1
|
Heibati B, Renz H, Lacy P. Wildfire and wood smoke effects on human airway epithelial cells: A scoping review. ENVIRONMENTAL RESEARCH 2025; 272:121153. [PMID: 39986423 DOI: 10.1016/j.envres.2025.121153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/29/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Wildfires, which occur naturally but are increasingly intensified by climate change, release a complex mixture of organic and inorganic pollutants. These emissions have significant public health implications, contributing to increased morbidity and mortality. Epidemiological and clinical studies have consistently shown that exposure to wildfire smoke exacerbates respiratory conditions such as chronic obstructive pulmonary disease (COPD) and asthma. There are many epidemiological studies of the potential impact of smoke on human health; however, there are remarkably few in vitro studies, and an investigation of the underlying mechanisms of wildfire and wood smoke exposure on airway epithelial cells is required to better understand their toxicity and significance. OBJECTIVES This scoping review aimed to critically examine studies on the association between wildfire and wood smoke exposure and airway epithelial cell responses. METHODS We conducted a systematic search of relevant studies that used a combination of keywords related to wood smoke, wildfire, and epithelial cells and were published up to May 2024. Studies were retrieved from MEDLINE, PubMed, Google Scholar, and Web of Science. RESULTS Twenty-three studies fulfilled our inclusion criteria and were included. This review highlights inflammation, oxidative stress, and cytotoxicity as key impacts of wildfire and wood smoke on airway epithelial cells, causing lung damage. More studies are needed to understand these effects and guide prevention strategies. DISCUSSION This scoping review underscores the need for further research to better understand the complex biological endpoints associated with exposure to wildfire/wood smoke, informing strategies to mitigate health effects, ultimately improving health and well-being of population exposed to wildfire/wood smoke.
Collapse
Affiliation(s)
- Behzad Heibati
- Alberta Respiratory Centre (ARC), Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Harald Renz
- Institute of Laboratory Medicine, Member of the German Center for Lung Research (DZL) and the Lung Center of the Universities of Giessen and Marburg (UGMLC), Philipps University Marburg, Marburg, Germany; Kilimanjaro Christian Medical University College (KCMUCo), Moshi, Tanzania
| | - Paige Lacy
- Alberta Respiratory Centre (ARC), Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
2
|
Li YJ, Geng WL, Li CC, Wu JH, Gao F, Wang Y. Progress of CCL20-CCR6 in the airways: a promising new therapeutic target. J Inflamm (Lond) 2024; 21:54. [PMID: 39731176 PMCID: PMC11681768 DOI: 10.1186/s12950-024-00427-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024] Open
Abstract
The chemokine CCL20, a small cytokine that belongs to the C-C chemokine family, interacts with its homologous receptor CCR6, which is expressed on wide range of cell types. According to current research, the CCL20-CCR6 has been established as acritical player in a diverse range of inflammatory, oncogenic, and autoimmune diseases. Within the respiratory system, CCL20-CCR6 demonstrates heightened expression in conditions such as allergic asthma, chronic airway inflammation, non-small cell lung cancer (NSCLC), chronic obstructive pulmonary disease (COPD), and other respiratory diseases, which is conducive to the inflammatory mediators recruitment and tumor microenvironment remodeling. Numerous studies have demonstrated that therapeutic interventions targeting CCL20 and CCR6, including antibodies and antagonists, have the potential to mitigate disease progression. Despite the promising research prospects surrounding the CCL20-CCR6 chemokine axis, the precise mechanisms underlying its action in respiratory diseases remain largely elusive. In this review, we delve into the potential roles of the CCL20-CCR6 axis within the respiratory system by synthesizing and analyzing current research findings. Our objective is to provide a comprehensive understanding of the CCL20-CCR6 axis and its implications for respiratory health and disease. And we aspire to propel research endeavors in this domain and furnish valuable insights for the development of future therapeutic strategies.
Collapse
Affiliation(s)
- Ya -Jing Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Wan-Li Geng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Chen-Chen Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Jia-Hao Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Fei Gao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Yong Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China.
| |
Collapse
|
3
|
Tian X, Wang S, Zhang C, Prakash YS, Vassallo R. Blocking IL-23 Signaling Mitigates Cigarette Smoke-Induced Murine Emphysema. ENVIRONMENTAL TOXICOLOGY 2024; 39:5334-5346. [PMID: 39221838 PMCID: PMC11567802 DOI: 10.1002/tox.24405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 05/15/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024]
Abstract
Inflammatory cell infiltration is a characteristic feature of COPD and correlates directly with the severity of the disease. Interleukin-23 (IL-23) is a pro-inflammatory cytokine that regulates Th-17 inflammation, which mediates many pathophysiological events in COPD. The primary goal of this study was to determine the role of IL-23 as a mediator of key pathologic processes in cigarette smoke-induced COPD. In this study, we report an increase in IL23 gene expression in the lung biopsies of COPD patients compared to controls and identified a positive correlation between IL23 gene expression and disease severity. In a cigarette smoke-induced murine emphysema model, the suppression of IL-23 with a monoclonal blocking antibody reduced the severity of cigarette smoke-induced murine emphysema. Mechanistically, the suppression of IL-23 was associated with a reduction in immune cell infiltration, oxidative stress injury, and apoptosis, suggesting a role for IL-23 as an essential immune mediator of the inflammatory processes in the pathogenesis of CS-induced emphysema.
Collapse
Affiliation(s)
- Xue Tian
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, 55905, USA
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Shaohua Wang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, 55905, USA
| | - Chujie Zhang
- Department of Cardiology, Xi-Jing Hospital, Fourth Military Medical University, Xi’an, 710000, China
| | - YS Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Robert Vassallo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, 55905, USA
| |
Collapse
|
4
|
Jia L, Li N, Abdelaal TRM, Guo N, IJsselsteijn ME, van Unen V, Lindelauf C, Jiang Q, Xiao Y, Pascutti MF, Hiemstra PS, Koning F, Stolk J, Khedoe PPSJ. High-Dimensional Mass Cytometry Reveals Emphysema-associated Changes in the Pulmonary Immune System. Am J Respir Crit Care Med 2024; 210:1002-1016. [PMID: 38536165 DOI: 10.1164/rccm.202303-0442oc] [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: 03/13/2023] [Accepted: 03/27/2024] [Indexed: 10/16/2024] Open
Abstract
Rationale: Chronic inflammation plays an important role in alveolar tissue damage in emphysema, but the underlying immune alterations and cellular interactions are incompletely understood. Objectives: To explore disease-specific pulmonary immune cell alterations and cellular interactions in emphysema. Methods: We used single-cell mass cytometry (CyTOF) to compare the immune compartment in alveolar tissue from 15 patients with severe emphysema and 5 control subjects. Imaging mass cytometry (IMC) was applied to identify altered cell-cell interactions in alveolar tissue from patients with emphysema (n = 12) compared with control subjects (n = 8). Measurements and Main Results: We observed higher percentages of central memory CD4 T cells in combination with lower proportions of effector memory CD4 T cells in emphysema. In addition, proportions of cytotoxic central memory CD8 T cells and CD127+CD27+CD69- T cells were higher in emphysema, the latter potentially reflecting an influx of circulating lymphocytes into the lungs. Central memory CD8 T cells, isolated from alveolar tissue from patients with emphysema, exhibited an IFN-γ response upon anti-CD3 and anti-CD28 activation. Proportions of CD1c+ dendritic cells, expressing migratory and costimulatory markers, were higher in emphysema. Importantly, IMC enabled us to visualize increased spatial colocalization of CD1c+ dendritic cells and CD8 T cells in emphysema in situ. Conclusions: Using CyTOF, we characterized the alterations of the immune cell signature in alveolar tissue from patients with chronic obstructive pulmonary disease stage III or IV emphysema versus control lung tissue. These data contribute to a better understanding of the pathogenesis of emphysema and highlight the feasibility of interrogating the immune cell signature using CyTOF and IMC in human lung tissue. Clinical trial registered with www.clinicaltrials.gov (NCT04918706).
Collapse
Affiliation(s)
- Li Jia
- Department of Immunology
- Department of Pulmonology, PulmoScience Lab, Leiden University Medical Center, Leiden, the Netherlands
| | - Na Li
- Department of Immunology
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory of Zoonosis Research of the Ministry of Education, Institute of Zoonosis and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Tamim R M Abdelaal
- Department of Radiology
- Systems and Biomedical Engineering Department, Faculty of Engineering, Cairo University, Giza, Egypt; and
- Pattern Recognition and Bioinformatics, Delft University of Technology, Delft, the Netherlands
| | | | | | | | | | | | | | | | - Pieter S Hiemstra
- Department of Pulmonology, PulmoScience Lab, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Jan Stolk
- Department of Pulmonology, PulmoScience Lab, Leiden University Medical Center, Leiden, the Netherlands
| | - P Padmini S J Khedoe
- Department of Pulmonology, PulmoScience Lab, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
5
|
Gutierrez-Chavez C, Aperrigue-Lira S, Ortiz-Saavedra B, Paz I. Chemokine receptors in COVID-19 infection. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 388:53-94. [PMID: 39260938 DOI: 10.1016/bs.ircmb.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Chemokine receptors play diverse roles in the immune response against pathogens by recruiting innate and adaptive immune cells to sites of infection. However, their involvement could also be detrimental, causing tissue damage and exacerbating respiratory diseases by triggering histological alterations such as fibrosis and remodeling. This chapter reviews the role of chemokine receptors in the immune defense against SARS-CoV-2 infection. In COVID-19, CXCR3 is expressed mainly in T cells, and its upregulation is related to an increase in SARS-CoV-2-specific antibodies but also to COVID-19 severity. CCR5 is a key player in T-cell recruitment, and its suppression leads to reduced inflammation and viremia levels. Conversely, CXCR6 is implicated in the aberrant migration of memory T cells within airways. On the other hand, increased CCR4+ cells in the blood and decreased CCR4+ cells in lung cells are associated with severe COVID-19. Additionally, CCR2 is associated with an increase in macrophage recruitment to lung tissues. Elevated levels of CXCR1 and CXCR2, which are predominantly expressed in neutrophils, are associated with the severity of the disease, and finally, the expression of CX3CR1 in cytotoxic T lymphocytes affects the retention of these cells in lung tissues, thereby impacting the severity of COVID-19. Despite the efforts of many clinical trials to find effective therapies for COVID-19 using chemokine receptor inhibitors, no conclusive results have been found due to the small number of patients, redundancy, and co-expression of chemokine receptors by immune cells, which explains the difficulty in finding a single therapeutic target or effective treatment.
Collapse
Affiliation(s)
| | - Shalom Aperrigue-Lira
- Universidad Nacional de San Agustín de Arequipa, Arequipa, Peru; Grupo de Investigación en Inmunología-GII, UNSA, Arequipa, Peru
| | - Brando Ortiz-Saavedra
- Universidad Nacional de San Agustín de Arequipa, Arequipa, Peru; Grupo de Investigación en Inmunología-GII, UNSA, Arequipa, Peru
| | - Irmia Paz
- Universidad Nacional de San Agustín de Arequipa, Arequipa, Peru.
| |
Collapse
|
6
|
Mengistu DT, Curtis JL, Freeman CM. A model of dysregulated crosstalk between dendritic, natural killer, and regulatory T cells in chronic obstructive pulmonary disease. Trends Immunol 2024; 45:428-441. [PMID: 38763820 PMCID: PMC11315412 DOI: 10.1016/j.it.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/21/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by infiltration of the airways and lung parenchyma by inflammatory cells. Lung pathology results from the cumulative effect of complex and aberrant interactions between multiple cell types. However, three cell types, natural killer cells (NK), dendritic cells (DCs), and regulatory T cells (Tregs), are understudied and underappreciated. We propose that their mutual interactions significantly contribute to the development of COPD. Here, we highlight recent advances in NK, DC, and Treg biology with relevance to COPD, discuss their pairwise bidirectional interactions, and identify knowledge gaps that must be bridged to develop novel therapies. Understanding their interactions will be crucial for therapeutic use of autologous Treg, an approach proving effective in other diseases with immune components.
Collapse
Affiliation(s)
- Dawit T Mengistu
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Jeffrey L Curtis
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA; Pulmonary & Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, USA; Pulmonary and Critical Care Medicine Section, VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Christine M Freeman
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA; Pulmonary & Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, USA; Research Service, VA Ann Arbor Healthcare System, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
Tesfaigzi Y, Curtis JL, Petrache I, Polverino F, Kheradmand F, Adcock IM, Rennard SI. Does Chronic Obstructive Pulmonary Disease Originate from Different Cell Types? Am J Respir Cell Mol Biol 2023; 69:500-507. [PMID: 37584669 PMCID: PMC10633838 DOI: 10.1165/rcmb.2023-0175ps] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/16/2023] [Indexed: 08/17/2023] Open
Abstract
The onset of chronic obstructive pulmonary disease (COPD) is heterogeneous, and current approaches to define distinct disease phenotypes are lacking. In addition to clinical methodologies, subtyping COPD has also been challenged by the reliance on human lung samples from late-stage diseases. Different COPD phenotypes may be initiated from the susceptibility of different cell types to cigarette smoke, environmental pollution, and infections at early stages that ultimately converge at later stages in airway remodeling and destruction of the alveoli when the disease is diagnosed. This perspective provides discussion points on how studies to date define different cell types of the lung that can initiate COPD pathogenesis, focusing on the susceptibility of macrophages, T and B cells, mast cells, dendritic cells, endothelial cells, and airway epithelial cells. Additional cell types, including fibroblasts, smooth muscle cells, neuronal cells, and other rare cell types not covered here, may also play a role in orchestrating COPD. Here, we discuss current knowledge gaps, such as which cell types drive distinct disease phenotypes and/or stages of the disease and which cells are primarily affected by the genetic variants identified by whole genome-wide association studies. Applying new technologies that interrogate the functional role of a specific cell type or a combination of cell types as well as single-cell transcriptomics and proteomic approaches are creating new opportunities to understand and clarify the pathophysiology and thereby the clinical heterogeneity of COPD.
