1
|
Gopalakrishnan V, Sparklin B, Kim JH, Bouquet J, Kehl M, Kenny T, Morehouse C, Caceres C, Warrener P, Hristova VA, Wilson S, Shandilya H, Barnes A, Ruzin A, Wang J, Oberg L, Angermann B, McCrae C, Platt A, Muthas D, Hess S, Tkaczyk C, Sellman BR, Ostridge K, Belvisi MG, Wilkinson TMA, Staples KJ, DiGiandomenico A. NTHi killing activity is reduced in COPD patients and is associated with a differential microbiome. Respir Res 2025; 26:45. [PMID: 39885466 PMCID: PMC11781068 DOI: 10.1186/s12931-025-03113-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/11/2025] [Indexed: 02/01/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic lung disease characterized by airway obstruction and inflammation. Non-typeable Haemophilus influenzae (NTHi) lung infections are common in COPD, promoting frequent exacerbations and accelerated lung function decline. The relationship with immune responses and NTHi are poorly understood. Herein, we comprehensively characterized the respiratory microbiome and mycobiome of patients while investigating microbial dynamics and host immune changes attributable to NTHi killing activity. Mild-to-moderate COPD patients encompassing frequent and infrequent exacerbators and healthy volunteers (HV) were enrolled. Microbial composition, proteomics and NTHi killing activity was analyzed using bronchoalveolar lavage fluid (BALF). In addition, antigen-antibody titers in sera to COPD pathogens were determined using a multiplex assay. Differential abundance analysis revealed an enrichment of Actinobacteria and Bacteroidetes in the BALF of COPD and HV subjects respectively. Significant differences in the IgA titer response were observed against NTHi antigens in COPD vs. HV. Notably, there was also significantly greater killing activity against NTHi in BALF from COPD vs. HV subjects (OR = 5.64; 95% CI = 1.75-20.20; p = 0.001). Stratification of COPD patients by NTHi killing activity identified unique microbial and protein signatures wherein Firmicutes, Actinobacteria and haptoglobin were enriched in patients with killing activity. We report that differences in host immune responses and NTHi-killing activity are associated with microbiome changes in mild-to-moderate COPD. This is suggestive of a potential link between the respiratory microbiome and immune activity against NTHi in the context of COPD pathogenesis even at this disease stage.
Collapse
Affiliation(s)
- Vancheswaran Gopalakrishnan
- Bioinformatics, Research and Early Development, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ben Sparklin
- Bioinformatics, Research and Early Development, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Jung Hwan Kim
- Bacterial Vaccines, Research and Early Development, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Jerome Bouquet
- Bioinformatics, Research and Early Development, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Margaret Kehl
- Bacterial Vaccines, Research and Early Development, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Tara Kenny
- Virology and Vaccine Discovery, Research and Early Development, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Christopher Morehouse
- Bioinformatics, Research and Early Development, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Carolina Caceres
- Translational Scientific Management, Research and Early Development, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Paul Warrener
- Bacterial Vaccines, Research and Early Development, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ventzislava A Hristova
- Dynamic Omics, Centre for Genomics Research (CGR), Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Susan Wilson
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Harini Shandilya
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Arnita Barnes
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Alexey Ruzin
- Translational Scientific Management, Research and Early Development, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Junmin Wang
- Quantitative Biology, Data Sciences and Quantitative Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Lisa Oberg
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Bastian Angermann
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Christopher McCrae
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Adam Platt
- VP and Head of Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Daniel Muthas
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Sonja Hess
- Dynamic Omics, Centre for Genomics Research (CGR), Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Christine Tkaczyk
- Microbial Antibodies and Technologies, Research and Early Development, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Bret R Sellman
- Bacterial Vaccines, Research and Early Development, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Kristoffer Ostridge
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Maria G Belvisi
- SVP and Head of Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Tom M A Wilkinson
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Karl J Staples
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Antonio DiGiandomenico
- Microbial Antibodies and Technologies, Research and Early Development, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA.
| |
Collapse
|
2
|
Pourrezaei S, Letafati A, Molaverdi G, Norouzi M, Mozhgani SH. RAB3GAP2 dysregulation in adult T-cell leukemia/lymphoma (ATLL) compared to acute lymphoblastic leukemia (ALL): a molecular perspective. BMC Res Notes 2025; 18:28. [PMID: 39838474 PMCID: PMC11752935 DOI: 10.1186/s13104-025-07084-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/03/2025] [Indexed: 01/23/2025] Open
Abstract
Adult T-cell leukemia/lymphoma (ATLL) is a type of blood cancer related to human T-cell lymphotropic virus type 1 (HTLV-1). The principal aim of this study was to investigate cellular processes related to innate immune response, intracellular protein transport, and translational initiation regulation in individuals afflicted with ATLL and Acute lymphoblastic leukemia (ALL). Whole blood samples and peripheral blood mononuclear cells were collected from 10 viral ATLL patients and 10 ALL subjects. Real-time quantitative PCR was then performed to quantify mRNA expression levels of SMC6, FANCM, EIF4H, WDR7, RAB3GAP2, and IFN α/β. The study revealed some distinctions between ATLL and ALL patients. Particularly, RAB3GAP2 level (P = 0.028) was found to be elevated in ATLL patients compared to ALL. Conversely, expression levels of IFN-β (P = 0.31), SMC6 (P = 0.68), WDR7 (P = 0.43), EIF4H (P = 0.38), and FANCM (P = 0.57) were diminished in ATLL patients in contrast to ALL. These proteins play a pivotal role in both translation and immune activation, suggesting a potential correlation between the observed disparities and the virus-mediated progression of cancer. However, it is worth noting that the expression differences in FANCM, EIF4H, SMC6, and WDR7 between ATLL and ALL were minimal. This proposes that the underlying molecular mechanisms governing ATLL and ALL may largely overlap concerning these cellular processes. However, considerable increased expression of RAB3GAP2 was observed in ATLL compared to ALL.
Collapse
Affiliation(s)
- Samira Pourrezaei
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Letafati
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghazale Molaverdi
- Student Research Committee, Alborz University of Medical Sciences, Alborz, Iran
| | - Mehdi Norouzi
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| | - Sayed-Hamidreza Mozhgani
- Department of Microbiology and Virology, School of Medicine, Alborz University of Medical Sciences, Alborz, Iran.
- Non-communicable Disease Research Center, Alborz University of Medical Sciences, Alborz, Iran.
| |
Collapse
|
3
|
Yalcin Mutlu M, Kleyer A, Kroenke G, Fagni F, Temiz SA, Meder C, Dietrich P, Orlemann T, Mößner J, Schoenau V, Bohr D, Schuster L, Hartmann F, Minopoulou I, Leppkes M, Ramming A, Pachowsky ML, Schuch F, Ronneberger M, Kleinert S, Hueber AJ, Manger K, Manger B, Atreya R, Berking C, Sticherling M, Neurath MF, Schett G, Simon D, Tascilar K. Paucity of viral infection symptoms in patients with immune-mediated inflammatory diseases. BMJ Open 2025; 15:e088486. [PMID: 39773803 PMCID: PMC11749532 DOI: 10.1136/bmjopen-2024-088486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/04/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVES Although patients with immune-mediated inflammatory diseases (IMID) are thought to be more susceptible to viral infections, it is unclear whether their presentation differs between patients with IMID and healthy controls. This study aimed to investigate the symptom pattern of common viral infections in patients with IMID and compare it with controls without IMIDs. DESIGN A cross-sectional study conducted between 1 February and 30 April 2020, using a questionnaire. SETTING Seven tertiary regional care centers in Germany, which specialised in the care of patients with IMID (namely, in gastroenterology, dermatology, rheumatology and immunology clinical care). PARTICIPANTS One thousand nine hundred nine participants completed the survey (757 patients with IMID; 1152 non-IMID controls). PRIMARY OUTCOME MEASURE The occurrence of 11 common viral illness symptoms within the preceding 3 months in patients with IMID and non-IMID controls. RESULTS Symptom data were clustered, based on number and co-occurrance, into 3 major clusters and 2 subclusters ranked by the average number of symptoms. Patients with inflammatory bowel disease and psoriasis were significantly overrepresented in the lower-frequency subcluster of the polysymptomatic cluster. Patients with rheumatoid arthritis were overrepresented in the lower-frequency subclusters of the intermediate and oligo-/asymptomatic clusters. Controls were over-represented only in the higher-frequency subclusters of each major cluster where none of the IMIDs were over-represented. Spondyloarthritis and other IMIDs were also overrepresented in the low-frequency subcluster, but the results were not significant. Overall, patients with rheumatoid arthritis patients reported fewer symptoms (rate ratio=0.68, 95% CI, 0.59 to 0.80) than non-IMID controls. CONCLUSION Patients with IMID are over-represented in low-frequency subclusters, even among individuals who have reported a broad range of viral infection symptoms. This pattern suggests that the manifestations of viral infections are different between patients with IMID and controls, thus challenging the accurate and early diagnosis of infections.
Collapse
Affiliation(s)
- Melek Yalcin Mutlu
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Arnd Kleyer
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Department of Rheumatology and Clinical Immunology, Charite - Universitatsmedizin Berlin, Berlin, Berlin, Germany
| | - Gerhard Kroenke
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Department of Rheumatology and Clinical Immunology, Charite - Universitatsmedizin Berlin, Berlin, Berlin, Germany
| | - Filippo Fagni
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Selahattin Alp Temiz
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Christine Meder
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen Department of Dermatology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Peter Dietrich
- Universitätsklinikum Erlangen Department of Medicine 1, Kussmaul Research Campus & Ludwig Demling Endoscopy Center of Excellence, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Till Orlemann
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen Department of Medicine 1, Kussmaul Research Campus & Ludwig Demling Endoscopy Center of Excellence, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Johanna Mößner
- Universitätsklinikum Erlangen Department of Medicine-2, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Verena Schoenau
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Daniela Bohr
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Louis Schuster
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Fabian Hartmann
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Ioanna Minopoulou
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Department of Rheumatology and Clinical Immunology, Charite - Universitatsmedizin Berlin, Berlin, Berlin, Germany
| | - Moritz Leppkes
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen Department of Medicine 1, Kussmaul Research Campus & Ludwig Demling Endoscopy Center of Excellence, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Andreas Ramming
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Milena L Pachowsky
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | | | | | | | - Axel J Hueber
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Division of Rheumatology, Klinikum Nürnberg, Paracelsus Private Medical University - Nuremberg Campus, Nurnberg, Bayern, Germany
- Rheumazentrum Erlangen-Nürnberg, UK
| | - Karin Manger
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Rheumatology Practice Bamberg, Bamberg, Bayern, Germany
| | - Bernhard Manger
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Raja Atreya
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen Department of Medicine 1, Kussmaul Research Campus & Ludwig Demling Endoscopy Center of Excellence, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Carola Berking
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen Department of Dermatology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Michael Sticherling
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen Department of Dermatology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Markus F Neurath
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen Department of Medicine 1, Kussmaul Research Campus & Ludwig Demling Endoscopy Center of Excellence, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - Georg Schett
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| | - David Simon
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Department of Rheumatology and Clinical Immunology, Charite - Universitatsmedizin Berlin, Berlin, Berlin, Germany
| | - Koray Tascilar
- Universitätsklinikum Erlangen Department of Medicine-3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Universitätsklinikum Erlangen, Deutsches Zentrum fuer Immunotherapie, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
| |
Collapse
|
4
|
de Castro MV, Cariste LM, Almeida RR, Sasahara GL, Silva MVR, Soares FB, Coria VR, Naslavsky MS, Santos KS, Cunha-Neto E, Kalil J, Zatz M. Potential protective role of interferon-induced protein with tetratricopeptide repeats 3 (IFIT3) in COVID-19. Front Cell Infect Microbiol 2024; 14:1464581. [PMID: 39664492 PMCID: PMC11631949 DOI: 10.3389/fcimb.2024.1464581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 11/07/2024] [Indexed: 12/13/2024] Open
Abstract
The COVID-19 pandemic has prompted a quest to understand why certain individuals remain uninfected or asymptomatic despite repetitive exposure to SARS-CoV-2. Here, we focused on six exposed females residing with their symptomatic and reinfected SARS-CoV-2 PCR-positive COVID-19 partners. Peripheral blood mononuclear cell samples from couples were analysed for poly (I:C)-induced mRNA expression of type I/III interferons and interferon-stimulated genes (ISGs). Remarkably, we found a significant upregulation of the ISG interferon-inducible protein with tetrapeptide repeats 3 (IFIT3) gene exclusively in exposed uninfected or asymptomatic females, suggesting a potential role in protective immunity against symptomatic COVID-19.
Collapse
Affiliation(s)
- Mateus V. de Castro
- Human Genome and Stem Cell Research Center, University of São Paulo, São Paulo, SP, Brazil
| | - Leonardo M. Cariste
- Laboratory of Immunology, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, (HCFMUSP), São Paulo, Brazil
- Instutute on Investigation in Immunology, - Instituto Nacional de Ciências e Tecnologia-iii-INCT, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Rafael R. Almeida
- Laboratory of Immunology, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, (HCFMUSP), São Paulo, Brazil
- Instutute on Investigation in Immunology, - Instituto Nacional de Ciências e Tecnologia-iii-INCT, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Greyce L. Sasahara
- Laboratory of Immunology, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, (HCFMUSP), São Paulo, Brazil
- Instutute on Investigation in Immunology, - Instituto Nacional de Ciências e Tecnologia-iii-INCT, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Monize V. R. Silva
- Human Genome and Stem Cell Research Center, University of São Paulo, São Paulo, SP, Brazil
| | - Flávia B. Soares
- Human Genome and Stem Cell Research Center, University of São Paulo, São Paulo, SP, Brazil
| | - Vivian R. Coria
- Human Genome and Stem Cell Research Center, University of São Paulo, São Paulo, SP, Brazil
| | - Michel S. Naslavsky
- Human Genome and Stem Cell Research Center, University of São Paulo, São Paulo, SP, Brazil
| | - Keity S. Santos
- Laboratory of Immunology, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, (HCFMUSP), São Paulo, Brazil
- Instutute on Investigation in Immunology, - Instituto Nacional de Ciências e Tecnologia-iii-INCT, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, (HCFMUSP), São Paulo, Brazil
- Instutute on Investigation in Immunology, - Instituto Nacional de Ciências e Tecnologia-iii-INCT, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, (HCFMUSP), São Paulo, Brazil
- Instutute on Investigation in Immunology, - Instituto Nacional de Ciências e Tecnologia-iii-INCT, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Mayana Zatz
- Human Genome and Stem Cell Research Center, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
5
|
Xie C, Yang J, Gul A, Li Y, Zhang R, Yalikun M, Lv X, Lin Y, Luo Q, Gao H. Immunologic aspects of asthma: from molecular mechanisms to disease pathophysiology and clinical translation. Front Immunol 2024; 15:1478624. [PMID: 39439788 PMCID: PMC11494396 DOI: 10.3389/fimmu.2024.1478624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
In the present review, we focused on recent translational and clinical discoveries in asthma immunology, facilitating phenotyping and stratified or personalized interventions for patients with this condition. The immune processes behind chronic inflammation in asthma exhibit marked heterogeneity, with diverse phenotypes defining discernible features and endotypes illuminating the underlying molecular mechanisms. In particular, two primary endotypes of asthma have been identified: "type 2-high," characterized by increased eosinophil levels in the airways and sputum of patients, and "type 2-low," distinguished by increased neutrophils or a pauci-granulocytic profile. Our review encompasses significant advances in both innate and adaptive immunities, with emphasis on the key cellular and molecular mediators, and delves into innovative biological and targeted therapies for all the asthma endotypes. Recognizing that the immunopathology of asthma is dynamic and continuous, exhibiting spatial and temporal variabilities, is the central theme of this review. This complexity is underscored through the innumerable interactions involved, rather than being driven by a single predominant factor. Integrated efforts to improve our understanding of the pathophysiological characteristics of asthma indicate a trend toward an approach based on disease biology, encompassing the combined examination of the clinical, cellular, and molecular dimensions of the disease to more accurately correlate clinical traits with specific disease mechanisms.
