) as well as with corresponding skin involvement extent by modified Rodnan skin score (r=0.481). In conclusion, defective DDR/R may exert a fuel-on-fire effect on type I interferon pathway activation and contribute to tissue fibrosis in SSc.
Collapse
Affiliation(s)
- Nikolaos I Vlachogiannis
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece.,Joint Academic Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Maria Pappa
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece.,Joint Academic Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Panagiotis A Ntouros
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece.,Joint Academic Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Adrianos Nezos
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Clio P Mavragani
- Joint Academic Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece.,Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vassilis L Souliotis
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece.,Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Petros P Sfikakis
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece.,Joint Academic Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| |
Collapse
|
28
|
Xu Z, Mo L, Feng X, Huang M, Li L. Using bioinformatics approach identifies key genes and pathways in idiopathic pulmonary fibrosis. Medicine (Baltimore) 2020; 99:e22099. [PMID: 32899090 PMCID: PMC7478566 DOI: 10.1097/md.0000000000022099] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a chronic and irreversible respiratory disease with a high incidence worldwide and no specific treatment. Currently, the etiology and pathogenesis of this disease remain largely unknown. In main purpose of this study, bioinformatics analysis was used to uncover key genes and pathways related to idiopathic pulmonary fibrosis (IPF). Gene expression profiles of GSE2052 and GSE35145 were obtained. After combining the 2 chip groups; then, we normalized the data, eliminating batch difference. R software was used to process and to screen differentially expressed genes (DEGs) between the IPF and normal tissues. Then, functional enrichment analysis of these DEGs was carried out, and a protein-protein interaction network (PPI) was also constructed. A total of 276 DEGs (152 up and 134 down-regulated genes) were identified in the IPF lung samples. The PPI network was established with 227 nodes and 763 edges. The top 10 hub genes were CAM1, CDH1, CXCL12, JUN, CTGF, SERPINE1, CXCL1, EDN1, COL1A2, and SPARC. Analyzing the PPI network modules with close interaction, the 3 key modules in the whole PPI network were screened out. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways enriched for the module containing DEGs contained the viral protein interaction with cytokine and the cytokine receptor, the TNF signaling pathway, and the chemokine signaling pathway. The identified key genes and pathways may play an important role in the occurrence and development of IPF, and may be expected to be biomarkers or therapeutic targets for the diagnosis of IPF.
Collapse
Affiliation(s)
- Zhongbo Xu
- Emergency Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine
| | - Lisha Mo
- Graduate College, Jiangxi University of Traditional Chinese Medicine
| | - Xin Feng
- Health Education Center, Maternal and Child Health Hospital of Jiangxi Province, Nanchang, Jiangxi, China
| | - Mingru Huang
- Graduate College, Jiangxi University of Traditional Chinese Medicine
| | - Lin Li
- Emergency Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine
| |
Collapse
|
29
|
Kinjo T, Kitaguchi Y, Droma Y, Yasuo M, Wada Y, Ueno F, Ota M, Hanaoka M. The Gly82Ser mutation in AGER contributes to pathogenesis of pulmonary fibrosis in combined pulmonary fibrosis and emphysema (CPFE) in Japanese patients. Sci Rep 2020; 10:12811. [PMID: 32732977 PMCID: PMC7393115 DOI: 10.1038/s41598-020-69184-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 07/08/2020] [Indexed: 11/09/2022] Open
Abstract
The dominant pathogenesis underlying the combined pulmonary fibrosis and emphysema (CPFE) remains unresolved. The receptor for advanced glycation end-products (RAGE) is highly expressed in lung tissues and interacts with distinct multiple ligands, implicating it in certain lung diseases. To elucidate the pathogenesis of CPFE, we genotyped three single nucleotide polymorphisms (SNPs: rs2070600, rs1800625, and rs2853807) of the gene encoding RAGE (AGER) in 111 CPFE patients and 337 chronic obstructive pulmonary disease (COPD) patients of Japanese by using StepOne Real-Time PCR System for SNP genotyping assay. Serum levels of soluble RAGE (sRAGE) were measured by ELISA. We found that the allele frequency of rs2070600 was significantly different between the two groups [corrected P (Pc) = 0.015]. In addition, the minor allele was associated with CPFE patients relative to COPD patients in a dominant effect model (Odds Ratio = 1.93; Pc = 0.018). Moreover, the serum sRAGE level was significantly lower in the CPFE group than the COPD group (P = 0.014). The rs2070600 minor allele was significantly associated with reduced sRAGE level in CPFE patients and independently affected sRAGE level reduction in this group (P = 0.020). We concluded that the AGER rs2070600 minor allele (Gly82Ser mutation) is associated with the pathogenesis of pulmonary fibrosis in CPFE in Japanese patients.
Collapse
Affiliation(s)
- Takumi Kinjo
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoshiaki Kitaguchi
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Yunden Droma
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Masanori Yasuo
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yosuke Wada
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Fumika Ueno
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Masao Ota
- Division of Hepatology and Gastroenterology, Department of Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Masayuki Hanaoka
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
30
|
Xu J, Xu X, Jiang L, Dua K, Hansbro PM, Liu G. SARS-CoV-2 induces transcriptional signatures in human lung epithelial cells that promote lung fibrosis. Respir Res 2020; 21:182. [PMID: 32664949 PMCID: PMC7359430 DOI: 10.1186/s12931-020-01445-6] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome (SARS)-CoV-2-induced coronavirus disease-2019 (COVID-19) is a pandemic disease that affects > 2.8 million people worldwide, with numbers increasing dramatically daily. However, there is no specific treatment for COVID-19 and much remains unknown about this disease. Angiotensin-converting enzyme (ACE)2 is a cellular receptor of SARS-CoV-2. It is cleaved by type II transmembrane serine protease (TMPRSS)2 and disintegrin and metallopeptidase domain (ADAM)17 to assist viral entry into host cells. Clinically, SARS-CoV-2 infection may result in acute lung injury and lung fibrosis, but the underlying mechanisms of COVID-19 induced lung fibrosis are not fully understood. METHODS The networks of ACE2 and its interacting molecules were identified using bioinformatic methods. Their gene and protein expressions were measured in human epithelial cells after 24 h SARS-CoV-2 infection, or in existing datasets of lung fibrosis patients. RESULTS We confirmed the binding of SARS-CoV-2 and ACE2 by bioinformatic analysis. TMPRSS2, ADAM17, tissue inhibitor of metalloproteinase (TIMP)3, angiotensinogen (AGT), transformation growth factor beta (TGFB1), connective tissue growth factor (CTGF), vascular endothelial growth factor (VEGF) A and fibronectin (FN) were interacted with ACE2, and the mRNA and protein of these molecules were expressed in lung epithelial cells. SARS-CoV-2 infection increased ACE2, TGFB1, CTGF and FN1 mRNA that were drivers of lung fibrosis. These changes were also found in lung tissues from lung fibrosis patients. CONCLUSIONS Therefore, SARS-CoV-2 binds with ACE2 and activates fibrosis-related genes and processes to induce lung fibrosis.