Collapse
Affiliation(s)
- Yohannes Tesfaigzi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jeffrey L. Curtis
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | - Irina Petrache
- Division of Pulmonary Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado
- University of Colorado, Denver, Colorado
| | - Francesca Polverino
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, Baylor University, Houston, Texas
| | - Farrah Kheradmand
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, Baylor University, Houston, Texas
| | - Ian M. Adcock
- Department of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Stephen I. Rennard
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
8
|
Nguyen HO, Tiberio L, Facchinetti F, Ripari G, Violi V, Villetti G, Salvi V, Bosisio D. Modulation of Human Dendritic Cell Functions by Phosphodiesterase-4 Inhibitors: Potential Relevance for the Treatment of Respiratory Diseases. Pharmaceutics 2023; 15:2254. [PMID: 37765223 PMCID: PMC10535230 DOI: 10.3390/pharmaceutics15092254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Inhibitors of phosphodiesterase-4 (PDE4) are small-molecule drugs that, by increasing the intracellular levels of cAMP in immune cells, elicit a broad spectrum of anti-inflammatory effects. As such, PDE4 inhibitors are actively studied as therapeutic options in a variety of human diseases characterized by an underlying inflammatory pathogenesis. Dendritic cells (DCs) are checkpoints of the inflammatory and immune responses, being responsible for both activation and dampening depending on their activation status. This review shows evidence that PDE4 inhibitors modulate inflammatory DC activation by decreasing the secretion of inflammatory and Th1/Th17-polarizing cytokines, although preserving the expression of costimulatory molecules and the CD4+ T cell-activating potential. In addition, DCs activated in the presence of PDE4 inhibitors induce a preferential Th2 skewing of effector T cells, retain the secretion of Th2-attracting chemokines and increase the production of T cell regulatory mediators, such as IDO1, TSP-1, VEGF-A and Amphiregulin. Finally, PDE4 inhibitors selectively induce the expression of the surface molecule CD141/Thrombomodulin/BDCA-3. The result of such fine-tuning is immunomodulatory DCs that are distinct from those induced by classical anti-inflammatory drugs, such as corticosteroids. The possible implications for the treatment of respiratory disorders (such as COPD, asthma and COVID-19) by PDE4 inhibitors will be discussed.
Collapse
Affiliation(s)
- Hoang Oanh Nguyen
- ImmunoConcEpT, CNRS UMR 5164, University of Bordeaux, 33000 Bordeaux, France;
| | - Laura Tiberio
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (L.T.); (G.R.); (V.V.)
| | - Fabrizio Facchinetti
- Department of Experimental Pharmacology and Translational Science, Corporate Pre-Clinical R&D, Chiesi Farmaceutici S.p.A., 43122 Parma, Italy; (F.F.); (G.V.)
| | - Giulia Ripari
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (L.T.); (G.R.); (V.V.)
| | - Valentina Violi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (L.T.); (G.R.); (V.V.)
| | - Gino Villetti
- Department of Experimental Pharmacology and Translational Science, Corporate Pre-Clinical R&D, Chiesi Farmaceutici S.p.A., 43122 Parma, Italy; (F.F.); (G.V.)
| | - Valentina Salvi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (L.T.); (G.R.); (V.V.)
| | - Daniela Bosisio
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (L.T.); (G.R.); (V.V.)
| |
Collapse
|
9
|
Raby KL, Michaeloudes C, Tonkin J, Chung KF, Bhavsar PK. Mechanisms of airway epithelial injury and abnormal repair in asthma and COPD. Front Immunol 2023; 14:1201658. [PMID: 37520564 PMCID: PMC10374037 DOI: 10.3389/fimmu.2023.1201658] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023] Open
Abstract
The airway epithelium comprises of different cell types and acts as a physical barrier preventing pathogens, including inhaled particles and microbes, from entering the lungs. Goblet cells and submucosal glands produce mucus that traps pathogens, which are expelled from the respiratory tract by ciliated cells. Basal cells act as progenitor cells, differentiating into different epithelial cell types, to maintain homeostasis following injury. Adherens and tight junctions between cells maintain the epithelial barrier function and regulate the movement of molecules across it. In this review we discuss how abnormal epithelial structure and function, caused by chronic injury and abnormal repair, drives airway disease and specifically asthma and chronic obstructive pulmonary disease (COPD). In both diseases, inhaled allergens, pollutants and microbes disrupt junctional complexes and promote cell death, impairing the barrier function and leading to increased penetration of pathogens and a constant airway immune response. In asthma, the inflammatory response precipitates the epithelial injury and drives abnormal basal cell differentiation. This leads to reduced ciliated cells, goblet cell hyperplasia and increased epithelial mesenchymal transition, which contribute to impaired mucociliary clearance and airway remodelling. In COPD, chronic oxidative stress and inflammation trigger premature epithelial cell senescence, which contributes to loss of epithelial integrity and airway inflammation and remodelling. Increased numbers of basal cells showing deregulated differentiation, contributes to ciliary dysfunction and mucous hyperproduction in COPD airways. Defective antioxidant, antiviral and damage repair mechanisms, possibly due to genetic or epigenetic factors, may confer susceptibility to airway epithelial dysfunction in these diseases. The current evidence suggests that a constant cycle of injury and abnormal repair of the epithelium drives chronic airway inflammation and remodelling in asthma and COPD. Mechanistic understanding of injury susceptibility and damage response may lead to improved therapies for these diseases.
Collapse
Affiliation(s)
- Katie Louise Raby
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - James Tonkin
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospital, London, United Kingdom
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospital, London, United Kingdom
| | - Pankaj Kumar Bhavsar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospital, London, United Kingdom
| |
Collapse
|
10
|
Kayongo A, Nyiro B, Siddharthan T, Kirenga B, Checkley W, Lutaakome Joloba M, Ellner J, Salgame P. Mechanisms of lung damage in tuberculosis: implications for chronic obstructive pulmonary disease. Front Cell Infect Microbiol 2023; 13:1146571. [PMID: 37415827 PMCID: PMC10320222 DOI: 10.3389/fcimb.2023.1146571] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/05/2023] [Indexed: 07/08/2023] Open
Abstract
Pulmonary tuberculosis is increasingly recognized as a risk factor for COPD. Severe lung function impairment has been reported in post-TB patients. Despite increasing evidence to support the association between TB and COPD, only a few studies describe the immunological basis of COPD among TB patients following successful treatment completion. In this review, we draw on well-elaborated Mycobacterium tuberculosis-induced immune mechanisms in the lungs to highlight shared mechanisms for COPD pathogenesis in the setting of tuberculosis disease. We further examine how such mechanisms could be exploited to guide COPD therapeutics.
Collapse
Affiliation(s)
- Alex Kayongo
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - Brian Nyiro
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Trishul Siddharthan
- Division of Pulmonary and Critical Care Medicine, University of Miami, Miami, FL, United States
| | - Bruce Kirenga
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - William Checkley
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States
- Center for Global Non-Communicable Disease Research and Training, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Moses Lutaakome Joloba
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - Jerrold Ellner
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Padmini Salgame
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
11
|
Bazzan E, Casara A, Radu CM, Tinè M, Biondini D, Faccioli E, Pezzuto F, Bernardinello N, Conti M, Balestro E, Calabrese F, Simioni P, Rea F, Turato G, Spagnolo P, Cosio MG, Saetta M. Macrophages-derived Factor XIII links coagulation to inflammation in COPD. Front Immunol 2023; 14:1131292. [PMID: 37180121 PMCID: PMC10166842 DOI: 10.3389/fimmu.2023.1131292] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 04/05/2023] [Indexed: 05/15/2023] Open
Abstract
Background The local, extravascular, activation of the coagulation system in response to injury is a key factor mediating the resulting inflammatory response. Coagulation Factor XIIIA (FXIIIA) found in alveolar macrophages (AM) and dendritic cells (DC), by influencing fibrin stability, might be an inflammatory modifier in COPD. Aims To study the expression of FXIIIA in AM and Langerin+DC (DC-1) and their relation to the inflammatory response and disease progression in COPD. Methods In 47 surgical lungs, 36 from smokers (22 COPD and 14 no-COPD) and 11 from non-smokers we quantified by immunohistochemistry FXIIIA expression in AM and DC-1 along with numbers of CD8+Tcells and CXCR3 expression in lung parenchyma and airways. Lung function was measured prior to surgery. Results The percentage of AM expressing FXIII (%FXIII+AM) was higher in COPD than no-COPD and non-smokers. DC-1 expressed FXIIIA and their numbers were higher in COPD than no-COPD and non-smokers. DC-1 positively correlated with %FXIII+AM (r=0.43; p<0.018). CD8+Tcells, which were higher in COPD than in no-COPD, were correlated with DC-1 (p<0.01) and %FXIII+AM. CXCR3+ cells were increased in COPD and correlated with %FXIII+AM (p<0.05). Both %FXIII+AM (r=-0.6; p=0.001) and DC-1 (r=-0.7; p=0.001) correlated inversely with FEV1. Conclusion FXIIIA, an important link between the extravascular coagulation cascade and inflammatory response, is significantly expressed in alveolar macrophages and dendritic cells of smokers with COPD, suggesting that it could play an important role in the adaptive inflammatory reaction characteristic of the disease.
Collapse
Affiliation(s)
- Erica Bazzan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Alvise Casara
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | | | - Mariaenrica Tinè
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Davide Biondini
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | - Eleonora Faccioli
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Nicol Bernardinello
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Maria Conti
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Elisabetta Balestro
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Paolo Simioni
- Department of Medicine, University of Padova, Padova, Italy
| | - Federico Rea
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Graziella Turato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Paolo Spagnolo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Manuel G. Cosio
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Meakins-Christie Laboratories, Respiratory Division, McGill University, Montreal, QC, Canada
| | - Marina Saetta
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| |
Collapse
|
12
|
Gómez-Melero S, Caballero-Villarraso J. CCR6 as a Potential Target for Therapeutic Antibodies for the Treatment of Inflammatory Diseases. Antibodies (Basel) 2023; 12:30. [PMID: 37092451 PMCID: PMC10123731 DOI: 10.3390/antib12020030] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/12/2023] [Accepted: 04/18/2023] [Indexed: 04/25/2023] Open
Abstract
The CC chemokine receptor 6 (CCR6) is a G protein-coupled receptor (GPCR) involved in a wide range of biological processes. When CCR6 binds to its sole ligand CCL20, a signaling network is produced. This pathway is implicated in mechanisms related to many diseases, such as cancer, psoriasis, multiple sclerosis, HIV infection or rheumatoid arthritis. The CCR6/CCL20 axis plays a fundamental role in immune homeostasis and activation. Th17 cells express the CCR6 receptor and inflammatory cytokines, including IL-17, IL-21 and IL-22, which are involved in the spread of inflammatory response. The CCL20/CCR6 mechanism plays a crucial role in the recruitment of these pro-inflammatory cells to local tissues. To date, there are no drugs against CCR6 approved, and the development of small molecules against CCR6 is complicated due to the difficulty in screenings. This review highlights the potential as a therapeutic target of the CCR6 receptor in numerous diseases and the importance of the development of antibodies against CCR6 that could be a promising alternative to small molecules in the treatment of CCR6/CCL20 axis-related pathologies.
Collapse
Affiliation(s)
- Sara Gómez-Melero
- Maimonides Biomedical Research Institute of Cordoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain
| | - Javier Caballero-Villarraso
- Maimonides Biomedical Research Institute of Cordoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain
| |
Collapse
|
13
|
Fuzo CA, Martins RB, Fraga‐Silva TFC, Amstalden MK, Canassa De Leo T, Souza JP, Lima TM, Faccioli LH, Okamoto DN, Juliano MA, França SC, Juliano L, Bonato VLD, Arruda E, Dias‐Baruffi M. Celastrol: A lead compound that inhibits SARS-CoV-2 replication, the activity of viral and human cysteine proteases, and virus-induced IL-6 secretion. Drug Dev Res 2022; 83:1623-1640. [PMID: 35989498 PMCID: PMC9539158 DOI: 10.1002/ddr.21982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 11/21/2022]
Abstract
The global emergence of coronavirus disease 2019 (COVID-19) has caused substantial human casualties. Clinical manifestations of this disease vary from asymptomatic to lethal, and the symptomatic form can be associated with cytokine storm and hyperinflammation. In face of the urgent demand for effective drugs to treat COVID-19, we have searched for candidate compounds using in silico approach followed by experimental validation. Here we identified celastrol, a pentacyclic triterpene isolated from Tripterygium wilfordii Hook F, as one of the best compounds out of 39 drug candidates. Celastrol reverted the gene expression signature from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected cells and irreversibly inhibited the recombinant forms of the viral and human cysteine proteases involved in virus invasion, such as Mpro (main protease), PLpro (papain-like protease), and recombinant human cathepsin L. Celastrol suppressed SARS-CoV-2 replication in human and monkey cell lines and decreased interleukin-6 (IL-6) secretion in the SARS-CoV-2-infected human cell line. Celastrol acted in a concentration-dependent manner, with undetectable signs of cytotoxicity, and inhibited in vitro replication of the parental and SARS-CoV-2 variant. Therefore, celastrol is a promising lead compound to develop new drug candidates to face COVID-19 due to its ability to suppress SARS-CoV-2 replication and IL-6 production in infected cells.
