1
|
dos S. P. Andrade AC, Lacasse E, Dubuc I, Gudimard L, Gravel A, Puhm F, Campolina-Silva G, Queiroz-Junior C, Allaeys I, Prunier J, Azeggouar Wallen O, Dumais É, Belleannée C, Droit A, Flamand N, Boilard É, Flamand L. Deficiency in platelet 12-lipoxygenase exacerbates inflammation and disease severity during SARS-CoV-2 infection. Proc Natl Acad Sci U S A 2025; 122:e2420441122. [PMID: 40100623 PMCID: PMC11962506 DOI: 10.1073/pnas.2420441122] [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: 10/15/2024] [Accepted: 01/27/2025] [Indexed: 03/20/2025] Open
Abstract
Platelets, known for maintaining blood balance, also participate in antimicrobial defense. Upon severeacute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, platelets become hyperactivated, releasing molecules such as cytokines, granule contents, and bioactive lipids. The key effector biolipids produced by platelets include 12-hydroxyeicosatetraenoic acid (12-HETE) and 12-hydroxyeicosatrienoic acid (12-HETrE), produced by 12-lipoxygenase (12-LOX), and prostaglandins and thromboxane, produced by cyclooxygenase-1. While prostaglandin E2 and thromboxane B2 were previously associated with lung inflammation in severe COVID-19, the role of platelet 12-LOX in SARS-CoV-2 infection remains unclear. Using mice deficient for platelets' 12-LOX, we report that SARS-CoV-2 infection resulted in higher lung inflammation characterized by histopathological tissue analysis, increased leukocyte infiltrates, and cytokine production relative to wild-type mice. In addition, distinct platelet and lung transcriptomic changes, including alterations in NOD-like receptor (NLR) family pyrin domain-containing 1 (NLRP1) inflammasome-related gene expression, were observed. Mass spectrometry lipidomic analysis in 12-LOX-deficient-infected mice revealed significant changes in bioactive lipid content, including reduced levels of 12-HETrE that inversely correlated with disease severity. Finally, platelet 12-LOX deficiency was associated with increased morbidity and lower survival rates relative to wild type (WT) mice. Overall, this study highlights the complex interplay between 12-LOX-related lipid metabolism and inflammatory responses during SARS-CoV-2 infection. The findings provide valuable insights into potential therapeutic targets aimed at mitigating severe outcomes, emphasizing the pivotal role of platelet enzymes in the host response to viral infections.
Collapse
Affiliation(s)
- Ana Claudia dos S. P. Andrade
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Emile Lacasse
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Isabelle Dubuc
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Leslie Gudimard
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Annie Gravel
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Florian Puhm
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Gabriel Campolina-Silva
- Division of Reproduction, mother and youth health, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Celso Queiroz-Junior
- Morphology Department, Universidade Federal de Minas Gerais, Belo Horizonte31270-901, Brazil
| | - Isabelle Allaeys
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Julien Prunier
- Division of Endocrinology and Nephrology, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Oumaima Azeggouar Wallen
- Centre de recherche de l’Institut Universitaire de cardiologie et pneumologie de Québec, Division of pneumology, Faculty of medicine, Université Laval, Québec City, QCG1V 4G5, Canada
| | - Élizabeth Dumais
- Centre de recherche de l’Institut Universitaire de cardiologie et pneumologie de Québec, Division of pneumology, Faculty of medicine, Université Laval, Québec City, QCG1V 4G5, Canada
| | - Clémence Belleannée
- Division of Reproduction, mother and youth health, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Arnaud Droit
- Division of Endocrinology and Nephrology, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Nicolas Flamand
- Centre de recherche de l’Institut Universitaire de cardiologie et pneumologie de Québec, Division of pneumology, Faculty of medicine, Université Laval, Québec City, QCG1V 4G5, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec City, QCG1V 4G5, Canada
| | - Éric Boilard
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
- Centre de Recherche ARThrite–Arthrite, Recherche, Traitements, Université Laval, Québec, QCG1V 4G2, Canada
- Department of microbiology, infectious disease and immunology, Faculty of Medicine, Université Laval, Québec City, QCG1V 0A6, Canada
| | - Louis Flamand
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
- Centre de Recherche ARThrite–Arthrite, Recherche, Traitements, Université Laval, Québec, QCG1V 4G2, Canada
- Department of microbiology, infectious disease and immunology, Faculty of Medicine, Université Laval, Québec City, QCG1V 0A6, Canada
| |
Collapse
|
2
|
Brown AC, Carroll OR, Mayall JR, Zounemat-Kermani N, Vinzenz SLE, Gomez HM, Mills EF, Kim RY, Donovan C, Baines KJ, Williams EJ, Berthon BS, Wynne K, Scott HA, Pinkerton JW, Guo Y, Hansbro PM, Foster PS, Wark PAB, Dahlen SE, Adcock IM, Wood LG, Horvat JC. Female sex hormones and the oral contraceptive pill modulate asthma severity through GLUT-1. Mucosal Immunol 2025:S1933-0219(25)00024-8. [PMID: 40021011 DOI: 10.1016/j.mucimm.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Females are disproportionately affected by asthma. An increased understanding of how female sex hormones influence key pathophysiological processes that underpin asthma may identify new, more effective asthma therapies, particularly for females with severe, poorly controlled asthma. We assessed the effects of oral ethinylestradiol/levonorgestrel (representing OCP use) and depot-medroxyprogesterone acetate (DMPA) and estradiol injections on key features of experimental asthma, and determined their effects on glucose transporter-1 (GLUT-1). The effects of OCP use on clinical asthma outcomes, and the relationships between estrogen receptors and type 2 (T2), non-T2, and GLUT-1 responses, in clinical asthma were also determined. OCP and DMPA reduce T2 responses, disease features, and lung expression of GLUT-1, whereas estradiol increases lung expression of GLUT-1, and results in severe, corticosteroid-insensitive, neutrophil-enriched disease, in experimental asthma. OCP use is associated with reduced T2 cytokine and GLUT-1 responses in clinical asthma. GLUT-1 expression is increased in sputum of severe asthmatics, and positively correlates with estrogen receptor expression and both T2 and non-T2 inflammatory responses. Significantly, OCP or GLUT-1 inhibition protects against obesity-associated or estradiol-induced, severe, experimental asthma, respectively. Together, these data show how female sex hormones and the OCP likely modulate asthma severity by modifying GLUT-1 responses in the airways.
Collapse
Affiliation(s)
- Alexandra C Brown
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Olivia R Carroll
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Jemma R Mayall
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | | | - Samantha L E Vinzenz
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Henry M Gomez
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Ed F Mills
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Richard Y Kim
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia; Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
| | - Chantal Donovan
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia; Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
| | - Katherine J Baines
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Evan J Williams
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Bronwyn S Berthon
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Katie Wynne
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Hayley A Scott
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - James W Pinkerton
- Respiratory Pharmacology & Toxicology Group, National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | - Yike Guo
- Hong Kong University of Science and Technology, Hong Kong
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Paul S Foster
- Woolcock Institute of Medical Research and Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Peter A B Wark
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia; School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Sven-Erik Dahlen
- Clinical Lung and Allergy Research Unit, Department of Medicine Huddinge, Karolinska Institutet, and, Department of Respiratory Medicine and Allergy, Karolinska University Hospital Huddinge, and Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ian M Adcock
- The Airway Disease Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | - Lisa G Wood
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Jay C Horvat
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.
| |
Collapse
|
3
|
Beltrami VA, Martins FRB, Martins DG, Queiroz-Junior CM, Félix FB, Resende LC, Santos FRDS, Lacerda LDSB, Costa VRDM, da Silva WN, Guimaraes PPG, Guimaraes G, Soriani FM, Teixeira MM, Costa VV, Pinho V. Selective phosphodiesterase 4 inhibitor roflumilast reduces inflammation and lung injury in models of betacoronavirus infection in mice. Inflamm Res 2025; 74:24. [PMID: 39862252 DOI: 10.1007/s00011-024-01985-3] [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: 07/31/2024] [Revised: 10/03/2024] [Accepted: 10/16/2024] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE We aimed to understand the potential therapeutic and anti-inflammatory effects of the phosphodiesterase-4 (PDE4) inhibitor roflumilast in models of pulmonary infection caused by betacoronaviruses. METHODS Mice were infected intranasally with murine hepatitis virus (MHV-3) or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Roflumilast was given to MHV-3-infected mice therapeutically at doses of 1 mg/kg or 10 mg/kg, or prophylactically at 10 mg/kg. In SARS-CoV-2-infected mice, roflumilast was given therapeutically at a dose of 10 mg/kg. Lung histopathology, chemokines (CXCL-1 and CCL2), cytokines (IL-1β, IL-6, TNF, IFN-γ, IL-10 and TGFβ), neutrophil immunohistochemical staining (Ly6G+ cells), macrophage immunofluorescence staining (F4/80+ cells), viral titration plaque assay, real-time PCR virus detection, and blood cell counts were examined. RESULTS Therapeutic treatment with roflumilast at 10 mg/kg reduced lung injury in SARS-CoV-2 or MHV-3-infected mice without compromising viral clearance. In MHV-3-infected mice, reduced lung injury was associated with decreased chemokines levels, prevention of neutrophil aggregates and reduced macrophage accumulation in the lung tissue. However, the prophylactic treatment strategy with roflumilast increased lung injury in MHV-3-infected mice. CONCLUSION Our findings indicate that therapeutic treatment with roflumilast reduced lung injury in MHV-3 and SARS-CoV-2 lung infections. Given the protection induced by roflumilast in inflammation, PDE4 targeting could be a promising therapeutic avenue worth exploring following severe viral infections of the lung.
Collapse
Affiliation(s)
- Vinícius Amorim Beltrami
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Flávia Rayssa Braga Martins
- Departamento Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Débora Gonzaga Martins
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Celso Martins Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Franciel Batista Félix
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Letícia Cassiano Resende
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Felipe Rocha da Silva Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Larisse de Souza Barbosa Lacerda
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Victor Rodrigues de Melo Costa
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Walison Nunes da Silva
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Pedro Pires Goulart Guimaraes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Goulart Guimaraes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Frederico Marianetti Soriani
- Departamento Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Vivian Vasconcelos Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Vanessa Pinho
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| |
Collapse
|
4
|
Vanders RL, Gomez HM, Daly K, Wark PA, Horvat JC, Hansbro PM. Immune checkpoints are suppressed during pregnancy following influenza A virus infection. Am J Physiol Lung Cell Mol Physiol 2024; 327:L890-L904. [PMID: 39254092 DOI: 10.1152/ajplung.00391.2023] [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: 12/14/2023] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Influenza A virus (IAV) infection is a major health risk during pregnancy. Although vaccination and antiviral agents are widely used and reduce IAV-induced symptoms, they are not sufficient to control IAV infections in pregnancy, especially during pandemics. Respiratory viruses like IAV exploit immune alterations that occur during pregnancy, including the upregulation of immune checkpoint proteins (ICPs) like programmed death ligand-1 (PDL1), programmed cell death receptor 1 (PD1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA4). We hypothesize that blocking expression of PDL1 on innate immune cells will improve maternal immunity following IAV infection. We used murine models of IAV infection during pregnancy with and without treatment with the immune checkpoint inhibitor (ICI), a-PDL1. Pregnant and nonpregnant mice were infected with mouse-adapted IAV (A/PR/8) and assessed at 3 days post infection (3 dpi). Lung cells were analyzed using flow cytometry. Lung mRNA expression of inflammatory and antiviral markers and histology was measured. Protein concentrations of inflammatory and antiviral markers, as well as viral titers were measured from lung bronchiolar lavage fluid (BALF). Lung function was also assessed. Following IAV infection, immune cells from pregnant mice had significant increases in the ICPs, PDL1, PD1, and CTLA4. a-PDL1 treatment effectively suppressed these ICPs and increased the activation marker, CD86. a-PDL1 treatment also reduced lung inflammatory cell infiltration and viral titers, increased antiviral responses, and improved lung function. Overall, IAV infection in pregnancy activates key inhibitory ICPs, leading to worsened disease outcomes. a-PDL1 treatment during IAV infection in pregnancy is an effective method to reduce ICP expression and improve overall immune cell responses.NEW & NOTEWORTHY Influenza infection worsens disease outcomes during pregnancy; however, treatment with anti-PDL1 can restore immune function during pregnancy.
Collapse
Affiliation(s)
- Rebecca L Vanders
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Henry M Gomez
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Katie Daly
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Peter A Wark
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Jay C Horvat
- Immune Health, Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
5
|
Donovan C, Thorpe AE, Yarak R, Coward-Smith M, Pillar AL, Gomez HM, Feng M, Bai X, Wang M, Xenaki D, Horvat JC, Chen H, Oliver BGG, Kim RY. Maternal thirdhand exposure to e-cigarette vapor alters lung and bone marrow immune cell responses in offspring in the absence or presence of influenza infection. Am J Physiol Lung Cell Mol Physiol 2024; 327:L796-L806. [PMID: 39316673 DOI: 10.1152/ajplung.00078.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/26/2024] Open
Abstract
There is increasing evidence that thirdhand exposure to e-cigarette vapor (e-vapor) can have detrimental effects on the lungs. However, whether maternal exposure during pregnancy results in harmful changes to the offspring is unknown. Using two different e-cigarette settings (low vs. high power), BALB/c mice were subjected to thirdhand e-vapor (e-vapor deposited onto towels, towels changed daily) in the absence or presence of nicotine, before, during, and after pregnancy. Male adult offspring were then infected with mouse-adapted influenza A virus (A/PR/8/34 H1N1; Flu) and lung and bone marrow immune cell responses were assessed 7 days postinfection. Maternal thirdhand exposure to low-power (MLP) or high-power (MHP) e-vapor with nicotine (MLP + NIC and MHP + NIC, respectively) increased the percentage of lung immune cells and neutrophils in the bone marrow. Interestingly, Flu-infected offspring from MLP + NIC and MHP + NIC groups had lower percentages of lung alveolar macrophages and more pronounced increases in neutrophils in the bone marrow, when compared with offspring from MSham Flu controls. Flu infection also decreased the percentage of lung CD4+ T cells and increased the percentage of lung CD8+ T cells, irrespective of maternal exposure (MLP -/+ NIC and MHP -/+ NIC). Significantly, both MLP + NIC and MHP + NIC resulted in blunted activation of lung CD4+ T cells, but only MLP + NIC caused blunted activation of lung CD8+ T cells. Together, we show for the first time that maternal thirdhand exposure to e-vapor results in significant, long-lived effects on lung and bone marrow immune cell responses in offspring at baseline and response to Flu infection.NEW & NOTEWORTHY Maternal exposure to environmental residues of e-cigarette use has significant effects on immune cell responses in the lungs and bone marrow of offspring at both baseline and in response to influenza A virus (Flu) infection.
Collapse
Affiliation(s)
- Chantal Donovan
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
- Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Andrew E Thorpe
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
- Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
| | - Rochelle Yarak
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Madison Coward-Smith
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Amber L Pillar
- Immune Health Program, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Henry M Gomez
- Immune Health Program, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Min Feng
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
- Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
| | - Xu Bai
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
- Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
| | - Meng Wang
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
- Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
| | - Dia Xenaki
- Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
| | - Jay C Horvat
- Immune Health Program, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Hui Chen
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Brian G G Oliver
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
- Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
| | - Richard Y Kim
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
- Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
6
|
Souza-Costa LP, Santos FRS, Pimenta JC, Queiroz-Junior CM, Tana FL, Teixeira DC, Couto MGG, Oliveira NFM, Pereira RD, Beltrami VA, Costa PAC, Lacerda LSB, Andrade-Chaves JT, Guimarães PPG, Aguiar RS, Teixeira MM, Costa VV, Franco LH. E3 Ubiquitin Ligase Smurf1 Regulates the Inflammatory Response in Macrophages and Attenuates Hepatic Damage during Betacoronavirus Infection. Pathogens 2024; 13:871. [PMID: 39452742 PMCID: PMC11510589 DOI: 10.3390/pathogens13100871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024] Open
Abstract
The E3 ubiquitin ligase Smurf1 catalyzes the ubiquitination and proteasomal degradation of several protein substrates related to inflammatory responses and antiviral signaling. This study investigated the role of Smurf1 in modulating inflammation induced by Betacoronavirus infection. Bone marrow-derived macrophages (BMDMs) from C57BL/6 (wild-type) or Smurf1-deficient (Smurf1-/-) mice were infected with MHV-A59 to evaluate the inflammatory response in vitro. Smurf1 was found to be required to downregulate the macrophage production of pro-inflammatory mediators, including TNF, and CXCL1; to control viral release from infected cells; and to increase cell viability. To assess the impact of Smurf 1 in vivo, we evaluated the infection of mice with MHV-A59 through the intranasal route. Smurf1-/- mice infected with a lethal inoculum of MHV-A59 succumbed earlier to infection. Intranasal inoculation with a 10-fold lower dose of MHV-A59 resulted in hematological parameter alterations in Smurf1-/- mice suggestive of exacerbated systemic inflammation. In the lung parenchyma, Smurf1 expression was essential to promote viral clearance, downregulating IFN-β mRNA and controlling the inflammatory profile of macrophages and neutrophils. Conversely, Smurf1 did not affect IFN-β mRNA regulation in the liver, but it was required to increase TNF and iNOS expression in neutrophils and decrease TNF expression in macrophages. In addition, Smurf1-/- mice exhibited augmented liver injuries, accompanied by high serum levels of alanine aminotransferase (ALT). These findings suggest that Smurf1 plays a critical role in regulating the inflammatory response in macrophages and attenuating systemic inflammation during Betacoronavirus infection.
