1
|
Russo RC, Ryffel B. The Chemokine System as a Key Regulator of Pulmonary Fibrosis: Converging Pathways in Human Idiopathic Pulmonary Fibrosis (IPF) and the Bleomycin-Induced Lung Fibrosis Model in Mice. Cells 2024; 13:2058. [PMID: 39768150 PMCID: PMC11674266 DOI: 10.3390/cells13242058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and lethal interstitial lung disease (ILD) of unknown origin, characterized by limited treatment efficacy and a fibroproliferative nature. It is marked by excessive extracellular matrix deposition in the pulmonary parenchyma, leading to progressive lung volume decline and impaired gas exchange. The chemokine system, a network of proteins involved in cellular communication with diverse biological functions, plays a crucial role in various respiratory diseases. Chemokine receptors trigger the activation, proliferation, and migration of lung-resident cells, including pneumocytes, endothelial cells, alveolar macrophages, and fibroblasts. Around 50 chemokines can potentially interact with 20 receptors, expressed by both leukocytes and non-leukocytes such as tissue parenchyma cells, contributing to processes such as leukocyte mobilization from the bone marrow, recirculation through lymphoid organs, and tissue influx during inflammation or immune response. This narrative review explores the complexity of the chemokine system in the context of IPF and the bleomycin-induced lung fibrosis mouse model. The goal is to identify specific chemokines and receptors as potential therapeutic targets. Recent progress in understanding the role of the chemokine system during IPF, using experimental models and molecular diagnosis, underscores the complex nature of this system in the context of the disease. Despite advances in experimental models and molecular diagnostics, discovering an effective therapy for IPF remains a significant challenge in both medicine and pharmacology. This work delves into microarray results from lung samples of IPF patients and murine samples at different stages of bleomycin-induced pulmonary fibrosis. By discussing common pathways identified in both IPF and the experimental model, we aim to shed light on potential targets for therapeutic intervention. Dysregulation caused by abnormal chemokine levels observed in IPF lungs may activate multiple targets, suggesting that chemokine signaling plays a central role in maintaining or perpetuating lung fibrogenesis. The highlighted chemokine axes (CCL8-CCR2, CCL19/CCL21-CCR7, CXCL9-CXCR3, CCL3/CCL4/CCL5-CCR5, and CCL20-CCR6) present promising opportunities for advancing IPF treatment research and uncovering new pharmacological targets within the chemokine system.
Collapse
Affiliation(s)
- Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais-UFMG, Belo Horizonte 31270-901, MG, Brazil
| | - Bernhard Ryffel
- Laboratory of Immuno-Neuro Modulation (INEM), UMR7355 Centre National de la Recherche Scientifique (CNRS), University of Orleans, 45071 Orleans, France
| |
Collapse
|
2
|
Morikka J, Federico A, Möbus L, Inkala S, Pavel A, Sani S, Vaani M, Peltola S, Serra A, Greco D. Toxicogenomic assessment of in vitro macrophages exposed to profibrotic challenge reveals a sustained transcriptomic immune signature. Comput Struct Biotechnol J 2024; 25:194-204. [PMID: 39430886 PMCID: PMC11490883 DOI: 10.1016/j.csbj.2024.10.010] [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: 06/20/2024] [Revised: 09/27/2024] [Accepted: 10/05/2024] [Indexed: 10/22/2024] Open
Abstract
Immune signalling is a crucial component in the progression of fibrosis. However, approaches for the safety assessment of potentially profibrotic substances, that provide information on mechanistic immune responses, are underdeveloped. This study aimed to develop a novel framework for assessing the immunotoxicity of fibrotic compounds. We exposed macrophages in vitro to multiple sublethal concentrations of the profibrotic agent bleomycin, over multiple timepoints, and generated RNA sequencing data. Using a toxicogenomic approach, we performed dose-dependent analysis to discover genes dysregulated by bleomycin exposure in a dose-responsive manner. A subset of immune genes displayed a sustained dose-dependent and differential expression response to profibrotic challenge. An immunoassay revealed cytokines and proteinases responding to bleomycin exposure that closely correlated to transcriptomic alterations, underscoring the integration between transcriptional immune response and external immune signalling activity. This study not only increases our understanding of the immunological mechanisms of fibrosis, but also offers an innovative framework for the toxicological evaluation of substances with potential fibrogenic effects on macrophage signalling. Our work brings a new immunotoxicogenomic direction for hazard assessment of fibrotic compounds, through the implementation of a time and resource efficient in vitro methodology.
Collapse
Affiliation(s)
- Jack Morikka
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
| | - Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Lena Möbus
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Simo Inkala
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Alisa Pavel
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Copenhagen, Denmark
| | - Saara Sani
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Maaret Vaani
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sanna Peltola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
3
|
Zhao AY, Unterman A, Abu Hussein NS, Sharma P, Nikola F, Flint J, Yan X, Adams TS, Justet A, Sumida TS, Zhao J, Schupp JC, Raredon MSB, Ahangari F, Deluliis G, Zhang Y, Buendia-Roldan I, Adegunsoye A, Sperling AI, Prasse A, Ryu C, Herzog E, Selman M, Pardo A, Kaminski N. Single-Cell Analysis Reveals Novel Immune Perturbations in Fibrotic Hypersensitivity Pneumonitis. Am J Respir Crit Care Med 2024; 210:1252-1266. [PMID: 38924775 PMCID: PMC11568434 DOI: 10.1164/rccm.202401-0078oc] [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: 01/10/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024] Open
Abstract
Rationale: Fibrotic hypersensitivity pneumonitis (FHP) is a debilitating interstitial lung disease driven by incompletely understood immune mechanisms. Objectives: To elucidate immune aberrations in FHP in single-cell resolution. Methods: Single-cell 5' RNA sequencing was conducted on peripheral blood mononuclear cells and BAL cells obtained from 45 patients with FHP, 63 patients with idiopathic pulmonary fibrosis (IPF), 4 patients with nonfibrotic hypersensitivity pneumonitis, and 36 healthy control subjects in the United States and Mexico. Analyses included differential gene expression (Seurat), TF (transcription factor) activity imputation (DoRothEA-VIPER), and trajectory analyses (Monocle3 and Velocyto-scVelo-CellRank). Measurements and Main Results: Overall, 501,534 peripheral blood mononuclear cells from 110 patients and control subjects and 88,336 BAL cells from 19 patients were profiled. Compared with control samples, FHP has elevated classical monocytes (adjusted-P = 2.5 × 10-3) and is enriched in CCL3hi/CCL4hi and S100Ahi classical monocytes (adjusted-P < 2.2 × 10-16). Trajectory analyses demonstrate that S100Ahi classical monocytes differentiate into SPP1hi lung macrophages associated with fibrosis. Compared with both control subjects and IPF, cells from patients with FHP are significantly enriched in GZMhi cytotoxic T cells. These cells exhibit TF activities indicative of TGFβ and TNFα and NFκB pathways. These results are publicly available at http://ildimmunecellatlas.com. Conclusions: Single-cell transcriptomics of patients with FHP uncovered novel immune perturbations, including previously undescribed increases in GZMhi cytotoxic CD4+ and CD8+ T cells-reflecting this disease's unique inflammatory T cell-driven nature-as well as increased S100Ahi and CCL3hi/CCL4hi classical monocytes also observed in IPF. Both cell populations may guide the development of new biomarkers and therapeutic interventions.
Collapse
Affiliation(s)
- Amy Y. Zhao
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Avraham Unterman
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
- Institute of Pulmonary Medicine, Tel Aviv Sourasky Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Prapti Sharma
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Fadi Nikola
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Jasper Flint
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Xiting Yan
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Taylor S. Adams
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Aurelien Justet
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
- Service de Pneumologie, Centre de Competences de Maladies Pulmonaires Rares, Centre Hospitalier Universitaire de Caen University of Caen Normandie, CEA, Centre National de la Recherche Scientifique, Imagerie et Stratégies Thérapeutiques pour les Cancers et Tissus Cérébraux/CERVOxy Group, GIP CYCERON, Normandie University, Caen, France
| | | | - Jiayi Zhao
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Jonas C. Schupp
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
- Department of Pulmonary and Infectious Diseases, Hannover Medical School, Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), German Center for Lung Research BREATH, Hannover, Germany
| | - Micha Sam B. Raredon
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
- Department of Anesthesiology, and
- Department of Immunobiology, Yale School of Medicine, New Haven, Conncecticut
| | - Farida Ahangari
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Giuseppe Deluliis
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Yingze Zhang
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ivette Buendia-Roldan
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Ayodeji Adegunsoye
- Section of Pulmonary/Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Anne I. Sperling
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Antje Prasse
- Section of Pulmonary Medicine, University Medical Center, Basel, Switzerland; and
| | - Changwan Ryu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Erica Herzog
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Moises Selman
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Annie Pardo
- Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| |
Collapse
|
4
|
Luo X, Xiang F. Acute exacerbation of idiopathic pulmonary fibrosis a narrative review primary focus on treatments. J Thorac Dis 2024; 16:4727-4741. [PMID: 39144320 PMCID: PMC11320219 DOI: 10.21037/jtd-23-1565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 05/17/2024] [Indexed: 08/16/2024]
Abstract
Background and Objective Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrotic interstitial pneumonia, which is the commonest type of idiopathic interstitial pneumonia in the clinic. For most patients, the course of the disease is slow and prolonged, but a percentage of them develop an acute respiratory worsening during the disease, known as an acute exacerbation of IPF (AE-IPF). The updated guidelines define AE-IPF as an acute worsening of dyspnea in an IPF patient within 1 month and exclude other conditions such as left heart failure and pulmonary embolism. However, the prevention and treatment of AE-IPF are still unclear. Based on the high mortality rate caused by AE, in this article, we will focus on the latest research advances in AE-IPF treatment strategies and provide a comprehensive review of its pathogenesis, risk factors, clinical features, and diagnosis. Methods This study searched for relevant literature published from 2018 to 2023 in the PubMed database. The search terms used were as follows: "Acute exacerbation", "Idiopathic pulmonary fibrosis", "Biomarker", "Pathogenesis", "Treatment", "HRCT", "Antifibrotic", "Infection", "Immunosuppressant", "Autoantibody", "Oxygen therapy", "Hemoperfusion", "Inflammation". Key Content and Findings The review found that corticosteroids are still the primary treatment strategy at present, although there is some controversy regarding the dosing and tapering of corticosteroids. However, corticosteroids combined with intravenous cyclophosphamide have been shown to be detrimental to the prognosis of patients with AE-IPF. Given its deadly high mortality rate, early intervention is crucial. Pirfenidone and nintedanib have been proven to reduce incidence of AE. Meanwhile, in the future, the lung microbiome may also be a break-through. Conclusions This study reviewed the pathogenesis and risk factors of AE-IPF and updated the current and potential treatment strategies regarding AE-IPF. The pathogenesis of AE-IPF is not exact, multiple mechanisms may be involved simultaneously. Corticosteroids remain the mainstream treatment modality in the medical treatment of AE-IFP. Many other treatment modalities have been proposed in succession, but no clear conclusions can be drawn about the effectiveness and safety of these interventions.
Collapse
Affiliation(s)
- Xiaohui Luo
- Department of Pulmonary and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Xiang
- Department of Pulmonary and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Ge Z, Chen Y, Ma L, Hu F, Xie L. Macrophage polarization and its impact on idiopathic pulmonary fibrosis. Front Immunol 2024; 15:1444964. [PMID: 39131154 PMCID: PMC11310026 DOI: 10.3389/fimmu.2024.1444964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lung disease that worsens over time, causing fibrosis in the lungs and ultimately resulting in respiratory failure and a high risk of death. Macrophages play a crucial role in the immune system, showing flexibility by transforming into either pro-inflammatory (M1) or anti-inflammatory (M2) macrophages when exposed to different stimuli, ultimately impacting the development of IPF. Recent research has indicated that the polarization of macrophages is crucial in the onset and progression of IPF. M1 macrophages secrete inflammatory cytokines and agents causing early lung damage and fibrosis, while M2 macrophages support tissue healing and fibrosis by releasing anti-inflammatory cytokines. Developing novel treatments for IPF relies on a thorough comprehension of the processes involved in macrophage polarization in IPF. The review outlines the regulation of macrophage polarization and its impact on the development of IPF, with the goal of investigating the possible therapeutic benefits of macrophage polarization in the advancement of IPF.
Collapse
Affiliation(s)
- Zhouling Ge
- Department of Respiratory Medicine, The Third Affiliated Hospital of Shanghai University (Wenzhou People’s Hospital), Wenzhou, China
| | - Yong Chen
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Leikai Ma
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fangjun Hu
- Department of Respiratory Medicine, The Third Affiliated Hospital of Shanghai University (Wenzhou People’s Hospital), Wenzhou, China
| | - Lubin Xie
- Department of Respiratory Medicine, The Third Affiliated Hospital of Shanghai University (Wenzhou People’s Hospital), Wenzhou, China
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
6
|
Kamiya M, Carter H, Espindola MS, Doyle TJ, Lee JS, Merriam LT, Zhang F, Kawano-Dourado L, Sparks JA, Hogaboam CM, Moore BB, Oldham WM, Kim EY. Immune mechanisms in fibrotic interstitial lung disease. Cell 2024; 187:3506-3530. [PMID: 38996486 PMCID: PMC11246539 DOI: 10.1016/j.cell.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 05/04/2024] [Accepted: 05/08/2024] [Indexed: 07/14/2024]
Abstract
Fibrotic interstitial lung diseases (fILDs) have poor survival rates and lack effective therapies. Despite evidence for immune mechanisms in lung fibrosis, immunotherapies have been unsuccessful for major types of fILD. Here, we review immunological mechanisms in lung fibrosis that have the potential to impact clinical practice. We first examine innate immunity, which is broadly involved across fILD subtypes. We illustrate how innate immunity in fILD involves a complex interplay of multiple cell subpopulations and molecular pathways. We then review the growing evidence for adaptive immunity in lung fibrosis to provoke a re-examination of its role in clinical fILD. We close with future directions to address key knowledge gaps in fILD pathobiology: (1) longitudinal studies emphasizing early-stage clinical disease, (2) immune mechanisms of acute exacerbations, and (3) next-generation immunophenotyping integrating spatial, genetic, and single-cell approaches. Advances in these areas are essential for the future of precision medicine and immunotherapy in fILD.