Collapse
Affiliation(s)
- Carlos A. Fuzo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão PretoUniversidade de São PauloRibeirão PretoSão PauloBrazil
| | - Ronaldo B. Martins
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão PretoUniversidade de São PauloRibeirão PretoSão PauloBrazil
| | - Thais F. C. Fraga‐Silva
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão PretoUniversidade de São PauloRibeirão PretoSão PauloBrazil
| | - Martin K. Amstalden
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão PretoUniversidade de São PauloRibeirão PretoSão PauloBrazil
| | - Thais Canassa De Leo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão PretoUniversidade de São PauloRibeirão PretoSão PauloBrazil
| | - Juliano P. Souza
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão PretoUniversidade de São PauloRibeirão PretoSão PauloBrazil
| | - Thais M. Lima
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão PretoUniversidade de São PauloRibeirão PretoSão PauloBrazil
| | - Lucia H. Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão PretoUniversidade de São PauloRibeirão PretoSão PauloBrazil
| | - Débora Noma Okamoto
- Departamento de Biofísica, Escola Paulista de MedicinaUniversidade Federal de São PauloSão PauloSão PauloBrazil
| | - Maria Aparecida Juliano
- Departamento de Biofísica, Escola Paulista de MedicinaUniversidade Federal de São PauloSão PauloSão PauloBrazil
| | - Suzelei C. França
- Unidade de BiotecnologiaUniversidade de Ribeirão PretoRibeirão PretoSão PauloBrazil
| | - Luiz Juliano
- Departamento de Biofísica, Escola Paulista de MedicinaUniversidade Federal de São PauloSão PauloSão PauloBrazil
| | - Vania L. D. Bonato
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão PretoUniversidade de São PauloRibeirão PretoSão PauloBrazil
| | - Eurico Arruda
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão PretoUniversidade de São PauloRibeirão PretoSão PauloBrazil
| | - Marcelo Dias‐Baruffi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão PretoUniversidade de São PauloRibeirão PretoSão PauloBrazil
| |
Collapse
|
14
|
Madrid DMDC, Gu W, Artiaga BL, Yang G, Loeb J, Hawkins IK, Castleman WL, Lednicky JA, Richt JA, Driver JP. Comparison of oseltamivir and α-galactosylceramide for reducing disease and transmission in pigs infected with 2009 H1N1 pandemic influenza virus. Front Vet Sci 2022; 9:999507. [PMID: 36337191 PMCID: PMC9635317 DOI: 10.3389/fvets.2022.999507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/30/2022] [Indexed: 11/24/2022] Open
Abstract
Influenza virus infections are a major cause of respiratory disease in humans. Neuraminidase inhibitors (NAIs) are the primary antiviral medication used to treat ongoing influenza infections. However, NAIs are not always effective for controlling virus shedding and lung inflammation. Other concerns are the emergence of NAI-resistant virus strains and the risk of side effects, which are occasionally severe. Consequently, additional anti-influenza therapies to replace or combine with NAIs are desirable. Here, we compared the efficacy of the NAI oseltamivir with the invariant natural killer T (iNKT) cell superagonist, α-galactosylceramide (α-GalCer), which induces innate immune responses that inhibit influenza virus replication in mouse models. We show that oseltamivir reduced lung lesions and lowered virus titers in the upper respiratory tract of pigs infected with A/California/04/2009 (CA04) pandemic H1N1pdm09. It also reduced virus transmission to influenza-naïve contact pigs. In contrast, α-GalCer had no impact on virus replication, lung disease, or virus transmission, even when used in combination with oseltamivir. This is significant as iNKT-cell therapy has been studied as an approach for treating humans with influenza.
Collapse
Affiliation(s)
| | - Weihong Gu
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Bianca L. Artiaga
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Guan Yang
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Julia Loeb
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, United States
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Ian K. Hawkins
- Department of Comparative, Diagnostic, and Population Medicine, University of Florida, Gainesville, FL, United States
| | - William L. Castleman
- Department of Comparative, Diagnostic, and Population Medicine, University of Florida, Gainesville, FL, United States
| | - John A. Lednicky
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, United States
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Jürgen A. Richt
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - John P. Driver
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States
| |
Collapse
|
15
|
The Role of CD40, CD86, and Glutathione S-Transferase Omega 1 in the Pathogenesis of Chronic Obstructive Pulmonary Disease. Can Respir J 2022; 2022:6810745. [PMID: 36051533 PMCID: PMC9427324 DOI: 10.1155/2022/6810745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/16/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022] Open
Abstract
Objective. The aim of the study was to explore the relevance of CD40, CD86, and GSTO1 with the pathogenesis of COPD. Methods. Patients with acute exacerbation of COPD were contrasted with the healthy and nonsmoking ones and smoking but without COPD ones. The changes of CD40, CD86, and GSTO1 in the peripheral blood, collected from different groups, were detected by flow cytometry and western blotting, respectively. Results. Compared with the nonsmoking group and smoking but without the COPD group, the expression of CD40 and CD86 of the patients with COPD increased significantly, but the expression of GSTO1 decreased. CD40 and CD86 were negatively correlated with FEV1%, while GSTO1 was positively correlated with FEV1% and negatively correlated with CD40 and CD86. Conclusion. CD40, CD86, and GSTO1 may play a role in the pathogenesis of COPD, and they are related to the severity of COPD and the degree of changes in the lung function.
Collapse
|
16
|
Yan L, Wu X, Wu P, Su B, Xiong Y, Rao Y, Chen X, Huang W, Cui T. Increased expression of Clec9A on cDC1s associated with cytotoxic CD8 + T cell response in COPD. Clin Immunol 2022; 242:109082. [PMID: 35901921 DOI: 10.1016/j.clim.2022.109082] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 07/12/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022]
Abstract
Although C-type lectin domain family 9A (Clec9A) on conventional type 1 dendritic cells (cDC1s) plays a critical role in cytotoxic CD8+ T cell response in cancers and viral infections, its role in chronic obstructive pulmonary disease (COPD) is unknown. We measured the expression of Clec9A in sera, BALF, and PBMCs from controls and COPD patients. The percentages of Clec9A+ DCs and cytotoxic CD8+ T cells in the BALF were determined by flow cytometry between patients with COPD and non-obstructive chronic bronchitis (NOCB). Compared with healthy individuals, the serum levels of Clec9A were increased at different stages of COPD patients, and the mRNA and protein levels of Clec9A were both increased in COPD patients at GOLD stages III-IV. The percentage of Clec9A+ DCs was also increased in the BALF of COPD patients compared with NOCB patients. Moreover, enhanced Clec9A+ DCs recruitment was positively correlated with cytotoxic CD8+ T cell response in the BALF of COPD patients. This study suggests that Clec9A+ DCs participate in the CD8+ T cell-mediated chronic airway inflammation in COPD.
Collapse
Affiliation(s)
- Li Yan
- Department of Laboratory Medicine, Wuhan No.1 Hospital, Wuhan, Hubei, China
| | - Xiaojie Wu
- Department of Respiratory and Critical Care Medicine, Wuhan No.1 Hospital, Wuhan, Hubei, China
| | - Ping Wu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bintao Su
- Department of Laboratory Medicine, Wuhan No.1 Hospital, Wuhan, Hubei, China
| | - Yin Xiong
- Department of Laboratory Medicine, Wuhan No.1 Hospital, Wuhan, Hubei, China
| | - Yahua Rao
- Department of Laboratory Medicine, Wuhan No.1 Hospital, Wuhan, Hubei, China
| | - Xing Chen
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Huang
- Department of Laboratory Medicine, Wuhan No.1 Hospital, Wuhan, Hubei, China.
| | - Tianpen Cui
- Department of Laboratory Medicine, Wuhan No.1 Hospital, Wuhan, Hubei, China.
| |
Collapse
|
17
|
Blaskovic S, Donati Y, Ruchonnet-Metrailler I, Avila Y, Schittny D, Schlepütz CM, Schittny JC, Barazzone-Argiroffo C. Early life exposure to nicotine modifies lung gene response after elastase-induced emphysema. Respir Res 2022; 23:44. [PMID: 35241086 PMCID: PMC8895880 DOI: 10.1186/s12931-022-01956-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 02/13/2022] [Indexed: 11/30/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is among the top 5 causes of mortality in the world and can develop as a consequence of genetic and/or environmental factors. Current efforts are focused on identifying early life insults and how these contribute to COPD development. In line with this, our study focuses on the influence of early life nicotine exposure and its potential impact on (a) lung pulmonary functions, and (b) elastase-induced emphysema in adulthood.
Methods To address this hypothesis, we developed a model of 2 hits, delivered at different time points: mouse pups were first exposed to nicotine/placebo in utero and during lactation, and then subsequently received elastase/placebo at the age of 11 weeks. The effect of nicotine pretreatment and elastase instillation was assessed by (a) measurement of pulmonary function at post-elastase day (ped) 21, and (b) transcriptomic profiling at ped3 and 21, and complementary protein determination. Statistical significance was determined by 3- and 2-way ANOVA for pulmonary functions, and RNAseq results were analyzed using the R project.
Results We did not observe any impact of nicotine pre- and early post-natal exposure compared to control samples on lung pulmonary functions in adulthood, as measured by FLEXIVENT technology. After elastase instillation, substantial lung damage was detected by x-ray tomography and was accompanied by loss in body weight at ped3 as well as an increase in cell numbers, inflammatory markers in BAL and lung volume at ped21. Lung functions showed a decrease in elastance and an increase in deep inflation volume and pressure volume (pv) loop area in animals with emphysema at ped21. Nicotine had no effect on elastance and deep inflation volume, but did affect the pv loop area in animals with emphysema at ped21. Extensive transcriptomic changes were induced by elastase at ped3 both in the nicotine-pretreated and the control samples, with several pathways common to both groups, such as for cell cycle, DNA adhesion and DNA damage. Nicotine pretreatment affected the number of lymphocytes present in BAL after elastase instillation and some of the complement pathway related proteins, arguing for a slight modification of the immune response, as well as changes related to general body metabolism. The majority of elastase-induced transcriptomic changes detected at ped3 had disappeared at ped21. In addition, transcriptomic profiling singled out a common gene pool that was independently activated by nicotine and elastase. Conclusions Our study reports a broad spectrum of transient transcriptomic changes in mouse emphysema and identifies nicotine as influencing the emphysema-associated immune system response. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-01956-4.
Collapse
Affiliation(s)
- Sanja Blaskovic
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, 4 rue Gabrielle-Perret-Gentil, 1211, Genève 14, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Yves Donati
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, 4 rue Gabrielle-Perret-Gentil, 1211, Genève 14, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Isabelle Ruchonnet-Metrailler
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, 4 rue Gabrielle-Perret-Gentil, 1211, Genève 14, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Yannick Avila
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, 4 rue Gabrielle-Perret-Gentil, 1211, Genève 14, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | | | | | - Constance Barazzone-Argiroffo
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, 4 rue Gabrielle-Perret-Gentil, 1211, Genève 14, Switzerland. .,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
18
|
Fernandes JR, Pinto TNC, Arruda LB, da Silva CCBM, de Carvalho CRF, Pinto RMC, da Silva Duarte AJ, Benard G. Age-associated phenotypic imbalance in TCD4 and TCD8 cell subsets: comparison between healthy aged, smokers, COPD patients and young adults. IMMUNITY & AGEING 2022; 19:9. [PMID: 35164774 PMCID: PMC8842531 DOI: 10.1186/s12979-022-00267-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022]
Abstract
Abstract
Background
COPD is associated with an abnormal lung immune response that leads to tissue damage and remodeling of the lung, but also to systemic effects that compromise immune responses. Cigarette smoking also impacts on innate and adaptative immune responses, exerting dual, pro- and anti-inflammatory effects. Previously, we showed that COPD patients presented accelerated telomere shortening and decreased telomerase activity, while, paradoxically, cigarette-smokers exhibited preserved telomerase activity and slower rate of telomere shortening.
Results
Here, we evaluated the naive, CM, EM and TEMRA subsets of TCD4 and TCD8 cells according to the expression of CCR7/CD45RA. We compared age-matched COPD patients, cigarette-smokers without clinical-laboratory evidence of pulmonary compromise, and healthy individuals. They were additionally compared with a group of young adults. For each subset we analysed the expression of markers associated with late differentiation, senescence and exhaustion (CD27/CD28/CD57/KLRG1/PD1). We show that COPD patients presented a drastically reduced naive cells pool, and, paradoxically, increased fractions of naive cells expressing late differentiation, senescence or exhaustion markers, likely impacting on their immunocompetence. Pronounced phenotypic alterations were also evidenced in their three memory T-cell subsets compared with the other aged and young groups, suggesting an also dysfunctional memory pool. Surprisingly, our smokers showed a profile closer to the Healthy aged than COPD patients. They exhibited the usual age-associated shift of naive to EM TCD4 and TCD8 cells, but not to CM or TEMRA T-cells. Nonetheless, their naive T-cells phenotypes were in general similar to those of the Youngs and Healthy aged, suggesting a rather phenotypically preserved subset, while the memory T-cells exhibited increased proportions of cells with the late-differentiation or senescence/exhaustion markers as in the Healthy aged.
Conclusion
Our study extends previous findings by showing that COPD patients have cells expressing a full range of late differentiated, senescent or exhausted phenotypes encompassing all TCD4 and TCD8 subsets, consistent with a premature immunosenescence phenotype. Surprisingly, the smokers group’s results suggest that moderate to heavy chronic cigarette smoking did not accelerate the pace of immunosenescence as compared with the Healthy aged.
