1
|
Xiao X, Ding Z, Shi Y, Zhang Q. Causal Role of Immune Cells in Chronic Obstructive Pulmonary Disease: A Two-Sample Mendelian Randomization Study. COPD 2024; 21:2327352. [PMID: 38573027 DOI: 10.1080/15412555.2024.2327352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/27/2024] [Indexed: 04/05/2024]
Abstract
Accumulating evidence has highlighted the importance of immune cells in the pathogenesis of chronic obstructive pulmonary disease (COPD). However, the understanding of the causal association between immunity and COPD remains incomplete due to the existence of confounding variables. In this study, we employed a two-sample Mendelian randomization (MR) analysis, utilizing the genome-wide association study database, to investigate the causal association between 731 immune-cell signatures and the susceptibility to COPD from a host genetics perspective. To validate the consistency of our findings, we utilized MR analysis results of lung function data to assess directional concordance. Furthermore, we employed MR-Egger intercept tests, Cochrane's Q test, MR-PRESSO global test, and "leave-one-out" sensitivity analyses to evaluate the presence of horizontal pleiotropy, heterogeneity, and stability, respectively. Inverse variance weighting results showed that seven immune phenotypes were associated with the risk of COPD. Analyses of heterogeneity and pleiotropy analysis confirmed the reliability of MR results. These results highlight the interactions between the immune system and the lungs. Further investigations into their mechanisms are necessary and will contribute to inform targeted prevention strategies for COPD.
Collapse
Affiliation(s)
- Xinru Xiao
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Ziqi Ding
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yujia Shi
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Qian Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| |
Collapse
|
2
|
Nemati H, de Graaf J. The cellular Potts model on disordered lattices. SOFT MATTER 2024; 20:8337-8352. [PMID: 39283268 PMCID: PMC11404401 DOI: 10.1039/d4sm00445k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/28/2024] [Indexed: 09/20/2024]
Abstract
The cellular Potts model, also known as the Glazier-Graner-Hogeweg model, is a lattice-based approach by which biological tissues at the level of individual cells can be numerically studied. Traditionally, a square or hexagonal underlying lattice structure is assumed for two-dimensional systems, and this is known to introduce artifacts in the structure and dynamics of the model tissues. That is, on regular lattices, cells can assume shapes that are dictated by the symmetries of the underlying lattice. Here, we developed a variant of this method that can be applied to a broad class of (ir)regular lattices. We show that on an irregular lattice deriving from a fluid-like configuration, two types of artifacts can be removed. We further report on the transition between a fluid-like disordered and a solid-like hexagonally ordered phase present for monodisperse confluent cells as a function of their surface tension. This transition shows the hallmarks of a first-order phase transition and is different from the glass/jamming transitions commonly reported for the vertex and active Voronoi models. We emphasize this by analyzing the distribution of shape parameters found in our state space. Our analysis provides a useful reference for the future study of epithelia using the (ir)regular cellular Potts model.
Collapse
Affiliation(s)
- Hossein Nemati
- Institute for Theoretical Physics, Center for Extreme Matter and Emergent Phenomena, Utrecht University, Princetonplein 5, 3584 CC Utrecht, The Netherlands.
| | - J de Graaf
- Institute for Theoretical Physics, Center for Extreme Matter and Emergent Phenomena, Utrecht University, Princetonplein 5, 3584 CC Utrecht, The Netherlands.
| |
Collapse
|
3
|
Zhang Y, Wu K, Mao D, Iberg CA, Yin-Declue H, Sun K, Wikfors HA, Keeler SP, Li M, Young D, Yantis J, Crouch EC, Chartock JR, Han Z, Byers DE, Brody SL, Romero AG, Holtzman MJ. A first-in-kind MAPK13 inhibitor that can correct stem cell reprogramming and post-injury disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.21.608990. [PMID: 39229202 PMCID: PMC11370402 DOI: 10.1101/2024.08.21.608990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The stress kinase MAPK13 (aka p38δ-MAPK) is an attractive entry point for therapeutic intervention because it regulates the structural remodeling that can develop after epithelial barrier injury in the lung and likely other tissue sites. However, a selective, safe, and effective MAPK13 inhibitor is not yet available for experimental or clinical application. Here we identify a first-in-kind MAPK13 inhibitor using structure-based drug design combined with a screening funnel for cell safety and molecular specificity. This inhibitor (designated NuP-4) down-regulates basal-epithelial stem cell reprogramming, structural remodeling, and pathophysiology equivalently to Mapk13 gene-knockout in mouse and mouse organoid models of post-viral lung disease. This therapeutic benefit persists after stopping treatment as a sign of disease modification and attenuates key aspects of inflammation and remodeling as an indication of disease reversal. Similarly, NuP-4 treatment can directly control cytokine-stimulated growth, immune activation, and mucinous differentiation in human basal-cell organoids. The data thereby provide a new tool and potential fix for long-term stem cell reprogramming after viral injury and related conditions that require MAPK13 induction-activation.
Collapse
Affiliation(s)
- Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Dailing Mao
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Courtney A Iberg
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Huiqing Yin-Declue
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kelly Sun
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Hallie A Wikfors
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Shamus P Keeler
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Ming Li
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Deanna Young
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Jennifer Yantis
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Erika C Crouch
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Joshua R Chartock
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Zhenfu Han
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Derek E Byers
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Steven L Brody
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Arthur G Romero
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
- NuPeak Therapeutics Inc., St. Louis, MO 63105
| |
Collapse
|
4
|
Wu K, Zhang Y, Mao D, Iberg CA, Yin-Declue H, Sun K, Keeler SP, Wikfors HA, Young D, Yantis J, Austin SR, Byers DE, Brody SL, Crouch EC, Romero AG, Holtzman MJ. MAPK13 controls structural remodeling and disease after epithelial injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596863. [PMID: 38895360 PMCID: PMC11185504 DOI: 10.1101/2024.05.31.596863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
All living organisms are charged with repair after injury particularly at epithelial barrier sites, but in some cases this response leads instead to structural remodeling and long-term disease. Identifying the molecular and cellular control of this divergence is key to disease modification. In that regard, stress kinase control of epithelial stem cells is a rational entry point for study. Here we examine the potential for mitogen-activated protein kinase 13 (MAPK13) regulation of epithelial stem cells using models of respiratory viral injury and post-viral lung disease. We show that Mapk13 gene-knockout mice handle acute infectious illness as expected but are protected against structural remodeling manifest as basal-epithelial stem cell (basal-ESC) hyperplasia-metaplasia, immune activation, and mucinous differentiation. In corresponding cell models, Mapk13-deficiency directly attenuates basal-ESC growth and organoid formation. Extension to human studies shows marked induction/activation of basal-cell MAPK13 in clinical samples of comparable remodeling found in asthma and COPD. Here again, MAPK13 gene-knockdown inhibits human basal-ESC growth in culture. Together, the data identify MAPK13 as a control for structural remodeling and disease after epithelial injury and as a suitable target for down-regulation as a disease-modifying strategy.
Collapse
Affiliation(s)
- Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Dailing Mao
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Courtney A. Iberg
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Huiqing Yin-Declue
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kelly Sun
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Shamus P. Keeler
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Hallie A. Wikfors
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Deanna Young
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Jennifer Yantis
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Stephen R. Austin
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Derek E Byers
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Steven L. Brody
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Erika C. Crouch
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Arthur G. Romero
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J. Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
- NuPeak Therapeutics Inc., St. Louis, MO 63105
| |
Collapse
|
5
|
Wu K, Zhang Y, Yin-DeClue H, Sun K, Mao D, Yang K, Austin SR, Crouch EC, Brody SL, Byers DE, Hoffmann CM, Hughes ME, Holtzman MJ. A correctable immune niche for epithelial stem cell reprogramming and post-viral lung diseases. J Clin Invest 2024; 134:e183092. [PMID: 39052353 PMCID: PMC11405052 DOI: 10.1172/jci183092] [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: 05/16/2024] [Accepted: 07/23/2024] [Indexed: 07/27/2024] Open
Abstract
Epithelial barriers are programmed for defense and repair but are also the site of long-term structural remodeling and disease. In general, this paradigm features epithelial stem cells (ESCs) that are called on to regenerate damaged tissues but can also be reprogrammed for detrimental remodeling. Here we identified a Wfdc21-dependent monocyte-derived dendritic cell (moDC) population that functioned as an early sentinel niche for basal ESC reprogramming in mouse models of epithelial injury after respiratory viral infection. Niche function depended on moDC delivery of ligand GPNMB to the basal ESC receptor CD44 so that properly timed antibody blockade of ligand or receptor provided long-lasting correction of reprogramming and broad disease phenotypes. These same control points worked directly in mouse and human basal ESC organoids. Together, the findings identify a mechanism to explain and modify what is otherwise a stereotyped but sometimes detrimental response to epithelial injury.
Collapse
Affiliation(s)
- Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine
| | | | - Kelly Sun
- Pulmonary and Critical Care Medicine, Department of Medicine
| | - Dailing Mao
- Pulmonary and Critical Care Medicine, Department of Medicine
| | - Kuangying Yang
- Pulmonary and Critical Care Medicine, Department of Medicine
| | | | | | - Steven L Brody
- Pulmonary and Critical Care Medicine, Department of Medicine
| | - Derek E Byers
- Pulmonary and Critical Care Medicine, Department of Medicine
| | | | - Michael E Hughes
- Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Genetics, and
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
6
|
Santus P, Signorello JC, Danzo F, Lazzaroni G, Saad M, Radovanovic D. Anti-Inflammatory and Anti-Oxidant Properties of N-Acetylcysteine: A Fresh Perspective. J Clin Med 2024; 13:4127. [PMID: 39064168 PMCID: PMC11278452 DOI: 10.3390/jcm13144127] [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: 05/30/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
N-acetyl-L-cysteine (NAC) was initially introduced as a treatment for mucus reduction and widely used for chronic respiratory conditions associated with mucus overproduction. However, the mechanism of action for NAC extends beyond its mucolytic activity and is complex and multifaceted. Contrary to other mucoactive drugs, NAC has been found to exhibit antioxidant, anti-infective, and anti-inflammatory activity in pre-clinical and clinical reports. These properties have sparked interest in its potential for treating chronic lung diseases, including chronic obstructive pulmonary disease (COPD), bronchiectasis (BE), cystic fibrosis (CF), and idiopathic pulmonary fibrosis (IPF), which are associated with oxidative stress, increased levels of glutathione and inflammation. NAC's anti-inflammatory activity is noteworthy, and it is not solely secondary to its antioxidant capabilities. In ex vivo models of COPD exacerbation, the anti-inflammatory effects have been observed even at very low doses, especially with prolonged treatment. The mechanism involves the inhibition of the activation of NF-kB and neurokinin A production, resulting in a reduction in interleukin-6 production, a cytokine abundantly present in the sputum and breath condensate of patients with COPD and correlates with the number of exacerbations. The unique combination of mucolytic, antioxidant, anti-infective, and anti-inflammatory properties positions NAC as a safe, cost-effective, and efficacious therapy for a plethora of respiratory conditions.
Collapse
Affiliation(s)
- Pierachille Santus
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
| | - Juan Camilo Signorello
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
| | - Fiammetta Danzo
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
| | - Giada Lazzaroni
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
| | - Marina Saad
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20122 Milano, Italy;
| | - Dejan Radovanovic
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20122 Milano, Italy;
| |
Collapse
|
7
|
Davila KMS, Nelli RK, Mora-Díaz JC, Sang Y, Miller LC, Giménez-Lirola LG. Transcriptome Analysis in Air-Liquid Interface Porcine Respiratory Epithelial Cell Cultures Reveals That the Betacoronavirus Porcine Encephalomyelitis Hemagglutinating Virus Induces a Robust Interferon Response to Infection. Viruses 2024; 16:939. [PMID: 38932231 PMCID: PMC11209522 DOI: 10.3390/v16060939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Porcine hemagglutinating encephalomyelitis virus (PHEV) replicates in the upper respiratory tract and tonsils of pigs. Using an air-liquid interface porcine respiratory epithelial cells (ALI-PRECs) culture system, we demonstrated that PHEV disrupts respiratory epithelia homeostasis by impairing ciliary function and inducing antiviral, pro-inflammatory cytokine, and chemokine responses. This study explores the mechanisms driving early innate immune responses during PHEV infection through host transcriptome analysis. Total RNA was collected from ALI-PRECs at 24, 36, and 48 h post inoculation (hpi). RNA-seq analysis was performed using an Illumina Hiseq 600 to generate 100 bp paired-end reads. Differential gene expression was analyzed using DeSeq2. PHEV replicated actively in ALI-PRECs, causing cytopathic changes and progressive mucociliary disruption. Transcriptome analysis revealed downregulation of cilia-associated genes such as CILK1, DNAH11, LRRC-23, -49, and -51, and acidic sialomucin CD164L2. PHEV also activated antiviral signaling pathways, significantly increasing the expression of interferon-stimulated genes (RSAD2, MX1, IFIT, and ISG15) and chemokine genes (CCL5 and CXCL10), highlighting inflammatory regulation. This study contributes to elucidating the molecular mechanisms of the innate immune response to PHEV infection of the airway epithelium, emphasizing the critical roles of the mucociliary, interferon, and chemokine responses.
