1
|
Morelli T, Freeman A, Staples KJ, Wilkinson TMA. Hidden in plain sight: the impact of human rhinovirus infection in adults. Respir Res 2025; 26:120. [PMID: 40155903 PMCID: PMC11954259 DOI: 10.1186/s12931-025-03178-w] [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: 06/15/2024] [Accepted: 03/02/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Human rhinovirus (HRV), a non-enveloped RNA virus, was first identified more than 70 years ago. It is highly infectious and easily transmitted through aerosols and direct contact. The advent of multiplex PCR has enhanced the detection of a diverse range of respiratory viruses, and HRV consistently ranks among the most prevalent respiratory pathogens globally. Circulation occurs throughout the year, with peak incidence in autumn and spring in temperate climates. Remarkably, during the SARS-CoV-2 pandemic, HRV transmission persisted, demonstrating its resistance to stringent public health measures aimed at curbing viral transmission. MAIN BODY HRV is characterised by its extensive genetic diversity, comprising three species and more than 170 genotypes. This diversity and substantial number of concurrently circulating strains allows HRVs to frequently escape the adaptive immune system and poses formidable challenges for the development of effective vaccines and antiviral therapies. There is currently a lack of specific treatments. Historically, HRV has been associated with self-limiting upper respiratory infection. However, there is now extensive evidence highlighting its significant role in severe lower respiratory disease in adults, including exacerbations of chronic airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD), as well as pneumonia. These severe manifestations can occur even in immunocompetent individuals, broadening the clinical impact of this ubiquitous virus. Consequently, the burden of rhinovirus infections extends across various healthcare settings, from primary care to general hospital wards and intensive care units. The impact of HRV in adults, in terms of morbidity and healthcare utilisation, rivals that of the other major respiratory viruses, including influenza and respiratory syncytial virus. Recognition of this substantial burden underscores the critical need for novel treatment strategies and effective management protocols to mitigate the impact of HRV infections on public health. CONCLUSION This review examines the epidemiology, clinical manifestations, and risk factors associated with severe HRV infection in adults. By drawing on contemporary literature, we aim to provide a comprehensive overview of the virus's significant health implications. Understanding the scope of this impact is essential for developing new, targeted interventions and improving patient outcomes in the face of this persistent and adaptable pathogen.
Collapse
Affiliation(s)
- Tommaso Morelli
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK.
| | - Anna Freeman
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Karl J Staples
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Tom M A Wilkinson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| |
Collapse
|
2
|
Li L, Zhang H, Xiong P, Liu C, Wan L, Liu M, Mao J, Li R, Shang M, Liu H, Luo Y, Yin J, Wu X, Chen J. Virus profiling of bronchoalveolar lavage fluid in hospitalized non-COVID-19 adult patients with pulmonary infection from November 2020 to November 2021. Virol Sin 2025:S1995-820X(25)00027-6. [PMID: 40120941 DOI: 10.1016/j.virs.2025.03.005] [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: 01/31/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
Identifying the cause of respiratory tract infections is important for reducing the burden of diagnosis and treatment. To assess viral etiologies of hospitalized patients with pulmonary infection (including patients with lower respiratory tract infection, tuberculosis, lung cancer, and pulmonary nodules), bronchoalveolar lavage fluid (BALF) specimens were collected from non-COVID-19 adult patients (n = 333) between November 2020 and November 2021. Multiple common respiratory pathogens were detected using multiplex reverse-transcription polymerase chain reaction. The result showed that at least one virus was identified in 35.44% (118/333) of the cases. Among these, influenza virus was the most commonly identified, followed by the parainfluenza virus, coronavirus, human rhinoviruses, and human respiratory syncytial viruses. The tuberculosis group demonstrated the highest viral detection rate, yet paradoxically exhibited the lowest co-infection rate. In contrast, the highest co-infection frequency was observed in the pulmonary nodules group. Patients with viral infections exhibited more severe clinical symptoms compared to those without detected viral infections. However, this observation was only noted in the lower respiratory tract infection group among the different disease groups. Notably, among patients infected with a specific virus, there were no significant differences in viral load between single and co-infections. Our study identified the major causative agents in hospitalized adult patients with pulmonary infection, offering insights for precise disease diagnosis and the prevention of unnecessary use of antimicrobial drugs.
Collapse
Affiliation(s)
- Liangyu Li
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Haiyue Zhang
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Pei Xiong
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chan Liu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine,Tongji University, Shanghai, 200433, China
| | - Lu Wan
- Tianjin University, Jinnan Hospital, Department of Respiratory and Critical Care Medicine, Tianjin, 200350, China
| | - Mengling Liu
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jieyu Mao
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ruiyun Li
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Min Shang
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hailing Liu
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yuchuan Luo
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jing Yin
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaojun Wu
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Jianjun Chen
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
3
|
Lewicki S, Bałan BJ, Stelmasiak M, Radomska-Leśniewska DM, Szymański Ł, Rios-Turek N, Bień-Kalinowska J, Szarpak Ł, Hajduk B. Immunological Insights and Therapeutic Advances in COPD: Exploring Oral Bacterial Vaccines for Immune Modulation and Clinical Improvement. Vaccines (Basel) 2025; 13:107. [PMID: 40006655 PMCID: PMC11861055 DOI: 10.3390/vaccines13020107] [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: 12/09/2024] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 02/27/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a prevalent chronic condition associated with substantial global morbidity and mortality. Primarily caused by prolonged exposure to harmful agents such as dust and gases, COPD is characterized by persistent airflow limitation, clinically manifesting as chronic cough, sputum production, and dyspnea. The disease course alternates between stable phases and exacerbations, with the latter often associated with pathogenic colonization of the respiratory tract. This review examines the immunological underpinnings of COPD, emphasizing the interplay between innate and adaptive immunity in disease pathogenesis. Dysregulated immune responses to environmental factors perpetuate chronic inflammation, resulting in progressive pulmonary epithelial damage and connective tissue hyperplasia, which compromise gas exchange. Exacerbations further exacerbate respiratory failure, aggravating patient symptoms and accelerating disease progression. Despite advances in COPD management, effective therapeutic options remain limited. Current treatments primarily aim to alleviate symptoms, reduce immune activation, and manage infections, yet many patients experience suboptimal outcomes. This review highlights the potential of novel therapeutic approaches targeting immune system cells and pathways. In particular, it explores the promise of oral bacterial vaccines as immunomodulatory agents to enhance immune responses and improve clinical outcomes in COPD, addressing critical gaps in current treatment paradigms.
Collapse
Affiliation(s)
- Sławomir Lewicki
- Institute of Outcomes Research, Maria Sklodowska-Curie Medical Academy, Pl. Żelaznej Bramy 10, 00-136 Warsaw, Poland; (J.B.-K.); (B.H.)
| | - Barbara Joanna Bałan
- Department of Environmental Threat Prevention, Allergology and Immunology, Faculty of Health Sciences, Medical University of Warsaw, Pawińskiego 3c, 02-106 Warsaw, Poland
| | - Marta Stelmasiak
- Department of Dietetics, Institute of Human Nutrition Science, Warsaw University of Life Sciences, Nowoursynowska 159c St., 02-776 Warsaw, Poland;
| | | | - Łukasz Szymański
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland;
| | - Natalia Rios-Turek
- Hull University Teaching Hospitals NHS Trust, Hull University, Anlaby Rd., Hull HU3 2JZ, UK;
| | - Justyna Bień-Kalinowska
- Institute of Outcomes Research, Maria Sklodowska-Curie Medical Academy, Pl. Żelaznej Bramy 10, 00-136 Warsaw, Poland; (J.B.-K.); (B.H.)
| | - Łukasz Szarpak
- Institute of Medicine Science, Collegium Medicum, The John Paul II Catholic University of Lublin, 20-708 Lublin, Poland;
- Department of Clinical Research and Development, LUXMED Group, 02-678 Warsaw, Poland
- Henry JN Taub Department of Emergency Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- TS Out-Patients Clinic for Cardiovascular and Pulmonary Diseases, 01-460 Warsaw, Poland;
| | - Bogdan Hajduk
- TS Out-Patients Clinic for Cardiovascular and Pulmonary Diseases, 01-460 Warsaw, Poland;
| |
Collapse
|
4
|
Wu L, Zhang E, Tu Y, Chen Y, Wang C, Ren Y, Fang B. Inherent immunity and adaptive immunity: Mechanism and role in AECOPD. Innate Immun 2025; 31:17534259251322612. [PMID: 40017227 PMCID: PMC11869301 DOI: 10.1177/17534259251322612] [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: 04/20/2024] [Revised: 12/20/2024] [Accepted: 02/04/2025] [Indexed: 03/01/2025] Open
Abstract
Acute exacerbation of chronic obstructive pulmonary disease (AECOPD) is the leading cause of hospitalization and mortality in COPD patients. The occurrence of antibiotic resistance and the progression of non-infectious diseases contribute to poor patient outcomes. Thus, a comprehensive understanding of the mechanisms underlying AECOPD is essential for effective prevention. It is widely acknowledged that the immune system plays a fundamental role in pathogen clearance and the development of inflammation. Immune dysregulation, either due to deficiency or hyperactivity, has been implicated in AECOPD pathogenesis. Therefore, the purpose of this review is to investigate the possible mechanisms underlying dysregulated immune function and disease progression in COPD patients, specifically focusing on the innate and adaptive immune responses. The ultimate aim is to provide new insights for clinical prevention and treatment strategies targeting AECOPD.
Collapse
Affiliation(s)
- Linguangjin Wu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Emergency Department, Shanghai, China
| | - Erxin Zhang
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yadan Tu
- Chongqing Hospital of Traditional Chinese Medicine, Classic Department of Traditional Chinese Medicine, Chongqing, China
| | - Yong Chen
- Chongqing Hospital of Traditional Chinese Medicine, Classic Department of Traditional Chinese Medicine, Chongqing, China
| | - Chenghu Wang
- Chongqing Hospital of Traditional Chinese Medicine, Classic Department of Traditional Chinese Medicine, Chongqing, China
| | - Yi Ren
- Chongqing Hospital of Traditional Chinese Medicine, Classic Department of Traditional Chinese Medicine, Chongqing, China
| | - Bangjiang Fang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Emergency Department, Shanghai, China
| |
Collapse
|
5
|
Finney LJ, Fenwick P, Kemp SV, Singanayagam A, Edwards MR, Belchamber KBR, Kebadze T, Regis E, Donaldson GD, Mallia P, Donnelly LE, Johnston SL, Wedzicha JA. Impaired antiviral immunity in frequent exacerbators of chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2025; 328:L120-L133. [PMID: 39560620 DOI: 10.1152/ajplung.00118.2024] [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: 04/03/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024] Open
Abstract
Respiratory viruses cause chronic obstructive pulmonary disease (COPD) exacerbations. Rhinoviruses (RVs) are the most frequently detected. Some patients with COPD experience frequent exacerbations (≥2 exacerbations/yr). The relationship between exacerbation frequency and antiviral immunity remains poorly understood. The objective of this study was to investigate the relationship between exacerbation frequency and antiviral immunity in COPD. Alveolar macrophages and bronchial epithelial cells (BECs) were obtained from patients with COPD and healthy participants. Alveolar macrophages were infected with RV-A16 multiplicity of infection (MOI) 5 and BECs infected with RV-A16 MOI 1 for 24. Interferons (IFNs) and proinflammatory cytokines IL-1β, IL-6, C-X-C motif chemokine ligand (CXCL)-8, and TNF were measured in cell supernatants using a mesoscale discovery platform. Viral load and interferon-stimulated genes were measured in cell lysates using quantitative PCR. Spontaneous and RV-induced IFN-β, IFN-γ, and CXCL-11 release were significantly reduced in alveolar macrophages from patients with COPD compared with healthy subjects. IFN-β was further impaired in uninfected alveolar macrophages from patients with COPD with frequent exacerbations 82.0 pg/mL versus infrequent exacerbators 234.7 pg/mL, P = 0.008 and RV-infected alveolar macrophages from frequent exacerbators 158.1 pg/mL versus infrequent exacerbators 279.5 pg/mL, P = 0.022. Release of proinflammatory cytokines CXCL-8, IL-6, TNF, and IL-1β was higher in uninfected BECs from patients with COPD compared with healthy subjects but there was no difference in proinflammatory response to RV between groups. IFN responses to RV were impaired in alveolar macrophages from patients with COPD and further reduced in patients with frequent exacerbations.NEW & NOTEWORTHY COPD exacerbations are commonly triggered by viral infections. Some patients with COPD have frequent exacerbations leading to rapid lung function decline and increased mortality. In this study, antiviral responses (interferons) from bronchial epithelial cells and alveolar macrophages were reduced in patients with COPD compared with healthy participants and further reduced in patients with COPD with frequent exacerbations. Impaired antiviral immunity may lead to frequent COPD exacerbations. Targeted vaccinations and antiviral therapy may reduce exacerbations in COPD.
Collapse
Affiliation(s)
- Lydia J Finney
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Peter Fenwick
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Samuel V Kemp
- Royal Brompton and Harefield Hospitals, London, United Kingdom
| | - Aran Singanayagam
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Michael R Edwards
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kylie B R Belchamber
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Tatiana Kebadze
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Eteri Regis
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Gavin D Donaldson
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Patrick Mallia
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Louise E Donnelly
- 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
| | - Jadwiga A Wedzicha
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
6
|
Antunes KH, Jackson MM, Mincham KT, Snelgrove RJ, Singanayagam A. Assessment of Rhinovirus Infection in a Mouse Model of COPD. Methods Mol Biol 2025; 2903:165-171. [PMID: 40016465 DOI: 10.1007/978-1-0716-4410-2_13] [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] [Indexed: 03/01/2025]
Abstract
A better understanding of the mechanisms governing virus-induced exacerbations of chronic obstructive pulmonary disease (COPD) is urgently needed to develop more effective therapeutic approaches. Animal models are invaluable tools for gaining mechanistic insight into innate immunity. Here, we describe the methods required to establish a short-term mouse model comprising elastase administration to induce features of COPD, combined with infection using the minor group rhinovirus serotype A1 (RV-A1). This model recapitulates the hallmark features of human virus-induced COPD exacerbation and facilitates the study of mechanisms that drive these episodes.
Collapse
Affiliation(s)
- Krist H Antunes
- Laboratory of Clinical and Experimental Immunology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Millie M Jackson
- Department of Infectious Disease, Imperial College London, London, UK
| | - Kyle T Mincham
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Aran Singanayagam
- Department of Infectious Disease, Imperial College London, London, UK.
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
7
|
Sajjan U. In Vivo Model to Assess the Nasal Infection by Minor Group Rhinovirus. Methods Mol Biol 2025; 2903:141-152. [PMID: 40016463 DOI: 10.1007/978-1-0716-4410-2_11] [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] [Indexed: 03/01/2025]
Abstract
Rhinovirus primarily infects humans via nasal epithelial cells and is spread through the inhalation of shed progeny virus via micro-droplets containing the virus. A handful of studies have investigated the nasal mucosal responses to rhinovirus in experimentally infected healthy volunteers. There are also studies that have investigated the effect of antiviral drugs in reducing symptoms induced by rhinovirus infection. It is extremely expensive to test the antiviral drugs in human volunteers, and therefore having preclinical in vivo models is necessary to confirm the antiviral effect of the drugs prior to conducting clinical trials. The existing in vivo models for RV infection are focused on lung inflammation, which may be relevant to patients with chronic lung diseases, in whom rhinovirus causes lower respiratory tract infections. In this chapter, we describe a mouse model of rhinovirus infection, which shows self-limiting inflammation of the sinonasal mucosa.