Collapse
|
19
|
Rahman ML, Bassig BA, Dai Y, Hu W, Wong JYY, Blechter B, Hosgood HD, Ren D, Duan H, Niu Y, Xu J, Fu W, Meliefste K, Zhou B, Yang J, Ye M, Jia X, Meng T, Bin P, Silverman DT, Vermeulen R, Rothman N, Zheng Y, Lan Q. Proteomic analysis of serum in workers exposed to diesel engine exhaust. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2022; 63:18-28. [PMID: 34894159 PMCID: PMC11812432 DOI: 10.1002/em.22469] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/25/2021] [Accepted: 12/09/2021] [Indexed: 06/14/2023]
Abstract
Diesel engine exhaust (DEE) is classified as a Group 1 human carcinogen. Using a targeted proteomics approach, we aimed to identify proteins associated with DEE and characterize these markers to understand the mechanisms of DEE-induced carcinogenicity. In this cross-sectional molecular epidemiology study, we measured elemental carbon (EC) using a personal air monitor and quantified 1317 targeted proteins in the serum using the SOMAScan assay (SOMALogic) among 19 diesel exposed factory workers and 19 unexposed controls. We used linear regressions to identify proteins associated with DEE and examined their exposure-response relationship across levels of EC using linear trend tests. We further examined pathway enrichment of DEE-related proteins using MetaCore. Occupational exposure to DEE was associated with altered levels of 22 serum proteins (permutation p < .01). Of these, 13 proteins (CXCL11, HAPLN1, FLT4, CD40LG, PES1, IGHE.IGK..IGL, TNFSF9, PGD, NAGK, CCL25, CCL4L1, PDXK, and PLA2G1B) showed an exposure-response relationship with EC (p trend < .01), with serum levels of all but PLA2G1B declining with increasing air levels of EC. For instance, C-X-C Motif Chemokine Ligand 11 (CXCL11) showed the most significant association with DEE (β = -0.25; permutation p = .00004), where mean serum levels were 4121.1, 2356.7, and 2298.8 relative fluorescent units among the unexposed, lower exposed (median, range : 56.9, 40.2-62.1 μg/m3 EC), and higher exposed (median, range of EC: 72.9, 66.9-107.7 μg/m3 EC) groups, respectively (p trend = .0005). Pathway analysis suggested that these proteins are enriched in pathways related to inflammation and immune regulation. Our study suggests that DEE exposure is associated with altered serum proteins, which play a role in inflammation and immune regulation.
Collapse
Affiliation(s)
- Mohammad L. Rahman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Bryan A. Bassig
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Yufei Dai
- Key laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Wei Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Jason YY. Wong
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Batel Blechter
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - H. Dean Hosgood
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Danzhi Ren
- Chaoyang Center for Disease Control and Prevention, Chaoyang, Liaoning, People’s Republic of China
| | - Huawei Duan
- Key laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Yong Niu
- Key laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Jun Xu
- School of Public Health, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Wei Fu
- Chaoyang Center for Disease Control and Prevention, Chaoyang, Liaoning, People’s Republic of China
| | - Kees Meliefste
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Baosen Zhou
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, People’s Republic of China
| | - Jufang Yang
- Chaoyang Center for Disease Control and Prevention, Chaoyang, Liaoning, People’s Republic of China
| | - Meng Ye
- Key laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Xiaowei Jia
- Key laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Tao Meng
- Key laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Ping Bin
- Key laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Debra T. Silverman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao, People’s Republic of China
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| |
Collapse
|
20
|
Aloufi N, Alluli A, Eidelman DH, Baglole CJ. Aberrant Post-Transcriptional Regulation of Protein Expression in the Development of Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2021; 22:ijms222111963. [PMID: 34769392 PMCID: PMC8584689 DOI: 10.3390/ijms222111963] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is an incurable and prevalent respiratory disorder that is characterized by chronic inflammation and emphysema. COPD is primarily caused by cigarette smoke (CS). CS alters numerous cellular processes, including the post-transcriptional regulation of mRNAs. The identification of RNA-binding proteins (RBPs), microRNAs (miRNAs), and long non-coding RNAs (lncRNAs) as main factors engaged in the regulation of RNA biology opens the door to understanding their role in coordinating physiological cellular processes. Dysregulation of post-transcriptional regulation by foreign particles in CS may lead to the development of diseases such as COPD. Here we review current knowledge about post-transcriptional events that may be involved in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Noof Aloufi
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
- Department of Medical Laboratory Technology, Applied Medical Science, Taibah University, Universities Road, Medina P.O. Box 344, Saudi Arabia
| | - Aeshah Alluli
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
| | - David H. Eidelman
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada;
| | - Carolyn J. Baglole
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada;
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada
- Correspondence:
| |
Collapse
|
21
|
Fung KY, Louis C, Metcalfe RD, Kosasih CC, Wicks IP, Griffin MDW, Putoczki TL. Emerging roles for IL-11 in inflammatory diseases. Cytokine 2021; 149:155750. [PMID: 34689057 DOI: 10.1016/j.cyto.2021.155750] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/16/2022]
Abstract
Interleukin-11 (IL-11) is a cytokine that has been strongly implicated in the pathogenesis of fibrotic diseases and solid malignancies. Elevated IL-11 expression is also associated with several non-malignant inflammatory diseases where its function remains less well-characterized. Here, we summarize current literature surrounding the contribution of IL-11 to the pathogenesis of autoimmune inflammatory diseases, including rheumatoid arthritis, multiple sclerosis, diabetes and systemic sclerosis, as well as other chronic inflammatory conditions such as periodontitis, asthma, chronic obstructive pulmonary disease, psoriasis and colitis.
Collapse
Affiliation(s)
- Ka Yee Fung
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Victoria 3053, Australia.
| | - Cynthia Louis
- Department of Medical Biology, University of Melbourne, Victoria 3053, Australia; Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Victoria 3052, Australia
| | - Riley D Metcalfe
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Technology Institute, University of Melbourne, Victoria 3010, Australia
| | - Clara C Kosasih
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Technology Institute, University of Melbourne, Victoria 3010, Australia
| | - Ian P Wicks
- Department of Medical Biology, University of Melbourne, Victoria 3053, Australia; Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Victoria 3052, Australia; Rheumatology Unit, The Royal Melbourne Hospital, Victoria 3050, Australia
| | - Michael D W Griffin
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Technology Institute, University of Melbourne, Victoria 3010, Australia
| | - Tracy L Putoczki
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Victoria 3053, Australia.
| |
Collapse
|
22
|
Dey S, Eapen MS, Chia C, Gaikwad AV, Wark PAB, Sohal SS. Pathogenesis, clinical features of asthma COPD overlap (ACO), and therapeutic modalities. Am J Physiol Lung Cell Mol Physiol 2021; 322:L64-L83. [PMID: 34668439 DOI: 10.1152/ajplung.00121.2021] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Both asthma and COPD are heterogeneous diseases identified by characteristic symptoms and functional abnormalities, with airway obstruction common in both diseases. Asthma COPD overlap (ACO) does not define a single disease but is a descriptive term for clinical use that includes several overlapping clinical phenotypes of chronic airways disease with different underlying mechanisms. This literature review was initiated to describe published studies, identify gaps in knowledge, and propose future research goals regarding the disease pathology of ACO, especially the airway remodelling changes and inflammation aspects. Airway remodelling occurs in asthma and COPD, but there are differences in the structures affected and the prime anatomic site at which they occur. Reticular basement membrane thickening and cellular infiltration with eosinophils and T-helper (CD4+) lymphocytes are prominent features of asthma. Epithelial squamous metaplasia, airway wall fibrosis, emphysema, bronchoalveolar lavage (BAL) neutrophilia and (CD8+) T-cytotoxic lymphocyte infiltrations in the airway wall are features of COPD. There is no universally accepted definition of ACO, nor are there clearly defined pathological characteristics to differentiate from asthma and COPD. Understanding etiological concepts within the purview of inflammation and airway remodelling changes in ACO would allow better management of these patients.
Collapse
Affiliation(s)
- Surajit Dey
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | - Mathew Suji Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | - Collin Chia
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia.,Department of Respiratory Medicine, Launceston General Hospital, Launceston, Tasmania, Australia
| | - Archana Vijay Gaikwad
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia.,Department of Respiratory and Sleep Medicine John Hunter Hospital, New Lambton Heights, Australia
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| |
Collapse
|
23
|
Cigarette Smoke Specifically Affects Small Airway Epithelial Cell Populations and Triggers the Expansion of Inflammatory and Squamous Differentiation Associated Basal Cells. Int J Mol Sci 2021; 22:ijms22147646. [PMID: 34299265 PMCID: PMC8305830 DOI: 10.3390/ijms22147646] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/24/2022] Open
Abstract
Smoking is a major risk factor for chronic obstructive pulmonary disease (COPD) and causes remodeling of the small airways. However, the exact smoke-induced effects on the different types of small airway epithelial cells (SAECs) are poorly understood. Here, using air–liquid interface (ALI) cultures, single-cell RNA-sequencing reveals previously unrecognized transcriptional heterogeneity within the small airway epithelium and cell type-specific effects upon acute and chronic cigarette smoke exposure. Smoke triggers detoxification and inflammatory responses and aberrantly activates and alters basal cell differentiation. This results in an increase of inflammatory basal-to-secretory cell intermediates and, particularly after chronic smoke exposure, a massive expansion of a rare inflammatory and squamous metaplasia associated KRT6A+ basal cell state and an altered secretory cell landscape. ALI cultures originating from healthy non-smokers and COPD smokers show similar responses to cigarette smoke exposure, although an increased pro-inflammatory profile is conserved in the latter. Taken together, the in vitro models provide high-resolution insights into the smoke-induced remodeling of the small airways resembling the pathological processes in COPD airways. The data may also help to better understand other lung diseases including COVID-19, as the data reflect the smoke-dependent variable induction of SARS-CoV-2 entry factors across SAEC populations.
Collapse
|
24
|
Takeda K, Kim SH, Joetham A, Petrache I, Gelfand EW. Therapeutic benefits of recombinant alpha1-antitrypsin IgG1 Fc-fusion protein in experimental emphysema. Respir Res 2021; 22:207. [PMID: 34271910 PMCID: PMC8283905 DOI: 10.1186/s12931-021-01784-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/24/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Alpha-1 antitrypsin (AAT) is a major serine protease inhibitor. AAT deficiency (AATD) is a genetic disorder characterized by early-onset severe emphysema. In well-selected AATD patients, therapy with plasma-derived AAT (pAAT), "augmentation therapy", provides modest clinical improvement but is perceived as cumbersome with weekly intravenous infusions. Using mouse models of emphysema, we compared the effects of a recombinant AAT-IgG1 Fc-fusion protein (AAT-Fc), which is expected to have a longer half-life following infusion, to those of pAAT. METHODS In an elastase model of emphysema, mice received a single intratracheal instillation of porcine pancreatic elastase (PPE) or human leucocyte elastase (hLE). AAT-Fc, pAAT, or vehicle was administered intraperitoneally 1 day prior to or 3 weeks following elastase instillation. Lung function and histology assessments were performed at 7 and 32 days after elastase instillation. In a cigarette smoke (CS) model of emphysema, mice were exposed to CS daily, 5 days a week, for 6 months and AAT-Fc, pAAT, or vehicle were administered every 10 days during the last 3 months of CS exposure. Assessments were performed 3 days after the last CS exposure. Immune responses to lung elastin peptide (EP) and the effects of AAT-Fc or pAAT treatment on dendritic cell (DC) function were determined ex vivo. RESULTS Both elastase instillation and CS exposure triggered emphysema-like alveolar enlargement, increased lung compliance, and increased markers of inflammation compared to controls. Administration of AAT-Fc either prior to or following elastase instillation or during CS exposure provided greater protection than pAAT against alveolar enlargement, lung dysfunction, and airway inflammation. When challenged ex vivo with EP, spleen mononuclear cells from elastase-exposed mice exhibited dose-dependent production of IFNγ and IL-17, suggesting immune reactivity. In co-culture experiments with splenic CD4+ T cells isolated from elastase-exposed mice, AAT-Fc treatment prior to EP-priming of bone marrow-derived dendritic cells inhibited the production of IFNγ and IL-17. CONCLUSIONS Compared to pAAT, AAT-Fc more effectively prevented or attenuated elastase- and CS-induced models of emphysema. These effects were associated with immunomodulatory effects on DC activity. AAT-Fc may provide a therapeutic option to individuals with AATD- and CS-induced emphysema.
Collapse
Affiliation(s)
- Katsuyuki Takeda
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA.
- Kyoritsu-Onsen Hospital, 1-39-1 Hirano, Kawanishi, 666-0121, Japan.
| | - Soo-Hyun Kim
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
- College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Anthony Joetham
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Irina Petrache
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| |
Collapse
|
25
|
Pathological and clinical relevance of selective recruitment of Langerhans cells in the respiratory bronchioles of smokers. Respir Investig 2021; 59:513-521. [PMID: 33839049 DOI: 10.1016/j.resinv.2021.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Smoking causes an influx of inflammatory cells including Langerhans cells (LCs) into the airways and lung parenchyma, thus inducing histological changes, such as emphysema and fibrosis. We examined the distribution and quantity of Langerhans cells in relation to clinical and pathological findings and explored the association between smoking and accumulation of Langerhans cells in the respiratory bronchioles. METHODS Fifty-three patients who underwent lung resection for primary diseases, including lung cancer, were recruited. Histological and immunohistochemistry analyses were utilized to identify CD1a-positive Langerhans cells in peripheral lung specimens separated from primary lesions. Clinical characteristics, pathological changes, and distribution of CD1a-positive Langerhans cells distribution were assessed. RESULTS Of the 53 patients, 35 were smokers and 18 were non-smokers. The number of Langerhans cells in the respiratory bronchioles was significantly increased in smokers as compared to that in non-smokers (p < 0.001). The number of Langerhans cells in smokers was significantly higher in patients with mild emphysema than in those without emphysema (p < 0.01). The high-LC group showed more frequent smoking-related histological changes, such as respiratory bronchiolitis, parenchymal fibrosis, accumulation of macrophages, and smoking-related interstitial fibrosis, than the low-LC group. However, there were no differences in the smoking indices and pulmonary functions of the groups. CONCLUSIONS Selective accumulation of Langerhans cells in the respiratory bronchioles of smokers may lead to the development of smoking-related pathological changes.
