1
|
Mayall JR, Horvat JC, Mangan NE, Chevalier A, McCarthy H, Hampsey D, Donovan C, Brown AC, Matthews AY, de Weerd NA, de Geus ED, Starkey MR, Kim RY, Daly K, Goggins BJ, Keely S, Maltby S, Baldwin R, Foster PS, Boyle MJ, Tanwar PS, Huntington ND, Hertzog PJ, Hansbro PM. Interferon-epsilon is a novel regulator of NK cell responses in the uterus. EMBO Mol Med 2024; 16:267-293. [PMID: 38263527 PMCID: PMC10897320 DOI: 10.1038/s44321-023-00018-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 01/25/2024] Open
Abstract
The uterus is a unique mucosal site where immune responses are balanced to be permissive of a fetus, yet protective against infections. Regulation of natural killer (NK) cell responses in the uterus during infection is critical, yet no studies have identified uterine-specific factors that control NK cell responses in this immune-privileged site. We show that the constitutive expression of IFNε in the uterus plays a crucial role in promoting the accumulation, activation, and IFNγ production of NK cells in uterine tissue during Chlamydia infection. Uterine epithelial IFNε primes NK cell responses indirectly by increasing IL-15 production by local immune cells and directly by promoting the accumulation of a pre-pro-like NK cell progenitor population and activation of NK cells in the uterus. These findings demonstrate the unique features of this uterine-specific type I IFN and the mechanisms that underpin its major role in orchestrating innate immune cell protection against uterine infection.
Collapse
Affiliation(s)
- Jemma R Mayall
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Jay C Horvat
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Niamh E Mangan
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Departments of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Anne Chevalier
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Huw McCarthy
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Daniel Hampsey
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Chantal Donovan
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2000, Australia
| | - Alexandra C Brown
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Antony Y Matthews
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Departments of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Nicole A de Weerd
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Departments of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Eveline D de Geus
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Departments of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Malcolm R Starkey
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
- Immunology and Pathology, Central Clinical School, Monash University, Clayton, VIC, 3168, Australia
| | - Richard Y Kim
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2000, Australia
| | - Katie Daly
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Bridie J Goggins
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Simon Keely
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Steven Maltby
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Rennay Baldwin
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Paul S Foster
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Michael J Boyle
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia
- Immunology and Infectious Diseases Unit, John Hunter Hospital, Newcastle, NSW, 2305, Australia
| | - Pradeep S Tanwar
- Gynecology Oncology Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Nicholas D Huntington
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3168, Australia
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Departments of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Philip M Hansbro
- Immune Health Program, Hunter Medical Research Institute and the University of Newcastle, Newcastle, NSW, 2308, Australia.
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2000, Australia.
| |
Collapse
|
2
|
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
|
3
|
Vanders RL, Gomez HM, Hsu AC, Daly K, Wark PAB, Horvat JC, Hansbro PM. Inflammatory and antiviral responses to influenza A virus infection are dysregulated in pregnant mice with allergic airway disease. Am J Physiol Lung Cell Mol Physiol 2023; 325:L385-L398. [PMID: 37463835 DOI: 10.1152/ajplung.00232.2022] [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: 07/24/2022] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/20/2023] Open
Abstract
Influenza A virus (IAV) infections are increased during pregnancy especially with asthma as a comorbidity, leading to asthma exacerbations, secondary bacterial infections, intensive care unit admissions, and mortality. We aimed to define the processes involved in increased susceptibility and severity of IAV infections during pregnancy, especially with asthma. We sensitized mice to house dust mite (HDM), induced pregnancy, and challenged with HDM to induce allergic airway disease (AAD). At midpregnancy, we induced IAV infection. We assessed viral titers, airway inflammation, lung antiviral responses, mucus hypersecretion, and airway hyperresponsiveness (AHR). During early IAV infection, pregnant mice with AAD had increased mRNA expression of the inflammatory markers Il13 and IL17 and reduced mRNA expression of the neutrophil chemoattractant marker Kc. These mice had increased mucous hyperplasia and increased AHR. miR155, miR574, miR223, and miR1187 were also reduced during early infection, as was mRNA expression of the antiviral β-defensins, Bd1, Bd2, and Spd and IFNs, Ifnα, Ifnβ, and Ifnλ. During late infection, Il17 was still increased as was eosinophil infiltration in the lungs. mRNA expression of Kc was reduced, as was neutrophil infiltration and mRNA expression of the antiviral markers Ifnβ, Ifnλ, and Ifnγ and Ip10, Tlr3, Tlr9, Pkr, and Mx1. Mucous hyperplasia was still significantly increased as was AHR. Early phase IAV infection in pregnancy with asthma heightens underlying inflammatory asthmatic phenotype and reduces antiviral responses.NEW & NOTEWORTHY Influenza A virus (IAV) infection during pregnancy with asthma is a major health concern leading to increased morbidity for both mother and baby. Using murine models, we show that IAV infection in pregnancy with allergic airway disease is associated with impaired global antiviral and antimicrobial responses, increased lung inflammation, mucus hypersecretion, and airway hyperresponsiveness (AHR). Targeting specific β-defensins or microRNAs (miRNAs) may prove useful in future treatments for IAV infection during pregnancy.
Collapse
Affiliation(s)
- Rebecca L Vanders
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Henry M Gomez
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Alan C Hsu
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Katie Daly
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
4
|
Mishkin N, Ricart Arbona RJ, Carrasco SE, Lawton S, Henderson KS, Momtsios P, Sigar IM, Ramsey KH, Cheleuitte-Nieves C, Monette S, Lipman NS. Reemergence of the Murine Bacterial Pathogen Chlamydia muridarum in Research Mouse Colonies. Comp Med 2022; 72:230-242. [PMID: 35803706 PMCID: PMC9413529 DOI: 10.30802/aalas-cm-22-000045] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Chlamydia muridarum (Cm) was detected in 2 colonies of mice with lymphoplasmacytic pulmonary infiltrates by using PCR and immunohistochemistry. This discovery was unexpected, as Cm infection had not been reported in laboratory mice since the 1940s. A Cm specific PCR assay was developed and testing implemented for the resident colonies of 8 vivaria from 3 academic institutions, 58 incoming mouse shipments from 39 academic institutions, and mice received from 55 commercial breeding colonies (4 vendors). To estimate Cm's global prevalence in research colonies, a database containing 11,387 metagenomic fecal microbiota samples from 120 institutions and a cohort of 900 diagnostic samples from 96 institutions were examined. Results indicate significant prevalence among academic institutions, with Cm detected in 63% of soiled bedding sentinels from 3 institutions; 33% of incoming mouse shipments from 39 academic institutions; 14% of 120 institutions submitting microbiota samples; and 16% of the diagnostic sample cohort. All samples from commercial breeding colonies were negative. In addition, naïve NOD. Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mice exposed to Cm-shedding mice and/or their soiled bedding developed clinical disease at 21 to 28 d after exposure. These mice had a moderate-to-severe histiocytic and neutro- philic bronchointerstitial pneumonia, with their respiratory epithelium demonstrating inclusions, chlamydial major outer membrane protein immunostaining, and hybridization with a Cm reference sequence (GenBank accession no. U68436). Cm was isolated from lungs, cecum, and feces of a Cm-infected NSG mouse by using HeLa 229 cells. The considerable prevalence of Cm is likely due to widespread global interinstitutional distribution of unique mouse strains and failure to recognize that some of these mice were from enzootically infected colonies. Given that experimental Cm colonization of mice results in a robust immune response and, on occasion, pathology, natural infection may confound experimental results. Therefore, Cm should be excluded and eradicated from enzootically infected mouse colonies.
Collapse
Key Words
- cm, chlamydia muridarum
- eb, elementary body
- ffpe, formalin-fixed paraffin embedded
- gem, genetically engineered mouse
- ib, inclusion body
- ifa, immunofluorescence
- ifu, inclusion forming units
- ihc, immunohistochemistry
- ish, in-situ hybridization
- momp, major outer membrane protein
- mopn, mouse pneumonitis virus
- msk, memorial sloan kettering
- nsg, nod.cg-prkdcscid il2rgtm1wjl/szj
- rb, reticulate body
- tlr, toll-like receptor
Collapse
Affiliation(s)
- Noah Mishkin
- Tri-Institutional Training Program in Laboratory Animal Medicine and Science, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, and The Rockefeller University, New York, New York,,Corresponding authors. Emails: ,
| | - Rodolfo J Ricart Arbona
- Tri-Institutional Training Program in Laboratory Animal Medicine and Science, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, and The Rockefeller University, New York, New York,,Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, New York
| | - Sebastian E Carrasco
- Tri-Institutional Training Program in Laboratory Animal Medicine and Science, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, and The Rockefeller University, New York, New York,,Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, New York
| | | | - Kenneth S Henderson
- Research Animal Diagnostic Services, Charles River Laboratories, Wilmington, Massachusetts, and
| | - Panagiota Momtsios
- Research Animal Diagnostic Services, Charles River Laboratories, Wilmington, Massachusetts, and
| | - Ira M Sigar
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, Illinois
| | - Kyle H Ramsey
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, Illinois
| | - Christopher Cheleuitte-Nieves
- Tri-Institutional Training Program in Laboratory Animal Medicine and Science, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, and The Rockefeller University, New York, New York,,Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, New York
| | - Sebastien Monette
- Tri-Institutional Training Program in Laboratory Animal Medicine and Science, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, and The Rockefeller University, New York, New York,,Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, New York
| | - Neil S Lipman
- Tri-Institutional Training Program in Laboratory Animal Medicine and Science, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, and The Rockefeller University, New York, New York,,Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, New York,,Corresponding authors. Emails: ,
| |
Collapse
|
5
|
Wang Y, Gu LF, Zhao X, Hu C, Chen Q. TFR1 expression in induced sputum is associated with asthma severity. PeerJ 2022; 10:e13474. [PMID: 35602900 PMCID: PMC9121881 DOI: 10.7717/peerj.13474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/29/2022] [Indexed: 01/14/2023] Open
Abstract
Background Asthma is characterized as a chronic inflammatory airway disease. Iron accumulation is related to asthma pathogenesis. Transferrin receptor 1(TFR1) expression is associated with intracellular iron overload in macrophages. In our study, we explored the association among TFR1 expression, the inflammatory macrophage phenotype, and asthma severity. Methods Induced sputum was collected from 50 asthma patients. Real-time PCR was used to evaluate mRNA expression. The status of inflammatory macrophage phenotype was assessed using flow cytometry. Results TFR1 levels were inversely correlated with forced expiratory volume in 1 s (FEV1)/forced vital capacity (FVC) and FEV1/vital capacity (VC). Among inflammatory cytokines, TFR1 expression was positively correlated with IL-1β, TNF-α, IL-6, IFN-γ, and IL-17A mRNA expression in induced sputum. Moreover, TFR1 expression was positively correlated with the number of proinflammatory M1 macrophages and iNOS expression in induced sputum. Neutrophil counts in induced sputum were significantly and positively related to TFR1 expression. Furthermore, TFR1 expression showed an increasing trend in asthma patients with no family history. Our findings indicated that TFR1 expression was consistent with the asthma severity index, especially the proinflammatory M1 macrophage phenotype. TFR1 expression may be a good marker to indicate asthma severity.
