Professor  Nathan Bartlett

Professor Nathan Bartlett

Assistant Dean- Industry Engagement

School of Biomedical Sciences and Pharmacy

The fast-evolving landscape of respiratory virus drug development

For a kid who was fascinated by viruses – he found them “pretty cool but a bit scary” – to now be at the forefront of research that could help protect us from a range of respiratory viruses is the realisation of a long-held ambition for viral immunologist Professor Nathan Bartlett.

Nathan Barlett standing in front of a brown wooden wall with his arms folded

Nathan found lots of fascinatingly scary viruses during an early-career scholarship with the Australian Centre for Disease Preparedness in Geelong, Victoria, an experience which reinforced his interest in working in virology. From there, he took up a postdoctoral position at Oxford University where his research focused primarily on developing viral vector vaccines using the vaccinia virus, a relative of the virus that causes smallpox. During this time, he learnt a great deal about genetic engineering and cemented his interest in molecular virology and immunology of viruses and vaccines.

But he knew there was something missing. He realised he wanted to be involved with clinical-facing translational research, so took up another postdoctoral position at Imperial College London with a researcher who concentrated primarily on viral exacerbations of asthma and chronic obstructive pulmonary disease (COPD). It became clear to Nathan that the logical next step was to shift his focus to respiratory medicine where, happily, he was able to continue working with those ever-fascinating viruses, particularly the most prevalent human respiratory pathogen, rhinovirus – otherwise known as the common cold virus.

Nathan’s experiences from 13 years in the UK set him on the trajectory to where he is now. He had collaborated with University of Newcastle researchers based at HMRI whilst in the UK, and eventually joined the organisation in 2015 after relocating to Newcastle, somewhere he viewed as more “climactically consistent than Geelong and London” where he could see himself settling and raising a family. Since that time, his translational research has centred on treatments for respiratory viruses, specifically rhinovirus.

“Respiratory viruses cause the clinically most impactful transmissible diseases that challenge societies. There is a huge global burden of respiratory viral infections, with some such as influenza attracting a great deal more research than others such as rhinovirus, even though rhinovirus causes a huge burden of illness,” explains Nathan.

From rhinovirus to coronavirus: a novel control and containment treatment

In his role as head of the Viral Immunology and Respiratory Disease group with HMRI, Nathan has been working to understand the early events that influence the outcome of a respiratory infection. What is the interface between the host’s airway and the virus? How do the airway’s epithelial cells sense and then respond to a virus? How is the innate immune response triggered to control, contain and prevent the viral illness from escalating? What determines whether an infection results in a mild cold, or develops into a more serious lung disease such as an asthma attack or COVID-19? He found that a lot of those early events, mediated by innate immunity, share commonalities across all viral pathogens, which enabled him to translate research that was initially focused on rhinovirus and influenza to other viruses, including coronavirus.

Since 2015, Nathan and his industry partner, Ena Respiratory, have been developing a drug to stimulate the innate immune response in the upper respiratory tract to contain a virus, and thus reduce the risk of it spreading from the upper respiratory tract into the lungs where it can escalate to a severe illness. To date, there are no products that can achieve this outcome. The translational research output is a nasal spray, with Phase 1 clinical trials starting in mid-2021.

Nathan describes his vision for the nasal spray as a protective treatment.

“I envisage a day when a person suffering from severe asthma, for example, can walk into a pharmacy and purchase a nasal spray to protect themselves. The product we’re developing will initially be for high-risk individuals including those at high risk of exposure, chronic respiratory disease or immune suppressed. They will administer the nasal spray which will immediately prime for an immune response that facilitates control of the infection in the upper respiratory tract before it has a chance to move through the airways to their lungs.”

When our global village was rapidly engulfed by the coronavirus pandemic early in 2020, Nathan quickly applied the knowledge gained studying the treatment for other respiratory viruses to help show that innate immunity can also be leveraged to tackle coronavirus, offering a potential new treatment approach for COVID-19.

Our community-based virus protective strategies are built around vaccines, but there is a large gap in respiratory virus infection control where vaccines are not available (eg rhinovirus, RSV) or don’t work as effectively as hoped (the elderly and immune suppressed) or are not being used as required (influenza, SARS-CoV-2). This is where the nasal spray will enter the market. It’s important to note that the nasal spray is not intended to take the place of a vaccine. Rather, it will help control and contain a virus, and prevent at-risk groups such as the immunosuppressed, elderly, unvaccinated or those who have not responded well to vaccination from becoming severely ill.

“The aim of the treatment is to keep respiratory viruses from travelling from the nose and throat and entering the lungs where, as we know, it can cause severe illness and death,” Nathan explains.

The real-world application of research: a gratifying experience

After 15 years of researching respiratory viruses, Nathan is proud to say that he has played a significant role in the development of a treatment targeting the upper airways which will prevent these viruses from generating a more serious disease as soon as it causes infection. He is quick to point out that, like all research endeavours, he did not achieve this alone.

He is grateful to the Hunter Medical Research Institute and Asthma Australia who provided the seed funding needed to develop the virus infection models that enabled him to work with Ena Respiratory.

Nathan believes that COVID-19 is going to change the landscape of respiratory virus drug development moving forward. Treatments are now a major global focus, and it’s imperative that research and investment in that area continue to improve.

“I have no doubt that there will be a lot of other medications developed on the back of the urgency around COVID-19. The offshoot of that is that we will soon see better treatments for other viral diseases including RSV bronchiolitis, influenza, and asthma and COPD exacerbations. Playing a role in that has been a gratifying experience.”

Nathan Barlett standing with his arms folded in front of a wooden wall

The fast-evolving landscape of respiratory virus drug development

For a kid who was fascinated by viruses – he found them “pretty cool but a bit scary” – to now be at the forefront of research that could help protect us from a range of respiratory viruses is the realisation of a long-held ambition for viral immunologist Associate Professor Nathan Bartlett.

Read more

Career Summary

Biography

Nathan Bartlett is Professor and head of the Viral Immunology and Respiratory Disease group and is based at the Hunter Medical Research Institute. He also retains an honorary academic appointment at Imperial College London, UK  to continue collaborative research.

Following the award of PhD, Dr Bartlett undertook a 5 year Postdoctoral research position, first at the Sir William Dunn School of Pathology, University of Oxford, then in the Department of Virology, Imperial College London. Dr Bartlett then undertook a second Postdoctoral position in the Department of Respiratory Medicine within the National Heart and Lung Institute (NHLI), also at Imperial.

After joining the NHLI, Dr Bartlett continued to build on his virology training leading to Bartlett et al, Nature Medicine 2008, the world's first mouse rhinovirus infection model. He has investigated the interaction of respiratory viral infection with type-2 immunity to uncover pathogenic mechanisms in asthma exacerbations. He was a co-applicant on several successful project and program grants and since his Lecturer appointment at Imperial in 2011 was successful as the Lead Investigator in achieving an MRC project grant to study the role of IL-25 in asthma exacerbations, the research from which was published as the featured cover story in Science Translational Medicine (October 2014). Dr Bartlett has contributed to several scholarly books including the Rhinovirus chapter for the Encyclopaedia of Virology (Elsevier) and ,edited Rhinovirus Infections: Rethinking Impact on Human Health and Disease (Elsevier). Dr Bartlett is considered a world expert on in vivo and human airway epithelial models of rhinovirus infection and consults for a number companies that are bringing novel therapies for respiratory infection and inflammatory airways diseases to the clinic. He is also an Associate Editor for the American Journal of Physiology - Lung, Cellular and Molecular Physiology (AJP-LUNG) and a member of the European Respiratory Society College of Experts.

In 2015 Dr Bartlett accepted an academic appointment at the University of Newcastle where he has received funding from HMRI, Asthma Australia, NH&MRC and multiple Industry Partners and associated Government grants. He work has led to the publishing of multiple patents related to  treatments currently in development for respiratory virus infections and exacerbations of chronic respiratory diseases.

He is now exploring novel therapies and treatments to fight COVID-19, a novel coronavirus which was declared a pandemic in March 2020. His team is exploring a range of anti-viral options for this virus, partnering with other researchers to explore different methods to fight the virus. For example Dr Bartlett is working with Professor Hubert Hondermarck to assess the safety and efficacy of a range of cancer drugs which have the potential to be repurposed.

Dr Bartlett continues to work with ENA Respiratory (Brandon BioCatalyst-funded company) who have developed an innate immune stimulant that protects the lungs against respiratory virus infections. Boosting lung innate immunity bridges the gap between vaccine mediated long term protection and anti-viral drug to treat active infection. He is now using coronavirus infections models to determine the efficacy of this approach.

He is also helping local industries that are focusing their business to produce disinfectants, sanitizers and virus inactivating materials that will become part of everyday life as we adapt to coexist with new respiratory viruses such as SARS-CoV-2.


Qualifications

  • PhD, Deakin University
  • Bachelor of Science (Honours), Deakin University

Keywords

  • Asthma
  • Epithelium
  • Respiratory virus
  • Rhinovirus
  • Type 2 immunity
  • allergy
  • anti-viral immunity
  • innate immunity

Fields of Research

Code Description Percentage
310706 Virology 40
320407 Innate immunity 40
321299 Ophthalmology and optometry not elsewhere classified 20

Professional Experience

UON Appointment

Title Organisation / Department
Professor University of Newcastle
School of Biomedical Sciences and Pharmacy
Australia

Academic appointment

Dates Title Organisation / Department
20/3/2011 -  Lecturer Imperial College London
United Kingdom
Edit

Publications

For publications that are currently unpublished or in-press, details are shown in italics.


Book (2 outputs)

Year Citation Altmetrics Link
2019 Rhinovirus infections: Rethinking the impact on human health and disease, Academic Press, London (2019)
DOI 10.1016/C2017-0-04472-5
Citations Scopus - 1
2019 Rhinovirus infections: Rethinking the impact on human health and disease, Academic Press, London (2019)
DOI 10.1016/C2017-0-04472-5
Citations Scopus - 1

Chapter (9 outputs)

Year Citation Altmetrics Link
2019 Girkin J, Maltby S, Singanayagam A, Bartlett N, Malia P, 'In vivo experimental models of infection and disease', Rhinovirus infections and disease: Rethinking impact on human health and disease, Elsevier, London 195-238 (2019) [B1]
DOI 10.1016/B978-0-12-816417-4.00008-1
Citations Scopus - 2
Co-authors Steven Maltby, Jason Girkin
2019 Bartlett N, Esneau C, Bochkov Y, 'Rhinovirus structure, replication, and classification', Rhinovirus infections: Rethinking the impact on human health and disease, Elsevier, London 1-23 (2019) [B1]
Citations Scopus - 6
Co-authors Camille Esneau
2019 Reid AT, Sutanto EN, Chander-Veerati P, Looi K, Li NF, Iosifidis T, et al., 'Ground zero-the airway epithelium', Rhinovirus Infections: Rethinking the Impact on Human Health and Disease, Academic Press, Cambridge, MS 61-98 (2019) [B1]
DOI 10.1016/B978-0-12-816417-4.00003-2
Citations Scopus - 4
Co-authors Suling Loo, Punnam Veerati, Andrew Reid
2019 Wark P, Williams T, Pathinayake P, 'The interplay of the host, virus, and the environment', Rhinovirus Infections: Rethinking the Impact on Human Health and Disease, Elsevier, London 169-194 (2019) [B1]
DOI 10.1016/B978-0-12-816417-4.00007-X
Co-authors Prabuddha Pathinayake
2019 McLean G, Girkin J, Solari R, 'Emerging therapeutic approaches', Rhinovirus Infections: Rethinking the Impact on Human Health and Disease, Elsevier, London 239-263 (2019) [B1]
DOI 10.1016/B978-0-12-816417-4.00009-3
Citations Scopus - 2
Co-authors Jason Girkin
2015 Bartlett NW, Singanayagam A, Johnston SL, 'Mouse models of rhinovirus infection and airways disease', Rhinoviruses: Methods and Protocols, Springer, New York 181-188 (2015) [B1]
DOI 10.1007/978-1-4939-1571-2_14
Citations Scopus - 15
2014 Bartlett NW, Johnston SL, 'Rhinoviruses', Reference Module in Biomedical Research (2014)

© 2014 Elsevier Inc. All rights reserved.Human rhinovirus (HRV) infections are the most frequent cause of the common cold, the most common illness affecting mankind with cases doc... [more]

© 2014 Elsevier Inc. All rights reserved.Human rhinovirus (HRV) infections are the most frequent cause of the common cold, the most common illness affecting mankind with cases documented as far back in human history as the ancient Egyptians. Morbidity associated with the illness is a major cause of lost productivity through absenteeism from school or work. More recently, rhinoviruses have been shown to play a significant role in precipitating severe respiratory disease syndromes such as acute exacerbations of asthma. Rhinoviruses belong to the family Picornaviridae. Initially 102 HRV serotypes were identified. Subsequent molecular-based genetic identification has extended the number to over 160 viruses which are divided into 3 species (HRV-A, -B and -C). The immense antigenic variation arising from such a large number of virus types has thwarted efforts to date to develop an anti-HRV vaccine. Viral replication occurs in the cytoplasm and the virions are small non-enveloped icosahedral particles which contain a single-stranded positive-sense RNA genome. The single polyprotein encoded by the RNA genome is cleaved by viral proteases to yield mature proteins. The majority of HRV serotypes require intercellular adhesion molecule 1 for cell binding and entry. Rhinoviruses exhibit a narrow host range infecting only humans and some primates although recently mouse infection models have been developed. Sensitive molecular assays such as polymerase chain reaction (PCR) have significantly increased the ability to detect HRVs direcetly in infected material. Transmitted by aerosol the virus enters via the nose and replicates in the nasal epithelium, inducing production of pro-inflammatory cytokines that contribute to cold symptoms with involvement of lower respiratory tract in susceptible individuals.

DOI 10.1016/B978-0-12-801238-3.02658-1
2008 Bartlett NW, Johnston SL, 'Rhinoviruses', Encyclopedia of Virology: Volume 1-5 V4-467-V4-475 (2008)

Human rhinovirus (HRV) infections are the most frequent cause of the common cold, the most common illness affecting mankind with cases documented as far back in human history as t... [more]

Human rhinovirus (HRV) infections are the most frequent cause of the common cold, the most common illness affecting mankind with cases documented as far back in human history as the ancient Egyptians. Morbidity associated with the illness is a major cause of lost productivity through absenteeism from school or work. More recently, rhinoviruses have been shown to play a significant role in precipitating severe respiratory disease syndromes such as acute exacerbations of asthma. Rhinoviruses belong to the family Picornaviridae. Over 100 serotypes of HRV have been identified, thwarting efforts to date to develop an anti-HRV vaccine. Viral replication occurs in the cytoplasm and the virions are small icosahedral particles which contain a single-stranded positive-sense RNA genome. The single polyprotein encoded by the RNA genome is cleaved by viral proteases to yield mature proteins. The majority of HRV serotypes require intercellular adhesion molecule 1 for cell binding and entry. Rhinoviruses exhibit a narrow host range infecting only humans and some primates. Recently, developed sensitive polymerase chain reaction assays have significantly increased the ability to detect HRVs in infected material. Transmitted by aerosol the virus enters via the nose and replicates in the nasal epithelium, inducing production of pro-inflammatory cytokines causing cold symptoms with involvement of lower respiratory tract in susceptible individuals.

