1
|
Li Y, Mao X, Shi P, Wan Z, Yang D, Ma T, Wang B, Wang J, Wang J, Zhu R. Microbiome-host interactions in the pathogenesis of acute exacerbation of chronic obstructive pulmonary disease. Front Cell Infect Microbiol 2024; 14:1386201. [PMID: 39091676 PMCID: PMC11291260 DOI: 10.3389/fcimb.2024.1386201] [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: 02/14/2024] [Accepted: 06/24/2024] [Indexed: 08/04/2024] Open
Abstract
Objective To explore the underlying mechanisms the airway microbiome contributes to Acute Exacerbation of Chronic Obstructive Pulmonary Disease(AECOPD). Methods We enrolled 31 AECOPD patients and 26 stable COPD patients, their sputum samples were collected for metagenomic and RNA sequencing, and then subjected to bioinformatic analyses. The expression of host genes was validated by Quantitative Real-time PCR(qPCR) using the same batch of specimens. Results Our results indicated a higher expression of Rothia mucilaginosa(p=0.015) in the AECOPD group and Haemophilus influenzae(p=0.005) in the COPD group. The Different expressed genes(DEGs) detected were significantly enriched in "type I interferon signaling pathway"(p<0.001, q=0.001) in gene function annotation, and "Cytosolic DNA-sensing pathway"(p=0.002, q=0.024), "Toll-like receptor signaling pathway"(p=0.006, q=0.045), and "TNF signaling pathway"(p=0.006, q=0.045) in KEGG enrichment analysis. qPCR amplification experiment verified that the expression of OASL and IL6 increased significantly in the AECOPD group. Conclusion Pulmonary bacteria dysbiosis may regulate the pathogenesis of AECOPD through innate immune system pathways like type I interferon signaling pathway and Toll-like receptor signaling pathway.
Collapse
Affiliation(s)
- Yao Li
- Department of Respiratory and Critical Care Medicine, Huaian Clinical College of Xuzhou Medical University, Huaian, China
| | - Xiaoyan Mao
- Department of Intensive Care Unit, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, China
| | - Pengfei Shi
- Department of Respiratory and Critical Care Medicine, Huaian Clinical College of Xuzhou Medical University, Huaian, China
| | - Zongren Wan
- Department of Respiratory and Critical Care Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Dan Yang
- Department of Respiratory and Critical Care Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Ting Ma
- Department of Respiratory and Critical Care Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Baolan Wang
- Department of Respiratory and Critical Care Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Jipeng Wang
- Department of Respiratory and Critical Care Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Jingjing Wang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rong Zhu
- Department of Respiratory and Critical Care Medicine, Huaian Clinical College of Xuzhou Medical University, Huaian, China
| |
Collapse
|
2
|
Kheradmand F, Zhang Y, Corry DB. Contribution of adaptive immunity to human COPD and experimental models of emphysema. Physiol Rev 2023; 103:1059-1093. [PMID: 36201635 PMCID: PMC9886356 DOI: 10.1152/physrev.00036.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 02/01/2023] Open
Abstract
The pathophysiology of chronic obstructive pulmonary disease (COPD) and the undisputed role of innate immune cells in this condition have dominated the field in the basic research arena for many years. Recently, however, compelling data suggesting that adaptive immune cells may also contribute to the progressive nature of lung destruction associated with COPD in smokers have gained considerable attention. The histopathological changes in the lungs of smokers can be limited to the large or small airways, but alveolar loss leading to emphysema, which occurs in some individuals, remains its most significant and irreversible outcome. Critically, however, the question of why emphysema progresses in a subset of former smokers remained a mystery for many years. The recognition of activated and organized tertiary T- and B-lymphoid aggregates in emphysematous lungs provided the first clue that adaptive immune cells may play a crucial role in COPD pathophysiology. Based on these findings from human translational studies, experimental animal models of emphysema were used to determine the mechanisms through which smoke exposure initiates and orchestrates adaptive autoreactive inflammation in the lungs. These models have revealed that T helper (Th)1 and Th17 subsets promote a positive feedback loop that activates innate immune cells, confirming their role in emphysema pathogenesis. Results from genetic studies and immune-based discoveries have further provided strong evidence for autoimmunity induction in smokers with emphysema. These new findings offer a novel opportunity to explore the mechanisms underlying the inflammatory landscape in the COPD lung and offer insights for development of precision-based treatment to halt lung destruction.
Collapse
Affiliation(s)
- Farrah Kheradmand
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas
| | - Yun Zhang
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - David B Corry
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas
| |
Collapse
|
3
|
Zamorina S, Timganova V, Bochkova M, Shardina K, Uzhviyuk S, Khramtsov P, Usanina D, Rayev M. The Effect of PEGylated Graphene Oxide Nanoparticles on the Th17-Polarization of Activated T Helpers. MATERIALS (BASEL, SWITZERLAND) 2023; 16:877. [PMID: 36676614 PMCID: PMC9865146 DOI: 10.3390/ma16020877] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 06/17/2023]
Abstract
We investigated the direct effect of PEGylated graphene oxide (P-GO) nanoparticles on the differentiation, viability, and cytokine profile of activated T helper type 17 (Th17) in vitro. The subject of the study were cultures of "naive" T-helpers (CD4+) isolated by immunomagnetic separation and polarized into the Th17 phenotype with a TCR activator and cytokines. It was found that P-GO at low concentrations (5 µg/mL) had no effect on the parameters studied. The presence of high concentrations of P-GO in T-helper cultures (25 μg/mL) did not affect the number and viability of these cells. However, the percentage of proliferating T-helpers in these cultures was reduced. GO nanoparticles modified with linear polyethylene glycol (PEG) significantly increased the percentage of Th17/22 cells in cultures of Th17-polarized T helpers and the production of IFN-γ, whereas those modified with branched PEG suppressed the synthesis of IL-17. Thus, a low concentration of PEGylated GO nanoparticles (5 μg/mL), in contrast to a concentration of 25 μg/mL, has no effect on the Th17-polarization of T helpers, allowing their further use for in-depth studies of the functions of T lymphocytes and other immune cells. Overall, we have studied for the first time the direct effect of P-GO nanoparticles on the conversion of T helper cells to the Th17 phenotype.
Collapse
Affiliation(s)
- Svetlana Zamorina
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| | - Valeria Timganova
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
| | - Maria Bochkova
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| | - Kseniya Shardina
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
| | - Sofya Uzhviyuk
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
| | - Pavel Khramtsov
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| | - Darya Usanina
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| | - Mikhail Rayev
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| |
Collapse
|
4
|
Weidinger D, Jamal Jameel K, Alisch D, Jacobsen J, Bürger P, Ruhe M, Yusuf F, Rohde S, Störtkuhl K, Kaufmann P, Kronsbein J, Peters M, Hatt H, Giannakis N, Knobloch J. OR2AT4 and OR1A2 counterregulate molecular pathophysiological processes of steroid-resistant inflammatory lung diseases in human alveolar macrophages. Mol Med 2022; 28:150. [PMID: 36503361 PMCID: PMC9743598 DOI: 10.1186/s10020-022-00572-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 11/08/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Therapeutic options for steroid-resistant non-type 2 inflammation in obstructive lung diseases are lacking. Alveolar macrophages are central in the progression of these diseases by releasing proinflammatory cytokines, making them promising targets for new therapeutic approaches. Extra nasal expressed olfactory receptors (ORs) mediate various cellular processes, but clinical data are lacking. This work investigates whether ORs in human primary alveolar macrophages could impact pathophysiological processes and could be considered as therapeutic targets. METHODS Human primary alveolar macrophages were isolated from bronchoalveolar lavages of 50 patients with pulmonary diseases. The expression of ORs was validated using RT-PCR, immunocytochemical staining, and Western blot. Changes in intracellular calcium levels were analyzed in real-time by calcium imaging. A luminescent assay was used to measure the cAMP concentration after OR stimulation. Cytokine secretion was measured in cell supernatants 24 h after stimulation by ELISA. Phagocytic ability was measured by the uptake of fluorescent-labeled beads by flow cytometry. RESULTS We demonstrated the expression of functional OR2AT4 and OR1A2 on mRNA and protein levels. Both ORs were primarily located in the plasma membrane. Stimulation with Sandalore, the ligand of OR2AT4, and Citronellal, the ligand of OR1A2, triggered a transient increase of intracellular calcium and cAMP. In the case of Sandalore, this calcium increase was based on a cAMP-dependent signaling pathway. Stimulation of alveolar macrophages with Sandalore and Citronellal reduced phagocytic capacity and release of proinflammatory cytokines. CONCLUSION These are the first indications for utilizing olfactory receptors as therapeutic target molecules in treating steroid-resistant lung diseases with non-type 2 inflammation.
Collapse
Affiliation(s)
- Daniel Weidinger
- grid.5570.70000 0004 0490 981XMedical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Kaschin Jamal Jameel
- grid.5570.70000 0004 0490 981XMedical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Desiree Alisch
- grid.5570.70000 0004 0490 981XMedical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Julian Jacobsen
- grid.5570.70000 0004 0490 981XMedical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Paul Bürger
- grid.5570.70000 0004 0490 981XMedical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Matthias Ruhe
- grid.5570.70000 0004 0490 981XMedical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Faisal Yusuf
- grid.5570.70000 0004 0490 981XMedical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Simon Rohde
- grid.5570.70000 0004 0490 981XMedical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Klemens Störtkuhl
- grid.5570.70000 0004 0490 981XAG Physiology of Senses, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Peter Kaufmann
- grid.5570.70000 0004 0490 981XMedical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Juliane Kronsbein
- grid.5570.70000 0004 0490 981XMedical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Marcus Peters
- grid.5570.70000 0004 0490 981XDepartment of Molecular Immunology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Hanns Hatt
- grid.5570.70000 0004 0490 981XDepartment of Cell Physiology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Nikolaos Giannakis
- grid.5570.70000 0004 0490 981XMedical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Jürgen Knobloch
- grid.5570.70000 0004 0490 981XMedical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| |
Collapse
|
5
|
Lee FFY, Alper S. Alternative pre-mRNA splicing as a mechanism for terminating Toll-like Receptor signaling. Front Immunol 2022; 13:1023567. [PMID: 36531997 PMCID: PMC9755862 DOI: 10.3389/fimmu.2022.1023567] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/11/2022] [Indexed: 12/03/2022] Open
Abstract
While inflammation induced by Toll-like receptor (TLR) signaling is required to combat infection, persistent inflammation can damage host tissues and contribute to a myriad of acute and chronic inflammatory disorders. Thus, it is essential not only that TLR signaling be activated in the presence of pathogens but that TLR signaling is ultimately terminated. One mechanism that limits persistent TLR signaling is alternative pre-mRNA splicing. In addition to encoding the canonical mRNAs that produce proteins that promote inflammation, many genes in the TLR signaling pathway also encode alternative mRNAs that produce proteins that are dominant negative inhibitors of signaling. Many of these negative regulators are induced by immune challenge, so production of these alternative isoforms represents a negative feedback loop that limits persistent inflammation. While these alternative splicing events have been investigated on a gene by gene basis, there has been limited systemic analysis of this mechanism that terminates TLR signaling. Here we review what is known about the production of negatively acting alternative isoforms in the TLR signaling pathway including how these inhibitors function, how they are produced, and what role they may play in inflammatory disease.