Collapse
Affiliation(s)
- Mari Kamiya
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Hannah Carter
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Milena S Espindola
- Division of Pulmonary and Critical Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Tracy J Doyle
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Joyce S Lee
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Louis T Merriam
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Fan Zhang
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA; Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Leticia Kawano-Dourado
- Hcor Research Institute, Hcor Hospital, Sao Paulo - SP 04004-030, Brazil; Pulmonary Division, Heart Institute (InCor), University of Sao Paulo, São Paulo - SP 05403-900, Brazil
| | - Jeffrey A Sparks
- Harvard Medical School, Boston, MA 02115, USA; Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Cory M Hogaboam
- Division of Pulmonary and Critical Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Bethany B Moore
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - William M Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA.
| | - Edy Y Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Enomoto N. Relationship between idiopathic interstitial pneumonias (IIPs) and connective tissue disease-related interstitial lung disease (CTD-ILD): A narrative review. Respir Investig 2024; 62:465-480. [PMID: 38564878 DOI: 10.1016/j.resinv.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/17/2024] [Accepted: 03/14/2024] [Indexed: 04/04/2024]
Abstract
While idiopathic interstitial pneumonia (IIP) centering on idiopathic pulmonary fibrosis (IPF) is the most prevalent interstitial lung disease (ILD), especially in the older adult population, connective tissue disease (CTD)-related ILD is the second most prevalent ILD. The pathogenesis of IPF is primarily fibrosis, whereas that of other ILDs, particularly CTD-ILD, is mainly inflammation. Therefore, a precise diagnosis is crucial for selecting appropriate treatments, such as antifibrotic or immunosuppressive agents. In addition, some patients with IIP have CTD-related features, such as arthritis and skin eruption, but do not meet the criteria for any CTD, this is referred to as interstitial pneumonia with autoimmune features (IPAF). IPAF is closely associated with idiopathic nonspecific interstitial pneumonia (iNSIP) and cryptogenic organizing pneumonia (COP). Furthermore, patients with iNSIP or those with NSIP with OP overlap frequently develop polymyositis/dermatomyositis after the diagnosis of IIP. Acute exacerbation of ILD, the most common cause of death, occurs more frequently in patients with IPF than in those with other ILDs. Although acute exacerbation of CTD-ILD occurs at a low rate of incidence, patients with rheumatoid arthritis, microscopic polyangiitis, or systemic sclerosis experience more acute exacerbation of CTD-ILD than those with other CTD. In this review, the features of each IIP, focusing on CTD-related signatures, are summarized, and the pathogenesis and appropriate treatments to improve the prognoses of patients with various ILDs are discussed.
Collapse
Affiliation(s)
- Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan; Health Administration Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan.
| |
Collapse
|
8
|
Read J, Reid AT, Thomson C, Plit M, Mejia R, Knight DA, Lize M, El Kasmi K, Grainge CL, Stahl H, Schuliga M. Alveolar epithelial cells of lung fibrosis patients are susceptible to severe virus-induced injury. Clin Sci (Lond) 2024; 138:537-554. [PMID: 38577922 DOI: 10.1042/cs20240220] [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: 01/31/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/06/2024]
Abstract
Patients with pulmonary fibrosis (PF) often experience exacerbations of their disease, characterised by a rapid, severe deterioration in lung function that is associated with high mortality. Whilst the pathobiology of such exacerbations is poorly understood, virus infection is a trigger. The present study investigated virus-induced injury responses of alveolar and bronchial epithelial cells (AECs and BECs, respectively) from patients with PF and age-matched controls (Ctrls). Air-liquid interface (ALI) cultures of AECs, comprising type I and II pneumocytes or BECs were inoculated with influenza A virus (H1N1) at 0.1 multiplicity of infection (MOI). Levels of interleukin-6 (IL-6), IL-36γ and IL-1β were elevated in cultures of AECs from PF patients (PF-AECs, n = 8-11), being markedly higher than Ctrl-AECs (n = 5-6), 48 h post inoculation (pi) (P<0.05); despite no difference in H1N1 RNA copy numbers 24 h pi. Furthermore, the virus-induced inflammatory responses of PF-AECs were greater than BECs (from either PF patients or controls), even though viral loads in the BECs were overall 2- to 3-fold higher than AECs. Baseline levels of the senescence and DNA damage markers, nuclear p21, p16 and H2AXγ were also significantly higher in PF-AECs than Ctrl-AECs and further elevated post-infection. Senescence induction using etoposide augmented virus-induced injuries in AECs (but not viral load), whereas selected senotherapeutics (rapamycin and mitoTEMPO) were protective. The present study provides evidence that senescence increases the susceptibility of AECs from PF patients to severe virus-induced injury and suggests targeting senescence may provide an alternative option to prevent or treat the exacerbations that worsen the underlying disease.
Collapse
Affiliation(s)
- Jane Read
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Andrew T Reid
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
| | - Claire Thomson
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
- Saint Vincent's Hospital, Sydney, NSW, Australia
| | | | - Ross Mejia
- John Hunter Hospital, Newcastle, NSW, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Providence Health Care Research Institute, Vancouver, British Columbia, Canada
| | - Muriel Lize
- Boehringer Ingelheim Pharma GmbH & Co. KG, Germany
| | | | - Christopher L Grainge
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
- John Hunter Hospital, Newcastle, NSW, Australia
| | - Heiko Stahl
- Boehringer Ingelheim Pharma GmbH & Co. KG, Germany
| | - Michael Schuliga
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
9
|
Luo J, Li P, Dong M, Zhang Y, Lu S, Chen M, Zhou H, Lin N, Jiang H, Wang Y. SLC15A3 plays a crucial role in pulmonary fibrosis by regulating macrophage oxidative stress. Cell Death Differ 2024; 31:417-430. [PMID: 38374230 PMCID: PMC11043330 DOI: 10.1038/s41418-024-01266-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/21/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal and irreversible disease with few effective treatments. Alveolar macrophages (AMs) are involved in the development of IPF from the initial stages due to direct exposure to air and respond to external oxidative damage (a major inducement of pulmonary fibrosis). Oxidative stress in AMs plays an indispensable role in promoting fibrosis development. The oligopeptide histidine transporter SLC15A3, mainly expressed on the lysosomal membrane of macrophages and highly expressed in the lung, has proved to be involved in innate immune and antiviral signaling pathways. In this study, we demonstrated that during bleomycin (BLM)- or radiation-induced pulmonary fibrosis, the recruitment of macrophages induced an increase of SLC15A3 in the lung, and the deficiency of SLC15A3 protected mice from pulmonary fibrosis and maintained the homeostasis of the pulmonary microenvironment. Mechanistically, deficiency of SLC15A3 resisted oxidative stress in macrophages, and SLC15A3 interacted with the scaffold protein p62 to regulate its expression and phosphorylation activation, thereby regulating p62-nuclear factor erythroid 2-related factor 2 (NRF2) antioxidant stress pathway protein, which is related to the production of reactive oxygen species (ROS). Overall, our data provided a novel mechanism for targeting SLC15A3 to regulate oxidative stress in macrophages, supporting the therapeutic potential of inhibiting or silencing SLC15A3 for the precautions and treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Jun Luo
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ping Li
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, China
| | - Minlei Dong
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yingqiong Zhang
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Shuanghui Lu
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Mingyang Chen
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hui Zhou
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Nengming Lin
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, China
| | - Huidi Jiang
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
| | - Yuqing Wang
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, China.
| |
Collapse
|
10
|
Dong Y, He L, Zhu Z, Yang F, Ma Q, Zhang Y, Zhang X, Liu X. The mechanism of gut-lung axis in pulmonary fibrosis. Front Cell Infect Microbiol 2024; 14:1258246. [PMID: 38362497 PMCID: PMC10867257 DOI: 10.3389/fcimb.2024.1258246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024] Open
Abstract
Pulmonary fibrosis (PF) is a terminal change of a lung disease that is marked by damage to alveolar epithelial cells, abnormal proliferative transformation of fibroblasts, excessive deposition of extracellular matrix (ECM), and concomitant inflammatory damage. Its characteristics include short median survival, high mortality rate, and limited treatment effectiveness. More in-depth studies on the mechanisms of PF are needed to provide better treatment options. The idea of the gut-lung axis has emerged as a result of comprehensive investigations into the microbiome, metabolome, and immune system. This theory is based on the material basis of microorganisms and their metabolites, while the gut-lung circulatory system and the shared mucosal immune system act as the connectors that facilitate the interplay between the gastrointestinal and respiratory systems. The emergence of a new view of the gut-lung axis is complementary and cross-cutting to the study of the mechanisms involved in PF and provides new ideas for its treatment. This article reviews the mechanisms involved in PF, the gut-lung axis theory, and the correlation between the two. Exploring the gut-lung axis mechanism and treatments related to PF from the perspectives of microorganisms, microbial metabolites, and the immune system. The study of the gut-lung axis and PF is still in its early stages. This review systematically summarizes the mechanisms of PF related to the gut-lung axis, providing ideas for subsequent research and treatment of related mechanisms.
Collapse
Affiliation(s)
- Yawei Dong
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Lanlan He
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Zhongbo Zhu
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Fan Yang
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Quan Ma
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Respiratory Medicine, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yanmei Zhang
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xuhui Zhang
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Respiratory Medicine, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xiping Liu
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
11
|
Yu H, Liu S, Wang S, Gu X. A narrative review of the role of HDAC6 in idiopathic pulmonary fibrosis. J Thorac Dis 2024; 16:688-695. [PMID: 38410580 PMCID: PMC10894383 DOI: 10.21037/jtd-23-1183] [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: 07/29/2023] [Accepted: 11/17/2023] [Indexed: 02/28/2024]
Abstract
Background and Objective Idiopathic pulmonary fibrosis (IPF) is a progressive and irreversible condition characterized by the deposition of extracellular matrix resulting from repetitive damage to the alveolar epithelium. These injuries, along with dysregulated wound repair and fibroblast dysfunction, lead to continuous tissue remodeling and fibrosis, eventually resulting in end-stage pulmonary fibrosis. Currently, there is no specific pharmacological treatment available for IPF. The role of inflammation in the development of IPF is still a topic of debate, and it is sometimes considered incidental to fibrosis. Over the past decade, macrophages have emerged as significant contributors to the pathogenesis of fibrosis. M1 macrophages are responsible for wound healing following alveolar epithelial injury, while M2 macrophages are involved in resolving wound repair and terminating the inflammatory response in the lungs. Various studies provide evidence that M2-like macrophages contribute to the abnormal fibrogenesis. In recent years, there has been growing interest in understanding macrophage polarization and its role in the development of pulmonary fibrosis. Histone deacetylase 6 (HDAC6), a member of the HDAC family with two functional deacetylase structural domains and a ubiquitin-binding zinc finger structural domain (ZnF-BUZ), plays a crucial role in pulmonary fibrosis. This article explores the role of HDAC6 in pulmonary fibrosis and evaluates its potential as a treatment approach for IPF. Methods PubMed, Cochrane Library, China National Knowledge Infrastructure (CNKI), Wanfang, China Biomedical Literature Service System (CBMdisc) and Web of Science were searched to obtain researches, published in English and Chinese, until July 2023. The search was performed using specific keywords such as Histone deacetylase 6, HDAC6, Idiopathic pulmonary fibrosis, IPF, fibrosis. Key Content and Findings HDAC6 has diverse effects on physiological processes, including the NLRP3 inflammasome, epithelial-mesenchymal transition, the TGFβ-PI3K-AKT pathway, macrophage polarization and TGF-β-Smad signaling pathway, due to its unique structure. HDAC6 has been found to enhance the inflammatory response and fibrosis of lung tissues, contributing to the development of IPF. Conclusions In the future, HDAC6 inhibitors are expected to play a crucial role in the treatment of fibrotic disorders and should be studied further deserves to pursue in future research.