DOI 10.1016/B978-012374410-4.00494-5
2008 Bartlett NW, 'Rhinoviruses', Encyclopedia of Virology, Elsevier, Oxford UK (2008)
Citations Scopus - 4
Show 6 more chapters

Journal article (92 outputs)

Year Citation Altmetrics Link
2023 Blackburn JB, Li NF, Bartlett NW, Richmond BW, 'An update in club cell biology and its potential relevance to chronic obstructive pulmonary disease.', Am J Physiol Lung Cell Mol Physiol, 324 L652-L665 (2023) [C1]
DOI 10.1152/ajplung.00192.2022
Citations Scopus - 3
2023 Girkin JLN, Bryant NE, Loo S-L, Hsu A, Kanwal A, Williams TC, et al., 'Upper Respiratory Tract OC43 Infection Model for Investigating Airway Immune-Modifying Therapies.', Am J Respir Cell Mol Biol, 69 614-622 (2023) [C1]
DOI 10.1165/rcmb.2023-0202MA
Co-authors Steven Maltby, Jason Girkin
2023 Veerati PC, Reid AT, Nichol KS, Wark PAB, Knight DA, Bartlett NW, Grainge CL, 'Mechanical forces suppress antiviral innate immune responses from asthmatic airway epithelial cells following rhinovirus infection.', Am J Physiol Lung Cell Mol Physiol, 325 L206-L214 (2023) [C1]
DOI 10.1152/ajplung.00074.2022
Citations Scopus - 2
Co-authors Punnam Veerati, Christopher Grainge, Andrew Reid
2023 Speck P, Mackenzie J, Bull RA, Slobedman B, Drummer H, Fraser J, et al., 'Statement in Support of: "Virology under the Microscope-a Call for Rational Discourse"', MBIO, 14 (2023)
DOI 10.1128/mbio.00815-23
Co-authors Camille Esneau, Alex Spencer
2023 Speck P, Mackenzie J, Bull RA, Slobedman B, Drummer H, Fraser J, et al., 'Statement in Support of: "Virology under the Microscope-a Call for Rational Discourse"', MSPHERE, 8 (2023)
DOI 10.1128/msphere.00165-23
Co-authors Alex Spencer, Camille Esneau
2023 Speck P, Mackenzie J, Bull RA, Slobedman B, Drummer H, Fraser J, et al., 'Statement in Support of: "Virology under the Microscope-a Call for Rational Discourse".', J Virol, 97 e0045123 (2023)
DOI 10.1128/jvi.00451-23
Co-authors Alex Spencer, Camille Esneau
2023 Bartlett NW, Feghali-Bostwick C, Gunst SJ, 'Call for Papers: "Targeting Airway Immunity in Lung Disease"', AMERICAN JOURNAL OF PHYSIOLOGY-LUNG CELLULAR AND MOLECULAR PHYSIOLOGY, 324 L48-L52 (2023)
DOI 10.1152/ajplung.00375.2022
2023 Antunes KH, Singanayagam A, Williams L, Faiez TS, Farias A, Jackson MM, et al., 'Airway-delivered short-chain fatty acid acetate boosts antiviral immunity during rhinovirus infection.', J Allergy Clin Immunol, 151 447-457.e5 (2023) [C1]
DOI 10.1016/j.jaci.2022.09.026
Citations Scopus - 15Web of Science - 4
Co-authors Lisa Wood, Punnam Veerati
2023 Dy ABC, Girkin J, Marrocco A, Collison A, Mwase C, O'Sullivan MJ, et al., 'Rhinovirus infection induces secretion of endothelin-1 from airway epithelial cells in both in vitro and in vivo models.', Respir Res, 24 205 (2023) [C1]
DOI 10.1186/s12931-023-02510-6
Citations Scopus - 3
Co-authors Adam Collison, Joerg Mattes, Jason Girkin
2023 Malik B, Bartlett NW, Upham JW, Nichol KS, Harrington J, Wark PAB, 'Severe asthma ILC2s demonstrate enhanced proliferation that is modified by biologics.', Respirology, 28 758-766 (2023) [C1]
DOI 10.1111/resp.14506
Citations Scopus - 3Web of Science - 1
2023 Akerman A, Milogiannakis V, Jean T, Esneau C, Silva MR, Ison T, et al., 'Emergence and antibody evasion of BQ, BA.2.75 and SARS-CoV-2 recombinant sub-lineages in the face of maturing antibody breadth at the population level.', EBioMedicine, 90 104545 (2023) [C1]
DOI 10.1016/j.ebiom.2023.104545
Citations Scopus - 16Web of Science - 1
Co-authors Camille Esneau
2022 Williams TC, Loo S-L, Nichol KS, Reid AT, Veerati PC, Esneau C, et al., 'IL-25 blockade augments antiviral immunity during respiratory virus infection', COMMUNICATIONS BIOLOGY, 5 (2022) [C1]
DOI 10.1038/s42003-022-03367-z
Citations Scopus - 8Web of Science - 2
Co-authors Jason Girkin, Punnam Veerati, Suling Loo, Andrew Reid, Camille Esneau, Christopher Grainge
2022 Singanayagam A, Footitt J, Marczynski M, Radicioni G, Cross MT, Finney LJ, et al., 'Airway mucins promote immunopathology in virus-exacerbated chronic obstructive pulmonary disease', JOURNAL OF CLINICAL INVESTIGATION, 132 (2022) [C1]
DOI 10.1172/JCI120901
Citations Scopus - 26Web of Science - 22
2022 Girkin JLN, Maltby S, Bartlett NW, 'Toll-like receptor-agonist-based therapies for respiratory viral diseases: thinking outside the cell', EUROPEAN RESPIRATORY REVIEW, 31 (2022) [C1]
DOI 10.1183/16000617.0274-2021
Citations Scopus - 7Web of Science - 1
Co-authors Steven Maltby, Jason Girkin
2022 Veerati PC, Nichol KS, Read JM, Bartlett NW, Wark PAB, Knight DA, et al., 'Conditionally reprogrammed asthmatic bronchial epithelial cells express lower
DOI 10.1152/ajplung.00230.2022
Citations Scopus - 2
Co-authors Andrew Reid, Punnam Veerati, Christopher Grainge
2022 Aggarwal A, Stella AO, Walker G, Akerman A, Esneau C, Milogiannakis V, et al., 'Platform for isolation and characterization of SARS-CoV-2 variants enables rapid characterization of Omicron in Australia', NATURE MICROBIOLOGY, 7 896-908 (2022) [C1]
DOI 10.1038/s41564-022-01135-7
Citations Scopus - 24Web of Science - 19
Co-authors Camille Esneau
2022 Esneau C, Duff AC, Bartlett NW, 'Understanding Rhinovirus Circulation and Impact on Illness', VIRUSES-BASEL, 14 (2022) [C1]
DOI 10.3390/v14010141
Citations Scopus - 27Web of Science - 6
Co-authors Camille Esneau
2022 Kan S, Grainge C, Nichol K, Reid A, Knight D, Sun Y, et al., 'TLR7 agonist loaded airway epithelial targeting nanoparticles stimulate innate immunity and suppress viral replication in human bronchial epithelial cells', INTERNATIONAL JOURNAL OF PHARMACEUTICS, 617 (2022) [C1]
DOI 10.1016/j.ijpharm.2022.121586
Citations Scopus - 1
Co-authors Christopher Grainge, Roger Liang, Andrew Reid
2022 George PM, Reed A, Desai SR, Devaraj A, Faiez TS, Laverty S, et al., 'A persistent neutrophil-associated immune signature characterizes post-COVID-19 pulmonary sequelae', SCIENCE TRANSLATIONAL MEDICINE, 14 (2022) [C1]
DOI 10.1126/scitranslmed.abo5795
Citations Scopus - 34Web of Science - 16
Co-authors Camille Esneau, Michael Schuliga
2022 Bartlett NW, Bastarache JA, Kuebler WM, Schmidt EP, 'Call for Papers: "In It for the Long Haul: Understanding the Lasting Impact of COVID-19 on Lung Health and Disease"', AMERICAN JOURNAL OF PHYSIOLOGY-LUNG CELLULAR AND MOLECULAR PHYSIOLOGY, 323 L683-L684 (2022)
DOI 10.1152/ajplung.00352.2022
2021 Contoli M, Papi A, Tomassetti L, Rizzo P, Vieceli Dalla Sega F, Fortini F, et al., 'Blood Interferon-a Levels and Severity, Outcomes, and Inflammatory Profiles in Hospitalized COVID-19 Patients', Frontiers in Immunology, 12 (2021) [C1]

Background: Deficient interferon responses have been proposed as one of the relevant mechanisms prompting severe manifestations of COVID-19. Objective: To evaluate the interferon ... [more]

Background: Deficient interferon responses have been proposed as one of the relevant mechanisms prompting severe manifestations of COVID-19. Objective: To evaluate the interferon (IFN)-a levels in a cohort of COVID-19 patients in relation to severity, evolution of the clinical manifestations and immune/inflammatory profile. Methods: This is prospective study recruiting consecutive hospitalized patients with respiratory failure associated with SARS-COV-2 infection and matched controls. After enrollment, patients were assessed every 7 ± 2 days for additional 2 consecutive visits, for a total of 21 days. The severity of the clinical condition was ranked based on the level of respiratory support required. At each time-point blood samples were obtained to assess immune cells and mediators by multiplex immunoassay. Results: Fifty-four COVD-19 and 11 control patients matched for severity were enrolled. At recruitment, lower levels of blood IFN-a were found in COVID-19 patients compared to controls (3.8-fold difference, p < 0.01). Improvements in COVID-19 severity were paralleled by a significant increase of blood IFN-a levels. A significant increase in blood IFN-a was found over the study period in survivors (70% of the study population). A similar trend was found for blood IFN-ß with IFN-ß levels below the threshold of detectability in a substantial proportion of subjects. Significantly higher values of blood lymphocytes and lower levels of IL-10 were found at each time point in patients who survived compared to patients who died. In patients who clinically improved and survived during the study, we found an inverse association between IL-10 and IFN-a levels. Conclusion: The study identifies a blood immune profile defined by deficient IFN-a levels associated with increased IL-10 expression in patients progressing to severe/life threatening COVID-19 conditions, suggesting the involvement of immunological pathways that could be target of pharmacological intervention. Clinical Trial Registration: ClinicalTrials.gov identifier NCT04343053.

DOI 10.3389/fimmu.2021.648004
Citations Scopus - 53Web of Science - 35
2021 Deliyannis G, Wong CY, McQuilten HA, Bachem A, Clarke M, Jia X, et al., 'TLR2-mediated activation of innate responses in the upper airways confers antiviral protection of the lungs', JCI Insight, 6 (2021) [C1]

The impact of respiratory virus infections on global health is felt not just during a pandemic, but endemic seasonal infections pose an equal and ongoing risk of severe disease. M... [more]

The impact of respiratory virus infections on global health is felt not just during a pandemic, but endemic seasonal infections pose an equal and ongoing risk of severe disease. Moreover, vaccines and antiviral drugs are not always effective or available for many respiratory viruses. We investigated how induction of effective and appropriate antigen-independent innate immunity in the upper airways can prevent the spread of respiratory virus infection to the vulnerable lower airways. Activation of TLR2, when restricted to the nasal turbinates, resulted in prompt induction of innate immune¿driven antiviral responses through action of cytokines, chemokines, and cellular activity in the upper but not the lower airways. We have defined how nasal epithelial cells and recruitment of macrophages work in concert and play pivotal roles to limit progression of influenza virus to the lungs and sustain protection for up to 7 days. These results reveal underlying mechanisms of how control of viral infection in the upper airways can occur and support the implementation of strategies that can activate TLR2 in nasal passages to provide rapid protection, especially for at-risk populations, against severe respiratory infection when vaccines and antiviral drugs are not always effective or available.

DOI 10.1172/jci.insight.140267
Citations Scopus - 15Web of Science - 7
Co-authors Jason Girkin
2021 Girkin J, Loo S-L, Esneau C, Maltby S, Mercuri F, Chua B, et al., 'TLR2-mediated innate immune priming boosts lung anti-viral immunity', EUROPEAN RESPIRATORY JOURNAL, 58 (2021) [C1]
DOI 10.1183/13993003.01584-2020
Citations Scopus - 14Web of Science - 9
Co-authors Steven Maltby, Suling Loo, Camille Esneau, Jason Girkin, Punnam Veerati, Christopher Grainge, Andrew Reid
2021 Shimoda LA, Bai C, Bartlett NW, Bastarache JA, Feghali-Bostwick C, Gunst SJ, et al., 'Promoting our early career members at AJP-Lung: the Editorial Board Fellowship Program and the Next Generation Physiologist Highlights section at our Journal', AMERICAN JOURNAL OF PHYSIOLOGY-LUNG CELLULAR AND MOLECULAR PHYSIOLOGY, 321 L844-L846 (2021)
DOI 10.1152/ajplung.00389.2021
Citations Scopus - 1
2021 Williams TC, Jackson DJ, Maltby S, Walton RP, Ching Y-M, Glanville N, et al., 'Rhinovirus-induced CCL17 and CCL22 in Asthma Exacerbations and Differential Regulation by STAT6', AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY, 64 344-356 (2021) [C1]
DOI 10.1165/rcmb.2020-0011OC
Citations Scopus - 13Web of Science - 10
Co-authors Suling Loo, Steven Maltby
2021 Schuliga M, Kanwal A, Read J, Blokland KEC, Burgess JK, Prele CM, et al., 'A cGAS-dependent response links DNA damage and senescence in alveolar epithelial cells: a potential drug target in IPF', AMERICAN JOURNAL OF PHYSIOLOGY-LUNG CELLULAR AND MOLECULAR PHYSIOLOGY, 321 L859-L871 (2021) [C1]
DOI 10.1152/ajplung.00574.2020
Citations Scopus - 17Web of Science - 8
Co-authors Michael Schuliga, Christopher Grainge
2021 Thomashow BM, Mannino DM, Tal-Singer R, Crapo JD, 'A rapidly changing understanding of COPD: World COPD Day from the COPD Foundation', American Journal of Physiology - Lung Cellular and Molecular Physiology, 321 L983-L987 (2021)
DOI 10.1152/ajplung.00400.2021
Citations Scopus - 8
Co-authors Christopher Grainge
2021 Proud PC, Tsitoura D, Watson RJ, Chua BY, Aram MJ, Bewley KR, et al., 'Prophylactic intranasal administration of a TLR2/6 agonist reduces upper respiratory tract viral shedding in a SARS-CoV-2 challenge ferret model', EBioMedicine, 63 (2021) [C1]
DOI 10.1016/j.ebiom.2020.103153
Citations Scopus - 64Web of Science - 48
2021 Wark PAB, Pathinayake PS, Kaiko G, Nichol K, Ali A, Chen L, et al., 'ACE2 expression is elevated in airway epithelial cells from older and male healthy individuals but reduced in asthma', Respirology, 26 442-451 (2021) [C1]

Background and objective: COVID-19 is complicated by acute lung injury, and death in some individuals. It is caused by SARS-CoV-2 that requires the ACE2 receptor and serine protea... [more]

Background and objective: COVID-19 is complicated by acute lung injury, and death in some individuals. It is caused by SARS-CoV-2 that requires the ACE2 receptor and serine proteases to enter AEC. We determined what factors are associated with ACE2 expression particularly in patients with asthma and COPD. Methods: We obtained lower AEC from 145 people from two independent cohorts, aged 2¿89 years, Newcastle (n = 115) and Perth (n = 30), Australia. The Newcastle cohort was enriched with people with asthma (n = 37) and COPD (n = 38). Gene expression for ACE2 and other genes potentially associated with SARS-CoV-2 cell entry was assessed by qPCR, and protein expression was confirmed with immunohistochemistry on endobronchial biopsies and cultured AEC. Results: Increased gene expression of ACE2 was associated with older age (P = 0.03) and male sex (P = 0.03), but not with pack-years smoked. When we compared gene expression between adults with asthma, COPD and healthy controls, mean ACE2 expression was lower in asthma patients (P = 0.01). Gene expression of furin, a protease that facilitates viral endocytosis, was also lower in patients with asthma (P = 0.02), while ADAM-17, a disintegrin that cleaves ACE2 from the surface, was increased (P = 0.02). ACE2 protein expression was also reduced in endobronchial biopsies from asthma patients. Conclusion: Increased ACE2 expression occurs in older people and males. Asthma patients have reduced expression. Altered ACE2 expression in the lower airway may be an important factor in virus tropism and may in part explain susceptibility factors and why asthma patients are not over-represented in those with COVID-19 complications.

DOI 10.1111/resp.14003
Citations Scopus - 53Web of Science - 52
Co-authors Andrew Reid, Christopher Oldmeadow, Ling Chen, Gerard Kaiko, Punnam Veerati, Ayesha Ayesha, Prabuddha Pathinayake
2021 Collison AM, Sokulsky LA, Kepreotes E, de Siqueira AP, Morten M, Edwards MR, et al., 'miR-122 promotes virus-induced lung disease by targeting SOCS1', JCI Insight, 6 (2021) [C1]

Virus-induced respiratory tract infections are a major health burden in childhood, and available treatments are supportive rather than disease modifying. Rhinoviruses (RVs), the c... [more]

Virus-induced respiratory tract infections are a major health burden in childhood, and available treatments are supportive rather than disease modifying. Rhinoviruses (RVs), the cause of approximately 80% of common colds, are detected in nearly half of all infants with bronchiolitis and the majority of children with an asthma exacerbation. Bronchiolitis in early life is a strong risk factor for the development of asthma. Here, we found that RV infection induced the expression of miRNA 122 (miR-122) in mouse lungs and in human airway epithelial cells. In vivo inhibition specifically in the lung reduced neutrophilic inflammation and CXCL2 expression, boosted innate IFN responses, and ameliorated airway hyperreactivity in the absence and in the presence of allergic lung inflammation. Inhibition of miR-122 in the lung increased the levels of suppressor of cytokine signaling 1 (SOCS1), which is an in vitro-validated target of miR-122. Importantly, gene silencing of SOCS1 in vivo completely reversed the protective effects of miR-122 inhibition on RV-induced lung disease. Higher miR-122 expression in nasopharyngeal aspirates was associated with a longer time on oxygen therapy and a higher rate of treatment failure in 87 infants hospitalized with moderately severe bronchiolitis. These results suggest that miR-122 promotes RV-induced lung disease via suppression of its target SOCS1 in vivo. Higher miR-122 expression was associated with worse clinical outcomes, highlighting the potential use of anti-miR-122 oligonucleotides, successfully trialed for treatment of hepatitis C, as potential therapeutics for RV-induced bronchiolitis and asthma exacerbations.