Collapse
Affiliation(s)
- Yang Wang
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - li Feng Gu
- Hunan Children’s Hospital, Changsha, Hunan, China
| | - Xincheng Zhao
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Chengping Hu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiong Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
6
|
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: 64] [Impact Index Per Article: 16.0] [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
|
7
|
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
|
8
|
Liu G, Philp AM, Corte T, Travis MA, Schilter H, Hansbro NG, Burns CJ, Eapen MS, Sohal SS, Burgess JK, Hansbro PM. Therapeutic targets in lung tissue remodelling and fibrosis. Pharmacol Ther 2021; 225:107839. [PMID: 33774068 DOI: 10.1016/j.pharmthera.2021.107839] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Structural changes involving tissue remodelling and fibrosis are major features of many pulmonary diseases, including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Abnormal deposition of extracellular matrix (ECM) proteins is a key factor in the development of tissue remodelling that results in symptoms and impaired lung function in these diseases. Tissue remodelling in the lungs is complex and differs between compartments. Some pathways are common but tissue remodelling around the airways and in the parenchyma have different morphologies. Hence it is critical to evaluate both common fibrotic pathways and those that are specific to different compartments; thereby expanding the understanding of the pathogenesis of fibrosis and remodelling in the airways and parenchyma in asthma, COPD and IPF with a view to developing therapeutic strategies for each. Here we review the current understanding of remodelling features and underlying mechanisms in these major respiratory diseases. The differences and similarities of remodelling are used to highlight potential common therapeutic targets and strategies. One central pathway in remodelling processes involves transforming growth factor (TGF)-β induced fibroblast activation and myofibroblast differentiation that increases ECM production. The current treatments and clinical trials targeting remodelling are described, as well as potential future directions. These endeavours are indicative of the renewed effort and optimism for drug discovery targeting tissue remodelling and fibrosis.
Collapse
Affiliation(s)
- Gang Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Ashleigh M Philp
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia; St Vincent's Medical School, UNSW Medicine, UNSW, Sydney, NSW, Australia
| | - Tamera Corte
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Mark A Travis
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre and Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom
| | - Heidi Schilter
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, NSW, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Chris J Burns
- Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mathew S Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Sukhwinder S Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Department of Pathology and Medical Biology, Groningen, The Netherlands; Woolcock Institute of Medical Research, Discipline of Pharmacology, The University of Sydney, Sydney, NSW, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
9
|
Tu X, Donovan C, Kim RY, Wark PAB, Horvat JC, Hansbro PM. Asthma-COPD overlap: current understanding and the utility of experimental models. Eur Respir Rev 2021; 30:30/159/190185. [PMID: 33597123 PMCID: PMC9488725 DOI: 10.1183/16000617.0185-2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 11/03/2020] [Indexed: 12/21/2022] Open
Abstract
Pathological features of both asthma and COPD coexist in some patients and this is termed asthma-COPD overlap (ACO). ACO is heterogeneous and patients exhibit various combinations of asthma and COPD features, making it difficult to characterise the underlying pathogenic mechanisms. There are no controlled studies that define effective therapies for ACO, which arises from the lack of international consensus on the definition and diagnostic criteria for ACO, as well as scant in vitro and in vivo data. There remain unmet needs for experimental models of ACO that accurately recapitulate the hallmark features of ACO in patients. The development and interrogation of such models will identify underlying disease-causing mechanisms, as well as enabling the identification of novel therapeutic targets and providing a platform for assessing new ACO therapies. Here, we review the current understanding of the clinical features of ACO and highlight the approaches that are best suited for developing representative experimental models of ACO. Understanding the pathogenesis of asthma-COPD overlap is critical for improving therapeutic approaches. We present current knowledge on asthma-COPD overlap and the requirements for developing an optimal animal model of disease.https://bit.ly/3lsjyvm
Collapse
Affiliation(s)
- Xiaofan Tu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Both authors contributed equally
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute, Camperdown, Australia.,University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia.,Both authors contributed equally
| | - Richard Y Kim
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute, Camperdown, Australia.,University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia .,Centre for Inflammation, Centenary Institute, Camperdown, Australia.,University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| |
Collapse
|
10
|
Shastri MD, Chong WC, Dua K, Peterson GM, Patel RP, Mahmood MQ, Tambuwala M, Chellappan DK, Hansbro NG, Shukla SD, Hansbro PM. Emerging concepts and directed therapeutics for the management of asthma: regulating the regulators. Inflammopharmacology 2020; 29:15-33. [PMID: 33152094 DOI: 10.1007/s10787-020-00770-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/17/2020] [Indexed: 12/19/2022]
Abstract
Asthma is a common, heterogeneous and serious disease, its prevalence has steadily risen in most parts of the world, and the condition is often inadequately controlled in many patients. Hence, there is a major need for new therapeutic approaches. Mild-to-moderate asthma is considered a T-helper cell type-2-mediated inflammatory disorder that develops due to abnormal immune responses to otherwise innocuous allergens. Prolonged exposure to allergens and persistent inflammation results in myofibroblast infiltration and airway remodelling with mucus hypersecretion, airway smooth muscle hypertrophy, and excess collagen deposition. The airways become hyper-responsive to provocation resulting in the characteristic wheezing and obstructed airflow experienced by patients. Extensive research has progressed the understanding of the underlying mechanisms and the development of new treatments for the management of asthma. Here, we review the basis of the disease, covering new areas such as the role of vascularisation and microRNAs, as well as associated potential therapeutic interventions utilising reports from animal and human studies. We also cover novel drug delivery strategies that are being developed to enhance therapeutic efficacy and patient compliance. Potential avenues to explore to improve the future of asthma management are highlighted.
Collapse
Affiliation(s)
- Madhur D Shastri
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Wai Chin Chong
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia.,Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia.,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia.,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Gregory M Peterson
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Rahul P Patel
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Malik Q Mahmood
- Faculty of Health, School of Medicine, Deakin University, Melbourne, Australia
| | - Murtaza Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Belfast, Northern Ireland, UK
| | - Dinesh K Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia.,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia.,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia. .,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia. .,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
11
|
Jones-Freeman B, Starkey MR. Bronchioalveolar stem cells in lung repair, regeneration and disease. J Pathol 2020; 252:219-226. [PMID: 32737996 DOI: 10.1002/path.5527] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/08/2020] [Accepted: 07/26/2020] [Indexed: 12/17/2022]
Abstract
Bronchioalveolar stem cells (BASCs) are a lung resident stem cell population located at bronchioalveolar duct junctions that contribute to the maintenance of bronchiolar club cells and alveolar epithelial cells of the distal lung. Their transformed counterparts are considered to be likely progenitors of lung adenocarcinomas, which has been a major area of research in relation to BASCs. A critical limitation in addressing the function of BASCs in vivo has been the lack of a unique BASC marker, which has prevented specific targeting of BASCs in animal models of respiratory conditions. Recently, there have been several studies describing genetically modified mice that allow in vivo quantification, tracing, and functional analysis of BASCs to address this long-standing issue. These cutting-edge experimental tools will likely have significant implications for future experimental studies involving BASCs and the elucidation of their role in various lung diseases. To date, this has been largely explored in models of lung injury including naphthalene-induced airway injury, bleomycin-induced alveolar injury, hyperoxia-induced models of bronchopulmonary dysplasia, and influenza virus infection. These novel experimental mouse tools will facilitate the assessment of the impact of BASC loss on additional respiratory conditions including infection-induced severe asthma and chronic obstructive pulmonary disease, as well as respiratory bacterial infections, both in early life and adulthood. These future studies may shed light on the potential broad applicability of targeting BASCs for a diverse range of respiratory conditions during lung development and in promoting effective regeneration and repair of the lung in respiratory diseases. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Bernadette Jones-Freeman
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - Malcolm R Starkey
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
12
|
Paudel KR, Dharwal V, Patel VK, Galvao I, Wadhwa R, Malyla V, Shen SS, Budden KF, Hansbro NG, Vaughan A, Yang IA, Kohonen-Corish MRJ, Bebawy M, Dua K, Hansbro PM. Role of Lung Microbiome in Innate Immune Response Associated With Chronic Lung Diseases. Front Med (Lausanne) 2020; 7:554. [PMID: 33043031 PMCID: PMC7530186 DOI: 10.3389/fmed.2020.00554] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
Respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD), lung fibrosis, and lung cancer, pose a huge socio-economic burden on society and are one of the leading causes of death worldwide. In the past, culture-dependent techniques could not detect bacteria in the lungs, therefore the lungs were considered a sterile environment. However, the development of culture-independent techniques, particularly 16S rRNA sequencing, allowed for the detection of commensal microbes in the lung and with further investigation, their roles in disease have since emerged. In healthy individuals, the predominant commensal microbes are of phylum Firmicutes and Bacteroidetes, including those of the genera Veillonella and Prevotella. In contrast, pathogenic microbes (Haemophilus, Streptococcus, Klebsiella, Pseudomonas) are often associated with lung diseases. There is growing evidence that microbial metabolites, structural components, and toxins from pathogenic and opportunistic bacteria have the capacity to stimulate both innate and adaptive immune responses, and therefore can contribute to the pathogenesis of lung diseases. Here we review the multiple mechanisms that are altered by pathogenic microbiomes in asthma, COPD, lung cancer, and lung fibrosis. Furthermore, we focus on the recent exciting advancements in therapies that can be used to restore altered microbiomes in the lungs.
Collapse
Affiliation(s)
- Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Vivek Dharwal
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Vyoma K Patel
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Izabela Galvao
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Ridhima Wadhwa
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - Vamshikrishna Malyla
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - Sj Sijie Shen
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Kurtis F Budden
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Annalicia Vaughan
- Faculty of Medicine, Thoracic Research Centre, The University of Queensland, Brisbane, QLD, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Ian A Yang
- Faculty of Medicine, Thoracic Research Centre, The University of Queensland, Brisbane, QLD, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Maija R J Kohonen-Corish
- Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.,Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia.,School of Medicine, Western Sydney University, Sydney, NSW, Australia.,St George and Sutherland Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Mary Bebawy
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - Kamal Dua
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
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: 60] [Impact Index Per Article: 10.0] [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
|
17
|
Pulmonary group 2 innate lymphoid cells: surprises and challenges. Mucosal Immunol 2019; 12:299-311. [PMID: 30664706 PMCID: PMC6436699 DOI: 10.1038/s41385-018-0130-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 02/04/2023]
Abstract
Group 2 innate lymphoid cells (ILC2s) are a recently described subset of innate lymphocytes with important immune and homeostatic functions at multiple tissue sites, especially the lung. These cells expand locally after birth and during postnatal lung maturation and are present in the lung and other peripheral organs. They are modified by a variety of processes and mediate inflammatory responses to respiratory pathogens, inhaled allergens and noxious particles. Here, we review the emerging roles of ILC2s in pulmonary homeostasis and discuss recent and surprising advances in our understanding of how hormones, age, neurotransmitters, environmental challenges, and infection influence ILC2s. We also review how these responses may underpin the development, progression and severity of pulmonary inflammation and chronic lung diseases and highlight some of the remaining challenges for ILC2 biology.