Collapse
Affiliation(s)
- Cong Xie
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Jingyan Yang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Aman Gul
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
- Department of Respiratory Medicine, Uyghur Medicines Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China
- College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Yifan Li
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Rui Zhang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Maimaititusun Yalikun
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Xiaotong Lv
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhan Lin
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qingli Luo
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Huijuan Gao
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
6
|
Ennadif B, Alaoui-Inboui FZ, Benmoussa AY, El Kettani A, Elmdaghri N, Slaoui B. Virological Profile of Asthma Exacerbation in Children: A Hospital-Based Retrospective Study. Cureus 2024; 16:e60261. [PMID: 38872674 PMCID: PMC11170309 DOI: 10.7759/cureus.60261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2024] [Indexed: 06/15/2024] Open
Abstract
Introduction Viruses are the most common triggering factors for asthma exacerbation during the autumn and winter seasons. Viruses, such as influenza A and rhinovirus, play a major role in the occurrence of severe exacerbation of asthma. This association between viral infection and asthma exacerbation in children is a result of the antiviral response of the immune system and various anti-inflammatory phenomena. In this work, we aimed to identify the virological profile of asthma exacerbation in children and analyze the correlation between viral infection type and the severity of exacerbation. Materials and methods This retrospective study was conducted from January 2016 to January 2024. The study included children hospitalized for asthma exacerbation associated with signs of viral-like respiratory infection with positive virological testing by multiplex real-time polymerase chain reaction or rapid test in the case of influenza A or respiratory syncytial virus (RSV). Data analysis was performed with Microsoft Excel and SPSS software using a previously established data collection sheet Results Thirty cases were collected for the study period. The mean age of the patients was 4 years and 8 months, with a male-to-female ratio of 3.3. Eighteen patients were known to have asthma, of which nine had uncontrolled asthma, and exacerbation was inaugural in 12 patients. Viral shedding was found in 14 patients. A viral agent was found in all patients, with coinfection of two or more viruses in three patients. The viruses found were influenza A (18 cases), coupled rhinovirus/enterovirus (eight cases), RSV (eight cases), human metapneumovirus (three patients), and parainfluenza type IV in only one inaugural patient. Asthma exacerbation was severe in 20 patients, moderate in eight patients, and two patients had severe acute asthma requiring intensive care management. We noted a higher frequency of severe exacerbation among those with an influenza A viral infection. All patients with RSV infection exhibited moderate exacerbation. No other significant correlation between asthma severity and other types of viruses was found. Conclusions Our results demonstrate the major role played by viruses in triggering asthma exacerbation, primarily influenza virus, followed by enterovirus, rhinovirus, RSV, and metapneumovirus. Larger-scale studies should be carried out to establish a more complete virological profile and further investigate the viral factor in the management of asthma in children.
Collapse
Affiliation(s)
- Basma Ennadif
- Department of Pediatrics, Faculté de Médecine et de Pharmacie, Université Hassan II, Casablanca, MAR
- Pediatric Pneumo-Allergology Unit, Pediatric Department 2, Hôpital Mère-Enfants Abderrahim Harouchi, Centre Hospitalier Universitaire Ibn Rochd, Casablanca, MAR
| | - Fatima Zahra Alaoui-Inboui
- Department of Pediatrics, Faculté de Médecine et de Pharmacie, Université Hassan II, Casablanca, MAR
- Pediatric Pneumo-Allergology Unit, Pediatric Department 2, Hôpital Mère-Enfants Abderrahim Harouchi, Centre Hospitalier Universitaire Ibn Rochd, Casablanca, MAR
| | - AbdelHakim Youssef Benmoussa
- Department of Pediatrics, Faculté de Médecine et de Pharmacie, Université Hassan II, Casablanca, MAR
- Pediatric Pneumo-Allergology Unit, Pediatric Department 2, Hôpital Mère-Enfants Abderrahim Harouchi, Centre Hospitalier Universitaire Ibn Rochd, Casablanca, MAR
| | - Assiya El Kettani
- Department of Microbiology, Faculté de Médecine et de Pharmacie, Université Hassan II, Casablanca, MAR
- Department of Microbiology, Centre Hospitalier Universitaire Ibn Rochd, Casablanca, MAR
| | - Naima Elmdaghri
- Department of Microbiology, Faculté de Médecine et de Pharmacie, Université Hassan II, Casablanca, MAR
- Department of Microbiology, Centre Hospitalier Universitaire Ibn Rochd, Casablanca, MAR
| | - Bouchra Slaoui
- Department of Pediatrics, Faculté de Médecine et de Pharmacie, Université Hassan II, Casablanca, MAR
- Pediatric Pneumo-Allergology Unit, Pediatric Department 2, Hôpital Mère-Enfants Abderrahim Harouchi, Centre Hospitalier Universitaire Ibn Rochd, Casablanca, MAR
| |
Collapse
|
7
|
Gil CH, Oh C, Lee J, Jang M, Han J, Cho SD, Park SH, Park JH, Kim HJ. Inhalation Delivery of Interferon-λ-Loaded Pulmonary Surfactant Nanoparticles Induces Rapid Antiviral Immune Responses in the Lung. ACS APPLIED MATERIALS & INTERFACES 2024; 16:11147-11158. [PMID: 38407048 DOI: 10.1021/acsami.3c13677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The interferon-λ (IFN-λ)-regulated innate immune responses in the airway expand our understanding toward antiviral strategies against influenza A virus (IAV). The application of IFN-λ as mucosal antiviral therapeutic is still challenging, and advanced research will be necessary to achieve more efficient delivery of recombinant IFN-λs to the damaged respiratory mucosa. In this study, we examine the capability of IFN-λ to stimulate the innate immune response, promoting the swift elimination of IAV in the lungs. Additionally, we develop IFN-λ-loaded nanoparticles incorporated into pulmonary surfactant for inhalation therapy aimed at treating lung infections caused by IAV. We found that inhaled delivery of IFNλ-PSNPs significantly restricted IAV replication in the lungs from 3 days after infection (dpi), and IAV-caused lung histopathologic findings were completely improved in response to IFNλ-PSNPs. More significant and rapid attenuation of viral RNA was observed in the lung of mice with inhaled delivery of IFNλ-PSNPs compared to mice with recombinant IFN-λs. Inhalation treatment of IFNλ-PSNPs to IAV-infected mice can result in the increase of monocyte frequency in concert with restoration of T and B cells composition. Furthermore, the transcriptional profiles of monocytes shifted toward heightened IFN responses following IFNλ-PSNP treatment. These results imply that IFN-λ could serve as a robust inducer of innate immunity in the lungs against IAV infection, and inhalation of IFN-λs encapsulated in PSNPs effectively resolves lung infections caused by IAV through rapid viral clearance. PSNPs facilitated improved delivery of IFN-λs to the lungs, triggering potent antiviral immune responses upon IAV infection onset.
Collapse
Affiliation(s)
- Chan Hee Gil
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Chanhee Oh
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Jeongsoo Lee
- Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Mincheol Jang
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Junhee Han
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Sung-Dong Cho
- Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Su-Hyung Park
- Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
- The Center for Epidemic Preparedness, KAIST Institute, Daejeon 34141, Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Hyun Jik Kim
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul 03080, Korea
- Seoul National University Hospital, Seoul 03080, Korea
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul 03080, Korea
| |
Collapse
|
8
|
Mihaescu G, Chifiriuc MC, Filip R, Bleotu C, Ditu LM, Constantin M, Cristian RE, Grigore R, Bertesteanu SV, Bertesteanu G, Vrancianu CO. Role of interferons in the antiviral battle: from virus-host crosstalk to prophylactic and therapeutic potential in SARS-CoV-2 infection. Front Immunol 2024; 14:1273604. [PMID: 38288121 PMCID: PMC10822962 DOI: 10.3389/fimmu.2023.1273604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 12/29/2023] [Indexed: 01/31/2024] Open
Abstract
Mammalians sense antigenic messages from infectious agents that penetrate the respiratory and digestive epithelium, as well as signals from damaged host cells through membrane and cytosolic receptors. The transduction of these signals triggers a personalized response, depending on the nature of the stimulus and the host's genetics, physiological condition, and comorbidities. Interferons (IFNs) are the primary effectors of the innate immune response, and their synthesis is activated in most cells within a few hours after pathogen invasion. IFNs are primarily synthesized in infected cells, but their anti-infective effect is extended to the neighboring cells by autocrine and paracrine action. The emergence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic in 2019 was a stark reminder of the potential threat posed by newly emerging viruses. This pandemic has also triggered an overwhelming influx of research studies aiming to unveil the mechanisms of protective versus pathogenic host immune responses induced by SARS-CoV-2. The purpose of this review is to describe the role of IFNs as vital players in the battle against SARS-CoV-2 infection. We will briefly characterize and classify IFNs, present the inductors of IFN synthesis, their sensors, and signaling pathways, and then discuss the role of IFNs in controlling the evolution of SARS-CoV-2 infection and its clinical outcome. Finally, we will present the perspectives and controversies regarding the prophylactic and therapeutic potential of IFNs in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Grigore Mihaescu
- Microbiology Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Mariana Carmen Chifiriuc
- Microbiology Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Department of Life, Medical and Agricultural Sciences, Biological Sciences Section, Academy of Romanian Scientists, Bucharest, Romania
| | - Roxana Filip
- Faculty of Medicine and Biological Sciences, Stefan cel Mare University of Suceava, Suceava, Romania
- Microbiology Department, Suceava Emergency County Hospital, Suceava, Romania
| | - Coralia Bleotu
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Cellular and Molecular Pathology Department, Stefan S. Nicolau Institute of Virology, Bucharest, Romania
| | - Lia Mara Ditu
- Microbiology Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Marian Constantin
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Institute of Biology of Romanian Academy, Bucharest, Romania
| | - Roxana-Elena Cristian
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Raluca Grigore
- ENT Department, University of Medicine and Pharmacy Carol Davila and Coltea Clinical Hospital, Bucharest, Romania
| | - Serban Vifor Bertesteanu
- ENT Department, University of Medicine and Pharmacy Carol Davila and Coltea Clinical Hospital, Bucharest, Romania
| | - Gloria Bertesteanu
- ENT Department, University of Medicine and Pharmacy Carol Davila and Coltea Clinical Hospital, Bucharest, Romania
| | - Corneliu Ovidiu Vrancianu
- Microbiology Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- DANUBIUS Department, National Institute of Research and Development for Biological Sciences, Bucharest, Romania
| |
Collapse
|
9
|
Wang C, Du Z, Li R, Luo Y, Zhu C, Ding N, Lei A. Interferons as negative regulators of ILC2s in allergic lung inflammation and respiratory viral infections. J Mol Med (Berl) 2023; 101:947-959. [PMID: 37414870 DOI: 10.1007/s00109-023-02345-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/08/2023]
Abstract
Group 2 innate lymphoid cells (ILC2s), characterized by a lack of antigen receptors, have been regarded as an important component of type 2 pulmonary immunity. Analogous to Th2 cells, ILC2s are capable of releasing type 2 cytokines and amphiregulin, thus playing an essential role in a variety of diseases, such as allergic diseases and virus-induced respiratory diseases. Interferons (IFNs), an important family of cytokines with potent antiviral effects, can be triggered by microbial products, microbial exposure, and pathogen infections. Interestingly, the past few years have witnessed encouraging progress in revealing the important role of IFNs and IFN-producing cells in modulating ILC2 responses in allergic lung inflammation and respiratory viral infections. This review underscores recent progress in understanding the role of IFNs and IFN-producing cells in shaping ILC2 responses and discusses disease phenotypes, mechanisms, and therapeutic targets in the context of allergic lung inflammation and infections with viruses, including influenza virus, rhinovirus (RV), respiratory syncytial virus (RSV), and severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2).
Collapse
Affiliation(s)
- Cui Wang
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Zhaoxiang Du
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Ranhui Li
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Ying Luo
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Cuiming Zhu
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Nan Ding
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Aihua Lei
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China.
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China.
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China.
| |
Collapse
|
10
|
Urbani F, Cometa M, Martelli C, Santoli F, Rana R, Ursitti A, Bonato M, Baraldo S, Contoli M, Papi A. Update on virus-induced asthma exacerbations. Expert Rev Clin Immunol 2023; 19:1259-1272. [PMID: 37470413 DOI: 10.1080/1744666x.2023.2239504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/01/2023] [Accepted: 07/18/2023] [Indexed: 07/21/2023]
Abstract
INTRODUCTION Viral infections are common triggers for asthma exacerbation. Subjects with asthma are more susceptible to viral infections and develop more severe or long-lasting lower respiratory tract symptoms than healthy individuals owing to impaired immune responses. Of the many viruses associated with asthma exacerbation, rhinovirus (RV) is the most frequently identified virus in both adults and children. AREAS COVERED We reviewed epidemiological and clinical links and mechanistic studies on virus-associated asthma exacerbations. We included sections on severe acute respiratory syndrome coronavirus 2 (SARS-CoV2), the latest evidence of coronavirus disease 2019 (COVID-19) in asthma patients, and past and future searches for therapeutic and prevention targets. EXPERT OPINION Early treatment or prevention of viral infections might significantly reduce the rate of asthma exacerbation, which is one of the key points of disease management. Although it is hypothetically possible nowadays to interfere with every step of the infectious cycle of respiratory tract viruses, vaccination development has provided some of the most encouraging results. Future research should proceed toward the development of a wider spectrum of vaccines to achieve a better quality of life for patients with asthma and to reduce the economic burden on the healthcare system.
Collapse
Affiliation(s)
- Francesca Urbani
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Marianna Cometa
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Chiara Martelli
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Federica Santoli
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Roberto Rana
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Antonio Ursitti
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Matteo Bonato
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Simonetta Baraldo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Marco Contoli
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Alberto Papi
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| |
Collapse
|
11
|
Kula A, Makuch E, Lisowska M, Reniewicz P, Lipiński T, Siednienko J. Pellino3 ligase negatively regulates influenza B dependent RIG-I signalling through downregulation of TRAF3-mediated induction of the transcription factor IRF3 and IFNβ production. Immunology 2023. [PMID: 36861386 DOI: 10.1111/imm.13637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 02/19/2023] [Indexed: 03/03/2023] Open
Abstract
Viral infection activates the innate immune system, which recognizes viral components by a variety of pattern recognition receptors and initiates signalling cascades leading to the production of pro-inflammatory cytokines. To date, signalling cascades triggered after virus recognition are not fully characterized and are investigated by many research groups. The critical role of the E3 ubiquitin ligase Pellino3 in antibacterial and antiviral response is now widely accepted, but the precise mechanism remains elusive. In this study, we sought to explore Pellino3 role in the retinoic acid-inducible gene I (RIG-I)-dependent signalling pathway. In this work, the molecular mechanisms of the innate immune response, regulated by Pellino3, were investigated in lung epithelial cells during influenza B virus infection. We used wild-type and Pellino3-deficient A549 cells as model cell lines to examine the role of Pellino3 ligase in the type I interferon (IFN) signalling pathway. Our results indicate that Pellino3 is involved in direct ubiquitination and degradation of the TRAF3, suppressing interferon regulatory factor 3 (IRF3) activation and interferon beta (IFNβ) production.
Collapse
Affiliation(s)
- Anna Kula
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland.,Laboratory of Medical Microbiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Edyta Makuch
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Marta Lisowska
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Patryk Reniewicz
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Tomasz Lipiński
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Jakub Siednienko
- Bioengineering Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| |
Collapse
|
12
|
Burke H, Cellura D, Freeman A, Hicks A, Ostridge K, Watson A, Williams NP, Spalluto CM, Staples KJ, Wilkinson TMA. Pulmonary EV miRNA profiles identify disease and distinct inflammatory endotypes in COPD. Front Med (Lausanne) 2022; 9:1039702. [PMID: 36590967 PMCID: PMC9797812 DOI: 10.3389/fmed.2022.1039702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/28/2022] [Indexed: 12/16/2022] Open
Abstract
Introduction Chronic obstructive pulmonary disease (COPD) is a heterogeneous condition without effective disease modifying therapies. Identification of novel inflammatory endotype markers such as extracellular vesicles (EVs), which are important intercellular messengers carrying microRNA (miRNA), may enable earlier diagnosis and disease stratification for a targeted treatment approach. Our aim was to identify differentially expressed EV miRNA in the lungs of COPD patients compared with healthy ex-smokers and determine whether they can help define inflammatory COPD endotypes. Methods EV miRNA were isolated and sequenced from ex-smoking COPD patients and healthy ex-smoker bronchoalveolar lavage fluid. Results were validated with RT-qPCR and compared to differential inflammatory cell counts. Results Expression analysis identified five upregulated miRNA in COPD (miR-223-3p, miR-2110, miR-182-5p, miR-200b-5p and miR-625-3p) and three downregulated miRNA (miR-138-5p, miR-338-3p and miR-204-5p), all with a log2 fold change of >1/-1, FDR < 0.05. These miRNAs correlated with disease defining characteristics such as FEF 25-75% (a small airways disease measure) and DLCO% (a surrogate measure of emphysema). Receiver operator curve analysis demonstrated miR-2110, miR-223-3p, and miR-182-5p showed excellent combinatory predictive ability (AUC 0.91, p < 0.0001) in differentiating between health and mild COPD. Furthermore, miR-223-3p and miR-338-3p correlated with airway eosinophilia and were able to distinguish "pure eosinophilic" COPD from other airway inflammatory subtypes (AUC 0.94 and 0.85, respectively). Discussion This is the first study to identify differentially expressed miRNA in COPD bronchoalveolar lavage fluid EVs. These findings suggest specific lung derived EV miRNA are a strong predictor of disease presence even in mild COPD. Furthermore, specific miRNA correlated with inflammatory cell numbers in COPD, and may have a role in defining inflammatory endotypes for future treatment stratification.