Collapse
Affiliation(s)
- Umadevi Sajjan
- Centre for Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA.
- Department of Microbiology, Immunology and Inflammation, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA.
- Department of Thoracic Medicine and Surgery, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Tabasi M, Markward T, Sajjan U. Culturing of Airway Stem Cells Obtained from COPD Patients to Assess the Effects of Rhinovirus Infection. Methods Mol Biol 2025; 2903:97-111. [PMID: 40016461 DOI: 10.1007/978-1-0716-4410-2_9] [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] [Indexed: 03/01/2025]
Abstract
Rhinovirus primarily infects airway epithelial cells lining the conductive airways. Mucociliary-differentiated airway epithelial cell cultures, established from airway basal cells, are relevant in vitro model systems to examine the rhinovirus-stimulated innate immune responses and changes in barrier function. The airway epithelium in patients with chronic respiratory diseases such as asthma, cystic fibrosis, and chronic obstructive pulmonary disease often shows remodeling, such as goblet cell metaplasia, squamous metaplasia, and basal cell hyperplasia. Such changes profoundly affect the airway epithelial responses to rhinovirus infection. Previously, we have demonstrated that mucociliary-differentiated cell cultures, established from airway basal cells isolated from COPD patients, show goblet cell and basal cell hyperplasia similar to that observed in patients. These cultures also show a pro-inflammatory phenotype and abnormal innate immune responses to rhinovirus infection. We describe a culturing method that maintains these in vivo features.
Collapse
Affiliation(s)
- Mohsen Tabasi
- Centre for Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA
| | - Tyler Markward
- Centre for Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA
| | - Umadevi Sajjan
- Centre for Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA.
- Department of Microbiology, Immunology and Inflammation, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA.
- Department of Thoracic Medicine and Surgery, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Sajjan U. Method to Generate High-Titer Rhinovirus C Suitable for In Vitro and In Vivo Studies. Methods Mol Biol 2025; 2903:21-30. [PMID: 40016455 DOI: 10.1007/978-1-0716-4410-2_3] [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] [Indexed: 03/01/2025]
Abstract
Rhinovirus (RV)-C, discovered in 2006, binds to human cadherin-related family member 3 expressed on ciliated cells. RV-C is the cause of severe respiratory illness in children with asthma. Unlike rhinovirus-A and B, very little is known about the RV-C biology or pathogenic mechanisms. This is because RV-C cannot be propagated using the conventional method that is described for RV-A and RV-B in Chapter 2 . H1HeLa cells or primary lung fibroblasts do not express the receptor for RV-C and, therefore, they are resistant to RV-C infection. Recently, human mucociliary-differentiated immortalized airway epithelial cells expressing cadherin-related family member 3 cells were shown to be suitable for propagating RV-C from clinical samples, but this method may not be sustainable due to the costs of culturing the cells for large-scale virus production. Another method to propagate the virus is to transfect the viral genome into H1HeLa or primary lung fibroblasts to bypass the initial infection step, which is the binding and endocytosis of the virus. RV-C virus propagated by this method was demonstrated to yield relatively high-titer infectious virus suitable for in vitro studies. Here, we describe a method to propagate and purify the RV-C suitable for in vitro and in vivo studies.
Collapse
Affiliation(s)
- Umadevi Sajjan
- Centre for Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA.
- Department of Microbiology, Immunology and Inflammation, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA.
- Department of Thoracic Medicine and Surgery, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Colaneri M, Fama F, Fassio F, Holmes D, Scaglione G, Mariani C, Galli L, Lai A, Antinori S, Gori A, Riva A, Schiavini M. Impact of early antiviral therapy on SARS-CoV-2 clearance time in high-risk COVID-19 subjects: A propensity score matching study. Int J Infect Dis 2024; 149:107265. [PMID: 39393523 DOI: 10.1016/j.ijid.2024.107265] [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: 05/02/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Effective treatments for COVID-19 are needed to mitigate disease progression and reduce the burden on healthcare systems. This study investigates the impact of early treatments on SARS-CoV-2 viral shedding duration among high-risk individuals with mild symptoms. METHODS A single-center, retrospective observational study was conducted at Luigi Sacco Hospital in Milan from December 2021 to March 2023. Hospitalized and nonhospitalized adults with a confirmed SARS-CoV-2 infection and at high-risk of disease progression were enrolled. Unadjusted and adjusted negative binomial regression models and a Random Forest regression model were performed before and after matching subjects based on their propensity of being treated or not. RESULTS Results from 518 subjects (428 treated and 90 untreated) revealed a significant reduction in SARS-CoV-2 viral shedding duration among those who received early treatment compared to untreated individuals. Propensity score matching and multivariable regression analyses confirmed this finding. Early treatment significantly reduced the risk of COVID-19-related hospitalization and pneumonia development. Subgroup analysis identified COPD as a potential factor influencing effectiveness of early treatments. CONCLUSIONS Early treatments play a crucial role in reducing SARS-CoV-2 viral shedding and preventing disease progression among high-risk individuals. Shorter viral shedding duration also contributes to improved healthcare resource utilization and infection control measures.
Collapse
Affiliation(s)
- Marta Colaneri
- Department of Clinical Sciences, Infectious Diseases and Immunopathology, University of Milan, Luigi Sacco Hospital, Milan, Italy; Centre for Multidisciplinary Research in Health Science (MACH), University of Milan, Milan, Italy
| | - Federico Fama
- Department of Clinical Sciences, Infectious Diseases and Immunopathology, University of Milan, Luigi Sacco Hospital, Milan, Italy; Centre for Multidisciplinary Research in Health Science (MACH), University of Milan, Milan, Italy.
| | - Federico Fassio
- Department of Public Health, Experimental and Forensic Medicine, Unit of Biostatistics and Clinical Epidemiology, University of Pavia, Pavia, Italy
| | - Darcy Holmes
- Infectious Disease Unit Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Giovanni Scaglione
- Department of Clinical Sciences, Infectious Diseases and Immunopathology, University of Milan, Luigi Sacco Hospital, Milan, Italy
| | - Chiara Mariani
- Department of Clinical Sciences, Infectious Diseases and Immunopathology, University of Milan, Luigi Sacco Hospital, Milan, Italy
| | - Lucia Galli
- Department of Clinical Sciences, Infectious Diseases and Immunopathology, University of Milan, Luigi Sacco Hospital, Milan, Italy
| | - Alessia Lai
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Spinello Antinori
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy; III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Andrea Gori
- Department of Clinical Sciences, Infectious Diseases and Immunopathology, University of Milan, Luigi Sacco Hospital, Milan, Italy; Centre for Multidisciplinary Research in Health Science (MACH), University of Milan, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy; III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Agostino Riva
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy; Infectious Diseases Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Monica Schiavini
- Department of Clinical Sciences, Infectious Diseases and Immunopathology, University of Milan, Luigi Sacco Hospital, Milan, Italy
| |
Collapse
|
11
|
Liu Y, Zhang W, Sun M, Liang X, Wang L, Zhao J, Hou Y, Li H, Yang X. The severity assessment and nucleic acid turning-negative-time prediction in COVID-19 patients with COPD using a fused deep learning model. BMC Pulm Med 2024; 24:515. [PMID: 39402509 PMCID: PMC11476205 DOI: 10.1186/s12890-024-03333-x] [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] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Previous studies have shown that patients with pre-existing chronic obstructive pulmonary diseases (COPD) were more likely to be infected with coronavirus disease (COVID-19) and lead to more severe lung lesions. However, few studies have explored the severity and prognosis of COVID-19 patients with different phenotypes of COPD. PURPOSE The aim of this study is to investigate the value of the deep learning and radiomics features for the severity evaluation and the nucleic acid turning-negative time prediction in COVID-19 patients with COPD including two phenotypes of chronic bronchitis predominant patients and emphysema predominant patients. METHODS A total of 281 patients were retrospectively collected from Hohhot First Hospital between October 2022 and January 2023. They were divided to three groups: COVID-19 group of 95 patients, COVID-19 with emphysema group of 94 patients, COVID-19 with chronic bronchitis group of 92 patients. All patients underwent chest computed tomography (CT) scans and recorded clinical data. The U-net model was pretrained to segment the pulmonary involvement area on CT images and the severity of pneumonia were evaluated by the percentage of pulmonary involvement volume to lung volume. The 107 radiomics features were extracted by pyradiomics package. The Spearman method was employed to analyze the correlation of the data and visualize it through a heatmap. Then we establish a deep learning model (model 1) and a fusion model (model 2) combined deep learning with radiomics features to predict nucleic acid turning-negative time. RESULTS COVID-19 patients with emphysema was lowest in the lymphocyte count compared to COVID-19 patients and COVID-19 companied with chronic bronchitis, and they have the most extensive range of pulmonary inflammation. The lymphocyte count was significantly correlated with pulmonary involvement and the time for nucleic acid turning negative (r=-0.145, P < 0.05). Importantly, our results demonstrated that model 2 achieved an accuracy of 80.9% in predicting nucleic acid turning-negative time. CONCLUSION The pre-existing emphysema phenotype of COPD severely aggravated the pulmonary involvement of COVID-19 patients. Deep learning and radiomics features may provide more information to accurately predict the nucleic acid turning-negative time, which is expected to play an important role in clinical practice.
Collapse
Affiliation(s)
- Yanhui Liu
- Medical Imaging Department, Hohhot First Hospital, Inner Mongolia, P.R. China
| | - Wenxiu Zhang
- Institute of Research and Clinical Innovations, Neusoft Medical Systems Co., Ltd, Shanghai, P.R. China
| | - Mengzhou Sun
- Institute of Research and Clinical Innovations, Neusoft Medical Systems Co., Ltd, Beijing, P.R. China
| | - Xiaoyun Liang
- Institute of Research and Clinical Innovations, Neusoft Medical Systems Co., Ltd, Shanghai, P.R. China
| | - Lu Wang
- Medical Imaging Department, Hohhot First Hospital, Inner Mongolia, P.R. China
| | - Jiaqi Zhao
- Medical Imaging Department, Hohhot First Hospital, Inner Mongolia, P.R. China
| | - Yongquan Hou
- Respiratory and Critical Care Medicine Department, Hohhot First Hospital, Inner Mongolia, P.R. China
| | - Haina Li
- Medical Imaging Department, Hohhot First Hospital, Inner Mongolia, P.R. China
| | - Xiaoguang Yang
- Medical Imaging Department, Hohhot First Hospital, Inner Mongolia, P.R. China.
| |
Collapse
|
12
|
Nguyen THO, Rowntree LC, Chua BY, Thwaites RS, Kedzierska K. Defining the balance between optimal immunity and immunopathology in influenza virus infection. Nat Rev Immunol 2024; 24:720-735. [PMID: 38698083 DOI: 10.1038/s41577-024-01029-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/05/2024]
Abstract
Influenza A viruses remain a global threat to human health, with continued pandemic potential. In this Review, we discuss our current understanding of the optimal immune responses that drive recovery from influenza virus infection, highlighting the fine balance between protective immune mechanisms and detrimental immunopathology. We describe the contribution of innate and adaptive immune cells, inflammatory modulators and antibodies to influenza virus-specific immunity, inflammation and immunopathology. We highlight recent human influenza virus challenge studies that advance our understanding of susceptibility to influenza and determinants of symptomatic disease. We also describe studies of influenza virus-specific immunity in high-risk groups following infection and vaccination that inform the design of future vaccines to promote optimal antiviral immunity, particularly in vulnerable populations. Finally, we draw on lessons from the COVID-19 pandemic to refocus our attention to the ever-changing, highly mutable influenza A virus, predicted to cause future global pandemics.
Collapse
Affiliation(s)
- Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Brendon Y Chua
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
13
|
Zhang Y, Wu K, Mao D, Iberg CA, Yin-Declue H, Sun K, Wikfors HA, Keeler SP, Li M, Young D, Yantis J, Crouch EC, Chartock JR, Han Z, Byers DE, Brody SL, Romero AG, Holtzman MJ. A first-in-kind MAPK13 inhibitor that can correct stem cell reprogramming and post-injury disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.21.608990. [PMID: 39229202 PMCID: PMC11370402 DOI: 10.1101/2024.08.21.608990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The stress kinase MAPK13 (aka p38δ-MAPK) is an attractive entry point for therapeutic intervention because it regulates the structural remodeling that can develop after epithelial barrier injury in the lung and likely other tissue sites. However, a selective, safe, and effective MAPK13 inhibitor is not yet available for experimental or clinical application. Here we identify a first-in-kind MAPK13 inhibitor using structure-based drug design combined with a screening funnel for cell safety and molecular specificity. This inhibitor (designated NuP-4) down-regulates basal-epithelial stem cell reprogramming, structural remodeling, and pathophysiology equivalently to Mapk13 gene-knockout in mouse and mouse organoid models of post-viral lung disease. This therapeutic benefit persists after stopping treatment as a sign of disease modification and attenuates key aspects of inflammation and remodeling as an indication of disease reversal. Similarly, NuP-4 treatment can directly control cytokine-stimulated growth, immune activation, and mucinous differentiation in human basal-cell organoids. The data thereby provide a new tool and potential fix for long-term stem cell reprogramming after viral injury and related conditions that require MAPK13 induction-activation.
Collapse
Affiliation(s)
- Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Dailing Mao
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Courtney A Iberg
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Huiqing Yin-Declue
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kelly Sun
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Hallie A Wikfors
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Shamus P Keeler
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Ming Li
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Deanna Young
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Jennifer Yantis
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Erika C Crouch
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Joshua R Chartock
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Zhenfu Han
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Derek E Byers
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Steven L Brody
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Arthur G Romero
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
- NuPeak Therapeutics Inc., St. Louis, MO 63105
| |
Collapse
|
14
|
Baraldo S, Bonato M, Cassia S, Casolari P, De Ferrari L, Tiné M, Baraldi F, Bigoni T, Riccio AM, Braido F, Saetta M, Papi A, Contoli M. Expression of human Interferon Regulatory Factor 3 (IRF-3) in alveolar macrophages relates to clinical and functional traits in COPD. Respir Res 2024; 25:315. [PMID: 39160551 PMCID: PMC11334339 DOI: 10.1186/s12931-024-02952-6] [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: 12/19/2023] [Accepted: 08/13/2024] [Indexed: 08/21/2024] Open
Abstract
INTRODUCTION Chronic obstructive pulmonary disease (COPD) is a frequent cause of morbidity and mortality. Dysregulated and enhanced immune-inflammatory responses have been described in COPD. Recent data showed impaired immune responses and, in particular, of interferon (IFNs) signaling pathway in these patients. AIM To evaluate in peripheral lung of COPD patients, the expression of some of the less investigated key components of the innate immune responses leading to IFN productions including: IFN-receptors (IFNAR1/IFNAR2), IRF-3 and MDA-5. Correlations with clinical traits and with the inflammatory cell profile have been assessed. METHODS Lung specimens were collected from 58 subjects undergoing thoracic surgery: 22 COPD patients, 21 smokers with normal lung function (SC) and 15 non-smoker controls (nSC). The expression of IFNAR1, IFNAR2, IRF-3 and MDA-5, of eosinophils and activated NK cells (NKp46+) were quantified in the peripheral lung by immunohistochemistry. RESULTS A significant increase of IRF-3 + alveolar macrophages were observed in COPD and SC compared with nSC subjects. However, in COPD patients, the lower the levels of IRF-3 + alveolar macrophages the lower the FEV1 and the higher the exacerbation rate. The presence of chronic bronchitis (CB) was also associated with low levels of IRF-3 + alveolar macrophages. NKp46 + cells, but not eosinophils, were increased in COPD patients compared to nSC patients (p < 0.0001). CONCLUSIONS Smoking is associated with higher levels of innate immune response as showed by higher levels of IRF-3 + alveolar macrophages and NKp46 + cells. In COPD, exacerbation rates, severe airflow obstruction and CB were associated with lower levels of IRF-3 expression, suggesting that innate immune responses characterize specific clinical traits of the disease.
