1
|
Russo RC, Ryffel B. The Chemokine System as a Key Regulator of Pulmonary Fibrosis: Converging Pathways in Human Idiopathic Pulmonary Fibrosis (IPF) and the Bleomycin-Induced Lung Fibrosis Model in Mice. Cells 2024; 13:2058. [PMID: 39768150 PMCID: PMC11674266 DOI: 10.3390/cells13242058] [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: 11/06/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and lethal interstitial lung disease (ILD) of unknown origin, characterized by limited treatment efficacy and a fibroproliferative nature. It is marked by excessive extracellular matrix deposition in the pulmonary parenchyma, leading to progressive lung volume decline and impaired gas exchange. The chemokine system, a network of proteins involved in cellular communication with diverse biological functions, plays a crucial role in various respiratory diseases. Chemokine receptors trigger the activation, proliferation, and migration of lung-resident cells, including pneumocytes, endothelial cells, alveolar macrophages, and fibroblasts. Around 50 chemokines can potentially interact with 20 receptors, expressed by both leukocytes and non-leukocytes such as tissue parenchyma cells, contributing to processes such as leukocyte mobilization from the bone marrow, recirculation through lymphoid organs, and tissue influx during inflammation or immune response. This narrative review explores the complexity of the chemokine system in the context of IPF and the bleomycin-induced lung fibrosis mouse model. The goal is to identify specific chemokines and receptors as potential therapeutic targets. Recent progress in understanding the role of the chemokine system during IPF, using experimental models and molecular diagnosis, underscores the complex nature of this system in the context of the disease. Despite advances in experimental models and molecular diagnostics, discovering an effective therapy for IPF remains a significant challenge in both medicine and pharmacology. This work delves into microarray results from lung samples of IPF patients and murine samples at different stages of bleomycin-induced pulmonary fibrosis. By discussing common pathways identified in both IPF and the experimental model, we aim to shed light on potential targets for therapeutic intervention. Dysregulation caused by abnormal chemokine levels observed in IPF lungs may activate multiple targets, suggesting that chemokine signaling plays a central role in maintaining or perpetuating lung fibrogenesis. The highlighted chemokine axes (CCL8-CCR2, CCL19/CCL21-CCR7, CXCL9-CXCR3, CCL3/CCL4/CCL5-CCR5, and CCL20-CCR6) present promising opportunities for advancing IPF treatment research and uncovering new pharmacological targets within the chemokine system.
Collapse
Affiliation(s)
- Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais-UFMG, Belo Horizonte 31270-901, MG, Brazil
| | - Bernhard Ryffel
- Laboratory of Immuno-Neuro Modulation (INEM), UMR7355 Centre National de la Recherche Scientifique (CNRS), University of Orleans, 45071 Orleans, France
| |
Collapse
|
2
|
Lucinian YA, Martineau P, Abikhzer G, Harel F, Pelletier-Galarneau M. Novel tracers to assess myocardial inflammation with radionuclide imaging. J Nucl Cardiol 2024; 42:102012. [PMID: 39069249 DOI: 10.1016/j.nuclcard.2024.102012] [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: 02/16/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
Myocardial inflammation plays a central role in the pathophysiology of various cardiac diseases. While FDG-PET is currently the primary method for molecular imaging of myocardial inflammation, its effectiveness is hindered by physiological myocardial uptake as well as its propensity for uptake by multiple disease-specific mechanisms. Novel radiotracers targeting diverse inflammatory immune cells and molecular pathways may provide unique insight through the visualization of underlying mechanisms central to the pathogenesis of inflammatory cardiac diseases, offering opportunities for increased understanding of immunocardiology. Moreover, the potentially enhanced specificity may lead to better quantification of disease activity, aiding in the guidance and monitoring of immunomodulatory therapy. This review aims to provide an update on advancements in non-FDG radiotracers for imaging myocardial inflammatory diseases, with a focus on cardiac sarcoidosis, myocarditis, and acute myocardial infarction.
Collapse
Affiliation(s)
| | | | - Gad Abikhzer
- Jewish General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
3
|
Shin J, Miaskowski C, Wong ML, Yates P, Olshen AB, Roy R, Dokiparthi V, Cooper B, Paul S, Conley YP, Levine JD, Hammer MJ, Kober K. Perturbations in inflammatory pathways are associated with shortness of breath profiles in oncology patients receiving chemotherapy. Support Care Cancer 2024; 32:250. [PMID: 38532105 PMCID: PMC11484916 DOI: 10.1007/s00520-024-08446-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 03/18/2024] [Indexed: 03/28/2024]
Abstract
PURPOSE One plausible mechanistic hypothesis is the potential contribution of inflammatory mechanisms to shortness of breath. This study was aimed to evaluate for associations between the occurrence of shortness of breath and perturbations in inflammatory pathways. METHODS Patients with cancer reported the occurrence of shortness of breath six times over two cycles of chemotherapy. Latent class analysis was used to identify subgroups of patients with distinct shortness of breath occurrence profiles (i.e., none (70.5%), decreasing (8.2%), increasing (7.8%), high (13.5%)). Using an extreme phenotype approach, whole transcriptome differential gene expression and pathway impact analyses were performed to evaluate for perturbed signaling pathways associated with shortness of breath between the none and high classes. Two independent samples (RNA-sequencing (n = 293) and microarray (n = 295) methodologies) were evaluated. Fisher's combined probability method was used to combine these results to obtain a global test of the null hypothesis. In addition, an unweighted knowledge network was created using the specific pathway maps to evaluate for interconnections among these pathways. RESULTS Twenty-nine Kyoto Encyclopedia of Genes and Genomes inflammatory signaling pathways were perturbed. The mitogen-activated protein kinase signaling pathway node had the highest closeness, betweenness, and degree scores. In addition, five common respiratory disease-related pathways, that may share mechanisms with cancer-related shortness of breath, were perturbed. CONCLUSIONS Findings provide preliminary support for the hypothesis that inflammation contribute to the occurrence of shortness of breath in patients with cancer. In addition, the mechanisms that underlie shortness of breath in oncology patients may be similar to other respiratory diseases.
Collapse
Affiliation(s)
- Joosun Shin
- School of Nursing, University of California, 2 Koret Way - N631Y, San Francisco, CA, 94143-0610, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Christine Miaskowski
- School of Nursing, University of California, 2 Koret Way - N631Y, San Francisco, CA, 94143-0610, USA
- School of Medicine, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Melisa L Wong
- School of Medicine, University of California, San Francisco, CA, USA
| | - Patsy Yates
- Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Adam B Olshen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Ritu Roy
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Vasuda Dokiparthi
- School of Nursing, University of California, 2 Koret Way - N631Y, San Francisco, CA, 94143-0610, USA
| | - Bruce Cooper
- School of Nursing, University of California, 2 Koret Way - N631Y, San Francisco, CA, 94143-0610, USA
| | - Steven Paul
- School of Nursing, University of California, 2 Koret Way - N631Y, San Francisco, CA, 94143-0610, USA
| | - Yvette P Conley
- School of Nursing, Univeristy of Pittsburgh, 3500 Victoria St, Pittsburgh, 15213, PA, USA
| | - Jon D Levine
- School of Medicine, University of California, San Francisco, CA, USA
| | | | - Kord Kober
- School of Nursing, University of California, 2 Koret Way - N631Y, San Francisco, CA, 94143-0610, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.
| |
Collapse
|
4
|
Russo RC, Quesniaux VFJ, Ryffel B. Homeostatic chemokines as putative therapeutic targets in idiopathic pulmonary fibrosis. Trends Immunol 2023; 44:1014-1030. [PMID: 37951789 DOI: 10.1016/j.it.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal chronic interstitial lung disease (ILD) that affects lung mechanical functions and gas exchange. IPF is caused by increased fibroblast activity and collagen deposition that compromise the alveolar-capillary barrier. Identifying an effective therapy for IPF remains a clinical challenge. Chemokines are key proteins in cell communication that have functions in immunity as well as in tissue homeostasis, damage, and repair. Chemokine receptor signaling induces the activation and proliferation of lung-resident cells, including alveolar macrophages (AMs) and fibroblasts. AMs are an important source of chemokines and cytokines during IPF. We highlight the complexity of this system and, based on insights from genetic and transcriptomic studies, propose a new role for homeostatic chemokine imbalance in IPF, with implications for putative therapeutic targets.