Collapse
|
18
|
Feasibility Analysis of Interleukin-13 as a Target for a Therapeutic Vaccine. Vaccines (Basel) 2019; 7:vaccines7010020. [PMID: 30759882 PMCID: PMC6466196 DOI: 10.3390/vaccines7010020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/04/2019] [Accepted: 02/07/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The development of therapeutic vaccines requires thorough knowledge of potential hazards associated with long-term inactivation of self-proteins. Among potential targets, interleukin 13 (IL-13) merits consideration, as monoclonal antibodies disrupting IL-13 signaling are proving to be exceedingly effective in common conditions such as atopic dermatitis. OBJECTIVE Given the mass publication of scientific data, an appraisal of safety aspects is challenging. METHODS We here provide a three-fold approach to survey clinically relevant information on off-target effects, both adverse and beneficial, that may potentially be encountered in patients undergoing long-term IL-13 inactivation. First, we review non-clinical data in vivo and in vitro. Second, we summarize safety data accumulating from patients dosed with anti-IL-13 drugs. Third, we exploit human mutation data as well as emerging large-scale genetic datasets (global exome data from 60,000 patients) to obtain information on any association of IL-13-inactivating genetic variants with disease states. In addition, we: (1) dissect the precise efficacy signals obtained with various drugs targeting IL-13 and/or IL-4, and (2) summarize unintended, but potentially beneficial effects of prolonged IL-13 inactivation on several functional systems. RESULTS Prolonged repression of IL-13 in several thousand patients so far has not uncovered any non-redundant functions of IL-13 in immune defense. Furthermore, missense mutations in the key genes IL-13, IL-13Rα1, IL-13Rα2, IL-4, IL-4Rα are common, while no case reports have been published on any immune deficiency or increased risk of neoplastic disease associated with such mutations, suggesting that these genes do not harbor non-redundant roles in adult outbred humans. In terms of efficacy, data from clinically used drugs strongly suggest that targeting IL-13 only, as opposed to IL-13 and IL-4, may be effective in eczema while being more selective. Importantly, several lines of evidence suggest that inhibition of IL-13 may in fact harbor potentially beneficial effects on non-targeted systems, including glucose metabolism, hepatic fibrosis, and atherosclerosis, suggesting that respective outcomes should be systematically captured in patients dosed with IL-13 interfering drugs. Collectively, available evidence suggests that IL-13 may fulfill safety requirements required for the target of a therapeutic vaccine.
Collapse
|
19
|
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: 32] [Impact Index Per Article: 5.3] [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
|
20
|
Donovan C, Starkey MR, Kim RY, Rana BMJ, Barlow JL, Jones B, Haw TJ, Mono Nair P, Budden K, Cameron GJM, Horvat JC, Wark PA, Foster PS, McKenzie ANJ, Hansbro PM. Roles for T/B lymphocytes and ILC2s in experimental chronic obstructive pulmonary disease. J Leukoc Biol 2018; 105:143-150. [PMID: 30260499 PMCID: PMC6487813 DOI: 10.1002/jlb.3ab0518-178r] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/03/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022] Open
Abstract
Pulmonary inflammation in chronic obstructive pulmonary disease (COPD) is characterized by both innate and adaptive immune responses; however, their specific roles in the pathogenesis of COPD are unclear. Therefore, we investigated the roles of T and B lymphocytes and group 2 innate lymphoid cells (ILC2s) in airway inflammation and remodelling, and lung function in an experimental model of COPD using mice that specifically lack these cells (Rag1−/− and Rorafl/flIl7rCre [ILC2‐deficient] mice). Wild‐type (WT) C57BL/6 mice, Rag1−/−, and Rorafl/flIl7rCre mice were exposed to cigarette smoke (CS; 12 cigarettes twice a day, 5 days a week) for up to 12 weeks, and airway inflammation, airway remodelling (collagen deposition and alveolar enlargement), and lung function were assessed. WT, Rag1−/−, and ILC2‐deficient mice exposed to CS had similar levels of airway inflammation and impaired lung function. CS exposure increased small airway collagen deposition in WT mice. Rag1−/− normal air‐ and CS‐exposed mice had significantly increased collagen deposition compared to similarly exposed WT mice, which was associated with increases in IL‐33, IL‐13, and ILC2 numbers. CS‐exposed Rorafl/flIl7rCre mice were protected from emphysema, but had increased IL‐33/IL‐13 expression and collagen deposition compared to WT CS‐exposed mice. T/B lymphocytes and ILC2s play roles in airway collagen deposition/fibrosis, but not inflammation, in experimental COPD.
Collapse
Affiliation(s)
- Chantal Donovan
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Malcolm R Starkey
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Richard Y Kim
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Batika M J Rana
- Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge, UK
| | - Jillian L Barlow
- Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge, UK
| | - Bernadette Jones
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Tatt Jhong Haw
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Prema Mono Nair
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Kurtis Budden
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Guy J M Cameron
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Jay C Horvat
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Peter A Wark
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Paul S Foster
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Andrew N J McKenzie
- Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge, UK
| | - Philip M Hansbro
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia.,The Centenary Institute and the School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
21
|
Therapeutic Effects of Kangzhi Syrup in a Guinea Pig Model of Ovalbumin-Induced Cough Variant Asthma. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:5867835. [PMID: 29951106 PMCID: PMC5989290 DOI: 10.1155/2018/5867835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/24/2018] [Accepted: 04/16/2018] [Indexed: 11/18/2022]
Abstract
Purpose This study aimed to investigate the possible effects and underlying mechanisms of Kangzhi syrup on ovalbumin- (OVA-) induced cough variant asthma (CVA) in guinea pigs. Methods All 48 guinea pigs were randomly assigned to four experimental groups: normal, OVA model with or without Kangzhi syrup (OVA and OVA + KZ), and OVA with Dexamethasone (OVA + DM). After sensitizing the guinea pigs, a cough challenge was performed by the inhalation of capsaicin. The antitussive effect, inflammatory cells, cytokines in bronchoalveolar lavage fluid (BALF) and lung tissue, and morphological changes were examined. Results Compared with model group, Kangzhi syrup effectively exerted an antitussive effect (p < 0.0001) and reduced the pneumonic anaphylacticitis by inhibiting the infiltration of total inflammatory cells (p < 0.0001) and reducing the percentage of eosinophil in BALF (p < 0.0001). Moreover, evidence from morphological studies also demonstrated that Kangzhi syrup inhibited the infiltration of inflammatory cells and ameliorated the structure changes. NF-κB and TGF-β1 expression were attenuated in the OVA + KZ group versus the OVA group (p < 0.0001). Additionally, a semiquantitative analysis of TGF-β1 expression also demonstrated that the Kangzhi syrup attenuated this profibrogenic growth factor (p < 0.001). Conclusions The results demonstrated that Kangzhi syrup exerted a considerable antitussive effect in CVA animal model, which depended on its marked impact on the anti-anaphylactic inflammation. Additionally, it could ameliorate the airway remodeling by inhibiting NF-κB and TGF-β1 signal pathway.
Collapse
|
22
|
Hansbro PM, Kim RY, Starkey MR, Donovan C, Dua K, Mayall JR, Liu G, Hansbro NG, Simpson JL, Wood LG, Hirota JA, Knight DA, Foster PS, Horvat JC. Mechanisms and treatments for severe, steroid-resistant allergic airway disease and asthma. Immunol Rev 2018; 278:41-62. [PMID: 28658552 DOI: 10.1111/imr.12543] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Severe, steroid-resistant asthma is clinically and economically important since affected individuals do not respond to mainstay corticosteroid treatments for asthma. Patients with this disease experience more frequent exacerbations of asthma, are more likely to be hospitalized, and have a poorer quality of life. Effective therapies are urgently required, however, their development has been hampered by a lack of understanding of the pathological processes that underpin disease. A major obstacle to understanding the processes that drive severe, steroid-resistant asthma is that the several endotypes of the disease have been described that are characterized by different inflammatory and immunological phenotypes. This heterogeneity makes pinpointing processes that drive disease difficult in humans. Clinical studies strongly associate specific respiratory infections with severe, steroid-resistant asthma. In this review, we discuss key findings from our studies where we describe the development of representative experimental models to improve our understanding of the links between infection and severe, steroid-resistant forms of this disease. We also discuss their use in elucidating the mechanisms, and their potential for developing effective therapeutic strategies, for severe, steroid-resistant asthma. Finally, we highlight how the immune mechanisms and therapeutic targets we have identified may be applicable to obesity-or pollution-associated asthma.
Collapse
Affiliation(s)
- Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Richard Y Kim
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Malcolm R Starkey
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Kamal Dua
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Jemma R Mayall
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Gang Liu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Jodie L Simpson
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Lisa G Wood
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Jeremy A Hirota
- James Hogg Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Darryl A Knight
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
23
|
Haw TJ, Starkey MR, Pavlidis S, Fricker M, Arthurs AL, Nair PM, Liu G, Hanish I, Kim RY, Foster PS, Horvat JC, Adcock IM, Hansbro PM. Toll-like receptor 2 and 4 have opposing roles in the pathogenesis of cigarette smoke-induced chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2018; 314:L298-L317. [PMID: 29025711 PMCID: PMC5866502 DOI: 10.1152/ajplung.00154.2017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 09/08/2017] [Accepted: 10/03/2017] [Indexed: 12/18/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of morbidity and death and imposes major socioeconomic burdens globally. It is a progressive and disabling condition that severely impairs breathing and lung function. There is a lack of effective treatments for COPD, which is a direct consequence of the poor understanding of the underlying mechanisms involved in driving the pathogenesis of the disease. Toll-like receptor (TLR)2 and TLR4 are implicated in chronic respiratory diseases, including COPD, asthma and pulmonary fibrosis. However, their roles in the pathogenesis of COPD are controversial and conflicting evidence exists. In the current study, we investigated the role of TLR2 and TLR4 using a model of cigarette smoke (CS)-induced experimental COPD that recapitulates the hallmark features of human disease. TLR2, TLR4, and associated coreceptor mRNA expression was increased in the airways in both experimental and human COPD. Compared with wild-type (WT) mice, CS-induced pulmonary inflammation was unaltered in TLR2-deficient ( Tlr2-/-) and TLR4-deficient ( Tlr4-/-) mice. CS-induced airway fibrosis, characterized by increased collagen deposition around small airways, was not altered in Tlr2-/- mice but was attenuated in Tlr4-/- mice compared with CS-exposed WT controls. However, Tlr2-/- mice had increased CS-induced emphysema-like alveolar enlargement, apoptosis, and impaired lung function, while these features were reduced in Tlr4-/- mice compared with CS-exposed WT controls. Taken together, these data highlight the complex roles of TLRs in the pathogenesis of COPD and suggest that activation of TLR2 and/or inhibition of TLR4 may be novel therapeutic strategies for the treatment of COPD.
