1
|
Gong KQ, Brune JE, Guo X, Manicone AM. MAP2K1 dampens cigarette smoke-induced inflammation via suppression of type I interferon pathway activation. Am J Physiol Lung Cell Mol Physiol 2024; 327:L740-L748. [PMID: 39316676 DOI: 10.1152/ajplung.00080.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD), comprised of chronic bronchitis and emphysema, is a leading cause of morbidity and mortality worldwide. Mitogen-activated protein 2 kinase (MAP2K) pathway activation is present in COPD lung tissue and a genetic polymorphism in Map2k1 associates with FEV1 decline in COPD, suggesting it may contribute to disease pathogenesis. To test the functional contribution of Map2k1 in cigarette smoke (CS)-induced lung inflammation, we used a short-term CS exposure model in mice deficient in myeloid Map2k1 (LysmCre+Mek1fl) and wild-type mice (Mek1fl). Mice deficient in myeloid Map2k1 had enhanced CS-induced lung inflammation characterized by increased neutrophil recruitment, vascular leak, augmented expression of elastolytic matrix metalloproteinases, and increased type I interferon-stimulated gene expression. The augmented neutrophilic inflammatory response could be abrogated by IFNAR1 blockade. These findings indicate that myeloid Map2k1 regulates the immune response to CS via inhibition of the type I interferon pathway. Overall, these results suggest that Map2k1 is a critical determinant in modulating the severity of CS-induced lung inflammation and its expression is protective.NEW & NOTEWORTHY Activation of the mitogen-activated protein kinases (MAPK)-ERK1/2 pathway is present in COPD lung tissue compared with healthy lungs. Our study using mice deficient in myeloid Map2k1 reveals that Map2k1 is a critical determinant in modulating the severity of CS-induced lung inflammation via suppression of type I interferon responses, and its expression is protective.
Collapse
Affiliation(s)
- Ke-Qin Gong
- Center for Lung Biology, University of Washington, Seattle, Washington, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington, United States
| | - Jourdan E Brune
- Center for Lung Biology, University of Washington, Seattle, Washington, United States
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States
| | - Xiaoyun Guo
- Center for Lung Biology, University of Washington, Seattle, Washington, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington, United States
| | - Anne M Manicone
- Center for Lung Biology, University of Washington, Seattle, Washington, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington, United States
| |
Collapse
|
2
|
Nicola T, Wenger N, Xu X, Evans M, Qiao L, Rezonzew G, Yang Y, Jilling T, Margaroli C, Genschmer K, Willis K, Ambalavanan N, Blalock JE, Gaggar A, Lal CV. A lactobacilli-based inhaled live biotherapeutic product attenuates pulmonary neutrophilic inflammation. Nat Commun 2024; 15:7113. [PMID: 39160214 PMCID: PMC11333600 DOI: 10.1038/s41467-024-51169-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/31/2024] [Indexed: 08/21/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease of prematurity. Exposure to noxious stimuli such as hyperoxia, volutrauma, and infection in infancy can have long-reaching impacts on lung health and predispose towards the development of conditions such as chronic obstructive pulmonary disease (COPD) in adulthood. BPD and COPD are both marked by lung tissue degradation, neutrophil influx, and decreased lung function. Both diseases also express a change in microbial signature characterized by firmicute depletion. However, the relationship between pulmonary bacteria and the mechanisms of downstream disease development has yet to be elucidated. We hypothesized that murine models of BPD would show heightened acetylated proline-glycine-proline (Ac-PGP) pathway and neutrophil activity, and through gain- and loss-of-function studies we show that Ac-PGP plays a critical role in driving BPD development. We further test a inhaled live biotherapeutic (LBP) using active Lactobacillus strains in in vitro and in vivo models of BPD and COPD. The Lactobacillus-based LBP is effective in improving lung structure and function, mitigating neutrophil influx, and reducing a broad swath of pro-inflammatory markers in these models of chronic pulmonary disease via the MMP-9/PGP (matrix metalloproteinase/proline-glycine-proline) pathway. Inhaled LBPs show promise in addressing common pathways of disease progression that in the future can be targeted in a variety of chronic lung diseases.
Collapse
Grants
- R01HL156275 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R35HL166433 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R35HL135710 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K08 HL141652 NHLBI NIH HHS
- R44HL164156 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL156275 NHLBI NIH HHS
- R35 HL135710 NHLBI NIH HHS
- R35 HL166433 NHLBI NIH HHS
- R44 HL164156 NHLBI NIH HHS
- U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
Collapse
Affiliation(s)
- Teodora Nicola
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nancy Wenger
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Xin Xu
- Division of Pulmonary, Allergy and Critical Care Medicine, Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael Evans
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Luhua Qiao
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gabriel Rezonzew
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Youfeng Yang
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tamas Jilling
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Camilla Margaroli
- Division of Pulmonary, Allergy and Critical Care Medicine, Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL, USA
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kristopher Genschmer
- Division of Pulmonary, Allergy and Critical Care Medicine, Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kent Willis
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J Edwin Blalock
- Division of Pulmonary, Allergy and Critical Care Medicine, Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amit Gaggar
- Division of Pulmonary, Allergy and Critical Care Medicine, Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL, USA
- Birmingham VA Medical Center, Birmingham, AL, USA
| | - Charitharth Vivek Lal
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA.
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL, USA.
- Marnix Heersink Institute of Biomedical Innovation, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
3
|
Li SH, Li QP, Chen WJ, Zhong YY, Sun J, Wu JF, Cao YX, Dong JC. Psoralen attenuates cigarette smoke extract-induced inflammation by modulating CD8 + T lymphocyte recruitment and chemokines via the JAK2/STAT1 signaling pathway. Heliyon 2024; 10:e32351. [PMID: 38988534 PMCID: PMC11233870 DOI: 10.1016/j.heliyon.2024.e32351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a respiratory inflammatory disease. Psoralen (PSO) is the main pharmacological component identified from Bu-Shen-Fang-Chuan formula which has been traditionally used in treatment of COPD, yet its efficacy in COPD inflammation were unreported. In this study, we aimed to elucidate the anti-inflammatory potential of PSO in COPD and unravel the underlying mechanisms, focusing on T lymphocyte recruitment and the modulation of chemokines, namely monokine induced by interferon-gamma (CXCL9), interferon inducible protein 10 (CXCL10), and interferon inducible T-Cell alpha chemoattractant (CXCL11). In vitro, RAW264.7 was stimulated by interferon (IFN)-γ + cigarette smoke extract (CSE) and were treated with PSO (2.5, 5, 10 μM), then the levels of chemokines and the activation of Janus kinase (JAK)/Signal transducer and activator of transcription 1 (STAT1) pathway were analyzed by real time PCR and western blot. In vivo, a murine model was established by intraperitoneal injection of CSE on day 1, 8, 15, and 22, then treated with PSO (10 mg/kg). Our experiments in vitro illustrated that PSO reduced the levels of CXCL9, CXCL10, and CXCL11, and decreased the protein phosphorylation levels of JAK2 and STAT1. Additionally, PSO effectively improved inflammatory infiltration and decreased the proportion of CD8+ T cells in CSE-exposed mice. Furthermore, PSO reduced the levels of CXCL9, CXCL10, and CXCL11 in bronchoalveolar lavage fluid (BALF) and lung tissue, and decreased the protein phosphorylation levels of JAK2 and STAT1. In conclusion, our results revealed the therapeutic potential of PSO for COPD inflammation, possibly mediated through the regulation of CD8+ T cell recruitment and chemokines via the JAK2/STAT1 signaling pathway.
Collapse
Affiliation(s)
- Shi-huan Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Qiu-ping Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Wen-jing Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yuan-yuan Zhong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jing Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Jin-feng Wu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yu-xue Cao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Jing-cheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| |
Collapse
|
4
|
Weary TE, Pappas T, Tusiime P, Tuhaise S, Ross E, Gern JE, Goldberg TL. High frequencies of nonviral colds and respiratory bacteria colonization among children in rural Western Uganda. Front Pediatr 2024; 12:1379131. [PMID: 38756971 PMCID: PMC11096560 DOI: 10.3389/fped.2024.1379131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
Introduction Respiratory illness is the most common childhood disease globally, especially in developing countries. Previous studies have detected viruses in approximately 70-80% of respiratory illnesses. Methods In a prospective cohort study of 234 young children (ages 3-11 years) and 30 adults (ages 22-51 years) in rural Western Uganda sampled monthly from May 2019 to August 2021, only 24.2% of nasopharyngeal swabs collected during symptomatic disease had viruses detectable by multiplex PCR diagnostics and metagenomic sequencing. In the remaining 75.8% of swabs from symptomatic participants, we measured detection rates of respiratory bacteria Haemophilus influenzae, Moraxella catarrhalis, and Streptococcus pneumoniae by quantitative PCR. Results 100% of children tested positive for at least one bacterial species. Detection rates were 87.2%, 96.8%, and 77.6% in children and 10.0%, 36.7%, and 13.3% for adults for H. influenzae, M. catarrhalis, and S. pneumoniae, respectively. In children, 20.8% and 70.4% were coinfected with two and three pathogens, respectively, and in adults 6.7% were coinfected with three pathogens but none were coinfected with two. Detection of any of the three pathogens was not associated with season or respiratory symptoms severity, although parsing detection status by symptoms was challenged by children experiencing symptoms in 80.3% of monthly samplings, whereas adults only reported symptoms 26.6% of the time. Pathobiont colonization in children in Western Uganda was significantly more frequent than in children living in high-income countries, including in a study of age-matched US children that utilized identical diagnostic methods. Detection rates were, however, comparable to rates in children living in other Sub-Saharan African countries. Discussion Overall, our results demonstrate that nonviral colds contribute significantly to respiratory disease burden among children in rural Uganda and that high rates of respiratory pathobiont colonization may play a role. These conclusions have implications for respiratory health interventions in the area, such as increasing childhood immunization rates and decreasing air pollutant exposure.
Collapse
Affiliation(s)
- Taylor E. Weary
- Department of Pathobiological Sciences, University of Wisconsin School of Veterinary Medicine, Madison, WI, United States
| | - Tressa Pappas
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | | | | | | | - James E. Gern
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Tony L. Goldberg
- Department of Pathobiological Sciences, University of Wisconsin School of Veterinary Medicine, Madison, WI, United States
| |
Collapse
|
5
|
Wohnhaas CT, Baßler K, Watson CK, Shen Y, Leparc GG, Tilp C, Heinemann F, Kind D, Stierstorfer B, Delić D, Brunner T, Gantner F, Schultze JL, Viollet C, Baum P. Monocyte-derived alveolar macrophages are key drivers of smoke-induced lung inflammation and tissue remodeling. Front Immunol 2024; 15:1325090. [PMID: 38348034 PMCID: PMC10859862 DOI: 10.3389/fimmu.2024.1325090] [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: 10/25/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024] Open
Abstract
Smoking is a leading risk factor of chronic obstructive pulmonary disease (COPD), that is characterized by chronic lung inflammation, tissue remodeling and emphysema. Although inflammation is critical to COPD pathogenesis, the cellular and molecular basis underlying smoking-induced lung inflammation and pathology remains unclear. Using murine smoke models and single-cell RNA-sequencing, we show that smoking establishes a self-amplifying inflammatory loop characterized by an influx of molecularly heterogeneous neutrophil subsets and excessive recruitment of monocyte-derived alveolar macrophages (MoAM). In contrast to tissue-resident AM, MoAM are absent in homeostasis and characterized by a pro-inflammatory gene signature. Moreover, MoAM represent 46% of AM in emphysematous mice and express markers causally linked to emphysema. We also demonstrate the presence of pro-inflammatory and tissue remodeling associated MoAM orthologs in humans that are significantly increased in emphysematous COPD patients. Inhibition of the IRAK4 kinase depletes a rare inflammatory neutrophil subset, diminishes MoAM recruitment, and alleviates inflammation in the lung of cigarette smoke-exposed mice. This study extends our understanding of the molecular signaling circuits and cellular dynamics in smoking-induced lung inflammation and pathology, highlights the functional consequence of monocyte and neutrophil recruitment, identifies MoAM as key drivers of the inflammatory process, and supports their contribution to pathological tissue remodeling.
Collapse
Affiliation(s)
- Christian T. Wohnhaas
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Kevin Baßler
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Carolin K. Watson
- Immunology & Respiratory Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Yang Shen
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Germán G. Leparc
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Cornelia Tilp
- Immunology & Respiratory Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Fabian Heinemann
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - David Kind
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Birgit Stierstorfer
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Denis Delić
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Thomas Brunner
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Florian Gantner
- Department of Biology, University of Konstanz, Konstanz, Germany
- Translational Medicine & Clinical Pharmacology, C. H. Boehringer Sohn AG & Co. KG, Biberach, Germany
| | - Joachim L. Schultze
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and University of Bonn, Bonn, Germany
| | - Coralie Viollet
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Patrick Baum
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
6
|
Linden DA, Guo-Parke H, McKelvey MC, Einarsson GG, Lee AJ, Fairley DJ, Brown V, Lundy G, Campbell C, Logan D, McFarland M, Singh D, McAuley DF, Taggart CC, Kidney JC. Valaciclovir for Epstein-Barr Virus Suppression in Moderate-to-Severe COPD: A Randomized Double-Blind Placebo-Controlled Trial. Chest 2023; 164:625-636. [PMID: 37011709 PMCID: PMC10808072 DOI: 10.1016/j.chest.2023.03.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/25/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND Epstein-Barr virus (EBV) frequently is measured at high levels in COPD using sputum quantitative polymerase chain reaction, whereas airway immunohistochemistry analysis has shown EBV detection to be common in severe disease. RESEARCH QUESTION Is valaciclovir safe and effective for EBV suppression in COPD? STUDY DESIGN AND METHODS The Epstein-Barr Virus Suppression in COPD (EViSCO) trial was a randomized double-blind placebo-controlled trial conducted at the Mater Hospital Belfast, Northern Ireland. Eligible patients had stable moderate-to-severe COPD and sputum EBV (measured using quantitative polymerase chain reaction) and were assigned randomly (1:1) to valaciclovir (1 g tid) or matching placebo for 8 weeks. The primary efficacy outcome was sputum EBV suppression (defined as ≥ 90% sputum viral load reduction) at week 8. The primary safety outcome was the incidence of serious adverse reactions. Secondary outcome measures were FEV1 and drug tolerability. Exploratory outcomes included changes in quality of life, sputum cell counts, and cytokines. RESULTS From November 2, 2018, through March 12, 2020, 84 patients were assigned randomly (n = 43 to valaciclovir). Eighty-one patients completed trial follow-up and were included in the intention-to-treat analysis of the primary outcome. A greater number of participants in the valaciclovir group achieved EBV suppression (n = 36 [87.8%] vs n = 17 [42.5%]; P < .001). Valaciclovir was associated with a significant reduction in sputum EBV titer compared with placebo (-90,404 copies/mL [interquartile range, -298,000 to -15,200 copies/mL] vs -3,940 copies/mL [interquartile range, -114,400 to 50,150 copies/mL]; P = .002). A statistically nonsignificant 24-mL numerical FEV1 increase was shown in the valaciclovir group (difference, -44 mL [95% CI, -150 to 62 mL]; P = .41). However, a reduction in sputum white cell count was noted in the valaciclovir group compared with the placebo group (difference, 2.89 [95% CI, 1.5 × 106-7.4 × 106]; P = .003). INTERPRETATION Valaciclovir is safe and effective for EBV suppression in COPD and may attenuate the sputum inflammatory cell infiltrate. The findings from the current study provide support for a larger trial to evaluate long-term clinical outcomes. TRIAL REGISTRY ClinicalTrials.gov; No.: NCT03699904; URL: www. CLINICALTRIALS gov.