Collapse
Affiliation(s)
- Remo C Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.
| | - Valerie F J Quesniaux
- Experimental and Molecular Immunology and Neurogenetics (INEM), Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7355, University of Orleans, Orleans 45071, France.
| | - Bernhard Ryffel
- Experimental and Molecular Immunology and Neurogenetics (INEM), Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7355, University of Orleans, Orleans 45071, France.
| |
Collapse
|
5
|
Yin Y, Mu C, Wang J, Wang Y, Hu W, Zhu W, Yu X, Hao W, Zheng Y, Li Q, Han W. CXCL17 Attenuates Diesel Exhaust Emissions Exposure-Induced Lung Damage by Regulating Macrophage Function. TOXICS 2023; 11:646. [PMID: 37624152 PMCID: PMC10459829 DOI: 10.3390/toxics11080646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 08/26/2023]
Abstract
Exposure to diesel exhaust emissions (DEE) is strongly linked to innate immune injury and lung injury, but the role of macrophage chemoattractant CXCL17 in the lung damage caused by DEE exposure remains unclear. In this study, whole-body plethysmography (WBP), inflammatory cell differential count, and histopathological analysis were performed to assess respiratory parameters, airway inflammation, and airway injury in C57BL/6 male mice exposed to DEE for 3 months. qRT-PCR, IHC (immunohistochemistry), and ELISA were performed to measure the CXCL17 expression in airway epithelium or BALF (bronchoalveolar lavage fluid) following DEE/Diesel exhaust particle (DEP) exposure. Respiratory parameters, airway inflammation, and airway injury were assessed in CXCL17-overexpressing mice through adeno-associated virus vector Type 5 (AAV5) infection. Additionally, an in vitro THP-1 and HBE co-culture system was constructed. Transwell assay was carried out to evaluate the effect of rh-CXCL17 (recombinant human protein-CXCL17) on THP-1 cell migration. Flow cytometry and qRT-PCR were conducted to assess the impacts of rh-CXCL17 on apoptosis and inflammation/remodeling of HBE cells. We found that the mice exposed to DEE showed abnormal respiratory parameters, accompanied by airway injury and remodeling (ciliary injury in airway epithelium, airway smooth muscle hyperplasia, and increased collagen deposition). Carbon content in airway macrophages (CCAM), but not the number of macrophages in BALF, increased significantly. CXCL17 expression significantly decreased in mice airways and HBE after DEE/DEP exposure. AAV5-CXCL17 enhanced macrophage recruitment and clearance of DEE in the lungs of mice, and it improved respiratory parameters, airway injury, and airway remodeling. In the THP-1/HBE co-culture system, rh-CXCL17 increased THP-1 cell migration while attenuating HBE cell apoptosis and inflammation/remodeling. Therefore, CXCL17 might attenuate DEE-induced lung damage by recruiting and activating pulmonary macrophages, which is expected to be a novel therapeutic target for DEE-associated lung diseases.
Collapse
Affiliation(s)
- Yize Yin
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of Public Health, Qingdao University, Qingdao 266071, China;
| | - Chaohui Mu
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China;
| | - Jiahui Wang
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao 266071, China; (J.W.); (W.H.)
| | - Yixuan Wang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao 266071, China;
- Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China; (W.Z.); (X.Y.)
| | - Wenmin Hu
- School of Medicine and Pharmacy, Ocean University of China, Department of Pulmonary and Critical Care Medicine, University of Health and Rehabilitation Science, Qingdao 266071, China;
| | - Wenjing Zhu
- Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China; (W.Z.); (X.Y.)
- Clinical Research Center, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao 266071, China
| | - Xinjuan Yu
- Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China; (W.Z.); (X.Y.)
- Clinical Research Center, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao 266071, China
| | - Wanming Hao
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao 266071, China; (J.W.); (W.H.)
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China;
| | - Qinghai Li
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao 266071, China; (J.W.); (W.H.)
- Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China; (W.Z.); (X.Y.)
| | - Wei Han
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao 266071, China; (J.W.); (W.H.)
- Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China; (W.Z.); (X.Y.)
- Clinical Research Center, Qingdao Municipal Hospital, University of Health and Rehabilitation Science, Qingdao 266071, China
| |
Collapse
|
6
|
Kadushkin AG, Tahanovich AD, Movchan LV, Zafranskaya MM, Dziadzichkina VV, Shman TV. [Population rearrangement of B-lymphocytes expressing chemokine receptors in patients with chronic obstructive pulmonary disease]. BIOMEDITSINSKAIA KHIMIIA 2022; 68:134-143. [PMID: 35485487 DOI: 10.18097/pbmc20226802134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
To date, there are no drugs that can prevent progressive destruction of lung tissue and small airway fibrosis in patients with chronic obstructive pulmonary disease (COPD). Therefore, molecular mechanisms of this disease are being studied. The aim of this study was to determine the chemokine receptor expression pattern of B-lymphocytes from peripheral blood and airways of patients with COPD. Peripheral blood was collected from 51 smokers with COPD, 21 healthy smokers, and 20 healthy non-smokers. Seven smokers with COPD and 7 healthy smokers were recruited to undergo bronchoscopy with bronchoalveolar lavage (BAL). The expression of chemokine receptors CCR5, CCR6, CCR7, CXCR3, CXCR4, and CXCR5 on the surface of blood and BAL B-lymphocytes was determined by flow cytometry. It was found that the percentage of blood B-lymphocytes expressing chemokine receptors CCR5 and CXCR3 was higher in smokers with COPD compared with healthy smokers and healthy non-smokers. The percentage of CD27⁺ B-cells expressing CCR5 and CXCR3 receptors exceeded the proportion of CD27⁻ B-lymphocytes expressing these receptors in peripheral blood of patients with COPD and healthy controls. In smoking patients with COPD, the percentage of BAL B-cells expressing receptors CCR5 and CXCR3 was also increased compared with healthy smokers. There were no differences in the percentage of B-lymphocytes expressing receptors CXCR4, CXCR5, CCR6, and CCR7 in both peripheral blood and BAL between smokers with COPD and healthy smokers. A greater percentage of CD27⁻ B-lymphocytes than CD27⁺ B-cells expressed receptors CXCR4, CXCR5, CCR6, and CCR7 in the peripheral blood of smokers with COPD and healthy controls. The results of this study indicate a modification in the chemokine receptor profile of B-lymphocytes in COPD.
Collapse
Affiliation(s)
| | | | - L V Movchan
- Republican Scientific and Practical Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | - M M Zafranskaya
- International Sakharov Environmental Institute of Belarusian State University, Minsk, Belarus
| | | | - T V Shman
- Republican Scientific and Practical Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| |
Collapse
|
7
|
Ferrero MR, Tavares LP, Garcia CC. The Dual Role of CCR5 in the Course of Influenza Infection: Exploring Treatment Opportunities. Front Immunol 2022; 12:826621. [PMID: 35126379 PMCID: PMC8810482 DOI: 10.3389/fimmu.2021.826621] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022] Open
Abstract
Influenza is one of the most relevant respiratory viruses to human health causing annual epidemics, and recurrent pandemics. Influenza disease is principally associated with inappropriate activation of the immune response. Chemokine receptor 5 (CCR5) and its cognate chemokines CCL3, CCL4 and CCL5 are rapidly induced upon influenza infection, contributing to leukocyte recruitment into the airways and a consequent effective antiviral response. Here we discuss the existing evidence for CCR5 role in the host immune responses to influenza virus. Complete absence of CCR5 in mice revealed the receptor’s role in coping with influenza via the recruitment of early memory CD8+ T cells, B cell activation and later recruitment of activated CD4+ T cells. Moreover, CCR5 contributes to inflammatory resolution by enhancing alveolar macrophages survival and reprogramming macrophages to pro-resolving phenotypes. In contrast, CCR5 activation is associated with excessive recruitment of neutrophils, inflammatory monocytes, and NK cells in models of severe influenza pneumonia. The available data suggests that, while CCL5 can play a protective role in influenza infection, CCL3 may contribute to an overwhelming inflammatory process that can harm the lung tissue. In humans, the gene encoding CCR5 might contain a 32-base pair deletion, resulting in a truncated protein. While discordant data in literature regarding this CCR5 mutation and influenza severity, the association of CCR5delta32 and HIV resistance fostered the development of different CCR5 inhibitors, now being tested in lung inflammation therapy. The potential use of CCR5 inhibitors to modulate the inflammatory response in severe human influenza infections is to be addressed.