Collapse
Affiliation(s)
- Luiz P. Souza-Costa
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (L.P.S.-C.)
| | - Felipe R. S. Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (L.P.S.-C.)
| | - Jordane C. Pimenta
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Celso M. Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Fernanda L. Tana
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (L.P.S.-C.)
| | - Danielle C. Teixeira
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Manoela G. G. Couto
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Natalia F. M. Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (L.P.S.-C.)
| | - Rafaela D. Pereira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (L.P.S.-C.)
| | - Vinicius A. Beltrami
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Pedro A. C. Costa
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Larisse S. B. Lacerda
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Josiane T. Andrade-Chaves
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (L.P.S.-C.)
| | - Pedro P. G. Guimarães
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Renato S. Aguiar
- Departamento de Genética e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Mauro M. Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (L.P.S.-C.)
| | - Vivian V. Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Luis H. Franco
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (L.P.S.-C.)
| |
Collapse
|
7
|
Hedley KE, Gomez HM, Kecelioglu E, Carroll OR, Jobling P, Horvat JC, Tadros MA. Neonatal Chlamydia muridarum respiratory infection causes neuroinflammation within the brainstem during the early postnatal period. J Neuroinflammation 2024; 21:158. [PMID: 38879567 PMCID: PMC11179230 DOI: 10.1186/s12974-024-03150-3] [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: 01/29/2024] [Accepted: 06/10/2024] [Indexed: 06/19/2024] Open
Abstract
Respiratory infections are one of the most common causes of illness and morbidity in neonates worldwide. In the acute phase infections are known to cause wide-spread peripheral inflammation. However, the inflammatory consequences to the critical neural control centres for respiration have not been explored. Utilising a well characterised model of neonatal respiratory infection, we investigated acute responses within the medulla oblongata which contains key respiratory regions. Neonatal mice were intranasally inoculated within 24 h of birth, with either Chlamydia muridarum or sham-infected, and tissue collected on postnatal day 15, the peak of peripheral inflammation. A key finding of this study is that, while the periphery appeared to show no sex-specific effects of a neonatal respiratory infection, sex had a significant impact on the inflammatory response of the medulla oblongata. There was a distinct sex-specific response in the medulla coincident with peak of peripheral inflammation, with females demonstrating an upregulation of anti-inflammatory cytokines and males showing very few changes. Microglia also demonstrated sex-specificity with the morphology of females and males differing based upon the nuclei. Astrocytes showed limited changes during the acute response to neonatal infection. These data highlight the strong sex-specific impact of a respiratory infection can have on the medulla in the acute inflammatory phase.
Collapse
Affiliation(s)
- Kateleen E Hedley
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Henry M Gomez
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Eda Kecelioglu
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Olivia R Carroll
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Phillip Jobling
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jay C Horvat
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Melissa A Tadros
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia.
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| |
Collapse
|
8
|
Oliveira VLS, Queiroz-Junior CM, Hoorelbeke D, Santos FRDS, Chaves IDM, Teixeira MM, Russo RDC, Proost P, Costa VV, Struyf S, Amaral FA. The glycosaminoglycan-binding chemokine fragment CXCL9(74-103) reduces inflammation and tissue damage in mouse models of coronavirus infection. Front Immunol 2024; 15:1378591. [PMID: 38686377 PMCID: PMC11056509 DOI: 10.3389/fimmu.2024.1378591] [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: 01/29/2024] [Accepted: 03/29/2024] [Indexed: 05/02/2024] Open
Abstract
Introduction Pulmonary diseases represent a significant burden to patients and the healthcare system and are one of the leading causes of mortality worldwide. Particularly, the COVID-19 pandemic has had a profound global impact, affecting public health, economies, and daily life. While the peak of the crisis has subsided, the global number of reported COVID-19 cases remains significantly high, according to medical agencies around the world. Furthermore, despite the success of vaccines in reducing the number of deaths caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), there remains a gap in the treatment of the disease, especially in addressing uncontrolled inflammation. The massive recruitment of leukocytes to lung tissue and alveoli is a hallmark factor in COVID-19, being essential for effectively responding to the pulmonary insult but also linked to inflammation and lung damage. In this context, mice models are a crucial tool, offering valuable insights into both the pathogenesis of the disease and potential therapeutic approaches. Methods Here, we investigated the anti-inflammatory effect of the glycosaminoglycan (GAG)-binding chemokine fragment CXCL9(74-103), a molecule that potentially decreases neutrophil transmigration by competing with chemokines for GAG-binding sites, in two models of pneumonia caused by coronavirus infection. Results In a murine model of betacoronavirus MHV-3 infection, the treatment with CXCL9(74-103) decreased the accumulation of total leukocytes, mainly neutrophils, to the alveolar space and improved several parameters of lung dysfunction 3 days after infection. Additionally, this treatment also reduced the lung damage. In the SARS-CoV-2 model in K18-hACE2-mice, CXCL9(74-103) significantly improved the clinical manifestations of the disease, reducing pulmonary damage and decreasing viral titers in the lungs. Discussion These findings indicate that CXCL9(74-103) resulted in highly favorable outcomes in controlling pneumonia caused by coronavirus, as it effectively diminishes the clinical consequences of the infections and reduces both local and systemic inflammation.
Collapse
Affiliation(s)
- Vivian Louise Soares Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Departament of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Celso Martins Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Delphine Hoorelbeke
- Departament of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Felipe Rocha da Silva Santos
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ian de Meira Chaves
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo de Castro Russo
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Paul Proost
- Departament of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Vivian Vasconcelos Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sofie Struyf
- Departament of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Flávio Almeida Amaral
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
9
|
Zhou B, Wang L, Yang S, Liang Y, Zhang Y, Liu X, Pan X, Li J. Pyrogallol protects against influenza A virus-triggered lethal lung injury by activating the Nrf2-PPAR-γ-HO-1 signaling axis. MedComm (Beijing) 2024; 5:e531. [PMID: 38617435 PMCID: PMC11014464 DOI: 10.1002/mco2.531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 02/04/2024] [Accepted: 02/26/2024] [Indexed: 04/16/2024] Open
Abstract
Pyrogallol, a natural polyphenol compound (1,2,3-trihydroxybenzene), has shown efficacy in the therapeutic treatment of disorders associated with inflammation. Nevertheless, the mechanisms underlying the protective properties of pyrogallol against influenza A virus infection are not yet established. We established in this study that pyrogallol effectively alleviated H1N1 influenza A virus-induced lung injury and reduced mortality. Treatment with pyrogallol was found to promote the expression and nuclear translocation of nuclear factor erythroid-2-related factor 2 (Nrf2) and peroxisome proliferator-activated receptor gamma (PPAR-γ). Notably, the activation of Nrf2 by pyrogallol was involved in elevating the expression of PPAR-γ, both of which act synergistically to enhance heme oxygenase-1 (HO-1) synthesis. Blocking HO-1 by zinc protoporphyrin (ZnPP) reduced the suppressive impact of pyrogallol on H1N1 virus-mediated aberrant retinoic acid-inducible gene-I-nuclear factor kappa B (RIG-I-NF-κB) signaling, which thus abolished the dampening effects of pyrogallol on excessive proinflammatory mediators and cell death (including apoptosis, necrosis, and ferroptosis). Furthermore, the HO-1-independent inactivation of janus kinase 1/signal transducers and activators of transcription (JAK1/STATs) and the HO-1-dependent RIG-I-augmented STAT1/2 activation were both abrogated by pyrogallol, resulting in suppression of the enhanced transcriptional activity of interferon-stimulated gene factor 3 (ISGF3) complexes, thus prominently inhibiting the amplification of the H1N1 virus-induced proinflammatory reaction and apoptosis in interferon-beta (IFN-β)-sensitized cells. The study provides evidence that pyrogallol alleviates excessive proinflammatory responses and abnormal cell death via HO-1 induction, suggesting it could be a potential agent for treating influenza.
Collapse
Affiliation(s)
- Beixian Zhou
- The People's Hospital of GaozhouGaozhouChina
- Cancer Center, Integrated Hospital of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouChina
| | | | - Sushan Yang
- The People's Hospital of GaozhouGaozhouChina
| | | | | | - Xuanyu Liu
- The People's Hospital of GaozhouGaozhouChina
| | | | - Jing Li
- State Key Laboratory of Respiratory DiseaseNational Clinical Research Center of Respiratory DiseaseGuangzhou Institute of Respiratory HealthInstitute of Chinese Integrative MedicineGuangdong‐Hongkong‐Macao Joint Laboratory of Infectious Respiratory Diseasethe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
10
|
Silva IS, Almeida AD, Lima Filho ACM, Fernandes-Braga W, Barra A, Oliveira HMC, Cassali GD, Capettini LSA, Menezes GB, Alvarez-Leite JI, Leite MF, Klein A. Platelet-activating factor and protease-activated receptor 2 cooperate to promote neutrophil recruitment and lung inflammation through nuclear factor-kappa B transactivation. Sci Rep 2023; 13:21637. [PMID: 38062077 PMCID: PMC10703791 DOI: 10.1038/s41598-023-48365-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Although it is well established that platelet-activated receptor (PAF) and protease-activated receptor 2 (PAR2) play a pivotal role in the pathophysiology of lung and airway inflammatory diseases, a role for a PAR2-PAFR cooperation in lung inflammation has not been investigated. Here, we investigated the role of PAR2 in PAF-induced lung inflammation and neutrophil recruitment in lungs of BALB/c mice. Mice were pretreated with the PAR2 antagonist ENMD1068, PAF receptor (PAFR) antagonist WEB2086, or aprotinin prior to intranasal instillation of carbamyl-PAF (C-PAF) or the PAR2 agonist peptide SLIGRL-NH2 (PAR2-AP). Leukocyte infiltration in bronchoalveolar lavage fluid (BALF), C-X-C motif ligand 1 (CXCL)1 and CXCL2 chemokines, myeloperoxidase (MPO), and N-acetyl-glycosaminidase (NAG) levels in BALF, or lung inflammation were evaluated. Intracellular calcium signaling, PAFR/PAR2 physical interaction, and the expression of PAR2 and nuclear factor-kappa B (NF-КB, p65) transcription factor were investigated in RAW 264.7 cells stimulated with C-PAF in the presence or absence of ENMD1068. C-PAF- or PAR2-AP-induced neutrophil recruitment into lungs was inhibited in mice pretreated with ENMD1068 and aprotinin or WEB2086, respectively. PAR2 blockade impaired C-PAF-induced neutrophil rolling and adhesion, lung inflammation, and production of MPO, NAG, CXCL1, and CXCL2 production in lungs of mice. PAFR activation reduced PAR2 expression and physical interaction of PAR2 and PAFR; co-activation is required for PAFR/PAR2 physical interaction. PAR2 blockade impaired C-PAF-induced calcium signal and NF-κB p65 translocation in RAW 264.7 murine macrophages. This study provides the first evidence for a cooperation between PAFR and PAR2 mediating neutrophil recruitment, lung inflammation, and macrophage activation.
Collapse
Affiliation(s)
- Irismara Sousa Silva
- Laboratory of Inflammation and Proteases, Department of Pharmacology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Aline D Almeida
- Laboratory of Inflammation and Proteases, Department of Pharmacology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | | | - Weslley Fernandes-Braga
- Laboratory of Atherosclerosis and Nutritional Biochemistry (LABIN-UFMG), Department of Biochemistry and Immunology, ICB/UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Ayslan Barra
- Laboratory of Inflammation and Proteases, Department of Pharmacology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Luciano S A Capettini
- Laboratory of Vascular Biology, Department of Pharmacology, ICB/UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Gustavo B Menezes
- Department of Morphology, ICB/UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Jacqueline I Alvarez-Leite
- Laboratory of Atherosclerosis and Nutritional Biochemistry (LABIN-UFMG), Department of Biochemistry and Immunology, ICB/UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Maria F Leite
- Department of Physiology and Biophysics, ICB/UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - André Klein
- Laboratory of Inflammation and Proteases, Department of Pharmacology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
11
|
Mendes de Almeida V, Engel DF, Ricci MF, Cruz CS, Lopes ÍS, Alves DA, d’ Auriol M, Magalhães J, Machado EC, Rocha VM, Carvalho TG, Lacerda LSB, Pimenta JC, Aganetti M, Zuccoli GS, Smith BJ, Carregari VC, da Silva Rosa E, Galvão I, Dantas Cassali G, Garcia CC, Teixeira MM, André LC, Ribeiro FM, Martins FS, Saia RS, Costa VV, Martins-de-Souza D, Hansbro PM, Marques JT, Aguiar ERGR, Vieira AT. Gut microbiota from patients with COVID-19 cause alterations in mice that resemble post-COVID symptoms. Gut Microbes 2023; 15:2249146. [PMID: 37668317 PMCID: PMC10481883 DOI: 10.1080/19490976.2023.2249146] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/19/2023] [Accepted: 08/14/2023] [Indexed: 09/06/2023] Open
Abstract
Long-term sequelae of coronavirus disease (COVID)-19 are frequent and of major concern. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection affects the host gut microbiota, which is linked to disease severity in patients with COVID-19. Here, we report that the gut microbiota of post-COVID subjects had a remarkable predominance of Enterobacteriaceae strains with an antibiotic-resistant phenotype compared to healthy controls. Additionally, short-chain fatty acid (SCFA) levels were reduced in feces. Fecal transplantation from post-COVID subjects to germ-free mice led to lung inflammation and worse outcomes during pulmonary infection by multidrug-resistant Klebsiella pneumoniae. transplanted mice also exhibited poor cognitive performance. Overall, we show prolonged impacts of SARS-CoV-2 infection on the gut microbiota that persist after subjects have cleared the virus. Together, these data demonstrate that the gut microbiota can directly contribute to post-COVID sequelae, suggesting that it may be a potential therapeutic target.
Collapse
Affiliation(s)
- Viviani Mendes de Almeida
- Laboratory of Microbiota and Immunomodulation - Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Daiane F. Engel
- Department of Clinical Analysis, School of Pharmacy, Universidade Federal de Ouro Preto - UFOP, Ouro Preto, Brazil
| | - Mayra F. Ricci
- Laboratory of Microbiota and Immunomodulation - Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Clênio Silva Cruz
- Laboratory of Microbiota and Immunomodulation - Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Ícaro Santos Lopes
- Laboratory of Virus Bioinformatics - Department of Biological Science, Center of Biotechnology and Genetics, Universidade Estadual de Santa Cruz - UESC, Ilhéus, Brazil
| | - Daniele Almeida Alves
- Laboratory of RNA Interference and Antiviral Immunity - Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Mirna d’ Auriol
- Laboratory of Toxicology - Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - João Magalhães
- Laboratory of Microbiota and Immunomodulation - Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Elayne C. Machado
- Laboratory of Microbiota and Immunomodulation - Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Victor M. Rocha
- Laboratory of Microbiota and Immunomodulation - Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Toniana G. Carvalho
- Laboratory of Neurobiochemistry - Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Larisse S. B. Lacerda
- Center for Research and Development of Drugs - Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Jordane C. Pimenta
- Center for Research and Development of Drugs - Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Mariana Aganetti
- Laboratory of Microbiota and Immunomodulation - Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Giuliana S. Zuccoli
- Laboratory of Neuroproteomics - Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade do Estado de Campinas - UNICAMP, Campinas, Brazil
| | - Bradley J. Smith
- Laboratory of Neuroproteomics - Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade do Estado de Campinas - UNICAMP, Campinas, Brazil
| | - Victor C. Carregari
- Laboratory of Neuroproteomics - Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade do Estado de Campinas - UNICAMP, Campinas, Brazil
| | - Erika da Silva Rosa
- Laboratory of Microbiota and Immunomodulation - Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Izabela Galvão
- Laboratory of Microbiota and Immunomodulation - Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Geovanni Dantas Cassali
- Laboratory of Comparative Pathology - Department of Pathology, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Cristiana C. Garcia
- Laboratory of Respiratory Viruses and Measles, Instituto Oswaldo Cruz - Fiocruz, Rio de Janeiro, Brazil
| | - Mauro Martins Teixeira
- Center for Research and Development of Drugs - Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Leiliane C. André
- Laboratory of Toxicology - Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Fabiola Mara Ribeiro
- Laboratory of Neurobiochemistry - Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Flaviano S. Martins
- Laboratory of Biotherapeutic Agents - Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Rafael Simone Saia
- Laboratory of Intestinal Physiology - Department of Physiology, Ribeirão Preto Medical School, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Vivian Vasconcelos Costa
- Center for Research and Development of Drugs - Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics - Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade do Estado de Campinas - UNICAMP, Campinas, Brazil
- D’Or Institute for Research and Education, São Paulo, Brazil
- Experimental Medicine Research Cluster, Universidade do Estado de Campinas - UNICAMP, Campinas, Brazil
- National Institute of Biomarkers in Neuropsychiatry, National Council for Scientific and Technological Development, São Paulo, Brazil
| | - Philip M. Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia
| | - João Trindade Marques
- Laboratory of RNA Interference and Antiviral Immunity - Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
- CNRS UPR9022, University of Strasbourg, Strasbourg, France
| | - Eric R. G. R. Aguiar
- Laboratory of Virus Bioinformatics - Department of Biological Science, Center of Biotechnology and Genetics, Universidade Estadual de Santa Cruz - UESC, Ilhéus, Brazil
| | - Angélica T. Vieira
- Laboratory of Microbiota and Immunomodulation - Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Brazil
| |
Collapse
|
12
|
Liu G, Haw TJ, Starkey MR, Philp AM, Pavlidis S, Nalkurthi C, Nair PM, Gomez HM, Hanish I, Hsu AC, Hortle E, Pickles S, Rojas-Quintero J, Estepar RSJ, Marshall JE, Kim RY, Collison AM, Mattes J, Idrees S, Faiz A, Hansbro NG, Fukui R, Murakami Y, Cheng HS, Tan NS, Chotirmall SH, Horvat JC, Foster PS, Oliver BG, Polverino F, Ieni A, Monaco F, Caramori G, Sohal SS, Bracke KR, Wark PA, Adcock IM, Miyake K, Sin DD, Hansbro PM. TLR7 promotes smoke-induced experimental lung damage through the activity of mast cell tryptase. Nat Commun 2023; 14:7349. [PMID: 37963864 PMCID: PMC10646046 DOI: 10.1038/s41467-023-42913-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/25/2023] [Indexed: 11/16/2023] Open
Abstract
Toll-like receptor 7 (TLR7) is known for eliciting immunity against single-stranded RNA viruses, and is increased in both human and cigarette smoke (CS)-induced, experimental chronic obstructive pulmonary disease (COPD). Here we show that the severity of CS-induced emphysema and COPD is reduced in TLR7-deficient mice, while inhalation of imiquimod, a TLR7-agonist, induces emphysema without CS exposure. This imiquimod-induced emphysema is reduced in mice deficient in mast cell protease-6, or when wild-type mice are treated with the mast cell stabilizer, cromolyn. Furthermore, therapeutic treatment with anti-TLR7 monoclonal antibody suppresses CS-induced emphysema, experimental COPD and accumulation of pulmonary mast cells in mice. Lastly, TLR7 mRNA is increased in pre-existing datasets from patients with COPD, while TLR7+ mast cells are increased in COPD lungs and associated with severity of COPD. Our results thus support roles for TLR7 in mediating emphysema and COPD through mast cell activity, and may implicate TLR7 as a potential therapeutic target.