Collapse
Affiliation(s)
- Frank Fang Yao Lee
- Department of Immunology and Genomic Medicine and Center for Genes, Environment, and Health, National Jewish Health, Denver, CO, United States,Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz, CO, United States
| | - Scott Alper
- Department of Immunology and Genomic Medicine and Center for Genes, Environment, and Health, National Jewish Health, Denver, CO, United States,Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz, CO, United States,*Correspondence: Scott Alper,
| |
Collapse
|
6
|
Lin L, Li J, Song Q, Cheng W, Chen P. The role of HMGB1/RAGE/TLR4 signaling pathways in cigarette smoke-induced inflammation in chronic obstructive pulmonary disease. Immun Inflamm Dis 2022; 10:e711. [PMID: 36301039 PMCID: PMC9552978 DOI: 10.1002/iid3.711] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/10/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common chronic respiratory disease with irreversible and continuous progression. It has become the fifth most burdensome disease and the third most deadly disease globally. Therefore, the prevention and treatment of COPD are urgent, and it is also important to clarify the pathogenesis of it. Smoking is the main and most common risk factor for COPD. Cigarette smoke (CS) can cause lung inflammation and other pathological mechanisms in the airways and lung tissue. Airway inflammation is one of the important mechanisms leading to the pathogenesis of COPD. Recent studies have shown that high mobility group box 1 (HMGB1) is involved in the occurrence and development of respiratory diseases, including COPD. HMGB1 is a typical damage-associated molecular pattern (DAMP) protein, which mainly exerts its activity by binding to the receptor for advanced glycation end products (RAGE) and toll-like receptor 4 (TLR4) and further participate in the process of airway inflammation. Studies have shown that the abnormal expression of HMGB1, RAGE, and TLR4 are related to inflammation in COPD. Herein, we discuss the roles of HMGB1, RAGE, and TLR4 in CS/cigarette smoke extract-induced inflammation in COPD, providing a new target for the diagnosis, treatment and prevention of COPD.
Collapse
Affiliation(s)
- Ling Lin
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Jing Li
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Qing Song
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Wei Cheng
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Ping Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| |
Collapse
|
7
|
Pei Y, Wei Y, Peng B, Wang M, Xu W, Chen Z, Ke X, Rong L. Combining single-cell RNA sequencing of peripheral blood mononuclear cells and exosomal transcriptome to reveal the cellular and genetic profiles in COPD. Respir Res 2022; 23:260. [PMID: 36127695 PMCID: PMC9490964 DOI: 10.1186/s12931-022-02182-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 09/09/2022] [Indexed: 11/30/2022] Open
Abstract
Background It has been a long-held consensus that immune reactions primarily mediate the pathology of chronic obstructive pulmonary disease (COPD), and that exosomes may participate in immune regulation in COPD. However, the relationship between exosomes and peripheral immune status in patients with COPD remains unclear. Methods In this study, we sequenced plasma exosomes and performed single-cell RNA sequencing on peripheral blood mononuclear cells (PBMCs) from patients with COPD and healthy controls. Finally, we constructed competing endogenous RNA (ceRNA) and protein–protein interaction (PPI) networks to delineate the interactions between PBMCs and exosomes within COPD. Results We identified 135 mRNAs, 132 lncRNAs, and 359 circRNAs from exosomes that were differentially expressed in six patients with COPD compared with four healthy controls. Functional enrichment analyses revealed that many of these differentially expressed RNAs were involved in immune responses including defending viral infection and cytokine–cytokine receptor interaction. We also identified 18 distinct cell clusters of PBMCs in one patient and one control by using an unsupervised cluster analysis called uniform manifold approximation and projection (UMAP). According to resultant cell identification, it was likely that the proportions of monocytes, dendritic cells, and natural killer cells increased in the COPD patient we tested, meanwhile the proportions of B cells, CD4 + T cells, and naïve CD8 + T cells declined. Notably, CD8 + T effector memory CD45RA + (Temra) cell and CD8 + effector memory T (Tem) cell levels were elevated in patient with COPD, which were marked by their lower capacity to differentiate due to their terminal differentiation state and lower reactive capacity to viral pathogens. Conclusions We generated exosomal RNA profiling and single-cell transcriptomic profiling of PBMCs in COPD, described possible connection between impaired immune function and COPD development, and finally determined the possible role of exosomes in mediating local and systemic immune reactions. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02182-8.
Collapse
Affiliation(s)
- Yanli Pei
- Respiratory Medicine Department, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yuxi Wei
- Peking Union Medical College (PUMC), PUMC and Chinese Academy of Medical Sciences, Beijing, China
| | - Boshizhang Peng
- Peking Union Medical College (PUMC), PUMC and Chinese Academy of Medical Sciences, Beijing, China
| | - Mengqi Wang
- Peking Union Medical College (PUMC), PUMC and Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Xu
- Respiratory Medicine Department, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhe Chen
- Laboratory of Cough, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu, China.
| | - Xindi Ke
- Peking Union Medical College (PUMC), PUMC and Chinese Academy of Medical Sciences, Beijing, China.
| | - Lei Rong
- Respiratory Medicine Department, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
8
|
Wang L, Zhao H, Raman I, Yan M, Chen Q, Li QZ. Peripheral Blood Mononuclear Cell Gene Expression in Chronic Obstructive Pulmonary Disease: miRNA and mRNA Regulation. J Inflamm Res 2022; 15:2167-2180. [PMID: 35392023 PMCID: PMC8983057 DOI: 10.2147/jir.s337894] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/16/2022] [Indexed: 01/01/2023] Open
Affiliation(s)
- Lijing Wang
- Departments of Geriatrics, Respiratory Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People’s Republic of China
| | - Hongjun Zhao
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People’s Republic of China
| | - Indu Raman
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Mei Yan
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Qiong Chen
- Departments of Geriatrics, Respiratory Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People’s Republic of China
| | - Quan-Zhen Li
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Correspondence: Quan-Zhen Li, Department of Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA, Tel +1 214-645-6071, Fax +1 214-645-6074, Email
| |
Collapse
|
9
|
Birru RL, Bein K, Bondarchuk N, Wells H, Lin Q, Di YP, Leikauf GD. Antimicrobial and Anti-Inflammatory Activity of Apple Polyphenol Phloretin on Respiratory Pathogens Associated With Chronic Obstructive Pulmonary Disease. Front Cell Infect Microbiol 2021; 11:652944. [PMID: 34881190 PMCID: PMC8645934 DOI: 10.3389/fcimb.2021.652944] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Bacterial infections contribute to accelerated progression and severity of chronic obstructive pulmonary disease (COPD). Apples have been associated with reduced symptoms of COPD and disease development due to their polyphenolic content. We examined if phloretin, an apple polyphenol, could inhibit bacterial growth and inflammation induced by the main pathogens associated with COPD. Phloretin displayed bacteriostatic and anti-biofilm activity against nontypeable Haemophilus influenzae (NTHi), Moraxella catarrhalis, Streptococcus pneumoniae, and to a lesser extent, Pseudomonas aeruginosa. In vitro, phloretin inhibited NTHi adherence to NCI-H292 cells, a respiratory epithelial cell line. Phloretin also exhibited anti-inflammatory activity in COPD pathogen-induced RAW 264.7 macrophages and human bronchial epithelial cells derived from normal and COPD diseased lungs. In mice, NTHi bacterial load and chemokine (C-X-C motif) ligand 1 (CXCL1), a neutrophil chemoattractant, was attenuated by a diet supplemented with phloretin. Our data suggests that phloretin is a promising antimicrobial and anti-inflammatory nutraceutical for reducing bacterial-induced injury in COPD.
Collapse
Affiliation(s)
- Rahel L Birru
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kiflai Bein
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Natalya Bondarchuk
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Heather Wells
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Qiao Lin
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Y Peter Di
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - George D Leikauf
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
10
|
Jamal Jameel K, Gallert WJ, Yanik SD, Panek S, Kronsbein J, Jungck D, Koch A, Knobloch J. Biomarkers for Comorbidities Modulate the Activity of T-Cells in COPD. Int J Mol Sci 2021; 22:ijms22137187. [PMID: 34281240 PMCID: PMC8269158 DOI: 10.3390/ijms22137187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/17/2021] [Accepted: 06/27/2021] [Indexed: 02/06/2023] Open
Abstract
In smoking-induced chronic obstructive pulmonary disease (COPD), various comorbidities are linked to systemic inflammation and infection-induced exacerbations. The underlying mechanisms are unclear but might provide therapeutic targets. T-cell activity is central in systemic inflammation and for infection-defense mechanisms and might be influenced by comorbidities. Hypothesis: Circulating biomarkers of comorbidities modulate the activity of T-cells of the T-helper type 1 (Th1) and/or T-cytotoxic type 1 (Tc1). T-cells in peripheral blood mononuclear cells (PBMCs) from non-smokers (NS), current smokers without COPD (S), and COPD subjects (total n = 34) were ex vivo activated towards Th1/Tc1 and were then stimulated with biomarkers for metabolic and/or cardiovascular comorbidities (Brain Natriuretic Peptide, BNP; chemokine (C-C motif) ligand 18, CCL18; C-X3-C motif chemokine ligand 1, CX3CL1; interleukin-18, IL-18) or for asthma- and/or cancer-related comorbidities (CCL22; epidermal growth factor, EGF; IL-17; periostin) each at 10 or 50 ng/mL. The Th1/Tc1 activation markers interferon-γ (IFNγ), tumor necrosis factor-α (TNFα), and granulocyte-macrophage colony-stimulating factor (GM-CSF) were analyzed in culture supernatants by Enzyme-Linked Immunosorbent Assay (ELISA). Ex-vivo activation induced IFNγ and TNFα without differences between the groups but GM-CSF more in S vs. NS. At 10 ng/mL, the different biomarkers increased or reduced the T-cell activation markers without a clear trend for one direction in the different categories of comorbidities or for the different T-cell activation markers. At 50 ng/mL, there was a clear shift towards suppressive effects, particularly for the asthma— and cancer-related biomarkers and in cells of S and COPD. Comorbidities might suppress T-cell immunity in COPD. This could explain the association of comorbidities with frequent exacerbations.
Collapse
Affiliation(s)
- Kaschin Jamal Jameel
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany; (K.J.J.); (W.-J.G.); (S.D.Y.); (S.P.); (J.K.)
| | - Willem-Jakob Gallert
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany; (K.J.J.); (W.-J.G.); (S.D.Y.); (S.P.); (J.K.)
| | - Sarah D. Yanik
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany; (K.J.J.); (W.-J.G.); (S.D.Y.); (S.P.); (J.K.)
| | - Susanne Panek
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany; (K.J.J.); (W.-J.G.); (S.D.Y.); (S.P.); (J.K.)
| | - Juliane Kronsbein
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany; (K.J.J.); (W.-J.G.); (S.D.Y.); (S.P.); (J.K.)
| | - David Jungck
- Department of Internal Medicine II, Pneumology, Allergology and Respiratory Medicine, Bethel Teaching Hospital, 12207 Berlin, Germany;
| | - Andrea Koch
- Pyhrn-Eisenwurzen-Klinikum Steyr, Klinik für Pneumologie, Lehrkrankenhaus der Uniklinik Linz, Sierninger Str. 170, 4400 Steyr, Austria;
- Ludwig-Maximilians-University of Munich (LMU) and DZL (German Center of Lung Science), 81377 Munich, Germany
| | - Jürgen Knobloch
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany; (K.J.J.); (W.-J.G.); (S.D.Y.); (S.P.); (J.K.)
- Correspondence: ; Tel.: +49-234-302-3404; Fax: +49-234-302-6420
| |
Collapse
|
11
|
Haider MB, Basida B, Bapatla A, Ismail R, Hafeez W. Herpes Simplex Virus Esophagitis in the Setting of Acute Use of Corticosteroids for COPD Exacerbation in an Immunocompetent Middle-Aged Woman. Case Rep Gastroenterol 2021; 15:616-620. [PMID: 34616265 PMCID: PMC8454247 DOI: 10.1159/000515413] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 02/19/2021] [Indexed: 11/19/2022] Open
Abstract
Herpes simplex (HSV) esophagitis is usually identified in patients with significant immunosuppressive conditions such as AIDS. Short course of immunosuppressive therapy is an uncommon risk factor for this condition. We present a case of acute gastrointestinal bleeding secondary to HSV type 1-induced esophageal ulcers. A 63-year-old woman developed acute hypoxic hypercapnic respiratory failure. Past medical history was significant for COPD for which the patient was taking short-acting bronchodilator inhalers. The patient was intubated and started on mechanical ventilation. Intravenous Solu-Medrol 40 mg Q6 was started. Hospital course was complicated by sepsis of unknown source. Empiric broad-spectrum antibiotic therapy was started. On the 11th hospital day, the patient experienced multiple episodes of coffee ground emesis. There was abdominal tenderness on physical examination. Significant laboratory results were lipase 1,911 U/L and lymphopenia (ALC = 300/mm3). Endoscopy revealed severe erosive esophagitis and multiple punched-out ulcerations of the esophagus. Empiric treatment with valacyclovir 500 mg OD was started. The patient required PEG tube insertion for dysphagia. Complete resolution of esophagitis was noted then. Immunohistochemical staining for HSV was strongly positive in the cells with inclusions. Short course of intravenous corticosteroids is an uncommon cause of HSV-1 esophagitis. Corticosteroid-induced lymphopenia impedes underlying cellular immunity, which might explain the reactivation of latent herpes and esophageal ulcer formation. Given the rarity of the disease, evidence of treatment is available from case reports only. We found complete resolution of esophageal ulcers after the patient received valacyclovir therapy for 10 days.