Collapse
Affiliation(s)
- Maximiliano Ruben Ferrero
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
- *Correspondence: Maximiliano Ruben Ferrero,
| | - Luciana Pádua Tavares
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Cristiana Couto Garcia
- Laboratory of Respiratory Virus and Measles, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| |
Collapse
|
8
|
Pouzol L, Sassi A, Baumlin N, Tunis M, Strasser DS, Lehembre F, Martinic MM. CXCR7 Antagonism Reduces Acute Lung Injury Pathogenesis. Front Pharmacol 2021; 12:748740. [PMID: 34803691 PMCID: PMC8602191 DOI: 10.3389/fphar.2021.748740] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022] Open
Abstract
Loss of control in the trafficking of immune cells to the inflamed lung tissue contributes to the pathogenesis of life-threatening acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS). Targeting CXCR7 has been proposed as a potential therapeutic approach to reduce pulmonary inflammation; however, its role and its crosstalk with the two chemokine receptors CXCR3 and CXCR4 via their shared ligands CXCL11 and CXCL12 is not yet completely understood. The present paper aimed to characterize the pathological role of the CXCR3/CXCR4/CXCR7 axis in a murine model of ALI. Lipopolysaccharide (LPS) inhalation in mice resulted in the development of key pathologic features of ALI/ARDS, including breathing dysfunctions, alteration in the alveolar capillary barrier, and lung inflammation. LPS inhalation induced immune cell infiltration into the bronchoalveolar space, including CXCR3+ and CXCR4+ cells, and enhanced the expression of the ligands of these two chemokine receptors. The first-in-class CXCR7 antagonist, ACT-1004-1239, increased levels of CXCL11 and CXCL12 in the plasma without affecting their levels in inflamed lung tissue, and consequently reduced CXCR3+ and CXCR4+ immune cell infiltrates into the bronchoalveolar space. In the early phase of lung inflammation, characterized by a massive influx of neutrophils, treatment with ACT-1004-1239 significantly reduced the LPS-induced breathing pattern alteration. Both preventive and therapeutic treatment with ACT-1004-1239 reduced lung vascular permeability and decreased inflammatory cell infiltrates. In conclusion, these results demonstrate a key pathological role of CXCR7 in ALI/ARDS and highlight the clinical potential of ACT-1004-1239 in ALI/ARDS pathogenesis.
Collapse
Affiliation(s)
| | - Anna Sassi
- Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | | | - Mélanie Tunis
- Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | | | | | | |
Collapse
|
9
|
Wong A, Liu M. Inflammatory responses in lungs from donation after brain death: Mechanisms and potential therapeutic targets. J Heart Lung Transplant 2021; 40:890-896. [PMID: 34167864 DOI: 10.1016/j.healun.2021.03.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 01/16/2023] Open
Abstract
The vast majority of lungs used in clinical transplantation are donated after brain death (DBD). The utilization of DBD lungs is low due to brain death-induced lung injury. Moreover, inflammatory responses in DBD lungs used for transplantation contribute to ischemia-reperfusion injury and primary graft dysfunction. Work from human observational studies has demonstrated overexpression of cytokines, activation of endothelial cells, and cell death in DBD lungs, are associated with the activation of signaling pathways. Animal models have characterized the pulmonary injury induced by brain death and identified potential strategies to improve donor management. Interestingly, transcriptomic studies comparing DBD and donated after circulatory death (DCD) lungs have found that inflammatory responses are elevated in DBD lungs, while cell death pathways are elevated in DCD lungs. Development of the ex vivo lung perfusion technique, has made it possible to assess donor lungs using inflammation and cell death biomarkers. In the future, identification of potential therapeutic targets and development of novel treatments strategies may allow for lung repair during EVLP prior to transplantation.
Collapse
Affiliation(s)
- Aaron Wong
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Ontario, Canada. https://twitter.com/aaronkkwong
| | - Mingyao Liu
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Ontario, Canada; Latner Thoracic Surgical Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Departments of Surgery, Medicine and Physiology, Temerty Faculty of Medicine, University of Toronto, Ontario, Canada.
| |
Collapse
|
10
|
Alon R, Sportiello M, Kozlovski S, Kumar A, Reilly EC, Zarbock A, Garbi N, Topham DJ. Leukocyte trafficking to the lungs and beyond: lessons from influenza for COVID-19. Nat Rev Immunol 2021; 21:49-64. [PMID: 33214719 PMCID: PMC7675406 DOI: 10.1038/s41577-020-00470-2] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2020] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19). Understanding of the fundamental processes underlying the versatile clinical manifestations of COVID-19 is incomplete without comprehension of how different immune cells are recruited to various compartments of virus-infected lungs, and how this recruitment differs among individuals with different levels of disease severity. As in other respiratory infections, leukocyte recruitment to the respiratory system in people with COVID-19 is orchestrated by specific leukocyte trafficking molecules, and when uncontrolled and excessive it results in various pathological complications, both in the lungs and in other organs. In the absence of experimental data from physiologically relevant animal models, our knowledge of the trafficking signals displayed by distinct vascular beds and epithelial cell layers in response to infection by SARS-CoV-2 is still incomplete. However, SARS-CoV-2 and influenza virus elicit partially conserved inflammatory responses in the different respiratory epithelial cells encountered early in infection and may trigger partially overlapping combinations of trafficking signals in nearby blood vessels. Here, we review the molecular signals orchestrating leukocyte trafficking to airway and lung compartments during primary pneumotropic influenza virus infections and discuss potential similarities to distinct courses of primary SARS-CoV-2 infections. We also discuss how an imbalance in vascular activation by leukocytes outside the airways and lungs may contribute to extrapulmonary inflammatory complications in subsets of patients with COVID-19. These multiple molecular pathways are potential targets for therapeutic interventions in patients with severe COVID-19.
Collapse
Affiliation(s)
- Ronen Alon
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel.
| | - Mike Sportiello
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Stav Kozlovski
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Ashwin Kumar
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Emma C Reilly
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Alexander Zarbock
- Department of Cellular Immunology, Institute of Experimental Immunology Medical Faculty, University of Bonn, Bonn, Germany
| | - Natalio Garbi
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
11
|
Lim JY, Ryu DB, Kim TW, Lee SE, Park G, Yoon HK, Min CK. CCL1 blockade alleviates human mesenchymal stem cell (hMSC)-induced pulmonary fibrosis in a murine sclerodermatous graft-versus-host disease (Scl-GVHD) model. Stem Cell Res Ther 2020; 11:254. [PMID: 32586381 PMCID: PMC7318460 DOI: 10.1186/s13287-020-01768-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/07/2020] [Accepted: 06/11/2020] [Indexed: 02/08/2023] Open
Abstract
Background Human chronic graft-versus-host disease (CGVHD) shares clinical characteristics with a murine sclerodermatous GVHD (Scl-GVHD, B10.D2 → BALB/c) model that is characterized by skin and lung fibrosis. In this study, bone marrow- or adipose tissue-derived human mesenchymal stem cells (hMSCs) were injected into the Scl-GVHD mice to address their therapeutic effect on CGVHD. Methods Lethally irradiated BALB/c mice were transplanted with B10.D2 T cell-depleted bone marrow with or without spleen cells to generate Scl-GVHD. hMSCs were intravenously treated on days 3, 5, and 7 post-transplantation, and the control antibody or CCL1 blocking antibody was subcutaneously injected according to the same schedule as the hMSCs. Fourteen days after transplantation, the recipient mice were sacrificed, and their skin and lungs were analyzed. Results After the early injection of hMSCs after transplantation, the clinical and pathological severity of Scl-GVHD in the skin was significantly attenuated, whereas the pathological score was exacerbated in the lungs. hMSCs had migrated into the lungs, but not into the skin. CD11b monocyte/macrophages and CD4 T cells were markedly decreased in skin tissues, whereas there was an early recruitment of CD11b cells, and subsequently increased infiltration of CD4 T cells, in the lungs. Importantly, hMSCs persistently upregulated the expression of CCL1 in the lungs, but not in the skin. Concurrent treatment of hMSCs with a CCL1-blocking antibody alleviated the severity of the lung histopathology score and fibrosis with the preservation of the cutaneous protective effect against CGVHD. Infiltration of CD3 T cells and CD68 macrophages and upregulation of chemokines were also decreased in lung tissues, along with the recruitment of eosinophils and tissue IgE expression. In the skin, chemokine expression was further reduced after CCL1 blockade. Conclusions These data demonstrate that despite a protective effect against Scl-GVHD in the skin, administration of hMSCs exacerbated lung fibrosis associated with eosinophilia and airway inflammation through persistent CCL1 upregulation. CCL1 blockade offers a potential treatment of pulmonary complications induced after treatment with hMSCs.