Collapse
Affiliation(s)
- Hanming Yu
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shi Liu
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shuo Wang
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiu Gu
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
He Y, Yao T, Zhang Y, Long L, Jiang G, Zhang X, Lv X, Han Y, Cheng X, Li M, Jiang M, Peng Z, Tao L, Meng J. Pyroptosis-related signatures predict immune characteristics and prognosis in IPF. Heliyon 2024; 10:e23683. [PMID: 38192798 PMCID: PMC10772192 DOI: 10.1016/j.heliyon.2023.e23683] [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: 04/11/2023] [Revised: 11/28/2023] [Accepted: 12/09/2023] [Indexed: 01/10/2024] Open
Abstract
The purpose of this work was to use integrated bioinformatics analysis to screen for pyroptosis-related genes (PRGs) and possible immunological phenotypes linked to the development and course of IPF. Transcriptome sequencing datasets GSE70866, GSE47460 and GSE150910 were obtained from GEO database. From the GSE70866 database, 34 PRGs with differential expression were found in IPF as compared to healthy controls. In addition, a diagnostic model containing 4 genes PRGs (CAMP, MKI67, TCEA3 and USP24) was constructed based on LASSO logistic regression. The diagnostic model showed good predictive ability to differentiate between IPF and healthy, with ROC-AUC ranging from 0.910 to 0.997 in GSE70866 and GSE150910 datasets. Moreover, based on a combined cohort of the Freiburg and the Siena cohorts from GSE70866 dataset, we identified ten PRGs that might predict prognosis for IPF. We constructed a prognostic model that included eight PRGs (CLEC5A, TREM2, MMP1, IRF2, SEZ6L2, ADORA3, NOS2, USP24) by LASSO Cox regression and validated it in the Leuven cohort. The risk model divided IPF patients from the combined cohort into high-risk and low-risk subgroups. There were significant differences between the two subgroups in terms of IPF survival and GAP stage. There is a close correlation between leukocyte migration, plasma membrane junction, and poor prognosis in a high-risk subgroup. Furthermore, a high-risk score was associated with more plasma cells, activated NK cells, monocytes, and activated mast cells. Additionally, we identified HDAC inhibitors in the cMAP database that might be therapeutic for IPF. To summarize, pyroptosis and its underlying immunological features are to blame for the onset and progression of IPF. PRG-based predictive models and drugs may offer new treatment options for IPF.
Collapse
Affiliation(s)
- Yijun He
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Tingting Yao
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Yan Zhang
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Lingzhi Long
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Guoliang Jiang
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Xiangyu Zhang
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Xin Lv
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanyuan Han
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyun Cheng
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Mengyu Li
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Mao Jiang
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhangzhe Peng
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
- National International Collaborative Research Center for Medical Metabolomics, Changsha, China
| | - Lijian Tao
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
- National International Collaborative Research Center for Medical Metabolomics, Changsha, China
| | - Jie Meng
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
- National International Collaborative Research Center for Medical Metabolomics, Changsha, China
| |
Collapse
|
13
|
Hirata M, Hara Y, Fujii H, Murohashi K, Saigusa Y, Zhao S, Kobayashi M, Nagasawa R, Tagami Y, Izawa A, Otsu Y, Watanabe K, Horita N, Kobayashi N, Kaneko T. ILD-GAP combined with the monocyte ratio could be a better prognostic prediction model than ILD-GAP in patients with interstitial lung diseases. BMC Pulm Med 2024; 24:16. [PMID: 38183005 PMCID: PMC10768524 DOI: 10.1186/s12890-023-02833-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 12/28/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND The ILD-GAP scoring system is known to be useful in predicting prognosis in patients with interstitial lung disease (ILD). An elevated monocyte count was associated with increased risks of IPF poor prognosis. We examined whether the ILD-GAP scoring system combined with the monocyte ratio (ILD-GAPM) is superior to the conventional ILD-GAP model in predicting ILD prognosis. METHODS In patients with ILD treated between April 2013 and April 2017, we were retrospectively assessed the relationships between baseline clinical parameters, including age, sex, Charlson Comorbidity Index score (CCIS), ILD diagnosis, blood biomarkers, pulmonary function test results, and disease outcomes. In ILD patients were included idiopathic pulmonary fibrosis (IPF), idiopathic nonspecific interstitial pneumonia (iNSIP), collagen vascular disease-related interstitial pneumonia (CVD-IP), chronic hypersensitivity pneumonitis (CHP), and unclassifiable ILD (UC-ILD). We also assessed the ability to predict prognosis was compared between the ILD-GAP and ILD-GAPM models. RESULTS A total of 179 patients (mean age, 73 years) were assessed. All of them were taken pulmonary function test, including percentage predicted diffusion capacity for carbon monoxide. ILD patients included 56 IPF cases, 112 iNSIP and CVD-IP cases, 6 CHP cases and 5 UC-ILD cases. ILD-GAPM provided a greater area under the receiver-operating characteristic curve (0.747) than ILD-GAP (0.710) for predicting 3-year ILD-related events. Furthermore, the log-rank test showed that the Kaplan-Meier curves in ILD-GAPM were significantly different by stage (P = 0.015), but not by stage in ILD-GAP (P = 0.074). CONCLUSIONS The ILD-GAPM model may be a more accurate predictor of prognosis for ILD patients than the ILD-GAP model.
Collapse
Affiliation(s)
- Momo Hirata
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Yu Hara
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan.
| | - Hiroaki Fujii
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Kota Murohashi
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Yusuke Saigusa
- Department of Biostatistics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shiqi Zhao
- Department of Biostatistics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Miyu Kobayashi
- Department of Biostatistics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryo Nagasawa
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Yoichi Tagami
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Ami Izawa
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Yukiko Otsu
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Keisuke Watanabe
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Nobuyuki Horita
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Nobuaki Kobayashi
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Takeshi Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| |
Collapse
|
14
|
Yang L, Xiang Z, Dai M, Zhang Q, Zhou Y. Prognosis of Lung Transplantation in Patients with Acute Exacerbations of Interstitial Lung Disease: A Meta-Analysis Based on Cohort Studies. Ann Thorac Cardiovasc Surg 2024; 30:24-00086. [PMID: 38972753 PMCID: PMC11237979 DOI: 10.5761/atcs.ra.24-00086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 06/14/2024] [Indexed: 07/09/2024] Open
Abstract
PURPOSE This meta-analysis aimed to examine the prognosis of patients with acute exacerbation of interstitial lung disease (AE-ILD) treated with lung transplantation compared to those with stable interstitial lung disease (ILD). METHODS We conducted a detailed search in PubMed, Embase, Web of Science, and the Cochrane Library, with the primary outcomes being overall survival (OS), acute cellular rejection (ACR), primary graft dysfunction (PGD), and length of stay (LOS). RESULTS Five cohort studies were included in this meta-analysis, with 183 patients enrolled in the AE-ILD group and 337 patients in the stable-ILD group. The results showed that in regard to perioperative outcomes, the AE-ILD group did not differ from the stable-ILD group in the incidence of ACR (relative risks [RR] = 0.34, p = 0.44) and the incidence of PGD Ⅲ (RR = 0.53, p = 0.43), but had a longer LOS (mean difference = 9.15, p = 0.02). Regarding prognosis, the two also did not differ in 90-day OS (RR = 0.97, p = 0.59), 1-year OS (RR = 1.05, p = 0.66), and 3-year OS (RR = 0.91, p = 0.76). CONCLUSION Our study concluded that the efficacy of lung transplantation in patients with AE-ILD is not inferior to that of patients with stable ILD. Lung transplantation is one of the potential treatments for patients with AE-ILD.
Collapse
Affiliation(s)
- Lei Yang
- Emergency Medical Center, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, China
| | - Zhiyi Xiang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Min Dai
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qiufeng Zhang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ying Zhou
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
15
|
Jannini-Sá YAP, Creyns B, Hogaboam CM, Parks WC, Hohmann MS. Macrophages in Lung Repair and Fibrosis. Results Probl Cell Differ 2024; 74:257-290. [PMID: 39406909 DOI: 10.1007/978-3-031-65944-7_10] [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] [Indexed: 11/01/2024]
Abstract
Macrophages are key regulators of tissue repair and fibrosis. Following injury, macrophages undergo marked phenotypic and functional changes to play crucial roles throughout the phases of tissue repair. Idiopathic Pulmonary Fibrosis, which is the most common fibrosing lung disease, has been described as an aberrant reparative response to repetitive alveolar epithelial injury in a genetically susceptible aging individual. The marked destruction of the lung architecture results from the excessive secretion of extracellular matrix by activated fibroblasts and myofibroblasts. Accumulating evidence suggests that macrophages have a pivotal regulatory role in pulmonary fibrosis. The origins and characteristics of macrophages in the lung and their role in regulating lung homeostasis, repair, and fibrosis are reviewed herein. We discuss recent studies that have employed single-cell RNA-sequencing to improve the identification and characterization of macrophage populations in the context of homeostatic and fibrotic conditions. We also discuss the current understanding of the macrophage-mediated mechanisms underlying the initiation and progression of pulmonary fibrosis, with a focus on the phenotypic and functional changes that aging macrophages acquire and how these changes ultimately contribute to age-related chronic lung diseases.
Collapse
Affiliation(s)
- Yago A P Jannini-Sá
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Brecht Creyns
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Cory M Hogaboam
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - William C Parks
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Miriam S Hohmann
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
16
|
Tagami Y, Hara Y, Murohashi K, Nagasawa R, Fujii H, Izawa A, Yabe A, Saigusa Y, Kobayashi M, Shiida M, Hirata M, Otsu Y, Watanabe K, Horita N, Kobayashi N, Kaneko T. Serum heme oxygenase-1 as a prognostic biomarker in patients with acute exacerbation of interstitial lung disease. Sci Rep 2023; 13:22639. [PMID: 38114539 PMCID: PMC10730846 DOI: 10.1038/s41598-023-49342-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023] Open
Abstract
Serum heme oxygenase (HO)-1 level has been reported as a clinically reliable diagnostic biomarker for acute exacerbation of interstitial lung disease (ILD); however, its utility for predicting mortality among these patients is unclear. Serum HO-1 levels of patients newly diagnosed with acute exacerbation of ILD were measured at the time of initiating steroid pulse therapy. The relationship between serum HO-1 and various other serum biomarkers, change in HRCT findings, and disease prognosis at 12 weeks after diagnosis of acute exacerbation was evaluated in 51 patients, of whom 17 (33%) had idiopathic pulmonary fibrosis (IPF). Serum HO-1 was higher in patients with acute exacerbation of IPF than in patients with acute exacerbation of other ILDs. Serum HO-1 levels were higher in patients who died within these 12 weeks than in survivors. Among age, sex, comorbidities, IPF diagnosis, HRCT findings, and blood biomarkers, serum HO-1 was a primary predictor of 12-week mortality. In 41 patients who underwent repeat HRCT, serum HO-1 was higher in patients with honeycomb progression than in those without. Serum HO-1 measurement could be useful for evaluating disease mortality and morbidity of patients with acute exacerbation of ILDs.
Collapse
Affiliation(s)
- Yoichi Tagami
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Yu Hara
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan.
| | - Kota Murohashi
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Ryo Nagasawa
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Hiroaki Fujii
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Ami Izawa
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Aya Yabe
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Yusuke Saigusa
- Department of Biostatistics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Miyu Kobayashi
- Department of Biostatistics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masafumi Shiida
- Research and Development Division, Minaris Medical Co., Ltd, 600-1 Minami-Ishiki, Nagaizumi-Cho, Sunto-Gun, Shizuoka, 411-0932, Japan
| | - Momo Hirata
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Yukiko Otsu
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Keisuke Watanabe
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Nobuyuki Horita
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Nobuaki Kobayashi
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Takeshi Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| |
Collapse
|
17
|
Kim HH, Kim HC, Shim TS, Ahn JH, Huh JW, Hong SB, Lee GD, Kim DK, Park SI, Choi S. Clinical impact of pre-existing acute exacerbation in patients with interstitial lung disease who underwent lung transplantation. Respir Res 2023; 24:307. [PMID: 38062465 PMCID: PMC10701919 DOI: 10.1186/s12931-023-02614-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Acute exacerbation of interstitial lung disease (AE-ILD) significantly impacts prognosis, leading to high mortality rates. Although lung transplantation is a life-saving treatment for selected patients with ILD, its outcomes in those presenting with AE-ILD have yielded conflicting results compared with those with stable ILD. This study aims to investigate the impact of pre-existing AE on the prognosis of ILD patients who underwent lung transplantation. METHOD We conducted a single-center retrospective study by reviewing the medical records of 108 patients who underwent lung transplantation for predisposing ILD at Asan Medical Center, Seoul, South Korea, between 2008 and 2022. The primary objective was to compare the survival of patients with AE-ILD at the time of transplantation with those without AE-ILD. RESULTS Among the 108 patients, 52 (48.1%) experienced AE-ILD at the time of lung transplantation, and 81 (75.0%) required pre-transplant mechanical ventilation. Although the type of ILD (IPF vs. non-IPF ILD) did not affect clinical outcomes after transplantation, AE-ILD was associated with worse survival outcomes. The survival probabilities at 90 days, 1 year, and 3 years post-transplant for patients with AE-ILD were 86.5%, 73.1%, and 60.1%, respectively, while those for patients without AE-ILD were higher, at 92.9%, 83.9%, and 79.6% (p = 0.032). In the multivariable analysis, pre-existing AE was an independent prognostic factor for mortality in ILD patients who underwent lung transplantation. CONCLUSIONS Although lung transplantation remains an effective treatment option for ILD patients with pre-existing AE, careful consideration is needed, especially in patients requiring pre-transplant mechanical respiratory support.
Collapse
Affiliation(s)
- Hyeon Hwa Kim
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ho Cheol Kim
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Tae Sun Shim
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jee Hwan Ahn
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Won Huh
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Bum Hong
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Geun Dong Lee
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Dong Kwan Kim
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Seung-Il Park
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Sehoon Choi
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
18
|
Koudstaal T, Funke-Chambour M, Kreuter M, Molyneaux PL, Wijsenbeek MS. Pulmonary fibrosis: from pathogenesis to clinical decision-making. Trends Mol Med 2023; 29:1076-1087. [PMID: 37716906 DOI: 10.1016/j.molmed.2023.08.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 09/18/2023]
Abstract
Pulmonary fibrosis (PF) encompasses a spectrum of chronic lung diseases that progressively impact the interstitium, resulting in compromised gas exchange, breathlessness, diminished quality of life (QoL), and ultimately respiratory failure and mortality. Various diseases can cause PF, with their underlying causes primarily affecting the lung interstitium, leading to their referral as interstitial lung diseases (ILDs). The current understanding is that PF arises from abnormal wound healing processes triggered by various factors specific to each disease, leading to excessive inflammation and fibrosis. While significant progress has been made in understanding the molecular mechanisms of PF, its pathogenesis remains elusive. This review provides an in-depth exploration of the latest insights into PF pathophysiology, diagnosis, treatment, and future perspectives.