Collapse
Affiliation(s)
- Hannah Burke
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Doriana Cellura
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Anna Freeman
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Alex Hicks
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Kris Ostridge
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Alastair Watson
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Nicholas P. Williams
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - C. Mirella Spalluto
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Karl J. Staples
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Tom M. A. Wilkinson
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| |
Collapse
|
13
|
Humbert MV, Spalluto CM, Bell J, Blume C, Conforti F, Davies ER, Dean LSN, Elkington P, Haitchi HM, Jackson C, Jones MG, Loxham M, Lucas JS, Morgan H, Polak M, Staples KJ, Swindle EJ, Tezera L, Watson A, Wilkinson TMA. Towards an artificial human lung: modelling organ-like complexity to aid mechanistic understanding. Eur Respir J 2022; 60:2200455. [PMID: 35777774 DOI: 10.1183/13993003.00455-2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/11/2022] [Indexed: 11/05/2022]
Abstract
Respiratory diseases account for over 5 million deaths yearly and are a huge burden to healthcare systems worldwide. Murine models have been of paramount importance to decode human lung biology in vivo, but their genetic, anatomical, physiological and immunological differences with humans significantly hamper successful translation of research into clinical practice. Thus, to clearly understand human lung physiology, development, homeostasis and mechanistic dysregulation that may lead to disease, it is essential to develop models that accurately recreate the extraordinary complexity of the human pulmonary architecture and biology. Recent advances in micro-engineering technology and tissue engineering have allowed the development of more sophisticated models intending to bridge the gap between the native lung and its replicates in vitro Alongside advanced culture techniques, remarkable technological growth in downstream analyses has significantly increased the predictive power of human biology-based in vitro models by allowing capture and quantification of complex signals. Refined integrated multi-omics readouts could lead to an acceleration of the translational pipeline from in vitro experimental settings to drug development and clinical testing in the future. This review highlights the range and complexity of state-of-the-art lung models for different areas of the respiratory system, from nasal to large airways, small airways and alveoli, with consideration of various aspects of disease states and their potential applications, including pre-clinical drug testing. We explore how development of optimised physiologically relevant in vitro human lung models could accelerate the identification of novel therapeutics with increased potential to translate successfully from the bench to the patient's bedside.
Collapse
Affiliation(s)
- Maria Victoria Humbert
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Cosma Mirella Spalluto
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- M.V. Humbert and C.M. Spalluto are co-first authors and contributed equally to this work
| | - Joseph Bell
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Cornelia Blume
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Franco Conforti
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Elizabeth R Davies
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Lareb S N Dean
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Paul Elkington
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Hans Michael Haitchi
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Claire Jackson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Mark G Jones
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Matthew Loxham
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Jane S Lucas
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Hywel Morgan
- Institute for Life Sciences, University of Southampton, Southampton, UK
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton, UK
| | - Marta Polak
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Karl J Staples
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Emily J Swindle
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Liku Tezera
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Department of Infection and Immunity, Faculty of Medicine, University College London, London, UK
| | - Alastair Watson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Tom M A Wilkinson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| |
Collapse
|
14
|
Farne H, Lin L, Jackson DJ, Rattray M, Simpson A, Custovic A, Joshi S, Wilson PA, Williamson R, Edwards MR, Singanayagam A, Johnston SL. In vivo bronchial epithelial interferon responses are augmented in asthma on day 4 following experimental rhinovirus infection. Thorax 2022; 77:929-932. [PMID: 35790388 DOI: 10.1136/thoraxjnl-2021-217389] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 03/15/2022] [Indexed: 11/03/2022]
Abstract
Despite good evidence of impaired innate antiviral responses in asthma, trials of inhaled interferon-β given during exacerbations showed only modest benefits in moderate/severe asthma. Using human experimental rhinovirus infection, we observe robust in vivo induction of bronchial epithelial interferon response genes 4 days after virus inoculation in 25 subjects with asthma but not 11 control subjects. This signature correlated with virus loads and lower respiratory symptoms. Our data indicate that the in vivo innate antiviral response is dysregulated in asthma and open up the potential that prophylactic rather than therapeutic interferon therapy may have greater clinical benefit.
Collapse
Affiliation(s)
- Hugo Farne
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Lijing Lin
- Division of Informatics, Imaging & Data Sciences, The University of Manchester, Manchester, UK
| | - David J Jackson
- National Heart and Lung Institute, Imperial College London, London, UK
- Guy's Severe Asthma Centre, Guy's and St Thomas' NHS Foundation Trust, London, UK
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - Magnus Rattray
- Division of Informatics, Imaging & Data Sciences, The University of Manchester, Manchester, UK
| | - Angela Simpson
- Division of Infection, Immunity & Respiratory Medicine, The University of Manchester, Manchester, UK
| | - Adnan Custovic
- Department of Paediatrics, Imperial College London, London, UK
| | | | | | | | - Michael R Edwards
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | | |
Collapse
|
15
|
Yun JH, Lee S, Srinivasa P, Morrow J, Chase R, Saferali A, Xu Z, Cho M, Castaldi P, Hersh CP. An interferon-inducible signature of airway disease from blood gene expression profiling. Eur Respir J 2022; 59:2100569. [PMID: 34649980 PMCID: PMC9245457 DOI: 10.1183/13993003.00569-2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 09/24/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND The molecular basis of airway remodelling in chronic obstructive pulmonary disease (COPD) remains poorly understood. We identified gene expression signatures associated with chest computed tomography (CT) scan airway measures to understand molecular pathways associated with airway disease. METHODS In 2396 subjects in the COPDGene Study, we examined the relationship between quantitative CT airway phenotypes and blood transcriptomes to identify airway disease-specific genes and to define an airway wall thickness (AWT) gene set score. Multivariable regression analyses were performed to identify associations of the AWT score with clinical phenotypes, bronchial gene expression and genetic variants. RESULTS Type 1 interferon (IFN)-induced genes were consistently associated with AWT, square root wall area of a hypothetical airway with 10 mm internal perimeter (Pi10) and wall area percentage, with the strongest enrichment in AWT. A score derived from 18 genes whose expression was associated with AWT was associated with COPD-related phenotypes including reduced lung function (forced expiratory volume in 1 s percentage predicted β= -3.4; p<0.05) and increased exacerbations (incidence rate ratio 1.7; p<0.05). The AWT score was reproducibly associated with AWT in bronchial samples from 23 subjects (β=3.22; p<0.05). The blood AWT score was associated with genetic variant rs876039, an expression quantitative trait locus for IKZF1, a gene that regulates IFN signalling and is associated with inflammatory diseases. CONCLUSIONS A gene expression signature with IFN-stimulated genes from peripheral blood and bronchial brushings is associated with CT AWT, lung function and exacerbations. Shared genes and genetic associations suggest viral responses and/or autoimmune dysregulation as potential underlying mechanisms of airway disease in COPD.
Collapse
Affiliation(s)
- Jeong H Yun
- Channing Division of Network Medicine, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sool Lee
- Channing Division of Network Medicine, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Pooja Srinivasa
- Channing Division of Network Medicine, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jarrett Morrow
- Channing Division of Network Medicine, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Robert Chase
- Channing Division of Network Medicine, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Aadbida Saferali
- Channing Division of Network Medicine, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Zhonghui Xu
- Channing Division of Network Medicine, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Michael Cho
- Channing Division of Network Medicine, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Peter Castaldi
- Channing Division of Network Medicine, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Chang JJY, Gleeson J, Rawlinson D, De Paoli-Iseppi R, Zhou C, Mordant FL, Londrigan SL, Clark MB, Subbarao K, Stinear TP, Coin LJM, Pitt ME. Long-Read RNA Sequencing Identifies Polyadenylation Elongation and Differential Transcript Usage of Host Transcripts During SARS-CoV-2 In Vitro Infection. Front Immunol 2022; 13:832223. [PMID: 35464437 PMCID: PMC9019466 DOI: 10.3389/fimmu.2022.832223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/14/2022] [Indexed: 12/04/2022] Open
Abstract
Better methods to interrogate host-pathogen interactions during Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infections are imperative to help understand and prevent this disease. Here we implemented RNA-sequencing (RNA-seq) using Oxford Nanopore Technologies (ONT) long-reads to measure differential host gene expression, transcript polyadenylation and isoform usage within various epithelial cell lines permissive and non-permissive for SARS-CoV-2 infection. SARS-CoV-2-infected and mock-infected Vero (African green monkey kidney epithelial cells), Calu-3 (human lung adenocarcinoma epithelial cells), Caco-2 (human colorectal adenocarcinoma epithelial cells) and A549 (human lung carcinoma epithelial cells) were analyzed over time (0, 2, 24, 48 hours). Differential polyadenylation was found to occur in both infected Calu-3 and Vero cells during a late time point (48 hpi), with Gene Ontology (GO) terms such as viral transcription and translation shown to be significantly enriched in Calu-3 data. Poly(A) tails showed increased lengths in the majority of the differentially polyadenylated transcripts in Calu-3 and Vero cell lines (up to ~101 nt in mean poly(A) length, padj = 0.029). Of these genes, ribosomal protein genes such as RPS4X and RPS6 also showed downregulation in expression levels, suggesting the importance of ribosomal protein genes during infection. Furthermore, differential transcript usage was identified in Caco-2, Calu-3 and Vero cells, including transcripts of genes such as GSDMB and KPNA2, which have previously been implicated in SARS-CoV-2 infections. Overall, these results highlight the potential role of differential polyadenylation and transcript usage in host immune response or viral manipulation of host mechanisms during infection, and therefore, showcase the value of long-read sequencing in identifying less-explored host responses to disease.
Collapse
Affiliation(s)
- Jessie J-Y Chang
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Josie Gleeson
- Centre for Stem Cell Systems, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Daniel Rawlinson
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Ricardo De Paoli-Iseppi
- Centre for Stem Cell Systems, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Chenxi Zhou
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Francesca L Mordant
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Sarah L Londrigan
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Michael B Clark
- Centre for Stem Cell Systems, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,World Health Organization (WHO) Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Lachlan J M Coin
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia.,Department of Infectious Disease, Imperial College London, London, United Kingdom.,Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Miranda E Pitt
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
17
|
Wu W, Tian L, Zhang W, Booth JL, Ritchey JW, Wu S, Xu C, Brown BR, Metcalf JP. Early IFN-β administration protects cigarette smoke exposed mice against lethal influenza virus infection without increasing lung inflammation. Sci Rep 2022; 12:4080. [PMID: 35260752 PMCID: PMC8902729 DOI: 10.1038/s41598-022-08066-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/25/2022] [Indexed: 11/21/2022] Open
Abstract
During influenza A virus (IAV) infection, it is unclear whether type I interferons (IFNs) have defensive antiviral effects or contribute to immunopathology in smokers. We treated nonsmoking (NS) and cigarette smoke (CS)-exposed mice intranasally with early (prophylactic) or late (therapeutic) IFN-β. We compared the mortality and innate immune responses of the treated mice following challenge with IAV. In NS mice, both early and late IFN-β administration decreased the survival rate in mice infected with IAV, with late IFN-β administration having the greatest effect on survival. In contrast, in CS-exposed mice, early IFN-β administration significantly increased survival during IAV infection while late IFN-β administration did not alter mortality. With regards to inflammation, in NS mice, IFN-β administration, especially late administration, significantly increased IAV-induced inflammation and lung injury. Early IFN-β administration to CS-exposed mice did not increase IAV-induced inflammation and lung injury as occurred in NS mice. Our results demonstrate, although IFN-β administration worsens the susceptibility of NS mice to influenza infection with increased immunopathology, early IFN-β administration to CS-exposed mice, which have suppression of the intrinsic IFN response, improved outcomes during influenza infection.
Collapse
Affiliation(s)
- Wenxin Wu
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Room 425, RP1, 800 N. Research Pkwy., Oklahoma City, OK, 73104, USA.
| | - Lili Tian
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Room 425, RP1, 800 N. Research Pkwy., Oklahoma City, OK, 73104, USA
| | - Wei Zhang
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Room 425, RP1, 800 N. Research Pkwy., Oklahoma City, OK, 73104, USA
| | - J Leland Booth
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Room 425, RP1, 800 N. Research Pkwy., Oklahoma City, OK, 73104, USA
| | - Jerry William Ritchey
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Shuhua Wu
- Division of Geriatrics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Chao Xu
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Brent R Brown
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Room 425, RP1, 800 N. Research Pkwy., Oklahoma City, OK, 73104, USA
| | - Jordan P Metcalf
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Room 425, RP1, 800 N. Research Pkwy., Oklahoma City, OK, 73104, USA.
- Veterans Affairs Medical Center, Oklahoma City, OK, USA.
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
18
|
Burke H, Freeman A, O'Regan P, Wysocki O, Freitas A, Dushianthan A, Celinski M, Batchelor J, Phan H, Borca F, Sheard N, Williams S, Watson A, Fitzpatrick P, Landers D, Wilkinson T. Biomarker identification using dynamic time warping analysis: a longitudinal cohort study of patients with COVID-19 in a UK tertiary hospital. BMJ Open 2022; 12:e050331. [PMID: 35168965 PMCID: PMC8852240 DOI: 10.1136/bmjopen-2021-050331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 01/27/2022] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES COVID-19 is a heterogeneous disease, and many reports have described variations in demographic, biochemical and clinical features at presentation influencing overall hospital mortality. However, there is little information regarding longitudinal changes in laboratory prognostic variables in relation to disease progression in hospitalised patients with COVID-19. DESIGN AND SETTING This retrospective observational report describes disease progression from symptom onset, to admission to hospital, clinical response and discharge/death among patients with COVID-19 at a tertiary centre in South East England. PARTICIPANTS Six hundred and fifty-one patients treated for SARS-CoV-2 between March and September 2020 were included in this analysis. Ethical approval was obtained from the HRA Specific Review Board (REC 20/HRA/2986) for waiver of informed consent. RESULTS The majority of patients presented within 1 week of symptom onset. The lowest risk patients had low mortality (1/45, 2%), and most were discharged within 1 week after admission (30/45, 67%). The highest risk patients, as determined by the 4C mortality score predictor, had high mortality (27/29, 93%), with most dying within 1 week after admission (22/29, 76%). Consistent with previous reports, most patients presented with high levels of C reactive protein (CRP) (67% of patients >50 mg/L), D-dimer (98%>upper limit of normal (ULN)), ferritin (65%>ULN), lactate dehydrogenase (90%>ULN) and low lymphocyte counts (81% CONCLUSIONS Serial measurement of routine blood tests may be a useful prognostic tool for monitoring treatment response in hospitalised patients with COVID-19. Changes in other biochemical parameters often included in a 'COVID-19 bundle' did not show significant association with outcome, suggesting there may be limited clinical benefit of serial sampling. This may have direct clinical utility in the context of escalating healthcare costs of the pandemic.