Collapse
Affiliation(s)
- Dermot A Linden
- Mater Hospital Belfast, Belfast Health and Social Care Trus, Belfast, Northern Ireland; Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Belfast, Northern Ireland.
| | - Hong Guo-Parke
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Belfast, Northern Ireland
| | - Michael C McKelvey
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Belfast, Northern Ireland
| | - Gisli G Einarsson
- Halo Research Group, School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland
| | - Andrew J Lee
- Halo Research Group, School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland
| | - Derek J Fairley
- Regional Virus Laboratory, Belfast Health and Social Care Trust, Belfast, Northern Ireland
| | - Vanessa Brown
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Belfast, Northern Ireland
| | - Gavin Lundy
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Belfast, Northern Ireland
| | | | - Danielle Logan
- Northern Ireland Clinical Trials Unit, Belfast, Northern Ireland
| | | | - Dave Singh
- Division of Infection and Immunity, University of Manchester, Manchester, England
| | - Daniel F McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Belfast, Northern Ireland; Royal Victoria Hospital, Belfast, Northern Ireland
| | - Clifford C Taggart
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Belfast, Northern Ireland
| | - Joseph C Kidney
- Mater Hospital Belfast, Belfast Health and Social Care Trus, Belfast, Northern Ireland
| |
Collapse
|
7
|
Kayongo A, Nyiro B, Siddharthan T, Kirenga B, Checkley W, Lutaakome Joloba M, Ellner J, Salgame P. Mechanisms of lung damage in tuberculosis: implications for chronic obstructive pulmonary disease. Front Cell Infect Microbiol 2023; 13:1146571. [PMID: 37415827 PMCID: PMC10320222 DOI: 10.3389/fcimb.2023.1146571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/05/2023] [Indexed: 07/08/2023] Open
Abstract
Pulmonary tuberculosis is increasingly recognized as a risk factor for COPD. Severe lung function impairment has been reported in post-TB patients. Despite increasing evidence to support the association between TB and COPD, only a few studies describe the immunological basis of COPD among TB patients following successful treatment completion. In this review, we draw on well-elaborated Mycobacterium tuberculosis-induced immune mechanisms in the lungs to highlight shared mechanisms for COPD pathogenesis in the setting of tuberculosis disease. We further examine how such mechanisms could be exploited to guide COPD therapeutics.
Collapse
Affiliation(s)
- Alex Kayongo
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - Brian Nyiro
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Trishul Siddharthan
- Division of Pulmonary and Critical Care Medicine, University of Miami, Miami, FL, United States
| | - Bruce Kirenga
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - William Checkley
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States
- Center for Global Non-Communicable Disease Research and Training, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Moses Lutaakome Joloba
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - Jerrold Ellner
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Padmini Salgame
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
8
|
Yi L, Zhao Y, Guo Z, Li Q, Zhang G, Tian X, Xu X, Luo Z. The role of small airway function parameters in preschool asthmatic children. BMC Pulm Med 2023; 23:219. [PMID: 37340433 DOI: 10.1186/s12890-023-02515-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/08/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND Small airways are the major sites of inflammation and airway remodeling in all severities of asthma patients. However, whether small airway function parameters could reflect the airway dysfunction feature in preschool asthmatic children remain unclear. We aim to investigate the role of small airway function parameters in evaluating airway dysfunction, airflow limitation and airway hyperresponsiveness (AHR). METHODS Eight hundred and fifty-one preschool children diagnosed with asthma were enrolled retrospectively to investigate the characteristics of small airway function parameters. Curve estimation analysis was applied to clarify the correlation between small and large airway dysfunction. Spearman's correlation and receiver-operating characteristic (ROC) curves were employed to evaluate the relationship between small airway dysfunction (SAD) and AHR. RESULTS The prevalence of SAD was 19.5% (166 of 851) in this cross-sectional cohort study. Small airway function parameters (FEF25-75%, FEF50%, FEF75%) showed strong correlations with FEV1% (r = 0.670, 0.658, 0.609, p<0.001, respectively), FEV1/FVC% (r = 0.812, 0.751, 0.871, p<0.001, respectively) and PEF% (r = 0.626, 0.635, 0.530, p<0.01, respectively). Moreover, small airway function parameters and large airway function parameters (FEV1%, FEV1/FVC%, PEF%) were curve-associated rather than linear-related (p<0.001). FEF25-75%, FEF50%, FEF75% and FEV1% demonstrated a positive correlation with PC20 (r = 0.282, 0.291, 0.251, 0.224, p<0.001, respectively). Interestingly, FEF25-75% and FEF50% exhibited a higher correlation coefficient with PC20 than FEV1% (0.282 vs. 0.224, p = 0.031 and 0.291 vs. 0.224, p = 0.014, respectively). ROC curve analysis for predicting moderate to severe AHR showed that the area under the curve (AUC) was 0.796, 0.783, 0.738, and 0.802 for FEF25-75%, FEF50%, FEF75%, and the combination of FEF25-75% and FEF75%, respectively. When Compared to children with normal lung function, patients with SAD were slightly older, more likely to have a family history of asthma and airflow obstruction with lower FEV1% and FEV1/FVC%, lower PEF% and more severe AHR with lower PC20 ( all p<0.05). CONCLUSION Small airway dysfunction is highly correlated with large airway function impairment, severe airflow obstruction and AHR in preschool asthmatic children. Small airway function parameters should be utilized in the management of preschool asthma.
Collapse
Affiliation(s)
- Liangqin Yi
- Chongqing Key Laboratory of Pediatrics, International Science and Technology Cooperation base of Child Development and Critical Disorders, Department of Children's Hospital of Chongqing Medical, Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Department of Clinical Laboratory center, University of Education, 400014, Chongqing, China
| | - Yan Zhao
- Chongqing Key Laboratory of Pediatrics, International Science and Technology Cooperation base of Child Development and Critical Disorders, Department of Children's Hospital of Chongqing Medical, Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Department of Clinical Laboratory center, University of Education, 400014, Chongqing, China
| | - Ziyao Guo
- Chongqing Key Laboratory of Pediatrics, International Science and Technology Cooperation base of Child Development and Critical Disorders, Department of Children's Hospital of Chongqing Medical, Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Department of Clinical Laboratory center, University of Education, 400014, Chongqing, China
| | - Qinyuan Li
- Chongqing Key Laboratory of Pediatrics, International Science and Technology Cooperation base of Child Development and Critical Disorders, Department of Children's Hospital of Chongqing Medical, Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Department of Clinical Laboratory center, University of Education, 400014, Chongqing, China
| | - Guangli Zhang
- National Clinical Research Center for Child Health and Disorders, Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyin Tian
- National Clinical Research Center for Child Health and Disorders, Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ximing Xu
- Big Data Center for Children's Medical Care, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhengxiu Luo
- National Clinical Research Center for Child Health and Disorders, Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
9
|
Bazzan E, Casara A, Radu CM, Tinè M, Biondini D, Faccioli E, Pezzuto F, Bernardinello N, Conti M, Balestro E, Calabrese F, Simioni P, Rea F, Turato G, Spagnolo P, Cosio MG, Saetta M. Macrophages-derived Factor XIII links coagulation to inflammation in COPD. Front Immunol 2023; 14:1131292. [PMID: 37180121 PMCID: PMC10166842 DOI: 10.3389/fimmu.2023.1131292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 04/05/2023] [Indexed: 05/15/2023] Open
Abstract
Background The local, extravascular, activation of the coagulation system in response to injury is a key factor mediating the resulting inflammatory response. Coagulation Factor XIIIA (FXIIIA) found in alveolar macrophages (AM) and dendritic cells (DC), by influencing fibrin stability, might be an inflammatory modifier in COPD. Aims To study the expression of FXIIIA in AM and Langerin+DC (DC-1) and their relation to the inflammatory response and disease progression in COPD. Methods In 47 surgical lungs, 36 from smokers (22 COPD and 14 no-COPD) and 11 from non-smokers we quantified by immunohistochemistry FXIIIA expression in AM and DC-1 along with numbers of CD8+Tcells and CXCR3 expression in lung parenchyma and airways. Lung function was measured prior to surgery. Results The percentage of AM expressing FXIII (%FXIII+AM) was higher in COPD than no-COPD and non-smokers. DC-1 expressed FXIIIA and their numbers were higher in COPD than no-COPD and non-smokers. DC-1 positively correlated with %FXIII+AM (r=0.43; p<0.018). CD8+Tcells, which were higher in COPD than in no-COPD, were correlated with DC-1 (p<0.01) and %FXIII+AM. CXCR3+ cells were increased in COPD and correlated with %FXIII+AM (p<0.05). Both %FXIII+AM (r=-0.6; p=0.001) and DC-1 (r=-0.7; p=0.001) correlated inversely with FEV1. Conclusion FXIIIA, an important link between the extravascular coagulation cascade and inflammatory response, is significantly expressed in alveolar macrophages and dendritic cells of smokers with COPD, suggesting that it could play an important role in the adaptive inflammatory reaction characteristic of the disease.
Collapse
Affiliation(s)
- Erica Bazzan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Alvise Casara
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | | | - Mariaenrica Tinè
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Davide Biondini
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | - Eleonora Faccioli
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Nicol Bernardinello
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Maria Conti
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Elisabetta Balestro
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Paolo Simioni
- Department of Medicine, University of Padova, Padova, Italy
| | - Federico Rea
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Graziella Turato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Paolo Spagnolo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Manuel G. Cosio
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Meakins-Christie Laboratories, Respiratory Division, McGill University, Montreal, QC, Canada
| | - Marina Saetta
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| |
Collapse
|
10
|
Nourian YH, Salimian J, Ahmadi A, Salehi Z, Karimi M, Emamvirdizadeh A, Azimzadeh Jamalkandi S, Ghanei M. cAMP-PDE signaling in COPD: Review of cellular, molecular and clinical features. Biochem Biophys Rep 2023; 34:101438. [PMID: 36865738 PMCID: PMC9971187 DOI: 10.1016/j.bbrep.2023.101438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/21/2023] [Accepted: 02/02/2023] [Indexed: 02/18/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the fourth leading cause of death among non-contagious diseases in the world. PDE inhibitors are among current medicines prescribed for COPD treatment of which, PDE-4 family is the predominant PDE isoform involved in hydrolyzing cyclic adenosine monophosphate (cAMP) that regulates the inflammatory responses in neutrophils, lymphocytes, macrophages and epithelial cells The aim of this study is to investigate the cellular and molecular mechanisms of cAMP-PDE signaling, as an important pathway in the treatment management of patients with COPD. In this review, a comprehensive literature review was performed about the effect of PDEs in COPD. Generally, PDEs are overexpressed in COPD patients, resulting in cAMP inactivation and decreased cAMP hydrolysis from AMP. At normal amounts, cAMP is one of the essential agents in regulating metabolism and suppressing inflammatory responses. Low amount of cAMP lead to activation of downstream inflammatory signaling pathways. PDE4 and PDE7 mRNA transcript levels were not altered in polymorphonuclear leukocytes and CD8 lymphocytes originating from the peripheral venous blood of stable COPD subjects compared to healthy controls. Therefore, cAMP-PDE signaling pathway is one of the most important signaling pathways involved in COPD. By examining the effects of different drugs in this signaling pathway critical steps can be taken in the treatment of this disease.
Collapse
Affiliation(s)
- Yazdan Hasani Nourian
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Salimian
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Ahmadi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Karimi
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Emamvirdizadeh
- Department of Molecular Genetics, Faculty of Bio Sciences, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Sadegh Azimzadeh Jamalkandi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran,Corresponding author.
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Huber ME, Larson E, Lust TN, Heisler CM, Harriff MJ. Chronic Obstructive Pulmonary Disease and Cigarette Smoke Lead to Dysregulated Mucosal-associated Invariant T-Cell Activation. Am J Respir Cell Mol Biol 2023; 68:90-102. [PMID: 36174211 PMCID: PMC9817907 DOI: 10.1165/rcmb.2022-0131oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 09/29/2022] [Indexed: 02/05/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is associated with airway inflammation, increased infiltration by CD8+ T lymphocytes, and infection-driven exacerbations. Although cigarette smoke is the leading risk factor for COPD, the mechanisms driving the development of COPD in only a subset of smokers are incompletely understood. Lung-resident mucosal-associated invariant T (MAIT) cells play a role in microbial infections and inflammatory diseases. The role of MAIT cells in COPD pathology is unknown. Here, we examined MAIT cell activation in response to cigarette smoke-exposed primary human bronchial epithelial cells (BECs) from healthy, COPD, or smoker donors. We observed significantly higher baseline MAIT cell responses to COPD BECs than healthy BECs. However, infected COPD BECs stimulated a smaller fold increase in MAIT cell response despite increased microbial infection. For all donor groups, cigarette smoke-exposed BECs elicited reduced MAIT cell responses; conversely, cigarette smoke exposure increased ligand-mediated MR1 surface translocation in healthy and COPD BECs. Our data demonstrate that MAIT cell activation is dysregulated in the context of cigarette smoke and COPD. MAIT cells could contribute to cigarette smoke- and COPD-associated inflammation through inappropriate activation and reduced early recognition of bacterial infection, contributing to microbial persistence and COPD exacerbations.