DOI 10.1172/jci.insight.127933
Citations Scopus - 15Web of Science - 6
Co-authors Ming Yang, Adam Collison, Joerg Mattes
2021 Finney LJ, Glanville N, Farne H, Aniscenko J, Fenwick P, Kemp SV, et al., 'Inhaled corticosteroids downregulate the SARS-CoV-2 receptor ACE2 in COPD through suppression of type I interferon', Journal of Allergy and Clinical Immunology, 147 510-519.e5 (2021) [C1]

Background: The mechanisms underlying altered susceptibility and propensity to severe Coronavirus disease 2019 (COVID-19) disease in at-risk groups such as patients with chronic o... [more]

Background: The mechanisms underlying altered susceptibility and propensity to severe Coronavirus disease 2019 (COVID-19) disease in at-risk groups such as patients with chronic obstructive pulmonary disease (COPD) are poorly understood. Inhaled corticosteroids (ICSs) are widely used in COPD, but the extent to which these therapies protect or expose patients to risk of severe COVID-19 is unknown. Objective: The aim of this study was to evaluate the effect of ICSs following pulmonary expression of the SARS-CoV-2 viral entry receptor angiotensin-converting enzyme-2 (ACE2). Methods: We evaluated the effect of ICS administration on pulmonary ACE2 expression in vitro in human airway epithelial cell cultures and in vivo in mouse models of ICS administration. Mice deficient in the type I IFN-a/ß receptor (Ifnar1-/-) and administration of exogenous IFN-ß were used to study the functional role of type-I interferon signaling in ACE2 expression. We compared sputum ACE2 expression in patients with COPD stratified according to use or nonuse of ICS. Results: ICS administration attenuated ACE2 expression in mice, an effect that was reversed by exogenous IFN-ß administration, and Ifnar1-/- mice had reduced ACE2 expression, indicating that type I interferon contributes mechanistically to this effect. ICS administration attenuated expression of ACE2 in airway epithelial cell cultures from patients with COPD and in mice with elastase-induced COPD-like changes. Compared with ICS nonusers, patients with COPD who were taking ICSs also had reduced sputum expression of ACE2. Conclusion: ICS therapies in COPD reduce expression of the SARS-CoV-2 entry receptor ACE2. This effect may thus contribute to altered susceptibility to COVID-19 in patients with COPD.

DOI 10.1016/j.jaci.2020.09.034
Citations Scopus - 105Web of Science - 70
Co-authors Suling Loo
2021 Shimoda LA, Bai C, Bartlett NW, Bastarache JA, Feghali-Bostwick C, Kuebler WM, et al., 'Announcing the Editorial Board Fellowship Program of the American Journal of Physiology-Lung Cellular and Molecular Physiology', AMERICAN JOURNAL OF PHYSIOLOGY-LUNG CELLULAR AND MOLECULAR PHYSIOLOGY, 321 L116-L118 (2021)
DOI 10.1152/ajplung.00239.2021
Citations Scopus - 1Web of Science - 1
2020 Sokulsky LA, Garcia-Netto K, Nguyen TH, Girkin JLN, Collison A, Mattes J, et al., 'A critical role for the CXCL3/CXCL5/CXCR2 neutrophilic chemotactic axis in the regulation of type 2 responses in a model of rhinoviral-induced asthma exacerbation', Journal of Immunology, 205 2468-2478 (2020) [C1]
DOI 10.4049/jimmunol.1901350
Citations Scopus - 31Web of Science - 17
Co-authors Ming Yang, Adam Collison, Gerard Kaiko, Jason Girkin, Joerg Mattes
2020 Davis JS, Chu G, Pathinayake P, Jones D, Giffard P, Macera L, et al., 'Seroprevalence of Torque Teno Virus in hemodialysis and renal transplant patients in Australia: A cross-sectional study', TRANSPLANT INFECTIOUS DISEASE, 22 (2020) [C1]
DOI 10.1111/tid.13400
Citations Scopus - 4Web of Science - 3
Co-authors G Chu, Prabuddha Pathinayake, Josh Davis
2020 Veerati PC, Mitchel JA, Reid AT, Knight DA, Bartlett NW, Park JA, Grainge CL, 'Airway mechanical compression: Its role in asthma pathogenesis and progression', European Respiratory Review, 29 1-13 (2020) [C1]
DOI 10.1183/16000617.0123-2019
Citations Scopus - 18Web of Science - 11
Co-authors Andrew Reid, Punnam Veerati, Christopher Grainge
2020 Reid AT, Nichol KS, Veerati PC, Moheimani F, Kicic A, Stick SM, et al., 'Blocking notch3 signaling abolishes MUC5AC production in airway epithelial cells from individuals with asthma', American Journal of Respiratory Cell and Molecular Biology, 62 513-523 (2020) [C1]

In asthma, goblet cell numbers are increased within the airway epithelium, perpetuating the production of mucus that is more difficult to clear and results in airway mucus pluggin... [more]

In asthma, goblet cell numbers are increased within the airway epithelium, perpetuating the production of mucus that is more difficult to clear and results in airway mucus plugging. Notch1, Notch2, or Notch3, or a combination of these has been shown to influence the differentiation of airway epithelial cells. How the expression of specific Notch isoforms differs in fully differentiated adult asthmatic epithelium and whether Notch influences mucin production after differentiation is currently unknown. We aimed to quantify different Notch isoforms in the airway epithelium of individuals with severe asthma and to examine the impact of Notch signaling on mucin MUC5AC. Human lung sections and primary bronchial epithelial cells from individuals with and without asthma were used in this study. Primary bronchial epithelial cells were differentiated at the air-liquid interface for 28 days. Notch isoform expression was analyzed by Taqman quantitative PCR. Immunohistochemistry was used to localize and quantify Notch isoforms in human airway sections. Notch signaling was inhibited in vitro using dibenzazepine or Notch3-specific siRNA, followed by analysis of MUC5AC. NOTCH3 was highly expressed in asthmatic airway epithelium compared with nonasthmatic epithelium. Dibenzazepine significantly reduced MUC5AC production in air-liquid interface cultures of primary bronchial epithelial cells concomitantly with suppression of NOTCH3 intracellular domain protein. Specific knockdown using NOTCH3 siRNA recapitulated the dibenzazepine-induced reduction in MUC5AC. We demonstrate that NOTCH3 is a regulator of MUC5AC production. Increased NOTCH3 signaling in the asthmatic airway epithelium may therefore be an underlying driver of excess MUC5AC production.

DOI 10.1165/rcmb.2019-0069OC
Citations Scopus - 29Web of Science - 19
Co-authors Punnam Veerati, Christopher Grainge, Andrew Reid
2020 Loo SL, Wark PAB, Esneau C, Nichol KS, Hsu ACY, Bartlett NW, 'Human coronaviruses 229E and OC43 replicate and induce distinct antiviral responses in differentiated primary human bronchial epithelial cells', American Journal of Physiology - Lung Cellular and Molecular Physiology, 319 L926-L931 (2020) [C1]

The recurrent emergence of novel, pathogenic coronaviruses (CoVs) severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1; 2002), Middle East respiratory syndrome (MERS)-CoV (... [more]

The recurrent emergence of novel, pathogenic coronaviruses (CoVs) severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1; 2002), Middle East respiratory syndrome (MERS)-CoV (2012), and most recently SARS-CoV-2 (2019) has highlighted the need for physiologically informative airway epithelial cell infection models for studying immunity to CoVs and development of antiviral therapies. To address this, we developed an in vitro infection model for two human coronaviruses; alphacoronavirus 229E-CoV (229E) and betacoronavirus OC43-CoV (OC43) in differentiated primary human bronchial epithelial cells (pBECs). Primary BECs from healthy subjects were grown at air-liquid interface (ALI) and infected with 229E or OC43, and replication kinetics and time-course expression of innate immune mediators were assessed. OC43 and 229E-CoVs replicated in differentiated pBECs but displayed distinct replication kinetics: 229E replicated rapidly with viral load peaking at 24 h postinfection, while OC43 replication was slower peaking at 96 h after infection. This was associated with diverse antiviral response profiles defined by increased expression of type I/III interferons and interferon-stimulated genes (ISGs) by 229E compared with no innate immune activation with OC43 infection. Understanding the host-virus interaction for previously established coronaviruses will give insight into pathogenic mechanisms underpinning SARS-CoV-2-induced respiratory disease and other future coronaviruses that may arise from zoonotic sources.

DOI 10.1152/AJPLUNG.00374.2020
Citations Scopus - 31Web of Science - 21
Co-authors Suling Loo, Camille Esneau
2020 Veerati PC, Troy NM, Reid AT, Li NF, Nichol KS, Kaur P, et al., 'Airway Epithelial Cell Immunity Is Delayed During Rhinovirus Infection in Asthma and COPD', FRONTIERS IN IMMUNOLOGY, 11 (2020) [C1]
DOI 10.3389/fimmu.2020.00974
Citations Scopus - 57Web of Science - 31
Co-authors Christopher Grainge, Punnam Veerati, Steven Maltby, Andrew Reid
2020 Kamal F, Glanville N, Xia W, Bakhsoliani E, Aniscenko J, Bartlett NW, et al., 'Beclomethasone Has Lesser Suppressive Effects on Inflammation and Antibacterial Immunity Than Fluticasone or Budesonide in Experimental Infection Models', Chest, 158 947-951 (2020) [C1]
DOI 10.1016/j.chest.2020.05.531
Citations Scopus - 4
2020 Kan S, Hariyadi DM, Grainge C, Knight DA, Bartlett NW, Liang M, 'Airway epithelial-targeted nanoparticles for asthma therapy', AMERICAN JOURNAL OF PHYSIOLOGY-LUNG CELLULAR AND MOLECULAR PHYSIOLOGY, 318 L500-L509 (2020) [C1]
DOI 10.1152/ajplung.00237.2019
Citations Scopus - 26Web of Science - 13
Co-authors Christopher Grainge, Roger Liang
2020 Hondermarck H, Bartlett NW, Nurcombe V, 'The role of growth factor receptors in viral infections: An opportunity for drug repurposing against emerging viral diseases such as COVID-19?', FASEB bioAdvances, 2 296-303 (2020) [C1]
DOI 10.1096/fba.2020-00015
Citations Scopus - 43Web of Science - 37
Co-authors Hubert Hondermarck
2019 Singanayagam A, Glanville N, Cuthbertson L, Bartlett NW, Finney LJ, Turek E, et al., 'Inhaled corticosteroid suppression of cathelicidin drives dysbiosis and bacterial infection in chronic obstructive pulmonary disease.', Science translational medicine, 11 13-13 (2019) [C1]
DOI 10.1126/scitranslmed.aav3879
Citations Scopus - 60Web of Science - 55
2019 Schuliga M, Bartlett N, 'Modeling the impact of low-dose particulate matter on lung health', AMERICAN JOURNAL OF PHYSIOLOGY-LUNG CELLULAR AND MOLECULAR PHYSIOLOGY, 317 L550-L553 (2019)
DOI 10.1152/ajplung.00343.2019
Citations Scopus - 2Web of Science - 1
Co-authors Michael Schuliga
2019 McVey MJ, Maishan M, Blokland KEC, Bartlett N, Kuebler WM, 'Extracellular vesicles in lung health, disease, and therapy.', American journal of physiology. Lung cellular and molecular physiology, 316 L977-L989 (2019) [C1]
DOI 10.1152/ajplung.00546.2018
Citations Scopus - 46Web of Science - 30
2019 Waters DW, Blokland KEC, Pathinayake PS, Wei L, Schuliga M, Jaffar J, et al., 'STAT3 Regulates the Onset of Oxidant-induced Senescence in Lung Fibroblasts.', Am J Respir Cell Mol Biol, 61 61-73 (2019) [C1]
DOI 10.1165/rcmb.2018-0328OC
Citations Scopus - 51Web of Science - 46
Co-authors Michael Schuliga, Christopher Grainge, Prabuddha Pathinayake
2019 Singanayagam A, Loo SL, Calderazzo M, Finney LJ, Torralbo MBT, Bakhsoliani E, et al., 'Antiviral immunity is impaired in COPD patients with frequent exacerbations', American Journal of Physiology - Lung Cellular and Molecular Physiology, 317 L893-L903 (2019) [C1]

Patients with frequent exacerbations represent a chronic obstructive pulmonary disease (COPD) subgroup requiring better treatment options. The aim of this study was to determine t... [more]

Patients with frequent exacerbations represent a chronic obstructive pulmonary disease (COPD) subgroup requiring better treatment options. The aim of this study was to determine the innate immune mechanisms that underlie susceptibility to frequent exacerbations in COPD. We measured sputum expression of immune mediators and bacterial loads in samples from patients with COPD at stable state and during virusassociated exacerbations. In vitro immune responses to rhinovirus infection in differentiated primary bronchial epithelial cells (BECs) sampled from patients with COPD were additionally evaluated. Patients were stratified as frequent exacerbators (=2 exacerbations in the preceding year) or infrequent exacerbators (<2 exacerbations in the preceding year) with comparisons made between these groups. Frequent exacerbators had reduced sputum cell mRNA expression of the antiviral immune mediators type I and III interferons and reduced interferon-stimulated gene (ISG) expression when clinically stable and during virus-associated exacerbation. A role for epithelial cellintrinsic innate immune dysregulation was identified: induction of interferons and ISGs during in vitro rhinovirus (RV) infection was also impaired in differentiated BECs from frequent exacerbators. Frequent exacerbators additionally had increased sputum bacterial loads at 2 wk following virus-associated exacerbation onset. These data implicate deficient airway innate immunity involving epithelial cells in the increased propensity to exacerbations observed in some patients with COPD. Therapeutic approaches to boost innate antimicrobial immunity in the lung could be a viable strategy for prevention and treatment of frequent exacerbations.

DOI 10.1152/ajplung.00253.2019
Citations Scopus - 44Web of Science - 36
Co-authors Christopher Grainge, Prabuddha Pathinayake, Punnam Veerati, Jason Girkin, Suling Loo, Andrew Reid
2018 Reid AT, Veerati PC, Gosens R, Bartlett NW, Wark PA, Grainge CL, et al., 'Persistent induction of goblet cell differentiation in the airways: Therapeutic approaches', Pharmacology and Therapeutics, 185 155-169 (2018) [C1]

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, ... [more]

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.

DOI 10.1016/j.pharmthera.2017.12.009
Citations Scopus - 21Web of Science - 19
Co-authors Andrew Reid, Punnam Veerati, Christopher Grainge
2018 Singanayagam A, Glanville N, Girkin JL, Ching YM, Marcellini A, Porter JD, et al., 'Corticosteroid suppression of antiviral immunity increases bacterial loads and mucus production in COPD exacerbations', NATURE COMMUNICATIONS, 9 (2018) [C1]
DOI 10.1038/s41467-018-04574-1
Citations Scopus - 136Web of Science - 116
Co-authors Punnam Veerati, Andrew Reid, Christopher Grainge, Prabuddha Pathinayake, Jason Girkin, Suling Loo
2018 Chairakaki A-D, Saridaki M-I, Pyrillou K, Mouratis M-A, Koltsida O, Walton RP, et al., 'Plasmacytoid dendritic cells drive acute exacerbations of asthma', Journal of Allergy and Clinical Immunology, 142 542-556 (2018) [C1]
DOI 10.1016/j.jaci.2017.08.032
Citations Scopus - 42Web of Science - 30
2018 Wark PAB, Ramsahai JM, Pathinayake P, Malik B, Bartlett NW, 'Respiratory Viruses and Asthma', Seminars in Respiratory and Critical Care Medicine, 39 45-55 (2018) [C1]

Asthma remains the most prevalent chronic respiratory disorder, affecting people of all ages. The relationship between respiratory virus infection and asthma has long been recogni... [more]

Asthma remains the most prevalent chronic respiratory disorder, affecting people of all ages. The relationship between respiratory virus infection and asthma has long been recognized, though remains incompletely understood. In this article, we will address key issues around this relationship. These will include the crucial role virus infection plays in early life, as a potential risk factor for the development of asthma and lung disease. We will assess the impact that virus infection has on those with established asthma as a trigger for acute disease and how this may influence asthma throughout life. Finally, we will explore the complex interaction that occurs between the airway and the immune responses that make those with asthma so susceptible to the effects of virus infection.