Collapse
Affiliation(s)
- Ji-Young Lim
- Hematology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, South Korea
| | - Da-Bin Ryu
- Hematology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, South Korea
| | - Tae Woo Kim
- Hematology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, South Korea
| | - Sung-Eun Lee
- Hematology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, South Korea
| | - Gyeongsin Park
- Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Hyoung Kyu Yoon
- Pulmonology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Chang-Ki Min
- Hematology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, South Korea.
| |
Collapse
|
12
|
The protective and pathogenic roles of CXCL17 in human health and disease: Potential in respiratory medicine. Cytokine Growth Factor Rev 2020; 53:53-62. [PMID: 32345516 PMCID: PMC7177079 DOI: 10.1016/j.cytogfr.2020.04.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023]
Abstract
C-X-C motif chemokine 17 (CXCL17), plays a functional role in maintaining homeostasis at mucosal barriers. CXCL17 expression is associated with both disease progression and protection in various diseases. The multifactorial mechanistic properties of CXCL17 could be exploited as a therapeutic target
C-X-C motif chemokine 17 (CXCL-17) is a novel chemokine that plays a functional role maintaining homeostasis at distinct mucosal barriers, including regulation of myeloid-cell recruitment, angiogenesis, and control of microorganisms. Particularly, CXCL17 is produced along the epithelium of the airways both at steady state and under inflammatory conditions. While increased CXCL17 expression is associated with disease progression in pulmonary fibrosis, asthma, and lung/hepatic cancer, it is thought to play a protective role in pancreatic cancer, autoimmune encephalomyelitis and viral infections. Thus, there is emerging evidence pointing to both a harmful and protective role for CXCL17 in human health and disease, with therapeutic potential for translational applications. In this review, we provide an overview of the discovery, characteristics and functions of CXCL17 emphasizing its clinical potential in respiratory disorders.
Collapse
|
13
|
Abstract
This review discusses nuclear imaging of inflammation using molecular probes beyond fluoro-d-glucose, is structured by cellular targets, and focuses on those tracers that have been successfully applied clinically.
Collapse
Affiliation(s)
- Malte Kircher
- Department of Nuclear Medicine, University Hospital Augsburg, Stenglinstr. 2, Würzburg 86156, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Augsburg, Stenglinstr. 2, Würzburg 86156, Germany.
| |
Collapse
|
14
|
Abstract
Asthma is a heterogeneous inflammatory disease of the airways that is associated with airway hyperresponsiveness and airflow limitation. Although asthma was once simply categorized as atopic or nonatopic, emerging analyses over the last few decades have revealed a variety of asthma endotypes that are attributed to numerous pathophysiological mechanisms. The classification of asthma by endotype is primarily routed in different profiles of airway inflammation that contribute to bronchoconstriction. Many asthma therapeutics target G protein-coupled receptors (GPCRs), which either enhance bronchodilation or prevent bronchoconstriction. Short-acting and long-acting β 2-agonists are widely used bronchodilators that signal through the activation of the β 2-adrenergic receptor. Short-acting and long-acting antagonists of muscarinic acetylcholine receptors are used to reduce bronchoconstriction by blocking the action of acetylcholine. Leukotriene antagonists that block the signaling of cysteinyl leukotriene receptor 1 are used as an add-on therapy to reduce bronchoconstriction and inflammation induced by cysteinyl leukotrienes. A number of GPCR-targeting asthma drug candidates are also in different stages of development. Among them, antagonists of prostaglandin D2 receptor 2 have advanced into phase III clinical trials. Others, including antagonists of the adenosine A2B receptor and the histamine H4 receptor, are in early stages of clinical investigation. In the past decade, significant research advancements in pharmacology, cell biology, structural biology, and molecular physiology have greatly deepened our understanding of the therapeutic roles of GPCRs in asthma and drug action on these GPCRs. This review summarizes our current understanding of GPCR signaling and pharmacology in the context of asthma treatment. SIGNIFICANCE STATEMENT: Although current treatment methods for asthma are effective for a majority of asthma patients, there are still a large number of patients with poorly controlled asthma who may experience asthma exacerbations. This review summarizes current asthma treatment methods and our understanding of signaling and pharmacology of G protein-coupled receptors (GPCRs) in asthma therapy, and discusses controversies regarding the use of GPCR drugs and new opportunities in developing GPCR-targeting therapeutics for the treatment of asthma.
Collapse
Affiliation(s)
- Stacy Gelhaus Wendell
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (S.G.W., C.Z.); Bioinformatics Institute, Agency for Science, Technology, and Research, Singapore (H.F.); and Department of Biological Sciences, National University of Singapore, and Center for Computational Biology, DUKE-NUS Medical School, Singapore (H.F.)
| | - Hao Fan
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (S.G.W., C.Z.); Bioinformatics Institute, Agency for Science, Technology, and Research, Singapore (H.F.); and Department of Biological Sciences, National University of Singapore, and Center for Computational Biology, DUKE-NUS Medical School, Singapore (H.F.)
| | - Cheng Zhang
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (S.G.W., C.Z.); Bioinformatics Institute, Agency for Science, Technology, and Research, Singapore (H.F.); and Department of Biological Sciences, National University of Singapore, and Center for Computational Biology, DUKE-NUS Medical School, Singapore (H.F.)
| |
Collapse
|
15
|
Mamazhakypov A, Viswanathan G, Lawrie A, Schermuly RT, Rajagopal S. The role of chemokines and chemokine receptors in pulmonary arterial hypertension. Br J Pharmacol 2019; 178:72-89. [PMID: 31399998 DOI: 10.1111/bph.14826] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by progressive pulmonary artery remodelling leading to increased right ventricular pressure overload, which results in right heart failure and premature death. Inflammation plays a central role in the development of PAH, and the recruitment and function of immune cells are tightly regulated by chemotactic cytokines called chemokines. A number of studies have shown that the development and progression of PAH are associated with the dysregulated expression of several chemokines and chemokine receptors in the pulmonary vasculature. Moreover, some chemokines are differentially regulated in the pressure-overloaded right ventricle. Recent studies have tested the efficacy of pharmacological agents targeting several chemokines and chemokine receptors for their effects on the development of PAH, suggesting that these receptors could serve as useful therapeutic targets. In this review, we provide recent insights into the role of chemokines and chemokine receptors in PAH and RV remodelling and the opportunities and roadblocks in targeting them. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.1/issuetoc.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Gayathri Viswanathan
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Allan Lawrie
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Ralph Theo Schermuly
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Sudarshan Rajagopal
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
16
|
Corry DB, Kheradmand F, Luong A, Pandit L. Immunological Mechanisms of Airway Diseases and Pathways to Therapy. Clin Immunol 2019. [DOI: 10.1016/b978-0-7020-6896-6.00041-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
17
|
Reversing CXCL10 Deficiency Ameliorates Kidney Disease in Diabetic Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2763-2773. [PMID: 30273603 DOI: 10.1016/j.ajpath.2018.08.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 08/04/2018] [Accepted: 08/23/2018] [Indexed: 12/11/2022]
Abstract
The excessive accumulation of extracellular matrix material in the kidney is a histopathologic hallmark of diabetic kidney disease that correlates closely with declining function. Although considerable research has focused on the role of profibrotic factors, comparatively little attention has been paid to the possibility that a diminution in endogenous antifibrotic factors may also contribute. Among the latter, the ELR- CXC chemokines, CXCL9, CXCL10, and CXCL11, have been shown to provide a stop signal to prevent excessive fibrosis. Although the plasma concentrations of CXCL9 and CXCL11 were similar, those of CXCL10 were markedly lower in diabetic db/db mice compared with control db/m mice. In cell culture, CXCL10 inhibited kidney fibroblast collagen production in response to high glucose and the prosclerotic growth factor, transforming growth factor-β. In vivo, recombinant murine CXCL10 reduced mesangial and peritubular matrix expansion, albuminuria, and glomerular hypertrophy in db/db mice. In bone marrow, a major source of circulating chemokines, the concentration of CXCL10 was lower in cells derived from diabetic mice than from their nondiabetic counterparts. Silencing of CXCR3, the cognate receptor for CXCL10, abrogated the antifibrotic effects of bone marrow-derived secretions. In conclusion, experimental diabetes is a state of CXCL10 deficiency and that restoration of CXCL10 abundance prevented fibrosis and the development of diabetic kidney disease in mice.