Collapse
Affiliation(s)
| | - Emily Larson
- Portland Veterans Affairs Research Foundation, Portland, Oregon; and
| | - Taylor N. Lust
- Division of Pulmonary, Allergy, and Critical Care Medicine, Oregon Health & Science University, Portland, Oregon
| | - Chelsea M. Heisler
- Division of Pulmonary, Allergy, and Critical Care Medicine, Oregon Health & Science University, Portland, Oregon
| | - Melanie J. Harriff
- Department of Molecular and Microbial Immunology and
- Division of Pulmonary, Allergy, and Critical Care Medicine, Oregon Health & Science University, Portland, Oregon
- Veterans Affairs Portland Health Care System, Portland, Oregon
| |
Collapse
|
12
|
Deng Z, Ding W, Li F, Shen S, Huang C, Lai K. Pulmonary IFN-γ Causes Lymphocytic Inflammation and Cough Hypersensitivity by Increasing the Number of IFN-γ-Secreting T Lymphocytes. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2022; 14:653-673. [PMID: 36426396 PMCID: PMC9709684 DOI: 10.4168/aair.2022.14.6.653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 07/13/2022] [Accepted: 07/26/2022] [Indexed: 07/25/2023]
Abstract
PURPOSE Respiratory viral infection increases the number of lung-resident T lymphocytes, which enhance cough sensitivity by producing interferon-γ (IFN-γ). It is poorly understood why IFN-γ-secreting T lymphocytes persist for a long time when the respiratory viruses have been removed. METHODS Repeated pulmonary administration of IFN-γ and intraperitoneal injection with different inhibitors were used to study the effects of pulmonary IFN-γ in mice and guinea pigs. RESULTS IFN-γ administration caused the increasing of IFN-γ-secreting T lymphocytes in both lung and blood, followed by the elevated physiological level of IFN-γ in the lung, the airway inflammation and the airway epithelial damage. IFN-γ administration also enhanced the cough sensitivity of guinea pigs. IFN-γ activated the STAT1 and extracellular signal-regulated kinase (ERK) pathways in lung tissues, released IFN-γ-inducible protein 10 (IP-10), and resulted in F-actin accumulation in lung-resident lymphocytes. The CXC chemokine receptor 3 (CXCR3) inhibitor potently suppressed all the IFN-γ-induced inflammatory changes. The STAT1 inhibitor mitigated IFN-γ-secreting T lymphocytes infiltration by inhibiting T lymphocytes proliferation. F-actin accumulation and the ERK1/2 pathway contributed to pulmonary IFN-γ-induced augmentation of the airway inflammation and increasing of IFN-γ-secreting T lymphocytes in blood. CONCLUSIONS High physiological levels of IFN-γ in the lung may cause pulmonary lymphocytic inflammation and cough hypersensitivity by increasing the number of IFN-γ-secreting T lymphocytes through the IP-10 and CXCR3 pathways.
Collapse
Affiliation(s)
- Zheng Deng
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenbin Ding
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fengying Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuirong Shen
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chuqin Huang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kefang Lai
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
13
|
Talukdar SN, Osan J, Ryan K, Grove B, Perley D, Kumar BD, Yang S, Dallman S, Hollingsworth L, Bailey KL, Mehedi M. RSV-induced Expanded Ciliated Cells Contribute to Bronchial Wall Thickening.. [DOI: 10.1101/2022.10.31.514471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
AbstractViral infection, particularly respiratory syncytial virus (RSV), causes inflammation in the bronchiolar airways (bronchial wall thickening, also known as bronchiolitis), reducing airflow through the bronchioles. This bronchial wall thickening is a common pathophysiological feature in RSV infection, but it causes more fatalities in infants than in children and adults. However, the molecular mechanism of RSV-induced bronchial wall thickening remains unknown, particularly in healthy adults. RSV infection in the airway epithelium of healthy adult bronchial cells reveals RSV-infects primarily ciliated cells. RSV infection expands the cell cytoskeleton substantially without compromising epithelial membrane integrity and ciliary functions. The RSV-induced actin cytoskeleton expansion increases ununiformly epithelial height, and cytoskeletal (actin polymerization), immunological (INF-L1, TNF-α, IP10/CXCL10), and viral (NS2) factors are probably responsible. Interestingly, RSV-infected cell cytoskeleton’s expansion resembles a noncanonical inflammatory phenotype, which contributes to bronchial wall thickening, and is termed cytoskeletal inflammation.Author SummaryRSV infects everyone. Although RSV-induced fatal pathophysiology (e.g., bronchiolitis) is more common in infants than adults, this bronchiolitis (or bronchial wall thickening) is common in the lower respiratory tract due to RSV infection in all ages. To determine the molecular mechanism of RSV-induced bronchial wall thickening, we infectedin vitroadult airway epithelium with RSV. We found that RSV-infection induced a substantial actin-cytoskeleton expansion, consequently increased the height of the epithelium. We identified actin polymerization, secretion of proinflammatory cytokines and chemokines, and viral proteins contribute to the RSV-induced cytoskeletal expansion. Our results suggest that RSV-induces a novel noncanonical epithelial host response termed cytoskeletal inflammation, which may contribute to bronchial wall thickening.
Collapse
|
14
|
Li DY, Chen L, Miao SY, Zhou M, Wu JH, Sun SW, Liu LL, Qi C, Xiong XZ. Inducible Costimulator-C-X-C Motif Chemokine Receptor 3 Signaling is Involved in Chronic Obstructive Pulmonary Disease Pathogenesis. Int J Chron Obstruct Pulmon Dis 2022; 17:1847-1861. [PMID: 35991707 PMCID: PMC9386059 DOI: 10.2147/copd.s371801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/31/2022] [Indexed: 11/23/2022] Open
Abstract
Background The role of inducible costimulator (ICOS) signaling in chronic obstructive pulmonary disease (COPD) has not been fully elucidated. Methods We compared the percentages of ICOS+ T cells and ICOS+ regulatory T (Treg) cells in CD4+ T cells and CD4+CD25+FOXP3+ Tregs, respectively, in the peripheral blood of smokers with or without COPD to those in healthy controls. We further characterized their phenotypes using flow cytometry. To investigate the influence of ICOS signaling on C-X-C motif chemokine receptor 3 (CXCR3) expression in COPD, we evaluated the expression levels of ICOS and CXCR3 in vivo and in vitro. Results ICOS expression was elevated on peripheral CD4+ T cells and CD4+ Tregs of COPD patients, which positively correlated with the severity of lung function impairment in patients with stable COPD (SCOPD), but not in patients with acute exacerbation of COPD (AECOPD). ICOS+CD4+ Tregs in patients with SCOPD expressed higher levels of coinhibitors, programmed cell death protein 1 (PD-1) and T-cell immunoreceptor with Ig and ITIM domains (TIGIT), than ICOS−CD4+ Tregs, whereas ICOS+CD4+ T cells mostly exhibited a central memory (CD45RA−CCR7+) or effector memory (CD45RA−CCR7−) phenotype, ensuring their superior potential to respond potently and quickly to pathogen invasion. Furthermore, increased percentages of CXCR3+CD4+ T cells and CXCR3+CD4+ Tregs were observed in the peripheral blood of patients with SCOPD, and the expression level of CXCR3 was higher in ICOS+CD4+ T cells than in ICOS−CD4+ T cells. The percentage of CXCR3+CD4+ T cells was even higher in the bronchoalveolar lavage fluid than in matched peripheral blood in SCOPD group. Lastly, in vitro experiments showed that ICOS induced CXCR3 expression on CD4+ T cells. Conclusions ICOS signaling is upregulated in COPD, which induces CXCR3 expression. This may contribute to increased numbers of CXCR3+ Th1 cells in the lungs of patients with COPD, causing inflammation and tissue damage.
Collapse
Affiliation(s)
- Dan-Yang Li
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Long Chen
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Shuai-Ying Miao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Department of Critical Care Medicine, General Hospital of Pingmei Shenma Medical Group, Pingdingshan, 467000, People's Republic of China
| | - Mei Zhou
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Jiang-Hua Wu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Sheng-Wen Sun
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Lan-Lan Liu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Chang Qi
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Xian-Zhi Xiong
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| |
Collapse
|
15
|
Caroff E, Meyer EA, Äänismaa P, Froidevaux S, Keller M, Piali L. Design, Synthesis, and Pharmacological Evaluation of Benzimidazolo-thiazoles as Potent CXCR3 Antagonists with Therapeutic Potential in Autoimmune Diseases: Discovery of ACT-672125. J Med Chem 2022; 65:11533-11549. [PMID: 35969159 DOI: 10.1021/acs.jmedchem.2c00676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The chemokine receptor CXCR3 allows the selective recruitment of innate and adaptive inflammatory immune cells into inflamed tissue. CXCR3 ligands are secreted after exposure to pro-inflammatory cytokines. Upon binding to CXCR3 ligands, CXCR3 expressing T-lymphocytes migrate toward sites of inflammation and can promote tissue damage. Therefore, antagonizing this receptor may provide clinical benefits for patients suffering from autoimmune diseases characterized by high concentrations of CXCR3 ligands. Herein, we report the second part of our CXCR3 discovery program where we explored the benzimidazolo-thiazole core scaffold. The optimization of potency and the mitigation of an hERG liability are described. Further pharmacokinetic considerations led to the identification of the potent CXCR3 antagonist ACT-672125 (29). The compound showed good physicochemical properties and safety profile. In a proof-of-mechanism model of lung inflammation, ACT-672125 inhibited the recruitment of CXCR3 expressing T cells into the inflamed lung in a dose-dependent manner.
Collapse
Affiliation(s)
- Eva Caroff
- Drug Discovery Chemistry Immunology, Idorsia Pharmaceuticals Ltd., Allschwil 4123, Switzerland
| | - Emmanuel A Meyer
- Drug Discovery Chemistry Immunology, Idorsia Pharmaceuticals Ltd., Allschwil 4123, Switzerland
| | - Päivi Äänismaa
- DMPK, Idorsia Pharmaceuticals Ltd., Allschwil 4123, Switzerland
| | | | - Marcel Keller
- Drug Discovery Biology Immunology, Idorsia Pharmaceuticals Ltd., Allschwil 4123, Switzerland
| | - Luca Piali
- Immunology, Infectious Diseases and Ophthalmology, pRED Roche, Basel 4070, Switzerland
| |
Collapse
|
16
|
Meyer EA, Äänismaa P, Froidevaux S, Keller M, Piali L, Caroff E. Discovery and In Vivo Evaluation of ACT-660602: A Potent and Selective Antagonist of the Chemokine Receptor CXCR3 for Autoimmune Diseases. J Med Chem 2022; 65:11513-11532. [PMID: 35947786 DOI: 10.1021/acs.jmedchem.2c00675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The chemokine receptor CXCR3 is a seven-transmembrane G-protein-coupled receptor (GPCR) involved in various pathologies, in particular autoimmune diseases. It is activated by the three chemokine ligands CXCL9, CXCL10, and CXCL11 and enables the recruitment of immune cell subsets leading to damage of inflamed tissues. Starting from a high-throughput screening hit, we describe the iterative optimization of a chemical series culminating in the discovery of the selective CXCR3 antagonist ACT-660602 (9j). The careful structural modifications during the lead optimization phase led to a compound with high biological potency in inhibiting cell migration together with improvements of the metabolic stability and hERG issue. In a LPS-induced lung inflammation model in mice, ACT-660602 led to significantly reduced recruitment of the CXCR3+ CD8+ T cell in the bronchoalveolar lavage compartment when administered orally at a dose of 30 mg/kg.
Collapse
Affiliation(s)
- Emmanuel A Meyer
- Drug Discovery Chemistry Immunology, Idorsia Pharmaceuticals Ltd., Allschwil 4123, Switzerland
| | - Päivi Äänismaa
- DMPK, Idorsia Pharmaceuticals Ltd., Allschwil 4123, Switzerland
| | | | - Marcel Keller
- Drug Discovery Biology Immunology, Idorsia Pharmaceuticals Ltd., Allschwil 4123, Switzerland
| | - Luca Piali
- Immunology, Infectious Diseases and Ophthalmology, pRED Roche, Basel 4070, Switzerland
| | - Eva Caroff
- Drug Discovery Chemistry Immunology, Idorsia Pharmaceuticals Ltd., Allschwil 4123, Switzerland
| |
Collapse
|
17
|
Aguilar D, Bosacoma A, Blanco I, Tura-Ceide O, Serrano-Mollar A, Barberà JA, Peinado VI. Differences and Similarities between the Lung Transcriptomic Profiles of COVID-19, COPD, and IPF Patients: A Meta-Analysis Study of Pathophysiological Signaling Pathways. Life (Basel) 2022; 12:887. [PMID: 35743918 PMCID: PMC9227224 DOI: 10.3390/life12060887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/02/2022] [Accepted: 06/11/2022] [Indexed: 11/20/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a pandemic respiratory disease associated with high morbidity and mortality. Although many patients recover, long-term sequelae after infection have become increasingly recognized and concerning. Among other sequelae, the available data indicate that many patients who recover from COVID-19 could develop fibrotic abnormalities over time. To understand the basic pathophysiology underlying the development of long-term pulmonary fibrosis in COVID-19, as well as the higher mortality rates in patients with pre-existing lung diseases, we compared the transcriptomic fingerprints among patients with COVID-19, idiopathic pulmonary fibrosis (IPF), and chronic obstructive pulmonary disease (COPD) using interactomic analysis. Patients who died of COVID-19 shared some of the molecular biological processes triggered in patients with IPF, such as those related to immune response, airway remodeling, and wound healing, which could explain the radiological images seen in some patients after discharge. However, other aspects of this transcriptomic profile did not resemble the profile associated with irreversible fibrotic processes in IPF. Our mathematical approach instead showed that the molecular processes that were altered in COVID-19 patients more closely resembled those observed in COPD. These data indicate that patients with COPD, who have overcome COVID-19, might experience a faster decline in lung function that will undoubtedly affect global health.
Collapse
Affiliation(s)
- Daniel Aguilar
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREDH), 28005 Madrid, Spain;
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (A.B.); (I.B.); (O.T.-C.); (A.S.-M.); (J.A.B.)
| | - Adelaida Bosacoma
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (A.B.); (I.B.); (O.T.-C.); (A.S.-M.); (J.A.B.)
- Biomedical Research Networking Center in Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| | - Isabel Blanco
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (A.B.); (I.B.); (O.T.-C.); (A.S.-M.); (J.A.B.)
- Biomedical Research Networking Center in Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Department of Pulmonary Medicine, Hospital Clínic, University of Barcelona, 08007 Barcelona, Spain
| | - Olga Tura-Ceide
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (A.B.); (I.B.); (O.T.-C.); (A.S.-M.); (J.A.B.)
- Biomedical Research Networking Center in Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Department of Pulmonary Medicine, Hospital Clínic, University of Barcelona, 08007 Barcelona, Spain
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
| | - Anna Serrano-Mollar
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (A.B.); (I.B.); (O.T.-C.); (A.S.-M.); (J.A.B.)