DOI 10.1055/s-0037-1617412
Citations Scopus - 22Web of Science - 17
Co-authors Prabuddha Pathinayake
2017 Hansel TT, Tunstall T, Trujillo-Torralbo MB, Shamji B, del-Rosario A, Dhariwal J, et al., 'A Comprehensive Evaluation of Nasal and Bronchial Cytokines and Chemokines Following Experimental Rhinovirus Infection in Allergic Asthma: Increased Interferons (IFN- and IFN- ) and Type 2 Inflammation (IL-5 and IL-13)', EBioMedicine, 19 128-138 (2017) [C1]

Background Rhinovirus infection is a major cause of asthma exacerbations. Objectives We studied nasal and bronchial mucosal inflammatory responses during experimental rhinovirus-i... [more]

Background Rhinovirus infection is a major cause of asthma exacerbations. Objectives We studied nasal and bronchial mucosal inflammatory responses during experimental rhinovirus-induced asthma exacerbations. Methods We used nasosorption on days 0, 2¿5 and 7 and bronchosorption at baseline and day 4 to sample mucosal lining fluid to investigate airway mucosal responses to rhinovirus infection in patients with allergic asthma (n¿=¿28) and healthy non-atopic controls (n¿=¿11), by using a synthetic absorptive matrix and measuring levels of 34 cytokines and chemokines using a sensitive multiplex assay. Results Following rhinovirus infection asthmatics developed more upper and lower respiratory symptoms and lower peak expiratory flows compared to controls (all P¿<¿0.05). Asthmatics also developed higher nasal lining fluid levels of an anti-viral pathway (including IFN-¿, IFN-¿/IL-29, CXCL11/ITAC, CXCL10/IP10 and IL-15) and a type 2 inflammatory pathway (IL-4, IL-5, IL-13, CCL17/TARC, CCL11/eotaxin, CCL26/eotaxin-3) (area under curve day 0¿7, all P¿<¿0.05). Nasal IL-5 and IL-13 were higher in asthmatics at day 0 (P¿<¿0.01) and levels increased by days 3 and 4 (P¿<¿0.01). A hierarchical correlation matrix of 24 nasal lining fluid cytokine and chemokine levels over 7¿days demonstrated expression of distinct interferon-related and type 2 pathways in asthmatics. In asthmatics IFN-¿, CXCL10/IP10, CXCL11/ITAC, IL-15 and IL-5 increased in bronchial lining fluid following viral infection (all P¿<¿0.05). Conclusions Precision sampling of mucosal lining fluid identifies robust interferon and type 2 responses in the upper and lower airways of asthmatics during an asthma exacerbation. Nasosorption and bronchosorption have potential to define asthma endotypes in stable disease and at exacerbation.

DOI 10.1016/j.ebiom.2017.03.033
Citations Scopus - 96Web of Science - 73
2017 Dhariwal J, Cameron A, Trujillo-Torralbo M-B, del Rosario A, Bakhsoliani E, Paulsen M, et al., 'Mucosal Type 2 Innate Lymphoid Cells Are a Key Component of the Allergic Response to Aeroallergens', American Journal of Respiratory and Critical Care Medicine, 195 1586-1596 (2017) [C1]
DOI 10.1164/rccm.201609-1846oc
2016 Tay H, Wark PAB, Bartlett NW, 'Advances in the treatment of virus-induced asthma', Expert Review of Respiratory Medicine, 10 629-641 (2016) [C1]

ABSTRACT: Viral exacerbations continue to represent the major burden in terms of morbidity, mortality and health care costs associated with asthma. Those at greatest risk for acut... [more]

ABSTRACT: Viral exacerbations continue to represent the major burden in terms of morbidity, mortality and health care costs associated with asthma. Those at greatest risk for acute asthma are those with more severe airways disease and poor asthma control. It is this group with established asthma in whom acute exacerbations triggered by virus infections remain a serious cause of increased morbidity. A range of novel therapies are emerging to treat asthma and in particular target this group with poor disease control, and in most cases their efficacy is now being judged by their ability to reduce the frequency of acute exacerbations. Critical for the development of new treatment approaches is an improved understanding of virus-host interaction in the context of the asthmatic airway. This requires research into the virology of the disease in physiological models in conjunction with detailed phenotypic characterisation of asthma patients to identify targets amenable to therapeutic intervention.

DOI 10.1080/17476348.2016.1180249
Citations Scopus - 10Web of Science - 4
2015 Jackson DJ, Trujillo-Torralbo M-B, del-Rosario J, Bartlett NW, Edwards MR, Mallia P, et al., 'The influence of asthma control on the severity of virus-induced asthma exacerbations', Journal of Allergy and Clinical Immunology, 136 497-500.e3 (2015) [C3]
DOI 10.1016/j.jaci.2015.01.028
Citations Scopus - 39Web of Science - 37
2015 Hatchwell L, Collison A, Girkin J, Parsons K, Li J, Zhang J, et al., 'Toll-like receptor 7 governs interferon and inflammatory responses to rhinovirus and is suppressed by IL-5-induced lung eosinophilia', Thorax, (2015) [C1]

© 2015 BMJ Publishing Group Ltd &amp; British Thoracic Society.Background Asthma exacerbations represent a significant disease burden and are commonly caused by rhinovirus (RV), w... [more]

© 2015 BMJ Publishing Group Ltd & British Thoracic Society.Background Asthma exacerbations represent a significant disease burden and are commonly caused by rhinovirus (RV), which is sensed by Toll-like receptors (TLR) such as TLR7. Some asthmatics have impaired interferon (IFN) responses to RV, but the underlying mechanisms of this clinically relevant observation are poorly understood. Objectives To investigate the importance of intact TLR7 signalling in vivo during RV exacerbation using mouse models of house dust mite (HDM)-induced allergic airways disease exacerbated by a superimposed RV infection. Methods Wild-type and TLR7-deficient (Tlr7<sup>-/-</sup>) BALB/c mice were intranasally sensitised and challenged with HDM prior to infection with RV1B. In some experiments, mice were administered recombinant IFN or adoptively transferred with plasmacytoid dendritic cells (pDC). Results Allergic Tlr7<sup>-/-</sup> mice displayed impaired IFN release upon RV1B infection, increased virus replication and exaggerated eosinophilic inflammation and airways hyper reactivity. Treatment with exogenous IFN or adoptive transfer of TLR7-competent pDCs blocked these exaggerated inflammatory responses and boosted IFN? release in the absence of host TLR7 signalling. TLR7 expression in the lungs was suppressed by allergic inflammation and by interleukin (IL)-5-induced eosinophilia in the absence of allergy. Subjects with moderate-to-severe asthma and eosinophilic but not neutrophilic airways inflammation, despite inhaled steroids, showed reduced TLR7 and IFN?2/3 expression in endobronchial biopsies. Furthermore, TLR7 expression inversely correlated with percentage of sputum eosinophils. Conclusions This implicates IL-5-induced airways eosinophilia as a negative regulator of TLR7 expression and antiviral responses, which provides a molecular mechanism underpinning the effect of eosinophil-targeting treatments for the prevention of asthma exacerbations.

DOI 10.1136/thoraxjnl-2014-205465
Citations Scopus - 89Web of Science - 76
Co-authors Adam Collison, Jason Girkin, Joerg Mattes
2015 Singanayagam A, Glanville N, Walton RP, Aniscenko J, Pearson RM, Pinkerton JW, et al., 'A short-term mouse model that reproduces the immunopathological features of rhinovirus-induced exacerbation of COPD.', Clin Sci (Lond), 129 245-258 (2015) [C1]
DOI 10.1042/CS20140654
Citations Scopus - 36Web of Science - 27
Co-authors Jay Horvat
2015 Girkin J, Hatchwell L, Foster P, Johnston SL, Bartlett N, Collison A, Mattes J, 'CCL7 and IRF-7 mediate hallmark inflammatory and IFN responses following rhinovirus 1B infection', Journal of Immunology, 194 4924-4930 (2015) [C1]

Rhinovirus (RV) infections are common and have the potential to exacerbate asthma. We have determined the lung transcriptome in RV strain 1B-infected naive BALB/c mice (nonallergi... [more]

Rhinovirus (RV) infections are common and have the potential to exacerbate asthma. We have determined the lung transcriptome in RV strain 1B-infected naive BALB/c mice (nonallergic) and identified CCL7 and IFN regulatory factor (IRF)-7 among the most upregulated mRNA transcripts in the lung. To investigate their roles we employed anti-CCL7 Abs and an IRF-7-targeting small interfering RNA in vivo. Neutralizing CCL7 or inhibiting IRF-7 limited neutrophil and macrophage influx and IFN responses in nonallergic mice. Neutralizing CCL7 also reduced activation of NF-¿B p65 and p50 subunits, as well as airway hyperreactivity (AHR) in nonallergic mice. However, neither NF-¿B subunit activation nor AHR was abolished with infection of allergic mice after neutralizing CCL7, despite a reduction in the number of neutrophils, macrophages, and eosinophils. IRF-7 small interfering RNA primarily suppressed IFN-a and IFN-b levels during infection of allergic mice. Our data highlight a pivotal role of CCL7 and IRF-7 in RV-induced inflammation and IFN responses and link NF-¿B signaling to the development of AHR.

DOI 10.4049/jimmunol.1401362
Citations Scopus - 34Web of Science - 33
Co-authors Jason Girkin, Adam Collison, Joerg Mattes
2014 George PM, Oliver E, Dorfmuller P, Dubois OD, Reed DM, Kirkby NS, et al., 'Evidence for the Involvement of Type I Interferon in Pulmonary Arterial Hypertension', CIRCULATION RESEARCH, 114 677-688 (2014) [C1]
DOI 10.1161/CIRCRESAHA.114.302221
Citations Scopus - 118Web of Science - 92
2014 Mahmutovic-Persson I, Akbarshahi H, Bartlett NW, Glanville N, Johnston SL, Brandelius A, Uller L, 'Inhaled dsRNA and rhinovirus evoke neutrophilic exacerbation and lung expression of thymic stromal lymphopoietin in allergic mice with established experimental asthma', Allergy: European Journal of Allergy and Clinical Immunology, 69 348-358 (2014) [C1]
DOI 10.1111/all.12329
Citations Scopus - 42Web of Science - 35
2014 Jackson DJ, Makrinioti H, Rana BMJ, Shamji BWH, Trujillo-Torralbo MB, Footitt J, et al., 'IL-33-Dependent type 2 inflammation during rhinovirus-induced asthma exacerbations in vivo', American Journal of Respiratory and Critical Care Medicine, 190 1373-1382 (2014) [C1]
DOI 10.1164/rccm.201406-1039OC
Citations Scopus - 467Web of Science - 391
2014 Beale J, Jayaraman A, Jackson DJ, Macintyre JDR, Edwards MR, Walton RP, et al., 'Rhinovirus-induced IL-25 in asthma exacerbation drives type 2 immunity and allergic pulmonary inflammation', Science Translational Medicine, 6 (2014) [C1]
DOI 10.1126/scitranslmed.3009124
Citations Scopus - 278Web of Science - 226
2014 Toussaint M, Singanayagam A, Johnston SL, Bartlett N, 'Role Of Interleukine-33 In Rhinovirus-Induced Allergic Asthma Exacerbation', JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, 133 AB52-AB52 (2014)
DOI 10.1016/j.jaci.2013.12.211
2014 Jayaraman A, Bartlett N, Johnston SL, 'Innate and Adaptive Lymphocyte Responses In a Mouse Model Of Rhinovirus-Induced Asthma Exacerbation', JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, 133 AB135-AB135 (2014)
DOI 10.1016/j.jaci.2013.12.501
2014 Jayaraman A, Jackson DJ, Message SD, Pearson RM, Aniscenko J, Caramori G, et al., 'IL-15 complexes induce NK- and T-cell responses independent of type I IFN signaling during rhinovirus infection', MUCOSAL IMMUNOLOGY, 7 1151-1164 (2014)
DOI 10.1038/mi.2014.2
Citations Scopus - 38Web of Science - 36
2013 McLean GR, Walton RP, Shetty S, Peel TJ, Paktiawal N, Kebadze T, et al., 'Corrigendum to: "Rhinovirus infections and immunisation induce cross-serotype reactive antibodies to VP1" [Antiviral Res. 95(3) (2012) 193-201]', Antiviral Research, 97 381 (2013)
DOI 10.1016/j.antiviral.2013.02.004
2013 Traub S, Nikonova A, Carruthers A, Dunmore R, Vousden KA, Gogsadze L, et al., 'An Anti-Human ICAM-1 Antibody Inhibits Rhinovirus-Induced Exacerbations of Lung Inflammation', PLoS Pathogens, 9 (2013) [C1]

Human rhinoviruses (HRV) cause the majority of common colds and acute exacerbations of asthma and chronic obstructive pulmonary disease (COPD). Effective therapies are urgently ne... [more]

Human rhinoviruses (HRV) cause the majority of common colds and acute exacerbations of asthma and chronic obstructive pulmonary disease (COPD). Effective therapies are urgently needed, but no licensed treatments or vaccines currently exist. Of the 100 identified serotypes, ~90% bind domain 1 of human intercellular adhesion molecule-1 (ICAM-1) as their cellular receptor, making this an attractive target for development of therapies; however, ICAM-1 domain 1 is also required for host defence and regulation of cell trafficking, principally via its major ligand LFA-1. Using a mouse anti-human ICAM-1 antibody (14C11) that specifically binds domain 1 of human ICAM-1, we show that 14C11 administered topically or systemically prevented entry of two major groups of rhinoviruses, HRV16 and HRV14, and reduced cellular inflammation, pro-inflammatory cytokine induction and virus load in vivo. 14C11 also reduced cellular inflammation and Th2 cytokine/chemokine production in a model of major group HRV-induced asthma exacerbation. Interestingly, 14C11 did not prevent cell adhesion via human ICAM-1/LFA-1 interactions in vitro, suggesting the epitope targeted by 14C11 was specific for viral entry. Thus a human ICAM-1 domain-1-specific antibody can prevent major group HRV entry and induction of airway inflammation in vivo. © 2013 Traub et al.

DOI 10.1371/journal.ppat.1003520
Citations Scopus - 75Web of Science - 62
2013 Glanville N, Mclean GR, Guy B, Lecouturier V, Berry C, Girerd Y, et al., 'Cross-Serotype Immunity Induced by Immunization with a Conserved Rhinovirus Capsid Protein', PLoS Pathogens, 9 (2013) [C1]

Human rhinovirus (RV) infections are the principle cause of common colds and precipitate asthma and COPD exacerbations. There is currently no RV vaccine, largely due to the existe... [more]

Human rhinovirus (RV) infections are the principle cause of common colds and precipitate asthma and COPD exacerbations. There is currently no RV vaccine, largely due to the existence of ~150 strains. We aimed to define highly conserved areas of the RV proteome and test their usefulness as candidate antigens for a broadly cross-reactive vaccine, using a mouse infection model. Regions of the VP0 (VP4+VP2) capsid protein were identified as having high homology across RVs. Immunization with a recombinant VP0 combined with a Th1 promoting adjuvant induced systemic, antigen specific, cross-serotype, cellular and humoral immune responses. Similar cross-reactive responses were observed in the lungs of immunized mice after infection with heterologous RV strains. Immunization enhanced the generation of heterosubtypic neutralizing antibodies and lung memory T cells, and caused more rapid virus clearance. Conserved domains of the RV capsid therefore induce cross-reactive immune responses and represent candidates for a subunit RV vaccine. © 2013 Glanville et al.

DOI 10.1371/journal.ppat.1003669
Citations Scopus - 64Web of Science - 54
2013 Glanville N, Message SD, Walton RP, Pearson RM, Parker HL, Laza-Stanca V, et al., ' dT cells suppress inflammation and disease during rhinovirus-induced asthma exacerbations', Mucosal Immunology, 6 1091-1100 (2013) [C1]

Most asthma exacerbations are triggered by virus infections, the majority being caused by human rhinoviruses (RV). In mouse models, ¿dT cells have been previously demonstrated to ... [more]

Most asthma exacerbations are triggered by virus infections, the majority being caused by human rhinoviruses (RV). In mouse models, ¿dT cells have been previously demonstrated to influence allergen-driven airways hyper-reactivity (AHR) and can have antiviral activity, implicating them as prime candidates in the pathogenesis of asthma exacerbations. To explore this, we have used human and mouse models of experimental RV-induced asthma exacerbations to examine ¿dT-cell responses and determine their role in the immune response and associated airways disease. In humans, airway ¿dT-cell numbers were increased in asthmatic vs. healthy control subjects during experimental infection. Airway and blood ¿dT-cell numbers were associated with increased airways obstruction and AHR. Airway ¿dT-cell number was also positively correlated with bronchoalveolar lavage (BAL) virus load and BAL eosinophils and lymphocytes during RV infection. Consistent with our observations of RV-induced asthma exacerbations in humans, infection of mice with allergic airways inflammation increased lung ¿dT-cell number and activation. Inhibiting ¿dT-cell responses using anti-¿dTCR (anti-¿dT-cell receptor) antibody treatment in the mouse asthma exacerbation model increased AHR and airway T helper type 2 cell recruitment and eosinophilia, providing evidence that ¿dT cells are negative regulators of airways inflammation and disease in RV-induced asthma exacerbations.