Collapse
Affiliation(s)
- Maryam Bilal Haider
- Department of Internal Medicine, Detroit Medical Center/Wayne State University Sinai Grace Hospital, Detroit, Michigan, USA
| | | | | | | | | |
Collapse
|
12
|
The association between bowel resection and the risk of nontyphoidal salmonella infection: a nationwide propensity score-matched cohort study. Sci Rep 2021; 11:1414. [PMID: 33446836 PMCID: PMC7809203 DOI: 10.1038/s41598-021-81224-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 01/05/2021] [Indexed: 11/08/2022] Open
Abstract
Nontyphoidal salmonella (NTS) infection has a high mortality rate. Bowel resections affect gut microbiota and immune function, and the association between bowel resection and NTS infection in human beings has not been addressed. We conducted a nationwide propensity score (PS)-matched cohort study to clarify this association. Data from the Longitudinal Health Insurance Database of Taiwan were used to establish a case-cohort with bowel resections from 2000 to 2013. Informed consent was waived by the Institutional Review Board of China Medical University Hospital (CMUH104-REC2-115) because all personal identifying information used had been de-identified. Each case was matched with one control without any bowel resection according to age, gender, index date, and propensity score (PS). Cumulative incidences of and hazard ratios (HRs) for NTS infection development were analyzed. The incidence of NTS infection was greater in patients with a bowel resection than in the control group (2.97 vs. 1.92 per 10,000 person-years), with an adjusted hazard ratio (aHR) of 1.64 (95% CI = 1.08-2.48). The incidence of NTS infection increased significantly for cases with small bowel resections and right hemicolectomies. Age (31-40 and > 50 years), hypertension, chronic kidney disease, chronic obstructive pulmonary disease, and autoimmune diseases were significant risk factors of NTS infection. Stratification analysis revealed that patients without comorbidities were prone to NTS infection after bowel resections. The increased risk of developing NTS infection could be related to the bowel resection. Specific age groups and comorbidities also contribute to increased risk of NTS infection.
Collapse
|
13
|
Knobloch J, Casjens S, Lehnert M, Yanik SD, Körber S, Lotz A, Rupp J, Raulf M, Zschiesche W, Weiss T, Kronsbein J, Koch A, Brüning T, Pesch B. Exposure to welding fumes suppresses the activity of T-helper cells. ENVIRONMENTAL RESEARCH 2020; 189:109913. [PMID: 32980007 DOI: 10.1016/j.envres.2020.109913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 06/11/2023]
Abstract
Welders have an increased susceptibility to airway infections with non-typeable Haemophilus influenzae (NTHi), which implicates immune defects and might promote pneumonia and chronic obstructive pulmonary disease (COPD). We hypothesized that welding-fume exposure suppresses Th1-lymphocyte activity. Non-effector CD4+ T-cells from blood of 45 welders (n = 23 gas metal arc welders, GMAW; n = 16 tungsten inert gas welders, TIG; n = 6 others) and 25 non-welders were ex vivo activated towards Th1 via polyclonal T-cell receptor stimulation and IL-12 (first activation step) and then stimulated with NTHi extract or lipopolysaccharide (LPS) (second activation step). IFNγ and IL-2 were measured by ELISA. In the first activation step, IFNγ was reduced in welders compared to non-welders and in the GMAW welders with higher concentrations of respirable particles compared to the lower exposed TIG welders. IFNγ was not influenced by tobacco smoking and correlated negatively with welding-fume exposure, respirable manganese, and iron. In the second activation step, NTHi and LPS induced additional IFNγ, which was reduced in current smokers compared to never smokers in welders as well as in non-welders. Analyzing both activation steps together, IFNγ production was lowest in smoking welders and highest in never smoking non-welders. IL-2 was not associated with any of these parameters. Welding-fume exposure might suppress Th1-based immune responses due to effects of particulate matter, which mainly consists of iron and manganese. For responses to NTHi this is strongest in smoking welders because welding fume suppresses T-cell activation towards Th1 and cigarette smoke suppresses the subsequent Th1-response to NTHi via LPS. Both effects are independent from IL-2-regulated T-cell proliferation. This might explain the increased susceptibility to infections and might promote COPD development.
Collapse
Affiliation(s)
- Jürgen Knobloch
- Medical Clinic III for Pneumology Allergology, Sleep-, and Respiratory Medicine, Bergmannsheil University Hospital, Ruhr University Bochum; Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany.
| | - Swaantje Casjens
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA); Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Martin Lehnert
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA); Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Sarah D Yanik
- Medical Clinic III for Pneumology Allergology, Sleep-, and Respiratory Medicine, Bergmannsheil University Hospital, Ruhr University Bochum; Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Sandra Körber
- Medical Clinic III for Pneumology Allergology, Sleep-, and Respiratory Medicine, Bergmannsheil University Hospital, Ruhr University Bochum; Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Anne Lotz
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA); Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Monika Raulf
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA); Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Wolfgang Zschiesche
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA); Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Tobias Weiss
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA); Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Juliane Kronsbein
- Medical Clinic III for Pneumology Allergology, Sleep-, and Respiratory Medicine, Bergmannsheil University Hospital, Ruhr University Bochum; Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Andrea Koch
- Zürcher RehaZentren Davos, Klinikstrasse 6, 7272 Davos-Clavadel, Switzerland; Ludwig-Maximilians-University of Munich (LMU) and DZL (German Center of Lung Science), 81377 Munich, Germany
| | - Thomas Brüning
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA); Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| | - Beate Pesch
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA); Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany
| |
Collapse
|
14
|
Sidletskaya K, Vitkina T, Denisenko Y. The Role of Toll-Like Receptors 2 and 4 in the Pathogenesis of Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2020; 15:1481-1493. [PMID: 32606656 PMCID: PMC7320879 DOI: 10.2147/copd.s249131] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/24/2020] [Indexed: 12/14/2022] Open
Abstract
Currently, chronic obstructive pulmonary disease (COPD) is one of the leading causes of morbidity and mortality worldwide. The determination of immune mechanisms of inflammation in the disease presents an important challenge for fundamental medical research. According to modern views, Toll-like receptors (TLRs), among which TLR2 and TLR4 play a key role, are one of the essential components of inflammatory process in COPD. This review focuses on following aspects: the role of TLR2 and TLR4 in the initiation of inflammatory process in COPD; the mechanisms of influence of various exogenous factors (cigarette smoke, suspended particulate matter, and bacteria) on the expression of TLR2 and TLR4; the contribution of these TLRs to the T-helper (Th) immune response development in COPD, in particular to the Th17 immune response, which contributes to the progression of the disease and therapeutic implications of TLR2 and TLR4 in COPD.
Collapse
Affiliation(s)
- Karolina Sidletskaya
- Vladivostok Branch of Federal State Budgetary Science Institution "Far Eastern Scientific Center of Physiology and Pathology of Respiration" - Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russia
| | - Tatyana Vitkina
- Vladivostok Branch of Federal State Budgetary Science Institution "Far Eastern Scientific Center of Physiology and Pathology of Respiration" - Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russia
| | - Yulia Denisenko
- Vladivostok Branch of Federal State Budgetary Science Institution "Far Eastern Scientific Center of Physiology and Pathology of Respiration" - Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russia
| |
Collapse
|
15
|
Lee FFY, Davidson K, Harris C, McClendon J, Janssen WJ, Alper S. NF-κB mediates lipopolysaccharide-induced alternative pre-mRNA splicing of MyD88 in mouse macrophages. J Biol Chem 2020; 295:6236-6248. [PMID: 32179652 DOI: 10.1074/jbc.ra119.011495] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Although a robust inflammatory response is needed to combat infection, this response must ultimately be terminated to prevent chronic inflammation. One mechanism that terminates inflammatory signaling is the production of alternative mRNA splice forms in the Toll-like receptor (TLR) signaling pathway. Whereas most genes in the TLR pathway encode positive mediators of inflammatory signaling, several, including that encoding the MyD88 signaling adaptor, also produce alternative spliced mRNA isoforms that encode dominant-negative inhibitors of the response. Production of these negatively acting alternatively spliced isoforms is induced by stimulation with the TLR4 agonist lipopolysaccharide (LPS); thus, this alternative pre-mRNA splicing represents a negative feedback loop that terminates TLR signaling and prevents chronic inflammation. In the current study, we investigated the mechanisms regulating the LPS-induced alternative pre-mRNA splicing of the MyD88 transcript in murine macrophages. We found that 1) the induction of the alternatively spliced MyD88 form is due to alternative pre-mRNA splicing and not caused by another RNA regulatory mechanism, 2) MyD88 splicing is regulated by both the MyD88- and TRIF-dependent arms of the TLR signaling pathway, 3) MyD88 splicing is regulated by the NF-κB transcription factor, and 4) NF-κB likely regulates MyD88 alternative pre-mRNA splicing per se rather than regulating splicing indirectly by altering MyD88 transcription. We conclude that alternative splicing of MyD88 may provide a sensitive mechanism that ensures robust termination of inflammation for tissue repair and restoration of normal tissue homeostasis once an infection is controlled.
Collapse
Affiliation(s)
- Frank Fang-Yao Lee
- Department of Biomedical Research, National Jewish Health, Denver, Colorado 80206; Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado 80206; Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Kevin Davidson
- Pulmonary and Critical Care, WakeMed Hospital, Raleigh, North Carolina 27610
| | - Chelsea Harris
- Department of Biomedical Research, National Jewish Health, Denver, Colorado 80206; Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado 80206; Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Jazalle McClendon
- Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - William J Janssen
- Department of Medicine, National Jewish Health, Denver, Colorado 80206; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Scott Alper
- Department of Biomedical Research, National Jewish Health, Denver, Colorado 80206; Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado 80206; Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado 80045.
| |
Collapse
|
16
|
Diminished stimulator of interferon genes production with cigarette smoke-exposure contributes to weakened anti-adenovirus vectors response and destruction of lung in chronic obstructive pulmonary disease model. Exp Cell Res 2019; 384:111545. [PMID: 31470016 DOI: 10.1016/j.yexcr.2019.111545] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/31/2019] [Accepted: 08/03/2019] [Indexed: 12/14/2022]
Abstract
Cigarette smoke (CS) is the primary risk factor for chronic obstructive pulmonary disease (COPD) and dampens antiviral response, which increases viral infections and leads to COPD acute exacerbation (AECOPD). Adenovirus, a nonenveloped DNA virus, is linked with AECOPD, whose DNAs trigger innate immune response via interacting with pattern recognition receptors (PRRs). Stimulator of interferon genes (STING), as a cytosolic DNA sensor, participates in adenovirus-induced interferon β (IFNβ)-dependent antiviral response. STING is involved in various pulmonary diseases, but role of STING in pathogenesis of AECOPD is not well documented. In the present study, we explored relationship between STING and AECOPD induced by recombinant adenovirus vectors (rAdVs) and CS in wild type (WT) and STING-/- mice; and also characterized the inhibition of STING- IFNβ pathway in pulmonary epithelium exposed to cigarette smoke extract (CSE). We found that CS or CSE exposure alone dramatically inhibited STING expression, but not significantly effected IFNβ production. Moreover, CS or CSE-exposed significantly suppressed activation of STING-IFNβ pathway induced by rAdVs and suppressed clearance of rAdVs DNA. Inflammation, fibrosis and emphysema of lung tissues were exaggerated when treated with CS plus rAdVs, which further deteriorate in absences of STING. In A549 cells with knockdown of STING, we also observed enhancing apoptosis related to emphysema, especially CSE and adenovirus vectors in combination. Therefore, STING may play a protective role in preventing the progress of COPD.