- Biomedical Research Networking Center in Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Department of Experimental Pathology, Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC-IDIBAPS, 08036 Barcelona, Spain
| | - Joan Albert Barberà
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (A.B.); (I.B.); (O.T.-C.); (A.S.-M.); (J.A.B.)
- Biomedical Research Networking Center in Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Department of Pulmonary Medicine, Hospital Clínic, University of Barcelona, 08007 Barcelona, Spain
| | - Victor Ivo Peinado
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (A.B.); (I.B.); (O.T.-C.); (A.S.-M.); (J.A.B.)
- Biomedical Research Networking Center in Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Department of Pulmonary Medicine, Hospital Clínic, University of Barcelona, 08007 Barcelona, Spain
- Department of Experimental Pathology, Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC-IDIBAPS, 08036 Barcelona, Spain
| |
Collapse
|
18
|
Angiogenesis, Lymphangiogenesis, and Inflammation in Chronic Obstructive Pulmonary Disease (COPD): Few Certainties and Many Outstanding Questions. Cells 2022; 11:cells11101720. [PMID: 35626756 PMCID: PMC9139415 DOI: 10.3390/cells11101720] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 02/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by chronic inflammation, predominantly affecting the lung parenchyma and peripheral airways, that results in progressive and irreversible airflow obstruction. COPD development is promoted by persistent pulmonary inflammation in response to several stimuli (e.g., cigarette smoke, bacterial and viral infections, air pollution, etc.). Angiogenesis, the formation of new blood vessels, and lymphangiogenesis, the formation of new lymphatic vessels, are features of airway inflammation in COPD. There is compelling evidence that effector cells of inflammation (lung-resident macrophages and mast cells and infiltrating neutrophils, eosinophils, basophils, lymphocytes, etc.) are major sources of a vast array of angiogenic (e.g., vascular endothelial growth factor-A (VEGF-A), angiopoietins) and/or lymphangiogenic factors (VEGF-C, -D). Further, structural cells, including bronchial and alveolar epithelial cells, endothelial cells, fibroblasts/myofibroblasts, and airway smooth muscle cells, can contribute to inflammation and angiogenesis in COPD. Although there is evidence that alterations of angiogenesis and, to a lesser extent, lymphangiogenesis, are associated with COPD, there are still many unanswered questions.
Collapse
|
19
|
Pournaras N, Andersson A, Kovach MA, Padra M, Che KF, Brundin B, Yoshihara S, Bozinovski S, Lindén SK, Jansson PA, Sköld MC, Qvarfordt I, Lindén A. Glucose Homeostasis in Relation to Neutrophil Mobilization in Smokers with COPD. Int J Chron Obstruct Pulmon Dis 2022; 17:1179-1194. [PMID: 35620349 PMCID: PMC9129100 DOI: 10.2147/copd.s353753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/03/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Nikolaos Pournaras
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
- Correspondence: Nikolaos Pournaras, Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden and Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden, Email
| | - Anders Andersson
- COPD Center, Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, Gothenburg, Sweden
- COPD Center, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Melissa A Kovach
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Médea Padra
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karlhans F Che
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Bettina Brundin
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Shigemi Yoshihara
- Pediatric Allergology and Respiratory Medicine, Department of Pediatrics, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Sara K Lindén
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per-Anders Jansson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magnus C Sköld
- Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
- Division for Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Ingemar Qvarfordt
- COPD Center, Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Lindén
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
20
|
Guo-Parke H, Linden D, Mousnier A, Scott IC, Killick H, Borthwick LA, Fisher AJ, Weldon S, Taggart CC, Kidney JC. Altered Differentiation and Inflammation Profiles Contribute to Enhanced Innate Responses in Severe COPD Epithelium to Rhinovirus Infection. Front Med (Lausanne) 2022; 9:741989. [PMID: 35280870 PMCID: PMC8916560 DOI: 10.3389/fmed.2022.741989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Background Respiratory viral infections are closely associated with COPD exacerbations, hospitalisations, and significant morbidity and mortality. The consequences of the persisting inflammation and differentiation status in virus associated severe disease is not fully understood. The aim of this study was to evaluate barrier function, cellular architecture, the inflammatory response in severe COPD bronchial epithelium to human rhinovirus (HRV) induced pathological changes and innate immune responses. Methods Well-differentiated primary bronchial epithelial cells (WD-PBECs) derived from severe COPD patients and age-matched healthy controls were cultured in the air-liquid interface (ALI) model. The differentiation phenotype, epithelial barrier integrity, pathological response and cytokine secreting profile of these cultures before and after HRV infection were investigated. Results WD-PBECs derived from severe COPD patients showed aberrant epithelium differentiation with a decreased proportion of ciliated cells but increased numbers of club cells and goblet cells compared with healthy controls. Tight junction integrity was compromised in both cultures following HRV infection, with heightened disruptions in COPD cultures. HRV induced increased epithelial cell sloughing, apoptosis and mucus hypersecretion in COPD cultures compared with healthy controls. A Th1/Th2 imbalance and a strong interferon and pro-inflammatory cytokine response was also observed in COPD cultures, characterized by increased levels of IFNγ, IFNβ, IP-10, IL-10 and decreased TSLP and IL-13 cytokine levels prior to HRV infection. Significantly enhanced basolateral secretion of eotaxin 3, IL-6, IL-8, GM-CSF were also observed in both mock and HRV infected COPD cultures compared with corresponding healthy controls. In response to HRV infection, all cultures displayed elevated levels of IFNλ1 (IL-29), IP-10 and TNFα compared with mock infected cultures. Interestingly, HRV infection dramatically reduced IFNλ levels in COPD cultures compared with healthy subjects. Conclusion An altered differentiation phenotype and cytokine response as seen in severe COPD WD-PBECs may contribute to increased disease susceptibility and an enhanced inflammatory response to HRV infection.
Collapse
Affiliation(s)
- Hong Guo-Parke
- Airway Innate Immunity Research Group, Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, United Kingdom
| | - Dermot Linden
- Airway Innate Immunity Research Group, Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, United Kingdom
| | - Aurelie Mousnier
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, United Kingdom
| | - Ian C. Scott
- Translational Sciences and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Helen Killick
- Translational Sciences and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Lee A. Borthwick
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrew J. Fisher
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- Institute of Transplantation, Newcastle upon Tyne Hospitals, NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Sinéad Weldon
- Airway Innate Immunity Research Group, Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, United Kingdom
| | - Clifford C. Taggart
- Airway Innate Immunity Research Group, Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, United Kingdom
| | - Joseph C. Kidney
- Department of Respiratory Medicine, Mater Hospital, Belfast, United Kingdom
- *Correspondence: Joseph C. Kidney
| |
Collapse
|
21
|
Facchinetti F, Civelli M, Singh D, Papi A, Emirova A, Govoni M. Tanimilast, A Novel Inhaled Pde4 Inhibitor for the Treatment of Asthma and Chronic Obstructive Pulmonary Disease. Front Pharmacol 2021; 12:740803. [PMID: 34887752 PMCID: PMC8650159 DOI: 10.3389/fphar.2021.740803] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic respiratory diseases are the third leading cause of death, behind cardiovascular diseases and cancer, affecting approximately 550 million of people all over the world. Most of the chronic respiratory diseases are attributable to asthma and chronic obstructive pulmonary disease (COPD) with this latter being the major cause of deaths. Despite differences in etiology and symptoms, a common feature of asthma and COPD is an underlying degree of airways inflammation. The nature and severity of this inflammation might differ between and within different respiratory conditions and pharmacological anti-inflammatory treatments are unlikely to be effective in all patients. A precision medicine approach is needed to selectively target patients to increase the chance of therapeutic success. Inhibitors of the phosphodiesterase 4 (PDE4) enzyme like the oral PDE4 inhibitor roflumilast have shown a potential to reduce inflammatory-mediated processes and the frequency of exacerbations in certain groups of COPD patients with a chronic bronchitis phenotype. However, roflumilast use is dampened by class related side effects as nausea, diarrhea, weight loss and abdominal pain, resulting in both substantial treatment discontinuation in clinical practice and withdrawal from clinical trials. This has prompted the search for PDE4 inhibitors to be given by inhalation to reduce the systemic exposure (and thus optimize the systemic safety) and maximize the therapeutic effect in the lung. Tanimilast (international non-proprietary name of CHF6001) is a novel highly potent and selective inhaled PDE4 inhibitor with proven anti-inflammatory properties in various inflammatory cells, including leukocytes derived from asthma and COPD patients, as well as in experimental rodent models of pulmonary inflammation. Inhaled tanimilast has reached phase III clinical development by showing promising pharmacodynamic results associated with a good tolerability and safety profile, with no evidence of PDE4 inhibitors class-related side effects. In this review we will discuss the main outcomes of preclinical and clinical studies conducted during tanimilast development, with particular emphasis on the characterization of the pharmacodynamic profile that led to the identification of target populations with increased therapeutic potential in inflammatory respiratory diseases.
Collapse
Affiliation(s)
| | | | - Dave Singh
- Medicines Evaluation Unit, Manchester University NHS Foundation Hospital Trust, Manchester, United Kingdom
| | - Alberto Papi
- Respiratory Medicine, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Aida Emirova
- Global Clinical Development, Chiesi, Parma, Italy
| | - Mirco Govoni
- Global Clinical Development, Chiesi, Parma, Italy
| |
Collapse
|
22
|
Pouzol L, Sassi A, Baumlin N, Tunis M, Strasser DS, Lehembre F, Martinic MM. CXCR7 Antagonism Reduces Acute Lung Injury Pathogenesis. Front Pharmacol 2021; 12:748740. [PMID: 34803691 PMCID: PMC8602191 DOI: 10.3389/fphar.2021.748740] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022] Open
Abstract
Loss of control in the trafficking of immune cells to the inflamed lung tissue contributes to the pathogenesis of life-threatening acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS). Targeting CXCR7 has been proposed as a potential therapeutic approach to reduce pulmonary inflammation; however, its role and its crosstalk with the two chemokine receptors CXCR3 and CXCR4 via their shared ligands CXCL11 and CXCL12 is not yet completely understood. The present paper aimed to characterize the pathological role of the CXCR3/CXCR4/CXCR7 axis in a murine model of ALI. Lipopolysaccharide (LPS) inhalation in mice resulted in the development of key pathologic features of ALI/ARDS, including breathing dysfunctions, alteration in the alveolar capillary barrier, and lung inflammation. LPS inhalation induced immune cell infiltration into the bronchoalveolar space, including CXCR3+ and CXCR4+ cells, and enhanced the expression of the ligands of these two chemokine receptors. The first-in-class CXCR7 antagonist, ACT-1004-1239, increased levels of CXCL11 and CXCL12 in the plasma without affecting their levels in inflamed lung tissue, and consequently reduced CXCR3+ and CXCR4+ immune cell infiltrates into the bronchoalveolar space. In the early phase of lung inflammation, characterized by a massive influx of neutrophils, treatment with ACT-1004-1239 significantly reduced the LPS-induced breathing pattern alteration. Both preventive and therapeutic treatment with ACT-1004-1239 reduced lung vascular permeability and decreased inflammatory cell infiltrates. In conclusion, these results demonstrate a key pathological role of CXCR7 in ALI/ARDS and highlight the clinical potential of ACT-1004-1239 in ALI/ARDS pathogenesis.
Collapse
Affiliation(s)
| | - Anna Sassi
- Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | | | - Mélanie Tunis
- Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | | | | | | |
Collapse
|
23
|
Baker JR, Donnelly LE. Leukocyte Function in COPD: Clinical Relevance and Potential for Drug Therapy. Int J Chron Obstruct Pulmon Dis 2021; 16:2227-2242. [PMID: 34354348 PMCID: PMC8331105 DOI: 10.2147/copd.s266394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/19/2021] [Indexed: 11/23/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive lung condition affecting 10% of the global population over 45 years. Currently, there are no disease-modifying treatments, with current therapies treating only the symptoms of the disease. COPD is an inflammatory disease, with a high infiltration of leukocytes being found within the lung of COPD patients. These leukocytes, if not kept in check, damage the lung, leading to the pathophysiology associated with the disease. In this review, we focus on the main leukocytes found within the COPD lung, describing how the release of chemokines from the damaged epithelial lining recruits these cells into the lung. Once present, these cells become active and may be driven towards a more pro-inflammatory phenotype. These cells release their own subtypes of inflammatory mediators, growth factors and proteases which can all lead to airway remodeling, mucus hypersecretion and emphysema. Finally, we describe some of the current therapies and potential new targets that could be utilized to target aberrant leukocyte function in the COPD lung. Here, we focus on old therapies such as statins and corticosteroids, but also look at the emerging field of biologics describing those which have been tested in COPD already and potential new monoclonal antibodies which are under review.
Collapse
Affiliation(s)
- Jonathan R Baker
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Louise E Donnelly
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
24
|
Li M, Xu Y, Pu K, Fan J, Cheng Z, Chen H, Zhou L. Genetic polymorphisms of chemokine (C-X-C motif) ligand 10 gene associated with hepatitis B virus infection in a Chinese Han population. Int Immunopharmacol 2021; 98:107888. [PMID: 34153670 DOI: 10.1016/j.intimp.2021.107888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND AIMS Chemokine (C-X-C motif) ligand 10 (CXCL10) has been recently shown to be associated with inflammatory diseases. However, the association between the genetic variation of this gene and the susceptibility to hepatitis B virus (HBV) infection remains unclear, especially in children. This study aimed to investigate the relationship between CXCL10 polymorphisms and the risk of chronic HBV infection in a Chinese Han population. METHODS A two-stage case-control study of 1048 adults and 627 children was performed. A total of 5 tagging SNPs in CXCL10 were genotyped. Dual-Luciferase Reporter Assay was used to assess the effect of the rs4508917 polymorphism on transcriptional activity of CXCL10. RESULTS CXCL10 rs4508917 and rs4256246 polymorphisms were significantly associated with an increased risk of chronic HBV infection in Chinese Han adults (p = 0.036 and p = 0.033), of which rs4508917 AA genotype could increase the serum CXCL10 level (p = 0.014). In addition, the rs4508917 AA genotype was identified to facilitate HBV persistent infection (p = 0.017) and breakthrough infection (p = 0.013) in children. Subsequent functional analysis indicated that rs4508917 A allele could promote the transcriptional activity of CXCL10. Additionally, we observed that the rs4508917 A allele carriers (AA and AG genotypes) had a limited HBV viral load suppression in patients treated with nucleos(t)ide analogues (NAs). CONCLUSION The A allele of the CXCL10 rs4508917 may be a risk factor of the persistent HBV infection both in adults and children, which may influence the response to NAs treatment.