Collapse
Affiliation(s)
- Simonetta Baraldo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Matteo Bonato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Pulmonology Unit, Ca' Foncello Hospital, Azienda Unità Locale Socio-Sanitaria 2 Marca Trevigiana, Treviso, Italy
| | - Sebastiano Cassia
- Respiratory Clinic, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Paolo Casolari
- Section of Respiratory Diseases, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Laura De Ferrari
- Respiratory Clinic, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Mariaenrica Tiné
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Federico Baraldi
- Section of Respiratory Diseases, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Tommaso Bigoni
- Section of Respiratory Diseases, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Anna Maria Riccio
- Respiratory Clinic, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Fulvio Braido
- Respiratory Clinic, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Marina Saetta
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Alberto Papi
- Section of Respiratory Diseases, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Azienda Ospedaliera Universitaria Ferrara and AUSL, Ferrara, Italy
| | - Marco Contoli
- Section of Respiratory Diseases, Department of Translational Medicine, University of Ferrara, Ferrara, Italy.
- Azienda Ospedaliera Universitaria Ferrara and AUSL, Ferrara, Italy.
| |
Collapse
|
15
|
Wu K, Zhang Y, Mao D, Iberg CA, Yin-Declue H, Sun K, Keeler SP, Wikfors HA, Young D, Yantis J, Austin SR, Byers DE, Brody SL, Crouch EC, Romero AG, Holtzman MJ. MAPK13 controls structural remodeling and disease after epithelial injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596863. [PMID: 38895360 PMCID: PMC11185504 DOI: 10.1101/2024.05.31.596863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
All living organisms are charged with repair after injury particularly at epithelial barrier sites, but in some cases this response leads instead to structural remodeling and long-term disease. Identifying the molecular and cellular control of this divergence is key to disease modification. In that regard, stress kinase control of epithelial stem cells is a rational entry point for study. Here we examine the potential for mitogen-activated protein kinase 13 (MAPK13) regulation of epithelial stem cells using models of respiratory viral injury and post-viral lung disease. We show that Mapk13 gene-knockout mice handle acute infectious illness as expected but are protected against structural remodeling manifest as basal-epithelial stem cell (basal-ESC) hyperplasia-metaplasia, immune activation, and mucinous differentiation. In corresponding cell models, Mapk13-deficiency directly attenuates basal-ESC growth and organoid formation. Extension to human studies shows marked induction/activation of basal-cell MAPK13 in clinical samples of comparable remodeling found in asthma and COPD. Here again, MAPK13 gene-knockdown inhibits human basal-ESC growth in culture. Together, the data identify MAPK13 as a control for structural remodeling and disease after epithelial injury and as a suitable target for down-regulation as a disease-modifying strategy.
Collapse
Affiliation(s)
- Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Dailing Mao
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Courtney A. Iberg
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Huiqing Yin-Declue
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kelly Sun
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Shamus P. Keeler
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Hallie A. Wikfors
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Deanna Young
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Jennifer Yantis
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Stephen R. Austin
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Derek E Byers
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Steven L. Brody
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Erika C. Crouch
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Arthur G. Romero
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J. Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
- NuPeak Therapeutics Inc., St. Louis, MO 63105
| |
Collapse
|
16
|
Wu K, Zhang Y, Yin-DeClue H, Sun K, Mao D, Yang K, Austin SR, Crouch EC, Brody SL, Byers DE, Hoffmann CM, Hughes ME, Holtzman MJ. A correctable immune niche for epithelial stem cell reprogramming and post-viral lung diseases. J Clin Invest 2024; 134:e183092. [PMID: 39052353 PMCID: PMC11405052 DOI: 10.1172/jci183092] [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: 05/16/2024] [Accepted: 07/23/2024] [Indexed: 07/27/2024] Open
Abstract
Epithelial barriers are programmed for defense and repair but are also the site of long-term structural remodeling and disease. In general, this paradigm features epithelial stem cells (ESCs) that are called on to regenerate damaged tissues but can also be reprogrammed for detrimental remodeling. Here we identified a Wfdc21-dependent monocyte-derived dendritic cell (moDC) population that functioned as an early sentinel niche for basal ESC reprogramming in mouse models of epithelial injury after respiratory viral infection. Niche function depended on moDC delivery of ligand GPNMB to the basal ESC receptor CD44 so that properly timed antibody blockade of ligand or receptor provided long-lasting correction of reprogramming and broad disease phenotypes. These same control points worked directly in mouse and human basal ESC organoids. Together, the findings identify a mechanism to explain and modify what is otherwise a stereotyped but sometimes detrimental response to epithelial injury.
Collapse
Affiliation(s)
- Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine
| | | | - Kelly Sun
- Pulmonary and Critical Care Medicine, Department of Medicine
| | - Dailing Mao
- Pulmonary and Critical Care Medicine, Department of Medicine
| | - Kuangying Yang
- Pulmonary and Critical Care Medicine, Department of Medicine
| | | | | | - Steven L Brody
- Pulmonary and Critical Care Medicine, Department of Medicine
| | - Derek E Byers
- Pulmonary and Critical Care Medicine, Department of Medicine
| | | | - Michael E Hughes
- Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Genetics, and
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
17
|
Katsoulis O, Toussaint M, Jackson MM, Mallia P, Footitt J, Mincham KT, Meyer GFM, Kebadze T, Gilmour A, Long M, Aswani AD, Snelgrove RJ, Johnston SL, Chalmers JD, Singanayagam A. Neutrophil extracellular traps promote immunopathogenesis of virus-induced COPD exacerbations. Nat Commun 2024; 15:5766. [PMID: 38982052 PMCID: PMC11233599 DOI: 10.1038/s41467-024-50197-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 07/03/2024] [Indexed: 07/11/2024] Open
Abstract
Respiratory viruses are a major trigger of exacerbations in chronic obstructive pulmonary disease (COPD). Airway neutrophilia is a hallmark feature of stable and exacerbated COPD but roles played by neutrophil extracellular traps (NETS) in driving disease pathogenesis are unclear. Here, using human studies of experimentally-induced and naturally-occurring exacerbations we identify that rhinovirus infection induces airway NET formation which is amplified in COPD and correlates with magnitude of inflammation and clinical exacerbation severity. We show that inhibiting NETosis protects mice from immunopathology in a model of virus-exacerbated COPD. NETs drive inflammation during exacerbations through release of double stranded DNA (dsDNA) and administration of DNAse in mice has similar protective effects. Thus, NETosis, through release of dsDNA, has a functional role in the pathogenesis of COPD exacerbations. These studies open up the potential for therapeutic targeting of NETs or dsDNA as a strategy for treating virus-exacerbated COPD.
Collapse
Affiliation(s)
- Orestis Katsoulis
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, UK
| | - Marie Toussaint
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Millie M Jackson
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, UK
| | - Patrick Mallia
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Joseph Footitt
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Kyle T Mincham
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Garance F M Meyer
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Tata Kebadze
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Amy Gilmour
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Merete Long
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Andrew D Aswani
- Department of Intensive Care Medicine, Guy's and St Thomas' NHS Foundation Trust, London, SE1 7EH, UK
| | | | | | - James D Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Aran Singanayagam
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, UK.
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
18
|
Pace E, Di Vincenzo S, Ferraro M, Lanata L, Scaglione F. Role of airway epithelium in viral respiratory infections: Can carbocysteine prevent or mitigate them? Immunology 2024; 172:329-342. [PMID: 38354831 DOI: 10.1111/imm.13762] [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: 10/14/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Alterations in airway epithelial homeostasis increase viral respiratory infections risk. Viral infections frequently are associated with chronic obstructive pulmonary disease (COPD) exacerbations, events that dramatically promote disease progression. Mechanism promoting the main respiratory viruses entry and virus-evocated innate and adaptive immune responses have now been elucidated, and an oxidative stress central role in these pathogenic processes has been recognized. Presence of reactive oxygen species in macrophages and other cells allows them to eliminate virus, but its excess alters the balance between innate and adaptive immune responses and proteases/anti-proteases and leads to uncontrolled inflammation, tissue damage, and hypercoagulability. Different upper and lower airway cell types also play a role in viral entry and infection. Carbocysteine is a muco-active drug with anti-oxidant and anti-inflammatory properties used for the management of several chronic respiratory diseases. Although the use of anti-oxidants has been proposed as an effective strategy in COPD exacerbations management, the molecular mechanisms that explain carbocysteine efficacy have not yet been fully clarified. The present review describes the most relevant features of the common respiratory virus pathophysiology with a focus on epithelial cells and oxidative stress role and reports data supporting a putative role of carbocysteine in viral respiratory infections.
Collapse
Affiliation(s)
- Elisabetta Pace
- Istituto di Farmacologia Traslazionale-Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Serena Di Vincenzo
- Istituto di Farmacologia Traslazionale-Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Maria Ferraro
- Istituto di Farmacologia Traslazionale-Consiglio Nazionale delle Ricerche, Palermo, Italy
| | | | - Francesco Scaglione
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| |
Collapse
|
19
|
Monk PD, Brookes JL, Tear VJ, Batten TN, Newall C, Mankowski M, Crooks MG, Singh D, Chaudhuri R, Leaker B, Lunn K, Reynolds S, Dudley S, Gabbay FJ, Holgate ST, Djukanovic R, Wilkinson TM. Nebulised interferon beta-1a (SNG001) in the treatment of viral exacerbations of COPD. Respir Res 2024; 25:228. [PMID: 38811970 PMCID: PMC11138078 DOI: 10.1186/s12931-024-02854-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/17/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Respiratory viral infections are major drivers of chronic obstructive pulmonary disease (COPD) exacerbations. Interferon-β is naturally produced in response to viral infection, limiting replication. This exploratory study aimed to demonstrate proof-of-mechanism, and evaluate the efficacy and safety of inhaled recombinant interferon-β1a (SNG001) in COPD. Part 1 assessed the effects of SNG001 on induced sputum antiviral interferon-stimulated gene expression, sputum differential cell count, and respiratory function. Part 2 compared SNG001 and placebo on clinical efficacy, sputum and serum biomarkers, and viral clearance. METHODS In Part 1, patients (N = 13) with stable COPD were randomised 4:1 to SNG001 or placebo once-daily for three days. In Part 2, patients (N = 109) with worsening symptoms and a positive respiratory viral test were randomised 1:1 to SNG001 or placebo once-daily for 14 days in two Groups: A (no moderate exacerbation); B (moderate COPD exacerbation [i.e., acute worsening of respiratory symptoms treated with antibiotics and/or oral corticosteroids]). RESULTS In Part 1, SNG001 upregulated sputum interferon gene expression. In Part 2, there were minimal SNG001-placebo differences in the efficacy endpoints; however, whereas gene expression was initially upregulated by viral infection, then declined on placebo, levels were maintained with SNG001. Furthermore, the proportion of patients with detectable rhinovirus (the most common virus) on Day 7 was lower with SNG001. In Group B, serum C-reactive protein and the proportion of patients with purulent sputum increased with placebo (suggesting bacterial infection), but not with SNG001. The overall adverse event incidence was similar with both treatments. CONCLUSIONS Overall, SNG001 was well-tolerated in patients with COPD, and upregulated lung antiviral defences to accelerate viral clearance. These findings warrant further investigation in a larger study. TRIAL REGISTRATION EU clinical trials register (2017-003679-75), 6 October 2017.
Collapse
Affiliation(s)
| | | | | | | | | | - Marcin Mankowski
- Synairgen Research Ltd, Southampton, UK
- tranScrip Ltd, Wokingham, UK
| | - Michael G Crooks
- Respiratory Research Group, Hull York Medical School, University of Hull, Kingston Upon Hull, Hull, UK
| | - Dave Singh
- Medicines Evaluation Unit, The University of Manchester, Manchester University NHS Foundation Trust, Manchester, UK
| | - Rekha Chaudhuri
- Gartnavel General Hospital, Glasgow, UK
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Brian Leaker
- Respiratory Clinical Trials Ltd, Fitzrovia Hospital, London, UK
| | | | | | | | | | - Stephen T Holgate
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, University of Southampton, Southampton, UK
| | - Ratko Djukanovic
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, University of Southampton, Southampton, UK
| | - Thomas Ma Wilkinson
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
20
|
Li K, Bartlett JA, Wohlford-Lenane CL, Xue B, Thurman AL, Gallagher TM, Pezzulo AA, McCray PB. Interleukin 13-Induced Inflammation Increases DPP4 Abundance but Does Not Enhance Middle East Respiratory Syndrome Coronavirus Replication in Airway Epithelia. J Infect Dis 2024; 229:1419-1429. [PMID: 37698016 PMCID: PMC11095549 DOI: 10.1093/infdis/jiad383] [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: 01/06/2023] [Revised: 08/18/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Chronic pulmonary conditions such as asthma and chronic obstructive pulmonary disease increase the risk of morbidity and mortality during infection with the Middle East respiratory syndrome coronavirus (MERS-CoV). We hypothesized that individuals with such comorbidities are more susceptible to MERS-CoV infection due to increased expression of its receptor, dipeptidyl peptidase 4 (DPP4). METHODS We modeled chronic airway disease by treating primary human airway epithelia with the Th2 cytokine interleukin 13 (IL-13), examining how this affected DPP4 protein levels with MERS-CoV entry and replication. RESULTS IL-13 exposure for 3 days led to greater DPP4 protein abundance, while a 21-day treatment raised DPP4 levels and caused goblet cell metaplasia. Surprisingly, despite this increase in receptor availability, MERS-CoV entry and replication were not significantly affected by IL-13 treatment. CONCLUSIONS Our results suggest that greater DPP4 abundance is likely not the primary mechanism leading to increased MERS severity in the setting of Th2 inflammation. Transcriptional profiling analysis highlighted the complexity of IL-13-induced changes in airway epithelia, including altered expression of genes involved in innate immunity, antiviral responses, and maintenance of the extracellular mucus barrier. These data suggest that additional factors likely interact with DPP4 abundance to determine MERS-CoV infection outcomes.
