1
|
Mineiro PCDO, Fraga-Junior VDS, Cardoso ADOP, Waters CM, Takiya CM, Benjamim CF, Valenca HDM, Lanzetti M, Moraes JA, Valenca SS. ASK1 inhibition by selonsertib attenuates elastase-induced emphysema in mice. Life Sci 2025; 372:123600. [PMID: 40189195 DOI: 10.1016/j.lfs.2025.123600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide, with its most severe form being pulmonary emphysema, for which no effective treatment currently exists. Apoptosis signal-regulating kinase 1 (ASK1) has been implicated in lung inflammation and injury. Here, we investigated the experimental treatment of elastase-induced emphysema in mice with selonsertib, an ASK1 inhibitor. Animals received intratracheal elastase and were subsequently treated with intranasal selonsertib at different doses. On day 21, bronchoalveolar lavage fluid and lung tissues were collected for histological and biochemical analyses. Results showed that elastase-instilled mice developed pulmonary emphysema, whereas treatment with selonsertib at a dose of 2 mg/kg significantly reduced mean alveolar diameter. Moreover, higher doses of selonsertib were effective in reducing inflammatory cytokines (CX3CL1, IL-6, CCL2, and IL-1β), reactive oxygen species, and apoptosis. These findings suggest that ASK1 plays a critical role in the development of elastase-induced emphysema in mice and could be a target for COPD treatment.
Collapse
Affiliation(s)
| | | | - Aline de Oliveira Pontes Cardoso
- Universidade Federal do Rio de Janeiro, Instituto de Microbiologia Paulo de Góes (Programa de Pós-graduação em Imunologia e Inflamação), Rio de Janeiro, RJ, Brazil
| | - Christopher Mark Waters
- Augusta University, Medical College of Georgia (Department of Physiology), Augusta, GA, USA.
| | - Christina Maeda Takiya
- Universidade Federal do Rio de Janeiro, Instituto de Biofísica Carlos Chagas Filho, Rio de Janeiro, RJ, Brazil.
| | - Cláudia Farias Benjamim
- Universidade Federal do Rio de Janeiro, Instituto de Biofísica Carlos Chagas Filho, Rio de Janeiro, RJ, Brazil.
| | - Helber da Maia Valenca
- Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Rio de Janeiro, RJ, Brazil
| | - Manuella Lanzetti
- Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Rio de Janeiro, RJ, Brazil.
| | - J A Moraes
- Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Rio de Janeiro, RJ, Brazil.
| | - S S Valenca
- Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
2
|
Chen Z, Gao N, Wang X, Chen X, Zeng Y, Li C, Yang X, Cai Q, Wang X. Shared genetic aetiology of respiratory diseases: a genome-wide multitraits association analysis. BMJ Open Respir Res 2024; 11:e002148. [PMID: 38834332 PMCID: PMC11163672 DOI: 10.1136/bmjresp-2023-002148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 05/10/2024] [Indexed: 06/06/2024] Open
Abstract
OBJECTIVE This study aims to explore the common genetic basis between respiratory diseases and to identify shared molecular and biological mechanisms. METHODS This genome-wide pleiotropic association study uses multiple statistical methods to systematically analyse the shared genetic basis between five respiratory diseases (asthma, chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, lung cancer and snoring) using the largest publicly available genome wide association studies summary statistics. The missions of this study are to evaluate global and local genetic correlations, to identify pleiotropic loci, to elucidate biological pathways at the multiomics level and to explore causal relationships between respiratory diseases. Data were collected from 27 November 2022 to 30 March 2023 and analysed from 14 April 2023 to 13 July 2023. MAIN OUTCOMES AND MEASURES The primary outcomes are shared genetic loci, pleiotropic genes, biological pathways and estimates of genetic correlations and causal effects. RESULTS Significant genetic correlations were found for 10 paired traits in 5 respiratory diseases. Cross-Phenotype Association identified 12 400 significant potential pleiotropic single-nucleotide polymorphism at 156 independent pleiotropic loci. In addition, multitrait colocalisation analysis identified 15 colocalised loci and a subset of colocalised traits. Gene-based analyses identified 432 potential pleiotropic genes and were further validated at the transcriptome and protein levels. Both pathway enrichment and single-cell enrichment analyses supported the role of the immune system in respiratory diseases. Additionally, five pairs of respiratory diseases have a causal relationship. CONCLUSIONS AND RELEVANCE This study reveals the common genetic basis and pleiotropic genes among respiratory diseases. It provides strong evidence for further therapeutic strategies and risk prediction for the phenomenon of respiratory disease comorbidity.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University Department of Thoracic Surgery, Changsha, Hunan, China
| | - Ning Gao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xuanye Wang
- Department of Oncology, Xi'an Jiaotong University Second Affiliated Hospital Department of Oncology, Xi'an, Shaanxi, China
| | - Xiangming Chen
- Department of Orthopaedics, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - YaQi Zeng
- Department of Psychiatry, Brain Hospital of Hunan Province, Changsha, Hunan, China
| | - Cong Li
- Department of Radiology, The Second Xiangya Hospital of Central South University Department of Radiology, Changsha, Hunan, China
| | - Xiahong Yang
- Department of Anesthesiology, The Second Xiangya Hospital of Central South University Department of Anesthesiology, Changsha, Hunan, China
| | - Qidong Cai
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University Department of Thoracic Surgery, Changsha, Hunan, China
| | - Xiang Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University Department of Thoracic Surgery, Changsha, Hunan, China
| |
Collapse
|
3
|
Janho dit Hreich S, Juhel T, Leroy S, Ghinet A, Brau F, Hofman V, Hofman P, Vouret-Craviari V. Activation of the P2RX7/IL-18 pathway in immune cells attenuates lung fibrosis. eLife 2024; 12:RP88138. [PMID: 38300690 PMCID: PMC10945561 DOI: 10.7554/elife.88138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is an aggressive interstitial lung disease associated with progressive and irreversible deterioration of respiratory functions that lacks curative therapies. Despite IPF being associated with a dysregulated immune response, current antifibrotics aim only at limiting fibroproliferation. Transcriptomic analyses show that the P2RX7/IL18/IFNG axis is downregulated in IPF patients and that P2RX7 has immunoregulatory functions. Using our positive modulator of P2RX7, we show that activation of the P2RX7/IL-18 axis in immune cells limits lung fibrosis progression in a mouse model by favoring an antifibrotic immune environment, with notably an enhanced IL-18-dependent IFN-γ production by lung T cells leading to a decreased production of IL-17 and TGFβ. Overall, we show the ability of the immune system to limit lung fibrosis progression by targeting the immunomodulator P2RX7. Hence, treatment with a small activator of P2RX7 may represent a promising strategy to help patients with lung fibrosis.
Collapse
Affiliation(s)
| | - Thierry Juhel
- Université Côte d’Azur, CNRS, INSERM, IRCANNiceFrance
| | - Sylvie Leroy
- FHU OncoAgeNiceFrance
- Université Côte d'Azur, CNRS, Institut Pharmacologie Moléculaire et CellulaireSophia-AntipolisFrance
- Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Pneumology DepartmentNiceFrance
| | - Alina Ghinet
- Inserm U995, LIRIC, Université de Lille, CHRU de Lille, Faculté de médecine – Pôle recherche, Place VerdunLilleFrance
- Hautes Etudes d’Ingénieur (HEI), JUNIA Hauts-de-France, UCLille, Laboratoire de chimie durable et santéLilleFrance
- ‘Al. I. Cuza’ University of Iasi, Faculty of ChemistryIasiRomania
| | - Frederic Brau
- Université Côte d'Azur, CNRS, Institut Pharmacologie Moléculaire et CellulaireSophia-AntipolisFrance
| | - Veronique Hofman
- Université Côte d’Azur, CNRS, INSERM, IRCANNiceFrance
- FHU OncoAgeNiceFrance
- Laboratory of Clinical and Experimental Pathology and Biobank, Pasteur HospitalNiceFrance
- Hospital-Related Biobank (BB-0033-00025), Pasteur HospitalNiceFrance
| | - Paul Hofman
- Université Côte d’Azur, CNRS, INSERM, IRCANNiceFrance
- FHU OncoAgeNiceFrance
- Laboratory of Clinical and Experimental Pathology and Biobank, Pasteur HospitalNiceFrance
- Hospital-Related Biobank (BB-0033-00025), Pasteur HospitalNiceFrance
| | | |
Collapse
|
4
|
Rajasekar N, Gandhi D, Sivanantham A, Ravikumar V, Raj D, Paramasivam SG, Mukhopadhyay S, Rajasekaran S. Dietary tannic acid attenuates elastase-induced pulmonary inflammation and emphysema in mice. Inflammopharmacology 2024; 32:747-761. [PMID: 37947914 DOI: 10.1007/s10787-023-01381-z] [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: 09/07/2023] [Accepted: 10/13/2023] [Indexed: 11/12/2023]
Abstract
Emphysema is one of the major components of chronic obstructive pulmonary disease (COPD), which is characterised by the destruction and enlargement of air spaces, leading to airflow limitation and dyspnoea, finally progressing to oxygen dependency. The alveolar wall destruction is due to chronic inflammation, oxidative stress, apoptosis, and proteinase/anti-proteinase imbalance. So far, there has been no effective therapy for patients with COPD. We evaluated the therapeutic efficacy of tannic acid (TA), a naturally occurring plant-derived polyphenol in the murine emphysema model. In C57BL/6 J mice, we established emphysema by intratracheal instillation of elastase (EL). Then, mice were treated with TA and evaluated 1 and 21 days post-EL instillation. After 24 h, TA treatment significantly reduced EL-induced histopathological alterations, infiltrating leukocytes, and gene expression of markers of inflammation and apoptosis. Similarly, after 21 days, TA treatment suppressed the mean linear intercept, gene expression of proteinases, and increased elastic fiber contents in the lungs when compared to the EL-alone group. Furthermore, EL induced the activation of p38 mitogen-activated protein kinase (MAPK) and nuclear factor kappa light chain enhancer of activated B cells (NF-kB) p65 pathways in the lungs was suppressed by TA treatment. In summary, TA has the potential to mitigate EL-induced inflammation, apoptosis, proteinase/anti-proteinase imbalance, and subsequent emphysema in mice.
Collapse
Affiliation(s)
- Nandhine Rajasekar
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu 620024, India
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Deepa Gandhi
- Division of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, 462030, India
| | - Ayyanar Sivanantham
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu 620024, India
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA, 02118, USA
| | - Vilwanathan Ravikumar
- Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620024, India
| | - Dharma Raj
- Division of Biostatistics and Bioinformatics, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh 462030, India
| | | | - Sramana Mukhopadhyay
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh 462026, India
| | - Subbiah Rajasekaran
- Division of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, 462030, India.
| |
Collapse
|
5
|
Gairola S, Sinha A, Kaundal RK. Linking NLRP3 inflammasome and pulmonary fibrosis: mechanistic insights and promising therapeutic avenues. Inflammopharmacology 2024; 32:287-305. [PMID: 37991660 DOI: 10.1007/s10787-023-01389-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/25/2023] [Indexed: 11/23/2023]
Abstract
Pulmonary fibrosis is a devastating disorder distinguished by redundant inflammation and matrix accumulation in the lung interstitium. The early inflammatory cascade coupled with recurring tissue injury orchestrates a set of events marked by perturbed matrix hemostasis, deposition of matrix proteins, and remodeling in lung tissue. Numerous investigations have corroborated a direct correlation between the NLR family pyrin domain-containing 3 (NLRP3) activation and the development of pulmonary fibrosis. Dysregulated activation of NLRP3 within the pulmonary microenvironment exacerbates inflammation and may incite fibrogenic responses. Nevertheless, the precise mechanisms through which the NLRP3 inflammasome elicits pro-fibrogenic responses remain inadequately defined. Contemporary findings suggest that the pro-fibrotic consequences stemming from NLRP3 signaling primarily hinge on the action of interleukin-1β (IL-1β). IL-1β instigates IL-1 receptor signaling, potentiating the activity of transforming growth factor-beta (TGF-β). This signaling cascade, in turn, exerts influence over various transcription factors, including SNAIL, TWIST, and zinc finger E-box-binding homeobox 1 (ZEB 1/2), which collectively foster myofibroblast activation and consequent lung fibrosis. Here, we have connected the dots to illustrate how the NLRP3 inflammasome orchestrates a multitude of signaling events, including the activation of transcription factors that facilitate myofibroblast activation and subsequent lung remodeling. In addition, we have highlighted the prominent role played by various cells in the formation of myofibroblasts, the primary culprit in lung fibrosis. We also provided a concise overview of various compounds that hold the potential to impede NLRP3 inflammasome signaling, thus offering a promising avenue for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Shobhit Gairola
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP, 226002, India
| | - Antarip Sinha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP, 226002, India
| | - Ravinder K Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP, 226002, India.
| |
Collapse
|
6
|
Nascimento M, Huot-Marchand S, Fanny M, Straube M, Le Bert M, Savigny F, Apetoh L, Van Snick J, Trovero F, Chamaillard M, Quesniaux VFJ, Ryffel B, Gosset P, Gombault A, Riteau N, Sokol H, Couillin I. NLRP6 controls pulmonary inflammation from cigarette smoke in a gut microbiota-dependent manner. Front Immunol 2023; 14:1224383. [PMID: 38146368 PMCID: PMC10749332 DOI: 10.3389/fimmu.2023.1224383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/20/2023] [Indexed: 12/27/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major health issue primarily caused by cigarette smoke (CS) and characterized by breathlessness and repeated airway inflammation. NLRP6 is a cytosolic innate receptor controlling intestinal inflammation and orchestrating the colonic host-microbial interface. However, its roles in the lungs remain largely unexplored. Using CS exposure models, our data show that airway inflammation is strongly impaired in Nlrp6-deficient mice with drastically fewer recruited neutrophils, a key cell subset in inflammation and COPD. We found that NLRP6 expression in lung epithelial cells is important to control airway and lung tissue inflammation in an inflammasome-dependent manner. Since gut-derived metabolites regulate NLRP6 inflammasome activation in intestinal epithelial cells, we investigated the link between NLRP6, CS-driven lung inflammation, and gut microbiota composition. We report that acute CS exposure alters gut microbiota in both wild-type (WT) and Nlrp6-deficient mice and that antibiotic treatment decreases CS-induced lung inflammation. In addition, gut microbiota transfer from dysbiotic Nlrp6-deficient mice to WT mice decreased airway lung inflammation in WT mice, highlighting an NLRP6-dependent gut-to-lung axis controlling pulmonary inflammation.