Collapse
Affiliation(s)
- Mengmeng Li
- Department of Epidemiology, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Ying Xu
- Department of Epidemiology, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Kexue Pu
- College of Medical Informatics, Chongqing Medical University, Chongqing, China
| | - Jie Fan
- Department of Epidemiology, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Zheng Cheng
- Department of Epidemiology, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Hao Chen
- Department of Epidemiology, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Li Zhou
- Department of Epidemiology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
25
|
Li L, Liu Y, Chiu C, Jin Y, Zhou W, Peng M, Chen LC, Sun Q, Gao J. A Regulatory Role of Chemokine Receptor CXCR3 in the Pathogenesis of Chronic Obstructive Pulmonary Disease and Emphysema. Inflammation 2021; 44:985-998. [PMID: 33415536 DOI: 10.1007/s10753-020-01393-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/11/2020] [Accepted: 12/03/2020] [Indexed: 10/22/2022]
Abstract
Chronic obstructive pulmonary disease (COPD)/pulmonary emphysema is driven by the dysregulated airway inflammation and primarily influenced by the interaction between cigarette smoking (CS) and the individual's susceptibility. The inflammation in COPD involves both innate and adaptive immunity. By binding to its specific ligands, chemokine receptor CXCR3 plays an important role in regulating tissue inflammation and damage. In acute animal model challenged with either CS or pathogens, CXCR3 knockout (KO) attenuated lung inflammation and pathology. However, the role of CXCR3 in CS-induced chronic airway inflammation and pulmonary emphysema remains unknown. In this present study, we investigated the effect of CXCR3 in CS-induced pulmonary emphysema in an animal model, and the association between CXCR3 single nucleotide polymorphisms (SNPs) and COPD susceptibility in human subjects. We found that after chronic exposure to side stream CS (SSCS) for 24 weeks, CXCR3 KO mice demonstrated significant airspace enlargement expressed by mean linear intercept (Lm) compared with the wild-type (WT) mice. Consistently, CXCR3 KO mice had significantly higher BAL fluid macrophages and neutrophils, TNFα, and lung homogenate MMP-9 and MMP-12. Through genetic analysis of CXCR3 polymorphisms in a cohort of COPD patients with Han Chinese ethnicity, one CXCR3 SNP, rs2280964, was found to be genetically related to COPD susceptibility. Furthermore, CXCR3 SNP rs2280964 was significantly associated with the levels of serum MMP-9 in COPD patients. Our data from both animal and human studies revealed a novel role of CXCR3 possibly via influencing MMP9 production in the pathogenesis and progression of CS-associated COPD/pulmonary emphysema.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Case-Control Studies
- China
- Disease Models, Animal
- Female
- Genetic Association Studies
- Genetic Predisposition to Disease
- Humans
- Lung/immunology
- Lung/metabolism
- Lung/pathology
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/metabolism
- Male
- Matrix Metalloproteinase 12/metabolism
- Matrix Metalloproteinase 9/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Middle Aged
- Neutrophils/immunology
- Neutrophils/metabolism
- Phenotype
- Polymorphism, Single Nucleotide
- Pulmonary Disease, Chronic Obstructive/genetics
- Pulmonary Disease, Chronic Obstructive/immunology
- Pulmonary Disease, Chronic Obstructive/metabolism
- Pulmonary Disease, Chronic Obstructive/pathology
- Pulmonary Emphysema/genetics
- Pulmonary Emphysema/immunology
- Pulmonary Emphysema/metabolism
- Pulmonary Emphysema/pathology
- Receptors, CXCR3/genetics
- Receptors, CXCR3/metabolism
- Tumor Necrosis Factor-alpha/metabolism
- Mice
Collapse
Affiliation(s)
- Lun Li
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Yi Liu
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
- Department of Respiratory Medicine, Civil Aviation General Hospital, Beijing, 100123, China
| | - Chin Chiu
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, NY, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA, USA
| | - Weixun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Min Peng
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Lung-Chi Chen
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, NY, USA
| | - Qinghua Sun
- Division of Environmental Health Sciences, College of Public Health, the Ohio State University, Columbus, OH, USA
| | - Jinming Gao
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
26
|
Reyes-García J, Montaño LM, Carbajal-García A, Wang YX. Sex Hormones and Lung Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:259-321. [PMID: 34019274 DOI: 10.1007/978-3-030-68748-9_15] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammation is a characteristic marker in numerous lung disorders. Several immune cells, such as macrophages, dendritic cells, eosinophils, as well as T and B lymphocytes, synthetize and release cytokines involved in the inflammatory process. Gender differences in the incidence and severity of inflammatory lung ailments including asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), lung cancer (LC), and infectious related illnesses have been reported. Moreover, the effects of sex hormones on both androgens and estrogens, such as testosterone (TES) and 17β-estradiol (E2), driving characteristic inflammatory patterns in those lung inflammatory diseases have been investigated. In general, androgens seem to display anti-inflammatory actions, whereas estrogens produce pro-inflammatory effects. For instance, androgens regulate negatively inflammation in asthma by targeting type 2 innate lymphoid cells (ILC2s) and T-helper (Th)-2 cells to attenuate interleukin (IL)-17A-mediated responses and leukotriene (LT) biosynthesis pathway. Estrogens may promote neutrophilic inflammation in subjects with asthma and COPD. Moreover, the activation of estrogen receptors might induce tumorigenesis. In this chapter, we summarize the most recent advances in the functional roles and associated signaling pathways of inflammatory cellular responses in asthma, COPD, PF, LC, and newly occurring COVID-19 disease. We also meticulously deliberate the influence of sex steroids on the development and progress of these common and severe lung diseases.
Collapse
Affiliation(s)
- Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico.,Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
27
|
Gerlza T, Nagele M, Gschwandtner M, Winkler S, Kungl A. Designing an improved T-cell mobilising CXCL10 mutant through enhanced GAG binding affinity. Protein Eng Des Sel 2020; 32:367-373. [DOI: 10.1093/protein/gzz043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/29/2019] [Indexed: 11/13/2022] Open
Abstract
Abstract
The chemokine CXCL10 is released by a plethora of cells, including immune and metastatic cancer cells, following stimulation with interferon-gamma. It acts via its GPC receptor on T-cells attracting them to various target tissues. Glycosaminoglycans (GAGs) are regarded as co-receptors of chemokines, which enable the establishment of a chemotactic gradient for target cell migration. We have engineered human CXCL10 towards improved T-cell mobilisation by implementing a single site-directed mutation N20K into the protein, which leads to a higher GAG binding affinity compared to the wild type. Interestingly, this mutation not only increased T-cell migration in a transendothelial migration assay, the mutant intensified T-cell chemotaxis also in a Boyden chamber set-up thereby indicating a strong role of T-cell-localised GAGs on leukocyte migration. A CXCL10 mutant with increased GAG-binding affinity could therefore potentially serve as a T-cell mobiliser in pathological conditions where the immune surveillance of the target tissue is impaired, as is the case for most solid tumors.
Collapse
Affiliation(s)
- Tanja Gerlza
- Institute of Pharmaceutical Sciences, Department of pharmaceutical chemistry, Karl-Franzens-University Graz, Universitätsplatz 1, Graz A-8010, Austria
| | - Michael Nagele
- Institute of Pharmaceutical Sciences, Department of pharmaceutical chemistry, Karl-Franzens-University Graz, Universitätsplatz 1, Graz A-8010, Austria
| | - Martha Gschwandtner
- Institute of Pharmaceutical Sciences, Department of pharmaceutical chemistry, Karl-Franzens-University Graz, Universitätsplatz 1, Graz A-8010, Austria
| | - Sophie Winkler
- Institute of Pharmaceutical Sciences, Department of pharmaceutical chemistry, Karl-Franzens-University Graz, Universitätsplatz 1, Graz A-8010, Austria
| | - Andreas Kungl
- Institute of Pharmaceutical Sciences, Department of pharmaceutical chemistry, Karl-Franzens-University Graz, Universitätsplatz 1, Graz A-8010, Austria
- Antagonis Biotherapeutics GmbH, Strasserhofweg 77a, Graz A-8045, Austria
| |
Collapse
|
28
|
Li Q, Sun J, Cao Y, Liu B, Li L, Mohammadtursun N, Zhang H, Dong J, Wu J. Bu-Shen-Fang-Chuan formula attenuates T-lymphocytes recruitment in the lung of rats with COPD through suppressing CXCL9/CXCL10/CXCL11-CXCR3 axis. Biomed Pharmacother 2019; 123:109735. [PMID: 31864210 DOI: 10.1016/j.biopha.2019.109735] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 12/20/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common respiratory disease characterized by irreversible airflow limitation. The current medications show limited effects on the decline of pulmonary function in COPD. Our multicenter clinical trial found that Bu-Shen-Fang-Chuan fomula (BSFCF), a Chinese herbal formula, markedly reduced the frequencies of acute exacerbation of COPD and delayed lung function decline. However, the underlying mechanisms are still unclear. In this study, we established a COPD rat model through a 6-month exposure to cigarette smoke (CS) and found that BSFCF (7.2 g/kg) effectively improved CS-induced reduction in pulmonary function and remarkably decreased the numbers of inflammatory cells in bronchoalveolar lavage fluid (BALF). Importantly, BSFCF treatment notably prevented the accumulation of T-lymphocytes (especially CD8+ T-cells) in the lung of COPD rats. RNA sequencing analysis of lung tissue demonstrated that CXCL9/CXCL10/CXCL11-CXCR3 chemokine axis in the lung of CS-exposed rats was significantly suppressed by BSFCF. Moreover, our Real-time PCR data verified that BSFCF evidently inhibited the mRNA expressions of CXCL9, CXCL10, CXCL11 and CXCR3. Conclusively, BSFCF markedly improved pulmonary function and attenuated CD8+ T-cells recruitment in the lung of CS-exposed rats, which were partially through inhibition of CXCL9/CXCL10/CXCL11-CXCR3 axis.
Collapse
Affiliation(s)
- Qiuping Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jing Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Yuxue Cao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Baojun Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Lulu Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Nabijan Mohammadtursun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Hu Zhang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China.
| | - Jinfeng Wu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
29
|
Soufan O, Ewald J, Viau C, Crump D, Hecker M, Basu N, Xia J. T1000: a reduced gene set prioritized for toxicogenomic studies. PeerJ 2019; 7:e7975. [PMID: 31681519 PMCID: PMC6824333 DOI: 10.7717/peerj.7975] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/02/2019] [Indexed: 12/12/2022] Open
Abstract
There is growing interest within regulatory agencies and toxicological research communities to develop, test, and apply new approaches, such as toxicogenomics, to more efficiently evaluate chemical hazards. Given the complexity of analyzing thousands of genes simultaneously, there is a need to identify reduced gene sets. Though several gene sets have been defined for toxicological applications, few of these were purposefully derived using toxicogenomics data. Here, we developed and applied a systematic approach to identify 1,000 genes (called Toxicogenomics-1000 or T1000) highly responsive to chemical exposures. First, a co-expression network of 11,210 genes was built by leveraging microarray data from the Open TG-GATEs program. This network was then re-weighted based on prior knowledge of their biological (KEGG, MSigDB) and toxicological (CTD) relevance. Finally, weighted correlation network analysis was applied to identify 258 gene clusters. T1000 was defined by selecting genes from each cluster that were most associated with outcome measures. For model evaluation, we compared the performance of T1000 to that of other gene sets (L1000, S1500, Genes selected by Limma, and random set) using two external datasets based on the rat model. Additionally, a smaller (T384) and a larger version (T1500) of T1000 were used for dose-response modeling to test the effect of gene set size. Our findings demonstrated that the T1000 gene set is predictive of apical outcomes across a range of conditions (e.g., in vitro and in vivo, dose-response, multiple species, tissues, and chemicals), and generally performs as well, or better than other gene sets available.
Collapse
Affiliation(s)
- Othman Soufan
- Institute of Parasitology, McGill University, Montreal, Canada
| | - Jessica Ewald
- Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Canada
| | - Charles Viau
- Institute of Parasitology, McGill University, Montreal, Canada
| | - Doug Crump
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, National Wildlife Research Centre, Carleton University, Ottawa, Canada
| | - Markus Hecker
- School of the Environment & Sustainability and Toxicology Centre, University of Saskatchewan, Saskatoon, Canada
| | - Niladri Basu
- Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Canada
| | - Jianguo Xia
- Institute of Parasitology, McGill University, Montreal, Canada.,Department of Animal Science, McGill University, Montreal, Canada
| |
Collapse
|
30
|
Hoffmann RF, Jonker MR, Brandenburg SM, de Bruin HG, Ten Hacken NHT, van Oosterhout AJM, Heijink IH. Mitochondrial dysfunction increases pro-inflammatory cytokine production and impairs repair and corticosteroid responsiveness in lung epithelium. Sci Rep 2019; 9:15047. [PMID: 31636329 PMCID: PMC6803636 DOI: 10.1038/s41598-019-51517-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 09/25/2019] [Indexed: 12/13/2022] Open
Abstract
COPD is characterized by chronic lung inflammation and irreversible lung tissue damage. Inhaled noxious gases, including cigarette smoke, are the major risk factor for COPD. Inhaled smoke first encounters the epithelial lining of the lungs, causing oxidative stress and mitochondrial dysfunction. We investigated whether a mitochondrial defect may contribute to increased lung epithelial pro-inflammatory responses, impaired epithelial repair and reduced corticosteroid sensitivity as observed in COPD. We used wild-type alveolar epithelial cells A549 and mitochondrial DNA-depleted A549 cells (A549 Rho-0) and studied pro-inflammatory responses using (multiplex) ELISA as well as epithelial barrier function and repair (real-time impedance measurements), in the presence and absence of the inhaled corticosteroid budesonide. We observed that A549 Rho-0 cells secrete higher levels of pro-inflammatory cytokines than wild-type A549 cells and display impaired repair upon wounding. Budesonide strongly suppressed the production of neutrophil attractant CXCL8, and promoted epithelial integrity in A549 wild-type cells, while A549 Rho-0 cells displayed reduced corticosteroid sensitivity compared to wild-type cells. The reduced corticosteroid responsiveness may be mediated by glycolytic reprogramming, specifically glycolysis-associated PI3K signaling, as PI3K inhibitor LY294002 restored the sensitivity of CXCL8 secretion to corticosteroids in A549 Rho-0 cells. In conclusion, mitochondrial defects may lead to increased lung epithelial pro-inflammatory responses, reduced epithelial repair and reduced corticosteroid responsiveness in lung epithelium, thus potentially contributing to the pathogenesis of COPD.