Collapse
Affiliation(s)
- Kaitlyn M. Sarlo Davila
- Infectious Bacterial Disease Research Unit, National Animal Disease Center, United States Department of Agriculture, Agricultural Research Service, Ames, IA 50010, USA;
| | - Rahul K. Nelli
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (R.K.N.); (J.C.M.-D.)
| | - Juan C. Mora-Díaz
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (R.K.N.); (J.C.M.-D.)
| | - Yongming Sang
- Department of Agricultural and Environmental Sciences, College of Agriculture, Tennessee State University, Nashville, TN 37209, USA;
| | - Laura C. Miller
- Virus and Prion Research Unit, National Animal Disease Center, United States Department of Agriculture, Agricultural Research Service, Ames, IA 50010, USA
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Luis G. Giménez-Lirola
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (R.K.N.); (J.C.M.-D.)
| |
Collapse
|
8
|
Hu Y, Wang M, Xie J, Jiao L, Ding Y, Luo Y. Exposure to ephedrine attenuates Th1/Th2 imbalance underlying OVA-induced asthma through airway epithelial cell-derived exosomal lnc-TRPM2-AS. Chin J Nat Med 2024; 22:530-540. [PMID: 38906600 DOI: 10.1016/s1875-5364(24)60554-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Indexed: 06/23/2024]
Abstract
Although various anti-inflammatory medications, such as ephedrine, are employed to manage cough-variant asthma, their underlying mechanisms are yet to be fully understood. Recent studies suggest that exosomes derived from airway epithelial cells (AECs) contain components like messenger RNAs (mRNAs), micro-RNAs (miRNAs), and long noncoding RNA (lncRNA), which play roles in the occurrence and progression of airway inflammation. This study investigates the influence of AEC-derived exosomes on the efficacy of ephedrine in treating cough-variant asthma. We established a mouse model of asthma and measured airway resistance and serum inflammatory cell levels. Real-time polymerase chain reaction (RT-qPCR), Western blotting, and enzyme-linked immunosorbent assay (ELISA) analyses were used to assess gene and protein expression levels. Exosomes were isolated and characterized. RNA immunoprecipitation (RIP) and RNA pull-down assays were conducted to examine the interaction between hnRNPA2B1 and lnc-TRPM2-AS1. In the ovalbumin (OVA)-challenged mouse model, ephedrine treatment reduced inflammatory responses, airway resistance, and Th1/Th2 cell imbalance. Exosomes from OVA-treated AECs showed elevated levels of lnc-TRPM2-AS1, which were diminished following ephedrine treatment. The exosomal lnc-TRPM2-AS1 mediated the Th1/Th2 imbalance in CD4+ T cells, with its packaging into exosomes being facilitated by hnRNPA2B1. This study unveils a novel mechanism by which ephedrine ameliorates OVA-induced CD4+ T cell imbalance by suppressing AEC-derived exosomal lnc-TRPM2-AS1. These findings could provide a theoretical framework for using ephedrine in asthma treatment.
Collapse
Affiliation(s)
- Yan Hu
- Department of Pediatrics, The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Mengqing Wang
- Department of Pediatrics, The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, China.
| | - Jing Xie
- Department of Pediatrics, The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Luojia Jiao
- Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yi Ding
- Department of Rehabilitation, Medical College, Changsha Civil Affairs Vocational and Technical College, Changsha 410004, Hunan Province, China
| | - Yinhe Luo
- Teaching and Research Office of Chinese and Western Combination, Hunan University of Chinese Medicine, Changsha 410208, China.
| |
Collapse
|
9
|
Raphael HE, Hassan GF, Osorio OA, Cohen LS, Payne MD, Katz-Kiriakos E, Tata I, Hicks J, Byers DE, Zhang B, Alexander-Brett J. Activator protein transcription factors coordinate human IL-33 expression from noncanonical promoters in chronic airway disease. JCI Insight 2024; 9:e174786. [PMID: 38456508 PMCID: PMC10972587 DOI: 10.1172/jci.insight.174786] [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: 08/14/2023] [Accepted: 01/22/2024] [Indexed: 03/09/2024] Open
Abstract
IL-33 is a cytokine central to type 2 immune pathology in chronic airway disease. This cytokine is abundantly expressed in the respiratory epithelium and increased in disease, but how expression is regulated is undefined. Here we show that increased IL33 expression occurs from multiple noncanonical promoters in human chronic obstructive pulmonary disease (COPD), and it facilitates production of alternatively spliced isoforms in airway cells. We found that phorbol 12-myristate 13-acetate (PMA) can activate IL33 promoters through protein kinase C in primary airway cells and lines. Transcription factor (TF) binding arrays combined with RNA interference identified activator protein (AP) TFs as regulators of baseline and induced IL33 promoter activity. ATAC-Seq and ChIP-PCR identified chromatin accessibility and differential TF binding as additional control points for transcription from noncanonical promoters. In support of a role for these TFs in COPD pathogenesis, we found that AP-2 (TFAP2A, TFAP2C) and AP-1 (FOS and JUN) family members are upregulated in human COPD specimens. This study implicates integrative and pioneer TFs in regulating IL33 promoters and alternative splicing in human airway basal cells. Our work reveals a potentially novel approach for targeting IL-33 in development of therapeutics for COPD.
Collapse
Affiliation(s)
- Heather E. Raphael
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
| | - Ghandi F. Hassan
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
| | - Omar A. Osorio
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
| | - Lucy S. Cohen
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
| | - Morgan D. Payne
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
| | - Ella Katz-Kiriakos
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
| | - Ishana Tata
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
| | - Jamie Hicks
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
| | - Derek E. Byers
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
| | - Bo Zhang
- Department of Developmental Biology, and
| | - Jen Alexander-Brett
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
10
|
Jantaruk P, Roytrakul S, Sistayanarain A, Kunthalert D. The pomegranate-derived peptide Pug-4 alleviates nontypeable Haemophilus influenzae-induced inflammation by suppressing NF-kB signaling and NLRP3 inflammasome activation. PeerJ 2024; 12:e16938. [PMID: 38406294 PMCID: PMC10885808 DOI: 10.7717/peerj.16938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 01/23/2024] [Indexed: 02/27/2024] Open
Abstract
The respiratory pathogen nontypeable Haemophilus influenzae (NTHi) is the most common cause of exacerbation of chronic obstructive pulmonary disease (COPD), of which an excessive inflammatory response is a hallmark. With the limited success of current medicines there is an urgent need for the development of novel therapeutics that are both safe and effective. In this study, we explored the regulatory potential of pomegranate-derived peptides Pug-1, Pug-2, Pug-3, and Pug-4 on NTHi-induced inflammation. Our results clearly showed that to varying degrees the Pug peptides inhibited NTHi-induced production of IL-1β, a pivotal cytokine in COPD, and showed that these effects were not related to cytotoxicity. Pug-4 peptide exhibited the most potent inhibitory activity. This was demonstrated in all studied cell types including murine (RAW264.7) and human (differentiated THP-1) macrophages as well as human lung epithelial cells (A549). Substantial reduction by Pug-4 of TNF-α, NO and PGE2 in NTHi-infected A549 cells was also observed. In addition, Pug-4 strongly inhibited the expression of nuclear-NF-κB p65 protein and the NF-κB target genes (determined by IL-1β, TNF-α, iNOS and COX-2 mRNA expression) in NTHi-infected A549 cells. Pug-4 suppressed the expression of NLRP3 and pro-IL-1β proteins and inhibited NTHi-mediated cleavage of caspase-1 and mature IL-1β. These results demonstrated that Pug-4 inhibited NTHi-induced inflammation through the NF-κB signaling and NLRP3 inflammasome activation. Our findings herein highlight the significant anti-inflammatory activity of Pug-4, a newly identified peptide from pomegranate, against NTHi-induced inflammation. We therefore strongly suggest the potential of the Pug-4 peptide as an anti-inflammatory medicine candidate for treatment of NTHi-mediated inflammation.
Collapse
Affiliation(s)
- Pornpimon Jantaruk
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Sittiruk Roytrakul
- National Science and Technology Development Agency, Thailand Science Park, National Center for Genetic Engineering and Biotechnology, Pathumthani, Thailand
| | - Anchalee Sistayanarain
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Duangkamol Kunthalert
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| |
Collapse
|
11
|
Brollo M, Salvator H, Grassin-Delyle S, Glorion M, Descamps D, Buenestado A, Naline E, Tenor H, Tiotiu A, Devillier P. The IL-4/13-induced production of M2 chemokines by human lung macrophages is enhanced by adenosine and PGE 2. Int Immunopharmacol 2024; 128:111557. [PMID: 38266451 DOI: 10.1016/j.intimp.2024.111557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND AND PURPOSE Lung macrophages (LMs) are critically involved in respiratory diseases. The primary objective of the present study was to determine whether or not an adenosine analog (NECA) and prostaglandin E2 (PGE2) affected the interleukin (IL)-4- and IL-13-induced release of M2a chemokines (CCL13, CCL17, CCL18, and CCL22) by human LMs. EXPERIMENTAL APPROACH Primary macrophages isolated from resected human lungs were incubated with NECA, PGE2, roflumilast, or vehicle and stimulated with IL-4 or IL-13 for 24 h. The levels of chemokines and PGE2 in the culture supernatants were measured using ELISAs and enzyme immunoassays. KEY RESULTS Exposure to IL-4 (10 ng/mL) and IL-13 (50 ng/mL) was associated with greater M2a chemokine production but not PGE2 production. PGE2 (10 ng/mL) and NECA (10-6 M) induced the production of M2a chemokines to a lesser extent but significantly enhanced the IL-4/IL-13-induced production of these chemokines. At either a clinically relevant concentration (10-9 M) or at a concentration (10-7 M) that fully inhibited phosphodiesterase 4 (PDE4) activity, roflumilast did not increase the production of M2a chemokines and did not modulate their IL-13-induced production, regardless of the presence or absence of PGE2. CONCLUSIONS NECA and PGE2 enhanced the IL-4/IL-13-induced production of M2a chemokines. The inhibition of PDE4 by roflumilast did not alter the production of these chemokines. These results contrast totally with the previously reported inhibitory effects of NECA, PGE2, and PDE4 inhibitors on the lipopolysaccharide-induced release of tumor necrosis factor alpha and M1 chemokines in human LMs.
Collapse
Affiliation(s)
- Marion Brollo
- Laboratory of Research in Respiratory Pharmacology, Faculté des Sciences de la Vie Simone Veil, VIM, UMR-0892, INRAE, UVSQ, Université Paris-Saclay, France
| | - Hélène Salvator
- Laboratory of Research in Respiratory Pharmacology, Faculté des Sciences de la Vie Simone Veil, VIM, UMR-0892, INRAE, UVSQ, Université Paris-Saclay, France; Department of Airway Diseases, Respiratory Pharmacology Unit, Foch Hospital, Suresnes, France
| | - Stanislas Grassin-Delyle
- Department of Airway Diseases, Respiratory Pharmacology Unit, Foch Hospital, Suresnes, France; Department of Airway Diseases, Thoracic surgery, Foch Hospital, Suresnes, France
| | - Mathieu Glorion
- Laboratory of Research in Respiratory Pharmacology, Faculté des Sciences de la Vie Simone Veil, VIM, UMR-0892, INRAE, UVSQ, Université Paris-Saclay, France; INSERM U1173, Infection & Inflammation, Département de Biotechnologie de la Santé, Université Paris-Saclay, Montigny-le-Bretonneux, France
| | - Delphyne Descamps
- VIM, UMR-0892, INRAE, UVSQ, Université Paris-Saclay, Jouy-en-Josas, France
| | - Amparo Buenestado
- Laboratory of Research in Respiratory Pharmacology, Faculté des Sciences de la Vie Simone Veil, VIM, UMR-0892, INRAE, UVSQ, Université Paris-Saclay, France
| | - Emmanuel Naline
- Laboratory of Research in Respiratory Pharmacology, Faculté des Sciences de la Vie Simone Veil, VIM, UMR-0892, INRAE, UVSQ, Université Paris-Saclay, France; Department of Airway Diseases, Respiratory Pharmacology Unit, Foch Hospital, Suresnes, France
| | | | - Angelica Tiotiu
- Department of Pulmonary Medicine, University Hospital Saint-Luc, Institut of Experimental and Clinical Research (IREC), University of Louvain, Brussels, Belgium
| | - Philippe Devillier
- Laboratory of Research in Respiratory Pharmacology, Faculté des Sciences de la Vie Simone Veil, VIM, UMR-0892, INRAE, UVSQ, Université Paris-Saclay, France; Department of Airway Diseases, Respiratory Pharmacology Unit, Foch Hospital, Suresnes, France.
| |
Collapse
|
12
|
Holtzman MJ, Zhang Y, Wu K, Romero AG. Mitogen-activated protein kinase-guided drug discovery for post-viral and related types of lung disease. Eur Respir Rev 2024; 33:230220. [PMID: 38417971 PMCID: PMC10900067 DOI: 10.1183/16000617.0220-2023] [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: 10/27/2023] [Accepted: 01/18/2024] [Indexed: 03/01/2024] Open
Abstract
Respiratory viral infections are a major public health problem, with much of their morbidity and mortality due to post-viral lung diseases that progress and persist after the active infection is cleared. This paradigm is implicated in the most common forms of chronic lung disease, such as asthma and COPD, as well as other virus-linked diseases including progressive and long-term coronavirus disease 2019. Despite the impact of these diseases, there is a lack of small-molecule drugs available that can precisely modify this type of disease process. Here we will review current progress in understanding the pathogenesis of post-viral and related lung disease with characteristic remodelling phenotypes. We will also develop how this data leads to mitogen-activated protein kinase (MAPK) in general and MAPK13 in particular as key druggable targets in this pathway. We will also explore recent advances and predict the future breakthroughs in structure-based drug design that will provide new MAPK inhibitors as drug candidates for clinical applications. Each of these developments point to a more effective approach to treating the distinct epithelial and immune cell based mechanisms, which better account for the morbidity and mortality of post-viral and related types of lung disease. This progress is vital given the growing prevalence of respiratory viruses and other inhaled agents that trigger stereotyped progression to acute illness and chronic disease.
