1
|
Ma Z, Tong S, Huang Y, Wang N, Chen G, Bai Q, Deng J, Zhou L, Luo Q, Wang J, Lu W, Chen L, Wang T. Development and Characterization of a Novel Rat Model for Emulating Chronic Obstructive Pulmonary Disease-Associated Cor Pulmonale. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:831-844. [PMID: 39889827 DOI: 10.1016/j.ajpath.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/02/2024] [Accepted: 01/07/2025] [Indexed: 02/03/2025]
Abstract
Cor pulmonale, a condition marked by right ventricular dysfunction, is frequently associated with chronic obstructive pulmonary disease (COPD) and significantly worsens COPD prognosis. Despite the clinical relevance of cor pulmonale, development of effective treatments is hindered by the lack of animal models that accurately replicate the complex interplay between cor pulmonale and COPD. This study introduces a novel rat model combining cigarette smoke exposure with left pulmonary artery ligation to better mimic the pathophysiological features of COPD-related cor pulmonale. Pulmonary function tests revealed impaired lung function, and histologic assessments indicated emphysematous changes and inflammatory infiltration, consistent with COPD pathology. Furthermore, the model exhibited hallmarks of cor pulmonale, including right ventricular hypertrophy, fibrosis, and capillary rarefaction, alongside hemodynamic alterations indicative of pulmonary hypertension. This study's findings underscore the potential of the left pulmonary artery ligation + cigarette smoke rat model to advance understanding of COPD-related cor pulmonale pathophysiology and facilitate the development of targeted therapeutics.
Collapse
Affiliation(s)
- Zhuoji Ma
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Suiyang Tong
- Department of Cardiology, Suizhou Hospital, Hubei University of Medicine, Suizhou, China
| | - Yuhang Huang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Neng Wang
- Department of Cardiology, Suizhou Hospital, Hubei University of Medicine, Suizhou, China
| | - Guanjin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qianwen Bai
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jia Deng
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liang Zhou
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qiao Luo
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lingdan Chen
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Tao Wang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Chavda VP, Bezbaruah R, Ahmed N, Alom S, Bhattacharjee B, Nalla LV, Rynjah D, Gadanec LK, Apostolopoulos V. Proinflammatory Cytokines in Chronic Respiratory Diseases and Their Management. Cells 2025; 14:400. [PMID: 40136649 PMCID: PMC11941495 DOI: 10.3390/cells14060400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Pulmonary homeostasis can be agitated either by external environmental insults or endogenous factors produced during respiratory/pulmonary diseases. The lungs counter these insults by initiating mechanisms of inflammation as a localized, non-specific first-line defense response. Cytokines are small signaling glycoprotein molecules that control the immune response. They are formed by numerous categories of cell types and induce the movement, growth, differentiation, and death of cells. During respiratory diseases, multiple proinflammatory cytokines play a crucial role in orchestrating chronic inflammation and structural changes in the respiratory tract by recruiting inflammatory cells and maintaining the release of growth factors to maintain inflammation. The issue aggravates when the inflammatory response is exaggerated and/or cytokine production becomes dysregulated. In such instances, unresolving and chronic inflammatory reactions and cytokine production accelerate airway remodeling and maladaptive outcomes. Pro-inflammatory cytokines generate these deleterious consequences through interactions with receptors, which in turn initiate a signal in the cell, triggering a response. The cytokine profile and inflammatory cascade seen in different pulmonary diseases vary and have become fundamental targets for advancement in new therapeutic strategies for lung diseases. There are considerable therapeutic approaches that target cytokine-mediated inflammation in pulmonary diseases; however, blocking specific cytokines may not contribute to clinical benefit. Alternatively, broad-spectrum anti-inflammatory approaches are more likely to be clinically effective. Herein, this comprehensive review of the literature identifies various cytokines (e.g., interleukins, chemokines, and growth factors) involved in pulmonary inflammation and the pathogenesis of respiratory diseases (e.g., asthma, chronic obstructive pulmonary, lung cancer, pneumonia, and pulmonary fibrosis) and investigates targeted therapeutic treatment approaches.
Collapse
Affiliation(s)
- Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India; (R.B.); (N.A.); (S.A.)
- Institute of Pharmacy, Assam Medical College and Hospital, Dibrugarh 786002, Assam, India
| | - Nasima Ahmed
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India; (R.B.); (N.A.); (S.A.)
| | - Shahnaz Alom
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India; (R.B.); (N.A.); (S.A.)
- Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India; (B.B.); (D.R.)
| | - Bedanta Bhattacharjee
- Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India; (B.B.); (D.R.)
| | - Lakshmi Vineela Nalla
- Department of Pharmacology, GITAM School of Pharmacy, GITAM (Deemed to be University), Rushikonda, Visakhapatnam 530045, Andhra Pradesh, India;
| | - Damanbhalang Rynjah
- Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India; (B.B.); (D.R.)
| | - Laura Kate Gadanec
- Institute for Health and Sport, Immunology and Translational Research Group, Victoria University, Werribee, VIC 3030, Australia;
| | - Vasso Apostolopoulos
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia;
| |
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
|
Thawanaphong S, Nair A, Volfson E, Nair P, Mukherjee M. IL-18 biology in severe asthma. Front Med (Lausanne) 2024; 11:1486780. [PMID: 39554494 PMCID: PMC11566457 DOI: 10.3389/fmed.2024.1486780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/18/2024] [Indexed: 11/19/2024] Open
Abstract
The role of interleukin-18 (IL-18) and inflammasomes in chronic inflammatory airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD), has garnered significant attention in recent years. This review aims to provide an overview of the current understanding of IL-18 biology, the associated signaling pathways, and the involvement of inflammasome complexes in airway diseases. We explore the multifaceted role of IL-18 in asthma pathophysiology, including its interactions with other cytokines and contributions to both T2 and non-T2 inflammation. Importantly, emerging evidence highlights IL-18 as a critical player in severe asthma, contributing to chronic airway inflammation, airway hyperresponsiveness (AHR), and mucus impaction. Furthermore, we discuss the emerging evidence of IL-18's involvement in autoimmunity and highlight potential therapeutic targets within the IL-18 and inflammasome pathways in severe asthma patients with evidence of infections and airway autoimmune responses. By synthesizing recent advancements and ongoing research, this review underscores the importance of IL-18 as a potential novel therapeutic target in the treatment of severe asthma and other related conditions.
Collapse
Affiliation(s)
- Sarita Thawanaphong
- Department of Medicine, McMAster University, Hamilton, ON, Canada
- Research Institute of St. Joe’s Hamilton, St. Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Aswathi Nair
- Department of Medicine, McMAster University, Hamilton, ON, Canada
- Research Institute of St. Joe’s Hamilton, St. Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
| | - Emily Volfson
- Department of Medicine, McMAster University, Hamilton, ON, Canada
- Research Institute of St. Joe’s Hamilton, St. Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
| | - Parameswaran Nair
- Department of Medicine, McMAster University, Hamilton, ON, Canada
- Research Institute of St. Joe’s Hamilton, St. Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
| | - Manali Mukherjee
- Department of Medicine, McMAster University, Hamilton, ON, Canada
- Research Institute of St. Joe’s Hamilton, St. Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
| |
Collapse
|
5
|
Zhang J, Zhao D, Zhang L, Feng X, Li B, Dong H, Qi Y, Jia Z, Liu F, Zhao S, Zhang J. Impact of HHIP gene polymorphisms on phenotypes, serum IL-17 and IL-18 in COPD patients of the Chinese Han population. Respir Res 2024; 25:386. [PMID: 39468530 PMCID: PMC11520666 DOI: 10.1186/s12931-024-03020-9] [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: 08/30/2024] [Accepted: 10/21/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Genetic factors, including the Hedgehog Interacting Protein (HHIP) gene, play a crucial role in Chronic Obstructive Pulmonary Disease (COPD) susceptibility. This study examines the association between HHIP gene polymorphisms and COPD susceptibility, phenotypes, and serum IL-17 and IL-18 levels in a Han Chinese population. METHODS A case-control study was conducted with 300 COPD patients and 300 healthy controls in Chinese Han population. Participants underwent genotyping for HHIP gene polymorphisms, pulmonary function tests, and quantitative CT scans. DNA samples were sequenced using a custom chip targeting the HHIP gene. Serum IL-17 and IL-18 levels were measured by enzyme-linked immunosorbent assay. Associations between SNPs, COPD susceptibility, and phenotypes were analyzed using logistic and multiple linear regression models, adjusting for confounders. RESULTS Our study identified the rs11100865 polymorphism in the HHIP gene as significantly associated with COPD susceptibility (OR 2.479, 95% CI 1.527-4.024, P = 2.39E-04) after screening 114 SNPs through rigorous quality control. Stratified analyses further indicated this association was particularly in individuals aged 60 or older. Serum levels of IL-17 and IL-18 were significantly elevated in COPD patients compared to controls, with rs11100865 showing a notable association with IL-18 levels (B = 49.654, SE = 19.627, P = 0.012). However, no significant associations were observed between rs11100865 and serum IL-17 levels, COPD-related imaging parameters, or clinical phenotypes. CONCLUSION This study identified a significant association between HHIP gene polymorphisms and COPD susceptibility in a Han Chinese population, with connections to inflammation, but found no significant associations between this SNP and COPD-related imaging or clinical phenotypes. TRIAL REGISTRATION www.chictr.org.cn ID: ChiCTR2300071579 2023-05-18.
Collapse
Affiliation(s)
- Jiajun Zhang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, People's Republic of China
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, People's Republic of China
| | - Di Zhao
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, People's Republic of China
| | - Lili Zhang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, People's Republic of China
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, People's Republic of China
| | - Xueyan Feng
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, People's Republic of China
| | - Beibei Li
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, People's Republic of China
| | - Hui Dong
- Center of Research Equipment Management, General Hospital of Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Yanchao Qi
- Department of Respiratory and Critical Care Medicine, The Second People's Hospital of Shizuishan, Shizuishan, 753000, People's Republic of China
| | - Zun Jia
- Department of Respiratory and Critical Care Medicine, The Fifth People's Hospital of Ningxia, Shizuishan, 753000, People's Republic of China
| | - Fuyun Liu
- Department of Respiratory and Critical Care Medicine, The Fifth People's Hospital of Ningxia, Shizuishan, 753000, People's Republic of China
| | - Shaohui Zhao
- Department of Respiratory and Critical Care Medicine, The Fifth People's Hospital of Ningxia, Shizuishan, 753000, People's Republic of China
| | - Jin Zhang
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, People's Republic of China.
| |
Collapse
|
6
|
Liu T, Woodruff PG, Zhou X. Advances in non-type 2 severe asthma: from molecular insights to novel treatment strategies. Eur Respir J 2024; 64:2300826. [PMID: 38697650 PMCID: PMC11325267 DOI: 10.1183/13993003.00826-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
Asthma is a prevalent pulmonary disease that affects more than 300 million people worldwide and imposes a substantial economic burden. While medication can effectively control symptoms in some patients, severe asthma attacks, driven by airway inflammation induced by environmental and infectious exposures, continue to be a major cause of asthma-related mortality. Heterogeneous phenotypes of asthma include type 2 (T2) and non-T2 asthma. Non-T2 asthma is often observed in patients with severe and/or steroid-resistant asthma. This review covers the molecular mechanisms, clinical phenotypes, causes and promising treatments of non-T2 severe asthma. Specifically, we discuss the signalling pathways for non-T2 asthma including the activation of inflammasomes, interferon responses and interleukin-17 pathways, and their contributions to the subtypes, progression and severity of non-T2 asthma. Understanding the molecular mechanisms and genetic determinants underlying non-T2 asthma could form the basis for precision medicine in severe asthma treatment.
Collapse
Affiliation(s)
- Tao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, China
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Mahajan A, Gunewardena S, Morris A, Clauss M, Dhillon NK. Analysis of MicroRNA Cargo in Circulating Extracellular Vesicles from HIV-Infected Individuals with Pulmonary Hypertension. Cells 2024; 13:886. [PMID: 38891019 PMCID: PMC11172129 DOI: 10.3390/cells13110886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/26/2024] [Accepted: 05/05/2024] [Indexed: 06/20/2024] Open
Abstract
The risk of developing pulmonary hypertension (PH) in people living with HIV is at least 300-fold higher than in the general population, and illicit drug use further potentiates the development of HIV-associated PH. The relevance of extracellular vesicles (EVs) containing both coding as well as non-coding RNAs in PH secondary to HIV infection and drug abuse is yet to be explored. We here compared the miRNA cargo of plasma-derived EVs from HIV-infected stimulant users with (HIV + Stimulants + PH) and without PH (HIV + Stimulants) using small RNA sequencing. The data were compared with 12 PH datasets available in the GEO database to identify potential candidate gene targets for differentially altered miRNAs using the following functional analysis tools: ingenuity pathway analysis (IPA), over-representation analysis (ORA), and gene set enrichment analysis (GSEA). MiRNAs involved in promoting cell proliferation and inhibition of intrinsic apoptotic signaling pathways were among the top upregulated miRNAs identified in EVs from the HIV + Stimulants + PH group compared to the HIV + Stimulants group. Alternatively, the downregulated miRNAs in the HIV + Stimulants + PH group suggested an association with the negative regulation of smooth muscle cell proliferation, IL-2 mediated signaling, and transmembrane receptor protein tyrosine kinase signaling pathways. The validation of significantly differentially expressed miRNAs in an independent set of HIV-infected (cocaine users and nondrug users) with and without PH confirmed the upregulation of miR-32-5p, 92-b-3p, and 301a-3p positively regulating cellular proliferation and downregulation of miR-5571, -4670 negatively regulating smooth muscle proliferation in EVs from HIV-PH patients. This increase in miR-301a-3p and decrease in miR-4670 were negatively correlated with the CD4 count and FEV1/FVC ratio, and positively correlated with viral load. Collectively, this data suggest the association of alterations in the miRNA cargo of circulating EVs with HIV-PH.