Collapse
Affiliation(s)
- R F Hoffmann
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - M R Jonker
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - S M Brandenburg
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - H G de Bruin
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - N H T Ten Hacken
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, The Netherlands
| | - A J M van Oosterhout
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - I H Heijink
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands.
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands.
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, The Netherlands.
| |
Collapse
|
31
|
Naz S, Bhat M, Ståhl S, Forsslund H, Sköld CM, Wheelock ÅM, Wheelock CE. Dysregulation of the Tryptophan Pathway Evidences Gender Differences in COPD. Metabolites 2019; 9:metabo9100212. [PMID: 31581603 PMCID: PMC6835831 DOI: 10.3390/metabo9100212] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 02/06/2023] Open
Abstract
Increased activity of indoleamine 2,3-dioxygenase (IDO) and tryptophan hydroxylase (TPH) have been reported in individuals with chronic obstructive pulmonary disease (COPD). We therefore investigated the effect of gender stratification upon the observed levels of tryptophan metabolites in COPD. Tryptophan, serotonin, kynurenine, and kynurenic acid were quantified in serum of never-smokers (n = 39), smokers (n = 40), COPD smokers (n = 27), and COPD ex-smokers (n = 11) by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). The individual metabolite associations with lung function, blood, and bronchoalveolar lavage (BAL) immune-cell composition, as well as chemokine and cytokine levels, were investigated. Stratification by gender and smoking status revealed that the observed alterations in kynurenine and kynurenic acid, and to a lesser extent serotonin, were prominent in males, irrespective of COPD status (kynurenine p = 0.005, kynurenic acid p = 0.009, and serotonin p = 0.02). Inferred serum IDO activity and kynurenine levels decreased in smokers relative to never-smokers (p = 0.005 and p = 0.004, respectively). In contrast, inferred tryptophan hydroxylase (TPH) activity and serotonin levels showed an increase with smoking that reached significance with COPD (p = 0.01 and p = 0.01, respectively). Serum IDO activity correlated with blood CXC chemokine ligand 9 (CXCL9, p = 0.0009, r = 0.93) and chemokine (C-C motif) ligand 4 (CCL4.(p = 0.04, r = 0.73) in female COPD smokers. Conversely, serum serotonin levels correlated with BAL CD4+ T-cells (%) (p = 0.001, r = 0.92) and CD8+ T-cells (%) (p = 0.002, r = -0.90) in female COPD smokers, but not in male COPD smokers (p = 0.1, r = 0.46 and p = 0.1, r = -0.50, respectively). IDO- and TPH-mediated tryptophan metabolites showed gender-based associations in COPD, which were primarily driven by smoking status.
Collapse
Affiliation(s)
- Shama Naz
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 171 77 Stockholm, Sweden;
| | - Maria Bhat
- Research and Development, Innovative Medicines, Personalised Healthcare and Biomarkers, Translational Science Centre, Science for Life Laboratory, AstraZeneca, SE 171 65 Solna, Sweden; (M.B.); (S.S.)
- Department of Clinical Neuroscience, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| | - Sara Ståhl
- Research and Development, Innovative Medicines, Personalised Healthcare and Biomarkers, Translational Science Centre, Science for Life Laboratory, AstraZeneca, SE 171 65 Solna, Sweden; (M.B.); (S.S.)
- Department of Clinical Neuroscience, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| | - Helena Forsslund
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, SE 171 77 Stockholm, Sweden; (H.F.); (C.M.S.)
| | - C. Magnus Sköld
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, SE 171 77 Stockholm, Sweden; (H.F.); (C.M.S.)
| | - Åsa M. Wheelock
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, SE 171 77 Stockholm, Sweden; (H.F.); (C.M.S.)
- Correspondence: (Å.M.W.); (C.E.W.); Tel.: +46-70-2200308 (Å.M.W.); +46-8-524-87630 (C.E.W.)
| | - Craig E. Wheelock
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 171 77 Stockholm, Sweden;
- Correspondence: (Å.M.W.); (C.E.W.); Tel.: +46-70-2200308 (Å.M.W.); +46-8-524-87630 (C.E.W.)
| |
Collapse
|
32
|
Landry ML, Foxman EF. Antiviral Response in the Nasopharynx Identifies Patients With Respiratory Virus Infection. J Infect Dis 2019; 217:897-905. [PMID: 29281100 PMCID: PMC5853594 DOI: 10.1093/infdis/jix648] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 12/12/2017] [Indexed: 12/19/2022] Open
Abstract
Background Despite the high burden of respiratory infection and the importance of early and accurate diagnosis, there is no simple diagnostic test to rule in viral infection as a cause of respiratory symptoms. Methods We performed RNA sequencing on human nasal epithelial cells following stimulation of the intracellular viral recognition receptor RIG-I. Next, we evaluated whether measuring identified host mRNAs and proteins from patient nasopharyngeal swabs could predict the presence of a respiratory virus in the sample. Results Our first study showed that a signature of 3 mRNAs, CXCL10, IFIT2, and OASL, predicted respiratory virus detection with an accuracy of 97% (95% confidence interval [CI], 0.9–1.0), and identified proteins correlating with virus detection. In a second study, elevated CXCL11 or CXCL10 protein levels identified samples containing respiratory viruses, including viruses not on the initial test panel. Overall area under the curve (AUC) values were: CXCL11 AUC = 0.901 (95% CI, 0.86–0.94); CXCL10 AUC = 0.85 (95% CI, 0.80–0.91). Conclusions Host antiviral mRNAs and single host proteins detectable using nasopharyngeal swabs accurately predict the presence of viral infection. This approach holds promise for developing rapid, cost-effective tests to improve management of patients with respiratory illnesses.
Collapse
Affiliation(s)
- Marie L Landry
- Department of Laboratory Medicine Yale University School of Medicine, New Haven, Connecticut.,Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Ellen F Foxman
- Department of Laboratory Medicine Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
33
|
Chemokines in COPD: From Implication to Therapeutic Use. Int J Mol Sci 2019; 20:ijms20112785. [PMID: 31174392 PMCID: PMC6600384 DOI: 10.3390/ijms20112785] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023] Open
Abstract
: Chronic Obstructive Pulmonary Disease (COPD) represents the 3rd leading cause of death in the world. The underlying pathophysiological mechanisms have been the focus of extensive research in the past. The lung has a complex architecture, where structural cells interact continuously with immune cells that infiltrate into the pulmonary tissue. Both types of cells express chemokines and chemokine receptors, making them sensitive to modifications of concentration gradients. Cigarette smoke exposure and recurrent exacerbations, directly and indirectly, impact the expression of chemokines and chemokine receptors. Here, we provide an overview of the evidence regarding chemokines involvement in COPD, and we hypothesize that a dysregulation of this tightly regulated system is critical in COPD evolution, both at a stable state and during exacerbations. Targeting chemokines and chemokine receptors could be highly attractive as a mean to control both chronic inflammation and bronchial remodeling. We present a special focus on the CXCL8-CXCR1/2, CXCL9/10/11-CXCR3, CCL2-CCR2, and CXCL12-CXCR4 axes that seem particularly involved in the disease pathophysiology.
Collapse
|
34
|
Cornwell WD, Kim C, Lastra AC, Dass C, Bolla S, Wang H, Zhao H, Ramsey FV, Marchetti N, Rogers TJ, Criner GJ. Inflammatory signature in lung tissues in patients with combined pulmonary fibrosis and emphysema. Biomarkers 2018; 24:232-239. [PMID: 30411980 DOI: 10.1080/1354750x.2018.1542458] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: The aetiology and inflammatory profile of combined pulmonary fibrosis and emphysema (CPFE) remain uncertain currently. Objective: We aimed to examine the levels of inflammatory proteins in lung tissue in a cohort of patients with emphysema, interstitial pulmonary fibrosis (IPF), and CPFE. Materials and methods: Explanted lungs were obtained from subjects with emphysema, IPF, CPFE, (or normal subjects), and tissue extracts were prepared. Thirty-four inflammatory proteins were measured in each tissue section. Results: The levels of all 34 proteins were virtually indistinguishable in IPF compared with CPFE tissues, and collectively, the inflammatory profile in the emphysematous tissues were distinct from IPF and CPFE. Moreover, inflammatory protein levels were independent of the severity of the level of diseased tissue. Conclusions: We find that emphysematous lung tissues have a distinct inflammatory profile compared with either IPF or CPFE. However, the inflammatory profile in CPFE lungs is essentially identical to lungs from patients with IPF. These data suggest that distinct inflammatory processes collectively contribute to the disease processes in patients with emphysema, when compared to IPF and CPFE.
Collapse
Affiliation(s)
- William D Cornwell
- a Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA.,b Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Cynthia Kim
- b Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Alejandra C Lastra
- b Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Chandra Dass
- c Department of Radiology, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Sudhir Bolla
- b Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - He Wang
- d Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Huaqing Zhao
- e Department of Clinical Sciences, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Frederick V Ramsey
- e Department of Clinical Sciences, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Nathaniel Marchetti
- a Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA.,b Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Thomas J Rogers
- a Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA.,b Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| | - Gerard J Criner
- a Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA.,b Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine , Temple University , Philadelphia , PA , USA
| |
Collapse
|
35
|
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
|
36
|
Aggarwal T, Wadhwa R, Thapliyal N, Sharma K, Rani V, Maurya PK. Oxidative, inflammatory, genetic, and epigenetic biomarkers associated with chronic obstructive pulmonary disorder. J Cell Physiol 2018; 234:2067-2082. [DOI: 10.1002/jcp.27181] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/17/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Taru Aggarwal
- Amity Institute of Biotechnology, Amity UniversityNoida India
| | - Ridhima Wadhwa
- Amity Institute of Biotechnology, Amity UniversityNoida India
| | | | - Kanishka Sharma
- Amity Education GroupOakdale, Long Island (Suffolk) New York
| | - Varsha Rani
- Amity Education GroupOakdale, Long Island (Suffolk) New York
| | - Pawan K. Maurya
- Amity Institute of Biotechnology, Amity UniversityNoida India
- Amity Education GroupOakdale, Long Island (Suffolk) New York
- Interdisciplinary Laboratory of Clinical Neuroscience (LINC), Department of PsychiatryFederal University of São PauloSão Paulo Brazil
| |
Collapse
|
37
|
Wei B, Sheng Li C. Changes in Th1/Th2-producing cytokines during acute exacerbation chronic obstructive pulmonary disease. J Int Med Res 2018; 46:3890-3902. [PMID: 29950127 PMCID: PMC6136028 DOI: 10.1177/0300060518781642] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Objective This study aimed to explore cytokine serum levels and the ratio of type 1 T helper (Th1)/Th2 cells in patients with acute exacerbation of chronic obstructive pulmonary disease (AECOPD). Methods A total 245 patients diagnosed with AECOPD and 193 patients who progressed to stable COPD after the initiation of treatment in hospital were selected, while a further 50 healthy individuals served as controls. All patients with COPD were diagnosed using Global Initiative for Chronic Obstructive Lung Disease criteria. Serum concentrations of interleukin (IL)-2, interferon (IFN)-γ, IL-4, IL-10, IL-17, and immunoglobulin (Ig)E were measured using enzyme-linked immunosorbent assays. Results AECOPD patients had higher levels of IL-2, IFN-γ, IL-4, IL-10, IL-17, and IgE than those with stable COPD or controls. Intriguingly, the ratios of Th1/Th2 and IL-17/IgE were lower in AECOPD patients compared with the other two groups. These data suggest that AECOPD patients produce more IgE and have more differentiated Th2 cells than other groups. Conclusion Our findings suggest that an imbalance of circulating CD4+ T cell subsets correlates with AECOPD, and that a shift of Th1/Th2 and IL-17/IgE ratios may be caused by increased Th2 cell production.
Collapse
Affiliation(s)
- Bing Wei
- 1 Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Chun Sheng Li
- 2 Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
38
|
Dysregulated Functions of Lung Macrophage Populations in COPD. J Immunol Res 2018; 2018:2349045. [PMID: 29670919 PMCID: PMC5835245 DOI: 10.1155/2018/2349045] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 11/29/2017] [Indexed: 01/02/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a diverse respiratory disease characterised by bronchiolitis, small airway obstruction, and emphysema. Innate immune cells play a pivotal role in the disease's progression, and in particular, lung macrophages exploit their prevalence and strategic localisation to orchestrate immune responses. To date, alveolar and interstitial resident macrophages as well as blood monocytes have been described in the lungs of patients with COPD contributing to disease pathology by changes in their functional repertoire. In this review, we summarise recent evidence from human studies and work with animal models of COPD with regard to altered functions of each of these myeloid cell populations. We primarily focus on the dysregulated capacity of alveolar macrophages to secrete proinflammatory mediators and proteases, induce oxidative stress, engulf microbes and apoptotic cells, and express surface and intracellular markers in patients with COPD. In addition, we discuss the differences in the responses between alveolar macrophages and interstitial macrophages/monocytes in the disease and propose how the field should advance to better understand the implications of lung macrophage functions in COPD.
Collapse
|
39
|
Kemény Á, Csekő K, Szitter I, Varga ZV, Bencsik P, Kiss K, Halmosi R, Deres L, Erős K, Perkecz A, Kereskai L, László T, Kiss T, Ferdinandy P, Helyes Z. Integrative characterization of chronic cigarette smoke-induced cardiopulmonary comorbidities in a mouse model. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 229:746-759. [PMID: 28648837 DOI: 10.1016/j.envpol.2017.04.098] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 02/24/2017] [Accepted: 04/06/2017] [Indexed: 12/16/2023]
Abstract
Cigarette smoke-triggered inflammatory cascades and consequent tissue damage are the main causes of chronic obstructive pulmonary disease (COPD). There is no effective therapy and the key mediators of COPD are not identified due to the lack of translational animal models with complex characterization. This integrative chronic study investigated cardiopulmonary pathophysiological alterations and mechanisms with functional, morphological and biochemical techniques in a 6-month-long cigarette smoke exposure mouse model. Some respiratory alterations characteristic of emphysema (decreased airway resistance: Rl; end-expiratory work and pause: EEW, EEP; expiration time: Te; increased tidal mid-expiratory flow: EF50) were detected in anaesthetized C57BL/6 mice, unrestrained plethysmography did not show changes. Typical histopathological signs were peribronchial/perivascular (PB/PV) edema at month 1, neutrophil/macrophage infiltration at month 2, interstitial leukocyte accumulation at months 3-4, and emphysema/atelectasis at months 5-6 quantified by mean linear intercept measurement. Emphysema was proven by micro-CT quantification. Leukocyte number in the bronchoalveolar lavage at month 2 and lung matrix metalloproteinases-2 and 9 (MMP-2/MMP-9) activities in months 5-6 significantly increased. Smoking triggered complex cytokine profile change in the lung with one characteristic inflammatory peak of C5a, interleukin-1α and its receptor antagonist (IL-1α, IL-1ra), monokine induced by gamma interferon (MIG), macrophage colony-stimulating factor (M-CSF), tissue inhibitor of matrix metalloproteinase-1 (TIMP-1) at months 2-3, and another peak of interferon-γ (IFN-γ), IL-4, 7, 13, 17, 27 related to tissue destruction. Transient systolic and diastolic ventricular dysfunction developed after 1-2 months shown by significantly decreased ejection fraction (EF%) and deceleration time, respectively. These parameters together with the tricuspid annular plane systolic excursion (TAPSE) decreased again after 5-6 months. Soluble intercellular adhesion molecule-1 (sICAM-1) significantly increased in the heart homogenates at month 6, while other inflammatory cytokines were undetectable. This is the first study demonstrating smoking duration-dependent, complex cardiopulmonary alterations characteristic to COPD, in which inflammatory cytokine cascades and MMP-2/9 might be responsible for pulmonary destruction and sICAM-1 for heart dysfunction.