Collapse
Affiliation(s)
- Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- NuPeak Therapeutics Inc., St. Louis, MO, USA
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Arthur G Romero
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
13
|
Nascimento M, Huot-Marchand S, Fanny M, Straube M, Le Bert M, Savigny F, Apetoh L, Van Snick J, Trovero F, Chamaillard M, Quesniaux VFJ, Ryffel B, Gosset P, Gombault A, Riteau N, Sokol H, Couillin I. NLRP6 controls pulmonary inflammation from cigarette smoke in a gut microbiota-dependent manner. Front Immunol 2023; 14:1224383. [PMID: 38146368 PMCID: PMC10749332 DOI: 10.3389/fimmu.2023.1224383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/20/2023] [Indexed: 12/27/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major health issue primarily caused by cigarette smoke (CS) and characterized by breathlessness and repeated airway inflammation. NLRP6 is a cytosolic innate receptor controlling intestinal inflammation and orchestrating the colonic host-microbial interface. However, its roles in the lungs remain largely unexplored. Using CS exposure models, our data show that airway inflammation is strongly impaired in Nlrp6-deficient mice with drastically fewer recruited neutrophils, a key cell subset in inflammation and COPD. We found that NLRP6 expression in lung epithelial cells is important to control airway and lung tissue inflammation in an inflammasome-dependent manner. Since gut-derived metabolites regulate NLRP6 inflammasome activation in intestinal epithelial cells, we investigated the link between NLRP6, CS-driven lung inflammation, and gut microbiota composition. We report that acute CS exposure alters gut microbiota in both wild-type (WT) and Nlrp6-deficient mice and that antibiotic treatment decreases CS-induced lung inflammation. In addition, gut microbiota transfer from dysbiotic Nlrp6-deficient mice to WT mice decreased airway lung inflammation in WT mice, highlighting an NLRP6-dependent gut-to-lung axis controlling pulmonary inflammation.
Collapse
Affiliation(s)
- Mégane Nascimento
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Sarah Huot-Marchand
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Manoussa Fanny
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Marjolène Straube
- Sorbonne Université, Institut National de la Recherche Médicale (INSERM), Centre de Recherche Saint-Antoine (CRSA), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint Antoine, Service de Gastroenterologie, Paris, France
| | - Marc Le Bert
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Florence Savigny
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | | | | | | | - Mathias Chamaillard
- Univ. Lille, Institut National de la Recherche Médicale (INSERM), U1003 - Laboratoire de physiologie cellulaire (PHYCEL) - Physiologie Cellulaire, Lille, France
| | - Valérie F. J. Quesniaux
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Bernhard Ryffel
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Philippe Gosset
- Institut PASTEUR INSERM U1019, Centre National de Recherche (CNRS) Unité Mixte de Recherche (UMR) 8204, Lille, France
| | - Aurélie Gombault
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Nicolas Riteau
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Harry Sokol
- Sorbonne Université, Institut National de la Recherche Médicale (INSERM), Centre de Recherche Saint-Antoine (CRSA), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint Antoine, Service de Gastroenterologie, Paris, France
- Institut national de la recherche agronomique (INRA), UMR1319 Micalis, AgroParisTech, Jouy-en-Josas, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Isabelle Couillin
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| |
Collapse
|
14
|
Fang Y, Xiao C, Wang L, Wang Y, Zeng J, Liang Y, Huang R, Shi Y, Wu S, Du X, Sun S, Li M, Zheng Y, Wu H, Guo Q, Yang W. Synergistic Enhancement of Isoforskolin and Dexamethasone Against Sepsis and Acute Lung Injury Mouse Models. J Inflamm Res 2023; 16:5989-6001. [PMID: 38088941 PMCID: PMC10712681 DOI: 10.2147/jir.s421232] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 10/09/2023] [Indexed: 11/01/2024] Open
Abstract
BACKGROUND Sepsis is initiated by the dysfunctional response of the host immune system to infection. Septic shock and acute lung injury (ALI) are the main etiology of death caused by sepsis. Glucocorticoids, which are commonly used in clinic to antagonize the inflammatory response of sepsis, may cause serious side effects. Isoforskolin (ISOF) from the plant Coleus forskohlii stimulates adenylyl cyclase, increases the cAMP level and inhibits inflammatory response. The aim of this study was to investigate the synergistic effect of ISOF with dexamethasone (DEX) to prevent and ameliorate septic inflammation. METHODS Lipopolysaccharide (LPS) of 30 and 5 mg/kg (iv.) was used to induce sepsis and ALI mice model respectively in vivo. BEAS-2B cells stimulated by LPS were applied as cell model in vitro. The cumulative survival of mice with LPS-induced sepsis and the histopathological changes of lungs in mice with acute lung injury were observed, and the secretion of pro-inflammatory cytokines was analyzed by ELISA. The expression of RGS2 in BEAS-2B cells was detected by immunoblotting assay and PCR. RESULTS In the sepsis mice model, ISOF (10 mg/kg) combined with DEX (10 mg/kg.) (ip.) pretreatment significantly increased mice survival rate from 33.3% to 58.3%, which was significantly higher than that of ISOF or DEX treated alone. In the ALI mice model, ISOF, DEX pretreatment alone and combined application attenuated pulmonary pathological changes in ALI mice. Furthermore, ISOF, DEX alone or combined administration decreased MPO, MDA, IL-6, and IL-8 levels, while significantly synergistic effects were observed in the combined treatment group compared with ISOF or DEX alone. In BEAS-2B cells, combined pretreatment with ISOF and DEX significantly decreased the expression of IL-8 and increased the expression of RGS2. CONCLUSION The results indicated that ISOF in combination with DEX synergistically improves survival rate and attenuates ALI in mice model through anti-inflammatory and antioxidant effects.
Collapse
Affiliation(s)
- Yan Fang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
- The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Chuang Xiao
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| | - Lueli Wang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| | - Youlan Wang
- Kunming Institute of Medical Sciences, Kunming, People’s Republic of China
| | - Jun Zeng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| | - Yaping Liang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| | - Rong Huang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| | - Yunke Shi
- The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Sha Wu
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| | - Xiaohua Du
- The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Shibo Sun
- The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Min Li
- The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Yuanyuan Zheng
- The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Hongxiang Wu
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| | - Qiuzhe Guo
- Fuwai Yunnan Cardiovascular Hospital, Kunming, People’s Republic of China
| | - Weimin Yang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, People’s Republic of China
| |
Collapse
|
15
|
Tesfaigzi Y, Curtis JL, Petrache I, Polverino F, Kheradmand F, Adcock IM, Rennard SI. Does Chronic Obstructive Pulmonary Disease Originate from Different Cell Types? Am J Respir Cell Mol Biol 2023; 69:500-507. [PMID: 37584669 PMCID: PMC10633838 DOI: 10.1165/rcmb.2023-0175ps] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/16/2023] [Indexed: 08/17/2023] Open
Abstract
The onset of chronic obstructive pulmonary disease (COPD) is heterogeneous, and current approaches to define distinct disease phenotypes are lacking. In addition to clinical methodologies, subtyping COPD has also been challenged by the reliance on human lung samples from late-stage diseases. Different COPD phenotypes may be initiated from the susceptibility of different cell types to cigarette smoke, environmental pollution, and infections at early stages that ultimately converge at later stages in airway remodeling and destruction of the alveoli when the disease is diagnosed. This perspective provides discussion points on how studies to date define different cell types of the lung that can initiate COPD pathogenesis, focusing on the susceptibility of macrophages, T and B cells, mast cells, dendritic cells, endothelial cells, and airway epithelial cells. Additional cell types, including fibroblasts, smooth muscle cells, neuronal cells, and other rare cell types not covered here, may also play a role in orchestrating COPD. Here, we discuss current knowledge gaps, such as which cell types drive distinct disease phenotypes and/or stages of the disease and which cells are primarily affected by the genetic variants identified by whole genome-wide association studies. Applying new technologies that interrogate the functional role of a specific cell type or a combination of cell types as well as single-cell transcriptomics and proteomic approaches are creating new opportunities to understand and clarify the pathophysiology and thereby the clinical heterogeneity of COPD.
Collapse
Affiliation(s)
- Yohannes Tesfaigzi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jeffrey L. Curtis
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | - Irina Petrache
- Division of Pulmonary Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado
- University of Colorado, Denver, Colorado
| | - Francesca Polverino
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, Baylor University, Houston, Texas
| | - Farrah Kheradmand
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, Baylor University, Houston, Texas
| | - Ian M. Adcock
- Department of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Stephen I. Rennard
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
16
|
Sankar P, Mishra BB. Early innate cell interactions with Mycobacterium tuberculosis in protection and pathology of tuberculosis. Front Immunol 2023; 14:1260859. [PMID: 37965344 PMCID: PMC10641450 DOI: 10.3389/fimmu.2023.1260859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/26/2023] [Indexed: 11/16/2023] Open
Abstract
Tuberculosis (TB) remains a significant global health challenge, claiming the lives of up to 1.5 million individuals annually. TB is caused by the human pathogen Mycobacterium tuberculosis (Mtb), which primarily infects innate immune cells in the lungs. These immune cells play a critical role in the host defense against Mtb infection, influencing the inflammatory environment in the lungs, and facilitating the development of adaptive immunity. However, Mtb exploits and manipulates innate immune cells, using them as favorable niche for replication. Unfortunately, our understanding of the early interactions between Mtb and innate effector cells remains limited. This review underscores the interactions between Mtb and various innate immune cells, such as macrophages, dendritic cells, granulocytes, NK cells, innate lymphocytes-iNKT and ILCs. In addition, the contribution of alveolar epithelial cell and endothelial cells that constitutes the mucosal barrier in TB immunity will be discussed. Gaining insights into the early cellular basis of immune reactions to Mtb infection is crucial for our understanding of Mtb resistance and disease tolerance mechanisms. We argue that a better understanding of the early host-pathogen interactions could inform on future vaccination approaches and devise intervention strategies.
Collapse
Affiliation(s)
| | - Bibhuti Bhusan Mishra
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| |
Collapse
|
17
|
Dotiwala F, Upadhyay AK. Next Generation Mucosal Vaccine Strategy for Respiratory Pathogens. Vaccines (Basel) 2023; 11:1585. [PMID: 37896988 PMCID: PMC10611113 DOI: 10.3390/vaccines11101585] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Inducing humoral and cytotoxic mucosal immunity at the sites of pathogen entry has the potential to prevent the infection from getting established. This is different from systemic vaccination, which protects against the development of systemic symptoms. The field of mucosal vaccination has seen fewer technological advances compared to nucleic acid and subunit vaccine advances for injectable vaccine platforms. The advent of the next-generation adenoviral vectors has given a boost to mucosal vaccine research. Basic research into the mechanisms regulating innate and adaptive mucosal immunity and the discovery of effective and safe mucosal vaccine adjuvants will continue to improve mucosal vaccine design. The results from clinical trials of inhaled COVID-19 vaccines demonstrate their ability to induce the proliferation of cytotoxic T cells and the production of secreted IgA and IgG antibodies locally, unlike intramuscular vaccinations. However, these mucosal vaccines induce systemic immune responses at par with systemic vaccinations. This review summarizes the function of the respiratory mucosa-associated lymphoid tissue and the advantages that the adenoviral vectors provide as inhaled vaccine platforms.
Collapse
Affiliation(s)
- Farokh Dotiwala
- Ocugen Inc., 11 Great Valley Parkway, Malvern, PA 19355, USA
| | | |
Collapse
|
18
|
Nashihah AK, Muhammad Firdaus FI, Fauzi MB, Mobarak NN, Lokanathan Y. Role of Biomaterials in the Development of Epithelial Support in 3D In Vitro Airway Epithelium Development: A Systematic Review. Int J Mol Sci 2023; 24:14935. [PMID: 37834382 PMCID: PMC10573735 DOI: 10.3390/ijms241914935] [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: 08/18/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
Respiratory diseases have a major impact on global health. The airway epithelium, which acts as a frontline defence, is one of the most common targets for inhaled allergens, irritants, or micro-organisms to enter the respiratory system. In the tissue engineering field, biomaterials play a crucial role. Due to the continuing high impact of respiratory diseases on society and the emergence of new respiratory viruses, in vitro airway epithelial models with high microphysiological similarities that are also easily adjustable to replicate disease models are urgently needed to better understand those diseases. Thus, the development of biomaterial scaffolds for the airway epithelium is important due to their function as a cell-support device in which cells are seeded in vitro and then are encouraged to lay down a matrix to form the foundations of a tissue for transplantation. Studies conducted in in vitro models are necessary because they accelerate the development of new treatments. Moreover, in comparatively controlled conditions, in vitro models allow for the stimulation of complex interactions between cells, scaffolds, and growth factors. Based on recent studies, the biomaterial scaffolds that have been tested in in vitro models appear to be viable options for repairing the airway epithelium and avoiding any complications. This review discusses the role of biomaterial scaffolds in in vitro airway epithelium models. The effects of scaffold, physicochemical, and mechanical properties in recent studies were also discussed.