Collapse
Affiliation(s)
- Jincheng Xu
- School of Stomatology, Bengbu Medical College, Bengbu, 2033, Anhui, China
| | - Xiaoyue Xu
- School of Public Health and Community Medicine, Faculty of Medicine, University of New South Wales, Kensington, 233000, NSW, Australia
- Faculty of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Lina Jiang
- School of Stomatology, Bengbu Medical College, Bengbu, 2033, Anhui, China
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Centre for Inflammation, Centenary Institute, Camperdown, NSW, 2050, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Camperdown, NSW, 2050, Australia
- School of Life Science, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Gang Liu
- Centre for Inflammation, Centenary Institute, Camperdown, NSW, 2050, Australia.
- School of Life Science, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
31
|
Yamaguchi K, Iwamoto H, Mazur W, Miura S, Sakamoto S, Horimasu Y, Masuda T, Miyamoto S, Nakashima T, Ohshimo S, Fujitaka K, Hamada H, Hattori N. Reduced endogenous secretory RAGE in blood and bronchoalveolar lavage fluid is associated with poor prognosis in idiopathic pulmonary fibrosis. Respir Res 2020; 21:145. [PMID: 32527263 PMCID: PMC7291663 DOI: 10.1186/s12931-020-01410-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 05/31/2020] [Indexed: 12/16/2022] Open
Abstract
Background The endogenous secretory receptor for advanced glycation end products (esRAGE) is a soluble isoform produced by alternative splicing of the RAGE gene. The isoform has anti-inflammatory properties due to its inhibition of the RAGE/ligand interaction and is reduced in the lung tissue of patients with idiopathic pulmonary fibrosis (IPF). This study aimed to investigate the association of esRAGE serum and bronchoalveolar lavage fluid (BALF) levels with progression of IPF. Methods This study included 79 IPF patients and 90 healthy controls. IPF and control serum esRAGE levels were compared, and the correlation between serum and BALF esRAGE levels was analyzed in 57 IPF patient samples. We also investigated the relationship of esRAGE serum and BALF levels with prognoses and lung function parameters in patients with IPF. Results Serum esRAGE levels in IPF patients were significantly lower than those in healthy controls (162.0 ± 102.4 ng/ml and 200.7 ± 107.3 ng/ml, p = 0.009), although the baseline characteristics of age and smoking history were not matched. Serum levels of esRAGE were correlated with BALF esRAGE levels (rs = 0.317). The BALF esRAGE levels were also correlated with diffusion capacity for carbon monoxide (rs = 0.406). A Kaplan-Meier curve analysis and univariate/multivariate Cox hazard proportion analysis revealed that lower levels of esRAGE in blood and BALF were significantly associated with poorer prognoses in patients with IPF. Conclusions Decreased esRAGE levels in BALF and blood were associated with poor prognoses in patients with IPF. These results suggest that esRAGE could be related to the pathophysiology of IPF and serum esRAGE could be a potential prognostic marker of IPF.
Collapse
Affiliation(s)
- Kakuhiro Yamaguchi
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hiroshi Iwamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Witold Mazur
- Heart and Lung Centre, Division of Pulmonary Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Shinichiro Miura
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shinjiro Sakamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yasushi Horimasu
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Takeshi Masuda
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shintaro Miyamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Taku Nakashima
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shinichiro Ohshimo
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazunori Fujitaka
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hironobu Hamada
- Department of Physical Analysis and Therapeutic Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
32
|
Wang D, Ma Y, Tong X, Zhang Y, Fan H. Diabetes Mellitus Contributes to Idiopathic Pulmonary Fibrosis: A Review From Clinical Appearance to Possible Pathogenesis. Front Public Health 2020; 8:196. [PMID: 32582606 PMCID: PMC7285959 DOI: 10.3389/fpubh.2020.00196] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 04/29/2020] [Indexed: 02/05/2023] Open
Abstract
Diabetes mellitus is a systematic metabolic disease characterized by persistent hyperglycemia, which complications often involve multiple organs and systems including vessels, kidneys, retinas, and nervous system. Idiopathic pulmonary fibrosis is a chronic, progressive, fibrotic disease with usual interstitial pneumonia patterns. With in-depth research, diabetic related lung injury has been confirmed, and the lung is also considered as one of the targeted organs of diabetes, which mainly manifests as the pulmonary fibrosis. Based on that, this review discusses the association between diabetes mellitus and idiopathic pulmonary fibrosis from clinical findings to possible mechanisms.