Collapse
|
26
|
Hadzic S, Wu CY, Gredic M, Kojonazarov B, Pak O, Kraut S, Sommer N, Kosanovic D, Grimminger F, Schermuly RT, Seeger W, Bellusci S, Weissmann N. The effect of long-term doxycycline treatment in a mouse model of cigarette smoke-induced emphysema and pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2021; 320:L903-L915. [PMID: 33760647 DOI: 10.1152/ajplung.00048.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of death and a still incurable disease, comprising emphysema and chronic bronchitis. In addition to airflow limitation, patients with COPD can suffer from pulmonary hypertension (PH). Doxycycline, an antibiotic from the tetracycline family, in addition to its pronounced antimicrobial activity, acts as a matrix metalloproteinase (MMP) inhibitor and has anti-inflammatory properties. Furthermore, doxycycline treatment exhibited a beneficial effect in several preclinical cardiovascular disease models. In preclinical research, doxycycline is frequently employed for gene expression modulation in Tet-On/Tet-Off transgenic animal models. Therefore, it is crucial to know whether doxycycline treatment in Tet-On/Tet-Off systems has effects independent of gene expression modulation by such systems. Against this background, we assessed the possible curative effects of long-term doxycycline administration in a mouse model of chronic CS exposure. Animals were exposed to cigarette smoke (CS) for 8 mo and then subsequently treated with doxycycline for additional 3 mo in room air conditions. Doxycycline decreased the expression of MMPs and general pro-inflammatory markers in the lungs from CS-exposed mice. This downregulation was, however, insufficient to ameliorate CS-induced emphysema or PH. Tet-On/Tet-Off induction by doxycycline in such models is a feasible genetic approach to study curative effects at least in established CS-induced emphysema and PH. However, we report several parameters that are influenced by doxycycline and use of a Tet-On/Tet-Off system when evaluating those parameters should be interpreted with caution.
Collapse
Affiliation(s)
- Stefan Hadzic
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Cheng-Yu Wu
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Marija Gredic
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Baktybek Kojonazarov
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany.,Institute for Lung Health (ILH), Justus-Liebig-University, Giessen, Germany
| | - Oleg Pak
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Simone Kraut
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Natascha Sommer
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Djuro Kosanovic
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany.,Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Friedrich Grimminger
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Ralph T Schermuly
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Werner Seeger
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany.,Institute for Lung Health (ILH), Justus-Liebig-University, Giessen, Germany.,Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Saverio Bellusci
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Norbert Weissmann
- Cardiopulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
27
|
Charting Extracellular Transcriptomes in The Human Biofluid RNA Atlas. Cell Rep 2020; 33:108552. [PMID: 33378673 DOI: 10.1016/j.celrep.2020.108552] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/14/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular RNAs present in biofluids have emerged as potential biomarkers for disease. Where most studies focus on blood-derived fluids, other biofluids may be more informative. We present an atlas of messenger, circular, and small RNA transcriptomes of a comprehensive collection of 20 human biofluids. By means of synthetic spike-in controls, we compare RNA content across biofluids, revealing a 10,000-fold difference in concentration. The circular RNA fraction is increased in most biofluids compared to tissues. Each biofluid transcriptome is enriched for RNA molecules derived from specific tissues and cell types. Our atlas enables an informed selection of the most relevant biofluid to monitor particular diseases. To verify the biomarker potential in these biofluids, four validation cohorts representing a broad spectrum of diseases were profiled, revealing numerous differential RNAs between case and control subjects. Spike-normalized data are publicly available in the R2 web portal for further exploration.
Collapse
|
28
|
Olloquequi J. COVID-19 Susceptibility in chronic obstructive pulmonary disease. Eur J Clin Invest 2020; 50:e13382. [PMID: 32780415 PMCID: PMC7435530 DOI: 10.1111/eci.13382] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023]
Abstract
In barely nine months, the pandemic known as COVID-19 has spread over 200 countries, affecting more than 22 million people and causing over than 786 000 deaths. Elderly people and patients with previous comorbidities such as hypertension and diabetes are at an increased risk to suffer a poor prognosis after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Although the same could be expected from patients with chronic obstructive pulmonary disease (COPD), current epidemiological data are conflicting. This could lead to a reduction of precautionary measures in these patients, in the context of a particularly complex global health crisis. Most COPD patients have a long history of smoking or exposure to other harmful particles or gases, capable of impairing pulmonary defences even years after the absence of exposure. Moreover, COPD is characterized by an ongoing immune dysfunction, which affects both pulmonary and systemic cellular and molecular inflammatory mediators. Consequently, increased susceptibility to viral respiratory infections have been reported in COPD, often worsened by bacterial co-infections and leading to serious clinical outcomes. The present paper is an up-to-date review that discusses the available research regarding the implications of coronavirus infection in COPD. Although validation in large studies is still needed, COPD likely increases SARS-CoV-2 susceptibility and increases COVID-19 severity. Hence, specific mechanisms to monitor and assess COPD patients should be addressed in the current pandemic.
Collapse
Affiliation(s)
- Jordi Olloquequi
- Laboratory of Cellular and Molecular PathologyFacultad de Ciencias de la SaludInstituto de Ciencias BiomédicasUniversidad Autónoma de ChileTalcaChile
| |
Collapse
|
29
|
Huang QQ, Tang HHF, Teo SM, Mok D, Ritchie SC, Nath AP, Brozynska M, Salim A, Bakshi A, Holt BJ, Khor CC, Sly PD, Holt PG, Holt KE, Inouye M. Neonatal genetics of gene expression reveal potential origins of autoimmune and allergic disease risk. Nat Commun 2020; 11:3761. [PMID: 32724101 PMCID: PMC7387553 DOI: 10.1038/s41467-020-17477-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic immune-mediated diseases of adulthood often originate in early childhood. To investigate genetic associations between neonatal immunity and disease, we map expression quantitative trait loci (eQTLs) in resting myeloid cells and CD4+ T cells from cord blood samples, as well as in response to lipopolysaccharide (LPS) or phytohemagglutinin (PHA) stimulation, respectively. Cis-eQTLs are largely specific to cell type or stimulation, and 31% and 52% of genes with cis-eQTLs have response eQTLs (reQTLs) in myeloid cells and T cells, respectively. We identified cis regulatory factors acting as mediators of trans effects. There is extensive colocalisation between condition-specific neonatal cis-eQTLs and variants associated with immune-mediated diseases, in particular CTSH had widespread colocalisation across diseases. Mendelian randomisation shows causal neonatal gene expression effects on disease risk for BTN3A2, HLA-C and others. Our study elucidates the genetics of gene expression in neonatal immune cells, and aetiological origins of autoimmune and allergic diseases. Some immune-mediated diseases may originate in early childhood. The authors mapped eQTLs and response eQTLs to various stimuli in neonatal myeloid cells and T cells, and revealed their potential role in immune-mediated diseases using colocalisation and Mendelian randomisation.
Collapse
Affiliation(s)
- Qin Qin Huang
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia. .,Department of Clinical Pathology, University of Melbourne, Parkville, VIC, 3010, Australia. .,Department of Human Genetics, Wellcome Sanger Institute, Cambridge, UK.
| | - Howard H F Tang
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,School of BioSciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Shu Mei Teo
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Danny Mok
- Telethon Kids Institute, The University of Western Australia, Perth, WA, 6009, Australia
| | - Scott C Ritchie
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK.,British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.,National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
| | - Artika P Nath
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Marta Brozynska
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Agus Salim
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.,School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, 3010, Australia.,Melbourne School of Population and Global Health, Carlton, VIC, 3053, Australia
| | - Andrew Bakshi
- Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Barbara J Holt
- Telethon Kids Institute, The University of Western Australia, Perth, WA, 6009, Australia
| | - Chiea Chuen Khor
- Human Genetics, Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, 138672, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Peter D Sly
- Child Health Research Centre, The University of Queensland, Brisbane, QLD, 4101, Australia
| | - Patrick G Holt
- Telethon Kids Institute, The University of Western Australia, Perth, WA, 6009, Australia.,Child Health Research Centre, The University of Queensland, Brisbane, QLD, 4101, Australia
| | - Kathryn E Holt
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,The London School of Hygiene and Tropical Medicine, London, WC1E 7TH, UK
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia. .,Department of Clinical Pathology, University of Melbourne, Parkville, VIC, 3010, Australia. .,Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK. .,British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK. .,National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK. .,The Alan Turing Institute, London, UK. .,British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK. .,Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK.
| |
Collapse
|
30
|
Rao W, Wang S, Duleba M, Niroula S, Goller K, Xie J, Mahalingam R, Neupane R, Liew AA, Vincent M, Okuda K, O'Neal WK, Boucher RC, Dickey BF, Wechsler ME, Ibrahim O, Engelhardt JF, Mertens TCJ, Wang W, Jyothula SSK, Crum CP, Karmouty-Quintana H, Parekh KR, Metersky ML, McKeon FD, Xian W. Regenerative Metaplastic Clones in COPD Lung Drive Inflammation and Fibrosis. Cell 2020; 181:848-864.e18. [PMID: 32298651 PMCID: PMC7294989 DOI: 10.1016/j.cell.2020.03.047] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/26/2019] [Accepted: 03/20/2020] [Indexed: 12/30/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive condition of chronic bronchitis, small airway obstruction, and emphysema that represents a leading cause of death worldwide. While inflammation, fibrosis, mucus hypersecretion, and metaplastic epithelial lesions are hallmarks of this disease, their origins and dependent relationships remain unclear. Here we apply single-cell cloning technologies to lung tissue of patients with and without COPD. Unlike control lungs, which were dominated by normal distal airway progenitor cells, COPD lungs were inundated by three variant progenitors epigenetically committed to distinct metaplastic lesions. When transplanted to immunodeficient mice, these variant clones induced pathology akin to the mucous and squamous metaplasia, neutrophilic inflammation, and fibrosis seen in COPD. Remarkably, similar variants pre-exist as minor constituents of control and fetal lung and conceivably act in normal processes of immune surveillance. However, these same variants likely catalyze the pathologic and progressive features of COPD when expanded to high numbers.
Collapse
Affiliation(s)
- Wei Rao
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | - Shan Wang
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | - Marcin Duleba
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | - Suchan Niroula
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | - Kristina Goller
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | - Jingzhong Xie
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | - Rajasekaran Mahalingam
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | - Rahul Neupane
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | - Audrey-Ann Liew
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | | | - Kenichi Okuda
- Marsico Lung Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Wanda K O'Neal
- Marsico Lung Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Richard C Boucher
- Marsico Lung Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Burton F Dickey
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Omar Ibrahim
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Tinne C J Mertens
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Wei Wang
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Soma S K Jyothula
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Christopher P Crum
- Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02215, USA
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Kalpaj R Parekh
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Surgery, Division of Cardiothoracic Surgery, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Mark L Metersky
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Frank D McKeon
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA.
| | - Wa Xian
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA.