Collapse
|
18
|
Weiskirchen R, Weiskirchen S, Tacke F. Organ and tissue fibrosis: Molecular signals, cellular mechanisms and translational implications. Mol Aspects Med 2018; 65:2-15. [PMID: 29958900 DOI: 10.1016/j.mam.2018.06.003] [Citation(s) in RCA: 381] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/25/2018] [Indexed: 02/08/2023]
Abstract
Fibrosis denotes excessive scarring, which exceeds the normal wound healing response to injury in many tissues. Although the extracellular matrix deposition appears unstructured disrupting the normal tissue architecture and subsequently impairing proper organ function, fibrogenesis is a highly orchestrated process determined by defined sequences of molecular signals and cellular response mechanisms. Persistent injury and parenchymal cell death provokes tissue inflammation, macrophage activation and immune cell infiltration. The release of biologically highly active soluble mediators (alarmins, cytokines, chemokines) lead to the local activation of collagen producing mesenchymal cells such as pericytes, myofibroblasts or Gli1 positive mesenchymal stem cell-like cells, to a transition of various cell types into myofibroblasts as well as to the recruitment of fibroblast precursors. Clinical observations and experimental models highlighted that fibrosis is not a one-way road. Specific mechanistic principles of fibrosis regression involve the resolution of chronic tissue injury, the shift of inflammatory processes towards recovery, deactivation of myofibroblasts and finally fibrolysis of excess matrix scaffold. The thorough understanding of common principles of fibrogenic molecular signals and cellular mechanisms in various organs - such as liver, kidney, lung, heart or skin - is the basis for developing improved diagnostics including biomarkers or imaging techniques and novel antifibrotic therapeutics.
Collapse
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Germany
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Germany
| | - Frank Tacke
- Dept. of Medicine III, University Hospital Aachen, Germany.
| |
Collapse
|
19
|
Wang F, Fu X, Wu X, Zhang J, Zhu J, Zou Y, Li J. Bone marrow derived M 2 macrophages protected against lipopolysaccharide-induced acute lung injury through inhibiting oxidative stress and inflammation by modulating neutrophils and T lymphocytes responses. Int Immunopharmacol 2018; 61:162-168. [PMID: 29883961 DOI: 10.1016/j.intimp.2018.05.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 05/06/2018] [Accepted: 05/20/2018] [Indexed: 01/31/2023]
Abstract
Acute lung injury (ALI) is characterized by aggravated inflammatory responses and the subsequent alveolar-capillary injury for which there are no specific therapies available currently. The present study was designed to investigate the protective roles of bone marrow derived M2 macrophages (M2 BMDMs) in lipopolysaccharide (LPS) induced ALI. M2 BMDMs were obtained from bone marrow cells stimulated with M-CSF and IL-4. Mice received M2 BMDMs intratracheally 3 h after LPS administration. Histology and wet/dry (W/D) weight ratio, activated immune cells and total protein were detected. Cytokines production were measured in vivo and vitro study. The effects of PD-L1 blockade on M2 BMDMs were calculated. The results showed that M2 BMDMs administration reduced the infiltration of neutrophils, inhibited the oxidative stress, while increased the counts of CD3+T lymphocytes as well as CD4+CD25+ regulatory T lymphocytes. Further, M2 BMDMs suppressed the TNF-α, IL-1β and IL-6 production, while increased the IL-10 production. Blockade of PD-L1/PD-1 pathway reversed cytokines production of M2 BMDMs in the BALF. These findings indicated that M2 BMDMs might be a promising therapeutic strategy for LPS-induced ALI through inhibiting oxidative stress and inflammation by modulating neutrophils and T lymphocytes responses.
Collapse
Affiliation(s)
- Fang Wang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20080, China; Department of Anesthesiology and Intensive Care, Changhai Hospital, the Second Military Medical University, Shanghai 200433, China
| | - Xiazhen Fu
- Department of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Xinwan Wu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20080, China
| | - Jianhai Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20080, China
| | - Jiali Zhu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20080, China
| | - Yun Zou
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20080, China; Department of Anesthesiology and Intensive Care, Changhai Hospital, the Second Military Medical University, Shanghai 200433, China.
| | - Jinbao Li
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20080, China.
| |
Collapse
|
20
|
Yang T, Li Y, Lyu Z, Huang K, Corrigan CJ, Ying S, Wang W, Wang C. Characteristics of Proinflammatory Cytokines and Chemokines in Airways of Asthmatics: Relationships with Disease Severity and Infiltration of Inflammatory Cells. Chin Med J (Engl) 2018; 130:2033-2040. [PMID: 28836545 PMCID: PMC5586170 DOI: 10.4103/0366-6999.213428] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Increased proinflammatory cytokines and chemokines might contribute to infiltration of inflammatory cells and remodeling in airways of asthma. Although these molecules may be associated with asthma, there is lack of systemic evidence showing which and how important these events are in the disease. We aimed to analyze the concentrations of these molecules in the airways and relationships with disease severity and with airway infiltration of inflammatory cells in a large cohort of asthmatics (n = 70, including 37 mild and 33 moderate/severe asthmatics) compared with controls (n = 30). METHODS Meso scale discovery system and commercial ELISA kits were used to measure the concentrations of proinflammatory cytokines interleukin (IL)-1β; tumor necrosis factor-alpha (TNF-α); IL-6; and IL-17 and CC and CXC chemokines CCL2, CCL4, CCL11, CCL13, CCL17, CCL22, and CCL26 and CXCL8, CXCL9, CXCL10, and CXCL11 in bronchoalveolar lavage fluid of asthmatics and controls. RESULTS The concentrations of IL-1, TNF-α, IL-6, CXCL8 and CXCL10, and CCL4, CCL11, CCL17, and CCL22 were significantly elevated in asthmatics compared with controls (P < 0.05). The concentrations of TNF-α and CXCL8, but not others, were negatively correlated with severity of disease (lung function forced expiratory volume in 1 s) (TNF-α vs. total: r = -0.359, P= 0.002 vs. moderate/severe: r= -0.541, P= 0.001; CXCL8 vs. total: r = -0.327, P= 0.006 vs. moderate/severe: r = -0.625, P= 0.0001, respectively). In addition, concentrations of these two molecules were also correlated with the absolute numbers of infiltrating eosinophils and neutrophils in asthmatic airways. CONCLUSIONS Increased concentrations of TNF-α and CXCL8 are associated with pathogenesis of asthma. Targeting these molecules might provide an alternative therapeutic for this disease.
Collapse
Affiliation(s)
- Ting Yang
- National Clinical Research Center for Respiratory Diseases, Center for Respiratory Diseases, China-Japan Friendship Hospital, Capital Medical University, Beijing 100029, China
| | - Yan Li
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Zhe Lyu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Kewu Huang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University and Beijing Institute of Respiratory Medicine, Beijing 100020, China
| | - Chris J Corrigan
- MRC and Asthma Centre in Allergic Mechanisms of Asthma, Division of Asthma, Allergy and Lung Biology, King's College London, London, UK
| | - Sun Ying
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, Division of Asthma, Allergy and Lung Biology, King's College London, London, UK
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Chen Wang
- National Clinical Research Center for Respiratory Diseases, Center for Respiratory Diseases, China-Japan Friendship Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
21
|
McCubbrey AL, Barthel L, Mohning MP, Redente EF, Mould KJ, Thomas SM, Leach SM, Danhorn T, Gibbings SL, Jakubzick CV, Henson PM, Janssen WJ. Deletion of c-FLIP from CD11b hi Macrophages Prevents Development of Bleomycin-induced Lung Fibrosis. Am J Respir Cell Mol Biol 2018; 58:66-78. [PMID: 28850249 DOI: 10.1165/rcmb.2017-0154oc] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a progressive lung disease with complex pathophysiology and fatal prognosis. Macrophages (MΦ) contribute to the development of lung fibrosis; however, the underlying mechanisms and specific MΦ subsets involved remain unclear. During lung injury, two subsets of lung MΦ coexist: Siglec-Fhi resident alveolar MΦ and a mixed population of CD11bhi MΦ that primarily mature from immigrating monocytes. Using a novel inducible transgenic system driven by a fragment of the human CD68 promoter, we targeted deletion of the antiapoptotic protein cellular FADD-like IL-1β-converting enzyme-inhibitory protein (c-FLIP) to CD11bhi MΦ. Upon loss of c-FLIP, CD11bhi MΦ became susceptible to cell death. Using this system, we were able to show that eliminating CD11bhi MΦ present 7-14 days after bleomycin injury was sufficient to protect mice from fibrosis. RNA-seq analysis of lung MΦ present during this time showed that CD11bhi MΦ, but not Siglec-Fhi MΦ, expressed high levels of profibrotic chemokines and growth factors. Human MΦ from patients with idiopathic pulmonary fibrosis expressed many of the same profibrotic chemokines identified in murine CD11bhi MΦ. Elimination of monocyte-derived MΦ may help in the treatment of fibrosis. We identify c-FLIP and the associated extrinsic cell death program as a potential pathway through which these profibrotic MΦ may be pharmacologically targeted.