Collapse
Affiliation(s)
- Thomas Koudstaal
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Manuela Funke-Chambour
- Department of Pulmonary Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Michael Kreuter
- Mainz Center for Pulmonary Medicine, Departments of Pneumology, Mainz University Medical Center and of Pulmonary, Critical Care & Sleep Medicine, Marienhaus Clinic Mainz, Mainz, Germany
| | - Philip L Molyneaux
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Marlies S Wijsenbeek
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
19
|
Larson-Casey JL, Saleem K, Surolia R, Pandey J, Mack M, Antony VB, Bodduluri S, Bhatt SP, Duncan SR, Carter AB. Myeloid Heterogeneity Mediates Acute Exacerbations of Pulmonary Fibrosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1714-1724. [PMID: 37782053 PMCID: PMC10843506 DOI: 10.4049/jimmunol.2300053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 09/15/2023] [Indexed: 10/03/2023]
Abstract
Epidemiological evidence indicates that exposure to particulate matter is linked to the development of idiopathic pulmonary fibrosis (IPF) and increases the incidence of acute exacerbations of IPF. In addition to accelerating the rate of lung function decline, exposure to fine particulate matter (particulate matter smaller than 2.5 μm [PM2.5]) is a risk factor for increased mortality in subjects with IPF. In this article, we show that exposure to PM2.5 mediates monocyte recruitment and fibrotic progression in mice with established fibrosis. In mice with established fibrosis, bronchoalveolar lavage cells showed monocyte/macrophage heterogeneity after exposure to PM2.5. These cells had a significant inflammatory and anti-inflammatory signature. The mixed heterogeneity of cells contributed to the proinflammatory and anti-inflammatory response. Although monocyte-derived macrophages were recruited to the lung in bleomycin-injured mice treated with PM2.5, recruitment of monocytes expressing Ly6Chi to the lung promoted progression of fibrosis, reduced lung aeration on computed tomography, and impacted lung compliance. Ly6Chi monocytes isolated from PM2.5-exposed fibrotic mice showed enhanced expression of proinflammatory markers compared with fibrotic mice exposed to vehicle. Moreover, IPF bronchoalveolar lavage cells treated ex vivo with PM2.5 showed an exaggerated inflammatory response. Targeting Ly6Chi monocyte recruitment inhibited fibrotic progression in mice. Moreover, the adoptive transfer of Ly6Chi monocytes exacerbated established fibrosis. These observations suggest that enhanced recruitment of Ly6Chi monocytes with a proinflammatory phenotype mediates acute exacerbations of pulmonary fibrosis, and targeting these cells may provide a potential novel therapeutic target to protect against acute exacerbations of IPF.
Collapse
Affiliation(s)
- Jennifer L. Larson-Casey
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Komal Saleem
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ranu Surolia
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jyotsana Pandey
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Matthias Mack
- Department of Nephrology, University of Regensburg, Regensburg, Germany
| | - Veena B. Antony
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sandeep Bodduluri
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- UAB Lung Imaging Lab, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Surya P. Bhatt
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- UAB Lung Imaging Lab, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Steven R. Duncan
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - A. Brent Carter
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Birmingham Veterans Administration Medical Center, Birmingham. AL, USA
| |
Collapse
|
20
|
Ye X, Zhang M, Gu H, Liu M, Zhao Y, Shi Y, Wu S, Jiang C, Ye X, Zhu H, Li Q, Huang X, Cao M. Animal models of acute exacerbation of pulmonary fibrosis. Respir Res 2023; 24:296. [PMID: 38007420 PMCID: PMC10675932 DOI: 10.1186/s12931-023-02595-z] [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: 07/05/2023] [Accepted: 11/07/2023] [Indexed: 11/27/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive scarring interstitial lung disease with an unknown cause. Some patients may experience acute exacerbations (AE), which result in severe lung damage visible on imaging or through examination of tissue samples, often leading to high mortality rates. However, the etiology and pathogenesis of AE-IPF remain unclear. AE-IPF patients exhibit diffuse lung damage, apoptosis of type II alveolar epithelial cells, and an excessive inflammatory response. Establishing a reliable animal model of AE is critical for investigating the pathogenesis. Recent studies have reported a variety of animal models for AE-IPF, each with its own advantages and disadvantages. These models are usually established in mice with bleomycin-induced pulmonary fibrosis, using viruses, bacteria, small peptides, or specific drugs. In this review, we present an overview of different AE models, hoping to provide a useful resource for exploring the mechanisms and targeted therapies for AE-IPF.
Collapse
Affiliation(s)
- Xu Ye
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Mingrui Zhang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huimin Gu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Mengying Liu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Yichao Zhao
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanchen Shi
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shufei Wu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Cheng Jiang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoling Ye
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Huihui Zhu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qi Li
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinmei Huang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
- Nanjing Institute of Respiratory Diseases, Nanjing, China.
| | - Mengshu Cao
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Nanjing Institute of Respiratory Diseases, Nanjing, China.
| |
Collapse
|
21
|
Young ON, Bourke JE, Widdop RE. Catch your breath: The protective role of the angiotensin AT 2 receptor for the treatment of idiopathic pulmonary fibrosis. Biochem Pharmacol 2023; 217:115839. [PMID: 37778444 DOI: 10.1016/j.bcp.2023.115839] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 10/03/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease whereby excessive deposition of extracellular matrix proteins (ECM) ultimately leads to respiratory failure. While there have been advances in pharmacotherapies for pulmonary fibrosis, IPF remains an incurable and irreversible disease. There remains an unmet clinical need for treatments that reverse fibrosis, or at the very least have a more tolerable side effect profile than currently available treatments. Transforming growth factor β1(TGFβ1) is considered the main driver of fibrosis in IPF. However, as our understanding of the role of the pulmonary renin-angiotensin system (PRAS) in the pathogenesis of IPF increases, it is becoming clear that targeting angiotensin receptors represents a potential novel treatment strategy for IPF - in particular, via activation of the anti-fibrotic angiotensin type 2 receptor (AT2R). This review describes the current understanding of the pathophysiology of IPF and the mediators implicated in its pathogenesis; focusing on TGFβ1, angiotensin II and related peptides in the PRAS and their contribution to fibrotic processes in the lung. Preclinical and clinical assessment of currently available AT2R agonists and the development of novel, highly selective ligands for this receptor will also be described, with a focus on compound 21, currently in clinical trials for IPF. Collectively, this review provides evidence of the potential of AT2R as a novel therapeutic target for IPF.
Collapse
Affiliation(s)
- Olivia N Young
- Department of Pharmacology and Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Jane E Bourke
- Department of Pharmacology and Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Robert E Widdop
- Department of Pharmacology and Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
22
|
Senoo S, Higo H, Taniguchi A, Kiura K, Maeda Y, Miyahara N. Pulmonary fibrosis and type-17 immunity. Respir Investig 2023; 61:553-562. [PMID: 37356133 DOI: 10.1016/j.resinv.2023.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/17/2023] [Accepted: 05/12/2023] [Indexed: 06/27/2023]
Abstract
Fibrosis of the lung can occur in idiopathic pulmonary fibrosis, collagen vascular diseases, and hypersensitivity pneumonitis, among other diseases. Transforming growth factor (TGF)-β, vascular epithelial growth factor, fibroblast growth factor, and platelet-derived growth factor contribute to the pathophysiology of fibrosis. TGF-β and other cytokines, including interleukin (IL)-1β, IL-6, and IL-23, activate type-17 immunity, which is involved in pulmonary fibrosis. The components of type-17 immunity include type-17 helper T cells, γδT cells, IL-17A-producing CD8-positive T cells, invariant NKT cells, and group 3 innate lymphoid cells. IL-17A, the main cytokine of type-17 immunity, is able to induce the epithelial-mesenchymal transition in epithelial cells via a production of TGF-β, directly stimulate fibroblasts and fibrocytes, and inhibit autophagy, which otherwise protects against pulmonary fibrosis. IL-23 induces type-17 immunity and plays an important role in the acute exacerbation of pulmonary fibrosis. Clinical studies have also linked type-17 immunity to the pathogenesis of pulmonary fibrosis. Consequently, targeting type-17 immunity may serve as a new therapeutic strategy to prevent the development or exacerbation of pulmonary fibrosis.
Collapse
Affiliation(s)
- Satoru Senoo
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Academic Field of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Hisao Higo
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan
| | - Akihiko Taniguchi
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Academic Field of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Katsuyuki Kiura
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan
| | - Yoshinobu Maeda
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Academic Field of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Nobuaki Miyahara
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan; Department of Medical Technology, Okayama University Academic Field of Health Sciences, Okayama, Japan.
| |
Collapse
|
23
|
Koudstaal T, Wijsenbeek MS. Idiopathic pulmonary fibrosis. Presse Med 2023; 52:104166. [PMID: 37156412 DOI: 10.1016/j.lpm.2023.104166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/14/2023] [Accepted: 05/02/2023] [Indexed: 05/10/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive devastating lung disease with substantial morbidity. It is associated with cough, dyspnea and impaired quality of life. If left untreated, IPF has a median survival of 3 years. IPF affects ∼3 million people worldwide, with increasing incidence in older patients. The current concept of pathogenesis is that pulmonary fibrosis results from repetitive injury to the lung epithelium, with fibroblast accumulation, myofibroblast activation, and deposition of matrix. These injuries, in combination with innate and adaptive immune responses, dysregulated wound repair and fibroblast dysfunction, lead to recurring tissue remodeling and self-perpetuating fibrosis as seen in IPF. The diagnostic approach includes the exclusion of other interstitial lung diseases or underlying conditions and depends on a multidisciplinary team-based discussion combining radiological and clinical features and well as in some cases histology. In the last decade, considerable progress has been made in the understanding of IPF clinical management, with the availability of two drugs, pirfenidone and nintedanib, that decrease pulmonary lung function decline. However, current IPF therapies only slow disease progression and prognosis remains poor. Fortunately, there are multiple clinical trials ongoing with potential new therapies targeting different disease pathways. This review provides an overview of IPF epidemiology, current insights in pathophysiology, diagnostic and therapeutic management approaches. Finally, a detailed description of current and evolving therapeutic approaches is also provided.
Collapse
Affiliation(s)
- Thomas Koudstaal
- Center for Interstitial Lung Diseases and Sarcoidosis, Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Marlies S Wijsenbeek
- Center for Interstitial Lung Diseases and Sarcoidosis, Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
24
|
Liu Y, Zhao J, Adams TS, Wang N, Schupp JC, Wu W, McDonough JE, Chupp GL, Kaminski N, Wang Z, Yan X. iDESC: identifying differential expression in single-cell RNA sequencing data with multiple subjects. BMC Bioinformatics 2023; 24:318. [PMID: 37608264 PMCID: PMC10463720 DOI: 10.1186/s12859-023-05432-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 07/18/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Single-cell RNA sequencing (scRNA-seq) technology has enabled assessment of transcriptome-wide changes at single-cell resolution. Due to the heterogeneity in environmental exposure and genetic background across subjects, subject effect contributes to the major source of variation in scRNA-seq data with multiple subjects, which severely confounds cell type specific differential expression (DE) analysis. Moreover, dropout events are prevalent in scRNA-seq data, leading to excessive number of zeroes in the data, which further aggravates the challenge in DE analysis. RESULTS We developed iDESC to detect cell type specific DE genes between two groups of subjects in scRNA-seq data. iDESC uses a zero-inflated negative binomial mixed model to consider both subject effect and dropouts. The prevalence of dropout events (dropout rate) was demonstrated to be dependent on gene expression level, which is modeled by pooling information across genes. Subject effect is modeled as a random effect in the log-mean of the negative binomial component. We evaluated and compared the performance of iDESC with eleven existing DE analysis methods. Using simulated data, we demonstrated that iDESC had well-controlled type I error and higher power compared to the existing methods. Applications of those methods with well-controlled type I error to three real scRNA-seq datasets from the same tissue and disease showed that the results of iDESC achieved the best consistency between datasets and the best disease relevance. CONCLUSIONS iDESC was able to achieve more accurate and robust DE analysis results by separating subject effect from disease effect with consideration of dropouts to identify DE genes, suggesting the importance of considering subject effect and dropouts in the DE analysis of scRNA-seq data with multiple subjects.
Collapse
Affiliation(s)
- Yunqing Liu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, 06520, USA
| | - Jiayi Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, 06520, USA
| | - Taylor S Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Ningya Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, 06520, USA
| | - Jonas C Schupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-Stage and Obstructive Lung Disease Hannover, German Center for Lung Research (DZL), Hannover, Germany
| | - Weimiao Wu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, 06520, USA
- Meta Platforms, Inc, Cambridge, USA
| | - John E McDonough
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Geoffrey L Chupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Zuoheng Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, 06520, USA.
| | - Xiting Yan
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, 06520, USA.