Collapse
|
17
|
The lung microbiome dynamics between stability and exacerbation in chronic obstructive pulmonary disease (COPD): Current perspectives. Respir Med 2019; 157:1-6. [PMID: 31450162 DOI: 10.1016/j.rmed.2019.08.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 12/13/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disorder with a course that is not uniform for all COPD patients. Although smoking is considered as the major cause of the disease, persistent or recurrent infections seem to play a particular role in the disease establishment and progression. COPD is characterized by dysregulated immunity that has been associated with the bacterial colonization and infections. The establishment of culture-independent techniques has shed new light on the relationships between bacterial ecology and health status and expanded our knowledge on the lung microbiome. Interactions between the host and lung microbiome result in inflammation and activation of resident cells. The lung microbiome contains populations of symbionts and pathobionts in balance which lose their equilibrium and disturb the balance of T-helper and regulatory T-cells (Treg) upon infection, or lung disease. In COPD factors such as disease severity, exacerbations, degree of inflammation, and type of treatment used (e.g inhaled or systemic steroids and antibiotics) affect the composition of lung microbiota. Recent data indicate that the presence of specific bacterial taxa in the airways has the potential to influence the host immune response and possibly to interfere with disease phenotype. Although, there is a growing body of evidence for the role of microbiome in COPD several unanswered questions still exist for its clinical relevance.
Collapse
|
18
|
Knobloch J, Panek S, Yanik SD, Jamal Jameel K, Bendella Z, Jungck D, Bürger P, Bülthoff E, Struck B, Giannakis N, Rupp J, Kronsbein J, Peters M, Koch A. The monocyte-dependent immune response to bacteria is suppressed in smoking-induced COPD. J Mol Med (Berl) 2019; 97:817-828. [PMID: 30929031 DOI: 10.1007/s00109-019-01778-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 03/04/2019] [Accepted: 03/13/2019] [Indexed: 12/20/2022]
Abstract
COPD patients have an increased susceptibility to bacterial airway infections that can induce exacerbations. In response to infections, circulating monocytes become recruited to the infected tissue and secrete cytokines. We hypothesized that this cytokine response is reduced in COPD. Cultured peripheral blood monocytes of never smokers (NS) and smokers without (S) and with COPD (3 study populations, n = 36-37) were stimulated with extracts of Haemophilus influenzae, Staphylococcus aureus, or Streptococcus pneumoniae or with four different pathogen-associated molecular patterns (PAMPs). Four cytokines and 9 PAMP-related signaling molecules were measured and compared between the groups. Granulocyte-macrophage-colony-stimulating-factor responses to all stimulants were reduced in S and COPD compared to NS. Tumor-necrosis-factor-α responses to all bacterial extracts, peptidoglycan, and lipopolysaccharide were reduced in S and/or COPD. Interleukin-10 responses to S. aureus and lipoteichoic acid were increased in COPD. Correlations to pack-years and lung function were found. The peptidoglycan-receptor NOD2 and the mRNA of the lipopolysaccharide-receptor TLR4 were reduced in S and COPD. Cytokine responses of monocytes to bacteria are suppressed by smoking and in COPD possibly due to NOD2 and TLR4 reduction and/or interleukin-10 increase. This might help to explain the increased susceptibility to bacterial infections. These systemic molecular pathologies might be targets for therapeutic strategies to prevent infection-induced exacerbations. KEY MESSAGES: COPD subjects have an increased susceptibility to bacterial infections. This implies defects in the immune response to bacteria and is critical for disease progression. The cytokine response of monocytes to bacteria is reduced in COPD. This might be due to a reduced NOD2 and TLR4 and an increased IL-10 expression. This can explain the increased susceptibility to infections and help to identify drug targets.
Collapse
Affiliation(s)
- Jürgen Knobloch
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany.
- Department of Pneumology, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany.
| | - Susanne Panek
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Sarah Derya Yanik
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Kaschin Jamal Jameel
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Zeynep Bendella
- Department of Pneumology, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Department of Radiology, University of Bonn Medical Center, Bonn, Germany
| | - David Jungck
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
- Department of Pneumology, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Department of Internal Medicine II, Pneumology, Allergology and Respiratory Medicine, Bethel Teaching Hospital, Berlin, Germany
| | - Paul Bürger
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Eike Bülthoff
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Birte Struck
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Nikolaos Giannakis
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Juliane Kronsbein
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Marcus Peters
- Department of Experimental Pneumology, Ruhr University Bochum, Bochum, Germany
| | - Andrea Koch
- Medical Clinic V, Ludwig-Maximilians-University LMU, Munich, Germany
- German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
19
|
Wu JH, Zhou M, Jin Y, Meng ZJ, Xiong XZ, Sun SW, Miao SY, Han HL, Tao XN. Generation and Immune Regulation of CD4 +CD25 -Foxp3 + T Cells in Chronic Obstructive Pulmonary Disease. Front Immunol 2019; 10:220. [PMID: 30842769 PMCID: PMC6392103 DOI: 10.3389/fimmu.2019.00220] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 01/25/2019] [Indexed: 11/13/2022] Open
Abstract
The imbalance of CD4+Foxp3+ T cell subsets is reportedly involved in abnormal inflammatory immune responses in patients with chronic obstructive pulmonary disease (COPD). However, the possible role of CD4+CD25-Foxp3+ T cells in immune regulation in COPD remains to be investigated. In the current study, distribution and phenotypic characteristics of CD4+CD25-Foxp3+ T cells from peripheral blood were determined by flow cytometry; the origin, immune function and ultimate fate of CD4+CD25-Foxp3+ T cells were further explored in vitro. It was observed that circulating CD4+CD25-Foxp3+ T cells were significantly increased in stable COPD patients (SCOPD) and resembled central memory or effector memory T cells. Compared with peripheral CD4+CD25+Foxp3+ T cells, peripheral CD4+CD25-Foxp3+ T cells showed a lower expression of Foxp3, CTLA-4, HELIOS, and TIGIT, but a higher expression of CD127 and KI-67, suggesting that CD4+CD25-Foxp3+ T cells lost the expression of Tregs-associated molecules following the reduction in CD25. Unexpectedly, our study found that transforming growth factor-β1 (TGFβ1) decreased CD25 expression and played a critical role in the generation of CD4+CD25-Foxp3+ T cells from CD4+CD25+Foxp3+ T cells. Phenotypic analysis further revealed that both inducible and peripheral CD4+CD25-Foxp3+ T cells exhibited the features of activated conventional T cells. Importantly, memory CD4+CD25-Foxp3+ T cells facilitated the proliferation and differentiation of naïve CD4+ T cells into Th17 cells in the presence of IL-1β, IL-6, IL-23, and TGFβ1. Finally, a fraction of CD4+CD25-Foxp3+ T cells, exhibiting instability and plasticity, were converted to Th17 cells when subjected to Th17 cell-polarizing condition. Taken together, we propose that TGFβ1 is responsible for the generation of CD4+CD25-Foxp3+ T cells, and these cells functionally exert an auxiliary effect on Th17 cells generation and might perpetuate chronic inflammation in COPD.
Collapse
Affiliation(s)
- Jiang-Hua Wu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mei Zhou
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao-Ji Meng
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian-Zhi Xiong
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng-Wen Sun
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai-Ying Miao
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Li Han
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Nan Tao
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Su YC, Jalalvand F, Thegerström J, Riesbeck K. The Interplay Between Immune Response and Bacterial Infection in COPD: Focus Upon Non-typeable Haemophilus influenzae. Front Immunol 2018; 9:2530. [PMID: 30455693 PMCID: PMC6230626 DOI: 10.3389/fimmu.2018.02530] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a debilitating respiratory disease and one of the leading causes of morbidity and mortality worldwide. It is characterized by persistent respiratory symptoms and airflow limitation due to abnormalities in the lower airway following consistent exposure to noxious particles or gases. Acute exacerbations of COPD (AECOPD) are characterized by increased cough, purulent sputum production, and dyspnea. The AECOPD is mostly associated with infection caused by common cold viruses or bacteria, or co-infections. Chronic and persistent infection by non-typeable Haemophilus influenzae (NTHi), a Gram-negative coccobacillus, contributes to almost half of the infective exacerbations caused by bacteria. This is supported by reports that NTHi is commonly isolated in the sputum from COPD patients during exacerbations. Persistent colonization of NTHi in the lower airway requires a plethora of phenotypic adaptation and virulent mechanisms that are developed over time to cope with changing environmental pressures in the airway such as host immuno-inflammatory response. Chronic inhalation of noxious irritants in COPD causes a changed balance in the lung microbiome, abnormal inflammatory response, and an impaired airway immune system. These conditions significantly provide an opportunistic platform for NTHi colonization and infection resulting in a "vicious circle." Episodes of large inflammation as the consequences of multiple interactions between airway immune cells and NTHi, accumulatively contribute to COPD exacerbations and may result in worsening of the clinical status. In this review, we discuss in detail the interplay and crosstalk between airway immune residents and NTHi, and their effect in AECOPD for better understanding of NTHi pathogenesis in COPD patients.
Collapse
Affiliation(s)
- Yu-Ching Su
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Farshid Jalalvand
- Department of Biology, Centre for Bacterial Stress Response and Persistence, University of Copenhagen, Copenhagen, Denmark
| | - John Thegerström
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Kristian Riesbeck
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
21
|
Effects of cigarette smoke on immunity, neuroinflammation and multiple sclerosis. J Neuroimmunol 2018; 329:24-34. [PMID: 30361070 DOI: 10.1016/j.jneuroim.2018.10.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 09/30/2018] [Accepted: 10/05/2018] [Indexed: 12/18/2022]
Abstract
Cigarette smoking is the most prominent significant cause of death and morbidity. It is recognised as a risk factor for a number of immune mediated, inflammatory diseases including multiple sclerosis (MS). Here, we review the complex immunological effects of smoking on the immune system, which include enhancement of inflammatory responses with a parallel reduction of some immune defences, resulting in an increased susceptibility to infection and a persistent proinflammatory environment. We discuss the effect of smoking on the susceptibility, clinical course, disability, and mortality in MS, the likely benefits of smoking cessation, and the specific immunological effects of smoking in MS. In conclusion, smoking is an important environmental risk factor for MS occurrence and outcome, and it acts in significant part through immunological mechanisms.
Collapse
|
22
|
Di Padova F, Quesniaux VFJ, Ryffel B. MyD88 as a therapeutic target for inflammatory lung diseases. Expert Opin Ther Targets 2018; 22:401-408. [PMID: 29658361 DOI: 10.1080/14728222.2018.1464139] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Myeloid differentiation primary response protein 88 (MyD88) is a critical adaptor protein involved in Toll-like and IL-1 receptor family signaling controlling innate immune responses and inflammation. Genetic deletion of MyD88 function results in profound suppression of inflammation and reduced resistance of the host to pathogens indicating non-redundant roles of MyD88. The TIR domain is critical for MyD88 dimerization and signaling for TLR and IL-1R family receptor. Areas covered: Emerging evidence suggests that chemical disruption of the TIR domain attenuates cell activation and inhibits in vivo MyD88-dependent inflammation. We review the development of MyD88 dimerization disruptors as a novel therapeutic approach of respiratory diseases with a focus on COPD. Expert opinion: There is a proof of concept that therapeutic targeting of MyD88 is feasible and first preclinical data are highly promising. This opens a great opportunity to treat exacerbations of COPD and other chronic respiratory diseases. However, extensive preclinical investigations and risk analyses are required with carefully evaluation of reduced host resistance and opportunistic infections.