Collapse
Affiliation(s)
- Alexandra L McCubbrey
- 1 Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado.,2 Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, and
| | - Lea Barthel
- 2 Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, and
| | - Michael P Mohning
- 1 Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado.,2 Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, and
| | - Elizabeth F Redente
- 1 Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado.,3 Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado.,4 Department of Research, Veterans Affairs Eastern Colorado Health Care System, Denver, Colorado
| | - Kara J Mould
- 1 Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado
| | - Stacey M Thomas
- 2 Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, and
| | - Sonia M Leach
- 5 Center for Genes, Environment, and Health, and.,6 Department of Biomedical Research, National Jewish Health, Denver, Colorado; and
| | - Thomas Danhorn
- 5 Center for Genes, Environment, and Health, and.,6 Department of Biomedical Research, National Jewish Health, Denver, Colorado; and
| | - Sophie L Gibbings
- 3 Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado
| | - Claudia V Jakubzick
- 3 Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado.,7 Integrated Department of Immunology, National Jewish Health and University of Colorado Denver Anshutz Campus, Denver, Colorado
| | - Peter M Henson
- 3 Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado
| | - William J Janssen
- 1 Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado.,2 Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, and
| |
Collapse
|
22
|
Toba H, Tomankova T, Wang Y, Bai X, Cho HR, Guan Z, Adeyi OA, Tian F, Keshavjee S, Liu M. XB130 deficiency enhances lipopolysaccharide-induced septic response and acute lung injury. Oncotarget 2018; 7:25420-31. [PMID: 27029000 PMCID: PMC5041914 DOI: 10.18632/oncotarget.8326] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 03/08/2016] [Indexed: 01/03/2023] Open
Abstract
XB130 is a novel oncoprotein that promotes cancer cell survival, proliferation and migration. Its physiological function in vivo is largely unknown. The objective of this study was to determine the role of XB130 in lipopolysaccharide (LPS)-induced septic responses and acute lung injury. LPS was intraperitoneally administrated to Xb130 knockout (KO) and wild type (WT) mice. There was a significant weight loss in KO mice at Day 2 and significantly higher disease scores during the 7 days of observation. The levels of tumor necrosis factor-alpha, monocyte chemoattractant protein-1, interleukin-6 and interleukin-10 in the serum were significantly higher in KO mice at Day 2. In KO mice there were a significantly higher lung injury score, higher wet/dry lung weight ratio, more apoptotic cells and less proliferative cells in the lung. Macrophage infiltration was significantly elevated in the lung of KO mice. There was significantly increased number of p-GSK-3β positive cells in KO mice, which were mainly neutrophils and macrophages. XB130 is expressed in alveolar type I and type II cells in the lung. The expression in these cells was significantly reduced after LPS challenge. XB130 deficiency delayed the recovery from systemic septic responses, and the presence of XB130 in the alveolar epithelial cells may provide protective mechanisms by reducing cell death and promoting cell proliferation, and reducing pulmonary permeability.
Collapse
Affiliation(s)
- Hiroaki Toba
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Universal Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Tereza Tomankova
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Universal Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Yingchun Wang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Universal Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Xiaohui Bai
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Universal Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Hae-Ra Cho
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Universal Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Zhehong Guan
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Universal Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Oyedele A Adeyi
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Feng Tian
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Universal Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Universal Health Network, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Universal Health Network, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Deng X, Zhou X, Deng Y, Liu F, Feng X, Yin Q, Gu Y, Shi S, Xu M. Thrombin Induces CCL2 Expression in Human Lung Fibroblasts via p300 Mediated Histone Acetylation and NF-KappaB Activation. J Cell Biochem 2017; 118:4012-4019. [PMID: 28407300 DOI: 10.1002/jcb.26057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/11/2017] [Indexed: 02/05/2023]
Abstract
Thrombin has been shown to play a key role in lung diseases such as pulmonary fibrosis via the induction of fibrotic cytokine- chemokine (CC motif) ligand-2 (CCL2) expression. We previously reported that transcription factor nuclear factor-κB (NF-κB) is responsible for thrombin-induced CCL2 expression in human lung fibroblasts (HLFs). Here, we extended our study to investigate the epigenetic regulation mechanism for thrombin-induced CCL2 expression in HLFs. HLFs were cultured in F-12 medium. CCL2 protein and mRNA levels were detected by ELISA and quantitative real-time PCR, respectively. Histone, histone acetyltransferases, and NF-κB binding to CCL2 promoter were detected by ChIP assay. NF-κB activation was detected by Western blotting. We revealed that increased binding of histone acetyltransferase p300 and acetylated histone H3 and H4 to CCL2 promoter are responsible for thrombin induced CCL2 expression in HLF cells. In addition, p300 inhibition attenuates both thrombin induced-CCL2 expression and histone H3 and H4 acetylation in HLFs, suggesting that p300 is involved in thrombin-induced CCL2 expression via hyperacetylating histone H3 and H4. Our data further showed that p300 also regulates CCL2 expression via interaction with NF-κB p65, as depletion of p300 inhibits both NF-κB p65 activation and its binding to CCL2 promoter. The findings strongly suggest that epigenetic dysregulation and the interaction between histone acetyltransferase and transcription factor may be responsible for thrombin induced-CCL2 expression in HLFs. Increased understanding of the epigenetic mechanisms of CCL2 regulation may provide opportunities for identifying novel molecular targets for therapeutic purposes. J. Cell. Biochem. 118: 4012-4019, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xiaoling Deng
- Department of Basic Medical Science, Xiamen University Medical College, Xiamen, 361102, Fujian Province, People's Republic of China
| | - Xiaoqiong Zhou
- Department of Basic Medical Science, Xiamen University Medical College, Xiamen, 361102, Fujian Province, People's Republic of China
| | - Yan Deng
- Department of Respiratory Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong Province, People's Republic of China
| | - Fan Liu
- Department of Basic Medical Science, Xiamen University Medical College, Xiamen, 361102, Fujian Province, People's Republic of China
| | - Xiaofan Feng
- Department of Basic Medical Science, Xiamen University Medical College, Xiamen, 361102, Fujian Province, People's Republic of China
| | - Qi Yin
- Department of Basic Medical Science, Xiamen University Medical College, Xiamen, 361102, Fujian Province, People's Republic of China
| | - Yinzhen Gu
- Department of Basic Medical Science, Xiamen University Medical College, Xiamen, 361102, Fujian Province, People's Republic of China
| | - Songlin Shi
- Department of Basic Medical Science, Xiamen University Medical College, Xiamen, 361102, Fujian Province, People's Republic of China
| | - Mingyan Xu
- Department of Oral Biology and Biomaterial, Xiamen Stomatological Research Institute, Xiamen Medical College, Xiamen, 361000, Fujian Province, People's Republic of China
| |
Collapse
|
24
|
Han CH, Zhang PX, Liu WW. Macrophage polarization is related to the pathogenesis of decompression induced lung injury. Med Gas Res 2017; 7:220-223. [PMID: 29152216 PMCID: PMC5674661 DOI: 10.4103/2045-9912.215753] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Studies have shown that blood bubbles may be detectable and there is ultrasonic evidence of acute interstitial lung edema even after diving without protocol violation. Macrophages play a central role in the inflammation, and macrophage polarization is closely related to the pathogenesis some lung diseases. Available findings indicate that decompression may induce the production of pro-inflammatory cytokines, chemokines, and adhesion molecules in the blood and tissues, which are associated with the macrophage polarization, and hyperbaric treatment may exert therapeutic effects on decompression related diseases via regulating these factors. Thus, we hypothesize that the polarization of circulating and/or resident macrophages is involved in the pathogenesis of decompression induced lung injury.