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
25
|
Mottais A, Riberi L, Falco A, Soccal S, Gohy S, De Rose V. Epithelial-Mesenchymal Transition Mechanisms in Chronic Airway Diseases: A Common Process to Target? Int J Mol Sci 2023; 24:12412. [PMID: 37569787 PMCID: PMC10418908 DOI: 10.3390/ijms241512412] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a reversible process, in which epithelial cells lose their epithelial traits and acquire a mesenchymal phenotype. This transformation has been described in different lung diseases, such as lung cancer, interstitial lung diseases, asthma, chronic obstructive pulmonary disease and other muco-obstructive lung diseases, such as cystic fibrosis and non-cystic fibrosis bronchiectasis. The exaggerated chronic inflammation typical of these pulmonary diseases can induce molecular reprogramming with subsequent self-sustaining aberrant and excessive profibrotic tissue repair. Over time this process leads to structural changes with progressive organ dysfunction and lung function impairment. Although having common signalling pathways, specific triggers and regulation mechanisms might be present in each disease. This review aims to describe the various mechanisms associated with fibrotic changes and airway remodelling involved in chronic airway diseases. Having better knowledge of the mechanisms underlying the EMT process may help us to identify specific targets and thus lead to the development of novel therapeutic strategies to prevent or limit the onset of irreversible structural changes.
Collapse
Affiliation(s)
- Angélique Mottais
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (A.M.); (S.G.)
| | - Luca Riberi
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Andrea Falco
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Simone Soccal
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Sophie Gohy
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (A.M.); (S.G.)
- Department of Pneumology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Cystic Fibrosis Reference Centre, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Virginia De Rose
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| |
Collapse
|
26
|
Salonen J, Jansa S, Vähänikkilä H, Kaarteenaho R. Re-hospitalisation predicts poor prognosis after acute exacerbation of interstitial lung disease. BMC Pulm Med 2023; 23:236. [PMID: 37393286 DOI: 10.1186/s12890-023-02534-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/24/2023] [Indexed: 07/03/2023] Open
Abstract
BACKGROUND Several markers have been identified to increase the risk for acute exacerbation of interstitial lung disease (AE-ILD) or mortality related to AE-ILD. However, less is known about the risk predictors of ILD patients who have survived AE. The aim of the study was to characterise AE-ILD survivors and investigate prognostic factors in this subpopulation. METHODS All AE-ILD patients (n = 95) who had been discharged alive from two hospitals located in Northern Finland were selected from a population of 128 AE-ILD patients. Clinical data related to the hospital treatment and six-month follow-up visit were collected retrospectively from medical records. RESULTS Fifty-three patients with idiopathic pulmonary fibrosis (IPF) and 42 patients with other ILD were identified. Two thirds of the patients had been treated without invasive or non-invasive ventilation support. The clinical features of six-month survivors (n = 65) and non-survivors (n = 30) did not differ in terms of medical treatment or oxygen requirements. Of the patients, 82.5% used corticosteroids at the six-month follow-up visit. Fifty-two patients experienced at least one non-elective respiratory re-hospitalisation before the six-month follow-up visit. In a univariate model, IPF diagnosis, high age and a non-elective respiratory re-hospitalisation increased the risk of death, although re-hospitalisation was the only independent risk factor in a multivariate model. In six-month survivors, there was no statistically significant decrease in pulmonary function test results (PFT) examined at the follow-up visit compared with earlier PFT examined near the time of AE-ILD. CONCLUSIONS The AE-ILD survivors were a heterogeneous group of patients both clinically and in terms of their outcome. A non-elective respiratory re-hospitalisation was identified as a marker of poor prognosis among AE-ILD survivors.
Collapse
Affiliation(s)
- Johanna Salonen
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulun Yliopisto, P.O. Box 8000, 90014, Oulu, Finland.
- Center of Internal and Respiratory Medicine and Medical Research Center (MRC) Oulu, Oulu University Hospital, Oulu, Finland.
| | - Sanna Jansa
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulun Yliopisto, P.O. Box 8000, 90014, Oulu, Finland
| | - Hannu Vähänikkilä
- Northern Finland Birth Cohorts, Arctic Biobank, Infrastructure for Population Studies, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Riitta Kaarteenaho
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulun Yliopisto, P.O. Box 8000, 90014, Oulu, Finland
- Center of Internal and Respiratory Medicine and Medical Research Center (MRC) Oulu, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
27
|
Wang Q, Xie Z, Wan N, Yang L, Jin Z, Jin F, Huang Z, Chen M, Wang H, Feng J. Potential biomarkers for diagnosis and disease evaluation of idiopathic pulmonary fibrosis. Chin Med J (Engl) 2023; 136:1278-1290. [PMID: 37130223 PMCID: PMC10309524 DOI: 10.1097/cm9.0000000000002171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Indexed: 05/04/2023] Open
Abstract
ABSTRACT Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease characterized by progressive lung fibrogenesis and histological features of usual interstitial pneumonia. IPF has a poor prognosis and presents a spectrum of disease courses ranging from slow evolving disease to rapid deterioration; thus, a differential diagnosis remains challenging. Several biomarkers have been identified to achieve a differential diagnosis; however, comprehensive reviews are lacking. This review summarizes over 100 biomarkers which can be divided into six categories according to their functions: differentially expressed biomarkers in the IPF compared to healthy controls; biomarkers distinguishing IPF from other types of interstitial lung disease; biomarkers differentiating acute exacerbation of IPF from stable disease; biomarkers predicting disease progression; biomarkers related to disease severity; and biomarkers related to treatment. Specimen used for the diagnosis of IPF included serum, bronchoalveolar lavage fluid, lung tissue, and sputum. IPF-specific biomarkers are of great clinical value for the differential diagnosis of IPF. Currently, the physiological measurements used to evaluate the occurrence of acute exacerbation, disease progression, and disease severity have limitations. Combining physiological measurements with biomarkers may increase the accuracy and sensitivity of diagnosis and disease evaluation of IPF. Most biomarkers described in this review are not routinely used in clinical practice. Future large-scale multicenter studies are required to design and validate suitable biomarker panels that have diagnostic utility for IPF.
Collapse
Affiliation(s)
- Qing Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Zhaoliang Xie
- Respiratory Department of Sanming Yong’an General Hospital, Sanming, Fujian 366000, China
| | - Nansheng Wan
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Lei Yang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhixian Jin
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Fang Jin
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhaoming Huang
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Min Chen
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Huiming Wang
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Jing Feng
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
28
|
Cha SR, Jang J, Park SM, Ryu SM, Cho SJ, Yang SR. Cigarette Smoke-Induced Respiratory Response: Insights into Cellular Processes and Biomarkers. Antioxidants (Basel) 2023; 12:1210. [PMID: 37371940 DOI: 10.3390/antiox12061210] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Cigarette smoke (CS) poses a significant risk factor for respiratory, vascular, and organ diseases owing to its high content of harmful chemicals and reactive oxygen species (ROS). These substances are known to induce oxidative stress, inflammation, apoptosis, and senescence due to their exposure to environmental pollutants and the presence of oxidative enzymes. The lung is particularly susceptible to oxidative stress. Persistent oxidative stress caused by chronic exposure to CS can lead to respiratory diseases such as chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), and lung cancer. Avoiding exposure to environmental pollutants, like cigarette smoke and air pollution, can help mitigate oxidative stress. A comprehensive understanding of oxidative stress and its impact on the lungs requires future research. This includes identifying strategies for preventing and treating lung diseases as well as investigating the underlying mechanisms behind oxidative stress. Thus, this review aims to investigate the cellular processes induced by CS, specifically inflammation, apoptosis, senescence, and their associated biomarkers. Furthermore, this review will delve into the alveolar response provoked by CS, emphasizing the roles of potential therapeutic target markers and strategies in inflammation and oxidative stress.
Collapse
Affiliation(s)
- Sang-Ryul Cha
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon 24341, Republic of Korea
| | - Jimin Jang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon 24341, Republic of Korea
| | - Sung-Min Park
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon 24341, Republic of Korea
| | - Se Min Ryu
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon 24341, Republic of Korea
| | - Seong-Joon Cho
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon 24341, Republic of Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon 24341, Republic of Korea
| |
Collapse
|
29
|
Kurniawan SV, Louisa M, Zaini J, Surini S, Soetikno V, Wuyung PE, Uli RCT. Acute exacerbation of idiopathic pulmonary fibrosis model in the rats using bleomycin and lipopolysaccharides. J Adv Vet Anim Res 2023; 10:196-204. [PMID: 37534065 PMCID: PMC10390678 DOI: 10.5455/javar.2023.j669] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 08/04/2023] Open
Abstract
Objective This study was conducted to establish a rat model of acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF) using the combination of bleomycin (BLM) and lipopolysaccharides (LPS). Materials and Method Twenty-four male Sprague Dawley rats were allocated into two equal groups: the sham or the bleomycin and lipopolysaccharides-induced AE-IPF group (BLM-LPS). On Day 7, BLM intratracheally and LPS intraperitoneally were both used to administer AE-IPF. The BLM-LPS group and its respective sham group were terminated on Days 8, 14, or 21. Samples of bronchoalveolar lavage fluid (BALF) and lungs were taken and investigated for cell count and histopathology. Results On Day 8, histological analysis revealed inflammatory cell infiltration with edema and hyaline membrane, and the BALF differential cell count revealed high neutrophil counts. By having a higher collagen density area and Ashcroft modified score than the sham group on Day 14, the BLM-LPS group displayed significantly lower oxygen saturation, alveolar air area, and a fibrotic appearance. However, there was a spontaneous resolution in inflammation and fibrotic appearance on Day 21 after the BLM administration. Conclusions By combining BLM and LPS, it was possible to create a successful rat model of AE-IPF. The present model showed the peak exacerbation on Day 8 and the fibrotic peak on Day 14, which gradually improved. The optimal time for the new AE-IPF therapeutic intervention was determined to be between Days 8 and 14.
Collapse
Affiliation(s)
- Sandy Vitria Kurniawan
- Doctoral Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Department of Pharmacology and Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Melva Louisa
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Jamal Zaini
- Department of Pulmonology and Respiratory Medicine Faculty of Medicine Universitas Indonesia, Persahabatan National Respiratory Referral Hospital, Jakarta, Indonesia
| | - Silvia Surini
- Laboratory of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Indonesia, Depok, Indonesia
| | - Vivian Soetikno
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Puspita Eka Wuyung
- Department of Anatomical Pathology, Faculty of Medicine Universitas Indonesia, Depok, Indonesia
- Animal Research Facilities, Indonesian Medical Education and Research Institute, Faculty of Medicine Universitas Indonesia, Depok, Indonesia
| | - Rosemary Ceria Tatap Uli
- Animal Research Facilities, Indonesian Medical Education and Research Institute, Faculty of Medicine Universitas Indonesia, Depok, Indonesia
| |
Collapse
|
30
|
Zinellu A, Fois AG, Pirina P, Carru C, Mangoni AA. A Systematic Review and Meta-analysis of Clinical, Respiratory, and Biochemical Risk Factors for Acute Exacerbation of idiopathic Pulmonary Fibrosis. Arch Med Res 2023:S0188-4409(23)00058-9. [PMID: 37137756 DOI: 10.1016/j.arcmed.2023.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/20/2023] [Accepted: 04/18/2023] [Indexed: 05/05/2023]
Abstract
BACKGROUND A better capacity to identify patients with idiopathic pulmonary fibrosis (IPF) at risk of acute exacerbation (AEIPF) might improve outcomes and reduce healthcare costs. AIMS We critically appraised the available evidence of the differences in clinical, respiratory, and biochemical parameters between AEIPF and IPF patients with stable disease (SIPF) by conducting a systematic review and meta-analysis. METHODS PubMed, Web of Science and Scopus were reviewed up until August 1, 2022, for studies reporting differences in clinical, respiratory, and biochemical parameters (including investigational biomarkers) between AEIPF and SIPF patients. The Joanna Briggs Institute Critical Appraisal Checklist was used to assess the risk of bias. RESULTS Twenty-nine cross-sectional studies published between 2010 and 2022 were identified (all with a low risk of bias). Of the 32 meta-analysed parameters, significant differences were observed between groups, assessed through standard mean differences or relative ratios, with age, forced vital capacity, vital capacity, carbon monoxide diffusion capacity, total lung capacity, oxygen partial pressure, alveolar-arterial oxygen gradient, P/F ratio, 6 min walk test distance, C-reactive protein, lactate dehydrogenase, white blood cell count, albumin, Krebs von den Lungen 6, surfactant protein D, high mobility group box 1 protein, and interleukin-1β, 6, and 8. CONCLUSIONS We identified significant differences between AEIPF and SIPF patients in age and specific parameters of respiratory function, inflammation, and epithelial lung damage. Prospective studies are warranted to determine the capacity of these parameters to predict AEIPF more accurately (PROSPERO registration number: CRD42022356640).