Collapse
Affiliation(s)
- Kun Li
- Department of Pediatrics, Pappajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Jennifer A Bartlett
- Department of Pediatrics, Pappajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Christine L Wohlford-Lenane
- Department of Pediatrics, Pappajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Biyun Xue
- Department of Internal Medicine, Pappajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Andrew L Thurman
- Department of Internal Medicine, Pappajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Thomas M Gallagher
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL
| | - Alejandro A Pezzulo
- Department of Internal Medicine, Pappajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Paul B McCray
- Department of Pediatrics, Pappajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Iowa City, IA
- Department of Microbiology, Pappajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Iowa City, IA
| |
Collapse
|
21
|
Jiang YZ, Huang XR, Chang J, Zhou Y, Huang XT. SIRT1: An Intermediator of Key Pathways Regulating Pulmonary Diseases. J Transl Med 2024; 104:102044. [PMID: 38452903 DOI: 10.1016/j.labinv.2024.102044] [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: 08/31/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024] Open
Abstract
Silent information regulator type-1 (SIRT1), a nicotinamide adenine dinucleotide+-dependent deacetylase, is a member of the sirtuins family and has unique protein deacetylase activity. SIRT1 participates in physiological as well as pathophysiological processes by targeting a wide range of protein substrates and signalings. In this review, we described the latest progress of SIRT1 in pulmonary diseases. We have introduced the basic information and summarized the prominent role of SIRT1 in several lung diseases, such as acute lung injury, acute respiratory distress syndrome, chronic obstructive pulmonary disease, lung cancer, and aging-related diseases.
Collapse
Affiliation(s)
- Yi-Zhu Jiang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xin-Ran Huang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jing Chang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xiao-Ting Huang
- Xiangya Nursing School, Central South University, Changsha, China.
| |
Collapse
|
22
|
Bourdin A, Ahmed E, Vachier I, Roche N, Pissarra J, Malafaye N, Molinari N. Hospitalizations for Chronic Obstructive Pulmonary Disease Exacerbation During COVID-19. JAMA Netw Open 2024; 7:e2412383. [PMID: 38771579 PMCID: PMC11109769 DOI: 10.1001/jamanetworkopen.2024.12383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/19/2024] [Indexed: 05/22/2024] Open
Abstract
This cross-sectional study investigates changes in the number of chronic obstructive pulmonary disease (COPD)–related admissions before, during, and after the COVID-19 pandemic in France.
Collapse
Affiliation(s)
- Arnaud Bourdin
- Department of Respiratory Diseases, University of Montpellier, Centre Hospitalier Universitaire (CHU) Montpellier, Montpellier, France
- Laboratoire de Physiologie et Médecine Expérimentale du Cœur et des Muscles, University of Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale (INSERM), CHU Montpellier, Montpellier, France
| | - Engi Ahmed
- Department of Respiratory Diseases, University of Montpellier, Centre Hospitalier Universitaire (CHU) Montpellier, Montpellier, France
- Laboratoire de Physiologie et Médecine Expérimentale du Cœur et des Muscles, University of Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale (INSERM), CHU Montpellier, Montpellier, France
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Isabelle Vachier
- Department of Respiratory Diseases, University of Montpellier, Centre Hospitalier Universitaire (CHU) Montpellier, Montpellier, France
- Laboratoire de Physiologie et Médecine Expérimentale du Cœur et des Muscles, University of Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale (INSERM), CHU Montpellier, Montpellier, France
| | - Nicolas Roche
- Assistance Publique–Hôpitaux de Paris, Centre–Université Paris Cité, Cochin Hospital and Institute (INSERM UMR1016), Respiratory Medicine, Paris, France
| | - Joana Pissarra
- Clinical Research and Epidemiology Unit, CHU Montpellier, University of Montpellier, Montpellier, France
| | - Nicolas Malafaye
- Clinical Research and Epidemiology Unit, CHU Montpellier, University of Montpellier, Montpellier, France
| | - Nicolas Molinari
- Institut Desbrest d’Épidémiologie et de Santé Publique, INSERM, Precision Medicine by Data Integration and Causal Learning, Institut National de Recherche en Informatique et en Automatique, University of Montpellier, CHU Montpellier, Montpellier, France
| |
Collapse
|
23
|
Baraldi F, Barrecheguren M, Papi A, Miravitlles M. Managing exacerbations of COPD: how much progress have we made? COPD IN THE 21ST CENTURY 2024. [DOI: 10.1183/2312508x.10008023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
24
|
Holtzman MJ, Zhang Y, Wu K, Romero AG. Mitogen-activated protein kinase-guided drug discovery for post-viral and related types of lung disease. Eur Respir Rev 2024; 33:230220. [PMID: 38417971 PMCID: PMC10900067 DOI: 10.1183/16000617.0220-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/18/2024] [Indexed: 03/01/2024] Open
Abstract
Respiratory viral infections are a major public health problem, with much of their morbidity and mortality due to post-viral lung diseases that progress and persist after the active infection is cleared. This paradigm is implicated in the most common forms of chronic lung disease, such as asthma and COPD, as well as other virus-linked diseases including progressive and long-term coronavirus disease 2019. Despite the impact of these diseases, there is a lack of small-molecule drugs available that can precisely modify this type of disease process. Here we will review current progress in understanding the pathogenesis of post-viral and related lung disease with characteristic remodelling phenotypes. We will also develop how this data leads to mitogen-activated protein kinase (MAPK) in general and MAPK13 in particular as key druggable targets in this pathway. We will also explore recent advances and predict the future breakthroughs in structure-based drug design that will provide new MAPK inhibitors as drug candidates for clinical applications. Each of these developments point to a more effective approach to treating the distinct epithelial and immune cell based mechanisms, which better account for the morbidity and mortality of post-viral and related types of lung disease. This progress is vital given the growing prevalence of respiratory viruses and other inhaled agents that trigger stereotyped progression to acute illness and chronic disease.
Collapse
Affiliation(s)
- Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- NuPeak Therapeutics Inc., St. Louis, MO, USA
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Arthur G Romero
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
25
|
Oxford JS, Catchpole A, Mann A, Bell A, Noulin N, Gill D, Oxford JR, Gilbert A, Balasingam S. A Brief History of Human Challenge Studies (1900-2021) Emphasising the Virology, Regulatory and Ethical Requirements, Raison D'etre, Ethnography, Selection of Volunteers and Unit Design. Curr Top Microbiol Immunol 2024; 445:1-32. [PMID: 35704095 DOI: 10.1007/82_2022_253] [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] [Indexed: 10/18/2022]
Abstract
Venetian quarantine 400 years ago was an important public health measure. Since 1900 this has been refined to include "challenge" or deliberate infection with pathogens be they viruses, bacteria, or parasites. Our focus is virology and ranges from the early experiments in Cuba with Yellow Fever Virus to the most widespread pathogen of our current times, COVID-19. The latter has so far caused over four million deaths worldwide and 190 million cases of the disease. Quarantine and challenge were also used to investigate the Spanish Influenza of 1918 which caused over 100 million deaths. We consider here the merits of the approach, that is the speeding up of knowledge in a practical sense leading to the more rapid licensing of vaccines and antimicrobials. At the core of quarantine and challenge initiatives is the design of the unit to allow safe confinement of the pathogen and protection of the staff. Most important though is the safety of volunteers. We can see now, as in 1900, that members of our society are prepared and willing to engage in these experiments for the public good. Our ethnology study, where the investigator observed the experiment from within the quarantine, gave us the first indication of changing attitudes amongst volunteers whilst in quarantine. These quarantine experiments, referred to as challenge studies, human infection studies, or "controlled human infection models" involve thousands of clinical samples taken over two to three weeks and can provide a wealth of immunological and molecular data on the infection itself and could allow the discovery of new targets for vaccines and therapeutics. The Yellow Fever studies from 121 years ago gave the impetus for development of a successful vaccine still used today whilst also uncovering the nature of the Yellow Fever agent, namely that it was a virus. We outline how carefully these experiments are approached and the necessity to have high quality units with self-contained air-flow along with extensive personal protective equipment for nursing and medical staff. Most important is the employment of highly trained scientific, medical and nursing staff. We face a future of emerging pathogens driven by the increasing global population, deforestation, climate change, antibiotic resistance and increased global travel. These emerging pathogens may be pathogens we currently are not aware of or have not caused outbreaks historically but could also be mutated forms of known pathogens including viruses such as influenza (H7N9, H5N1 etc.) and coronaviruses. This calls for challenge studies to be part of future pandemic preparedness as an additional tool to assist with the rapid development of broad-spectrum antimicrobials, immunomodulators and new vaccines.
Collapse
Affiliation(s)
- J S Oxford
- Blizzard Institute of Cell and Molecular Science, Queen Mary University of London, London, E1 2AT, UK
| | | | | | | | | | - D Gill
- Blizzard Institute of Cell and Molecular Science, Queen Mary University of London, London, E1 2AT, UK
| | - J R Oxford
- Inveresk Medical Practice, Edinburgh, E21 7BP, UK
| | | | | |
Collapse
|
26
|
Peng T. Bad Neighbors or Bad Neighborhoods: Pathogenic Residency of T Cells in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2023; 208:1148-1150. [PMID: 37855741 PMCID: PMC10868362 DOI: 10.1164/rccm.202310-1760ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 10/18/2023] [Indexed: 10/20/2023] Open
Affiliation(s)
- Tien Peng
- Department of Medicine Cardiovascular Institute and Division of Pulmonary, Critical Care, Allergy and Sleep Medicine
- Bakar ImmunoX Initiative University of California, San Francisco San Francisco, California
| |
Collapse
|
27
|
Bhatt SP, Agusti A, Bafadhel M, Christenson SA, Bon J, Donaldson GC, Sin DD, Wedzicha JA, Martinez FJ. Phenotypes, Etiotypes, and Endotypes of Exacerbations of Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2023; 208:1026-1041. [PMID: 37560988 PMCID: PMC10867924 DOI: 10.1164/rccm.202209-1748so] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 08/04/2023] [Indexed: 08/11/2023] Open
Abstract
Chronic obstructive pulmonary disease is a major health problem with a high prevalence, a rising incidence, and substantial morbidity and mortality. Its course is punctuated by acute episodes of increased respiratory symptoms, termed exacerbations of chronic obstructive pulmonary disease (ECOPD). ECOPD are important events in the natural history of the disease, as they are associated with lung function decline and prolonged negative effects on quality of life. The present-day therapy for ECOPD with short courses of antibiotics and steroids and escalation of bronchodilators has resulted in only modest improvements in outcomes. Recent data indicate that ECOPD are heterogeneous, raising the need to identify distinct etioendophenotypes, incorporating traits of the acute event and of patients who experience recurrent events, to develop novel and targeted therapies. These characterizations can provide a complete clinical picture, the severity of which will dictate acute pharmacological treatment, and may also indicate whether a change in maintenance therapy is needed to reduce the risk of future exacerbations. In this review we discuss the latest knowledge of ECOPD types on the basis of clinical presentation, etiology, natural history, frequency, severity, and biomarkers in an attempt to characterize these events.
Collapse
Affiliation(s)
- Surya P. Bhatt
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Alvar Agusti
- Institut Respiratori (Clinic Barcelona), Càtedra Salut Respiratoria (Universitat de Barcelona), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS-Barcelona), Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), España
| | - Mona Bafadhel
- Faculty of Life Sciences and Medicine, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Stephanie A. Christenson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, California
| | - Jessica Bon
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Gavin C. Donaldson
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Don D. Sin
- Centre for Heart Lung Innovation and
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- St. Paul’s Hospital, Vancouver, British Columbia, Canada; and
| | - Jadwiga A. Wedzicha
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | |
Collapse
|
28
|
Hyams C, Qian G, Nava G, Challen R, Begier E, Southern J, Lahuerta M, Nguyen JL, King J, Morley A, Clout M, Maskell N, Jodar L, Oliver J, Ellsbury G, McLaughlin JM, Gessner BD, Finn A, Danon L, Dodd JW. Impact of SARS-CoV-2 infective exacerbation of chronic obstructive pulmonary disease on clinical outcomes in a prospective cohort study of hospitalised adults. J R Soc Med 2023; 116:371-385. [PMID: 37404021 PMCID: PMC10686205 DOI: 10.1177/01410768231184162] [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: 01/05/2023] [Accepted: 06/04/2023] [Indexed: 07/06/2023] Open
Abstract
OBJECTIVES To determine whether acute exacerbations of chronic obstructive pulmonary disease (AECOPD) triggered by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), have worse outcomes than AECOPD caused by other infectious agents or non-infective AECOPD (NI-COPD). DESIGN A two-hospital prospective cohort study of adults hospitalised with acute respiratory disease. We compared outcomes with AECOPD and a positive test for SARS-CoV-2 (n = 816), AECOPD triggered by other infections (n = 3038) and NI-COPD (n = 994). We used multivariable modelling to adjust for potential confounders and assessed variation by seasons associated with different SARS-CoV-2 variants. SETTING Bristol UK, August 2020-May 2022. PARTICIPANTS Adults (≥18 y) hospitalised with AECOPD. MAIN OUTCOME MEASURES We determined the risk of positive pressure support, longer hospital admission and mortality following hospitalisation with AECOPD due to non-SARS-CoV-2 infection compared with SARS-CoV-2 AECOPD and NI-COPD. RESULTS Patients with SARS-CoV-2 AECOPD, in comparison to non-SARS-CoV-2 infective AECOPD or NI-COPD, more frequently required positive pressure support (18.5% and 7.5% vs. 11.7%, respectively), longer hospital stays (median [interquartile range, IQR]: 7 [3-15] and 5 [2-10] vs. 4 [2-9] days, respectively) and had higher 30-day mortality (16.9% and 11.1% vs. 5.9%, respectively) (all p < 0.001). In adjusted analyses, SARS-CoV-2 AECOPD was associated with a 55% (95% confidence interval [95% CI]: 24-93), 26% (95% CI: 15-37) and 35% (95% CI: 10-65) increase in the risk of positive pressure support, hospitalisation length and 30-day mortality, respectively, relative to non-SARS-CoV-2 infective AECOPD. The difference in risk remained similar during periods of wild-type, Alpha and Delta SARS-CoV-2 strain dominance, but diminished during Omicron dominance. CONCLUSIONS SARS-CoV-2-related AECOPD had worse patient outcomes compared with non-SARS-CoV-2 AECOPD or NI-AECOPD, although the difference in risks was less pronounced during Omicron dominance.