Collapse
Affiliation(s)
- Cui-Hong Han
- Department of Pathology, the First Hospital of Jining City, Jining City, Shandong Province, China
| | - Pei-Xi Zhang
- Department of Cardiothoracic Surgery, the First Hospital of Jining City, Jining City, Shandong Province, China
| | - Wen-Wu Liu
- Department of Diving and Hyperbaric Medicine, Secondary Military Medical University, Shanghai, China
| |
Collapse
|
25
|
Dyskova T, Gallo J, Kriegova E. The Role of the Chemokine System in Tissue Response to Prosthetic By-products Leading to Periprosthetic Osteolysis and Aseptic Loosening. Front Immunol 2017; 8:1026. [PMID: 28883822 PMCID: PMC5573717 DOI: 10.3389/fimmu.2017.01026] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/08/2017] [Indexed: 12/27/2022] Open
Abstract
Millions of total joint replacements are performed annually worldwide, and the number is increasing every year. The overall proportion of patients achieving a successful outcome is about 80–90% in a 10–20-years time horizon postoperatively, periprosthetic osteolysis (PPOL) and aseptic loosening (AL) being the most frequent reasons for knee and hip implant failure and reoperations. The chemokine system (chemokine receptors and chemokines) is crucially involved in the inflammatory and osteolytic processes leading to PPOL/AL. Thus, the modulation of the interactions within the chemokine system may influence the extent of PPOL. Indeed, recent studies in murine models reported that (i) blocking the CCR2–CCL2 or CXCR2–CXCL2 axis or (ii) activation of the CXCR4–CXCL12 axis attenuate the osteolysis of artificial joints. Importantly, chemokines, inhibitory mutant chemokines, antagonists of chemokine receptors, or neutralizing antibodies to the chemokine system attached to or incorporated into the implant surface may influence the tissue responses and mitigate PPOL, thus increasing prosthesis longevity. This review summarizes the current state of the art of the knowledge of the chemokine system in human PPOL/AL. Furthermore, the potential for attenuating cell trafficking to the bone–implant interface and influencing tissue responses through modulation of the chemokine system is delineated. Additionally, the prospects of using immunoregenerative biomaterials (including chemokines) for the prevention of failed implants are discussed. Finally, this review highlights the need for a more sophisticated understanding of implant debris-induced changes in the chemokine system to mitigate this response effectively.
Collapse
Affiliation(s)
- Tereza Dyskova
- Faculty of Medicine and Dentistry, Department of Immunology, Palacky University Olomouc, Olomouc, Czechia
| | - Jiri Gallo
- Faculty of Medicine and Dentistry, Department of Orthopaedics, Palacky University Olomouc, University Hospital Olomouc, Olomouc, Czechia
| | - Eva Kriegova
- Faculty of Medicine and Dentistry, Department of Immunology, Palacky University Olomouc, Olomouc, Czechia
| |
Collapse
|
26
|
Liu Y, Gunsten SP, Sultan DH, Luehmann HP, Zhao Y, Blackwell TS, Bollermann-Nowlis Z, Pan JH, Byers DE, Atkinson JJ, Kreisel D, Holtzman MJ, Gropler RJ, Combadiere C, Brody SL. PET-based Imaging of Chemokine Receptor 2 in Experimental and Disease-related Lung Inflammation. Radiology 2017; 283:758-768. [PMID: 28045644 PMCID: PMC5452886 DOI: 10.1148/radiol.2016161409] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Purpose To characterize a chemokine receptor type 2 (CCR2)-binding peptide adapted for use as a positron emission tomography (PET) radiotracer for noninvasive detection of lung inflammation in a mouse model of lung injury and in human tissues from subjects with lung disease. Materials and Methods The study was approved by institutional animal and human studies committees. Informed consent was obtained from patients. A 7-amino acid CCR2 binding peptide (extracellular loop 1 inverso [ECL1i]) was conjugated to tetraazacyclododecane tetraacetic acid (DOTA) and labeled with copper 64 (64Cu) or fluorescent dye. Lung inflammation was induced with intratracheal administration of lipopolysaccharide (LPS) in wild-type (n = 19) and CCR2-deficient (n = 4) mice, and these mice were compared with wild-type mice given control saline (n = 5) by using PET performed after intravenous injection of 64Cu-DOTA-ECL1i. Lung immune cells and those binding fluorescently labeled ECL1i in vivo were detected with flow cytometry. Lung inflammation in tissue from subjects with nondiseased lungs donated for lung transplantation (n = 11) and those with chronic obstructive pulmonary disease (COPD) who were undergoing lung transplantation (n = 16) was evaluated for CCR2 with immunostaining and autoradiography (n = 6, COPD) with 64Cu-DOTA-ECL1i. Groups were compared with analysis of variance, the Mann-Whitney U test, or the t test. Results Signal on PET images obtained in mouse lungs after injury with LPS was significantly greater than that in the saline control group (mean = 4.43% of injected dose [ID] per gram of tissue vs 0.99% of injected dose per gram of tissue; P < .001). PET signal was significantly diminished with blocking studies using nonradiolabeled ECL1i in excess (mean = 0.63% ID per gram of tissue; P < .001) and in CCR2-deficient mice (mean = 0.39% ID per gram of tissue; P < .001). The ECL1i signal was associated with an elevated level of mouse lung monocytes. COPD lung tissue displayed significantly elevated CCR2 levels compared with nondiseased tissue (median = 12.8% vs 1.2% cells per sample; P = .002), which was detected with 64Cu-DOTA-ECL1i by using autoradiography. Conclusion 64Cu-DOTA-ECL1i is a promising tool for PET-based detection of CCR2-directed inflammation in an animal model and in human tissues as a step toward clinical translation. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Yongjian Liu
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Sean P. Gunsten
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Deborah H. Sultan
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Hannah P. Luehmann
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Yongfeng Zhao
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - T. Scott Blackwell
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Zachary Bollermann-Nowlis
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Jie-hong Pan
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Derek E. Byers
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Jeffrey J. Atkinson
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Daniel Kreisel
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Michael J. Holtzman
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Robert J. Gropler
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Christophe Combadiere
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| | - Steven L. Brody
- From the Mallinckrodt Institute of Radiology (Y.L., D.H.S., H.P.L., Y.Z., R.J.G., S.L.B.) and Departments of Medicine (S.P.G., T.S.B., Z.B.N., J.H.P., D.E.B., J.J.A., M.J.H., R.J.G., S.L.B.), Surgery (D.K.), Pathology and Immunology (D.K.), and Cell Biology (M.J.H.), Washington University School of Medicine, 660 S Euclid Ave, Box 8052, St Louis, MO 63110; and Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Faculté de Médecine Pitié-Salpêtrière, Paris INSERM, Paris, France (C.C.)
| |
Collapse
|
27
|
CCR9 Is a Key Regulator of Early Phases of Allergic Airway Inflammation. Mediators Inflamm 2016; 2016:3635809. [PMID: 27795621 PMCID: PMC5067335 DOI: 10.1155/2016/3635809] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/07/2016] [Indexed: 11/24/2022] Open
Abstract
Airway inflammation is the most common hallmark of allergic asthma. Chemokine receptors involved in leukocyte recruitment are closely related to the pathology in asthma. CCR9 has been described as a homeostatic and inflammatory chemokine receptor, but its role and that of its ligand CCL25 during lung inflammation remain unknown. To investigate the role of CCR9 as a modulator of airway inflammation, we established an OVA-induced allergic inflammation model in CCR9-deficient mice. Here, we report the expression of CCR9 and CCL25 as early as 6 hours post-OVA challenge in eosinophils and T-lymphocytes. Moreover, in challenged CCR9-deficient mice, cell recruitment was impaired at peribronchial and perivenular levels. OVA-administration in CCR9-deficient mice leads to a less inflammatory cell recruitment, which modifies the expression of IL-10, CCL11, and CCL25 at 24 hours after OVA challenge. In contrast, the secretion of IL-4 and IL-5 was not affected in CCR9-deficient mice compared to WT mice. These results demonstrate for the first time that CCR9 and CCL25 expressions are induced in the early stages of airway inflammation and they have an important role modulating eosinophils and lymphocytes recruitment at the first stages of inflammatory process, suggesting that they might be a potential target to regulate inflammation in asthma.