Collapse
Affiliation(s)
- Aatish Mahajan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Kansas Medical Center, Mail Stop 3007, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Sumedha Gunewardena
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Alison Morris
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Matthias Clauss
- Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Navneet K. Dhillon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Kansas Medical Center, Mail Stop 3007, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| |
Collapse
|
8
|
Zhang M, Wang S, Guan Q, Wang J, Yan B, Zhang L, Li D. A bidirectional Mendelian randomization study investigating the relationship between genetically predicted systemic inflammatory regulators and chronic obstructive pulmonary disease. Heliyon 2024; 10:e24109. [PMID: 38268600 PMCID: PMC10806290 DOI: 10.1016/j.heliyon.2024.e24109] [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: 08/06/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 01/26/2024] Open
Abstract
Research has shown a connection between inflammation and chronic obstructive pulmonary disease (COPD), however the relationship between inflammation mediators and COPD causation remains unknown. To investigate the causal relationship of mediators of inflammation and COPD, we conducted a two-sample Mendelian randomization (MR) study. In our study, we incorporated 41 regulators of inflammation from 8293 Finnish individuals from genome-wide association studies (GWASs) of COPD corresponding to GWAS summary data for 2115 cases and 454,233 healthy individuals in Europe. Our research validated that higher levels of interleukin 8 (IL-8) are related with a decrease occurrence of COPD (OR = 0.795, 95 % CI = 0.642-0.984, p = 0.035) but that elevated levels of interleukin 18(IL-18) and interleukin 2 (IL-2) may be connected to an amplified risk of COPD (OR = 1.247, 95 % CI = 1.011-1.538; p = 0.039; OR = 1.257, 95 % CI = 1.037-1.523, p = 0.020, respectively). According to our research, cytokines play a crucial role in the development of COPD, and further investigation is necessary to explore the potential of utilizing these cytokines as targets for treatment and prevention of COPD.
Collapse
Affiliation(s)
- Mengyuan Zhang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Shengnan Wang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qingtian Guan
- First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Jianglong Wang
- First Operating Room, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Bailing Yan
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Li Zhang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Dan Li
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
9
|
Cai X, Li Y, Liu Q, Gao X, Li J. Exploration of the Shared Gene Signatures and Molecular Mechanisms between Chronic Bronchitis and Antineutrophil Cytoplasmic Antibody-associated Glomerulonephritis: Evidence from Transcriptome Data. Curr Pharm Des 2024; 30:1966-1984. [PMID: 38847168 DOI: 10.2174/0113816128297623240521070426] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/18/2024] [Indexed: 09/21/2024]
Abstract
BACKGROUND Chronic Bronchitis (CB) is a recurrent and persistent pulmonary inflammation disease. Growing evidence suggests an association between CB and Anti-neutrophil Cytoplasmic Antibody-associated Glomerulonephritis (ANCA-GN). However, the precise mechanisms underlying their association remain unclear. AIMS The purpose of this study was to further explore the molecular mechanism of the occurrence of chronic bronchitis (CB) associated with anti-neutrophil cytoplasmic antibody-associated glomerulonephritis (ANCA- GN). OBJECTIVE Our study aimed to investigate the potential shared pathogenesis of CB-associated ANCA-GN. METHODS Datasets of ANCA (GSE108113 and GSE104948) and CB (GSE151052 and GSE162635) were obtained from the Gene Expression Omnibus (GEO) datasets. Firstly, GSE108113 and GSE151052 were analyzed to identify common differentially expressed genes (DEGs) by Limma package. Based on common DEGs, protein-protein interaction (PPI) network and functional enrichment analyses, including GO, KEGG, and GSEA, were performed. Then, hub genes were identified by degree algorithm and validated in GSE104948 and GSE162635. Further PPI network and functional enrichment analyses were performed on hub genes. Additionally, a competitive ceRNA network was constructed through miRanda and spongeScan. Transcription factors (TFs) were predicted and verified using the TRRUST database. Furthermore, the CIBERSORT algorithm was employed to explore immune cell infiltration. The Drug Gene Interaction Database (DGIDB) was utilized to predict small-molecular compounds of CB and ANCA-GN. RESULTS A total of 963 DEGs were identified in the integrated CB dataset, and 610 DEGs were identified in the integrated ANCA-GN dataset. Totally, we identified 22 common DEGs, of which 10 hub genes (LYZ, IRF1, PIK3CG, IL2RG, NT5E, ARG2, HBEGF, NFATC2, ALPL, and FKBP5) were primarily involved in inflammation and immune responses. Focusing on hub genes, we constructed a ceRNA network composed of 323 miRNAs and 348 lncRNAs. Additionally, five TFs (SP1, RELA, NFKB1, HIF1A, and SP3) were identified to regulate the hub genes. Furthermore, immune cell infiltration results revealed immunoregulation in CB and ANCA-GN. Finally, some small-molecular compounds (Daclizumab, Aldesleukin, and NT5E) were predicted to predominantly regulate inflammation and immunity, especially IL-2. CONCLUSION Our study explores the inflammatory-immune pathways underlying CB-associated ANCA-GN and emphasizes the importance of NETs and lymphocyte differentiation, providing novel insights into the shared pathogenesis and therapeutic targets.
Collapse
Affiliation(s)
- Xiaojing Cai
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yueqiang Li
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingquan Liu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Gao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junhua Li
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Panek I, Liczek M, Gabryelska A, Rakoczy I, Kuna P, Panek M. Inflammasome signalling pathway in the regulation of inflammation - its involvement in the development and exacerbation of asthma and chronic obstructive pulmonary disease. Postepy Dermatol Alergol 2023; 40:487-495. [PMID: 37692274 PMCID: PMC10485761 DOI: 10.5114/ada.2022.118077] [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: 05/10/2021] [Accepted: 09/13/2021] [Indexed: 09/12/2023] Open
Abstract
Inflammasomes are multiprotein oligomers, whose main function is the recruitment and activation of caspase-1, which cleaves the precursor forms of interleukin (IL)-1β and IL-18, generating biologically active cytokines. Activation of inflammasome is an essential component of the innate immune response, and according to recent reports it is involved in epithelial homeostasis and type 2 T helper cell (Th2) differentiation. In recent years, the contribution of inflammasome dependent signalling pathways to the development of inflammatory diseases became a topic of multiple research studies. Asthma and chronic obstructive pulmonary disease (COPD) are the most prevalent obstructive lung diseases. Recent studies have focused on inflammatory aspects of asthma and COPD development, demonstrating the key role of inflammasome-dependent processes. Factors responsible for activation of inflammasome complex are similar in both asthma and COPD and include bacteria, viruses, cigarette smoke, and particulate matter. Some recent studies have revealed that NLRP3 inflammasome plays a crucial role, particularly in the development of acute exacerbations of COPD (AECOPD). Activation of NLRP3 inflammasome has been linked with neutrophilic severe steroid-resistant asthma. Although most of the studies on inflammasomes in asthma and COPD focused on the NLRP3 inflammasome, there are scarce scientific reports linking other inflammasomes such as AIM2 and NLRP1 with obstructive lung diseases. In this mini review we focus on the role of molecular pathways associated with inflammasome in the most prevalent lung diseases such as asthma and COPD. Furthermore, we will try to answer the question of whether inhibition of inflammasome can occur as a modern therapy in these diseases.
Collapse
Affiliation(s)
- Iga Panek
- Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, Lodz, Poland
| | - Maciej Liczek
- Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, Lodz, Poland
| | - Agata Gabryelska
- Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, Lodz, Poland
| | - Igor Rakoczy
- Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, Lodz, Poland
| | - Piotr Kuna
- Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, Lodz, Poland
| | - Michał Panek
- Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
11
|
Kim SH, Adams TS, Hu Q, Shin HJ, Chae G, Lee SE, Sharma L, Kwon HK, Lee FY, Park HJ, Huh WJ, Manning E, Kaminski N, Sauler M, Chen L, Song JW, Kim TK, Kang MJ. VISTA (PD-1H) Is a Crucial Immune Regulator to Limit Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2023; 69:22-33. [PMID: 36450109 PMCID: PMC10324045 DOI: 10.1165/rcmb.2022-0219oc] [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: 05/25/2022] [Accepted: 11/30/2022] [Indexed: 09/09/2023] Open
Abstract
VISTA (V domain immunoglobulin suppressor of T cell activation, also called PD-1H [programmed death-1 homolog]), a novel immune regulator expressed on myeloid and T lymphocyte lineages, is upregulated in mouse and human idiopathic pulmonary fibrosis (IPF). However, the significance of VISTA and its therapeutic potential in regulating IPF has yet to be defined. To determine the role of VISTA and its therapeutic potential in IPF, the expression profile of VISTA was evaluated from human single-cell RNA sequencing data (IPF Cell Atlas). Inflammatory response and lung fibrosis were assessed in bleomycin-induced experimental pulmonary fibrosis models in VISTA-deficient mice compared with wild-type littermates. In addition, these outcomes were evaluated after VISTA agonistic antibody treatment in the wild-type pulmonary fibrosis mice. VISTA expression was increased in lung tissue-infiltrating monocytes of patients with IPF. VISTA was induced in the myeloid population, mainly circulating monocyte-derived macrophages, during bleomycin-induced pulmonary fibrosis. Genetic ablation of VISTA drastically promoted pulmonary fibrosis, and bleomycin-induced fibroblast activation was dependent on the interaction between VISTA-expressing myeloid cells and fibroblasts. Treatment with VISTA agonistic antibody reduced fibrotic phenotypes accompanied by the suppression of lung innate immune and fibrotic mediators. In conclusion, these results suggest that VISTA upregulation in pulmonary fibrosis may be a compensatory mechanism to limit inflammation and fibrosis, and stimulation of VISTA signaling using VISTA agonists effectively limits the fibrotic innate immune landscape and consequent tissue fibrosis. Further studies are warranted to test VISTA as a novel therapeutic target for the IPF treatment.
Collapse
Affiliation(s)
- Sang-Hun Kim
- Section of Pulmonary, Critical Care, and Sleep Medicine
| | | | - Qianni Hu
- Division of Hematology and Oncology, Department of Medicine at Vanderbilt University Medical Center, Nashville, Tennessee; and
| | | | - Ganghee Chae
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang Eun Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care, and Sleep Medicine
| | | | | | - Hong-Jai Park
- Section of Rheumatology, Allergy, and Immunology, Department of Internal Medicine
| | | | | | | | - Maor Sauler
- Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Lieping Chen
- Department of Immunobiology, and
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Jin Woo Song
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Tae Kon Kim
- Division of Hematology and Oncology, Department of Medicine at Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Min-Jong Kang
- Section of Pulmonary, Critical Care, and Sleep Medicine
| |
Collapse
|
12
|
Huber ME, Larson E, Lust TN, Heisler CM, Harriff MJ. Chronic Obstructive Pulmonary Disease and Cigarette Smoke Lead to Dysregulated Mucosal-associated Invariant T-Cell Activation. Am J Respir Cell Mol Biol 2023; 68:90-102. [PMID: 36174211 PMCID: PMC9817907 DOI: 10.1165/rcmb.2022-0131oc] [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: 03/28/2022] [Accepted: 09/29/2022] [Indexed: 02/05/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is associated with airway inflammation, increased infiltration by CD8+ T lymphocytes, and infection-driven exacerbations. Although cigarette smoke is the leading risk factor for COPD, the mechanisms driving the development of COPD in only a subset of smokers are incompletely understood. Lung-resident mucosal-associated invariant T (MAIT) cells play a role in microbial infections and inflammatory diseases. The role of MAIT cells in COPD pathology is unknown. Here, we examined MAIT cell activation in response to cigarette smoke-exposed primary human bronchial epithelial cells (BECs) from healthy, COPD, or smoker donors. We observed significantly higher baseline MAIT cell responses to COPD BECs than healthy BECs. However, infected COPD BECs stimulated a smaller fold increase in MAIT cell response despite increased microbial infection. For all donor groups, cigarette smoke-exposed BECs elicited reduced MAIT cell responses; conversely, cigarette smoke exposure increased ligand-mediated MR1 surface translocation in healthy and COPD BECs. Our data demonstrate that MAIT cell activation is dysregulated in the context of cigarette smoke and COPD. MAIT cells could contribute to cigarette smoke- and COPD-associated inflammation through inappropriate activation and reduced early recognition of bacterial infection, contributing to microbial persistence and COPD exacerbations.
