1
|
Kreger JE, Sliwicki AL, Essoh SN, Li Y, Jayachandran C, Czapla JA, Lewis TC, Kirkham EM, Vergotine RJ, Popova AP, Flori HR, Hershenson MB. Upper Airway Gene Expression in Hospitalized Children with Rhinovirus-induced Respiratory Illnesses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.29.651288. [PMID: 40370957 PMCID: PMC12077871 DOI: 10.1101/2025.04.29.651288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Background The precise mechanisms underlying rhinovirus (RV)-induced respiratory illnesses are not completely known. Objective We sought to obtain nasal transcriptomic data from hospitalized children with respiratory viral infections. Methods We obtained nasal swabs from 46 children with RV (16 RV-A, 30 RV-C). For comparison, we examined swabs from 12 children with RSV and six controls. Subjects ranged in age from 1 month to 18 years. Viral detection, genotyping and copy number were determined by PCR. RNA transcripts were measured by next generation sequencing and differences in gene expression calculated using DESeq2. Results Compared to controls, 1232 transcripts were upregulated (adjusted p<0.05, fold change >1.5) by all three viruses, including genes regulating granulocyte chemotaxis, cysteinyl leukotriene production, epithelial remodeling and antiviral responses. Cilium-related genes were downregulated. Compared to RSV, RV induced greater expression of 207 genes including those regulating eosinophilic inflammation, mucus secretion and mast cell function.RSV induced greater upregulation of 674 genes including those regulating neutrophilic inflammation and type 1 IFN response. Computational deconvolution of RNA-seq profiles revealed that viral infection decreased ciliated cells while increasing neutrophils, natural killer cells, monocytes (all viral species) and goblet cells (RV only). RV-C infections increased mast cells and IFN-λ mRNA expression. RV copy number correlated with the expression of mast cell proteases and numerous pro-inflammatory and IFN-stimulated genes. Conclusion Children hospitalized with RV and RSV infections mount robust inflammatory responses, but virus-specific differences exist. Clinical Implication These data provide insight into mechanisms by which RV, and in particular, RV-C, trigger respiratory illnesses. Capsule summary Nasal transcriptomics demonstrate that RV infections in hospitalized children induce expression of genes regulating eosinophilic inflammation, mucus secretion and mast cell function, with RV-C in particular increasing IFN-λ expression.
Collapse
|
2
|
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] [Download PDF] [Figures] [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
|
3
|
Iyadorai T, Wong PL, Sii HL, P'ng CK, Ee SS, Tan MP, Hanafi NS, Pang YK, Ng KT, Chook JB, Takebe Y, Chan KG, Singh S, Sam IC, Lim SH, Tee KK. Respiratory Symptoms and Health Outcomes of Rhinovirus and Influenza Virus Infections: Implications of Asthma, Diabetes Mellitus and Allergic Rhinitis in Rhinovirus C Infection. J Med Virol 2025; 97:e70281. [PMID: 40022583 DOI: 10.1002/jmv.70281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 02/11/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Rhinovirus (RV), classified into RV-A, RV-B, and RV-C, is a prevalent cause of respiratory tract infections (RTIs). Here, we analysed RV infection and its clinical implications among outpatients with acute upper RTIs. Demographic data, baseline comorbidities, clinical symptoms, and health outcomes of RV-infected patients (n = 849) were compared with influenza (n = 417). Multivariable logistic regression was employed to evaluate predictors and health outcomes over a 1-year follow-up period. RV infections predominantly presented with cough, nasal discharge, and sore throat, whereas fever was more prevalent in influenza cases. RV-C-infected individuals with diabetes mellitus (adjusted odds ratio [aOR] 3.6; 95% CI 1.7-7.2; p = 0.001) and asthma (aOR 1.9; 95% CI 1.0-3.5; p = 0.047) showed a higher likelihood of experiencing severe acute respiratory symptoms. RV-C patients with comorbidities were twice more likely to have primary care visits due to RTIs within 1 year (aOR 2.4; 95% CI 1.4-4.4; p = 0.003). Asthma (aOR 3.8; 95% CI 1.9-7.2; p < 0.0001) and allergic rhinitis (aOR 2.9; 95% CI 1.0-8.0; p = 0.042) were key predictors of increased RTI-related primary care visits. RV infection, particularly in individuals with asthma, allergic rhinitis and diabetes, poses a significant disease burden similar to that of influenza.
Collapse
Affiliation(s)
- Thevambiga Iyadorai
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Pui Li Wong
- Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Hoe Leong Sii
- Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Chun Keat P'ng
- Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Soon Sean Ee
- Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Maw Pin Tan
- Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Nik Sherina Hanafi
- Department of Primary Care Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Yong Kek Pang
- Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Kim Tien Ng
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Jack Bee Chook
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | - Yutaka Takebe
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- AIDS Research Center, National Institute of Infectious Diseases, Toyama, Shinjuku-ku, Tokyo, Japan
| | - Kok-Gan Chan
- Division of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia
- Microbiome Research Group, Research Center for Life Science and Healthcare, China
- Beacons of Excellence Research and Innovation Institute (CBI), University of Nottingham, Ningbo China, Zhejiang, China
| | - Sarbhan Singh
- Biomedical Epidemiology Unit, Special Resource Centre, Institute for Medical Research, Ministry of Health, Shah Alam, Malaysia
| | - I-Ching Sam
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Sin How Lim
- Department of Social and Preventive Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Kok Keng Tee
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
4
|
Zhou Z, Zhu R, Yang H, Deng W, Zhang Z, Li Y, Xu J, Yan Z, Wang R, Chang S, Yin Z, Wu Y, Zhang D, Fang M, Liu C, Que Y, Zhang J, Xia N, Wang Y, Xu L, Cheng T. Transgenic mice expressing the human CDHR3 receptor: A sensitive RV-C infection model for the evaluation of vaccines and therapeutics. Antiviral Res 2025; 235:106102. [PMID: 39922540 DOI: 10.1016/j.antiviral.2025.106102] [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: 10/27/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/10/2025]
Abstract
Rhinovirus C (RV-C) is the primary causative agent of severe acute respiratory illnesses (ARTIs) in infants and young children. The limited availability of animal models complicates the development of prophylactic and therapeutic strategies targeting RV-C. Previous studies have identified human cadherin-related family member 3 (hCDHR3) as the cellular receptor for RV-C, with its expression enabling previously unsusceptible cells to support both viral entry and replication. Recently, an adult hCDHR3 transgenic mouse model was developed to investigate the role of human stimulator of interferon genes (hSTING) in RV-C15 infection in vivo. However, adult mice do not support efficient RV-C15 infection. Here, we report a transgenic mouse line expressing hCDHR3 constitutively that is highly susceptible to early-life infections by multiple serotypes of RV-C, including RV-C15, RV-C2, and RV-C41. Neonatal transgenic mice infected with various RV-C strains via the intraperitoneal (i.p.) route exhibit similar symptoms, such as severe inflammation, limb paralysis, and death. Moreover, passive immunization with antisera or therapeutic antibodies can protect against lethal RV-C infection in these transgenic mice. Overall, this study provides a valuable animal model for the in vivo antiviral evaluation against RV-C.
Collapse
Affiliation(s)
- Zhenhong Zhou
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Rui Zhu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Hongwei Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Weixi Deng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Zijie Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Yue Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Jiaxin Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Ziyang Yan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Ruoxi Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Sijia Chang
- Beijing Wantai Biological Pharmacy Enterprise Co., Ltd., Beijing, 102206, PR China
| | - Zhichao Yin
- Beijing Wantai Biological Pharmacy Enterprise Co., Ltd., Beijing, 102206, PR China
| | - Yuanyuan Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Dongqing Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Mujin Fang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Che Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Yuqiong Que
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Yingbin Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China.
| | - Longfa Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China.
| | - Tong Cheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China.
| |
Collapse
|
5
|
Fu H, De R, Sun Y, Yao Y, Zhu R, Chen D, Zhou Y, Guo Q, Zhao L. Association between cadherin-related family member 3 rs6967330-A and human rhinovirus-C induced wheezing in children. Virol J 2025; 22:29. [PMID: 39915850 PMCID: PMC11804036 DOI: 10.1186/s12985-025-02644-7] [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: 10/26/2024] [Accepted: 01/28/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND The heterogeneity of childhood wheezing illnesses is associated with viral and host factors. Human rhinoviruses (HRV) are the major pathogens in severe wheezing in young children. The single nucleotide polymorphism (SNP) rs6967330 G > A proved to heighten the risk of wheezing. However, the relation between rs6967330 variants of cadherin-related family member 3 (CDHR3) and wheezing induced by human rhinovirus (HRV)-C has not been determined. METHODS A total of 11,756 respiratory specimens collected from hospitalized children with acute respiratory infections (ARIs) between September 2017 and March 2023 were screened for enterovirus (EV)/HRVs by the capillary electrophoresis-based multiplex PCR (CEMP) assay, and those positive only for HRVs were amplified and sequenced for HRV and CDHR3 genotyping. The clinical data of the enrolled patients were obtained and analyzed. RESULTS EV/HRVs (15.2%; 1,616/10,608) were the more common viruses detected in inpatients with ARIs. Among the enrolled samples, 148 were positive for HRV-A (49.83%; 148/297), 129 for HRV-C (43.4%; 129/297), and 20 for HRV-B (6.7%; 20/297). More patients infected with HRV-C had history of allergy (P = 0.004), family history of asthma (P = 0.001), wheezing (P = 0.005) and asthma (P = 0.001) than those infected with HRV-A or HRV-B, while patients infected with HRV-C were less likely to have older siblings compared to those infected with HRV-A (P = 0.014). The rs6967330-A variant was related to a high incidence of the three concave signs (P = 0.047), asthma exacerbation (P = 0.025), a higher risk of HRV-C infection determined by the dominant model (OR 1.91, 95% confidence interval 1.05-3.48; P = 0.033), and a high proportion of wheezing (56.67%) in patients infected with HRV-C. CONCLUSIONS HRV-C is the dominant species responsible for HRV-induced wheezing. The rs6967330-A variant is a risk factor for HRV-C infection, and was associated with the high rate of wheezing induced by HRV-C.
Collapse
Affiliation(s)
- Hanhaoyu Fu
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ri De
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yu Sun
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yao Yao
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Runan Zhu
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Dongmei Chen
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yutong Zhou
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Qi Guo
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Linqing Zhao
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, 100020, China.
| |
Collapse
|
6
|
Palumbo S, Irish J, Narendran N, Stern DA, Volpe S, Le CH, Starks R, Bosco A, Martinez FD, Chang EH. The rs6967330 minor allele in CDHR3 is a significant risk factor for severe acute exacerbations in chronic rhinosinusitis. J Allergy Clin Immunol 2025; 155:583-593. [PMID: 39389125 PMCID: PMC11805668 DOI: 10.1016/j.jaci.2024.09.025] [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: 01/22/2024] [Revised: 09/12/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Acute exacerbations of chronic rhinosinusitis (AECRS) are commonly triggered by rhinovirus (RV) infections with secondary bacterial infections. Risk factors for AECRS are not well understood. OBJECTIVE We sought to determine whether carriers of the minor allele rs6967330 (AA/AG) in the cadherin-related family member 3 (CDHR3) gene have an increased risk for RV infections in AECRS in vivo and identify CDHR3 genotype-dependent host responses to RV infection in differentiated nasal airway-liquid interface (ALI) cultures ex vivo. METHODS We performed a prospective year-long study of adult subjects with chronic rhinosinusitis by the rs6967330 genotype (AA/AG, n = 16; GG, n = 38). We contacted subjects every 2 weeks, and if they reported AECRS, then clinical data were collected. ALI cultures of adults with chronic rhinosinusitis (AG/AA, n = 19; GG, n = 19) were challenged with RV-A and RV-C. We measured viral copy numbers at 4 and 48 hours postinfection and RNA transcriptomes and cytokines at 48 hours postinfection. RESULTS Subjects with the minor allele had significantly higher rates of RV and bacterial infections than those with the major allele. ALI minor allele cultures had higher viral copy numbers of RV-A and RV-C after 48 hours compared with the major allele. Differentially expressed genes and pathways identified an upregulation of IL-10 and IL-4/IL-13 pathways and a significant downregulation of Toll-like receptor pathways in the minor allele cultures after RV-A and RV-C infection. Unsupervised hierarchical analysis of all differentially expressed genes suggested that allergic rhinitis had an additive effect on this response. CONCLUSIONS The rs6967330 minor allele is associated with increased RV-A and RV-C replication, downregulation of Toll-like receptor-mediated responses, and increased type-2 and cytokine and chemokine responses during RV infection.