Collapse
Affiliation(s)
- Ágnes Kemény
- Department of Pharmacology and Pharmacotherapy, University of Pécs, Faculty of Medicine, H-7624 Pécs, Szigeti út 12., Hungary; Department of Medical Biology, University of Pécs, Faculty of Medicine, H-7624 Pécs, Szigeti út 12., Hungary; Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Ifjúság útja 20., Hungary.
| | - Kata Csekő
- Department of Pharmacology and Pharmacotherapy, University of Pécs, Faculty of Medicine, H-7624 Pécs, Szigeti út 12., Hungary; Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Ifjúság útja 20., Hungary.
| | - István Szitter
- Department of Pharmacology and Pharmacotherapy, University of Pécs, Faculty of Medicine, H-7624 Pécs, Szigeti út 12., Hungary; Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Ifjúság útja 20., Hungary.
| | - Zoltán V Varga
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Faculty of Medicine, H-1089 Budapest, Nagyvárad tér 4., Hungary.
| | - Péter Bencsik
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Faculty of Medicine, H-6720 Szeged, Dóm tér 9., Hungary; Pharmahungary Group, H-6722 Szeged, Hajnóczy u. 6., Hungary.
| | - Krisztina Kiss
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Faculty of Medicine, H-6720 Szeged, Dóm tér 9., Hungary.
| | - Róbert Halmosi
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Ifjúság útja 20., Hungary; I(st) Department of Internal Medicine, University of Pécs, Faculty of Medicine, H-7624 Pécs, Ifjúság útja 13., Hungary.
| | - László Deres
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Ifjúság útja 20., Hungary; I(st) Department of Internal Medicine, University of Pécs, Faculty of Medicine, H-7624 Pécs, Ifjúság útja 13., Hungary.
| | - Krisztián Erős
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Ifjúság útja 20., Hungary; I(st) Department of Internal Medicine, University of Pécs, Faculty of Medicine, H-7624 Pécs, Ifjúság útja 13., Hungary; Department of Biochemistry and Medical Chemistry, University of Pécs, Faculty of Medicine, H-7624 Pécs, Szigeti út 12., Hungary.
| | - Anikó Perkecz
- Department of Pharmacology and Pharmacotherapy, University of Pécs, Faculty of Medicine, H-7624 Pécs, Szigeti út 12., Hungary.
| | - László Kereskai
- Department of Pathology, University of Pécs, Faculty of Medicine, H-7624 Pécs, Szigeti út 12., Hungary.
| | - Terézia László
- Department of Pathology, University of Pécs, Faculty of Medicine, H-7624 Pécs, Szigeti út 12., Hungary.
| | - Tamás Kiss
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Ifjúság útja 20., Hungary.
| | - Péter Ferdinandy
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Faculty of Medicine, H-1089 Budapest, Nagyvárad tér 4., Hungary; Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Faculty of Medicine, H-6720 Szeged, Dóm tér 9., Hungary; Pharmahungary Group, H-6722 Szeged, Hajnóczy u. 6., Hungary.
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, University of Pécs, Faculty of Medicine, H-7624 Pécs, Szigeti út 12., Hungary; Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Ifjúság útja 20., Hungary; MTA-PTE NAP B Chronic Pain Research Group, University of Pécs, Faculty of Medicine, H-7624 Pécs, Szigeti út 12., Hungary; PharmInVivo Ltd, H-7629 Pécs, Szondi György út 10., Hungary.
| |
Collapse
|
40
|
Cellular and molecular mechanisms of asthma and COPD. Clin Sci (Lond) 2017; 131:1541-1558. [PMID: 28659395 DOI: 10.1042/cs20160487] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 12/19/2022]
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) both cause airway obstruction and are associated with chronic inflammation of the airways. However, the nature and sites of the inflammation differ between these diseases, resulting in different pathology, clinical manifestations and response to therapy. In this review, the inflammatory and cellular mechanisms of asthma and COPD are compared and the differences in inflammatory cells and profile of inflammatory mediators are highlighted. These differences account for the differences in clinical manifestations of asthma and COPD and their response to therapy. Although asthma and COPD are usually distinct, there are some patients who show an overlap of features, which may be explained by the coincidence of two common diseases or distinct phenotypes of each disease. It is important to better understand the underlying cellular and molecular mechanisms of asthma and COPD in order to develop new treatments in areas of unmet need, such as severe asthma, curative therapy for asthma and effective anti-inflammatory treatments for COPD.
Collapse
|
41
|
Briend E, Ferguson GJ, Mori M, Damera G, Stephenson K, Karp NA, Sethi S, Ward CK, Sleeman MA, Erjefält JS, Finch DK. IL-18 associated with lung lymphoid aggregates drives IFNγ production in severe COPD. Respir Res 2017; 18:159. [PMID: 28830544 PMCID: PMC5568255 DOI: 10.1186/s12931-017-0641-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 08/10/2017] [Indexed: 11/16/2022] Open
Abstract
Background Increased interferon gamma (IFNγ) release occurs in Chronic Obstructive Pulmonary Disease (COPD) lungs. IFNγ supports optimal viral clearance, but if dysregulated could increase lung tissue destruction. Methods The present study investigates which mediators most closely correlate with IFNγ in sputum in stable and exacerbating disease, and seeks to shed light on the spatial requirements for innate production of IFNγ, as reported in mouse lymph nodes, to observe whether such microenvironmental cellular organisation is relevant to IFNγ production in COPD lung. Results We show tertiary follicle formation in severe disease alters the dominant mechanistic drivers of IFNγ production, because cells producing interleukin-18, a key regulator of IFNγ, are highly associated with such structures. Interleukin-1 family cytokines correlated with IFNγ in COPD sputum. We observed that the primary source of IL-18 in COPD lungs was myeloid cells within lymphoid aggregates and IL-18 was increased in severe disease. IL-18 released from infected epithelium or from activated myeloid cells, was more dominant in driving IFNγ when IL-18-producing and responder cells were in close proximity. Conclusions Unlike tight regulation to control infection spread in lymphoid organs, this local interface between IL-18-expressing and responder cell is increasingly supported in lung as disease progresses, increasing its potential to increase tissue damage via IFNγ. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0641-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emmanuel Briend
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.,Present address: Agenus Ltd, Cambridge, UK
| | | | - Michiko Mori
- Department of Experimental Medical Science, BMC D12, Lund University, SE-221 84, Lund, Sweden
| | - Gautam Damera
- MedImmune LLC, 1 MedImmune Way, Gaithersburg, MD, USA
| | - Katherine Stephenson
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.,Present address: University of Nottingham, Nottingham, UK
| | - Natasha A Karp
- Quantitative Biology IMED, AstraZeneca R&D, Cambridge, UK
| | - Sanjay Sethi
- Department of Medicine, University at Buffalo, 3495 Bailey Avenue, Buffalo, NY, 14215, USA
| | - Christine K Ward
- MedImmune LLC, 1 MedImmune Way, Gaithersburg, MD, USA.,Present address: Bristol-Myers Squibb, Princeton, NJ, USA
| | - Matthew A Sleeman
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.,Present address: Regeneron Pharmaceuticals Inc, Tarrytown, NY, USA
| | - Jonas S Erjefält
- Department of Experimental Medical Science, BMC D12, Lund University, SE-221 84, Lund, Sweden.,Department of Respiratory Medicine and Allergology, Lund University Hospital, Lund, Sweden
| | - Donna K Finch
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.
| |
Collapse
|
42
|
Carreño E, Portero A, Herreras JM, García-Vázquez C, Whitcup SM, Stern ME, Calonge M, Enríquez-de-Salamanca A. Cytokine and chemokine tear levels in patients with uveitis. Acta Ophthalmol 2017; 95:e405-e414. [PMID: 27873479 DOI: 10.1111/aos.13292] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 09/13/2016] [Indexed: 12/14/2022]
Abstract
PURPOSE To determine whether the levels of cytokines and chemokines in tears differ in uveitis patients and healthy subjects. METHODS Ninety-two uveitis patients (mean age 46.4 years) and 157 control healthy subjects (mean age 49.5 years) were recruited. Subjects with ocular surface diseases such as dry eye were excluded from the study. Using multiplex bead-based assays, tears (4 μl) were analysed for the concentration of interleukin (IL)-1β, IL-1RA, IL-2, IL-6, IL-7, IL-8/CXCL8, IL-10, IL-12p70, IL-15, IL-17A, IL-23, epidermal growth factor (EGF), fractalkine/CX3CL1, interferon-γ, IP-10/CXCL10, monocyte chemo-attractant protein (MCP)-1/CCL2, tumour necrosis factor-α, vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-β1, TGF-β2 and TGF-β3. Tear molecule levels were compared between the groups and among the different forms of uveitis and disease severity. RESULTS Epidermal growth factor, IL-1RA, IL-7, IL-8/CXCL8, IP-10/CXCL10, MCP-1/CCL2, TGF-β2 and VEGF were detected in more than 75% of the samples in both groups. Statistically significant differences in percentage of detection between control and patient groups were found for IL-23, IL-1β, IL-15, EGF, fractalkine/CX3CL1 and MCP-1/CCL2. The concentrations of IL-1RA, IL-8/CXCL8, fractalkine/CX3CL1, IP-10/CXCL10, VEGF and TGF-β2 in uveitis tear samples were elevated compared to controls (p < 0.05). Significant differences in tear levels of those molecules and also EGF were also present depending on the anatomic classification of uveitis. CONCLUSION There were significant differences in the levels of several cytokines and chemokines in tears of patients with uveitis compared with healthy subjects. These results can help understand the underlying pathophysiology of the uveitis and could potentially aid in diagnosis.
Collapse
Affiliation(s)
- Ester Carreño
- IOBA (Institute of Applied OphthalmoBiology); University of Valladolid; Valladolid Spain
| | - Alejandro Portero
- IOBA (Institute of Applied OphthalmoBiology); University of Valladolid; Valladolid Spain
| | - José M. Herreras
- IOBA (Institute of Applied OphthalmoBiology); University of Valladolid; Valladolid Spain
- University Clinic Hospital; Valladolid Spain
- CIBER-BBN (Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine); Valladolid Spain
| | - Carmen García-Vázquez
- IOBA (Institute of Applied OphthalmoBiology); University of Valladolid; Valladolid Spain
| | | | | | - Margarita Calonge
- IOBA (Institute of Applied OphthalmoBiology); University of Valladolid; Valladolid Spain
- CIBER-BBN (Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine); Valladolid Spain
| | - Amalia Enríquez-de-Salamanca
- IOBA (Institute of Applied OphthalmoBiology); University of Valladolid; Valladolid Spain
- CIBER-BBN (Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine); Valladolid Spain
| |
Collapse
|
43
|
COPD monocytes demonstrate impaired migratory ability. Respir Res 2017; 18:90. [PMID: 28494757 PMCID: PMC5425971 DOI: 10.1186/s12931-017-0569-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 05/01/2017] [Indexed: 12/20/2022] Open
Abstract
Background Increased lung macrophage numbers in COPD may arise from upregulation of blood monocyte recruitment into the lungs. CCR5 is a monocyte chemokine receptor regulated by interleukin-6 (IL-6); the concentration of CCR5 ligands are known to be elevated in COPD lungs. The objective of this study was to investigate mechanisms of monocyte recruitment to the lung in COPD, including the role of CCR5 signalling. Methods Ninety one COPD patients, 29 smokers (S) and 37 non-smokers (NS) underwent sputum induction, plasma sampling (to measure IL-6 and soluble IL-6 receptor [sIL-6R] by immunoassay), monocyte characterization (by flow cytometry) and monocyte isolation for cell migration and quantitative polymerase chain reaction studies. Lung tissue was used for immunohistochemistry. Results Plasma IL-6 and sIL-6R levels were increased in COPD. Greater proportions of COPD CD14++CD16+ monocytes expressed CCR5 compared to controls. Monocyte stimulation with IL-6 and sIL-6R increased CCR5 gene expression. COPD monocytes demonstrated impaired migration towards sputum supernatant compared to NS (% migration, 4.4 vs 11.5, respectively; p < 0.05). Pulmonary microvessels showed reduced monocyte recruitment (% marginated cells) in COPD compared to NS, (9.3% vs 83.1%, respectively). The proportion of replicating Ki67+ alveolar macrophages was reduced in COPD compared to NS. All alveolar macrophages from COPD and S expressed the anti-apoptosis marker BCL2; this protein was not present in non-smokers or COPD ex-smokers. Conclusion COPD monocytes show decreased migratory ability despite increased CCR5 expression. Increased COPD lung macrophage numbers may be due to delayed apoptosis. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0569-y) contains supplementary material, which is available to authorized users.