Collapse
Affiliation(s)
- Gang Liu
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Tatt Jhong Haw
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Malcolm R Starkey
- Depatrment of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Ashleigh M Philp
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare clinical campus, UNSW, Sydney, Australia
| | - Stelios Pavlidis
- The Airways Disease Section, National Heart & Lung Institute, Imperial College London, London, UK
| | - Christina Nalkurthi
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Prema M Nair
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Henry M Gomez
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Irwan Hanish
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Alan Cy Hsu
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Elinor Hortle
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Sophie Pickles
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | | | - Raul San Jose Estepar
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Jacqueline E Marshall
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Richard Y Kim
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Adam M Collison
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Joerg Mattes
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Sobia Idrees
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Alen Faiz
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Ryutaro Fukui
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minatoku, Tokyo, Japan
| | - Yusuke Murakami
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Musashino University, Nishitokyo-shi, Tokyo, Japan
| | - Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Jay C Horvat
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Paul S Foster
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Brian Gg Oliver
- Woolcock Institute of Medical Research, University of Sydney & School of Life Sciences, University of Technology, Sydney, Australia
| | | | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Section of Anatomic Pathology, Università di Messina, Messina, Italy
| | - Francesco Monaco
- Thoracic Surgery, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Gaetano Caramori
- Pneumologia, Dipartimento BIOMORF and Dipartimento di Medicina e Chirurgia, Universities of Messina and Parma, Messina, Italy
| | - Sukhwinder S Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, Australia
| | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Peter A Wark
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Ian M Adcock
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare clinical campus, UNSW, Sydney, Australia
| | - Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minatoku, Tokyo, Japan
| | - Don D Sin
- The University of British Columbia Centre for Heart Lung Innovation, St Paul's Hospital & Respiratory Division, Dept of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia.
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia.
| |
Collapse
|
13
|
Pereira RDD, Rabelo RAN, Oliveira NFDM, Porto SLT, Andrade ACDSP, Queiroz-Junior CM, Barbosa CLN, de Souza-Costa LP, Santos FRDS, Oliveira FBR, da Silva BLV, Umezu HL, Ferreira R, da Silva GSF, Cruz JS, Teixeira MM, Costa VV, Machado FS. A 5-Lipoxygenase Inhibitor, Zileuton, Modulates Host Immune Responses and Improves Lung Function in a Model of Severe Acute Respiratory Syndrome (SARS) Induced by Betacoronavirus. Viruses 2023; 15:2049. [PMID: 37896826 PMCID: PMC10611395 DOI: 10.3390/v15102049] [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: 09/15/2023] [Revised: 09/30/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
Exacerbated inflammatory responses are a hallmark of severe coronavirus disease 2019 (COVID-19). Zileuton (Zi) is a selective inhibitor of 5-lipoxygenase, an enzyme involved in the production of several inflammatory/pro-resolving lipid mediators. Herein, we investigated the effect of Zi treatment in a severe acute respiratory syndrome (SARS) model. Mouse hepatitis virus (MHV)3-infected mice treated with Zi significantly improved the clinical score, weight loss, cardiopulmonary function, and survival rates compared with infected untreated animals. The protection observed in Zi-treated mice was associated with a lower inflammatory score, reduced dendritic cell-producing tumor necrosis factor (TNF), and increased neutrophil-producing interleukin (IL)-10 in the lungs three days after infection (dpi). At 5 dpi, the lungs of treated mice showed an increase in Th2-, Treg CD4+-, and Treg CD8+-producing IL-10 and reduced Th1 infiltrating cells. Furthermore, similar results were found upon Zi treatment after SARS-CoV-2 infection in transgenic mice expressing the human angiotensin I-converting enzyme 2 (ACE2) receptor driven by the cytokeratin-18 (K18) gene promoter (K18-hACE2), significantly improving the clinical score, weight loss, and lung inflammatory score compared with untreated animals. Our data suggest that Zi protects against developing severe lung disease during SARS induced by betacoronavirus without affecting the host's capacity to deal with infection.
Collapse
Affiliation(s)
- Rafaela das Dores Pereira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.d.D.P.); (R.A.N.R.); (N.F.d.M.O.); (S.L.T.P.); (L.P.d.S.-C.); (F.R.d.S.S.); (F.B.R.O.); (R.F.); (J.S.C.); (M.M.T.)
| | - Rayane Aparecida Nonato Rabelo
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.d.D.P.); (R.A.N.R.); (N.F.d.M.O.); (S.L.T.P.); (L.P.d.S.-C.); (F.R.d.S.S.); (F.B.R.O.); (R.F.); (J.S.C.); (M.M.T.)
| | - Natália Fernanda de Melo Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.d.D.P.); (R.A.N.R.); (N.F.d.M.O.); (S.L.T.P.); (L.P.d.S.-C.); (F.R.d.S.S.); (F.B.R.O.); (R.F.); (J.S.C.); (M.M.T.)
| | - Samuel Luiz Teixeira Porto
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.d.D.P.); (R.A.N.R.); (N.F.d.M.O.); (S.L.T.P.); (L.P.d.S.-C.); (F.R.d.S.S.); (F.B.R.O.); (R.F.); (J.S.C.); (M.M.T.)
| | - Ana Claudia dos Santos Pereira Andrade
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (A.C.d.S.P.A.); (C.M.Q.-J.); (B.L.V.d.S.)
| | - Celso M. Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (A.C.d.S.P.A.); (C.M.Q.-J.); (B.L.V.d.S.)
| | - César Luís Nascimento Barbosa
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.d.D.P.); (R.A.N.R.); (N.F.d.M.O.); (S.L.T.P.); (L.P.d.S.-C.); (F.R.d.S.S.); (F.B.R.O.); (R.F.); (J.S.C.); (M.M.T.)
- Program in Health Sciences: Infectious Diseases and Tropical Medicine, Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Luiz Pedro de Souza-Costa
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.d.D.P.); (R.A.N.R.); (N.F.d.M.O.); (S.L.T.P.); (L.P.d.S.-C.); (F.R.d.S.S.); (F.B.R.O.); (R.F.); (J.S.C.); (M.M.T.)
| | - Felipe Rocha da Silva Santos
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.d.D.P.); (R.A.N.R.); (N.F.d.M.O.); (S.L.T.P.); (L.P.d.S.-C.); (F.R.d.S.S.); (F.B.R.O.); (R.F.); (J.S.C.); (M.M.T.)
| | - Fernando Bento Rodrigues Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.d.D.P.); (R.A.N.R.); (N.F.d.M.O.); (S.L.T.P.); (L.P.d.S.-C.); (F.R.d.S.S.); (F.B.R.O.); (R.F.); (J.S.C.); (M.M.T.)
| | - Bárbara Luísa Vieira da Silva
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (A.C.d.S.P.A.); (C.M.Q.-J.); (B.L.V.d.S.)
| | - Hanna L. Umezu
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (H.L.U.); (G.S.F.d.S.)
| | - Raquel Ferreira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.d.D.P.); (R.A.N.R.); (N.F.d.M.O.); (S.L.T.P.); (L.P.d.S.-C.); (F.R.d.S.S.); (F.B.R.O.); (R.F.); (J.S.C.); (M.M.T.)
| | - Glauber S. F. da Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (H.L.U.); (G.S.F.d.S.)
| | - Jader Santos Cruz
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.d.D.P.); (R.A.N.R.); (N.F.d.M.O.); (S.L.T.P.); (L.P.d.S.-C.); (F.R.d.S.S.); (F.B.R.O.); (R.F.); (J.S.C.); (M.M.T.)
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.d.D.P.); (R.A.N.R.); (N.F.d.M.O.); (S.L.T.P.); (L.P.d.S.-C.); (F.R.d.S.S.); (F.B.R.O.); (R.F.); (J.S.C.); (M.M.T.)
| | - Vivian Vasconcelos Costa
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (A.C.d.S.P.A.); (C.M.Q.-J.); (B.L.V.d.S.)
| | - Fabiana Simão Machado
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (R.d.D.P.); (R.A.N.R.); (N.F.d.M.O.); (S.L.T.P.); (L.P.d.S.-C.); (F.R.d.S.S.); (F.B.R.O.); (R.F.); (J.S.C.); (M.M.T.)
- Program in Health Sciences: Infectious Diseases and Tropical Medicine, Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| |
Collapse
|
14
|
Zhang C, Xu H, Netto KG, Sokulsky LA, Miao Y, Mo Z, Meng Y, Du Y, Wu C, Han L, Zhang L, Liu C, Zhang G, Li F, Yang M. Inhibition of γ-glutamyl transferase suppresses airway hyperresponsiveness and airway inflammation in a mouse model of steroid resistant asthma exacerbation. Front Immunol 2023; 14:1132939. [PMID: 37377967 PMCID: PMC10292800 DOI: 10.3389/fimmu.2023.1132939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Introduction Despite recent advances, there are limited treatments available for acute asthma exacerbations. Here, we investigated the therapeutic potential of GGsTop, a γ-glutamyl transferase inhibitor, on the disease with a murine model of asthma exacerbation. Methods GGsTop was administered to mice that received lipopolysaccharide (LPS) and ovalbumin (OVA) challenges. Airway hyperresponsiveness (AHR), lung histology, mucus hypersecretion, and collagen deposition were analyzed to evaluate the hallmark features of asthma exacerbation. The level of proinflammatory cytokines and glutathione were determined with/without GGsTop. The transcription profiles were also examined. Results GGsTop attenuates hallmark features of the disease with a murine model of LPS and OVA driven asthma exacerbation. Airway hyperresponsiveness (AHR), mucus hypersecretion, collagen deposition, and expression of inflammatory cytokines were dramatically inhibited by GGsTop treatment. Additionally, GGsTop restored the level of glutathione. Using RNA-sequencing and pathway analysis, we demonstrated that the activation of LPS/NFκB signaling pathway in airway was downregulated by GGsTop. Interestingly, further analysis revealed that GGsTop significantly inhibited not only IFNγ responses but also the expression of glucocorticoid-associated molecules, implicating that GGsTop profoundly attenuates inflammatory pathways. Conclusions Our study suggests that GGsTop is a viable treatment for asthma exacerbation by broadly inhibiting the activation of multiple inflammatory pathways.
Collapse
Affiliation(s)
- Cancan Zhang
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Huisha Xu
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Keilah G. Netto
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Leon A. Sokulsky
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Yiyan Miao
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhongyuan Mo
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yan Meng
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yingying Du
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chengyong Wu
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Liyou Han
- Institute for Liberal Arts and Sciences, Kyoto University, Kyoto, Japan
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chi Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Guojun Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fuguang Li
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ming Yang
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
15
|
Martins FRB, de Oliveira MD, Souza JAM, Queiroz-Junior CM, Lobo FP, Teixeira MM, Malacco NL, Soriani FM. Chronic ethanol exposure impairs alveolar leukocyte infiltration during pneumococcal pneumonia, leading to an increased bacterial burden despite increased CXCL1 and nitric oxide levels. Front Immunol 2023; 14:1175275. [PMID: 37275853 PMCID: PMC10235596 DOI: 10.3389/fimmu.2023.1175275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/05/2023] [Indexed: 06/07/2023] Open
Abstract
Ethanol abuse is a risk factor for the development of pneumonia caused by Streptococcus pneumoniae, a critical pathogen for public health. The aim of this article was to investigate the inflammatory mechanisms involved in pneumococcal pneumonia that may be associated with chronic ethanol exposure. Male C57BL6/J-Unib mice were exposed to 20% (v/v) ethanol for twelve weeks and intranasally infected with 5x104 CFU of S. pneumoniae. Twenty-four hours after infection, lungs, bronchoalveolar lavage and blood samples were obtained to assess the consequences of chronic ethanol exposure during infection. Alcohol-fed mice showed increased production of nitric oxide and CXCL1 in alveoli and plasma during pneumococcal pneumonia. Beside this, ethanol-treated mice exhibited a decrease in leukocyte infiltration into the alveoli and reduced frequency of severe lung inflammation, which was associated with an increase in bacterial load. Curiously, no changes were observed in survival after infection. Taken together, these results demonstrate that chronic ethanol exposure alters the inflammatory response during S. pneumoniae lung infection in mice with a reduction in the inflammatory infiltrate even in the presence of higher levels of the chemoattractant CXCL1.
Collapse
Affiliation(s)
- Flávia Rayssa Braga Martins
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maycon Douglas de Oliveira
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jéssica Amanda Marques Souza
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Francisco Pereira Lobo
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Frederico Marianetti Soriani
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
16
|
Zhou B, Wang L, Yang S, Liang Y, Zhang Y, Pan X, Li J. Diosmetin alleviates benzo[ a]pyrene-exacerbated H1N1 influenza virus-induced acute lung injury and dysregulation of inflammation through modulation of the PPAR-γ-NF-κB/P38 MAPK signaling axis. Food Funct 2023; 14:3357-3378. [PMID: 36942763 DOI: 10.1039/d2fo02590f] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
The severity of a viral respiratory illness was greatly exacerbated after exposure to a contaminant containing benzo[a]pyrene (B[a]P). Flavonoid-rich fruit intake has gained intense interest due to their health-promoting benefits for viral respiratory diseases, including influenza viruses. In our study, diosmetin (3',5,7-trihydroxy-4'-methoxyflavone), a naturally occurring hydroxylated methoxyflavone that is abundant in Citrus fruits, was explored for its effects on B[a]P-exacerbated H1N1 influenza virus-mediated inflammation and lung injury. Initially, in vivo results demonstrated that diosmetin protected against H1N1 virus-elicited acute lung injury. Simultaneously, H1N1 virus or B[a]P-stimulated A549 cells treated with diosmetin inhibited NF-κB and P-P38 activation, resulting in suppression of pro-inflammatory cytokines and apoptosis. Interestingly, diosmetin obviously promoted the expression of PPAR-γ as well as nuclear translocation of PPAR-γ, whereas, PPAR-γ inhibition by GW9662 weakened the inhibitory effects of diosmetin on H1N1 virus or B[a]P-mediated activation of NF-κB and P-P38, elevated expression of pro-inflammatory mediators as well as apoptosis. Furthermore, it was surprising to discover that mice exposed to both B[a]P and H1N1 viruses contributed to exacerbated acute lung injury, which were significantly ameliorated by diosmetin administration. In vitro studies showed that A549 cells with the combination of B[a]P and H1N1 virus augmented NF-κB and P-P38 activation, accompanied by higher levels of pro-inflammatory mediators and apoptosis, all of which were also significantly reduced by diosmetin treatment. Repressing PPAR-γ abrogated the inhibitory effects of diosmetin on B[a]P-exacerbated H1N1 virus-mediated NF-κB and P-P38 activation, inflammation, and apoptosis in A549 cells. Our findings suggest that diosmetin protected against B[a]P-exacerbated H1N1 virus-mediated lung injury by suppressing the exacerbation of NF-κB and P38 kinase activation in a PPAR-γ-dependent manner, suggesting potential benefits for B[a]P-exacerbated influenza-related illness therapeutics.