Collapse
Affiliation(s)
- Catherine Hyams
- Academic Respiratory Unit and Bristol Vaccine Centre, University of Bristol, Bristol, BS15, UK
| | - George Qian
- Engineering Mathematics, University of Bristol, Bristol, Bristol, BS8, UK
| | - George Nava
- Academic Respiratory Unit, University of Bristol, Southmead Hospital, Bristol, Bristol, BS15, UK
| | - Robert Challen
- Engineering Mathematics, University of Bristol, Bristol, Bristol, BS8, UK
| | - Elizabeth Begier
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer Inc., Collegeville, PA 19426, USA
| | - Jo Southern
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer Inc., Collegeville, PA 19426, USA
| | - Maria Lahuerta
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer Inc., Collegeville, PA 19426, USA
| | - Jennifer L Nguyen
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer Inc., Collegeville, PA 19426, USA
| | - Jade King
- Clinical Research and Imaging Centre, UHBW NHS Trust, Bristol, Bristol, BS2, UK
| | - Anna Morley
- Academic Respiratory Unit, Southmead Hospital, Bristol, Bristol, BS15, UK
| | - Madeleine Clout
- Bristol Vaccine Centre and Population Health Sciences, University of Bristol, Bristol, BS2, UK
| | - Nick Maskell
- Academic Respiratory Unit, University of Bristol, Southmead Hospital, Bristol, Bristol, BS15, UK
| | - Luis Jodar
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer Inc., Collegeville, PA 19426, USA
| | - Jennifer Oliver
- Bristol Vaccine Centre and Population Health Sciences, University of Bristol, Bristol, BS2, UK
| | | | - John M McLaughlin
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer Inc., Collegeville, PA 19426, USA
| | - Bradford D Gessner
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer Inc., Collegeville, PA 19426, USA
| | - Adam Finn
- Bristol Vaccine Centre, Cellular and Molecular Medicine and Population Health Sciences, University of Bristol, Bristol, BS2, UK
| | - Leon Danon
- Engineering Mathematics, University of Bristol, Bristol, Bristol, BS8, UK
| | - James W Dodd
- Academic Respiratory Unit and Population Health Sciences, University of Bristol, Southmead Hospital, Bristol, BS15, UK
| | - The Avon CAP Research Group
- Academic Respiratory Unit and Bristol Vaccine Centre, University of Bristol, Bristol, BS15, UK
- Engineering Mathematics, University of Bristol, Bristol, Bristol, BS8, UK
- Academic Respiratory Unit, University of Bristol, Southmead Hospital, Bristol, Bristol, BS15, UK
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer Inc., Collegeville, PA 19426, USA
- Clinical Research and Imaging Centre, UHBW NHS Trust, Bristol, Bristol, BS2, UK
- Academic Respiratory Unit, Southmead Hospital, Bristol, Bristol, BS15, UK
- Bristol Vaccine Centre and Population Health Sciences, University of Bristol, Bristol, BS2, UK
- Vaccines Medical Affairs, Pfizer Ltd, Tadworth, KT20, UK
- Bristol Vaccine Centre, Cellular and Molecular Medicine and Population Health Sciences, University of Bristol, Bristol, BS2, UK
- Academic Respiratory Unit and Population Health Sciences, University of Bristol, Southmead Hospital, Bristol, BS15, UK
| |
Collapse
|
29
|
Almond M, Farne HA, Jackson MM, Jha A, Katsoulis O, Pitts O, Tunstall T, Regis E, Dunning J, Byrne AJ, Mallia P, Kon OM, Saunders KA, Simpson KD, Snelgrove RJ, Openshaw PJM, Edwards MR, Barclay WS, Heaney LM, Johnston SL, Singanayagam A. Obesity dysregulates the pulmonary antiviral immune response. Nat Commun 2023; 14:6607. [PMID: 37857661 PMCID: PMC10587167 DOI: 10.1038/s41467-023-42432-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023] Open
Abstract
Obesity is a well-recognized risk factor for severe influenza infections but the mechanisms underlying susceptibility are poorly understood. Here, we identify that obese individuals have deficient pulmonary antiviral immune responses in bronchoalveolar lavage cells but not in bronchial epithelial cells or peripheral blood dendritic cells. We show that the obese human airway metabolome is perturbed with associated increases in the airway concentrations of the adipokine leptin which correlated negatively with the magnitude of ex vivo antiviral responses. Exogenous pulmonary leptin administration in mice directly impaired antiviral type I interferon responses in vivo and ex vivo in cultured airway macrophages. Obese individuals hospitalised with influenza showed dysregulated upper airway immune responses. These studies provide insight into mechanisms driving propensity to severe influenza infections in obesity and raise the potential for development of leptin manipulation or interferon administration as novel strategies for conferring protection from severe infections in obese higher risk individuals.
Collapse
Affiliation(s)
- Mark Almond
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Hugo A Farne
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Millie M Jackson
- Centre for Bacterial Resistance Biology. Section of Molecular Microbiology. Department of Infectious Disease, Imperial College London, London, UK
| | - Akhilesh Jha
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Orestis Katsoulis
- Centre for Bacterial Resistance Biology. Section of Molecular Microbiology. Department of Infectious Disease, Imperial College London, London, UK
| | - Oliver Pitts
- Centre for Bacterial Resistance Biology. Section of Molecular Microbiology. Department of Infectious Disease, Imperial College London, London, UK
| | | | - Eteri Regis
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Jake Dunning
- Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | - Adam J Byrne
- National Heart and Lung Institute, Imperial College London, London, UK
- School of Medicine and Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, 4, Ireland
| | - Patrick Mallia
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Onn Min Kon
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | | | | | - Michael R Edwards
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Wendy S Barclay
- Section of Virology, Department of Infectious Disease, Imperial College London, London, UK
| | - Liam M Heaney
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | | | - Aran Singanayagam
- Centre for Bacterial Resistance Biology. Section of Molecular Microbiology. Department of Infectious Disease, Imperial College London, London, UK.
| |
Collapse
|
30
|
Alipour Khabir Y, Alipour Khabir S, Anari H, Mohammadzadeh B, Hoseininia S, Aslani MR. Chest computed tomography severity score is a reliable predictor of mortality in patients with chronic obstructive pulmonary disease co-infected with COVID-19. Eur J Med Res 2023; 28:346. [PMID: 37715265 PMCID: PMC10503086 DOI: 10.1186/s40001-023-01336-8] [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: 03/09/2023] [Accepted: 09/01/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) pandemic is considered a global health crisis. The data related to chronic obstructive pulmonary disease (COPD) patients with COVID-19 are incomplete, especially the findings of the chest computed tomography (CT). The aim of the current study was to investigate the severity of the disease of COVID-19 in patients with COPD based on CT severity score and to evaluate its predictive power in the mortality of patients. METHODS In a retrospective study, demographic, clinical, and CT scan findings of COPD patients with COVID-19 were extracted from March 2020 to February 2022. CT severity score was determined based on the extent and nature of involvement of lungs in CT scan findings. By performing receiver operating characteristics (ROC) and Kaplan-Meier survival analysis were determined the disease severity and survival probability. RESULTS The most frequent radiological findings in chest CT scan included ground glass opacities (89.3%), consolidations (51.8%), crazy-paving pattern (46.4%), and septal thickening (35.7%). The mean CT severity score of deceased patients (34.61 ± 18.73) was significantly higher than recovered patients (16.71 ± 14.01, p < 0.001). Based on the ROC and Kaplan-Meier survival curves, it was revealed that CT severity score was a valuable criteria in the diagnosis of mortality in COPD patients with COVID-19. CONCLUSION The findings of this study revealed that the CT severity scoring in COPD patients with COVID-19 was valuable in identifying poor prognosis, although further studies are needed.
Collapse
Affiliation(s)
- Yalda Alipour Khabir
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Sevda Alipour Khabir
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hassan Anari
- Department of Radiology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Bahman Mohammadzadeh
- Department of Radiology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Saeed Hoseininia
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Reza Aslani
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Physiology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
31
|
Wang Y, Ninaber DK, Faiz A, van der Linden AC, van Schadewijk A, Lutter R, Hiemstra PS, van der Does AM, Ravi A. Acute cigarette smoke exposure leads to higher viral infection in human bronchial epithelial cultures by altering interferon, glycolysis and GDF15-related pathways. Respir Res 2023; 24:207. [PMID: 37612597 PMCID: PMC10464373 DOI: 10.1186/s12931-023-02511-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/13/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Acute exacerbations of chronic inflammatory lung diseases, such as chronic obstructive pulmonary disease (COPD), are frequently associated with rhinovirus (RV) infections. Despite these associations, the pathogenesis of virus-induced exacerbations is incompletely understood. We aimed to investigate effects of cigarette smoke (CS), a primary risk factor for COPD, on RV infection in airway epithelium and identify novel mechanisms related to these effects. METHODS Primary bronchial epithelial cells (PBEC) from COPD patients and controls were differentiated by culture at the air-liquid interface (ALI) and exposed to CS and RV-A16. Bulk RNA sequencing was performed using samples collected at 6 and 24 h post infection (hpi), and viral load, mediator and L-lactate levels were measured at 6, 24 and 48hpi. To further delineate the effect of CS on RV-A16 infection, we performed growth differentiation factor 15 (GDF15) knockdown, L-lactate and interferon pre-treatment in ALI-PBEC. We performed deconvolution analysis to predict changes in the cell composition of ALI-PBEC after the various exposures. Finally, we compared transcriptional responses of ALI-PBEC to those in nasal epithelium after human RV-A16 challenge. RESULTS CS exposure impaired antiviral responses at 6hpi and increased viral replication at 24 and 48hpi in ALI-PBEC. At 24hpi, CS exposure enhanced expression of RV-A16-induced epithelial interferons, inflammation-related genes and CXCL8. CS exposure increased expression of oxidative stress-related genes, of GDF15, and decreased mitochondrial membrane potential. GDF15 knockdown experiments suggested involvement of this pathway in the CS-induced increase in viral replication. Expression of glycolysis-related genes and L-lactate production were increased by CS exposure, and was demonstrated to contribute to higher viral replication. No major differences were demonstrated between COPD and non-COPD-derived cultures. However, cellular deconvolution analysis predicted higher secretory cells in COPD-derived cultures at baseline. CONCLUSION Altogether, our findings demonstrate that CS exposure leads to higher viral infection in human bronchial epithelium by altering not only interferon responses, but likely also through a switch to glycolysis, and via GDF15-related pathways.
Collapse
Affiliation(s)
- Ying Wang
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Dennis K Ninaber
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Alen Faiz
- Respiratory Bioinformatics and Molecular Biology (RBMB), School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW, 2007, Australia
| | - Abraham C van der Linden
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Annemarie van Schadewijk
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - René Lutter
- Department of Pulmonary Medicine, Amsterdam University Medical Center, University of Amsterdam, 1081HV, Amsterdam, The Netherlands
| | - Pieter S Hiemstra
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Anne M van der Does
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Abilash Ravi
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
32
|
Pratt AJ, Purssell A, Zhang T, Luks VPJ, Bauza X, Mulpuru S, Kirby M, Aaron SD, Cowan J. Complexity in clinical diagnoses of acute exacerbation of chronic obstructive pulmonary disease. BMC Pulm Med 2023; 23:298. [PMID: 37580731 PMCID: PMC10426055 DOI: 10.1186/s12890-023-02587-1] [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: 05/14/2023] [Accepted: 07/28/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Acute exacerbation of chronic obstructive pulmonary disease (AECOPD) is a clinical syndrome with various causes. It is not uncommon that COPD patients presenting with dyspnea have multiple causes for their symptoms including AECOPD, pneumonia, or congestive heart failure occurring concurrently. METHODS To identify clinical, radiographic, and laboratory characteristics that might help distinguish AECOPD from another dominant disease in patients with a history of COPD, we conducted a retrospective cohort study of hospitalized patients with admitting diagnosis of AECOPD who were screened for a prospective randomized controlled trial from Sep 2016 to Mar 2018. Clinical characteristics, course in hospital, and final diagnosis at discharge were reviewed and adjudicated by two authors. The final diagnosis of each patient was determined based on the synthesis of all presenting signs and symptoms, imaging, and laboratory results. We adhered to AECOPD diagnosis definitions based on the GOLD guidelines. Univariate and multivariate analyses were performed to identify any associated features of AECOPD with and without other acute processes contributing to dyspnea. RESULTS Three hundred fifteen hospitalized patients with admitting diagnosis of AECOPD were included. Mean age was 72.5 (SD 10.6) years. Two thirds (65.4%) had spirometry defined COPD. The most common presenting symptom was dyspnea (96.5%), followed by cough (67.9%), and increased sputum (57.5%). One hundred and eighty (57.1%) had a final diagnosis of AECOPD alone whereas 87 (27.6%) had AECOPD with other conditions and 48 (15.2%) did not have AECOPD after adjudication. Increased sputum purulence (OR 3.35, 95%CI 1.68-6.69) and elevated venous pCO2 (OR 1.04, 95%CI 1.01 - 1.07) were associated with a diagnosis of AECOPD but these were not associated with AECOPD alone without concomitant conditions. Radiographic evidence of pleural effusion (OR 0.26, 95%CI 0.12 - 0.58) was negatively associated with AECOPD with or without other conditions while radiographic evidence of pulmonary edema (OR 0.31; 95%CI 0.11 - 0.91) and lobar pneumonia (OR 0.13, 95%CI 0.07 - 0.25) suggested against the diagnosis of AECOPD alone. CONCLUSION The study highlighted the complexity and difficulty of AECOPD diagnosis. A more specific clinical tool to diagnose AECOPD is needed.
Collapse
Affiliation(s)
| | - Andrew Purssell
- Division of Infectious Diseases, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Tinghua Zhang
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Vanessa P J Luks
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Division of Respirology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Xavier Bauza
- Department of Physics, Toronto Metropolitan University, Ottawa, ON, Canada
| | - Sunita Mulpuru
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Division of Respirology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Miranda Kirby
- Department of Physics, Toronto Metropolitan University, Ottawa, ON, Canada
| | - Shawn D Aaron
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Division of Respirology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Juthaporn Cowan
- Division of Infectious Diseases, Department of Medicine, University of Ottawa, Ottawa, ON, Canada.
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
33
|
Awatade NT, Wark PAB, Chan ASL, Mamun SMAA, Mohd Esa NY, Matsunaga K, Rhee CK, Hansbro PM, Sohal SS. The Complex Association between COPD and COVID-19. J Clin Med 2023; 12:jcm12113791. [PMID: 37297985 DOI: 10.3390/jcm12113791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is significant cause of morbidity and mortality worldwide. There is mounting evidence suggesting that COPD patients are at increased risk of severe COVID-19 outcomes; however, it remains unclear whether they are more susceptible to acquiring SARS-CoV-2 infection. In this comprehensive review, we aim to provide an up-to-date perspective of the intricate relationship between COPD and COVID-19. We conducted a thorough review of the literature to examine the evidence regarding the susceptibility of COPD patients to COVID-19 infection and the severity of their disease outcomes. While most studies have found that pre-existing COPD is associated with worse COVID-19 outcomes, some have yielded conflicting results. We also discuss confounding factors such as cigarette smoking, inhaled corticosteroids, and socioeconomic and genetic factors that may influence this association. Furthermore, we review acute COVID-19 management, treatment, rehabilitation, and recovery in COPD patients and how public health measures impact their care. In conclusion, while the association between COPD and COVID-19 is complex and requires further investigation, this review highlights the need for careful management of COPD patients during the pandemic to minimize the risk of severe COVID-19 outcomes.