Collapse
Affiliation(s)
- Hannah Burke
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Anna Freeman
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Paul O'Regan
- Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Oskar Wysocki
- Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Andre Freitas
- Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | | | | | - James Batchelor
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Hang Phan
- Clinical Informatics Research Unit, University of Southampton Faculty of Medicine, Southampton, UK
- University of Southampton, Southampton, UK
| | - Florina Borca
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Natasha Sheard
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Sarah Williams
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Alastair Watson
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Paul Fitzpatrick
- University of Manchester, Cancer Biomarker Centre, Cancer Research UK Manchester Institute, Manchester, UK
| | - Dónal Landers
- Digital Experimental Cancer Medicine Team, University of Manchester, Cancer Biomarker Centre, Cancer Research UK Manchester Institute, Alderley Edge, Cheshire, UK
| | - Tom Wilkinson
- Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
19
|
Guo-Parke H, Linden D, Weldon S, Kidney JC, Taggart CC. Deciphering Respiratory-Virus-Associated Interferon Signaling in COPD Airway Epithelium. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:121. [PMID: 35056429 PMCID: PMC8781535 DOI: 10.3390/medicina58010121] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/08/2022] [Accepted: 01/11/2022] [Indexed: 01/08/2023]
Abstract
COPD is a chronic lung disorder characterized by a progressive and irreversible airflow obstruction, and persistent pulmonary inflammation. It has become a global epidemic affecting 10% of the population, and is the third leading cause of death worldwide. Respiratory viruses are a primary cause of COPD exacerbations, often leading to secondary bacterial infections in the lower respiratory tract. COPD patients are more susceptible to viral infections and associated severe disease, leading to accelerated lung function deterioration, hospitalization, and an increased risk of mortality. The airway epithelium plays an essential role in maintaining immune homeostasis, and orchestrates the innate and adaptive responses of the lung against inhaled and pathogen insults. A healthy airway epithelium acts as the first line of host defense by maintaining barrier integrity and the mucociliary escalator, secreting an array of inflammatory mediators, and initiating an antiviral state through the interferon (IFN) response. The airway epithelium is a major site of viral infection, and the interaction between respiratory viruses and airway epithelial cells activates host defense mechanisms, resulting in rapid virus clearance. As such, the production of IFNs and the activation of IFN signaling cascades directly contributes to host defense against viral infections and subsequent innate and adaptive immunity. However, the COPD airway epithelium exhibits an altered antiviral response, leading to enhanced susceptibility to severe disease and impaired IFN signaling. Despite decades of research, there is no effective antiviral therapy for COPD patients. Herein, we review current insights into understanding the mechanisms of viral evasion and host IFN antiviral defense signaling impairment in COPD airway epithelium. Understanding how antiviral mechanisms operate in COPD exacerbations will facilitate the discovery of potential therapeutic interventions to reduce COPD hospitalization and disease severity.
Collapse
Affiliation(s)
- Hong Guo-Parke
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queens University Belfast, Belfast BT9 7AE, UK; (H.G.-P.); (D.L.); (S.W.)
| | - Dermot Linden
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queens University Belfast, Belfast BT9 7AE, UK; (H.G.-P.); (D.L.); (S.W.)
| | - Sinéad Weldon
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queens University Belfast, Belfast BT9 7AE, UK; (H.G.-P.); (D.L.); (S.W.)
| | - Joseph C. Kidney
- Department of Respiratory Medicine, Mater Hospital Belfast, Belfast BT14 6AB, UK;
| | - Clifford C. Taggart
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queens University Belfast, Belfast BT9 7AE, UK; (H.G.-P.); (D.L.); (S.W.)
| |
Collapse
|
20
|
Freeman A, Watson A, O'Regan P, Wysocki O, Burke H, Freitas A, Livingstone R, Dushianthan A, Celinski M, Batchelor J, Phan H, Borca F, Fitzpatrick P, Landers D, Wilkinson TM. Wave comparisons of clinical characteristics and outcomes of COVID-19 admissions - Exploring the impact of treatment and strain dynamics. J Clin Virol 2022; 146:105031. [PMID: 34844145 PMCID: PMC8608665 DOI: 10.1016/j.jcv.2021.105031] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 11/11/2021] [Accepted: 11/21/2021] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Dexamethasone has now been incorporated into the standard of care for COVID-19 hospital patients. However, larger intensive care unit studies have failed to show discernible improvements in mortality in the recent wave. We aimed to investigate the impacts of these factors on disease outcomes in a UK hospital study. METHODS This retrospective observational study reports patient characteristics, interventions and outcomes in COVID-19 patients from a UK teaching hospital; cohort 1, pre 16th June-2020 (pre-dexamethasone); cohort 2, 17th June to 30th November-2020 (post-dexamethasone, pre-VOC 202,012/01 as dominant strain); cohort 3, 1st December-2020 to 3rd March-2021 (during establishment of VOC202012/01 as the dominant strain). RESULTS Dexamethasone treatment was more common in cohorts 2 and 3 (42.7% and 51.6%) compared with cohort 1 (2.5%). After adjusting for risk, odds of death within 28 days were 2-fold lower in cohort 2 vs 1 (OR:0.47,[0.27,0.79],p = 0.006). Mortality was higher cohort 3 vs 2 (20% vs 14%); but not significantly different to cohort 1 (OR: 0.86,[0.64, 1.15],p = 0.308). CONCLUSIONS The real world finding of lower mortality following dexamethasone supports the published trial evidence and highlights ongoing need for research with introduction of new treatments and ongoing concern over new COVID-19 variants.
Collapse
Affiliation(s)
- Anna Freeman
- Faculty of Medicine, University of Southampton, United Kingdom; University Hospitals Southampton NHS Foundation Trust, United Kingdom.
| | - Alastair Watson
- Faculty of Medicine, University of Southampton, United Kingdom; University Hospitals Southampton NHS Foundation Trust, United Kingdom
| | - Paul O'Regan
- Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, Cancer Research UK Manchester Institute, The University of Manchester, United Kingdom
| | - Oskar Wysocki
- Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, Cancer Research UK Manchester Institute, The University of Manchester, United Kingdom; Department of Computer Science, The University of Manchester, United Kingdom
| | - Hannah Burke
- Faculty of Medicine, University of Southampton, United Kingdom; University Hospitals Southampton NHS Foundation Trust, United Kingdom
| | - Andre Freitas
- Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, Cancer Research UK Manchester Institute, The University of Manchester, United Kingdom; Department of Computer Science, The University of Manchester, United Kingdom; Idiap Research Institute, Switzerland
| | - Robert Livingstone
- Faculty of Medicine, University of Southampton, United Kingdom; University Hospitals Southampton NHS Foundation Trust, United Kingdom
| | - Ahilanadan Dushianthan
- Faculty of Medicine, University of Southampton, United Kingdom; University Hospitals Southampton NHS Foundation Trust, United Kingdom; NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, United Kingdom
| | - Michael Celinski
- Faculty of Medicine, University of Southampton, United Kingdom; University Hospitals Southampton NHS Foundation Trust, United Kingdom
| | - James Batchelor
- Faculty of Medicine, University of Southampton, United Kingdom; University Hospitals Southampton NHS Foundation Trust, United Kingdom; Institute for Life Sciences, University of Southampton, United Kingdom; Clinical Informatics Research Unit Faculty of Medicine, University of Southampton,United Kingdom
| | - Hang Phan
- Faculty of Medicine, University of Southampton, United Kingdom; Clinical Informatics Research Unit Faculty of Medicine, University of Southampton,United Kingdom; NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, United Kingdom
| | - Florina Borca
- Faculty of Medicine, University of Southampton, United Kingdom; University Hospitals Southampton NHS Foundation Trust, United Kingdom; Clinical Informatics Research Unit Faculty of Medicine, University of Southampton,United Kingdom; NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, United Kingdom
| | - Paul Fitzpatrick
- Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, Cancer Research UK Manchester Institute, The University of Manchester, United Kingdom
| | - Donal Landers
- Digital Experimental Cancer Medicine Team, Cancer Biomarker Centre, Cancer Research UK Manchester Institute, The University of Manchester, United Kingdom
| | - Tom Ma Wilkinson
- Faculty of Medicine, University of Southampton, United Kingdom; University Hospitals Southampton NHS Foundation Trust, United Kingdom
| |
Collapse
|
21
|
Watson A, Wilkinson TM. Digital healthcare in COPD management: a narrative review on the advantages, pitfalls, and need for further research. Ther Adv Respir Dis 2022; 16:17534666221075493. [PMID: 35234090 PMCID: PMC8894614 DOI: 10.1177/17534666221075493] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/07/2022] [Indexed: 12/27/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) remains a leading cause of morbidity and mortality despite current treatment strategies which focus on smoking cessation, pulmonary rehabilitation, and symptomatic relief. A focus of COPD care is to encourage self-management, particularly during COVID-19, where much face-to-face care has been reduced or ceased. Digital health solutions may offer affordable and scalable solutions to support COPD patient education and self-management, such solutions could improve clinical outcomes and expand service reach for limited additional cost. However, optimal ways to deliver digital medicine are still in development, and there are a number of important considerations for clinicians, commissioners, and patients to ensure successful implementation of digitally augmented care. In this narrative review, we discuss advantages, pitfalls, and future prospects of digital healthcare, which offer a variety of tools including self-management plans, education videos, inhaler training videos, feedback to patients and healthcare professionals (HCPs), exacerbation monitoring, and pulmonary rehabilitation. We discuss the key issues with sustaining patient and HCP engagement and limiting attrition of use, interoperability with devices, integration into healthcare systems, and ensuring inclusivity and accessibility. We explore the essential areas of research beyond determining safety and efficacy to understand the acceptability of digital healthcare solutions to patients, clinicians, and healthcare systems, and hence ways to improve this and sustain engagement. Finally, we explore the regulatory challenges to ensure quality and engagement and effective integration into current healthcare systems and care pathways, while maintaining patients' autonomy and privacy. Understanding and addressing these issues and successful incorporation of an acceptable, simple, scalable, affordable, and future-proof digital solution into healthcare systems could help remodel global chronic disease management and fractured healthcare systems to provide best patient care and optimisation of healthcare resources to meet the global burden and unmet clinical need of COPD.
Collapse
Affiliation(s)
- Alastair Watson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UKNIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UKCollege of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Tom M.A. Wilkinson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK. NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| |
Collapse
|
22
|
Reid LV, Spalluto CM, Watson A, Staples KJ, Wilkinson TMA. The Role of Extracellular Vesicles as a Shared Disease Mechanism Contributing to Multimorbidity in Patients With COPD. Front Immunol 2021; 12:754004. [PMID: 34925327 PMCID: PMC8675939 DOI: 10.3389/fimmu.2021.754004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/04/2021] [Indexed: 01/27/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the leading causes of death worldwide. Individuals with COPD typically experience a progressive, debilitating decline in lung function as well as systemic manifestations of the disease. Multimorbidity, is common in COPD patients and increases the risk of hospitalisation and mortality. Central to the genesis of multimorbidity in COPD patients is a self-perpetuating, abnormal immune and inflammatory response driven by factors including ageing, pollutant inhalation (including smoking) and infection. As many patients with COPD have multiple concurrent chronic conditions, which require an integrative management approach, there is a need to greater understand the shared disease mechanisms contributing to multimorbidity. The intercellular transfer of extracellular vesicles (EVs) has recently been proposed as an important method of local and distal cell-to-cell communication mediating both homeostatic and pathological conditions. EVs have been identified in many biological fluids and provide a stable capsule for the transfer of cargo including proteins, lipids and nucleic acids. Of these cargo, microRNAs (miRNAs), which are short 17-24 nucleotide non-coding RNA molecules, have been amongst the most extensively studied. There is evidence to support that miRNA are selectively packaged into EVs and can regulate recipient cell gene expression including major pathways involved in inflammation, apoptosis and fibrosis. Furthermore changes in EV cargo including miRNA have been reported in many chronic diseases and in response to risk factors including respiratory infections, noxious stimuli and ageing. In this review, we discuss the potential of EVs and EV-associated miRNA to modulate shared pathological processes in chronic diseases. Further delineating these may lead to the identification of novel biomarkers and therapeutic targets for patients with COPD and multimorbidities.
Collapse
Affiliation(s)
- Laura V Reid
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - C Mirella Spalluto
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,National Institute for Health Research Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - Alastair Watson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,National Institute for Health Research Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom.,Birmingham Medical School, University of Birmingham, Birmingham, United Kingdom
| | - Karl J Staples
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,National Institute for Health Research Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - Tom M A Wilkinson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,National Institute for Health Research Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| |
Collapse
|
23
|
Cojocaru E, Cojocaru C, Antoniu SA, Stafie CS, Rajnoveanu A, Rajnoveanu RM. Inhaled interferons beta and SARS-COV2 infection: a preliminary therapeutic perspective. Expert Rev Respir Med 2021; 16:257-261. [PMID: 34793285 PMCID: PMC8726005 DOI: 10.1080/17476348.2022.2008910] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Introduction SARS-COV2 infection represents a therapeutic challenge due to the limited number of effective therapies available and due to the fact that it is not clear which host response in terms of inflammation pattern is the most predictive for an optimal (and rapid) recovery. Interferon β pathway is impaired in SARS-COV2 infection and this is associated with a bigger disease burden. Exogenous inhaled interferon might be beneficial in this setting. Areas covered Nebulized interferon-β is currently investigated as a potential therapy for SARS-COV2 because the available data from a phase II study demonstrate that this medication is able to accelerate the recovery from disease. Expert opinion Further clinical studies are needed in order to better document the efficacy of this therapy especially in severe forms of COVID-19, the optimal duration of therapy and if such a medication is appropriate for domiciliary use. Also combined regimens with antivirals or with compounds which are able to enhance the endogenous production of interferon might be of promise.
Collapse
Affiliation(s)
- Elena Cojocaru
- Morpho-Functional Sciences II Department, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
| | - Cristian Cojocaru
- Medical III Department, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
| | | | - Celina Silvia Stafie
- Preventive Medicine and Interdisciplinarity Department, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
| | - Armand Rajnoveanu
- Occupational Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | |
Collapse
|
24
|
Cumpstey AF, Clark AD, Santolini J, Jackson AA, Feelisch M. COVID-19: A Redox Disease-What a Stress Pandemic Can Teach Us About Resilience and What We May Learn from the Reactive Species Interactome About Its Treatment. Antioxid Redox Signal 2021; 35:1226-1268. [PMID: 33985343 DOI: 10.1089/ars.2021.0017] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Significance: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus causing coronavirus disease 2019 (COVID-19), affects every aspect of human life by challenging bodily, socioeconomic, and political systems at unprecedented levels. As vaccines become available, their distribution, safety, and efficacy against emerging variants remain uncertain, and specific treatments are lacking. Recent Advances: Initially affecting the lungs, COVID-19 is a complex multisystems disease that disturbs the whole-body redox balance and can be long-lasting (Long-COVID). Numerous risk factors have been identified, but the reasons for variations in susceptibility to infection, disease severity, and outcome are poorly understood. The reactive species interactome (RSI) was recently introduced as a framework to conceptualize how cells and whole organisms sense, integrate, and accommodate stress. Critical Issues: We here consider COVID-19 as a redox disease, offering a holistic perspective of its effects on the human body, considering the vulnerability of complex interconnected systems with multiorgan/multilevel interdependencies. Host/viral glycan interactions underpin SARS-CoV-2's extraordinary efficiency in gaining cellular access, crossing the epithelial/endothelial barrier to spread along the vascular/lymphatic endothelium, and evading antiviral/antioxidant defences. An inflammation-driven "oxidative storm" alters the redox landscape, eliciting epithelial, endothelial, mitochondrial, metabolic, and immune dysfunction, and coagulopathy. Concomitantly reduced nitric oxide availability renders the sulfur-based redox circuitry vulnerable to oxidation, with eventual catastrophic failure in redox communication/regulation. Host nutrient limitations are crucial determinants of resilience at the individual and population level. Future Directions: While inflicting considerable damage to health and well-being, COVID-19 may provide the ultimate testing ground to improve the diagnosis and treatment of redox-related stress diseases. "Redox phenotyping" of patients to characterize whole-body RSI status as the disease progresses may inform new therapeutic approaches to regain redox balance, reduce mortality in COVID-19 and other redox diseases, and provide opportunities to tackle Long-COVID. Antioxid. Redox Signal. 35, 1226-1268.