Collapse
Affiliation(s)
| | - Emily Larson
- Portland Veterans Affairs Research Foundation, Portland, Oregon; and
| | - Taylor N. Lust
- Division of Pulmonary, Allergy, and Critical Care Medicine, Oregon Health & Science University, Portland, Oregon
| | - Chelsea M. Heisler
- Division of Pulmonary, Allergy, and Critical Care Medicine, Oregon Health & Science University, Portland, Oregon
| | - Melanie J. Harriff
- Department of Molecular and Microbial Immunology and
- Division of Pulmonary, Allergy, and Critical Care Medicine, Oregon Health & Science University, Portland, Oregon
- Veterans Affairs Portland Health Care System, Portland, Oregon
| |
Collapse
|
13
|
Zhang Y, Wang S, Xia H, Guo J, He K, Huang C, Luo R, Chen Y, Xu K, Gao H, Sheng J, Li L. Identification of Monocytes Associated with Severe COVID-19 in the PBMCs of Severely Infected Patients Through Single-Cell Transcriptome Sequencing. ENGINEERING (BEIJING, CHINA) 2022; 17:161-169. [PMID: 34150352 PMCID: PMC8196473 DOI: 10.1016/j.eng.2021.05.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/07/2021] [Accepted: 05/17/2021] [Indexed: 05/15/2023]
Abstract
Understanding the immunological characteristics of monocytes-including the characteristics associated with fibrosis-in severe coronavirus disease 2019 (COVID-19) is crucial for understanding the pathogenic mechanism of the disease and preventing disease severity. In this study, we performed single-cell transcriptomic sequencing of peripheral blood samples collected from six healthy controls and 14 COVID-19 samples including severe, moderate, and convalescent samples from three severely/critically ill and four moderately ill patients. We found that the monocytes were strongly remodeled in the severely/critically ill patients with COVID-19, with an increased proportion of monocytes and seriously reduced diversity. In addition, we discovered two novel severe-disease-specific monocyte subsets: Mono 0 and Mono 5. These subsets expressed amphiregulin (AREG), epiregulin (EREG), and cytokine interleukin-18 (IL-18) gene, exhibited an enriched erythroblastic leukemia viral oncogene homolog (ErbB) signaling pathway, and appeared to exhibit pro-fibrogenic and pro-inflammation characteristics. We also found metabolic changes in Mono 0 and Mono 5, including increased glycolysis/gluconeogenesis and an increased hypoxia inducible factor-1 (HIF-1) signaling pathway. Notably, one pre-severe sample displayed a monocyte atlas similar to that of the severe/critical samples. In conclusion, our study discovered two novel severe-disease-specific monocyte subsets as potential predictors and therapeutic targets for severe COVID-19. Overall, this study provides potential predictors for severe disease and therapeutic targets for COVID-19 and thus provides a resource for further studies on COVID-19.
Collapse
Affiliation(s)
- Yan Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Shuting Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - He Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jing Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Kangxin He
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Chenjie Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Rui Luo
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yanfei Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Kaijin Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Hainv Gao
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou 310022, China
| | - Jifang Sheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
14
|
Sun J, Li Y. Pyroptosis and respiratory diseases: A review of current knowledge. Front Immunol 2022; 13:920464. [PMID: 36248872 PMCID: PMC9561627 DOI: 10.3389/fimmu.2022.920464] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Pyroptosis is a relatively newly discovered programmed cell death accompanied by an inflammatory response. In the classical view, pyroptosis is mediated by caspases-1,-4,-5,-11 and executed by GSDMD, however, recently it was demonstrated that caspase-3 and-8 also participate in the process of pyroptosis, by cleaving GSDMD/E and GSDMD respectively. Different from autophagy and apoptosis, many pores are formed on the cell membrane during pyroptosis, which makes the cell membrane lose its integrity, eventually leading to the release of cytokines interleukin(IL)-1β and IL-18. When the body is infected with pathogens or exposed to some stimulations, pyroptosis could play an immune defense role. It is found that pyroptosis exists widely in infectious and inflammatory respiratory diseases such as acute lung injury, bronchial dysplasia, chronic obstructive pulmonary disease, and asthma. Excessive pyroptosis may accompany airway inflammation, tissue injury, and airway damage, and induce an inflammatory reaction, leading to more serious damage and poor prognosis of respiratory diseases. This review summarizes the relationship between pyroptosis and related respiratory diseases.
Collapse
|
15
|
Cross-talk between IL-6 trans-signaling and AIM2 inflammasome/IL-1β axes bridge innate immunity and epithelial apoptosis to promote emphysema. Proc Natl Acad Sci U S A 2022; 119:e2201494119. [PMID: 36037355 PMCID: PMC9457334 DOI: 10.1073/pnas.2201494119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pulmonary emphysema is associated with dysregulated innate immune responses that promote chronic pulmonary inflammation and alveolar apoptosis, culminating in lung destruction. However, the molecular regulators of innate immunity that promote emphysema are ill-defined. Here, we investigated whether innate immune inflammasome complexes, comprising the adaptor ASC, Caspase-1 and specific pattern recognition receptors (PRRs), promote the pathogenesis of emphysema. In the lungs of emphysematous patients, as well as spontaneous gp130F/F and cigarette smoke (CS)-induced mouse models of emphysema, the expression (messenger RNA and protein) and activation of ASC, Caspase-1, and the inflammasome-associated PRR and DNA sensor AIM2 were up-regulated. AIM2 up-regulation in emphysema coincided with the biased production of the mature downstream inflammasome effector cytokine IL-1β but not IL-18. These observations were supported by the genetic blockade of ASC, AIM2, and the IL-1 receptor and therapy with AIM2 antagonistic suppressor oligonucleotides, which ameliorated emphysema in gp130F/F mice by preventing elevated alveolar cell apoptosis. The functional requirement for AIM2 in driving apoptosis in the lung epithelium was independent of its expression in hematopoietic-derived immune cells and the recruitment of infiltrating immune cells in the lung. Genetic and inhibitor-based blockade of AIM2 also protected CS-exposed mice from pulmonary alveolar cell apoptosis. Intriguingly, IL-6 trans-signaling via the soluble IL-6 receptor, facilitated by elevated levels of IL-6, acted upstream of the AIM2 inflammasome to augment AIM2 expression in emphysema. Collectively, we reveal cross-talk between the AIM2 inflammasome/IL-1β and IL-6 trans-signaling axes for potential exploitation as a therapeutic strategy for emphysema.
Collapse
|
16
|
Sánchez‐Ovando S, Pavlidis S, Kermani NZ, Baines KJ, Barker D, Gibson PG, Wood LG, Adcock IM, Chung KF, Simpson JL, Wark PA. Pathways linked to unresolved inflammation and airway remodelling characterize the transcriptome in two independent severe asthma cohorts. Respirology 2022; 27:730-738. [PMID: 35673765 PMCID: PMC9540453 DOI: 10.1111/resp.14302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 05/09/2022] [Indexed: 12/12/2022]
Abstract
Background and objective Severe asthma (SA) is a heterogeneous disease. Transcriptomic analysis contributes to the understanding of pathogenesis necessary for developing new therapies. We sought to identify and validate mechanistic pathways of SA across two independent cohorts. Methods Transcriptomic profiles from U‐BIOPRED and Australian NOVocastrian Asthma cohorts were examined and grouped into SA, mild/moderate asthma (MMA) and healthy controls (HCs). Differentially expressed genes (DEGs), canonical pathways and gene sets were identified as central to SA mechanisms if they were significant across both cohorts in either endobronchial biopsies or induced sputum. Results Thirty‐six DEGs and four pathways were shared across cohorts linking to tissue remodelling/repair in biopsies of SA patients, including SUMOylation, NRF2 pathway and oxidative stress pathways. MMA presented a similar profile to HCs. Induced sputum demonstrated IL18R1 as a shared DEG in SA compared with healthy subjects. We identified enrichment of gene sets related to corticosteroid treatment; immune‐related mechanisms; activation of CD4+ T cells, mast cells and IL18R1; and airway remodelling in SA. Conclusion Our results identified differentially expressed pathways that highlight the role of CD4+ T cells, mast cells and pathways linked to ongoing airway remodelling, such as IL18R1, SUMOylation and NRF2 pathways, as likely active mechanisms in the pathogenesis of SA. Transcriptome analysis from endobronchial biopsies and induced sputum from two independent cohorts of adults with severe asthma (SA) (U‐BIOPRED and Australian NOVocastrian Asthma cohort) demonstrated shared differentially expressed pathways previously linked to persistent unresolved inflammation and novel mechanisms of airway remodelling, which may represent potential novel mechanistic pathways involved in the pathogenesis of SA. See relatededitorial
Collapse
Affiliation(s)
- Stephany Sánchez‐Ovando
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine University of Newcastle Newcastle New South Wales Australia
| | | | | | - Katherine Joanne Baines
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine University of Newcastle Newcastle New South Wales Australia
| | - Daniel Barker
- Faculty of Health and Medicine University of Newcastle Newcastle New South Wales Australia
| | - Peter G. Gibson
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine University of Newcastle Newcastle New South Wales Australia
- Respiratory and Sleep Medicine John Hunter Hospital NSW New Lambton Heights New South Wales Australia
| | - Lisa G. Wood
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine University of Newcastle Newcastle New South Wales Australia
| | - Ian M. Adcock
- National Heart and Lung Institute Imperial College London London UK
| | - Kian Fan Chung
- National Heart and Lung Institute Imperial College London London UK
| | - Jodie Louise Simpson
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine University of Newcastle Newcastle New South Wales Australia
| | - Peter A.B. Wark
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine University of Newcastle Newcastle New South Wales Australia
- Respiratory and Sleep Medicine John Hunter Hospital NSW New Lambton Heights New South Wales Australia
| |
Collapse
|
17
|
Matarazzo L, Hernandez Santana YE, Walsh PT, Fallon PG. The IL-1 cytokine family as custodians of barrier immunity. Cytokine 2022; 154:155890. [DOI: 10.1016/j.cyto.2022.155890] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/31/2022] [Accepted: 04/13/2022] [Indexed: 12/12/2022]
|
18
|
Mishra A, Majid D, Kandikattu HK, Yadavalli CS, Upparahalli Venkateshaiah S. Role of IL-18-transformed CD274-expressing eosinophils in promoting airway obstruction in experimental asthma. Allergy 2022; 77:1165-1179. [PMID: 34800294 DOI: 10.1111/all.15180] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/30/2021] [Accepted: 10/18/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND IL-5-dependent residential and IL-18-transformed pathogenic eosinophils have been reported; however, the role of IL-18-transformed CD274-expressing pathogenic eosinophils compared to IL-5-generated eosinophils in promoting airway obstruction in asthma has not yet been examined. METHODS Eosinophils are detected by tissue anti-MBP and anti-EPX immunostaining, CD274 expression by flow cytometry, and airway resistance using the Buxco FinePointe RC system. RESULTS We show that A. fumigatus-challenged wild-type mice, and different gene-deficient mice including naïve CC10-IL-18-transgenic mice, accumulate mostly peribronchial and perivascular CD274-expressing eosinophils except naïve CD2-IL-5-transgenic mice. Additionally, we show that CD2-IL-5 transgenic mice following rIL-18 treatment accumulate high number of CD274-expressing perivascular and peribronchial eosinophils with induced collagen, goblet cell hyperplasia and airway resistance compared to saline-challenged CD2-IL5 transgenic mice. Furthermore, we also show that even A. fumigatus-challenged IL-5 -/- mice and rIL-18 given ΔdblGATA mice accumulate CD274-expressing eosinophil-associated asthma pathogenesis including airway obstruction. Most importantly, we provide evidence that neutralization of CD274 and IL-18 in A. fumigatus-challenged mice ameliorate experimental asthma. Taken together, the data presented are clinically significant in establishing that anti-IL-18 neutralization is a novel immunotherapy to restrict asthma pathogenesis. CONCLUSIONS We demonstrate that IL-18 is critical for inducing asthma pathogenesis, and neutralization of CD274 is a potential immunotherapeutic strategy for asthma.
Collapse
Affiliation(s)
- Anil Mishra
- John W. Deming Department of Medicine Tulane Eosinophilic Disorders Center (TEDC) Section of Pulmonary Diseases Tulane University School of Medicine New Orleans Louisina USA
| | - Dewan Majid
- John W. Deming Department of Medicine Tulane Eosinophilic Disorders Center (TEDC) Section of Pulmonary Diseases Tulane University School of Medicine New Orleans Louisina USA
| | - Hemanth Kumar Kandikattu
- John W. Deming Department of Medicine Tulane Eosinophilic Disorders Center (TEDC) Section of Pulmonary Diseases Tulane University School of Medicine New Orleans Louisina USA
| | - Chandra Sekhar Yadavalli
- John W. Deming Department of Medicine Tulane Eosinophilic Disorders Center (TEDC) Section of Pulmonary Diseases Tulane University School of Medicine New Orleans Louisina USA
| | - Sathisha Upparahalli Venkateshaiah
- John W. Deming Department of Medicine Tulane Eosinophilic Disorders Center (TEDC) Section of Pulmonary Diseases Tulane University School of Medicine New Orleans Louisina USA
| |
Collapse
|
19
|
Majumder D, Sarkar C, Debnath R, Tribedi P, Maiti D. Mechanistic insight into the synergism of IL-27 and IL-28B in regulation of benzo(a)pyrene-induced lung carcinogenesis associated ROS/NF-κB/NLRP3 crosstalk. Chem Biol Interact 2022; 354:109807. [PMID: 34999049 DOI: 10.1016/j.cbi.2022.109807] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/18/2021] [Accepted: 01/05/2022] [Indexed: 01/04/2023]
Abstract
AIM Our previous work depicted that benzo(a)pyrene (BaP)-induced lung cancer associated pulmonary redox imbalance and inflammation were effectively regulated by the combinatorial treatment of IL-27 and IL-28B. So in continuation of that finding the present study was designed to reveal the inflammation regulating signaling network modulated by IL-27 and IL-28B treatment related to BaP-induced lung cancer. METHODS Male Swiss albino mice were treated with BaP to induce lung tumor. Then they received individual as well as combinatorial treatment of IL-27 and IL-28B. At the end of the experimental schedule, the expression of NF-κB signaling proteins, the formation of NLRP3 inflammasome complex and IL-18; IL-17A expression in the lung were observed using Western blot and RT-PCR. The tissue and serum levels of some proinflammatory cytokines were also studied using ELISA. Mast cell density was also studied using toluidine blue staining procedure. RESULTS Treatment with IL-27 or IL-28B alone was successful to regulate the expression of NF-κB signaling proteins and NLRP3 complex in some cases but best attenuation was observed in animals who received both IL-27 and IL-28B in combination. In combination, it was successful in down-regulating the expression of p-ERK1/2 and in reducing the accumulation of mast cells in the lung tissue associated with BaP-induced lung carcinogenesis. The impaired PPARγ expression was also reinstated upon combination treatment. CONCLUSION Altogether, the treatment in combination with IL-27 and IL-28B is an effective regimen to attenuate the ROS/NF-κB/NLRP3 axis associated with BaP-induced lung carcinogenesis.