Collapse
Affiliation(s)
- Beixian Zhou
- The People's Hospital of Gaozhou, Gaozhou 525200, China
| | | | - Sushan Yang
- The People's Hospital of Gaozhou, Gaozhou 525200, China
| | - Yueyun Liang
- The People's Hospital of Gaozhou, Gaozhou 525200, China
| | - Yuehan Zhang
- The People's Hospital of Gaozhou, Gaozhou 525200, China
| | | | - Jing Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
- Institute of Chinese Integrative Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
17
|
Tim-3 blockade enhances the clearance of Chlamydia psittaci in the lung by promoting a cell-mediated immune response. Int Immunopharmacol 2023; 116:109780. [PMID: 36720194 DOI: 10.1016/j.intimp.2023.109780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/10/2023] [Accepted: 01/20/2023] [Indexed: 01/30/2023]
Abstract
Chlamydia psittaci is remarkable at disrupting immunity and thus poses a great risk to the animal industry and public health. Immune inhibitory molecule upregulation and the accumulation of specialized cells play key roles in chlamydial clearance. It is clear that the T-cell immunoglobulin and mucin domain protein 3 receptor (Tim-3) can regulate effector T cells in infectious disease. However, the immunomodulatory effect of Tim-3 in C. psittaci infection remains unknown. Thus, the expression of Tim-3 in effector T cells and its immune regulatory function during C. psittaci infection were investigated. The level of Tim-3 on CD4+ and CD8+ T cells was meaningfully higher in C. psittaci-infected mice. Blockade of Tim-3 signaling by anti-Tim-3 antibody showed accelerated C. psittaci clearance and less pathological changes in the lung than isotype immunoglobulin treatment. Furthermore, treatment with anti-Tim-3 antibody greatly enhanced the levels of IFN-γ and interleukin (IL)-22/IL-17, which were correlated with an improved Th1- and Th17-mediated immune response, and decreased IL-10, which were related with a decreased Treg immune response. In conclusion, Tim-3 expression in effector T cells negatively regulates Th1 and Th17 immune responses against C. psittaci respiratory infection.
Collapse
|
18
|
Absence of CCR2 Promotes Proliferation of Alveolar Macrophages That Control Lung Inflammation in Acute Respiratory Distress Syndrome in Mice. Int J Mol Sci 2022; 23:ijms232112920. [DOI: 10.3390/ijms232112920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/13/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) consists of uncontrolled inflammation that causes hypoxemia and reduced lung compliance. Since it is a complex process, not all details have been elucidated yet. In a well-controlled experimental murine model of lipopolysaccharide (LPS)-induced ARDS, the activity and viability of macrophages and neutrophils dictate the beginning and end phases of lung inflammation. C-C chemokine receptor type 2 (CCR2) is a critical chemokine receptor that mediates monocyte/macrophage activation and recruitment to the tissues. Here, we used CCR2-deficient mice to explore mechanisms that control lung inflammation in LPS-induced ARDS. CCR2−/− mice presented higher total numbers of pulmonary leukocytes at the peak of inflammation as compared to CCR2+/+ mice, mainly by enhanced influx of neutrophils, whereas we observed two to six-fold lower monocyte or interstitial macrophage numbers in the CCR2−/−. Nevertheless, the time needed to control the inflammation was comparable between CCR2+/+ and CCR2−/−. Interestingly, CCR2−/− mice presented higher numbers and increased proliferative rates of alveolar macrophages from day 3, with a more pronounced M2 profile, associated with transforming growth factor (TGF)-β and C-C chemokine ligand (CCL)22 production, decreased inducible nitric oxide synthase (Nos2), interleukin (IL)-1β and IL-12b mRNA expression and increased mannose receptor type 1 (Mrc1) mRNA and CD206 protein expression. Depletion of alveolar macrophages significantly delayed recovery from the inflammatory insult. Thus, our work shows that the lower number of infiltrating monocytes in CCR2−/− is partially compensated by increased proliferation of resident alveolar macrophages during the inflammation control of experimental ARDS.
Collapse
|
19
|
Kim K, Shin D, Lee G, Bae H. Loss of SP-A in the Lung Exacerbates Pulmonary Fibrosis. Int J Mol Sci 2022; 23:ijms23105292. [PMID: 35628104 PMCID: PMC9141401 DOI: 10.3390/ijms23105292] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/04/2022] [Accepted: 05/07/2022] [Indexed: 02/01/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating and common chronic lung disease that is pathologically characterized by the destruction of lung architecture and the accumulation of extracellular matrix in the lung. Previous studies have shown an association between lung surfactant protein (SP) and the pathogenesis of IPF, as demonstrated by mutations and the altered expression of SP in patients with IPF. However, the role of SP in the development of lung fibrosis is poorly understood. In this study, the role of surfactant protein A (SP-A) was explored in experimental lung fibrosis induced with a low or high dose of bleomycin (BLM) and CRISPR/Cas9-mediated genetic deletion of SP-A. Our results showed that lung SP-A deficiency in mice promoted the development of fibrotic damage and exacerbated inflammatory responses to the BLM challenge. In vitro experiments with murine lung epithelial LA-4 cells demonstrated that in response to transforming growth factor-β1 (TGF-β1), LA-4 cells had a decreased protein expression of SP-A. Furthermore, exogenous SP administration to LA-4 cells inhibited the TGF-β1-induced upregulation of fibrotic markers. Overall, these findings suggest a novel antifibrotic mechanism of SP-A in the development of lung fibrosis, which indicates the therapeutic potential of the lung SP-A in preventing the development of IPF.
Collapse
Affiliation(s)
- Kyunghwa Kim
- Department of Health Sciences, The Graduate School of Dong-A University, 840 Hadan-dong, Saha-gu, Busan 49315, Korea; (K.K.); (G.L.)
| | - Dasom Shin
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea;
| | - Gaheon Lee
- Department of Health Sciences, The Graduate School of Dong-A University, 840 Hadan-dong, Saha-gu, Busan 49315, Korea; (K.K.); (G.L.)
| | - Hyunsu Bae
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea;
- Correspondence:
| |
Collapse
|
20
|
Itaconate and itaconate derivatives target JAK1 to suppress alternative activation of macrophages. Cell Metab 2022; 34:487-501.e8. [PMID: 35235776 DOI: 10.1016/j.cmet.2022.02.002] [Citation(s) in RCA: 159] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/24/2021] [Accepted: 02/02/2022] [Indexed: 12/11/2022]
Abstract
The Krebs cycle-derived metabolite itaconate and its derivatives suppress the inflammatory response in pro-inflammatory "M1" macrophages. However, alternatively activated "M2" macrophages can take up itaconate. We therefore examined the effect of itaconate and 4-octyl itaconate (OI) on M2 macrophage activation. We demonstrate that itaconate and OI inhibit M2 polarization and metabolic remodeling. Examination of IL-4 signaling revealed inhibition of JAK1 and STAT6 phosphorylation by both itaconate and OI. JAK1 activation was also inhibited by OI in response to IL-13, interferon-β, and interferon-γ in macrophages and in T helper 2 (Th2) cells. Importantly, JAK1 was directly modified by itaconate derivatives at multiple residues, including cysteines 715, 816, 943, and 1130. Itaconate and OI also inhibited JAK1 kinase activity. Finally, OI treatment suppressed M2 macrophage polarization and JAK1 phosphorylation in vivo. We therefore identify itaconate and OI as JAK1 inhibitors, suggesting a new strategy to inhibit JAK1 in M2 macrophage-driven diseases.
Collapse
|
21
|
Liu X, Li X, Chen L, Hsu ACY, Asquith KL, Liu C, Laurie K, Barr I, Foster PS, Yang M. Proteomic Analysis Reveals a Novel Therapeutic Strategy Using Fludarabine for Steroid-Resistant Asthma Exacerbation. Front Immunol 2022; 13:805558. [PMID: 35280986 PMCID: PMC8913936 DOI: 10.3389/fimmu.2022.805558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/04/2022] [Indexed: 11/30/2022] Open
Abstract
Virus-induced asthma exacerbation is a health burden worldwide and lacks effective treatment. To better understand the disease pathogenesis and find novel therapeutic targets, we established a mouse model of steroid (dexamethasone (DEX)) resistant asthma exacerbation using ovalbumin (OVA) and influenza virus (FLU) infection. Using liquid chromatography-tandem mass spectrometry (LC-MC/MS), we performed a shotgun proteomics assay coupled with label-free quantification to define all dysregulated proteins in the lung proteome of asthmatic mice. Compared to control, 71, 89, and 30 proteins were found significantly upregulated by at least two-fold (p-value ≤ 0.05) in OVA-, OVA/FLU-, and OVA/FLU/DEX-treated mice, respectively. We then applied a Z-score transformed hierarchical clustering analysis and Ingenuity Pathway Analysis (IPA) to highlight the key inflammation pathways underlying the disease. Within all these upregulated proteins, 64 proteins were uniquely highly expressed in OVA/FLU mice compared to OVA mice; and 11 proteins were DEX-refractory. IPA assay revealed two of the most enriched pathways associated with these over-expressed protein clusters were those associated with MHC class I (MHC-I) antigen-presentation and interferon (IFN) signaling. Within these pathways, signal-transducer-and-activator-of-transcription-1 (STAT1) protein was identified as the most significantly changed protein contributing to the pathogenesis of exacerbation and the underlying steroid resistance based on the label-free quantification; and this was further confirmed by both Parallel Reaction Monitoring (PRM) proteomics assay and western blots. Further, the pharmacological drug Fludarabine decreased STAT1 expression, restored the responsiveness of OVA/FLU mice to DEX and markedly suppressed disease severity. Taken together, this study describes the proteomic profile underpinning molecular mechanisms of FLU-induced asthma exacerbation and identifies STAT1 as a potential therapeutic target, more importantly, we provided a novel therapeutic strategy that may be clinically translated into practice.
Collapse
Affiliation(s)
- Xiaoming Liu
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Health Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
| | - Xiang Li
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Health Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
| | - Ling Chen
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Health Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
| | - Alan Chen-Yu Hsu
- Priority Research Centre for Health Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Kelly L. Asquith
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Health Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
| | - Chi Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Karen Laurie
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Ian Barr
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Paul S. Foster
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Health Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
- *Correspondence: Ming Yang, ; Paul S. Foster,
| | - Ming Yang
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Health Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
- *Correspondence: Ming Yang, ; Paul S. Foster,
| |
Collapse
|
22
|
Zhao L, Li J, Zhou X, Pan Q, Zhao W, Yang X, Wang H. Natural Killer Cells Regulate Pulmonary Macrophages Polarization in Host Defense Against Chlamydial Respiratory Infection. Front Cell Infect Microbiol 2022; 11:775663. [PMID: 35059323 PMCID: PMC8764407 DOI: 10.3389/fcimb.2021.775663] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/07/2021] [Indexed: 12/22/2022] Open
Abstract
NK cells and pulmonary macrophages both are important components of innate immunity. The interaction between NK cells and pulmonary macrophages during chlamydial infection is poorly understood. In this study, we explored the effect of NK cells on regulation of pulmonary macrophage function during chlamydial respiratory infection. We found that NK depletion led to polarization of pulmonary macrophages from M1 to M2 phenotype, and it is related to reduced miR-155 expression in lung macrophage. Using adoptive transfer approach, we found that the recipients receiving lung macrophages isolated from C. muridarum-infected NK-cell-depleted mice exhibited an increased bacterial load and severe inflammation in the lung upon chlamydial challenge infection when compared with the recipients of lung macrophages from infected isotype control antibody treated mice. Herein, the effects of NK cells on macrophage polarization were examined in vitro. We found that NK cells from chlamydial-infected mice (iNK) significantly induced M1 polarization compared to that from uninfected mice (uNK). Inhibition of miR-155 expression in macrophages reduced M1 polarization induced by iNK, while miR-155 over-expression enhanced it. Furthermore, neutralization of IFN-γ in the coculture system decreased the expression of miR-155 by macrophages, and resulted in weakened M1 polarization. The data indicates that NK cells promote M1 polarization through up-regulation of miR-155 in macrophages by producing IFN-γ during chlamydial infection, and NK-regulated macrophage polarization is functionally relevant to host defense against the infection.
Collapse
Affiliation(s)
- Lei Zhao
- Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Li
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, China.,Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xiaoqing Zhou
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Qianqian Pan
- Department of Respiratory, Laiwu Central Hospital, Jinan, China
| | - Weiming Zhao
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xi Yang
- Departments of Immunology and Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Hong Wang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| |
Collapse
|
23
|
Matos NAD, Reis DCD, Rocha LK, Mattos MSD, Cassali GD, Russo RC, Perez ADC, Klein A. Pharmacological blockade of protease-Activated Receptor 2 improves airway remodeling and lung inflammation in experimental allergic asthma. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e201089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
24
|
Single-cell RNA transcriptomic analysis identifies Creb5 and CD11b-DCs as regulator of asthma exacerbations. Mucosal Immunol 2022; 15:1363-1374. [PMID: 36038770 PMCID: PMC9705253 DOI: 10.1038/s41385-022-00556-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 07/20/2022] [Accepted: 08/08/2022] [Indexed: 02/04/2023]
Abstract
Immune responses that result in asthma exacerbation are associated with allergen or viral exposure. Identification of common immune factors will be beneficial for the development of uniformed targeted therapy. We employed a House Dust Mite (HDM) mouse model of asthma and challenged allergic HDM mice with allergens (HDM, cockroach extract (CRE)) or respiratory syncytial virus (RSV). Purified lung immune cells underwent high-dimensional single-cell RNA deep sequencing (scRNA-seq) to generate an RNA transcriptome. Gene silencing with siRNA was employed to confirm the efficacy of scRNA-seq analysis. scRNA-seq UMAP analysis portrayed an array of cell markers within individual immune clusters. SCENIC R analysis showed an increase in regulon number and activity in CD11b- DC cells. Analysis of conserved regulon factors further identified Creb5 as a shared regulon between the exacerbation groups. Creb5 siRNAs attenuated HDM, CRE or RSV-induced asthma exacerbation. scRNA-seq multidimensional analysis of immune clusters identified gene pathways that were conserved between the exacerbation groups. We propose that these analyses provide a strong framework that could be used to identify specific therapeutic targets in multifaceted pathologies.
Collapse
|
25
|
Kim RY, Sunkara KP, Bracke KR, Jarnicki AG, Donovan C, Hsu AC, Ieni A, Beckett EL, Galvão I, Wijnant S, Ricciardolo FL, Di Stefano A, Haw TJ, Liu G, Ferguson AL, Palendira U, Wark PA, Conickx G, Mestdagh P, Brusselle GG, Caramori G, Foster PS, Horvat JC, Hansbro PM. A microRNA-21-mediated SATB1/S100A9/NF-κB axis promotes chronic obstructive pulmonary disease pathogenesis. Sci Transl Med 2021; 13:eaav7223. [PMID: 34818056 DOI: 10.1126/scitranslmed.aav7223] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Richard Y Kim
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales 2007, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Krishna P Sunkara
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia.,Graduate School of Health, Discipline of Pharmacy, University of Technology Sydney, Sydney, New South Wales 2007, Australia.,Intensive Care Unit, John Hunter Hospital, Newcastle, New South Wales 2308, Australia
| | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - Andrew G Jarnicki
- Department of Biochemistry and Pharmacology, University of Melbourne, Victoria 3010, Australia
| | - Chantal Donovan
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales 2007, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Alan C Hsu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Section of Anatomic Pathology, University of Messina, Messina 98100, Italy
| | - Emma L Beckett
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Izabela Galvão
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Sara Wijnant
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - Fabio Lm Ricciardolo
- Rare Lung Disease Unit, Department of Clinical and Biological Sciences, University of Torino, San Luigi Gonzaga University Hospital Orbassano, Torino 10043, Italy
| | - Antonino Di Stefano
- Istituti Clinici Scientifici Maugeri, IRCCS, SpA Società Benefit, Divisione di Pneumologia e Laboratorio di Citoimmunopatologia dell'Apparato Cardio Respiratorio, Veruno, Novara 28100, Italy
| | - Tatt Jhong Haw
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Gang Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Angela L Ferguson
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2006, Australia.,Centenary Institute and University of Technology Sydney, Sydney, New South Wales 2006, Australia
| | - Umamainthan Palendira
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Peter A Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Griet Conickx
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent 9000, Belgium.,Ablynx N.V., a Sanofi company, Ghent 9052, Belgium
| | - Pieter Mestdagh
- Center for Medical Genetics and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Guy G Brusselle
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - Gaetano Caramori
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina 98100, Italy
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales 2007, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia
| |
Collapse
|
26
|
Andrade ACDSP, Campolina-Silva GH, Queiroz-Junior CM, de Oliveira LC, Lacerda LDSB, Pimenta JC, de Souza FRO, de Meira Chaves I, Passos IB, Teixeira DC, Bittencourt-Silva PG, Valadão PAC, Rossi-Oliveira L, Antunes MM, Figueiredo AFA, Wnuk NT, Temerozo JR, Ferreira AC, Cramer A, Oliveira CA, Durães-Carvalho R, Weis Arns C, Guimarães PPG, Costa GMJ, de Menezes GB, Guatimosim C, da Silva GSF, Souza TML, Barrioni BR, Pereira MDM, de Sousa LP, Teixeira MM, Costa VV. A Biosafety Level 2 Mouse Model for Studying Betacoronavirus-Induced Acute Lung Damage and Systemic Manifestations. J Virol 2021; 95:e0127621. [PMID: 34495692 PMCID: PMC8549505 DOI: 10.1128/jvi.01276-21] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/01/2021] [Indexed: 12/29/2022] Open
Abstract
The emergence of life-threatening zoonotic diseases caused by betacoronaviruses, including the ongoing coronavirus disease 19 (COVID-19) pandemic, has highlighted the need for developing preclinical models mirroring respiratory and systemic pathophysiological manifestations seen in infected humans. Here, we showed that C57BL/6J wild-type mice intranasally inoculated with the murine betacoronavirus murine hepatitis coronavirus 3 (MHV-3) develop a robust inflammatory response leading to acute lung injuries, including alveolar edema, hemorrhage, and fibrin thrombi. Although such histopathological changes seemed to resolve as the infection advanced, they efficiently impaired respiratory function, as the infected mice displayed restricted lung distention and increased respiratory frequency and ventilation. Following respiratory manifestation, the MHV-3 infection became systemic, and a high virus burden could be detected in multiple organs along with morphological changes. The systemic manifestation of MHV-3 infection was also marked by a sharp drop in the number of circulating platelets and lymphocytes, besides the augmented concentration of the proinflammatory cytokines interleukin 1 beta (IL-1β), IL-6, IL-12, gamma interferon (IFN-γ), and tumor necrosis factor (TNF), thereby mirroring some clinical features observed in moderate and severe cases of COVID-19. Importantly, both respiratory and systemic changes triggered by MHV-3 infection were greatly prevented by blocking TNF signaling, either via genetic or pharmacologic approaches. In line with this, TNF blockage also diminished the infection-mediated release of proinflammatory cytokines and virus replication of human epithelial lung cells infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Collectively, results show that MHV-3 respiratory infection leads to a large range of clinical manifestations in mice and may constitute an attractive, lower-cost, biosafety level 2 (BSL2) in vivo platform for evaluating the respiratory and multiorgan involvement of betacoronavirus infections. IMPORTANCE Mouse models have long been used as valuable in vivo platforms to investigate the pathogenesis of viral infections and effective countermeasures. The natural resistance of mice to the novel betacoronavirus SARS-CoV-2, the causative agent of COVID-19, has launched a race toward the characterization of SARS-CoV-2 infection in other animals (e.g., hamsters, cats, ferrets, bats, and monkeys), as well as adaptation of the mouse model, by modifying either the host or the virus. In the present study, we utilized a natural pathogen of mice, MHV, as a prototype to model betacoronavirus-induced acute lung injure and multiorgan involvement under biosafety level 2 conditions. We showed that C57BL/6J mice intranasally inoculated with MHV-3 develops severe disease, which includes acute lung damage and respiratory distress that precede systemic inflammation and death. Accordingly, the proposed animal model may provide a useful tool for studies regarding betacoronavirus respiratory infection and related diseases.