Collapse
Affiliation(s)
- Sunny Palumbo
- Department of Otolaryngology, University of Arizona, Tucson, Ariz; Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Joseph Irish
- Department of Otolaryngology, University of Arizona, Tucson, Ariz; Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Nirushan Narendran
- Department of Otolaryngology, University of Arizona, Tucson, Ariz; Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Debra A Stern
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Sophia Volpe
- Department of Otolaryngology, University of Arizona, Tucson, Ariz; Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Christopher H Le
- Department of Otolaryngology, University of Arizona, Tucson, Ariz
| | - Rebekah Starks
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz; Department of Immunobiology, University of Arizona College of Medicine, Tucson, Ariz
| | - Anthony Bosco
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz; Department of Immunobiology, University of Arizona College of Medicine, Tucson, Ariz
| | - Fernando D Martinez
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Eugene H Chang
- Department of Otolaryngology, University of Arizona, Tucson, Ariz; Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz.
| |
Collapse
|
7
|
Renouf B, Sutanto EN, Kidd C, Lim J, Amin M, Berry L, Hoyne GF, D'Vaz N, Kicic-Starcevich E, Stick SM, Iosifidis T. Profiling epithelial viral receptor expression in amniotic membrane and nasal epithelial cells at birth. Placenta 2025; 160:82-88. [PMID: 39778257 DOI: 10.1016/j.placenta.2024.12.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/17/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION Children with wheeze and asthma present with airway epithelial vulnerabilities, such as impaired responses to viral infection. It is postulated that the in utero environment may contribute to the development of airway epithelial vulnerabilities. The aims of the study were to establish whether the receptors for rhinovirus (RV), respiratory syncytial virus (RSV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are expressed in the amniotic membrane and whether the pattern of expression is similar to newborn nasal epithelium. METHODS Placenta were collected (n = 33) from newborns in AERIAL, a sub-study nested under the ORIGINS birth cohort. Using purified RNA from amniotic samples (n = 33), along with previously extracted RNA from nasal epithelial cells from newborns (n = 20), real-time quantitative polymerase chain reaction (qPCR) was performed to determine gene expression of viral receptors for RV, RSV and SARS-CoV-2 in both amniotic and newborn nasal epithelial samples. In addition, receptor protein expression was quantified through Western blot and localised using immunohistochemical staining in amniotic samples. RESULTS Amniotic and newborn nasal samples expressed various receptors for RV (ICAM-1, LDLR, CDHR3), RSV (NCL, CX3CR1) and SARS-CoV-2 (ACE2, TMPRSS2) at the gene level, although the magnitude of expression varied. In addition, protein expression of these receptors was confirmed in the amniotic samples. These proteins were localised to the epithelial layer of the amniotic membrane. CONCLUSION This proof-of-concept study indicates the potential of amniotic samples to facilitate investigation into the interactions between the in utero environment and prenatal programming of epithelial innate immune responses to viruses.
Collapse
Affiliation(s)
- Bailee Renouf
- Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, 6009, Western Australia, Australia; School of Health Sciences, The University of Notre Dame Australia, Fremantle, 6160, Western Australia, Australia
| | - Erika N Sutanto
- Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, 6009, Western Australia, Australia; Centre for Child Health Research, The University of Western Australia, Nedlands, 6009, Western Australia, Australia; School of Public Health, Curtin University, Bentley, 6102, Western Australia, Australia
| | - Courtney Kidd
- Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, 6009, Western Australia, Australia
| | - James Lim
- Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, 6009, Western Australia, Australia
| | - Minda Amin
- Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, 6009, Western Australia, Australia
| | - Luke Berry
- Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, 6009, Western Australia, Australia
| | - Gerard F Hoyne
- School of Health Sciences, The University of Notre Dame Australia, Fremantle, 6160, Western Australia, Australia; Institute of Respiratory Health, QEII Medical Centre, Nedlands, 6009, Western Australia, Australia
| | - Nina D'Vaz
- Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, 6009, Western Australia, Australia
| | | | - Stephen M Stick
- Centre for Cell Therapy and Regenerative Medicine, Medical School, The University of Western Australia, Nedlands, 6009, Western Australia, Australia; Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, 6009, Western Australia, Australia
| | - Thomas Iosifidis
- Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, 6009, Western Australia, Australia; School of Public Health, Curtin University, Bentley, 6102, Western Australia, Australia; Centre for Cell Therapy and Regenerative Medicine, Medical School, The University of Western Australia, Nedlands, 6009, Western Australia, Australia.
| |
Collapse
|
8
|
McCluskey E, Sajjan U. In Vitro Model to Test Therapeutic Agents Against Rhinovirus Infection. Methods Mol Biol 2025; 2903:185-191. [PMID: 40016467 DOI: 10.1007/978-1-0716-4410-2_15] [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
To develop an antiviral drug, it is extremely important to have a relevant cell culture model system. The airway epithelial cells lining the conductive airways are the primary target for all three classes of rhinoviruses. However, rhinovirus replication is not very robust in airway epithelial cells; therefore, it does not cause cytopathic effects. However, rhinovirus induces replication-dependent cytokines such as type I and type III interferons and CXCL-10, which can be used as surrogates for viral load. Here, we describe a method using the immortalized normal human bronchial epithelial cell line, BEAS-2B cells. These cells readily grow in serum-free medium and can be infected with rhinovirus A and B. The binding, endocytosis, and replication of the virus induce CXCL-8 in these cells. These cells also respond to replicating virus by showing robust CXCL-10 production. Both CXCL-8 and CXCL-10 secreted into the medium can be readily measured by ELISA. Therefore, this method can be used to examine the drugs that can inhibit binding, endocytosis, and replication of the virus.
Collapse
Affiliation(s)
- Elizabeth McCluskey
- 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
|
Watkinson RL, Bochkov YA, Gern JE, Iosifidis T, Looi K, Laing IA, Kicic A. Investigation of Differentiated Nasal Epithelial Responses to Infection with Clinical Isolates of Rhinovirus A and C. Methods Mol Biol 2025; 2903:113-139. [PMID: 40016462 DOI: 10.1007/978-1-0716-4410-2_10] [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
The nasal epithelium is the primary point of contact for inhaled respiratory viruses such as rhinovirus, respiratory syncytial virus, influenza, and coronavirus, among others. In order to establish infection, these viruses must engage their respective receptors located on host epithelial cells and begin replication. However, the nasal epithelium is also a pivotal orchestrator of both structural and innate immune defenses against these pathogens and thus mounts a broad antiviral response to halt the progression of the infection into the lower airways. Of note, the most common virus found in the airways of children presenting to the hospital emergency department with acute wheezing and asthma is rhinovirus C (RV-C), followed by rhinovirus A (RV-A). Here, we illustrate infection of a preclinical differentiated nasal epithelial model with clinical isolates of RV-A and -C, in conjunction with several methods utilized for characterization of epithelial responses post-infection in vitro.
Collapse
Affiliation(s)
- Rebecca L Watkinson
- Division of Paediatrics, School of Medicine, The University of Western Australia, Crawley, WA, Australia
- Wal-Yan Respiratory Research Centre, The Kids Research Institute Australia, Nedlands, WA, Australia
| | - Yury A Bochkov
- Department of Paediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - James E Gern
- Department of Paediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Thomas Iosifidis
- Division of Paediatrics, School of Medicine, The University of Western Australia, Crawley, WA, Australia
- Wal-Yan Respiratory Research Centre, The Kids Research Institute Australia, Nedlands, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia, Crawley, WA, Australia
| | - Kevin Looi
- Division of Paediatrics, School of Medicine, The University of Western Australia, Crawley, WA, Australia
- Wal-Yan Respiratory Research Centre, The Kids Research Institute Australia, Nedlands, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
| | - Ingrid A Laing
- Division of Paediatrics, School of Medicine, The University of Western Australia, Crawley, WA, Australia
- Wal-Yan Respiratory Research Centre, The Kids Research Institute Australia, Nedlands, WA, Australia
- Division of Cardiovascular and Respiratory Sciences, School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
| | - Anthony Kicic
- Wal-Yan Respiratory Research Centre, The Kids Research Institute Australia, Nedlands, WA, Australia.
- School of Population Health, Curtin University, Bentley, WA, Australia.
- Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia, Crawley, WA, Australia.
| |
Collapse
|
10
|
Tabasi M, Ganjian H, Sajjan U. Quantification of Infectious Rhinovirus A and B Serotypes by Plaque Assay. Methods Mol Biol 2025; 2903:31-38. [PMID: 40016456 DOI: 10.1007/978-1-0716-4410-2_4] [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
Plaque assay is a quantitative assay that determines the number of infective virions in the viral stock or the infected cells. Plaques are essentially virus-infected cells that produce progeny virus and infect adjacent cells. The cells producing progeny virus die and detach from the dish, leaving an empty space in the confluent monolayer of cells. Unlike other viruses, rhinovirus does not form characteristic round plaques but shows small clear areas of different shapes surrounded by dying cells. Rhinoviruses form plaques only in highly susceptible H1HeLa cells but not in their primary target, airway epithelial cells. The plaque assay to determine the number of infective virions works only for rhinovirus species A and B, but not C, because the latter does not infect H1HeLa cells. Here we describe a method to quantify the infective virions by plaque assay for rhinovirus speciesA and B.
Collapse
Affiliation(s)
- Mohsen Tabasi
- Centre for Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA
| | - Haleh Ganjian
- 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
|
11
|
Ganjian H, Sajjan U. Laboratory Protocol for Propagation and Purification of Rhinovirus A and B Suitable for In Vitro and In Vivo Infection. Methods Mol Biol 2025; 2903:9-19. [PMID: 40016454 DOI: 10.1007/978-1-0716-4410-2_2] [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
H1HeLa cell line has become the most utilized cell culture system for in vitro studies of the rhinovirus life cycle. These cells are also widely used to propagate the rhinovirus to study host responses. Unlike other viruses, high-titer virus is required for studies focusing on understanding the host responses to rhinovirus infection in vivo and in vitro. Therefore, after propagation of rhinovirus, H1HeLa cells should be concentrated and purified. In this chapter, we describe the methods used for the propagation of rhinovirus A and B that are adapted to H1HeLa cells.
Collapse
Affiliation(s)
- Haleh Ganjian
- 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, Temple University, Philadelphia, PA, USA.
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Sajjan U. Determination of Binding and Endocytosis of Rhinovirus by Flow Cytometry. Methods Mol Biol 2025; 2903:51-63. [PMID: 40016458 DOI: 10.1007/978-1-0716-4410-2_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] [Indexed: 03/01/2025]
Abstract
Viruses are dependent on their host cells for replication. The first step in viral infection is the binding of the virus to the host cell membrane, followed by internalization or endocytosis of the virus. Rhinoviruses utilize several cellular membrane receptors to cross the plasma membrane by endocytosis. Three major glycoprotein receptors that are utilized by rhinoviruses to gain entry into the cells are intracellular adhesion molecules, low-density lipoprotein receptor family members, and cadherin-related family member-3. Rhinovirus is endocytosed by a clathrin- or dynamin-dependent mechanism. Upon uptake into an acidic endosomal environment, rhinovirus uncoats and injects its genome into the cytoplasm, where the viral replication begins. It is often difficult to differentiate between bound and the endocytosed rhinovirus in the cells. Here, we describe a method to quantify bound and endocytosed virus by flow cytometry.
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
|
13
|
Horton K, Wing PAC, Jackson CL, McCormick CJ, Carroll MP, Lucas JS. Interplay between respiratory viruses and cilia in the airways. Eur Respir Rev 2025; 34:240224. [PMID: 40107662 PMCID: PMC11920889 DOI: 10.1183/16000617.0224-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/19/2025] [Indexed: 03/22/2025] Open
Abstract
The airway epithelium is the first point of contact for inhaled pathogens. The role of epithelial cells in clearance, infection and colonisation of bacteria is established. The interactions of respiratory viruses and cilia is less understood, but viruses are known to target ciliated epithelial cells for entry, replication and dissemination. Furthermore, some respiratory viruses impair and/or enhance ciliary activity. This review examines what is known about the interactions between cilia and viral infection and how respiratory viruses effect cilia function with subsequent consequences for human health. We discuss the models which can be used to investigate the relationship between respiratory viruses and the host airway.
Collapse
Affiliation(s)
- Katie Horton
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- These authors contributed equally to this work
| | - Peter A C Wing
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- These authors contributed equally to this work
| | - Claire L Jackson
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- These authors contributed equally to this work
| | - Christopher J McCormick
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Mary P Carroll
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jane S Lucas
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
14
|
Brasier AR. Interactions between epithelial mesenchymal plasticity, barrier dysfunction and innate immune pathways shape the genesis of allergic airway disease. Expert Rev Respir Med 2025; 19:29-41. [PMID: 39745473 PMCID: PMC11757041 DOI: 10.1080/17476348.2024.2449079] [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: 10/21/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025]
Abstract
INTRODUCTION In genetically predisposed individuals, exposure to aeroallergens and infections from RNA viruses shape epithelial barrier function, leading to Allergic Asthma (AA). Here, activated pattern recognition receptors (PRRs) in lower airway sentinel cells signal epithelial injury-repair pathways leading to cell-state changes [epithelial mesenchymal plasticity (EMP)], barrier disruption and sensitization. AREAS COVERED 1. Characteristics of sentinel epithelial cells of the bronchoalveolar junction, 2. The effect of aeroallergens on epithelial PRRs, 3. Role of tight junctions (TJs) in barrier function and how aeroallergens disrupt their function, 4. Induction of mucosal TGF autocrine loops activating type-2 innate lymphoid cells (ICL2s) leading to Th2 polarization, 5. How respiratory syncytial virus (RSV) directs goblet cell hyperplasia, and 6. Coupling of endoplasmic reticulum (ER) stress to metabolic adaptations and effects on basal lamina remodeling. EXPERT OPINION When aeroallergens or viral infections activate innate immunity in sentinel cells of the bronchoalveolar junction, normal barrier function is disrupted, promoting chronic inflammation and Th2 responses. An improved mechanistic understanding of how activated PRRs induce EMP couples with TJ disruption, metabolic reprogramming and ECM deposition provides new biologically validated targets to restore barrier function, reduce sensitization, and remodeling in AA.