Collapse
Affiliation(s)
- Debabrata Majumder
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura, 799022 , India.
| | - Chaitali Sarkar
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura, 799022 , India.
| | - Rahul Debnath
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura, 799022 , India.
| | - Prosun Tribedi
- Department of Biotechnology, Jhinger Pole, Diamond Harbour Rd, Sarisha, West Bengal, 743368, India.
| | - Debasish Maiti
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura, 799022 , India.
| |
Collapse
|
20
|
Azargoon A, Kharazmkia A, Kordalivand N, Birjandi M, Mir S. Evaluation of exposure to secondhand smoke and serum level of interleukin 18 in non-smokers. Ann Med Surg (Lond) 2022; 73:103238. [PMID: 35079372 PMCID: PMC8767295 DOI: 10.1016/j.amsu.2021.103238] [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: 10/26/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 11/24/2022] Open
Abstract
Objective Smoking is one significant global health care problems, that not only affects the users but also endangers the health of people inhaling the smoke (passive smoking/secondhand smoke). The serum level of IL-18, an important regulator of inherent and acquired immune response, is affected by cigarette smoking. The aim of this study was to evaluate the effect of secondhand smoke (SHS) exposure on IL-18 serum level in non-smoker adults. Methods In a case-control study, using easy sampling method, 76 non-smokers who were exposed to cigarette smoke for at least 1 h daily during the past year were considered as exposure group, while 76 of their companions without exposure to cigarette smoke (after matching age) were considered as non-exposure group. Serum IL-18 levels were measured for all participants and finally compared between the two groups using Chi-square test. P value < 0.05 was considered to be statistically significant. Results The exposure and non-exposure groups included 58 (76.3%) and 25 (32.9%) males, respectively (P < 0.001). The mean ± SD of age for the exposure and non-exposure groups was 35.42 ± 10.37 and 38.47 ± 12.49 years, respectively (P = 0.102). There was no significant difference between the mean serum levels of IL-18 in the exposure (54.81 ± 57.03 ng/ml) and non-exposure (41.49 ± 42.14 ng/ml) groups (P = 0.104). Conclusion The exposure to secondhand smoke has no significant effect on serum level of IL-18 in exposed adult individuals. However, more studies with larger sample sizes on different populations are required to confirm these results. Smoking is one significant global health care problems. That not only affects the users but also endangers the health of people inhaling the smoke. The serum level of IL-18, an important regulator of inherent and acquired immune response. The exposure to secondhand smoke has no significant effect on serum level of IL-18.
Collapse
Affiliation(s)
- Alireza Azargoon
- Department of Internal Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Kharazmkia
- Department of Clinical Pharmacy, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Nazanin Kordalivand
- Department of Internal Medicine, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mehdi Birjandi
- Department of Biostatistics and Epidemiology, School of Health and Nutrition, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Samareh Mir
- Nutritional Health Research Center, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
21
|
N-Acetylcysteine (NAC) Inhibits Synthesis of IL-18 in Macrophage by Suppressing NLRP3 Expression to Reduce the Production of IFN- γ from NK Cells. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:7596343. [PMID: 34899969 PMCID: PMC8664516 DOI: 10.1155/2021/7596343] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/25/2021] [Indexed: 12/28/2022]
Abstract
Background N-Acetylcysteine (NAC) had exerted antioxidation and anti-inflammation effects on chronic obstructive pulmonary disease (COPD) patients. However, its effect in regulating interleukin- (IL-) 18 was not fully understood. This study was designed to evaluate the specific mechanism of NAC regulating IL-18. Materials and Methods A total of 112 COPD patients and 103 health individuals were recruited in the study. Cytokine level in patients' serum was measured by enzyme-linked immunosorbent assay (ELISA). A COPD mouse model was established by administration of lipopolysaccharide (LPS) and cigarette smoke. The expression of cytokines was measured by ELISA and flow cytometry. Inflammasome-related protein was measured by Western blot. Result NAC could effectively improve the immune status of COPD patients as well as the COPD mouse model by downregulating proinflammation and inflammation cytokines including IL-1β, interferon- (IFN-) γ, tumor necrosis factor- (TNF-) α, and IL-18. It also had the capability to suppress synthesis of IL-18 in macrophage to inhibit the secretion of IFN-γ from natural killer (NK) cells through influencing the inflammasome-related protein in macrophages. Conclusion NAC could effectively inhibit the production of IL-18 by suppressing NLRP3 expression in macrophages to reduce the production of IFN-γ in NK cells.
Collapse
|
22
|
Yun JH, Lee C, Liu T, Liu S, Kim EY, Xu S, Curtis JL, Pinello L, Bowler RP, Silverman EK, Hersh CP, Zhou X. Hedgehog interacting protein-expressing lung fibroblasts suppress lymphocytic inflammation in mice. JCI Insight 2021; 6:e144575. [PMID: 34375314 PMCID: PMC8492352 DOI: 10.1172/jci.insight.144575] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 07/21/2021] [Indexed: 11/30/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is mainly caused by cigarette smoking and characterized by chronic inflammation in vulnerable individuals. However, it is unknown how genetic factors may shape chronic inflammation in COPD. To understand how hedgehog interacting protein, encoded by HHIP gene identified in the genome-wide association study in COPD, plays a role in inflammation, we utilized Hhip+/– mice that present persistent inflammation and emphysema upon aging similar to that observed in human COPD. By performing single-cell RNA sequencing of the whole lung from mice at different ages, we found that Hhip+/– mice developed a cytotoxic immune response with a specific increase in killer cell lectin-like receptor G1–positive CD8+ T cells with upregulated Ifnγ expression recapitulating human COPD. Hhip expression was restricted to a lung fibroblast subpopulation that had increased interaction with CD8+ T lymphocytes in Hhip+/– compared with Hhip+/+ during aging. Hhip-expressing lung fibroblasts had upregulated IL-18 pathway genes in Hhip+/– lung fibroblasts, which was sufficient to drive increased levels of IFN-γ in CD8+ T cells ex vivo. Our finding provides insight into how a common genetic variation contributes to the amplified lymphocytic inflammation in COPD.
Collapse
Affiliation(s)
- Jeong H Yun
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - ChangHee Lee
- Department of Genetics, Harvard Medical School, Boston, United States of America
| | - Tao Liu
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Siqi Liu
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Edy Y Kim
- Department of Medicine, Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Shuang Xu
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Jeffrey L Curtis
- VA Center, University of Michigan Medical School, Ann Arbor, United States of America
| | - Luca Pinello
- Department of Pathology, Massachusetts General Hospital, Boston, United States of America
| | - Russell P Bowler
- Department of Medicine, National Jewish Health, Denver, United States of America
| | - Edwin K Silverman
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Craig P Hersh
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, United States of America
| | - Xiaobo Zhou
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, United States of America
| |
Collapse
|
23
|
Interleukin-18: A Novel Participant in the Occurrence, Development, and Drug Therapy of Obliterative Bronchiolitis Postlung Transplantation. DISEASE MARKERS 2021; 2021:5586312. [PMID: 34367377 PMCID: PMC8337162 DOI: 10.1155/2021/5586312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 07/15/2021] [Indexed: 12/03/2022]
Abstract
Background Obliterative bronchiolitis (OB) was a main cause of deterioration of long-term prognosis in lung transplant recipients after the first posttransplant year. Proinflammatory cytokine interleukin-18 (IL-18) strengthened both the natural immunity and acquired immunity and played an important role in organ transplantation. The roles of IL-18 in the occurrence, development, and drug treatment of OB remained unclear. Methods Small interfering RNA (siRNA) against mouse IL-18 (siRNA-IL-18) was used to silence IL-18 expression. Mouse heterotopic tracheal transplantation model was used to simulate OB. Recipient mice were divided into 5 groups (n = 12) according to donor mouse strains and drug treatment: isograft group, allograft group, allograft+tacrolimus group, allograft+azithromycin group, and allograft+tacrolimus+azithromycin group. The luminal obliteration rates were pathological evaluation. Expressions of cytokines and MMPs were detected by real-time PCR, western blot, and enzyme chain immunosorbent assay (ELISA). Results The luminal obliteration rates of IL-18 of the siRNA-IL-18 group were significantly lower than those of the negative control group (p < 0.0001) and the blank control group (p = 0.0002). mRNA expressions of IFN-γ, EMMPRIN, MMP-8, and MMP-9 of the siRNA-IL-18 group were significantly lower than those of the negative and blank control groups. No tracheal occlusion occurred in grafts of the isograft group. The rates of tracheal occlusion of the allograft group, allograft+tacrolimus group, allograft+azithromycin group, and allograft+tacrolimus+azithromycin group were 72.17 ± 4.66%, 40.33 ± 3.00%, 38.50 ± 2.08%, and 23.33 ± 3.24%, respectively. There were significant differences between the 4 groups (p < 0.001). Serum protein expressions of IL-17 (p = 0.0017), IL-18 (p = 0.0036), IFN-γ (p = 0.0102), and MMP-9 (p = 0.0194) were significantly decreased in the allograft+tacrolimus+azithromycin group compared to the allograft group. Conclusions IL-18 could be a novel molecular involved in the occurrence, development, and drug treatment of OB.
Collapse
|
24
|
Bao W, Zhang X, Jin Y, Hao H, Yang F, Yin D, Chen X, Xue Y, Han L, Zhang M. Factors Associated with the Expression of ACE2 in Human Lung Tissue: Pathological Evidence from Patients with Normal FEV 1 and FEV 1/FVC. J Inflamm Res 2021; 14:1677-1687. [PMID: 33953595 PMCID: PMC8091594 DOI: 10.2147/jir.s300747] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/24/2021] [Indexed: 01/08/2023] Open
Abstract
Background Whether COVID-19 comorbidities and risk factors such as old age, male gender, smoking, obesity, eosinophils and blood types have direct contact with expression of ACE2 and pro-inflammation cytokines in human lung tissues were still unclear. Patients and Methods Sixty-four patients with normal FEV1 and FEV1/FVC underwent thoracotomy for pulmonary nodules were included. Blinded histological assessments were performed by two pathologists. Clinical features and results of the immunohistochemical staining of ACE2 were collected and analyzed. Results ACE2 expressed in alveolar macrophages (most obvious), alveolar epithelia and vascular endothelia, but not in small-airway epithelia. ACE2 expressions are positively related to age (r =0.26, P =0.040), weight (r =0.43, P<0.001), as well as BMI (r = 0.38, P =0.002), and male patients show higher expressions of ACE2 in lungs (P <0.05). ACE2 expressions are negatively related to peripheral eosinophils (r = -0.30, P =0.017). There was no correlation between ABO blood types and ACE2 expression in normal lung tissues (P > 0.05). IL-13 and IL-6R expression in lung tissue increased with age (r =0.26, P <0.05, for both). Conclusion Our pathological evidences showed that the alveolar epithelia, vascular endothelia, and alveolar macrophages are susceptible in human lungs for SARS-CoV-2 infection. The risk factors such as high body weight/BMI, old age, male gender, and eosinopenia may be related to ACE2 expression in human lungs, and associated with more chance to develop the severe cases. IL-6R expression in lung tissue also increased with age. Therefore, weight control and smoking cessation are essential to reduce the susceptibility of SARS-CoV-2 infection, especially in obesity, old or male patients. Peripheral eosinophils monitor is also quite necessary to detect severe tendency in COVID-19 patients.
Collapse
Affiliation(s)
- Wuping Bao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xue Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yubiao Jin
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Huijuan Hao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Fu Yang
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Dongning Yin
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xi Chen
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yishu Xue
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Lei Han
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Min Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
25
|
Camargo LDN, Dos Santos TM, de Andrade FCP, Fukuzaki S, Dos Santos Lopes FDTQ, de Arruda Martins M, Prado CM, Leick EA, Righetti RF, Tibério IDFLC. Bronchial Vascular Remodeling Is Attenuated by Anti-IL-17 in Asthmatic Responses Exacerbated by LPS. Front Pharmacol 2020; 11:1269. [PMID: 33013361 PMCID: PMC7500412 DOI: 10.3389/fphar.2020.01269] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/31/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction Although the major alterations associated with asthma are related to the airways, there is also evidence of the importance of peribronchial vascular inflammation and remodeling in its pathophysiology. Objectives To determine the effects of anti-IL-17 therapy on peribronchial vessels of an asthma model exacerbated by lipopolysaccharide. Methods We evaluated several factors, including lung function, inflammation, oxidative stress, vascular remodeling, and signaling pathways present in the peribronchial vessels of 66 male BALB/c mice exposed to ovalbumin and treated (or not) treated with anti-IL-17. Twenty-four hours before the end of the experimental protocol, groups of sensitized animals (OVA–LPS and OVA–LPS anti-IL-17) also received LPS. Results The OVA–LPS-anti-IL-17 group presented a decrease in several factors [airway resistance and elastance, bronchoalveolar lavage fluid (BALF) cell counts, inflammatory response, eosinophils, TSLP, IL-33, TARC, TNF-α, CD4+, CD8+, IL-4, IL-6, IL-10, IL-17, and VEGF positive cells/104μm2, peribronchovascular edema, and angiogenesis], including remodeling (MMP-9, MMP-12, TIMP-1 and TGF-β positive cells and volume fraction of collagen fibers I, collagen fibers III, collagen fibers V, decorin, lumican, actin, biglycan, fibronectin, and integrin), oxidative stress (iNOS positive cells and volume fraction of PGF2α), and signaling pathways (FoxP3), as well as dendritic cells, NF-kB, ROCK-1, ROCK-2, STAT-1, and phosphor-STAT1-positive cells compared to OVA–LPS (p < 0.05). Conclusions In this model of LPS-induced asthma exacerbation, IL-17 inhibition represents a promising therapeutic strategy, indicating the potential of bronchial vascular control of Th2 and Th17 responses and the activation of the remodeling and oxidative stress pathways, associated with the control of signaling pathways.