Collapse
Affiliation(s)
- Tatt Jhong Haw
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Malcolm R Starkey
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
- Priority Research Centre for Grow Up Well, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Stelios Pavlidis
- The Airways Disease Section, National Heart and Lung Institute, Imperial College London , London , United Kingdom
| | - Michael Fricker
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Anya L Arthurs
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Prema M Nair
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Gang Liu
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Irwan Hanish
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor , Malaysia
| | - Richard Y Kim
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Ian M Adcock
- The Airways Disease Section, National Heart and Lung Institute, Imperial College London , London , United Kingdom
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| |
Collapse
|
24
|
Reid AT, Veerati PC, Gosens R, Bartlett NW, Wark PA, Grainge CL, Stick SM, Kicic A, Moheimani F, Hansbro PM, Knight DA. Persistent induction of goblet cell differentiation in the airways: Therapeutic approaches. Pharmacol Ther 2017; 185:155-169. [PMID: 29287707 DOI: 10.1016/j.pharmthera.2017.12.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dysregulated induction of goblet cell differentiation results in excessive production and retention of mucus and is a common feature of several chronic airways diseases. To date, therapeutic strategies to reduce mucus accumulation have focused primarily on altering the properties of the mucus itself, or have aimed to limit the production of mucus-stimulating cytokines. Here we review the current knowledge of key molecular pathways that are dysregulated during persistent goblet cell differentiation and highlights both pre-existing and novel therapeutic strategies to combat this pathology.
Collapse
Affiliation(s)
- Andrew T Reid
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia.
| | - Punnam Chander Veerati
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nathan W Bartlett
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia
| | - Peter A Wark
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Chris L Grainge
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Stephen M Stick
- School of Paediatrics and Child Health, University of Western Australia, Nedlands 6009, Western Australia, Australia; Telethon Kids Institute, University of Western Australia, Nedlands 6009, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth 6001, Western Australia, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands 6009, Western Australia, Australia
| | - Anthony Kicic
- School of Paediatrics and Child Health, University of Western Australia, Nedlands 6009, Western Australia, Australia; Telethon Kids Institute, University of Western Australia, Nedlands 6009, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth 6001, Western Australia, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands 6009, Western Australia, Australia; Occupation and Environment, School of Public Health, Curtin University, Bentley 6102, Western Australia, Australia
| | - Fatemeh Moheimani
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
25
|
Liu G, Cooley MA, Nair PM, Donovan C, Hsu AC, Jarnicki AG, Haw TJ, Hansbro NG, Ge Q, Brown AC, Tay H, Foster PS, Wark PA, Horvat JC, Bourke JE, Grainge CL, Argraves WS, Oliver BG, Knight DA, Burgess JK, Hansbro PM. Airway remodelling and inflammation in asthma are dependent on the extracellular matrix protein fibulin-1c. J Pathol 2017; 243:510-523. [PMID: 28862768 DOI: 10.1002/path.4979] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 01/08/2023]
Abstract
Asthma is a chronic inflammatory disease of the airways. It is characterized by allergic airway inflammation, airway remodelling, and airway hyperresponsiveness (AHR). Asthma patients, in particular those with chronic or severe asthma, have airway remodelling that is associated with the accumulation of extracellular matrix (ECM) proteins, such as collagens. Fibulin-1 (Fbln1) is an important ECM protein that stabilizes collagen and other ECM proteins. The level of Fbln1c, one of the four Fbln1 variants, which predominates in both humans and mice, is increased in the serum and airways fluids in asthma but its function is unclear. We show that the level of Fbln1c was increased in the lungs of mice with house dust mite (HDM)-induced chronic allergic airway disease (AAD). Genetic deletion of Fbln1c and therapeutic inhibition of Fbln1c in mice with chronic AAD reduced airway collagen deposition, and protected against AHR. Fbln1c-deficient (Fbln1c-/- ) mice had reduced mucin (MUC) 5 AC levels, but not MUC5B levels, in the airways as compared with wild-type (WT) mice. Fbln1c interacted with fibronectin and periostin that was linked to collagen deposition around the small airways. Fbln1c-/- mice with AAD also had reduced numbers of α-smooth muscle actin-positive cells around the airways and reduced airway contractility as compared with WT mice. After HDM challenge, these mice also had fewer airway inflammatory cells, reduced interleukin (IL)-5, IL-13, IL-33, tumour necrosis factor (TNF) and CXCL1 levels in the lungs, and reduced IL-5, IL-33 and TNF levels in lung-draining lymph nodes. Therapeutic targeting of Fbln1c reduced the numbers of GATA3-positive Th2 cells in the lymph nodes and lungs after chronic HDM challenge. Treatment also reduced the secretion of IL-5 and IL-13 from co-cultured dendritic cells and T cells restimulated with HDM extract. Human epithelial cells cultured with Fbln1c peptide produced more CXCL1 mRNA than medium-treated controls. Our data show that Fbln1c may be a therapeutic target in chronic asthma. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Gang Liu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Marion A Cooley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Prema M Nair
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Alan C Hsu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Andrew G Jarnicki
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Tatt Jhong Haw
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Qi Ge
- Woolcock Institute of Medical Research, Discipline of Pharmacology, University of Sydney, Sydney, New South Wales, Australia
| | - Alexandra C Brown
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Hock Tay
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Peter A Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Jane E Bourke
- Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Chris L Grainge
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - W Scott Argraves
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Brian G Oliver
- Woolcock Institute of Medical Research, Discipline of Pharmacology, University of Sydney, Sydney, New South Wales, Australia.,School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Darryl A Knight
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Janette K Burgess
- Woolcock Institute of Medical Research, Discipline of Pharmacology, University of Sydney, Sydney, New South Wales, Australia.,University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Groningen Research Institute of Asthma and COPD, Groningen, The Netherlands
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
26
|
Nair PM, Starkey MR, Haw TJ, Liu G, Horvat JC, Morris JC, Verrills NM, Clark AR, Ammit AJ, Hansbro PM. Targeting PP2A and proteasome activity ameliorates features of allergic airway disease in mice. Allergy 2017; 72:1891-1903. [PMID: 28543283 DOI: 10.1111/all.13212] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Asthma is an allergic airway disease (AAD) caused by aberrant immune responses to allergens. Protein phosphatase-2A (PP2A) is an abundant serine/threonine phosphatase with anti-inflammatory activity. The ubiquitin proteasome system (UPS) controls many cellular processes, including the initiation of inflammatory responses by protein degradation. We assessed whether enhancing PP2A activity with fingolimod (FTY720) or 2-amino-4-(4-(heptyloxy) phenyl)-2-methylbutan-1-ol (AAL(S) ), or inhibiting proteasome activity with bortezomib (BORT), could suppress experimental AAD. METHODS Acute AAD was induced in C57BL/6 mice by intraperitoneal sensitization with ovalbumin (OVA) in combination with intranasal (i.n) exposure to OVA. Chronic AAD was induced in mice with prolonged i.n exposure to crude house dust mite (HDM) extract. Mice were treated with vehicle, FTY720, AAL(S) , BORT or AAL(S) +BORT and hallmark features of AAD assessed. RESULTS AAL(S) reduced the severity of acute AAD by suppressing tissue eosinophils and inflammation, mucus-secreting cell (MSC) numbers, type 2-associated cytokines (interleukin (IL)-33, thymic stromal lymphopoietin, IL-5 and IL-13), serum immunoglobulin (Ig)E and airway hyper-responsiveness (AHR). FTY720 only suppressed tissue inflammation and IgE. BORT reduced bronchoalveolar lavage fluid (BALF) and tissue eosinophils and inflammation, IL-5, IL-13 and AHR. Combined treatment with AAL(S) +BORT had complementary effects and suppressed BALF and tissue eosinophils and inflammation, MSC numbers, reduced the production of type 2 cytokines and AHR. AAL(S) , BORT and AAL(S) +BORT also reduced airway remodelling in chronic AAD. CONCLUSION These findings highlight the potential of combination therapies that enhance PP2A and inhibit proteasome activity as novel therapeutic strategies for asthma.