Collapse
Affiliation(s)
- Ab Karim Nashihah
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia (F.I.M.F.); (M.B.F.)
| | - Fairuz Izan Muhammad Firdaus
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia (F.I.M.F.); (M.B.F.)
| | - Mh. Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia (F.I.M.F.); (M.B.F.)
| | - Nadhratun Naiim Mobarak
- School of Chemical Sciences and Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia UKM, Bangi 43600, Malaysia;
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia (F.I.M.F.); (M.B.F.)
| |
Collapse
|
19
|
Durnell LA, Hippee CE, Cattaneo R, Bartlett JA, Singh BK, Sinn PL. Interferon-independent processes constrain measles virus cell-to-cell spread in primary human airway epithelial cells. Microbiol Spectr 2023; 11:e0136123. [PMID: 37724882 PMCID: PMC10580916 DOI: 10.1128/spectrum.01361-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/27/2023] [Indexed: 09/21/2023] Open
Abstract
Amplification of measles virus (MeV) in human airway epithelia may contribute to its extremely high contagious nature. We use well-differentiated primary cultures of human airway epithelial cells (HAE) to model ex vivo how MeV spreads in human airways. In HAE, MeV spreads cell-to-cell for 3-5 days, but then, infectious center growth is arrested. What stops MeV spread in HAE is not understood, but interferon (IFN) is known to slow MeV spread in other in vitro and in vivo models. Here, we assessed the role of type I and type III IFN in arresting MeV spread in HAE. The addition of IFN-β or IFN-λ1 to the medium of infected HAE slowed MeV infectious center growth, but when IFN receptor signaling was blocked, infectious center size was not affected. In contrast, blocking type-I IFN receptor signaling enhanced respiratory syncytial virus spread. HAE were also infected with MeV mutants defective for the V protein. The V protein has been demonstrated to interact with both MDA5 and STAT2 to inhibit activation of innate immunity; however, innate immune reactions were unexpectedly muted against the V-defective MeV in HAE. Minimal innate immunity activation was confirmed by deep sequencing, quantitative RT-PCR, and single-cell RNA-seq analyses of the transcription of IFN and IFN-stimulated genes. We conclude that in HAE, IFN-signaling can contribute to slowing infectious center growth; however, IFN-independent processes are most important for limiting cell-to-cell spread. IMPORTANCE Fundamental biological questions remain about the highly contagious measles virus (MeV). MeV amplifies within airway epithelial cells before spreading to the next host. This final step likely contributes to the ability of MeV to spread host-to-host. Over the course of 3-5 days post-infection of airway epithelial cells, MeV spreads directly cell-to-cell and forms infectious centers. Infectious center formation is unique to MeV. In this study, we show that interferon (IFN) signaling does not explain why MeV cell-to-cell spread is ultimately impeded within the cell layer. The ability of MeV to spread cell-to-cell in airway cells without appreciable IFN induction may contribute to its highly contagious nature. This study contributes to the understanding of a significant global health concern by demonstrating that infectious center formation occurs independent of the simplest explanation for limiting viral transmission within a host.
Collapse
Affiliation(s)
- Lorellin A. Durnell
- Department of Microbiology and Immunology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Camilla E. Hippee
- Department of Microbiology and Immunology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jennifer A. Bartlett
- Stead Family Department of Pediatrics, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Brajesh K. Singh
- Stead Family Department of Pediatrics, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Patrick L. Sinn
- Department of Microbiology and Immunology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
- Stead Family Department of Pediatrics, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
20
|
Wang Q, Fu R, Li G, Xiong S, Zhu Y, Zhang H. Hedgehog receptors exert immune-surveillance roles in the epidermis across species. Cell Rep 2023; 42:112929. [PMID: 37527037 DOI: 10.1016/j.celrep.2023.112929] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 04/29/2023] [Accepted: 07/19/2023] [Indexed: 08/03/2023] Open
Abstract
Hedgehog signaling plays pivotal roles in the development and homeostasis of epithelial barrier tissues. However, whether and how Hedgehog signaling directly regulates innate immunity in epithelial cells remains unknown. By utilizing C. elegans epidermis as the model, we found that several Hedgehog receptors are involved in cell-autonomous regulation of the innate immune response in the epidermis. Particularly, loss of the Patched family receptor induces aberrant up-regulation of epidermal antimicrobial peptides in a STAT-dependent manner. External or internal insult to the epidermis triggers rapid rearrangement of Patched distribution along the plasma membrane, indicating that the Hedgehog (Hh) receptor is likely involved in recognition and defense against epidermal damage. Loss of PTCH1 function in primary human keratinocytes and intact mouse skin also results in STAT-dependent immune activation. These findings reveal an evolutionally conserved immune-surveillance function of Hedgehog receptors and an insult-sensing and response strategy of epithelial tissues.
Collapse
Affiliation(s)
- Qin Wang
- Jiangsu Key Laboratory of Infection and Immunity, Institute of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Rong Fu
- Jiangsu Key Laboratory of Infection and Immunity, Institute of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Gang Li
- Jiangsu Key Laboratory of Infection and Immunity, Institute of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Shaojie Xiong
- Jiangsu Key Laboratory of Infection and Immunity, Institute of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Yi Zhu
- Jiangsu Key Laboratory of Infection and Immunity, Institute of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Huimin Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institute of Biology and Medical Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
21
|
Dy ABC, Girkin J, Marrocco A, Collison A, Mwase C, O'Sullivan MJ, Phung TKN, Mattes J, Koziol-White C, Gern JE, Bochkov YA, Bartlett NW, Park JA. Rhinovirus infection induces secretion of endothelin-1 from airway epithelial cells in both in vitro and in vivo models. Respir Res 2023; 24:205. [PMID: 37598152 PMCID: PMC10440034 DOI: 10.1186/s12931-023-02510-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/11/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Rhinovirus (RV) infection of airway epithelial cells triggers asthma exacerbations, during which airway smooth muscle (ASM) excessively contracts. Due to ASM contraction, airway epithelial cells become mechanically compressed. We previously reported that compressed human bronchial epithelial (HBE) cells are a source of endothelin-1 (ET-1) that causes ASM contraction. Here, we hypothesized that epithelial sensing of RV by TLR3 and epithelial compression induce ET-1 secretion through a TGF-β receptor (TGFβR)-dependent mechanism. METHODS To test this, we used primary HBE cells well-differentiated in air-liquid interface culture and two mouse models (ovalbumin and house dust mite) of allergic airway disease (AAD). HBE cells were infected with RV-A16, treated with a TLR3 agonist (poly(I:C)), or exposed to compression. Thereafter, EDN1 (ET-1 protein-encoding gene) mRNA expression and secreted ET-1 protein were measured. We examined the role of TGFβR in ET-1 secretion using either a pharmacologic inhibitor of TGFβR or recombinant TGF-β1 protein. In the AAD mouse models, allergen-sensitized and allergen-challenged mice were subsequently infected with RV. We then measured ET-1 in bronchoalveolar lavage fluid (BALF) and airway hyperresponsiveness (AHR) following methacholine challenge. RESULTS Our data reveal that RV infection induced EDN1 expression and ET-1 secretion in HBE cells, potentially mediated by TLR3. TGFβR activation was partially required for ET-1 secretion, which was induced by RV, poly(I:C), or compression. TGFβR activation alone was sufficient to increase ET-1 secretion. In AAD mouse models, RV induced ET-1 secretion in BALF, which positively correlated with AHR. CONCLUSIONS Our data provide evidence that RV infection increased epithelial-cell ET-1 secretion through a TGFβR-dependent mechanism, which contributes to bronchoconstriction during RV-induced asthma exacerbations.
Collapse
Affiliation(s)
- Alane Blythe C Dy
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Jason Girkin
- College of Health, Medicine and Wellbeing, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Antonella Marrocco
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Adam Collison
- College of Health, Medicine and Wellbeing, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Chimwemwe Mwase
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Michael J O'Sullivan
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Thien-Khoi N Phung
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Joerg Mattes
- College of Health, Medicine and Wellbeing, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Australia
| | | | - James E Gern
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Yury A Bochkov
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Nathan W Bartlett
- College of Health, Medicine and Wellbeing, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Jin-Ah Park
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA.
| |
Collapse
|
22
|
Malik B, Bartlett NW, Upham JW, Nichol KS, Harrington J, Wark PAB. Severe asthma ILC2s demonstrate enhanced proliferation that is modified by biologics. Respirology 2023; 28:758-766. [PMID: 37114915 PMCID: PMC10946917 DOI: 10.1111/resp.14506] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 03/22/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND AND OBJECTIVE Type 2 (T2) innate lymphoid cells (ILC2s) contribute to airway inflammation and disease in asthma. We hypothesize that ILC2s isolated from people with severe allergic and eosinophilic asthma would exhibit an enhanced T2 inflammatory activity that would be altered following treatment with mepolizumab and omalizumab. We compare peripheral blood (PB) isolated ILC2's proliferative capacity, IL-5 and IL-13 secretion and phenotype between healthy without asthma (HC), non-asthma allergic (NAA), mild asthma (MA) and severe allergic and eosinophilic asthma (SA) subjects. We then determined the impact of 6 months treatment with either mepolizumab or omalizumab on ILC2s physiology of SA subjects. METHODS ILC2s were sorted and cultured in the presence of IL-2, IL-25, IL-33 and thymic stromal lymphopoietin (TSLP) for 14 days. ILC2s proliferation, phenotypes and functions were assessed using flowcytometry. The ILC2s response was then reassessed following clinically successful treatment of SA subjects with mepolizumab and omalizumab. RESULTS SA ILC2s demonstrated increased proliferative capacity, TSLP receptor (TSLPR), GATA3 and NFATc1 protein expressions and increased IL-5 and IL-13 release. ILC2s were also capable of releasing IL-6 in response to stimulation. Mepolizumab treatment reduced ILC2s proliferative capacity and expression of TSLPR, GATA3 and NFATc1. Both mepolizumab and omalizumab were associated with reduced ILC2s release of IL-5 and IL-13, only mepolizumab reduced IL-6. CONCLUSION ILC2s from severe allergic and eosinophilic asthma demonstrated an active phenotype typified by increased proliferation, TSLPR, GATA3 and NFATc1 expression and increased IL-5, IL-13 and IL-6 release. Mepolizumab reduced markers of ILC2s activation.
Collapse
Affiliation(s)
- Bilal Malik
- Immune Health Program, Hunter Medical Research InstituteUniversity of NewcastleCallaghanNew South WalesAustralia
| | - Nathan W. Bartlett
- Immune Health Program, Hunter Medical Research InstituteUniversity of NewcastleCallaghanNew South WalesAustralia
| | - John W. Upham
- Department of Respiratory MedicinePrincess Alexandra HospitalBrisbaneQueenslandAustralia
| | - Kristy S. Nichol
- Immune Health Program, Hunter Medical Research InstituteUniversity of NewcastleCallaghanNew South WalesAustralia
| | - John Harrington
- Department of Respiratory and Sleep MedicineJohn Hunter HospitalNew Lambton HeightsNew South WalesAustralia
| | - Peter A. B. Wark
- Immune Health Program, Hunter Medical Research InstituteUniversity of NewcastleCallaghanNew South WalesAustralia
- Department of Respiratory and Sleep MedicineJohn Hunter HospitalNew Lambton HeightsNew South WalesAustralia
| |
Collapse
|
23
|
Lee JY, Lee SM, Lee WK, Park JY, Kim DS. NAA10 Hypomethylation is associated with particulate matter exposure and worse prognosis for patients with non-small cell lung cancer. Anim Cells Syst (Seoul) 2023; 27:72-82. [PMID: 37033451 PMCID: PMC10075488 DOI: 10.1080/19768354.2023.2189934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/03/2023] [Indexed: 04/07/2023] Open
Abstract
Airborne particulate matter (PM) is a major health hazard worldwide and is a key factor in lung cancer, which remains the most common type of malignancy and the leading cause of cancer-related deaths. DNA methylation is a critical mechanism underlying the detrimental effects of PM, however, the molecular link between PM exposure and lung cancer remains to be elucidated. N-α-acetyltransferase 10 (NAA10) is involved in the cell cycle, migration, apoptosis, differentiation, and proliferation. In order to investigate the role of NAA10 in PM-induced pathogenesis processes leading to lung cancer, we determined the expression and methylation of NAA10 in normal human bronchial epithelial (NHBE) cells treated with PM10, PM10-polycyclic aromatic hydrocarbons (PAH), and PM2.5 and evaluated the prognostic value of the NAA10 methylation status in lung cancer patients. Exposure to all PM types significantly increased the expression of NAA10 mRNA and decreased the methylation of the NAA10 promoter in NHBE cells compared with the mock-treated control. NAA10 hypomethylation was observed in 9.3% (13/140) of lung cancer tissue samples and correlated with NAA10 transcriptional upregulation. Univariate and multivariate analyses revealed that NAA10 hypomethylation was associated with decreased survival of patients with lung cancer. Therefore, these results suggest that PM-induced hypomethylation of the NAA10 may play an important role in the pathogenesis of lung cancer and may be used as a potential prognostic biomarker for lung cancer progression. Further studies with large numbers of patients are warranted to confirm our findings.
Collapse
Affiliation(s)
- Ji Yun Lee
- Department of Anatomy, BK21 Plus Biomedical Convergence Program, Daegu, Republic of Korea
| | - Su Man Lee
- Department of Graduate School for Biomedical Science & Engineering, Hanyang University, Seoul, Republic of Korea
| | - Won Kee Lee
- Department of Preventive Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jae Yong Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Dong Sun Kim
- Department of Anatomy, BK21 Plus Biomedical Convergence Program, Daegu, Republic of Korea
| |
Collapse
|
24
|
Morens DM, Taubenberger JK, Fauci AS. Rethinking next-generation vaccines for coronaviruses, influenzaviruses, and other respiratory viruses. Cell Host Microbe 2023; 31:146-157. [PMID: 36634620 PMCID: PMC9832587 DOI: 10.1016/j.chom.2022.11.016] [Citation(s) in RCA: 77] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/26/2022] [Accepted: 11/29/2022] [Indexed: 01/13/2023]
Abstract
Viruses that replicate in the human respiratory mucosa without infecting systemically, including influenza A, SARS-CoV-2, endemic coronaviruses, RSV, and many other "common cold" viruses, cause significant mortality and morbidity and are important public health concerns. Because these viruses generally do not elicit complete and durable protective immunity by themselves, they have not to date been effectively controlled by licensed or experimental vaccines. In this review, we examine challenges that have impeded development of effective mucosal respiratory vaccines, emphasizing that all of these viruses replicate extremely rapidly in the surface epithelium and are quickly transmitted to other hosts, within a narrow window of time before adaptive immune responses are fully marshaled. We discuss possible approaches to developing next-generation vaccines against these viruses, in consideration of several variables such as vaccine antigen configuration, dose and adjuventation, route and timing of vaccination, vaccine boosting, adjunctive therapies, and options for public health vaccination polices.