Collapse
Affiliation(s)
- Leandro do Nascimento Camargo
- Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, Brazil.,Serviço de Reabilitação, Hospital Sírio-Libanês, São Paulo, Brazil
| | - Tabata Maruyama Dos Santos
- Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, Brazil.,Serviço de Reabilitação, Hospital Sírio-Libanês, São Paulo, Brazil
| | | | - Silvia Fukuzaki
- Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, Brazil
| | | | | | - Carla Máximo Prado
- Department of Bioscience, Federal University of São Paulo, Santos, Brazil
| | | | - Renato Fraga Righetti
- Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, Brazil.,Serviço de Reabilitação, Hospital Sírio-Libanês, São Paulo, Brazil
| | | |
Collapse
|
26
|
Lee HG, Cho MZ, Choi JM. Bystander CD4 + T cells: crossroads between innate and adaptive immunity. Exp Mol Med 2020; 52:1255-1263. [PMID: 32859954 PMCID: PMC8080565 DOI: 10.1038/s12276-020-00486-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/21/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
T cells are the central mediators of both humoral and cellular adaptive immune responses. Highly specific receptor-mediated clonal selection and expansion of T cells assure antigen-specific immunity. In addition, encounters with cognate antigens generate immunological memory, the capacity for long-term, antigen-specific immunity against previously encountered pathogens. However, T-cell receptor (TCR)-independent activation, termed “bystander activation”, has also been found. Bystander-activated T cells can respond rapidly and secrete effector cytokines even in the absence of antigen stimulation. Recent studies have rehighlighted the importance of antigen-independent bystander activation of CD4+ T cells in infection clearance and autoimmune pathogenesis, suggesting the existence of a distinct innate-like immunological function performed by conventional T cells. In this review, we discuss the inflammatory mediators that activate bystander CD4+ T cells and the potential physiological roles of these cells during infection, autoimmunity, and cancer. Immune cells that become activated in the absence of antigen stimulation could be harnessed in the fight against infection, autoimmunity, and cancer. Je-Min Choi and colleagues from Hanyang University in Seoul, South Korea, review how the immune system can deploy helper T cells through an unusual process called bystander activation. Most T cells become activated only after receptors on their surface bind to specific cognate antigen. In contrast, bystander T cells are activated non-specifically in response to cytokines and other pro-inflammatory mediators. Studies have shown that this cell population has a variety of protective and pathogenic functions, for example, guarding against multiple sclerosis, aggravating the symptoms of parasitic infections and promoting antitumor immunity. A better understanding of these immune cells could lead to new therapeutic options for these diseases.
Collapse
Affiliation(s)
- Hong-Gyun Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Min-Zi Cho
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea. .,Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea. .,Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Relationship of Serum Levels of IL-17, IL-18, TNF- α, and Lung Function Parameters in Patients with COPD, Asthma-COPD Overlap, and Bronchial Asthma. Mediators Inflamm 2020; 2020:4652898. [PMID: 32733164 PMCID: PMC7372292 DOI: 10.1155/2020/4652898] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 12/26/2019] [Accepted: 01/21/2020] [Indexed: 12/16/2022] Open
Abstract
Determination of markers of systemic inflammation is one of the important directions in the study of pathogenesis and improvement of diagnosis of chronic obstructive pulmonary disease (COPD), asthma-COPD overlap (ACO), and bronchial asthma (BA). The aim of our work was a comparative study of the features of changes in serum levels of IL-17, IL-18, and TNF-α in patients with COPD, ACO, and BA with various severity of the disease, as well as evaluation of the relationship between the level of these cytokines and lung ventilation function. A total of 147 patients with COPD (n = 58), ACO (n = 57), and BA (n = 32) during a stable period have been examined in this study. The control group included 21 healthy nonsmokers with similar sex-age indicators. Serum levels of IL-17, IL-18, and TNF-α were determined by ELISA. The concentrations of these cytokines in the circulation in the studied patients with COPD, ACO, and BA were higher than those in healthy nonsmokers (p ≤ 0.001). IL-17 and IL-18 levels in the blood serum were comparable in all examined patients. The mean TNF-α concentrations in the circulation in COPD and ACO were significantly higher than those in BA (p < 0.001). In patients with COPD, the levels of IL-17 and TNF-α increased progressively against the background of a decrease in numerous spirometric indicators, which allows us to consider these cytokines as systemic biomarkers of disease severity. In BA, the inverse correlations between the level of IL-17 and FEV1/FVC (%) and FEV1 have been found. In patients with ACO, the increase in IL-18 levels was associated with a decrease in FEV1 and TNF-α with FEV1/FVC (%). These findings indicate that IL-17, IL-18, and TNF-α can participate in the mechanisms of systemic inflammation and the genesis of disorders of airway obstruction in COPD, AСO, and BA. An increase in the levels of IL-17 and TNF-α may be associated with impaired bronchial patency in COPD and BA. The established associations of the IL-18 concentration in the blood serum and FEV1 only in patients with ACO allow using the level of IL-18 as a potential marker of the degree of impaired airway obstruction in this disease.
Collapse
|
28
|
Domingo-Gonzalez R, Zanini F, Che X, Liu M, Jones RC, Swift MA, Quake SR, Cornfield DN, Alvira CM. Diverse homeostatic and immunomodulatory roles of immune cells in the developing mouse lung at single cell resolution. eLife 2020; 9:e56890. [PMID: 32484158 PMCID: PMC7358008 DOI: 10.7554/elife.56890] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
At birth, the lungs rapidly transition from a pathogen-free, hypoxic environment to a pathogen-rich, rhythmically distended air-liquid interface. Although many studies have focused on the adult lung, the perinatal lung remains unexplored. Here, we present an atlas of the murine lung immune compartment during early postnatal development. We show that the late embryonic lung is dominated by specialized proliferative macrophages with a surprising physical interaction with the developing vasculature. These macrophages disappear after birth and are replaced by a dynamic mixture of macrophage subtypes, dendritic cells, granulocytes, and lymphocytes. Detailed characterization of macrophage diversity revealed an orchestration of distinct subpopulations across postnatal development to fill context-specific functions in tissue remodeling, angiogenesis, and immunity. These data both broaden the putative roles for immune cells in the developing lung and provide a framework for understanding how external insults alter immune cell phenotype during a period of rapid lung growth and heightened vulnerability.
Collapse
Affiliation(s)
- Racquel Domingo-Gonzalez
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| | - Fabio Zanini
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South WalesSydneyAustralia
| | - Xibing Che
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
| | - Min Liu
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| | - Robert C Jones
- Department of Bioengineering, Stanford UniversityStanfordUnited States
| | - Michael A Swift
- Department of Chemical and Systems Biology, Stanford UniversityStanfordUnited States
| | - Stephen R Quake
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
- Department of Applied Physics, Stanford UniversityStanfordUnited States
| | - David N Cornfield
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
| | - Cristina M Alvira
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
29
|
Ding L, Wang H, Geng H, Cui N, Huang F, Zhu X, Zhu X. Prediction of Bronchopulmonary Dysplasia in Preterm Infants Using Postnatal Risk Factors. Front Pediatr 2020; 8:349. [PMID: 32676490 PMCID: PMC7333538 DOI: 10.3389/fped.2020.00349] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/26/2020] [Indexed: 01/12/2023] Open
Abstract
Objective: To identify postnatal risk factors for bronchopulmonary dysplasia (BPD) development in preterm infants with gestational age ≤32 weeks. Methods: Seventy-two preterm infants(30 with BPD and 42 non-BPD controls) admitted in the neonatal intensive care unit (NICU) of the Children's Hospital of Soochow University during 2017 were enrolled in this prospective longitudinal study. Perinatal clinical data, a neonatal critical illness score (NCIS), different soluble B7-H3(sB7-H3), and interleukin-18 (IL-18) levels by days after birth were collected. An early predictive model for BPD development was established based on clinical data using multiple logistic regression analysis. And the sensitivity and specificity of the model were assesed by ROC curve. Results: Electrolyte disturbances, hemodynamically significant patent ductus arteriosus (hs-PDA), and the age that infants achieved 120 kcal/kg.d via enteral feeding ≥40 days after birth were found to be associated with the BPD pathogenesis. Serum sB7-H3, IL-18, and NCIS were significantly higher in the BPD group compared to the non-BPD group (p < 0.05). BPD group had significantly lower enteral fluid and caloric intake compared to the non-BPD group at 1, 7, 14, and 28 days after birth. The risk factors were analyzed by multiple logistic regression and a predictive model of a combination of sB7-H3 (day 7), IL-18 (day 14), NCIS, and clinical risk factors was evaluated via ROC curve with an area under the curve (AUC) of 0.960 having sensitivity of 86.7% and a specificity of 97.6%, respectively. Conclusion: The causes of BPD are multifactorial postnatal risk factors. And the combination of sB7-H3 (day 7), IL-18 (day 14), NCIS, and clinical risk factors (electrolyte disturbances, hs-PDA, and the age that infants achieved 120 kcal/kg.d via enteral feeding ≥40 days after birth) might be served as an optimal predictive model for the occurrence of BPD.
Collapse
Affiliation(s)
- Li Ding
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Huawei Wang
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Haifeng Geng
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Ningxun Cui
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Fengxia Huang
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Xueping Zhu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Xiaoli Zhu
- Department of Intervention, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
30
|
Ding L, Zhao X, Zhu N, Zhao M, Hu Q, Ni Y. The balance of serum IL-18/IL-37 levels is disrupted during the development of oral squamous cell carcinoma. Surg Oncol 2019; 32:99-107. [PMID: 31983439 DOI: 10.1016/j.suronc.2019.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/07/2019] [Accepted: 12/05/2019] [Indexed: 11/24/2022]
Abstract
Growing evidences have demonstrated a pivotal role of chronic inflammation in oral squamous cell carcinoma (OSCC) through the modulation of inflammatory cells and cytokine production. IL-37 is newly discovered anti-inflammatory member of IL-1 family and can bind to IL-18 receptor to inhibit IL-18 (pro-inflammatory member of IL-1 family) function. Investigation on the balance of IL-18/IL-37 would provide new insights into the function of IL-1 family in OSCC. Thus, serum IL-18 and IL-37 levels of OSCC patients (n = 108), leukoplakia patients (n = 40), and healthy donors (n = 36) were collected to analyze the balance of IL-18 and IL-37, and also determine their diagnostic value and prognostic significance in OSCC. The results showed that OSCC patients had high IL-18 and low IL-37 levels in serum and peripheral blood mononuclear cell (PBMC). The ratio of IL-18/IL-37 in serum efficiently distinguished non-cancer individuals from OSCC patients (cut off value: 2.15). Moreover, patients with high IL-18 and low IL-37 were susceptible to develop advanced tumor stage and lymph node metastasis (Odd ratios of IL-18/IL-37 is 4.903 and 12.613, respectively). Meanwhile, higher IL-18/IL-37 ratio could predict shorter overall survival and disease-free survival of OSCC patients, although it was not an independent prognostic factor. We further analyze the correlations of serum IL-18/IL-37 with immunocytes in peripheral blood and found that high IL-18 level was associated with more CD19+ B cells, while serum IL-37 seem to be associated with reduced percentage of CD3+CD8+ T cells, indicating its balance could change the adaptive immune response. Unexpectedly, we first revealed the different function of IL-18/IL-37 in serum and tumor tissues. High mRNA expression of IL-18 in tumor tissues correlated with low lymph node metastasis rate and low tumor stage, which was contradictory to the pro-tumor role of IL-18 in serum. In conclusion, enhanced ratio of IL-18/IL-37 level in serum could be an efficient biomarker for OSCC. Its balance might regulate CD19+ B cells and CD3+ CD8+ T cells for OSCC progression.