Collapse
Affiliation(s)
- Dongguang Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Yao Ma
- The Center of Gerontology and Geriatrics, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Xiang Tong
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Yonggang Zhang
- Department of Periodical Press, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Fan
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Halu A, Liu S, Baek SH, Hobbs BD, Hunninghake GM, Cho MH, Silverman EK, Sharma A. Exploring the cross-phenotype network region of disease modules reveals concordant and discordant pathways between chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. Hum Mol Genet 2020; 28:2352-2364. [PMID: 30997486 DOI: 10.1093/hmg/ddz069] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 03/12/2019] [Accepted: 03/23/2019] [Indexed: 12/16/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) are two pathologically distinct chronic lung diseases that are associated with cigarette smoking. Genetic studies have identified shared loci for COPD and IPF, including several loci with opposite directions of effect. The existence of additional shared genetic loci, as well as potential shared pathobiological mechanisms between the two diseases at the molecular level, remains to be explored. Taking a network-based approach, we built disease modules for COPD and IPF using genome-wide association studies-implicated genes. The two disease modules displayed strong disease signals in an independent gene expression data set of COPD and IPF lung tissue and showed statistically significant overlap and network proximity, sharing 19 genes, including ARHGAP12 and BCHE. To uncover pathways at the intersection of COPD and IPF, we developed a metric, NetPathScore, which prioritizes the pathways of a disease by their network overlap with another disease. Applying NetPathScore to the COPD and IPF disease modules enabled the determination of concordant and discordant pathways between these diseases. Concordant pathways between COPD and IPF included extracellular matrix remodeling, Mitogen-activated protein kinase (MAPK) signaling and ALK pathways, whereas discordant pathways included advanced glycosylation end product receptor signaling and telomere maintenance and extension pathways. Overall, our findings reveal shared molecular interaction regions between COPD and IPF and shed light on the congruent and incongruent biological processes lying at the intersection of these two complex diseases.
Collapse
Affiliation(s)
- Arda Halu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shikang Liu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Seung Han Baek
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian D Hobbs
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gary M Hunninghake
- Division of Pulmonary and Critical Care, 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.,Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amitabh Sharma
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
34
|
McDonough JE, Ahangari F, Li Q, Jain S, Verleden SE, Herazo-Maya J, Vukmirovic M, DeIuliis G, Tzouvelekis A, Tanabe N, Chu F, Yan X, Verschakelen J, Homer RJ, Manatakis DV, Zhang J, Ding J, Maes K, De Sadeleer L, Vos R, Neyrinck A, Benos PV, Bar-Joseph Z, Tantin D, Hogg JC, Vanaudenaerde BM, Wuyts WA, Kaminski N. Transcriptional regulatory model of fibrosis progression in the human lung. JCI Insight 2019; 4:131597. [PMID: 31600171 PMCID: PMC6948862 DOI: 10.1172/jci.insight.131597] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/04/2019] [Indexed: 11/17/2022] Open
Abstract
To develop a systems biology model of fibrosis progression within the human lung we performed RNA sequencing and microRNA analysis on 95 samples obtained from 10 idiopathic pulmonary fibrosis (IPF) and 6 control lungs. Extent of fibrosis in each sample was assessed by microCT-measured alveolar surface density (ASD) and confirmed by histology. Regulatory gene expression networks were identified using linear mixed-effect models and dynamic regulatory events miner (DREM). Differential gene expression analysis identified a core set of genes increased or decreased before fibrosis was histologically evident that continued to change with advanced fibrosis. DREM generated a systems biology model (www.sb.cs.cmu.edu/IPFReg) that identified progressively divergent gene expression tracks with microRNAs and transcription factors that specifically regulate mild or advanced fibrosis. We confirmed model predictions by demonstrating that expression of POU2AF1, previously unassociated with lung fibrosis but proposed by the model as regulator, is increased in B lymphocytes in IPF lungs and that POU2AF1-knockout mice were protected from bleomycin-induced lung fibrosis. Our results reveal distinct regulation of gene expression changes in IPF tissue that remained structurally normal compared with moderate or advanced fibrosis and suggest distinct regulatory mechanisms for each stage.
Collapse
Affiliation(s)
- John E. McDonough
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Farida Ahangari
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Qin Li
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Siddhartha Jain
- Carnegie Mellon University of Computer Science, Pittsburgh, Pennsylvania, USA
| | - Stijn E. Verleden
- Department of Chronic Diseases, Metabolism, and Ageing, KU Leuven, Leuven Belgium
| | - Jose Herazo-Maya
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Milica Vukmirovic
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Giuseppe DeIuliis
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Argyrios Tzouvelekis
- Division of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Athens, Greece
| | - Naoya Tanabe
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
| | - Fanny Chu
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
| | - Xiting Yan
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Johny Verschakelen
- Department of Chronic Diseases, Metabolism, and Ageing, KU Leuven, Leuven Belgium
| | - Robert J. Homer
- Department of Pathology, Yale University School of Medicine, New Haven,Connecticut, USA
- Pathology and Laboratory Medicine Service, VA CT HealthCare System, West Haven, Connecticut, USA
| | - Dimitris V. Manatakis
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Junke Zhang
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jun Ding
- Carnegie Mellon University of Computer Science, Pittsburgh, Pennsylvania, USA
| | - Karen Maes
- Department of Chronic Diseases, Metabolism, and Ageing, KU Leuven, Leuven Belgium
| | - Laurens De Sadeleer
- Department of Chronic Diseases, Metabolism, and Ageing, KU Leuven, Leuven Belgium
| | - Robin Vos
- Department of Chronic Diseases, Metabolism, and Ageing, KU Leuven, Leuven Belgium
| | - Arne Neyrinck
- Department of Chronic Diseases, Metabolism, and Ageing, KU Leuven, Leuven Belgium
| | - Panayiotis V. Benos
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ziv Bar-Joseph
- Carnegie Mellon University of Computer Science, Pittsburgh, Pennsylvania, USA
| | - Dean Tantin
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - James C. Hogg
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
| | | | - Wim A. Wuyts
- Department of Chronic Diseases, Metabolism, and Ageing, KU Leuven, Leuven Belgium
| | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
35
|
Living on the Edge: Efferocytosis at the Interface of Homeostasis and Pathology. Immunity 2019; 50:1149-1162. [PMID: 31117011 DOI: 10.1016/j.immuni.2019.04.018] [Citation(s) in RCA: 248] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/13/2019] [Accepted: 04/29/2019] [Indexed: 01/23/2023]
Abstract
Nearly every tissue in the body undergoes routine turnover of cells as part of normal healthy living. The majority of these cells undergoing turnover die via apoptosis, and then are rapidly removed by phagocytes by the process of efferocytosis that is anti-inflammatory. However, a number of pathologies have recently been linked to defective clearance of apoptotic cells. Perturbed clearance arises for many reasons, including overwhelming of the clearance machinery, disruptions at different stages of efferocytosis, and responses of phagocytes during efferocytosis, all of which can alter the homeostatic tissue environment. This review covers linkages of molecules involved in the different phases of efferocytosis to disease pathologies that can arise due to their loss or altered function.