DOI 10.1038/mi.2013.3
Citations Scopus - 37Web of Science - 35
2013 Brignull LM, Czimmerer Z, Saidi H, Daniel B, Villela I, Bartlett NW, et al., 'Reprogramming of lysosomal gene expression by interleukin-4 and Stat6', BMC Genomics, 14 (2013) [C1]

Background: Lysosomes play important roles in multiple aspects of physiology, but the problem of how the transcription of lysosomal genes is coordinated remains incompletely under... [more]

Background: Lysosomes play important roles in multiple aspects of physiology, but the problem of how the transcription of lysosomal genes is coordinated remains incompletely understood. The goal of this study was to illuminate the physiological contexts in which lysosomal genes are coordinately regulated and to identify transcription factors involved in this control.Results: As transcription factors and their target genes are often co-regulated, we performed meta-analyses of array-based expression data to identify regulators whose mRNA profiles are highly correlated with those of a core set of lysosomal genes. Among the ~50 transcription factors that rank highest by this measure, 65% are involved in differentiation or development, and 22% have been implicated in interferon signaling. The most strongly correlated candidate was Stat6, a factor commonly activated by interleukin-4 (IL-4) or IL-13. Publicly available chromatin immunoprecipitation (ChIP) data from alternatively activated mouse macrophages show that lysosomal genes are overrepresented among Stat6-bound targets. Quantification of RNA from wild-type and Stat6-deficient cells indicates that Stat6 promotes the expression of over 100 lysosomal genes, including hydrolases, subunits of the vacuolar H+ ATPase and trafficking factors. While IL-4 inhibits and activates different sets of lysosomal genes, Stat6 mediates only the activating effects of IL-4, by promoting increased expression and by neutralizing undefined inhibitory signals induced by IL-4.Conclusions: The current data establish Stat6 as a broadly acting regulator of lysosomal gene expression in mouse macrophages. Other regulators whose expression correlates with lysosomal genes suggest that lysosome function is frequently re-programmed during differentiation, development and interferon signaling. © 2013 Brignull et al.; licensee BioMed Central Ltd.

DOI 10.1186/1471-2164-14-853
Citations Scopus - 16Web of Science - 13
2013 Collison AM, Hatchwell LM, Verrills NM, Wark PA, Pereira De Siqueira AL, Tooze MK, et al., 'The E3 ubiquitin ligase midline 1 promotes allergen and rhinovirus-induced asthma by inhibiting protein phosphatase 2A activity', Nature Medicine, 19 232-237 (2013) [C1]
Citations Scopus - 124Web of Science - 114
Co-authors Adam Collison, Nikki Verrills, Joerg Mattes
2012 Edwards MR, Bartlett NW, Hussell T, Openshaw P, Johnston SL, 'The microbiology of asthma', Nature Reviews Microbiology, 10 459-471 (2012) [C1]
DOI 10.1038/nrmicro2801
Citations Scopus - 161Web of Science - 138
2012 Bartlett NW, Slater L, Glanville N, Haas JJ, Caramori G, Casolari P, et al., 'Defining critical roles for NF- B p65 and type I interferon in innate immunity to rhinovirus', EMBO Molecular Medicine, 4 1244-1260 (2012) [C1]
DOI 10.1002/emmm.201201650
Citations Scopus - 72Web of Science - 66
2012 Hewson CA, Haas JJ, Bartlett NW, Message SD, Laza-Stanca V, Kebadze T, et al., 'Rhinovirus induces MUC5AC in a human infection model and in vitro via NF- B and EGFR pathways (European Respiratory Journal (2010) 36, (1425-1435))', European Respiratory Journal, 39 793 (2012)
DOI 10.1183/09031936.50026910
Citations Scopus - 2
2012 Kuo C, Lim S, King NJC, Bartlett NW, Walton RP, Zhu J, et al., 'Erratum: Rhinovirus infection induces expression of airway remodelling factors in vitro and in vivo (Respirology (2011) 16 (367-377))', Respirology, 17 192 (2012)
DOI 10.1111/j.1440-1843.2011.02110.x
2012 McLean GR, Walton RP, Shetty S, Paktiawal N, Kebadze T, Gogsadze L, et al., 'Rhinovirus infections and immunisation induce cross-serotype reactive antibodies to VP1', ANTIVIRAL RESEARCH, 95 193-201 (2012)
DOI 10.1016/j.antiviral.2012.06.006
Citations Scopus - 37Web of Science - 32
2012 Slater L, Bartlett NW, Haas JJ, Zhu J, Message SD, Walton RP, et al., 'Correction: Co-ordinated Role of TLR3, RIG-I and MDA5 in the Innate Response to Rhinovirus in Bronchial Epithelium', PLoS pathogens, 8 (2012)
2011 Telcian AG, Laza-Stanca V, Edwards MR, Harker JA, Wang H, Bartlett NW, et al., 'RSV-induced bronchial epithelial cell PD-L1 expression inhibits CD8
DOI 10.1093/infdis/jiq020
Citations Scopus - 62Web of Science - 58
2011 Kuo C, Lim S, King NJC, Bartlett NW, Walton RP, Zhu J, et al., 'Rhinovirus infection induces expression of airway remodelling factors in vitro and in vivo', Respirology, 16 367-377 (2011) [C1]
DOI 10.1111/j.1440-1843.2010.01918.x
Citations Scopus - 44Web of Science - 40
2010 Hewson CA, Haas JJ, Bartlett NW, Message SD, Laza-Stanca V, Kebadze T, et al., 'Rhinovirus induces MUC5AC in a human infection model and in vitro via NF-kappa B and EGFR pathways', EUROPEAN RESPIRATORY JOURNAL, 36 1425-1435 (2010)
DOI 10.1183/09031936.00026910
Citations Scopus - 96Web of Science - 80
2010 Slater L, Bartlett NW, Haas JJ, Zhu J, Message SD, Walton RP, et al., 'Co-ordinated Role of TLR3, RIG-I and MDA5 in the Innate Response to Rhinovirus in Bronchial Epithelium', PLOS PATHOGENS, 6 (2010)
DOI 10.1371/journal.ppat.1001178
Citations Scopus - 260Web of Science - 235
2009 Edwards MR, Bartlett NW, Clarke D, Birrell M, Belvisi M, Johnston SL, 'Targeting the NF-kappa B pathway in asthma and chronic obstructive pulmonary disease', PHARMACOLOGY & THERAPEUTICS, 121 1-13 (2009)
DOI 10.1016/j.pharmthera.2008.09.003
Citations Scopus - 334Web of Science - 292
2009 Bartlett NW, McLean GR, Chang Y-S, Johnston SL, 'Genetics and epidemiology: asthma and infection', CURRENT OPINION IN ALLERGY AND CLINICAL IMMUNOLOGY, 9 395-400 (2009)
DOI 10.1097/ACI.0b013e32833066fa
Citations Scopus - 47Web of Science - 39
2008 Jacobs N, Bartlett NW, Clark RH, Smith GL, 'Vaccinia virus lacking the Bcl-2-like protein N1 induces a stronger natural killer cell response to infection', JOURNAL OF GENERAL VIROLOGY, 89 2877-2881 (2008)
DOI 10.1099/vir.0.2008/004119-0
Citations Scopus - 27Web of Science - 24
2008 Bartlett NW, Walton RP, Edwards MR, Aniscenko J, Caramori G, Zhu J, et al., 'Mouse models of rhinovirus-induced disease and exacerbation of allergic airway inflammation', NATURE MEDICINE, 14 199-204 (2008)
DOI 10.1038/nm1713
Citations Scopus - 308Web of Science - 277
2007 Cooray S, Bahar MW, Abrescia NGA, McVey CE, Bartlett NW, Chen RA-J, et al., 'Functional and structural studies of the vaccinia virus virulence factor N1 reveal a Bcl-2-like anti-apoptotic protein', JOURNAL OF GENERAL VIROLOGY, 88 1656-1666 (2007)
DOI 10.1099/vir.0.82772-0
Citations Scopus - 155Web of Science - 144
2006 Clark RH, Kenyon JC, Bartlett NW, Tscharke DC, Smith GL, 'Deletion of gene A41L enhances vaccinia virus immunogenicity and vaccine efficacy', JOURNAL OF GENERAL VIROLOGY, 87 29-38 (2006)
DOI 10.1099/vir.0.81417-0
Citations Scopus - 69Web of Science - 70
2006 Contoli M, Message SD, Laza-Stanca V, Edwards MR, Wark PA, Bartlett N, et al., 'Role of eficient type III interferon-lambda production in asthma exacerbations', Nature Medicine, 12 1023-1026 (2006) [C1]
DOI 10.1038/nm1462
Citations Scopus - 897Web of Science - 783
2005 Stack J, Haga IR, Schroder M, Bartlett NW, Maloney G, Reading PC, et al., 'Vaccinia virus protein Toll-like-interleukin-1 A46R targets multiple receptor adaptors and contributes to virulence', JOURNAL OF EXPERIMENTAL MEDICINE, 201 1007-1018 (2005)
DOI 10.1084/jem.20041442
Citations Scopus - 312Web of Science - 288
2005 Bartlett NW, Buttigieg K, Kotenko SV, Smith GL, 'Murine interferon lambdas (type III interferons) exhibit potent antiviral activity in vivo in a poxvirus infection model', JOURNAL OF GENERAL VIROLOGY, 86 1589-1596 (2005)
DOI 10.1099/vir.0.80904-0
Citations Scopus - 98Web of Science - 89
2004 Bartlett NW, Dumoutier L, Renauld JC, Kotenko SV, McVey CE, Lee HJ, Smith GL, 'A new member of the interleukin 10-related cytokine family encoded by a poxvirus', JOURNAL OF GENERAL VIROLOGY, 85 1401-1412 (2004)
DOI 10.1099/vir.0.79980-0
Citations Scopus - 23Web of Science - 19
2003 Harte MT, Haga IR, Maloney G, Gray P, Reading PC, Bartlett NW, et al., 'The poxvirus protein A52R targets toll-like receptor signaling complexes to suppress host defense', JOURNAL OF EXPERIMENTAL MEDICINE, 197 343-351 (2003)
DOI 10.1084/jem.20021652
Citations Scopus - 315Web of Science - 290
2002 Bartlett N, Symons JA, Tscharke DC, Smith GL, 'The vaccinia virus N1L protein is an intracellular homodimer that promotes virulence', JOURNAL OF GENERAL VIROLOGY, 83 1965-1976 (2002)
DOI 10.1099/0022-1317-83-8-1965
Citations Scopus - 108Web of Science - 104
Show 89 more journal articles

Conference (35 outputs)

Year Citation Altmetrics Link
2023 Williams L, Berthon B, Bartlett N, Wark P, Wood L, 'Bacterial metabolites of dietary fibre fermentation, propionate and butyrate, reduce type 2 cytokine responses by peripheral blood mononuclear cells from subjects with asthma', PROCEEDINGS OF THE NUTRITION SOCIETY (2023)
DOI 10.1017/S0029665123000873
Co-authors Bronwyn Berthon
2023 Kanwal A, Bartlett NW, Schuliga M, Reid AT, Jackson C, 'Increased IL-25 Expression in Idiopathic Pulmonary Fibrosis and Role in Fibroblast Proliferation and Activation', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, DC, Washington (2023)
Co-authors Andrew Reid
2023 Dy AC, Girkin J, Marrocco A, Collison A, Mwase C, O'Sullivan MJ, et al., 'Rhinovirus Infection Induces Endothelin-1 Secretion From Human Bronchial Epithelial Cells and in Mouse Models of Allergic Airway Disease', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, DC, Washington (2023)
Co-authors Adam Collison, Joerg Mattes
2022 Bartlett NW, Williams T, Loo S, Girkin J, 'IL-25 Inhibits Airway Anti-Viral Immunity and Promotes Virus Exacerbation of Allergic Airways Disease', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, San Francisco, CA (2022)
Co-authors Suling Loo
2022 Anderson GP, Jarnicki A, Loo S, Ciccotosto J, Girkin J, O'Donoghue R, et al., 'Development of TLR2/6 Agonist (INNA-051) to Protect the Elderly Against Respiratory Virus Infection', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, CA, San Francisco (2022)
Co-authors Suling Loo, Jason Girkin
2022 Girkin J, Bryant NE, Loo S, Demaison C, Mercuri F, Bartlett NW, 'TLR2/6 Agonist Treatment Enhances Antiviral Innate Immune Responses in a Novel Mouse Coronavirus Respiratory Infection Model', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, San Francisco, CA (2022)
Co-authors Suling Loo
2021 Ngan FL, Reid A, Nichol K, Grainge C, Wark P, Knight D, Bartlett N, 'Dysregulated actin cytoskeleton associated with barrier dysfunction in asthma', FASEB JOURNAL, ELECTR NETWORK (2021)
DOI 10.1096/fasebj.2021.35.S1.03196
Co-authors Christopher Grainge, Andrew Reid
2021 Malik B, Mckerrow R, Harrington J, Upham J, Bartlett N, Wark P, 'A COMPARISON OF GROUP 2 INNATE LYMPHOID CELLS (ILC2s) BETWEEN SEVERE ASTHMA, HEALTHY CONTROLS WITH ATOPY AND NON-ATOPIC HEALTHY CONTROLS', RESPIROLOGY (2021)
2020 Wark P, Pathinyake P, Kaiko G, Sohal S, Oldmeadow C, Bartlett N, et al., 'Late Breaking Abstract-ACE2 expression in lower airway epithelial cells is increased with age and in males, but is less in asthma', EUROPEAN RESPIRATORY JOURNAL (2020)
DOI 10.1183/13993003.congress-2020.4150
Co-authors Gerard Kaiko, Christopher Oldmeadow, Prabuddha Pathinayake
2020 Wei L, Read J, Reid AT, Bartlett NW, Grainge C, Knight DA, 'Intrinsic Asthma and Type-2 Cytokines Mediated STAT1 Response to Rhinovirus in Bronchial Epithelial Cells', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, ELECTR NETWORK (2020)
Co-authors Christopher Grainge, Andrew Reid
2020 Kanwal A, Grainge C, Knight DA, Schuliga M, Bartlett NW, 'The Fibrogenic Actions of IL-25 and Its Potential Role in Idiopathic Pulmonary Fibrosis (IPF)', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, ELECTR NETWORK (2020)
Co-authors Christopher Grainge, Michael Schuliga
2020 Malik B, Bartlett N, Mckerrow R, Harrington J, Upham J, Wark P, 'A COMPARISON OF BLOOD INNATE LYMPHOID CELLS BETWEEN SEVERE ASTHMA, HEALTHY CONTROLS WITH ATOPY AND NON-ATOPIC HEALTHY CONTROLS', RESPIROLOGY (2020)
2020 Malik B, Bartlett N, Mckerrow R, Harrington J, Upham JW, Wark PAB, 'Comparison of blood ILC2 cell numbers, phenotypes and proliferation between atopic severe asthma participants, healthy controls with atopy and non-atopic healthy controls', EUROPEAN RESPIRATORY JOURNAL (2020)
DOI 10.1183/13993003.congress-2020.319
2019 Williams T, Girkin J, Nichol K, Knight D, Alton K, Shimkets R, Bartlett N, 'IL-25 BLOCKADE AUGMENTS EPITHELIAL ANTIVIRAL IMMUNITY DURING RHINOVIRUS INFECTION', RESPIROLOGY (2019)
Co-authors Jason Girkin
2019 Li N, Reid AT, Nichol KS, Grainge C, Wark PAB, Knight DA, Bartlett NW, 'Differentiating Bronchial Epithelial Cells from Patients with Asthma Display Prolonged Repair and Delayed Barrier Formation', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, Dallas, TX (2019)
Co-authors Christopher Grainge, Andrew Reid
2019 Veerati PC, Reid A, Nichol K, Wark P, Bartlett N, Knight D, Grainge C, 'Asthmatic airway epithelial cells subjected to apical mechanical stress exhibit suppressed interferon release following viral infection', EUROPEAN RESPIRATORY JOURNAL, Madrid, SPAIN (2019)
DOI 10.1183/13993003.congress-2019.PA3337
Citations Web of Science - 1
Co-authors Punnam Veerati, Andrew Reid, Christopher Grainge
2019 Bartlett NW, Loo S, Girkin J, Jackson DC, Mercuri F, Demaison C, 'Airway Epithelial Cells from Patients with Asthma and COPD Exhibit Improved Resistance to Rhinovirus Infection Following Treatment with TLR2 Immune Modulators', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, Dallas, TX (2019)
Co-authors Suling Loo
2019 Schuliga M, Jaffar J, Westall G, Blokland K, Waters D, Burgess J, et al., 'CGAS IS A PIVOTAL MEDIATOR OF IPF LUNG FIBROBLAST SENESCENCE', RESPIROLOGY (2019)
Co-authors Christopher Grainge, Michael Schuliga
2018 Bartlett N, Girkin J, Williams T, Vincent T, Jackson C, Alton K, Shimkets R, 'ABM125 Anti-IL-25 Antibody Pre-Clinical Development for Viral Asthma Exacerbations Identifies IL-25 Mediated Regulation of Type-2-and Anti-Viral Immunity', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, San Diego, CA (2018)
Citations Web of Science - 2
Co-authors Jason Girkin
2018 Loo S, Nichol K, Hsu A, Londrigan S, Reading P, Bartlett N, Wark P, 'AIRWAY EPITHELIAL INNATE IMMUNE RESPONSES TO CORONAVIRUSES', RESPIROLOGY (2018)
Citations Web of Science - 1
Co-authors Suling Loo
2018 Reid A, Nichol K, Wei L, Moheimani F, Bartlett N, Hansbro P, et al., 'NOTCH3 INHIBITION SIGNIFICANTLY REDUCES MUC5AC IN HUMAN AIRWAY EPITHELIAL CELLS', RESPIROLOGY (2018)
Co-authors Christopher Grainge
2018 Reid AT, Nichol KS, Wei L, Moheimani F, Bartlett NW, Hansbro PM, et al., 'Inhibition of NOTCH3 Signaling Abolishes MUC5AC Production in Human Airway Epithelial Cells', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, San Diego, CA (2018)
Co-authors Christopher Grainge, Andrew Reid
2018 Veerati P, Reid A, Nichol K, Ngan L, Teresa W, Wark PAB, et al., 'A Physiological Relevant Rhinovirus Infection Model in Differentiated Human Primary Bronchial Epithelial Cells from Healthy, Asthmatic and COPD Donors', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, San Diego, CA (2018)
Co-authors Punnam Veerati, Christopher Grainge
2018 Veerati P, Bartlett NW, Nichol K, Wark PAB, Knight DA, Grainge CL, 'Mechanical Forces Suppress Innate Anti-Viral Immunity in Primary Human Airway Epithelial Cells Obtained from Asthma Donors', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, San Diego, CA (2018)
Co-authors Christopher Grainge, Punnam Veerati
2018 Bartlett NW, Girkin J, Wong C, Deliyannis G, Zeng W, Demaison C, Jackson DC, 'Upper Airway TLR2 Immune Modulators Prime Broad Respiratory Immunity Against Rhinovirus and Influenza Infection and Inhibit Subsequent Lung Inflammation', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, San Diego, CA (2018)
Co-authors Jason Girkin
2017 Loo S-L, Hsu A, Bartlett N, Wark P, 'AIRWAY EPITHELIAL INNATE IMMUNE RESPONSES TO CORONAVIRUSES', RESPIROLOGY (2017)
Co-authors Suling Loo
2017 Veerati P, Bartlett N, Parsons K, Moheimani F, Wark P, Knight D, Grainge C, 'MECHANICAL FORCES ATTENUATE ANTI-VIRAL IMMUNITY IN PRIMARY HUMAN AIRWAY EPITHELIAL CELLS FROM ASTHMATIC DONORS', RESPIROLOGY (2017)
Co-authors Christopher Grainge, Punnam Veerati
2017 Reid A, Moheimani F, Nichol K, Bartlett N, Wark P, Grainge C, Knight D, 'ACUTE INHIBITION OF NOTCH SIGNALLING ABLATES MUC5AC PRODUCTION IN HUMAN AIRWAY EPITHELIAL CELLS FROM ASTHMATIC, NON-ASTHMATIC AND COPD DONORS.', RESPIROLOGY (2017)
Citations Web of Science - 1
Co-authors Christopher Grainge, Andrew Reid
2017 Reid AT, Moheimani F, Nichol K, Bartlett N, Wark PAB, Grainge C, et al., 'Short-Term Inhibition Of Notch Signalling Ablates Muc5ac Production In Human Airway Epithelial Cells From Asthmatic, Non-Asthmatic And COPD Donors', AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, Washington, DC (2017)
Co-authors Christopher Grainge, Andrew Reid
2016 Bartlett N, Singanayagam A, Johnston S, 'INHALED CORTICOSTEROIDS SUPPRESS INNATE AND ADAPTIVE ANTI-VIRAL IMMUNE RESPONSES IN THE AIRWAYS', RESPIROLOGY (2016)
2016 Tay H, Yang M, Hsu A, Nguyen T-H, Plank M, Maltby S, et al., 'Role of interleukin-36 gamma in regulating lung inflammation', EUROPEAN JOURNAL OF IMMUNOLOGY, Melbourne, AUSTRALIA (2016)
Co-authors Steven Maltby, Ming Yang
2014 Collison A, Hatchwell L, Girkin J, Parsons K, Li J, Zhang J, et al., 'Late-breaking abstract: IL-5-induced airways eosinophilia as a negative regulator of TLR7 expression may impair the interferon response to rhinovirus in allergic airways', EUROPEAN RESPIRATORY JOURNAL (2014)
Co-authors Adam Collison, Joerg Mattes, Jason Girkin
2012 Collison AM, Hatchwell LM, Siqueira AP, Bartlett NW, Johnston SL, Foster PS, Mattes J, 'Antagonism of microRNA-122 is comparible to azithromycin treatment in a mouse model of rhinovirus-induced exacerbation of allergic airways disease', Respirology, Canberra, ACT (2012) [E3]
Co-authors Joerg Mattes, Adam Collison
2012 Hatchwell LM, Collison AM, Siqueira AP, Bartlett NW, Johnston SL, Foster PS, Mattes J, 'Toll-like receptor 7 mediates anti-viral responses to rhinovirus while suppressing exacerbation of asthma', Respirology, Canberra, ACT (2012) [E3]
Co-authors Adam Collison, Joerg Mattes
2012 Hatchwell LM, Collison AM, Siqueira AP, Bartlett NW, Johnston SL, Foster PS, Mattes J, 'TRAIL regulates inflammatory responses to rhinovirus and rhinovirus-induced exacerbation of asthma', Respirology, Canberra, ACT (2012) [E3]
Co-authors Adam Collison, Joerg Mattes
Show 32 more conferences
Edit