Collapse
Affiliation(s)
- Liang Ding
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xingxing Zhao
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Nisha Zhu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Mengxiang Zhao
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qingang Hu
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Yanhong Ni
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
31
|
Chen S, Zhang Z, Chen L, Zhang J. miRNA‑101‑3p.1 as an independent diagnostic biomarker aggravates chronic obstructive pulmonary disease via activation of the EGFR/PI3K/AKT signaling pathway. Mol Med Rep 2019; 20:4293-4302. [PMID: 31545413 DOI: 10.3892/mmr.2019.10657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/28/2019] [Indexed: 11/06/2022] Open
Abstract
Exploring independent biomarkers and delineating pathogenic mechanisms could improve the early diagnosis and treatment of chronic obstructive pulmonary disease (COPD). In the present study, a study was conducted to determine the diagnostic potential of miRNA‑101‑3p.1 in identifying stable COPD (SCOPD) and acute exacerbation of COPD (AECOPD) patients and to reveal the molecular mechanism by which miRNA‑101‑3p.1 regulates COPD progression. miRNA‑101‑3p.1 profiles in peripheral blood mononuclear cells of COPD patients were evaluated. Subsequently, receiver operating characteristic curves were created to demonstrate the diagnostic accuracy of miRNA‑101‑3p.1 in discriminating SCOPD and AECOPD. Finally, the molecular mechanism by which miRNA‑101‑3p.1 regulates COPD progression was explored. The present study revealed that patients with COPD, and especially patients with AECOPD, had significantly increased levels of miRNA‑101‑3p.1 and the level of miRNA‑101‑3p.1 was closely correlated with CAT score and FEV1% predicted. Notably, miRNA‑101‑3p.1 accurately discriminated SCOPD and AECOPD. Furthermore, increasing miRNA‑101‑3p.1 promoted cell proliferation and induced the expression of inflammatory cytokines. Mechanistic investigations revealed that miRNA‑101‑3p.1 inhibited the expression of von Hippel‑Lindau tumor suppressor (pVHL) and ubiquitin conjugating enzyme E2 D1 (UBE2D1). pVHL and UBE2D1 co‑upregulated HIF‑1α, and HIF‑1α mediated activation of the EGFR/PI3K/AKT signaling pathway. The present results collectively demonstrated that miRNA‑101‑3p.1 could act as an independent biomarker for the diagnosis of SCOPD and AECOPD, and that miRNA‑101‑3p.1 facilitates COPD progression by activating the EGFR/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Shuifang Chen
- Respiratory Department of Internal Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Zeying Zhang
- Respiratory Department of Internal Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Lina Chen
- Respiratory Department of Internal Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Jianli Zhang
- Respiratory Department of Internal Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
32
|
Ke Q, Yang L, Cui Q, Diao W, Zhang Y, Xu M, He B. Ciprofibrate attenuates airway remodeling in cigarette smoke-exposed rats. Respir Physiol Neurobiol 2019; 271:103290. [PMID: 31525465 DOI: 10.1016/j.resp.2019.103290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023]
Abstract
Airway remodeling is a key pathological lesion in chronic obstructive pulmonary disease (COPD), and it leads to poorly reversible airway obstruction. Current pharmacological interventions are ineffective at controlling airway remodeling. To address this issue, we queried the Connectivity Map (cMap) database to screen for drug candidates that had the potential to dilate the bronchus and inhibit airway smooth muscle (ASM) proliferation. We identified ciprofibrate as a drug candidate. Ciprofibrate inhibited cigarette smoke extract-induced rat ASM cell contraction and proliferation in vitro. We exposed Sprague-Dawley (SD) rats to clean air or cigarette smoke (CS) and treated the rats with ciprofibrate. Ciprofibrate improved pulmonary function, inhibited airway hypercontraction, and ameliorated morphological small airway remodeling, including airway smooth muscle proliferation, in CS-exposed rats. Ciprofibrate also significantly reduced IL-1β, IL-12p70, IL-17A and IL-18 expression, which are related to airway remodeling, in the sera of CS-exposed rats. These findings indicate that ciprofibrate could attenuate airway remodeling in CS-exposed rats.
Collapse
Affiliation(s)
- Qian Ke
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China.
| | - Lin Yang
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China.
| | - Qinghua Cui
- Department of Biomedical Informatics, Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Wenqi Diao
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China.
| | - Youyi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China; Beijing Key Laboratory of Cardiovascular Receptors Research. Beijing, China.
| | - Ming Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China; Beijing Key Laboratory of Cardiovascular Receptors Research. Beijing, China; Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Bei He
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
33
|
Assessment of Cytokines, Biochemical Markers of Malnutrition and Frailty Syndrome Patients Considered for Lung Transplantation. Transplant Proc 2019; 51:2009-2013. [DOI: 10.1016/j.transproceed.2019.04.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 04/05/2019] [Indexed: 11/21/2022]
|
34
|
Cade BE, Chen H, Stilp AM, Louie T, Ancoli-Israel S, Arens R, Barfield R, Below JE, Cai J, Conomos MP, Evans DS, Frazier-Wood AC, Gharib SA, Gleason KJ, Gottlieb DJ, Hillman DR, Johnson WC, Lederer DJ, Lee J, Loredo JS, Mei H, Mukherjee S, Patel SR, Post WS, Purcell SM, Ramos AR, Reid KJ, Rice K, Shah NA, Sofer T, Taylor KD, Thornton TA, Wang H, Yaffe K, Zee PC, Hanis CL, Palmer LJ, Rotter JI, Stone KL, Tranah GJ, Wilson JG, Sunyaev SR, Laurie CC, Zhu X, Saxena R, Lin X, Redline S. Associations of variants In the hexokinase 1 and interleukin 18 receptor regions with oxyhemoglobin saturation during sleep. PLoS Genet 2019; 15:e1007739. [PMID: 30990817 PMCID: PMC6467367 DOI: 10.1371/journal.pgen.1007739] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 10/03/2018] [Indexed: 12/12/2022] Open
Abstract
Sleep disordered breathing (SDB)-related overnight hypoxemia is associated with cardiometabolic disease and other comorbidities. Understanding the genetic bases for variations in nocturnal hypoxemia may help understand mechanisms influencing oxygenation and SDB-related mortality. We conducted genome-wide association tests across 10 cohorts and 4 populations to identify genetic variants associated with three correlated measures of overnight oxyhemoglobin saturation: average and minimum oxyhemoglobin saturation during sleep and the percent of sleep with oxyhemoglobin saturation under 90%. The discovery sample consisted of 8,326 individuals. Variants with p < 1 × 10(-6) were analyzed in a replication group of 14,410 individuals. We identified 3 significantly associated regions, including 2 regions in multi-ethnic analyses (2q12, 10q22). SNPs in the 2q12 region associated with minimum SpO2 (rs78136548 p = 2.70 × 10(-10)). SNPs at 10q22 were associated with all three traits including average SpO2 (rs72805692 p = 4.58 × 10(-8)). SNPs in both regions were associated in over 20,000 individuals and are supported by prior associations or functional evidence. Four additional significant regions were detected in secondary sex-stratified and combined discovery and replication analyses, including a region overlapping Reelin, a known marker of respiratory complex neurons.These are the first genome-wide significant findings reported for oxyhemoglobin saturation during sleep, a phenotype of high clinical interest. Our replicated associations with HK1 and IL18R1 suggest that variants in inflammatory pathways, such as the biologically-plausible NLRP3 inflammasome, may contribute to nocturnal hypoxemia.
Collapse
Affiliation(s)
- Brian E. Cade
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, United States of America
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX United States of America
- Center for Precision Health, School of Public Health and School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX United States of America
| | - Adrienne M. Stilp
- Department of Biostatistics, University of Washington, Seattle, WA United States of America
| | - Tin Louie
- Department of Biostatistics, University of Washington, Seattle, WA United States of America
| | - Sonia Ancoli-Israel
- Department of Psychiatry, University of California, San Diego, CA, United States of America
| | - Raanan Arens
- The Children’s Hospital at Montefiore, Division of Respiratory and Sleep Medicine, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Richard Barfield
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
| | - Jennifer E. Below
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Jianwen Cai
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, United States of America
| | - Matthew P. Conomos
- Department of Biostatistics, University of Washington, Seattle, WA United States of America
| | - Daniel S. Evans
- California Pacific Medical Center Research Institute, San Francisco, CA, United States of America
| | - Alexis C. Frazier-Wood
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States of America
| | - Sina A. Gharib
- Computational Medicine Core, Center for Lung Biology, UW Medicine Sleep Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle WA, United States of America
| | - Kevin J. Gleason
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America
- Department of Public Health Sciences, University of Chicago, Chicago, IL, United States of America
| | - Daniel J. Gottlieb
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America
- VA Boston Healthcare System, Boston, MA, United States of America
| | - David R. Hillman
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - W. Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA United States of America
| | - David J. Lederer
- Departments of Medicine and Epidemiology, Columbia University, New York, NY, United States of America
| | - Jiwon Lee
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America
| | - Jose S. Loredo
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, UC San Diego School of Medicine, La Jolla, CA, United States of America
| | - Hao Mei
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Sutapa Mukherjee
- Sleep Health Service, Respiratory and Sleep Services, Southern Adelaide Local Health Network, Adelaide, South Australia
- Adelaide Institute for Sleep Health, Flinders University, Adelaide, South Australia
| | - Sanjay R. Patel
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Wendy S. Post
- Division of Cardiology, Johns Hopkins University, Baltimore, MD, United States of America
| | - Shaun M. Purcell
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, United States of America
| | - Alberto R. Ramos
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Kathryn J. Reid
- Department of Neurology, Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Ken Rice
- Department of Biostatistics, University of Washington, Seattle, WA United States of America
| | - Neomi A. Shah
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America
- Department of Biostatistics, University of Washington, Seattle, WA United States of America
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Timothy A. Thornton
- Department of Biostatistics, University of Washington, Seattle, WA United States of America
| | - Heming Wang
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, United States of America
| | - Kristine Yaffe
- Department of Psychiatry, Neurology, and Epidemiology and Biostatistics, University of California at San Francisco, San Francisco, CA, United States of America
- San Francisco VA Medical Center, San Francisco, CA, United States of America
| | - Phyllis C. Zee
- Department of Neurology, Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Craig L. Hanis
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX United States of America
| | - Lyle J. Palmer
- School of Public Health, University of Adelaide, South Australia, Australia
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Katie L. Stone
- California Pacific Medical Center Research Institute, San Francisco, CA, United States of America
| | - Gregory J. Tranah
- California Pacific Medical Center Research Institute, San Francisco, CA, United States of America
| | - James G. Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson MS, United States of America
| | - Shamil R. Sunyaev
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, United States of America
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, United States of America
- Division of Medical Sciences, Harvard Medical School, Boston, MA, United States of America
| | - Cathy C. Laurie
- Department of Biostatistics, University of Washington, Seattle, WA United States of America
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States of America
| | - Richa Saxena
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, United States of America
- Center for Genomic Medicine and Department of Anesthesia, Pain, and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, United States of America
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America
- Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| |
Collapse
|
35
|
Pathogenic function of bystander-activated memory-like CD4 + T cells in autoimmune encephalomyelitis. Nat Commun 2019; 10:709. [PMID: 30755603 PMCID: PMC6372661 DOI: 10.1038/s41467-019-08482-w] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 01/10/2019] [Indexed: 12/22/2022] Open
Abstract
T cells generate antigen-specific immune responses to their cognate antigen as a hallmark of adaptive immunity. Despite the importance of antigen-specific T cells, here we show that antigen non-related, bystander memory-like CD4+ T cells also significantly contribute to autoimmune pathogenesis. Transcriptome analysis demonstrates that interleukin (IL)-1β- and IL-23-prime T cells that express pathogenic TΗ17 signature genes such as RORγt, CCR6, and granulocyte macrophage colony-stimulating factor (GM-CSF). Importantly, when co-transferred with myelin-specific 2D2 TCR-transgenic naive T cells, unrelated OT-II TCR-transgenic memory-like TH17 cells infiltrate the spinal cord and produce IL-17A, interferon (IFN)-γ, and GM-CSF, increasing the susceptibility of the recipients to experimental autoimmune encephalomyelitis in an IL-1 receptor-dependent manner. In humans, IL-1R1high memory CD4+ T cells are major producers of IL-17A and IFN-γ in response to IL-1β and IL-23. Collectively, our findings reveal the innate-like pathogenic function of antigen non-related memory CD4+ T cells, which contributes to the development of autoimmune diseases. T cells express specific T cell receptors (TCR) to recognise antigens and initiate adaptive immune responses. Here the authors show, in a mouse model of autoimmune encephalomyelitis, that memory-like CD4 T cells expressing unrelated TCR can also infiltrate the spinal cord and contribute to autoimmunity.
Collapse
|
36
|
Matsuo T, Hashimoto M, Ito I, Kubo T, Uozumi R, Furu M, Ito H, Fujii T, Tanaka M, Terao C, Kono H, Mori M, Hamaguchi M, Yamamoto W, Ohmura K, Morita S, Mimori T. Interleukin-18 is associated with the presence of interstitial lung disease in rheumatoid arthritis: a cross-sectional study. Scand J Rheumatol 2018; 48:87-94. [PMID: 30269670 DOI: 10.1080/03009742.2018.1477989] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Serum interleukin-18 (IL-18) levels are increased in patients with interstitial lung disease (ILD). In addition, IL-18 levels are increased in patients with rheumatoid arthritis (RA) and are associated with arthritis activity. We determined whether increased IL-18 levels are associated with ILD in RA. METHOD RA patients were enrolled using an RA cohort database. Plasma IL-18 levels were measured by enzyme-linked immunosorbent assay. ILD was determined by a pulmonologist and a radiologist based on chest radiography and computed tomography findings. IL-18 levels for RA with ILD and RA without ILD were compared. Associations between ILD and various markers including IL-18 and confounding factors (e.g. smoking history) were investigated by logistic regression analysis. Diagnostic values of IL-18 for the presence of ILD were investigated using receiver operating characteristics curve analysis. RESULTS ILD was complicated in 8.2% (n = 26) of the study population (N = 312). Plasma IL-18 levels were higher for RA patients with ILD than for RA patients without ILD (721.0 ± 481.4 vs 436.8 ± 438.9 pg/mL, p < 0.001). IL-18, Krebs von den Lungen-6, and anti-cyclic citrullinated peptide antibody titre and glucocorticoid doses were independently associated with the presence of ILD during multivariate logistic regression analysis. Sensitivity and specificity of IL-18 levels for the detection of ILD in RA patients were 65.3% and 76.3%, respectively (area under the curve = 0.73). CONCLUSION Plasma IL-18 levels were higher for RA patients with ILD than for those without ILD. Increased IL-18 levels were associated with the presence of ILD.