Collapse
Affiliation(s)
- Allan R Brasier
- School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States
- The Institute for Clinical and Translational Research, Madison, Wisconsin, United States
| |
Collapse
|
15
|
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
|
16
|
Li C, Yu Y, Wan Z, Chiu MC, Huang J, Zhang S, Zhu X, Lan Q, Deng Y, Zhou Y, Xue W, Yue M, Cai JP, Yip CCY, Wong KKY, Liu X, Yu Y, Huang L, Chu H, Chan JFW, Clevers H, Yuen KY, Zhou J. Human respiratory organoids sustained reproducible propagation of human rhinovirus C and elucidation of virus-host interaction. Nat Commun 2024; 15:10772. [PMID: 39738014 PMCID: PMC11686133 DOI: 10.1038/s41467-024-55076-2] [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: 04/16/2024] [Accepted: 11/26/2024] [Indexed: 01/01/2025] Open
Abstract
The lack of a robust system to reproducibly propagate HRV-C, a family of viruses refractory to cultivation in standard cell lines, has substantially hindered our understanding of this common respiratory pathogen. We sought to develop an organoid-based system to reproducibly propagate HRV-C, and characterize virus-host interaction using respiratory organoids. We demonstrate that airway organoids sustain serial virus passage with the aid of CYT387-mediated immunosuppression, whereas nasal organoids that more closely simulate the upper airway achieve this without any intervention. Nasal organoids are more susceptible to HRV-C than airway organoids. Intriguingly, upon HRV-C infection, we observe an innate immune response that is stronger in airway organoids than in nasal organoids, which is reproduced in a Poly(I:C) stimulation assay. Treatment with α-CDHR3 and antivirals significantly reduces HRV-C viral growth in airway and nasal organoids. Additionally, an organoid-based immunofluorescence assay is established to titrate HRV-C infectious particles. Collectively, we develop an organoid-based system to reproducibly propagate the poorly cultivable HRV-C, followed by a comprehensive characterization of HRV-C infection and innate immunity in physiologically active respiratory organoids. The organoid-based HRV-C infection model can be extended for developing antiviral strategies. More importantly, our study has opened an avenue for propagating and studying other uncultivable human and animal viruses.
Collapse
Affiliation(s)
- Cun Li
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yifei Yu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Zhixin Wan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Man Chun Chiu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Jingjing Huang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Shuxin Zhang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xiaoxin Zhu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Qiaoshuai Lan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Yanlin Deng
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Ying Zhou
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Wei Xue
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ming Yue
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jian-Piao Cai
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Cyril Chik-Yan Yip
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kenneth Kak-Yuen Wong
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, and Queen Mary Hospital, Hong Kong, China
| | - Xiaojuan Liu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Yang Yu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | | | - Hin Chu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Jasper Fuk-Woo Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), and University Medical Center (UMC) Utrecht, Utrecht, the Netherlands
- Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Kwok Yung Yuen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jie Zhou
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China.
- BiomOrgan Ltd, Hong Kong, China.
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
17
|
Welch SR, Bilello JP, Carter K, Delang L, Dirr L, Durantel D, Feng JY, Gowen BB, Herrero LJ, Janeba Z, Kleymann G, Lee AA, Meier C, Moffat J, Schang LM, Schiffer JT, Seley-Radtke KL, Sheahan TP, Spengler JR. Meeting report of the 37th International Conference on Antiviral Research in Gold Coast, Australia, May 20-24, 2024, organized by the International Society for Antiviral Research. Antiviral Res 2024; 232:106037. [PMID: 39542140 PMCID: PMC11871649 DOI: 10.1016/j.antiviral.2024.106037] [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: 10/12/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
The 37th International Conference on Antiviral Research (ICAR) was held in Gold Coast, Australia, May 20-24, 2024. ICAR 2024 featured over 75 presentations along with two poster sessions and special events, including those specifically tailored for trainees and early-career scientists. The meeting served as a platform for the exchange of cutting-edge research, with presentations and discussions covering novel antiviral compounds, vaccine development, clinical trials, and therapeutic advancements. A comprehensive array of topics in antiviral science was covered, from the latest breakthroughs in antiviral drug development to innovative strategies for combating emerging viral threats. The keynote presentations provided fascinating insight into two diverse areas fundamental to medical countermeasure development and use, including virus emergence at the human-animal interface and practical considerations for bringing antivirals to the clinic. Additional sessions addressed a variety of timely post-pandemic topics, such as the hunt for broad spectrum antivirals, combination therapy, pandemic preparedness, application of in silico tools and AI in drug discovery, the virosphere, and more. Here, we summarize all the presentations and special sessions of ICAR 2024 and introduce the 38th ICAR, which will be held in Las Vegas, USA, March 17-21, 2025.
Collapse
Affiliation(s)
- Stephen R Welch
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | | | | | - Leen Delang
- Virus-Host Interactions & Therapeutic Approaches Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Larissa Dirr
- Institute for Biomedicine and Glycomics, Griffith University, Southport, QLD, Australia
| | - David Durantel
- Centre International de Recherche en Infectiologie (CIRI), Inserm_U1111, CNRS_UMR5308, Université Claude Bernard Lyon 1, F-69007, Lyon, France
| | - Joy Y Feng
- Division of the Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Brian B Gowen
- Institute for Antiviral Research and Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Lara J Herrero
- Institute for Biomedicine and Glycomics, Griffith University, Southport, QLD, Australia
| | - Zlatko Janeba
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 160 00, Prague, Czech Republic
| | - Gerald Kleymann
- Innovative Molecules GmbH, Lipowsky Str. 10, 81373, Munich, Bavaria, Germany
| | | | - Chris Meier
- Organic Chemistry, Department of Chemistry, Faculty of Sciences, University of Hamburg, Martin-Luther-King-Platz 6, Hamburg, Germany
| | - Jennifer Moffat
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Luis M Schang
- Baker Institute and Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Joshua T Schiffer
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, WA, USA; Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Katherine L Seley-Radtke
- Department of Chemistry & Biochemistry, University of Maryland, Baltimore County, Baltimore, MD, USA
| | - Timothy P Sheahan
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA; Rapidly Emerging Antiviral Drug Development Initiative, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jessica R Spengler
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
18
|
Gil-Cantero D, Mata CP, Valiente L, Rodríguez-Huete A, Valbuena A, Twarock R, Stockley PG, Mateu MG, Castón JR. Cryo-EM of human rhinovirus reveals capsid-RNA duplex interactions that provide insights into virus assembly and genome uncoating. Commun Biol 2024; 7:1501. [PMID: 39537894 PMCID: PMC11561273 DOI: 10.1038/s42003-024-07213-2] [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: 08/02/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
The cryo-EM structure of the human rhinovirus B14 determined in this study reveals 13-bp RNA duplexes symmetrically bound to regions around each of the 30 two-fold axes in the icosahedral viral capsid. The RNA duplexes (~12% of the ssRNA genome) define a quasi-dodecahedral cage that line a substantial part of the capsid interior surface. The RNA duplexes establish a complex network of non-covalent interactions with pockets in the capsid inner wall, including coulombic interactions with a cluster of basic amino acid residues that surround each RNA duplex. A direct comparison was made between the cryo-EM structure of RNA-filled virions and that of RNA-free (empty) capsids that resulted from genome release from a small fraction of viruses. The comparison reveals that some specific residues involved in capsid-duplex RNA interactions in the virion undergo remarkable conformational rearrangements upon RNA release from the capsid. RNA release is also associated with the asynchronous opening of channels at the 30 two-fold axes. The results provide further insights into the molecular mechanisms leading to assembly of rhinovirus particles and their genome uncoating during infection. They may also contribute to development of novel antiviral strategies aimed at interfering with viral capsid-genome interactions during the infectious cycle.
Collapse
Affiliation(s)
- David Gil-Cantero
- Department of Structure of Macromolecules, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Madrid, Spain
| | - Carlos P Mata
- Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- Biocomputing Unit, Department of Structure of Macromolecules, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Madrid, Spain
| | - Luis Valiente
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Alicia Rodríguez-Huete
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Alejandro Valbuena
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Reidun Twarock
- Department of Mathematics and Department of Biology, University of York, York, UK
| | - Peter G Stockley
- Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Mauricio G Mateu
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain.
| | - José R Castón
- Department of Structure of Macromolecules, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Madrid, Spain.
- Nanobiotechnology Associated Unit CNB-CSIC-IMDEA, Campus Cantoblanco, Madrid, Spain.
| |
Collapse
|
19
|
See WR, Yousefi M, Ooi YS. A review of virus host factor discovery using CRISPR screening. mBio 2024; 15:e0320523. [PMID: 39422472 PMCID: PMC11559068 DOI: 10.1128/mbio.03205-23] [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] [Indexed: 10/19/2024] Open
Abstract
The emergence of genome-scale forward genetic screening techniques, such as Haploid Genetic screen and clustered regularly interspaced short palindromic repeats (CRISPR) knockout screen has opened new horizons in our understanding of virus infection biology. CRISPR screening has become a popular tool for the discovery of novel host factors for several viruses due to its specificity and efficiency in genome editing. Here, we review how CRISPR screening has revolutionized our understanding of virus-host interactions from scientific and technological viewpoints. A summary of the published screens conducted thus far to uncover virus host factors is presented, highlighting their experimental design and significant findings. We will outline relevant methods for customizing the CRISPR screening process to answer more specific hypotheses and compile a glossary of conducted CRISPR screens to show their design aspects. Furthermore, using flaviviruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as examples, we hope to offer a broad-based perspective on the capabilities of CRISPR screening to serve as a reference point to guide future unbiased discovery of virus host factors.
Collapse
Affiliation(s)
- Wayne Ren See
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Meisam Yousefi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Yaw Shin Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
20
|
Tanyaratsrisakul S, Bochkov YA, White V, Lee H, Loeffler J, Everman J, Schiltz AM, Freeman KL, Hamlington KL, Secor EA, Jackson ND, Chu HW, Liu AH, Ledford JG, Kraft M, Seibold MA, Voelker DR, Numata M. Surfactant Protein A Inhibits Human Rhinovirus C Binding and Infection of Airway Epithelial Cells from Pediatric Asthma. Viruses 2024; 16:1709. [PMID: 39599822 PMCID: PMC11598966 DOI: 10.3390/v16111709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Rhinovirus C (RV-C) infection can trigger asthma exacerbations in children and adults, and RV-C-induced wheezing illnesses in preschool children correlate with the development of childhood asthma. Surfactant protein A (SP-A) plays a critical role in regulating pulmonary innate immunity by binding to numerous respiratory pathogens. Mature SP-A consists of multiple isoforms that form the hetero-oligomers of SP-A1 and SP-A2, organized in 18-mers. In this report, we examined the efficacy of SP-A to antagonize RV-C infection using the wild-type (RV-C15) and reporter-expressing (RV-C15-GFP) viruses in differentiated nasal epithelial cells (NECs) from asthmatic and non-asthmatic children. We also determined the antiviral mechanism of action of SP-A on RV-C15 infection. The native SP-A was purified from alveolar proteinosis patients. The recombinant (r) SP-A1 and SP-A2 variants were expressed in FreeStyle™ 293-F cells. SP-A reduced the fluorescent focus-forming units (FFUs) after RV-C15-GFP infection of NECs by 99%. Both simultaneous and 4 h post-infection treatment with SP-A inhibited RV-C15 and RV-C15-GFP viral RNA load by 97%. In addition, the antiviral genes and chemokines (IFN-λ, IRF-7, MDA-5, and CXLC11) were not induced in the infected NECs due to the inhibition of RV-C propagation by SP-A. Furthermore, SP-A bound strongly to RV-C15 in a dose- and Ca2+-dependent manner, and this interaction inhibited RV-C15 binding to NECs. In contrast, rSP-A1 did not bind to solid-phase RV-C15, whereas the rSP-A2 variants, [A91, K223] and [P91, Q223], had strong binding affinities to RV-C15, similar to native SP-A. This study demonstrates that SP-A might have potential as an antiviral for RV infection and RV-induced asthma exacerbations.
Collapse
Affiliation(s)
- Sasipa Tanyaratsrisakul
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA; (S.T.); (V.W.); (H.L.); (J.L.); (H.W.C.); (D.R.V.)