Grants and Funding

Summary

Number of grants 48
Total funding $21,514,090

Click on a grant title below to expand the full details for that specific grant.


20241 grants / $276,000

PREVENT (Pandemic Respiratory Virus survEillaNce Trial)$276,000

Funding body: NHMRC (National Health & Medical Research Council)

Funding body NHMRC (National Health & Medical Research Council)
Project Team Professor Josh Davis, Professor Nathan Bartlett, Dr Craig Dalton, Dr James Fielding, Dr Adrian Marcato, Peter Massey, Professor Jodie McVernon, Professor Kanta Subbarao
Scheme Partnership Projects
Role Investigator
Funding Start 2024
Funding Finish 2024
GNo G2400245
Type Of Funding Scheme excluded from IGS
Category EXCL
UON Y

20235 grants / $257,315

Development of antiviral RNA therapeutics targeting SARS-CoV-2 infection$124,666

Funding body: Department of Health and Aged Care

Funding body Department of Health and Aged Care
Project Team Doctor Roger Liang, Professor Nathan Bartlett, Professor Anthony Kelleher, Dr Chantelle Ahlenstiel, Professor Maria Kavallaris, Professor Philip Hansbro, Professor Daniela Traini, Professor Pall Thordarson, Associate Professor Kathy Petoumenos, A/P Stuart Turville, Cees van Rijin
Scheme MRFF - COVID-19 Treatment Access and Public Health Activities
Role Investigator
Funding Start 2023
Funding Finish 2024
GNo G2200959
Type Of Funding C1300 - Aust Competitive - Medical Research Future Fund
Category 1300
UON Y

TLR2/6 agonist boosting immunogenicity of tumour-targeting RNAi-based nanomedicines$80,000

Funding body: NSW Ministry of Health

Funding body NSW Ministry of Health
Project Team Professor Nathan Bartlett, Doctor Roger Liang, Miss Ellie Doubleday, Dr Anna Galkin
Scheme RNA Future Leaders PhD Program
Role Lead
Funding Start 2023
Funding Finish 2026
GNo G2200072
Type Of Funding C2400 – Aust StateTerritoryLocal – Other
Category 2400
UON Y

EXercise for Exacerbations and Rhinovirus Treatment (EXERT): human in vitro and mouse in vivo COPD models$27,700

Funding body: Lung Foundation Australia

Funding body Lung Foundation Australia
Project Team Professor Nathan Bartlett, Professor Anne Holland
Scheme Early Career Fellowship for Lung Cancer Research
Role Lead
Funding Start 2023
Funding Finish 2024
GNo G2300239
Type Of Funding C1700 - Aust Competitive - Other
Category 1700
UON Y

Intranasal lactoferrin to inhibit coronavirus infection$20,000

Funding body: Noumi Operations Pty Ltd

Funding body Noumi Operations Pty Ltd
Project Team Professor Nathan Bartlett, Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2023
Funding Finish 2023
GNo G2300957
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Improving arbovirus surveillance in the Hunter region through the development of PCR based assay to survey mosquito species and arboviruses.$4,949

Funding body: University of Newcastle

Funding body University of Newcastle
Project Team Doctor Camille Esneau, Professor Nathan Bartlett, Doctor Toby Mills
Scheme Pilot Funding Scheme
Role Investigator
Funding Start 2023
Funding Finish 2023
GNo G2300473
Type Of Funding Internal
Category INTE
UON Y

20226 grants / $275,225

LNR125 anti-IL-25 blocking rhinovirus induced airway neutrophilic inflammation during allergic airways disease exacerbation$135,600

Funding body: Lanier Biotherapeutics, Inc

Funding body Lanier Biotherapeutics, Inc
Project Team Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2022
Funding Finish 2022
GNo G2201153
Type Of Funding C3400 – International For Profit
Category 3400
UON Y

Lactorferrin (PUREnFERRIN) - In Vitro Studies & POF Model$50,000

Funding body: Pactum Dairy Group Pty Ltd

Funding body Pactum Dairy Group Pty Ltd
Project Team Professor Nathan Bartlett
Scheme Entrepreneurs' Programme: Innovation Connections
Role Lead
Funding Start 2022
Funding Finish 2022
GNo G2101405
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Lactorferrin (PUREnFERRIN) - In Vitro Studies & POF Model$50,000

Funding body: Department of Industry, Science, Energy and Resources

Funding body Department of Industry, Science, Energy and Resources
Project Team Professor Nathan Bartlett
Scheme Entrepreneurs' Programme: Innovation Connections
Role Lead
Funding Start 2022
Funding Finish 2022
GNo G2101406
Type Of Funding C2200 - Aust Commonwealth – Other
Category 2200
UON Y

Anitviral Efficacy of Exintech Face Masks for Pathogen Mitigation$15,000

Funding body: Exintech Pty Ltd

Funding body Exintech Pty Ltd
Project Team Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2022
Funding Finish 2022
GNo G2200899
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Testing of SARS-CoV-2 inactivation protocols$13,375

Funding body: Avicena Systems Ltd

Funding body Avicena Systems Ltd
Project Team Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2022
Funding Finish 2022
GNo G2200811
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Testing the antiviral efficacy of Dulux coatings for pathogen mitigation$11,250

Funding body: Dulux Group (Australia) Pty Ltd

Funding body Dulux Group (Australia) Pty Ltd
Project Team Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2022
Funding Finish 2022
GNo G2200951
Type Of Funding Scheme excluded from IGS
Category EXCL
UON Y

20218 grants / $522,043

Lactoferrin supplementation, Immune Function & Respiratory Virus Infection$199,986

Funding body: Freedom Foods Group Nutritionals Pty Ltd

Funding body Freedom Foods Group Nutritionals Pty Ltd
Project Team Professor Lisa Wood, Professor Nathan Bartlett, Doctor Bronwyn Berthon, Doctor Evan Williams
Scheme Entrepreneurs' Programme: Innovation Connections
Role Investigator
Funding Start 2021
Funding Finish 2022
GNo G2100982
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Efficacy of UV-C devices designed and built by GERMII on known pathogens $61,200

Funding body: Germii Pathogen Mitigation Pty Ltd

Funding body Germii Pathogen Mitigation Pty Ltd
Project Team Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2021
Funding Finish 2021
GNo G2101118
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Assessment of UV lamp sterilisation effectiveness$50,637

Funding body: Fusion HVAC Australia Pty Ltd

Funding body Fusion HVAC Australia Pty Ltd
Project Team Professor Nathan Bartlett
Scheme Entrepreneurs' Programme: Innovation Connections
Role Lead
Funding Start 2021
Funding Finish 2021
GNo G2100685
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Assessment of UV lamp sterilisation effectiveness$50,000

Funding body: Department of Industry, Innovation and Science

Funding body Department of Industry, Innovation and Science
Project Team Professor Nathan Bartlett
Scheme Entrepreneurs' Programme: Innovation Connections
Role Lead
Funding Start 2021
Funding Finish 2021
GNo G2100739
Type Of Funding C2200 - Aust Commonwealth – Other
Category 2200
UON Y

Lactoferrin supplementation, Immune Function & Respiratory Virus Infection$50,000

Funding body: Department of Industry, Innovation and Science

Funding body Department of Industry, Innovation and Science
Project Team Professor Lisa Wood, Professor Nathan Bartlett, Doctor Bronwyn Berthon, Doctor Evan Williams
Scheme Entrepreneurs' Programme: Innovation Connections
Role Investigator
Funding Start 2021
Funding Finish 2022
GNo G2101004
Type Of Funding C2200 - Aust Commonwealth – Other
Category 2200
UON Y

Airway-targeting anti-viral treatments for COVID-19$49,035

Funding body: Hunter Medical Research Institute

Funding body Hunter Medical Research Institute
Project Team Professor Nathan Bartlett, A/P Stuart Turville
Scheme Research Grant
Role Lead
Funding Start 2021
Funding Finish 2021
GNo G2100149
Type Of Funding C3300 – Aust Philanthropy
Category 3300
UON Y

Transcriptomics to define host-coronavirus interaction$37,435

Funding body: Hunter Medical Research Institute

Funding body Hunter Medical Research Institute
Project Team Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2021
Funding Finish 2022
GNo G2101051
Type Of Funding C3300 – Aust Philanthropy
Category 3300
UON Y

Pathopen: a Rapid Saliva Point-of-Care Diagnostic for COVID-19$23,750

Funding body: Hunter Medical Research Institute

Funding body Hunter Medical Research Institute
Project Team Laureate Professor Roger Smith, Professor Nathan Bartlett, Professor Paul Dastoor, Doctor Kaushik Maiti
Scheme Research Grant
Role Investigator
Funding Start 2021
Funding Finish 2021
GNo G2101061
Type Of Funding C3300 – Aust Philanthropy
Category 3300
UON Y

20209 grants / $423,439

INNA compounds for the prevention of respiratory virus-induced disease$136,350

Funding body: Ena Respiratory Pty Ltd

Funding body Ena Respiratory Pty Ltd
Project Team Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2020
Funding Finish 2020
GNo G2000959
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Pre-clinical respiratory virus infection models for screening N-myristoyltransferase inhibitors for anti-viral activity $73,600

Funding body: Myricx Pharma Limited

Funding body Myricx Pharma Limited
Project Team Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2020
Funding Finish 2020
GNo G2000562
Type Of Funding C3400 – International For Profit
Category 3400
UON Y

In vitro studies of PUREnFERRIN™ Lactoferrin$49,418

Funding body: Pactum Dairy Group Pty Ltd

Funding body Pactum Dairy Group Pty Ltd
Project Team Professor Nathan Bartlett
Scheme Entrepreneurs' Programme: Innovation Connections
Role Lead
Funding Start 2020
Funding Finish 2020
GNo G2001275
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

In vitro studies of PUREnFERRIN™ Lactoferrin$49,418

Funding body: Department of Industry, Innovation and Science

Funding body Department of Industry, Innovation and Science
Project Team Professor Nathan Bartlett
Scheme Entrepreneurs' Programme: Innovation Connections
Role Lead
Funding Start 2020
Funding Finish 2020
GNo G2001362
Type Of Funding C2200 - Aust Commonwealth – Other
Category 2200
UON Y

Zexa Sure Shield Sanitiser coronavirus inactivity research$41,600

Funding body: Sirron Holdings Group Pty Ltd

Funding body Sirron Holdings Group Pty Ltd
Project Team Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2020
Funding Finish 2020
GNo G2000576
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Antiviral Efficacy testing for Silicone (PDMS) embedded with Nano silver$32,020

Funding body: Silfresh Pty Ltd

Funding body Silfresh Pty Ltd
Project Team Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2020
Funding Finish 2021
GNo G2001198
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Test Clear Diamond (self-adhesive, silver ion coated plastic film) against coronavirus at various time intervals to determine how quickly and how effective the product is at destroying this virus$24,533

Funding body: Schaffer and Co Pty Ltd

Funding body Schaffer and Co Pty Ltd
Project Team Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2020
Funding Finish 2020
GNo G2000849
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Metis fabric coronavirus inactivating activity$11,500

Funding body: Metis Technologies Pty Ltd

Funding body Metis Technologies Pty Ltd
Project Team Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2020
Funding Finish 2020
GNo G2000976
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

RNAi nanomedicine for coronavirus infection $5,000

Funding body: Hunter Medical Research Institute

Funding body Hunter Medical Research Institute
Project Team Professor Nathan Bartlett, Doctor Roger Liang
Scheme Research Grant
Role Lead
Funding Start 2020
Funding Finish 2020
GNo G2000216
Type Of Funding C3200 – Aust Not-for Profit
Category 3200
UON Y