Collapse
Affiliation(s)
- Mégane Nascimento
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Sarah Huot-Marchand
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Manoussa Fanny
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Marjolène Straube
- Sorbonne Université, Institut National de la Recherche Médicale (INSERM), Centre de Recherche Saint-Antoine (CRSA), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint Antoine, Service de Gastroenterologie, Paris, France
| | - Marc Le Bert
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Florence Savigny
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | | | | | | | - Mathias Chamaillard
- Univ. Lille, Institut National de la Recherche Médicale (INSERM), U1003 - Laboratoire de physiologie cellulaire (PHYCEL) - Physiologie Cellulaire, Lille, France
| | - Valérie F. J. Quesniaux
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Bernhard Ryffel
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Philippe Gosset
- Institut PASTEUR INSERM U1019, Centre National de Recherche (CNRS) Unité Mixte de Recherche (UMR) 8204, Lille, France
| | - Aurélie Gombault
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Nicolas Riteau
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| | - Harry Sokol
- Sorbonne Université, Institut National de la Recherche Médicale (INSERM), Centre de Recherche Saint-Antoine (CRSA), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint Antoine, Service de Gastroenterologie, Paris, France
- Institut national de la recherche agronomique (INRA), UMR1319 Micalis, AgroParisTech, Jouy-en-Josas, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Isabelle Couillin
- University of Orleans and Centre National de Recherche scientifique (CNRS), Experimental and Molecular Immunology and Neurogenetics (INEM)-UMR7355, Orleans, France
| |
Collapse
|
7
|
Maruyama D, Liao WI, Tian X, Bredon M, Knapp J, Tat C, Doan TNM, Chassaing B, Bhargava A, Sokol H, Prakash A. Regulation of Lung Immune Tone by the Gut-Lung Axis via Dietary Fiber, Gut Microbiota, and Short-Chain Fatty Acids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.552964. [PMID: 37662303 PMCID: PMC10473695 DOI: 10.1101/2023.08.24.552964] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Lung immune tone, i.e. the immune state of the lung, can vary between individuals and over a single individual's lifetime, and its basis and regulation in the context of inflammatory responses to injury is poorly understood. The gut microbiome, through the gut-lung axis, can influence lung injury outcomes but how the diet and microbiota affect lung immune tone is also unclear. We hypothesized that lung immune tone would be influenced by the presence of fiber-fermenting short-chain fatty acid (SCFA)-producing gut bacteria. To test this hypothesis, we conducted a fiber diet intervention study followed by lung injury in mice and profiled gut microbiota using 16S sequencing, metabolomics, and lung immune tone. We also studied germ-free mice to evaluate lung immune tone in the absence of microbiota and performed in vitro mechanistic studies on immune tone and metabolic programming of alveolar macrophages exposed to the SCFA propionate (C3). Mice on high-fiber diet were protected from sterile lung injury compared to mice on a fiber-free diet. This protection strongly correlated with lower lung immune tone, elevated propionate levels and enrichment of specific fecal microbiota taxa; conversely, lower levels of SCFAs and an increase in other fatty acid metabolites and bacterial taxa correlated with increased lung immune tone and increased lung injury in the fiber-free group. In vitro , C3 reduced lung alveolar macrophage immune tone (through suppression of IL-1β and IL-18) and metabolically reprogrammed them (switching from glycolysis to oxidative phosphorylation after LPS challenge). Overall, our findings reveal that the gut-lung axis, through dietary fiber intake and enrichment of SCFA-producing gut bacteria, can regulate innate lung immune tone via IL-1β and IL-18 pathways. These results provide a rationale for the therapeutic development of dietary interventions to preserve or enhance specific aspects of host lung immunity.
Collapse
|
8
|
Gbotosho OT, Li W, Joiner CH, Brown LAS, Hyacinth HI. The inflammatory profiles of pulmonary alveolar macrophages and alveolar type 2 cells in SCD. Exp Biol Med (Maywood) 2023; 248:1013-1023. [PMID: 37012678 PMCID: PMC10581160 DOI: 10.1177/15353702231157940] [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: 11/05/2022] [Accepted: 01/15/2023] [Indexed: 04/05/2023] Open
Abstract
The lung microenvironment plays a crucial role in maintaining lung homeostasis as well as the initiation and resolution of both acute and chronic lung injury. Acute chest syndrome (ACS) is a complication of sickle cell disease (SCD) like acute lung injury. Both the endothelial cells and peripheral blood mononuclear cells are known to secrete proinflammatory cytokines elevated during ACS episodes. However, in SCD, the lung microenvironment that may favor excessive production of proinflammatory cytokines and the contribution of other lung resident cells, such as alveolar macrophages and alveolar type 2 epithelial (AT-2) cells, to ACS pathogenesis is not completely understood. Here, we sought to understand the pulmonary microenvironment and the proinflammatory profile of lung alveolar macrophages (LAMs) and AT-2 cells at steady state in Townes sickle cell (SS) mice compared to control mice (AA). In addition, we examined lung function and micromechanics molecules essential for pulmonary epithelial barrier function in these mice. Our results showed that bronchoalveolar lavage (BAL) fluid in SS mice had elevated protein levels of pro-inflammatory cytokines interleukin (IL)-1β and IL-12 (p ⩽ 0.05) compared to AA controls. We showed for the first time, significantly increased protein levels of inflammatory mediators (Human antigen R (HuR), Toll-like receptor 4 (TLR4), MyD88, and PU.1) in AT-2 cells (1.4 to 2.2-fold) and LAM (17-21%) isolated from SS mice compared to AA control mice at steady state. There were also low levels of anti-inflammatory transcription factors (Nrf2 and PPARy) in SS mice compared to AA controls (p ⩽ 0.05). Finally, we found impaired lung function and a dysregulated composition of surfactant proteins (B and C). Our results demonstrate that SS mice at steady state had a compromised lung microenvironment with elevated expression of proinflammatory cytokines by AT-2 cells and LAM, as well as dysregulated expression of surfactant proteins necessary for maintaining the alveolar barrier integrity and lung function.
Collapse
Affiliation(s)
- Oluwabukola T Gbotosho
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Wei Li
- Aflac Cancer & Blood Disorders Center of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Clinton H Joiner
- Aflac Cancer & Blood Disorders Center of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lou Ann S Brown
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hyacinth I Hyacinth
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
9
|
Chen P, Zhou Y, Li X, Yang J, Zheng Z, Zou Y, Li X, Liao J, Dai J, Xu Y, Yin L, Chen G, Gu J, Ouyang Q, Cho WJ, Tang Q, Liang G. Design, Synthesis, and Bioevaluation of Novel MyD88 Inhibitor c17 against Acute Lung Injury Derived from the Virtual Screen. J Med Chem 2023; 66:6938-6958. [PMID: 37130331 DOI: 10.1021/acs.jmedchem.3c00359] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Myeloid differentiation primary response protein 88 (MyD88) is crucial to immune cascades mediated by Toll-like receptors (TLRs) and interleukin-1 receptors (IL-1Rs). MyD88 dysregulation has been linked to a wide variety of inflammatory diseases, making it a promising new target for anti-inflammatory and cancer therapy development. In this study, 46 compounds were designed and synthesized inspired by virtual screen hit. The anti-inflammatory activity of designed compounds was evaluated biologically, and c17 was discovered to have a high binding affinity with MyD88. It inhibited the interaction of TLR4 and MyD88 and suppressed the NF-κB pathway. In addition, c17 treatment led to the accumulation in the lungs of rats and attenuated LPS-induced ALI mice model. Furthermore, c17 showed negligible toxicity in vivo. Together, these findings suggest that c17 may serve as a potential therapeutical method for the treatment of ALI and as a lead structure for the continued development of MyD88 inhibitors.
Collapse
Affiliation(s)
- Pan Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, China
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Ying Zhou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Xiaobo Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Jun Yang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Zhiwei Zheng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Yu Zou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Xiang Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Jing Liao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Jintian Dai
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Yuye Xu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Lina Yin
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, China
| | - Gaozhi Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jing Gu
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Qin Ouyang
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Won-Jea Cho
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea
| | - Qidong Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| |
Collapse
|
10
|
Kokturk N, Khodayari N, Lascano J, Riley EL, Brantly ML. Lung Inflammation in alpha-1-antitrypsin deficient individuals with normal lung function. Respir Res 2023; 24:40. [PMID: 36732772 PMCID: PMC9893669 DOI: 10.1186/s12931-023-02343-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/24/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Alpha-1-antitrypsin deficient (AATD) individuals are prone to develop early age of onset chronic obstructive pulmonary disease (COPD) more severe than non-genetic COPD. Here, we investigated the characteristics of lower respiratory tract of AATD individuals prior to the onset of clinically significant COPD. METHODS Bronchoalveolar lavage was performed on 22 AATD with normal lung function and 14 healthy individuals. Cell counts and concentrations of proteases, alpha-1-antitrypsin and proinflammatory mediators were determined in the bronchoalveolar lavage fluid from study subjects. In order to determine the airway inflammation, we also analyzed immune cell components of the large airways from bronchial biopsies using immunohistochemistry in both study subjects. Finally, we made comparisons between airway inflammation and lung function rate of decline using four repeated lung function tests over one year in AATD individuals. RESULTS AATD individuals with normal lung function had 3 folds higher neutrophil counts, 2 folds increase in the proteases levels, and 2-4 folds higher levels of IL-8, IL-6, IL-1β, and leukotriene B4 in their epithelial lining fluid compared to controls. Neutrophil elastase levels showed a positive correlation with the levels of IL-8 and neutrophils in AATD epithelial lining fluid. AATD individuals also showed a negative correlation of baseline FEV1 with neutrophil count, neutrophil elastase, and cytokine levels in epithelial lining fluid (p < 0.05). In addition, we observed twofold increase in the number of lymphocytes, macrophages, neutrophils, and mast cells of AATD epithelial lining fluid as compared to controls. CONCLUSION Mild inflammation is present in the lower respiratory tract and airways of AATD individuals despite having normal lung function. A declining trend was also noticed in the lung function of AATD individuals which was correlated with pro-inflammatory phenotype of their lower respiratory tract. This results suggest the presence of proinflammatory phenotype in AATD lungs. Therefore, early anti-inflammatory therapies may be a potential strategy to prevent progression of lung disease in AATD individuals.
Collapse
Affiliation(s)
- Nurdan Kokturk
- Division of Pulmonary, Critical Care and Sleep Medicine, J. Hillis Miller Health Science Center, University of Florida College of Medicine, P.O. Box 100225, Gainesville, FL, 32610-0225, USA
- Department of Pulmonary and Critical Care, Gazi University School of Medicine, Ankara, Turkey
| | - Nazli Khodayari
- Division of Pulmonary, Critical Care and Sleep Medicine, J. Hillis Miller Health Science Center, University of Florida College of Medicine, P.O. Box 100225, Gainesville, FL, 32610-0225, USA
| | - Jorge Lascano
- Division of Pulmonary, Critical Care and Sleep Medicine, J. Hillis Miller Health Science Center, University of Florida College of Medicine, P.O. Box 100225, Gainesville, FL, 32610-0225, USA
| | | | - Mark L Brantly
- Division of Pulmonary, Critical Care and Sleep Medicine, J. Hillis Miller Health Science Center, University of Florida College of Medicine, P.O. Box 100225, Gainesville, FL, 32610-0225, USA.
| |
Collapse
|
11
|
Cross-talk between IL-6 trans-signaling and AIM2 inflammasome/IL-1β axes bridge innate immunity and epithelial apoptosis to promote emphysema. Proc Natl Acad Sci U S A 2022; 119:e2201494119. [PMID: 36037355 PMCID: PMC9457334 DOI: 10.1073/pnas.2201494119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pulmonary emphysema is associated with dysregulated innate immune responses that promote chronic pulmonary inflammation and alveolar apoptosis, culminating in lung destruction. However, the molecular regulators of innate immunity that promote emphysema are ill-defined. Here, we investigated whether innate immune inflammasome complexes, comprising the adaptor ASC, Caspase-1 and specific pattern recognition receptors (PRRs), promote the pathogenesis of emphysema. In the lungs of emphysematous patients, as well as spontaneous gp130F/F and cigarette smoke (CS)-induced mouse models of emphysema, the expression (messenger RNA and protein) and activation of ASC, Caspase-1, and the inflammasome-associated PRR and DNA sensor AIM2 were up-regulated. AIM2 up-regulation in emphysema coincided with the biased production of the mature downstream inflammasome effector cytokine IL-1β but not IL-18. These observations were supported by the genetic blockade of ASC, AIM2, and the IL-1 receptor and therapy with AIM2 antagonistic suppressor oligonucleotides, which ameliorated emphysema in gp130F/F mice by preventing elevated alveolar cell apoptosis. The functional requirement for AIM2 in driving apoptosis in the lung epithelium was independent of its expression in hematopoietic-derived immune cells and the recruitment of infiltrating immune cells in the lung. Genetic and inhibitor-based blockade of AIM2 also protected CS-exposed mice from pulmonary alveolar cell apoptosis. Intriguingly, IL-6 trans-signaling via the soluble IL-6 receptor, facilitated by elevated levels of IL-6, acted upstream of the AIM2 inflammasome to augment AIM2 expression in emphysema. Collectively, we reveal cross-talk between the AIM2 inflammasome/IL-1β and IL-6 trans-signaling axes for potential exploitation as a therapeutic strategy for emphysema.