| |
Collapse
|
31
|
Zhao Q, Li Y, Li Y, Ji X, Li H, Wu D, Wei W, Xinchun W. Silencing EPB41 Gene Expression Leads to Cell Cycle Arrest, Migration Inhibition, and Upregulation of Cell Surface Antigen in DC2.4 Cells. Med Sci Monit 2020; 26:e920594. [PMID: 32157074 PMCID: PMC7085237 DOI: 10.12659/msm.920594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Protein 4.1R (EPB41) is the main cytoskeleton component of the erythrocyte membrane and may be involved in cell migration and adhesion. Previous research discovered overexpression of 4.1R in the thymus of patients with myasthenia gravis (MG). The protein 4.1R on dendritic cells may play a pivotal role in MG pathogenesis. This research investigated the effects of small interfering RNA 4.1R-siRNA on cell migration, cell cycle, and surface antigen expression of DC2.4 mouse dendritic cells, thus providing a new direction for the study of MG pathogenesis. MATERIAL AND METHODS Three 4.1R-specific siRNAs were designed, and the expression of 4.1R was detected by real-time PCR at the mRNA level and Western blot analysis at the protein level to select out the most efficient siRNAs. Changes in cell morphology were observed and cell migration ability was analyzed by Transwell assay. Cell cycle and surface antigen were both analyzed by flow cytometry. RESULTS The cell bodies of DC2.4 diminished, the synapses were increased, and protuberance became more obvious after being transfected with 4.1R-siRNA. After knockdown of 4.1R, cell migration ability decreased and the proportion of cells in S phase significantly increased (both P<0.05). The expression levels of MHCII, CD80, and CD86 were all increased in DC2.4 cells (all <0.05). CONCLUSIONS Silencing the expression of 4.1R in dendritic cells resulted in inhibition of migration ability, cell cycle arrest, and increase in surface antigens, which suggest that 4.1R participates in MG autoimmunity.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Rheumatism, Huaihe Hosptial of Henan University, Kaifeng, Henan, China (mainland)
| | - Yongqiang Li
- Biochemisty and Molecular Teaching and Research Office, School of Basic Medical Science, Henan University, Kaifeng, Henan, China (mainland)
| | - Yanhong Li
- Department of General Medicine, The First Affiliated Hospital of Henan University, Kaifeng, Henan, China (mainland)
| | - Xinying Ji
- Department of Medical Microbiology, School of Basic Medical Science, Henan University, Kaifeng, Henan, China (mainland)
| | - Huimin Li
- Human Anatomy Teaching and Research Room, School of Basic Medical Science, Henan University, Kaifeng, Henan, China (mainland)
| | - Dongdong Wu
- Department of Physiology, School of Basic Medical Science, Henan University, Kaifeng, Henan, China (mainland)
| | - Wenqiang Wei
- Department of Medical Microbiology, School of Basic Medical Science, Henan University, Kaifeng, Henan, China (mainland)
| | - Wang Xinchun
- Molecular Biology Laboratory, The First Affiliated Hospital of Henan University, Kaifeng, Henan, China (mainland)
| |
Collapse
|
32
|
Liu H, Osterburg AR, Flury J, Swank Z, McGraw DW, Gupta N, Wikenheiser-Brokamp KA, Kumar A, Tazi A, Inoue Y, Hirose M, McCormack FX, Borchers MT. MAPK mutations and cigarette smoke promote the pathogenesis of pulmonary Langerhans cell histiocytosis. JCI Insight 2020; 5:132048. [PMID: 31961828 DOI: 10.1172/jci.insight.132048] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 01/15/2020] [Indexed: 12/12/2022] Open
Abstract
Pulmonary Langerhans cell histiocytosis (PLCH) is a rare smoking-related lung disease characterized by dendritic cell (DC) accumulation, bronchiolocentric nodule formation, and cystic lung remodeling. Approximately 50% of patients with PLCH harbor somatic BRAF-V600E mutations in cells of the myeloid/monocyte lineage. However, the rarity of the disease and lack of animal models have impeded the study of PLCH pathogenesis. Here, we establish a cigarette smoke-exposed (CS-exposed) BRAF-V600E-mutant mouse model that recapitulates many hallmark characteristics of PLCH. We show that CD11c-targeted expression of BRAF-V600E increases DC responsiveness to stimuli, including the chemokine CCL20, and that mutant cell accumulation in the lungs of CS-exposed mice is due to both increased cellular viability and enhanced recruitment. Moreover, we report that the chemokine CCL7 is secreted from DCs and human peripheral blood monocytes in a BRAF-V600E-dependent manner, suggesting a possible mechanism for recruitment of cells known to dominate PLCH lesions. Inflammatory lesions and airspace dilation in BRAF-V600E mice in response to CS are attenuated by transitioning animals to filtered air and treatment with a BRAF-V600E inhibitor, PLX4720. Collectively, this model provides mechanistic insights into the role of myelomonocytic cells and the BRAF-V600E mutation and CS exposure in PLCH pathogenesis and provides a platform to develop biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Huan Liu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Andrew R Osterburg
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jennifer Flury
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Zulma Swank
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Dennis W McGraw
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Nishant Gupta
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Kathryn A Wikenheiser-Brokamp
- Division of Pathology and Laboratory Medicine and.,Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ashish Kumar
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Abdellatif Tazi
- INSERM UMR-S 976, University Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | - Yoshikazu Inoue
- National Hospital Organization Kinki-Chuo Chest Medical Center, Osaka, Japan
| | - Masaki Hirose
- National Hospital Organization Kinki-Chuo Chest Medical Center, Osaka, Japan
| | - Francis X McCormack
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Michael T Borchers
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
33
|
Osei ET, Brandsma CA, Timens W, Heijink IH, Hackett TL. Current perspectives on the role of interleukin-1 signalling in the pathogenesis of asthma and COPD. Eur Respir J 2020; 55:13993003.00563-2019. [PMID: 31727692 DOI: 10.1183/13993003.00563-2019] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 11/05/2019] [Indexed: 12/12/2022]
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) cause significant morbidity and mortality worldwide. In the context of disease pathogenesis, both asthma and COPD involve chronic inflammation of the lung and are characterised by the abnormal release of inflammatory cytokines, dysregulated immune cell activity and remodelling of the airways. To date, current treatments still only manage symptoms and do not reverse the primary disease processes. In recent work, interleukin (IL)-1α and IL-1β have been suggested to play important roles in both asthma and COPD. In this review, we summarise overwhelming pre-clinical evidence for dysregulated signalling of IL-1α and IL-1β contributing to disease pathogenesis and discuss the paradox of IL-1 therapeutic studies in asthma and COPD. This is particularly important given recent completed and ongoing clinical trials with IL-1 biologics that have had varying degrees of failure and success as therapeutics for disease modification in asthma and COPD.
Collapse
Affiliation(s)
- Emmanuel T Osei
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada .,Dept of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Corry-Anke Brandsma
- Dept of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute of Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wim Timens
- Dept of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute of Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Irene H Heijink
- Dept of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute of Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Dept of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tillie-Louise Hackett
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.,Dept of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
34
|
Hoffmann RF, Jonker MR, Brandenburg SM, de Bruin HG, Ten Hacken NHT, van Oosterhout AJM, Heijink IH. Mitochondrial dysfunction increases pro-inflammatory cytokine production and impairs repair and corticosteroid responsiveness in lung epithelium. Sci Rep 2019; 9:15047. [PMID: 31636329 PMCID: PMC6803636 DOI: 10.1038/s41598-019-51517-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 09/25/2019] [Indexed: 12/13/2022] Open
Abstract
COPD is characterized by chronic lung inflammation and irreversible lung tissue damage. Inhaled noxious gases, including cigarette smoke, are the major risk factor for COPD. Inhaled smoke first encounters the epithelial lining of the lungs, causing oxidative stress and mitochondrial dysfunction. We investigated whether a mitochondrial defect may contribute to increased lung epithelial pro-inflammatory responses, impaired epithelial repair and reduced corticosteroid sensitivity as observed in COPD. We used wild-type alveolar epithelial cells A549 and mitochondrial DNA-depleted A549 cells (A549 Rho-0) and studied pro-inflammatory responses using (multiplex) ELISA as well as epithelial barrier function and repair (real-time impedance measurements), in the presence and absence of the inhaled corticosteroid budesonide. We observed that A549 Rho-0 cells secrete higher levels of pro-inflammatory cytokines than wild-type A549 cells and display impaired repair upon wounding. Budesonide strongly suppressed the production of neutrophil attractant CXCL8, and promoted epithelial integrity in A549 wild-type cells, while A549 Rho-0 cells displayed reduced corticosteroid sensitivity compared to wild-type cells. The reduced corticosteroid responsiveness may be mediated by glycolytic reprogramming, specifically glycolysis-associated PI3K signaling, as PI3K inhibitor LY294002 restored the sensitivity of CXCL8 secretion to corticosteroids in A549 Rho-0 cells. In conclusion, mitochondrial defects may lead to increased lung epithelial pro-inflammatory responses, reduced epithelial repair and reduced corticosteroid responsiveness in lung epithelium, thus potentially contributing to the pathogenesis of COPD.
Collapse
Affiliation(s)
- R F Hoffmann
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - M R Jonker
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - S M Brandenburg
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - H G de Bruin
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - N H T Ten Hacken
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, The Netherlands
| | - A J M van Oosterhout
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - I H Heijink
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands.
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands.
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, The Netherlands.
| |
Collapse
|
35
|
Wilke CA, Chadwick MM, Chan PR, Moore BB, Zhou X. Stem cell transplantation impairs dendritic cell trafficking and herpesvirus immunity. JCI Insight 2019; 4:130210. [PMID: 31479426 PMCID: PMC6795288 DOI: 10.1172/jci.insight.130210] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/21/2019] [Indexed: 12/25/2022] Open
Abstract
Long-term survivors after hematopoietic stem cell transplantation are at high risk of infection, which accounts for one-third of all deaths related to stem cell transplantation. Little is known about the cause of inferior host defense after immune cell reconstitution. Here, we exploited a murine syngeneic BM transplantation (BMT) model of late infection with murine gammaherpesvirus 68 (MHV-68) to determine the role of conventional DC (cDC) trafficking in adaptive immunity in BMT mice. After infection, the expression of chemokine Ccl21 in the lung is reduced and the migration of cDCs into lung draining lymph nodes (dLNs) is impaired in BMT mice, limiting the opportunity for cDCs to prime Th cells in the dLNs. While cDC subsets are redundant in priming Th1 cells, Notch2 functions in cDC2s are required for priming increased Th17 responses in BMT mice, and cDC1s can lessen this activity. Importantly, Th17 cells can be primed both in the lungs and dLNs, allowing for increased Th17 responses without optimum cDC trafficking in BMT mice. Taken together, impaired cDC trafficking in BMT mice reduces protective Th1 responses and allows increased pathogenic Th17 responses. Thus, we have revealed a previously unknown mechanism for BMT procedures to cause long-term inferior immune responses to herpes viral infection.
Collapse
Affiliation(s)
- Carol A. Wilke
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Mathew M. Chadwick
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Paul R. Chan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Bethany B. Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Xiaofeng Zhou
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
36
|
Patel VI, Metcalf JP. Airway Macrophage and Dendritic Cell Subsets in the Resting Human Lung. Crit Rev Immunol 2019; 38:303-331. [PMID: 30806245 DOI: 10.1615/critrevimmunol.2018026459] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Dendritic cells (DCs) and macrophages (MΦs) are antigen-presenting phagocytic cells found in many peripheral tissues of the human body, including the blood, lymph nodes, skin, and lung. They are vital to maintaining steady-state respiration in the human lung based on their ability to clear airways while also directing tolerogenic or inflammatory responses based on specific stimuli. Over the past three decades, studies have determined that there are multiple subsets of these two general cell types that exist in the airways and interstitium. Identifying these numerous subsets has proven challenging, especially with the unique microenvironments present in the lung. Cells found in the vasculature are not the same subsets found in the skin or the lung, as demonstrated by surface marker expression. By transcriptional profiling, these subsets show similarities but also major differences. Primary human lung cells and/ or tissues are difficult to acquire, particularly in a healthy condition. Additionally, surface marker screening and transcriptional profiling are continually identifying new DC and MΦ subsets. While the overall field is moving forward, we emphasize that more attention needs to focus on replicating the steady-state microenvironment of the lung to reveal the physiological functions of these subsets.
Collapse
Affiliation(s)
- Vineet Indrajit Patel
- Pulmonary and Critical Care Division of the Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jordan Patrick Metcalf
- Pulmonary and Critical Care Division of the Department of Medicine and Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
37
|
Chemokines in COPD: From Implication to Therapeutic Use. Int J Mol Sci 2019; 20:ijms20112785. [PMID: 31174392 PMCID: PMC6600384 DOI: 10.3390/ijms20112785] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023] Open
Abstract
: Chronic Obstructive Pulmonary Disease (COPD) represents the 3rd leading cause of death in the world. The underlying pathophysiological mechanisms have been the focus of extensive research in the past. The lung has a complex architecture, where structural cells interact continuously with immune cells that infiltrate into the pulmonary tissue. Both types of cells express chemokines and chemokine receptors, making them sensitive to modifications of concentration gradients. Cigarette smoke exposure and recurrent exacerbations, directly and indirectly, impact the expression of chemokines and chemokine receptors. Here, we provide an overview of the evidence regarding chemokines involvement in COPD, and we hypothesize that a dysregulation of this tightly regulated system is critical in COPD evolution, both at a stable state and during exacerbations. Targeting chemokines and chemokine receptors could be highly attractive as a mean to control both chronic inflammation and bronchial remodeling. We present a special focus on the CXCL8-CXCR1/2, CXCL9/10/11-CXCR3, CCL2-CCR2, and CXCL12-CXCR4 axes that seem particularly involved in the disease pathophysiology.
Collapse
|
38
|
Su YC, Jalalvand F, Thegerström J, Riesbeck K. The Interplay Between Immune Response and Bacterial Infection in COPD: Focus Upon Non-typeable Haemophilus influenzae. Front Immunol 2018; 9:2530. [PMID: 30455693 PMCID: PMC6230626 DOI: 10.3389/fimmu.2018.02530] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a debilitating respiratory disease and one of the leading causes of morbidity and mortality worldwide. It is characterized by persistent respiratory symptoms and airflow limitation due to abnormalities in the lower airway following consistent exposure to noxious particles or gases. Acute exacerbations of COPD (AECOPD) are characterized by increased cough, purulent sputum production, and dyspnea. The AECOPD is mostly associated with infection caused by common cold viruses or bacteria, or co-infections. Chronic and persistent infection by non-typeable Haemophilus influenzae (NTHi), a Gram-negative coccobacillus, contributes to almost half of the infective exacerbations caused by bacteria. This is supported by reports that NTHi is commonly isolated in the sputum from COPD patients during exacerbations. Persistent colonization of NTHi in the lower airway requires a plethora of phenotypic adaptation and virulent mechanisms that are developed over time to cope with changing environmental pressures in the airway such as host immuno-inflammatory response. Chronic inhalation of noxious irritants in COPD causes a changed balance in the lung microbiome, abnormal inflammatory response, and an impaired airway immune system. These conditions significantly provide an opportunistic platform for NTHi colonization and infection resulting in a "vicious circle." Episodes of large inflammation as the consequences of multiple interactions between airway immune cells and NTHi, accumulatively contribute to COPD exacerbations and may result in worsening of the clinical status. In this review, we discuss in detail the interplay and crosstalk between airway immune residents and NTHi, and their effect in AECOPD for better understanding of NTHi pathogenesis in COPD patients.