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ 85724, USA;
| | - Yury A. Bochkov
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA;
| | - Vanessa White
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA; (S.T.); (V.W.); (H.L.); (J.L.); (H.W.C.); (D.R.V.)
| | - Heejung Lee
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA; (S.T.); (V.W.); (H.L.); (J.L.); (H.W.C.); (D.R.V.)
| | - Jessica Loeffler
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA; (S.T.); (V.W.); (H.L.); (J.L.); (H.W.C.); (D.R.V.)
| | - Jamie Everman
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206, USA; (J.E.); (E.A.S.); (N.D.J.); (M.A.S.)
| | - Allison M. Schiltz
- Section of Pediatric Pulmonary & Sleep Medicine, Children’s Hospital Colorado and University of Colorado School of Medicine, Aurora, CO 80045, USA; (A.M.S.); (K.L.H.); (A.H.L.)
| | - Kristy L. Freeman
- Section of Pediatric Pulmonary & Sleep Medicine, Children’s Hospital Colorado and University of Colorado School of Medicine, Aurora, CO 80045, USA; (A.M.S.); (K.L.H.); (A.H.L.)
| | - Katharine L. Hamlington
- Section of Pediatric Pulmonary & Sleep Medicine, Children’s Hospital Colorado and University of Colorado School of Medicine, Aurora, CO 80045, USA; (A.M.S.); (K.L.H.); (A.H.L.)
| | - Elizabeth A. Secor
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206, USA; (J.E.); (E.A.S.); (N.D.J.); (M.A.S.)
| | - Nathan D. Jackson
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206, USA; (J.E.); (E.A.S.); (N.D.J.); (M.A.S.)
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA; (S.T.); (V.W.); (H.L.); (J.L.); (H.W.C.); (D.R.V.)
| | - Andrew H. Liu
- Section of Pediatric Pulmonary & Sleep Medicine, Children’s Hospital Colorado and University of Colorado School of Medicine, Aurora, CO 80045, USA; (A.M.S.); (K.L.H.); (A.H.L.)
| | - Julie G. Ledford
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ 85724, USA;
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85719, USA
| | - Monica Kraft
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Max A. Seibold
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206, USA; (J.E.); (E.A.S.); (N.D.J.); (M.A.S.)
- Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado-AMC, Aurora, CO 80045, USA
| | - Dennis R. Voelker
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA; (S.T.); (V.W.); (H.L.); (J.L.); (H.W.C.); (D.R.V.)
| | - Mari Numata
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA; (S.T.); (V.W.); (H.L.); (J.L.); (H.W.C.); (D.R.V.)
| |
Collapse
|
21
|
Djeddi S, Fernandez-Salinas D, Huang GX, Aguiar VRC, Mohanty C, Kendziorski C, Gazal S, Boyce JA, Ober C, Gern JE, Barrett NA, Gutierrez-Arcelus M. Rhinovirus infection of airway epithelial cells uncovers the non-ciliated subset as a likely driver of genetic risk to childhood-onset asthma. CELL GENOMICS 2024; 4:100636. [PMID: 39197446 PMCID: PMC11480861 DOI: 10.1016/j.xgen.2024.100636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 06/11/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024]
Abstract
Asthma is a complex disease caused by genetic and environmental factors. Studies show that wheezing during rhinovirus infection correlates with childhood asthma development. Over 150 non-coding risk variants for asthma have been identified, many affecting gene regulation in T cells, but the effects of most risk variants remain unknown. We hypothesized that airway epithelial cells could also mediate genetic susceptibility to asthma given they are the first line of defense against respiratory viruses and allergens. We integrated genetic data with transcriptomics of airway epithelial cells subject to different stimuli. We demonstrate that rhinovirus infection significantly upregulates childhood-onset asthma-associated genes, particularly in non-ciliated cells. This enrichment is also observed with influenza infection but not with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or cytokine activation. Overall, our results suggest that rhinovirus infection is an environmental factor that interacts with genetic risk factors through non-ciliated airway epithelial cells to drive childhood-onset asthma.
Collapse
Affiliation(s)
- Sarah Djeddi
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniela Fernandez-Salinas
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Licenciatura en Ciencias Genómicas, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos 62210, México
| | - George X Huang
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Vitor R C Aguiar
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Chitrasen Mohanty
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - Steven Gazal
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90007, USA; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90007, USA
| | - Joshua A Boyce
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - James E Gern
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53726, USA; Departments of Pediatrics and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Nora A Barrett
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Maria Gutierrez-Arcelus
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
22
|
Goldstein ME, Ignacio MA, Loube JM, Whorton MR, Scull MA. Human Stimulator of Interferon Genes Promotes Rhinovirus C Replication in Mouse Cells In Vitro and In Vivo. Viruses 2024; 16:1282. [PMID: 39205256 PMCID: PMC11358906 DOI: 10.3390/v16081282] [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: 05/02/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Rhinovirus C (RV-C) infects airway epithelial cells and is an important cause of acute respiratory disease in humans. To interrogate the mechanisms of RV-C-mediated disease, animal models are essential. Towards this, RV-C infection was recently reported in wild-type (WT) mice, yet, titers were not sustained. Therefore, the requirements for RV-C infection in mice remain unclear. Notably, prior work has implicated human cadherin-related family member 3 (CDHR3) and stimulator of interferon genes (STING) as essential host factors for virus uptake and replication, respectively. Here, we report that even though human (h) and murine (m) CDHR3 orthologs have similar tissue distribution, amino acid sequence homology is limited. Further, while RV-C can replicate in mouse lung epithelial type 1 (LET1) cells and produce infectious virus, we observed a significant increase in the frequency and intensity of dsRNA-positive cells following hSTING expression. Based on these findings, we sought to assess the impact of hCDHR3 and hSTING on RV-C infection in mice in vivo. Thus, we developed hCDHR3 transgenic mice, and utilized adeno-associated virus (AAV) to deliver hSTING to the murine airways. Subsequent challenge of these mice with RV-C15 revealed significantly higher titers 24 h post-infection in mice expressing both hCDHR3 and hSTING-compared to either WT mice, or mice with hCDHR3 or hSTING alone, indicating more efficient infection. Ultimately, this mouse model can be further engineered to establish a robust in vivo model, recapitulating viral dynamics and disease.
Collapse
Affiliation(s)
- Monty E. Goldstein
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, 3134 Biosciences Research Building, University of Maryland, College Park, MD 20742, USA
| | - Maxinne A. Ignacio
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, 3134 Biosciences Research Building, University of Maryland, College Park, MD 20742, USA
| | - Jeffrey M. Loube
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, 3134 Biosciences Research Building, University of Maryland, College Park, MD 20742, USA
| | - Matthew R. Whorton
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Margaret A. Scull
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, 3134 Biosciences Research Building, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
23
|
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
|
24
|
Xiao M, Banu A, Jia Y, Chang M, Wang G, An J, Huang Y, Hu X, Tang C, Li Z, Niu Y, Tian X, Deng W, Tang C, Du J, Cui X, Chan JFW, Peng R, Yin F. Circulation pattern and genetic variation of rhinovirus infection among hospitalized children on Hainan Island, before and after the dynamic zero-COVID policy, from 2021 to 2023. J Med Virol 2024; 96:e29755. [PMID: 38922896 DOI: 10.1002/jmv.29755] [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: 04/03/2024] [Revised: 05/17/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
Throughout the COVID-19 pandemic, rhinovirus (RV) remained notable persistence, maintaining its presence while other seasonal respiratory viruses were largely suppressed by pandemic restrictions during national lockdowns. This research explores the epidemiological dynamics of RV infections among pediatric populations on Hainan Island, China, specifically focusing on the impact before and after the zero-COVID policy was lifted. From January 2021 to December 2023, 19 680 samples were collected from pediatric patients hospitalized with acute lower respiratory tract infections (ARTIs) at the Hainan Maternal and Child Health Hospital. The infection of RV was detected by tNGS. RV species and subtypes were identified in 32 RV-positive samples representing diverse time points by analyzing the VP4/VP2 partial regions. Among the 19 680 pediatric inpatients with ARTIs analyzed, 21.55% were found to be positive for RV infection, with notable peaks observed in April 2021 and November 2022. A gradual annual decline in RV infections was observed, alongside a seasonal pattern of higher prevalence during the colder months. The highest proportion of RV infections was observed in the 0-1-year age group. Phylogenetic analysis on 32 samples indicated a trend from RV-A to RV-C in 2022. This observation suggests potential evolving dynamics within the RV species although further studies are needed due to the limited sample size. The research emphasizes the necessity for ongoing surveillance and targeted management, particularly for populations highly susceptible to severe illnesses caused by RV infections.
Collapse
Affiliation(s)
- Meifang Xiao
- Department of Clinical Laboratory, Center for Laboratory Medicine, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
- Department of Microbiology, Faculty of Medicine, Lincoln University College, Petaling Jaya, Malaysia
| | - Afreen Banu
- Department of Microbiology, Faculty of Medicine, Lincoln University College, Petaling Jaya, Malaysia
| | - Yibo Jia
- Medical Administration Division, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
- International School of Public Health and One Health, Hainan Medical College, Haikou, Hainan, China
| | - Meng Chang
- Department of Clinical Laboratory, Center for Laboratory Medicine, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Gaoyu Wang
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Jing An
- Department of Clinical Laboratory, Center for Laboratory Medicine, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Yi Huang
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Xiaoyuan Hu
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Chuanning Tang
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Zihan Li
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Yi Niu
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Xiuying Tian
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Wanxin Deng
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Cheng Tang
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Jiang Du
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiuji Cui
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
- Department of Pathogen Biology, Hainan Medical University, Haikou, Hainan, China
| | - Jasper Fuk-Woo Chan
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, and Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Ruoyan Peng
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Feifei Yin
- Department of Clinical Laboratory, Center for Laboratory Medicine, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
- Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
- Department of Pathogen Biology, Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
25
|
Sang L, Gong X, Huang Y, Zhang L, Sun J. Immunotherapeutic implications on targeting the cytokines produced in rhinovirus-induced immunoreactions. FRONTIERS IN ALLERGY 2024; 5:1427762. [PMID: 38859875 PMCID: PMC11163110 DOI: 10.3389/falgy.2024.1427762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 05/13/2024] [Indexed: 06/12/2024] Open
Abstract
Rhinovirus is a widespread virus associated with several respiratory diseases, especially asthma exacerbation. Currently, there are no accurate therapies for rhinovirus. Encouragingly, it is found that during rhinovirus-induced immunoreactions the levels of certain cytokines in patients' serum will alter. These cytokines may have pivotal pro-inflammatory or anti-inflammatory effects via their specific mechanisms. Thus far, studies have shown that inhibitions of cytokines such as IL-1, IL-4, IL-5, IL-6, IL-13, IL-18, IL-25, and IL-33 may attenuate rhinovirus-induced immunoreactions, thereby relieving rhinovirus infection. Furthermore, such therapeutics for rhinovirus infection can be applied to viruses of other species, with certain practicability.
Collapse
Affiliation(s)
- Le Sang
- Department of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province, China
| | - Xia Gong
- Department of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province, China
| | - Yunlei Huang
- Department of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province, China
| | - Linling Zhang
- Department of Respiratory Medicine, Shaoxing People’s Hospital, Shaoxing City, Zhejiang Province, China
| | - Jian Sun
- Department of Respiratory Medicine, Shaoxing People’s Hospital, Shaoxing City, Zhejiang Province, China
| |
Collapse
|
26
|
Naeem A, Alkadi HS, Manzoor MU, Yousaf I, Awadalla M, Alturaiki W, AlYami AS, Zafar A, Alosaimi B. Mutations at the conserved N-Terminal of the human Rhinovirus capsid gene VP4, and their impact on the immune response. J Immunoassay Immunochem 2024; 45:271-291. [PMID: 38551181 DOI: 10.1080/15321819.2024.2323460] [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: 06/25/2024]
Abstract
Rhinoviruses (RV) are the major cause of chronic obstructive pulmonary disease and are associated with exacerbation development as well as community-acquired pneumonia in children, leading to substantial morbidity, mortality, and hospital admission. Here we have examined how changes at the amino terminal of the conserved VP4 epitope of different RV serotypes may affect pulmonary cytokine and chemokine responses and disease severity. Samples positive for rhinovirus were used for genetic characterization, followed by profiling gene expression of pulmonary Th1 and Th2 cytokines/chemokines by RT-PCR arrays. Genetic sequencing and homology 3D modeling revealed changes at the amino terminal of the conserved viral protein 4 (VP4) epitope in the RV-A101 serotype, especially serine at several positions that are important for interactive binding with the host immune cells. We found dysregulation of pulmonary gene expression of Th1- and Th2-related cytokines and chemokines in RV-A 101 and RV-C 8 pneumonia patients. These findings might contribute to a better understanding of RV immunity and the potential mechanisms underlying the pathogenesis of severe RV infections, but further functional studies are needed to confirm the causal relationship.