Collapse
Affiliation(s)
- T Matsuo
- a Department of Rheumatology and Clinical Immunology, Graduate School of Medicine , Kyoto University , Kyoto , Japan
| | - M Hashimoto
- b Department of Advanced Medicine for Rheumatic Diseases , Graduate School of Medicine, Kyoto University , Kyoto , Japan
| | - I Ito
- c Department of Respiratory Medicine, Graduate School of Medicine , Kyoto University , Kyoto , Japan
| | - T Kubo
- d Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine , Kyoto University , Kyoto , Japan
| | - R Uozumi
- e Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine , Kyoto University , Kyoto , Japan
| | - M Furu
- b Department of Advanced Medicine for Rheumatic Diseases , Graduate School of Medicine, Kyoto University , Kyoto , Japan
| | - H Ito
- b Department of Advanced Medicine for Rheumatic Diseases , Graduate School of Medicine, Kyoto University , Kyoto , Japan
| | - T Fujii
- f Department of Rheumatology and Clinical Immunology , Wakayama Medical University , Wakayama , Japan
| | - M Tanaka
- b Department of Advanced Medicine for Rheumatic Diseases , Graduate School of Medicine, Kyoto University , Kyoto , Japan
| | - C Terao
- g Center for Genomic Medicine, Graduate School of Medicine , Kyoto University , Kyoto , Japan.,h Center for the Promotion of Interdisciplinary Education and Research , Kyoto University , Kyoto , Japan
| | - H Kono
- i Department of Internal Medicine , Teikyo University School of Medicine , Tokyo , Japan
| | - M Mori
- a Department of Rheumatology and Clinical Immunology, Graduate School of Medicine , Kyoto University , Kyoto , Japan
| | - M Hamaguchi
- j Department of Diabetology , Kameoka Municipal Hospital , Kyoto , Japan
| | - W Yamamoto
- k Department of Health Information Management , Kurashiki Sweet Hospital , Kurashiki , Japan
| | - K Ohmura
- a Department of Rheumatology and Clinical Immunology, Graduate School of Medicine , Kyoto University , Kyoto , Japan
| | - S Morita
- e Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine , Kyoto University , Kyoto , Japan
| | - T Mimori
- a Department of Rheumatology and Clinical Immunology, Graduate School of Medicine , Kyoto University , Kyoto , Japan
| |
Collapse
|
37
|
Abstract
Initially described as an interferon (IFN)γ‐inducing factor, interleukin (IL)‐18 is indeed involved in Th1 and NK cell activation, but also in Th2, IL‐17‐producing γδ T cells and macrophage activation. IL‐18, a member of the IL‐1 family, is similar to IL‐1β for being processed by caspase 1 to an 18 kDa‐biologically active mature form. IL‐18 binds to its specific receptor (IL‐18Rα, also known as IL‐1R7) forming a low affinity ligand chain. This is followed by recruitment of the IL‐18Rβ chain. IL‐18 then uses the same signaling pathway as IL‐1 to activate NF‐kB and induce inflammatory mediators such as adhesion molecules, chemokines and Fas ligand. IL‐18 also binds to the circulating high affinity IL‐18 binding protein (BP), such as only unbound free IL‐18 is active. IL‐18Rα may also bind IL‐37, another member of the IL‐1 family, but in association with the negative signaling chain termed IL‐1R8, which transduces an anti‐inflammatory signal. IL‐18BP also binds IL‐37 and this acts as a sink for the anti‐inflammatory properties of IL‐37. There is now ample evidence for a role of IL‐18 in various infectious, metabolic or inflammatory diseases such as influenza virus infection, atheroma, myocardial infarction, chronic obstructive pulmonary disease, or Crohn's disease. However, IL‐18 plays a very specific role in the pathogenesis of hemophagocytic syndromes (HS) also termed Macrophage Activation Syndrome. In children affected by NLRC4 gain‐of‐function mutations, IL‐18 circulates in the range of tens of nanograms/mL. HS is treated with the IL‐1 Receptor antagonist (anakinra) but also specifically with IL‐18BP. Systemic juvenile idiopathic arthritis or adult‐onset Still's disease are also characterized by high serum IL‐18 concentrations and are treated by IL‐18BP.
Collapse
Affiliation(s)
- Gilles Kaplanski
- Assistance Publique-Hôpitaux de Marseille, Centre Hospitalier Universitaire Conception, Service de Médecine Interne et Immunologie Clinique, Aix-Marseille Université, Marseille, France.,Vascular Research Center Marseille, Faculté de Pharmacie, Aix-Marseille Université, INSERM UMR_S1076, Marseille, France
| |
Collapse
|
38
|
Kim DH, Park HJ, Lim S, Koo JH, Lee HG, Choi JO, Oh JH, Ha SJ, Kang MJ, Lee CM, Lee CG, Elias JA, Choi JM. Regulation of chitinase-3-like-1 in T cell elicits Th1 and cytotoxic responses to inhibit lung metastasis. Nat Commun 2018; 9:503. [PMID: 29403003 PMCID: PMC5799380 DOI: 10.1038/s41467-017-02731-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 12/20/2017] [Indexed: 01/30/2023] Open
Abstract
Chitinase-3-like-1 (Chi3l1) is known to play a significant role in the pathogenesis of Type 2 inflammation and cancer. However, the function of Chi3l1 in T cell and its clinical implications are largely unknown. Here we show that Chi3l1 expression was increased in activated T cells, especially in Th2 cells. In addition, Chi3l1-deficient T cells are hyper-responsive to TcR stimulation and are prone to differentiating into Th1 cells. Chi3l1-deficient Th1 cells show increased expression of anti-tumor immunity genes and decreased Th1 negative regulators. Deletion of Chi3l1 in T cells in mice show reduced melanoma lung metastasis with increased IFNγ and TNFα-producing T cells in the lung. Furthermore, silencing of Chi3l1 expression in the lung using peptide-siRNA complex (dNP2-siChi3l1) efficiently inhibit lung metastasis with enhanced Th1 and CTL responses. Collectively, this study demonstrates Chi3l1 is a regulator of Th1 and CTL which could be a therapeutic target to enhance anti-tumor immunity.
Collapse
Affiliation(s)
- Do-Hyun Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Hong-Jai Park
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Sangho Lim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Ja-Hyun Koo
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Hong-Gyun Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Jin Ouk Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Ji Hoon Oh
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Min-Jong Kang
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Chang-Min Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, 04763, Korea
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
- Division of Medical and Biological Sciences, Warren Alpert Medical School, Brown University, Providence, RI, 02903, USA
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea.
- Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea.
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon, 16419, Korea.
| |
Collapse
|
39
|
Cordero MD, Alcocer-Gómez E. Inflammasome in the Pathogenesis of Pulmonary Diseases. EXPERIENTIA SUPPLEMENTUM (2012) 2018; 108:111-151. [PMID: 30536170 PMCID: PMC7123416 DOI: 10.1007/978-3-319-89390-7_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Lung diseases are common and significant causes of illness and death around the world. Inflammasomes have emerged as an important regulator of lung diseases. The important role of IL-1 beta and IL-18 in the inflammatory response of many lung diseases has been elucidated. The cleavage to turn IL-1 beta and IL-18 from their precursors into the active forms is tightly regulated by inflammasomes. In this chapter, we structurally review current evidence of inflammasome-related components in the pathogenesis of acute and chronic lung diseases, focusing on the "inflammasome-caspase-1-IL-1 beta/IL-18" axis.
Collapse
Affiliation(s)
- Mario D. Cordero
- Department of Physiology, Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center (CIBM), University of Granada, Armilla, Spain
| | - Elísabet Alcocer-Gómez
- Departamento de Psicología Experimental, Facultad de Psicología, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
40
|
Mussarat A, Manohar M, Verma AK, Upparahalli Venkateshaiah S, Zaidi A, Sanders NL, Zhu X, Mishra A. Intestinal overexpression of interleukin (IL)-15 promotes tissue eosinophilia and goblet cell hyperplasia. Immunol Cell Biol 2017; 96:273-283. [PMID: 29363170 DOI: 10.1111/imcb.1036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/21/2017] [Accepted: 11/21/2017] [Indexed: 12/30/2022]
Abstract
Interleukin (IL)-15 overexpression in eosinophilic gastrointestinal disorders is reported, but IL-15's role in promoting eosinophilic gastroenteritis is largely unknown. Therefore, we generated enterocyte-overexpressed IL-15 transgenic mice using Fabpi promoter. The Fabpi-IL-15 (iIL-15) transgenic mice showed induced IL-15 levels in the jejunum with a marked increase in jejunum eosinophils. However, no induction of eosinophilia in the blood or any other gastrointestinal segment was observed. Eosinophilia in the jejunum villus was substantially higher in iIL-15 mice compared to wild-type mice. In addition, goblet cell hyperplasia was also observed in the jejunum of iIL-15 mice. Furthermore, a significant correlation between induced IL-15 transcript and the IL-18 transcripts was observed. Therefore, to further understand the role of IL-18 in IL-15 mice associated gastrointestinal disorders, we generated iIL-15/IL-18Rα-/- mice. Using these mice, we found that IL-18 has an important role in promoting IL-15-induced eosinophilia. As intestinal IL-15 overexpression is reported in food intolerance, we examined OVA intolerance in iIL-15 mice. The OVA-sensitized and challenged iIL-15 mice experienced weight loss, diarrhea and eosinophilia in the jejunum. Taken together, our findings demonstrate that intestinal IL-15 overexpression induces IL-18-dependent eosinophilia and immunoglobulins in the intestine that promotes food allergic responses.
Collapse
Affiliation(s)
- Ahad Mussarat
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorder Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Murli Manohar
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorder Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Alok K Verma
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorder Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Sathisha Upparahalli Venkateshaiah
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorder Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Asifa Zaidi
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorder Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Nathan L Sanders
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorder Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Xiang Zhu
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorder Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Anil Mishra
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorder Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| |
Collapse
|
41
|
Colarusso C, Terlizzi M, Molino A, Pinto A, Sorrentino R. Role of the inflammasome in chronic obstructive pulmonary disease (COPD). Oncotarget 2017; 8:81813-81824. [PMID: 29137224 PMCID: PMC5669850 DOI: 10.18632/oncotarget.17850] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/19/2017] [Indexed: 01/05/2023] Open
Abstract
Inflammation is central to the development of chronic obstructive pulmonary disease (COPD), a pulmonary disorder characterized by chronic bronchitis, chronic airway obstruction, emphysema, associated to progressive and irreversible decline of lung function. Emerging genetic and pharmacological evidence suggests that IL-1-like cytokines are highly detected in the sputum and broncho-alveolar lavage (BAL) of COPD patients, implying the involvement of the multiprotein complex inflammasome. So far, scientific evidence has focused on nucleotide-binding oligomerization domain-like receptors protein 3 (NLRP3) inflammasome, a specialized inflammatory signaling platform that governs the maturation and secretion of IL-1-like cytokines through the regulation of caspase-1-dependent proteolytic processing. Some studies revealed that it is involved during airway inflammation typical of COPD. Based on the influence of cigarette smoke in various respiratory diseases, including COPD, in this view we report its effects in inflammatory and immune responses in COPD mouse models and in human subjects affected by COPD. In sharp contrast to what reported on experimental and clinical studies, randomized clinical trials show that indirect inflammasome inhibitors did not have any beneficial effect in moderate to severe COPD patients.
Collapse
Affiliation(s)
- Chiara Colarusso
- Department of Pharmacy, University of Salerno, ImmunePharma s.r.l., Fisciano, Salerno, Italy
| | - Michela Terlizzi
- Department of Pharmacy, University of Salerno, ImmunePharma s.r.l., Fisciano, Salerno, Italy
| | - Antonio Molino
- Department of Medicine and Surgery, Respiratory Division, University of Naples “Federico II”, Naples, Italy
| | - Aldo Pinto
- Department of Pharmacy, University of Salerno, ImmunePharma s.r.l., Fisciano, Salerno, Italy
| | - Rosalinda Sorrentino
- Department of Pharmacy, University of Salerno, ImmunePharma s.r.l., Fisciano, Salerno, Italy
| |
Collapse
|
42
|
Roos AB, Stampfli MR. Targeting Interleukin-17 signalling in cigarette smoke-induced lung disease: Mechanistic concepts and therapeutic opportunities. Pharmacol Ther 2017; 178:123-131. [PMID: 28438639 DOI: 10.1016/j.pharmthera.2017.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
It is widely accepted that compromised lung function in chronic obstructive pulmonary disease (COPD) is, at least in part, a consequence of persistent airway inflammation caused by particles and noxious gases present in cigarette smoke and indoor air pollution from burning biomass fuel. Currently, the World Health Organization estimates that 80 million people have moderate or severe COPD worldwide. While there is a global need for effective medical treatment, current therapeutic interventions have shown limited success in preventing disease pathology and progression. This is, in large part, due to the complexity and heterogeneity of COPD, and an incomplete understanding of the molecular mechanisms governing inflammatory processes in individual patients. This review discusses recent discoveries related to the pro-inflammatory cytokine interleukin (IL)-17A, and its potential role in the pathogenesis of COPD. We propose that an intervention strategy targeting IL-17 signalling offers an exciting opportunity to mitigate inflammatory processes, and prevent the progression of tissue pathologies associated with COPD.