Collapse
Affiliation(s)
- David M. Morens
- Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeffery K. Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA,Corresponding author
| | - Anthony S. Fauci
- Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
25
|
Wijsman PC, Goorsenberg AWM, Ravi A, d’Hooghe JNS, Dierdorp BS, Dekker T, van Schaik CCLM, ten Hacken NHT, Shah PL, Weersink EJM, Bel EH, Annema JT, Lutter R, Bonta PI. Airway Inflammation Before and After Bronchial Thermoplasty in Severe Asthma. J Asthma Allergy 2022; 15:1783-1794. [PMID: 36560975 PMCID: PMC9767029 DOI: 10.2147/jaa.s383418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/30/2022] [Indexed: 12/23/2022] Open
Abstract
Background Bronchial thermoplasty (BT) is a bronchoscopic treatment for severe asthma, of which the working mechanism and responder profile are partly unknown. The aim of this study is to analyse whether BT alters airway inflammation by epithelial gene expression, inflammatory cell counts and cytokines, and whether this relates to treatment response. Methods In this clinical trial, 28 severe asthma patients underwent bronchoscopy before and after treatment to obtain bronchial brushes and bronchoalveolar lavage fluid (BALF) from treated and untreated airways. RNA was extracted from bronchial brushes for transcriptome analysis, and BALF cells and cytokines were analysed. Asthma quality of life questionnaires were used to distinguish responders from non-responders. We compared results before and after treatment, between treated and untreated airways, and between responders and non-responders. Results Gene expression of airway epithelium related to airway inflammation gene set was significantly downregulated in treated airways compared to untreated airways, although this did not differ for patients before and after treatment. No differences were observed in cell counts and cytokines, neither from the untreated compared to treated airways, nor before and after treatment. At baseline, compared to non-responders, the expression of genes related to glycolysis in bronchial epithelium was downregulated and both BALF and blood eosinophil counts were higher in responders. Conclusion Local differences in gene sets pertaining to epithelial inflammatory status were identified between treated and untreated airways after treatment, not resulting in changes in differential cell counts and cytokine analyses in BALF. Secondly, baseline epithelial glycolysis genes and eosinophil counts in BALF and blood were different between responders and non-responders. The observations from this study demonstrate the potential impact of BT on epithelial gene expression related to airway inflammation while also identifying a possible responder profile.
Collapse
Affiliation(s)
- Pieta C Wijsman
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Annika W M Goorsenberg
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Abilash Ravi
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Julia N S d’Hooghe
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Barbara S Dierdorp
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Tamara Dekker
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Nick H T ten Hacken
- Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Pallav L Shah
- Department of Pulmonology, Royal Brompton Hospital, London, UK
- National Heart & Lung Institute, Imperial College, London, UK
- Department of Pulmonology, Chelsea & Westminster Hospital, London, UK
| | - Els J M Weersink
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Elisabeth H Bel
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jouke T Annema
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - René Lutter
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Peter I Bonta
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
26
|
Chen H, Durinck S, Patel H, Foreman O, Mesh K, Eastham J, Caothien R, Newman RJ, Roose-Girma M, Darmanis S, Warming S, Lattanzi A, Liang Y, Haley B. Population-wide gene disruption in the murine lung epithelium via AAV-mediated delivery of CRISPR-Cas9 components. MOLECULAR THERAPY - METHODS & CLINICAL DEVELOPMENT 2022; 27:431-449. [DOI: 10.1016/j.omtm.2022.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/27/2022] [Indexed: 11/13/2022]
|
27
|
Dickey BF, Chen J, Peebles RS. Airway Mucus Dysfunction in COVID-19. Am J Respir Crit Care Med 2022; 206:1304-1306. [PMID: 35830305 PMCID: PMC9746853 DOI: 10.1164/rccm.202207-1306ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Burton F. Dickey
- Department of Pulmonary MedicineThe University of Texas MD Anderson Cancer CenterHouston, Texas
| | - Jichao Chen
- Department of Pulmonary MedicineThe University of Texas MD Anderson Cancer CenterHouston, Texas
| | - R. Stokes Peebles
- Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashville, Tennessee
| |
Collapse
|
28
|
Jia J, Zeng M, Zhu D, Jiao X, Zhang B, Yang R, Feng W, Zheng X. An Amide Alkaloid Isolated from Ephedra sinica Ameliorates OVA-Induced Allergic Asthma by Inhibiting Mast Cell Activation and Dendritic Cell Maturation. Int J Mol Sci 2022; 23:13541. [PMID: 36362328 PMCID: PMC9655655 DOI: 10.3390/ijms232113541] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/30/2022] [Accepted: 11/01/2022] [Indexed: 09/14/2023] Open
Abstract
Asthma, which is a chronic inflammatory disease of the airways, is usually caused by allergens in which various structures and immune cells are involved. Ephedra sinica, the most commonly used Chinese medicine, has significant clinical effects on asthma, but its components are complex and the mechanism of action has not been fully elucidated. Among its components, we identified an amide alkaloid (EB-A) and investigated its anti-asthmatic activity and the underlying mechanisms. In this study, we replicated an OVA-sensitized/challenged allergic asthma mouse model, and divided the mice into a model (OVA) group, positive drug (Y, 0.5 mg/kg/day) group, and EB-A treatment with low (Low, 10 mg/kg/day) and high dose (High, 20 mg/kg/day) groups. Asthma-related features were analyzed through the airway hyperresponsiveness (AHR), cough and wheeze indexes, allergen-specific IgE, prostaglandin D2 (PDG2), and lung histology in mice. The levels of apoptosis and reactive oxygen species (ROS) in the primary lung cells, cytokines in the serum and broncho-alveolar lavage fluid (BALF), and proteinase-activated receptor-2 (PAR2) pathway activation in the lung tissue were measured to evaluate the inflammatory injury and lung epithelial barrier damage in the mice. Dendritic cell (DC) maturation and mast cell (MC) activation were verified in vitro and in vivo. Furthermore, the effect of a PAR2 activation in lung epithelial cells on the maturation of DCs was evaluated by the co-culture system of (human bronchial epithelial cell lines) 16HBE and bone marrow-derived dendritic cells (BMDCs). The results showed that EB-A inhibited the typical asthmatic phenotypes, as well as lung injury and inflammation, MC activation and degranulation, and DC maturation in the OVA-sensitized/challenged BALB/c mice. In addition, EB-A inhibited the expression of PAR2 in the lung epithelial cells and significantly interfered with the maturation of DCs after inhibiting PAR2. Taken together, our study firstly demonstrated that EB-A could ameliorate OVA-induced allergic asthma by inhibiting MC activation and DC maturation, and the molecular mechanism of EB-A's anti-asthmatic activity might be mediated by inhibiting PAR2. Our data provide a molecular justification for the use of EB-A in the treatment of allergic asthma.
Collapse
Affiliation(s)
- Jufang Jia
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Mengnan Zeng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Denghui Zhu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Xinmian Jiao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Beibei Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Ruolan Yang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| |
Collapse
|
29
|
Didriksen H, Molberg Ø, Mehta A, Jordan S, Palchevskiy V, Fretheim H, Gude E, Ueland T, Brunborg C, Garen T, Midtvedt Ø, Andreassen AK, Lund-Johansen F, Distler O, Belperio J, Hoffmann-Vold AM. Target organ expression and biomarker characterization of chemokine CCL21 in systemic sclerosis associated pulmonary arterial hypertension. Front Immunol 2022; 13:991743. [PMID: 36211384 PMCID: PMC9541617 DOI: 10.3389/fimmu.2022.991743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Systemic sclerosis (SSc) is a heterogenous disorder that appears to result from interplay between vascular pathologies, tissue fibrosis and immune processes, with evidence for deregulation of chemokines, which normally control immune trafficking. We recently identified altered levels of chemokine CCL21 in SSc associated pulmonary arterial hypertension (PAH). Here, we aimed to define target organ expression and biomarker characteristics of CCL21. Materials and methods To investigate target organ expression of CCL21, we performed immunohistochemistry (IHC) on explanted lung tissues from SSc-PAH patients. We assessed serum levels of CCL21 by ELISA and Luminex in two well-characterized SSc cohorts from Oslo (OUH, n=552) and Zurich (n=93) University hospitals and in 168 healthy controls. For detection of anti-CCl21 antibodies, we performed protein array analysis applying serum samples from SSc patients (n=300) and healthy controls. To characterize circulating CCL21 in SSc, we applied immunoprecipitation (IP) with antibodies detecting both full length and tailless and a custom-made antibody detecting only the C-terminal of CCL21. IP products were analyzed by SDS-PAGE/western blot and Mass spectrometry (MS). Results By IHC, we found that CCL21 was mainly expressed in the airway epithelial cells of SSc patients with PAH. In the analysis of serum levels of CCL21 we found weak correlation between Luminex and ELISA (r=0.515, p<0.001). Serum levels of anti-CCL21 antibodies were higher in SSc patients than in healthy controls (p<0.001), but only 5% of the SSc population were positive for anti-CCL21 antibodies in SSc, and we found no correlation between anti-CCl21 and serum levels of CCL21. By MS, we only identified peptides located within amino acid (aa) 23-102 of CCL21, indicating that CCL21 in SSc circulate as a truncated protein without the C-terminal tail. Conclusion This study demonstrates expression of CCL21 in epithelial lung tissue from SSc patients with PAH, and indicate that CCL21 in SSc circulates as a truncated protein. We extend previous observations indicating biomarker potential of CCL21, but find that Luminex is not suitable as platform for biomarker analyses. Finally, in vivo generated anti-CCL21 antibodies exist in SSc, but do not appear to modify serum CCL21 levels in patients with SSc-PAH.
Collapse
Affiliation(s)
- Henriette Didriksen
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Øyvind Molberg
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Adi Mehta
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Suzana Jordan
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Vyacheslav Palchevskiy
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Håvard Fretheim
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Einar Gude
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital – Rikshospitalet, Oslo, Norway
| | - Cathrine Brunborg
- Oslo Centre for Biostatistics and Epidemiology, Research Support Services, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Torhild Garen
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Øyvind Midtvedt
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Arne K. Andreassen
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | | | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - John Belperio
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Anna-Maria Hoffmann-Vold
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- *Correspondence: Anna-Maria Hoffmann-Vold,
| |
Collapse
|
30
|
Tumor-polarized GPX3 + AT2 lung epithelial cells promote premetastatic niche formation. Proc Natl Acad Sci U S A 2022; 119:e2201899119. [PMID: 35914155 PMCID: PMC9371733 DOI: 10.1073/pnas.2201899119] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The cellular and molecular components required for the formation of premetastatic niche (PMN) to promote lung metastasis need to be further investigated. Lung epithelial cells have been reported to exhibit immunomodulatory roles in lung homeostasis and also to mediate immunosuppressive PMN formation in lung metastasis. Here, by single-cell sequencing, we identified a tumor-polarized subpopulation of alveolar type 2 (AT2) epithelial cells with increased expression of glutathione peroxidase 3 (GPX3) and high production of interleukin (IL)-10 in the PMN. IL-10-producing GPX3+ AT2 cells inhibited CD4+ T cell proliferation but enhanced regulatory T cell generation. Mechanistically, tumor exosome-inducing GPX3 expression is required for GPX3+ AT2 cells to preferentially produce IL-10 by stabilizing hypoxia-inducible factor 1 (HIF-1α) and promoting HIF-1α-induced IL-10 production. Accordingly, conditional knockout of GPX3 in AT2 cells suppressed lung metastasis in spontaneous metastatic models. Together, our findings reveal a role of tumor-polarized GPX3+ AT2 cells in promoting lung PMN formation, adding insights into immune evasion in lung metastasis and providing potential targets for the intervention of tumor metastasis.
Collapse
|
31
|
Agrawal S, Monteiro C, Baca CF, Mohammadi R, Subramanian V, de Melo Bento CA, Agrawal A. Metabolites and growth factors produced by airway epithelial cells induce tolerance in macrophages. Life Sci 2022; 302:120659. [PMID: 35623392 PMCID: PMC10081865 DOI: 10.1016/j.lfs.2022.120659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/13/2022] [Accepted: 05/21/2022] [Indexed: 11/27/2022]
Abstract
Macrophages play a role in preventing inflammation in the respiratory tract. To investigate the mechanisms that lead to tolerance in macrophages, we examined the crosstalk between airway epithelial cells (AECs) and macrophages using a 2D coculture model. Culture of macrophages with AECs led to a significant inhibition of LPS induced pro-inflammatory responses. More importantly, AECs induced the secretion of TGF-β and IL-10 from macrophages even in the absence of LPS stimulation. In addition, the expression of inhibitory molecule, CD200R was also upregulated on AEC exposed macrophages. Furthermore, the AECs exposed macrophages induced significantly increased level of T regulatory cells. Investigation into the possible mechanisms indicated that a combination of growth factor, G-CSF, and metabolites, Kynurenine and lactic acid produced by AECs is responsible for inducing tolerance in macrophages. Interestingly, all these molecules had differential effect on macrophages with G-CSF inducing TGF-β, Kynurenine elevating IL-10, and lactic acid upregulating CD200R. Furthermore, a cocktail of these factors/metabolites induced similar changes in macrophages as AEC exposure. Altogether, these data identify factors secreted by AECs that enhance tolerance in the respiratory tract. These mediators thus have the potential to be used for therapeutic purposes to modulate respiratory inflammation following local viral infections and lung diseases.