Collapse
Affiliation(s)
- Asif Naeem
- Department of Research Labs, Research Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Haitham S Alkadi
- Department of Research Labs, Research Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Muhammad U Manzoor
- Department of Medical Imaging, Diagnostic & Interventional Neuroradiology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Imran Yousaf
- Department of Medical Imaging, Diagnostic & Interventional Neuroradiology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Maaweya Awadalla
- Department of Research Labs, Research Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Wael Alturaiki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Riyadh Region, Saudi Arabia
| | - Ahmad S AlYami
- Pathology and Clinical Laboratory Medicine Administration, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Adnan Zafar
- Pediatric Department, John Hopkins Aramco Healthcare, Al-Ahsa, Saudi Arabia
| | - Bandar Alosaimi
- Department of Research Labs, Research Center, King Fahad Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
27
|
An Q, Cao Y, Guo W, Jiang Z, Luo H, Liu H, Zhan X. Identification of common genes of rhinovirus single/double‑stranded RNA‑induced asthma deterioration by bioinformatics analysis. Exp Ther Med 2024; 27:210. [PMID: 38590566 PMCID: PMC11000450 DOI: 10.3892/etm.2024.12498] [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: 08/28/2023] [Accepted: 01/29/2024] [Indexed: 04/10/2024] Open
Abstract
Rhinovirus (RV) is the most common respiratory virus affecting humans. The majority of asthma deteriorations are triggered by RV infections. However, whether the effects of RV single- and double-stranded RNA on asthma deterioration have common target genes needs to be further studied. In the present study, two datasets (GSE51392 and GSE30326) were used to screen for common differentially expressed genes (cDEGs). The molecular function, signaling pathways, interaction networks, hub genes, key modules and regulatory molecules of cDEGs were systematically analyzed using online tools such as Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, STRING and NetworkAnalyst. Finally, the hub genes STAT1 and IFIH1 were verified in clinical samples using reverse transcription-quantitative PCR (RT-qPCR). A total of 85 cDEGs were identified. Function analysis revealed that cDEGs served an important role in the innate immune response to viruses and its regulation. Signal transducer and activator of transcription 1 (STAT1), interferon induced with helicase C domain 1 (IFIH1), interferon regulatory factor 7 (IRF7), DExD/H box helicase 58 (DDX58) and interferon-stimulating gene 15 (ISG15) were detected to be hub genes based on the protein-protein interactions and six topological algorithms. A key module involved in influenza A, the Toll-like receptor signaling pathway, was identified using Cytoscape software. The hub genes were regulated by GATA-binding factor 2 and microRNA-146a-5p. In addition, RT-qPCR indicated that the expression levels of the hub genes STAT1 and IFIH1 were low during asthma deterioration compared with post-treatment recovery samples. The present study enhanced the understanding of the mechanism of RV-induced asthma deterioration.
Collapse
Affiliation(s)
- Qian An
- Department of Respiratory and Critical Care Medicine, Wuhu Hospital of Traditional Chinese Medicine, Wuhu, Anhui 241000, P.R. China
| | - Yi Cao
- Department of Medical Parasitology, School of Basic Medicine, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Wei Guo
- Department of Medical Parasitology, School of Basic Medicine, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Ziyun Jiang
- Department of Medical Parasitology, School of Basic Medicine, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Hui Luo
- Department of Medical Parasitology, School of Basic Medicine, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Hui Liu
- Department of Medical Parasitology, School of Basic Medicine, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Xiaodong Zhan
- Department of Medical Parasitology, School of Basic Medicine, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| |
Collapse
|
28
|
Miah M, Davis AM, Hannoun C, Said JS, Fitzek M, Preston M, Smith D, Uwamariya C, Kärmander A, Lundbäck T, Bergström T, Trybala E. Identification of epidermal growth factor receptor-tyrosine kinase inhibitor targeting the VP1 pocket of human rhinovirus. Antimicrob Agents Chemother 2024; 68:e0106423. [PMID: 38349161 PMCID: PMC10916396 DOI: 10.1128/aac.01064-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 01/10/2024] [Indexed: 03/07/2024] Open
Abstract
Screening a library of 1,200 preselected kinase inhibitors for anti-human rhinovirus 2 (HRV-2) activity in HeLa cells identified a class of epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKI) as effective virus blockers. These were based on the 4-anilinoquinazoline-7-oxypiperidine scaffold, with the most potent representative AZ5385 inhibiting the virus with EC50 of 0.35 µM. Several structurally related analogs confirmed activity in the low µM range, while interestingly, other TKIs targeting EGFR lacked anti-HRV-2 activity. To further probe this lack of association between antiviral activity and EGFR inhibition, we stained infected cells with antibodies specific for activated EGFR (Y1068) and did not observe a dependency on EGFR-TK activity. Instead, consecutive passages of HRV-2 in HeLa cells in the presence of a compound and subsequent nucleotide sequence analysis of resistant viral variants identified the S181T and T210A alterations in the major capsid VP1 protein, with both residues located in the vicinity of a known hydrophobic pocket on the viral capsid. Further characterization of the antiviral effects of AZ5385 showed a modest virus-inactivating (virucidal) activity, while anti-HRV-2 activity was still evident when the inhibitor was added as late as 10 h post infection. The RNA copy/infectivity ratio of HRV-2 propagated in AZ5385 presence was substantially higher than that of control HRV indicating that the compound preferentially targeted HRV progeny virions during their maturation in infected cells. Besides HRV, the compound showed anti-respiratory syncytial virus activity, which warrants its further studies as a candidate compound against viral respiratory infections.
Collapse
Affiliation(s)
- Masum Miah
- Department of Infectious Disease, Section for Clinical Virology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Andrew M. Davis
- Discovery Sciences, BioPharmaceutical R&D, AstraZeneca, Mölndal, Sweden
- Discovery Sciences, BioPharmaceutical R&D, AstraZeneca, Cambridge, United Kingdom
| | - Charles Hannoun
- Department of Infectious Disease, Section for Clinical Virology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Joanna S. Said
- Department of Infectious Disease, Section for Clinical Virology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Martina Fitzek
- HTS Discovery Sciences, BioPharmaceutical R&D, AstraZeneca, Macclesfield, United Kingdom
| | - Marian Preston
- HTS Discovery Sciences, BioPharmaceutical R&D, AstraZeneca, Macclesfield, United Kingdom
| | - Dave Smith
- Discovery Sciences, BioPharmaceutical R&D, AstraZeneca, Cambridge, United Kingdom
| | - Colores Uwamariya
- Department of Infectious Disease, Section for Clinical Virology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Ambjörn Kärmander
- Department of Infectious Disease, Section for Clinical Virology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Thomas Lundbäck
- Discovery Sciences, BioPharmaceutical R&D, AstraZeneca, Mölndal, Sweden
| | - Tomas Bergström
- Department of Infectious Disease, Section for Clinical Virology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Edward Trybala
- Department of Infectious Disease, Section for Clinical Virology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
29
|
Hizawa N. Common Pathogeneses Underlying Asthma and Chronic Obstructive Pulmonary Disease -Insights from Genetic Studies. Int J Chron Obstruct Pulmon Dis 2024; 19:633-642. [PMID: 38464563 PMCID: PMC10922945 DOI: 10.2147/copd.s441992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/21/2024] [Indexed: 03/12/2024] Open
Abstract
Neither asthma nor chronic obstructive pulmonary disease (COPD) is a single disease consisting of a uniform pathogenesis; rather, they are both syndromes that result from a variety of basic distinct pathogeneses. Many of the basic pathogeneses overlap between the two diseases, and multiple basic pathogeneses are simultaneously involved at varying proportions in individual patients. The specific combination of different basic pathogeneses in each patient determines the phenotype of the patient, and it varies widely from patient to patient. For example, type 2 airway inflammation and neutrophilic airway inflammation may coexist in the same patient, and quite a few patients have clinical characteristics of both asthma and COPD. Even in the same patient, the contribution of each pathogenesis is expected to differ at different life stages (eg, childhood, adolescence, middle age, and older), during different seasons (eg, high seasons for hay fever and rhinovirus infection), and depending on the nature of treatments. This review describes several basic pathogeneses commonly involved in both asthma and COPD, including chronic non-type 2 inflammation, type 2 inflammation, viral infections, and lung development. Understanding of the basic molecular pathogeneses in individual patients, rather than the use of clinical diagnosis, such as asthma, COPD, or even asthma COPD overlap, will enable us to better deal with the diversity seen in disease states, and lead to optimal treatment practices tailored for each patient with less disease burden, such as drug-induced side effects, and improved prognosis. Furthermore, we can expect to focus on these molecular pathways as new drug discovery targets.
Collapse
Affiliation(s)
- Nobuyuki Hizawa
- Department of Pulmonary Medicine, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
30
|
Lang J, Soddemann M, Edwards MJ, Wilson GC, Lang KS, Gulbins E. Sphingosine Prevents Rhinoviral Infections. Int J Mol Sci 2024; 25:2486. [PMID: 38473734 DOI: 10.3390/ijms25052486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 03/14/2024] Open
Abstract
Rhinoviral infections cause approximately 50% of upper respiratory tract infections and novel treatment options are urgently required. We tested the effects of 10 μM to 20 μM sphingosine on the infection of cultured and freshly isolated human cells with minor and major group rhinovirus in vitro. We also performed in vivo studies on mice that were treated with an intranasal application of 10 μL of either a 10 μM or a 100 μM sphingosine prior and after infection with rhinovirus strains 1 and 2 and determined the infection of nasal epithelial cells in the presence or absence of sphingosine. Finally, we determined and characterized a direct binding of sphingosine to rhinovirus. Our data show that treating freshly isolated human nasal epithelial cells with sphingosine prevents infections with rhinovirus strains 2 (minor group) and 14 (major group). Nasal infection of mice with rhinovirus 1b and 2 is prevented by the intranasal application of sphingosine before or as long as 8 h after infection with rhinovirus. Nasal application of the same doses of sphingosine exerts no adverse effects on epithelial cells as determined by hemalaun and TUNEL stainings. The solvent, octylglucopyranoside, was without any effect in vitro and in vivo. Mechanistically, we demonstrate that the positively charged lipid sphingosine binds to negatively charged molecules in the virus, which seems to prevent the infection of epithelial cells. These findings indicate that exogenous sphingosine prevents infections with rhinoviruses, a finding that could be therapeutically exploited. In addition, we demonstrated that sphingosine has no obvious adverse effects on the nasal mucosa. Sphingosine prevents rhinoviral infections by a biophysical mode of action, suggesting that sphingosine could serve to prevent many viral infections of airways and epithelial cells in general. Future studies need to determine the molecular mechanisms of how sphingosine prevents rhinoviral infections and whether sphingosine also prevents infections with other viruses inducing respiratory tract infections. Furthermore, our studies do not provide detailed pharmacokinetics that are definitely required before the further development of sphingosine.
Collapse
Affiliation(s)
- Judith Lang
- Department of Immunology, University Clinic, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Matthias Soddemann
- Department of Molecular Biology, University Clinic, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Michael J Edwards
- Department of Molecular Biology, University Clinic, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Gregory C Wilson
- Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Karl S Lang
- Department of Immunology, University Clinic, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Erich Gulbins
- Department of Molecular Biology, University Clinic, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
- Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
31
|
Djeddi S, Fernandez-Salinas D, Huang GX, Aguiar VRC, Mohanty C, Kendziorski C, Gazal S, Boyce J, Ober C, Gern J, Barrett N, Gutierrez-Arcelus M. Rhinovirus infection of airway epithelial cells uncovers the non-ciliated subset as a likely driver of genetic susceptibility to childhood-onset asthma. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.02.24302068. [PMID: 38370648 PMCID: PMC10871459 DOI: 10.1101/2024.02.02.24302068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Asthma is a complex disease caused by genetic and environmental factors. Epidemiological studies have shown that in children, wheezing during rhinovirus infection (a cause of the common cold) is associated with asthma development during childhood. This has led scientists to hypothesize there could be a causal relationship between rhinovirus infection and asthma or that RV-induced wheezing identifies individuals at increased risk for asthma development. However, not all children who wheeze when they have a cold develop asthma. Genome-wide association studies (GWAS) have identified hundreds of genetic variants contributing to asthma susceptibility, with the vast majority of likely causal variants being non-coding. Integrative analyses with transcriptomic and epigenomic datasets have indicated that T cells drive asthma risk, which has been supported by mouse studies. However, the datasets ascertained in these integrative analyses lack airway epithelial cells. Furthermore, large-scale transcriptomic T cell studies have not identified the regulatory effects of most non-coding risk variants in asthma GWAS, indicating there could be additional cell types harboring these "missing regulatory effects". Given that airway epithelial cells are the first line of defense against rhinovirus, we hypothesized they could be mediators of genetic susceptibility to asthma. Here we integrate GWAS data with transcriptomic datasets of airway epithelial cells subject to stimuli that could induce activation states relevant to asthma. We demonstrate that epithelial cultures infected with rhinovirus significantly upregulate childhood-onset asthma-associated genes. We show that this upregulation occurs specifically in non-ciliated epithelial cells. This enrichment for genes in asthma risk loci, or 'asthma heritability enrichment' is also significant for epithelial genes upregulated with influenza infection, but not with SARS-CoV-2 infection or cytokine activation. Additionally, cells from patients with asthma showed a stronger heritability enrichment compared to cells from healthy individuals. Overall, our results suggest that rhinovirus infection is an environmental factor that interacts with genetic risk factors through non-ciliated airway epithelial cells to drive childhood-onset asthma.