Collapse
Affiliation(s)
- Yu-Ching Su
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Farshid Jalalvand
- Department of Biology, Centre for Bacterial Stress Response and Persistence, University of Copenhagen, Copenhagen, Denmark
| | - John Thegerström
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Kristian Riesbeck
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
39
|
Finch DK, Stolberg VR, Ferguson J, Alikaj H, Kady MR, Richmond BW, Polosukhin VV, Blackwell TS, McCloskey L, Curtis JL, Freeman CM. Lung Dendritic Cells Drive Natural Killer Cytotoxicity in Chronic Obstructive Pulmonary Disease via IL-15Rα. Am J Respir Crit Care Med 2018; 198:1140-1150. [PMID: 29676596 PMCID: PMC6221577 DOI: 10.1164/rccm.201712-2513oc] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/19/2018] [Indexed: 02/02/2023] Open
Abstract
RATIONALE Lung natural killer cells (NKs) kill a greater percentage of autologous lung parenchymal cells in chronic obstructive pulmonary disease (COPD) than in nonobstructed smokers. To become cytotoxic, NKs require priming, typically by dendritic cells (DCs), but whether priming occurs in the lungs in COPD is unknown. METHODS We used lung tissue and in some cases peripheral blood from patients undergoing clinically indicated resections to determine in vitro killing of CD326+ lung epithelial cells by isolated lung CD56+ NKs. We also measured the cytotoxicity of unprimed blood NKs after preincubation with lung DCs. To investigate mechanisms of DC-mediated priming, we used murine models of COPD induced by cigarette smoke (CS) exposure or by polymeric immunoglobulin receptor (pIgR) deficiency, and blocked IL-15Rα (IL-15 receptor α subunit) trans-presentation by genetic and antibody approaches. RESULTS Human lung NKs killed isolated autologous lung epithelial cells; cytotoxicity was increased (P = 0.0001) in COPD, relative to smokers without obstruction. Similarly, increased lung NK cytotoxicity compared with control subjects was observed in CS-exposed mice and pIgR-/- mice. Blood NKs both from smokers without obstruction and subjects with COPD showed minimal epithelial cell killing, but in COPD, preincubation with lung DCs increased cytotoxicity. NKs were primed by CS-exposed murine DCs in vitro and in vivo. Inhibiting IL-15Rα trans-presentation eliminated NK priming both by murine CS-exposed DCs and by lung DCs from subjects with COPD. CONCLUSIONS Heightened NK cytotoxicity against lung epithelial cells in COPD results primarily from lung DC-mediated priming via IL-15 trans-presentation on IL-15Rα. Future studies are required to test whether increased NK cytotoxicity contributes to COPD pathogenesis.
Collapse
Affiliation(s)
- Donna K. Finch
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd., Cambridge, United Kingdom
| | - Valerie R. Stolberg
- Research Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - John Ferguson
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd., Cambridge, United Kingdom
| | - Henrih Alikaj
- Research Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Mohamed R. Kady
- Research Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Bradley W. Richmond
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | | | - Timothy S. Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Cell and Developmental Biology and
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Lisa McCloskey
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Jeffrey L. Curtis
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan; and
- Pulmonary and Critical Care Medicine Section, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Christine M. Freeman
- Research Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan; and
| |
Collapse
|
40
|
Effects of cigarette smoke on immunity, neuroinflammation and multiple sclerosis. J Neuroimmunol 2018; 329:24-34. [PMID: 30361070 DOI: 10.1016/j.jneuroim.2018.10.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 09/30/2018] [Accepted: 10/05/2018] [Indexed: 12/18/2022]
Abstract
Cigarette smoking is the most prominent significant cause of death and morbidity. It is recognised as a risk factor for a number of immune mediated, inflammatory diseases including multiple sclerosis (MS). Here, we review the complex immunological effects of smoking on the immune system, which include enhancement of inflammatory responses with a parallel reduction of some immune defences, resulting in an increased susceptibility to infection and a persistent proinflammatory environment. We discuss the effect of smoking on the susceptibility, clinical course, disability, and mortality in MS, the likely benefits of smoking cessation, and the specific immunological effects of smoking in MS. In conclusion, smoking is an important environmental risk factor for MS occurrence and outcome, and it acts in significant part through immunological mechanisms.
Collapse
|
41
|
Zheng X, Zhang L, Chen J, Gu Y, Xu J, Ouyang Y. Dendritic cells and Th17/Treg ratio play critical roles in pathogenic process of chronic obstructive pulmonary disease. Biomed Pharmacother 2018; 108:1141-1151. [PMID: 30372815 DOI: 10.1016/j.biopha.2018.09.113] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 09/11/2018] [Accepted: 09/19/2018] [Indexed: 12/22/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common disorder of respiratory system. This study aimed to evaluate changes of mature dendritic cells (DCs) and regulatory T cells (Treg) in lung tissues and peripheral blood of COPD patients. For lung tissue analysis, patients were divided into no-smoking and no-COPD (CS-COPD-), smoking and no-COPD (CS + COPD-) and COPD group. For peripheral blood analysis, patients were divided into CS-COPD-, CS + COPD-, stable COPD (SCOPD) and acute exacerbation of COPD (AECOPD) group. Hematoxylin and eosin (HE) staining was used to evaluate inflammation of lung tissues. Immunohistochemistry assay was employed to examine CD80, CCR6, IL-17 A, FoxP3 in lung tissues. DCs and Treg cells were isolated from lung tissues and peripheral blood. Levels of CD80, FoxP3+ Treg, CCR6 and IL-17 A were detected by using flow cytometry. Results showed that FEV%, FVC% and FEV1/FVC were significantly reduced and Bosken scores were remarkably increased in COPD patients compared to non-COPD patients (p < 0.05). CD80 and FoxP3 levels were lower, and CCR6 and IL-17A levels were higher obviously in COPD compared to non-COPD patients (p < 0.05). COPD patients illustrated reduced mDCs levels and enhanced imDCs levels. COPD patients exhibited remarkably higher Th17 levels compared to no-smoking patients (p < 0.05). COPD patients illustrated obviously lower Treg levels and significantly higher Th17/Treg ratio compared to non-smoking patients (p < 0.05). Th17% (Th17/Treg) negatively and Treg% was positively correlated with FEV1%, FEVC%, FEV1/FEVC (p < 0.05). In conclusion, dendritic cells and Th17/Treg ratio play critical roles for pathogenic process of chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Xiangru Zheng
- Department of Respiratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lanying Zhang
- Department of Respiratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jie Chen
- Department of Respiratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yanhui Gu
- Department of Respiratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jingqing Xu
- Tongji Medical College of HUST, Wuhan, China
| | - Yao Ouyang
- Department of Respiratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
42
|
Qiu SL, Kuang LJ, Tang QY, Duan MC, Bai J, He ZY, Zhang JQ, Li MH, Deng JM, Liu GN, Zhong XN. Enhanced activation of circulating plasmacytoid dendritic cells in patients with Chronic Obstructive Pulmonary Disease and experimental smoking-induced emphysema. Clin Immunol 2018; 195:107-118. [DOI: 10.1016/j.clim.2017.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 08/14/2017] [Accepted: 11/07/2017] [Indexed: 12/24/2022]
|
43
|
González-Guerrero C, Morgado-Pascual JL, Cannata-Ortiz P, Ramos-Barron MA, Gómez-Alamillo C, Arias M, Mezzano S, Egido J, Ruiz-Ortega M, Ortiz A, Ramos AM. CCL20 blockade increases the severity of nephrotoxic folic acid-induced acute kidney injury. J Pathol 2018; 246:191-204. [PMID: 29984403 DOI: 10.1002/path.5132] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 06/11/2018] [Accepted: 06/22/2018] [Indexed: 01/19/2023]
Abstract
The chemokine CCL20 activates the CCR6 receptor and has been implicated in the pathogenesis of glomerular injury. However, it is unknown whether it contributes to acute kidney injury (AKI). We identified CCL20 as upregulated in a systems biology strategy combining transcriptomics of kidney tissue from experimental toxic folic acid-induced AKI and from stressed cultured tubular cells and have explored the expression and function of CCL20 in experimental and clinical AKI. CCL20 upregulation was confirmed in three models of kidney injury induced by a folic acid overdose, cisplatin or unilateral ureteral obstruction. In injured kidneys, CCL20 was expressed by tubular, endothelial, and interstitial cells, and was also upregulated in human kidneys with AKI. Urinary CCL20 was increased in human AKI and was associated with severity. The function of CCL20 in nephrotoxic folic acid-induced AKI was assessed by using neutralising anti-CCL20 antibodies or CCR6-deficient mice. CCL20/CCR6 targeting increased the severity of kidney failure and mortality. This was associated with more severe histological injury, nephrocalcinosis, capillary rarefaction, and fibrosis, as well as higher expression of tubular injury-associated genes. Surprisingly, mice with CCL20 blockade had a lower tubular proliferative response and a higher number of cells in the G2/M phase, suggesting impaired repair mechanisms. This may be related to a lower influx of Tregs, despite a milder inflammatory response in terms of chemokine expression and infiltration by IL-17+ cells and neutrophils. In conclusion, CCL20 has a nephroprotective role during AKI, both by decreasing tissue injury and by facilitating repair. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Cristian González-Guerrero
- Laboratory of Nephrology, IIS-Fundación Jiménez Díaz, School of Medicine, UAM, Madrid, Spain.,Red de Investigación Renal (REDINREN)
| | | | - Pablo Cannata-Ortiz
- Pathology, IIS-Fundación Jiménez Díaz, School of Medicine, UAM, Madrid, Spain
| | - María Angeles Ramos-Barron
- Nephrology Investigation Unit, University Hospital Marqués de Valdecilla, IDIVAL (Instituto de Investigacion Valdecilla), Santander, Spain
| | - Carlos Gómez-Alamillo
- Red de Investigación Renal (REDINREN).,Nephrology Investigation Unit, University Hospital Marqués de Valdecilla, IDIVAL (Instituto de Investigacion Valdecilla), Santander, Spain
| | - Manuel Arias
- Red de Investigación Renal (REDINREN).,Nephrology Investigation Unit, University Hospital Marqués de Valdecilla, IDIVAL (Instituto de Investigacion Valdecilla), Santander, Spain
| | - Sergio Mezzano
- Division of Nephrology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Jesús Egido
- IIS-Fundación Jiménez Díaz, School of Medicine, UAM, Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Spain
| | - Marta Ruiz-Ortega
- Red de Investigación Renal (REDINREN).,Cellular Biology in Renal Diseases Laboratory. School of Medicine, UAM, Madrid, Spain
| | - Alberto Ortiz
- Laboratory of Nephrology, IIS-Fundación Jiménez Díaz, School of Medicine, UAM, Madrid, Spain.,Red de Investigación Renal (REDINREN)
| | - Adrián M Ramos
- Laboratory of Nephrology, IIS-Fundación Jiménez Díaz, School of Medicine, UAM, Madrid, Spain.,Red de Investigación Renal (REDINREN)
| |
Collapse
|
44
|
Faiz A, Weckmann M, Tasena H, Vermeulen CJ, Van den Berge M, Ten Hacken NHT, Halayko AJ, Ward JPT, Lee TH, Tjin G, Black JL, Haghi M, Xu CJ, King GG, Farah CS, Oliver BG, Heijink IH, Burgess JK. Profiling of healthy and asthmatic airway smooth muscle cells following interleukin-1β treatment: a novel role for CCL20 in chronic mucus hypersecretion. Eur Respir J 2018; 52:13993003.00310-2018. [PMID: 29946002 DOI: 10.1183/13993003.00310-2018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 05/25/2018] [Indexed: 02/07/2023]
Abstract
Chronic mucus hypersecretion (CMH) contributes to the morbidity and mortality of asthma, and remains uncontrolled by current therapies in the subset of patients with severe, steroid-resistant disease. Altered cross-talk between airway epithelium and airway smooth muscle cells (ASMCs), driven by pro-inflammatory cytokines such as interleukin (IL)-1β, provides a potential mechanism that influences CMH. This study investigated mechanisms underlying CMH by comparing IL-1β-induced gene expression profiles between asthma and control-derived ASMCs and the subsequent paracrine influence on airway epithelial mucus production in vitroIL-1β-treated ASMCs from asthmatic patients and healthy donors were profiled using microarray analysis and ELISA. Air-liquid interface (ALI)-cultured CALU-3 and primary airway epithelial cells were treated with identified candidates and mucus production assessed.The IL-1β-induced CCL20 expression and protein release was increased in ASMCs from moderate compared with mild asthmatic patients and healthy controls. IL-1β induced lower MIR146A expression in asthma-derived ASMCs compared with controls. Decreased MIR146A expression was validated in vivo in bronchial biopsies from 16 asthmatic patients versus 39 healthy donors. miR-146a-5p overexpression abrogated CCL20 release in ASMCs. CCL20 treatment of ALI-cultured CALU-3 and primary airway epithelial cells induced mucus production, while CCL20 levels in sputum were associated with increased levels of CMH in asthmatic patients.Elevated CCL20 production by ASMCs, possibly resulting from dysregulated expression of the anti-inflammatory miR-146a-5p, may contribute to enhanced mucus production in asthma.