Collapse
|
12
|
Philip N, Priya SP, Jumah Badawi AH, Mohd Izhar MH, Mohtarrudin N, Tengku Ibrahim TA, Sekawi Z, Neela VK. Pulmonary haemorrhage as the earliest sign of severe leptospirosis in hamster model challenged with Leptospira interrogans strain HP358. PLoS Negl Trop Dis 2022; 16:e0010409. [PMID: 35584087 PMCID: PMC9116642 DOI: 10.1371/journal.pntd.0010409] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/10/2022] [Indexed: 11/18/2022] Open
Abstract
Background Severe leptospirosis is challenging as it could evolve rapidly and potentially fatal if appropriate management is not performed. An understanding of the progression and pathophysiology of Leptospira infection is important to determine the early changes that could be potentially used to predict the severe occurrence of leptospirosis. This study aimed to understand the kinetics pathogenesis of Leptospira interrogans strain HP358 in the hamster model and identify the early parameters that could be used as biomarkers to predict severe leptospirosis. Methodology/Principal findings Male Syrian hamsters were infected with Leptospira interrogans strain HP358 and euthanized after 24 hours, 3, 4, 5, 6 and 7 days post-infection. Blood, lungs, liver and kidneys were collected for leptospiral detection, haematology, serum biochemistry and differential expression of pro- and anti-inflammatory markers. Macroscopic and microscopic organ damages were investigated. Leptospira interrogans strain HP358 was highly pathogenic and killed hamsters within 6–7 days post-infection. Pulmonary haemorrhage and blood vessel congestion in organs were noticed as the earliest pathological changes. The damages in organs and changes in biochemistry value were preceded by changes in haematology and immune gene expression. Conclusion/Significance This study deciphered haemorrhage as the earliest manifestation of severe leptospirosis and high levels of IL-1β, CXCL10/IP-10, CCL3/MIP-α, neutrophils and low levels of lymphocytes and platelets serve as a cumulative panel of biomarkers in severe leptospirosis. As the severe form of leptospirosis could progress rapidly and be potentially fatal if not treated earlier, deciphering the pathophysiology kinetics of infection is crucial to determine the parameters of disease severity. To understand this, we challenged hamsters with the highly virulent Leptospira interrogans strain HP358. Pulmonary haemorrhage was observed as the earliest pathological change followed by liver and kidneys damages. The increased expression of IL-1β, CXCL10/IP-10, CCL3/MIP-α, high neutrophils and low lymphocytes and platelets production observed in the present study indicate that these parameters could serve as a cumulative panel of biomarkers in severe leptospirosis.
Collapse
Affiliation(s)
- Noraini Philip
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Sivan Padma Priya
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- RAK College of Dental Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Ahmad Hussein Jumah Badawi
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Mohd Hafidz Mohd Izhar
- Comparative Medicine and Technology Unit, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Norhafizah Mohtarrudin
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Tengku Azmi Tengku Ibrahim
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Zamberi Sekawi
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Vasantha Kumari Neela
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- * E-mail:
| |
Collapse
|
13
|
Artlett CM. The Mechanism and Regulation of the NLRP3 Inflammasome during Fibrosis. Biomolecules 2022; 12:biom12050634. [PMID: 35625564 PMCID: PMC9138796 DOI: 10.3390/biom12050634] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 02/01/2023] Open
Abstract
Fibrosis is often the end result of chronic inflammation. It is characterized by the excessive deposition of extracellular matrix. This leads to structural alterations in the tissue, causing permanent damage and organ dysfunction. Depending on the organ it effects, fibrosis can be a serious threat to human life. The molecular mechanism of fibrosis is still not fully understood, but the NLRP3 (NOD-, LRR- and pyrin–domain–containing protein 3) inflammasome appears to play a significant role in the pathogenesis of fibrotic disease. The NLRP3 inflammasome has been the most extensively studied inflammatory pathway to date. It is a crucial component of the innate immune system, and its activation mediates the secretion of interleukin (IL)-1β and IL-18. NLRP3 activation has been strongly linked with fibrosis and drives the differentiation of fibroblasts into myofibroblasts by the chronic upregulation of IL-1β and IL-18 and subsequent autocrine signaling that maintains an activated inflammasome. Both IL-1β and IL-18 are profibrotic, however IL-1β can have antifibrotic capabilities. NLRP3 responds to a plethora of different signals that have a common but unidentified unifying trigger. Even after 20 years of extensive investigation, regulation of the NLRP3 inflammasome is still not completely understood. However, what is known about NLRP3 is that its regulation and activation is complex and not only driven by various activators but controlled by numerous post-translational modifications. More recently, there has been an intensive attempt to discover NLRP3 inhibitors to treat chronic diseases. This review addresses the role of the NLRP3 inflammasome in fibrotic disorders across many different tissues. It discusses the relationships of various NLRP3 activators to fibrosis and covers different therapeutics that have been developed, or are currently in development, that directly target NLRP3 or its downstream products as treatments for fibrotic disorders.
Collapse
Affiliation(s)
- Carol M Artlett
- Department of Microbiology & Immunology, College of Medicine, Drexel University, Philadelphia, PA 19129, USA
| |
Collapse
|
14
|
Rasul TF, Bergholz DR, Rovinski R, Gulraiz S, Fonts E. Combined Pulmonary Fibrosis and Emphysema and Digital Clubbing. Cureus 2022; 14:e24231. [PMID: 35602819 PMCID: PMC9115620 DOI: 10.7759/cureus.24231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2022] [Indexed: 11/23/2022] Open
Abstract
Combined pulmonary fibrosis and emphysema (CPFE) is an underrecognized syndrome that involves simultaneous restrictive-obstructive lung disease. The prognosis is poor, and it frequently occurs with comorbidities. Heavy or former smoking is a major risk factor, and computed tomography (CT) typically shows lower zone fibrosis and upper zone emphysema. Chronic respiratory failure, pulmonary hypertension, and lung carcinoma are major causes of mortality. Diagnosis of CPFE should be combined with palliative care due to the high mortality of the condition, especially in the case of delayed diagnosis. We present the case of a 73-year-old male with a history of non-small cell lung cancer, 50 pack-year smoking, and cervical spine injury (CSI) with a late diagnosis of CPFE. After presenting to the emergency department for an acute exacerbation of dyspnea and hypoxia, he was initially treated with a congestive heart failure protocol. Further examination showed mixed pulmonary function tests as well as digital clubbing, and a CT scan showed changes indicative of advanced bullous emphysema diffusely throughout both lungs with an upper lobe predominance and basilar fibrosis. He was diagnosed with CPFE and immediately treated for both restrictive and obstructive lung diseases with supplemental oxygen, albuterol, ipratropium, corticosteroids, systemic antibiotics, as well as provided with palliative consultation. His previous history and CSI delayed diagnosis, as his lung restriction was likely assumed to be from impaired chest wall mobility rather than CPFE. This case highlights the presentation of a relatively rare disease that was confounded by comorbidities.
Collapse
|
15
|
Pinar AA, S Samuel CS. Immune Mechanisms and Related Targets for the Treatment of Fibrosis in Various Organs. Curr Mol Med 2022; 22:240-249. [PMID: 35034593 DOI: 10.2174/1566524022666220114122839] [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: 07/25/2021] [Revised: 11/08/2021] [Accepted: 01/05/2022] [Indexed: 11/22/2022]
Abstract
Inflammation and fibrosis are two inter-related disease pathologies with several overlapping components. Three specific cell types, macrophages, T helper cells and myofibroblasts, each play important roles in regulating both processes. Following tissue injury, an inflammatory stimulus is often necessary to initiate tissue repair, where cytokines released from infiltrating and resident immune and inflammatory cells stimulate the proliferation and activation of extracellular matrix-producing myofibroblasts. However, persistent tissue injury drives an inappropriate pro-fibrotic response. Additionally, activated myofibroblasts can take on the role of traditional antigen-presenting cells, secrete pro-inflammatory cytokines, and recruit inflammatory cells to fibrotic foci, amplifying the fibrotic response in a vicious cycle. Moreover, inflammatory cells have been shown to play contradictory roles in the initiation, amplification and resolution of fibrotic disease processes. The central role of the inflammasome molecular platform in contributing to fibrosis is only beginning to be fully appreciated. In this review, we discuss the immune mechanisms that can lead to fibrosis, the inflammasomes that have been implicated in the fibrotic process in the context of the immune response to injury, and also discuss current and emerging therapies that target inflammasome-induced collagen deposition to treat organ fibrosis.
Collapse
Affiliation(s)
- Anita A Pinar
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Chrishan S S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
16
|
Pera T, Loblundo C, Penn RB. Pharmacological Management of Asthma and COPD. COMPREHENSIVE PHARMACOLOGY 2022:762-802. [DOI: 10.1016/b978-0-12-820472-6.00095-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
17
|
Wortmann M, Peters AS, Erhart P, Körfer D, Böckler D, Dihlmann S. Inflammasomes in the Pathophysiology of Aortic Disease. Cells 2021; 10:cells10092433. [PMID: 34572082 PMCID: PMC8468335 DOI: 10.3390/cells10092433] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/26/2021] [Accepted: 08/29/2021] [Indexed: 12/27/2022] Open
Abstract
Aortic diseases comprise aneurysms, dissections, and several other pathologies. In general, aging is associated with a slow but progressive dilation of the aorta, along with increased stiffness and pulse pressure. The progression of aortic disease is characterized by subclinical development or acute presentation. Recent evidence suggests that inflammation participates causally in different clinical manifestations of aortic diseases. As of yet, diagnostic imaging and surveillance is mainly based on ultrasonography, computed tomography (CT), and magnetic resonance imaging (MRI). Little medical therapy is available so far to prevent or treat the majority of aortic diseases. Endovascular therapy by the introduction of covered stentgrafts provides the main treatment option, although open surgery and implantation of synthetic grafts remain necessary in many situations. Because of the risks associated with surgery, there is a need for identification of pharmaceutical targets interfering with the pathophysiology of aortic remodeling. The participation of innate immunity and inflammasome activation in different cell types is common in aortic diseases. This review will thus focus on inflammasome activities in vascular cells of different chronic and acute aortic diseases and discuss their role in development and progression. We will also identify research gaps and suggest promising therapeutic targets, which may be used for future medical interventions.
Collapse
|
18
|
Zhang J, Xu Q, Sun W, Zhou X, Fu D, Mao L. New Insights into the Role of NLRP3 Inflammasome in Pathogenesis and Treatment of Chronic Obstructive Pulmonary Disease. J Inflamm Res 2021; 14:4155-4168. [PMID: 34471373 PMCID: PMC8405160 DOI: 10.2147/jir.s324323] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/06/2021] [Indexed: 11/23/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is an inflammatory lung disease characterized by chronic airway obstruction and emphysema. Accumulating studies have shown that the onset and development of COPD are related to an aberrant immune response induced by the dysregulation of a number of genetic and environmental factors, while the exact pathogenesis of this disease is not well defined. Emerging studies based on tests on samples from COPD patients, animal models, pharmacological and genetic data suggest that the NLR family pyrin domain containing 3 (NLRP3) inflammasome activation is required in the lung inflammatory responses in the development of COPD. Although the available clinical studies targeting the inflammasome effector cytokine, IL-1β, or IL-1 signaling do not show positive outcomes for COPD treatment, many alternative strategies have been proposed by recent emerging studies. Here, we highlight the recent progress in our understanding of the role of the NLRP3 inflammasome in COPD and propose possible future studies that may further elucidate the roles of the inflammasome in the pathogenesis or the intervention of this inflammatory lung disease.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Immunology, School of Medicine, Nantong University, Nantong, Jiangsu, 226019, People's Republic of China.,Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226019, People's Republic of China
| | - Qiuyun Xu
- Department of Immunology, School of Medicine, Nantong University, Nantong, Jiangsu, 226019, People's Republic of China
| | - Weichen Sun
- Department of Immunology, School of Medicine, Nantong University, Nantong, Jiangsu, 226019, People's Republic of China
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, Jiangsu, 226019, People's Republic of China
| | - Da Fu
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, People's Republic of China
| | - Liming Mao
- Department of Immunology, School of Medicine, Nantong University, Nantong, Jiangsu, 226019, People's Republic of China.,Basic Medical Research Center, School of Medicine, Nantong University, Nantong, Jiangsu, 226019, People's Republic of China
| |
Collapse
|
19
|
Schiffers C, van de Wetering C, Bauer RA, Habibovic A, Hristova M, Dustin CM, Lambrichts S, Vacek PM, Wouters EF, Reynaert NL, van der Vliet A. Downregulation of epithelial DUOX1 in chronic obstructive pulmonary disease. JCI Insight 2021; 6:142189. [PMID: 33301419 PMCID: PMC7934842 DOI: 10.1172/jci.insight.142189] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic respiratory disease characterized by small airway remodeling and alveolar emphysema due to environmental stresses such as cigarette smoking (CS). Oxidative stress is commonly implicated in COPD pathology, but recent findings suggest that one oxidant-producing NADPH oxidase homolog, dual oxidase 1 (DUOX1), is downregulated in the airways of patients with COPD. We evaluated lung tissue sections from patients with COPD for small airway epithelial DUOX1 protein expression, in association with measures of lung function and small airway and alveolar remodeling. We also addressed the impact of DUOX1 for lung tissue remodeling in mouse models of COPD. Small airway DUOX1 levels were decreased in advanced COPD and correlated with loss of lung function and markers of emphysema and remodeling. Similarly, DUOX1 downregulation in correlation with extracellular matrix remodeling was observed in a genetic model of COPD, transgenic SPC-TNF-α mice. Finally, development of subepithelial airway fibrosis in mice due to exposure to the CS-component acrolein, or alveolar emphysema induced by administration of elastase, were in both cases exacerbated in Duox1-deficient mice. Collectively, our studies highlight that downregulation of DUOX1 may be a contributing feature of COPD pathogenesis, likely related to impaired DUOX1-mediated innate injury responses involved in epithelial homeostasis.
Collapse
Affiliation(s)
- Caspar Schiffers
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.,Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Cheryl van de Wetering
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.,Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Robert A Bauer
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Aida Habibovic
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Milena Hristova
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Christopher M Dustin
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Sara Lambrichts
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Pamela M Vacek
- Department of Medical Biostatistics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Emiel Fm Wouters
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands.,Ludwig Boltzman Institute for Lung Health, Vienna, Austria
| | - Niki L Reynaert
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
20
|
Adini A, Wu H, Dao DT, Ko VH, Yu LJ, Pan A, Puder M, Mitiku SZ, Potla R, Chen H, Rice JM, Matthews BD. PR1P Stabilizes VEGF and Upregulates Its Signaling to Reduce Elastase-induced Murine Emphysema. Am J Respir Cell Mol Biol 2020; 63:452-463. [PMID: 32663413 PMCID: PMC7528927 DOI: 10.1165/rcmb.2019-0434oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 06/04/2020] [Indexed: 12/21/2022] Open
Abstract
Emphysema is a progressive and fatal lung disease with no cure that is characterized by thinning, enlargement, and destruction of alveoli, leading to impaired gas exchange. Disease progression is due in part to dysregulation of VEGF (vascular endothelial growth factor) signaling in the lungs and increased lung-cell apoptosis. Here we asked whether PR1P (Prominin-1-derived peptide), a novel short peptide we designed that increases VEGF binding to endothelial cells, could be used to improve outcome in in vitro and in vivo models of emphysema. We used computer simulation and in vitro and in vivo studies to show that PR1P upregulated endogenous VEGF receptor-2 signaling by binding VEGF and preventing its proteolytic degradation. In so doing, PR1P mitigated toxin-induced lung-cell apoptosis, including from cigarette-smoke extract in vitro and from LPS in vivo in mice. Remarkably, inhaled PR1P led to significantly increased VEGF concentrations in murine lungs within 30 minutes that remained greater than twofold above that of control animals 24 hours later. Finally, inhaled PR1P reduced acute lung injury in 4- and 21-day elastase-induced murine emphysema models. Taken together, these results highlight the potential of PR1P as a novel therapeutic agent for the treatment of emphysema or other lung diseases characterized by VEGF signaling dysregulation.