Collapse
|
32
|
Carneiro J, Tavendale R, Mukhopadhyay S, Soares P. Does CDHR3 gene polymorphism affect paediatric asthma and its treatment response? Clin Exp Allergy 2024; 54:159-161. [PMID: 38017357 DOI: 10.1111/cea.14430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/18/2023] [Accepted: 11/02/2023] [Indexed: 11/30/2023]
Affiliation(s)
- Joana Carneiro
- NOVA National School of Public Health, NOVA University Lisbon, Lisbon, Portugal
| | - Roger Tavendale
- Division of Population and Health Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Somnath Mukhopadhyay
- Academic Department of Paediatrics, Brighton & Sussex Medical School, Royal Alexandra Children's Hospital, Brighton, UK
| | - Patrícia Soares
- NOVA National School of Public Health, Public Health Research Center, Comprehensive Health Research Center, CHRC, NOVA University Lisbon, Lisbon, Portugal
| |
Collapse
|
33
|
Bakhache W, Orr W, McCormick L, Dolan PT. Uncovering Structural Plasticity of Enterovirus A through Deep Insertional and Deletional Scanning. RESEARCH SQUARE 2024:rs.3.rs-3835307. [PMID: 38410474 PMCID: PMC10896406 DOI: 10.21203/rs.3.rs-3835307/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Insertions and deletions (InDels) are essential sources of novelty in protein evolution. In RNA viruses, InDels cause dramatic phenotypic changes contributing to the emergence of viruses with altered immune profiles and host engagement. This work aimed to expand our current understanding of viral evolution and explore the mutational tolerance of RNA viruses to InDels, focusing on Enterovirus A71 (EV-A71) as a prototype for Enterovirus A species (EV-A). Using newly described deep InDel scanning approaches, we engineered approximately 45,000 insertions and 6,000 deletions at every site across the viral proteome, quantifying their effects on viral fitness. As a general trend, most InDels were lethal to the virus. However, our screen reproducibly identified a set of InDel-tolerant regions, demonstrating our ability to comprehensively map tolerance to these mutations. Tolerant sites highlighted structurally flexible and mutationally plastic regions of viral proteins that avoid core structural and functional elements. Phylogenetic analysis on EV-A species infecting diverse mammalian hosts revealed that the experimentally-identified hotspots overlapped with sites of InDels across the EV-A species, suggesting structural plasticity at these sites is an important function for InDels in EV speciation. Our work reveals the fitness effects of InDels across EV-A71, identifying regions of evolutionary capacity that require further monitoring, which could guide the development of Enterovirus vaccines.
Collapse
Affiliation(s)
- William Bakhache
- Quantitative Virology and Evolution Unit, Laboratory of Viral Diseases, NIH-NIAID Division of Intramural Research, Bethesda, MD, USA
| | - Walker Orr
- Quantitative Virology and Evolution Unit, Laboratory of Viral Diseases, NIH-NIAID Division of Intramural Research, Bethesda, MD, USA
| | - Lauren McCormick
- Quantitative Virology and Evolution Unit, Laboratory of Viral Diseases, NIH-NIAID Division of Intramural Research, Bethesda, MD, USA
- Department of Biology, University of Oxford, Oxford, UK
| | - Patrick T. Dolan
- Quantitative Virology and Evolution Unit, Laboratory of Viral Diseases, NIH-NIAID Division of Intramural Research, Bethesda, MD, USA
| |
Collapse
|
34
|
Kageyama T, Ito T, Tanaka S, Nakajima H. Physiological and immunological barriers in the lung. Semin Immunopathol 2024; 45:533-547. [PMID: 38451292 PMCID: PMC11136722 DOI: 10.1007/s00281-024-01003-y] [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: 10/04/2023] [Accepted: 02/10/2024] [Indexed: 03/08/2024]
Abstract
The lungs serve as the primary organ for respiration, facilitating the vital exchange of gases with the bloodstream. Given their perpetual exposure to external particulates and pathogens, they possess intricate protective barriers. Cellular adhesion in the lungs is robustly maintained through tight junctions, adherens junctions, and desmosomes. Furthermore, the pulmonary system features a mucociliary clearance mechanism that synthesizes mucus and transports it to the outside. This mucus is enriched with chemical barriers like antimicrobial proteins and immunoglobulin A (IgA). Additionally, a complex immunological network comprising epithelial cells, neural cells, and immune cells plays a pivotal role in pulmonary defense. A comprehensive understanding of these protective systems offers valuable insights into potential pathologies and their therapeutic interventions.
Collapse
Affiliation(s)
- Takahiro Kageyama
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan.
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan.
| | - Takashi Ito
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba, Japan
| | - Shigeru Tanaka
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba, Japan
| |
Collapse
|
35
|
Wolters AAB, Kersten ETG, Koppelman GH. Genetics of preschool wheeze and its progression to childhood asthma. Pediatr Allergy Immunol 2024; 35:e14067. [PMID: 38284918 DOI: 10.1111/pai.14067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 01/30/2024]
Abstract
Wheezing is a common and heterogeneous condition in preschool children. In some countries, the prevalence can be as high as 30% and up to 50% of all children experience wheezing before the age of 6. Asthma often starts with preschool wheeze, but not all wheezing children will develop asthma at school age. At this moment, it is not possible to accurately predict which wheezing children will develop asthma. Recently, studying the genetics of wheeze and the childhood-onset of asthma have grown in interest. Childhood-onset asthma has a stronger heritability in comparison with adult-onset asthma. In early childhood asthma exacerbations, CDHR3, which encodes the receptor for Rhinovirus C, was identified, as well as IL33, and the 17q locus that includes GSDMB and ORMDL3 genes. The 17q locus is the strongest wheeze and childhood-onset asthma locus, and was shown to interact with many environmental factors, including smoking and infections. Finally, ANXA1 was recently associated with early-onset, persistent wheeze. ANXA1 may help resolve eosinophilic inflammation. Overall, despite its complexities, genetic approaches to unravel the early-onset of wheeze and asthma are promising, since these shed more light on mechanisms of childhood asthma-onset. Implicated genes point toward airway epithelium and its response to external factors, such as viral infections. However, the heterogeneity of wheeze phenotypes complicates genetic studies. It is therefore important to define accurate wheezing phenotypes and forge larger international collaborations to gain a better understanding of the pathways underlying early-onset asthma.
Collapse
Affiliation(s)
- Alba A B Wolters
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Elin T G Kersten
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
36
|
Zhao P, Shao N, Dong J, Su H, Sui H, Zhang T, Yang F. Genetic diversity and characterization of rhinoviruses from Chinese clinical samples with a global perspective. Microbiol Spectr 2023; 11:e0084023. [PMID: 37733296 PMCID: PMC10715137 DOI: 10.1128/spectrum.00840-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 08/03/2023] [Indexed: 09/22/2023] Open
Abstract
IMPORTANCE Based on clinical samples collected in China, we detected and reported 22 types for the first time in China, as well as three types for the first time in Asia, and reported their genetic characteristics and diversity. We identified a novel type of Rhinovirus (RV), A110, highlighting its unique genetic features. We annotated the genomic structure and serotype of all the existing RV sequences in the database, and four novel RV types were identified and their genetic diversity reported. Combined with the sequence annotation, we constructed a complete VP1 data set of RV and conducted the first large-scale evolutionary dynamics analysis of RV. Based on a high-quality data set, we conducted a comprehensive analysis of the guanine-cytosine (GC) content variations among serotypes of RVs. This study provides crucial theoretical support and valuable data for understanding RV's genetic diversity and developing antiviral strategies.
Collapse
Affiliation(s)
- Peng Zhao
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Shao
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Dong
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haoxiang Su
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongtao Sui
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting Zhang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fan Yang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China
| |
Collapse
|
37
|
Kan-O K, Washio Y, Oki T, Fujimoto T, Ninomiya T, Yoshida M, Fujita M, Nakanishi Y, Matsumoto K. Effects of treatment with corticosteroids on human rhinovirus-induced asthma exacerbations in pediatric inpatients: a prospective observational study. BMC Pulm Med 2023; 23:487. [PMID: 38053068 DOI: 10.1186/s12890-023-02798-6] [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: 07/02/2023] [Accepted: 11/30/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND Human rhinoviruses (HRVs) infection is a common cause of exacerbations in pediatric patients with asthma. However, the effects of corticosteroids on HRV-induced exacerbations in pediatric asthma are unknown. We conducted a prospective observational study to determine the viral pathogens in school-age pediatric inpatients with asthma exacerbations. We assessed the effects of maintenance inhaled corticosteroids (ICS) on the detection rates of HRV species and treatment periods of systemic corticosteroids during exacerbations on pulmonary lung function after exacerbations. METHODS Nasopharyngeal samples and clinical information were collected from 59 patients with asthma exacerbations between April 2018 and March 2020. Pulmonary function tests were carried out 3 months after exacerbations in 18 HRV-positive patients. Changes in forced expiratory volume in 1 second (FEV1)% predicted from baseline in a stable state were compared according to the treatment periods of systemic corticosteroids. RESULTS Fifty-four samples collected from hospitalized patients were analyzed, and viral pathogens were identified in 45 patients (83.3%) using multiplex PCR assay. HRV-A, -B, and -C were detected in 16 (29.6%), one (1.9%), and 16 (29.6%) patients, respectively. The detection rates of HRV-C were lower in the ICS-treated group compared with those in the ICS-untreated group (p = 0.01), whereas maintenance ICS treatment did not affect the detection rate for viral pathogens in total and HRV-A. Changes in FEV1% predicted in patients treated with systemic corticosteroids for 6-8 days (n = 10; median, 4.90%) were higher than those in patients treated for 3-5 days (n = 8; median, - 10.25%) (p = 0.0085). CONCLUSIONS Maintenance ICS reduced the detection rates of HRV (mainly HRV-C) in school-age inpatients with asthma exacerbations, and the treatment periods of systemic corticosteroids during exacerbations affected lung function after HRV-induced exacerbations. The protective effects of corticosteroids on virus-induced asthma exacerbations may be dependent upon the types of viral pathogen.
Collapse
Affiliation(s)
- Keiko Kan-O
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Yasuyoshi Washio
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takeshi Oki
- Division of Pediatrics, National Hospital Organization Fukuoka National Hospital, Fukuoka, Japan
| | - Tsuguto Fujimoto
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takahito Ninomiya
- Division of Pediatrics, National Hospital Organization Fukuoka National Hospital, Fukuoka, Japan
| | - Makoto Yoshida
- Division of Respiratory Medicine, National Hospital Organization Fukuoka National Hospital, Fukuoka, Japan
| | - Masaki Fujita
- Department of Respiratory Medicine, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Yoichi Nakanishi
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koichiro Matsumoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Division of Respirology, Department of Medicine, Fukuoka Dental College, Fukuoka, Japan
| |
Collapse
|
38
|
Xing Y, Leung ASY, Wong GWK. From preschool wheezing to asthma: Environmental determinants. Pediatr Allergy Immunol 2023; 34:e14049. [PMID: 38010001 DOI: 10.1111/pai.14049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 11/08/2023] [Indexed: 11/29/2023]
Abstract
Wheezing is common among preschool children, representing a group of highly heterogeneous conditions with varying natural history. Several phenotypes of wheezing have been proposed to facilitate the identification of young children who are at risk of subsequent development of asthma. Epidemiological and immunological studies across different populations have revealed the key role of environmental factors in influencing the progression from preschool wheezing to childhood asthma. Significant risk factors include severe respiratory infections, allergic sensitization, and exposure to tobacco smoke. In contrast, a farming/rural environment has been linked to asthma protection in both human and animal studies. Early and intense exposures to microorganisms and microbial metabolites have been demonstrated to alter host immune responses to allergens and viruses, thereby driving the trajectory away from wheezing illness and asthma. Ongoing clinical trials of candidate microbes and microbial products have shown promise in shaping the immune function to reduce episodes of viral-induced wheezing. Moreover, restoring immune training may be especially important for young children who had reduced microbial exposure due to pandemic restrictions. A comprehensive understanding of the role of modifiable environmental factors will pave the way for developing targeted prevention strategies for preschool wheezing and asthma.
Collapse
Affiliation(s)
- Yuhan Xing
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Special Administrative Region, China
| | - Agnes Sze-Yin Leung
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Special Administrative Region, China
| | - Gary Wing-Kin Wong
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Special Administrative Region, China
| |
Collapse
|
39
|
Medeleanu MV, Qian YC, Moraes TJ, Subbarao P. Early-immune development in asthma: A review of the literature. Cell Immunol 2023; 393-394:104770. [PMID: 37837916 DOI: 10.1016/j.cellimm.2023.104770] [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: 06/29/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/16/2023]
Abstract
This review presents a comprehensive examination of the various factors contributing to the immunopathogenesis of asthma from the prenatal to preschool period. We focus on the contributions of genetic and environmental components as well as the role of the nasal and gut microbiome on immune development. Predisposing genetic factors, including inherited genes associated with increased susceptibility to asthma, are discussed alongside environmental factors such as respiratory viruses and pollutant exposure, which can trigger or exacerbate asthma symptoms. Furthermore, the intricate interplay between the nasal and gut microbiome and the immune system is explored, emphasizing their influence on allergic immune development and response to environmental stimuli. This body of literature underscores the necessity of a comprehensive approach to comprehend and manage asthma, as it emphasizes the interactions of multiple factors in immune development and disease progression.