Collapse
Affiliation(s)
- Abraham B Roos
- Respiratory, Inflammation and Autoimmunity, Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden and
| | - Martin R Stampfli
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada; Department of Medicine, Firestone Institute of Respiratory Health at St. Joseph's Health Care, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
43
|
Deficiency of Psgl-1 accelerates bleomycin (BLM)-induced lung fibrosis and inflammation in mice through activating PI3K/AKT. Biochem Biophys Res Commun 2017; 491:558-565. [DOI: 10.1016/j.bbrc.2017.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 03/01/2017] [Indexed: 12/16/2022]
|
44
|
Gurczynski SJ, Moore BB. IL-17 in the lung: the good, the bad, and the ugly. Am J Physiol Lung Cell Mol Physiol 2017; 314:L6-L16. [PMID: 28860146 DOI: 10.1152/ajplung.00344.2017] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The IL-17 family of cytokines has emerged over the last two decades as a pleiotropic group of molecules that function in a wide variety of both beneficial and detrimental (pathological) processes, mainly in mucosal barrier tissue. The beneficial effects of IL-17 expression are especially important in the lung, where exposure to foreign agents is abundant. IL-17A plays an important role in protection from both extracellular bacteria and fungi, as well as viruses that infect cells of the mucosal tracts. IL-17 coregulated cytokines, such as IL-22, are involved in maintaining epithelial cell homeostasis and participate in epithelial cell repair/regeneration following inflammatory insults. Thus, the IL-17/IL-22 axis is important in both responding to, and recovering from, pathogens. However, aberrant expression or overexpression of IL-17 cytokines contributes to a number of pathological outcomes, including asthma, pneumonitis, and generation or exacerbation of pulmonary fibrosis. This review covers the good, bad, and ugly aspects of IL-17 in the lung.
Collapse
Affiliation(s)
- Stephen J Gurczynski
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan , Ann Arbor, Michigan
| | - Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan , Ann Arbor, Michigan.,Department of Microbiology and Immunology, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
45
|
Briend E, Ferguson GJ, Mori M, Damera G, Stephenson K, Karp NA, Sethi S, Ward CK, Sleeman MA, Erjefält JS, Finch DK. IL-18 associated with lung lymphoid aggregates drives IFNγ production in severe COPD. Respir Res 2017; 18:159. [PMID: 28830544 PMCID: PMC5568255 DOI: 10.1186/s12931-017-0641-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 08/10/2017] [Indexed: 11/16/2022] Open
Abstract
Background Increased interferon gamma (IFNγ) release occurs in Chronic Obstructive Pulmonary Disease (COPD) lungs. IFNγ supports optimal viral clearance, but if dysregulated could increase lung tissue destruction. Methods The present study investigates which mediators most closely correlate with IFNγ in sputum in stable and exacerbating disease, and seeks to shed light on the spatial requirements for innate production of IFNγ, as reported in mouse lymph nodes, to observe whether such microenvironmental cellular organisation is relevant to IFNγ production in COPD lung. Results We show tertiary follicle formation in severe disease alters the dominant mechanistic drivers of IFNγ production, because cells producing interleukin-18, a key regulator of IFNγ, are highly associated with such structures. Interleukin-1 family cytokines correlated with IFNγ in COPD sputum. We observed that the primary source of IL-18 in COPD lungs was myeloid cells within lymphoid aggregates and IL-18 was increased in severe disease. IL-18 released from infected epithelium or from activated myeloid cells, was more dominant in driving IFNγ when IL-18-producing and responder cells were in close proximity. Conclusions Unlike tight regulation to control infection spread in lymphoid organs, this local interface between IL-18-expressing and responder cell is increasingly supported in lung as disease progresses, increasing its potential to increase tissue damage via IFNγ. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0641-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emmanuel Briend
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.,Present address: Agenus Ltd, Cambridge, UK
| | | | - Michiko Mori
- Department of Experimental Medical Science, BMC D12, Lund University, SE-221 84, Lund, Sweden
| | - Gautam Damera
- MedImmune LLC, 1 MedImmune Way, Gaithersburg, MD, USA
| | - Katherine Stephenson
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.,Present address: University of Nottingham, Nottingham, UK
| | - Natasha A Karp
- Quantitative Biology IMED, AstraZeneca R&D, Cambridge, UK
| | - Sanjay Sethi
- Department of Medicine, University at Buffalo, 3495 Bailey Avenue, Buffalo, NY, 14215, USA
| | - Christine K Ward
- MedImmune LLC, 1 MedImmune Way, Gaithersburg, MD, USA.,Present address: Bristol-Myers Squibb, Princeton, NJ, USA
| | - Matthew A Sleeman
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.,Present address: Regeneron Pharmaceuticals Inc, Tarrytown, NY, USA
| | - Jonas S Erjefält
- Department of Experimental Medical Science, BMC D12, Lund University, SE-221 84, Lund, Sweden.,Department of Respiratory Medicine and Allergology, Lund University Hospital, Lund, Sweden
| | - Donna K Finch
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.
| |
Collapse
|
46
|
Abstract
Objectives: To characterize overlap syndrome, investigate its impact on airflow limitation and blood oxygen condition, and detect the risk factors that affect its airway resistance. Methods: We reviewed retrospectively the clinical data of 158 patients with overlap syndrome (OS), chronic obstructive pulmonary disease (COPD), and obstructive sleep apnea (OSA), treated in the Critical Care Medicine Department of the People’s Hospital of Liaocheng, Liaocheng, China from May 2014 to March 2015. The lowest and average oxyhemoglobin saturation were measured using polysomnography. The pulmonary functions were tested using the cardiopulmonary measuring instruments, and the viscous resistance at oscillation frequencies of 5, 10, 15, 20 Hz was measured using the impulse oscillation system for all the patients. Results: The values of forced expiratory volume (FEV)1/FVC, FEV1% predicted, and the lowest SaO2 in the OS group were significantly lower than those in the OSA (p<0.01, p<0.01, p=0.01), or the COPD group (p=0.03, p=0.02, p=0.03), but the value of viscous resistance at 5 Hz was significantly higher than that in the 2 groups (p<0.01). Old age, body mass index, and smoking history were significantly correlated with the viscous resistance in OS patients, at an oscillation frequency of 5Hz (p=0.03, p=0.04). Conclusion: The OS patients present with higher viscous resistance and more severe oxygen deficit, when compared with OSA and COPD patients, and weight decrease and smoking cessation are necessary for these patients.
Collapse
Affiliation(s)
- He Huang
- Department of Critical Care Medicine, People's Hospital of Liaocheng, Liaocheng City, Shandong Province, China. E-mail.
| | | |
Collapse
|
47
|
Lakshmi SP, Reddy AT, Reddy RC. Emerging pharmaceutical therapies for COPD. Int J Chron Obstruct Pulmon Dis 2017; 12:2141-2156. [PMID: 28790817 PMCID: PMC5531723 DOI: 10.2147/copd.s121416] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
COPD, for which cigarette smoking is the major risk factor, remains a worldwide burden. Current therapies provide only limited short-term benefit and fail to halt progression. A variety of potential therapeutic targets are currently being investigated, including COPD-related proinflammatory mediators and signaling pathways. Other investigational compounds target specific aspects or complications of COPD such as mucus hypersecretion and pulmonary hypertension. Although many candidate therapies have shown no significant effects, other emerging therapies have improved lung function, pulmonary hypertension, glucocorticoid sensitivity, and/or the frequency of exacerbations. Among these are compounds that inhibit the CXCR2 receptor, mitogen-activated protein kinase/Src kinase, myristoylated alanine-rich C kinase substrate, selectins, and the endothelin receptor. Activation of certain transcription factors may also be relevant, as a large retrospective cohort study of COPD patients with diabetes found that the peroxisome proliferator-activated receptor γ (PPARγ) agonists rosiglitazone and pioglitazone were associated with reduced COPD exacerbation rate. Notably, several therapies have shown efficacy only in identifiable subgroups of COPD patients, suggesting that subgroup identification may become more important in future treatment strategies. This review summarizes the status of emerging therapeutic pharmaceuticals for COPD and highlights those that appear most promising.
Collapse
Affiliation(s)
- Sowmya P Lakshmi
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine.,Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Aravind T Reddy
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine.,Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Raju C Reddy
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine.,Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| |
Collapse
|
48
|
Zheng Z, Qi Y, Xu X, Jiang H, Li Z, Yang Q, Zhang C, Zhang K, Chen R, Wang J, Lu W. Sputum mucin 1 is increased during the acute phase of chronic obstructive pulmonary disease exacerbation. J Thorac Dis 2017; 9:1873-1882. [PMID: 28839985 DOI: 10.21037/jtd.2017.06.63] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Mucin 1 (MUC1) is a membrane tethered protein on airway epithelial cells. This protein is upregulated and plays an important anti-inflammatory role during acute lung inflammation. However, the relationship between sputum MUC1 level and acute exacerbation of chronic obstructive pulmonary disease (AECOPD) is unknown. METHODS The levels of MUC1, IL-8, and TNF-α in induced sputum from 78 COPD patients were assessed by ELISA. The association between COPD exacerbation and MUC1 fragment levels was analyzed. An acute airway inflammation mouse model was established by intranasal LPS inhalation. The expression of Muc1 in lung and the levels of Muc1, TNF-α and KC in BAL fluid from mice were determined with western blotting and ELISA, respectively. RESULTS Higher levels of MUC1 membrane-tethered (CT) and extracellular (EC) fragments, cytokines TNF-α and IL-8, more leucocyte and neutrophil counts were found in sputum from COPD patients in acute than in remission phase. Linear regression analysis confirmed that the level of sputum MUC1 CT fragment is positively correlated with sputum neutrophil number and patients' age; whereas the sputum EC fragment level is correlated inversely with FEV1/FVC value and positively with patients' age. Inhalation of lipopolysaccharide (LPS) induced acute lung inflammation in mice which exhibited increased levels of Muc1 CT fragment in lung and only Muc1 EC fragment increase in BAL fluid. CONCLUSIONS Unlike pure bacterial induced lung inflammation, both sputum MUC1 CT and EC fragments are increased during acute exacerbation of COPD. The clinical benefits from measuring the changes of various sputum MUC1 fragments in AECOPD need to be elucidated in future studies.
Collapse
Affiliation(s)
- Zeguang Zheng
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China
| | - Yafei Qi
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China.,Central Hospital of Panyu District, Guangzhou 511470, China
| | - Xiaoming Xu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China
| | - Hua Jiang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China
| | - Zhengtu Li
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China
| | - Quan Yang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China
| | - Chenting Zhang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China
| | - Kedong Zhang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China
| | - Rongchang Chen
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China.,Department of Laboratory Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China
| |
Collapse
|
49
|
Zarei S, Mirtar A, Morrow JD, Castaldi PJ, Belloni P, Hersh CP. Subtyping Chronic Obstructive Pulmonary Disease Using Peripheral Blood Proteomics. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2017; 4:97-108. [PMID: 28848918 DOI: 10.15326/jcopdf.4.2.2016.0147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous disorder. COPD patients may have different clinical features, imaging characteristics and natural history. Multiple studies have investigated heterogeneity using statistical methods such as unsupervised clustering to define different subgroups of COPD based largely on clinical phenotypes. Some studies have performed clustering using genetic data or limited numbers of blood biomarkers. Our primary goal was to use proteomic data to find subtypes of COPD within clinically similar individuals. In the Treatment of Emphysema with a gamma-Selective Retinoid Agonist (TESRA) study, multiplex biomarker panels were run in serum samples collected prior to randomization. After implementing an algorithm to minimize missing values, the dataset included 396 COPD individuals and 87 biomarkers. Using hierarchical clustering, we identified 3 COPD subgroups, containing 267 (67.4%), 104 (26.3%), and 25 (6.3%) individuals, respectively. The third cluster had less emphysema on quantitative analysis of chest computed tomography scans (p=0.03) and worse disease-related quality of life based on the St. George's Respiratory Questionnaire (total score cluster 1: 45.6, cluster 2: 45.4, cluster 3: 56.6; p=0.01), despite similar levels of lung function impairment (forced expiratory volume in 1 second (49.2%, 49.2%, 48.2 % predicted, respectively). Enrichment analysis showed the biomarkers distinguishing cluster 3 mapped to platelet alpha granule and cell chemotaxis pathways. Thus, we identified a subgroup which has less emphysema but may have greater inflammation, which could be potentially targeted with anti-inflammatory therapies.
Collapse
Affiliation(s)
- Sara Zarei
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.,San Juan Bautista School of Medicine, Caguas, Puerto Rico
| | | | - Jarrett D Morrow
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
50
|
Han S, Jerome JA, Gregory AD, Mallampalli RK. Cigarette smoke destabilizes NLRP3 protein by promoting its ubiquitination. Respir Res 2017; 18:2. [PMID: 28056996 PMCID: PMC5217194 DOI: 10.1186/s12931-016-0485-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/08/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Cigarette smoke suppresses innate immunity, making smokers more susceptible to infection. The NLRP3 inflammasome is a multi-protein complex that releases interleukin (IL) -1β and IL -18. These cytokines are critical for a timely host response to pathogens. Whether cigarette smoke affects NLRP3 protein levels, and its ability to form an inflammasome, is not known. METHODS AND RESULTS Using the human monocyte THP1 cell line and C57BL/6 mice, we show that cigarette smoke decreases NLRP3 levels in cells by increasing ubiquitin-mediated proteasomal processing. Half-life of NLRP3 is shortened with the exposure to cigarette smoke extract. Cigarette smoke extract reduces cellular NLRP3 protein abundance in the presence of lipopolysaccharide, a known inducer of NLRP3 protein, thereby decreasing the formation of NLRP3 inflammasomes. The release of IL-1β and IL-18 by inflammasome activation is also decreased with the exposure to cigarette smoke extract both in THP1 cells and primary human peripheral blood macrophages. CONCLUSIONS Cigarette smoke extract decreased NLRP3 protein abundance via increased ubiquitin-mediated proteasomal processing. The release of IL-1β and IL-18 is also decreased with cigarette smoke extract. Our findings may provide mechanistic insights on immunosuppression in smokers and unique opportunities to develop a strategy to modulate immune function.
Collapse
Affiliation(s)
- SeungHye Han
- Department of Medicine, The Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jacob A Jerome
- Department of Medicine, The Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alyssa D Gregory
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, 15213, Pittsburgh, PA, USA
| | - Rama K Mallampalli
- Department of Medicine, The Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, USA. .,Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, 15213, Pittsburgh, PA, USA. .,Medical Specialty Service Line, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|