Collapse
Affiliation(s)
- Avner Adini
- Vascular Biology Program
- Department of Pathology
- Department of Surgery, and
- Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hao Wu
- Vascular Biology Program
- Department of Pathology
- Department of Surgery, and
| | - Duy T. Dao
- Vascular Biology Program
- Department of Surgery, and
| | | | - Lumeng J. Yu
- Vascular Biology Program
- Department of Surgery, and
| | - Amy Pan
- Vascular Biology Program
- Department of Surgery, and
| | - Mark Puder
- Vascular Biology Program
- Department of Surgery, and
| | - Selome Z. Mitiku
- Vascular Biology Program
- Department of Pathology
- Department of Surgery, and
- Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ratnakar Potla
- Vascular Biology Program
- Department of Pathology
- Department of Surgery, and
| | - Hong Chen
- Vascular Biology Program
- Department of Pathology
- Department of Surgery, and
| | - James M. Rice
- Vascular Biology Program
- Department of Pathology
- Department of Surgery, and
| | - Benjamin D. Matthews
- Vascular Biology Program
- Department of Pathology
- Department of Surgery, and
- Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
21
|
Nascimento M, Huot-Marchand S, Gombault A, Panek C, Bourinet M, Fanny M, Savigny F, Schneider P, Le Bert M, Ryffel B, Riteau N, Quesniaux VFJ, Couillin I. B-Cell Activating Factor Secreted by Neutrophils Is a Critical Player in Lung Inflammation to Cigarette Smoke Exposure. Front Immunol 2020; 11:1622. [PMID: 32849550 PMCID: PMC7405926 DOI: 10.3389/fimmu.2020.01622] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/17/2020] [Indexed: 12/18/2022] Open
Abstract
Cigarette smoke (CS) is the major cause of chronic lung injuries, such as chronic obstructive pulmonary disease (COPD). In patients with severe COPD, tertiary lymphoid follicles containing B lymphocytes and B cell-activating factor (BAFF) overexpression are associated with disease severity. In addition, BAFF promotes adaptive immunity in smokers and mice chronically exposed to CS. However, the role of BAFF in the early phase of innate immunity has never been investigated. We acutely exposed C57BL/6J mice to CS and show early BAFF expression in the bronchoalveolar space and lung tissue that correlates to airway neutrophil and macrophage influx. Immunostaining analysis revealed that neutrophils are the major source of BAFF. We confirmed in vitro that neutrophils secrete BAFF in response to cigarette smoke extract (CSE) stimulation. Antibody-mediated neutrophil depletion significantly dampens lung inflammation to CS exposure but only partially decreases BAFF expression in lung tissue and bronchoalveolar space suggesting additional sources of BAFF. Importantly, BAFF deficient mice displayed decreased airway neutrophil recruiting chemokines and neutrophil influx while the addition of exogenous BAFF significantly enhanced this CS-induced neutrophilic inflammation. This demonstrates that BAFF is a key proinflammatory cytokine and that innate immune cells in particular neutrophils, are an unconsidered source of BAFF in early stages of CS-induced innate immunity.
Collapse
Affiliation(s)
| | | | | | - Corinne Panek
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | - Manon Bourinet
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | - Manoussa Fanny
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | | | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Épalinges, Switzerland
| | - Marc Le Bert
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | - Bernhard Ryffel
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | - Nicolas Riteau
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | | | | |
Collapse
|
22
|
Gonzalez J, Rivera-Ortega P, Rodríguez-Fraile M, Restituto P, Colina I, Calleja MDLD, Alcaide AB, Campo A, Bertó J, Seijo L, Pérez-Warnisher MT, Zulueta JJ, Varo N, de-Torres JP. Exploring the Association Between Emphysema Phenotypes and Low Bone Mineral Density in Smokers with and without COPD. Int J Chron Obstruct Pulmon Dis 2020; 15:1823-1829. [PMID: 32801680 PMCID: PMC7401322 DOI: 10.2147/copd.s257918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 07/02/2020] [Indexed: 12/18/2022] Open
Abstract
Rationale Emphysema and osteoporosis are tobacco-related diseases. Many studies have shown that emphysema is a strong and independent predictor of low bone mineral density (BMD) in smokers; however, none of them explored its association with different emphysema subtypes. Objective To explore the association between the different emphysema subtypes and the presence of low bone mineral density in a population of active or former smokers with and without chronic obstructive pulmonary disease (COPD). Methods One hundred and fifty-three active and former smokers from a pulmonary clinic completed clinical questionnaires, pulmonary function tests, a low-dose chest computed tomography (LDCT) and a dual-energy absorptiometry (DXA) scans. Subjects were classified as having normal BMD or low BMD (osteopenia or osteoporosis). Emphysema was classified visually for its subtype and severity. Logistic regression analysis explored the relationship between the different emphysema subtypes and the presence of low BMD adjusting for other important factors. Results Seventy-five percent of the patients had low BMD (78 had osteopenia and 37 had osteoporosis). Emphysema was more frequent (66.1 vs 26.3%, p=<0.001) and severe in those with low BMD. Multivariable analysis adjusting for other significant cofactors (age, sex, FEV1, and severity of emphysema) showed that BMI (OR=0.91, 95% CI: 0.76–0.92) and centrilobular emphysema (OR=26.19, 95% CI: 1.71 to 399.44) were associated with low BMD. Conclusion Low BMD is highly prevalent in current and former smokers. BMI and centrilobular emphysema are strong and independent predictors of its presence, which suggests that they should be considered when evaluating smokers at risk for low BMD.
Collapse
Affiliation(s)
- Jessica Gonzalez
- Pulmonary Department, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| | | | | | - Patricia Restituto
- Biochemistry Department, Clínica Universitaria de Navarra, Pamplona, Spain
| | - Inmaculada Colina
- Department of Internal Medicine, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Ana B Alcaide
- Pulmonary Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Aránzazu Campo
- Pulmonary Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Juan Bertó
- Pulmonary Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Luis Seijo
- Pulmonary Department, Clínica Universidad de Navarra, Madrid, Spain
| | | | - Javier J Zulueta
- Pulmonary Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Nerea Varo
- Biochemistry Department, Clínica Universitaria de Navarra, Pamplona, Spain
| | | |
Collapse
|
23
|
Akita T, Hirokawa M, Yamashita C. The effects of 1α,25-dihydroxyvitamin D3 on alveolar repair and bone mass in adiponectin-deficient mice. J Steroid Biochem Mol Biol 2020; 201:105696. [PMID: 32407869 DOI: 10.1016/j.jsbmb.2020.105696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 03/24/2020] [Accepted: 05/06/2020] [Indexed: 11/23/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of death worldwide. However, no drugs can regenerate lung tissue in COPD patients, and differentiation-inducing drugs that can effectively treat damaged alveoli are needed. In addition, the presence of systemic comorbidities is also considered problematic. Our previous study revealed that a retinoic acid derivative improved emphysema in elastase-induced COPD model mice at a dose of 1.0 mg/kg, whereas 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) showed an emphysema-improving effect in the same model at 0.1 μg/kg. Elastase-induced COPD model mice do not exhibit a systemic disease state, so evaluation in a model that better reflects the human disease state is considered necessary. To solve this problem, we focused on the adiponectin-deficient mouse and examined the effects of 1,25(OH)2D3 on alveolar regeneration. Fifty-week-old adiponectin-deficient mice were treated with 1,25(OH)2D3 (0.1 μg/kg) twice a week, for 30 weeks. The effects of pulmonary administration on alveolar repair were evaluated according to the distance between alveolar walls (Lm values) and computed tomography (CT) parameters. Bone density was evaluated based on CT. The administration of 1,25(OH)2D3 was confirmed to show a significant therapeutic effect. The Lm values in the control and 1,25(OH)2D3-treated groups were 98 ± 4 μm and 63 ± 1 μm, respectively. However, on CT, the average CT value and % of low attenuation area showed no significant change. In adiponectin-deficient mice, the reduction of bone density (cortical, spongy, and total bone), which is a systemic symptom of COPD, was significantly suppressed by 1,25(OH)2D3 at 80 weeks of age. The present study suggests that 1,25(OH)2D3 could be a potential candidate drug that may provide a radical cure for the lung disease and comorbidities of COPD patients. This work can lead to the development drugs that may provide a radical cure for COPD.
Collapse
Affiliation(s)
- Tomomi Akita
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences, 2641 Yamazaki, Noda, Chiba 278-8510, Japan; Fusion of Regenerative Medicine With DDS, Research Institute for Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Mai Hirokawa
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Chikamasa Yamashita
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences, 2641 Yamazaki, Noda, Chiba 278-8510, Japan; Fusion of Regenerative Medicine With DDS, Research Institute for Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
| |
Collapse
|
24
|
Wada T, Jaw JE, Tsuruta M, Moritani K, Tsutsui M, Tam A, Vasilescu DM, Cheung CY, Yamasaki K, Lichtenstein S, Machan L, Gelbart D, Man SP, Sin DD. External radiofrequency as a novel extracorporeal therapy for emphysema. Eur Respir J 2020; 56:13993003.01422-2020. [PMID: 32471932 DOI: 10.1183/13993003.01422-2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 05/13/2020] [Indexed: 11/05/2022]
Affiliation(s)
- Takeyuki Wada
- Centre for Heart Lung Innovation, St Paul's Hospital, and Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Both authors contributed equally to this work
| | - Jen-Erh Jaw
- Centre for Heart Lung Innovation, St Paul's Hospital, and Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Both authors contributed equally to this work
| | - Masashi Tsuruta
- Centre for Heart Lung Innovation, St Paul's Hospital, and Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Konosuke Moritani
- Centre for Heart Lung Innovation, St Paul's Hospital, and Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Mai Tsutsui
- Centre for Heart Lung Innovation, St Paul's Hospital, and Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Anthony Tam
- Centre for Heart Lung Innovation, St Paul's Hospital, and Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Dragoş M Vasilescu
- Centre for Heart Lung Innovation, St Paul's Hospital, and Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Chung Yan Cheung
- Centre for Heart Lung Innovation, St Paul's Hospital, and Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Kei Yamasaki
- Centre for Heart Lung Innovation, St Paul's Hospital, and Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Samuel Lichtenstein
- Ikomed Technologies Inc., Vancouver, BC, Canada.,Division of Cardiac Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Lindsay Machan
- Ikomed Technologies Inc., Vancouver, BC, Canada.,Dept of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Dan Gelbart
- Ikomed Technologies Inc., Vancouver, BC, Canada
| | - S Paul Man
- Centre for Heart Lung Innovation, St Paul's Hospital, and Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Don D Sin
- Centre for Heart Lung Innovation, St Paul's Hospital, and Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
25
|
Fysikopoulos A, Seimetz M, Hadzic S, Knoepp F, Wu CY, Malkmus K, Wilhelm J, Pichl A, Bednorz M, Tadele Roxlau E, Ghofrani HA, Sommer N, Gierhardt M, Schermuly RT, Seeger W, Grimminger F, Weissmann N, Kraut S. Amelioration of elastase-induced lung emphysema and reversal of pulmonary hypertension by pharmacological iNOS inhibition in mice. Br J Pharmacol 2020; 178:152-171. [PMID: 32201936 DOI: 10.1111/bph.15057] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 02/12/2020] [Accepted: 03/10/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Chronic obstructive pulmonary disease, encompassing chronic airway obstruction and lung emphysema, is a major worldwide health problem and a severe socio-economic burden. Evidence previously provided by our group has shown that inhibition of inducible NOS (iNOS) prevents development of mild emphysema in a mouse model of chronic tobacco smoke exposure and can even trigger lung regeneration. Moreover, we could demonstrate that pulmonary hypertension is not only abolished in cigarette smoke-exposed iNOS-/- mice but also precedes emphysema development. Possible regenerative effects of pharmacological iNOS inhibition in more severe models of emphysema not dependent on tobacco smoke, however, are hitherto unknown. EXPERIMENTAL APPROACH We have established a mouse model using a single dose of porcine pancreatic elastase or saline, intratracheally instilled in C57BL/6J mice. Emphysema, as well as pulmonary hypertension development was determined by both structural and functional measurements. KEY RESULTS Our data revealed that (i) emphysema is fully established after 21 days, with the same degree of emphysema after 21 and 28 days post instillation, (ii) emphysema is stable for at least 12 weeks and (iii) pulmonary hypertension is evident, in contrast to smoke models, only after emphysema development. Oral treatment with the iNOS inhibitor N(6)-(1-iminoethyl)-l-lysine (L-NIL) was started after emphysema establishment and continued for 12 weeks. This resulted in significant lung regeneration, evident in the improvement of emphysema and reversal of pulmonary hypertension. CONCLUSION AND IMPLICATIONS Our data indicate that iNOS is a potential new therapeutic target to treat severe emphysema and associated pulmonary hypertension. LINKED ARTICLES This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.1/issuetoc.
Collapse
Affiliation(s)
- Athanasios Fysikopoulos
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Michael Seimetz
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Stefan Hadzic
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Fenja Knoepp
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Cheng-Yu Wu
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Kathrin Malkmus
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Jochen Wilhelm
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Alexandra Pichl
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Mariola Bednorz
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Elsa Tadele Roxlau
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Hossein A Ghofrani
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Natascha Sommer
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Mareike Gierhardt
- Max-Planck Heart and Lung Laboratory, Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Ralph T Schermuly
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Werner Seeger
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany.,Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Friedrich Grimminger
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Norbert Weissmann
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Simone Kraut
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
26
|
Self-DNA release and STING-dependent sensing drives inflammation to cigarette smoke in mice. Sci Rep 2019; 9:14848. [PMID: 31619733 PMCID: PMC6795997 DOI: 10.1038/s41598-019-51427-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 09/30/2019] [Indexed: 02/07/2023] Open
Abstract
Cigarette smoke exposure is a leading cause of chronic obstructive pulmonary disease (COPD), a major health issue characterized by airway inflammation with fibrosis and emphysema. Here we demonstrate that acute exposure to cigarette smoke causes respiratory barrier damage with the release of self-dsDNA in mice. This triggers the DNA sensor cGAS (cyclic GMP-AMP synthase) and stimulator of interferon genes (STING), driving type I interferon (IFN I) dependent lung inflammation, which are attenuated in cGAS, STING or type I interferon receptor (IFNAR) deficient mice. Therefore, we demonstrate a critical role of self-dsDNA release and of the cGAS-STING-type I interferon pathway upon cigarette smoke-induced damage, which may lead to therapeutic targets in COPD.