Collapse
Affiliation(s)
- P. M. Nair
- Priority Research Centres for Healthy Lungs; Grow up Well and Cancer Research, Innovation and Translation; University of Newcastle & Hunter Medical Research Institute; New Lambton Heights NSW Australia
- Faculty of Health and Medicine; School of Biomedical Sciences and Pharmacy; University of Newcastle; Callaghan NSW Australia
| | - M. R. Starkey
- Priority Research Centres for Healthy Lungs; Grow up Well and Cancer Research, Innovation and Translation; University of Newcastle & Hunter Medical Research Institute; New Lambton Heights NSW Australia
- Faculty of Health and Medicine; School of Biomedical Sciences and Pharmacy; University of Newcastle; Callaghan NSW Australia
| | - T. J. Haw
- Priority Research Centres for Healthy Lungs; Grow up Well and Cancer Research, Innovation and Translation; University of Newcastle & Hunter Medical Research Institute; New Lambton Heights NSW Australia
- Faculty of Health and Medicine; School of Biomedical Sciences and Pharmacy; University of Newcastle; Callaghan NSW Australia
| | - G. Liu
- Priority Research Centres for Healthy Lungs; Grow up Well and Cancer Research, Innovation and Translation; University of Newcastle & Hunter Medical Research Institute; New Lambton Heights NSW Australia
- Faculty of Health and Medicine; School of Biomedical Sciences and Pharmacy; University of Newcastle; Callaghan NSW Australia
| | - J. C. Horvat
- Priority Research Centres for Healthy Lungs; Grow up Well and Cancer Research, Innovation and Translation; University of Newcastle & Hunter Medical Research Institute; New Lambton Heights NSW Australia
- Faculty of Health and Medicine; School of Biomedical Sciences and Pharmacy; University of Newcastle; Callaghan NSW Australia
| | - J. C. Morris
- School of Chemistry; University of New South Wales; Sydney NSW Australia
| | - N. M. Verrills
- Priority Research Centres for Healthy Lungs; Grow up Well and Cancer Research, Innovation and Translation; University of Newcastle & Hunter Medical Research Institute; New Lambton Heights NSW Australia
- Faculty of Health and Medicine; School of Biomedical Sciences and Pharmacy; University of Newcastle; Callaghan NSW Australia
| | - A. R. Clark
- Institute of Inflammation and Ageing; College of Medical and Dental Sciences; University of Birmingham; Birmingham UK
| | - A. J. Ammit
- Woolcock Emphysema Centre; Woolcock Institute of Medical Research; University of Sydney; Sydney NSW Australia
- Faculty of Science; School of Life Sciences; University of Technology Sydney; Sydney NSW Australia
| | - P. M. Hansbro
- Priority Research Centres for Healthy Lungs; Grow up Well and Cancer Research, Innovation and Translation; University of Newcastle & Hunter Medical Research Institute; New Lambton Heights NSW Australia
- Faculty of Health and Medicine; School of Biomedical Sciences and Pharmacy; University of Newcastle; Callaghan NSW Australia
| |
Collapse
|
27
|
Chotirmall SH, Gellatly SL, Budden KF, Mac Aogain M, Shukla SD, Wood DLA, Hugenholtz P, Pethe K, Hansbro PM. Microbiomes in respiratory health and disease: An Asia-Pacific perspective. Respirology 2017; 22:240-250. [PMID: 28102970 DOI: 10.1111/resp.12971] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/30/2016] [Accepted: 11/17/2016] [Indexed: 02/06/2023]
Abstract
There is currently enormous interest in studying the role of the microbiome in health and disease. Microbiome's role is increasingly being applied to respiratory diseases, in particular COPD, asthma, cystic fibrosis and bronchiectasis. The changes in respiratory microbiomes that occur in these diseases and how they are modified by environmental challenges such as cigarette smoke, air pollution and infection are being elucidated. There is also emerging evidence that gut microbiomes play a role in lung diseases through the modulation of systemic immune responses and can be modified by diet and antibiotic treatment. There are issues that are particular to the Asia-Pacific region involving diet and prevalence of specific respiratory diseases. Each of these issues is further complicated by the effects of ageing. The challenges now are to elucidate the cause and effect relationships between changes in microbiomes and respiratory diseases and how to translate these into new treatments and clinical care. Here we review the current understanding and progression in these areas.
Collapse
Affiliation(s)
- Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Shaan L Gellatly
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Kurtis F Budden
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Micheál Mac Aogain
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - David L A Wood
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Philip Hugenholtz
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Kevin Pethe
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| |
Collapse
|
28
|
Kim RY, Pinkerton JW, Essilfie AT, Robertson AAB, Baines KJ, Brown AC, Mayall JR, Ali MK, Starkey MR, Hansbro NG, Hirota JA, Wood LG, Simpson JL, Knight DA, Wark PA, Gibson PG, O'Neill LAJ, Cooper MA, Horvat JC, Hansbro PM. Role for NLRP3 Inflammasome-mediated, IL-1β-Dependent Responses in Severe, Steroid-Resistant Asthma. Am J Respir Crit Care Med 2017; 196:283-297. [PMID: 28252317 DOI: 10.1164/rccm.201609-1830oc] [Citation(s) in RCA: 324] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
RATIONALE Severe, steroid-resistant asthma is the major unmet need in asthma therapy. Disease heterogeneity and poor understanding of pathogenic mechanisms hampers the identification of therapeutic targets. Excessive nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome and concomitant IL-1β responses occur in chronic obstructive pulmonary disease, respiratory infections, and neutrophilic asthma. However, the direct contributions to pathogenesis, mechanisms involved, and potential for therapeutic targeting remain poorly understood, and are unknown in severe, steroid-resistant asthma. OBJECTIVES To investigate the roles and therapeutic targeting of the NLRP3 inflammasome and IL-1β in severe, steroid-resistant asthma. METHODS We developed mouse models of Chlamydia and Haemophilus respiratory infection-mediated, ovalbumin-induced severe, steroid-resistant allergic airway disease. These models share the hallmark features of human disease, including elevated airway neutrophils, and NLRP3 inflammasome and IL-1β responses. The roles and potential for targeting of NLRP3 inflammasome, caspase-1, and IL-1β responses in experimental severe, steroid-resistant asthma were examined using a highly selective NLRP3 inhibitor, MCC950; the specific caspase-1 inhibitor Ac-YVAD-cho; and neutralizing anti-IL-1β antibody. Roles for IL-1β-induced neutrophilic inflammation were examined using IL-1β and anti-Ly6G. MEASUREMENTS AND MAIN RESULTS Chlamydia and Haemophilus infections increase NLRP3, caspase-1, IL-1β responses that drive steroid-resistant neutrophilic inflammation and airway hyperresponsiveness. Neutrophilic airway inflammation, disease severity, and steroid resistance in human asthma correlate with NLRP3 and IL-1β expression. Treatment with anti-IL-1β, Ac-YVAD-cho, and MCC950 suppressed IL-1β responses and the important steroid-resistant features of disease in mice, whereas IL-1β administration recapitulated these features. Neutrophil depletion suppressed IL-1β-induced steroid-resistant airway hyperresponsiveness. CONCLUSIONS NLRP3 inflammasome responses drive experimental severe, steroid-resistant asthma and are potential therapeutic targets in this disease.
Collapse
Affiliation(s)
- Richard Y Kim
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - James W Pinkerton
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Ama T Essilfie
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Avril A B Robertson
- 2 Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Katherine J Baines
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Alexandra C Brown
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Jemma R Mayall
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - M Khadem Ali
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Malcolm R Starkey
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Nicole G Hansbro
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Jeremy A Hirota
- 3 James Hogg Research Centre, University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Lisa G Wood
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Jodie L Simpson
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Darryl A Knight
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Peter A Wark
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Peter G Gibson
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Luke A J O'Neill
- 4 School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Matthew A Cooper
- 2 Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Jay C Horvat
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- 1 Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
29
|
Dua K, Hansbro NG, Hansbro PM. Steroid resistance and concomitant respiratory infections: A challenging battle in pulmonary clinic. EXCLI JOURNAL 2017; 16:981-985. [PMID: 28900378 PMCID: PMC5579404 DOI: 10.17179/excli2017-425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 06/17/2017] [Indexed: 12/02/2022]
Affiliation(s)
- Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology,Sydney, Ultimo NSW 2007, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW2308, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, J Lot 1 Kookaburra Circuit, New Lambton Heights, Newcastle, NSW 2305, Australia
| | - Nicole G. Hansbro
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW2308, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, J Lot 1 Kookaburra Circuit, New Lambton Heights, Newcastle, NSW 2305, Australia
| | - Philip M. Hansbro
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW2308, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, J Lot 1 Kookaburra Circuit, New Lambton Heights, Newcastle, NSW 2305, Australia
| |
Collapse
|
30
|
Al-Kouba J, Wilkinson AN, Starkey MR, Rudraraju R, Werder RB, Liu X, Law SC, Horvat JC, Brooks JF, Hill GR, Davies JM, Phipps S, Hansbro PM, Steptoe RJ. Allergen-encoding bone marrow transfer inactivates allergic T cell responses, alleviating airway inflammation. JCI Insight 2017; 2:85742. [PMID: 28570267 DOI: 10.1172/jci.insight.85742] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 04/25/2017] [Indexed: 01/09/2023] Open
Abstract
Memory Th2 cell responses underlie the development and perpetuation of allergic diseases. Because these states result from immune dysregulation, established Th2 cell responses represent a significant challenge for conventional immunotherapies. New approaches that overcome the detrimental effects of immune dysregulation are required. We tested whether memory Th2 cell responses were silenced using a therapeutic approach where allergen expression in DCs is transferred to sensitized recipients using BM cells as a vector for therapeutic gene transfer. Development of allergen-specific Th2 responses and allergen-induced airway inflammation was blocked by expression of allergen in DCs. Adoptive transfer studies showed that Th2 responses were inactivated by a combination of deletion and induction of T cell unresponsiveness. Transfer of BM encoding allergen expression targeted to DCs terminated, in an allergen-specific manner, Th2 responses in sensitized recipients. Importantly, when preexisting airway inflammation was present, there was effective silencing of Th2 cell responses, airway inflammation was alleviated, and airway hyperreactivity was reversed. The effectiveness of DC-targeted allergen expression to terminate established Th2 responses in sensitized animals indicates that exploiting cell-intrinsic T cell tolerance pathways could lead to development of highly effective immunotherapies.
Collapse
Affiliation(s)
- Jane Al-Kouba
- The University of Queensland Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Andrew N Wilkinson
- The University of Queensland Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Malcolm R Starkey
- Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Rajeev Rudraraju
- The University of Queensland Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Rhiannon B Werder
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Xiao Liu
- The University of Queensland Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Soi-Cheng Law
- The University of Queensland Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Jay C Horvat
- Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Jeremy F Brooks
- The University of Queensland Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Geoffrey R Hill
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Janet M Davies
- School of Medicine, University of Queensland, Brisbane, Australia
| | - Simon Phipps
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Philip M Hansbro
- Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Raymond J Steptoe
- The University of Queensland Diamantina Institute, University of Queensland, Brisbane, Australia
| |
Collapse
|
31
|
Ali MK, Kim RY, Karim R, Mayall JR, Martin KL, Shahandeh A, Abbasian F, Starkey MR, Loustaud-Ratti V, Johnstone D, Milward EA, Hansbro PM, Horvat JC. Role of iron in the pathogenesis of respiratory disease. Int J Biochem Cell Biol 2017; 88:181-195. [PMID: 28495571 DOI: 10.1016/j.biocel.2017.05.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 05/01/2017] [Accepted: 05/03/2017] [Indexed: 12/13/2022]
Abstract
Iron is essential for many biological processes, however, too much or too little iron can result in a wide variety of pathological consequences, depending on the organ system, tissue or cell type affected. In order to reduce pathogenesis, iron levels are tightly controlled in throughout the body by regulatory systems that control iron absorption, systemic transport and cellular uptake and storage. Altered iron levels and/or dysregulated homeostasis have been associated with several lung diseases, including chronic obstructive pulmonary disease, lung cancer, cystic fibrosis, idiopathic pulmonary fibrosis and asthma. However, the mechanisms that underpin these associations and whether iron plays a key role in the pathogenesis of lung disease are yet to be fully elucidated. Furthermore, in order to survive and replicate, pathogenic micro-organisms have evolved strategies to source host iron, including freeing iron from cells and proteins that store and transport iron. To counter these microbial strategies, mammals have evolved immune-mediated defence mechanisms that reduce iron availability to pathogens. This interplay between iron, infection and immunity has important ramifications for the pathogenesis and management of human respiratory infections and diseases. An increased understanding of the role that iron plays in the pathogenesis of lung disease and respiratory infections may help inform novel therapeutic strategies. Here we review the clinical and experimental evidence that highlights the potential importance of iron in respiratory diseases and infections.