Collapse
|
36
|
Eichhorst A, Daniel C, Rzepka R, Sehnert B, Nimmerjahn F, Voll RE, Chevalier N. Relevance of Receptor for Advanced Glycation end Products (RAGE) in Murine Antibody-Mediated Autoimmune Diseases. Int J Mol Sci 2019; 20:ijms20133234. [PMID: 31266174 PMCID: PMC6651235 DOI: 10.3390/ijms20133234] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 12/24/2022] Open
Abstract
It is incompletely understood how self-antigens become targets of humoral immunity in antibody-mediated autoimmune diseases. In this context, alarmins are discussed as an important level of regulation. Alarmins are recognized by various receptors, such as receptor for advanced glycation end products (RAGE). As RAGE is upregulated under inflammatory conditions, strongly binds nucleic acids and mediates pro-inflammatory responses upon alarmin recognition, our aim was to examine its contribution to immune complex-mediated autoimmune diseases. This question was addressed employing RAGE−/− animals in murine models of pristane-induced lupus, collagen-induced, and serum-transfer arthritis. Autoantibodies were assessed by enzyme-linked immunosorbent assay, renal disease by quantification of proteinuria and histology, arthritis by scoring joint inflammation. The associated immune status was determined by flow cytometry. In both disease entities, we detected tendentiously decreased autoantibody levels in RAGE−/− mice, however no differences in clinical outcome. In accordance with autoantibody levels, a subgroup of the RAGE−/− animals showed a decrease in plasma cells, and germinal center B cells and an increase in follicular B cells. Based on our results, we suggest that RAGE deficiency alone does not significantly affect antibody-mediated autoimmunity. RAGE may rather exert its effects along with other receptors linking environmental factors to auto-reactive immune responses.
Collapse
Affiliation(s)
- Alexandra Eichhorst
- Department of Rheumatology and Clinical Immunology, Medical Centre-University of Freiburg, Faculty of Medicine, 79106 Freiburg, Baden-Wuerttemberg, Germany
| | - Christoph Daniel
- Department of Nephropathology, Friedrich-Alexander University (FAU) of Erlangen-Nuremberg, 91054 Erlangen, Bavaria, Germany
| | - Rita Rzepka
- Department of Rheumatology and Clinical Immunology, Medical Centre-University of Freiburg, Faculty of Medicine, 79106 Freiburg, Baden-Wuerttemberg, Germany
| | - Bettina Sehnert
- Department of Rheumatology and Clinical Immunology, Medical Centre-University of Freiburg, Faculty of Medicine, 79106 Freiburg, Baden-Wuerttemberg, Germany
| | - Falk Nimmerjahn
- Department of Biology, Friedrich-Alexander University (FAU) of Erlangen-Nuremberg, 91058 Erlangen, Bavaria, Germany
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, Medical Centre-University of Freiburg, Faculty of Medicine, 79106 Freiburg, Baden-Wuerttemberg, Germany
| | - Nina Chevalier
- Department of Rheumatology and Clinical Immunology, Medical Centre-University of Freiburg, Faculty of Medicine, 79106 Freiburg, Baden-Wuerttemberg, Germany.
| |
Collapse
|
37
|
Correll KA, Edeen KE, Zemans RL, Redente EF, Serban KA, Curran-Everett D, Edelman BL, Mikels-Vigdal A, Mason RJ. Transitional human alveolar type II epithelial cells suppress extracellular matrix and growth factor gene expression in lung fibroblasts. Am J Physiol Lung Cell Mol Physiol 2019; 317:L283-L294. [PMID: 31166130 DOI: 10.1152/ajplung.00337.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Epithelial-fibroblast interactions are thought to be very important in the adult lung in response to injury, but the specifics of these interactions are not well defined. We developed coculture systems to define the interactions of adult human alveolar epithelial cells with lung fibroblasts. Alveolar type II cells cultured on floating collagen gels reduced the expression of type 1 collagen (COL1A1) and α-smooth muscle actin (ACTA2) in fibroblasts. They also reduced fibroblast expression of hepatocyte growth factor (HGF), fibroblast growth factor 7 (FGF7, KGF), and FGF10. When type II cells were cultured at an air-liquid interface to maintain high levels of surfactant protein expression, this inhibitory activity was lost. When type II cells were cultured on collagen-coated tissue culture wells to reduce surfactant protein expression further and increase the expression of some type I cell markers, the epithelial cells suppressed transforming growth factor-β (TGF-β)-stimulated ACTA2 and connective tissue growth factor (CTGF) expression in lung fibroblasts. Our results suggest that transitional alveolar type II cells and likely type I cells but not fully differentiated type II cells inhibit matrix and growth factor expression in fibroblasts. These cells express markers of both type II cells and type I cells. This is probably a normal homeostatic mechanism to inhibit the fibrotic response in the resolution phase of wound healing. Defining how transitional type II cells convert activated fibroblasts into a quiescent state and inhibit the effects of TGF-β may provide another approach to limiting the development of fibrosis after alveolar injury.