Collapse
|
28
|
Liu Y, Li W, Luehmann HP, Zhao Y, Detering L, Sultan DE, Hsiao HM, Krupnick AS, Gelman AE, Combadiere C, Gropler RJ, Brody SL, Kreisel D. Noninvasive Imaging of CCR2 + Cells in Ischemia-Reperfusion Injury After Lung Transplantation. Am J Transplant 2016; 16:3016-3023. [PMID: 27273836 PMCID: PMC5143208 DOI: 10.1111/ajt.13907] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/27/2016] [Accepted: 05/29/2016] [Indexed: 01/25/2023]
Abstract
Ischemia-reperfusion injury-mediated primary graft dysfunction substantially hampers short- and long-term outcomes after lung transplantation. This condition continues to be diagnosed based on oxygen exchange parameters as well as radiological appearance, and therapeutic strategies are mostly supportive in nature. Identifying patients who may benefit from targeted therapy would therefore be highly desirable. Here, we show that C-C chemokine receptor type 2 (CCR2) expression in murine lung transplant recipients promotes monocyte infiltration into pulmonary grafts and mediates graft dysfunction. We have developed new positron emission tomography imaging agents using a CCR2 binding peptide, ECLi1, that can be used to monitor inflammatory responses after organ transplantation. Both 64 Cu-radiolabeled ECL1i peptide radiotracer (64 Cu-DOTA-ECL1i) and ECL1i-conjugated gold nanoclusters doped with 64 Cu (64 CuAuNCs-ECL1i) showed specific detection of CCR2, which is upregulated during ischemia-reperfusion injury after lung transplantation. Due to its fast pharmacokinetics, 64 Cu-DOTA-ECL1i functioned efficiently for rapid and serial imaging of CCR2. The multivalent 64 CuAuNCs-ECL1i with extended pharmacokinetics is favored for long-term CCR2 detection and potential targeted theranostics. This imaging may be applicable for diagnostic and therapeutic purposes for many immune-mediated diseases.
Collapse
Affiliation(s)
- Yongjian Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA,Correspondence to: Yongjian Liu, Ph.D., Assistant Professor of Radiology, 510 S. Kingshighway Blvd, Campus Box 8225, Washington University School of Medicine, St. Louis, MO 63110, Tel: (314) 362-8431, Fax: (314) 362-9940, or Daniel Kreisel, M.D., Ph.D., Professor of Surgery, Pathology & Immunology, Campus Box 8234, 660 South Euclid Avenue, Washington University School of Medicine, St. Louis, MO 63110, Tel: (314) 362-6021, Fax: (314) 367-8459,
| | - Wenjun Li
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Hannah P. Luehmann
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yongfeng Zhao
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lisa Detering
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Deborah E. Sultan
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hsi-Min Hsiao
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexander S. Krupnick
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA,Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew E. Gelman
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA,Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Christophe Combadiere
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Boulevard de l’Hôpital, F-75013 Paris, France
| | - Robert J. Gropler
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Steven L. Brody
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA,Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA,Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA,Correspondence to: Yongjian Liu, Ph.D., Assistant Professor of Radiology, 510 S. Kingshighway Blvd, Campus Box 8225, Washington University School of Medicine, St. Louis, MO 63110, Tel: (314) 362-8431, Fax: (314) 362-9940, or Daniel Kreisel, M.D., Ph.D., Professor of Surgery, Pathology & Immunology, Campus Box 8234, 660 South Euclid Avenue, Washington University School of Medicine, St. Louis, MO 63110, Tel: (314) 362-6021, Fax: (314) 367-8459,
| |
Collapse
|
29
|
Polverino F, Seys LJM, Bracke KR, Owen CA. B cells in chronic obstructive pulmonary disease: moving to center stage. Am J Physiol Lung Cell Mol Physiol 2016; 311:L687-L695. [PMID: 27542809 DOI: 10.1152/ajplung.00304.2016] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 08/15/2016] [Indexed: 12/22/2022] Open
Abstract
Chronic inflammatory responses in the lungs contribute to the development and progression of chronic obstructive pulmonary disease (COPD). Although research studies focused initially on the contributions of the innate immune system to the pathogenesis of COPD, more recent studies have implicated adaptive immune responses in COPD. In particular, studies have demonstrated increases in B cell counts and increases in the number and size of B cell-rich lymphoid follicles in COPD lungs that correlate directly with COPD severity. There are also increases in lung levels of mediators that promote B cell maturation, activation, and survival in COPD patients. B cell products such as autoantibodies directed against lung cells, components of cells, and extracellular matrix proteins are also present in COPD lungs. These autoantibodies may contribute to lung inflammation and injury in COPD patients, in part, by forming immune complexes that activate complement components. Studies of B cell-deficient mice and human COPD patients have linked B cells most strongly to the emphysema phenotype. However, B cells have protective activities during acute exacerbations of COPD by promoting adaptive immune responses that contribute to host defense against pathogens. This review outlines the evidence that links B cells and B cell-rich lymphoid follicles to the pathogenesis of COPD and the mechanisms involved. It also reviews the potential and limitations of B cells as therapeutic targets to slow the progression of human COPD.
Collapse
Affiliation(s)
- Francesca Polverino
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; COPD Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico; University of Parma, Parma, Italy; and
| | - Leen J M Seys
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; COPD Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico;
| |
Collapse
|
30
|
Development of therapeutic antibodies to G protein-coupled receptors and ion channels: Opportunities, challenges and their therapeutic potential in respiratory diseases. Pharmacol Ther 2016; 169:113-123. [PMID: 27153991 DOI: 10.1016/j.pharmthera.2016.04.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The development of recombinant antibody therapeutics continues to be a significant area of growth in the pharmaceutical industry with almost 50 approved monoclonal antibodies on the market in the US and Europe. Therapeutic drug targets such as soluble cytokines, growth factors and single transmembrane spanning receptors have been successfully targeted by recombinant monoclonal antibodies and the development of new product candidates continues. Despite this growth, however, certain classes of important disease targets have remained intractable to therapeutic antibodies due to the complexity of the target molecules. These complex target molecules include G protein-coupled receptors and ion channels which represent a large target class for therapeutic intervention with monoclonal antibodies. Although these targets have typically been addressed by small molecule approaches, the exquisite specificity of antibodies provides a significant opportunity to provide selective modulation of these important regulators of cell function. Given this opportunity, a significant effort has been applied to address the challenges of targeting these complex molecules and a number of targets are linked to the pathophysiology of respiratory diseases. In this review, we provide a summary of the importance of GPCRs and ion channels involved in respiratory disease and discuss advantages offered by antibodies as therapeutics at these targets. We highlight some recent GPCRs and ion channels linked to respiratory disease mechanisms and describe in detail recent progress made in the strategies for discovery of functional antibodies against challenging membrane protein targets such as GPCRs and ion channels.
Collapse
|
31
|
Shelfoon C, Shariff S, Traves SL, Kooi C, Leigh R, Proud D. Chemokine release from human rhinovirus-infected airway epithelial cells promotes fibroblast migration. J Allergy Clin Immunol 2016; 138:114-122.e4. [PMID: 26883463 DOI: 10.1016/j.jaci.2015.12.1308] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 11/05/2015] [Accepted: 12/05/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Thickening of the lamina reticularis, a feature of remodeling in the asthmatic airways, is now known to be present in young children who wheeze. Human rhinovirus (HRV) infection is a common trigger for childhood wheezing, which is a risk factor for subsequent asthma development. We hypothesized that HRV-infected epithelial cells release chemoattractants to recruit fibroblasts that could potentially contribute to thickening of the lamina reticularis. OBJECTIVE We sought to investigate whether conditioned medium from HRV-infected epithelial cells can trigger directed migration of fibroblasts. METHODS Human bronchial epithelial cells were exposed to medium alone or infected with HRV-16. Conditioned medium from both conditions were tested as chemoattractants for human bronchial fibroblasts in the xCELLigence cell migration apparatus. RESULTS HRV-conditioned medium was chemotactic for fibroblasts. Treatment of fibroblasts with pertussis toxin, an inhibitor of Gαi-coupled receptors, prevented their migration. Production of epithelial chemoattractants required HRV replication. Multiplex analysis of epithelial supernatants identified CXCL10, CXCL8, and CCL5 as Gαi-coupled receptor agonists of potential interest. Subsequent analysis confirmed that fibroblasts express CXCR3 and CXCR1 receptors and that CXCL10 and, to a lesser extent, CXCL8, but not CCL5, are major contributors to fibroblast migration caused by HRV-conditioned medium. CONCLUSION CXCL10 and CXCL8 produced from HRV-infected epithelial cells are chemotactic for fibroblasts. This raises the possibility that repeated HRV infections in childhood could contribute to the initiation and progression of airway remodeling in asthmatic patients by recruiting fibroblasts that produce matrix proteins and thicken the lamina reticularis.
Collapse
Affiliation(s)
- Christopher Shelfoon
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sami Shariff
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Suzanne L Traves
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Cora Kooi
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Richard Leigh
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Medicine, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - David Proud
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
32
|
|