Collapse
Affiliation(s)
- Md Khadem Ali
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Richard Y Kim
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Rafia Karim
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Jemma R Mayall
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Kristy L Martin
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Ali Shahandeh
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Firouz Abbasian
- Global Centre for Environmental Remediation, Faculty of Science, the University of Newcastle, Callaghan, NSW 2308, Australia
| | - Malcolm R Starkey
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | | | - Daniel Johnstone
- Bosch Institute and Discipline of Physiology, The University of Sydney, Sydney NSW 2000, Australia
| | - Elizabeth A Milward
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Philip M Hansbro
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia
| | - Jay C Horvat
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan NSW 2308, Australia.
| |
Collapse
|
32
|
Hsu ACY, Dua K, Starkey MR, Haw TJ, Nair PM, Nichol K, Zammit N, Grey ST, Baines KJ, Foster PS, Hansbro PM, Wark PA. MicroRNA-125a and -b inhibit A20 and MAVS to promote inflammation and impair antiviral response in COPD. JCI Insight 2017; 2:e90443. [PMID: 28405612 DOI: 10.1172/jci.insight.90443] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Influenza A virus (IAV) infections lead to severe inflammation in the airways. Patients with chronic obstructive pulmonary disease (COPD) characteristically have exaggerated airway inflammation and are more susceptible to infections with severe symptoms and increased mortality. The mechanisms that control inflammation during IAV infection and the mechanisms of immune dysregulation in COPD are unclear. We found that IAV infections lead to increased inflammatory and antiviral responses in primary bronchial epithelial cells (pBECs) from healthy nonsmoking and smoking subjects. In pBECs from COPD patients, infections resulted in exaggerated inflammatory but deficient antiviral responses. A20 is an important negative regulator of NF-κB-mediated inflammatory but not antiviral responses, and A20 expression was reduced in COPD. IAV infection increased the expression of miR-125a or -b, which directly reduced the expression of A20 and mitochondrial antiviral signaling (MAVS), and caused exaggerated inflammation and impaired antiviral responses. These events were replicated in vivo in a mouse model of experimental COPD. Thus, miR-125a or -b and A20 may be targeted therapeutically to inhibit excessive inflammatory responses and enhance antiviral immunity in IAV infections and in COPD.
Collapse
Affiliation(s)
- Alan C-Y Hsu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Kamal Dua
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Malcolm R Starkey
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Tatt-Jhong Haw
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Prema M Nair
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Kristy Nichol
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Nathan Zammit
- Transplantation Immunology Group, Immunology Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Shane T Grey
- Transplantation Immunology Group, Immunology Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Katherine J Baines
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Peter A Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia
| |
Collapse
|
33
|
Shukla SD, Budden KF, Neal R, Hansbro PM. Microbiome effects on immunity, health and disease in the lung. Clin Transl Immunology 2017; 6:e133. [PMID: 28435675 PMCID: PMC5382435 DOI: 10.1038/cti.2017.6] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 02/02/2017] [Accepted: 02/05/2017] [Indexed: 12/14/2022] Open
Abstract
Chronic respiratory diseases, including asthma, chronic obstructive pulmonary disease (COPD) and cystic fibrosis (CF), are among the leading causes of mortality and morbidity worldwide. In the past decade, the interest in the role of microbiome in maintaining lung health and in respiratory diseases has grown exponentially. The advent of sophisticated multiomics techniques has enabled the identification and characterisation of microbiota and their roles in respiratory health and disease. Furthermore, associations between the microbiome of the lung and gut, as well as the immune cells and mediators that may link these two mucosal sites, appear to be important in the pathogenesis of lung conditions. Here we review the recent evidence of the role of normal gastrointestinal and respiratory microbiome in health and how dysbiosis affects chronic pulmonary diseases. The potential implications of host and environmental factors such as age, gender, diet and use of antibiotics on the composition and overall functionality of microbiome are also discussed. We summarise how microbiota may mediate the dynamic process of immune development and/or regulation focusing on recent data from both clinical human studies and translational animal studies. This furthers the understanding of the pathogenesis of chronic pulmonary diseases and may yield novel avenues for the utilisation of microbiota as potential therapeutic interventions.
Collapse
Affiliation(s)
- Shakti D Shukla
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Kurtis F Budden
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Rachael Neal
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
34
|
Inflammasomes in the lung. Mol Immunol 2017; 86:44-55. [PMID: 28129896 DOI: 10.1016/j.molimm.2017.01.014] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 12/11/2022]
Abstract
Innate immune responses act as first line defences upon exposure to potentially noxious stimuli. The innate immune system has evolved numerous intracellular and extracellular receptors that undertake surveillance for potentially damaging particulates. Inflammasomes are intracellular innate immune multiprotein complexes that form and are activated following interaction with these stimuli. Inflammasome activation leads to the cleavage of pro-IL-1β and release of the pro-inflammatory cytokine, IL-1β, which initiates acute phase pro-inflammatory responses, and other responses are also involved (IL-18, pyroptosis). However, excessive activation of inflammasomes can result in chronic inflammation, which has been implicated in a range of chronic inflammatory diseases. The airways are constantly exposed to a wide variety of stimuli. Inflammasome activation and downstream responses clears these stimuli. However, excessive activation may drive the pathogenesis of chronic respiratory diseases such as severe asthma and chronic obstructive pulmonary disease. Thus, there is currently intense interest in the role of inflammasomes in chronic inflammatory lung diseases and in their potential for therapeutic targeting. Here we review the known associations between inflammasome-mediated responses and the development and exacerbation of chronic lung diseases.
Collapse
|
35
|
Gangwar RS, Landolina N, Arpinati L, Levi-Schaffer F. Mast cell and eosinophil surface receptors as targets for anti-allergic therapy. Pharmacol Ther 2016; 170:37-63. [PMID: 27773785 DOI: 10.1016/j.pharmthera.2016.10.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Roopesh Singh Gangwar
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Nadine Landolina
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Ludovica Arpinati
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Francesca Levi-Schaffer
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel.
| |
Collapse
|
36
|
Starkey MR, Nguyen DH, Brown AC, Essilfie AT, Kim RY, Yagita H, Horvat JC, Hansbro PM. Programmed Death Ligand 1 Promotes Early-Life Chlamydia Respiratory Infection-Induced Severe Allergic Airway Disease. Am J Respir Cell Mol Biol 2016; 54:493-503. [PMID: 26378990 DOI: 10.1165/rcmb.2015-0204oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Chlamydia infections are frequent causes of respiratory illness, particularly pneumonia in infants, and are linked to permanent reductions in lung function and the induction of asthma. However, the immune responses that protect against early-life infection and the mechanisms that lead to chronic lung disease are incompletely understood. In the current study, we investigated the role of programmed death (PD)-1 and its ligands PD-L1 and PD-L2 in promoting early-life Chlamydia respiratory infection, and infection-induced airway hyperresponsiveness (AHR) and severe allergic airway disease in later life. Infection increased PD-1 and PD-L1, but not PD-L2, mRNA expression in the lung. Flow cytometric analysis of whole lung homogenates identified monocytes, dendritic cells, CD4(+), and CD8(+) T cells as major sources of PD-1 and PD-L1. Inhibition of PD-1 and PD-L1, but not PD-L2, during infection ablated infection-induced AHR in later life. Given that PD-L1 was the most highly up-regulated and its targeting prevented infection-induced AHR, subsequent analyses focused on this ligand. Inhibition of PD-L1 had no effect on Chlamydia load but suppressed infection-induced pulmonary inflammation. Infection decreased the levels of the IL-13 decoy receptor in the lung, which were restored to baseline levels by inhibition of PD-L1. Finally, inhibition of PD-L1 during infection prevented subsequent infection-induced severe allergic airways disease in later life by decreasing IL-13 levels, Gob-5 expression, mucus production, and AHR. Thus, early-life Chlamydia respiratory infection-induced PD-L1 promotes severe inflammation during infection, permanent reductions in lung function, and the development of more severe allergic airway disease in later life.
Collapse
Affiliation(s)
- Malcolm R Starkey
- 1 Center for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia; and
| | - Duc H Nguyen
- 1 Center for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia; and
| | - Alexandra C Brown
- 1 Center for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia; and
| | - Ama-Tawiah Essilfie
- 1 Center for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia; and
| | - Richard Y Kim
- 1 Center for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia; and
| | - Hideo Yagita
- 2 Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Jay C Horvat
- 1 Center for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia; and
| | - Philip M Hansbro
- 1 Center for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia; and
| |
Collapse
|
37
|
Kim RY, Rae B, Neal R, Donovan C, Pinkerton J, Balachandran L, Starkey MR, Knight DA, Horvat JC, Hansbro PM. Elucidating novel disease mechanisms in severe asthma. Clin Transl Immunology 2016; 5:e91. [PMID: 27525064 PMCID: PMC4973321 DOI: 10.1038/cti.2016.37] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/05/2016] [Accepted: 05/05/2016] [Indexed: 02/06/2023] Open
Abstract
Corticosteroids are broadly active and potent anti-inflammatory agents that, despite the introduction of biologics, remain as the mainstay therapy for many chronic inflammatory diseases, including inflammatory bowel diseases, nephrotic syndrome, rheumatoid arthritis, chronic obstructive pulmonary disease and asthma. Significantly, there are cohorts of these patients with poor sensitivity to steroid treatment even with high doses, which can lead to many iatrogenic side effects. The dose-limiting toxicity of corticosteroids, and the lack of effective therapeutic alternatives, leads to substantial excess morbidity and healthcare expenditure. We have developed novel murine models of respiratory infection-induced, severe, steroid-resistant asthma that recapitulate the hallmark features of the human disease. These models can be used to elucidate novel disease mechanisms and identify new therapeutic targets in severe asthma. Hypothesis-driven studies can elucidate the roles of specific factors and pathways. Alternatively, 'Omics approaches can be used to rapidly generate new targets. Similar approaches can be used in other diseases.