Collapse
Affiliation(s)
- Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Alessandro G Fois
- Department of Respiratory Diseases, University Hospital Sassari, Sassari, Italy; Department of Clinical and Experimental Medicine, University of Sassari, Sassari, Italy
| | - Pietro Pirina
- Department of Respiratory Diseases, University Hospital Sassari, Sassari, Italy; Department of Clinical and Experimental Medicine, University of Sassari, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia; Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia.
| |
Collapse
|
31
|
Holton SE, Mitchem M, Pipavath S, Morrell ED, Bhatraju PK, Hamerman JA, Speake C, Malhotra U, Wurfel MM, Ziegler S, Mikacenic C. Mediators of monocyte chemotaxis and matrix remodeling are associated with the development of fibrosis in patients with COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.28.23289261. [PMID: 37205332 PMCID: PMC10187320 DOI: 10.1101/2023.04.28.23289261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Acute respiratory distress syndrome (ARDS) has a fibroproliferative phase that may be followed by pulmonary fibrosis. This has been described in patients with COVID-19 pneumonia, but the underlying mechanisms have not been completely defined. We hypothesized that protein mediators of tissue remodeling and monocyte chemotaxis are elevated in the plasma and endotracheal aspirates of critically ill patients with COVID-19 who subsequently develop radiographic fibrosis. We enrolled COVID-19 patients admitted to the ICU who had hypoxemic respiratory failure, were hospitalized and alive for at least 10 days, and had chest imaging done during hospitalization ( n = 119). Plasma was collected within 24h of ICU admission and at 7d. In mechanically ventilated patients, endotracheal aspirates (ETA) were collected at 24h and 48-96h. Protein concentrations were measured by immunoassay. We tested for associations between protein concentrations and radiographic evidence of fibrosis using logistic regression adjusting for age, sex, and APACHE score. We identified 39 patients (33%) with features of fibrosis. Within 24h of ICU admission, plasma proteins related to tissue remodeling (MMP-9, Amphiregulin) and monocyte chemotaxis (CCL-2/MCP-1, CCL-13/MCP-4) were associated with the subsequent development of fibrosis whereas markers of inflammation (IL-6, TNF-α) were not. After 1 week, plasma MMP-9 increased in patients without fibrosis. In ETAs, only CCL-2/MCP-1 was associated with fibrosis at the later timepoint. This cohort study identifies proteins of tissue remodeling and monocyte recruitment that may identify early fibrotic remodeling following COVID-19. Measuring changes in these proteins over time may allow for early detection of fibrosis in patients with COVID-19.
Collapse
|
32
|
Classical monocyte-derived macrophages as therapeutic targets of umbilical cord mesenchymal stem cells: comparison of intratracheal and intravenous administration in a mouse model of pulmonary fibrosis. Respir Res 2023; 24:68. [PMID: 36870972 PMCID: PMC9985859 DOI: 10.1186/s12931-023-02357-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 02/01/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease that has no cure. Although mesenchymal stem cells (MSCs) have been reported to ameliorate lung inflammation and fibrosis in mouse models, their mechanisms of action remain unknown. Therefore, we aimed to determine the changes in various immune cells, especially macrophages and monocytes, involved in the effects of MSC treatment on pulmonary fibrosis. METHODS We collected and analyzed explanted lung tissues and blood from patients with IPF who underwent lung transplantation. After establishing a pulmonary fibrosis model via the intratracheal administration of bleomycin (BLM) to 8-week-old mice, MSCs derived from human umbilical cords were administered intravenously or intratracheally on day 10 and the lungs were immunologically analyzed on days 14 and 21. Flow cytometry was performed to analyze the immune cell characteristics, and gene expression levels were examined using quantitative reverse transcription-polymerase chain reaction. RESULTS In the histological analysis of explanted human lung tissues, the terminally fibrotic areas contained a larger number of macrophages and monocytes than the early fibrotic areas of the lungs. When human monocyte-derived macrophages (MoMs) were stimulated with interleukin-13 in vitro, the expression of type 2 macrophage (M2) markers was more prominent in MoMs from the classical monocyte subset than in those from intermediate or non-classical monocyte subsets, and MSCs suppressed M2 marker expression independent of MoM subsets. In the mouse model, the increased number of inflammatory cells in the bronchoalveolar lavage fluid and the degree of lung fibrosis observed in BLM-treated mice were significantly reduced by MSC treatment, which tended to be more prominent with intravenous administration than intratracheal administration. Both M1 and M2 MoMs were upregulated in BLM-treated mice. The M2c subset of M2 MoMs was significantly reduced by MSC treatment. Among M2 MoMs, M2 MoMs derived from Ly6C+ monocytes were most effectively regulated by the intravenous administration, not intratracheal administration, of MSCs. CONCLUSIONS Inflammatory classical monocytes may play a role in lung fibrosis in human IPF and BLM-induced pulmonary fibrosis. Intravenous rather than intratracheal administration of MSCs may ameliorate pulmonary fibrosis by inhibiting monocyte differentiation into M2 macrophages.
Collapse
|
33
|
Liu S, Liu C, Wang Q, Liu S, Min J. CC Chemokines in Idiopathic Pulmonary Fibrosis: Pathogenic Role and Therapeutic Potential. Biomolecules 2023; 13:biom13020333. [PMID: 36830702 PMCID: PMC9953349 DOI: 10.3390/biom13020333] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 02/12/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF), characterized by progressive worsening of dyspnea and irreversible decline in lung function, is a chronic and progressive respiratory disease with a poor prognosis. Chronic or repeated lung injury results in inflammation and an excessive injury-repairing response that drives the development of IPF. A number of studies have shown that the development and progression of IPF are associated with dysregulated expression of several chemokines and chemokine receptors, several of which have been used as predictors of IPF outcome. Chemokines of the CC family play significant roles in exacerbating IPF progression by immune cell attraction or fibroblast activation. Modulating levels of detrimental CC chemokines and interrupting the corresponding transduction axis by neutralizing antibodies or antagonists are potential treatment options for IPF. Here, we review the roles of different CC chemokines in the pathogenesis of IPF, and their potential use as biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Shanshan Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Correspondence:
| | - Chang Liu
- Drug Clinical Trial Institution, Children’s Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Qianrong Wang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Suosi Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Jiali Min
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| |
Collapse
|
34
|
Coelho DB, Santos V, Alexandre AT, Bastos HNE, Mota PC, Morais A, Melo N. Risk Factors for Acute Exacerbations of Fibrotic Hypersensitivity Pneumonitis-Is Exposure a Trigger That We're Missing? Arch Bronconeumol 2023; 59:59-60. [PMID: 35945072 DOI: 10.1016/j.arbres.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 01/05/2023]
Affiliation(s)
- David Barros Coelho
- Pulmonology Department, Centro Hospitalar Universitário de São João, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Vanessa Santos
- Pulmonology Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| | | | - Helder Novais E Bastos
- Pulmonology Department, Centro Hospitalar Universitário de São João, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde I3S, University of Porto, Porto, Portugal
| | - Patrícia Caetano Mota
- Pulmonology Department, Centro Hospitalar Universitário de São João, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde I3S, University of Porto, Porto, Portugal
| | - António Morais
- Pulmonology Department, Centro Hospitalar Universitário de São João, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde I3S, University of Porto, Porto, Portugal
| | - Natália Melo
- Pulmonology Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| |
Collapse
|
35
|
Iommi M, Faragalli A, Bonifazi M, Mei F, Latini LL, Pompili M, Carle F, Gesuita R. Prognosis and Survival in Idiopathic Pulmonary Fibrosis in the Era of Antifibrotic Therapy in Italy: Evidence from a Longitudinal Population Study Based on Healthcare Utilization Databases. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:16689. [PMID: 36554568 PMCID: PMC9779053 DOI: 10.3390/ijerph192416689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 06/17/2023]
Abstract
The aim was to evaluate the determinants of acute exacerbation (AE) and death in new cases of idiopathic pulmonary fibrosis (IPF) using administrative databases in the Marche Region. Adults at their first prescription of antifibrotics or hospitalization with a diagnosis of IPF occurring in 2014-2019 were considered as new cases. Multiple Cox regression was used to estimate the risk of AE and of all-cause mortality adjusted by demographic and clinical characteristics, stratifying patients according to antifibrotic treatment. Overall, 676 new cases of IPF were identified and 276 deaths and 248 AE events occurred. In never-treated patients, the risk of AE was higher in patients with poor health conditions at diagnosis; the risk of death was higher in males, in patients aged ≥75 and in those with poor health conditions at baseline. The increasing number of AEs increased the risk of death in treated and never-treated patients. Within the limits of an observational study based on secondary data, the combined use of healthcare administrative databases allows the accurate analysis of progression and survival of IPF from the beginning of the antifibrotic therapy era, suggesting that timely and early diagnosis is critical to prescribing the most suitable treatment to increase survival and maintain a healthy life expectancy.
Collapse
Affiliation(s)
- Marica Iommi
- Center of Epidemiology Biostatistics and Medical Information Technology, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60121 Ancona, Italy
| | - Andrea Faragalli
- Center of Epidemiology Biostatistics and Medical Information Technology, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60121 Ancona, Italy
| | - Martina Bonifazi
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60121 Ancona, Italy
- Respiratory Diseases Unit, Azienda Ospedaliero-Universitaria “Ospedali Riuniti”, 60166 Ancona, Italy
| | - Federico Mei
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60121 Ancona, Italy
- Respiratory Diseases Unit, Azienda Ospedaliero-Universitaria “Ospedali Riuniti”, 60166 Ancona, Italy
| | - Lara Letizia Latini
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60121 Ancona, Italy
- Respiratory Diseases Unit, Azienda Ospedaliero-Universitaria “Ospedali Riuniti”, 60166 Ancona, Italy
| | - Marco Pompili
- Regional Health Agency of Marche, 60121 Ancona, Italy
| | - Flavia Carle
- Center of Epidemiology Biostatistics and Medical Information Technology, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60121 Ancona, Italy
- National Centre for Healthcare Research and Pharmacoepidemiology, 20126 Milano, Italy
| | - Rosaria Gesuita
- Center of Epidemiology Biostatistics and Medical Information Technology, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60121 Ancona, Italy
- National Centre for Healthcare Research and Pharmacoepidemiology, 20126 Milano, Italy
| |
Collapse
|
36
|
Broens B, Duitman JW, Zwezerijnen GJC, Nossent EJ, van der Laken CJ, Voskuyl AE. Novel tracers for molecular imaging of interstitial lung disease: A state of the art review. Autoimmun Rev 2022; 21:103202. [PMID: 36150433 DOI: 10.1016/j.autrev.2022.103202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/16/2022] [Indexed: 12/14/2022]
Abstract
Interstitial lung disease is an overarching term for a wide range of disorders characterized by inflammation and/or fibrosis in the lungs. Most prevalent forms, among others, include idiopathic pulmonary fibrosis (IPF) and connective tissue disease associated interstitial lung disease (CTD-ILD). Currently, only disease modifying treatment options are available for IPF and progressive fibrotic CTD-ILD, leading to reduction or stabilization in the rate of lung function decline at best. Management of these patients would greatly advance if we identify new strategies to improve (1) early detection of ILD, (2) predicting ILD progression, (3) predicting response to therapy and (4) understanding pathophysiology. Over the last years, positron emission tomography (PET) and single photon emission computed tomography (SPECT) have emerged as promising molecular imaging techniques to improve ILD management. Both are non-invasive diagnostic tools to assess molecular characteristics of an individual patient with the potential to apply personalized treatment. In this review, we encompass the currently available pre-clinical and clinical studies on molecular imaging with PET and SPECT in IPF and CTD-ILD. We provide recommendations for potential future clinical applications of these tracers and directions for future research.
Collapse
Affiliation(s)
- Bo Broens
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Rheumatology and Clinical Immunology, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Infection & Immunity, Inflammatory diseases, Amsterdam, the Netherlands.
| | - Jan-Willem Duitman
- Amsterdam Infection & Immunity, Inflammatory diseases, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Pulmonary Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Experimental Immunology (EXIM), Meibergdreef 9, Amsterdam, the Netherlands.
| | - Gerben J C Zwezerijnen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, the Netherlands.
| | - Esther J Nossent
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences Research Institute, Amsterdam, the Netherlands..
| | - Conny J van der Laken
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Rheumatology and Clinical Immunology, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Infection & Immunity, Inflammatory diseases, Amsterdam, the Netherlands.
| | - Alexandre E Voskuyl
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Rheumatology and Clinical Immunology, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Infection & Immunity, Inflammatory diseases, Amsterdam, the Netherlands.
| |
Collapse
|
37
|
Enomoto N. Pathological Roles of Pulmonary Cells in Acute Lung Injury: Lessons from Clinical Practice. Int J Mol Sci 2022; 23:ijms232315027. [PMID: 36499351 PMCID: PMC9736972 DOI: 10.3390/ijms232315027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/23/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022] Open
Abstract
Interstitial lung diseases (ILD) are relatively rare and sometimes become life threatening. In particular, rapidly progressive ILD, which frequently presents as acute lung injury (ALI) on lung histopathology, shows poor prognosis if proper and immediate treatments are not initiated. These devastating conditions include acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF), clinically amyopathic dermatomyositis (CADM), epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI)-induced lung injury, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infection named coronavirus disease 2019 (COVID-19). In this review, clinical information, physical findings, laboratory examinations, and findings on lung high-resolution computed tomography and lung histopathology are presented, focusing on majorly damaged cells in each disease. Furthermore, treatments that should be immediately initiated in clinical practice for each disease are illustrated to save patients with these diseases.