Collapse
Affiliation(s)
| | - Valerie F J Quesniaux
- b Laboratory of experimental and molecular immunology and neurogenetics (INEM) , UMR 7355 CNRS-University of Orleans , Orleans , France
| | - Bernhard Ryffel
- b Laboratory of experimental and molecular immunology and neurogenetics (INEM) , UMR 7355 CNRS-University of Orleans , Orleans , France.,c IDM, Institute of Infectious Disease and Molecular Medicine , University of Cape Town , Cape Town , RSA
| |
Collapse
|
23
|
Ahearn CP, Gallo MC, Murphy TF. Insights on persistent airway infection by non-typeable Haemophilus influenzae in chronic obstructive pulmonary disease. Pathog Dis 2017; 75:3753446. [PMID: 28449098 PMCID: PMC5437125 DOI: 10.1093/femspd/ftx042] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/28/2017] [Indexed: 12/21/2022] Open
Abstract
Non-typeable Haemophilus influenzae (NTHi) is the most common bacterial cause of infection of the lower airways in adults with chronic obstructive pulmonary disease (COPD). Infection of the COPD airways causes acute exacerbations, resulting in substantial morbidity and mortality. NTHi has evolved multiple mechanisms to establish infection in the hostile environment of the COPD airways, allowing the pathogen to persist in the airways for months to years. Persistent infection of the COPD airways contributes to chronic airway inflammation that increases symptoms and accelerates the progressive loss of pulmonary function, which is a hallmark of the disease. Persistence mechanisms of NTHi include the expression of multiple redundant adhesins that mediate binding to host cellular and extracellular matrix components. NTHi evades host immune recognition and clearance by invading host epithelial cells, forming biofilms, altering gene expression and displaying surface antigenic variation. NTHi also binds host serum factors that confer serum resistance. Here we discuss the burden of COPD and the role of NTHi infections in the course of the disease. We provide an overview of NTHi mechanisms of persistence that allow the pathogen to establish a niche in the hostile COPD airways.
Collapse
Affiliation(s)
- Christian P. Ahearn
- Department of Microbiology and Immunology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
- Clinical and Translational Research Center, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Mary C. Gallo
- Department of Microbiology and Immunology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
- Clinical and Translational Research Center, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Timothy F. Murphy
- Department of Microbiology and Immunology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
- Clinical and Translational Research Center, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
- Division of Infectious Disease, Department of Medicine, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| |
Collapse
|
24
|
Tan DBA, Teo TH, Setiawan AM, Ong NE, Zimmermann M, Price P, Kirkham LAS, Moodley YP. Increased CTLA-4 + T cells may contribute to impaired T helper type 1 immune responses in patients with chronic obstructive pulmonary disease. Immunology 2017; 151:219-226. [PMID: 28190271 DOI: 10.1111/imm.12725] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/17/2017] [Accepted: 02/05/2017] [Indexed: 11/29/2022] Open
Abstract
Impaired T helper type 1 (Th1) function is implicated in the susceptibility of patients with chronic obstructive pulmonary disease (COPD) to respiratory infections, which are common causes of acute exacerbations of COPD (AECOPD). To understand the underlying mechanisms, we assessed regulatory T (Treg) cells and the expression of an inhibitory T-cell receptor, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4). Cryopreserved peripheral blood mononuclear cells (PBMC) from patients with AECOPD (n = 17), patients with stable COPD (sCOPD; n = 24) and age-matched healthy non-smoking controls (n = 26) were cultured for 24 hr with brefeldin-A or monensin to detect intracellular or surface CTLA-4 (respectively) by flow cytometry. T cells in PBMC from AECOPD (n = 9), sCOPD (n = 14) and controls (n = 12) were stimulated with anti-CD3 with and without anti-CTLA-4 blocking antibodies and cytokines were quantified by ELISA. Frequencies of circulating T cells expressing intracellular CTLA-4 were higher in sCOPD (P = 0·01), whereas patients with AECOPD had more T cells expressing surface CTLA-4 than healthy controls (P = 0·03). Increased frequencies of surface CTLA-4+ CD4+ T cells and CTLA-4+ Treg cells paralleled increases in plasma soluble tumour necrosis factor receptor-1 levels (r = 0·32, P = 0·01 and r = 0·29, P = 0·02, respectively) in all subjects. Interferon-γ responses to anti-CD3 stimulation were inversely proportional to frequencies of CD4+ T cells expressing intracellular CTLA-4 (r = -0·43, P = 0·01). Moreover, CTLA-4 blockade increased the induction of interferon-γ, tumour necrosis factor-α and interleukin-6 in PBMC stimulated with anti-CD3. Overall, chronic inflammation may expand sub-populations of T cells expressing CTLA-4 in COPD patients and therefore impair T-cell function. CTLA-4 blockade may restore Th1 function in patients with COPD and so aid the clearance of bacterial pathogens responsible for AECOPD.
Collapse
Affiliation(s)
- Dino B A Tan
- Centre for Respiratory Health, School of Medicine & Pharmacology, University of Western Australia, Perth, WA, Australia.,Stem Cell Unit, Institute of Respiratory Health, Perth, WA, Australia
| | - Teck-Hui Teo
- Centre for Respiratory Health, School of Medicine & Pharmacology, University of Western Australia, Perth, WA, Australia
| | - Abdul M Setiawan
- Centre for Respiratory Health, School of Medicine & Pharmacology, University of Western Australia, Perth, WA, Australia
| | - Nathanael E Ong
- Stem Cell Unit, Institute of Respiratory Health, Perth, WA, Australia
| | - Maja Zimmermann
- Stem Cell Unit, Institute of Respiratory Health, Perth, WA, Australia
| | - Patricia Price
- School of Biomedical Science, Curtin University, Perth, WA, Australia
| | - Lea-Ann S Kirkham
- School of Paediatrics and Child Health, University of Western Australia, Perth, WA, Australia.,Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Yuben P Moodley
- Centre for Respiratory Health, School of Medicine & Pharmacology, University of Western Australia, Perth, WA, Australia.,Stem Cell Unit, Institute of Respiratory Health, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Fiona Stanley Hospital, Perth, WA, Australia
| |
Collapse
|
25
|
Peters M, Bonowitz P, Bufe A. A Bioassay for the Determination of Lipopolysaccharides and Lipoproteins. Methods Mol Biol 2017; 1600:143-150. [PMID: 28478565 DOI: 10.1007/978-1-4939-6958-6_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The availability of convenient assays for the detection and quantification of pathogen-associated molecular patterns (PAMPs) is limited. In the case of lipopolysaccharide (LPS) the so-called LAL (limulus amebocyte lysate) test is available, an assay that is performed with the lysate of the blood of the horse shoe crab. Although a sensitive and convenient assay, it lacks specificity, since it is affected by other endotoxins like, for instance, fungal cell walls as well. Here, we describe a bioassay that can be used to detect and quantitate PAMPs in environmental samples. More specific we demonstrate the usage of TLR2 and TLR4/CD14/MD2 transfected Hek293 cells to quantitatively determine bacterial lipoproteins and LPS, respectively. We show the usefulness of these assays to measure LPS in tobacco before and after combustion.
Collapse
Affiliation(s)
- Marcus Peters
- Department of Experimental Pneumology, Ruhr University Bochum, Bochum, Germany.
| | - Petra Bonowitz
- Department of Experimental Pneumology, Ruhr University Bochum, Bochum, Germany
| | - Albrecht Bufe
- Department of Experimental Pneumology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
26
|
Gomez C, Chanez P. The lung microbiome: the perfect culprit for COPD exacerbations? Eur Respir J 2016; 47:1034-6. [PMID: 27037309 DOI: 10.1183/13993003.00270-2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Carine Gomez
- Dept of Respiratory Diseases, Lung Transplant Team APHM, URMITE CNRS IRD UMR 6236, IHU Méditerranée Infection, Aix-Marseille University, Marseille, France
| | - Pascal Chanez
- Dept of Respiratory Diseases APHM, INSERM CNRS U 1067, UMR7333, Aix-Marseille University, Marseille France
| |
Collapse
|
27
|
An evaluation of CD39 as a novel immunoregulatory mechanism invoked by COPD. Hum Immunol 2016; 77:916-920. [PMID: 27430193 DOI: 10.1016/j.humimm.2016.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 06/23/2016] [Accepted: 07/14/2016] [Indexed: 12/23/2022]
Abstract
Acute exacerbations of chronic obstructive pulmonary disease (AECOPD) are characterized by increased pulmonary and systemic inflammation and commonly caused by bacterial and/or viral infection. Little is known about the T-cell dysregulation in AECOPD that promotes these outcomes. CD39 is an ectonucleotidase able to hydrolyse adenosine triphosphate to create adenosine that may inhibit T-cell responses in patients with AECOPD. Here T-cell expression of CD39 measured by flow cytometry was higher in AECOPD patients than stable COPD patients or healthy controls. Higher expression of CD39 was associated with higher levels of plasma soluble tumor necrosis factor receptor but lower interferon-γ (IFNγ) levels in supernatants from staphylococcal enterotoxin-B stimulated peripheral blood mononuclear cells. This links increased expression of CD39 with systemic inflammation and impaired T-cell responses (e.g. IFNγ). The blockade of CD39 pathways may be a novel approach to the control of AECOPD, reducing the dependency on antibiotics.
Collapse
|
28
|
Knobloch J, Yakin Y, Körber S, Grensemann B, Bendella Z, Boyaci N, Gallert WJ, Yanik SD, Jungck D, Koch A. Simvastatin requires activation in accessory cells to modulate T-cell responses in asthma and COPD. Eur J Pharmacol 2016; 788:294-305. [PMID: 27343379 DOI: 10.1016/j.ejphar.2016.06.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 06/17/2016] [Accepted: 06/22/2016] [Indexed: 01/09/2023]
Abstract
T-cell-dependent airway and systemic inflammation triggers the progression of chronic obstructive pulmonary disease (COPD) and asthma. Retrospective studies suggest that simvastatin has anti-inflammatory effects in both diseases but it is unclear, which cell types are targeted. We hypothesized that simvastatin modulates T-cell activity. Circulating CD4+ and CD8+ T-cells, either pure, co-cultured with monocytes or alveolar macrophages (AM) or in peripheral blood mononuclear cells (PBMCs), were ex vivo activated towards Th1/Tc1 or Th2/Tc2 and incubated with simvastatin. Markers for Th1/Tc1 (IFNγ) and Th2/Tc2 (IL-5, IL-13) were measured by ELISA; with PBMCs this was done comparative between 11 healthy never-smokers, 11 current smokers without airflow limitation, 14 smokers with COPD and 11 never-smokers with atopic asthma. T-cell activation induced IFNγ, IL-5 and IL-13 in the presence and absence of accessory cells. Simvastatin did not modulate cytokine expression in pure T-cell fractions. β-hydroxy-simvastatin acid (activated simvastatin) suppressed IL-5 and IL-13 in pure Th2- and Tc2-cells. Simvastatin suppressed IL-5 and IL-13 in Th2-cells co-cultivated with monocytes or AM, which was partially reversed by the carboxylesterase inhibitor benzil. Simvastatin suppressed IL-5 production of Th2/Tc2-cells in PBMCs without differences between cohorts and IL-13 stronger in never-smokers and asthma compared to COPD. Simvastatin induced IFNγ in Th1/Tc1-cells in PBMCs of all cohorts except asthmatics. Simvastatin requires activation in accessory cells likely by carboxylesterase to suppress IL-5 and IL-13 in Th2/Tc2-cells. The effects on Il-13 are partially reduced in COPD. Asthma pathogenesis prevents simvastatin-induced IFNγ up-regulation. Simvastatin has anti-inflammatory effects that could be of interest for asthma therapy.