Collapse
Affiliation(s)
- Sudhanshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, CA, USA 92617
| | - Clarice Monteiro
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, CA, USA 92617; Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil; Department of Microbiology, Immunology and Parasitology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | - Rezaa Mohammadi
- Department of Materials Science and Engineering, University of California Irvine, CA 92617, USA; Sue and Bill Stem Cell Center, University of California Irvine, CA 92617, USA
| | - Veedamali Subramanian
- Division of Gastroenterology, Department of Medicine, University of California Irvine, CA 92617, USA
| | - Cleonice Alves de Melo Bento
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil; Department of Microbiology, Immunology and Parasitology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Anshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, CA, USA 92617.
| |
Collapse
|
32
|
Sun H, Damania A, Mair ML, Otukoya E, Li YD, Polsky K, Zeng Y, Alt JA, Citardi MJ, Corry DB, Luong AU, Knight JM. STAT6 Blockade Abrogates Aspergillus-Induced Eosinophilic Chronic Rhinosinusitis and Asthma, A Model of Unified Airway Disease. Front Immunol 2022; 13:818017. [PMID: 35281012 PMCID: PMC8904741 DOI: 10.3389/fimmu.2022.818017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/28/2022] [Indexed: 12/25/2022] Open
Abstract
Unified airway disease, including concurrent asthma and chronic rhinosinusitis (CRS), is a common, but poorly understood disorder with no curative treatment options. To establish a murine model of chronic unified eosinophilic airway inflammation, mice were challenged with Aspergillus niger, and sinonasal mucosa and lung tissue were evaluated by immunohistochemistry, flow cytometry, and gene expression. Inhalation of A niger conidia resulted in a Th2-biased lung and sinus inflammation that typifies allergic asthma and CRS. Gene network and pathway analysis correlated with human disease with upregulation of not only the JAK-STAT and helper T-cell pathways, but also less expected pathways governing the spliceosome, osteoclast differentiation, and coagulation pathways. Utilizing a specific inhibitor and gene-deficient mice, we demonstrate that STAT6 is required for mycosis-induced sinus inflammation. These findings confirm the relevance of this new model and portend future studies that further extend our understanding of the immunopathologic basis of airway mycosis and unified airway disease.
Collapse
Affiliation(s)
- Hua Sun
- Center for Immunology and Autoimmune Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Department of Otorhinolaryngology-Head and Neck Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Ashish Damania
- Department of Pediatrics-Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Megan L Mair
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Eniola Otukoya
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Yi-Dong Li
- Center for Immunology and Autoimmune Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Department of Otorhinolaryngology-Head and Neck Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Katherine Polsky
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Yuying Zeng
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jeremiah A Alt
- Division of Otolaryngology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Martin J Citardi
- Department of Otorhinolaryngology-Head and Neck Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - David B Corry
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States.,Michael E. Debakey VA Center for Translational Research in Inflammatory Diseases, Houston, TX, United States
| | - Amber U Luong
- Center for Immunology and Autoimmune Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Department of Otorhinolaryngology-Head and Neck Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - John Morgan Knight
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
33
|
Morris CR, Habibovic A, Dustin CM, Schiffers C, Lin MC, Ather JL, Janssen-Heininger YMW, Poynter ME, Utermohlen O, Krönke M, van der Vliet A. Macrophage-intrinsic DUOX1 contributes to type 2 inflammation and mucus metaplasia during allergic airway disease. Mucosal Immunol 2022; 15:977-989. [PMID: 35654836 PMCID: PMC9391268 DOI: 10.1038/s41385-022-00530-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 02/24/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023]
Abstract
The NADPH oxidase DUOX1 contributes to epithelial production of alarmins, including interleukin (IL)-33, in response to injurious triggers such as airborne protease allergens, and mediates development of mucus metaplasia and airway remodeling in chronic allergic airways diseases. DUOX1 is also expressed in non-epithelial lung cell types, including macrophages that play an important role in airway remodeling during chronic lung disease. We therefore conditionally deleted DUOX1 in either lung epithelial or monocyte/macrophage lineages to address its cell-specific actions in innate airway responses to acute airway challenge with house dust mite (HDM) allergen, and in chronic HDM-driven allergic airway inflammation. As expected, acute responses to airway challenge with HDM, as well as type 2 inflammation and related features of airway remodeling during chronic HDM-induced allergic inflammation, were largely driven by DUOX1 with the respiratory epithelium. However, in the context of chronic HDM-driven inflammation, DUOX1 deletion in macrophages also significantly impaired type 2 cytokine production and indices of mucus metaplasia. Further studies revealed a contribution of macrophage-intrinsic DUOX1 in macrophage recruitment upon chronic HDM challenge, as well as features of macrophage activation that impact on type 2 inflammation and remodeling.
Collapse
Affiliation(s)
- Carolyn R Morris
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, USA
| | - Aida Habibovic
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, USA
| | - Christopher M Dustin
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, USA
| | - Caspar Schiffers
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, USA
| | - Miao-Chong Lin
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, USA
| | - Jennifer L Ather
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Yvonne M W Janssen-Heininger
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, USA
| | - Matthew E Poynter
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Olaf Utermohlen
- Institute for Medical Microbiology, Immunology and Hygiene, University Hospital Cologne, Cologne, Germany
| | - Martin Krönke
- Institute for Medical Microbiology, Immunology and Hygiene, University Hospital Cologne, Cologne, Germany
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, USA.
| |
Collapse
|
34
|
Qiu M, Tang Y, Chen J, Muriph R, Ye Z, Huang C, Evans J, Henske EP, Xu Q. Lung-selective mRNA delivery of synthetic lipid nanoparticles for the treatment of pulmonary lymphangioleiomyomatosis. Proc Natl Acad Sci U S A 2022; 119:e2116271119. [PMID: 35173043 PMCID: PMC8872770 DOI: 10.1073/pnas.2116271119] [Citation(s) in RCA: 215] [Impact Index Per Article: 107.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/18/2022] [Indexed: 11/30/2022] Open
Abstract
Safe and efficacious systemic delivery of messenger RNA (mRNA) to specific organs and cells in vivo remains the major challenge in the development of mRNA-based therapeutics. Targeting of systemically administered lipid nanoparticles (LNPs) coformulated with mRNA has largely been confined to the liver and spleen. Using a library screening approach, we identified that N-series LNPs (containing an amide bond in the tail) are capable of selectively delivering mRNA to the mouse lung, in contrast to our previous discovery that O-series LNPs (containing an ester bond in the tail) that tend to deliver mRNA to the liver. We analyzed the protein corona on the liver- and lung-targeted LNPs using liquid chromatography-mass spectrometry and identified a group of unique plasma proteins specifically absorbed onto the surface that may contribute to the targetability of these LNPs. Different pulmonary cell types can also be targeted by simply tuning the headgroup structure of N-series LNPs. Importantly, we demonstrate here the success of LNP-based RNA therapy in a preclinical model of lymphangioleiomyomatosis (LAM), a destructive lung disease caused by loss-of-function mutations in the Tsc2 gene. Our lung-targeting LNP exhibited highly efficient delivery of the mouse tuberous sclerosis complex 2 (Tsc2) mRNA for the restoration of TSC2 tumor suppressor in tumor and achieved remarkable therapeutic effect in reducing tumor burden. This research establishes mRNA LNPs as a promising therapeutic intervention for the treatment of LAM.
Collapse
Affiliation(s)
- Min Qiu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155
| | - Yan Tang
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115;
| | - Jinjin Chen
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155
| | - Rachel Muriph
- Department of Chemistry, University of Massachusetts Boston, Boston, MA 02125
| | - Zhongfeng Ye
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155
| | - Changfeng Huang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155
| | - Jason Evans
- Department of Chemistry, University of Massachusetts Boston, Boston, MA 02125
| | - Elizabeth P Henske
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115;
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155;
| |
Collapse
|
35
|
Lai HC, Lin TL, Chen TW, Kuo YL, Chang CJ, Wu TR, Shu CC, Tsai YH, Swift S, Lu CC. Gut microbiota modulates COPD pathogenesis: role of anti-inflammatory Parabacteroides goldsteinii lipopolysaccharide. Gut 2022; 71:309-321. [PMID: 33687943 DOI: 10.1136/gutjnl-2020-322599] [Citation(s) in RCA: 142] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 02/11/2021] [Accepted: 02/27/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Chronic obstructive pulmonary disease (COPD) is a global disease characterised by chronic obstruction of lung airflow interfering with normal breathing. Although the microbiota of respiratory tract is established to be associated with COPD, the causality of gut microbiota in COPD development is not yet established. We aimed to address the connection between gut microbiota composition and lung COPD development, and characterise bacteria and their derived active components for COPD amelioration. DESIGN A murine cigarette smoking (CS)-based model of COPD and strategies evaluating causal effects of microbiota were performed. Gut microbiota structure was analysed, followed by isolation of target bacterium. Single cell RNA sequencing, together with sera metabolomics analyses were performed to identify host responsive molecules. Bacteria derived active component was isolated, followed by functional assays. RESULTS Gut microbiota composition significantly affects CS-induced COPD development, and faecal microbiota transplantation restores COPD pathogenesis. A commensal bacterium Parabacteroides goldsteinii was isolated and shown to ameliorate COPD. Reduction of intestinal inflammation and enhancement of cellular mitochondrial and ribosomal activities in colon, systematic restoration of aberrant host amino acids metabolism in sera, and inhibition of lung inflammations act as the important COPD ameliorative mechanisms. Besides, the lipopolysaccharide derived from P. goldsteinii is anti-inflammatory, and significantly ameliorates COPD by acting as an antagonist of toll-like receptor 4 signalling pathway. CONCLUSION The gut microbiota-lung COPD axis was connected. A potentially benefial bacterial strain and its functional component may be developed and used as alternative agents for COPD prevention or treatment.
Collapse
Affiliation(s)
- Hsin-Chih Lai
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Microbiota Research Center and Emerging Viral Infections Research Center, Chang Gung University, Taoyuan, Taiwan.,Central Research Laboratory, Xiamen Chang Gung Hospital, XiaMen, China.,Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Tzu-Lung Lin
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ting-Wen Chen
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan.,Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.,Center For Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao Tung University, Hsinchu, Taiwan
| | - Yu-Lun Kuo
- Biotools, Co, Ltd, New Taipei City, Taiwan
| | - Chih-Jung Chang
- Central Research Laboratory, Xiamen Chang Gung Hospital, XiaMen, China
| | - Tsung-Ru Wu
- Institute of Biomedical Science, Academia Sinica, Taipei, Taiwan
| | - Ching-Chung Shu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ying-Huang Tsai
- Central Research Laboratory, Xiamen Chang Gung Hospital, XiaMen, China
| | - Simon Swift
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Chia-Chen Lu
- Department of Respiratory Therapy, Fu Jen Catholic University, New Taipei City, Taiwan .,Department of Chest Medicine, Internal Medicine, Fu Jen Catholic University Hospital, New Taipei City, Taiwan
| |
Collapse
|
36
|
Unravelling the molecular mechanisms underlying chronic respiratory diseases for the development of novel therapeutics via in vitro experimental models. Eur J Pharmacol 2022; 919:174821. [DOI: 10.1016/j.ejphar.2022.174821] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/01/2022] [Accepted: 02/09/2022] [Indexed: 12/11/2022]
|
37
|
Johansson K, Woodruff PG, Ansel KM. Regulation of airway immunity by epithelial miRNAs. Immunol Rev 2021; 304:141-153. [PMID: 34549450 PMCID: PMC9135676 DOI: 10.1111/imr.13028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 02/07/2023]
Abstract
The airway epithelium is essential to protect the host from inhaled pathogens and particles. It maintains immune homeostasis and mediates tissue repair after injury. Inflammatory diseases of the airways are associated with failure of epithelial functions, including loss of barrier integrity that results in increased tissue permeability and immune activation; excessive mucus secretion and impaired mucociliary clearance that leads to airflow obstruction and microbial overgrowth; and dysregulation of cellular signals that promotes inflammation and alters tissue structure and airway reactivity. MicroRNAs play crucial roles in mounting appropriate cellular responses to environmental stimuli and preventing disease, using a common machinery and mechanism to regulate gene expression in epithelial cells, immune cells of hematopoietic origin, and other cellular components of the airways. Respiratory diseases are accompanied by dramatic changes in epithelial miRNA expression that drive persistent immune dysregulation. In this review, we discuss responses of the epithelium that promote airway immunopathology, with a focus on miRNAs that contribute to the breakdown of essential epithelial functions. We emphasize the emerging role of miRNAs in regulation of epithelial responses in respiratory health and their value as diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Kristina Johansson
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
- Sandler Asthma Basic Research Center, University of California, San Francisco, California, USA
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of California, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| | - Prescott G. Woodruff
- Sandler Asthma Basic Research Center, University of California, San Francisco, California, USA
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of California, San Francisco, California, USA
- Cardiovascular Research Institute, University of California, San Francisco, California, USA
| | - K. Mark Ansel
- Sandler Asthma Basic Research Center, University of California, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| |
Collapse
|
38
|
Wu K, Kamimoto K, Zhang Y, Yang K, Keeler SP, Gerovac BJ, Agapov EV, Austin SP, Yantis J, Gissy KA, Byers DE, Alexander-Brett J, Hoffmann CM, Wallace M, Hughes ME, Crouch EC, Morris SA, Holtzman MJ. Basal epithelial stem cells cross an alarmin checkpoint for postviral lung disease. J Clin Invest 2021; 131:e149336. [PMID: 34343135 PMCID: PMC8483760 DOI: 10.1172/jci149336] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Epithelial cells are charged with protection at barrier sites, but whether this normally beneficial response might sometimes become dysfunctional still needs definition. Here, we recognized a pattern of imbalance marked by basal epithelial cell growth and differentiation that replaced normal airspaces in a mouse model of progressive postviral lung disease due to the Sendai virus. Single-cell and lineage-tracing technologies identified a distinct subset of basal epithelial stem cells (basal ESCs) that extended into gas-exchange tissue to form long-term bronchiolar-alveolar remodeling regions. Moreover, this cell subset was selectively expanded by crossing a cell-growth and survival checkpoint linked to the nuclear-localized alarmin IL-33 that was independent of IL-33 receptor signaling and instead connected to autocrine chromatin accessibility. This mechanism creates an activated stem-progenitor cell lineage with potential for physiological or pathological function. Thus, conditional loss of Il33 gene function in basal epithelial cells disrupted the homeostasis of the epithelial barrier at skin and gut sites but also markedly attenuated postviral disease in the lung based on the downregulation of remodeling and inflammation. Thus, we define a basal ESC strategy to deploy innate immune machinery that appears to overshoot the primordial goal of self-defense. Our findings reveal new targets to stratify and correct chronic and often deadly postviral disease.