Collapse
|
27
|
Shiraishi K, Shichino S, Tsukui T, Hashimoto S, Ueha S, Matsushima K. Engraftment and proliferation potential of embryonic lung tissue cells in irradiated mice with emphysema. Sci Rep 2019; 9:3657. [PMID: 30842492 PMCID: PMC6403395 DOI: 10.1038/s41598-019-40237-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 02/08/2019] [Indexed: 12/26/2022] Open
Abstract
Recently, there has been increasing interest in stem cell transplantation therapy, to treat chronic respiratory diseases, using lung epithelial cells or alveolospheres derived from endogenous lung progenitor cells. However, optimal transplantation strategy of these cells has not been addressed. To gain insight into the optimization of stem cell transplantation therapy, we investigated whether lung cell engraftment potential differ among different developmental stages. After preconditioning with irradiation and elastase to induce lung damage, we infused embryonic day 15.5 (E15.5) CAG-EGFP whole lung cells, and confirmed the engraftment of epithelial cells, endothelial cells, and mesenchymal cells. The number of EGFP-positive epithelial cells increased from day 7 to 28 after infusion. Among epithelial cells derived from E13.5, E15.5, E18.5, P7, P14, and P56 mice, E15.5 cells demonstrated the most efficient engraftment. In vitro, E15.5 epithelial cells showed high proliferation potential. Transcriptome analyses of sorted epithelial cells from E13.5, E15.5, E18.5, P14, and P56 mice revealed that cell cycle and cell-cell adhesion genes were highly enriched in E15.5 epithelial cells. Our findings suggest that cell therapy for lung diseases might be most effective when epithelial cells with transcriptional traits similar to those of E15.5 epithelial cells are used.
Collapse
Affiliation(s)
- Kazushige Shiraishi
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.,Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Noda, 278-0022, Japan
| | - Shigeyuki Shichino
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.,Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Noda, 278-0022, Japan
| | - Tatsuya Tsukui
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Shinichi Hashimoto
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.,Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Noda, 278-0022, Japan.,Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-8641, Japan
| | - Satoshi Ueha
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.,Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Noda, 278-0022, Japan
| | - Kouji Matsushima
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan. .,Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Noda, 278-0022, Japan.
| |
Collapse
|
28
|
Chen L, Chen H, Chen P, Zhang W, Wu C, Sun C, Luo W, Zheng L, Liu Z, Liang G. Development of 2-amino-4-phenylthiazole analogues to disrupt myeloid differentiation factor 88 and prevent inflammatory responses in acute lung injury. Eur J Med Chem 2019; 161:22-38. [PMID: 30342423 DOI: 10.1016/j.ejmech.2018.09.068] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/11/2018] [Accepted: 09/26/2018] [Indexed: 11/16/2022]
|
29
|
The early detection of asthma based on blood gene expression. Mol Biol Rep 2018; 46:217-223. [PMID: 30421126 DOI: 10.1007/s11033-018-4463-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/01/2018] [Indexed: 01/10/2023]
Abstract
Asthma is a complex heterogeneous disorder with hereditary tendency and the most widely used therapy is inhalation of anti-inflammatory corticosteroids. But it has systemic side effects. If the chronic inflammation can be detected in early stage, the dosage of corticosteroids will be low and the side effects can be avoided. Therefore, to discover the early stage blood biomarkers for asthma, we analyzed the gene expression profiles in the blood of 77 moderate asthma patients and 87 healthy controls. With advanced feature selection methods, minimal Redundancy Maximal Relevance and Incremental Feature Selection, we identified 31 genes, such as MYD88, ZFP36, CCR3 and CYP3A5, as the optimal asthma biomarker. The sensitivity, specificity and accuracy of the 31-gene Support Vector Machine predictor evaluated with Leave-One-Out Cross Validation were 0.870, 0.816 and 0.841, respectively. Through literature survey, many biomarker genes have asthma associated functions. Our results not only provided the easy-to-apply blood gene expression biomarkers for early detection of asthma, but also an explainable qualitative model with biological significance.
Collapse
|
30
|
Proteases and Their Inhibitors in Chronic Obstructive Pulmonary Disease. J Clin Med 2018; 7:jcm7090244. [PMID: 30154365 PMCID: PMC6162857 DOI: 10.3390/jcm7090244] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/24/2018] [Accepted: 08/25/2018] [Indexed: 12/21/2022] Open
Abstract
In the context of respiratory disease, chronic obstructive pulmonary disease (COPD) is the leading cause of mortality worldwide. Despite much development in the area of drug development, currently there are no effective medicines available for the treatment of this disease. An imbalance in the protease: Antiprotease ratio in the COPD lung remains an important aspect of COPD pathophysiology and several studies have shown the efficacy of antiprotease therapy in both in vitro and in vivo COPD models. However more in-depth studies will be required to validate the efficacy of lead drug molecules targeting these proteases. This review discusses the current status of protease-directed drugs used for treating COPD and explores the future prospects of utilizing the potential of antiprotease-based therapeutics as a treatment for this disease.
Collapse
|
31
|
Di Padova F, Quesniaux VFJ, Ryffel B. MyD88 as a therapeutic target for inflammatory lung diseases. Expert Opin Ther Targets 2018; 22:401-408. [PMID: 29658361 DOI: 10.1080/14728222.2018.1464139] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Myeloid differentiation primary response protein 88 (MyD88) is a critical adaptor protein involved in Toll-like and IL-1 receptor family signaling controlling innate immune responses and inflammation. Genetic deletion of MyD88 function results in profound suppression of inflammation and reduced resistance of the host to pathogens indicating non-redundant roles of MyD88. The TIR domain is critical for MyD88 dimerization and signaling for TLR and IL-1R family receptor. Areas covered: Emerging evidence suggests that chemical disruption of the TIR domain attenuates cell activation and inhibits in vivo MyD88-dependent inflammation. We review the development of MyD88 dimerization disruptors as a novel therapeutic approach of respiratory diseases with a focus on COPD. Expert opinion: There is a proof of concept that therapeutic targeting of MyD88 is feasible and first preclinical data are highly promising. This opens a great opportunity to treat exacerbations of COPD and other chronic respiratory diseases. However, extensive preclinical investigations and risk analyses are required with carefully evaluation of reduced host resistance and opportunistic infections.
Collapse
Affiliation(s)
| | - Valerie F J Quesniaux
- b Laboratory of experimental and molecular immunology and neurogenetics (INEM) , UMR 7355 CNRS-University of Orleans , Orleans , France
| | - Bernhard Ryffel
- b Laboratory of experimental and molecular immunology and neurogenetics (INEM) , UMR 7355 CNRS-University of Orleans , Orleans , France.,c IDM, Institute of Infectious Disease and Molecular Medicine , University of Cape Town , Cape Town , RSA
| |
Collapse
|
32
|
Song J, Kim W, Kim YB, Kim B, Lee K. Time course of polyhexamethyleneguanidine phosphate-induced lung inflammation and fibrosis in mice. Toxicol Appl Pharmacol 2018; 345:94-102. [PMID: 29476863 DOI: 10.1016/j.taap.2018.02.013] [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: 11/06/2017] [Revised: 02/17/2018] [Accepted: 02/20/2018] [Indexed: 12/15/2022]
Abstract
Pulmonary fibrosis is a chronic progressive disease with unknown etiology and has poor prognosis. Polyhexamethyleneguanidine phosphate (PHMG-P) causes acute interstitial pneumonia and pulmonary fibrosis in humans when it exposed to the lung. In a previous study, when rats were exposed to PHMG-P through inhalation for 3 weeks, lung inflammation and fibrosis was observed even after 3 weeks of recovery. In this study, we aimed to determine the time course of PHMG-P-induced lung inflammation and fibrosis. We compared pathological action of PHMG-P with that of bleomycin (BLM) and investigated the mechanism underlying PHMG-P-induced lung inflammation and fibrosis. PHMG-P (0.9 mg/kg) or BLM (1.5 mg/kg) was intratracheally administered to mice. At weeks 1, 2, 4 and 10 after instillation, the levels of inflammatory and fibrotic markers and the expression of inflammasome proteins were measured. The inflammatory and fibrotic responses were upregulated until 10 and 4 weeks in the PHMG-P and BLM groups, respectively. Immune cell infiltration and considerable collagen deposition in the peribronchiolar and interstitial areas of the lungs, fibroblast proliferation, and hyperplasia of type II epithelial cells were observed. NALP3 inflammasome activation was detected in the PHMG-P group until 4 weeks, which is suspected to be the main reason for the persistent inflammatory response and exacerbation of fibrotic changes. Most importantly, the pathological changes in the PHMG-P group were similar to those observed in humidifier disinfectant-associated patients. A single exposure of PHMG-P led to persistent pulmonary inflammation and fibrosis for at least 10 weeks.
Collapse
Affiliation(s)
- Jeongah Song
- Systems Toxicology Center, Predictive Toxicology Department, Korea Institute of Toxicology, Daejeon 305-343, Republic of Korea
| | - Woojin Kim
- Pathology Analytical Research Center, Korea Institute of Toxicology, Daejeon 305-343, Republic of Korea
| | - Yong-Bum Kim
- Pathology Analytical Research Center, Korea Institute of Toxicology, Daejeon 305-343, Republic of Korea
| | - Bumseok Kim
- Biosafety Research Institute and Laboratory of Pathology (BK21 Plus Program), College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea.
| | - Kyuhong Lee
- Inhalation Toxicology Research Center, Korea Institute of Toxicology, Jeonbuk 580-185, Republic of Korea; Human and Environment Toxicology, University of Science and Technology, Daejeon 305-350, Republic of Korea.
| |
Collapse
|
33
|
Abstract
Inflammasomes are large innate cytoplasmic complexes that play a major role in promoting inflammation in the lung in response to a range of environmental and infectious stimuli. Inflammasomes are critical for driving acute innate immune responses that resolve infection and maintain tissue homeostasis. However, dysregulated or excessive inflammasome activation can be detrimental. Here, we discuss the plethora of recent data from clinical studies and small animal disease models that implicate excessive inflammasome responses in the pathogenesis of a number of acute and chronic respiratory inflammatory diseases. Understanding of the role of inflammasomes in lung disease is of great therapeutic interest.
Collapse
Affiliation(s)
- Saleela M Ruwanpura
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Sarah Rosli
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Michelle D Tate
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
34
|
Talikka M, Martin F, Sewer A, Vuillaume G, Leroy P, Luettich K, Chaudhary N, Peck MJ, Peitsch MC, Hoeng J. Mechanistic Evaluation of the Impact of Smoking and Chronic Obstructive Pulmonary Disease on the Nasal Epithelium. CLINICAL MEDICINE INSIGHTS-CIRCULATORY RESPIRATORY AND PULMONARY MEDICINE 2017; 11:1179548417710928. [PMID: 28620266 PMCID: PMC5466113 DOI: 10.1177/1179548417710928] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 04/04/2017] [Indexed: 12/27/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the major causes of chronic morbidity and mortality worldwide. The development of markers of COPD onset is hampered by the lack of accessibility to the primary target tissue, and there is a need to consider other sample sources as surrogates for biomarker research. Airborne toxicants pass through the nasal epithelium before reaching the lower airways, and the similarity with bronchial histology makes it an attractive surrogate for lower airways. In this work, we describe the transcriptomics findings from the nasal epithelia of subjects enrolled in a clinical study focusing on the identification of COPD biomarkers. Transcriptomic data were analyzed using the biological network approach that enabled us to pinpoint the biological processes affected in the upper respiratory tract in response to smoking and mild-to-moderate COPD. Our results indicated that nasal and lower airway immune responses were considerably different in COPD subjects and caution should be exercised when using upper airway samples as a surrogate for the lower airway. Nevertheless, the network approach described here could present a sensitive means of identifying smokers at risk of developing COPD.
Collapse
Affiliation(s)
- Marja Talikka
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Florian Martin
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Alain Sewer
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Grégory Vuillaume
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Patrice Leroy
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Karsta Luettich
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Nveed Chaudhary
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Michael J Peck
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Manuel C Peitsch
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| | - Julia Hoeng
- Philip Morris Products SA and Research & Development (R&D), Philip Morris International, Neuchâtel, Switzerland
| |
Collapse
|
35
|
Sakai H, Horiguchi M, Akita T, Ozawa C, Hirokawa M, Oiso Y, Kumagai H, Takeda Y, Tachibana I, Maeda N, Yamashita C. Effect of 4-[(5,6,7,8-Tetrahydro-5,5,8,8-Tetramethyl-2-Naphthalenyl)Carbamoyl]Benzoic Acid (Am80) on Alveolar Regeneration in Adiponectin Deficient-Mice Showing a Chronic Obstructive Pulmonary Disease-Like Pathophysiology. J Pharmacol Exp Ther 2017; 361:501-505. [PMID: 28432078 DOI: 10.1124/jpet.117.240515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/27/2017] [Indexed: 11/22/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is an intractable pulmonary disease that causes widespread and irreversible alveolar collapse. Although COPD occurs worldwide, only symptomatic therapy is currently available. Our objective is the development of therapeutic agents to eradicate COPD. Therefore, we focused on 4-[(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl) carbamoyl] benzoic acid (Am80), which is a derivative of all-trans retinoic acid. We evaluated the effects of Am80 on alveolar repair in a novel COPD model of adiponectin-deficient mice. This mouse model has more symptoms similar to human COPD than the classic elastase-induced emphysema mouse model. Lung volume, computed tomography (CT) values, low-attenuation area ratios, and bone and fat mass were measured by CT. However, the administration of Am80 did not affect these results. To examine the degree of destruction in the alveoli, the mean linear intercept of the alveolar walls was calculated, and assessment of this value confirmed that there was a significant difference between the control (46.3 ± 2.3 μm) and 0.5 mg/kg Am80-treated group (34.4 ± 1.7 µm). All mice survived the treatment, which lasted for more than 6 months, and we did not observe any abnormalities in autopsies performed at 80 weeks of age. These results suggested that Am80 was effective as a novel therapeutic compound for the treatment of COPD.