Collapse
Affiliation(s)
| | - Gabriel Henrique Campolina-Silva
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Leonardo Camilo de Oliveira
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Jordane C Pimenta
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Ian de Meira Chaves
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ingredy Beatriz Passos
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danielle Cunha Teixeira
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Paloma Graziele Bittencourt-Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Leonardo Rossi-Oliveira
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maisa Mota Antunes
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - André Felipe Almeida Figueiredo
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Natália Teixeira Wnuk
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jairo R. Temerozo
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, RJ, Brazil
- National Institute for Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - André Costa Ferreira
- National Institute for Science and Technology on Innovation on Diseases of Neglected Populations (INCT/IDNP), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, RJ, Brazil
- Immunopharmacology Laboratory, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, RJ, Brazil
- Laboratório de Pesquisas Pré-clínicas, Universidade Iguaçu (UNIG), Rio de Janeiro, RJ, Brazil
| | - Allysson Cramer
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Cleida Aparecida Oliveira
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Clarice Weis Arns
- Laboratory of Virology, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Pedro Pires Goulart Guimarães
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Guilherme Mattos Jardim Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gustavo Batista de Menezes
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Cristina Guatimosim
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Glauber Santos Ferreira da Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Thiago Moreno L. Souza
- National Institute for Science and Technology on Innovation on Diseases of Neglected Populations (INCT/IDNP), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, RJ, Brazil
- Immunopharmacology Laboratory, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Breno Rocha Barrioni
- Department of Metallurgical Engineering and Materials, Federal University of Minas Gerais, School of Engineering, Belo Horizonte, Brazil
| | - Marivalda de Magalhães Pereira
- Department of Metallurgical Engineering and Materials, Federal University of Minas Gerais, School of Engineering, Belo Horizonte, Brazil
| | - Lirlândia Pires de Sousa
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vivian Vasconcelos Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
27
|
Liu X, Nguyen TH, Sokulsky L, Li X, Garcia Netto K, Hsu ACY, Liu C, Laurie K, Barr I, Tay H, Eyers F, Foster PS, Yang M. IL-17A is a common and critical driver of impaired lung function and immunopathology induced by influenza virus, rhinovirus and respiratory syncytial virus. Respirology 2021; 26:1049-1059. [PMID: 34472161 DOI: 10.1111/resp.14141] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/08/2021] [Accepted: 08/09/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVE Influenza virus (FLU), rhinovirus (RV) and respiratory syncytial virus (RSV) are the most common acute respiratory infections worldwide. Infection can cause severe health outcomes, while therapeutic options are limited, primarily relieving symptoms without attenuating the development of lesions or impaired lung function. We therefore examined the inflammatory response to these infections with the intent to identify common components that are critical drivers of immunopathogenesis and thus represent potential therapeutic targets. METHODS BALB/c mice were infected with FLU, RV or RSV, and lung function, airway inflammation and immunohistopathology were measured over a 10-day period. Anti-IL-17A mAb was administered to determine the impact of attenuating this cytokine's function on the development and severity of disease. RESULTS All three viruses induced severe airway constriction and inflammation at 2 days post-infection (dpi). However, only FLU induced prolonged inflammation till 10 dpi. Increased IL-17A expression was correlated with the alterations in lung function and its persistence. Neutralization of IL-17A did not affect the viral replication but led to the resolution of airway hyperresponsiveness. Furthermore, anti-IL-17A treatment resulted in reduced infiltration of neutrophils (in RV- and FLU-infected mice at 2 dpi) and lymphocytes (in RSV-infected mice at 2 dpi and FLU-infected mice at 10 dpi), and attenuated the severity of immunopathology. CONCLUSION IL-17A is a common pathogenic molecule regulating disease induced by three prevalent respiratory viruses. Targeting the IL-17A pathway may provide a unified approach to the treatment of these respiratory infections alleviating both inflammation-induced lesions and difficulties in breathing.
Collapse
Affiliation(s)
- Xiaoming Liu
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Thi Hiep Nguyen
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Leon Sokulsky
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
| | - Xiang Li
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Keilah Garcia Netto
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Alan Chen-Yu Hsu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, New South Wales, Australia.,School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia.,Programme in Emerging Infectious Diseases, Duke - National University of Singapore (NUS) Medical School, Singapore
| | - Chi Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Karen Laurie
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Ian Barr
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hock Tay
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Fiona Eyers
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Paul S Foster
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Ming Yang
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
28
|
Kuchibhotla VNS, Starkey MR, Reid AT, Heijink IH, Nawijn MC, Hansbro PM, Knight DA. Inhibition of β-Catenin/CREB Binding Protein Signaling Attenuates House Dust Mite-Induced Goblet Cell Metaplasia in Mice. Front Physiol 2021; 12:690531. [PMID: 34385933 PMCID: PMC8353457 DOI: 10.3389/fphys.2021.690531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 07/05/2021] [Indexed: 11/26/2022] Open
Abstract
Excessive mucus production is a major feature of allergic asthma. Disruption of epithelial junctions by allergens such as house dust mite (HDM) results in the activation of β-catenin signaling, which has been reported to stimulate goblet cell differentiation. β-catenin interacts with various co-activators including CREB binding protein (CBP) and p300, thereby regulating the expression of genes involved in cell proliferation and differentiation, respectively. We specifically investigated the role of the β-catenin/CBP signaling pathway in goblet cell metaplasia in a HDM-induced allergic airway disease model in mice using ICG-001, a small molecule inhibitor that blocks the binding of CBP to β-catenin. Female 6- 8-week-old BALB/c mice were sensitized to HDM/saline on days 0, 1, and 2, followed by intranasal challenge with HDM/saline with or without subcutaneous ICG-001/vehicle treatment from days 14 to 17, and samples harvested 24 h after the last challenge/treatment. Differential inflammatory cells in bronchoalveolar lavage (BAL) fluid were enumerated. Alcian blue (AB)/Periodic acid–Schiff (PAS) staining was used to identify goblet cells/mucus production, and airway hyperresponsiveness (AHR) was assessed using invasive plethysmography. Exposure to HDM induced airway inflammation, goblet cell metaplasia and increased AHR, with increased airway resistance in response to the non-specific spasmogen methacholine. Inhibition of the β-catenin/CBP pathway using treatment with ICG-001 significantly attenuated the HDM-induced goblet cell metaplasia and infiltration of macrophages, but had no effect on eosinophils, neutrophils, lymphocytes or AHR. Increased β-catenin/CBP signaling may promote HDM-induced goblet cell metaplasia in mice.
Collapse
Affiliation(s)
- Virinchi N S Kuchibhotla
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,GRIAC Research Institute, University Medical Center Groningen, Groningen, Netherlands.,Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research, University of Groningen, Groningen, Netherlands
| | - Malcolm R Starkey
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Priority Research Centre GrowUpWell and Hunter Medical Research Institute, Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Andrew T Reid
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
| | - Irene H Heijink
- GRIAC Research Institute, University Medical Center Groningen, Groningen, Netherlands.,Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research, University of Groningen, Groningen, Netherlands.,Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Martijn C Nawijn
- GRIAC Research Institute, University Medical Center Groningen, Groningen, Netherlands.,Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research, University of Groningen, Groningen, Netherlands
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Darryl A Knight
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Providence Health Care Research Institute, Vancouver, BC, Canada.,Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
29
|
Skerrett-Byrne DA, Bromfield EG, Murray HC, Jamaluddin MFB, Jarnicki AG, Fricker M, Essilfie AT, Jones B, Haw TJ, Hampsey D, Anderson AL, Nixon B, Scott RJ, Wark PAB, Dun MD, Hansbro PM. Time-resolved proteomic profiling of cigarette smoke-induced experimental chronic obstructive pulmonary disease. Respirology 2021; 26:960-973. [PMID: 34224176 DOI: 10.1111/resp.14111] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/01/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVE Chronic obstructive pulmonary disease (COPD) is the third leading cause of illness and death worldwide. Current treatments aim to control symptoms with none able to reverse disease or stop its progression. We explored the major molecular changes in COPD pathogenesis. METHODS We employed quantitative label-based proteomics to map the changes in the lung tissue proteome of cigarette smoke-induced experimental COPD that is induced over 8 weeks and progresses over 12 weeks. RESULTS Quantification of 7324 proteins enabled the tracking of changes to the proteome. Alterations in protein expression profiles occurred in the induction phase, with 18 and 16 protein changes at 4- and 6-week time points, compared to age-matched controls, respectively. Strikingly, 269 proteins had altered expression after 8 weeks when the hallmark pathological features of human COPD emerge, but this dropped to 27 changes at 12 weeks with disease progression. Differentially expressed proteins were validated using other mouse and human COPD bronchial biopsy samples. Major changes in RNA biosynthesis (heterogeneous nuclear ribonucleoproteins C1/C2 [HNRNPC] and RNA-binding protein Musashi homologue 2 [MSI2]) and modulators of inflammatory responses (S100A1) were notable. Mitochondrial dysfunction and changes in oxidative stress proteins also occurred. CONCLUSION We provide a detailed proteomic profile, identifying proteins associated with the pathogenesis and disease progression of COPD establishing a platform to develop effective new treatment strategies.
Collapse
Affiliation(s)
- David A Skerrett-Byrne
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Elizabeth G Bromfield
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia.,Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Heather C Murray
- University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - M Fairuz B Jamaluddin
- University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Andrew G Jarnicki
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Michael Fricker
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Ama T Essilfie
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Queensland Institute of Medical Research, Herston, Queensland, Australia
| | - Bernadette Jones
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Tatt J Haw
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Daniel Hampsey
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Amanda L Anderson
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Brett Nixon
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Rodney J Scott
- University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Matthew D Dun
- University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
30
|
Vieira-Santos F, Leal-Silva T, de Lima Silva Padrão L, Ruas ACL, Nogueira DS, Kraemer L, Oliveira FMS, Caliari MV, Russo RC, Fujiwara RT, Bueno LL. Concomitant experimental coinfection by Plasmodium berghei NK65-NY and Ascaris suum downregulates the Ascaris-specific immune response and potentiates Ascaris-associated lung pathology. Malar J 2021; 20:296. [PMID: 34210332 PMCID: PMC8248286 DOI: 10.1186/s12936-021-03824-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 06/17/2021] [Indexed: 12/22/2022] Open
Abstract
Background Ascariasis and malaria are highly prevalent parasitic diseases in tropical regions and often have overlapping endemic areas, contributing to high morbidity and mortality rates in areas with poor sanitary conditions. Several studies have previously aimed to correlate the effects of Ascaris-Plasmodium coinfections but have obtained contradictory and inconclusive results. Therefore, the present study aimed to investigate parasitological and immunopathological aspects of the lung during murine experimental concomitant coinfection by Plasmodium berghei and Ascaris suum during larvae ascariasis. Methods C57BL/6J mice were inoculated with 1 × 104P. berghei strain NK65-NY-infected red blood cells (iRBCs) intraperitoneally and/or 2500 embryonated eggs of A. suum by oral gavage. P. berghei parasitaemia, morbidity and the survival rate were assessed. On the seventh day postinfection (dpi), A. suum lung burden analysis; bronchoalveolar lavage (BAL); histopathology; NAG, MPO and EPO activity measurements; haematological analysis; and respiratory mechanics analysis were performed. The concentrations of interleukin (IL)-1β, IL-12/IL-23p40, IL-6, IL-4, IL-33, IL-13, IL-5, IL-10, IL-17A, IFN-γ, TNF and TGF-β were assayed by sandwich ELISA. Results Animals coinfected with P. berghei and A. suum show decreased production of type 1, 2, and 17 and regulatory cytokines; low leukocyte recruitment in the tissue; increased cellularity in the circulation; and low levels of NAG, MPO and EPO activity that lead to an increase in larvae migration, as shown by the decrease in larvae recovered in the lung parenchyma and increase in larvae recovered in the airway. This situation leads to severe airway haemorrhage and, consequently, an impairment respiratory function that leads to high morbidity and early mortality. Conclusions This study demonstrates that the Ascaris-Plasmodium interaction is harmful to the host and suggests that this coinfection may potentiate Ascaris-associated pathology by dampening the Ascaris-specific immune response, resulting in the early death of affected animals. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-03824-w.
Collapse
Affiliation(s)
- Flaviane Vieira-Santos
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thaís Leal-Silva
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiza de Lima Silva Padrão
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Cristina Loiola Ruas
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Denise Silva Nogueira
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas Kraemer
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabrício Marcus Silva Oliveira
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcelo Vidigal Caliari
- Laboratory of Protozooses, Institute of Biological Sciences, Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Institute of Biological Sciences, Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lilian Lacerda Bueno
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
31
|
Lu Z, Van Eeckhoutte HP, Liu G, Nair PM, Jones B, Gillis CM, Nalkurthi BC, Verhamme F, Buyle-Huybrecht T, Vandenabeele P, Berghe TV, Brusselle GG, Horvat JC, Murphy JM, Wark PA, Bracke KR, Fricker M, Hansbro PM. Necroptosis Signalling Promotes Inflammation, Airway Remodelling and Emphysema in COPD. Am J Respir Crit Care Med 2021; 204:667-681. [PMID: 34133911 DOI: 10.1164/rccm.202009-3442oc] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Necroptosis, mediated by RIPK3 and MLKL, is a form of regulated necrosis that can drive tissue inflammation and destruction, however its contribution to COPD pathogenesis is poorly understood. OBJECTIVES To determine the role of necroptosis in COPD. METHODS Levels of RIPK3, MLKL and activated phospho-MLKL were measured in lung tissues of COPD patients and non-COPD controls. Necroptosis-related mRNA and proteins and cell death were examined in the lungs and pulmonary macrophages of mice with cigarette smoke (CS)-induced experimental COPD. The responses of Ripk3- and Mlkl-deficient (-/-) mice to CS exposure were compared to wild-type mice. Combined inhibition of apoptosis (pan-caspase inhibitor qVD-OPh) and necroptosis (Mlkl-/- mice) was assessed. MEASUREMENTS AND MAIN RESULTS Protein levels of MLKL and pMLKL but not RIPK3 were increased in lung tissues of COPD patients compared to never smokers or smoker non-COPD controls. Necroptosis-related mRNA and protein levels were increased in lung tissue and macrophages in CS-exposed mice/experimental COPD. Ripk3 or Mlkl deletion prevented airway inflammation in response to acute CS-exposure. Ripk3 deficiency reduced airway inflammation and remodelling and development of emphysematous pathology following chronic CS-exposure. Mlkl deletion and qVD-OPh treatment reduced chronic CS-induced airway inflammation, but only Mlkl deletion prevented airway remodelling and emphysema. Ripk3 or Mlkl deletion and qVD-OPh treatment reduced CS-induced lung cell death. CONCLUSIONS Necroptosis is induced by CS exposure and increased in COPD patient lungs and experimental COPD. Inhibiting necroptosis attenuates CS-induced airway inflammation, airway remodelling and emphysema. Targeted inhibition of necroptosis is a potential therapeutic strategy in COPD.