Collapse
Affiliation(s)
- Richard Y Kim
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, University of Newcastle , Newcastle, New South Wales, Australia
| | - Brittany Rae
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, University of Newcastle , Newcastle, New South Wales, Australia
| | - Rachel Neal
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, University of Newcastle , Newcastle, New South Wales, Australia
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, University of Newcastle , Newcastle, New South Wales, Australia
| | - James Pinkerton
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, University of Newcastle , Newcastle, New South Wales, Australia
| | - Lohis Balachandran
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, University of Newcastle , Newcastle, New South Wales, Australia
| | - Malcolm R Starkey
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, University of Newcastle , Newcastle, New South Wales, Australia
| | - Darryl A Knight
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, University of Newcastle , Newcastle, New South Wales, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, University of Newcastle , Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, University of Newcastle , Newcastle, New South Wales, Australia
| |
Collapse
|
38
|
A pathogenic role for tumor necrosis factor-related apoptosis-inducing ligand in chronic obstructive pulmonary disease. Mucosal Immunol 2016; 9:859-72. [PMID: 26555706 DOI: 10.1038/mi.2015.111] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 09/18/2015] [Indexed: 02/04/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a life-threatening inflammatory respiratory disorder, often induced by cigarette smoke (CS) exposure. The development of effective therapies is impaired by a lack of understanding of the underlining mechanisms. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a cytokine with inflammatory and apoptotic properties. We interrogated a mouse model of CS-induced experimental COPD and human tissues to identify a novel role for TRAIL in COPD pathogenesis. CS exposure of wild-type mice increased TRAIL and its receptor messenger RNA (mRNA) expression and protein levels, as well as the number of TRAIL(+)CD11b(+) monocytes in the lung. TRAIL and its receptor mRNA were also increased in human COPD. CS-exposed TRAIL-deficient mice had decreased pulmonary inflammation, pro-inflammatory mediators, emphysema-like alveolar enlargement, and improved lung function. TRAIL-deficient mice also developed spontaneous small airway changes with increased epithelial cell thickness and collagen deposition, independent of CS exposure. Importantly, therapeutic neutralization of TRAIL, after the establishment of early-stage experimental COPD, reduced pulmonary inflammation, emphysema-like alveolar enlargement, and small airway changes. These data provide further evidence for TRAIL being a pivotal inflammatory factor in respiratory diseases, and the first preclinical evidence to suggest that therapeutic agents that target TRAIL may be effective in COPD therapy.
Collapse
|
39
|
Thorburn AN, Tseng HY, Donovan C, Hansbro NG, Jarnicki AG, Foster PS, Gibson PG, Hansbro PM. TLR2, TLR4 AND MyD88 Mediate Allergic Airway Disease (AAD) and Streptococcus pneumoniae-Induced Suppression of AAD. PLoS One 2016; 11:e0156402. [PMID: 27309732 PMCID: PMC4911048 DOI: 10.1371/journal.pone.0156402] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 05/15/2016] [Indexed: 12/25/2022] Open
Abstract
Background Exposure to non-pathogenic Streptococcus pneumoniae and vaccination are inversely associated with asthma. Studies in animal models demonstrate that airway administration of S. pneumoniae (live or killed), or its vaccines or components, suppresses the characteristic features of asthma in mouse models of allergic airway disease (AAD). These components could be developed into immunoregulatory therapies. S. pneumoniae components are recognized by Toll-like receptors (TLR) 2 and TLR4, and both induce inflammatory cell responses through the adaptor protein myeloid differentiation primary response gene 88 (MyD88). The involvement of TLR2, TLR4 and MyD88 in the pathogenesis of AAD and asthma is incompletely understood, and has not been studied in S. pneumoniae-mediated suppression of AAD. We investigated the role of TLR2, TLR4 and MyD88 in the development of AAD and S. pneumoniae-mediated suppression of AAD. Methods and Findings OVA-induced AAD and killed S. pneumoniae-mediated suppression of AAD were assessed in wild-type, TLR2-/-, TLR4-/-, TLR2/4-/- and MyD88-/- BALB/c mice. During OVA-induced AAD, TLR2, TLR4 and MyD88 were variously involved in promoting eosinophil accumulation in bronchoalveolar lavage fluid and blood, and T-helper type (Th)2 cytokine release from mediastinal lymph node T cells and splenocytes. However, all were required for the induction of airways hyperresponsiveness (AHR). In S. pneumoniae-mediated suppression of AAD, TLR2, TLR4 and MyD88 were variously involved in the suppression of eosinophilic and splenocyte Th2 responses but all were required for the reduction in AHR. Conclusions These results highlight important but complex roles for TLR2, TLR4 and MyD88 in promoting the development of OVA-induced AAD, but conversely in the S. pneumoniae-mediated suppression of AAD, with consistent and major contributions in both the induction and suppression of AHR. Thus, TLR signaling is likely required for both the development of asthma and the suppression of asthma by S. pneumoniae, and potentially other immunoregulatory therapies.
Collapse
Affiliation(s)
- Alison N. Thorburn
- The Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Hsin-Yi Tseng
- The Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Chantal Donovan
- The Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Nicole G. Hansbro
- The Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Andrew G. Jarnicki
- The Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Paul S. Foster
- The Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Peter G. Gibson
- The Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Philip M. Hansbro
- The Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
- * E-mail:
| |
Collapse
|
40
|
Kim RY, Horvat JC, Pinkerton JW, Starkey MR, Essilfie AT, Mayall JR, Nair PM, Hansbro NG, Jones B, Haw TJ, Sunkara KP, Nguyen TH, Jarnicki AG, Keely S, Mattes J, Adcock IM, Foster PS, Hansbro PM. MicroRNA-21 drives severe, steroid-insensitive experimental asthma by amplifying phosphoinositide 3-kinase-mediated suppression of histone deacetylase 2. J Allergy Clin Immunol 2016; 139:519-532. [PMID: 27448447 DOI: 10.1016/j.jaci.2016.04.038] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 04/17/2016] [Accepted: 04/29/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND Severe steroid-insensitive asthma is a substantial clinical problem. Effective treatments are urgently required, however, their development is hampered by a lack of understanding of the mechanisms of disease pathogenesis. Steroid-insensitive asthma is associated with respiratory tract infections and noneosinophilic endotypes, including neutrophilic forms of disease. However, steroid-insensitive patients with eosinophil-enriched inflammation have also been described. The mechanisms that underpin infection-induced, severe steroid-insensitive asthma can be elucidated by using mouse models of disease. OBJECTIVE We sought to develop representative mouse models of severe, steroid-insensitive asthma and to use them to identify pathogenic mechanisms and investigate new treatment approaches. METHODS Novel mouse models of Chlamydia, Haemophilus influenzae, influenza, and respiratory syncytial virus respiratory tract infections and ovalbumin-induced, severe, steroid-insensitive allergic airway disease (SSIAAD) in BALB/c mice were developed and interrogated. RESULTS Infection induced increases in the levels of microRNA (miRNA)-21 (miR-21) expression in the lung during SSIAAD, whereas expression of the miR-21 target phosphatase and tensin homolog was reduced. This was associated with an increase in levels of phosphorylated Akt, an indicator of phosphoinositide 3-kinase (PI3K) activity, and decreased nuclear histone deacetylase (HDAC)2 levels. Treatment with an miR-21-specific antagomir (Ant-21) increased phosphatase and tensin homolog levels. Treatment with Ant-21, or the pan-PI3K inhibitor LY294002, reduced PI3K activity and restored HDAC2 levels. This led to suppression of airway hyperresponsiveness and restored steroid sensitivity to allergic airway disease. These observations were replicated with SSIAAD associated with 4 different pathogens. CONCLUSION We identify a previously unrecognized role for an miR-21/PI3K/HDAC2 axis in SSIAAD. Our data highlight miR-21 as a novel therapeutic target for the treatment of this form of asthma.
Collapse
Affiliation(s)
- Richard Y Kim
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - James W Pinkerton
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Malcolm R Starkey
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Ama T Essilfie
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Jemma R Mayall
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Prema M Nair
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Bernadette Jones
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Tatt Jhong Haw
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Krishna P Sunkara
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Thi Hiep Nguyen
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Andrew G Jarnicki
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Simon Keely
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Joerg Mattes
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Ian M Adcock
- Airways Disease Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia.
| |
Collapse
|
41
|
Abstract
Chronic bacterial infection is implicated in both the development and severity of asthma. The atypical bacteria Mycoplasma pneumoniae and Chlamydophila pneumoniae have been identified in the airways of asthmatics and correlated with clinical features such as adult onset, exacerbation risks, steroid sensitivity, and symptom control. Asthmatic patients with evidence of bacterial infection may benefit from antibiotic treatment directed towards these atypical organisms. Examination of the airway microbiome may identify microbial communities that confer risk for or protection from severe asthma.
Collapse
|
42
|
Zhang C, Zhang LH, Wu YF, Lai TW, Wang HS, Xiao H, Che LQ, Ying SM, Li W, Chen ZH, Shen HH. Suhuang antitussive capsule at lower doses attenuates airway hyperresponsiveness, inflammation, and remodeling in a murine model of chronic asthma. Sci Rep 2016; 6:21515. [PMID: 26861679 PMCID: PMC4748281 DOI: 10.1038/srep21515] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 01/25/2016] [Indexed: 11/09/2022] Open
Abstract
Suhuang antitussive capsule (Suhuang), a traditional Chinese medication, is found effective in treating chronic cough and cough variant asthma (CVA). This study aimed to determine the possible effects and underlying mechanisms of Suhuang on chronic ovalbumin (OVA)-induced airway hyperresponsiveness (AHR), inflammation, and remodeling in mice. Mice were randomly assigned to six experimental groups: control, OVA model with or without Suhuang (low dose: 3.5 g/kg, middle dose: 7.0 g/kg, high dose: 14.0 g/kg), or dexamethasone (2.5 mg/kg). AHR, inflammatory cells, cytokines in bronchoalveolar lavage fluid (BALF), lung pathology, mucus production, and airway remodeling were examined. We found Suhuang treated at lower doses effectively inhibited OVA-induced AHR, airway inflammation, mucus production and collagen deposition around the airway. High dose of Suhuang reduced most of the inflammatory hallmarks while exerted inconsiderable effects on the number of macrophages in BALF and AHR. At all doses, Suhuang significantly reduced the levels of interlukin (IL) -13 and transforming growth factor (TGF)-β1, but had little effects on IL-4, IL-5, IL-17A and interferon (IFN)-γ. Thus, Suhuang administration alleviates the pathological changes of chronic asthma likely through inhibition of IL-13 and TGF-β1. Suhuang might be a promising therapy for patients with allergic asthma in the future.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Lan-Hong Zhang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Yin-Fang Wu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian-Wen Lai
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Hai-Sheng Wang
- Yangtze River Pharmaceutical Group Beijing Haiyan Pharmaceutical Co., Ltd, Beijing, China
| | - Hui Xiao
- Yangtze River Pharmaceutical Group Beijing Haiyan Pharmaceutical Co., Ltd, Beijing, China
| | - Luan-Qing Che
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Song-Min Ying
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Wen Li
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi-Hua Chen
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Hua-Hao Shen
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China.,State Key Lab of Respiratory Disease, Guangzhou, China
| |
Collapse
|
43
|
Abstract
Chlamydia pneumoniae, an obligate intracellular bacterial pathogen, has long been investigated as a potential developmental or exacerbating factor in various pathologies. Its unique lifestyle and ability to disseminate throughout the host while persisting in relative safety from the immune response has placed this obligate intracellular pathogen in the crosshairs as a potentially mitigating factor in chronic inflammatory diseases. Many animal model and human correlative studies have been performed to confirm or deny a role for C. pneumoniae infection in these disorders. In some cases, antibiotic clinical trials were conducted to prove a link between bacterial infections and atherosclerosis. In this review, we detail the latest information regarding the potential role that C. pneumoniae infection may have in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Rebecca A Porritt
- Division of Pediatric Infectious Diseases and Immunology, Burns and Allen Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Timothy R Crother
- Division of Pediatric Infectious Diseases and Immunology, Burns and Allen Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| |
Collapse
|
44
|
Pulmonary immunity during respiratory infections in early life and the development of severe asthma. Ann Am Thorac Soc 2015; 11 Suppl 5:S297-302. [PMID: 25525736 DOI: 10.1513/annalsats.201402-086aw] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Asthma affects 10% of the population in Westernized countries, being most common in children. It is a heterogeneous condition characterized by chronic allergic airway inflammation, mucus hypersecretion, and airway hyperresponsiveness (AHR) to normally innocuous antigens. Combination therapies with inhaled corticosteroids and bronchodilators effectively manage mild to moderate asthma, but there are no cures, and patients with severe asthma do not respond to these treatments. The inception of asthma is linked to respiratory viral (respiratory syncytial virus, rhinovirus) and bacterial (Chlamydia, Mycoplasma) infections. The examination of mouse models of early-life infections and allergic airway disease (AAD) provides valuable insights into the mechanisms of disease inception that may lead to the development of more effective therapeutics. For example, early-life, but not adult, Chlamydia respiratory infections in mice permanently modify immunity and lung physiology. This increases the severity of AAD by promoting IL-13 expression, mucus hypersecretion, and AHR. We have identified novel roles for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and IL-13 in promoting infection-induced pathology in early life and subsequent chronic lung disease. Genetic deletion of TRAIL or IL-13 variously protected against neonatal infection-induced inflammation, mucus hypersecretion, altered lung structure, AHR, and impaired lung function. Therapeutic neutralization of these factors prevented infection-induced severe AAD. Other novel mechanisms and avenues for intervention are also being explored. Such studies indicate the immunological mechanisms that may underpin the association between early-life respiratory infections and the development of more severe asthma and may facilitate the development of tailored preventions and treatments.