Collapse
Affiliation(s)
- Nikhil T Awatade
- Immune Health Program, Hunter Medical Research Institute and University of Newcastle, Newcastle 2305, Australia
| | - Peter A B Wark
- Immune Health Program, Hunter Medical Research Institute and University of Newcastle, Newcastle 2305, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle 2305, Australia
| | - Andrew S L Chan
- Department of Respiratory and Sleep Medicine, Royal North Shore Hospital, St. Leonards 2065, Australia
- Northern Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
| | - S M Abdullah Al Mamun
- Department of Respiratory Medicine & Sleep Medicine, Evercare Hospitals Dhaka, Dhaka 1229, Bangladesh
| | | | - Kazuto Matsunaga
- Department of Respiratory Medicine and Infectious Disease Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-kogushi, Ube 755-8505, Japan
| | - Chin Kook Rhee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Philip M Hansbro
- Immune Health Program, Hunter Medical Research Institute and University of Newcastle, Newcastle 2305, Australia
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney 2050, Australia
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston 7248, Australia
| |
Collapse
|
34
|
Yang N, Zhang L, Tian D, Wang P, Men K, Ge Y, Zhang C. Tanshinone increases Hemopexin expression in lung cells and macrophages to protect against cigarette smoke-induced COPD and enhance antiviral responses. Cell Cycle 2023; 22:645-665. [PMID: 36218263 PMCID: PMC9980497 DOI: 10.1080/15384101.2022.2129933] [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: 05/10/2022] [Revised: 09/01/2022] [Accepted: 09/26/2022] [Indexed: 11/03/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease, while respiratory infections can elicit exacerbations in COPD patients to mediate increased mortality. Administration of Tanshinones (TS) derivatives has been demonstrated to protect against cigarette smoking (CS) and lipopolysaccharide (LPS)-induced COPD progression. However, the underlying molecular mechanisms and the roles of TS in mitigating the severity of viral-mediated exacerbations of COPD have not been elucidated. Here, we found that TS treatments significantly attenuated lung function decline, inflammatory responses and oxidative stress in CS and LPS-induced COPD mice. Subsequent RNA-seq analysis revealed significantly upregulated Hemopexin expression and enriched interferons (IFNs) signaling pathways in lung tissues of COPD mice upon TS treatments. Moreover, TS administration demonstrated Hemopexin-dependent beneficial roles in BEAS-2B lung cells and RAW264.7 macrophages, which was associated with the suppression of oxidative stress and ERK, NF-κB, and NLRP3 inflammasome signaling pathways-mediated inflammation. Furthermore, TS promoted IFN signaling and rescued impaired antiviral responses in CS and LPS-exposed lung cells that were infected by influenza virus. Notably, hemopexin over-expression in lung cells and macrophages recapitulated the pharmacological activities of TS. Taken together, these results indicate that TS administration is a promising and potential therapeutic strategy for treating COPD and preventing COPD exacerbations.
Collapse
Affiliation(s)
- Ning Yang
- Department of Respiratory, the Affiliated Hospital of Yan’an University, Yan’an, Shaanxi Province, China
| | - Liang Zhang
- Department of Respiratory, the Affiliated Hospital of Yan’an University, Yan’an, Shaanxi Province, China
| | - Dongdong Tian
- Department of Respiratory, the Affiliated Hospital of Yan’an University, Yan’an, Shaanxi Province, China
| | - Ping Wang
- Department of Respiratory, the Affiliated Hospital of Yan’an University, Yan’an, Shaanxi Province, China
| | - Kai Men
- Department of Respiratory, the Affiliated Hospital of Yan’an University, Yan’an, Shaanxi Province, China
| | - Yiliang Ge
- Hengyang Medical School, University of South China, Hunan
| | - Cailian Zhang
- Department of Respiratory, the Affiliated Hospital of Yan’an University, Yan’an, Shaanxi Province, China
| |
Collapse
|
35
|
Nourian YH, Salimian J, Ahmadi A, Salehi Z, Karimi M, Emamvirdizadeh A, Azimzadeh Jamalkandi S, Ghanei M. cAMP-PDE signaling in COPD: Review of cellular, molecular and clinical features. Biochem Biophys Rep 2023; 34:101438. [PMID: 36865738 PMCID: PMC9971187 DOI: 10.1016/j.bbrep.2023.101438] [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: 11/26/2022] [Revised: 01/21/2023] [Accepted: 02/02/2023] [Indexed: 02/18/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the fourth leading cause of death among non-contagious diseases in the world. PDE inhibitors are among current medicines prescribed for COPD treatment of which, PDE-4 family is the predominant PDE isoform involved in hydrolyzing cyclic adenosine monophosphate (cAMP) that regulates the inflammatory responses in neutrophils, lymphocytes, macrophages and epithelial cells The aim of this study is to investigate the cellular and molecular mechanisms of cAMP-PDE signaling, as an important pathway in the treatment management of patients with COPD. In this review, a comprehensive literature review was performed about the effect of PDEs in COPD. Generally, PDEs are overexpressed in COPD patients, resulting in cAMP inactivation and decreased cAMP hydrolysis from AMP. At normal amounts, cAMP is one of the essential agents in regulating metabolism and suppressing inflammatory responses. Low amount of cAMP lead to activation of downstream inflammatory signaling pathways. PDE4 and PDE7 mRNA transcript levels were not altered in polymorphonuclear leukocytes and CD8 lymphocytes originating from the peripheral venous blood of stable COPD subjects compared to healthy controls. Therefore, cAMP-PDE signaling pathway is one of the most important signaling pathways involved in COPD. By examining the effects of different drugs in this signaling pathway critical steps can be taken in the treatment of this disease.
Collapse
Affiliation(s)
- Yazdan Hasani Nourian
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Salimian
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Ahmadi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Karimi
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Emamvirdizadeh
- Department of Molecular Genetics, Faculty of Bio Sciences, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Sadegh Azimzadeh Jamalkandi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran,Corresponding author.
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Antunes KH, Singanayagam A, Williams L, Faiez TS, Farias A, Jackson MM, Faizi FK, Aniscenko J, Kebadze T, Chander Veerati P, Wood L, Bartlett NW, Duarte de Souza AP, Johnston SL. Airway-delivered short-chain fatty acid acetate boosts antiviral immunity during rhinovirus infection. J Allergy Clin Immunol 2023; 151:447-457.e5. [PMID: 36216081 DOI: 10.1016/j.jaci.2022.09.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 08/03/2022] [Accepted: 09/09/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Microbiota are recognized to play a major role in regulation of immunity through release of immunomodulatory metabolites such as short-chain fatty acids (SCFAs). Rhinoviruses (RVs) induce upper respiratory tract illnesses and precipitate exacerbations of asthma and chronic obstructive pulmonary disease through poorly understood mechanisms. Local interactions between SCFAs and antiviral immune responses in the respiratory tract have not been previously investigated. OBJECTIVE We sought to investigate whether pulmonary metabolite manipulation through lung-delivered administration of SCFAs can modulate antiviral immunity to RV infection. METHODS We studied the effects of intranasal administration of the SCFAs acetate, butyrate, and propionate on basal expression of antiviral signatures, and of acetate in a mouse model of RV infection and in RV-infected lung epithelial cell lines. We additionally assessed the effects of acetate, butyrate, and propionate on RV infection in differentiated human primary bronchial epithelial cells. RESULTS Intranasal acetate administration induced basal upregulation of IFN-β, an effect not observed with other SCFAs. Butyrate induced RIG-I expression. Intranasal acetate treatment of mice increased interferon-stimulated gene and IFN-λ expression during RV infection and reduced lung virus loads at 8 hours postinfection. Acetate ameliorated virus-induced proinflammatory responses with attenuated pulmonary mucin and IL-6 expression observed at day 4 and 6 postinfection. This interferon-enhancing effect of acetate was confirmed in human bronchial and alveolar epithelial cell lines. In differentiated primary bronchial epithelial cells, butyrate treatment better modulated IFN-β and IFN-λ gene expression during RV infection. CONCLUSIONS SCFAs augment antiviral immunity and reduce virus load and proinflammatory responses during RV infection.
Collapse
Affiliation(s)
- Krist Helen Antunes
- Laboratory of Clinical and Experimental Immunology - Pontifical Catholic University of Rio Grande do Sul, Porto Alegre; National Heart and Lung Institute and, Department of Infectious Disease, Imperial College London, London
| | - Aran Singanayagam
- Centre for Molecular Bacteriology and Infection, Department of Infectious Disease, Imperial College London, London
| | - Lily Williams
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle
| | - Tasnim Syakirah Faiez
- Centre for Molecular Bacteriology and Infection, Department of Infectious Disease, Imperial College London, London
| | - Ana Farias
- National Heart and Lung Institute and, Department of Infectious Disease, Imperial College London, London
| | - Millie M Jackson
- Centre for Molecular Bacteriology and Infection, Department of Infectious Disease, Imperial College London, London
| | - Fatima K Faizi
- Centre for Molecular Bacteriology and Infection, Department of Infectious Disease, Imperial College London, London
| | - Julia Aniscenko
- National Heart and Lung Institute and, Department of Infectious Disease, Imperial College London, London
| | - Tatiana Kebadze
- National Heart and Lung Institute and, Department of Infectious Disease, Imperial College London, London
| | | | - Lisa Wood
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle
| | - Nathan W Bartlett
- National Heart and Lung Institute and, Department of Infectious Disease, Imperial College London, London; School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle
| | - Ana Paula Duarte de Souza
- Laboratory of Clinical and Experimental Immunology - Pontifical Catholic University of Rio Grande do Sul, Porto Alegre; National Heart and Lung Institute and, Department of Infectious Disease, Imperial College London, London.
| | - Sebastian L Johnston
- National Heart and Lung Institute and, Department of Infectious Disease, Imperial College London, London; Asthma UK Centre in Allergic Mechanisms of Asthma, London.
| |
Collapse
|
37
|
Zsichla L, Müller V. Risk Factors of Severe COVID-19: A Review of Host, Viral and Environmental Factors. Viruses 2023; 15:175. [PMID: 36680215 PMCID: PMC9863423 DOI: 10.3390/v15010175] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
The clinical course and outcome of COVID-19 are highly variable, ranging from asymptomatic infections to severe disease and death. Understanding the risk factors of severe COVID-19 is relevant both in the clinical setting and at the epidemiological level. Here, we provide an overview of host, viral and environmental factors that have been shown or (in some cases) hypothesized to be associated with severe clinical outcomes. The factors considered in detail include the age and frailty, genetic polymorphisms, biological sex (and pregnancy), co- and superinfections, non-communicable comorbidities, immunological history, microbiota, and lifestyle of the patient; viral genetic variation and infecting dose; socioeconomic factors; and air pollution. For each category, we compile (sometimes conflicting) evidence for the association of the factor with COVID-19 outcomes (including the strength of the effect) and outline possible action mechanisms. We also discuss the complex interactions between the various risk factors.
Collapse
Affiliation(s)
- Levente Zsichla
- Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary
- National Laboratory for Health Security, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Viktor Müller
- Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary
- National Laboratory for Health Security, Eötvös Loránd University, 1117 Budapest, Hungary
| |
Collapse
|
38
|
Kapusta J, Chudzik M, Kałuzińska-Kołat Ż, Kołat D, Burzyńska M, Jankowski P, Babicki M. Do selected lifestyle parameters affect the severity and symptoms of COVID-19 among elderly patients? The retrospective evaluation of individuals from the STOP-COVID registry of the PoLoCOV study. J Infect Public Health 2023; 16:143-153. [PMID: 36521330 PMCID: PMC9743693 DOI: 10.1016/j.jiph.2022.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/30/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Older individuals tend to include less physical activity in their routine and are more prone to chronic diseases and severe medical complications, making them the most burdened group that is losing years of life due to pandemic-related premature mortality. This research aimed to assess the lifestyle factors that affect the COVID-19 course among patients ≥ 65 years old. METHODS The study included 568 convalescents (64.1% women and 35.9% men) with persistent clinical symptoms after isolation. The mean age was 70.41 ± 4.64 years (minimum: 65 years; maximum: 89 years). The patients completed the questionnaire during their in-person visit to the medical center. The survey included questions regarding their health status when suffering from COVID-19, basic sociodemographic data, and medical history concerning chronic conditions and lifestyle. RESULTS Physical inactivity (p < 0.001) and feeling nervous (p = 0.026) increased the risk of having a severe disease course. Coronary artery disease raised both the risk of a severe disease course (p = 0.002) and the number of present symptoms up to 4 weeks (p = 0.039). Sleep disturbances increased the number of symptoms during infection (p = 0.001). The occurrence of any symptoms was also associated with the female sex (p = 0.004). The severity of the course was associated with longer persistent symptoms (p < 0.001) and a greater number of symptoms (p = 0.004); those with a more severe course were also at a greater risk of persistent symptoms for up to 4 weeks (p = 0.006). Senior citizens in the third pandemic wave suffered with more severe disease (p = 0.004), while illness during the fourth (p = 0.001) and fifth (p < 0.001) waves was associated with a lower risk of persistent symptoms for up to 4 weeks. The disease duration was significantly shorter among vaccinated patients (p = 0.042). CONCLUSIONS Elderly COVID-19 patients should re-think their lifestyle habits to consider a physical activity level that is adjusted to their abilities, in order to decrease the risk of a severe disease course and to further limit both the number and duration of symptoms. The research was carried out in accordance with the Declaration of Helsinki, and approval from the Bioethics Committee of Lodz Regional Medical Chamber to conduct the study was obtained (approval number 0115/2021). The PoLoCOV-Study ClinicalTrials.gov identifier is NCT05018052.
Collapse
Affiliation(s)
- Joanna Kapusta
- Department of Internal Medicine and Cardiac Rehabilitation, Medical University of Lodz, 70–445 Lodz, Poland,Corresponding author
| | - Michał Chudzik
- Department of Internal Medicine and Geriatric Cardiology, Medical Centre for Postgraduate Education, 01–813 Warsaw, Poland,Boruta Medical Center, 95–100 Zgierz, Poland,Corresponding author at: Department of Internal Medicine and Geriatric Cardiology, Medical Centre for Postgraduate Education, 01–813 Warsaw, Poland
| | - Żaneta Kałuzińska-Kołat
- Boruta Medical Center, 95–100 Zgierz, Poland,Department of Experimental Surgery, Medical University of Lodz, 90–136 Lodz, Poland
| | - Damian Kołat
- Boruta Medical Center, 95–100 Zgierz, Poland,Department of Experimental Surgery, Medical University of Lodz, 90–136 Lodz, Poland
| | - Monika Burzyńska
- Department of Epidemiology and Biostatistics, Social and Preventive Medicine of the Medical University of Lodz, 90–752 Lodz, Poland
| | - Piotr Jankowski
- Department of Internal Medicine and Geriatric Cardiology, Medical Centre for Postgraduate Education, 01–813 Warsaw, Poland
| | - Mateusz Babicki
- Department of Family Medicine, Wroclaw Medical University, 51–141 Wroclaw, Poland
| |
Collapse
|
39
|
The establishment of COPD organoids to study host-pathogen interaction reveals enhanced viral fitness of SARS-CoV-2 in bronchi. Nat Commun 2022; 13:7635. [PMID: 36496442 PMCID: PMC9735280 DOI: 10.1038/s41467-022-35253-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterised by airflow limitation and infective exacerbations, however, in-vitro model systems for the study of host-pathogen interaction at the individual level are lacking. Here, we describe the establishment of nasopharyngeal and bronchial organoids from healthy individuals and COPD that recapitulate disease at the individual level. In contrast to healthy organoids, goblet cell hyperplasia and reduced ciliary beat frequency were observed in COPD organoids, hallmark features of the disease. Single-cell transcriptomics uncovered evidence for altered cellular differentiation trajectories in COPD organoids. SARS-CoV-2 infection of COPD organoids revealed more productive replication in bronchi, the key site of infection in severe COVID-19. Viral and bacterial exposure of organoids induced greater pro-inflammatory responses in COPD organoids. In summary, we present an organoid model that recapitulates the in vivo physiological lung microenvironment at the individual level and is amenable to the study of host-pathogen interaction and emerging infectious disease.