Collapse
Affiliation(s)
- Zhe Lu
- The University of Newcastle Hunter Medical Research Institute, 454568, Priority Research Centre for Healthy Lungs, New Lambton, New South Wales, Australia
| | | | - Gang Liu
- The University of Newcastle Hunter Medical Research Institute, 454568, Priority Research Centre for Healthy Lungs, New Lambton, New South Wales, Australia.,University of Technology Sydney Faculty of Science, 170529, Centre for Inflammation, Centenary Institute, Sydney, New South Wales, Australia
| | - Prema M Nair
- University of Newcastle Hunter Medical Research Institute, 454568, Priority Research Centres for Healthy Lungs and GrowUpWell, New Lambton, New South Wales, Australia.,The University of Newcastle Faculty of Health and Medicine, 64834, School of Biomedical Sciences and Pharmacy, Callaghan, New South Wales, Australia
| | - Bernadette Jones
- The University of Newcastle, 5982, Centre for Asthma & Respiratory Disease, Callaghan, New South Wales, Australia
| | - Caitlin M Gillis
- University of Technology Sydney Faculty of Science, 170529, Centre for Inflammation, Centenary Institute, Sydney, New South Wales, Australia.,Ghent University, 26656, VIB Center for Inflammation Research, Department for Biomedical Molecular Biology, Gent, Belgium.,Ghent University, 26656, Methusalem program CEDAR-IC, Gent, Belgium
| | - B Christina Nalkurthi
- University of Technology Sydney Faculty of Science, 170529, Centre for Inflammation, Centenary Institute, Sydney, New South Wales, Australia
| | | | - Tamariche Buyle-Huybrecht
- University Hospital Ghent, 60200, Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Gent, Belgium
| | - Peter Vandenabeele
- University Hospital Ghent, 60200, Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Gent, Belgium
| | - Tom Vanden Berghe
- Ghent University, 26656, VIB Center for Inflammation Research, Department for Biomedical Molecular Biology, Gent, Belgium.,University of Antwerp, 26660, Department Biomedical Sciences, Antwerpen, Belgium
| | - Guy G Brusselle
- University Hospital Ghent, 60200, Respiratory Medicine, Gent, Belgium
| | - Jay C Horvat
- Hunter Medical Research Institute, Vaccines, Immunity, Viruses and Asthma Group, Newcastle, New South Wales, Australia
| | - James M Murphy
- Walter and Eliza Hall Institute of Medical Research, 5388, Department of Medical Biology University of Melbourne , Melbourne, Victoria, Australia
| | - Peter A Wark
- The University of Newcastle, 5982, Centre for Asthma & Respiratory Disease, Callaghan, New South Wales, Australia.,The University of Newcastle Hunter Medical Research Institute, 454568, Vaccines, Infection, Viruses & Asthma, New Lambton, New South Wales, Australia
| | - Ken R Bracke
- University Hospital Ghent, 60200, Respiratory Medicine, Gent, Belgium
| | - Michael Fricker
- The University of Newcastle Hunter Medical Research Institute, 454568, Priority Research Centres for Healthy Lungs & Grow Up Well, New Lambton, New South Wales, Australia
| | - Philip M Hansbro
- University of Technology Sydney, 1994, Sydney, New South Wales, Australia;
| |
Collapse
|
32
|
Sencio V, Barthelemy A, Tavares LP, Machado MG, Soulard D, Cuinat C, Queiroz-Junior CM, Noordine ML, Salomé-Desnoulez S, Deryuter L, Foligné B, Wahl C, Frisch B, Vieira AT, Paget C, Milligan G, Ulven T, Wolowczuk I, Faveeuw C, Le Goffic R, Thomas M, Ferreira S, Teixeira MM, Trottein F. Gut Dysbiosis during Influenza Contributes to Pulmonary Pneumococcal Superinfection through Altered Short-Chain Fatty Acid Production. Cell Rep 2021; 30:2934-2947.e6. [PMID: 32130898 DOI: 10.1016/j.celrep.2020.02.013] [Citation(s) in RCA: 229] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/13/2019] [Accepted: 02/04/2020] [Indexed: 02/07/2023] Open
Abstract
Secondary bacterial infections often complicate viral respiratory infections. We hypothesize that perturbation of the gut microbiota during influenza A virus (IAV) infection might favor respiratory bacterial superinfection. Sublethal infection with influenza transiently alters the composition and fermentative activity of the gut microbiota in mice. These changes are attributed in part to reduced food consumption. Fecal transfer experiments demonstrate that the IAV-conditioned microbiota compromises lung defenses against pneumococcal infection. In mechanistic terms, reduced production of the predominant short-chain fatty acid (SCFA) acetate affects the bactericidal activity of alveolar macrophages. Following treatment with acetate, mice colonized with the IAV-conditioned microbiota display reduced bacterial loads. In the context of influenza infection, acetate supplementation reduces, in a free fatty acid receptor 2 (FFAR2)-dependent manner, local and systemic bacterial loads. This translates into reduced lung pathology and improved survival rates of double-infected mice. Lastly, pharmacological activation of the SCFA receptor FFAR2 during influenza reduces bacterial superinfection.
Collapse
Affiliation(s)
- Valentin Sencio
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Adeline Barthelemy
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Luciana P Tavares
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marina G Machado
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France; Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daphnée Soulard
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Céline Cuinat
- Micalis Institute, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | | | - Marie-Louise Noordine
- Micalis Institute, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Sophie Salomé-Desnoulez
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Lucie Deryuter
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Benoit Foligné
- Université de Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), CHU Lille, U995, Lille Inflammation Research International Center (LIRIC), 59000 Lille, France
| | | | - Benoit Frisch
- Centre National de la Recherche Scientifique, Université de Strasbourg, Faculté de Pharmacie, 67400 Illkirch, France
| | - Angelica T Vieira
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Christophe Paget
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, University of Glasgow, G12 8QQ Glasgow, Scotland, UK
| | - Trond Ulven
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Isabelle Wolowczuk
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Christelle Faveeuw
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Ronan Le Goffic
- Molecular Virology and Immunology, Institut National de la Recherche Agronomique, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Muriel Thomas
- Micalis Institute, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | | | - Mauro M Teixeira
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - François Trottein
- Université de Lille, U1019 UMR 9017, Centre d'Infection et d'Immunité de Lille (CIIL), 59000 Lille, France; Centre National de la Recherche Scientifique, UMR 9017, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale, U1019, 59000 Lille, France; Centre Hospitalier Universitaire de Lille, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France.
| |
Collapse
|
33
|
Nazareth L, Walkden H, Chacko A, Delbaz A, Shelper T, Armitage CW, Reshamwala R, Trim LK, St John JA, Beagley KW, Ekberg JAK. Chlamydia muridarum Can Invade the Central Nervous System via the Olfactory and Trigeminal Nerves and Infect Peripheral Nerve Glial Cells. Front Cell Infect Microbiol 2021; 10:607779. [PMID: 33489937 PMCID: PMC7819965 DOI: 10.3389/fcimb.2020.607779] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Chlamydia pneumoniae can infect the brain and has been linked to late-onset dementia. Chlamydia muridarum, which infects mice, is often used to model human chlamydial infections. While it has been suggested to be also important for modelling brain infection, nervous system infection by C. muridarum has not been reported in the literature. C. pneumoniae has been shown to infect the olfactory bulb in mice after intranasal inoculation, and has therefore been suggested to invade the brain via the olfactory nerve; however, nerve infection has not been shown to date. Another path by which certain bacteria can reach the brain is via the trigeminal nerve, but it remains unknown whether Chlamydia species can infect this nerve. Other bacteria that can invade the brain via the olfactory and/or trigeminal nerve can do so rapidly, however, whether Chlamydia spp. can reach the brain earlier than one-week post inoculation remains unknown. In the current study, we showed that C. muridarum can within 48 h invade the brain via the olfactory nerve, in addition to infecting the trigeminal nerve. We also cultured the glial cells of the olfactory and trigeminal nerves and showed that C. muridarum readily infected the cells, constituting a possible cellular mechanism explaining how the bacteria can invade the nerves without being eliminated by glial immune functions. Further, we demonstrated that olfactory and trigeminal glia differed in their responses to C. muridarum, with olfactory glia showing less infection and stronger immune response than trigeminal glia.
Collapse
Affiliation(s)
- Lynn Nazareth
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Heidi Walkden
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Anu Chacko
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Ali Delbaz
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Todd Shelper
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Charles W Armitage
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ronak Reshamwala
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Logan K Trim
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - James A St John
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Kenneth W Beagley
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Jenny A K Ekberg
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| |
Collapse
|
34
|
A monoclonal antibody to Siglec-8 suppresses non-allergic airway inflammation and inhibits IgE-independent mast cell activation. Mucosal Immunol 2021; 14:366-376. [PMID: 32814824 PMCID: PMC7946634 DOI: 10.1038/s41385-020-00336-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/30/2020] [Accepted: 07/27/2020] [Indexed: 02/04/2023]
Abstract
In addition to their well characterized role in mediating IgE-dependent allergic diseases, aberrant accumulation and activation of mast cells (MCs) is associated with many non-allergic inflammatory diseases, whereby their activation is likely triggered by non-IgE stimuli (e.g., IL-33). Siglec-8 is an inhibitory receptor expressed on MCs and eosinophils that has been shown to inhibit IgE-mediated MC responses and reduce allergic inflammation upon ligation with a monoclonal antibody (mAb). Herein, we evaluated the effects of an anti-Siglec-8 mAb (anti-S8) in non-allergic disease models of experimental cigarette-smoke-induced chronic obstructive pulmonary disease and bleomycin-induced lung injury in Siglec-8 transgenic mice. Therapeutic treatment with anti-S8 inhibited MC activation and reduced recruitment of immune cells, airway inflammation, and lung fibrosis. Similarly, using a model of MC-dependent, IL-33-induced inflammation, anti-S8 treatment suppressed neutrophil influx, and cytokine production through MC inhibition. Transcriptomic profiling of MCs further demonstrated anti-S8-mediated downregulation of MC signaling pathways induced by IL-33, including TNF signaling via NF-κB. Collectively, these findings demonstrate that ligating Siglec-8 with an antibody reduces non-allergic inflammation and inhibits IgE-independent MC activation, supporting the evaluation of an anti-Siglec-8 mAb as a therapeutic approach in both allergic and non-allergic inflammatory diseases in which MCs play a role.
Collapse
|
35
|
Pinkerton JW, Kim RY, Koeninger L, Armbruster NS, Hansbro NG, Brown AC, Jayaraman R, Shen S, Malek N, Cooper MA, Nordkild P, Horvat JC, Jensen BAH, Wehkamp J, Hansbro PM. Human β-defensin-2 suppresses key features of asthma in murine models of allergic airways disease. Clin Exp Allergy 2020; 51:120-131. [PMID: 33098152 DOI: 10.1111/cea.13766] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 09/23/2020] [Accepted: 10/03/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Asthma is an airway inflammatory disease and a major health problem worldwide. Anti-inflammatory steroids and bronchodilators are the gold-standard therapy for asthma. However, they do not prevent the development of the disease, and critically, a subset of asthmatics are resistant to steroid therapy. OBJECTIVE To elucidate the therapeutic potential of human β-defensins (hBD), such as hBD2 mild to moderate and severe asthma. METHODS We investigated the role of hBD2 in a steroid-sensitive, house dust mite-induced allergic airways disease (AAD) model and a steroid-insensitive model combining ovalbumin-induced AAD with C muridarum (Cmu) respiratory infection. RESULTS In both models, we demonstrated that therapeutic intranasal application of hBD2 significantly reduced the influx of inflammatory cells into the bronchoalveolar lavage fluid. Furthermore, key type 2 asthma-related cytokines IL-9 and IL-13, as well as additional immunomodulating cytokines, were significantly decreased after administration of hBD2 in the steroid-sensitive model. The suppression of inflammation was associated with improvements in airway physiology and treatment also suppressed airway hyper-responsiveness (AHR) in terms of airway resistance and compliance to methacholine challenge. CONCLUSIONS AND CLINICAL RELEVANCE These data indicate that hBD2 reduces the hallmark features and has potential as a new therapeutic agent in allergic and especially steroid-resistant asthma.
Collapse
Affiliation(s)
- James W Pinkerton
- Priority Research Centre for Healthy Lungs, University of Newcastle, & Hunter Medical Research Institute, Newcastle, NSW, Australia.,National Heart & Lung Institute, Imperial College London, London, UK
| | - Richard Y Kim
- Priority Research Centre for Healthy Lungs, University of Newcastle, & Hunter Medical Research Institute, Newcastle, NSW, Australia.,Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Louis Koeninger
- Department of Internal Medicine, University of Tübingen, Tübingen, Germany
| | | | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, University of Newcastle, & Hunter Medical Research Institute, Newcastle, NSW, Australia.,Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Alexandra C Brown
- Priority Research Centre for Healthy Lungs, University of Newcastle, & Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Ranjith Jayaraman
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Sijie Shen
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Nisar Malek
- Department of Internal Medicine, University of Tübingen, Tübingen, Germany
| | - Matthew A Cooper
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Qld, Australia
| | - Peter Nordkild
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, University of Newcastle, & Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Benjamin A H Jensen
- Section for Human Genomics and Metagenomics in Metabolism, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jan Wehkamp
- Department of Internal Medicine, University of Tübingen, Tübingen, Germany
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, University of Newcastle, & Hunter Medical Research Institute, Newcastle, NSW, Australia.,Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
36
|
Li J, Zheng J, Wang M, Zhang Y, Jiang Y, Zhang X, Guo P. [Inhibition of CD96 enhances interferon-γ secretion by natural killer cells to alleviate lung injury in mice with pulmonary Chlamydia muridarum infection]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:930-935. [PMID: 32895152 DOI: 10.12122/j.issn.1673-4254.2020.07.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To assess the effect of neutralizing CD96 on natural killer (NK) cell functions in mice with pulmonary Chlamydia muridarum infection and explore the possible mechanism. METHODS Male BALB/c mice were randomly divided into infection group (Cm group), anti-CD96 treatment group (anti-CD96 group) and control group (n=5). In the former two groups, C. muridarum was inoculated via intranasal administration to establish mouse models of pulmonary C. muridarum infection, and the mice in the control group received intranasal administration of the inhalation buffer. In anti-CD96 group, the mice were injected with anti-CD96 antibody intraperitoneally at the dose of 250 μg every 3 days after the infection; the mice in Cm group received intraperitoneal injections of saline. The body weight of the mice was recorded daily. The mice were sacrificed 5 days after C. muridarum infection, and CD96 expression was detected by quantitative real-time PCR and Western blotting. HE staining and pathological scores were used to evaluate pneumonia of the mice. The inclusion body forming units (IFUs) were detected in the lung tissue homogenates to assess lung tissue chlamydia load. Flow cytometry and ELISA were used to assess the capacity of the lung NK cells to produce interferon-γ (IFN-γ) and regulate macrophages and Th1 cells. RESULTS C. muridarum infection inhibited CD96 expression in NK cells of the mice. Compared with those in Cm group, the mice in antiCD96 mice showed significantly milder lung inflammation (P < 0.05) and reduced chlamydia load in the lung tissue (P < 0.05). Neutralizing CD96 with anti-CD96 significantly enhanced IFN-γ secretion by the NK cells (P < 0.05) and augmented the immunoregulatory effect of the NK cells shown by enhanced responses of the lung macrophages (P < 0.05) and Th1 cells (P < 0.05). CONCLUSIONS Inhibition of CD96 alleviates pneumonia in C. muridarum-infected mice possibly by enhancing IFN-γ secretion by NK cells and augmenting the immunoregulatory effect of the NK cells on innate and adaptive immunity.
Collapse
Affiliation(s)
- Jing Li
- Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Jing Zheng
- Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Minda Wang
- Anhui Provincial Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233030, China
| | - Yan Zhang
- Anhui Provincial Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233030, China
| | - Yifan Jiang
- Anhui Provincial Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233030, China
| | - Xiaofeng Zhang
- Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Pu Guo
- Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| |
Collapse
|
37
|
Kavian N, Hachim A, Poon LLM, Valkenburg SA. Vaccination with ADCC activating HA peptide epitopes provides partial protection from influenza infection. Vaccine 2020; 38:5885-5890. [PMID: 32718818 PMCID: PMC7524583 DOI: 10.1016/j.vaccine.2020.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/11/2020] [Accepted: 07/06/2020] [Indexed: 12/17/2022]
Abstract
Influenza-specific antibody dependent cellular cytotoxicity (ADCC) antibodies have a broad cross reactivity and potential as an immune correlate for universal vaccines. Peptide-mapping for ADCC reactivity of H1-HA and H7-HA proteins from human serum samples identified high ADCC-inducing peptides in both the HA1 and HA2 regions. Vaccination of mice with single ADCC-peptides induced ADCC activity leading to partial protection from lethal influenza challenge, with increased survival, reduced viral loads, and reduced activation of NK cells in the lungs. Targeted vaccination strategies to elicit ADCC responses may provide an approach for universal vaccines.