Collapse
Affiliation(s)
- Andrew F Cumpstey
- Respiratory and Critical Care Research Group, Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Anna D Clark
- Respiratory and Critical Care Research Group, Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jérôme Santolini
- Institute for Integrative Biology of the Cell (I2BC), Biochemistry, Biophysics and Structural Biology, CEA, CNRS, Université Paris-Sud, Universite Paris-Saclay, Gif-sur-Yvette, France
| | - Alan A Jackson
- Human Nutrition, University of Southampton and University Hospital Southampton, Southampton, United Kingdom
| | - Martin Feelisch
- Respiratory and Critical Care Research Group, Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
25
|
Ackland J, Watson A, Wilkinson TMA, Staples KJ. Interrupting the Conversation: Implications for Crosstalk Between Viral and Bacterial Infections in the Asthmatic Airway. FRONTIERS IN ALLERGY 2021; 2:738987. [PMID: 35386999 PMCID: PMC8974750 DOI: 10.3389/falgy.2021.738987] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/20/2021] [Indexed: 12/20/2022] Open
Abstract
Asthma is a heterogeneous, chronic respiratory disease affecting 300 million people and is thought to be driven by different inflammatory endotypes influenced by a myriad of genetic and environmental factors. The complexity of asthma has rendered it challenging to develop preventative and disease modifying therapies and it remains an unmet clinical need. Whilst many factors have been implicated in asthma pathogenesis and exacerbations, evidence indicates a prominent role for respiratory viruses. However, advances in culture-independent detection methods and extensive microbial profiling of the lung, have also demonstrated a role for respiratory bacteria in asthma. In particular, airway colonization by the Proteobacteria species Nontypeable Haemophilus influenzae (NTHi) and Moraxella catarrhalis (Mcat) is associated with increased risk of developing recurrent wheeze and asthma in early life, poor clinical outcomes in established adult asthma and the development of more severe inflammatory phenotypes. Furthermore, emerging evidence indicates that bacterial-viral interactions may influence exacerbation risk and disease severity, highlighting the need to consider the impact chronic airway colonization by respiratory bacteria has on influencing host responses to viral infection. In this review, we first outline the currently understood role of viral and bacterial infections in precipitating asthma exacerbations and discuss the underappreciated potential impact of bacteria-virus crosstalk in modulating host responses. We discuss the mechanisms by which early life infection may predispose to asthma development. Finally, we consider how infection and persistent airway colonization may drive different asthma phenotypes, with a view to identifying pathophysiological mechanisms that may prove tractable to new treatment modalities.
Collapse
Affiliation(s)
- Jodie Ackland
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
| | - Alastair Watson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Tom M. A. Wilkinson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
- Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, United Kingdom
| | - Karl J. Staples
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
- Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, United Kingdom
- *Correspondence: Karl J. Staples
| |
Collapse
|
26
|
Coultas JA, Cafferkey J, Mallia P, Johnston SL. Experimental Antiviral Therapeutic Studies for Human Rhinovirus Infections. J Exp Pharmacol 2021; 13:645-659. [PMID: 34276229 PMCID: PMC8277446 DOI: 10.2147/jep.s255211] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/01/2021] [Indexed: 12/17/2022] Open
Abstract
Rhinovirus infection is common and usually causes mild, self-limiting upper respiratory tract symptoms. Rhinoviruses can cause exacerbation of chronic respiratory diseases, such as asthma or chronic obstructive pulmonary disease, leading to a significant burden of morbidity and mortality. There has been a great deal of progress in efforts to understand the immunological basis of rhinovirus infection. However, despite a number of in vitro and in vivo attempts, there have been no effective treatments developed. This review article summarises the up to date virological and immunological understanding of these infections. We discuss the challenges researchers face, and key solutions, in their work to investigate potential therapies including in vivo rhinovirus challenge studies. Finally, we explore past and present experimental therapeutic strategies employed in the treatment of rhinovirus infections and highlight promising areas of future work.
Collapse
Affiliation(s)
- James A Coultas
- National Heart and Lung Institute, Imperial College London, London, UK
| | - John Cafferkey
- Respiratory Medicine, St Mary's Hospital, Imperial College Healthcare Foundation Trust, London, UK
| | - Patrick Mallia
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
27
|
Watson A, Öberg L, Angermann B, Spalluto CM, Hühn M, Burke H, Cellura D, Freeman A, Muthas D, Etal D, Belfield G, Karlsson F, Nordström K, Ostridge K, Staples KJ, Wilkinson T. Dysregulation of COVID-19 related gene expression in the COPD lung. Respir Res 2021; 22:164. [PMID: 34051791 PMCID: PMC8164067 DOI: 10.1186/s12931-021-01755-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/17/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) patients are at increased risk of poor outcome from Coronavirus disease (COVID-19). Early data suggest elevated Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) receptor angiotensin converting enzyme 2 (ACE2) expression, but relationships to disease phenotype and downstream regulators of inflammation in the Renin-Angiotensin system (RAS) are unknown. We aimed to determine the relationship between RAS gene expression relevant to SARS-CoV-2 infection in the lung with disease characteristics in COPD, and the regulation of newly identified SARS-CoV-2 receptors and spike-cleaving proteases, important for SARS-CoV-2 infection. METHODS We quantified gene expression using RNA sequencing of epithelial brushings and bronchial biopsies from 31 COPD and 37 control subjects. RESULTS ACE2 gene expression (log2-fold change (FC)) was increased in COPD compared to ex-smoking (HV-ES) controls in epithelial brushings (0.25, p = 0.042) and bronchial biopsies (0.23, p = 0.050), and correlated with worse lung function (r = - 0.28, p = 0.0090). ACE2 was further increased in frequent exacerbators compared to infrequent exacerbators (0.51, p = 0.00045) and associated with use of ACE inhibitors (ACEi) (0.50, p = 0.0034), having cardiovascular disease (0.23, p = 0.048) or hypertension (0.34, p = 0.0089), and inhaled corticosteroid use in COPD subjects in bronchial biopsies (0.33, p = 0.049). Angiotensin II receptor type (AGTR)1 and 2 expression was decreased in COPD bronchial biopsies compared to HV-ES controls with log2FC of -0.26 (p = 0.033) and - 0.40, (p = 0.0010), respectively. However, the AGTR1:2 ratio was increased in COPD subjects compared with HV-ES controls, log2FC of 0.57 (p = 0.0051). Basigin, a newly identified potential SARS-CoV-2 receptor was also upregulated in both brushes, log2FC of 0.17 (p = 0.0040), and bronchial biopsies, (log2FC of 0.18 (p = 0.017), in COPD vs HV-ES. Transmembrane protease, serine (TMPRSS)2 was not differentially regulated between control and COPD. However, various other spike-cleaving proteases were, including TMPRSS4 and Cathepsin B, in both epithelial brushes (log2FC of 0.25 (p = 0.0012) and log2FC of 0.56 (p = 5.49E-06), respectively) and bronchial biopsies (log2FC of 0.49 (p = 0.00021) and log2FC of 0.246 (p = 0.028), respectively). CONCLUSION This study identifies key differences in expression of genes related to susceptibility and aetiology of COVID-19 within the COPD lung. Further studies to understand the impact on clinical course of disease are now required.
Collapse
MESH Headings
- Aged
- Angiotensin-Converting Enzyme 2/genetics
- Angiotensin-Converting Enzyme 2/metabolism
- Basigin/genetics
- Basigin/metabolism
- COVID-19/diagnosis
- COVID-19/genetics
- COVID-19/metabolism
- COVID-19/physiopathology
- Case-Control Studies
- Female
- Forced Expiratory Volume
- Gene Expression Regulation
- Humans
- Lung/metabolism
- Lung/physiopathology
- Male
- Middle Aged
- Prognosis
- Pulmonary Disease, Chronic Obstructive/diagnosis
- Pulmonary Disease, Chronic Obstructive/genetics
- Pulmonary Disease, Chronic Obstructive/metabolism
- Pulmonary Disease, Chronic Obstructive/physiopathology
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
- Transcriptome
- Vital Capacity
Collapse
Affiliation(s)
- Alastair Watson
- Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Lisa Öberg
- Translational Science and Experimental Medicine, Research and Early Development, AstraZeneca, Respiratory & Immunology, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Bastian Angermann
- Translational Science and Experimental Medicine, Research and Early Development, AstraZeneca, Respiratory & Immunology, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - C Mirella Spalluto
- Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Michael Hühn
- Translational Science and Experimental Medicine, Research and Early Development, AstraZeneca, Respiratory & Immunology, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Hannah Burke
- Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Doriana Cellura
- Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Anna Freeman
- Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Daniel Muthas
- Translational Science and Experimental Medicine, Research and Early Development, AstraZeneca, Respiratory & Immunology, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Damla Etal
- Translational Genomics, Discovery Biology, Discovery Sciences, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Graham Belfield
- Translational Genomics, Discovery Biology, Discovery Sciences, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Fredrik Karlsson
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Karl Nordström
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Kris Ostridge
- Faculty of Medicine, University of Southampton, Southampton, UK
- Translational Science and Experimental Medicine, Research and Early Development, AstraZeneca, Respiratory & Immunology, BioPharmaceuticals R&D, Gothenburg, Sweden
- Clinical Development, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Karl J Staples
- Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Tom Wilkinson
- Faculty of Medicine, University of Southampton, Southampton, UK.
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK.
| |
Collapse
|
28
|
Barber C, Lau L, Ward JA, Daniels T, Watson A, Staples KJ, Wilkinson TMA, Howarth PH. Sputum processing by mechanical dissociation: A rapid alternative to traditional sputum assessment approaches. CLINICAL RESPIRATORY JOURNAL 2021; 15:800-807. [PMID: 33749082 DOI: 10.1111/crj.13365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Sputum cytology is currently the gold standard to evaluate cellular inflammation in the airways and phenotyping patients with airways diseases. Sputum eosinophil proportions have been used to guide treatment for moderate to severe asthma. Furthermore, raised sputum neutrophils are associated with poor disease control and impaired lung function in both asthma and COPD and small airways disease in cystic fibrosis. However, induced-sputum analysis is subjective and resource heavy, requiring dedicated specialist processing and assessment; this limits its utility in most clinical settings. Indirect blood eosinophil measures have been adopted in clinical care. However, there are currently no good peripheral blood biomarkers of airway neutrophils. A resource-light sputum processing approach could thus help integrate induced sputum more readily into routine clinical care. New mechanical disruption (MD) methods can rapidly obtain viable single cell suspensions from sputum samples. AIMS The aim of this study was to compare MD sputum processing to traditional methods for cell viability, granulocyte proportions and sputum cytokine analysis. METHODS Sputum plugs were split and processed using traditional methods and the MD method, and samples were then compared. RESULTS The MD method produced a homogeneous cell suspension in 62 s; 70 min faster than the standard method used. No significant difference was seen between the cell viability (p = 0.09), or the concentration of eosinophils (p = 0.83), neutrophils (p = 0.99) or interleukin-8 (p = 0.86) using MD. CONCLUSION This cost-effective method of sputum processing could provide a more pragmatic, sustainable means of directly monitoring the airway milieu. Therefore, we recommend this method be taken forward for further investigation.
Collapse
Affiliation(s)
- Clair Barber
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton, UK
| | - Laurie Lau
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Jonathan A Ward
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Thomas Daniels
- Cystic Fibrosis Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Alastair Watson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton, UK
| | - Karl J Staples
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton, UK
| | - Tom M A Wilkinson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton, UK
| | - Peter H Howarth
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Cystic Fibrosis Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Respiratory Medicine, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
29
|
Page LK, Staples KJ, Spalluto CM, Watson A, Wilkinson TMA. Influence of Hypoxia on the Epithelial-Pathogen Interactions in the Lung: Implications for Respiratory Disease. Front Immunol 2021; 12:653969. [PMID: 33868294 PMCID: PMC8044850 DOI: 10.3389/fimmu.2021.653969] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Under normal physiological conditions, the lung remains an oxygen rich environment. However, prominent regions of hypoxia are a common feature of infected and inflamed tissues and many chronic inflammatory respiratory diseases are associated with mucosal and systemic hypoxia. The airway epithelium represents a key interface with the external environment and is the first line of defense against potentially harmful agents including respiratory pathogens. The protective arsenal of the airway epithelium is provided in the form of physical barriers, and the production of an array of antimicrobial host defense molecules, proinflammatory cytokines and chemokines, in response to activation by receptors. Dysregulation of the airway epithelial innate immune response is associated with a compromised immunity and chronic inflammation of the lung. An increasing body of evidence indicates a distinct role for hypoxia in the dysfunction of the airway epithelium and in the responses of both innate immunity and of respiratory pathogens. Here we review the current evidence around the role of tissue hypoxia in modulating the host-pathogen interaction at the airway epithelium. Furthermore, we highlight the work needed to delineate the role of tissue hypoxia in the pathophysiology of chronic inflammatory lung diseases such as asthma, cystic fibrosis, and chronic obstructive pulmonary disease in addition to novel respiratory diseases such as COVID-19. Elucidating the molecular mechanisms underlying the epithelial-pathogen interactions in the setting of hypoxia will enable better understanding of persistent infections and complex disease processes in chronic inflammatory lung diseases and may aid the identification of novel therapeutic targets and strategies.
Collapse
Affiliation(s)
- Lee K. Page
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
| | - Karl J. Staples
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - C. Mirella Spalluto
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - Alastair Watson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
- Birmingham Medical School, University of Birmingham, Birmingham, United Kingdom
| | - Tom M. A. Wilkinson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| |
Collapse
|
30
|
Ray E, Culliford D, Kruk H, Gillett K, North M, Astles CM, Hicks A, Johnson M, Lin SX, Orlando R, Thomas M, Jordan RE, Price D, Konstantin M, Wilkinson TMA. Specialist respiratory outreach: a case-finding initiative for identifying undiagnosed COPD in primary care. NPJ Prim Care Respir Med 2021; 31:7. [PMID: 33574260 PMCID: PMC7878732 DOI: 10.1038/s41533-021-00219-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 01/06/2021] [Indexed: 12/18/2022] Open
Abstract
COPD remains largely undiagnosed or is diagnosed late in the course of disease. We report findings of a specialist outreach programme to identify undiagnosed COPD in primary care. An electronic case-finding algorithm identified 1602 at-risk patients from 12 practices who were invited to attend the clinic. Three hundred and eighty-three (23.9%) responded and 288 were enrolled into the study. Forty-eight (16.6%) had undiagnosed mild and 28 (9.7%) had moderate airway obstruction, meeting spirometric diagnostic criteria for COPD. However, at 12 months only 8 suspected COPD patients (10.6%) had received a diagnostic label in their primary care record. This constituted 0.38% of the total patient population, as compared with 0.31% of control practices, p = 0.306. However, if all patients with airway obstruction received a coding of COPD, then the diagnosis rate in the intervention group would have risen by 0.84%. Despite the low take-up and diagnostic yield, this programme suggests that integrated case-finding strategies could improve COPD recognition.
Collapse
Affiliation(s)
- Emma Ray
- NIHR ARC Wessex, Faculty of Health Sciences, University of Southampton, Southampton, UK
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - David Culliford
- NIHR ARC Wessex, Faculty of Health Sciences, University of Southampton, Southampton, UK
| | - Helen Kruk
- NIHR ARC Wessex, Faculty of Health Sciences, University of Southampton, Southampton, UK
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Kate Gillett
- NIHR ARC Wessex, Faculty of Health Sciences, University of Southampton, Southampton, UK
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Mal North
- NIHR ARC Wessex, Faculty of Health Sciences, University of Southampton, Southampton, UK
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Carla M Astles
- NIHR ARC Wessex, Faculty of Health Sciences, University of Southampton, Southampton, UK
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Alexander Hicks
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Matthew Johnson
- NIHR ARC Wessex, Faculty of Health Sciences, University of Southampton, Southampton, UK
| | - Sharon Xiaowen Lin
- NIHR ARC Wessex, Faculty of Health Sciences, University of Southampton, Southampton, UK
| | - Rosanna Orlando
- NIHR ARC Wessex, Faculty of Health Sciences, University of Southampton, Southampton, UK
| | - Mike Thomas
- Department of Primary Care and Population Sciences, University of Southampton, Southampton, UK
| | - Rachel E Jordan
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - David Price
- Observational and Pragmatic Research Institute, Singapore, Singapore
- Centre of Academic Primary Care, Division of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - Mita Konstantin
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Tom M A Wilkinson
- Faculty of Medicine, University of Southampton, Southampton, UK.
- NIHR Biomedical Research Centre, University Hospitals Southampton NHS Foundation Trust, Southampton, UK.
- Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK.
| |
Collapse
|
31
|
Rose D, Ray E, Summers RH, Taylor M, Kruk H, North M, Gillett K, Thomas M, Wilkinson TMA. Case-finding for COPD clinic acceptability to patients in GPs across Hampshire: a qualitative study. NPJ Prim Care Respir Med 2021; 31:4. [PMID: 33542246 PMCID: PMC7862661 DOI: 10.1038/s41533-021-00216-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 11/17/2020] [Indexed: 11/23/2022] Open
Abstract
Despite high mortality and morbidity, COPD remains under-diagnosed. Case-finding strategies are possible, but patients' perspectives are unexplored. Using qualitative methods, we explored the patient perspective of a case-finding intervention among at-risk patients in primary care. Semi-structured telephone interviews were transcribed and thematic analysis utilised. Seven patients without (mean age 64.5 years (58-74), n = 4) and 8 with obstructed spirometry (mean age 63.5 (53-75), n = 4) were interviewed. Themes identified were motives, challenges and concerns regarding attending the clinic. These included wanting to be well; to help with research; concern over negative impact to life from COPD diagnosis; perceived utility of the clinic; quality of information given; staff manner, approachability and knowledge; and perceived effects of the clinic on lifestyle, self-management and symptoms. The intervention was generally deemed useful and reassuring, although shared information was too detailed or irrelevant for some. Several reported positive lifestyle changes, improved symptoms and improved self-management.
Collapse
Affiliation(s)
| | - Emma Ray
- NIHR ARC Wessex, Faculty of Health Sciences, University of Southampton, Southampton, UK
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Rachael H Summers
- Faculty of Health Sciences, University of Southampton, Southampton, UK
| | - Melinda Taylor
- NIHR ARC Wessex, Faculty of Health Sciences, University of Southampton, Southampton, UK
| | - Helen Kruk
- NIHR ARC Wessex, Faculty of Health Sciences, University of Southampton, Southampton, UK
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Mal North
- NIHR ARC Wessex, Faculty of Health Sciences, University of Southampton, Southampton, UK
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Kate Gillett
- NIHR ARC Wessex, Faculty of Health Sciences, University of Southampton, Southampton, UK
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Mike Thomas
- Department of Primary Care and Population Sciences, University of Southampton, Southampton, UK
| | - Tom M A Wilkinson
- Faculty of Medicine, University of Southampton, Southampton, UK.
- NIHR Biomedical Research Centre, University Hospitals Southampton NHS Foundation Trust, Southampton, UK.
- Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK.
| |
Collapse
|
32
|
McCrae C, Olsson M, Gustafson P, Malmgren A, Aurell M, Fagerås M, Da Silva CA, Cavallin A, Paraskos J, Karlsson K, Wingren C, Monk P, Marsden R, Harrison T. INEXAS: A Phase 2 Randomized Trial of On-demand Inhaled Interferon Beta-1a in Severe Asthmatics. Clin Exp Allergy 2021; 51:273-283. [PMID: 33091192 PMCID: PMC7984268 DOI: 10.1111/cea.13765] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/18/2020] [Accepted: 10/17/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Upper respiratory tract infections (URTIs) are important triggers for asthma exacerbations. We hypothesized that inhalation of the anti-viral cytokine, interferon (IFN)-β, during URTI, could prevent these exacerbations. OBJECTIVE To evaluate the efficacy of on-demand inhaled IFN-β1a (AZD9412) to prevent severe asthma exacerbations following symptomatic URTI. METHODS This was a randomized, double-blind, placebo-controlled trial in which patients with severe asthma (GINA 4-5; n = 121) reporting URTI symptoms were randomized to 14 days of once-daily nebulized AZD9412 or placebo. The primary endpoint was severe exacerbations during treatment. Secondary endpoints included 6-item asthma control questionnaire (ACQ-6) and lung function. Exploratory biomarkers included IFN-response markers in serum and sputum, blood leucocyte counts and serum inflammatory cytokines. RESULTS Following a pre-planned interim analysis, the trial was terminated early due to an unexpectedly low exacerbation rate. Asthma worsenings were generally mild and tended to peak at randomization, possibly contributing to the lack of benefit of AZD9412 on other asthma endpoints. Numerically, AZD9412 did not reduce severe exacerbation rate, ACQ-6, asthma symptom scores or reliever medication use. AZD9412 improved lung function (morning peak expiratory flow; mPEF) by 19.7 L/min. Exploratory post hoc analyses indicated a greater mPEF improvement by AZD9412 in patients with high blood eosinophils (>0.3 × 109 /L) at screening and low serum interleukin-18 relative change at pre-treatment baseline. Pharmacodynamic effect of AZD9412 was confirmed using IFN-response markers. CONCLUSIONS & CLINICAL RELEVANCE Colds did not have the impact on asthma patients that was expected and, due to the low exacerbation rate, the trial was stopped early. On-demand AZD9412 treatment did not numerically reduce the number of exacerbations, but did attenuate URTI-induced worsening of mPEF. Severe asthma patients with high blood eosinophils or low serum interleukin-18 response are potential subgroups for further investigation of inhaled IFN-β1a.
Collapse
Affiliation(s)
- Christopher McCrae
- Translational Science and Experimental Medicine, Research and Early DevelopmentRespiratory & Immunology, BioPharmaceuticals R&DAstraZenecaGaithersburgMarylandUSA
- Krefting Research CentreDepartment of Internal Medicine and Clinical NutritionInstitute of MedicineUniversity of GothenburgGothenburgSweden
| | - Marita Olsson
- Early Biometrics and Statistical InnovationData Science and AI, BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Per Gustafson
- BioPharmaceutical MedicalAstraZenecaGothenburgSweden
| | - Anna Malmgren
- Early Respiratory & Immunology Projects DepartmentBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Malin Aurell
- Early Respiratory & Immunology Clinical DevelopmentBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Malin Fagerås
- BioPharmaceutical MedicalAstraZenecaGothenburgSweden
| | - Carla A. Da Silva
- Early Respiratory & Immunology Clinical DevelopmentBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Anders Cavallin
- Translational Science and Experimental MedicineEarly Cardiovascular, Renal and Metabolism (CVRM)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Jonathan Paraskos
- Point of Care Diagnostics, Precision MedicineOncology R&DAstraZenecaCambridgeUK
| | - Karin Karlsson
- Translational Science and Experimental MedicineEarly Cardiovascular, Renal and Metabolism (CVRM)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Cecilia Wingren
- Translational Science and Experimental Medicine, Research and Early DevelopmentRespiratory & Immunology, BioPharmaceuticals R&DAstraZenecaGaithersburgMarylandUSA
| | - Phillip Monk
- Synairgen Research LtdSouthampton University HospitalSouthamptonUK
| | - Richard Marsden
- Synairgen Research LtdSouthampton University HospitalSouthamptonUK
| | - Tim Harrison
- Nottingham NIHR Biomedical Research CentreUniversity of NottinghamNottingham City HospitalNottinghamUK
| |
Collapse
|
33
|
Watson A, Madsen J, Clark HW. SP-A and SP-D: Dual Functioning Immune Molecules With Antiviral and Immunomodulatory Properties. Front Immunol 2021; 11:622598. [PMID: 33542724 PMCID: PMC7851053 DOI: 10.3389/fimmu.2020.622598] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/14/2020] [Indexed: 01/08/2023] Open
Abstract
Surfactant proteins A (SP-A) and D (SP-D) are soluble innate immune molecules which maintain lung homeostasis through their dual roles as anti-infectious and immunomodulatory agents. SP-A and SP-D bind numerous viruses including influenza A virus, respiratory syncytial virus (RSV) and human immunodeficiency virus (HIV), enhancing their clearance from mucosal points of entry and modulating the inflammatory response. They also have diverse roles in mediating innate and adaptive cell functions and in clearing apoptotic cells, allergens and other noxious particles. Here, we review how the properties of these first line defense molecules modulate inflammatory responses, as well as host-mediated immunopathology in response to viral infections. Since SP-A and SP-D are known to offer protection from viral and other infections, if their levels are decreased in some disease states as they are in severe asthma and chronic obstructive pulmonary disease (COPD), this may confer an increased risk of viral infection and exacerbations of disease. Recombinant molecules of SP-A and SP-D could be useful in both blocking respiratory viral infection while also modulating the immune system to prevent excessive inflammatory responses seen in, for example, RSV or coronavirus disease 2019 (COVID-19). Recombinant SP-A and SP-D could have therapeutic potential in neutralizing both current and future strains of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus as well as modulating the inflammation-mediated pathology associated with COVID-19. A recombinant fragment of human (rfh)SP-D has recently been shown to neutralize SARS-CoV-2. Further work investigating the potential therapeutic role of SP-A and SP-D in COVID-19 and other infectious and inflammatory diseases is indicated.
Collapse
Affiliation(s)
- Alastair Watson
- Clinical and Experimental Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, United Kingdom
- Birmingham Medical School, University of Birmingham, Birmingham, United Kingdom
| | - Jens Madsen
- Neonatology, EGA Institute for Women’s Health, Faculty of Population Health Sciences, University College London, London, United Kingdom
| | - Howard William Clark
- Neonatology, EGA Institute for Women’s Health, Faculty of Population Health Sciences, University College London, London, United Kingdom
- NIHR Biomedical Research Centre, University College London Hospital (UCLH), University College London (UCL), London, United Kingdom
| |
Collapse
|
34
|
Gao X, Chan PKS, Lui GCY, Hui DSC, Chu IMT, Sun X, Tsang MSM, Chan BCL, Lam CWK, Wong CK. Interleukin-38 ameliorates poly(I:C) induced lung inflammation: therapeutic implications in respiratory viral infections. Cell Death Dis 2021; 12:53. [PMID: 33414457 PMCID: PMC7790341 DOI: 10.1038/s41419-020-03283-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022]
Abstract
Interleukin-38 has recently been shown to have anti-inflammatory properties in lung inflammatory diseases. However, the effects of IL-38 in viral pneumonia remains unknown. In the present study, we demonstrate that circulating IL-38 concentrations together with IL-36α increased significantly in influenza and COVID-19 patients, and the level of IL-38 and IL-36α correlated negatively and positively with disease severity and inflammation, respectively. In the co-cultured human respiratory epithelial cells with macrophages to mimic lung microenvironment in vitro, IL-38 was able to alleviate inflammatory responses by inhibiting poly(I:C)-induced overproduction of pro-inflammatory cytokines and chemokines through intracellular STAT1, STAT3, p38 MAPK, ERK1/2, MEK, and NF-κB signaling pathways. Intriguingly, transcriptomic profiling revealed that IL-38 targeted genes were associated with the host innate immune response to virus. We also found that IL-38 counteracts the biological processes induced by IL-36α in the co-culture. Furthermore, the administration of recombinant IL-38 could mitigate poly I:C-induced lung injury, with reduced early accumulation of neutrophils and macrophages in bronchoalveolar lavage fluid, activation of lymphocytes, production of pro-inflammatory cytokines and chemokines and permeability of the alveolar-epithelial barrier. Taken together, our study indicates that IL-38 plays a crucial role in protection from exaggerated pulmonary inflammation during poly(I:C)-induced pneumonia, thereby providing the basis of a novel therapeutic target for respiratory viral infections.
Collapse
Affiliation(s)
- Xun Gao
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Paul Kay Sheung Chan
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, China.,Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong, China
| | - Grace Chung Yan Lui
- Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong, China.,Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - David Shu Cheong Hui
- Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong, China.,Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Ida Miu-Ting Chu
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoyu Sun
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Miranda Sin-Man Tsang
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Ben Chung Lap Chan
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Christopher Wai-Kei Lam
- Faculty of Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Chun-Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China. .,Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China. .,Li Dak Sum Yip Yio Chin R & D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
35
|
Watson A, Wilkinson TMA. Respiratory viral infections in the elderly. Ther Adv Respir Dis 2021; 15:1753466621995050. [PMID: 33749408 PMCID: PMC7989115 DOI: 10.1177/1753466621995050] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
With the global over 60-year-old population predicted to more than double over the next 35 years, caring for this aging population has become a major global healthcare challenge. In 2016 there were over 1 million deaths in >70 year olds due to lower respiratory tract infections; 13-31% of these have been reported to be caused by viruses. Since then, there has been a global COVID-19 pandemic, which has caused over 2.3 million deaths so far; increased age has been shown to be the biggest risk factor for morbidity and mortality. Thus, the burden of respiratory viral infections in the elderly is becoming an increasing unmet clinical need. Particular challenges are faced due to the interplay of a variety of factors including complex multimorbidities, decreased physiological reserve and an aging immune system. Moreover, their atypical presentation of symptoms may lead to delayed necessary care, prescription of additional drugs and prolonged hospital stay. This leads to morbidity and mortality and further nosocomial spread. Clinicians currently have limited access to sensitive detection methods. Furthermore, a lack of effective antiviral treatments means there is little incentive to diagnose and record specific non-COVID-19 viral infections. To meet this unmet clinical need, it is first essential to fully understand the burden of respiratory viruses in the elderly. Doing this through prospective screening research studies for all respiratory viruses will help guide preventative policies and clinical trials for emerging therapeutics. The implementation of multiplex point-of-care diagnostics as a mainstay in all healthcare settings will be essential to understand the burden of respiratory viruses, diagnose patients and monitor outbreaks. The further development of novel targeted vaccinations as well as anti-viral therapeutics and new ways to augment the aging immune system is now also essential.The reviews of this paper are available via the supplemental material section.
Collapse
Affiliation(s)
- Alastair Watson
- Faculty of Medicine, Clinical & Experimental Sciences, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Birmingham Medical School, University of Birmingham, Birmingham, UK
| | - Tom M. A. Wilkinson
- Faculty of Medicine, Clinical and Experimental Sciences, Southampton University, Mailpoint 810, Level F, South Block, Southampton General Hospital, Southampton, Hampshire, SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
36
|
Kazakov AS, Sofin AD, Avkhacheva NV, Denesyuk AI, Deryusheva EI, Rastrygina VA, Sokolov AS, Permyakova ME, Litus EA, Uversky VN, Permyakov EA, Permyakov SE. Interferon Beta Activity Is Modulated via Binding of Specific S100 Proteins. Int J Mol Sci 2020; 21:ijms21249473. [PMID: 33322098 PMCID: PMC7764042 DOI: 10.3390/ijms21249473] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 12/18/2022] Open
Abstract
Interferon-β (IFN-β) is a pleiotropic cytokine used for therapy of multiple sclerosis, which is also effective in suppression of viral and bacterial infections and cancer. Recently, we reported a highly specific interaction between IFN-β and S100P lowering IFN-β cytotoxicity to cancer cells (Int J Biol Macromol. 2020; 143: 633–639). S100P is a member of large family of multifunctional Ca2+-binding proteins with cytokine-like activities. To probe selectivity of IFN-β—S100 interaction with respect to S100 proteins, we used surface plasmon resonance spectroscopy, chemical crosslinking, and crystal violet assay. Among the thirteen S100 proteins studied S100A1, S100A4, and S100A6 proteins exhibit strictly Ca2+-dependent binding to IFN-β with equilibrium dissociation constants, Kd, of 0.04–1.5 µM for their Ca2+-bound homodimeric forms. Calcium depletion abolishes the S100—IFN-β interactions. Monomerization of S100A1/A4/A6 decreases Kd values down to 0.11–1.0 nM. Interferon-α is unable of binding to the S100 proteins studied. S100A1/A4 proteins inhibit IFN-β-induced suppression of MCF-7 cells viability. The revealed direct influence of specific S100 proteins on IFN-β activity uncovers a novel regulatory role of particular S100 proteins, and opens up novel approaches to enhancement of therapeutic efficacy of IFN-β.