Collapse
Affiliation(s)
- Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; ; Tel.: +81-53-435-2263; Fax: +81-53-435-2354
- Health Administration Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu 431-3192, Japan
| |
Collapse
|
38
|
Dai X, Yang Z, Zhang W, Liu S, Zhao Q, Liu T, Chen L, Li L, Wang Y, Shao R. Identification of diagnostic gene biomarkers related to immune infiltration in patients with idiopathic pulmonary fibrosis based on bioinformatics strategies. Front Med (Lausanne) 2022; 9:959010. [PMID: 36507532 PMCID: PMC9729277 DOI: 10.3389/fmed.2022.959010] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 11/10/2022] [Indexed: 11/25/2022] Open
Abstract
Objective The study aims to identify potential diagnostic markers of idiopathic pulmonary fibrosis (IPF) and analyze the significance of immune cell infiltration in this pathology. Materials and methods Download two publicly available gene expression profiles (GSE10667 and GSE24206 datasets) from the GEO database including 48 Idiopathic pulmonary fibrosis (IPF) samples and 21 human control samples and select for distinctly expressed genes (DEG) from them. Lasso regression model and support vector machine recursive feature elimination S,V,R,F analysis were used to check candidate biomarkers. The area under the subject's work characteristic curve (AUC) value is used to evaluate its recognition ability. The GSE53845 dataset (40 IPF patients and 8 controls) continue to validate the expression level and diagnostic value of biomarkers in IPF. Comprehensive analysis of immune infiltrated cells of IPF was performed using R software and immune cell infiltration estimation analysis tool- deconvolution algorithm (CIBERSORT). Results 43 DEGs were identified in total. The identified DEGs mostly involve pneumonia, lung disease, collagen disease, obstructive pulmonary disease and other diseases. The activation of IL-17 signaling pathways, amoebic disease, interaction of viral proteins with cytokines and cytokine receptors, protein digestion and absorption, and flaccid hormone signaling pathways in IPF were different from the control group. The expression degree of CRTAC1, COL10A1, COMP, RPS4Y1, IGFL2, NECAB1, SCG5, SLC6A4, and SPP1 in IPF tissue were prominently higher than the normal group. Immune cell infiltration analysis showed that CRTAC1, COL10A1, COMP, IGFL2, NECAB1, SCG5, SLC6A4, and SPP1 were associated with monocytes, plasma cells, neutrophils, and regulatory (treg) T cells. Conclusion CRTAC1, COL10A1, COMP, IGFL2, NECAB1, SCG5, SLC6A4, and SPP1 can be used as diagnostic markers for IPF, providing new ideas for the future study of IPF occurrence and molecular mechanisms.
Collapse
Affiliation(s)
- Xiangdong Dai
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihua Yang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenjing Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shuai Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qianru Zhao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Tao Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lu Chen
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Yi Wang,
| | - Rui Shao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China,*Correspondence: Rui Shao,
| |
Collapse
|
39
|
Fikry H, Saleh LA, Gawad SA. Therapeutic effect of adipose-derived mesenchymal stem cells (AD-MSCs) compared to pirfenidone on corticosteroid resistance in a mouse model of acute exacerbation of idiopathic pulmonary fibrosis. Histol Histopathol 2022; 37:1065-1083. [PMID: 35816024 DOI: 10.14670/hh-18-493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Acute exacerbation-idiopathic pulmonary fibrosis (AE-IPF) is a life-threatening condition. In the treatment of AE-IPF, corticosteroid medication is commonly utilized. However, there is insufficient evidence to justify its usage. Pirfenidone (PFD) has recently been discovered to be effective in the treatment of AE-IPF patients. However, regenerative therapy, such as stem cell therapy or tissue engineering, is necessary due to ineffective and limited therapies. Combining MSC transplantation with pharmacological therapy may also give additional benefits; nevertheless, its use must be proven experimentally. As a result, the goal of this study was to assess the therapeutic effects of adipose-derived mesenchymal stem cells (AD-MSCs) on corticosteroid resistance in an animal model of AE-IPF caused by bleomycin compared to PFD. MATERIALS AND METHODS Seventy C57BL/6J male mice were randomly divided into seven groups, control, BLM, methylprednisolone (MP), PFD, AD-MSCs, PFD +MP, and AD-MSCs +MP. RESULTS In terms of survival, collagen deposition, the acute lung injury score (ALI), and the Ashcroft score, AD-MSCs exceeded PFD. AD-MSCs + MP provided protection and preserved the lung's architecture in BLM-induced AE. In addition, AD-MSCs successfully decreased chemokine (CC motif) ligand-2 (CCL2) positive cells and lower pro-fibrotic and pro-inflammatory cytokines. CONCLUSIONS AD-MSCs enhanced histological structure, Ashcroft and ALI scores, lung collagen deposition, survival, and cytokines in an animal model of AE-IPF. As a result, we believe that AD-MSCs may be more therapeutically helpful for AE-IPF than presently available therapies, either alone or in conjunction with MP.
Collapse
Affiliation(s)
- Heba Fikry
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Lobna A Saleh
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sara Abdel Gawad
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
40
|
Lamichhane R, Patial S, Saini Y. Higher susceptibility of males to bleomycin-induced pulmonary inflammation is associated with sex-specific transcriptomic differences in myeloid cells. Toxicol Appl Pharmacol 2022; 454:116228. [PMID: 36087614 PMCID: PMC9793441 DOI: 10.1016/j.taap.2022.116228] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/24/2022] [Accepted: 09/02/2022] [Indexed: 12/30/2022]
Abstract
Idiopathic pulmonary fibrosis, a condition with likely genetic and environmental etiology, is relatively more prevalent with poor prognosis in human males. However, the underlying mechanisms for these gender-associated differences in the severity of fibrosis remain unknown. Here, we tested the hypothesis that the transcriptomic repertoire of myeloid cells determines the higher susceptibility of male mice to bleomycin (BLM)-induced lung fibrosis. Adult mice were oropharyngeally challenged with saline or BLM. Lung injury, inflammation, and fibrosis outcomes were assessed, and airspace myeloid-cells were subjected to RNA-sequencing. As compared with the female mice, the male mice manifested significantly increased lung injury, inflammation, proinflammatory cytokines (IL-6, IL-1β, IL-7, and IP-10), and fibrosis in response to BLM challenge. Interestingly, several pro-inflammatory and extracellular matrix-associated genes were significantly up-regulated in male myeloid-cells compared to female myeloid-cells in the saline-control group. Similarly, BLM challenge resulted in greater pro-inflammatory and pro-fibrotic transcriptomic changes in male compared to female myeloid cells. On the other hand, anti-inflammatory and regulatory cytokine, Il10 and Ifng respectively, were uniquely upregulated in BLM-challenged female but not in male myeloid cells when compared to their respective saline-control groups. Further, cross-sex bone marrow transplantation experiments revealed that male hematopoietic progenitor cells (HPCs) increased the granulocytic infiltration in female mice while female HPCs decreased the granulocytic infiltration in male mice post-BLM challenge. These findings suggest that there are inherent transcriptomic differences between the male and female lung myeloid cells and that the pro-inflammatory nature of male myeloid cells is sufficient to increase the susceptibility of female mice to BLM-induced inflammation.
Collapse
|
41
|
Awano N, Jo T, Izumo T, Inomata M, Morita K, Matsui H, Fushimi K, Urushiyama H, Nagase T, Yasunaga H. Efficacy of initial high- versus low-dose intravenous corticosteroid therapy in patients with acute exacerbation of idiopathic interstitial pneumonia: A nationwide observational study. ANNALS OF CLINICAL EPIDEMIOLOGY 2022; 5:37-47. [PMID: 38505731 PMCID: PMC10944999 DOI: 10.37737/ace.23006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/23/2022] [Indexed: 03/21/2024]
Abstract
BACKGROUND Acute exacerbation of idiopathic interstitial pneumonias (AE-IIPs) has a high mortality. However, there is no established treatment for AE-IIPs. Therefore, we aimed to compare the efficacy of high- and low-dose corticosteroid therapies in AE-IIPs patients. METHODS Data were retrospectively collected from the Japanese Diagnosis Procedure Combination database from July 2010 to March 2018. Adult patients with AE-IIPs who received high-dose (methylprednisolone at a dose of 500-1000 mg/day for 3 days starting within 4 days after admission) or low-dose (methylprednisolone at a dose of 100-200 mg/day for at least 5 days starting within 4 days after admission) corticosteroid therapy were identified. Eligible patients (n = 17,317) were divided into the high-dose (n = 16,998) and low-dose (n = 319) groups. A stabilized inverse probability of treatment weighting using propensity scores was performed to compare outcomes between the groups. RESULTS The primary outcome was in-hospital mortality, and the secondary outcomes were 28-day mortality, infections during hospitalization, length of hospitalization, duration of steroid use, and discharge to home. The in-hospital mortality rates of the high- and low-dose corticosteroid groups were 50.6% and 47.0%, respectively. In-hospital mortality did not significantly differ between the two groups after stabilized inverse probability of treatment weighting, and the odds ratio in the low-dose corticosteroid group was 0.86 (95% confidence interval: 0.64-1.16; p = 0.33). The secondary outcomes also did not significantly differ between the groups. CONCLUSIONS There was no significant difference in outcomes between patients with AE-IIPs who received high- and low-dose corticosteroid therapies.
Collapse
Affiliation(s)
- Nobuyasu Awano
- Department of Respiratory Medicine, Japanese Red Cross Medical Center
| | - Taisuke Jo
- Department of Health Services Research, Graduate School of Medicine, The University of Tokyo
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo
| | - Takehiro Izumo
- Department of Respiratory Medicine, Japanese Red Cross Medical Center
| | - Minoru Inomata
- Department of Respiratory Medicine, Japanese Red Cross Medical Center
| | - Kojiro Morita
- Department of Clinical Epidemiology and Health Economics, School of Public Health, The University of Tokyo
- Department of Health Services Research, Faculty of Medicine, University of Tsukuba
| | - Hiroki Matsui
- Department of Clinical Epidemiology and Health Economics, School of Public Health, The University of Tokyo
| | - Kiyohide Fushimi
- Department of Health Policy and Informatics, Tokyo Medical and Dental University Graduate School of Medicine
| | - Hirokazu Urushiyama
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo
| | - Takahide Nagase
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo
| | - Hideo Yasunaga
- Department of Clinical Epidemiology and Health Economics, School of Public Health, The University of Tokyo
| |
Collapse
|
42
|
Yegen CH, Haine L, Da Costa Ferreira K, Marchant D, Bernaudin JF, Planès C, Voituron N, Boncoeur E. A New Model of Acute Exacerbation of Experimental Pulmonary Fibrosis in Mice. Cells 2022; 11:3379. [PMID: 36359778 PMCID: PMC9654438 DOI: 10.3390/cells11213379] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/07/2022] [Accepted: 10/23/2022] [Indexed: 11/27/2023] Open
Abstract
RATIONALE idiopathic pulmonary fibrosis (IPF) is the most severe form of fibrosing interstitial lung disease, characterized by progressive respiratory failure leading to death. IPF's natural history is heterogeneous, and its progression unpredictable. Most patients develop a progressive decline of respiratory function over years; some remain stable, but others present a fast-respiratory deterioration without identifiable cause, classified as acute exacerbation (AE). OBJECTIVES to develop and characterize an experimental mice model of lung fibrosis AE, mimicking IPF-AE at the functional, histopathological, cellular and molecular levels. METHODS we established in C57BL/6 male mice a chronic pulmonary fibrosis using a repetitive low-dose bleomycin (BLM) intratracheal (IT) instillation regimen (four instillations of BLM every 2 weeks), followed by two IT instillations of a simple or double-dose BLM challenge to induce AE. Clinical follow-up and histological and molecular analyses were done for fibrotic and inflammatory lung remodeling analysis. MEASUREMENTS AND MAIN RESULTS as compared with a low-dose BLM regimen, this AE model induced a late burst of animal mortality, worsened lung fibrosis and remodeling, and superadded histopathological features as observed in humans IPF-AE. This was associated with stronger inflammation, increased macrophage infiltration of lung tissue and increased levels of pro-inflammatory cytokines in lung homogenates. Finally, it induced in the remodeled lung a diffuse expression of hypoxia-inducible factor 1α, a hallmark of tissular hypoxia response and a major player in the progression of IPF. CONCLUSION this new model is a promising model of AE in chronic pulmonary fibrosis that could be relevant to mimic IPF-AE in preclinical trials.
Collapse
Affiliation(s)
- Céline-Hivda Yegen
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
| | - Liasmine Haine
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
| | - Kevin Da Costa Ferreira
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
| | - Dominique Marchant
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
| | - Jean-Francois Bernaudin
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
- Faculté de Médecine, Sorbonne Université, 75006 Paris, France
- Service de Physiologie et d’Explorations Fonctionnelles, Hôpital Avicenne, APHP, 93000 Bobigny, France
| | - Carole Planès
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
- Service de Physiologie et d’Explorations Fonctionnelles, Hôpital Avicenne, APHP, 93000 Bobigny, France
| | - Nicolas Voituron
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
- Département STAPS, Université Sorbonne Paris-Nord, 93000 Bobigny, France
| | - Emilie Boncoeur
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, 93000 Bobigny, France
| |
Collapse
|
43
|
Patel H, Shah JR, Patel DR, Avanthika C, Jhaveri S, Gor K. Idiopathic pulmonary fibrosis: Diagnosis, biomarkers and newer treatment protocols. Dis Mon 2022:101484. [DOI: 10.1016/j.disamonth.2022.101484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
44
|
Bomb K, Pradhan L, Zhang Q, Jarai BM, Bhattacharjee A, Burris DL, Kloxin AM, Fromen CA. Destructive fibrotic teamwork: how both microenvironment stiffness and profibrotic interleukin 13 impair alveolar macrophage phenotype and function. Biomater Sci 2022; 10:5689-5706. [PMID: 36018297 PMCID: PMC9632634 DOI: 10.1039/d2bm00828a] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The pulmonary fibrotic microenvironment is characterized by increased stiffness of lung tissue and enhanced secretion of profibrotic soluble cues contributing to a feedback loop that leads to dysregulated wound healing and lung failure. Pinpointing the individual and tandem effects of profibrotic stimuli in impairing immune cell response remains difficult and is needed for improved therapeutic strategies. We utilized a statistical design of experiment (DOE) to investigate how microenvironment stiffness and interleukin 13 (IL13), a profibrotic soluble factor linked with disease severity, contribute to the impaired macrophage response commonly observed in pulmonary fibrosis. We used engineered bioinspired hydrogels of different stiffness, ranging from healthy to fibrotic lung tissue, and cultured murine alveolar macrophages (MH-S cells) with or without IL13 to quantify cell response and analyze independent and synergistic effects. We found that, while both stiffness and IL13 independently influence macrophage morphology, phenotype, phagocytosis and efferocytosis, these factors work synergistically to exacerbate impaired macrophage phenotype and efferocytosis. These unique findings provide insights into how macrophages in fibrotic conditions are not as effective in clearing debris, contributing to fibrosis initiation/progression, and more broadly inform how underlying drivers of fibrosis modulate immune cell response to facilitate therapeutic strategies.