Collapse
|
40
|
Cecchini A, Othman A, Kaur K, Richardson A, Cecchini A. Enterovirus-Human-Rhinovirus Infection Leading to Acute Respiratory Distress Syndrome: A Case Report. Cureus 2022; 14:e31615. [DOI: 10.7759/cureus.31615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2022] [Indexed: 11/18/2022] Open
|
41
|
Johansen MD, Mahbub RM, Idrees S, Nguyen DH, Miemczyk S, Pathinayake P, Nichol K, Hansbro NG, Gearing LJ, Hertzog PJ, Gallego-Ortega D, Britton WJ, Saunders BM, Wark PA, Faiz A, Hansbro PM. Increased SARS-CoV-2 Infection, Protease, and Inflammatory Responses in Chronic Obstructive Pulmonary Disease Primary Bronchial Epithelial Cells Defined with Single-Cell RNA Sequencing. Am J Respir Crit Care Med 2022; 206:712-729. [PMID: 35549656 PMCID: PMC9799113 DOI: 10.1164/rccm.202108-1901oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 05/12/2022] [Indexed: 01/01/2023] Open
Abstract
Rationale: Patients with chronic obstructive pulmonary disease (COPD) develop more severe coronavirus disease (COVID-19); however, it is unclear whether they are more susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and what mechanisms are responsible for severe disease. Objectives: To determine whether SARS-CoV-2 inoculated primary bronchial epithelial cells (pBECs) from patients with COPD support greater infection and elucidate the effects and mechanisms involved. Methods: We performed single-cell RNA sequencing analysis on differentiated pBECs from healthy subjects and patients with COPD 7 days after SARS-CoV-2 inoculation. We correlated changes with viral titers, proinflammatory responses, and IFN production. Measurements and Main Results: Single-cell RNA sequencing revealed that COPD pBECs had 24-fold greater infection than healthy cells, which was supported by plaque assays. Club/goblet and basal cells were the predominant populations infected and expressed mRNAs involved in viral replication. Proteases involved in SARS-CoV-2 entry/infection (TMPRSS2 and CTSB) were increased, and protease inhibitors (serpins) were downregulated more so in COPD. Inflammatory cytokines linked to COPD exacerbations and severe COVID-19 were increased, whereas IFN responses were blunted. Coexpression analysis revealed a prominent population of club/goblet cells with high type 1/2 IFN responses that were important drivers of immune responses to infection in both healthy and COPD pBECs. Therapeutic inhibition of proteases and inflammatory imbalances reduced viral titers and cytokine responses, particularly in COPD pBECs. Conclusions: COPD pBECs are more susceptible to SARS-CoV-2 infection because of increases in coreceptor expression and protease imbalances and have greater inflammatory responses. A prominent cluster of IFN-responsive club/goblet cells emerges during infection, which may be important drivers of immunity. Therapeutic interventions suppress SARS-CoV-2 replication and consequent inflammation.
Collapse
Affiliation(s)
- Matt D. Johansen
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Rashad M. Mahbub
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Sobia Idrees
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Duc H. Nguyen
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Stefan Miemczyk
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Prabuddha Pathinayake
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Kristy Nichol
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Nicole G. Hansbro
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Linden J. Gearing
- Department of Molecular and Translational Sciences, School of Clinical Sciences at Monash Health, Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Paul J. Hertzog
- Department of Molecular and Translational Sciences, School of Clinical Sciences at Monash Health, Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - David Gallego-Ortega
- Faculty of Engineering and Information Technology, School of Biomedical Engineering, Centre for Single Cell Technology, University of Technology Sydney, Ultimo, New South Wales, Australia
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales Sydney, Kensington, New South Wales, Australia; and
| | - Warwick J. Britton
- Centenary Institute, University of Sydney and Department of Clinical Immunology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Bernadette M. Saunders
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Peter A. Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Alen Faiz
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Philip M. Hansbro
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
42
|
Conway FM, Bloom CI, Shah PL. Susceptibility of Patients with Airway Disease to SARS-CoV-2 Infection. Am J Respir Crit Care Med 2022; 206:696-703. [PMID: 35549839 PMCID: PMC9799128 DOI: 10.1164/rccm.202111-2547pp] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Francesca M. Conway
- Royal Brompton Hospital, London, United Kingdom;,Chelsea & Westminster Hospital, London, United Kingdom; and,National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Chloe I. Bloom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pallav L. Shah
- Royal Brompton Hospital, London, United Kingdom;,Chelsea & Westminster Hospital, London, United Kingdom; and,National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
43
|
Kartsiouni E, Chatzipanagiotou S, Tamvakeras P, Douros K. The role of viral infections in pulmonary exacerbations of patients with non-cystic fibrosis bronchiectasis: A systematic review. Respir Investig 2022; 60:625-632. [PMID: 35811289 DOI: 10.1016/j.resinv.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Bronchiectasis is a cause of increased morbidity of the respiratory system. Exacerbations among patients with non-CF (cystic fibrosis) bronchiectasis result in reduced pulmonary function and poor quality of life. While the role of bacteria in triggering exacerbations in patients with non- CF bronchiectasis has been well studied, little is known about viral infections in these patients. We aimed to review the evidence on the role of respiratory viruses in the exacerbations of non-CF bronchiectasis. METHODS Relevant literature was searched on the MEDLINE/PubMed database. Seven studies satisfied the criteria and were included in this review. RESULTS According to the included articles, respiratory viruses are often identified in exacerbations of patients with non-CF bronchiectasis with the most frequent being human rhinovirus and influenza viruses. When a virus is isolated during an exacerbation patients have more symptoms from the upper respiratory tract. One study showed that detection of Epstein- Barr virus among patients with non-CF bronchiectasis is correlated with faster reduction of pulmonary function and progression of the disease. CONCLUSION Viruses seem to have a role in the exacerbation of patients with non-CF bronchiectasis. However, the exact nature and importance of this role remain elusive. Viruses are also isolated during the stable period of the disease. Further well-designed studies are necessary to clarify this complex issue.
Collapse
Affiliation(s)
- Elpiniki Kartsiouni
- Pediatric Allergy and Respiratory Unit, 3rd Department of Pediatrics, "Attikon" University Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Stylianos Chatzipanagiotou
- Department of Medical Biopathology, National and Kapodistrian University of Athens, School of Medicine, "Eginition" Hospital, Athens, Greece
| | | | - Konstantinos Douros
- Pediatric Allergy and Respiratory Unit, 3rd Department of Pediatrics, "Attikon" University Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.
| |
Collapse
|
44
|
Wu W, Alexander JS, Metcalf JP. In Vivo and In Vitro Studies of Cigarette Smoke Effects on Innate Responses to Influenza Virus: A Matter of Models? Viruses 2022; 14:1824. [PMID: 36016446 PMCID: PMC9415757 DOI: 10.3390/v14081824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 11/26/2022] Open
Abstract
Cigarette smoke (CS) is a significant public health problem and a leading risk factor for the development of chronic obstructive pulmonary disease (COPD) in the developed world. Respiratory viral infections, such as the influenza A virus (IAV), are associated with acute exacerbations of COPD and are more severe in cigarette smokers. To fight against viral infection, the host has developed an innate immune system, which has complicated mechanisms regulating the expression and activation of cytokines and chemokines to maximize the innate and adaptive antiviral response, as well as limiting the immunopathology that leads to exaggerated lung damage. In the case of IAV, responders include airway and alveolar epithelia, lung macrophages and dendritic cells. To achieve a successful infection, IAV must overcome these defenses. In this review, we summarize the detrimental role of CS in influenza infections. This includes both immunosuppressive and proinflammatory effects on innate immune responses during IAV infection. Some of the results, with respect to CS effects in mouse models, appear to have discordant results, which could be at least partially addressed by standardization of animal viral infection models to evaluate the effect of CS exposure in this context.
Collapse
Affiliation(s)
- Wenxin Wu
- Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jeremy S. Alexander
- Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jordan P. Metcalf
- Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Microbiology and Immunology, the University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
45
|
De Luca G, Nardin M, Algowhary M, Uguz B, Oliveira DC, Ganyukov V, Zimbakov Z, Cercek M, Okkels Jensen L, Loh PH, Calmac L, Roura Ferrer G, Quadros A, Milewski M, Scotto di Uccio F, von Birgelen C, Versaci F, Ten Berg J, Casella G, Wong Sung Lung A, Kala P, Díez Gil JL, Carrillo X, Dirksen M, Becerra-Munoz VM, Lee MKY, Arifa Juzar D, de Moura Joaquim R, Paladino R, Milicic D, Davlouros P, Bakraceski N, Zilio F, Donazzan L, Kraaijeveld A, Galasso G, Lux A, Marinucci L, Guiducci V, Menichelli M, Scoccia A, Yamac AH, Ugur Mert K, Flores Rios X, Kovarnik T, Kidawa M, Moreu J, Flavien V, Fabris E, Martínez-Luengas IL, Boccalatte M, Bosa Ojeda F, Arellano-Serrano C, Caiazzo G, Cirrincione G, Kao HL, Sanchis Forés J, Vignali L, Pereira H, Manzo S, Ordoñez S, Özkan AA, Scheller B, Lehtola H, Teles R, Mantis C, Antti Y, Brum Silveira JA, Zoni R, Bessonov I, Savonitto S, Kochiadakis G, Alexopoulos D, Uribe CE, Kanakakis J, Faurie B, Gabrielli G, Gutierrez Barrios A, Bachini JP, Rocha A, Tam FCC, Rodriguez A, Lukito AA, Saint-Joy V, Pessah G, Tuccillo A, Cortese G, Parodi G, Bouraghda MA, Kedhi E, Lamelas P, Suryapranata H, Verdoia M. Impact of chronic obstructive pulmonary disease on short-term outcome in patients with ST-elevation myocardial infarction during COVID-19 pandemic: insights from the international multicenter ISACS-STEMI registry. Respir Res 2022; 23:207. [PMID: 35971173 PMCID: PMC9376902 DOI: 10.1186/s12931-022-02128-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 05/02/2022] [Indexed: 11/10/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is projected to become the third cause of mortality worldwide. COPD shares several pathophysiological mechanisms with cardiovascular disease, especially atherosclerosis. However, no definite answers are available on the prognostic role of COPD in the setting of ST elevation myocardial infarction (STEMI), especially during COVID-19 pandemic, among patients undergoing primary angioplasty, that is therefore the aim of the current study. Methods In the ISACS-STEMI COVID-19 registry we included retrospectively patients with STEMI treated with primary percutaneous coronary intervention (PCI) between March and June of 2019 and 2020 from 109 high-volume primary PCI centers in 4 continents. Results A total of 15,686 patients were included in this analysis. Of them, 810 (5.2%) subjects had a COPD diagnosis. They were more often elderly and with a more pronounced cardiovascular risk profile. No preminent procedural dissimilarities were noticed except for a lower proportion of dual antiplatelet therapy at discharge among COPD patients (98.9% vs. 98.1%, P = 0.038). With regards to short-term fatal outcomes, both in-hospital and 30-days mortality occurred more frequently among COPD patients, similarly in pre-COVID-19 and COVID-19 era. However, after adjustment for main baseline differences, COPD did not result as independent predictor for in-hospital death (adjusted OR [95% CI] = 0.913[0.658–1.266], P = 0.585) nor for 30-days mortality (adjusted OR [95% CI] = 0.850 [0.620–1.164], P = 0.310). No significant differences were detected in terms of SARS-CoV-2 positivity between the two groups. Conclusion This is one of the largest studies investigating characteristics and outcome of COPD patients with STEMI undergoing primary angioplasty, especially during COVID pandemic. COPD was associated with significantly higher rates of in-hospital and 30-days mortality. However, this association disappeared after adjustment for baseline characteristics. Furthermore, COPD did not significantly affect SARS-CoV-2 positivity. Trial registration number: NCT 04412655 (2nd June 2020).
Collapse
Affiliation(s)
- Giuseppe De Luca
- Division of Clinical and Experimental Cardiology, AOU Sassari, Sassari, Italy. .,University of Sassari, Sassari, Italy.