Collapse
Affiliation(s)
| | | | - Rachel L Zemans
- National Jewish Health, Denver, Colorado.,Division of Pulmonary and Critical Care Medicine/Department of Medicine, University of Michigan, Ann Arbor, Michigan
| | | | | | | | | | | | | |
Collapse
|
38
|
P2X7 Receptor Indirectly Regulates the JAM-A Protein Content via Modulation of GSK-3β. Int J Mol Sci 2019; 20:ijms20092298. [PMID: 31075901 PMCID: PMC6539570 DOI: 10.3390/ijms20092298] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/30/2019] [Accepted: 05/08/2019] [Indexed: 12/20/2022] Open
Abstract
The alveolar epithelial cells represent an important part of the alveolar barrier, which is maintained by tight junction proteins, particularly JAM-A, occludin, and claudin-18, which regulate paracellular permeability. In this study, we report on a strong increase in epithelial JAM-A expression in P2X7 receptor knockout mice when compared to the wildtype. Precision-cut lung slices of wildtype and knockout lungs and immortal epithelial lung E10 cells were treated with bleomycin, the P2X7 receptor inhibitor oxATP, and the agonist BzATP, respectively, to evaluate early changes in JAM-A expression. Biochemical and immunohistochemical data showed evidence for P2X7 receptor-dependent JAM-A expression in vitro. Inhibition of the P2X7 receptor using oxATP increased JAM-A, whereas activation of the receptor decreased the JAM-A protein level. In order to examine the role of GSK-3β in the expression of JAM-A in alveolar epithelial cells, we used lithium chloride for GSK-3β inhibiting experiments, which showed a modulating effect on bleomycin-induced alterations in JAM-A levels. Our data suggest that an increased constitutive JAM-A protein level in P2X7 receptor knockout mice may have a protective effect against bleomycin-induced lung injury. Bleomycin-treated precision-cut lung slices from P2X7 receptor knockout mice responded with a lower increase in mRNA expression of JAM-A than bleomycin-treated precision-cut lung slices from wildtype mice.
Collapse
|
39
|
Personnaz J, Piccolo E, Branchereau M, Filliol A, Paccoud R, Moreau E, Calise D, Riant E, Gourdy P, Heymes C, Schwabe RF, Dray C, Valet P, Pradère J. Macrophage-derived HMGB1 is dispensable for tissue fibrogenesis. FASEB Bioadv 2019; 1:227-245. [PMID: 32123829 PMCID: PMC6996376 DOI: 10.1096/fba.2018-00035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/11/2018] [Accepted: 12/14/2018] [Indexed: 12/19/2022] Open
Abstract
Alarmins and damage-associated molecular patterns (DAMPs) are powerful inflammatory mediators, capable of initiating and maintaining sterile inflammation during acute or chronic tissue injury. Recent evidence suggests that alarmins/DAMPs may also trigger tissue regeneration and repair, suggesting a potential contribution to tissue fibrogenesis. High mobility group B1 (HMGB1), a bona fide alarmin/DAMP, may be released passively by necrotic cells or actively secreted by innate immune cells. Macrophages can release large amounts of HMGB1 and play a key role in wound healing and regeneration processes. Here, we hypothesized that macrophages may be a key source of HMGB1 and thereby contribute to wound healing and fibrogenesis. Surprisingly, cell-specific deletion approaches, demonstrated that macrophage-derived HMGB1 is not involved in tissue fibrogenesis in multiple organs with different underlying pathologies. Compared to control HMGB1Flox mice, mice with macrophage-specific HMGB1 deletion (HMGB1ΔMac) do not display any modification of fibrogenesis in the liver after CCL4 or thioacetamide treatment and bile duct ligation; in the kidney following unilateral ureter obstruction; and in the heart after transverse aortic constriction. Of note, even under thermoneutral housing, known to exacerbate inflammation and fibrosis features, HMGB1ΔMac mice do not show impairment of fibrogenesis. In conclusion, our study clearly establishes that macrophage-derived HMGB1 does not contribute to tissue repair and fibrogenesis.
Collapse
Affiliation(s)
- Jean Personnaz
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de ToulouseToulouseFrance
| | - Enzo Piccolo
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de ToulouseToulouseFrance
| | - Maxime Branchereau
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de ToulouseToulouseFrance
| | | | - Romain Paccoud
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de ToulouseToulouseFrance
| | - Elsa Moreau
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de ToulouseToulouseFrance
| | - Denis Calise
- UMS006, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (INSERM) U1048, Institute of Cardiovascular and Metabolic DiseaseToulouseFrance
| | - Elodie Riant
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de ToulouseToulouseFrance
| | - Pierre Gourdy
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de ToulouseToulouseFrance
- Service de Diabétologie, Maladies Métaboliques et Nutrition, CHU de ToulouseToulouseFrance
| | - Christophe Heymes
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de ToulouseToulouseFrance
| | | | - Cédric Dray
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de ToulouseToulouseFrance
| | - Philippe Valet
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de ToulouseToulouseFrance
| | - Jean‐Philippe Pradère
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de ToulouseToulouseFrance
| |
Collapse
|
40
|
Perkins TN, Oczypok EA, Dutz RE, Donnell ML, Myerburg MM, Oury TD. The receptor for advanced glycation end products is a critical mediator of type 2 cytokine signaling in the lungs. J Allergy Clin Immunol 2019; 144:796-808.e12. [PMID: 30940519 DOI: 10.1016/j.jaci.2019.03.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 03/05/2019] [Accepted: 03/18/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND Asthma is estimated to effect more than 300 million persons worldwide, leading to nearly 250,000 deaths annually. The majority of patients with mild-to-severe asthma have what is deemed "type-2 high" asthma, which is driven by the prototypical type 2 cytokines IL-4, IL-5, and IL-13. Studies have indicated that the receptor for advanced glycation end products (RAGE) is a critical molecule in the pathogenesis of experimental asthma/allergic airway inflammation. More specifically, RAGE expressed on stromal cells, rather than hematopoietic cells, is critical to induction of asthma/allergic airway inflammation by driving type 2 inflammatory responses. However, the role of RAGE in directly mediating type 2 cytokine signaling has never been investigated. OBJECTIVE The goal of this study was to test the hypothesis that RAGE mediates type 2 cytokine-induced signal transduction, airway inflammation, and mucus metaplasia in the lungs. METHODS Wild-type (WT) and RAGE knockout (RAGE-/-) mice, were intranasally administered rIL-5/rIL-13 or rIL-4 alone, and signal transducer and activator of transcription 6 (STAT6) signaling, airway inflammation, and mucus metaplasia were assessed. A RAGE small-molecule antagonist was used to determine the effects of pharmacologically inhibiting RAGE on type 2 cytokine-induced effects. RESULTS Administration of type 2 cytokines induced pronounced airway inflammation and mucus metaplasia in WT mice, which was nearly completely abrogated in RAGE-/- mice. In addition, treatment with a RAGE-specific antagonist diminished the effects of type 2 cytokines in WT mice and in primary human bronchial epithelial cell cultures. Genetic ablation or pharmacologic inhibition of RAGE blocks the effects of IL-13 and IL-4 by inhibiting sustained STAT6 activation and downstream target gene expression in mice and in human bronchial epithelial cells. CONCLUSIONS This study is the first to indicate that RAGE is a critical component of type 2 cytokine signal transduction mechanisms, which is a driving force behind type 2-high asthma.