Collapse
|
45
|
Essilfie AT, Horvat JC, Kim RY, Mayall JR, Pinkerton JW, Beckett EL, Starkey MR, Simpson JL, Foster PS, Gibson PG, Hansbro PM. Macrolide therapy suppresses key features of experimental steroid-sensitive and steroid-insensitive asthma. Thorax 2015; 70:458-67. [PMID: 25746630 DOI: 10.1136/thoraxjnl-2014-206067] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 02/12/2015] [Indexed: 11/04/2022]
Abstract
BACKGROUND Steroid-insensitive endotypes of asthma are an important clinical problem and effective therapies are required. They are associated with bacterial infection and non-eosinophilic inflammatory responses in the asthmatic lung. Macrolide therapy is effective in steroid-insensitive endotypes, such as non-eosinophilic asthma. However, whether the effects of macrolides are due to antimicrobial or anti-inflammatory mechanisms is not known. OBJECTIVE To determine and assess the efficacy of macrolide (ie, clarithromycin) and non-macrolide (ie, amoxicillin) antibiotic treatments in experimental models of infection-induced, severe, steroid-insensitive neutrophilic allergic airways disease (SSIAAD), compared with steroid-sensitive AAD and to delineate the antimicrobial and anti-inflammatory effects of macrolide therapy. METHODS We developed and used novel mouse models of Chlamydia and Haemophilus lung infection-induced SSIAAD. We used these models to investigate the effects of clarithromycin and amoxicillin treatment on immune responses and airways hyper-responsiveness (AHR) in Ova-induced, T helper lymphocyte (Th) 2 -associated steroid-sensitive AAD and infection-induced Th1/Th17-associated SSIAAD compared with dexamethasone treatment. RESULTS Clarithromycin and amoxicillin had similar antimicrobial effects on infection. Amoxicillin did attenuate some features, but did not broadly suppress either form of AAD. It did restore steroid sensitivity in SSIAAD by reducing infection. In contrast, clarithromycin alone widely suppressed inflammation and AHR in both steroid-sensitive AAD and SSIAAD. This occurred through reductions in Th2 responses that drive steroid-sensitive eosinophilic AAD and tumour necrosis factor α and interleukin 17 responses that induce SSIAAD. CONCLUSIONS Macrolides have broad anti-inflammatory effects in AAD that are likely independent of their antimicrobial effects. The specific responses that are suppressed are dependent upon the responses that dominate during AAD.
Collapse
Affiliation(s)
- Ama-Tawiah Essilfie
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Jay C Horvat
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Richard Y Kim
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Jemma R Mayall
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - James W Pinkerton
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Emma L Beckett
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Malcolm R Starkey
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Jodie L Simpson
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Paul S Foster
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Peter G Gibson
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
46
|
Ranasinghe C, Trivedi S, Wijesundara DK, Jackson RJ. IL-4 and IL-13 receptors: Roles in immunity and powerful vaccine adjuvants. Cytokine Growth Factor Rev 2014; 25:437-42. [DOI: 10.1016/j.cytogfr.2014.07.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 07/15/2014] [Indexed: 01/01/2023]
|
47
|
Starkey MR, Nguyen DH, Essilfie AT, Kim RY, Hatchwell LM, Collison AM, Yagita H, Foster PS, Horvat JC, Mattes J, Hansbro PM. Tumor necrosis factor-related apoptosis-inducing ligand translates neonatal respiratory infection into chronic lung disease. Mucosal Immunol 2014; 7:478-88. [PMID: 24045576 DOI: 10.1038/mi.2013.65] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Revised: 07/24/2013] [Accepted: 08/13/2013] [Indexed: 02/04/2023]
Abstract
Respiratory infections in early life can lead to chronic respiratory disease. Chlamydia infections are common causes of respiratory disease, particularly pneumonia in neonates, and are linked to permanent reductions in pulmonary function and the induction of asthma. However, the immune responses that protect against early-life infection and the mechanisms that lead to chronic lung disease are incompletely understood. Here we identify novel roles for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in promoting Chlamydia respiratory infection-induced pathology in early life, and subsequent chronic lung disease. By infecting TRAIL-deficient neonatal mice and using neutralizing antibodies against this factor and its receptors in wild-type mice, we demonstrate that TRAIL is critical in promoting infection-induced histopathology, inflammation, and mucus hypersecretion, as well as subsequent alveolar enlargement and impaired lung function. This suggests that therapeutic agents that target TRAIL or its receptors may be effective treatments for early-life respiratory infections and associated chronic lung disease.
Collapse
Affiliation(s)
- M R Starkey
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - D H Nguyen
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - A T Essilfie
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - R Y Kim
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - L M Hatchwell
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - A M Collison
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - H Yagita
- Department of Immunology, Juntendo University School of Medicine, Hongo, Bunkyo-ku, Tokyo, Japan
| | - P S Foster
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - J C Horvat
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| | - J Mattes
- 1] Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia [2] Pediatric Respiratory and Sleep Medicine Unit, Newcastle Children's Hospital, Kaleidoscope, New Lambton Heights, Newcastle, New South Wales, Australia
| | - P M Hansbro
- Priority Research Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Newcastle, New South Wales, Australia
| |
Collapse
|
48
|
Heijink IH, Nawijn MC, Hackett TL. Airway epithelial barrier function regulates the pathogenesis of allergic asthma. Clin Exp Allergy 2014; 44:620-30. [DOI: 10.1111/cea.12296] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- I. H. Heijink
- Department of Pathology and Medical Biology; Experimental Pulmonology and Inflammation Research; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
- Department of Pulmonology; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
- GRIAC Research Institute; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
| | - M. C. Nawijn
- Department of Pathology and Medical Biology; Experimental Pulmonology and Inflammation Research; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
- GRIAC Research Institute; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
| | - T.-L. Hackett
- Centre for Heart Lung Innovation; St Paul's Hospital; University of British Columbia; Vancouver BC Canada
| |
Collapse
|
49
|
Hallstrand TS, Hackett TL, Altemeier WA, Matute-Bello G, Hansbro PM, Knight DA. Airway epithelial regulation of pulmonary immune homeostasis and inflammation. Clin Immunol 2014; 151:1-15. [PMID: 24503171 DOI: 10.1016/j.clim.2013.12.003] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Accepted: 12/04/2013] [Indexed: 11/23/2022]
Abstract
Recent genetic, structural and functional studies have identified the airway and lung epithelium as a key orchestrator of the immune response. Further, there is now strong evidence that epithelium dysfunction is involved in the development of inflammatory disorders of the lung. Here we review the characteristic immune responses that are orchestrated by the epithelium in response to diverse triggers such as pollutants, cigarette smoke, bacterial peptides, and viruses. We focus in part on the role of epithelium-derived interleukin (IL)-25, IL-33 and thymic stromal lymphopoietin (TSLP), as well as CC family chemokines as critical regulators of the immune response. We cite examples of the function of the epithelium in host defense and the role of epithelium dysfunction in the development of inflammatory diseases.
Collapse
Affiliation(s)
- Teal S Hallstrand
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA, USA.
| | - Tillie L Hackett
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - William A Altemeier
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA, USA
| | - Gustavo Matute-Bello
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA, USA
| | - Philip M Hansbro
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
50
|
Starkey MR, Jarnicki AG, Essilfie AT, Gellatly SL, Kim RY, Brown AC, Foster PS, Horvat JC, Hansbro PM. Murine models of infectious exacerbations of airway inflammation. Curr Opin Pharmacol 2013; 13:337-44. [PMID: 23566696 DOI: 10.1016/j.coph.2013.03.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 03/08/2013] [Accepted: 03/14/2013] [Indexed: 12/24/2022]
Abstract
Airway inflammation underpins the pathogenesis of the major human chronic respiratory diseases. It is now well recognized that respiratory infections with bacteria and viruses are important in the induction, progression and exacerbation of these diseases. There are no effective therapies that prevent or reverse these events. The development and use of mouse models are proving valuable in understanding the role of infection in disease pathogenesis. They have recently been used to show that infections in early life alter immune responses and lung structure to increase asthma severity, and alter immune responses in later life to induce steroid resistance. Infection following smoke exposure or in experimental chronic obstructive pulmonary disease exacerbates inflammation and remodeling, and worsens cystic fibrosis. Further exploration of these models will facilitate the identification of new therapeutic approaches and the testing of new preventions and treatments.
Collapse
Affiliation(s)
- Malcolm Ronald Starkey
- Centre for Asthma and Respiratory Disease, The Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|