Collapse
Affiliation(s)
- Maria V Medeleanu
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Canada; Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Canada
| | - Yu Chen Qian
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Canada; Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Canada
| | - Theo J Moraes
- Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Canada; Laboratory Medicine and Pathology, Temerty Faculty of Medicine, University of Toronto, Canada; Department of Paediatrics, Temerty Faculty of Medicine, University of Toronto, Canada; Division of Respiratory Medicine, Hospital for Sick Children, Canada
| | - Padmaja Subbarao
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Canada; Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Canada; Department of Paediatrics, Temerty Faculty of Medicine, University of Toronto, Canada; Division of Respiratory Medicine, Hospital for Sick Children, Canada; Epidemiology Division, Dalla Lana School of Public Health, University of Toronto, Canada.
| |
Collapse
|
40
|
Forsström V, Toivonen L, Homil K, Waris M, Pedersen CET, Bønnelykke K, Jartti T, Peltola V. Association of Asthma Risk Alleles With Acute Respiratory Tract Infections and Wheezing Illnesses in Young Children. J Infect Dis 2023; 228:990-998. [PMID: 36967681 PMCID: PMC10582910 DOI: 10.1093/infdis/jiad075] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/03/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Genome-wide association studies have identified several risk alleles for early childhood asthma, particularly in the 17q21 locus and in the cadherin-related family member 3 (CDHR3) gene. Contribution of these alleles to the risk of acute respiratory tract infections (ARI) in early childhood is unclear. METHODS We analyzed data from the STEPS birth-cohort study of unselected children and the VINKU and VINKU2 studies on children with severe wheezing illness. Genome-wide genotyping was performed on 1011 children. We analyzed the association between 11 preselected asthma risk alleles and the risk of ARIs and wheezing illnesses of various viral etiologies. RESULTS The asthma risk alleles in CDHR3, GSDMA, and GSDMB were associated with an increased rate of ARIs (for CDHR3, incidence rate ratio [IRR], 1.06; 95% confidence interval [CI], 1.01-1.12; P = .02), and risk allele in CDHR3 gene with rhinovirus infections (IRR, 1.10; 95% CI, 1.01-1.20, P = .03). Asthma risk alleles in GSDMA, GSDMB, IKZF3, ZPBP2, and ORMDL3 genes were associated with wheezing illnesses in early childhood, especially rhinovirus-positive wheezing illnesses. CONCLUSIONS Asthma risk alleles were associated with an increased rate of ARIs and an increased risk of viral wheezing illnesses. Nonwheezing and wheezing ARIs and asthma may have shared genetic risk factors. Clinical Trials Registration. NCT00494624 and NCT00731575.
Collapse
Affiliation(s)
- Ville Forsström
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Laura Toivonen
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Kiara Homil
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Matti Waris
- Virology Unit, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Casper-Emil T Pedersen
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Tuomas Jartti
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
- Research Unit for Pediatrics, Dermatology, Clinical Genetics, Obstetrics and Gynecology, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, University of Oulu, Oulu, Finland
| | - Ville Peltola
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
41
|
Volpe S, Irish J, Palumbo S, Lee E, Herbert J, Ramadan I, Chang EH. Viral infections and chronic rhinosinusitis. J Allergy Clin Immunol 2023; 152:819-826. [PMID: 37574080 PMCID: PMC10592176 DOI: 10.1016/j.jaci.2023.07.018] [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: 05/26/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023]
Abstract
Viral infections are the most common cause of upper respiratory infections; they frequently infect adults once or twice and children 6 to 8 times annually. In most cases, these infections are self-limiting and resolve. However, many patients with chronic rhinosinusitis (CRS) relay that their initiating event began with an upper respiratory infection that progressed in both symptom severity and duration. Viruses bind to sinonasal epithelia through specific receptors, thereby entering cells and replicating within them. Viral infections stimulate interferon-mediated innate immune responses. Recent studies suggest that viral infections may also induce type 2 immune responses and stimulate the aberrant production of cytokines that can result in loss of barrier function, which is a hallmark in CRS. The main purpose of this review will be to highlight common viruses and their associated binding receptors and highlight pathophysiologic mechanisms associated with alterations in mucociliary clearance, epithelial barrier function, and dysfunctional immune responses that might lead to a further understanding of the pathogenesis of CRS.
Collapse
Affiliation(s)
- Sophia Volpe
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arizona, Tucson, Ariz
| | - Joseph Irish
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arizona, Tucson, Ariz
| | - Sunny Palumbo
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arizona, Tucson, Ariz
| | - Eric Lee
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arizona, Tucson, Ariz
| | - Jacob Herbert
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arizona, Tucson, Ariz
| | - Ibrahim Ramadan
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arizona, Tucson, Ariz
| | - Eugene H Chang
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arizona, Tucson, Ariz.
| |
Collapse
|
42
|
Spector C, De Sanctis CM, Panettieri RA, Koziol-White CJ. Rhinovirus induces airway remodeling: what are the physiological consequences? Respir Res 2023; 24:238. [PMID: 37773065 PMCID: PMC10540383 DOI: 10.1186/s12931-023-02529-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Rhinovirus infections commonly evoke asthma exacerbations in children and adults. Recurrent asthma exacerbations are associated with injury-repair responses in the airways that collectively contribute to airway remodeling. The physiological consequences of airway remodeling can manifest as irreversible airway obstruction and diminished responsiveness to bronchodilators. Structural cells of the airway, including epithelial cells, smooth muscle, fibroblasts, myofibroblasts, and adjacent lung vascular endothelial cells represent an understudied and emerging source of cellular and extracellular soluble mediators and matrix components that contribute to airway remodeling in a rhinovirus-evoked inflammatory environment. MAIN BODY While mechanistic pathways associated with rhinovirus-induced airway remodeling are still not fully characterized, infected airway epithelial cells robustly produce type 2 cytokines and chemokines, as well as pro-angiogenic and fibroblast activating factors that act in a paracrine manner on neighboring airway cells to stimulate remodeling responses. Morphological transformation of structural cells in response to rhinovirus promotes remodeling phenotypes including induction of mucus hypersecretion, epithelial-to-mesenchymal transition, and fibroblast-to-myofibroblast transdifferentiation. Rhinovirus exposure elicits airway hyperresponsiveness contributing to irreversible airway obstruction. This obstruction can occur as a consequence of sub-epithelial thickening mediated by smooth muscle migration and myofibroblast activity, or through independent mechanisms mediated by modulation of the β2 agonist receptor activation and its responsiveness to bronchodilators. Differential cellular responses emerge in response to rhinovirus infection that predispose asthmatic individuals to persistent signatures of airway remodeling, including exaggerated type 2 inflammation, enhanced extracellular matrix deposition, and robust production of pro-angiogenic mediators. CONCLUSIONS Few therapies address symptoms of rhinovirus-induced airway remodeling, though understanding the contribution of structural cells to these processes may elucidate future translational targets to alleviate symptoms of rhinovirus-induced exacerbations.
Collapse
Affiliation(s)
- Cassandra Spector
- Rutgers Institute for Translation Medicine and Science, New Brunswick, NJ, USA
| | - Camden M De Sanctis
- Rutgers Institute for Translation Medicine and Science, New Brunswick, NJ, USA
| | | | | |
Collapse
|
43
|
Song YP, Tang MF, Leung ASY, Tao KP, Chan OM, Wong GWK, Chan PKS, Chan RWY, Leung TF. Interactive effects between CDHR3 genotype and rhinovirus species for diagnosis and severity of respiratory tract infections in hospitalized children. Microbiol Spectr 2023; 11:e0118123. [PMID: 37750685 PMCID: PMC10581227 DOI: 10.1128/spectrum.01181-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/08/2023] [Indexed: 09/27/2023] Open
Abstract
Rhinovirus (RV) is the leading pathogen causing childhood wheezing, with rhinovirus C (RV-C) species reported to cause asthma exacerbation. Allele A of single-nucleotide polymorphism (SNP) CDHR3_rs6967330 upregulates epithelial expression of RV-C receptors which results in more severe asthma exacerbations in children. Nevertheless, there are limited data on interactions between CDHR3 variants and their impact on severity of RV-related pediatric respiratory tract infections (RTIs). Medical records of RV-related RTIs in children aged below 18 years who were hospitalized in two public hospitals in 2015-2016 were independently reviewed by two paediatricians. Archived nasopharyngeal aspirates were retrieved for RV detection and sequencing as well as CDHR3 genotyping. HaploView v.5.0 and generalized multifactor dimensionality reduction (GMDR) analysis were employed for haplotypic assignment and gene-environment interaction analyses. Among 1019 studied cases, our results confirmed the relationship between RV-C species and more severe RTIs. Besides the top risk variant rs6967330-A, we identified rs140154310-T to be associated with RV-C susceptibility under the additive model [odds ratio (OR) 2.53, 95% CI 1.15-5.56; P = 0.021]. Rs140154310 was associated with wheezing illness (OR 2.38, 95% CI 1.12-5.04; P = 0.024), with such association being stronger in subjects who wheezed due to RV-C infections (OR 2.71, 95% CI 1.32-5.58; P = 0.007). Haplotype GAG constructed from rs4730125, rs6967330, and rs73195665 was associated with increased risk of RV-C infection (OR 1.71, 95% CI 1.11-2.65; P = 0.016) and oxygen supplementation (OR 1.93, 95% CI 1.13-3.30; P = 0.016). GMDR analyses revealed epistatic interaction between rs140154310 and rs6967330 of CDHR3 for RV-C infection (P = 0.001), RV-C-associated lower RTI (P = 0.004), and RV-C-associated wheeze (P = 0.007). There was synergistic gene-environmental interaction between rs3887998 and RV-C for more severe clinical outcomes (P < 0.001). To conclude, rs140154310-T is another risk variant for RV-C susceptibility and more severe RTIs. Synergistic epistatic interaction is found between CDHR3 SNPs and RV-C for RTI severity, which is likely mediated by susceptibility to RV-C. Haplotypic analysis and GMDR should be included in identifying prediction models of CDHR3 for childhood asthma and RTIs. IMPORTANCE This case-control study investigated the interaction between CDHR3 genotypes and rhinovirus (RV) species on disease severity in Hong Kong children hospitalized for respiratory tract infection (RTI). There were synergistic effects between RV-C and CDHR3 SNPs for RTI severity, which was mainly driven by RV-C. Specifically, rs6967330 and rs140154310 alone and their epistatic interaction were associated with RV-C-related and severe RTIs in our subjects. Therefore, genotyping of CDHR3 SNPs may help physicians formulate prediction models for severity of RV-associated RTIs.
Collapse
Affiliation(s)
- Yu P. Song
- Department of Paediatrics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Man F. Tang
- Department of Paediatrics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong, China
| | - Agnes S. Y. Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong, China
| | - Kin P. Tao
- Department of Paediatrics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong, China
- The Chinese University of Hong Kong-University Medical Center Utrecht Joint Research Laboratory of Respiratory Virus and Immunobiology, The Chinese University of Hong Kong, Hong Kong, China
| | - Oi M. Chan
- Department of Paediatrics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Gary W. K. Wong
- Department of Paediatrics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Paul K. S. Chan
- Department of Microbiology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Renee W. Y. Chan
- Department of Paediatrics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong, China
- The Chinese University of Hong Kong-University Medical Center Utrecht Joint Research Laboratory of Respiratory Virus and Immunobiology, The Chinese University of Hong Kong, Hong Kong, China
| | - Ting F. Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong, China
- The Chinese University of Hong Kong-University Medical Center Utrecht Joint Research Laboratory of Respiratory Virus and Immunobiology, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
44
|
Bakakos A, Sotiropoulou Z, Vontetsianos A, Zaneli S, Papaioannou AI, Bakakos P. Epidemiology and Immunopathogenesis of Virus Associated Asthma Exacerbations. J Asthma Allergy 2023; 16:1025-1040. [PMID: 37791040 PMCID: PMC10543746 DOI: 10.2147/jaa.s277455] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/16/2023] [Indexed: 10/05/2023] Open
Abstract
Asthma is a common airway disease, affecting millions of people worldwide. Although most asthma patients experience mild symptoms, it is characterized by variable airflow limitation, which can occasionally become life threatening in the case of a severe exacerbation. The commonest triggers of asthma exacerbations in both children and adults are viral infections. In this review article, we will try to investigate the most common viruses triggering asthma exacerbations and their role in asthma immunopathogenesis, since viral infections in young adults are thought to trigger the development of asthma either right away after the infection or at a later stage of their life. The commonest viral pathogens associated with asthma include the respiratory syncytial virus, rhinoviruses, influenza and parainfluenza virus, metapneumovirus and coronaviruses. All these viruses exploit different molecular pathways to infiltrate the host. Asthmatics are more prone to severe viral infections due to their unique inflammatory response, which is mostly characterized by T2 cytokines. Unlike the normal T1 high response to viral infection, asthmatics with T2 high inflammation are less potent in containing a viral infection. Inhaled and/or systematic corticosteroids and bronchodilators remain the cornerstone of asthma exacerbation treatment, and although many targeted therapies which block molecules that viruses use to infect the host have been used in a laboratory level, none has been yet approved for clinical use. Nevertheless, further understanding of the unique pathway that each virus follows to infect an individual may be crucial in the development of targeted therapies for the commonest viral pathogens to effectively prevent asthma exacerbations. Finally, biologic therapies resulted in a complete change of scenery in the treatment of severe asthma, especially with a T2 high phenotype. All available data suggest that monoclonal antibodies are safe and able to drastically reduce the rate of viral asthma exacerbations.