Collapse
Affiliation(s)
- Timothy N Perkins
- Department of Pathology, University of Pittsburgh, School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pa; Department of Pediatrics, Division of Pulmonary, Allergy, and Clinical Immunology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pa.
| | - Elizabeth A Oczypok
- Department of Medicine, University of Pittsburgh, School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Regina E Dutz
- Department of Pathology, University of Pittsburgh, School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Mason L Donnell
- Department of Pathology, University of Pittsburgh, School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Michael M Myerburg
- Department of Medicine, University of Pittsburgh, School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh, School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pa.
| |
Collapse
|
41
|
Khaket TP, Kang SC, Mukherjee TK. The Potential of Receptor for Advanced Glycation End Products (RAGE) as a Therapeutic Target for Lung Associated Diseases. Curr Drug Targets 2019; 20:679-689. [DOI: 10.2174/1389450120666181120102159] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/17/2018] [Accepted: 11/02/2018] [Indexed: 12/27/2022]
Abstract
The receptor for advanced glycation end products (RAGE) is a multi-ligand pattern recognition
receptor that is highly expressed in lung epithelial cells. It helps alveolar epithelial cells to
maintain their morphology and specific architecture. However, in various pathophysiological conditions,
pulmonary tissues express a supraphysiological level of RAGE and its ligands including advanced
glycation end products, high mobility group box 1 proteins, and S100 proteins. On interaction
with RAGE, these ligands stimulate downstream signaling that generates inflammation and oxidative
stress leading to asthma, chronic obstructive pulmonary disease, lung cancers, idiopathic pulmonary
fibrosis, acute lung injury, pneumonia, bronchopulmonary dysplasia, cystic fibrosis, and sepsis. Thus,
pharmacological agents that can either suppress the production of RAGE or block its biological activity
would offer promising therapeutic value against pathogenesis of the aforementioned lungassociated
diseases. This review presents a comprehensive overview of the recent progress made in
defining the functions of RAGE in lung-associated diseases.
Collapse
Affiliation(s)
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, Korea
| | - Tapan Kumar Mukherjee
- Department of Biotechnology, Maharishi Markandeshwar University, Mullana, Haryana, India
| |
Collapse
|
42
|
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
|
43
|
Perkins TN, Oczypok EA, Milutinovic PS, Dutz RE, Oury TD. RAGE-dependent VCAM-1 expression in the lung endothelium mediates IL-33-induced allergic airway inflammation. Allergy 2019; 74:89-99. [PMID: 29900561 DOI: 10.1111/all.13500] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND The receptor for advanced glycation endproducts (RAGE) has been implicated as a critical molecule in the pathogenesis of experimental asthma/allergic airway inflammation (AAI). It has been previously shown that RAGE acts both upstream of interleukin-33 (IL-33) release and downstream of IL-33 release via RAGE-dependent IL-33-induced accumulation of type 2 innate lymphoid cells (ILC2s) in the lungs, which perpetuate type 2 inflammation and mucus metaplasia. However, the mechanism by which RAGE mediates downstream IL-33-induced type 2 inflammatory responses is unknown. OBJECTIVE This study tested the hypothesis that ILC2s are recruited to the lungs via RAGE-dependent vascular cell adhesion molecule 1 (VCAM-1) expression on lung endothelial cells. METHODS House dust mite extract, Alternaria alternata extract, or rIL-33 was used to induce AAI/VCAM-1 expression in wild-type (WT) and RAGE-knockout (RAGE-KO) mice. Intravenous (i.v.) anti-VCAM-1 or intraperitoneal (i.p.) β7 blocking antibody administration was used to determine the role of VCAM-1 in IL-33-induced AAI. RESULTS Enhanced VCAM-1 expression in the lungs by HDM, AA, or rIL-33 exposure was found to be RAGE-dependent. In addition, stimulation of primary mouse lung endothelial cells with IL-33 induced VCAM-1 expression in WT, but not RAGE-KO cells. Administration of VCAM-1 and β7-integrin blocking antibodies reduced IL-33-induced eosinophilic inflammation, mucus metaplasia, and type 2 inflammatory responses. CONCLUSION This study demonstrates that allergen- and cytokine-induced VCAM-1 expression is RAGE-dependent and contributes to lung ILC2 accumulation and downstream eosinophilic inflammation, mucus metaplasia, and type 2 inflammatory responses.