Collapse
Affiliation(s)
- Alexey S. Kazakov
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya str., 7, 142290 Pushchino, Russia; (A.S.K.); (A.D.S.); (N.V.A.); (A.I.D.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Alexander D. Sofin
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya str., 7, 142290 Pushchino, Russia; (A.S.K.); (A.D.S.); (N.V.A.); (A.I.D.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Nadezhda V. Avkhacheva
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya str., 7, 142290 Pushchino, Russia; (A.S.K.); (A.D.S.); (N.V.A.); (A.I.D.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Alexander I. Denesyuk
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya str., 7, 142290 Pushchino, Russia; (A.S.K.); (A.D.S.); (N.V.A.); (A.I.D.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland
| | - Evgenia I. Deryusheva
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya str., 7, 142290 Pushchino, Russia; (A.S.K.); (A.D.S.); (N.V.A.); (A.I.D.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Victoria A. Rastrygina
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya str., 7, 142290 Pushchino, Russia; (A.S.K.); (A.D.S.); (N.V.A.); (A.I.D.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Andrey S. Sokolov
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya str., 7, 142290 Pushchino, Russia; (A.S.K.); (A.D.S.); (N.V.A.); (A.I.D.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Maria E. Permyakova
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya str., 7, 142290 Pushchino, Russia; (A.S.K.); (A.D.S.); (N.V.A.); (A.I.D.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Ekaterina A. Litus
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya str., 7, 142290 Pushchino, Russia; (A.S.K.); (A.D.S.); (N.V.A.); (A.I.D.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Vladimir N. Uversky
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya str., 7, 142290 Pushchino, Russia; (A.S.K.); (A.D.S.); (N.V.A.); (A.I.D.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Correspondence: (V.N.U.); (S.E.P.); Tel.: +7-(495)-143-7741 (S.E.P.); Fax: +7-(4967)-33-05-22 (S.E.P.)
| | - Eugene A. Permyakov
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya str., 7, 142290 Pushchino, Russia; (A.S.K.); (A.D.S.); (N.V.A.); (A.I.D.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
| | - Sergei E. Permyakov
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institutskaya str., 7, 142290 Pushchino, Russia; (A.S.K.); (A.D.S.); (N.V.A.); (A.I.D.); (E.I.D.); (V.A.R.); (A.S.S.); (M.E.P.); (E.A.L.); (E.A.P.)
- Correspondence: (V.N.U.); (S.E.P.); Tel.: +7-(495)-143-7741 (S.E.P.); Fax: +7-(4967)-33-05-22 (S.E.P.)
| |
Collapse
|
37
|
Day K, Ostridge K, Conway J, Cellura D, Watson A, Spalluto CM, Staples KJ, Thompson B, Wilkinson T. Interrelationships Among Small Airways Dysfunction, Neutrophilic Inflammation, and Exacerbation Frequency in COPD. Chest 2020; 159:1391-1399. [PMID: 33245876 DOI: 10.1016/j.chest.2020.11.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 11/03/2020] [Accepted: 11/08/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Small airways disease (SAD) is a key component of COPD and is a main contributing factor to lung function decline. RESEARCH QUESTION Is SAD a key feature of frequent COPD exacerbators and is this related to airway inflammation? STUDY DESIGN AND METHODS Thirty-nine COPD patients defined as either frequent exacerbator (FE) group (≥ 2 exacerbations/y; n = 17) and infrequent exacerbator (IFE) group (≤ 1 exacerbation/y; n = 22) underwent the forced oscillation technique (resistance at 5 Hz minus 19 Hz [R5-R19], area of reactance [AX]), multiple breath nitrogen washout (conducting airways ventilation heterogeneity, acinar ventilation heterogeneity [Sacin]), plethysmography (ratio of residual volume to total lung capacity), single-breath transfer factor of the lung for carbon monoxide, spirometry (FEV1, FEV1/FVC), and paired inspiratory-expiratory CT scans to ascertain SAD. A subpopulation underwent bronchoscopy to enable enumeration of BAL cell proportions. RESULTS Sacin was significantly higher in the COPD FE group compared with the IFE group (P = .027). In the FE group, markers of SAD were associated strongly with BAL neutrophil proportions, R5-R19 (P = .001, r = 0.795), AX (P = .049, ρ = 0.560), residual volume to total lung capacity ratio (P = .004, r = 0.730), and the mean lung density of the paired CT scans (P = .018, r = 0.639). INTERPRETATION Increased Sacin may be a consequence of previous exacerbations or may highlight a group of patients prone to exacerbations. Measures of SAD were associated strongly with neutrophilic inflammation in the small airways of FE patients, supporting the hypothesis that frequent exacerbations are associated with SAD related to increased cellular inflammation.
Collapse
Affiliation(s)
- Kerry Day
- Faculty of Medicine, University of Southampton, Southampton; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton.
| | - Kristoffer Ostridge
- Faculty of Medicine, University of Southampton, Southampton; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton; Clinical Development, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | | | | | | | - Karl J Staples
- Faculty of Medicine, University of Southampton, Southampton; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton
| | - Bruce Thompson
- Swinburne University of Technology, Melbourne, Australia
| | - Tom Wilkinson
- Faculty of Medicine, University of Southampton, Southampton; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton
| |
Collapse
|
38
|
Ortega H, Nickle D, Carter L. Rhinovirus and asthma: Challenges and opportunities. Rev Med Virol 2020; 31:e2193. [PMID: 33217098 PMCID: PMC8365703 DOI: 10.1002/rmv.2193] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/11/2022]
Abstract
Human rhinoviruses (RVs) are the primary aetiological agent of the common cold. Generally, the associated infection is mild and self‐limiting, but may also be associated with bronchiolitis in infants, pneumonia in the immunocompromised and exacerbation in patients with pulmonary conditions such as asthma or chronic obstructive pulmonary disease. Viral infection accounts for as many as two thirds of asthma exacerbations in children and more than half in adults. Allergy and asthma are major risk factors for more frequent and severe RV‐related illnesses. The prevalence of RV‐induced wheezing will likely continue to increase given that asthma affects a significant proportion of the population, with allergic asthma accounting for the majority. Several new respiratory viruses and their subgroups have been discovered, with various degrees of relevance. This review will focus on RV infection in the context of the epidemiologic evidence, genetic variability, pathobiology, clinical studies in the context of asthma, differences with other viruses including COVID‐19 and current treatment interventions.
Collapse
|
39
|
Monk PD, Marsden RJ, Tear VJ, Brookes J, Batten TN, Mankowski M, Gabbay FJ, Davies DE, Holgate ST, Ho LP, Clark T, Djukanovic R, Wilkinson TMA. Safety and efficacy of inhaled nebulised interferon beta-1a (SNG001) for treatment of SARS-CoV-2 infection: a randomised, double-blind, placebo-controlled, phase 2 trial. THE LANCET RESPIRATORY MEDICINE 2020; 9:196-206. [PMID: 33189161 PMCID: PMC7836724 DOI: 10.1016/s2213-2600(20)30511-7] [Citation(s) in RCA: 309] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection carries a substantial risk of severe and prolonged illness; treatment options are currently limited. We assessed the efficacy and safety of inhaled nebulised interferon beta-1a (SNG001) for the treatment of patients admitted to hospital with COVID-19. METHODS We did a randomised, double-blind, placebo-controlled, phase 2 pilot trial at nine UK sites. Adults aged 18 years or older and admitted to hospital with COVID-19 symptoms, with a positive RT-PCR or point-of-care test, or both, were randomly assigned (1:1) to receive SNG001 (6 MIU) or placebo by inhalation via a mouthpiece daily for 14 days. The primary outcome was the change in clinical condition on the WHO Ordinal Scale for Clinical Improvement (OSCI) during the dosing period in the intention-to-treat population (all randomised patients who received at least one dose of the study drug). The OSCI is a 9-point scale, where 0 corresponds to no infection and 8 corresponds to death. Multiple analyses were done to identify the most suitable statistical method for future clinical trials. Safety was assessed by monitoring adverse events for 28 days. This trial is registered with Clinicaltrialsregister.eu (2020-001023-14) and ClinicalTrials.gov (NCT04385095); the pilot trial of inpatients with COVID-19 is now completed. FINDINGS Between March 30 and May 30, 2020, 101 patients were randomly assigned to SNG001 (n=50) or placebo (n=51). 48 received SNG001 and 50 received placebo and were included in the intention-to-treat population. 66 (67%) patients required oxygen supplementation at baseline: 29 in the placebo group and 37 in the SNG001 group. Patients receiving SNG001 had greater odds of improvement on the OSCI scale (odds ratio 2·32 [95% CI 1·07-5·04]; p=0·033) on day 15 or 16 and were more likely than those receiving placebo to recover to an OSCI score of 1 (no limitation of activities) during treatment (hazard ratio 2·19 [95% CI 1·03-4·69]; p=0·043). SNG001 was well tolerated. The most frequently reported treatment-emergent adverse event was headache (seven [15%] patients in the SNG001 group and five [10%] in the placebo group). There were three deaths in the placebo group and none in the SNG001 group. INTERPRETATION Patients who received SNG001 had greater odds of improvement and recovered more rapidly from SARS-CoV-2 infection than patients who received placebo, providing a strong rationale for further trials. FUNDING Synairgen Research.
Collapse
Affiliation(s)
- Phillip D Monk
- Synairgen Research, Southampton General Hospital, Southampton, UK
| | | | - Victoria J Tear
- Synairgen Research, Southampton General Hospital, Southampton, UK
| | - Jody Brookes
- Synairgen Research, Southampton General Hospital, Southampton, UK
| | | | | | | | - Donna E Davies
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton, UK
| | - Stephen T Holgate
- Synairgen Research, Southampton General Hospital, Southampton, UK; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton, UK
| | - Ling-Pei Ho
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, UK
| | - Tristan Clark
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton, UK
| | - Ratko Djukanovic
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton, UK
| | - Tom M A Wilkinson
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton, UK.
| | | |
Collapse
|
40
|
North M, Bourne S, Green B, Chauhan AJ, Brown T, Winter J, Jones T, Neville D, Blythin A, Watson A, Johnson M, Culliford D, Elkes J, Cornelius V, Wilkinson TMA. A randomised controlled feasibility trial of E-health application supported care vs usual care after exacerbation of COPD: the RESCUE trial. NPJ Digit Med 2020; 3:145. [PMID: 33145441 PMCID: PMC7603326 DOI: 10.1038/s41746-020-00347-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 09/17/2020] [Indexed: 01/17/2023] Open
Abstract
Exacerbations of COPD are one of the commonest causes of admission and readmission to hospital. The role of digital interventions to support self-management in improving outcomes is uncertain. We conducted an open, randomised controlled trial of a digital health platform application (app) in 41 COPD patients recruited following hospital admission with an acute exacerbation. Subjects were randomised to either receive usual care, including a written self-management plan (n = 21), or the myCOPD app (n = 20) for 90 days. The primary efficacy outcome was recovery rate of symptoms measured by COPD assessment test (CAT) score. Exacerbations, readmission, inhaler technique quality of life and patient activation (PAM) scores were also captured by a blinded team. The app was acceptable in this care setting and was used by 17 of the 20 patients with sustained use over the study period. The treatment effect on the CAT score was 4.49 (95% CI: -8.41, -0.58) points lower in the myCOPD arm. Patients' inhaler technique improved in the digital intervention arm (101 improving to 20 critical errors) compared to usual care (100 to 72 critical errors). Exacerbations tended to be less frequent in the digital arm compared to usual care; 34 vs 18 events. Hospital readmissions risk was numerically lower in the digital intervention arm: OR for readmission 0.383 (95% CI: 0.074, 1.987; n = 35). In this feasibility study of the digital self-management platform myCOPD, the app has proven acceptable to patients to use and use has improved exacerbation recovery rates, with strong signals of lower re-exacerbation and readmission rates over 90 days. myCOPD reduced the number of critical errors in inhaler technique compared to usual care with written self-management. This provides a strong basis for further exploration of the use of app interventions in the context of recently hospitalised patients with COPD and informs the potential design of a large multi-centre trial.
Collapse
Affiliation(s)
| | | | - Ben Green
- Portsmouth Hospitals NHS Trust, Portsmouth, UK
| | | | - Tom Brown
- Portsmouth Hospitals NHS Trust, Portsmouth, UK
| | | | - Tom Jones
- Portsmouth Hospitals NHS Trust, Portsmouth, UK
| | - Dan Neville
- Portsmouth Hospitals NHS Trust, Portsmouth, UK
| | | | - Alastair Watson
- NIHR ARC Wessex, University of Southampton, Southampton, UK
- University of Southampton Faculty of Medicine, Southampton, UK
| | | | | | | | | | - Tom M. A. Wilkinson
- my mhealth Limited, Bournemouth, UK
- NIHR ARC Wessex, University of Southampton, Southampton, UK
- University of Southampton Faculty of Medicine, Southampton, UK
| |
Collapse
|
41
|
Farne H, Kumar K, Ritchie AI, Finney LJ, Johnston SL, Singanayagam A. Repurposing Existing Drugs for the Treatment of COVID-19. Ann Am Thorac Soc 2020; 17:1186-1194. [PMID: 32692580 PMCID: PMC7640626 DOI: 10.1513/annalsats.202005-566fr] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
The rapid global spread and significant mortality associated with the coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral infection has spurred an urgent race to find effective treatments. Repurposing existing drugs is a particularly attractive approach as pharmacokinetic and safety data already exist; thus, development can leapfrog straight to clinical trials of efficacy, generating results far more quickly than de novo drug development. This review summarizes the state of play for the principle drugs identified as candidates to be repurposed for treating COVID-19 grouped by broad mechanism of action: antiviral, immune enhancing, and antiinflammatory or immunomodulatory. Patient selection, particularly with regard to disease stage, is likely to be key. To date, only dexamethasone and remdesivir have been shown to be effective, but several other promising candidates are in trials.
Collapse
Affiliation(s)
- Hugo Farne
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kartik Kumar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Andrew I Ritchie
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Lydia J Finney
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sebastian L Johnston
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Aran Singanayagam
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
42
|
Burke H, Freeman A, Cellura DC, Stuart BL, Brendish NJ, Poole S, Borca F, Phan HTT, Sheard N, Williams S, Spalluto CM, Staples KJ, Clark TW, Wilkinson TMA. Inflammatory phenotyping predicts clinical outcome in COVID-19. Respir Res 2020; 21:245. [PMID: 32962703 PMCID: PMC7506817 DOI: 10.1186/s12931-020-01511-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 09/14/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The COVID-19 pandemic has led to more than 760,000 deaths worldwide (correct as of 16th August 2020). Studies suggest a hyperinflammatory response is a major cause of disease severity and death. Identitfying COVID-19 patients with hyperinflammation may identify subgroups who could benefit from targeted immunomodulatory treatments. Analysis of cytokine levels at the point of diagnosis of SARS-CoV-2 infection can identify patients at risk of deterioration. METHODS We used a multiplex cytokine assay to measure serum IL-6, IL-8, TNF, IL-1β, GM-CSF, IL-10, IL-33 and IFN-γ in 100 hospitalised patients with confirmed COVID-19 at admission to University Hospital Southampton (UK). Demographic, clinical and outcome data were collected for analysis. RESULTS Age > 70 years was the strongest predictor of death (OR 28, 95% CI 5.94, 139.45). IL-6, IL-8, TNF, IL-1β and IL-33 were significantly associated with adverse outcome. Clinical parameters were predictive of poor outcome (AUROC 0.71), addition of a combined cytokine panel significantly improved the predictability (AUROC 0.85). In those ≤70 years, IL-33 and TNF were predictive of poor outcome (AUROC 0.83 and 0.84), addition of a combined cytokine panel demonstrated greater predictability of poor outcome than clinical parameters alone (AUROC 0.92 vs 0.77). CONCLUSIONS A combined cytokine panel improves the accuracy of the predictive value for adverse outcome beyond standard clinical data alone. Identification of specific cytokines may help to stratify patients towards trials of specific immunomodulatory treatments to improve outcomes in COVID-19.
Collapse
Affiliation(s)
- H Burke
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, LF13A, South Academic Block, Southampton, SO16 6YD, UK.