Collapse
Affiliation(s)
- Jürgen Knobloch
- Department of Internal Medicine III, Bergmannsheil University Hospital, Bochum, Germany; Department of Pneumology, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany.
| | - Yakup Yakin
- Department of Internal Medicine III, Bergmannsheil University Hospital, Bochum, Germany; Department of Pneumology, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
| | - Sandra Körber
- Department of Internal Medicine III, Bergmannsheil University Hospital, Bochum, Germany
| | - Barbara Grensemann
- Department of Pneumology, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
| | - Zeynep Bendella
- Department of Pneumology, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
| | - Niyazi Boyaci
- Department of Pneumology, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
| | - Willem-Jakob Gallert
- Department of Internal Medicine III, Bergmannsheil University Hospital, Bochum, Germany
| | - Sarah Derya Yanik
- Department of Internal Medicine III, Bergmannsheil University Hospital, Bochum, Germany
| | - David Jungck
- Department of Internal Medicine III, Bergmannsheil University Hospital, Bochum, Germany; Department of Pneumology, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
| | - Andrea Koch
- Department of Internal Medicine III, Bergmannsheil University Hospital, Bochum, Germany; Department of Pneumology, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
29
|
Zuo L, Lucas K, Fortuna CA, Chuang CC, Best TM. Molecular Regulation of Toll-like Receptors in Asthma and COPD. Front Physiol 2015; 6:312. [PMID: 26617525 PMCID: PMC4637409 DOI: 10.3389/fphys.2015.00312] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 10/19/2015] [Indexed: 11/13/2022] Open
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) have both been historically associated with significant morbidity and financial burden. These diseases can be induced by several exogenous factors, such as pathogen-associated molecular patterns (PAMPs) (e.g., allergens and microbes). Endogenous factors, including reactive oxygen species, and damage-associated molecular patterns (DAMPs) recognized by toll-like receptors (TLRs), can also result in airway inflammation. Asthma is characterized by the dominant presence of eosinophils, mast cells, and clusters of differentiation (CD)4+ T cells in the airways, while COPD typically results in the excessive formation of neutrophils, macrophages, and CD8+ T cells in the airways. In both asthma and COPD, in the respiratory tract, TLRs are the primary proteins of interest associated with the innate and adaptive immune responses; hence, multiple treatment options targeting TLRs are being explored in an effort to reduce the severity of the symptoms of these disorders. TLR-mediated pathways for both COPD and asthma have their similarities and differences with regards to cell types and the pro-inflammatory cytotoxins present in the airway. Because of the complex TLR cascade, a variety of treatments have been used to minimize airway hypersensitivity and promote bronchodilation. Although unsuccessful at completely alleviating COPD and severe asthmatic symptoms, new studies are focused on possible targets within the TLR cascade to ameliorate airway inflammation.
Collapse
Affiliation(s)
- Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, The Ohio State University Wexner Medical Center, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, Ohio State University Columbus, OH, USA ; Interdisciplinary Biophysics Graduate Program, The Ohio State University Columbus, OH, USA
| | - Kurt Lucas
- Multiphase Chemistry Department, Max Planck Institute for Chemistry Mainz, Germany
| | - Christopher A Fortuna
- Radiologic Sciences and Respiratory Therapy Division, The Ohio State University Wexner Medical Center, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, Ohio State University Columbus, OH, USA
| | - Chia-Chen Chuang
- Radiologic Sciences and Respiratory Therapy Division, The Ohio State University Wexner Medical Center, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, Ohio State University Columbus, OH, USA ; Interdisciplinary Biophysics Graduate Program, The Ohio State University Columbus, OH, USA
| | - Thomas M Best
- Division of Sports Medicine, Department of Family Medicine, Sports Health and Performance Institute, The Ohio State University Wexner Medical Center Columbus, OH, USA
| |
Collapse
|
30
|
Knobloch J, Chikosi SJ, Yanik S, Rupp J, Jungck D, Koch A. A systemic defect in Toll-like receptor 4 signaling increases lipopolysaccharide-induced suppression of IL-2-dependent T-cell proliferation in COPD. Am J Physiol Lung Cell Mol Physiol 2015; 310:L24-39. [PMID: 26498252 DOI: 10.1152/ajplung.00367.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 10/21/2015] [Indexed: 02/06/2023] Open
Abstract
The susceptibility to bacterial infections is increased in chronic obstructive pulmonary disease (COPD). This promotes exacerbations. IL-2 triggers CD4(+)/Th1-cell proliferation, which is important for infection defense. Bacterial endotoxin (LPS) activates MyD88/IRAK and TRIF/IKKε/TBK1 pathways via Toll-like receptor-4 (TLR4) in Th1 cells. Systemic defects in TLR pathways in CD4(+)/Th1 cells cause an impairment of IL-2-dependent immune responses to bacterial infections in COPD. Peripheral blood CD4(+) T cells of never smokers, smokers without COPD, and smokers with COPD (each n = 10) were ex vivo activated towards Th1 and stimulated with LPS. IL-2, MyD88, and TRIF expression, and cell proliferation was analyzed by ELISA, quantitative RT-PCR, and bromodeoxyuridine (BrdU) and trypan blue staining comparative among the cohorts. IL-2 release from activated T cells was increased in COPD vs. smokers and never smokers. LPS reduced IL-2 expression and T-cell proliferation. These effects were increased in COPD vs. never smokers and inversely correlated with FEV1 (%predicted). The MyD88/TRIF ratio was decreased in Th1 cells of COPD. The suppression of IL-2 by LPS was abolished by MyD88/IRAK blockade in never smokers but by TRIF/IKKε/TBK1 blockade in COPD. Moxifloxacin restored IL-2 expression and T-cell proliferation in the presence of LPS by blocking p38 MAPK. The increased IL-2 release from Th1 cells in COPD might contribute to airway inflammation in disease exacerbations. A switch from MyD88/IRAK to TRIF/IKKε/TBK1 signaling amplifies the suppression of IL-2-dependent proliferation of CD4(+) T cells by LPS in COPD. This molecular pathology is of systemic origin, might impair adaptive immune responses, and could explain the increased susceptibility to bacterial infections in COPD. Targeting TLR4-downstream signaling, for example, with moxifloxacin, might reduce exacerbation rates.
Collapse
Affiliation(s)
- Jürgen Knobloch
- Medical Clinic III for Pneumology, Allergology, Sleep and Respiratory Medicine, Bergmannsheil University Hospital, Bochum, Germany; and
| | - Sarah-Jane Chikosi
- Medical Clinic III for Pneumology, Allergology, Sleep and Respiratory Medicine, Bergmannsheil University Hospital, Bochum, Germany; and
| | - Sarah Yanik
- Medical Clinic III for Pneumology, Allergology, Sleep and Respiratory Medicine, Bergmannsheil University Hospital, Bochum, Germany; and
| | - Jan Rupp
- Department of Medical Microbiology and Hygiene, University of Lübeck, Lübeck, Germany
| | - David Jungck
- Medical Clinic III for Pneumology, Allergology, Sleep and Respiratory Medicine, Bergmannsheil University Hospital, Bochum, Germany; and
| | - Andrea Koch
- Medical Clinic III for Pneumology, Allergology, Sleep and Respiratory Medicine, Bergmannsheil University Hospital, Bochum, Germany; and
| |
Collapse
|
31
|
Roberts MEP, Higgs BW, Brohawn P, Pilataxi F, Guo X, Kuziora M, Bowler RP, White WI. CD4+ T-Cell Profiles and Peripheral Blood Ex-Vivo Responses to T-Cell Directed Stimulation Delineate COPD Phenotypes. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2015; 2:268-280. [PMID: 28848849 DOI: 10.15326/jcopdf.2.4.2015.0131] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The heterogeneous clinical phenotypes of chronic obstructive pulmonary disease (COPD) challenge successful drug development. To identify COPD subgroups beyond clinical phenotypes, we interrogated blood immune cell profiles and ex-vivo responses of current and former smokers, with or without COPD, in the longitudinal COPD Genetic Epidemiology study (COPDGene) cohort. CD4+ and CD8+ T cells and monocytes were profiled by flow cytometry. Microarray analysis was performed on the RNA from the aforementioned isolated cells. T-cell directed whole blood ex-vivo stimulation was used to assess functional responses. Blood CD4+ T-cell transcript analysis distinguished patients with COPD from control smokers and also enriched for a subset of patients with COPD that had a history of exacerbations of the disease. Analogous analyses of CD8+ T cells and monocytes failed to discriminate patients with COPD from the control population. Patients with COPD had a diminished cytokine response, compared to control smokers, characterized by low levels of granulocyte-monocyte colony stimulation factor (GM-CSF), interferon gamma (IFN-ɣ), interleukin one-alpha (IL-1α), tumor necrosis factor-alpha (TNF-α) and tumor necrosis factor-beta (TNF-β) secreted in response to T-cell directed ex-vivo stimulation. This cytokine response associated with baseline disease severity (forced expiratory volume in 1 second [FEV1]% predicted), rapidly declining lung function, and emphysema. Our observations indicate that COPD phenotypes can be further differentiated based on blood CD4+ T-cell profiles and resultant immune responses, suggesting a role for these cells in COPD pathophysiology.
Collapse
Affiliation(s)
| | - Brandon W Higgs
- MedImmune, Department of Translational Sciences, Gaithersburg, Maryland
| | - Philip Brohawn
- MedImmune, Department of Translational Sciences, Gaithersburg, Maryland
| | - Fernanda Pilataxi
- MedImmune, Department of Translational Sciences, Gaithersburg, Maryland
| | - Xiang Guo
- MedImmune, Department of Translational Sciences, Gaithersburg, Maryland
| | - Michael Kuziora
- MedImmune, Department of Translational Sciences, Gaithersburg, Maryland
| | - Russell P Bowler
- National Jewish Health, Department of Medicine, Denver, Colorado
| | - Wendy I White
- MedImmune, Department of Translational Sciences, Gaithersburg, Maryland
| |
Collapse
|
32
|
King PT, Sharma R. The Lung Immune Response to Nontypeable Haemophilus influenzae (Lung Immunity to NTHi). J Immunol Res 2015; 2015:706376. [PMID: 26114124 PMCID: PMC4465770 DOI: 10.1155/2015/706376] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/12/2015] [Accepted: 05/13/2015] [Indexed: 11/18/2022] Open
Abstract
Haemophilus influenzae is divided into typeable or nontypeable strains based on the presence or absence of a polysaccharide capsule. The typeable strains (such as type b) are an important cause of systemic infection, whilst the nontypeable strains (designated as NTHi) are predominantly respiratory mucosal pathogens. NTHi is present as part of the normal microbiome in the nasopharynx, from where it may spread down to the lower respiratory tract. In this context it is no longer a commensal and becomes an important respiratory pathogen associated with a range of common conditions including bronchitis, bronchiectasis, pneumonia, and particularly chronic obstructive pulmonary disease. NTHi induces a strong inflammatory response in the respiratory tract with activation of immune responses, which often fail to clear the bacteria from the lung. This results in recurrent/persistent infection and chronic inflammation with consequent lung pathology. This review will summarise the current literature about the lung immune response to nontypeable Haemophilus influenzae, a topic that has important implications for patient management.
Collapse
Affiliation(s)
- Paul T. King
- Monash Lung and Sleep, Monash Medical Centre, Melbourne, VIC 3168, Australia
- Monash University Department of Medicine, Monash Medical Centre, Melbourne, VIC 3168, Australia
| | - Roleen Sharma
- Monash Lung and Sleep, Monash Medical Centre, Melbourne, VIC 3168, Australia
- Monash University Department of Medicine, Monash Medical Centre, Melbourne, VIC 3168, Australia
| |
Collapse
|
33
|
Bracke KR, Brusselle GG. Chronic Obstructive Pulmonary Disease. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00097-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
34
|
Xu D, Zhao M, Song Y, Song J, Huang Y, Wang J. Novel insights in preventing Gram-negative bacterial infection in cirrhotic patients: review on the effects of GM-CSF in maintaining homeostasis of the immune system. Hepatol Int 2014; 9:28-34. [DOI: 10.1007/s12072-014-9588-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 10/20/2014] [Indexed: 02/08/2023]
|
35
|
Understanding nontypeable Haemophilus influenzae and chronic obstructive pulmonary disease. Curr Opin Pulm Med 2014; 20:159-64. [PMID: 24441573 DOI: 10.1097/mcp.0000000000000023] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW Bacteria are frequently implicated in acute exacerbations of chronic obstructive pulmonary disease (COPD), but their influence on airway inflammation remains unclear. This review will focus on nontypeable Haemophilus influenzae (NTHi), its impact on host immune responses, and the potential for vaccination strategies in COPD. RECENT FINDINGS NTHi is associated with impaired immune function in patients with COPD. Features of the bacterium itself potentiate its ability to colonize the lower airways. An imbalance between bacterial load and host immunity may lend itself to the development of exacerbations. Oral immunotherapy may be a method of augmenting the host immune response and could provide protection from exacerbations. SUMMARY A causal link between NTHi and COPD exacerbations has not been clearly established. However, colonization of the lower airways by NTHi likely plays a significant role in the inflammatory state of COPD.