Collapse
Affiliation(s)
- T. N. Perkins
- Department of Pathology University of Pittsburgh School of Medicine University of Pittsburgh Medical Center Pittsburgh PA USA
- Department of Pediatrics Division of Pulmonary, Allergy, and Clinical Immunology Children's Hospital of Pittsburgh of UPMC Pittsburgh PA USA
| | - E. A. Oczypok
- Department of Pathology University of Pittsburgh School of Medicine University of Pittsburgh Medical Center Pittsburgh PA USA
| | - P. S. Milutinovic
- Department of Pediatrics Duke University Medical Center Durham NC USA
- Department of Medicine Duke University Medical Center Durham NC USA
| | - R. E. Dutz
- Department of Pathology University of Pittsburgh School of Medicine University of Pittsburgh Medical Center Pittsburgh PA USA
| | - T. D. Oury
- Department of Pathology University of Pittsburgh School of Medicine University of Pittsburgh Medical Center Pittsburgh PA USA
| |
Collapse
|
44
|
Evolving Genomics of Pulmonary Fibrosis. Respir Med 2019. [DOI: 10.1007/978-3-319-99975-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
45
|
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
|
46
|
Sagheddu R, Chiappalupi S, Salvadori L, Riuzzi F, Donato R, Sorci G. Targeting RAGE as a potential therapeutic approach to Duchenne muscular dystrophy. Hum Mol Genet 2018; 27:3734-3746. [DOI: 10.1093/hmg/ddy288] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 07/30/2018] [Indexed: 12/21/2022] Open
Affiliation(s)
- Roberta Sagheddu
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
- Interuniversity Institute of Myology (IIM)
| | - Sara Chiappalupi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
- Interuniversity Institute of Myology (IIM)
| | - Laura Salvadori
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
- Interuniversity Institute of Myology (IIM)
| | - Francesca Riuzzi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
- Interuniversity Institute of Myology (IIM)
| | - Rosario Donato
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
- Interuniversity Institute of Myology (IIM)
- Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, Perugia, Italy
| | - Guglielmo Sorci
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
- Interuniversity Institute of Myology (IIM)
| |
Collapse
|
47
|
de Carvalho GC, Hirata FYA, Domingues R, Figueiredo CA, Zaniboni MC, Pereira NV, Sotto MN, Aoki V, da Silva Duarte AJ, Sato MN. Up-regulation of HMGB1 and TLR4 in skin lesions of lichen planus. Arch Dermatol Res 2018; 310:523-528. [PMID: 29728859 DOI: 10.1007/s00403-018-1837-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 04/17/2018] [Accepted: 04/25/2018] [Indexed: 01/27/2023]
Abstract
Lichen planus (LP) is a chronic, mucocutaneous inflammatory disease of an unknown aetiology. The disease has been associated with certain viruses, and the factors such as DAMPs (damage-associated molecular patterns) and PAMPs (pathogen-associated molecular patterns) may also contribute to the inflammatory response in LP. HMGB1 (high mobility group box 1 protein) is one of the major DAMPs that induces inflammation and could trigger LP disease. The present study was aimed to examine TLR4, RAGE and HMGB1 production in epidermis or dermis by immunohistochemistry and the respective expression of these targets in the skin lesions of patients with LP. Moreover, we measured HMGB1 serum levels by ELISA. The results showed similar profile of expression by HMGB1 and TLR4, which are decreased at epidermis and up-regulated at dermis of skin lesions of LP patients that was sustained by intense cellular infiltration. RAGE expression was also increased in dermis of LP. Although there is increased RAGE protein levels, a decreased RAGE transcript levels was detected. Similar HMGB1 serum levels were detected in the LP and control groups. This study demonstrates that HMGB1 and TLR4 could contribute to the inflammatory LP process in skin.
Collapse
Affiliation(s)
- Gabriel Costa de Carvalho
- Laboratory of Dermatology and Immunodeficiencies, Medical School of the University of São Paulo, LIM-56, Av. Dr. Enéas de Carvalho Aguiar, 470, 3rd floor, São Paulo, 05403-000, Brazil
| | - Fabiana Yasumoto Araujo Hirata
- Laboratory of Dermatology and Immunodeficiencies, Medical School of the University of São Paulo, LIM-56, Av. Dr. Enéas de Carvalho Aguiar, 470, 3rd floor, São Paulo, 05403-000, Brazil
| | - Rosana Domingues
- Laboratory of Dermatology and Immunodeficiencies, Medical School of the University of São Paulo, LIM-56, Av. Dr. Enéas de Carvalho Aguiar, 470, 3rd floor, São Paulo, 05403-000, Brazil
| | | | - Mariana Colombini Zaniboni
- Laboratory of Dermatology and Immunodeficiencies, Medical School of the University of São Paulo, LIM-56, Av. Dr. Enéas de Carvalho Aguiar, 470, 3rd floor, São Paulo, 05403-000, Brazil
| | - Naiura Vieira Pereira
- Laboratory of Dermatology and Immunodeficiencies, Medical School of the University of São Paulo, LIM-56, Av. Dr. Enéas de Carvalho Aguiar, 470, 3rd floor, São Paulo, 05403-000, Brazil
| | | | - Valéria Aoki
- Laboratory of Dermatology and Immunodeficiencies, Medical School of the University of São Paulo, LIM-56, Av. Dr. Enéas de Carvalho Aguiar, 470, 3rd floor, São Paulo, 05403-000, Brazil
| | - Alberto José da Silva Duarte
- Laboratory of Dermatology and Immunodeficiencies, Medical School of the University of São Paulo, LIM-56, Av. Dr. Enéas de Carvalho Aguiar, 470, 3rd floor, São Paulo, 05403-000, Brazil.,Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| | - Maria Notomi Sato
- Laboratory of Dermatology and Immunodeficiencies, Medical School of the University of São Paulo, LIM-56, Av. Dr. Enéas de Carvalho Aguiar, 470, 3rd floor, São Paulo, 05403-000, Brazil.