Collapse
Affiliation(s)
- Kartik Bomb
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | - Lina Pradhan
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | - Qi Zhang
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | - Bader M Jarai
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | | | - David L Burris
- Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - April M Kloxin
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
- Material Science and Engineering, University of Delaware, Newark, DE, USA
| | - Catherine A Fromen
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
45
|
Kletukhina S, Mutallapova G, Titova A, Gomzikova M. Role of Mesenchymal Stem Cells and Extracellular Vesicles in Idiopathic Pulmonary Fibrosis. Int J Mol Sci 2022; 23:ijms231911212. [PMID: 36232511 PMCID: PMC9569825 DOI: 10.3390/ijms231911212] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial fibrotic disease that leads to disability and death within 5 years of diagnosis. Pulmonary fibrosis is a disease with a multifactorial etiology. The concept of aberrant regeneration of the pulmonary epithelium reveals the pathogenesis of IPF, according to which repeated damage and death of alveolar epithelial cells is the main mechanism leading to the development of progressive IPF. Cell death provokes the migration, proliferation and activation of fibroblasts, which overproduce extracellular matrix, resulting in fibrotic deformity of the lung tissue. Mesenchymal stem cells (MSCs) and extracellular vesicles (EVs) are promising therapies for pulmonary fibrosis. MSCs, and EVs derived from MSCs, modulate the activity of immune cells, inhibit the expression of profibrotic genes, reduce collagen deposition and promote the repair of damaged lung tissue. This review considers the molecular mechanisms of the development of IPF and the multifaceted role of MSCs in the therapy of IPF. Currently, EVs-MSCs are regarded as a promising cell-free therapy tool, so in this review we discuss the results available to date of the use of EVs-MSCs for lung tissue repair.
Collapse
Affiliation(s)
- Sevindzh Kletukhina
- Laboratory of Intercellular Communication, Kazan Federal University, 420008 Kazan, Russia
| | - Guzel Mutallapova
- Laboratory of Intercellular Communication, Kazan Federal University, 420008 Kazan, Russia
| | - Angelina Titova
- Morphology and General Pathology Department, Kazan Federal University, 420008 Kazan, Russia
| | - Marina Gomzikova
- Laboratory of Intercellular Communication, Kazan Federal University, 420008 Kazan, Russia
- Correspondence: ; Tel.: +7-917-8572269
| |
Collapse
|
46
|
Otsuka J, Yoshizawa S, Ikematsu Y, Kudo K, Osoreda H, Ishimatsu A, Taguchi K, Moriwaki A, Wakamatsu K, Iwanaga T, Yoshida M. Acute exacerbation in antineutrophil cytoplasmic antibody-associated interstitial lung disease: Clinical features and risk factors. Respir Med 2022; 203:106992. [DOI: 10.1016/j.rmed.2022.106992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/30/2022] [Accepted: 09/16/2022] [Indexed: 10/31/2022]
|
47
|
Spagnolo P, Tonelli R, Samarelli AV, Castelli G, Cocconcelli E, Petrarulo S, Cerri S, Bernardinello N, Clini E, Saetta M, Balestro E. The role of immune response in the pathogenesis of idiopathic pulmonary fibrosis: far beyond the Th1/Th2 imbalance. Expert Opin Ther Targets 2022; 26:617-631. [PMID: 35983984 DOI: 10.1080/14728222.2022.2114897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION . Idiopathic pulmonary fibrosis (IPF) is a chronic disease of unknown origin characterized by progressive scarring of the lung leading to irreversible loss of function. Despite the availability of two drugs that are able to slow down disease progression, IPF remains a deadly disease. The pathogenesis of IPF is poorly understood, but a dysregulated wound healing response following recurrent alveolar epithelial injury is thought to be crucial. Areas covered. In the last few years, the role of the immune system in IPF pathobiology has been reconsidered; indeed, recent data suggest that a dysfunctional immune system may promote and unfavorable interplay with pro-fibrotic pathways thus acting as a cofactor in disease development and progression. In this article, we review and critically discuss the role of T cells in the pathogenesis and progression of IPF in the attempt to highlight ways in which further research in this area may enable the development of targeted immunomodulatory therapies for this dreadful disease. EXPERT OPINION A better understanding of T cells interactions has the potential to facilitate the development of immune modulators targeting multiple T cell-mediated pathways thus halting disease initiation and progression.
Collapse
Affiliation(s)
- Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Roberto Tonelli
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults University Hospital of Modena and Reggio Emilia, Modena, Italy.,University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Anna Valeria Samarelli
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults University Hospital of Modena and Reggio Emilia, Modena, Italy.,University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Gioele Castelli
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Elisabetta Cocconcelli
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Simone Petrarulo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Stefania Cerri
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults University Hospital of Modena and Reggio Emilia, Modena, Italy.,University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Nicol Bernardinello
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Enrico Clini
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults University Hospital of Modena and Reggio Emilia, Modena, Italy.,University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marina Saetta
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Elisabetta Balestro
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| |
Collapse
|
48
|
Rui Y, Han X, Jiang A, Hu J, Li M, Liu B, Qian F, Huang L. Eucalyptol prevents bleomycin-induced pulmonary fibrosis and M2 macrophage polarization. Eur J Pharmacol 2022; 931:175184. [PMID: 35964659 DOI: 10.1016/j.ejphar.2022.175184] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 07/23/2022] [Accepted: 08/01/2022] [Indexed: 11/03/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrosing interstitial pneumonia with limited therapeutic options. Eucalyptol, a terpenoid oxide isolated from eucalyptus species, reportedly exhibits various biological activities such as anti-inflammatory and antioxidant effects. In the present study, we aimed to determine whether eucalyptol could alleviate bleomycin (BLM)-induced pulmonary fibrosis and inhibit interleukin (IL)-13-induced M2 macrophage polarization. Upon treatment with eucalyptol, BLM-induced pulmonary fibrosis and lung inflammation were significantly reduced. The pulmonary neutrophil accumulation and pulmonary permeability were inhibited and the expression of hydroxyproline, alpha-smooth muscle actin, and fibronectin was significantly down-regulated. Eucalyptol also markedly inhibited the expression of arginase-1, Ym-1, IL-13, and transforming growth factor (TGF)-β1, reduced the production of IL-13, IL-6, tumor necrosis factor (TNF)-α, and attenuated the activity of TGF-β1 in bronchoalveolar lavage fluid (BALF). Furthermore, the in vitro assay revealed that eucalyptol disturbed M2 macrophage polarization and reduced the macrophage-mediated secretion of the profibrotic factor TGF-β1. Eucalyptol inhibited the nuclear location of signal transducer and activator of transcription 6 (STAT6) and the phosphorylation of STAT6 and p38 mitogen-activated protein kinase (p38 MAPK), and reduced the expression of their downstream transcription factors, krupple-like factor 4 (KLF4) and peroxisome proliferator-activated receptor gamma (PPAR-γ). These findings indicated that eucalyptol alleviates BLM-induced pulmonary fibrosis by regulating M2 macrophage polarization, which, in turn, inhibits the activation of signaling molecules (e.g., STAT6 and p38 MAPK) and the expression of transcription factors (e.g., KLF4 and PPAR-γ). Thus, eucalyptol might be a potential therapeutic agent for IPF.
Collapse
Affiliation(s)
- Yan Rui
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, China; The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Department of Respiration and Critical Care Medicine, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Xiaojing Han
- Department of Clinical Laboratory Diagnostics, School of Laboratory Medicine, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Anbang Jiang
- Department of Respiration and Critical Care Medicine, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Junfeng Hu
- Department of Respiration and Critical Care Medicine, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Miao Li
- Department of General Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Bangzhu Liu
- Department of Respiratory Medicine, The Second People's Hospital of Anhui, Wuhu, Anhui, 233000, China
| | - Feng Qian
- Department of Clinical Laboratory Diagnostics, School of Laboratory Medicine, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui, 233000, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Linian Huang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, China; The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Department of Respiration and Critical Care Medicine, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China.
| |
Collapse
|
49
|
Humphries DC, Mills R, Boz C, McHugh BJ, Hirani N, Rossi AG, Pedersen A, Schambye HT, Slack RJ, Leffler H, Nilsson UJ, Wang W, Sethi T, Mackinnon AC. Galectin-3 inhibitor GB0139 protects against acute lung injury by inhibiting neutrophil recruitment and activation. Front Pharmacol 2022; 13:949264. [PMID: 36003515 PMCID: PMC9393216 DOI: 10.3389/fphar.2022.949264] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Rationale: Galectin-3 (Gal-3) drives fibrosis during chronic lung injury, however, its role in acute lung injury (ALI) remains unknown. Effective pharmacological therapies available for ALI are limited; identifying novel concepts in treatment is essential. GB0139 is a Gal-3 inhibitor currently under clinical investigation for the treatment of idiopathic pulmonary fibrosis. We investigate the role of Gal-3 in ALI and evaluate whether its inhibition with GB0139 offers a protective role. The effect of GB0139 on ALI was explored in vivo and in vitro. Methods: The pharmacokinetic profile of intra-tracheal (i.t.) GB0139 was investigated in C57BL/6 mice to support the daily dosing regimen. GB0139 (1–30 µg) was then assessed following acute i.t. lipopolysaccharide (LPS) and bleomycin administration. Histology, broncho-alveolar lavage fluid (BALf) analysis, and flow cytometric analysis of lung digests and BALf were performed. The impact of GB0139 on cell activation and apoptosis was determined in vitro using neutrophils and THP-1, A549 and Jurkat E6 cell lines. Results: GB0139 decreased inflammation severity via a reduction in neutrophil and macrophage recruitment and neutrophil activation. GB0139 reduced LPS-mediated increases in interleukin (IL)-6, tumor necrosis factor alpha (TNFα) and macrophage inflammatory protein-1-alpha. In vitro, GB0139 inhibited Gal-3-induced neutrophil activation, monocyte IL-8 secretion, T cell apoptosis and the upregulation of pro-inflammatory genes encoding for IL-8, TNFα, IL-6 in alveolar epithelial cells in response to mechanical stretch. Conclusion: These data indicate that Gal-3 adopts a pro-inflammatory role following the early stages of lung injury and supports the development of GB0139, as a potential treatment approach in ALI.
Collapse
Affiliation(s)
- Duncan C. Humphries
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
- Galecto Inc. Nine Edinburgh BioQuarter, Edinburgh, United Kingdom
| | - Ross Mills
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Cecilia Boz
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Brian J. McHugh
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Nikhil Hirani
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Adriano G. Rossi
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | - Hakon Leffler
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | | | - Wei Wang
- Department of Asthma, Allergy and Respiratory Science, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Tariq Sethi
- Galecto Inc, Copenhagen, Denmark
- Department of Asthma, Allergy and Respiratory Science, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Alison C. Mackinnon
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
- Galecto Inc. Nine Edinburgh BioQuarter, Edinburgh, United Kingdom
- *Correspondence: Alison C. Mackinnon,
| |
Collapse
|
50
|
Assessment of diagnostic utility of serum hemeoxygenase-1 measurement for acute exacerbation of interstitial pneumonias. Sci Rep 2022; 12:12935. [PMID: 35902685 PMCID: PMC9334264 DOI: 10.1038/s41598-022-17290-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/22/2022] [Indexed: 11/08/2022] Open
Abstract
The present study aimed to evaluate whether serum heme oxygenase (HO)-1 could be a reliable blood biomarker for diagnosing acute exacerbations (AEs) of both idiopathic interstitial pneumonia (IIP) and secondary interstitial pneumonia (SIP). Serum HO-1 levels of newly diagnosed patients with IP were measured, and the relationships between serum HO-1 and other serum biomarkers and high-resolution CT scores, were evaluated. Blood samples were collected from 90 patients with IIP, including 32 having an AE, and 32 with SIP, including 9 having an AE. The patients having an AE had significantly higher HO-1 levels than those not having an AE (35.2 ng/mL vs. 16.4 ng/mL; p < 0.001). On receiver operating characteristics (ROC) curve analysis for serum HO-1 ability to detect an AE, the area under the ROC curve (AUC) was 0.87 in patients with IIPs and 0.86 in those with SIPs. Also, in patients with both IIPs and SIPs, the combination of the serum HO-1 level and the GGO score showed favorable AUCs (IIPs: 0.92, SIPs: 0.83), though HO-1-not-including model (combination of LDH and GGO) also showed acceptable AUCs. Serum HO-1 could be a clinically useful biomarker for the accurate diagnosis of patients with AEs.
Collapse
|