20195 grants / $1,087,705

In vitro human corneal epithelial cell culture models for screening novel AT-API conjugates for the treatment of dry eye disease$886,745

Funding body: Foray Therapeutics Pty Ltd

Funding body Foray Therapeutics Pty Ltd
Project Team Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2019
Funding Finish 2023
GNo G1900809
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Confirm the mechanism of action (MOA) and determine the efficacy of the Company’s therapeutic INNA-X$50,699

Funding body: Ena Therapeutics Pty Ltd

Funding body Ena Therapeutics Pty Ltd
Project Team Professor Nathan Bartlett
Scheme Entrepreneurs' Programme: Innovation Connections
Role Lead
Funding Start 2019
Funding Finish 2020
GNo G1901545
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Define the mechanism of action and determine the efficacy of the Company’s therapeutic drug$50,261

Funding body: Ena Therapeutics Pty Ltd

Funding body Ena Therapeutics Pty Ltd
Project Team Professor Nathan Bartlett, Doctor Su Ling Loo
Scheme Entrepreneurs' Programme: Innovation Connections
Role Lead
Funding Start 2019
Funding Finish 2019
GNo G1900110
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Define the mechanism of action and determine the efficacy of the Company’s therapeutic drug$50,000

Funding body: Department of Industry, Innovation and Science

Funding body Department of Industry, Innovation and Science
Project Team Professor Nathan Bartlett, Doctor Su Ling Loo
Scheme Entrepreneurs' Programme: Innovation Connections
Role Lead
Funding Start 2019
Funding Finish 2019
GNo G1900266
Type Of Funding C2200 - Aust Commonwealth – Other
Category 2200
UON Y

Confirm the mechanism of action (MOA) and determine the efficacy of the Company’s therapeutic INNA-X$50,000

Funding body: Department of Industry, Innovation and Science

Funding body Department of Industry, Innovation and Science
Project Team Professor Nathan Bartlett
Scheme Entrepreneurs' Programme: Innovation Connections
Role Lead
Funding Start 2019
Funding Finish 2020
GNo G2000024
Type Of Funding C2200 - Aust Commonwealth – Other
Category 2200
UON Y

20181 grants / $1,104,379

How does bronchoconstriction worsen asthma? $1,104,379

Funding body: NHMRC (National Health & Medical Research Council)

Funding body NHMRC (National Health & Medical Research Council)
Project Team Conjoint Associate Professor Christopher Grainge, Professor Nathan Bartlett, Professor Darryl Knight, Conjoint Professor Peter Wark, Professor Alastair Stewart, Stewart, Alastair
Scheme Project Grant
Role Investigator
Funding Start 2018
Funding Finish 2021
GNo G1700343
Type Of Funding C1100 - Aust Competitive - NHMRC
Category 1100
UON Y

20174 grants / $1,422,397

Shared innate immune mechanisms underpin-steroid resistant pathogen-induced asthma exacerbations$844,726

Funding body: NHMRC (National Health & Medical Research Council)

Funding body NHMRC (National Health & Medical Research Council)
Project Team Professor Paul Foster, Associate Professor Ming Yang, Professor Nathan Bartlett
Scheme Project Grant
Role Investigator
Funding Start 2017
Funding Finish 2020
GNo G1600084
Type Of Funding C1100 - Aust Competitive - NHMRC
Category 1100
UON Y

Pre-clinical studies with REG3500 and dupilumab$421,307

Funding body: Sanofi US Services Inc.

Funding body Sanofi US Services Inc.
Project Team Professor Nathan Bartlett, Conjoint Professor Peter Wark
Scheme Research Project
Role Lead
Funding Start 2017
Funding Finish 2019
GNo G1701373
Type Of Funding C3400 – International For Profit
Category 3400
UON Y

Novel epithelial targets and targeting strategies to prevent asthma exacerbations$136,364

Funding body: Asthma Australia

Funding body Asthma Australia
Project Team Professor Nathan Bartlett, Conjoint Professor Peter Wark, Doctor Roger Liang, Professor Darryl Knight
Scheme National Research Program
Role Lead
Funding Start 2017
Funding Finish 2018
GNo G1601217
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Plasma Torque Teno Virus load as a novel tool to monitor intensity of immunosuppression in renal transplant recipients$20,000

Funding body: Hunter Medical Research Institute

Funding body Hunter Medical Research Institute
Project Team Professor Josh Davis, Professor Nathan Bartlett, Dr Peter Choi
Scheme Project Grant
Role Investigator
Funding Start 2017
Funding Finish 2017
GNo G1701566
Type Of Funding C3300 – Aust Philanthropy
Category 3300
UON Y

20163 grants / $1,471,783

Inhibition of rhinovirus-induced disease by TLR2 agonist$726,548

Funding body: Ena Therapeutics Pty Ltd

Funding body Ena Therapeutics Pty Ltd
Project Team Professor Nathan Bartlett, Conjoint Professor Peter Wark
Scheme Research Grant
Role Lead
Funding Start 2016
Funding Finish 2019
GNo G1600747
Type Of Funding C3100 – Aust For Profit
Category 3100
UON Y

Collaborative Research Agreement: New pathways and targets in severe asthma and COPD$611,171

Funding body: Boehringer Ingelheim Pharma GmbH & Co KG

Funding body Boehringer Ingelheim Pharma GmbH & Co KG
Project Team Professor Darryl Knight, Conjoint Associate Professor Christopher Grainge, Conjoint Professor Peter Wark, Professor Nathan Bartlett
Scheme Research Grant
Role Investigator
Funding Start 2016
Funding Finish 2019
GNo G1601257
Type Of Funding C3400 – International For Profit
Category 3400
UON Y

Efficacy of ABM109 in blocking rhinovirus induced asthma exacerbation: preclinical proof of concept$134,064

Funding body: Abeome Corporation

Funding body Abeome Corporation
Project Team Professor Nathan Bartlett
Scheme Research Grant
Role Lead
Funding Start 2016
Funding Finish 2016
GNo G1600775
Type Of Funding C3400 – International For Profit
Category 3400
UON Y

20152 grants / $50,238

Novel anti-viral agent for rhinovirus infection$25,238

Determining the efficacy of a novel anti-viral compound for rhinovirus infection.

Funding body: 3E Therapeutics

Funding body 3E Therapeutics
Project Team

Nathan Bartlett

Scheme Commercial
Role Lead
Funding Start 2015
Funding Finish 2015
GNo
Type Of Funding External
Category EXTE
UON N

Specifically targeting the airways to prevent virus-induced asthma attacks$25,000

Funding body: Hunter Medical Research Institute

Funding body Hunter Medical Research Institute
Project Team Professor Nathan Bartlett, Professor Darryl Knight
Scheme Project Grant
Role Lead
Funding Start 2015
Funding Finish 2016
GNo G1501378
Type Of Funding Grant - Aust Non Government
Category 3AFG
UON Y

20141 grants / $23,566

Miltenyi Biotec GentleMACS Octo Dissociator with Heaters $23,566

Funding body: NHMRC (National Health & Medical Research Council)

Funding body NHMRC (National Health & Medical Research Council)
Project Team Professor Phil Hansbro, Professor Paul Foster, Professor Darryl Knight, Professor Dirk Van Helden, Professor Joerg Mattes, Professor Jodie Simpson, Professor Lisa Wood, Prof LIZ Milward, Dr NATHAN Bartlett, Professor Simon Keely, Doctor Steven Maltby, Doctor Andrew Jarnicki, Doctor Malcolm Starkey, Associate Professor Adam Collison, Doctor Shaan Gellatly
Scheme Equipment Grant
Role Investigator
Funding Start 2014
Funding Finish 2014
GNo G1500861
Type Of Funding Other Public Sector - Commonwealth
Category 2OPC
UON Y

20122 grants / $4,600,000

Mechanisms of interplay between allergy and viruses in asthma$4,000,000

We apply to form an Alliance between GSK and the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma to discover and develop new approaches for prevention/treatment of asthma and asthma attacks. We will integrate research in areas of major strength for the Centre with the strengths in asthma drug discovery at GSK. The overall aim is to investigate mechanisms of interplay between allergy and virus infection in development of asthma and in acute asthma attacks. 
The major cause of asthma attacks are human rhinovirus (RV) infections, these are also related to development of asthma when they occur in early life. We will investigate gene expression and how expression is regulated in lung cells during RV infection to identify molecules/pathways induced by RV infection that are implicated in promoting allergic responses.
We will determine whether RV induction/modification of these molecules/processes are related to pre-existing allergic responses, virus load and clinical severity of asthma exacerbation. 
A major cause of asthma is failure of development of immunological tolerance to allergens. We have shown that tolerance can be reversed by a virus infection, but the mechanisms are unknown. We will purify lung cells from the virus-induced breakdown of tolerance model to identify genes implicated in tolerance breakdown and determine whether these are replicated in lung cells in the human.
Finally we will determine whether blocking or inducing molecules implicated in virus induced worsening of allergic responses/breakdown of tolerance, determines severity of RV induced allergic airway inflammation.
Accomplishment of these aims will identify targets for development of novel therapies for asthma and asthma attacks.

Funding body: Medical Research Council of the United Kingdom

Funding body Medical Research Council of the United Kingdom
Project Team

Sebastian Johnston, Nathan Bartlett, CIs in MRC Centre for Allergic Mechanisms in Asthma

Scheme Program Grant
Role Investigator
Funding Start 2012
Funding Finish 2017
GNo
Type Of Funding International - Competitive
Category 3IFA
UON N

Defining the importance of IL-25 in rhinovirus induced asthma exacerbations$600,000

Rhinovirus infection induces production of IL-25, augmenting Th2-type immune responses and exacerbating allergic airways inflammation. Thus IL-25 plays a central role in the pathogenesis of RV-induced asthma exacerbations and is a good candidate target for the development of new therapies for asthma aexacerbations. 

The aim of this application is to investigate rhinovirus-induced IL-25 and define its role in RV-induced asthma exacerbations using a combination of patient tissue analysis and experimental mouse models. If IL-25 expression is induced and related to disease outcomes in the human model, AND causally related to Th2 mediated disease outcomes in the mouse in vivo models this cytokine will be a very strong candidate for immediate translation of approaches to inhibit expression or function in pivotal proof of concept human intervention studies. 

Funding body: Medical Research Council of the United Kingdom

Funding body Medical Research Council of the United Kingdom
Project Team

Nathan Bartlett, Sebastian Johnston

Scheme Project Grant
Role Lead
Funding Start 2012
Funding Finish 2014
GNo
Type Of Funding International - Competitive
Category 3IFA
UON N

20101 grants / $10,000,000

Post infectious immune reprogramming and its association with persistence and chronicity of respiratory allergic diseases (PreDicta)$10,000,000

The PreDicta project is based on the observation that childhood asthma usually occurs after a viral respiratory tract infection. Thus, the central hypothesis of this programme is that repeated, acute infection-mediated events may reprogram the innate, adaptive and/or regulatory immune responses to predispose towards a chronic inflammation pattern.

To analyze the relationship between these infections with persistence of asthma, Predicta will employ the latest technologies of molecular biology, virology, and cytology.  The Consortium is privileged to include groups with an excellent track record in clinical research and cohort-based studies that have significantly contributed to the field of allergy and asthma research internationally.

Funding body: European Commission, European Union

Funding body European Commission, European Union
Project Team

Nikolaos Papadopoulos, Sebastian Johnston, Nathan Bartlett, PreDicta Consortium

Scheme 7th Research Framework Programme
Role Investigator
Funding Start 2010
Funding Finish 2016
GNo
Type Of Funding International - Competitive
Category 3IFA
UON N
Edit

Research Supervision

Number of supervisions

Completed15
Current6

Current Supervision

Commenced Level of Study Research Title Program Supervisor Type
2023 PhD Aptamer Optimisation And Selection For Non-Invasive Monitoring Of Cancer Using A Breathalyser PhD (Biological Sciences), College of Engineering, Science and Environment, The University of Newcastle Co-Supervisor
2023 PhD TLR2 Agonist Boosting Immunogenicity of Tumour-Targeting RNAi-Based Nanomedicines PhD (Immunology & Microbiol), College of Health, Medicine and Wellbeing, The University of Newcastle Principal Supervisor
2023 PhD Smartphone Aptasensor For The Detection Of Infectious Diseases PhD (Physics), College of Engineering, Science and Environment, The University of Newcastle Co-Supervisor
2022 PhD Airway Epithelial Cell-Targeting Nanoparticles to Harness RNA Interference Therapeutics for Respiratory Virus Induced Diseases PhD (Immunology & Microbiol), College of Health, Medicine and Wellbeing, The University of Newcastle Principal Supervisor
2021 PhD Topical, Sustained Release Drug Delivery Platform for the Treatment of Ocular Pain PhD (Immunology & Microbiol), College of Health, Medicine and Wellbeing, The University of Newcastle Principal Supervisor
2021 PhD Targeting Mitochondrial ROS in Airway Epithelial Cells to Prevent Virus Induced Disease in Asthma and COPD PhD (Pharmacy), College of Health, Medicine and Wellbeing, The University of Newcastle Co-Supervisor