Collapse
|
36
|
Finney LJ, Ritchie A, Pollard E, Johnston SL, Mallia P. Lower airway colonization and inflammatory response in COPD: a focus on Haemophilus influenzae. Int J Chron Obstruct Pulmon Dis 2014; 9:1119-32. [PMID: 25342897 DOI: 10.2147/copd.s54477] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Bacterial infection of the lower respiratory tract in chronic obstructive pulmonary disease (COPD) patients is common both in stable patients and during acute exacerbations. The most frequent bacteria detected in COPD patients is Haemophilus influenzae, and it appears this organism is uniquely adapted to exploit immune deficiencies associated with COPD and to establish persistent infection in the lower respiratory tract. The presence of bacteria in the lower respiratory tract in stable COPD is termed colonization; however, there is increasing evidence that this is not an innocuous phenomenon but is associated with airway inflammation, increased symptoms, and increased risk for exacerbations. In this review, we discuss host immunity that offers protection against H. influenzae and how disturbance of these mechanisms, combined with pathogen mechanisms of immune evasion, promote persistence of H. influenzae in the lower airways in COPD. In addition, we examine the role of H. influenzae in COPD exacerbations, as well as interactions between H. influenzae and respiratory virus infections, and review the role of treatments and their effect on COPD outcomes. This review focuses predominantly on data derived from human studies but will refer to animal studies where they contribute to understanding the disease in humans.
Collapse
Affiliation(s)
- Lydia J Finney
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Andrew Ritchie
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | | | - Sebastian L Johnston
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Patrick Mallia
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| |
Collapse
|
37
|
Leung JM, Liu JC, Mtambo A, Ngan D, Nashta N, Guillemi S, Harris M, Lima VD, Mattman A, Shaipanich T, Raju R, Hague C, Leipsic JA, Sin DD, Montaner JS, Man SP. The determinants of poor respiratory health status in adults living with human immunodeficiency virus infection. AIDS Patient Care STDS 2014; 28:240-7. [PMID: 24742270 DOI: 10.1089/apc.2013.0373] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The increased longevity afforded by combination antiretroviral therapy in developed countries has led to an increased concern regarding senescence-related diseases in patients with human immunodeficiency virus (HIV) infection. Previous epidemiologic analyses have demonstrated an increased risk of chronic obstructive pulmonary disease, as well as a significant burden of respiratory symptoms in HIV-infected patients. We performed the St. George's Respiratory Questionnaire (SGRQ) in 199 HIV-positive men, and determined the predominant factors contributing to poor respiratory-related health status. In univariate analyses, worse SGRQ scores were associated with respiratory-related variables such as greater smoking pack-year history (p=0.028), lower forced expiratory volume in 1 second (FEV1) (p<0.001), and worse emphysema severity as quantified by computed tomographic imaging (p=0.017). In addition, HIV-specific variables, such as a history of plasma viral load >100,000 copies/mL (p=0.043), lower nadir CD4 cell count (p=0.040), and current CD4 cell count ≤350 cells/μL (p=0.005), as well as elevated levels of inflammatory markers, specifically plasma interleukin (IL)-6 (p=0.002) and alpha-1 antitrypsin (p=0.005) were also associated with worse SGRQ scores. In a multiple regression model, FEV1, current CD4 count ≤350 cells/μL, and IL-6 levels remained significant contributors to reduced respiratory-related health status. HIV disease activity as measured by HIV-related immunosuppression in conjunction with the triggering of key inflammatory pathways may be important determinants of worse respiratory health status among HIV-infected individuals. Limitations of this analysis include the absence of available echocardiograms, diffusion capacity and lung volume testing, and an all-male cohort due to the demographics of the clinic population.
Collapse
Affiliation(s)
| | - Joseph C. Liu
- UBC James Hogg Research Centre, Vancouver, BC, Canada
| | - Andy Mtambo
- AIDS Research Program, St. Paul's Hospital, Vancouver, BC, Canada
| | - David Ngan
- UBC James Hogg Research Centre, Vancouver, BC, Canada
| | - Negar Nashta
- AIDS Research Program, St. Paul's Hospital, Vancouver, BC, Canada
| | - Silvia Guillemi
- AIDS Research Program, St. Paul's Hospital, Vancouver, BC, Canada
- Department of Family Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Division of HIV/AIDS, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Marianne Harris
- AIDS Research Program, St. Paul's Hospital, Vancouver, BC, Canada
- Department of Family Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Division of HIV/AIDS, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Viviane D. Lima
- BC Centre for Excellence in HIV/AIDS, St. Paul's Hospital, Vancouver, BC, Canada
| | - Andre Mattman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tawimas Shaipanich
- UBC Department of Medicine and Division of Respiratory Medicine, St. Paul's Hospital, Vancouver, BC, Canada
| | - Rekha Raju
- Department of Radiology and Diagnostic Imaging, St. Paul's Hospital, Vancouver, BC, Canada
| | - Cameron Hague
- Department of Radiology and Diagnostic Imaging, St. Paul's Hospital, Vancouver, BC, Canada
| | - Jonathon A. Leipsic
- Department of Radiology and Diagnostic Imaging, St. Paul's Hospital, Vancouver, BC, Canada
| | - Don D. Sin
- UBC James Hogg Research Centre, Vancouver, BC, Canada
- UBC Department of Medicine and Division of Respiratory Medicine, St. Paul's Hospital, Vancouver, BC, Canada
| | - Julio S. Montaner
- BC Centre for Excellence in HIV/AIDS, St. Paul's Hospital, Vancouver, BC, Canada
| | - S.F. Paul Man
- UBC James Hogg Research Centre, Vancouver, BC, Canada
- UBC Department of Medicine and Division of Respiratory Medicine, St. Paul's Hospital, Vancouver, BC, Canada
| |
Collapse
|
38
|
The proportion and function of peripheral myeloid-derived suppressor cells do not correlate with systemic inflammation in chronic obstructive pulmonary disease. Hum Immunol 2013; 75:5-9. [PMID: 24090682 DOI: 10.1016/j.humimm.2013.09.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 09/17/2013] [Accepted: 09/20/2013] [Indexed: 11/21/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) have been implicated in the regulation of chronic inflammation. Chronic obstructive pulmonary disease (COPD) involves persistent inflammation, but the role of MDSC has not been explored. Here, proportions of MDSC (CD14(-)HLA-DR(-)CD33(+)CD11b(+) cells) were quantified in peripheral blood mononuclear cells (PBMC) isolated from patients with 'stable' COPD (n = 12), smokers with no evidence of COPD (n = 11) and healthy non-smokers (n = 11). The proportions of MDSC were similar in all groups. MDSC function was assessed by comparing T-cell and cytokine responses of whole and MDSC-depleted PBMC stimulated with Staphylococcus enterotoxin-B (SEB). Depletion of MDSC did not enhance CD4(+) or CD8(+) T-cell activation and proliferation, or alter IFNγ and IL-17 production in response to SEB. However production of TGFβ decreased after depletion of MDSC, so MDSC may be a source of this cytokine. In conclusion, COPD was not associated with perturbations in the proportion or function of MDSC in peripheral blood.
Collapse
|
39
|
Knobloch J, Wahl C, Feldmann M, Jungck D, Strauch J, Stoelben E, Koch A. Resveratrol attenuates the release of inflammatory cytokines from human bronchial smooth muscle cells exposed to lipoteichoic acid in chronic obstructive pulmonary disease. Basic Clin Pharmacol Toxicol 2013; 114:202-9. [PMID: 23981542 DOI: 10.1111/bcpt.12129] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Accepted: 08/12/2013] [Indexed: 12/21/2022]
Abstract
During bacterial infections, pathogen-associated molecular patterns (PAMPs) induce cytokine/chemokine release in immunoactive cells. This increases corticosteroid-resistant airway inflammation in chronic obstructive pulmonary disease (COPD) and leads to exacerbations. Anti-inflammatory therapies other than corticosteroids are required and resveratrol is currently under discussion. Resveratrol is an activator of sirtuins, which are class III histone deacetylases (HDACs). We suggested that human airway smooth muscle cells (HASMCs) release COPD-associated cytokines/chemokines in response to lipoteichoic acid (LTA), a major PAMP of gram-positive bacteria and that resveratrol is superior to the corticosteroid dexamethasone in suppressing these cytokines/chemokines. Cultivated HASMCs of patients with COPD were pre-incubated with resveratrol or dexamethasone before stimulation with LTA. CCL2, GM-CSF, IL-6 and IL-8 were analysed in culture supernatants by enzyme-linked immunosorbent assay. Drug effects were investigated in the absence and presence of trichostatin A (TSA), an inhibitor of class I/II HDACs, and EX527, an inhibitor of the sirtuin SIRT1. LTA induced robust cytokine/chemokine release. Resveratrol was superior to dexamethasone in reducing CCL-2, IL-6 and IL-8 in LTA-exposed HASMCs of patients with COPD. Both drugs were equally effective in reducing GM-CSF. Resveratrol effects were partially reversed by EX527 but not by TSA. Dexamethasone effects were partially reversed by TSA but not by EX527. We conclude that HASMCs contribute to the increase in airway inflammation in COPD exacerbations caused by gram-positive bacterial infections. Our data suggest resveratrol as an alternative anti-inflammatory therapy in infection-induced COPD exacerbations. Resveratrol and corticosteroids suppress cytokine/chemokine expression through activation of SIRT1 or interaction with class I/II HDACs, respectively, in HASMCs.
Collapse
Affiliation(s)
- Jürgen Knobloch
- Department of Internal Medicine III for Pneumology, Allergology, Sleep- and Respiratory Medicine, University Hospital Bergmannsheil, Bochum, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Knobloch J, Lin Y, Konradi J, Jungck D, Behr J, Strauch J, Stoelben E, Koch A. Inflammatory responses of airway smooth muscle cells and effects of endothelin receptor antagonism. Am J Respir Cell Mol Biol 2013; 49:114-27. [PMID: 23590298 DOI: 10.1165/rcmb.2012-0287oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Endothelin receptor antagonists (ETRAs), authorized for pulmonary hypertension, have failed to prove their utility in chronic lung diseases with corticosteroid-resistant airway inflammation when applied at late disease stages with emphysema/fibrosis. Earlier administration might prove effective by targeting the interaction between airway inflammation and tissue remodeling. We hypothesized that human airway smooth muscle cells (HASMCs) participate in linking inflammation with remodeling and that associated genes become differentially suppressed by ambrisentan (A-receptor selective ETRA) and bosentan (nonselective/dual ETRA). Inflammatory responses of ex vivo-cultivated HASMCs to TNF-α were investigated by whole-genome microarray analyses. qRT-PCR and ELISA were used to test inflammatory and remodeling genes for sensitivity to bosentan and ambrisentan and to investigate differential sensitivities mechanistically. ETRA and corticosteroid effects were compared in HASMCs from patients with chronic obstructive pulmonary disease. TNF-α induced the expression of 18 cytokines/chemokines and five tissue remodeling genes involved in severe, corticosteroid-insensitive asthma, chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and/or pulmonary hypertension. Thirteen cytokines/chemokines, MMP13, and WISP1 were suppressed by ETRAs. Eight genes had differential sensitivity to bosentan and ambrisentan depending on the endothelin-B receptor impact on transcriptional regulation and mRNA stabilization. Chemokine (C-C motif) ligands 2 and 5, granulocyte macrophage colony-stimulating factor, and MMP13 had increased sensitivity to bosentan or bosentan/dexamethasone combination versus dexamethasone alone. Suppression of cytokine and remodeling gene expression by ETRAs was confirmed in TNF-α-activated human bronchial epithelial cells. HASMCs and human bronchial epithelial cells participate in the interaction of inflammation and tissue remodeling. This interaction is targeted differentially by selective and nonselective ETRAs, which could be used in therapies of chronic lung diseases with corticosteroid-resistant airway inflammation at early disease stages to attenuate inflammation-induced airway remodeling.