Collapse
Affiliation(s)
- Niloufar Kavian
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region; Faculté de Médecine Université Paris Descartes, Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Universitaire Paris Centre, Centre Hospitalier Universitaire Cochin, Serviced'Immunologie Biologique, Paris, France; Institut Cochin, INSERM U1016, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Asmaa Hachim
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Leo L M Poon
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region; Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region.
| | - Sophie A Valkenburg
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
38
|
Galvão I, Kim RY, Shen S, Budden KF, Vieira AT, Hansbro PM. Emerging therapeutic targets and preclinical models for severe asthma. Expert Opin Ther Targets 2020; 24:845-857. [PMID: 32569487 DOI: 10.1080/14728222.2020.1786535] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Asthma is a heterogeneous disease with complex multifactorial causes. It is possible to subclassify asthma into different phenotypes that have distinct immunological features. Eosinophilic asthma is a well-known phenotype of severe asthma; however, a large body of clinical and experimental evidence strongly associates persistent airway inflammation, including the accumulation of neutrophils in the bronchial mucosa, and resistance to corticosteroid therapy and non-Type-2 immune responses with severe asthma. Importantly, mainstay therapies are often ineffective in severe asthma and effective alternatives are urgently needed. AREAS COVERED Here, we discussed recently developed mouse models of severe asthma that recapitulates key features of the disease in humans. We also provide findings from clinically relevant experimental models that have identified potential therapeutic targets for severe asthma. The most relevant publications on the topic of interest were selected from PubMed. EXPERT COMMENTARY Increasing the understanding of disease-causing mechanisms in severe asthma may lead to the identification of novel therapeutic targets and the development of more effective therapies. Intense research interest into investigating the pathophysiological mechanisms of severe asthma has driven the development and interrogation of a myriad of mouse models that aim to replicate hallmark features of severe asthma in humans.
Collapse
Affiliation(s)
- Izabela Galvão
- Centre for Inflammation, Centenary Institute and University of Technology Sydney , Sydney, Australia
| | - Richard Y Kim
- Centre for Inflammation, Centenary Institute and University of Technology Sydney , Sydney, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and the University of Newcastle , Newcastle, Australia
| | - Sijie Shen
- Centre for Inflammation, Centenary Institute and University of Technology Sydney , Sydney, Australia
| | - Kurtis F Budden
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and the University of Newcastle , Newcastle, Australia
| | - Angélica T Vieira
- Laboratory of Microbiota and Immunomodulation, Department of Biochemistry and Immunology, Instituto De Ciências Biológicas, Federal University of Minas Gerais , Belo Horizonte, Brazil
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney , Sydney, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and the University of Newcastle , Newcastle, Australia
| |
Collapse
|
39
|
de Almeida AD, Silva IS, Fernandes-Braga W, LimaFilho ACM, Florentino ROM, Barra A, de Oliveira Andrade L, Leite MF, Cassali GD, Klein A. A role for mast cells and mast cell tryptase in driving neutrophil recruitment in LPS-induced lung inflammation via protease-activated receptor 2 in mice. Inflamm Res 2020; 69:1059-1070. [PMID: 32632517 DOI: 10.1007/s00011-020-01376-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/04/2020] [Accepted: 07/02/2020] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE This study aims to investigate the role of protease-activated receptor (PAR) 2 and mast cell (MC) tryptase in LPS-induced lung inflammation and neutrophil recruitment in the lungs of C57BL/6 mice. METHODS C57BL/6 mice were pretreated with the PAR2 antagonist ENMD-1068, compound 48/80 or aprotinin prior to intranasal instillation of MC tryptase or LPS. Blood leukocytes, C-X-C motif chemokine ligand (CXCL) 1 production leukocytes recovered from bronchoalveolar lavage fluid (BALF), and histopathological analysis of the lung were evaluated 4 h later. Furthermore, we performed experiments to determine intracellular calcium signaling in RAW 264.7 cells stimulated with LPS in the presence or absence of a protease inhibitor cocktail or ENMD-1068 and evaluated PAR2 expression in the lungs of LPS-treated mice. RESULTS Pharmacological blockade of PAR2 or inhibition of proteases reduced neutrophils recovered in BALF and LPS-induced calcium signaling. PAR2 blockade impaired LPS-induced lung inflammation, PAR2 expression in the lung and CXCL1 release in BALF, and increased circulating blood neutrophils. Intranasal instillation of MC tryptase increased the number of neutrophils recovered in BALF, and MC depletion with compound 48/80 impaired LPS-induced neutrophil migration. CONCLUSION Our study provides, for the first time, evidence of a pivotal role for MCs and MC tryptase in neutrophil migration, lung inflammation and macrophage activation triggered by LPS, by a mechanism dependent on PAR2 activation.
Collapse
Affiliation(s)
- Aline Dias de Almeida
- Laboratory of Pain and Inflammation, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Presidente Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-010, Brazil
| | - Irismara Sousa Silva
- Laboratory of Pain and Inflammation, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Presidente Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-010, Brazil
| | - Weslley Fernandes-Braga
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Antônio Carlos Melo LimaFilho
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - R Odrigo Machado Florentino
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ayslan Barra
- Laboratory of Pain and Inflammation, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Presidente Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-010, Brazil
| | - Luciana de Oliveira Andrade
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - M Fátima Leite
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Geovanni Dantas Cassali
- Department of Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - André Klein
- Laboratory of Pain and Inflammation, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Presidente Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-010, Brazil.
| |
Collapse
|
40
|
Wang H, Li J, Dong X, Zhou X, Zhao L, Wang X, Rashu R, Zhao W, Yang X. NK Cells Contribute to Protective Memory T Cell Mediated Immunity to Chlamydia muridarum Infection. Front Cell Infect Microbiol 2020; 10:296. [PMID: 32626664 PMCID: PMC7311576 DOI: 10.3389/fcimb.2020.00296] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/19/2020] [Indexed: 12/23/2022] Open
Abstract
We previously reported that NK cells can promote type 1 T cell immune response that is essential for protection to a primary infection of Chlamydia muridarum. In this study, we have investigated the contribution of NK cells to memory T cells associated immunity during chlamydial infection. We have found that NK cell depletion led to impaired production of IFN-γ by memory T cells upon re-stimulation with chlamydial antigens in vitro. Mice with depleted NK cells also exhibited reduced type 1 T cell recall responses, with increased production of IL-4 from CD4+ T cells and a lower level of Chlamydia-specific IgG2a titers compared to control mice. In addition, Tregs response was significantly increased in mice with NK cell depletion. Moreover, NK cell-depleted mice showed an increased bacterial loads and more severe inflammatory pathological changes than control mice. These findings indicate that NK cells contribute to protective memory T cell associated immunity to chlamydial re-infection through modulating the cytokine pattern of T cell and inhibition of Tregs response.
Collapse
Affiliation(s)
- Hong Wang
- Department of Pathogenic Biology & Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Jing Li
- Department of Pathogenic Biology & Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiaojing Dong
- Department of Pathogenic Biology & Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xaoqing Zhou
- Department of Pathogenic Biology & Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Lei Zhao
- Institute of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao Wang
- Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Rasheduzzaman Rashu
- Department of Immunology and Department of Medical Microbiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Weiming Zhao
- Department of Pathogenic Biology & Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xi Yang
- Department of Immunology and Department of Medical Microbiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
41
|
Ali MK, Kim RY, Brown AC, Donovan C, Vanka KS, Mayall JR, Liu G, Pillar AL, Jones-Freeman B, Xenaki D, Borghuis T, Karim R, Pinkerton JW, Aryal R, Heidari M, Martin KL, Burgess JK, Oliver BG, Trinder D, Johnstone DM, Milward EA, Hansbro PM, Horvat JC. Critical role for iron accumulation in the pathogenesis of fibrotic lung disease. J Pathol 2020; 251:49-62. [PMID: 32083318 DOI: 10.1002/path.5401] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 12/19/2019] [Accepted: 02/13/2020] [Indexed: 12/18/2022]
Abstract
Increased iron levels and dysregulated iron homeostasis, or both, occur in several lung diseases. Here, the effects of iron accumulation on the pathogenesis of pulmonary fibrosis and associated lung function decline was investigated using a combination of murine models of iron overload and bleomycin-induced pulmonary fibrosis, primary human lung fibroblasts treated with iron, and histological samples from patients with or without idiopathic pulmonary fibrosis (IPF). Iron levels are significantly increased in iron overloaded transferrin receptor 2 (Tfr2) mutant mice and homeostatic iron regulator (Hfe) gene-deficient mice and this is associated with increases in airway fibrosis and reduced lung function. Furthermore, fibrosis and lung function decline are associated with pulmonary iron accumulation in bleomycin-induced pulmonary fibrosis. In addition, we show that iron accumulation is increased in lung sections from patients with IPF and that human lung fibroblasts show greater proliferation and cytokine and extracellular matrix responses when exposed to increased iron levels. Significantly, we show that intranasal treatment with the iron chelator, deferoxamine (DFO), from the time when pulmonary iron levels accumulate, prevents airway fibrosis and decline in lung function in experimental pulmonary fibrosis. Pulmonary fibrosis is associated with an increase in Tfr1+ macrophages that display altered phenotype in disease, and DFO treatment modified the abundance of these cells. These experimental and clinical data demonstrate that increased accumulation of pulmonary iron plays a key role in the pathogenesis of pulmonary fibrosis and lung function decline. Furthermore, these data highlight the potential for the therapeutic targeting of increased pulmonary iron in the treatment of fibrotic lung diseases such as IPF. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Md Khadem Ali
- Division of Pulmonary and Critical Care Medicine, School of Medicine, Stanford University, Stanford, CA, USA.,Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Richard Y Kim
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia
| | - Alexandra C Brown
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia
| | - Kanth S Vanka
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Jemma R Mayall
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Gang Liu
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia
| | - Amber L Pillar
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Bernadette Jones-Freeman
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Dikaia Xenaki
- Woolcock Institute of Medical Research, University of Sydney and School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Theo Borghuis
- Department of Pathology and Medical Biology, Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Rafia Karim
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - James W Pinkerton
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Respiratory Pharmacology & Toxicology Group, National Heart & Lung Institute, Imperial College London, London, UK
| | - Ritambhara Aryal
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Brain and Mental Health and School of Biomedical Sciences, University of Newcastle, Newcastle, Australia
| | - Moones Heidari
- Priority Research Centre for Brain and Mental Health and School of Biomedical Sciences, University of Newcastle, Newcastle, Australia
| | - Kristy L Martin
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Brain and Mental Health and School of Biomedical Sciences, University of Newcastle, Newcastle, Australia
| | - Janette K Burgess
- Woolcock Institute of Medical Research, University of Sydney and School of Life Sciences, University of Technology Sydney, Sydney, Australia.,Department of Pathology and Medical Biology, Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Brian G Oliver
- Woolcock Institute of Medical Research, University of Sydney and School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Debbie Trinder
- Medical School and, Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Australia
| | - Daniel M Johnstone
- Discipline of Physiology and Bosch Institute, University of Sydney, Sydney, Australia
| | - Elizabeth A Milward
- Priority Research Centre for Brain and Mental Health and School of Biomedical Sciences, University of Newcastle, Newcastle, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| |
Collapse
|
42
|
Woods JJ, Skelding KA, Martin KL, Aryal R, Sontag E, Johnstone DM, Horvat JC, Hansbro PM, Milward EA. Assessment of evidence for or against contributions of Chlamydia pneumoniae infections to Alzheimer's disease etiology. Brain Behav Immun 2020; 83:22-32. [PMID: 31626972 DOI: 10.1016/j.bbi.2019.10.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/26/2019] [Accepted: 10/14/2019] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease, the most common form of dementia, was first formally described in 1907 yet its etiology has remained elusive. Recent proposals that Aβ peptide may be part of the brain immune response have revived longstanding contention about the possibility of causal relationships between brain pathogens and Alzheimer's disease. Research has focused on infectious pathogens that may colonize the brain such as herpes simplex type I. Some researchers have proposed the respiratory bacteria Chlamydia pneumoniae may also be implicated in Alzheimer's disease, however this remains controversial. This review aims to provide a balanced overview of the current evidence and its limitations and future approaches that may resolve controversies. We discuss the evidence from in vitro, animal and human studies proposed to implicate Chlamydia pneumoniae in Alzheimer's disease and other neurological conditions, the potential mechanisms by which the bacterium may contribute to pathogenesis and limitations of previous studies that may explain the inconsistencies in the literature.
Collapse
Affiliation(s)
- Jason J Woods
- School of Biomedical Sciences and Pharmacy, University Drive, University of Newcastle, Callaghan NSW 2308, Australia.
| | - Kathryn A Skelding
- School of Biomedical Sciences and Pharmacy, University Drive, University of Newcastle, Callaghan NSW 2308, Australia
| | - Kristy L Martin
- School of Biomedical Sciences and Pharmacy, University Drive, University of Newcastle, Callaghan NSW 2308, Australia; Discipline of Physiology and Bosch Institute, Anderson Stuart Building F13, University of Sydney, NSW 2006, Australia
| | - Ritambhara Aryal
- School of Biomedical Sciences and Pharmacy, University Drive, University of Newcastle, Callaghan NSW 2308, Australia
| | - Estelle Sontag
- School of Biomedical Sciences and Pharmacy, University Drive, University of Newcastle, Callaghan NSW 2308, Australia
| | - Daniel M Johnstone
- Discipline of Physiology and Bosch Institute, Anderson Stuart Building F13, University of Sydney, NSW 2006, Australia
| | - Jay C Horvat
- Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton Heights NSW 2305, Australia
| | - Philip M Hansbro
- School of Biomedical Sciences and Pharmacy, University Drive, University of Newcastle, Callaghan NSW 2308, Australia; Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton Heights NSW 2305, Australia; Centre for Inflammation, Centenary Institute, Camperdown NSW 2050, Australia; Centre for Inflammation, Faculty of Science, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Elizabeth A Milward
- School of Biomedical Sciences and Pharmacy, University Drive, University of Newcastle, Callaghan NSW 2308, Australia
| |
Collapse
|
43
|
Wadhwa R, Dua K, Adcock IM, Horvat JC, Kim RY, Hansbro PM. Cellular mechanisms underlying steroid-resistant asthma. Eur Respir Rev 2019; 28:28/153/190096. [PMID: 31636089 DOI: 10.1183/16000617.0096-2019] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 09/19/2019] [Indexed: 01/04/2023] Open
Abstract
Severe steroid-resistant asthma is clinically important, as patients with this form of the disease do not respond to mainstay corticosteroid therapies. The heterogeneity of this form of asthma and poor understanding of the pathological mechanisms involved hinder the identification of therapeutic targets and the development of more effective therapies. A major limiting factor in the understanding of severe steroid-resistant asthma is the existence of multiple endotypes represented by different immunological and inflammatory phenotypes, particularly in adults. Several clinical and experimental studies have revealed associations between specific respiratory infections and steroid-resistant asthma in adults. Here, we discuss recent findings from other authors as well as our own studies that have developed novel experimental models for interrogating the association between respiratory infections and severe steroid-resistant asthma. These models have enabled the identification of new therapies using macrolides, as well as several novel disease mechanisms, including the microRNA-21/phosphoinositide 3-kinase/histone deacetylase 2 axis and NLRP3 inflammasomes, and highlight the potential of these mechanisms as therapeutic targets.
Collapse
Affiliation(s)
- Ridhima Wadhwa
- Centre for Inflammation, Centenary Institute, Sydney, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, Australia.,Both authors contributed equally
| | - Kamal Dua
- Centre for Inflammation, Centenary Institute, Sydney, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,Both authors contributed equally
| | - Ian M Adcock
- The Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Richard Y Kim
- Centre for Inflammation, Centenary Institute, Sydney, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,Faculty of Science, University of Technology Sydney, Sydney, Australia
| |
Collapse
|
44
|
Hadjigol S, Netto KG, Maltby S, Tay HL, Nguyen TH, Hansbro NG, Eyers F, Hansbro PM, Yang M, Foster PS. Lipopolysaccharide induces steroid-resistant exacerbations in a mouse model of allergic airway disease collectively through IL-13 and pulmonary macrophage activation. Clin Exp Allergy 2019; 50:82-94. [PMID: 31579973 DOI: 10.1111/cea.13505] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/26/2019] [Accepted: 09/15/2019] [Indexed: 01/09/2023]
Abstract
BACKGROUND Acute exacerbations of asthma represent a major burden of disease and are often caused by respiratory infections. Viral infections are recognized as significant triggers of exacerbations; however, less is understood about the how microbial bioproducts such as the endotoxin (lipopolysaccharide (LPS)) trigger episodes. Indeed, increased levels of LPS have been linked to asthma onset, severity and steroid resistance. OBJECTIVE The goal of this study was to identify mechanisms underlying bacterial-induced exacerbations by employing LPS as a surrogate for infection. METHODS We developed a mouse model of LPS-induced exacerbation on the background of pre-existing type-2 allergic airway disease (AAD). RESULTS LPS-induced exacerbation was characterized by steroid-resistant airway hyperresponsiveness (AHR) and an exaggerated inflammatory response distinguished by increased numbers of infiltrating neutrophils/macrophages and elevated production of lung inflammatory cytokines, including TNFα, IFNγ, IL-27 and MCP-1. Expression of the type-2 associated inflammatory factors such as IL-5 and IL-13 were elevated in AAD but not altered by LPS exposure. Furthermore, AHR and airway inflammation were no longer suppressed by corticosteroid (dexamethasone) treatment after LPS exposure. Depletion of pulmonary macrophages by administration of 2-chloroadenosine into the lungs suppressed AHR and reduced IL-13, TNFα and IFNγ expression. Blocking IL-13 function, through either IL-13-deficiency or administration of specific blocking antibodies, also suppressed AHR and airway inflammation. CONCLUSIONS & CLINICAL RELEVANCE We present evidence that IL-13 and innate immune pathways (in particular pulmonary macrophages) contribute to LPS-induced exacerbation of pre-existing AAD and provide insight into the complex molecular processes potentially underlying microbial-induced exacerbations.