Collapse
Affiliation(s)
- Hitomi Sakai
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences (H.S., M.Ho., T.A., C.O., M.Hi., O.Y., H.K., C.Y.), and Fusion of Regenerative Medicine with DDS, Research Institute for Science and Technology (M.Ho., C.Y.), Tokyo University of Science, Chiba; Respiratory Medicine, Allergy and Rheumatic Diseases (Y.T., I.T., N.M.), and Department of Metabolism and Atherosclerosis (N.M.), Graduate School of Medicine, Osaka University, Osaka; Department of Medicine, Nissay Hospital, Nippon Life Saiseikai Public Interest Incorporated Foundation, Osaka (I.T.), Japan
| | - Michiko Horiguchi
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences (H.S., M.Ho., T.A., C.O., M.Hi., O.Y., H.K., C.Y.), and Fusion of Regenerative Medicine with DDS, Research Institute for Science and Technology (M.Ho., C.Y.), Tokyo University of Science, Chiba; Respiratory Medicine, Allergy and Rheumatic Diseases (Y.T., I.T., N.M.), and Department of Metabolism and Atherosclerosis (N.M.), Graduate School of Medicine, Osaka University, Osaka; Department of Medicine, Nissay Hospital, Nippon Life Saiseikai Public Interest Incorporated Foundation, Osaka (I.T.), Japan
| | - Tomomi Akita
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences (H.S., M.Ho., T.A., C.O., M.Hi., O.Y., H.K., C.Y.), and Fusion of Regenerative Medicine with DDS, Research Institute for Science and Technology (M.Ho., C.Y.), Tokyo University of Science, Chiba; Respiratory Medicine, Allergy and Rheumatic Diseases (Y.T., I.T., N.M.), and Department of Metabolism and Atherosclerosis (N.M.), Graduate School of Medicine, Osaka University, Osaka; Department of Medicine, Nissay Hospital, Nippon Life Saiseikai Public Interest Incorporated Foundation, Osaka (I.T.), Japan
| | - Chihiro Ozawa
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences (H.S., M.Ho., T.A., C.O., M.Hi., O.Y., H.K., C.Y.), and Fusion of Regenerative Medicine with DDS, Research Institute for Science and Technology (M.Ho., C.Y.), Tokyo University of Science, Chiba; Respiratory Medicine, Allergy and Rheumatic Diseases (Y.T., I.T., N.M.), and Department of Metabolism and Atherosclerosis (N.M.), Graduate School of Medicine, Osaka University, Osaka; Department of Medicine, Nissay Hospital, Nippon Life Saiseikai Public Interest Incorporated Foundation, Osaka (I.T.), Japan
| | - Mai Hirokawa
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences (H.S., M.Ho., T.A., C.O., M.Hi., O.Y., H.K., C.Y.), and Fusion of Regenerative Medicine with DDS, Research Institute for Science and Technology (M.Ho., C.Y.), Tokyo University of Science, Chiba; Respiratory Medicine, Allergy and Rheumatic Diseases (Y.T., I.T., N.M.), and Department of Metabolism and Atherosclerosis (N.M.), Graduate School of Medicine, Osaka University, Osaka; Department of Medicine, Nissay Hospital, Nippon Life Saiseikai Public Interest Incorporated Foundation, Osaka (I.T.), Japan
| | - Yuki Oiso
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences (H.S., M.Ho., T.A., C.O., M.Hi., O.Y., H.K., C.Y.), and Fusion of Regenerative Medicine with DDS, Research Institute for Science and Technology (M.Ho., C.Y.), Tokyo University of Science, Chiba; Respiratory Medicine, Allergy and Rheumatic Diseases (Y.T., I.T., N.M.), and Department of Metabolism and Atherosclerosis (N.M.), Graduate School of Medicine, Osaka University, Osaka; Department of Medicine, Nissay Hospital, Nippon Life Saiseikai Public Interest Incorporated Foundation, Osaka (I.T.), Japan
| | - Harumi Kumagai
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences (H.S., M.Ho., T.A., C.O., M.Hi., O.Y., H.K., C.Y.), and Fusion of Regenerative Medicine with DDS, Research Institute for Science and Technology (M.Ho., C.Y.), Tokyo University of Science, Chiba; Respiratory Medicine, Allergy and Rheumatic Diseases (Y.T., I.T., N.M.), and Department of Metabolism and Atherosclerosis (N.M.), Graduate School of Medicine, Osaka University, Osaka; Department of Medicine, Nissay Hospital, Nippon Life Saiseikai Public Interest Incorporated Foundation, Osaka (I.T.), Japan
| | - Yoshito Takeda
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences (H.S., M.Ho., T.A., C.O., M.Hi., O.Y., H.K., C.Y.), and Fusion of Regenerative Medicine with DDS, Research Institute for Science and Technology (M.Ho., C.Y.), Tokyo University of Science, Chiba; Respiratory Medicine, Allergy and Rheumatic Diseases (Y.T., I.T., N.M.), and Department of Metabolism and Atherosclerosis (N.M.), Graduate School of Medicine, Osaka University, Osaka; Department of Medicine, Nissay Hospital, Nippon Life Saiseikai Public Interest Incorporated Foundation, Osaka (I.T.), Japan
| | - Isao Tachibana
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences (H.S., M.Ho., T.A., C.O., M.Hi., O.Y., H.K., C.Y.), and Fusion of Regenerative Medicine with DDS, Research Institute for Science and Technology (M.Ho., C.Y.), Tokyo University of Science, Chiba; Respiratory Medicine, Allergy and Rheumatic Diseases (Y.T., I.T., N.M.), and Department of Metabolism and Atherosclerosis (N.M.), Graduate School of Medicine, Osaka University, Osaka; Department of Medicine, Nissay Hospital, Nippon Life Saiseikai Public Interest Incorporated Foundation, Osaka (I.T.), Japan
| | - Norikazu Maeda
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences (H.S., M.Ho., T.A., C.O., M.Hi., O.Y., H.K., C.Y.), and Fusion of Regenerative Medicine with DDS, Research Institute for Science and Technology (M.Ho., C.Y.), Tokyo University of Science, Chiba; Respiratory Medicine, Allergy and Rheumatic Diseases (Y.T., I.T., N.M.), and Department of Metabolism and Atherosclerosis (N.M.), Graduate School of Medicine, Osaka University, Osaka; Department of Medicine, Nissay Hospital, Nippon Life Saiseikai Public Interest Incorporated Foundation, Osaka (I.T.), Japan
| | - Chikamasa Yamashita
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences (H.S., M.Ho., T.A., C.O., M.Hi., O.Y., H.K., C.Y.), and Fusion of Regenerative Medicine with DDS, Research Institute for Science and Technology (M.Ho., C.Y.), Tokyo University of Science, Chiba; Respiratory Medicine, Allergy and Rheumatic Diseases (Y.T., I.T., N.M.), and Department of Metabolism and Atherosclerosis (N.M.), Graduate School of Medicine, Osaka University, Osaka; Department of Medicine, Nissay Hospital, Nippon Life Saiseikai Public Interest Incorporated Foundation, Osaka (I.T.), Japan
| |
Collapse
|
36
|
Zou Y, Chen X, Liu J, Zhou DB, Kuang X, Xiao J, Yu Q, Lu X, Li W, Xie B, Chen Q. Serum IL-1β and IL-17 levels in patients with COPD: associations with clinical parameters. Int J Chron Obstruct Pulmon Dis 2017; 12:1247-1254. [PMID: 28490868 PMCID: PMC5413485 DOI: 10.2147/copd.s131877] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
COPD is a chronic airway inflammatory disease characterized mainly by neutrophil airway infiltrations. Interleukin (IL)-1β and IL-17 are the key mediators of neutrophilic airway inflammation in COPD. This study was undertaken to evaluate the serum IL-1β and IL-17 levels and associations between these two key mediators with clinical parameters in COPD patients. Serum samples were collected from 60 COPD subjects during the acute exacerbation of COPD, 60 subjects with stable COPD and 40 healthy control subjects. Commercial enzyme-linked immunosorbent assay kits were used to measure the serum IL-1β and IL-17 concentrations. The association between serum IL-1β and IL-17 with FEV1% predicted, C-reactive protein, neutrophil percentage and smoking status (pack-years) was assessed in the COPD patients. We found that serum IL-1β and IL-17 levels in acute exacerbation of COPD subjects were significantly higher than that in stable COPD or control subjects and were positively correlated to serum C-reactive protein levels, neutrophil % and smoking status (pack-years) but negatively correlated with FEV1% predicted in COPD patients. More importantly, serum IL-1β levels were markedly positively associated with serum IL-17 levels in patients with COPD (P=0.741, P<0.001). In conclusion, elevated serum IL-1β and IL-17 levels may be used as a biomarker for indicating persistent neutrophilic airway inflammation and potential ongoing exacerbation of COPD.
Collapse
Affiliation(s)
- Yong Zou
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Xi Chen
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Jie Liu
- Department of Emergency, The First People's Hospital of Changsha, Changsha, People's Republic of China
| | - Dong Bo Zhou
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Xiao Kuang
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Jian Xiao
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Qiao Yu
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Xiaoxiao Lu
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Wei Li
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Bin Xie
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Qiong Chen
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| |
Collapse
|
37
|
Craig JM, Scott AL, Mitzner W. Immune-mediated inflammation in the pathogenesis of emphysema: insights from mouse models. Cell Tissue Res 2017; 367:591-605. [PMID: 28164246 PMCID: PMC5366983 DOI: 10.1007/s00441-016-2567-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/21/2016] [Indexed: 12/31/2022]
Abstract
The cellular mechanisms that result in the initiation and progression of emphysema are clearly complex. A growing body of human data combined with discoveries from mouse models utilizing cigarette smoke exposure or protease administration have improved our understanding of emphysema development by implicating specific cell types that may be important for the pathophysiology of chronic obstructive pulmonary disease. The most important aspects of emphysematous damage appear to be oxidative or protease stress and sustained macrophage activation and infiltration of other immune cells leading to epithelial damage and cell death. Despite the identification of these associated processes and cell types in many experimental studies, the reasons why cigarette smoke and other pollutants result in unremitting damage instead of injury resolution are still uncertain. We propose an important role for macrophages in the sequence of events that lead and maintain this chronic tissue pathologic process in emphysema. This model involves chronic activation of macrophage subtypes that precludes proper healing of the lung. Further elucidation of the cross-talk between epithelial cells that release damage-associated signals and the cellular immune effectors that respond to these cues is a critical step in the development of novel therapeutics that can restore proper lung structure and function to those afflicted with emphysema.
Collapse
Affiliation(s)
- John M Craig
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe St., Baltimore, MD, USA
| | - Alan L Scott
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Wayne Mitzner
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe St., Baltimore, MD, USA.
| |
Collapse
|
38
|
Fujita T, Yoshioka K, Umezawa H, Tanaka K, Naito Y, Nakayama T, Hatano M, Tatsumi K, Kasuya Y. Role of CD69 in the pathogenesis of elastase-induced pulmonary inflammation and emphysema. Biochem Biophys Rep 2016; 7:400-407. [PMID: 28955931 PMCID: PMC5613653 DOI: 10.1016/j.bbrep.2016.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 06/19/2016] [Accepted: 07/13/2016] [Indexed: 01/04/2023] Open
Abstract
Cluster of differentiation 69 (CD69), known as an early activation marker of lymphocytes, has been demonstrated to regulate inflammatory events in various disease models. Although the increased number of CD69-expressed T lymphocytes in the lungs of patients with chronic obstructive pulmonary disease (COPD) has been reported, a functional role of CD69 in the pathogenesis of COPD remains unknown. To address to this question, CD69-deficient (CD69KO) mice and wild-type (WT) mice were subjected to a mouse model of porcine pancreatic elastase (PPE)-induced pulmonary inflammation and emphysema. In the two genotypes, PPE increased counts of macrophages, neutrophils and lymphocytes in bronchoalveolar lavage fluid (BALF) and induced emphysematous changes in the lung, whereas those two pathological signs were significantly enhanced in CD69KO mice compared to WT mice. Moreover, the PPE-induced levels of IL-17 and IL-6 in BALF were significantly higher in CD69KO mice than in WT mice at the acute inflammatory phase. Immunofluorescent studies showed that IL-17 and IL-6 were predominantly expressed in CD4+ and γδ T cells and macrophages, respectively. Concomitant administration of IL-17- and IL-6-neutralizing antibodies significantly attenuated the PPE-induced emphysematous changes in the two genotypes. These findings suggest that CD69 negatively regulates the development of PPE-induced emphysema in part at least through modulating function of IL-17-producing T cells.
Collapse
Affiliation(s)
- Tetsuo Fujita
- Department of Respirology, Chiba University, Chiba 260-8670, Japan.,Department of Biochemistry and Molecular Pharmacology, Chiba University, Chiba 260-8670, Japan
| | - Kento Yoshioka
- Department of Biochemistry and Molecular Pharmacology, Chiba University, Chiba 260-8670, Japan.,Department of Biomedical Science, Chiba University, Chiba 260-8670, Japan
| | - Hiroki Umezawa
- Department of Respirology, Chiba University, Chiba 260-8670, Japan.,Department of Biochemistry and Molecular Pharmacology, Chiba University, Chiba 260-8670, Japan
| | - Kensuke Tanaka
- Department of Respirology, Chiba University, Chiba 260-8670, Japan.,Department of Biochemistry and Molecular Pharmacology, Chiba University, Chiba 260-8670, Japan
| | - Yusuke Naito
- Department of Respirology, Chiba University, Chiba 260-8670, Japan.,Department of Biochemistry and Molecular Pharmacology, Chiba University, Chiba 260-8670, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Masahiko Hatano
- Department of Biomedical Science, Chiba University, Chiba 260-8670, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Chiba University, Chiba 260-8670, Japan
| | - Yoshitoshi Kasuya
- Department of Biochemistry and Molecular Pharmacology, Chiba University, Chiba 260-8670, Japan.,Department of Biomedical Science, Chiba University, Chiba 260-8670, Japan
| |
Collapse
|
39
|
Involvement of matrix metalloproteinases (MMPs) and inflammasome pathway in molecular mechanisms of fibrosis. Biosci Rep 2016; 36:BSR20160107. [PMID: 27247426 PMCID: PMC4945993 DOI: 10.1042/bsr20160107] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 05/31/2016] [Indexed: 12/18/2022] Open
Abstract
Fibrosis is a basic connective tissue lesion defined by the increase in the fibrillar extracellular matrix (ECM) components in tissue or organ. Matrix metalloproteinases (MMPs) are a major group of proteases known to regulate the turn-over of ECM and so they are suggested to be important in tissue remodelling observed during fibrogenic process associated with chronic inflammation. Tissue remodelling is the result of an imbalance in the equilibrium of the normal processes of synthesis and degradation of ECM components markedly controlled by the MMPs/TIMP imbalance. We previously showed an association of the differences in collagen deposition in the lungs of bleomycin-treated mice with a reduced molar pro-MMP-9/TIMP-1 ratio. Using the carbon tetrachloride (CCl4) preclinical model of liver fibrosis in mice, we observed a significant increase in collagen deposition with increased expression and release of tissue inhibitors of metalloproteinase (TIMP)-1 both at 24 h and 3 weeks later. This suggests an early altered regulation of matrix turnover involved in the development of fibrosis. We also demonstrated an activation of NLRP3-inflammasome pathway associated with the IL-1R/MyD88 signalling in the development of experimental fibrosis both in lung and liver. This was also associated with an increased expression of purinergic receptors mainly P2X7. Finally, these observations emphasize those effective therapies for these disorders must be given early in the natural history of the disease, prior to the development of tissue remodelling and fibrosis.