Collapse
Affiliation(s)
- Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine
| | - Kenji Kamimoto
- Department of Genetics
- Department of Developmental Biology
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine
| | - Kuangying Yang
- Pulmonary and Critical Care Medicine, Department of Medicine
- Division of Biostatistics
| | | | | | | | | | - Jennifer Yantis
- Pulmonary and Critical Care Medicine, Department of Medicine
| | - Kelly A. Gissy
- Pulmonary and Critical Care Medicine, Department of Medicine
| | - Derek E. Byers
- Pulmonary and Critical Care Medicine, Department of Medicine
| | - Jennifer Alexander-Brett
- Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Pathology and Immunology
| | | | - Matthew Wallace
- Pulmonary and Critical Care Medicine, Department of Medicine
| | - Michael E. Hughes
- Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Genetics
| | | | | | - Michael J. Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
39
|
Balkrishna A, Solleti SK, Singh H, Singh R, Sharma N, Varshney A. Biotite-Calx Based Traditional Indian Medicine Sahastraputi-Abhrak-Bhasma Prophylactically Mitigates Allergic Airway Inflammation in a Mouse Model of Asthma by Amending Cytokine Responses. J Inflamm Res 2021; 14:4743-4760. [PMID: 34557016 PMCID: PMC8455516 DOI: 10.2147/jir.s313955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/25/2021] [Indexed: 12/15/2022] Open
Abstract
Purpose Asthma is a heterogeneous airway inflammatory disease with limited therapeutic options. Traditional medicine is extensively used for treating various ailments including asthma. Sahastraputi-Abhrak-Bhasma (SPAB) is a biotite-calx based Indian medicine. Methods We have tested for the anti-inflammatory and anti-asthmatic properties of SPAB, using a mouse model of ovalbumin-induced allergic asthma in-vivo and cell-based assays in-vitro. Histological analysis, qPCR and ELISA were performed to assess the pathology. SEM, EDX and XRD-analysis were performed to characterize the SPAB particles. Results SEM, EDX and XRD-analysis identified the presence of SPAB particle of 100 nm–~1µm diameter and contains annite-1M, aluminium silicate, kyanite, aluminium oxide, magnesium silicate, and maghemite in the samples. Ova-challenge resulted in severe inflammatory responses, airway remodelling and increased oxidative burden in lungs. Importantly, prophylactic treatment with SPAB significantly attenuated allergen induced leukocyte infiltration specifically eosinophils, lymphocytes, macrophages and neutrophils in BALF. Ova-induced mucus hypersecretion, peri-bronchial collagen deposition, inflammatory cell infiltration and bronchial epithelial thickening were significantly abrogated upon SPAB treatment. qPCR and ELISA analysis identified that allergen induced increases in IL-5, IL-13, IL-33, IFN-γ and IL-1β cytokines mRNA in whole lungs and the levels of IL-6, IL-1β and TNF-α proteins in BALF were significantly attenuated upon oral SPAB treatment. SPAB restored allergen induced decreases in anti-oxidant markers in lungs. In-vitro, SPAB attenuated the secretion of IL-6, and TNF-α from human bronchial epithelial cells and modestly inhibited NF-kB/AP-1 pathway in HEK cells. Conclusion Taken together, our results experimentally validated the prophylactic ameliorative potential of the Indian classical medicine Sahastraputi-Abhrak-Bhasma against asthma associated airway inflammation.
Collapse
Affiliation(s)
- Acharya Balkrishna
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, Uttarakhand, India.,Department of Allied and Applied Sciences, University of Patanjali, Patanjali Yog Peeth, Haridwar, Uttarakhand, India.,Patanjali UK Trust, Glasgow, UK
| | - Siva Kumar Solleti
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, Uttarakhand, India
| | - Hoshiyar Singh
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, Uttarakhand, India
| | - Rani Singh
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, Uttarakhand, India
| | - Niti Sharma
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, Uttarakhand, India
| | - Anurag Varshney
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, Uttarakhand, India.,Department of Allied and Applied Sciences, University of Patanjali, Patanjali Yog Peeth, Haridwar, Uttarakhand, India.,Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
40
|
Medina CB, Chiu YH, Stremska ME, Lucas CD, Poon I, Tung KS, Elliott MR, Desai B, Lorenz UM, Bayliss DA, Ravichandran KS. Pannexin 1 channels facilitate communication between T cells to restrict the severity of airway inflammation. Immunity 2021; 54:1715-1727.e7. [PMID: 34283971 PMCID: PMC8363584 DOI: 10.1016/j.immuni.2021.06.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/21/2021] [Accepted: 06/21/2021] [Indexed: 01/14/2023]
Abstract
Allergic airway inflammation is driven by type-2 CD4+ T cell inflammatory responses. We uncover an immunoregulatory role for the nucleotide release channel, Panx1, in T cell crosstalk during airway disease. Inverse correlations between Panx1 and asthmatics and our mouse models revealed the necessity, specificity, and sufficiency of Panx1 in T cells to restrict inflammation. Global Panx1-/- mice experienced exacerbated airway inflammation, and T-cell-specific deletion phenocopied Panx1-/- mice. A transgenic designed to re-express Panx1 in T cells reversed disease severity in global Panx1-/- mice. Panx1 activation occurred in pro-inflammatory T effector (Teff) and inhibitory T regulatory (Treg) cells and mediated the extracellular-nucleotide-based Treg-Teff crosstalk required for suppression of Teff cell proliferation. Mechanistic studies identified a Salt-inducible kinase-dependent phosphorylation of Panx1 serine 205 important for channel activation. A genetically targeted mouse expressing non-phosphorylatable Panx1S205A phenocopied the exacerbated inflammation in Panx1-/- mice. These data identify Panx1-dependent Treg:Teff cell communication in restricting airway disease.
Collapse
Affiliation(s)
- Christopher B Medina
- Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA; Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Yu-Hsin Chiu
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu, 30071, Taiwan
| | - Marta E Stremska
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Christopher D Lucas
- Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA
| | - Ivan Poon
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, VIC 3083, Australia
| | - Kenneth S Tung
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Michael R Elliott
- Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA; Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Bimal Desai
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Ulrike M Lorenz
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Kodi S Ravichandran
- Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA; Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA; VIB/UGent Inflammation Research Centre and the Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
41
|
Teafatiller T, Agrawal S, De Robles G, Rahmatpanah F, Subramanian VS, Agrawal A. Vitamin C Enhances Antiviral Functions of Lung Epithelial Cells. Biomolecules 2021; 11:1148. [PMID: 34439814 PMCID: PMC8394979 DOI: 10.3390/biom11081148] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
Vitamin C is well documented to have antiviral functions; however, there is limited information about its effect on airway epithelial cells-the first cells to encounter infections. Here, we examined the effect of vitamin C on human bronchial epithelium transformed with Ad12-SV40 2B (BEAS-2B) cells, and observed that sodium-dependent vitamin C transporter 2 (SVCT2) was the primary vitamin C transporter. Transcriptomic analysis revealed that treating BEAS-2B cells with vitamin C led to a significant upregulation of several metabolic pathways and interferon-stimulated genes (ISGs) along with a downregulation of pathways involved in lung injury and inflammation. Remarkably, vitamin C also enhanced the expression of the viral-sensing receptors retinoic acid-inducible gene 1 (RIG-1) and melanoma differentiation-associated protein 5 (MDA-5), which was confirmed at the protein and functional levels. In addition, the lungs of l-gulono-γ-lactone oxidase knockout (GULO-KO) mice also displayed a marked decrease in these genes compared to wild-type controls. Collectively, our findings indicate that vitamin C acts at multiple levels to exert its antiviral and protective functions in the lungs.
Collapse
Affiliation(s)
- Trevor Teafatiller
- Division of Gastroenterology, Department of Medicine, University of California, Irvine, CA 92697, USA;
| | - Sudhanshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA 92697, USA;
| | - Gabriela De Robles
- Department of Pathology, University of California, Irvine, CA 92697, USA; (G.D.R.); (F.R.)
| | - Farah Rahmatpanah
- Department of Pathology, University of California, Irvine, CA 92697, USA; (G.D.R.); (F.R.)
| | - Veedamali S. Subramanian
- Division of Gastroenterology, Department of Medicine, University of California, Irvine, CA 92697, USA;
| | - Anshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA 92697, USA;
| |
Collapse
|
42
|
Warren KJ, Poole JA, Sweeter JM, DeVasure JM, Dickinson JD, Peebles RS, Wyatt TA. Neutralization of IL-33 modifies the type 2 and type 3 inflammatory signature of viral induced asthma exacerbation. Respir Res 2021; 22:206. [PMID: 34266437 PMCID: PMC8281667 DOI: 10.1186/s12931-021-01799-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022] Open
Abstract
Background Respiratory viral infections are one of the leading causes of need for emergency care and hospitalizations in asthmatic individuals, and airway-secreted cytokines are released within hours of viral infection to initiate these exacerbations. IL-33, specifically, contributes to these allergic exacerbations by amplifying type 2 inflammation. We hypothesized that blocking IL-33 in RSV-induced exacerbation would significantly reduce allergic inflammation. Methods Sensitized BALB/c mice were challenged with aerosolized ovalbumin (OVA) to establish allergic inflammation, followed by RSV-A2 infection to yield four treatment groups: saline only (Saline), RSV-infected alone (RSV), OVA alone (OVA), and OVA-treated with RSV infection (OVA-RSV). Lung outcomes included lung mRNA and protein markers of allergic inflammation, histology for mucus cell metaplasia and lung immune cell influx by cytospin and flow cytometry. Results While thymic stromal lymphopoietin (TSLP) and IL-33 were detected 6 h after RSV infection in the OVA-RSV mice, IL-23 protein was uniquely upregulated in RSV-infected mice alone. OVA-RSV animals varied from RSV- or OVA-treated mice as they had increased lung eosinophils, neutrophils, group 2 innate lymphoid cells (ILC2) and group 3 innate lymphoid cells (ILC3) detectable as early as 6 h after RSV infection. Neutralized IL-33 significantly reduced ILC2 and eosinophils, and the prototypical allergic proteins, IL-5, IL-13, CCL17 and CCL22 in OVA-RSV mice. Numbers of neutrophils and ILC3 were also reduced with anti-IL-33 treatment in both RSV and OVA-RSV treated animals as well. Conclusions Taken together, our findings indicate a broad reduction in allergic-proinflammatory events mediated by IL-33 neutralization in RSV-induced asthma exacerbation.
Collapse
Affiliation(s)
- Kristi J Warren
- Division of Pulmonary Medicine, Department of Internal Medicine, University of Utah Health, 26 N 1900 E, Salt Lake City, UT, 84132, USA. .,VA Salt Lake City Health Care System, Salt Lake City, UT, 84148, USA.
| | - Jill A Poole
- Critical Care and Sleep Division, Department of Internal Medicine, Pulmonary, University of Nebraska Medical Center, Omaha, USA
| | - Jenea M Sweeter
- Critical Care and Sleep Division, Department of Internal Medicine, Pulmonary, University of Nebraska Medical Center, Omaha, USA
| | - Jane M DeVasure
- Critical Care and Sleep Division, Department of Internal Medicine, Pulmonary, University of Nebraska Medical Center, Omaha, USA
| | - John D Dickinson
- Critical Care and Sleep Division, Department of Internal Medicine, Pulmonary, University of Nebraska Medical Center, Omaha, USA
| | - R Stokes Peebles
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, USA
| | - Todd A Wyatt
- Critical Care and Sleep Division, Department of Internal Medicine, Pulmonary, University of Nebraska Medical Center, Omaha, USA.,Department of Environmental, Agricultural and Occupational Health, University of Nebraska Medical Center, Omaha, NE, 68198-5910, USA.,VA Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
| |
Collapse
|
43
|
Chiarella SE, Cardet JC, Prakash YS. Sex, Cells, and Asthma. Mayo Clin Proc 2021; 96:1955-1969. [PMID: 34218868 PMCID: PMC8262071 DOI: 10.1016/j.mayocp.2020.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/19/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022]
Abstract
There are marked sex differences in asthma prevalence and severity. Sex hormones play a central role in these sex biases and directly interact with multiple key cells involved in the pathogenesis of asthma. Here we review the known effects of estrogen, progesterone, and testosterone on airway epithelial cells, airway smooth muscle cells, the mononuclear phagocyte system, innate lymphoid cells, eosinophils, mast cells, T cells, and B cells, all in the context of asthma. Furthermore, we explore unresolved clinical questions, such as the role of sex hormones in the link between asthma and obesity.