Collapse
Affiliation(s)
- Sara Hadjigol
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Keilah G Netto
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Steven Maltby
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Hock L Tay
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Thi H Nguyen
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Fiona Eyers
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Ming Yang
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
45
|
Shukla SD, Shastri MD, Chong WC, Dua K, Budden KF, Mahmood MQ, Hansbro NG, Keely S, Eri R, Patel RP, Peterson GM, Hansbro PM. Microbiome-focused asthma management strategies. Curr Opin Pharmacol 2019; 46:143-149. [PMID: 31357048 DOI: 10.1016/j.coph.2019.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/24/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022]
Abstract
Asthma is a common, heterogeneous and serious disease with high prevalence globally. Poorly controlled, steroid-resistant asthma is particularly important as there are no effective therapies and it exerts substantial healthcare and societal burden. The role of microbiomes, particularly in chronic diseases has generated considerable interest in recent times. Existing evidence clearly demonstrates an association between asthma initiation and the microbiome, both respiratory and gastro-intestinal, although its' roles are poorly understood when assessing the asthma progression or heterogeneity (i.e. phenotypes/endotypes) across different geographical locations. Moreover, modulating microbiomes could be preventive and/or therapeutic in patients with asthma warrants urgent attention. Here, we review recent advances in assessing the role of microbiomes in asthma and present the challenges associated with the potential therapeutic utility of modifying microbiomes in management.
Collapse
Affiliation(s)
- Shakti D Shukla
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia
| | - Madhur D Shastri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia
| | - Wai Chin Chong
- Department of Molecular and Translational Science, Monash University, Clayton, Australia; Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Australia
| | - Kamal Dua
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia
| | - Kurtis F Budden
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia
| | - Malik Quasir Mahmood
- Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia; Centre for inflammation, Centenary Institute, Sydney, and School of Life Sciences, University of Technology, Ultimo, NSW, Australia
| | - Simon Keely
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia
| | - Rajaraman Eri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia
| | - Rahul P Patel
- Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Gregory M Peterson
- Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia; Centre for inflammation, Centenary Institute, Sydney, and School of Life Sciences, University of Technology, Ultimo, NSW, Australia.
| |
Collapse
|
46
|
Starkey MR, Plank MW, Casolari P, Papi A, Pavlidis S, Guo Y, Cameron GJM, Haw TJ, Tam A, Obiedat M, Donovan C, Hansbro NG, Nguyen DH, Nair PM, Kim RY, Horvat JC, Kaiko GE, Durum SK, Wark PA, Sin DD, Caramori G, Adcock IM, Foster PS, Hansbro PM. IL-22 and its receptors are increased in human and experimental COPD and contribute to pathogenesis. Eur Respir J 2019; 54:1800174. [PMID: 31196943 PMCID: PMC8132110 DOI: 10.1183/13993003.00174-2018] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/19/2019] [Indexed: 12/24/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of morbidity and death globally. The lack of effective treatments results from an incomplete understanding of the underlying mechanisms driving COPD pathogenesis.Interleukin (IL)-22 has been implicated in airway inflammation and is increased in COPD patients. However, its roles in the pathogenesis of COPD is poorly understood. Here, we investigated the role of IL-22 in human COPD and in cigarette smoke (CS)-induced experimental COPD.IL-22 and IL-22 receptor mRNA expression and protein levels were increased in COPD patients compared to healthy smoking or non-smoking controls. IL-22 and IL-22 receptor levels were increased in the lungs of mice with experimental COPD compared to controls and the cellular source of IL-22 included CD4+ T-helper cells, γδ T-cells, natural killer T-cells and group 3 innate lymphoid cells. CS-induced pulmonary neutrophils were reduced in IL-22-deficient (Il22 -/-) mice. CS-induced airway remodelling and emphysema-like alveolar enlargement did not occur in Il22 -/- mice. Il22 -/- mice had improved lung function in terms of airway resistance, total lung capacity, inspiratory capacity, forced vital capacity and compliance.These data highlight important roles for IL-22 and its receptors in human COPD and CS-induced experimental COPD.
Collapse
Affiliation(s)
- Malcolm R Starkey
- Priority Research Centres GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
| | - Maximilian W Plank
- Priority Research Centres GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
| | - Paolo Casolari
- Interdepartmental Study Center for Inflammatory and Smoke-related Airway Diseases (CEMICEF), Cardiorespiratory and Internal Medicine Section, University of Ferrara, Ferrara, Italy
| | - Alberto Papi
- Interdepartmental Study Center for Inflammatory and Smoke-related Airway Diseases (CEMICEF), Cardiorespiratory and Internal Medicine Section, University of Ferrara, Ferrara, Italy
| | - Stelios Pavlidis
- The Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Yike Guo
- The Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Guy J M Cameron
- Priority Research Centres GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
| | - Tatt Jhong Haw
- Priority Research Centres GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
| | - Anthony Tam
- The University of British Columbia Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada
- Respiratory Division, Dept of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Ma'en Obiedat
- The University of British Columbia Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada
- Respiratory Division, Dept of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Chantal Donovan
- Priority Research Centres GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
| | - Nicole G Hansbro
- Priority Research Centres GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
- Centre for inflammation, Centenary Institute, Sydney, Australia
- School of Life Sciences, University of Technology, Ultimo, Australia
| | - Duc H Nguyen
- Priority Research Centres GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
| | - Prema Mono Nair
- Priority Research Centres GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
| | - Richard Y Kim
- Priority Research Centres GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
| | - Jay C Horvat
- Priority Research Centres GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
| | - Gerard E Kaiko
- Priority Research Centres GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
| | - Scott K Durum
- Laboratory of Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Peter A Wark
- Priority Research Centres GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
| | - Don D Sin
- The University of British Columbia Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada
- Respiratory Division, Dept of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gaetano Caramori
- UOC di Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Ian M Adcock
- The Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Paul S Foster
- Priority Research Centres GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
| | - Philip M Hansbro
- Priority Research Centres GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, Australia
- Centre for inflammation, Centenary Institute, Sydney, Australia
- School of Life Sciences, University of Technology, Ultimo, Australia
| |
Collapse
|
47
|
Loering S, Cameron GJM, Starkey MR, Hansbro PM. Lung development and emerging roles for type 2 immunity. J Pathol 2019; 247:686-696. [PMID: 30506724 DOI: 10.1002/path.5211] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/06/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
Lung development is a complex process mediated through the interaction of multiple cell types, factors and mediators. In mice, it starts as early as embryonic day 9 and continues into early adulthood. The process can be separated into five different developmental stages: embryonic, pseudoglandular, canalicular, saccular, and alveolar. Whilst lung bud formation and branching morphogenesis have been studied extensively, the mechanisms of alveolarisation are incompletely understood. Aberrant lung development can lead to deleterious consequences for respiratory health such as bronchopulmonary dysplasia (BPD), a disease primarily affecting preterm neonates, which is characterised by increased pulmonary inflammation and disturbed alveolarisation. While the deleterious effects of type 1-mediated inflammatory responses on lung development have been well established, the role of type 2 responses in postnatal lung development remains poorly understood. Recent studies indicate that type 2-associated immune cells, such as group 2 innate lymphoid cells and alveolar macrophages, are increased in number during postnatal alveolarisation. Here, we present the current state of understanding of the postnatal stages of lung development and the key cell types and mediators known to be involved. We also provide an overview of how stem cells are involved in lung development and regeneration, and the negative influences of respiratory infections. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Svenja Loering
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Guy J M Cameron
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Malcolm R Starkey
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Center for Inflammation, Centenary Institute and The School of Life Sciences, University of Technology, Sydney, New South Wales, Australia
| |
Collapse
|
48
|
Neonatal Streptococcus pneumoniae Pneumonia Induces an Aberrant Airway Smooth Muscle Phenotype and AHR in Mice Model. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1948519. [PMID: 30723734 PMCID: PMC6339730 DOI: 10.1155/2019/1948519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/12/2018] [Accepted: 12/23/2018] [Indexed: 12/21/2022]
Abstract
Our previous study showed that neonatal S. pneumoniae infection aggravated airway inflammation and airway hyperresponsiveness (AHR) in an OVA-induced allergic asthma model. As airway smooth muscle (ASM) plays a pivotal role in AHR development, we aim to investigate the effects of neonatal S. pneumoniae pneumonia on ASM structure and AHR development. Non-lethal neonatal pneumonia was established by intranasally infecting 1-week-old BALB/C mice with the S. pneumoniae strain D39. Five weeks after infection, the lungs were collected to assess the levels of α-SMA and the contractile proteins of ASM. Our results indicate that neonatal S. pneumoniae pneumonia significantly increased adulthood lung α-SMA and SMMHC proteins production and aggravated airway inflammatory cells infiltration and cytokines release. In addition, the neonatal S. pneumoniae pneumonia group had significantly higher Penh values compared to the uninfected controls. These data suggest that neonatal S. pneumoniae pneumonia promoted an aberrant ASM phenotype and AHR development in mice model.
Collapse
|
49
|
Campa CC, Silva RL, Margaria JP, Pirali T, Mattos MS, Kraemer LR, Reis DC, Grosa G, Copperi F, Dalmarco EM, Lima-Júnior RCP, Aprile S, Sala V, Dal Bello F, Prado DS, Alves-Filho JC, Medana C, Cassali GD, Tron GC, Teixeira MM, Ciraolo E, Russo RC, Hirsch E. Inhalation of the prodrug PI3K inhibitor CL27c improves lung function in asthma and fibrosis. Nat Commun 2018; 9:5232. [PMID: 30542075 PMCID: PMC6290777 DOI: 10.1038/s41467-018-07698-6] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 11/14/2018] [Indexed: 12/11/2022] Open
Abstract
PI3K activation plays a central role in the development of pulmonary inflammation and tissue remodeling. PI3K inhibitors may thus offer an improved therapeutic opportunity to treat non-resolving lung inflammation but their action is limited by unwanted on-target systemic toxicity. Here we present CL27c, a prodrug pan-PI3K inhibitor designed for local therapy, and investigate whether inhaled CL27c is effective in asthma and pulmonary fibrosis. Mice inhaling CL27c show reduced insulin-evoked Akt phosphorylation in lungs, but no change in other tissues and no increase in blood glycaemia, in line with a local action. In murine models of acute or glucocorticoid-resistant neutrophilic asthma, inhaled CL27c reduces inflammation and improves lung function. Finally, inhaled CL27c administered in a therapeutic setting protects from bleomycin-induced lung fibrosis, ultimately leading to significantly improved survival. Therefore, local delivery of a pan-PI3K inhibitor prodrug reduces systemic on-target side effects but effectively treats asthma and irreversible pulmonary fibrosis. Activation of PI3K plays a role in pulmonary inflammation. Here, the authors develop a drug inhibitor of PI3K, and show that it inhibits lung inflammation and damage in mouse models of asthma and lung fibrosis.
Collapse
Affiliation(s)
- Carlo C Campa
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Rangel L Silva
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, 14049-900, Ribeirao Preto, Brazil
| | - Jean P Margaria
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Tracey Pirali
- Dipartimento di Scienze del Farmaco, Università degli Studi del Piemonte Orientale "A. Avogadro", Largo Donegani 2, 28100, Novara, Italy
| | - Matheus S Mattos
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais/UFMG, Avenida Antonio Carlos 6627, Belo Horizonte, 31270-901, Brazil
| | - Lucas R Kraemer
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais/UFMG, Avenida Antonio Carlos 6627, Belo Horizonte, 31270-901, Brazil
| | - Diego C Reis
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais/UFMG, Avenida Antonio Carlos 6627, Belo Horizonte, 31270-901, Brazil.,Laboratory of Comparative Pathology, Department of General Pathology Institute of Biological Sciences, Universidade Federal de Minas Gerais/UFMG, Avenida Antonio Carlos 6627, Belo Horizonte, 31270-901, Brazil
| | - Giorgio Grosa
- Dipartimento di Scienze del Farmaco, Università degli Studi del Piemonte Orientale "A. Avogadro", Largo Donegani 2, 28100, Novara, Italy
| | - Francesca Copperi
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Eduardo M Dalmarco
- Department of Clinical Analysis, Universidade Federal de Santa Catarina/UFSC, Rua Delfino Conti, S/N, Florianopolis, 88040-370, Brazil
| | - Roberto C P Lima-Júnior
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy.,Laboratory of Pharmacology of Inflammation and Cancer, Department of Physiology and Pharmacology, Universidade Federal do Ceará/UFC, Rua Cel Nunes de Melo 1127, Fortaleza, 60430-270, Brazil
| | - Silvio Aprile
- Dipartimento di Scienze del Farmaco, Università degli Studi del Piemonte Orientale "A. Avogadro", Largo Donegani 2, 28100, Novara, Italy
| | - Valentina Sala
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Federica Dal Bello
- Department of Molecular Biotechnology and Health Sciences, Mass Spectrometry Unit, University of Torino, Via Giuria 5, 10125, Torino, Italy
| | - Douglas Silva Prado
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, 14049-900, Ribeirao Preto, Brazil
| | - Jose Carlos Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, 14049-900, Ribeirao Preto, Brazil
| | - Claudio Medana
- Department of Molecular Biotechnology and Health Sciences, Mass Spectrometry Unit, University of Torino, Via Giuria 5, 10125, Torino, Italy
| | - Geovanni D Cassali
- Laboratory of Comparative Pathology, Department of General Pathology Institute of Biological Sciences, Universidade Federal de Minas Gerais/UFMG, Avenida Antonio Carlos 6627, Belo Horizonte, 31270-901, Brazil
| | - Gian Cesare Tron
- Dipartimento di Scienze del Farmaco, Università degli Studi del Piemonte Orientale "A. Avogadro", largo Donegani 2, 28100, Novara, Italy.,Kither Biotech S.r.l., Via Nizza 52, 10126, Torino, Italy
| | - Mauro M Teixeira
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais/UFMG, Avenida Antonio Carlos 6627, Belo Horizonte, 31270-901, Brazil
| | - Elisa Ciraolo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy. .,Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany.
| | - Remo C Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais/UFMG, Avenida Antonio Carlos 6627, Belo Horizonte, 31270-901, Brazil.,Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais/UFMG, Avenida Antonio Carlos 6627, Belo Horizonte, 31270-901, Brazil
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy.,Kither Biotech S.r.l., Via Nizza 52, 10126, Torino, Italy
| |
Collapse
|
50
|
Kim KH, Lee S, Lee H, Shin D, Min D, Kim M, Ryu B, Kim HW, Bae H. A standardized herbal extract PM014 ameliorates pulmonary fibrosis by suppressing the TGF-β1 pathway. Sci Rep 2018; 8:16860. [PMID: 30443024 PMCID: PMC6237877 DOI: 10.1038/s41598-018-35320-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 11/01/2018] [Indexed: 12/13/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating and common chronic lung disease pathologically characterized by loss of epithelial cells and activation of fibroblasts and myofibroblasts. The etiology of IPF remains unclear and the disease pathology is poorly understood with no known efficacious therapy. PM014 is an herbal extract that has been shown to have beneficial effects in pulmonary diseases, which are likely to exert anti-inflammatory bioactions. In the present study, we observed that bleomycin (BLM) caused increased inflammatory infiltration as well as collagen deposition in lungs of mice on day 14 after treatment. Administration of PM014 suppressed BLM-induced inflammatory responses and fibrotic changes in dose-dependent manner in mice. Additionally, we provided in vitro evidence suggesting that PM014 inhibited TGF-β1-induced epithelial-mesenchymal transition (EMT) and fibroblast activation in alveolar epithelial cells and human lung fibroblasts from healthy donor and IPF patients. PM014 appeared to target TGF-β1 signaling via Smad-dependent pathways and p38 mitogen-activated protein kinases (MAPKs) pathways. Taken together, our data suggest that PM014 administration exerts a protective effect against lung fibrosis and highlight PM014 as a viable treatment option that may bring benefits to patient with IPF.
Collapse
Affiliation(s)
- Kyung Hwa Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, South Korea
| | - Sujin Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, South Korea
| | - Hyunji Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, South Korea
| | - Dasom Shin
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, South Korea
| | - Daeun Min
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, South Korea
| | - Miran Kim
- Central Research Institute, Hanlim Pharm. Co. Ltd., Yongin, 17040, South Korea
| | - Byeol Ryu
- College of Pharmacy and Research Institute of Pharmaceutical Science, Seoul National University, Seoul, 08826, South Korea
| | - Hyeon Woo Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science, Seoul National University, Seoul, 08826, South Korea
| | - Hyunsu Bae
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, South Korea.
| |
Collapse
|