Collapse
|
44
|
Ishii T, Hosoki K, Nikura Y, Yamashita N, Nagase T, Yamashita N. IFN Regulatory Factor 3 Potentiates Emphysematous Aggravation by Lipopolysaccharide. THE JOURNAL OF IMMUNOLOGY 2017; 198:3637-3649. [PMID: 28363903 DOI: 10.4049/jimmunol.1601069] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 02/23/2017] [Indexed: 11/19/2022]
Abstract
Acute exacerbation of chronic obstructive pulmonary disease (COPD) is often induced by infection and often has a poor prognosis. Bacterial LPS activates innate immune receptor TLR4 followed by activation of a transcriptional factor IFN regulatory factor-3 (IRF3) as well as NF-κB, resulting in upregulation of various inflammatory mediators. To clarify the role of IRF3 in the pathogenesis of LPS-triggered COPD exacerbation, porcine pancreatic elastase (PPE) followed by LPS was administered intranasally to wild-type (WT) or IRF3-/- male mice. Sequential quantitative changes in emphysema were evaluated by microcomputed tomography, and lung histology was evaluated at the sixth week. WT mice treated with PPE and LPS exhibited enlarged alveolar spaces, whereas this feature was attenuated in similarly treated IRF3-/- mice. Moreover, LPS-induced emphysema aggravation was detected only in WT mice. Analysis of acute inflammation induced by PPE plus LPS revealed that the lungs of treated IRF3-/- mice had decreased mRNA transcripts for MCP-1, MIP-1α, TNF-α, and IFN-γ-inducible protein-10 but had increased neutrophils. IRF3 was involved in the production of mediators from macrophages, alveolar epithelial cells, and neutrophils. Furthermore, compared with isolated WT neutrophils from inflamed lung, those of IRF3-/- neutrophils exhibited impaired autophagic activation, phagocytosis, and apoptosis. These results suggest that IRF3 accelerated emphysema formation based on distinct profiles of mediators involved in LPS-induced COPD exacerbation. Regulation of the IRF3 pathway can affect multiple cell types and contribute to ameliorate pathogenesis of infection-triggered exacerbation of COPD.
Collapse
Affiliation(s)
- Takashi Ishii
- Department of Pharmacotherapy, Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo 202-8585, Japan.,Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan; and
| | - Keisuke Hosoki
- Department of Pharmacotherapy, Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo 202-8585, Japan.,Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan; and
| | - Yuichi Nikura
- Department of Pharmacotherapy, Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo 202-8585, Japan
| | - Naohide Yamashita
- Department of Advanced Medical Science, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Takahide Nagase
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan; and
| | - Naomi Yamashita
- Department of Pharmacotherapy, Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo 202-8585, Japan;
| |
Collapse
|
45
|
Hayney MS, Henriquez KM, Barnet JH, Ewers T, Champion HM, Flannery S, Barrett B. Serum IFN-γ-induced protein 10 (IP-10) as a biomarker for severity of acute respiratory infection in healthy adults. J Clin Virol 2017; 90:32-37. [PMID: 28334685 PMCID: PMC5408957 DOI: 10.1016/j.jcv.2017.03.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 02/20/2017] [Accepted: 03/05/2017] [Indexed: 01/08/2023]
Abstract
Serum IP-10 concentrations from 225 ARI episodes correlated with ARI severity for the episode. IP-10 concentrations varied with the viral pathogen that was identified. IP-10 may be a biomarker for ARI severity and for presence of a viral pathogen.
Background The inflammatory chemokine, interferon-gamma inducible protein of 10 kDa (IP-10), is a biomarker associated with several conditions. Objectives This study investigated serum concentrations of IP-10 in healthy individuals who developed acute respiratory infection (ARI). The hypothesis is that serum IP-10 concentrations correlate with ARI severity and detection of viral pathogens. Study design Data come from a randomized controlled trial measuring the effects of mindfulness meditation or exercise on ARI (Clinical Trials ID: NCT01654289). Healthy adults ages 30–69 were followed for a single season for ARI incidence and severity. This trial is ongoing, and the investigators are still blinded. When a participant reported ARI symptoms, nasal swab and lavage for PCR-based viral identification and blood samples were collected within the first 72 h of ARI symptoms. Serum IP-10 concentrations were measured by ELISA (R&D Systems, Inc., Quantikine ELISA, Minneapolis, MN). ARI severity was measured using the validated Wisconsin Upper Respiratory Symptom Survey (WURSS-24) until the ARI episode resolved. Results Serum IP-10 concentrations from 225 ARI episodes correlated with ARI global severity (rho 0.28 [95% CI: 0.15–0.39]; p < 0.001). IP-10 concentrations were higher with an ARI in which a viral pathogen was detected compared to no viral pathogen detected (median 366 pg/ml [IQR: 227–486] vs 163 pg/ml [IQR: 127–295], p < 0.0001). Influenza infections had higher IP-10 concentrations than coronavirus, enterovirus or rhinovirus, and paramyxovirus. Conclusion Serum IP-10 concentration correlates with ARI global severity. Also, IP-10 concentration measured early in the course of the ARI correlates with the daily severity, duration, and illness symptoms.
Collapse
Affiliation(s)
- Mary S Hayney
- School of Pharmacy, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States.
| | - Kelsey M Henriquez
- School of Pharmacy, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| | - Jodi H Barnet
- Department of Family Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| | - Tola Ewers
- Department of Family Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| | - Heather M Champion
- School of Pharmacy, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| | - Sean Flannery
- School of Pharmacy, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| | - Bruce Barrett
- Department of Family Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
46
|
Forsslund H, Yang M, Mikko M, Karimi R, Nyrén S, Engvall B, Grunewald J, Merikallio H, Kaarteenaho R, Wahlström J, Wheelock ÅM, Sköld CM. Gender differences in the T-cell profiles of the airways in COPD patients associated with clinical phenotypes. Int J Chron Obstruct Pulmon Dis 2016; 12:35-48. [PMID: 28053515 PMCID: PMC5191844 DOI: 10.2147/copd.s113625] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
T lymphocytes are believed to play an important role in the pathogenesis of chronic obstructive pulmonary disease (COPD). How T cells are recruited to the lungs and contribute to the inflammatory process is largely unknown. COPD is a heterogeneous disease, and discriminating disease phenotypes based on distinct molecular and cellular pathways may provide new approaches for individualized diagnosis and therapies. Bronchoalveolar lavage (BAL) and blood samples were obtained from 40 never-smokers, 40 smokers with normal lung function, and 38 COPD patients. T-cell chemokine receptor expression was analyzed with flow cytometry, and soluble BAL cytokines and chemokines were measured using a cytokine multiplex assay. Correlations with gender and clinical characteristics including lung imaging were investigated using multivariate modeling. Th1/Tc1- and Th2/Tc2-associated soluble analytes and T-cell chemokine receptors were analyzed as cumulative Th1/Tc1 and Th2/Tc2 immune responses. A higher expression of chemokine receptor CCR5 on CD8+ T cells in BAL and higher percentage of CXCR3+CD8+ T cells in blood was found in female smokers with COPD compared to those without COPD. CCR5 expression on CD4+ and CD8+ T cells was lower in BAL from male smokers with COPD compared to those without COPD. Among female smokers with COPD, Th1/Tc1 immune response was linked to BAL macrophage numbers and goblet cell density, and Th2/Tc2 response was associated with the measures of emphysema on high-resolution computed tomography. The highly gender-dependent T-cell profile in COPD indicates different links between cellular events and clinical manifestations in females compared to males. Our findings may reveal mechanisms of importance for the difference in clinical course in female COPD patients compared to males.
Collapse
Affiliation(s)
- Helena Forsslund
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - Mingxing Yang
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - Mikael Mikko
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - Reza Karimi
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - Sven Nyrén
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Benita Engvall
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - Johan Grunewald
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - Heta Merikallio
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit; Respiratory Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Riitta Kaarteenaho
- Respiratory Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland; Unit of Medicine and Clinical Research, Pulmonary Division, University of Eastern Finland; Center for Medicine and Clinical Research, Division of Respiratory Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Jan Wahlström
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - Åsa M Wheelock
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| | - C Magnus Sköld
- Department of Medicine Solna and Centre for Molecular Medicine, Respiratory Medicine Unit
| |
Collapse
|
47
|
Barnes PJ. Inflammatory mechanisms in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol 2016; 138:16-27. [PMID: 27373322 DOI: 10.1016/j.jaci.2016.05.011] [Citation(s) in RCA: 899] [Impact Index Per Article: 112.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 12/15/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is associated with chronic inflammation affecting predominantly the lung parenchyma and peripheral airways that results in largely irreversible and progressive airflow limitation. This inflammation is characterized by increased numbers of alveolar macrophages, neutrophils, T lymphocytes (predominantly TC1, TH1, and TH17 cells), and innate lymphoid cells recruited from the circulation. These cells and structural cells, including epithelial and endothelial cells and fibroblasts, secrete a variety of proinflammatory mediators, including cytokines, chemokines, growth factors, and lipid mediators. Although most patients with COPD have a predominantly neutrophilic inflammation, some have an increase in eosinophil counts, which might be orchestrated by TH2 cells and type 2 innate lymphoid cells though release of IL-33 from epithelial cells. These patients might be more responsive to corticosteroids and bronchodilators. Oxidative stress plays a key role in driving COPD-related inflammation, even in ex-smokers, and might result in activation of the proinflammatory transcription factor nuclear factor κB (NF-κB), impaired antiprotease defenses, DNA damage, cellular senescence, autoantibody generation, and corticosteroid resistance though inactivation of histone deacetylase 2. Systemic inflammation is also found in patients with COPD and can worsen comorbidities, such as cardiovascular diseases, diabetes, and osteoporosis. Accelerated aging in the lungs of patients with COPD can also generate inflammatory protein release from senescent cells in the lung. In the future, it will be important to recognize phenotypes of patients with optimal responses to more specific therapies, and development of biomarkers that identify the therapeutic phenotypes will be important.
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart and Lung Institute, Imperial College, London, United Kingdom.
| |
Collapse
|
48
|
Admas TH, Bernat V, Heinrich MR, Tschammer N. Development of Photoactivatable Allosteric Modulators for the Chemokine Receptor CXCR3. ChemMedChem 2016; 11:575-84. [PMID: 26880380 DOI: 10.1002/cmdc.201500573] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Indexed: 11/05/2022]
Abstract
The CXCR3 receptor, a class A G protein-coupled receptor (GPCR), is involved in the regulation and trafficking of various immune cells. CXCR3 antagonists have been proposed to be beneficial for the treatment of a wide range of disorders including but not limited to inflammatory and autoimmune diseases. The structure-based design of CXCR3 ligands remains, however, hampered by a lack of structural information describing in detail the interactions between an allosteric ligand and the receptor. We designed and synthesized photoactivatable probes for the structural and functional characterization, using photoaffinity labeling followed by mass spectrometry, of the CXCR3 allosteric binding pocket of AMG 487 and RAMX3, two potent and selective CXCR3 negative allosteric modulators. Photoaffinity labeling is a common approach to elucidate binding modes of small-molecule ligands of GPCRs through the aid of photoactivatable probes that convert to extremely reactive intermediates upon photolysis. The photolabile probe N-[({1-[3-(4-ethoxyphenyl)-4-oxo-3,4-dihydropyrido[2,3-d]pyrimidin-2-yl]ethyl}-2-[4-fluoro-3-(trifluoromethyl)phenyl]-N-{1-[4-(3-(trifluoromethyl)-3H-diazirin-3-yl]benzyl}piperidin-4-yl)methyl]acetamide (10) showed significant labeling of the CXCR3 receptor (80%) in a [(3) H]RAMX3 radioligand displacement assay. Compound 10 will serve as an important tool compound for the detailed investigation of the binding pocket of CXCR3 by mass spectrometry.
Collapse
Affiliation(s)
- Tizita Haimanot Admas
- Department of Chemistry & Pharmacy, Emil Fischer Center, Friedrich Alexander University Erlangen-Nürnberg, Schuhstr. 19, 91052, Erlangen, Germany
| | - Viachaslau Bernat
- Department of Chemistry & Pharmacy, Emil Fischer Center, Friedrich Alexander University Erlangen-Nürnberg, Schuhstr. 19, 91052, Erlangen, Germany.,Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, 3A1, 33458, Jupiter, FL, USA
| | - Markus R Heinrich
- Department of Chemistry & Pharmacy, Emil Fischer Center, Friedrich Alexander University Erlangen-Nürnberg, Schuhstr. 19, 91052, Erlangen, Germany
| | - Nuska Tschammer
- Department of Chemistry & Pharmacy, Emil Fischer Center, Friedrich Alexander University Erlangen-Nürnberg, Schuhstr. 19, 91052, Erlangen, Germany. .,NanoTemper Technologies GmbH, Flößergasse 4, 81369, München, Germany.
| |
Collapse
|
49
|
de Oliveira MR. Phloretin-induced cytoprotective effects on mammalian cells: A mechanistic view and future directions. Biofactors 2016; 42:13-40. [PMID: 26826024 DOI: 10.1002/biof.1256] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/19/2015] [Indexed: 11/11/2022]
Abstract
Phloretin (C15 H14 O5 ), a dihydrochalcone flavonoid, is mainly found in fruit, leaves, and roots of apple tree. Phloretin exerts antioxidant, anti-inflammatory, and anti-tumor activities in mammalian cells through mechanisms that have been partially elucidated throughout the years. Phloretin bioavailability is well known in humans, but still remains to be better studied in experimental animals, such as mouse and rat. The focus of the present review is to gather information regarding the mechanisms involved in the phloretin-elicited effects in different in vitro and in vivo experimental models. Several manuscripts were analyzed and data raised by authors were described and discussed here in a mechanistic manner. Comparisons between the effects elicited by phloretin and phloridzin were made whenever possible, as well as with other polyphenols, clarifying questions about the use of phloretin as a potential therapeutic agent. Toxicological aspects associated to phloretin exposure were also discussed here. Furthermore, a special section containing future directions was created as a suggestive guide towards the elucidation of phloretin-related actions in mammalian cells and tissues.
Collapse
Affiliation(s)
- Marcos Roberto de Oliveira
- Department of Chemistry/ICET, Postgraduate Program in Chemistry (PPGQ), Federal University of Mato Grosso (UFMT), CEP, Cuiaba, MT, Brazil
| |
Collapse
|
50
|
Abstract
Chemokines and their receptors are known to play important roles in disease. More than 40 chemokine ligands and 20 chemokine receptors have been identified, but, to date, only two small molecule chemokine receptor antagonists have been approved by the FDA. The chemokine receptor CXCR3 was identified in 1996, and nearly 20 years later, new areas of CXCR3 disease biology continue to emerge. Several classes of small molecule CXCR3 antagonists have been developed, and two have shown efficacy in preclinical models of inflammatory disease. However, only one CXCR3 antagonist has been evaluated in clinical trials, and there remain many opportunities to further investigate known classes of CXCR3 antagonists and to identify new chemotypes. This Perspective reviews the known CXCR3 antagonists and considers future opportunities for the development of small molecules for clinical evaluation.
Collapse
Affiliation(s)
- Stephen P Andrews
- Heptares Therapeutics , BioPark, Broadwater Road, Welwyn Garden City, AL7 3AX, United Kingdom
| | - Rhona J Cox
- Respiratory, Inflammation & Autoimmunity iMed, AstraZeneca, Respiratory, Inflammation & Autoimmunity IMED , Pepparedsleden, 431 83 Mölndal, Sweden
| |
Collapse
|