Collapse
Affiliation(s)
- Sergio E Chiarella
- Division of Allergic Diseases, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Juan Carlos Cardet
- Division of Allergy and Immunology, Department of Internal Medicine, University of South Florida, Tampa
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN.
| |
Collapse
|
44
|
Ramu S, Akbarshahi H, Mogren S, Berlin F, Cerps S, Menzel M, Hvidtfeldt M, Porsbjerg C, Uller L, Andersson CK. Direct effects of mast cell proteases, tryptase and chymase, on bronchial epithelial integrity proteins and anti-viral responses. BMC Immunol 2021; 22:35. [PMID: 34078278 PMCID: PMC8170739 DOI: 10.1186/s12865-021-00424-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mast cells (MCs) are known to contribute to both acute and chronic inflammation. Bronchial epithelial cells are the first line of defence against pathogens and a deficient anti-viral response has been suggested to play a role in the pathogenesis of asthma exacerbations. However, effects of MC mediators on bronchial epithelial immune response have been less studied. The aim of this study is to investigate the direct effects of stimulation with MC proteases, tryptase and chymase, on inflammatory and anti-viral responses in human bronchial epithelial cells (HBECs). METHOD Cultured BEAS-2b cells and primary HBECs from 3 asthmatic patients were stimulated with tryptase or chymase (0.1 to 0.5 μg/ml) for 1, 3, 6 and 24 h. To study the effects of MC mediators on the anti-viral response, cells were stimulated with 10 μg/ml of viral mimic Poly (I:C) for 3 and 24 h following pre-treatment with 0.5 μg/ml tryptase or chymase for 3 h. Samples were analysed for changes in pro-inflammatory and anti-viral mediators and receptors using RT-qPCR, western blot and Luminex. RESULTS Tryptase and chymase induced release of the alarmin ATP and pro-inflammatory mediators IL-8, IL-6, IL-22 and MCP-1 from HBECs. Moreover, tryptase and chymase decreased the expression of E-cadherin and zonula occludens-1 expression from HBECs. Pre-treatment of HBECs with tryptase and chymase further increased Poly (I:C) induced IL-8 release at 3 h. Furthermore, tryptase significantly reduced type-I and III interferons (IFNs) and pattern recognition receptor (PRR) expression in HBECs. Tryptase impaired Poly (I:C) induced IFN and PRR expression which was restored by treatment of a serine protease inhibitor. Similar effects of tryptase on inflammation and anti-viral responses were also confirmed in primary HBECs from asthmatic patients. CONCLUSION MC localization within the epithelium and the release of their proteases may play a critical role in asthma pathology by provoking pro-inflammatory and alarmin responses and downregulating IFNs. Furthermore, MC proteases induce downregulation of epithelial junction proteins which may lead to barrier dysfunction. In summary, our data suggests that mast cells may contribute towards impaired anti-viral epithelial responses during asthma exacerbations mediated by the protease activity of tryptase.
Collapse
Affiliation(s)
- Sangeetha Ramu
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Hamid Akbarshahi
- Department of Experimental Medical Science, Lund University, Lund, Sweden.,Department of Respiratory Medicine and Allergology, Lund University, Lund, Sweden
| | - Sofia Mogren
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Frida Berlin
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Samuel Cerps
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Mandy Menzel
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Morten Hvidtfeldt
- Department of Respiratory Medicine, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Celeste Porsbjerg
- Department of Respiratory Medicine, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Lena Uller
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | |
Collapse
|
45
|
Kwon EK, Choi Y, Yoon IH, Won HK, Sim S, Lee HR, Kim HS, Ye YM, Shin YS, Park HS, Ban GY. Oleoylethanolamide induces eosinophilic airway inflammation in bronchial asthma. Exp Mol Med 2021; 53:1036-1045. [PMID: 34079051 PMCID: PMC8257664 DOI: 10.1038/s12276-021-00622-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 12/31/2022] Open
Abstract
Asthma is a chronic eosinophilic inflammatory disease with an increasing prevalence worldwide. Endocannabinoids are known to have immunomodulatory biological effects. However, the contribution of oleoylethanolamide (OEA) to airway inflammation remains to be elucidated. To investigate the effect of OEA, the expression of proinflammatory cytokines was measured by RT-qPCR and ELISA in airway epithelial (A549) cells. The numbers of airway inflammatory cells and cytokine levels in bronchoalveolar lavage fluid, airway hyperresponsiveness, and type 2 innate lymphoid cells (ILC2s) were examined in BALB/c mice after 4 days of OEA treatment. Furthermore, eosinophil activation after OEA treatment was evaluated by measuring cellular CD69 levels in eosinophils from human peripheral eosinophils using flow cytometry. OEA induced type 2 inflammatory responses in vitro and in vivo. OEA increased the levels of proinflammatory cytokines, such as IL-6, IL-8, and IL-33, in A549 cells. In addition, it also induced eosinophilic inflammation, the production of IL-4, IL-5, IL-13, and IL-33 in bronchoalveolar lavage fluid, and airway hyperresponsiveness. OEA increased the numbers of IL-5- or IL-13-producing ILC2s in a mouse model. Finally, we confirmed that OEA increased CD69 expression (an eosinophil activation marker) on purified eosinophils from patients with asthma compared to those from healthy controls. OEA may play a role in the pathogenesis of asthma by activating ILC2s and eosinophils.
Collapse
Affiliation(s)
- Eun-Kyung Kwon
- Department of Pulmonary, Allergy and Critical Care Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Youngwoo Choi
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Il-Hee Yoon
- VHS Veterans Medical Research Institute, VHS Medical Center, Seoul, Korea
| | - Ha-Kyeong Won
- Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Korea
| | - Soyoon Sim
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | | | - Hyoung Su Kim
- Department of Internal Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Young-Min Ye
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Yoo Seob Shin
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Ga-Young Ban
- Department of Pulmonary, Allergy and Critical Care Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea. .,Allergy and Clinical Immunology Research Center, Hallym University College of Medicine, Dongtan, Korea.
| |
Collapse
|
46
|
The interplay between airway epithelium and the immune system - A primer for the respiratory clinician. Paediatr Respir Rev 2021; 38:2-8. [PMID: 33812796 PMCID: PMC8178232 DOI: 10.1016/j.prrv.2021.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023]
Abstract
The respiratory epithelium is one of the primary interfaces between the body's immune system and the external environment. This review discusses the innate and adaptive immunomodulatory effects of the respiratory epithelium, highlighting the physiologic immune responses associated with health and the disease-causing sequelae when these physiologic responses go awry. Airway macrophages, dendritic cells, and innate lymphoid cells are discussed as orchestrators of physiological and pathological innate immune responses and T cells, B cells, mast cells, and granulocytes (eosinophils and neutrophils) as orchestrators of physiologic and pathologic adaptive immune responses. The interplay between the airway epithelium and the varied immune cells as well as the interplay between these immune cells is discussed, highlighting the importance of the dose of noxious stimuli and pathogens in immune programming and the timing of their interaction with the immune cells that determine the pattern of immune responses. Although each cell type has been researched individually, this review highlights the need for simultaneous temporal investigation of immune responses from these varied cells to noxious stimuli and pathogens.
Collapse
|
47
|
Wang HX, Qin XH, Shen J, Liu QH, Shi YB, Xue L. Proteomic Analysis Reveals That Placenta-Specific Protein 9 Inhibits Proliferation and Stimulates Motility of Human Bronchial Epithelial Cells. Front Oncol 2021; 11:628480. [PMID: 34123785 PMCID: PMC8194706 DOI: 10.3389/fonc.2021.628480] [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: 11/13/2020] [Accepted: 04/14/2021] [Indexed: 11/13/2022] Open
Abstract
Placenta-specific protein 9 (PLAC9) is a putative secretory protein that was initially identified in the placenta and is involved in cell proliferation and motility. Bioinformatics analyses revealed that PLAC9 is repressed in lung cancers (LCs), especially lung adenocarcinomas, compared to that in the paired adjacent normal tissues, indicating that PLAC9 might be involved in the pathogenesis of pulmonary diseases. To investigate the potential role of PLAC9 in the abnormal reprogramming of airway epithelial cells (AECs), a key cause of pulmonary diseases, we constructed a stable PLAC9-overexpressing human bronchial epithelial cell line (16HBE-GFP-Plac9). We utilized the proteomic approach isobaric tag for relative and absolute quantification (iTRAQ) to analyze the effect of PLAC9 on cellular protein composition. Gene ontology (GO) and pathway analyses revealed that GO terms and pathways associated with cell proliferation, cell cycle progression, and cell motility and migration were significantly enriched among the proteins regulated by PLAC9. Our in vitro results showed that PLAC9 overexpression reduced cell proliferation, altered cell cycle progression, and increased cell motility, including migration and invasion. Our findings suggest that PLAC9 inhibits cell proliferation through S phase arrest by altering the expression levels of cyclin/cyclin-dependent kinases (CDKs) and promotes cell motility, likely via the concerted actions of cyclins, E-cadherin, and vimentin. Since these mechanisms may underlie PLAC9-mediated abnormal human bronchial pathogenesis, our study provides a basis for the development of molecular targeted treatments for LCs.
Collapse
Affiliation(s)
- Hai-Xia Wang
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Xu-Hui Qin
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Jinhua Shen
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Qing-Hua Liu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Lu Xue
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China.,Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Allergic asthma reflects the interplay between inflammatory mediators and immune, airway epithelial, and other cells. This review summarizes key insights in these areas over the past year. RECENT FINDINGS Key findings over the past year demonstrate that epithelial cells mediate tight junction breakdown to facilitate the development of asthma-like disease in mice. Innate lymph lymphoid cells (ILC), while previously shown to promote allergic airway disease, have now been shown to inhibit the development of severe allergic disease in mice. Fibrinogen cleavage products (previously shown to mediate allergic airway disease and macrophage fungistatic immunity by signaling through Toll-like receptor 4) have now been shown to first bind to the integrin Mac-1 (CD11c/CD18). Therapeutically, recent discoveries include the development of the antiasthma drug PM-43I that inhibits the allergy-related transcription factors STAT5 and STAT6 in mice, and confirmatory evidence of the efficacy of the antifungal agent voriconazole in human asthma. SUMMARY Studies over the past year provide critical new insight into the mechanisms by which epithelial cells, ILC, and coagulation factors contribute to the expression of asthma-like disease and further support the development antiasthma drugs that block STAT factors and inhibit fungal growth in the airways.
Collapse
|
49
|
Bonner K, Scotney E, Saglani S. Factors and mechanisms contributing to the development of preschool wheezing disorders. Expert Rev Respir Med 2021; 15:745-760. [PMID: 33881953 DOI: 10.1080/17476348.2021.1913057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Half of all children will experience an episode of wheezing by their sixth birthday and acute episodes of wheezing in preschool children account for the majority of all childhood hospital admissions for wheeze. Recurrent preschool wheezing associates with early loss of lung function and a life-long impact on lung health. AREAS COVERED We reviewed the literature on PubMed from August 2010-2020 focussing on factors associated with wheeze inception and persistence, paying specific attention to mechanistic studies that have investigated the impact of early life exposures in shaping immune responses in children with underlying susceptibility to wheezing. In particular, the role of early allergen sensitization, respiratory infections, and the impact of the environment on shaping the airway microbiome and resulting immune responses are discussed. EXPERT OPINION There is an abundance of associative data showing the role of in utero and postnatal factors influencing wheeze onset and persistence. However, mechanistic and stratified, biomarker-based interventional studies that confirm these associations are now needed if we are to impact the significant healthcare burden resulting from preschool wheezing disorders.
Collapse
Affiliation(s)
- Katie Bonner
- Inflammation, Repair & Development Section, National Heart & Lung Institute, Imperial College London, London, UK.,Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, London, UK
| | - Elizabeth Scotney
- Inflammation, Repair & Development Section, National Heart & Lung Institute, Imperial College London, London, UK.,Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, London, UK
| | - Sejal Saglani
- Inflammation, Repair & Development Section, National Heart & Lung Institute, Imperial College London, London, UK.,Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, London, UK
| |
Collapse
|
50
|
Wang X, Wu K, Keeler SP, Mao D, Agapov EV, Zhang Y, Holtzman MJ. TLR3-Activated Monocyte-Derived Dendritic Cells Trigger Progression from Acute Viral Infection to Chronic Disease in the Lung. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:1297-1314. [PMID: 33514511 PMCID: PMC7946811 DOI: 10.4049/jimmunol.2000965] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 01/01/2021] [Indexed: 11/19/2022]
Abstract
Acute infection is implicated as a trigger for chronic inflammatory disease, but the full basis for this switch is uncertain. In this study, we examine this issue using a mouse model of chronic lung disease that develops after respiratory infection with a natural pathogen (Sendai virus). We investigate this model using a combination of TLR3-deficient mice and adoptive transfer of immune cells into these mice versus the comparable responses in wild-type mice. We found that acute and transient expression of TLR3 on monocyte-derived dendritic cells (moDCs) was selectively required to induce long-term expression of IL-33 and consequent type 2 immune-driven lung disease. Unexpectedly, moDC participation was not based on canonical TLR3 signaling and relied instead on a trophic effect to expand the alveolar epithelial type 2 cell population beyond repair of tissue injury and thereby provide an enriched and persistent cell source of IL-33 required for progression to a disease phenotype that includes lung inflammation, hyperreactivity, excess mucus production, and remodeling. The findings thereby provide a framework wherein viral infection activates TLR3 in moDCs as a front-line immune cell niche upstream of lung epithelial cells to drive the type 2 immune response, leading to chronic inflammatory diseases of the lung (such as asthma and chronic obstructive pulmonary disease in humans) and perhaps progressive and long-term postviral disease in general.
Collapse
Affiliation(s)
- Xinyu Wang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Shamus P Keeler
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Dailing Mao
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Eugene V Agapov
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|