Collapse
|
40
|
The sbv IMPROVER project team and challenge best performers, Namasivayam AA, Morales AF, Lacave ÁMF, Tallam A, Simovic B, Alfaro DG, Bobbili DR, Martin F, Androsova G, Shvydchenko I, Park J, Calvo JV, Hoeng J, Peitsch MC, Racero MGV, Biryukov M, Talikka M, Pérez MB, Rohatgi N, Díaz-Díaz N, Mandarapu R, Ruiz RA, Davidyan S, Narayanasamy S, Boué S, Guryanova S, Arbas SM, Menon S, Xiang Y. Community-Reviewed Biological Network Models for Toxicology and Drug Discovery Applications. GENE REGULATION AND SYSTEMS BIOLOGY 2016; 10:51-66. [PMID: 27429547 PMCID: PMC4944831 DOI: 10.4137/grsb.s39076] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/31/2016] [Accepted: 04/12/2016] [Indexed: 12/13/2022]
Abstract
Biological network models offer a framework for understanding disease by describing the relationships between the mechanisms involved in the regulation of biological processes. Crowdsourcing can efficiently gather feedback from a wide audience with varying expertise. In the Network Verification Challenge, scientists verified and enhanced a set of 46 biological networks relevant to lung and chronic obstructive pulmonary disease. The networks were built using Biological Expression Language and contain detailed information for each node and edge, including supporting evidence from the literature. Network scoring of public transcriptomics data inferred perturbation of a subset of mechanisms and networks that matched the measured outcomes. These results, based on a computable network approach, can be used to identify novel mechanisms activated in disease, quantitatively compare different treatments and time points, and allow for assessment of data with low signal. These networks are periodically verified by the crowd to maintain an up-to-date suite of networks for toxicology and drug discovery applications.
Collapse
Affiliation(s)
| | - Aishwarya Alex Namasivayam
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, Esch-sur-Alzette, Luxembourg
| | | | | | - Aravind Tallam
- TWINCORE, Zentrum für Experimentelle und Klinische Infektionsforschung, Hannover, Germany
| | | | | | - Dheeraj Reddy Bobbili
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, Esch-sur-Alzette, Luxembourg
| | - Florian Martin
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud, Neuchâtel, Switzerland (part of Philip Morris International group of companies)
| | - Ganna Androsova
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, Esch-sur-Alzette, Luxembourg
| | - Irina Shvydchenko
- Kuban State University of Physical Education, Sport and Tourism, Krasnodar, Russia
| | | | - Jorge Val Calvo
- Center for Molecular Biology, “Severo Ochoa” – Spanish National Research Council, Madrid, Spain
| | - Julia Hoeng
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud, Neuchâtel, Switzerland (part of Philip Morris International group of companies)
| | - Manuel C. Peitsch
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud, Neuchâtel, Switzerland (part of Philip Morris International group of companies)
| | | | - Maria Biryukov
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, Esch-sur-Alzette, Luxembourg
| | - Marja Talikka
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud, Neuchâtel, Switzerland (part of Philip Morris International group of companies)
| | | | - Neha Rohatgi
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland
| | | | - Rajesh Mandarapu
- Prakhya Research Laboratories, Lakshminagar, Selaiyur, Chennai, Tamil Nadu, India
| | | | - Sergey Davidyan
- Institute of Biochemical Physics Russian Academy of Sciences, Moscow, Russia
| | - Shaman Narayanasamy
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, Esch-sur-Alzette, Luxembourg
| | - Stéphanie Boué
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud, Neuchâtel, Switzerland (part of Philip Morris International group of companies)
| | - Svetlana Guryanova
- Institute of Bioorganic Chemistry Russian Academy of Sciences, Moscow, Russia
| | - Susana Martínez Arbas
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, Esch-sur-Alzette, Luxembourg
| | - Swapna Menon
- AnalyzeDat Consulting Services, Edapally Byepass Junction, Kochi, Kerala, India
| | - Yang Xiang
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud, Neuchâtel, Switzerland (part of Philip Morris International group of companies)
| |
Collapse
|
41
|
Foronjy RF, Salathe MA, Dabo AJ, Baumlin N, Cummins N, Eden E, Geraghty P. TLR9 expression is required for the development of cigarette smoke-induced emphysema in mice. Am J Physiol Lung Cell Mol Physiol 2016; 311:L154-66. [PMID: 27288485 PMCID: PMC4967186 DOI: 10.1152/ajplung.00073.2016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/04/2016] [Indexed: 11/22/2022] Open
Abstract
The expression of Toll-like receptor (TLR)-9, a pathogen recognition receptor that recognizes unmethylated CpG sequences in microbial DNA molecules, is linked to the pathogenesis of several lung diseases. TLR9 expression and signaling was investigated in animal and cell models of chronic obstructive pulmonary disease (COPD). We observed enhanced TLR9 expression in mouse lungs following exposure to cigarette smoke. Tlr9(-/-) mice were resistant to cigarette smoke-induced loss of lung function as determined by mean linear intercept, total lung capacity, lung compliance, and tissue elastance analysis. Tlr9 expression also regulated smoke-mediated immune cell recruitment to the lung; apoptosis; expression of granulocyte-colony stimulating factor (G-CSF), the CXCL5 protein, and matrix metalloproteinase-2 (MMP-2); and protein tyrosine phosphatase 1B (PTP1B) activity in the lung. PTP1B, a phosphatase with anti-inflammatory abilities, was identified as binding to TLR9. In vivo delivery of a TLR9 agonist enhanced TLR9 binding to PTP1B, which inactivated PTP1B. Ptp1b(-/-) mice had elevated lung concentrations of G-CSF, CXCL5, and MMP-2, and tissue expression of type-1 interferon following TLR9 agonist administration, compared with wild-type mice. TLR9 responses were further determined in fully differentiated normal human bronchial epithelial (NHBE) cells isolated from nonsmoker, smoker, and COPD donors, and then cultured at air liquid interface. NHBE cells from smokers and patients with COPD expressed more TLR9 and secreted greater levels of G-CSF, IL-6, CXCL5, IL-1β, and MMP-2 upon TLR9 ligand stimulation compared with cells from nonsmoker donors. Although TLR9 combats infection, our results indicate that TLR9 induction can affect lung function by inactivating PTP1B and upregulating expression of proinflammatory cytokines.
Collapse
Affiliation(s)
- Robert F Foronjy
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, New York; Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Matthias A Salathe
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Miami, Miami, Florida; and
| | - Abdoulaye J Dabo
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, New York; Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Nathalie Baumlin
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Miami, Miami, Florida; and
| | - Neville Cummins
- Division of Pulmonary and Critical Care Medicine, Mount Sinai Roosevelt, Mount Sinai Health System, New York, New York
| | - Edward Eden
- Division of Pulmonary and Critical Care Medicine, Mount Sinai Roosevelt, Mount Sinai Health System, New York, New York
| | - Patrick Geraghty
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, New York; Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, New York;
| |
Collapse
|
42
|
Xia X, Fu J, Song X, Shi Q, Su C, Song E, Song Y. Neohesperidin dihydrochalcone down-regulates MyD88-dependent and -independent signaling by inhibiting endotoxin-induced trafficking of TLR4 to lipid rafts. Free Radic Biol Med 2015; 89:522-32. [PMID: 26453923 DOI: 10.1016/j.freeradbiomed.2015.08.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/25/2015] [Accepted: 08/26/2015] [Indexed: 02/07/2023]
Abstract
Fulminant hepatic failure (FHF) is a lethal clinical syndrome characterized by the activation of macrophages and the increased production of inflammatory mediators. The purpose of this study was to investigate the effects of neohesperidin dihydrochalcone (NHDC), a widely-used low caloric artificial sweetener against FHF. An FHF experimental model was established in mice by intraperitoneal injection of D-galactosamine (d-GalN) (400mg/kg)/lipopolysaccharides (LPS) (10 μg/kg). Mice were orally administered NHDC for 6 continuous days and at 1h before d-GalN/LPS administration. RAW264.7 macrophages were used as an in vitro model. Cells were pre-treated with NHDC for 1h before stimulation with LPS (10 μg/ml) for 6h. d-GalN/LPS markedly increased the serum transaminase activities and levels of oxidative and inflammatory markers, which were significantly attenuated by NHDC. Mechanistic analysis indicated that NHDC inhibited LPS-induced myeloid differentiation factor 88 (MyD88) and TIR-containing adapter molecule (TRIF)-dependent signaling. Transient transfection of TLR4 or MyD88 siRNA inhibited the downstream inflammatory signaling. This effect could also be achieved by the pretreatment with NHDC. The fluorescence microscopy and flow cytometry results suggested that NHDC potently inhibited the binding of LPS to TLR4 in RAW264.7 macrophages. In addition, the inhibitory effect of NHDC on LPS-induced translocation of TLR4 into lipid raft domains played an important role in the amelioration of production of downstream pro-inflammatory molecules. Furthermore, the activation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) by NHDC inhibited TLR4 signaling. In conclusion, our results suggest that NHDC attenuates d-GalN/LPS-induced FHF by inhibiting the TLR4-mediated inflammatory pathway, demonstrating a new application of NHDC as a hepatoprotective agent.
Collapse
Affiliation(s)
- Xiaomin Xia
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China, 400715
| | - Juanli Fu
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China, 400715
| | - Xiufang Song
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China, 400715
| | - Qiong Shi
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China, 400715
| | - Chuanyang Su
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China, 400715
| | - Erqun Song
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China, 400715
| | - Yang Song
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China, 400715.
| |
Collapse
|
43
|
Limjunyawong N, Craig JM, Lagassé HAD, Scott AL, Mitzner W. Experimental progressive emphysema in BALB/cJ mice as a model for chronic alveolar destruction in humans. Am J Physiol Lung Cell Mol Physiol 2015; 309:L662-76. [PMID: 26232300 PMCID: PMC4593839 DOI: 10.1152/ajplung.00214.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 07/29/2015] [Indexed: 01/09/2023] Open
Abstract
Emphysema, one of the major components of chronic obstructive pulmonary disease (COPD), is characterized by the progressive and irreversible loss of alveolar lung tissue. Even though >80% of COPD cases are associated with cigarette smoking, only a relatively small proportion of smokers develop emphysema, suggesting a potential role for genetic factors in determining individual susceptibility to emphysema. Although strain-dependent effects have been shown in animal models of emphysema, the molecular basis underlying this intrinsic susceptibility is not fully understood. In this present study, we investigated emphysema development using the elastase-induced experimental emphysema model in two commonly used mouse strains, C57BL/6J and BALB/cJ. The results demonstrate that mice with different genetic backgrounds show disparate susceptibility to the development of emphysema. BALB/cJ mice were found to be much more sensitive than C57BL/6J to elastase injury in both a dose-dependent and time-dependent manner, as measured by significantly higher mortality, greater body weight loss, greater decline in lung function, and a greater loss of alveolar tissue. The more susceptible BALB/cJ strain also showed the persistence of inflammatory cells in the lung, especially macrophages and lymphocytes. A comparative gene expression analysis following elastase-induced injury showed BALB/cJ mice had elevated levels of il17A mRNA and a number of classically (M1) and alternatively (M2) activated macrophage genes, whereas the C57BL/6J mice demonstrated augmented levels of interferon-γ. These findings suggest a possible role for these cellular and molecular mediators in modulating the severity of emphysema and highlight the possibility that they might contribute to the heterogeneity observed in clinical emphysema outcomes.
Collapse
Affiliation(s)
- Nathachit Limjunyawong
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland; and
| | - John M Craig
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland; and
| | - H A Daniel Lagassé
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Alan L Scott
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Wayne Mitzner
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland; and
| |
Collapse
|
44
|
Poole JA, Wyatt TA, Romberger DJ, Staab E, Simet S, Reynolds SJ, Sisson JH, Kielian T. MyD88 in lung resident cells governs airway inflammatory and pulmonary function responses to organic dust treatment. Respir Res 2015; 16:111. [PMID: 26376975 PMCID: PMC4574163 DOI: 10.1186/s12931-015-0272-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 09/03/2015] [Indexed: 11/10/2022] Open
Abstract
Inhalation of organic dusts within agriculture environments contributes to the development and/or severity of airway diseases, including asthma and chronic bronchitis. MyD88 KO (knockout) mice are nearly completely protected against the inflammatory and bronchoconstriction effects induced by acute organic dust extract (ODE) treatments. However, the contribution of MyD88 in lung epithelial cell responses remains unclear. In the present study, we first addressed whether ODE-induced changes in epithelial cell responses were MyD88-dependent by quantitating ciliary beat frequency and cell migration following wounding by electric cell-substrate impedance sensing. We demonstrate that the normative ciliary beat slowing response to ODE is delayed in MyD88 KO tracheal epithelial cells as compared to wild type (WT) control. Similarly, the normative ODE-induced slowing of cell migration in response to wound repair was aberrant in MyD88 KO cells. Next, we created MyD88 bone marrow chimera mice to investigate the relative contribution of MyD88-dependent signaling in lung resident (predominately epithelial cells) versus hematopoietic cells. Importantly, we demonstrate that ODE-induced airway hyperresponsiveness is MyD88-dependent in lung resident cells, whereas MyD88 action in hematopoietic cells is mainly responsible for ODE-induced TNF-α release. MyD88 signaling in lung resident and hematopoietic cells are necessary for ODE-induced IL-6 and neutrophil chemoattractant (CXCL1 and CXCL2) release and neutrophil influx. Collectively, these findings underscore an important role for MyD88 in lung resident cells for regulating ciliary motility, wound repair and inflammatory responses to ODE, and moreover, show that airway hyperresponsiveness appears uncoupled from airway inflammatory consequences to organic dust challenge in terms of MyD88 involvement.