Past Supervision

Year Level of Study Research Title Program Supervisor Type
2023 PhD Soluble Fibre as an Anti-Inflammatory Treatment for Asthma PhD (Nutritional Biochemistry), College of Health, Medicine and Wellbeing, The University of Newcastle Co-Supervisor
2023 PhD The Fibrogenic Actions of IL-25 in Idiopathic Pulmonary Fibrosis (IPF) PhD (Immunology & Microbiol), College of Health, Medicine and Wellbeing, The University of Newcastle Principal Supervisor
2022 PhD Type 2 Innate Lymphoid Cell Dysfunction in Severe Asthma and Response to Treatment with Mepolizumab and Omalizumab PhD (Medicine), College of Health, Medicine and Wellbeing, The University of Newcastle Co-Supervisor
2022 PhD The Role of STATs in the Interaction of Virus and Type 2 Cytokines in Airway Epithelial Cells PhD (Medical Biochemistry), College of Health, Medicine and Wellbeing, The University of Newcastle Principal Supervisor
2021 PhD The Impact of Respiratory Virus Infection on Airway Epithelial Cell Differentiation and Barrier Formation in Asthma PhD (Immunology & Microbiol), College of Health, Medicine and Wellbeing, The University of Newcastle Principal Supervisor
2021 PhD Development of Airway Epithelial Targeted Nanoparticles Loaded with TLR7 agonist for Asthma Therapy PhD (Pharmacy), College of Health, Medicine and Wellbeing, The University of Newcastle Co-Supervisor
2021 PhD The Interplay between Epithelial-derived Type-2 Inflammation and Rhinovirus Infection in Asthma PhD (Immunology & Microbiol), College of Health, Medicine and Wellbeing, The University of Newcastle Principal Supervisor
2020 PhD Rhinovirus Diversity and Replication in Differentiated Airway Epithelial Cells PhD (Immunology & Microbiol), College of Health, Medicine and Wellbeing, The University of Newcastle Principal Supervisor
2019 PhD Role of Mechanical Forces in Asthma Pathogenesis PhD (Medicine), College of Health, Medicine and Wellbeing, The University of Newcastle Co-Supervisor
2018 PhD Innate Anti-Viral Responses of Airway Epithelial Cells to Infection with Rhinovirus and Coronavirus PhD (Medicine), College of Health, Medicine and Wellbeing, The University of Newcastle Co-Supervisor
2015 PhD Innate Th2 and anti-viral immune responses in rhinovirus-induced asthma exacerbations
&lt;p style="margin-bottom:0cm;margin-bottom:.0001pt;text-autospace:none;"&gt;&lt;span lang="EN-US" style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;W&lt;/span&gt;&lt;span lang="EN-US" style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;&lt;span style="font-size:small;font-family:Arial, Helvetica, sans-serif;"&gt;e recently used type I IFN receptor knockout (IFNAR1KO) mice to show that IFN signalling regulates both allergic and innate anti-viral immune responses in a model of RV-induced exacerbation of allergic airways inflammation. In the absence of type I IFN signalling, RV infection profoundly exacerbated allergen-induced Th2 cell activation (Figure 1). This suggests that type I IFN signalling regulates Th2 immune responses during RV infection. However, RV infection did not exacerbate allergic airways inflammation in strain-matched wild type C57 Bl/6 mice, which is in contrast to the model established in the Th2-biased Balb/c strain whereby RV increases allergen-driven inflammation (Bartlett &lt;/span&gt;&lt;em&gt;et al&lt;/em&gt;&lt;span style="font-size:small;font-family:Arial, Helvetica, sans-serif;"&gt;., 2008). Together, this suggests that RV-induced exacerbation of allergic airways inflammation may be dependent on impaired type I IFN signalling and augmented Th2 immunity.&amp;nbsp;&lt;/span&gt;&lt;/span&gt;&lt;/p&gt;&lt;p style="margin-bottom:0cm;margin-bottom:.0001pt;text-autospace:none;"&gt;&lt;strong&gt;&lt;span lang="EN-US" style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;Hypothesis:&lt;/span&gt;&lt;/strong&gt;&lt;/p&gt;&lt;p style="margin-bottom:0cm;margin-bottom:.0001pt;text-autospace:none;"&gt;&lt;strong&gt;&lt;span lang="EN-US" style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;Type I IFN signalling modulates Th2-mediated allergic airways inflammation during RV-induced asthma exacerbations. &lt;/span&gt;&lt;/strong&gt;&lt;/p&gt;&lt;p style="margin-bottom:0cm;margin-bottom:.0001pt;text-autospace:none;"&gt;&lt;strong&gt;&lt;span lang="EN-US" style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;Aims:&lt;/span&gt;&lt;/strong&gt;&lt;strong&gt;&lt;span lang="EN-US" style="font-family:'Times New Roman',serif;"&gt;&lt;/span&gt;&lt;/strong&gt;&lt;/p&gt;&lt;p style="margin-bottom:0cm;margin-bottom:.0001pt;"&gt;&lt;span lang="EN-US" style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;Mouse models of OVA-induced allergic airways inflammation and RV-induced exacerbation of allergic airways inflammation will be used to determine the role of type I IFN signalling in RV-induced asthma exacerbations. &lt;/span&gt;&lt;/p&gt;&lt;p style="margin-bottom:0cm;margin-bottom:.0001pt;text-indent:-18.0pt;"&gt;&lt;span lang="EN-US" style="font-family:'Times New Roman',serif;"&gt;&lt;span style="font-size:small;font-family:Arial, Helvetica, sans-serif;"&gt;-&lt;/span&gt;&lt;span style="font-variant-numeric:normal;font-size:7pt;line-height:normal;font-family:'Times New Roman';"&gt;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp; &lt;/span&gt;&lt;/span&gt;&lt;span lang="EN-US" style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;To characterise and compare innate anti-viral and early Th2 responses in a model of RV-induced asthma exacerbation in both wild type Balb/C and C57 Bl/6 mouse strains.&lt;/span&gt;&lt;/p&gt;&lt;p style="margin-bottom:0cm;margin-bottom:.0001pt;text-indent:-18.0pt;"&gt;&lt;span lang="EN-US" style="font-family:'Times New Roman',serif;"&gt;&lt;span style="font-size:small;font-family:Arial, Helvetica, sans-serif;"&gt;-&lt;/span&gt;&lt;span style="font-variant-numeric:normal;font-size:7pt;line-height:normal;font-family:'Times New Roman';"&gt;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp; &lt;/span&gt;&lt;/span&gt;&lt;span lang="EN-US" style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;To use type I IFN receptor knock out (IFNAR1KO) mice in a model of RV-induced asthma exacerbation to determine how type I IFN regulates Th2 immune responses during allergen challenge and RV infection. &lt;/span&gt;&lt;/p&gt;&lt;p style="margin-bottom:0cm;margin-bottom:.0001pt;text-indent:-18.0pt;"&gt;&lt;span lang="EN-US" style="font-family:'Times New Roman',serif;"&gt;&lt;span style="font-size:small;font-family:Arial, Helvetica, sans-serif;"&gt;-&lt;/span&gt;&lt;span style="font-variant-numeric:normal;font-size:7pt;line-height:normal;font-family:'Times New Roman';"&gt;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp; &lt;/span&gt;&lt;/span&gt;&lt;span lang="EN-US" style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;To investigate the effects of recombinant mouse IFN&amp;beta; treatment in mouse models of asthma and RV-induced asthma exacerbation to determine whether type I IFN signalling can inhibit Th2 immune responses in both the absence and presence of RV infection. &lt;/span&gt;&lt;/p&gt;&lt;p style="margin-bottom:0cm;margin-bottom:.0001pt;"&gt;&lt;span lang="EN-US" style="font-family:'Times New Roman',serif;"&gt;&lt;span style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;&lt;/span&gt;&lt;/span&gt;&lt;/p&gt;&lt;p style="margin-bottom:0cm;margin-bottom:.0001pt;"&gt;&lt;span lang="EN-US" style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;&lt;span style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;Human &lt;/span&gt;&lt;em style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;in vitro&lt;/em&gt;&lt;span style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt; cell-based assays will be used to investigate the molecular mechanisms of type I IFN signalling.&amp;nbsp;&lt;/span&gt;&lt;/span&gt;&lt;/p&gt;&lt;p style="margin-bottom:0cm;margin-bottom:.0001pt;text-indent:-18.0pt;"&gt;&lt;span lang="EN-US" style="font-family:'Times New Roman',serif;"&gt;&lt;span style="font-size:small;font-family:Arial, Helvetica, sans-serif;"&gt;-&lt;/span&gt;&lt;span style="font-variant-numeric:normal;font-size:7pt;line-height:normal;font-family:'Times New Roman';"&gt;&amp;nbsp;&amp;nbsp;&amp;nbsp;&lt;/span&gt;&lt;/span&gt;&lt;span lang="EN-US" style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;TTo determine the role of type I IFN signalling on epithelial-mediated Th2 immune responses in a model of RV- and IL-4-stimulated bronchial epithelial cell culture. &lt;/span&gt;&lt;/p&gt;&lt;p style="margin-bottom:0cm;margin-bottom:.0001pt;text-indent:-18.0pt;"&gt;&lt;span lang="EN-US" style="font-family:'Times New Roman',serif;"&gt;&lt;span style="font-size:small;font-family:Arial, Helvetica, sans-serif;"&gt;-&lt;/span&gt;&lt;span style="font-variant-numeric:normal;font-size:7pt;line-height:normal;font-family:'Times New Roman';"&gt;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp;&amp;nbsp; &lt;/span&gt;&lt;/span&gt;&lt;span lang="EN-US" style="font-family:Arial, Helvetica, sans-serif;font-size:small;"&gt;To investigate the mechanisms of type I IFN-mediated regulation Th2 cells in a Th2 polarised CD4+ T cell model.&lt;/span&gt;&lt;/p&gt;
Medical Science, Imperial College London Principal Supervisor
2014 PhD Effect of inhaled corticosteroids on viral and bacterial infection in chronic obstructive pulmonary disease
Rhinovirus (RV) infections trigger exacerbations of chronic obstructive pulmonary disease (COPD) exacerbations and may precipitate secondary bacterial infections. Inhaled corticosteroids (ICS) are used commonly in COPD but are relatively ineffective in the context of virus-induced exacerbations and may also increase the risk of pneumonia. We hypothesised that, in a mouse model, ICS would suppress anti-viral and anti-bacterial immune responses leading to alteration of the airway microbiota and secondary bacterial infection following RV-induced exacerbation of COPD.<br />Despite extensive optimisation, we were unable to define a representative mouse model of the deficient anti-viral and anti-bacterial responses that are indicative of human COPD. For this reason, and because of difficulties in measuring the airway microbiota in mice, we employed models of primary RV1B and Streptococcus pneumoniae infection as surrogates for viral exacerbation and bacterial colonisation in COPD. Fluticasone propionate (FP) administration prior to RV1B infection suppressed innate and adaptive immune responses leading to impaired virus control, in a dose dependent manner. This effect was causally related to suppression of type I interferon (IFN) as administration of recombinant IFN-&beta; reconstituted IFN-stimulated gene expression and restored virus control. FP suppressed RV-induced airway inflammation but led to enhanced airway mucin production, effects that were unaltered by recombinant IFN-&beta;. FP administration also suppressed innate responses to S. pneumoniae including expression of anti-bacterial cytokines and cathelicidin-related anti-microbial peptide. High dose FP increased lung tissue bacterial loads with the opposite effect observed with lower dose FP despite similar anti-inflammatory effects.<br />Our findings demonstrate beneficial anti-inflammatory effects of ICS during virus-induced COPD exacerbations but reveal some previously unrecognised detrimental effects including increased virus replication and enhanced mucin production. Additionally, we show that high dose ICS administration may increase bacterial loads and thus increase pneumonia risk but lower doses may conversely reduce bacterial loads and therefore could be safer in COPD.
Medical Science, Imperial College London Co-Supervisor
2012 PhD The role of IL-25 in rhinovirus-induced asthma exacerbations
Background and hypothesis: Rhinovirus (RV) infections are the principal cause of asthma exacerbations. While Th2-mediated inflammation is clearly implicated in the asthmatic response, it is unknown how the immune response to RV infection interacts with Th2 immunity to enhance disease pathogenesis. The epithelial-derived cytokine, IL-25, has been identified as an initiator and regulator of Th2 immunity and plays a role in asthma pathogenesis. Based on the fact that bronchial epithelial cells are the primary site of RV infection, we hypothesized that RV induces IL-25 production providing a link between infection and Th2 driven allergic inflammation. Aims and methods: RV-induced IL-25 expression was measured in human bronchial epithelial cells (HBECs) obtained from bronchoscopic brushings from atopic asthmatics and healthy patients. Mouse models of RV infection and RV-induced allergic airways disease were also employed to examine IL-25 induction in response to RV infection and/or OVA sensitisation and challenge. Finally, to define a mechanistic role for RV-induced IL-25, signalling mediated by IL-25 was blocked in our model of RV-induced allergic airways disease by neutralising the IL-25 receptor. Results: RV-infected HBECs from asthmatics expressed significantly greater IL-25 gene and protein compared with cells from healthy controls. Furthermore, RV infection of mice induced IL-25 expression in the airway epithelium as well as in inflammatory cells in the airway lamina propia. Using a mouse model of RV-induced allergic disease, we demonstrated that RV enhanced allergen-driven IL-25 gene and protein expression which was associated with increased Th2 inflammation in the lung. Finally, by blocking IL-25 signalling in an RV-infected and OVA-sensitised and challenged mouse, several key features of the exacerbation phenotype were significantly reduced including airway leukocyte infiltration, BAL Th2 cytokines and chemokines and Th2 cells. Conclusions: These novel findings indicate that RV-induced IL-25 plays an important role in enhancing Th2 inflammation associated with the exacerbation phenotype which is mediated by binding to the IL-25 receptor.
Medical Science, Imperial College London Principal Supervisor
2011 PhD The role of IL-15 in response to rhinovirus infections
Rhinoviruses (RV) cause the common cold and are major precipitants of asthma exacerbations. The underlying mechanisms of RV-induced airways disease are unclear. IL-15 is a proinflammatory cytokine produced during viral infections and plays a key role in the regulation of NK cells. Using mouse models of RV infection and RV-induced asthma exacerbation we examined the role of IL-15 and its importance for NK cell responses during RV infections in allergic and non-allergic airways. We demonstrate RV-induced IL-15 upregulation in the airway and lungs of BALB/c mice at day 1 after infection and accumulation of NK cells in the airway and lungs at days 1-2 and 2-4 respectively. The NK cells exhibited an activated phenotype characterised by upregulated CD69, IFN-&gamma; and GranzymeB expression. Blocking IL-15 upon intranasal administration of an IL-15 neutralising antibody inhibited the NK cell response to RV infection, which was associated with deficient IFN-&gamma; production and increased expression of Th2 mediators. IL-15R&alpha; knockout mice lack NK cells and also demonstrated deficient IFN-&gamma; and increased Th2 responses to RV infection; these mice also exhibited deficient CD8+ T cell responses and an increased viral load. Similar results were observed in RV infected IFNAR1 ko mice, which was associated with deficient IL-15 upregulation. We suggest that RV-induced IL-15 is mediated by type I interferon signalling, and is necessary for NK cell responses and early IFN-&gamma; production during RV-1B infection, which drives development of appropriate Th1 antiviral responses. In the absence of this pathway, Th2 responses result and are associated with impaired antiviral immunity. To examine the interaction between allergen driven Th2 immunity and RV infection, we employed a RV-induced asthma exacerbation model. Unexpectedly, RV infected allergen challenged mice, despite having increased viral load, demonstrated increased IL-15 expression and NK cell responses, revealing a novel interaction between allergic responses and antiviral immunity.
Microbiology, Imperial College London Co-Supervisor
2010 PhD T cell responses in models of rhinovirus-induced airways disease
<p style="text-align:justify;line-height:150%;"><span lang="EN-GB" style="font-family:'Arial',sans-serif;">Human Rhinoviruses (HRV) cause the common cold and are associated wtih more severe respiratory diseases such as asthma exacerbations. There is good evidence that T lymphocytes (T cells) play a central role in the pathogenesis of atopic asthma however little is known about T cell responses to HRV infection or how HRV infection modulates T cell responses in asthma. One T cell subset expresses the &gamma;&delta; TCR and are capable of modulating inflammation &amp; AHR in mouse models of infection and asthma. Furthermore we have observed increased &gamma;&delta; T cells in the airways of human atopic asthmatics following experimental HRV infection which was associated with worse symptoms, enhanced airway inflammation and diminished lung function. Despite the association of &gamma;&delta; T cells with disease in asthma the functional role of these cells is unknown (Message et al., unpublished data). The aims of this thesis were to use recently developed mouse models and characterise the T cell responses to HRV infection in healthy and in ovalbumin (OVA) sensitised &amp; challenged (asthmatic) mice, and to define a role for &gamma;&delta; T cells in RV infection and RV-induced asthma exacerbations... </span></p><p style="line-height:150%;"><span lang="EN-GB">Consistent with observations in human studies, mouse HRV infection was characterised by neutrophilic and lymphocytic airways inflammation. The airway and lung-tissue lymphocyte response to HRV infection was composed of activated CD4+, CD8+ and CD4<sup>-</sup>/CD8<sup>-</sup> &gamma;&delta; TCR+ T cells<span style="color:#999999;"> </span>and was weakly Th1 orientated.<span style="color:#999999;"> </span>In HRV induced asthma exacerbation, HRV infection modestly enhanced numbers of multiple T cell populations in the airway and lung tissue as well as levels of Th2 cytokines such as IL-4. Systemic depletion of &gamma;&delta; T cells with a monoclonal anti-TCR&gamma;&delta; antibody in the HRV infection model modestly enhanced some aspects of airways inflammation such as neutrophil recruitment but had no effect on AHR. In both the asthma and the HRV induced asthma exacerbation models, &gamma;&delta; T cell depletion enhanced AHR at 48hrs post allergen challenge/ HRV infection. In the allergic asthma model &gamma;&delta; T cell depletion also enhanced some aspects of airways inflammation such as&hellip; </span></p><p style="text-align:justify;line-height:150%;"><span lang="EN-GB" style="font-family:'Arial',sans-serif;">This thesis has demonstrated that HRV infection is capable of increasing multiple T cell populations and associated cytokine production in the lung, Antibody-mediated depletion studies specifically defined a function in disease for &gamma;&delta; T cells indicating that they perform a regulatory function, limiting both virus and allergen induced airway inflammation and, in asthma and asthma exacerbation models, also limiting AHR.</span></p>
Medical Studies, Imperial College London Co-Supervisor
Edit

News

Citizen of Year recipients WIDGET

News • 23 Jan 2024

Citizen of the Year honoured for pioneering COVID-19 treatment

Two leading University of Newcastle health researchers, Professor Nathan Bartlett and Emeritus Professor Julie Byles have each been honoured in the 2024 City of Newcastle Citizen of the Year Awards.

Associate Professor Nathan Bartlett

News • 7 Oct 2021

COVID-19 nasal spray moves to Phase 2 study

Encouraging preliminary results from an ongoing phase 1 study have triggered the announcement of phase 2 studies for a first-in-class nasal spray to protect people from respiratory viral diseases such as COVID-19 and influenza.

News • 2 Feb 2021

Research shows nasal spray that protects against COVID-19 is also effective against the common cold

Research into a new drug which primes the immune system in the respiratory tract and is in development for COVID-19 shows it is also effective against rhinovirus.

Nathan Barlett and Hubert Hondermarck in a labratory looking at the camera

News • 16 Apr 2020

Drug repurposing potential for COVID-19

The COVID-19 pandemic has highlighted the need for anti-viral therapies to treat respiratory virus infections. With a vaccine probably at least 12 months away, drug repurposing (using clinically approved drugs which also have anti-viral activity) offers hope in fast-tracking therapies to possibly treat infected people and save lives.

News • 16 Jul 2019

The perils of the common cold

Nathan Bartlett

News • 14 Jun 2018

Research highlights need for a new approach to COPD management

Researchers at the University of Newcastle and Imperial College London have provided evidence that a conventional treatment for common respiratory diseases such as chronic obstructive pulmonary disease (COPD) can actually lead to worse health outcomes when used to treat symptoms caused by respiratory virus infections.

Professor Nathan Bartlett

Position

Assistant Dean- Industry Engagement
Bartlett Lab
School of Biomedical Sciences and Pharmacy
College of Health, Medicine and Wellbeing

Contact Details

Email nathan.bartlett@newcastle.edu.au
Phone (02) 4042 0171

Office

Room HMRI2405
Building HMRI
Location New Lambton Heights

,
Edit