| | - Matteo Nardin
- Third Medicine Division, ASST Spedali Civili, Brescia, Italy
| | - Magdy Algowhary
- Division of Cardiology, Assiut University Heart Hospital, Assiut University, Asyut, Egypt
| | - Berat Uguz
- Division of Cardiology, Bursa City Hospital, Bursa, Turkey
| | - Dinaldo C Oliveira
- Pronto de Socorro Cardiologico Prof. Luis Tavares, Centro PROCAPE, Federal University of Pernambuco, Recife, Brazil
| | - Vladimir Ganyukov
- Department of Heart and Vascular Surgery, State Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Zan Zimbakov
- Medical Faculty, University Clinic for Cardiology, Ss' Cyril and Methodius University, Skopje, North Macedonia
| | - Miha Cercek
- Centre for Intensive Internal Medicine, University Medical Centre, Ljubljana, Slovenia
| | | | - Poay Huan Loh
- Department of Cardiology, National University Hospital, Singapore, Singapore
| | - Lucian Calmac
- Clinic Emergency Hospital of Bucharest, Bucharest, Romania
| | - Gerard Roura Ferrer
- Interventional Cardiology Unit, Heart Disease Institute, Hospital Universitari de Bellvitge, Barcelona, Spain
| | | | - Marek Milewski
- Division of Cardiology, Medical University of Silezia, Katowice, Poland
| | | | - Clemens von Birgelen
- Department of Cardiology, Medisch Spectrum Twente, Thoraxcentrum Twente, Enschede, The Netherlands
| | - Francesco Versaci
- Division of Cardiology, Ospedale Santa Maria Goretti Latina, Latina, Italy
| | - Jurrien Ten Berg
- Division of Cardiology, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Gianni Casella
- Division of Cardiology, Ospedale Maggiore Bologna, Bologna, Italy
| | | | - Petr Kala
- University Hospital Brno, Medical Faculty of Masaryk University, Brno, Czech Republic
| | | | | | - Maurits Dirksen
- Division of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | | | - Michael Kang-Yin Lee
- Department of Cardiology, Queen Elizabeth Hospital, University of Hong Kong, Yau Ma Tei, Hong Kong
| | - Dafsah Arifa Juzar
- Department of Cardiology and Vascular Medicine, University of Indonesia National Cardiovascular Center "Harapan Kita", Jakarta, Indonesia
| | | | | | - Davor Milicic
- Department of Cardiology, University Hospital Centre, University of Zagreb, Zagreb, Croatia
| | - Periklis Davlouros
- Invasive Cardiology and Congenital Heart Disease, Patras University Hospital, Patras, Greece
| | | | - Filippo Zilio
- Division of Cardiology, Ospedale Santa Chiara di Trento, Trento, Italy
| | - Luca Donazzan
- Division of Cardiology, Ospedale "S. Maurizio" Bolzano, Bolzano, Italy
| | | | - Gennaro Galasso
- Division of Cardiology, Ospedale San Giovanni di Dio e Ruggi d'Aragona, Salerno, Italy
| | - Arpad Lux
- Maastricht University Medical Center, Maastricht, The Netherlands
| | - Lucia Marinucci
- Division of Cardiology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", Pesaro, Italy
| | - Vincenzo Guiducci
- Division of Cardiology, AUSL-IRCCS Reggio Emilia, Reggio Emilia, Italy
| | | | | | - Aylin Hatice Yamac
- Department of Cardiology, Hospital Bezmialem Vakıf University, İstanbul, Turkey
| | - Kadir Ugur Mert
- Division of Cardiology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | | | | | - Michal Kidawa
- Central Hospital of Medical University of Lodz, Lodz, Poland
| | - Josè Moreu
- Division of Cardiology, Complejo Hospitalario de Toledo, Toledo, Spain
| | - Vincent Flavien
- Division of Cardiology, Center Hospitalier Universitaire de Lille, Lille, France
| | - Enrico Fabris
- Azienda Ospedaliero-Universitaria Ospedali Riuniti Trieste, Trieste, Italy
| | | | - Marco Boccalatte
- Division of Cardiology, Ospedale Santa Maria delle Grazie, Pozzuoli, Italy
| | - Francisco Bosa Ojeda
- Division of Cardiology, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | | | | | | | - Hsien-Li Kao
- Cardiology Division, Department of Internal Medicine, National Taiwan University Hospital, Tapei, Taiwan
| | - Juan Sanchis Forés
- Division of Cardiology, Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - Luigi Vignali
- Interventional Cardiology Unit, Azienda Ospedaliera Sanitaria, Parma, Italy
| | - Helder Pereira
- Cardiology Department, Hospital Garcia de Orta, Pragal, Almada, Portugal
| | - Stephane Manzo
- Division of Cardiology, CHU Lariboisière, AP-HP, Paris VII University, INSERM UMRS 942, Paris, France
| | - Santiago Ordoñez
- Instituto Cardiovascular de Buenos Aires, Buenos Aires, Argentina
| | | | - Bruno Scheller
- Division of Cardiology Clinical and Experimental Interventional Cardiology, University of Saarland, Saarbrücken, Germany
| | - Heidi Lehtola
- Division of Cardiology, Oulu University Hospital, Oulu, Finland
| | - Rui Teles
- Division of Cardiology, Hospital de Santa Cruz, CHLO-Nova Medical School, CEDOC, Lisbon, Portugal
| | - Christos Mantis
- Division of Cardiology, Kontantopoulion Hospital, Athens, Greece
| | - Ylitalo Antti
- Division of Cardiology, Heart Centre Turku, Turku, Finland
| | | | - Rodrigo Zoni
- Department of Teaching and Research, Instituto de Cardiología de Corrientes "Juana F. Cabral", Corrientes, Argentina
| | | | | | | | | | - Carlos E Uribe
- Division of Cardiology, Universidad UPB, Universidad CES, Medellin, Colombia
| | - John Kanakakis
- Division of Cardiology, Alexandra Hospital, Athens, Greece
| | - Benjamin Faurie
- Division of Cardiology, Groupe Hospitalier Mutualiste de Grenoble, Grenoble, France
| | - Gabriele Gabrielli
- Interventional Cardiology Unit, Azienda Ospedaliero Universitaria"Ospedali Riuniti", Ancona, Italy
| | | | | | - Alex Rocha
- Department of Cardiology and Cardiovascular Interventions, Instituto Nacional de Cirugía Cardíaca, Montevideo, Uruguay
| | - Frankie Chor-Cheung Tam
- Department of Cardiology, Queen Mary Hospital, University of Hong Kong, Pok Fu Lam, Hong Kong
| | | | - Antonia Anna Lukito
- Cardiovascular Department Pelita, Harapan University/Heart Center Siloam Lippo Village Hospital, Tangerang, Banten, Indonesia
| | | | - Gustavo Pessah
- Division of Cardiology, Hospiatl Cordoba, Cordoba, Argentina
| | | | - Giuliana Cortese
- Department of Statistical Sciences, University of Padova, Padua, Italy
| | - Guido Parodi
- Department of Cardiology, ASL 4 Liguria, Lavagna, Italy
| | | | - Elvin Kedhi
- Division of Cardiology, Hopital Erasmus, Universitè Libre de Bruxelles, Brussels, Belgium
| | - Pablo Lamelas
- Instituto Cardiovascular de Buenos Aires, Buenos Aires, Argentina
| | - Harry Suryapranata
- Division of Cardiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Monica Verdoia
- Division of Cardiology, Ospedale degli Infermi, ASL Biella, Ponderano, Italy
| |
Collapse
|
46
|
Calabrese C, Annunziata A, Mariniello DF, Coppola A, Mirizzi AI, Simioli F, Pelaia C, Atripaldi L, Pugliese G, Guarino S, Fiorentino G. Evolution of the Clinical Profile and Outcomes of Unvaccinated Patients Affected by Critical COVID-19 Pneumonia from the Pre-Vaccination to the Post-Vaccination Waves in Italy. Pathogens 2022; 11:pathogens11070793. [PMID: 35890037 PMCID: PMC9323253 DOI: 10.3390/pathogens11070793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 02/04/2023] Open
Abstract
The vaccination campaign and the new SARS-CoV-2 variants may have changed the clinical profile and outcomes of patients admitted to sub-intensive unit care. We conducted a retrospective study aimed to compare the clinical and radiological features of unvaccinated critical COVID-19 patients hospitalized during the last pandemic wave (December 2021−February 2022, No-Vax group) and before starting the vaccination campaign (March−December 2020, Pre-Vax group). The No-Vax group was also compared with vaccinated patients of the same pandemic wave (Vax group). With respect to the Pre-Vax group, the No-Vax group contained a higher percentage of smokers (p = 0.0007) and a lower prevalence of males (p = 0.0003). At admission, the No-Vax patients showed both a higher CT score of pneumonia and a worse severe respiratory failure (p < 0.0001). In the No-Vax group, a higher percentage of deaths occurred, though this was not significant. In comparison with the No-Vax group, the Vax patients were older (p = 0.0097), with a higher Charlson comorbidity index (p < 0.0001) and a significantly lower HRCT score (p = 0.0015). The percentage of deaths was not different between the two groups. The No-Vax patients showed a more severe disease in comparison with the Pre-Vax patients, and were younger and had fewer comorbidities than the Vax patients.
Collapse
Affiliation(s)
- Cecilia Calabrese
- Department of Translational Medical Sciences, University of Campania ‘Luigi Vanvitelli’, A.O.R.N. Ospedali dei Colli, 80131 Napoli, Italy; (D.F.M.); (L.A.); (G.P.)
- Correspondence:
| | - Anna Annunziata
- Department of Intensive Care, A.O.R.N. Ospedali dei Colli, 80131 Napoli, Italy; (A.A.); (A.C.); (A.I.M.); (F.S.); (G.F.)
| | - Domenica Francesca Mariniello
- Department of Translational Medical Sciences, University of Campania ‘Luigi Vanvitelli’, A.O.R.N. Ospedali dei Colli, 80131 Napoli, Italy; (D.F.M.); (L.A.); (G.P.)
| | - Antonietta Coppola
- Department of Intensive Care, A.O.R.N. Ospedali dei Colli, 80131 Napoli, Italy; (A.A.); (A.C.); (A.I.M.); (F.S.); (G.F.)
| | - Angela Irene Mirizzi
- Department of Intensive Care, A.O.R.N. Ospedali dei Colli, 80131 Napoli, Italy; (A.A.); (A.C.); (A.I.M.); (F.S.); (G.F.)
| | - Francesca Simioli
- Department of Intensive Care, A.O.R.N. Ospedali dei Colli, 80131 Napoli, Italy; (A.A.); (A.C.); (A.I.M.); (F.S.); (G.F.)
| | - Corrado Pelaia
- Respiratory Medicine Unit, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy;
| | - Lidia Atripaldi
- Department of Translational Medical Sciences, University of Campania ‘Luigi Vanvitelli’, A.O.R.N. Ospedali dei Colli, 80131 Napoli, Italy; (D.F.M.); (L.A.); (G.P.)
| | - Gaia Pugliese
- Department of Translational Medical Sciences, University of Campania ‘Luigi Vanvitelli’, A.O.R.N. Ospedali dei Colli, 80131 Napoli, Italy; (D.F.M.); (L.A.); (G.P.)
| | - Salvatore Guarino
- Department of Radiology, Monaldi Hospital, A.O.R.N. Ospedali dei Colli, 80131 Napoli, Italy;
| | - Giuseppe Fiorentino
- Department of Intensive Care, A.O.R.N. Ospedali dei Colli, 80131 Napoli, Italy; (A.A.); (A.C.); (A.I.M.); (F.S.); (G.F.)
| |
Collapse
|
47
|
Kumar Biswas B, Soo Shin J, Malpani YR, Hwang D, Jung E, Bong Han S, Vishakantegowda AG, Jung YS. Enteroviral replication inhibition by N-Alkyl triazolopyrimidinone derivatives through a non-capsid binding mode. Bioorg Med Chem Lett 2022; 64:128673. [PMID: 35292344 DOI: 10.1016/j.bmcl.2022.128673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/14/2022] [Accepted: 03/10/2022] [Indexed: 11/20/2022]
Abstract
Small-molecule inhibitors exhibiting broad-spectrum enteroviral inhibition by targeting viral replication proteins are highly desirable in antiviral drug discovery. We used the previously identified antiviral compound 1 as the starting material to develop a novel compound series with high efficacy against human rhinovirus (hRV). Further optimization of N-substituted triazolopyrimidinone derivatives revealed that the N-alkyl triazolopyrimidinone derivatives (2) had more potent antiviral activity against hRVs than compound 1. The new compounds showed improved selectivity index values, and compound 2c (KR-25210) displayed broad anti-hRV activity, with half-maximal effective concentration values ≤ 2 µM against all tested hRVs. In addition, 2c showed notable activity against other enteroviruses. Drug-likeness elucidation showed that 2c exhibited reasonable human and rat liver microsomal phase-I stability and safe CYP inhibition. Replication studies revealed that 2c is not a capsid inhibitor, and a time-of-addition assay indicated that 2c targets the virus replication stages.
Collapse
Affiliation(s)
- Bishyajit Kumar Biswas
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, 141 Gajeongro, Yuseong, Daejeon 34114, Republic of Korea; Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, 217 Gajeongro, Yuseong, Daejeon 34113, Republic of Korea
| | - Jin Soo Shin
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, 141 Gajeongro, Yuseong, Daejeon 34114, Republic of Korea
| | - Yashwardhan R Malpani
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, 141 Gajeongro, Yuseong, Daejeon 34114, Republic of Korea; Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, 217 Gajeongro, Yuseong, Daejeon 34113, Republic of Korea
| | - Dasom Hwang
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, 141 Gajeongro, Yuseong, Daejeon 34114, Republic of Korea
| | - Eunhye Jung
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, 141 Gajeongro, Yuseong, Daejeon 34114, Republic of Korea
| | - Soo Bong Han
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, 141 Gajeongro, Yuseong, Daejeon 34114, Republic of Korea; Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, 217 Gajeongro, Yuseong, Daejeon 34113, Republic of Korea
| | - Avinash G Vishakantegowda
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, 141 Gajeongro, Yuseong, Daejeon 34114, Republic of Korea; Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, 217 Gajeongro, Yuseong, Daejeon 34113, Republic of Korea
| | - Young-Sik Jung
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, 141 Gajeongro, Yuseong, Daejeon 34114, Republic of Korea; Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, 217 Gajeongro, Yuseong, Daejeon 34113, Republic of Korea.
| |
Collapse
|
48
|
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
|
49
|
Love ME, Proud D. Respiratory Viral and Bacterial Exacerbations of COPD—The Role of the Airway Epithelium. Cells 2022; 11:cells11091416. [PMID: 35563722 PMCID: PMC9099594 DOI: 10.3390/cells11091416] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 12/14/2022] Open
Abstract
COPD is a leading cause of death worldwide, with acute exacerbations being a major contributor to disease morbidity and mortality. Indeed, exacerbations are associated with loss of lung function, and exacerbation frequency predicts poor prognosis. Respiratory infections are important triggers of acute exacerbations of COPD. This review examines the role of bacterial and viral infections, along with co-infections, in the pathogenesis of COPD exacerbations. Because the airway epithelium is the initial site of exposure both to cigarette smoke (or other pollutants) and to inhaled pathogens, we will focus on the role of airway epithelial cell responses in regulating the pathophysiology of exacerbations of COPD. This will include an examination of the interactions of cigarette smoke alone, and in combination with viral and bacterial exposures in modulating epithelial function and inflammatory and host defense pathways in the airways during COPD. Finally, we will briefly examine current and potential medication approaches to treat acute exacerbations of COPD triggered by respiratory infections.
Collapse
|
50
|
Singanayagam A, Footitt J, Marczynski M, Radicioni G, Cross MT, Finney LJ, Trujillo-Torralbo MB, Calderazzo M, Zhu J, Aniscenko J, Clarke TB, Molyneaux PL, Bartlett NW, Moffatt MF, Cookson WO, Wedzicha J, Evans CM, Boucher RC, Kesimer M, Lieleg O, Mallia P, Johnston SL. Airway mucins promote immunopathology in virus-exacerbated chronic obstructive pulmonary disease. J Clin Invest 2022; 132:e120901. [PMID: 35239513 PMCID: PMC9012283 DOI: 10.1172/jci120901] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 03/01/2022] [Indexed: 11/18/2022] Open
Abstract
The respiratory tract surface is protected from inhaled pathogens by a secreted layer of mucus rich in mucin glycoproteins. Abnormal mucus accumulation is a cardinal feature of chronic respiratory diseases, but the relationship between mucus and pathogens during exacerbations is poorly understood. We identified elevations in airway mucin 5AC (MUC5AC) and MUC5B concentrations during spontaneous and experimentally induced chronic obstructive pulmonary disease (COPD) exacerbations. MUC5AC was more sensitive to changes in expression during exacerbation and was therefore more predictably associated with viral load, inflammation, symptom severity, decrements in lung function, and secondary bacterial infections. MUC5AC was functionally related to inflammation, as Muc5ac-deficient (Muc5ac-/-) mice had attenuated RV-induced (RV-induced) airway inflammation, and exogenous MUC5AC glycoprotein administration augmented inflammatory responses and increased the release of extracellular adenosine triphosphate (ATP) in mice and human airway epithelial cell cultures. Hydrolysis of ATP suppressed MUC5AC augmentation of RV-induced inflammation in mice. Therapeutic suppression of mucin production using an EGFR antagonist ameliorated immunopathology in a mouse COPD exacerbation model. The coordinated virus induction of MUC5AC and MUC5B expression suggests that non-Th2 mechanisms trigger mucin hypersecretion during exacerbations. Our data identified a proinflammatory role for MUC5AC during viral infection and suggest that MUC5AC inhibition may ameliorate COPD exacerbations.
Collapse
Affiliation(s)
- Aran Singanayagam
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Joseph Footitt
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Matthias Marczynski
- School of Engineering and Design, Department of Materials Engineering and
- Center for Protein Assemblies, Technical University of Munich, Munich, Germany
| | - Giorgia Radicioni
- Marsico Lung Institute/Cystic Fibrosis and Pulmonary Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael T. Cross
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lydia J. Finney
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - Maria Calderazzo
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jie Zhu
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Julia Aniscenko
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Thomas B. Clarke
- Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Philip L. Molyneaux
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nathan W. Bartlett
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- College of Health, Medicine and Wellbeing, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Miriam F. Moffatt
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - William O. Cookson
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jadwiga Wedzicha
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Christopher M. Evans
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard C. Boucher
- Marsico Lung Institute/Cystic Fibrosis and Pulmonary Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mehmet Kesimer
- Marsico Lung Institute/Cystic Fibrosis and Pulmonary Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Oliver Lieleg
- School of Engineering and Design, Department of Materials Engineering and
- Center for Protein Assemblies, Technical University of Munich, Munich, Germany
| | - Patrick Mallia
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | |
Collapse
|