| |
Collapse
|
48
|
Hagiwara S, Sourris K, Ziemann M, Tieqiao W, Mohan M, McClelland AD, Brennan E, Forbes J, Coughlan M, Harcourt B, Penfold S, Wang B, Higgins G, Pickering R, El-Osta A, Thomas MC, Cooper ME, Kantharidis P. RAGE Deletion Confers Renoprotection by Reducing Responsiveness to Transforming Growth Factor-β and Increasing Resistance to Apoptosis. Diabetes 2018; 67:960-973. [PMID: 29449307 DOI: 10.2337/db17-0538] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 02/07/2018] [Indexed: 11/13/2022]
Abstract
Signaling via the receptor of advanced glycation end products (RAGE)-though complex and not fully elucidated in the setting of diabetes-is considered a key injurious pathway in the development of diabetic nephropathy (DN). We report here that RAGE deletion resulted in increased expression of fibrotic markers (collagen I and IV, fibronectin) and the inflammatory marker MCP-1 in primary mouse mesangial cells (MCs) and in kidney cortex. RNA sequencing analysis in MCs from RAGE-/- and wild-type mice confirmed these observations. Nevertheless, despite these gene expression changes, decreased responsiveness to transforming growth factor-β was identified in RAGE-/- mice. Furthermore, RAGE deletion conferred a more proliferative phenotype in MCs and reduced susceptibility to staurosporine-induced apoptosis. RAGE restoration experiments in RAGE-/- MCs largely reversed these gene expression changes, resulting in reduced expression of fibrotic and inflammatory markers. This study highlights that protection against DN in RAGE knockout mice is likely to be due in part to the decreased responsiveness to growth factor stimulation and an antiapoptotic phenotype in MCs. Furthermore, it extends our understanding of the role of RAGE in the progression of DN, as RAGE seems to play a key role in modulating the sensitivity of the kidney to injurious stimuli such as prosclerotic cytokines.
Collapse
Affiliation(s)
- Shinji Hagiwara
- Department of Diabetes, Monash University, Melbourne, Australia
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Karly Sourris
- Department of Diabetes, Monash University, Melbourne, Australia
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Mark Ziemann
- Department of Diabetes, Monash University, Melbourne, Australia
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Wu Tieqiao
- Department of Diabetes, Monash University, Melbourne, Australia
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Muthukumar Mohan
- Department of Diabetes, Monash University, Melbourne, Australia
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Aaron D McClelland
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Eoin Brennan
- Department of Diabetes, Monash University, Melbourne, Australia
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Josephine Forbes
- Mater Clinical School, University of Queensland, St. Lucia, Brisbane, Australia
| | - Melinda Coughlan
- Department of Diabetes, Monash University, Melbourne, Australia
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Brooke Harcourt
- Centre for Hormone Research, Murdoch Children's Research Institute, Melbourne, Australia
| | - Sally Penfold
- Department of Diabetes, Monash University, Melbourne, Australia
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Bo Wang
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Kidney Regeneration and Stem Cell Laboratory, Monash University, Melbourne, Australia
| | - Gavin Higgins
- Department of Diabetes, Monash University, Melbourne, Australia
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Raelene Pickering
- Department of Diabetes, Monash University, Melbourne, Australia
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Assam El-Osta
- Department of Diabetes, Monash University, Melbourne, Australia
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Merlin C Thomas
- Department of Diabetes, Monash University, Melbourne, Australia
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Mark E Cooper
- Department of Diabetes, Monash University, Melbourne, Australia
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Phillip Kantharidis
- Department of Diabetes, Monash University, Melbourne, Australia
- JDRF Danielle Alberti Memorial Centre for Diabetes Complications, Diabetes Division, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
49
|
Vukmirovic M, Kaminski N. Impact of Transcriptomics on Our Understanding of Pulmonary Fibrosis. Front Med (Lausanne) 2018; 5:87. [PMID: 29670881 PMCID: PMC5894436 DOI: 10.3389/fmed.2018.00087] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 03/20/2018] [Indexed: 12/22/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal fibrotic lung disease characterized by aberrant remodeling of the lung parenchyma with extensive changes to the phenotypes of all lung resident cells. The introduction of transcriptomics, genome scale profiling of thousands of RNA transcripts, caused a significant inversion in IPF research. Instead of generating hypotheses based on animal models of disease, or biological plausibility, with limited validation in humans, investigators were able to generate hypotheses based on unbiased molecular analysis of human samples and then use animal models of disease to test their hypotheses. In this review, we describe the insights made from transcriptomic analysis of human IPF samples. We describe how transcriptomic studies led to identification of novel genes and pathways involved in the human IPF lung such as: matrix metalloproteinases, WNT pathway, epithelial genes, role of microRNAs among others, as well as conceptual insights such as the involvement of developmental pathways and deep shifts in epithelial and fibroblast phenotypes. The impact of lung and transcriptomic studies on disease classification, endotype discovery, and reproducible biomarkers is also described in detail. Despite these impressive achievements, the impact of transcriptomic studies has been limited because they analyzed bulk tissue and did not address the cellular and spatial heterogeneity of the IPF lung. We discuss new emerging technologies and applications, such as single-cell RNAseq and microenvironment analysis that may address cellular and spatial heterogeneity. We end by making the point that most current tissue collections and resources are not amenable to analysis using the novel technologies. To take advantage of the new opportunities, we need new efforts of sample collections, this time focused on access to all the microenvironments and cells in the IPF lung.
Collapse
Affiliation(s)
- Milica Vukmirovic
- Section of Pulmonary, Critical Care and Sleep Medicine, Precision Pulmonary Medicine Center (P2MED), Yale University School of Medicine, New Haven, CT, United States
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Precision Pulmonary Medicine Center (P2MED), Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
50
|
Potential contribution of alveolar epithelial type I cells to pulmonary fibrosis. Biosci Rep 2017; 37:BSR20171301. [PMID: 29026006 PMCID: PMC5696455 DOI: 10.1042/bsr20171301] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/28/2017] [Accepted: 09/29/2017] [Indexed: 12/13/2022] Open
Abstract
Pulmonary fibrosis (PF) is characterized by inflammation and fibrosis of the interstitium and destruction of alveolar histoarchitecture ultimately leading to a fatal impairment of lung function. Different concepts describe either a dominant role of inflammatory pathways or a disturbed remodeling of resident cells of the lung parenchyma during fibrogenesis. Further, a combination of both the mechanisms has been postulated. The present review emphasizes the particular involvement of alveolar epithelial type I cells in all these processes, their contribution to innate immune/inflammatory functions and maintenance of proper alveolar barrier functions. Amongst the different inflammatory and repair events the purinergic receptor P2X7, an ATP-gated cationic channel that regulates not only apoptosis, necrosis, autophagy, and NLPR3 inflammosome activation, but also the turnover of diverse tight junction (TJ) and water channel proteins, seems to be essential for the stability of alveolar barrier integrity and for the interaction with protective factors during lung injury.
Collapse
|