Collapse
Affiliation(s)
- Jürgen Knobloch
- Department of Internal Medicine III, University Hospital Bergmannsheil, Bochum, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Jetté-Côté I, Ouellette D, Béliveau C, Mitchell A. Total dysphagia after short course of systemic corticotherapy: Herpes simplex virus esophagitis. World J Gastroenterol 2013; 19:5178-5181. [PMID: 23964155 PMCID: PMC3746393 DOI: 10.3748/wjg.v19.i31.5178] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 04/27/2013] [Accepted: 06/10/2013] [Indexed: 02/06/2023] Open
Abstract
A 72 year-old female developed a herpetic esophagitis after 3 d of oral corticotherapy for an acute exacerbation of chronic obstructive pulmonary disease, presenting as odynophagia and total dysphagia. Biospies were taken during a first esophagogastroduodenoscopy (EGD) and the patient was referred to the thoracic surgery service with a presumptive diagnosis of esophageal cancer. A second EGD was planned for dilatation, but by that time the stenosis was completely resolved. The biopsies taken during the first EGD revealed multiple herpetic viral inclusions and ulcerations without any dysplasia or neoplasia. In front of a severe esophageal stenosis, one must still exclude the usual differential diagnosis peptic stenosis and cancer. Visualization of endoscopic lesions can suggest the diagnosis but must be promptly confirmed by biopsy, viral culture or polymerase chain reaction. Although immune systemic effects of corticotherapy are well known and herpetic esophagitis occurs most frequently in immunocompromised individuals, this case emphasizes the importance of clinical awareness concerning short courses of corticotherapy for immunocompetent individuals. This article discusses the reactivation process of herpetic infection in this context and addresses its diagnostic and therapeutic issues.
Collapse
|
42
|
Infectious Mechanisms Regulating Susceptibility to Acute Exacerbations of COPD. SMOKING AND LUNG INFLAMMATION 2013. [PMCID: PMC7115011 DOI: 10.1007/978-1-4614-7351-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Acute exacerbations of COPD (AECOPD) are defined by clinical criteria, outlined in the Global Initiative for Chronic Obstructive Lung Disease (GOLD) guidelines [1]. These include an acute increase in one or more of the following cardinal symptoms, beyond day to day variability: dyspnea, increased frequency or severity of cough and increased volume or change in character of sputum, which represent an acute increase in airway inflammation. The role of infection in the pathogenesis of COPD, acute exacerbation and disease progression has been a clinical and research question for many years, and the pendulum has swung from infection as a major cause of acute exacerbation and COPD (British Hypothesis) [2], to infection as an unrelated epiphomenon in acute exacerbation [3–5], and back again to infection as integral in the development of AECOPD and likely an important contributor to COPD progression [6–19]. Upwards of 80 % of AECOPD are driven by infectious stimuli, with 40–50 % associated with bacterial infection and 30–50 % associated with acute viral infection, with some exacerbations having dual bacterial and viral causation [20]. Much of the advancement in our understanding of the role of infection is AECOPD is due to the advancement of clinical and research tools that have allowed researchers to accurately characterize the microbial pathogens, and better understand the host-pathogen interactions (Table 1).
Collapse
|
43
|
Abstract
Evidence has increasingly shown that the lungs are a major site of immune regulation. A robust and highly regulated immune response in the lung protects the host from pathogen infection, whereas an inefficient or deleterious response can lead to various pulmonary diseases. Many cell types, such as epithelial cells, dendritic cells, macrophages, neutrophils, eosinophils, and B and T lymphocytes, contribute to lung immunity. This review focuses on the recent advances in understanding how T lymphocytes mediate pulmonary host defenses against bacterial, viral, and fungal pathogens.
Collapse
Affiliation(s)
- Kong Chen
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15201, USA
| | | |
Collapse
|
44
|
Knobloch J, Feldmann M, Wahl C, Jungck D, Behr J, Stoelben E, Koch A. Endothelin Receptor Antagonists Attenuate the Inflammatory Response of Human Pulmonary Vascular Smooth Muscle Cells to Bacterial Endotoxin. J Pharmacol Exp Ther 2013; 346:290-9. [DOI: 10.1124/jpet.112.202358] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
45
|
Lee SW, Kim DR, Kim TJ, Paik JH, Chung JH, Jheon S, Huh JW, Lee JH, Lee CT. The association of down-regulated toll-like receptor 4 expression with airflow limitation and emphysema in smokers. Respir Res 2012; 13:106. [PMID: 23170858 PMCID: PMC3546871 DOI: 10.1186/1465-9921-13-106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Accepted: 10/29/2012] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND An association between innate immunity including Toll-like receptors (TLRs) and COPD is reported recently; TLR4 deficiency in lung can cause emphysema in animals, which is not evident in humans. We analyzed the association of TLR4 expression, airflow limitation and emphysema in smokers. METHODS We enrolled patients of ≥40years old with smoking histories of ≥10 pack-years and who had undergone lung resection. We measured TLR4 expression in lung lysates. The severity of emphysema was evaluated on computed tomography. TLR4 expression was also evaluated immunohistochemically. RESULTS In total, 53 patients were enrolled. Forced expiratory volume in one second per forced vital capacity (FEV1/FVC) increased (P=0.03) and emphysema score decreased (P=0.01) as TLR4 expression increased. These were still significant, in multiple regression analysis including sex, age, tuberculosis history, smoking history and inhaled corticosteroid (ICS) usage. We also classified patients as high, intermediate, and low expressers according to TLR4 expression. Although no differences in age, gender, tuberculosis, or smoking history were observed among the groups, emphysema severity increased significantly (P = 0.02) and FEV1/FVC decreased significantly (P = 0.006) in TLR4 low expresser. The difference in TLR4 expression based on immunohistochemistry was most prominent in bronchial and alveolar epithelial cells. CONCLUSION Down-regulated TLR4 expression in lung was associated with emphysema and airflow limitation in smokers.
Collapse
Affiliation(s)
- Sei Won Lee
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Beasley V, Joshi PV, Singanayagam A, Molyneaux PL, Johnston SL, Mallia P. Lung microbiology and exacerbations in COPD. Int J Chron Obstruct Pulmon Dis 2012; 7:555-69. [PMID: 22969296 PMCID: PMC3437812 DOI: 10.2147/copd.s28286] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the most common chronic respiratory condition in adults and is characterized by progressive airflow limitation that is not fully reversible. The main etiological agents linked with COPD are cigarette smoking and biomass exposure but respiratory infection is believed to play a major role in the pathogenesis of both stable COPD and in acute exacerbations. Acute exacerbations are associated with more rapid decline in lung function and impaired quality of life and are the major causes of morbidity and mortality in COPD. Preventing exacerbations is a major therapeutic goal but currently available treatments for exacerbations are not very effective. Historically, bacteria were considered the main infective cause of exacerbations but with the development of new diagnostic techniques, respiratory viruses are also frequently detected in COPD exacerbations. This article aims to provide a state-of-the art review of current knowledge regarding the role of infection in COPD, highlight the areas of ongoing debate and controversy, and outline emerging technologies and therapies that will influence future diagnostic and therapeutic pathways in COPD.
Collapse
|
47
|
Agustí A, Barnes PJ. Update in chronic obstructive pulmonary disease 2011. Am J Respir Crit Care Med 2012; 185:1171-6. [PMID: 22661523 DOI: 10.1164/rccm.201203-0505up] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Alvar Agustí
- Institut del Torax, Hospital Clinic, Villarroel 170, Barcelona, Spain.
| | | |
Collapse
|
48
|
Kheradmand F, Shan M, Xu C, Corry DB. Autoimmunity in chronic obstructive pulmonary disease: clinical and experimental evidence. Expert Rev Clin Immunol 2012; 8:285-92. [PMID: 22390492 DOI: 10.1586/eci.12.7] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Over the past few decades, neutrophils and macrophages had co-occupied center stage as the critical innate immune cells underlying the pathobiology of cigarette smoke-induced chronic obstructive pulmonary disease and lung parenchymal destruction (i.e., emphysema). While chronic exposure to smoke facilitates the recruitment of innate immune cells into the lung, a clear role for adaptive immunity in emphysema has emerged. Evidence from human studies specifically point to a role for recruitment and activation of pathogenic lymphocytes and lung antigen-presenting cells in emphysema; similarly, animal models have confirmed a significant role for autoimumnity in progressive smoke-induced emphysema. Increased numbers of activated antigen-presenting cells, Th1 and Th17 cells, have been associated with smoke-induced lung inflammation and production of the canonical cytokines of these cells, IFN-γ and IL-17, correlates with disease severity. These exciting new breakthroughs could open new avenues for developing effective new therapies for smoke-induced emphysema.
Collapse
Affiliation(s)
- Farrah Kheradmand
- Department of Medicine, Sections of Pulmonary and Critical Care, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | |
Collapse
|
49
|
King P. Haemophilus influenzae and the lung (Haemophilus and the lung). Clin Transl Med 2012; 1:10. [PMID: 23369277 PMCID: PMC3567431 DOI: 10.1186/2001-1326-1-10] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 06/14/2012] [Indexed: 12/26/2022] Open
Abstract
Haemophilus influenzae is present as a commensal organism in the nasopharynx of most healthy adults from where it can spread to cause both systemic and respiratory tract infection. This bacterium is divided into typeable forms (such as type b) or nontypeable forms based on the presence or absence of a tough polysaccharide capsule. Respiratory disease is predominantly caused by the nontypeable forms (NTHi). Haemophilus influenzae has evolved a number of strategies to evade the host defense including the ability to invade into local tissue. Pathogenic properties of this bacterium as well as defects in host defense may result in the spread of this bacterium from the upper airway to the bronchi of the lung. This can result in airway inflammation and colonization particularly in chronic obstructive pulmonary disease. Treatment of respiratory tract infection with Haemophilus influenzae is often only partially successful with ongoing infection and inflammation. Improvement in patient outcome will be dependent on a better understanding of the pathogenesis and host immune response to this bacterium.
Collapse
Affiliation(s)
- Paul King
- Department of Respiratory and Sleep Medicine and Department of Medicine, Monash University, Monash Medical Centre, 246 Clayton Rd, Clayton, Melbourne, 3168, Australia.
| |
Collapse
|
50
|
Mat Z, Grensemann B, Yakin Y, Knobloch J, Koch A. Effect of lipoteichoic acid on IL-2 and IL-5 release from T lymphocytes in asthma and COPD. Int Immunopharmacol 2012; 13:284-91. [PMID: 22542695 DOI: 10.1016/j.intimp.2012.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 03/19/2012] [Accepted: 04/11/2012] [Indexed: 12/28/2022]
Abstract
Susceptibility to infections with gram-positive bacteria, which are an important trigger of exacerbations, is increased in COPD and asthma. Unraveling the underlying mechanisms may help developing therapeutic strategies to reduce exacerbation rates. The aim of this study was to evaluate the effects of lipoteichoic acid (LTA), a danger signal from gram-positive bacteria, on T cell cytokines related to bacterial infection defense in COPD and asthma. T cell populations within peripheral blood mononuclear cells (PBMCs) were ex-vivo activated towards T(H)2/T(C)2 subtypes and subsequently stimulated with LTA. IL-2 and IL-5 concentrations in cell culture supernatants were measured by ELISA comparative between non-smokers (NS), current smokers without airflow limitation (S), smokers with moderate to severe COPD and mild to moderate asthmatics (A) (each n=10). IL-2 and IL-5 baseline levels were without differences between the cohorts. After T cell activation, IL-2 and IL-5 releases were increased in all cohorts, however, for IL-2 this increase was significantly higher in S and by trend in COPD compared to the other groups. LTA time-dependently suppressed IL-2 release in NS, S and COPD but not in A. LTA reduced IL-5 release in COPD and A but not in NS and S. Summarized, LTA reduces T(H)2/T(C)2 cytokines indicating immunosuppressive effects, which are dysregulated in COPD and asthma. This implies a misguided response to gram-positive bacterial infections, which might help to explain the increased susceptibility to bacterial infections in COPD and asthma.
Collapse
Affiliation(s)
- Zeynep Mat
- Department of Pneumology, Medical Clinic III for Internal Medicine, University of Cologne, Germany
| | | | | | | | | |
Collapse
|