- University Hospitals Southampton NHS Foundation Trust, Southampton, UK.
| | - A Freeman
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, LF13A, South Academic Block, Southampton, SO16 6YD, UK
- University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| | - D C Cellura
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, LF13A, South Academic Block, Southampton, SO16 6YD, UK
| | - B L Stuart
- Southampton Clinical Trials Unit, University of Southampton, Southampton, UK
| | - N J Brendish
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, LF13A, South Academic Block, Southampton, SO16 6YD, UK
- University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| | - S Poole
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, LF13A, South Academic Block, Southampton, SO16 6YD, UK
- University Hospitals Southampton NHS Foundation Trust, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - F Borca
- University Hospitals Southampton NHS Foundation Trust, Southampton, UK
- Clinical Informatics Research Unit Faculty of Medicine, University of Southampton, Southampton, UK
| | - H T T Phan
- Southampton Clinical Trials Unit, University of Southampton, Southampton, UK
- Clinical Informatics Research Unit Faculty of Medicine, University of Southampton, Southampton, UK
| | - N Sheard
- University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| | - S Williams
- University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| | - C M Spalluto
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, LF13A, South Academic Block, Southampton, SO16 6YD, UK
| | - K J Staples
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, LF13A, South Academic Block, Southampton, SO16 6YD, UK
- University Hospitals Southampton NHS Foundation Trust, Southampton, UK
- Southampton Clinical Trials Unit, University of Southampton, Southampton, UK
- Wessex Investigational Sciences Hub, University Of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - T W Clark
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, LF13A, South Academic Block, Southampton, SO16 6YD, UK
- University Hospitals Southampton NHS Foundation Trust, Southampton, UK
- Southampton Clinical Trials Unit, University of Southampton, Southampton, UK
- NIHR Post-Doctoral Fellowship Programme, Southampton, UK
| | - T M A Wilkinson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, LF13A, South Academic Block, Southampton, SO16 6YD, UK
- University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
43
|
Bartel S, Deshane J, Wilkinson T, Gabrielsson S. Extracellular Vesicles as Mediators of Cellular Cross Talk in the Lung Microenvironment. Front Med (Lausanne) 2020; 7:326. [PMID: 32850874 PMCID: PMC7417309 DOI: 10.3389/fmed.2020.00326] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/03/2020] [Indexed: 12/27/2022] Open
Abstract
The human lung is a complex tissue subdivided into several regions that differ in size, function, and resident cell types. Despite years of intensive research, we still do not fully understand the cross talk between these different regions and diverse cell populations in the lung and how this is altered in the development of chronic respiratory disease. The discovery of extracellular vesicles (EVs), small membrane vesicles released from cells for intercellular communication, has added another layer of complexity to cellular cross talk in the complex lung microenvironment. EVs from patients with chronic obstructive pulmonary disease, asthma, or sarcoidosis have been shown to carry microRNAs, proteins, and lipids that may contribute to inflammation or tissue degeneration. Here, we summarize the contribution of these small vesicles in the interplay of several different cell types in the lung microenvironment, with a focus on the development of chronic respiratory diseases. Although there are already many studies demonstrating the adverse effects of EVs in the diseased lung, we still have substantial knowledge gaps regarding the concrete role of EV involvement in lung disease, which should be addressed in future studies.
Collapse
Affiliation(s)
- Sabine Bartel
- Department of Pathology and Medical Biology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jessy Deshane
- Pulmonary Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Tom Wilkinson
- Clinical and Experimental Science, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Susanne Gabrielsson
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
44
|
She L, Alanazi HH, Yan L, Brooks EG, Dube PH, Xiang Y, Zhang F, Sun Y, Liu Y, Zhang X, Li XD. Sensing and signaling of immunogenic extracellular RNAs restrain group 2 innate lymphoid cell-driven acute lung inflammation and airway hyperresponsiveness. PLoS One 2020; 15:e0236744. [PMID: 32730309 PMCID: PMC7392318 DOI: 10.1371/journal.pone.0236744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/13/2020] [Indexed: 01/02/2023] Open
Abstract
Repeated exposures to environmental allergens in susceptible individuals drive the development of type 2 inflammatory conditions such as asthma, which have been traditionally considered to be mainly mediated by Th2 cells. However, emerging evidence suggest that a new innate cell type, group 2 innate lymphoid cells (ILC2), plays a central role in initiating and amplifying a type 2 response, even in the absence of adaptive immunity. At present, the regulatory mechanisms for controlling ILC2 activation remain poorly understood. Here we report that respiratory delivery of immunogenic extracellular RNA (exRNAs) derived from RNA- and DNA-virus infected cells, was able to activate a protective response against acute type 2 lung immunopathology and airway hyperresponsiveness (AHR) induced by IL-33 and a fungal allergen, A. flavus, in mice. Mechanistically, we found that the innate immune responses triggered by exRNAs had a potent suppressive effect in vivo on the proliferation and function of ILC2 without the involvement of adaptive immunity. We further provided the loss-of-function genetic evidence that the TLR3- and MAVS-mediated signaling axis is essential for the inhibitory effects of exRNAs in mouse lungs. Thus, our results indicate that the host detection of extracellular immunostimulatory RNAs generated during respiratory viral infections have an important function in the regulation of ILC2-driven acute lung inflammation.
Collapse
Affiliation(s)
- Li She
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hamad H. Alanazi
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Liping Yan
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Edward G. Brooks
- Division of Immunology and Infectious Disease, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States of America
| | - Peter H. Dube
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Yan Xiang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Fushun Zhang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Yilun Sun
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Yong Liu
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Zhang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiao-Dong Li
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
- * E-mail:
| |
Collapse
|
45
|
Ganjian H, Rajput C, Elzoheiry M, Sajjan U. Rhinovirus and Innate Immune Function of Airway Epithelium. Front Cell Infect Microbiol 2020; 10:277. [PMID: 32637363 PMCID: PMC7316886 DOI: 10.3389/fcimb.2020.00277] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Airway epithelial cells, which lines the respiratory mucosa is in direct contact with the environment. Airway epithelial cells are the primary target for rhinovirus and other inhaled pathogens. In response to rhinovirus infection, airway epithelial cells mount both pro-inflammatory responses and antiviral innate immune responses to clear the virus efficiently. Some of the antiviral responses include the expression of IFNs, endoplasmic reticulum stress induced unfolded protein response and autophagy. Airway epithelial cells also recruits other innate immune cells to establish antiviral state and resolve the inflammation in the lungs. In patients with chronic lung disease, these responses may be either defective or induced in excess leading to deficient clearing of virus and sustained inflammation. In this review, we will discuss the mechanisms underlying antiviral innate immunity and the dysregulation of some of these mechanisms in patients with chronic lung diseases.
Collapse
Affiliation(s)
- Haleh Ganjian
- Department of Thoracic Medicine and Surgery, Lewis Katz Medical School, Temple University, Philadelphia, PA, United States
| | - Charu Rajput
- Department of Thoracic Medicine and Surgery, Lewis Katz Medical School, Temple University, Philadelphia, PA, United States
| | - Manal Elzoheiry
- Department of Thoracic Medicine and Surgery, Lewis Katz Medical School, Temple University, Philadelphia, PA, United States
| | - Umadevi Sajjan
- Department of Thoracic Medicine and Surgery, Lewis Katz Medical School, Temple University, Philadelphia, PA, United States
- Department of Physiology, Lewis Katz Medical School, Temple University, Philadelphia, PA, United States
| |
Collapse
|
46
|
Freeman AT, Hill D, Newell C, Moyses H, Azim A, Knight D, Presland L, Harvey M, Haitchi HM, Watson A, Staples KJ, Kurukulaaratchy RJ, Wilkinson TMA. Patient perceived barriers to exercise and their clinical associations in difficult asthma. Asthma Res Pract 2020; 6:5. [PMID: 32537235 PMCID: PMC7285728 DOI: 10.1186/s40733-020-00058-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Exercise is recommended in guidelines for asthma management and has beneficial effects on symptom control, inflammation and lung function in patients with sub-optimally controlled asthma. Despite this, physical activity levels in patients with difficult asthma are often impaired. Understanding the barriers to exercise in people with difficult asthma is crucial for increasing their activity, and in implementing successful, disease modifying, and holistic approaches to improve their health. METHODS 62 Patients within the WATCH Difficult Asthma Cohort (Southampton, UK) completed an Exercise Therapy Burden Questionnaire (ETBQ). The results were analyzed with contemporaneous asthma-related data to determine relationships between perceived exercise barriers and asthma and comorbidity characteristics. RESULTS Patients were reflective of a difficult asthma cohort, 66% were female, and 63% were atopic. They had a high BMI (median [inter-quartile range]) of 29.3 [25.5-36.2], age of 53.5 [38.75, 65.25], impaired spirometry with FEV1 73% predicted [59.5, 86.6%] and FEV/FVC ratio of 72 [56.5, 78.0] and poor symptom control, as defined by an Asthma Control Questionnaire (ACQ6) result of 2.4 [1.28, 3.2]. A high perceived barriers to exercise score was significantly correlated with increased asthma symptoms (r = 0.452, p < 0.0001), anxiety (r = 0.375, p = 0.005) and depression (r = 0.363, p = 0.008), poor quality of life (r = 0.345, p = 0.015) and number of rescue oral steroid courses in the past 12 months (r = 0.257, p = 0.048). Lung function, blood eosinophil count, FeNO, Njimegen and SNOT22 scores, BMI and hospitalisations in the previous year were not related to exercise perceptions. CONCLUSION In difficult asthma, perceived barriers to exercise are related to symptom burden and psychological morbidity. Therefore, exercise interventions combined with psychological input such as CBT to restructure thought processes around these perceived barriers may be useful in facilitating adoption of exercise.
Collapse
Affiliation(s)
- Anna T. Freeman
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
- Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, UK
| | - David Hill
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, UK
| | - Colin Newell
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, UK
| | - Helen Moyses
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, UK
| | - Adnan Azim
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, UK
- Asthma, Allergy and Clinical Immunology Department, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Deborah Knight
- Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, UK
| | - Laura Presland
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, UK
| | - Matthew Harvey
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, UK
| | - Hans Michael Haitchi
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, UK
- Asthma, Allergy and Clinical Immunology Department, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Alastair Watson
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
- Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, UK
| | - Karl J. Staples
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
- Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, UK
| | - Ramesh J. Kurukulaaratchy
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, UK
- Asthma, Allergy and Clinical Immunology Department, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- The David Hide Asthma & Allergy Research Centre, St Mary’s Hospital, Newport, Isle of Wight UK
| | - Tom M. A. Wilkinson
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
- Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
- Southampton NIHR Respiratory Biomedical Research Centre, Southampton General Hospital, Southampton, UK
| |
Collapse
|
47
|
Cafferkey J, Coultas JA, Mallia P. Human rhinovirus infection and COPD: role in exacerbations and potential for therapeutic targets. Expert Rev Respir Med 2020; 14:777-789. [PMID: 32498634 DOI: 10.1080/17476348.2020.1764354] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Respiratory virus infections (predominantly rhinoviruses) are the commonly identified in COPD exacerbations but debate about their role as a trigger of exacerbations continues. Experimental infection studies have provided significant new evidence establishing a causal relationship between virus infection and COPD exacerbations and contributed to a better understanding of the mechanisms of virus-induced exacerbations. However as yet no anti-viral treatments have undergone clinical trials in COPD patients. AREAS COVERED This review discusses the evidence for and against respiratory viruses being the main trigger of COPD exacerbations from both epidemiological studies and experimental infection studies. The host immune response to rhinovirus infection and how abnormalities in host immunity may underlie increased susceptibility to virus infection in COPD are discussed and the role of dual viral-bacterial infection in COPD exacerbations. Finally the current state of anti-viral therapy is discussed and how these may be used in the future treatment of COPD exacerbations. EXPERT OPINION Respiratory virus infections are the trigger of a substantial proportion of COPD exacerbations and rhinoviruses are the most common virus type. Clinical trials of anti-viral agents are needed in COPD patients to determine whether they are effective in virus-induced COPD exacerbations.
Collapse
Affiliation(s)
- John Cafferkey
- Department of Respiratory Medicine, Imperial College Healthcare NHS Trust , London, UK
| | | | - Patrick Mallia
- Department of Respiratory Medicine, Imperial College Healthcare NHS Trust , London, UK.,National Heart and Lung Institute, Imperial College London , London, UK
| |
Collapse
|
48
|
Watson A, Sørensen GL, Holmskov U, Whitwell HJ, Madsen J, Clark H. Generation of novel trimeric fragments of human SP-A and SP-D after recombinant soluble expression in E. coli. Immunobiology 2020; 225:151953. [PMID: 32747028 PMCID: PMC7422833 DOI: 10.1016/j.imbio.2020.151953] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 04/28/2020] [Indexed: 01/19/2023]
Abstract
Surfactant treatment for neonatal respiratory distress syndrome has dramatically improved survival of preterm infants. However, this has resulted in a markedly increased incidence of sequelae such as neonatal chronic inflammatory lung disease. The current surfactant preparations in clinical use lack the natural lung defence proteins surfactant proteins (SP)-A and D. These are known to have anti-inflammatory and anti-infective properties essential for maintaining healthy non-inflamed lungs. Supplementation of currently available animal derived surfactant therapeutics with these anti-inflammatory proteins in the first few days of life could prevent the development of inflammatory lung disease in premature babies. However, current systems for production of recombinant versions of SP-A and SP-D require a complex solubilisation and refolding protocol limiting expression at scale for drug development. Using a novel solubility tag, we describe the expression and purification of recombinant fragments of human (rfh) SP-A and SP-D using Escherichia coli without the need for refolding. We obtained a mean (± SD) of 23.3 (± 5.4) mg and 86 mg (± 3.5) per litre yield of rfhSP-A and rfhSP-D, respectively. rfhSP-D was trimeric and 68% bound to a ManNAc-affinity column, giving a final yield of 57.5 mg/litre of highly pure protein, substantially higher than the 3.3 mg/litre obtained through the standard refolding protocol. Further optimisation of this novel lab based method could potentially make rfhSP-A and rfhSP-D production more commercially feasible to enable development of novel therapeutics for the treatment of lung infection and inflammation.
Collapse
MESH Headings
- Cloning, Molecular
- Escherichia coli/genetics
- Escherichia coli/metabolism
- Gene Expression
- Humans
- Models, Molecular
- Protein Conformation
- Protein Multimerization
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/isolation & purification
- Receptors, Cell Surface/metabolism
- Receptors, Immunologic/chemistry
- Receptors, Immunologic/genetics
- Receptors, Immunologic/isolation & purification
- Receptors, Immunologic/metabolism
- Recombinant Proteins
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Alastair Watson
- Department of Child Health, Division of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom.
| | - Grith L Sørensen
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Uffe Holmskov
- Department of Cancer and Inflammation, University of Southern Denmark, Odense, Denmark
| | - Harry J Whitwell
- Department of Chemical Engineering, Faculty of Engineering, Imperial College London, London, UK; Department of Metabolomics, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Jens Madsen
- Department of Child Health, Division of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Howard Clark
- Department of Child Health, Division of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| |
Collapse
|
49
|
Johnston SL. IFN Therapy in Airway Disease: Is Prophylaxis a New Approach in Exacerbation Prevention? Am J Respir Crit Care Med 2020; 201:9-11. [PMID: 31577905 PMCID: PMC6938143 DOI: 10.1164/rccm.201909-1850ed] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Sebastian L Johnston
- National Heart and Lung InstituteImperial College LondonLondon, United Kingdomand.,Asthma UK Centre in Allergic Mechanisms of AsthmaLondon, United Kingdom
| |
Collapse
|
50
|
Kong CW, Wilkinson TM. Predicting and preventing hospital readmission for exacerbations of COPD. ERJ Open Res 2020; 6:00325-2019. [PMID: 32420313 PMCID: PMC7211949 DOI: 10.1183/23120541.00325-2019] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/06/2020] [Indexed: 12/17/2022] Open
Abstract
More than a third of patients hospitalised for acute exacerbation of COPD are readmitted to hospital within 90 days. Healthcare professionals and service providers are expected to collaboratively drive efforts to improve hospital readmission rates, which can be challenging due to the lack of clear consensus and guidelines on how best to predict and prevent readmissions. This review identifies these risk factors, highlighting the contribution of multimorbidity, frailty and poor socioeconomic status. Predictive models of readmission that address the multifactorial nature of readmissions and heterogeneity of the disease are reviewed, recognising that in an era of precision medicine, in-depth understanding of the intricate biological mechanisms that heighten the risk of COPD exacerbation and re-exacerbation is needed to derive modifiable biomarkers that can stratify accurately the highest risk groups for targeted treatment. We evaluate conventional and emerging strategies to reduce these potentially preventable readmissions. Here, early recognition of exacerbation symptoms and the delivery of prompt treatment can reduce risk of hospital admissions, while patient education can improve treatment adherence as a key component of self-management strategies. Care bundles are recommended to ensure high-quality care is provided consistently, but evidence for their benefit is limited to date. The search continues for interventions which are effective, sustainable and applicable to a diverse population of patients with COPD exacerbations. Further research into mechanisms that drive exacerbation and affect recovery is crucial to improve our understanding of this complex, highly prevalent disease and to advance the development of more effective treatments.
Collapse
Affiliation(s)
- Chia Wei Kong
- Southampton NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University Hospital Southampton, Southampton, UK
| | - Tom M.A. Wilkinson
- Southampton NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University Hospital Southampton, Southampton, UK
| |
Collapse
|