Collapse
Affiliation(s)
- Agamemnon Bakakos
- 1st University Department of Respiratory Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - Zoi Sotiropoulou
- 1st University Department of Respiratory Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - Angelos Vontetsianos
- 1st University Department of Respiratory Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - Stavroula Zaneli
- 1st University Department of Respiratory Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - Andriana I Papaioannou
- 1st University Department of Respiratory Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - Petros Bakakos
- 1st University Department of Respiratory Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece
| |
Collapse
|
45
|
Gebski EB, Parikh V, Lam H, Kim N, Bochkov YA, Cao G, Panettieri RA, Kurten R, Gern J, An SS, Koziol-White CJ. Rhinovirus C15 Attenuates Relaxation and cAMP Production in Human Airways and Smooth Muscle. Am J Respir Cell Mol Biol 2023; 69:172-181. [PMID: 37098126 PMCID: PMC10399146 DOI: 10.1165/rcmb.2021-0526oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 04/25/2023] [Indexed: 04/27/2023] Open
Abstract
Rhinoviruses (RVs) evoke as many as 85% of acute asthma exacerbations in children and 50% in adults and can induce airway hyperresponsiveness and decrease efficacy of current therapeutics to provide symptom relief. Using human precision-cut lung slices (hPCLSs), primary human air-liquid interface-differentiated airway epithelial cells (HAECs), and human airway smooth muscle (HASM) as preclinical experimental models, we demonstrated that RV-C15 attenuates agonist-induced bronchodilation. Specifically, airway relaxation to formoterol and cholera toxin, but not forskolin (Fsk), was attenuated following hPCLS exposure to RV-C15. In isolated HASM cells, exposure to conditioned media from RV-exposed HAECs decreased cellular relaxation in response to isoproterenol and prostaglandin E2, but not Fsk. Additionally, cAMP generation elicited by formoterol and isoproterenol, but not Fsk, was attenuated following HASM exposure to RV-C15-conditioned HAEC media. HASM exposure to RV-C15-conditioned HAEC media modulated expression of components of relaxation pathways, specifically GNAI1 and GRK2. Strikingly, similar to exposure to intact RV-C15, hPCLS exposed to UV-inactivated RV-C15 showed markedly attenuated airway relaxation in response to formoterol, suggesting that the mechanism(s) of RV-C15-mediated loss of bronchodilation is independent of virus replication pathways. Further studies are warranted to identify soluble factor(s) regulating the epithelial-driven smooth muscle loss of β2-adrenergic receptor function.
Collapse
Affiliation(s)
- Eric B. Gebski
- Rutgers Institute for Translational Medicine and Science, New Brunswick, New Jersey
| | - Vishal Parikh
- Rutgers Institute for Translational Medicine and Science, New Brunswick, New Jersey
| | - Hong Lam
- Rutgers Institute for Translational Medicine and Science, New Brunswick, New Jersey
| | - Nicholas Kim
- Rutgers Institute for Translational Medicine and Science, New Brunswick, New Jersey
| | - Yury A. Bochkov
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine and Science, New Brunswick, New Jersey
| | | | - Richard Kurten
- Division of Pediatric Allergy & Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Lung Cell Biology Laboratory, Arkansas Children’s Research Institute, Rogers, Arkansas; and
| | - James Gern
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Steven S. An
- Rutgers Institute for Translational Medicine and Science, New Brunswick, New Jersey
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, The State University of New Jersey, Piscataway, New Jersey
| | - Cynthia J. Koziol-White
- Rutgers Institute for Translational Medicine and Science, New Brunswick, New Jersey
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, The State University of New Jersey, Piscataway, New Jersey
| |
Collapse
|
46
|
Bochkov YA, Devries M, Tetreault K, Gangnon R, Lee S, Bacharier LB, Busse WW, Camargo CA, Choi T, Cohen R, De R, DeMuri GP, Fitzpatrick AM, Gergen PJ, Grindle K, Gruchalla R, Hartert T, Hasegawa K, Khurana Hershey GK, Holt P, Homil K, Jartti T, Kattan M, Kercsmar C, Kim H, Laing IA, Le Souëf PN, Liu AH, Mauger DT, Pappas T, Patel SJ, Phipatanakul W, Pongracic J, Seroogy C, Sly PD, Tisler C, Wald ER, Wood R, Lemanske RF, Jackson DJ, Gern JE. Rhinoviruses A and C elicit long-lasting antibody responses with limited cross-neutralization. J Med Virol 2023; 95:e29058. [PMID: 37638498 PMCID: PMC10484091 DOI: 10.1002/jmv.29058] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/29/2023]
Abstract
Rhinoviruses (RVs) can cause severe wheezing illnesses in young children and patients with asthma. Vaccine development has been hampered by the multitude of RV types with little information about cross-neutralization. We previously showed that neutralizing antibody (nAb) responses to RV-C are detected twofold to threefold more often than those to RV-A throughout childhood. Based on those findings, we hypothesized that RV-C infections are more likely to induce either cross-neutralizing or longer-lasting antibody responses compared with RV-A infections. We pooled RV diagnostic data from multiple studies of children with respiratory illnesses and compared the expected versus observed frequencies of sequential infections with RV-A or RV-C types using log-linear regression models. We tested longitudinally collected plasma samples from children to compare the duration of RV-A versus RV-C nAb responses. Our models identified limited reciprocal cross-neutralizing relationships for RV-A (A12-A75, A12-A78, A20-A78, and A75-A78) and only one for RV-C (C2-C40). Serologic analysis using reference mouse sera and banked human plasma samples confirmed that C40 infections induced nAb responses with modest heterotypic activity against RV-C2. Mixed-effects regression modeling of longitudinal human plasma samples collected from ages 2 to 18 years demonstrated that RV-A and RV-C illnesses induced nAb responses of similar duration. These results indicate that both RV-A and RV-C nAb responses have only modest cross-reactivity that is limited to genetically similar types. Contrary to our initial hypothesis, RV-C species may include even fewer cross-neutralizing types than RV-A, whereas the duration of nAb responses during childhood is similar between the two species. The modest heterotypic responses suggest that RV vaccines must have a broad representation of prevalent types.
Collapse
Affiliation(s)
| | - Mark Devries
- University of Wisconsin-Madison, Madison, WI, United States
| | | | - Ronald Gangnon
- University of Wisconsin-Madison, Madison, WI, United States
| | - Sujin Lee
- Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, GA, United States
| | | | | | - Carlos A. Camargo
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Timothy Choi
- University of Wisconsin-Madison, Madison, WI, United States
| | - Robyn Cohen
- Boston University, Boston, MA, United States
| | - Ramyani De
- Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, GA, United States
| | | | - Anne M. Fitzpatrick
- Department of Pediatrics, Emory University and Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Peter J. Gergen
- National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, United States
| | | | | | - Tina Hartert
- Vanderbilt University, Nashville, TN, United States
| | - Kohei Hasegawa
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | | | - Patrick Holt
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Kiara Homil
- University of Turku and Turku University Hospital, Turku, Finland
| | - Tuomas Jartti
- University of Turku and Turku University Hospital, Turku, Finland
- PEDEGO Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Meyer Kattan
- Columbia University, New York, NY, United States
| | | | - Haejin Kim
- Henry Ford Health Systems, Detroit, MI, United States
| | | | | | - Andrew H. Liu
- Children’s Hospital Colorado, University of Colorado, Aurora, CO, United States
| | | | - Tressa Pappas
- University of Wisconsin-Madison, Madison, WI, United States
| | | | | | | | | | - Peter D. Sly
- Child Health Research Centre, The University of Queensland, South Brisbane, Australia
| | | | - Ellen R. Wald
- University of Wisconsin-Madison, Madison, WI, United States
| | - Robert Wood
- Johns Hopkins University, Baltimore, MD, United States
| | | | | | - James E. Gern
- University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
47
|
Basnet S, Mohanty C, Bochkov YA, Brockman-Schneider RA, Kendziorski C, Gern JE. Rhinovirus C causes heterogeneous infection and gene expression in airway epithelial cell subsets. Mucosal Immunol 2023; 16:386-398. [PMID: 36796588 PMCID: PMC10629931 DOI: 10.1016/j.mucimm.2023.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 01/27/2023] [Indexed: 02/16/2023]
Abstract
Rhinoviruses infect ciliated airway epithelial cells, and rhinoviruses' nonstructural proteins quickly inhibit and divert cellular processes for viral replication. However, the epithelium can mount a robust innate antiviral immune response. Therefore, we hypothesized that uninfected cells contribute significantly to the antiviral immune response in the airway epithelium. Using single-cell RNA sequencing, we demonstrate that both infected and uninfected cells upregulate antiviral genes (e.g. MX1, IFIT2, IFIH1, and OAS3) with nearly identical kinetics, whereas uninfected non-ciliated cells are the primary source of proinflammatory chemokines. Furthermore, we identified a subset of highly infectable ciliated epithelial cells with minimal interferon responses and determined that interferon responses originate from distinct subsets of ciliated cells with moderate viral replication. These findings suggest that the composition of ciliated airway epithelial cells and coordinated responses of infected and uninfected cells could determine the risk of more severe viral respiratory illnesses in children with asthma, chronic obstructive pulmonary disease, and genetically susceptible individuals.
Collapse
Affiliation(s)
- Sarmila Basnet
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA.
| | - Chitrasen Mohanty
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA
| | - Yury A Bochkov
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | | | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA
| | - James E Gern
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
48
|
Li Y, Hershenson MB. Remember the Airway Smooth Muscle! How Rhinovirus Impairs Bronchodilator Responses. Am J Respir Cell Mol Biol 2023; 69:121-122. [PMID: 37163760 PMCID: PMC10399143 DOI: 10.1165/rcmb.2023-0146ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Affiliation(s)
- Yiran Li
- Departments of Pediatrics and Molecular and Integrative Physiology University of Michigan Medical School Ann Arbor, Michigan
| | - Marc B Hershenson
- Departments of Pediatrics and Molecular and Integrative Physiology University of Michigan Medical School Ann Arbor, Michigan
| |
Collapse
|
49
|
Boboltz A, Kumar S, Duncan GA. Inhaled drug delivery for the targeted treatment of asthma. Adv Drug Deliv Rev 2023; 198:114858. [PMID: 37178928 PMCID: PMC10330872 DOI: 10.1016/j.addr.2023.114858] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/14/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023]
Abstract
Asthma is a chronic lung disease affecting millions worldwide. While classically acknowledged to result from allergen-driven type 2 inflammatory responses leading to IgE and cytokine production and the influx of immune cells such as mast cells and eosinophils, the wide range in asthmatic pathobiological subtypes lead to highly variable responses to anti-inflammatory therapies. Thus, there is a need to develop patient-specific therapies capable of addressing the full spectrum of asthmatic lung disease. Moreover, delivery of targeted treatments for asthma directly to the lung may help to maximize therapeutic benefit, but challenges remain in design of effective formulations for the inhaled route. In this review, we discuss the current understanding of asthmatic disease progression as well as genetic and epigenetic disease modifiers associated with asthma severity and exacerbation of disease. We also overview the limitations of clinically available treatments for asthma and discuss pre-clinical models of asthma used to evaluate new therapies. Based on the shortcomings of existing treatments, we highlight recent advances and new approaches to treat asthma via inhalation for monoclonal antibody delivery, mucolytic therapy to target airway mucus hypersecretion and gene therapies to address underlying drivers of disease. Finally, we conclude with discussion on the prospects for an inhaled vaccine to prevent asthma.
Collapse
Affiliation(s)
- Allison Boboltz
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, United States
| | - Sahana Kumar
- Biological Sciences Graduate Program, University of Maryland, College Park, MD 20742, United States
| | - Gregg A Duncan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, United States; Biological Sciences Graduate Program, University of Maryland, College Park, MD 20742, United States.
| |
Collapse
|
50
|
Urbani F, Cometa M, Martelli C, Santoli F, Rana R, Ursitti A, Bonato M, Baraldo S, Contoli M, Papi A. Update on virus-induced asthma exacerbations. Expert Rev Clin Immunol 2023; 19:1259-1272. [PMID: 37470413 DOI: 10.1080/1744666x.2023.2239504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/01/2023] [Accepted: 07/18/2023] [Indexed: 07/21/2023]
Abstract
INTRODUCTION Viral infections are common triggers for asthma exacerbation. Subjects with asthma are more susceptible to viral infections and develop more severe or long-lasting lower respiratory tract symptoms than healthy individuals owing to impaired immune responses. Of the many viruses associated with asthma exacerbation, rhinovirus (RV) is the most frequently identified virus in both adults and children. AREAS COVERED We reviewed epidemiological and clinical links and mechanistic studies on virus-associated asthma exacerbations. We included sections on severe acute respiratory syndrome coronavirus 2 (SARS-CoV2), the latest evidence of coronavirus disease 2019 (COVID-19) in asthma patients, and past and future searches for therapeutic and prevention targets. EXPERT OPINION Early treatment or prevention of viral infections might significantly reduce the rate of asthma exacerbation, which is one of the key points of disease management. Although it is hypothetically possible nowadays to interfere with every step of the infectious cycle of respiratory tract viruses, vaccination development has provided some of the most encouraging results. Future research should proceed toward the development of a wider spectrum of vaccines to achieve a better quality of life for patients with asthma and to reduce the economic burden on the healthcare system.
Collapse
Affiliation(s)
- Francesca Urbani
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Marianna Cometa
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Chiara Martelli
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Federica Santoli
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Roberto Rana
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Antonio Ursitti
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Matteo Bonato
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Simonetta Baraldo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Marco Contoli
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Alberto Papi
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| |
Collapse
|