Collapse
Affiliation(s)
- Alen Faiz
- Woolcock Institute of Medical Research, The University of Sydney, Glebe, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia.,Dept of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Dept of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Markus Weckmann
- Woolcock Institute of Medical Research, The University of Sydney, Glebe, Australia.,Section for Pediatric Pneumology and Allergology, University Medical Center Schleswig-Holstein, Campus Centrum Luebeck, Airway Research Centre North (ARCN), Member of the German Centre of Lung Research (DZL), Luebeck, Germany
| | - Haitatip Tasena
- Dept of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Dept of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Corneel J Vermeulen
- Dept of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maarten Van den Berge
- Dept of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Nick H T Ten Hacken
- Dept of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andrew J Halayko
- University of Manitoba/Manitoba Institute of Child Health - Winnipeg, Winnipeg, MB, Canada
| | | | - Tak H Lee
- Dept of Physiology, Kings College London, London, UK
| | - Gavin Tjin
- Woolcock Institute of Medical Research, The University of Sydney, Glebe, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia.,Discipline of Pharmacology, Faculty of Medicine, The University of Sydney, Sydney, Australia
| | - Judith L Black
- Woolcock Institute of Medical Research, The University of Sydney, Glebe, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia.,Discipline of Pharmacology, Faculty of Medicine, The University of Sydney, Sydney, Australia
| | - Mehra Haghi
- Graduate School of Health, University of Technology Sydney, Sydney, Australia
| | - Cheng-Jian Xu
- GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Dept of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands
| | - Gregory G King
- Woolcock Institute of Medical Research, The University of Sydney, Glebe, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia.,Dept of Respiratory Medicine, Royal North Shore Hospital, St Leonards, Australia
| | - Claude S Farah
- Woolcock Institute of Medical Research, The University of Sydney, Glebe, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia.,Dept of Respiratory Medicine, Concord Hospital, Concord, Australia
| | - Brian G Oliver
- Woolcock Institute of Medical Research, The University of Sydney, Glebe, Australia.,School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, Australia
| | - Irene H Heijink
- Dept of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Dept of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Janette K Burgess
- Woolcock Institute of Medical Research, The University of Sydney, Glebe, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia.,GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Discipline of Pharmacology, Faculty of Medicine, The University of Sydney, Sydney, Australia.,Dept of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
45
|
Martin JG. Airway smooth muscle may drive mucus hypersecretion in asthma. Eur Respir J 2018; 52:52/2/1801166. [PMID: 30093557 DOI: 10.1183/13993003.01166-2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 07/02/2018] [Indexed: 02/03/2023]
Affiliation(s)
- James G Martin
- Dept of Medicine, Division of Respiratory Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada .,Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
46
|
Ni L, Dong C. Roles of Myeloid and Lymphoid Cells in the Pathogenesis of Chronic Obstructive Pulmonary Disease. Front Immunol 2018; 9:1431. [PMID: 29977245 PMCID: PMC6021485 DOI: 10.3389/fimmu.2018.01431] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/08/2018] [Indexed: 12/15/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is currently the third largest cause of human mortality in the world after stroke and heart disease. COPD is characterized by sustained inflammation of the airways, leading to destruction of lung tissue and declining pulmonary function. The main risk factor is known to be cigarette smoke currently. However, the strategies for prevention and treatment have not altered significantly for many years. A growing body of evidences indicates that the immune system plays a pivotal role in the pathogenesis of COPD. The repeated and progressive activation of immune cells is at least in part the source of this chronic inflammation. In this review paper, we have conducted an extensive literature search of the studies of immune cells in COPD patients. The objective is to assess the contributions of different immune cell types, the imbalance of pro/anti-inflammatory immune cells, such as M1/M2 macrophages, Tc1/Tc10, and Th17/Treg, and their mediators in the peripheral blood as well as in the lung to the pathogenesis of COPD. Therefore, understanding their roles in COPD development will help us find the potential target to modify this disease. This review focuses predominantly on data derived from human studies but will refer to animal studies where they help understand the disease in humans.
Collapse
Affiliation(s)
- Ling Ni
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
47
|
De Rose V, Molloy K, Gohy S, Pilette C, Greene CM. Airway Epithelium Dysfunction in Cystic Fibrosis and COPD. Mediators Inflamm 2018; 2018:1309746. [PMID: 29849481 PMCID: PMC5911336 DOI: 10.1155/2018/1309746] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 01/15/2018] [Accepted: 02/01/2018] [Indexed: 12/22/2022] Open
Abstract
Cystic fibrosis is a genetic disease caused by mutations in the CFTR gene, whereas chronic obstructive pulmonary disease (COPD) is mainly caused by environmental factors (mostly cigarette smoking) on a genetically susceptible background. Although the etiology and pathogenesis of these diseases are different, both are associated with progressive airflow obstruction, airway neutrophilic inflammation, and recurrent exacerbations, suggesting common mechanisms. The airway epithelium plays a crucial role in maintaining normal airway functions. Major molecular and morphologic changes occur in the airway epithelium in both CF and COPD, and growing evidence suggests that airway epithelial dysfunction is involved in disease initiation and progression in both diseases. Structural and functional abnormalities in both airway and alveolar epithelium have a relevant impact on alteration of host defences, immune/inflammatory response, and the repair process leading to progressive lung damage and impaired lung function. In this review, we address the evidence for a critical role of dysfunctional airway epithelial cells in chronic airway inflammation and remodelling in CF and COPD, highlighting the common mechanisms involved in the epithelial dysfunction as well as the similarities and differences of the two diseases.
Collapse
Affiliation(s)
- Virginia De Rose
- Department of Clinical and Biological Sciences, University of Torino, A.O.U. S. Luigi Gonzaga, Regione Gonzole 10, 10043 Orbassano, Torino, Italy
| | - Kevin Molloy
- Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Dublin, Ireland
| | - Sophie Gohy
- Institute of Experimental and Clinical Research, Pole of Pneumology, ENT and Dermatology, Université Catholique de Louvain (UCL), Brussels, Belgium
- Department of Pneumology, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Charles Pilette
- Institute of Experimental and Clinical Research, Pole of Pneumology, ENT and Dermatology, Université Catholique de Louvain (UCL), Brussels, Belgium
- Department of Pneumology, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Catherine M. Greene
- Lung Biology Group, Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Dublin, Ireland
| |
Collapse
|
48
|
Di Vincenzo S, Heijink IH, Noordhoek JA, Cipollina C, Siena L, Bruno A, Ferraro M, Postma DS, Gjomarkaj M, Pace E. SIRT1/FoxO3 axis alteration leads to aberrant immune responses in bronchial epithelial cells. J Cell Mol Med 2018; 22:2272-2282. [PMID: 29411515 PMCID: PMC5867095 DOI: 10.1111/jcmm.13509] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/25/2017] [Indexed: 12/13/2022] Open
Abstract
Inflammation and ageing are intertwined in chronic obstructive pulmonary disease (COPD). The histone deacetylase SIRT1 and the related activation of FoxO3 protect from ageing and regulate inflammation. The role of SIRT1/FoxO3 in COPD is largely unknown. This study evaluated whether cigarette smoke, by modulating the SIRT1/FoxO3 axis, affects airway epithelial pro-inflammatory responses. Human bronchial epithelial cells (16HBE) and primary bronchial epithelial cells (PBECs) from COPD patients and controls were treated with/without cigarette smoke extract (CSE), Sirtinol or FoxO3 siRNA. SIRT1, FoxO3 and NF-κB nuclear accumulation, SIRT1 deacetylase activity, IL-8 and CCL20 expression/release and the release of 12 cytokines, neutrophil and lymphocyte chemotaxis were assessed. In PBECs, the constitutive FoxO3 expression was lower in patients with COPD than in controls. Furthermore, CSE reduced FoxO3 expression only in PBECs from controls. In 16HBE, CSE decreased SIRT1 activity and nuclear expression, enhanced NF-κB binding to the IL-8 gene promoter thus increasing IL-8 expression, decreased CCL20 expression, increased the neutrophil chemotaxis and decreased lymphocyte chemotaxis. Similarly, SIRT1 inhibition reduced FoxO3 expression and increased nuclear NF-κB. FoxO3 siRNA treatment increased IL-8 and decreased CCL20 expression in 16HBE. In conclusion, CSE impairs the function of SIRT1/FoxO3 axis in bronchial epithelium, dysregulating NF-κB activity and inducing pro-inflammatory responses.
Collapse
Affiliation(s)
- Serena Di Vincenzo
- Istituto di Biomedicina e Immunologia Molecolare‐Consiglio Nazionale delle RicerchePalermoItaly
| | - Irene H. Heijink
- Department of Pathology and Medical BiologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Jacobien A. Noordhoek
- Department of Pathology and Medical BiologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Chiara Cipollina
- Istituto di Biomedicina e Immunologia Molecolare‐Consiglio Nazionale delle RicerchePalermoItaly
- Fondazione Ri.MEDPalermoItaly
| | - Liboria Siena
- Istituto di Biomedicina e Immunologia Molecolare‐Consiglio Nazionale delle RicerchePalermoItaly
| | - Andreina Bruno
- Istituto di Biomedicina e Immunologia Molecolare‐Consiglio Nazionale delle RicerchePalermoItaly
| | - Maria Ferraro
- Istituto di Biomedicina e Immunologia Molecolare‐Consiglio Nazionale delle RicerchePalermoItaly
| | - Dirkje S. Postma
- Department of Pathology and Medical BiologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Mark Gjomarkaj
- Istituto di Biomedicina e Immunologia Molecolare‐Consiglio Nazionale delle RicerchePalermoItaly
| | - Elisabetta Pace
- Istituto di Biomedicina e Immunologia Molecolare‐Consiglio Nazionale delle RicerchePalermoItaly
| |
Collapse
|
49
|
Brogden KA, Parashar D, Hallier AR, Braun T, Qian F, Rizvi NA, Bossler AD, Milhem MM, Chan TA, Abbasi T, Vali S. Genomics of NSCLC patients both affirm PD-L1 expression and predict their clinical responses to anti-PD-1 immunotherapy. BMC Cancer 2018; 18:225. [PMID: 29486723 PMCID: PMC5897943 DOI: 10.1186/s12885-018-4134-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 02/15/2018] [Indexed: 12/21/2022] Open
Abstract
Background Programmed Death Ligand 1 (PD-L1) is a co-stimulatory and immune checkpoint protein. PD-L1 expression in non-small cell lung cancers (NSCLC) is a hallmark of adaptive resistance and its expression is often used to predict the outcome of Programmed Death 1 (PD-1) and PD-L1 immunotherapy treatments. However, clinical benefits do not occur in all patients and new approaches are needed to assist in selecting patients for PD-1 or PD-L1 immunotherapies. Here, we hypothesized that patient tumor cell genomics influenced cell signaling and expression of PD-L1, chemokines, and immunosuppressive molecules and these profiles could be used to predict patient clinical responses. Methods We used a recent dataset from NSCLC patients treated with pembrolizumab. Deleterious gene mutational profiles in patient exomes were identified and annotated into a cancer network to create NSCLC patient-specific predictive computational simulation models. Validation checks were performed on the cancer network, simulation model predictions, and PD-1 match rates between patient-specific predicted and clinical responses. Results Expression profiles of these 24 chemokines and immunosuppressive molecules were used to identify patients who would or would not respond to PD-1 immunotherapy. PD-L1 expression alone was not sufficient to predict which patients would or would not respond to PD-1 immunotherapy. Adding chemokine and immunosuppressive molecule expression profiles allowed patient models to achieve a greater than 85.0% predictive correlation among predicted and reported patient clinical responses. Conclusions Our results suggested that chemokine and immunosuppressive molecule expression profiles can be used to accurately predict clinical responses thus differentiating among patients who would and would not benefit from PD-1 or PD-L1 immunotherapies. Electronic supplementary material The online version of this article (10.1186/s12885-018-4134-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kim A Brogden
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA.
| | - Deepak Parashar
- Cellworks Research India Ltd., Whitefield, Bangalore, 560066, India
| | - Andrea R Hallier
- Biomedical Engineering, The University of Iowa, 5318 SC, Iowa City, IA, 52242, USA
| | - Terry Braun
- Biomedical Engineering, The University of Iowa, 5318 SC, Iowa City, IA, 52242, USA
| | - Fang Qian
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA.,Division of Biostatistics and Research Design, College of Dentistry, The University of Iowa, 801 Newton Road, Iowa City, IA, 52242, USA
| | - Naiyer A Rizvi
- Division of Hematology/Oncology, Columbia University Medical Center, 177 Fort Washington Avenue, New York, NY, 10032, USA
| | - Aaron D Bossler
- Molecular Pathology Laboratory, Department of Pathology, University of Iowa Hospitals and Clinics, 200 Hawkins Dr., C606GH, Iowa City, IA, 52242, USA
| | - Mohammed M Milhem
- Clinical Services, Experimental Therapeutics, Melanoma and Sarcoma Program, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, 52242, USA
| | - Timothy A Chan
- Department of Radiation Oncology, Human Oncology and Pathogenesis Program, Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Taher Abbasi
- Cellworks Group, Inc., 2033 Gateway Place Suite 500, San Jose, CA, 95110, USA
| | - Shireen Vali
- Cellworks Group, Inc., 2033 Gateway Place Suite 500, San Jose, CA, 95110, USA
| |
Collapse
|
50
|
Clinical implications of oncogenic mutations in pulmonary Langerhans cell histiocytosis. Curr Opin Pulm Med 2018; 24:281-286. [PMID: 29470255 DOI: 10.1097/mcp.0000000000000470] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE OF REVIEW Langerhans cell histiocytosis (LCH) is a neoplasm of dendritic cells with a wide clinical spectrum. Localized pulmonary LCH occurs in young adults with a history of smoking and can either resolve spontaneously or lead to progressive decline in pulmonary function. Young children can also present with localized disease - frequently bone or skin - or with multifocal or multisystem disease. Clinical outcomes in these patients also vary widely, ranging from spontaneous resolution to multiorgan failure and death. This review describes recent developments in our understanding of the underlying pathogenesis of LCH and how these discoveries and other research are affecting how the disease is classified, treated and monitored. RECENT FINDINGS Somatic mutations resulting in activation of the mitogen-activated protein kinase (MAPK) pathway were recently identified as a key pathogenetic mechanism in both pediatric and pulmonary LCH. SUMMARY Knowledge of underlying pathogenetic mechanisms of LCH transforming how this disease and other histocytic/dendritic disorders are classified, treated and monitored.
Collapse
|