Collapse
Affiliation(s)
- Jill A Poole
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE, 68198-5990, USA.
| | - Todd A Wyatt
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE, 68198-5990, USA.,Department of Environmental, Agricultural & Occupational Health, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE, 68198-5990, USA.,VA Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
| | - Debra J Romberger
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE, 68198-5990, USA.,Department of Environmental, Agricultural & Occupational Health, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE, 68198-5990, USA
| | - Elizabeth Staab
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE, 68198-5990, USA
| | - Samantha Simet
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE, 68198-5990, USA
| | - Stephen J Reynolds
- High Plains Intermountain Center for Agricultural Health and Safety, Department of Environmental and Radiological Health Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Joseph H Sisson
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE, 68198-5990, USA
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE, 68198-5990, USA
| |
Collapse
|
45
|
Alfaidi M, Wilson H, Daigneault M, Burnett A, Ridger V, Chamberlain J, Francis S. Neutrophil elastase promotes interleukin-1β secretion from human coronary endothelium. J Biol Chem 2015; 290:24067-78. [PMID: 26269588 DOI: 10.1074/jbc.m115.659029] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Indexed: 11/06/2022] Open
Abstract
The endothelium is critically involved in the pathogenesis of atherosclerosis by producing pro-inflammatory mediators, including IL-1β. Coronary arteries from patients with ischemic heart disease express large amounts of IL-1β in the endothelium. However, the mechanism by which endothelial cells (ECs) release IL-1β remains to be elucidated. We investigated neutrophil elastase (NE), a potent serine protease detected in vulnerable areas of human carotid plaques, as a potential "trigger" for IL-1β processing and release. This study tested the hypothesis that NE potentiates the processing and release of IL-1β from human coronary endothelium. We found that NE cleaves the pro-isoform of IL-1β in ECs and causes significant secretion of bioactive IL-1β via extracellular vesicles. This release was attenuated significantly by inhibition of neutrophil elastase but not caspase-1. Transient increases in intracellular Ca(2+) levels were observed prior to secretion. Inside ECs, and after NE treatment only, IL-1β was detected within LAMP-1-positive multivesicular bodies. The released vesicles contained bioactive IL-1β. In vivo, in experimental atherosclerosis, NE was detected in mature atherosclerotic plaques, predominantly in the endothelium, alongside IL-1β. This study reveals a novel mechanistic link between NE expression in atherosclerotic plaques and concomitant pro-inflammatory bioactive IL-1β secretion from ECs. This could reveal additional potential anti-IL-1β therapeutic targets and provide further insights into the inflammatory process by which vascular disease develops.
Collapse
Affiliation(s)
- Mabruka Alfaidi
- From the Department of Cardiovascular Science, Medical School, University of Sheffield, Sheffield S10 2RX, United Kingdom
| | - Heather Wilson
- From the Department of Cardiovascular Science, Medical School, University of Sheffield, Sheffield S10 2RX, United Kingdom
| | - Marc Daigneault
- From the Department of Cardiovascular Science, Medical School, University of Sheffield, Sheffield S10 2RX, United Kingdom
| | - Amanda Burnett
- From the Department of Cardiovascular Science, Medical School, University of Sheffield, Sheffield S10 2RX, United Kingdom
| | - Victoria Ridger
- From the Department of Cardiovascular Science, Medical School, University of Sheffield, Sheffield S10 2RX, United Kingdom
| | - Janet Chamberlain
- From the Department of Cardiovascular Science, Medical School, University of Sheffield, Sheffield S10 2RX, United Kingdom
| | - Sheila Francis
- From the Department of Cardiovascular Science, Medical School, University of Sheffield, Sheffield S10 2RX, United Kingdom
| |
Collapse
|
46
|
Solleti SK, Simon DM, Srisuma S, Arikan MC, Bhattacharya S, Rangasamy T, Bijli KM, Rahman A, Crossno JT, Shapiro SD, Mariani TJ. Airway epithelial cell PPARγ modulates cigarette smoke-induced chemokine expression and emphysema susceptibility in mice. Am J Physiol Lung Cell Mol Physiol 2015; 309:L293-304. [PMID: 26024894 DOI: 10.1152/ajplung.00287.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 05/26/2015] [Indexed: 11/22/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a highly prevalent, chronic inflammatory lung disease with limited existing therapeutic options. While modulation of peroxisome proliferator-activating receptor (PPAR)-γ activity can modify inflammatory responses in several models of lung injury, the relevance of the PPARG pathway in COPD pathogenesis has not been previously explored. Mice lacking Pparg specifically in airway epithelial cells displayed increased susceptibility to chronic cigarette smoke (CS)-induced emphysema, with excessive macrophage accumulation associated with increased expression of chemokines, Ccl5, Cxcl10, and Cxcl15. Conversely, treatment of mice with a pharmacological PPARγ activator attenuated Cxcl10 and Cxcl15 expression and macrophage accumulation in response to CS. In vitro, CS increased lung epithelial cell chemokine expression in a PPARγ activation-dependent fashion. The ability of PPARγ to regulate CS-induced chemokine expression in vitro was not specifically associated with peroxisome proliferator response element (PPRE)-mediated transactivation activity but was correlated with PPARγ-mediated transrepression of NF-κB activity. Pharmacological or genetic activation of PPARγ activity abrogated CS-dependent induction of NF-κB activity. Regulation of NF-κB activity involved direct PPARγ-NF-κB interaction and PPARγ-mediated effects on IKK activation, IκBα degradation, and nuclear translocation of p65. Our data indicate that PPARG represents a disease-relevant pathophysiological and pharmacological target in COPD. Its activation state likely contributes to NF-κB-dependent, CS-induced chemokine-mediated regulation of inflammatory cell accumulation.
Collapse
Affiliation(s)
- Siva Kumar Solleti
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York
| | - Dawn M Simon
- Emory-Children's Center Pulmonary, Apnea, Cystic Fibrosis and Sleep Clinic, Atlanta, Georgia
| | - Sorachai Srisuma
- Faculty of Medicine, Department of Physiology, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Meltem C Arikan
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Soumyaroop Bhattacharya
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York;
| | - Tirumalai Rangasamy
- Division of Pulmonary & Critical Care Medicine, University of Rochester Medical Center, Rochester, New York
| | - Kaiser M Bijli
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York; Atlanta VA and Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University, Atlanta, Georgia
| | - Arshad Rahman
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York
| | - Joseph T Crossno
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Health Sciences Center, Denver, Colorado
| | - Steven D Shapiro
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Thomas J Mariani
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York;
| |
Collapse
|
47
|
Artlett CM, Thacker JD. Molecular activation of the NLRP3 Inflammasome in fibrosis: common threads linking divergent fibrogenic diseases. Antioxid Redox Signal 2015; 22:1162-75. [PMID: 25329971 DOI: 10.1089/ars.2014.6148] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Over the past 10 years, there has been a plethora of investigations centering on the NLRP3 inflammasome and its role in fibrosis and other disease pathologies. To date, the signaling pathways from the inflammasome to myofibroblast differentiation and chronic collagen synthesis have not been fully elucidated, and many questions are left to be answered. RECENT ADVANCES Recent studies have demonstrated the significant and critical role of reactive oxygen species (ROS) and calcium signaling in the assembly of the inflammasome, and this may result in autocrine signaling maintaining the myofibroblast phenotype, leading to fibrotic disease. CRITICAL ISSUES Traditionally, myofibroblasts under tight regulation aid in wound healing and then, once the wound has closed, undergo apoptosis and the collagen in the wound remodels. During fibrosis, however, the myofibroblast maintains an activated state via a chronically activated inflammasome, leading to the continual synthesis of collagens and other extracellular matrix proteins that result in damage to the tissue or organ. The mechanism that is driving this abnormality has not been fully elucidated. FUTURE DIRECTIONS However, studies have been conducted to suggest that modulating the calcium or the ROS axis may be of therapeutic value in regulating inflammasome activation. A number of novel drugs are currently being developed that may prove beneficial to patients suffering from fibrotic diseases.
Collapse
Affiliation(s)
- Carol M Artlett
- 1 Department of Microbiology and Immunology, Drexel University College of Medicine , Philadelphia, Pennsylvania
| | | |
Collapse
|
48
|
Baron L, Gombault A, Fanny M, Villeret B, Savigny F, Guillou N, Panek C, Le Bert M, Lagente V, Rassendren F, Riteau N, Couillin I. The NLRP3 inflammasome is activated by nanoparticles through ATP, ADP and adenosine. Cell Death Dis 2015; 6:e1629. [PMID: 25654762 PMCID: PMC4669808 DOI: 10.1038/cddis.2014.576] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 11/28/2014] [Accepted: 12/03/2014] [Indexed: 12/11/2022]
Abstract
The NLR pyrin domain containing 3 (NLRP3) inflammasome is a major component of the innate immune system, but its mechanism of activation by a wide range of molecules remains largely unknown. Widely used nano-sized inorganic metal oxides such as silica dioxide (nano-SiO2) and titanium dioxide (nano-TiO2) activate the NLRP3 inflammasome in macrophages similarly to silica or asbestos micro-sized particles. By investigating towards the molecular mechanisms of inflammasome activation in response to nanoparticles, we show here that active adenosine triphosphate (ATP) release and subsequent ATP, adenosine diphosphate (ADP) and adenosine receptor signalling are required for inflammasome activation. Nano-SiO2 or nano-TiO2 caused a significant increase in P2Y1, P2Y2, A2A and/or A2B receptor expression, whereas the P2X7 receptor was downregulated. Interestingly, IL-1β secretion in response to nanoparticles is increased by enhanced ATP and ADP hydrolysis, whereas it is decreased by adenosine degradation or selective A2A or A2B receptor inhibition. Downstream of these receptors, our results show that nanoparticles activate the NLRP3 inflammasome via activation of PLC-InsP3 and/or inhibition of adenylate cyclase (ADCY)-cAMP pathways. Finally, a high dose of adenosine triggers inflammasome activation and IL-1β secretion through adenosine cellular uptake by nucleotide transporters and by its subsequent transformation in ATP by adenosine kinase. In summary, we show for the first time that extracellular adenosine activates the NLRP3 inflammasome by two ways: by interacting with adenosine receptors at nanomolar/micromolar concentrations and through cellular uptake by equilibrative nucleoside transporters at millimolar concentrations. These findings provide new molecular insights on the mechanisms of NLRP3 inflammasome activation and new therapeutic strategies to control inflammation.
Collapse
Affiliation(s)
- L Baron
- INEM, CNRS, UMR7355, University of Orleans, France
| | - A Gombault
- INEM, CNRS, UMR7355, University of Orleans, France
| | - M Fanny
- INEM, CNRS, UMR7355, University of Orleans, France
| | - B Villeret
- INEM, CNRS, UMR7355, University of Orleans, France
| | - F Savigny
- INEM, CNRS, UMR7355, University of Orleans, France
| | - N Guillou
- INEM, CNRS, UMR7355, University of Orleans, France
| | - C Panek
- INEM, CNRS, UMR7355, University of Orleans, France
| | - M Le Bert
- INEM, CNRS, UMR7355, University of Orleans, France
| | - V Lagente
- INSERM U991, University of Rennes, France
| | - F Rassendren
- IGF, CNRS, UMR 5203 and INSERM U661, University of Montpellier, France
| | - N Riteau
- INEM, CNRS, UMR7355, University of Orleans, France
| | - I Couillin
- INEM, CNRS, UMR7355, University of Orleans, France
| |
Collapse
|
49
|
Uncoupling between inflammatory and fibrotic responses to silica: evidence from MyD88 knockout mice. PLoS One 2014; 9:e99383. [PMID: 25050810 PMCID: PMC4106757 DOI: 10.1371/journal.pone.0099383] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 05/14/2014] [Indexed: 12/21/2022] Open
Abstract
The exact implication of innate immunity in granuloma formation and irreversible lung fibrosis remains to be determined. In this study, we examined the lung inflammatory and fibrotic responses to silica in MyD88-knockout (KO) mice. In comparison to wild-type (WT) mice, we found that MyD88-KO animals developed attenuated lung inflammation, neutrophil accumulation and IL-1β release in response to silica. Granuloma formation was also less pronounced in MyD88-KO mice after silica. This limited inflammatory response was not accompanied by a concomitant attenuation of lung collagen accumulation after silica. Histological analyses revealed that while pulmonary fibrosis was localized in granulomas in WT animals, it was diffusely distributed throughout the parenchyma in MyD88-KO mice. Robust collagen accumulation was also observed in mice KO for several other components of innate immunity (IL-1R, IL-1, ASC, NALP3, IL-18R, IL-33R, TRIF, and TLR2-3-4,). We additionally show that pulmonary fibrosis in MyD88-KO mice was associated with the accumulation of pro-fibrotic regulatory T lymphocytes (T regs) and pro-fibrotic cytokine expression (TGF-β, IL-10 and PDGF-B), not with T helper (Th) 17 cell influx. Our findings indicate that the activation of MyD88-related innate immunity is central in the establishment of particle-induced lung inflammatory and granuloma responses. The development of lung fibrosis appears uncoupled from inflammation and may be orchestrated by a T reg-associated pathway.
Collapse
|
50
|
Targeting IL-1β and IL-17A driven inflammation during influenza-induced exacerbations of chronic lung inflammation. PLoS One 2014; 9:e98440. [PMID: 24918427 PMCID: PMC4053370 DOI: 10.1371/journal.pone.0098440] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 05/04/2014] [Indexed: 12/05/2022] Open
Abstract
For patients with chronic lung diseases, such as chronic obstructive pulmonary disease (COPD), exacerbations are life-threatening events causing acute respiratory distress that can even lead to hospitalization and death. Although a great deal of effort has been put into research of exacerbations and potential treatment options, the exact underlying mechanisms are yet to be deciphered and no therapy that effectively targets the excessive inflammation is available. In this study, we report that interleukin-1β (IL-1β) and interleukin-17A (IL-17A) are key mediators of neutrophilic inflammation in influenza-induced exacerbations of chronic lung inflammation. Using a mouse model of disease, our data shows a role for IL-1β in mediating lung dysfunction, and in driving neutrophilic inflammation during the whole phase of viral infection. We further report a role for IL-17A as a mediator of IL-1β induced neutrophilia at early time points during influenza-induced exacerbations. Blocking of IL-17A or IL-1 resulted in a significant abrogation of neutrophil recruitment to the airways in the initial phase of infection or at the peak of viral replication, respectively. Therefore, IL-17A and IL-1β are potential targets for therapeutic treatment of viral exacerbations of chronic lung inflammation
Collapse
|