1
|
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 DOI: 10.1038/s41467-024-55076-2] [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: 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
|
2
|
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
|
3
|
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
|
4
|
Dy ABC, Girkin J, Marrocco A, Collison A, Mwase C, O'Sullivan MJ, Phung TKN, Mattes J, Koziol-White C, Gern JE, Bochkov YA, Bartlett NW, Park JA. Rhinovirus infection induces secretion of endothelin-1 from airway epithelial cells in both in vitro and in vivo models. Respir Res 2023; 24:205. [PMID: 37598152 PMCID: PMC10440034 DOI: 10.1186/s12931-023-02510-6] [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: 05/28/2023] [Accepted: 08/11/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Rhinovirus (RV) infection of airway epithelial cells triggers asthma exacerbations, during which airway smooth muscle (ASM) excessively contracts. Due to ASM contraction, airway epithelial cells become mechanically compressed. We previously reported that compressed human bronchial epithelial (HBE) cells are a source of endothelin-1 (ET-1) that causes ASM contraction. Here, we hypothesized that epithelial sensing of RV by TLR3 and epithelial compression induce ET-1 secretion through a TGF-β receptor (TGFβR)-dependent mechanism. METHODS To test this, we used primary HBE cells well-differentiated in air-liquid interface culture and two mouse models (ovalbumin and house dust mite) of allergic airway disease (AAD). HBE cells were infected with RV-A16, treated with a TLR3 agonist (poly(I:C)), or exposed to compression. Thereafter, EDN1 (ET-1 protein-encoding gene) mRNA expression and secreted ET-1 protein were measured. We examined the role of TGFβR in ET-1 secretion using either a pharmacologic inhibitor of TGFβR or recombinant TGF-β1 protein. In the AAD mouse models, allergen-sensitized and allergen-challenged mice were subsequently infected with RV. We then measured ET-1 in bronchoalveolar lavage fluid (BALF) and airway hyperresponsiveness (AHR) following methacholine challenge. RESULTS Our data reveal that RV infection induced EDN1 expression and ET-1 secretion in HBE cells, potentially mediated by TLR3. TGFβR activation was partially required for ET-1 secretion, which was induced by RV, poly(I:C), or compression. TGFβR activation alone was sufficient to increase ET-1 secretion. In AAD mouse models, RV induced ET-1 secretion in BALF, which positively correlated with AHR. CONCLUSIONS Our data provide evidence that RV infection increased epithelial-cell ET-1 secretion through a TGFβR-dependent mechanism, which contributes to bronchoconstriction during RV-induced asthma exacerbations.
Collapse
Affiliation(s)
- Alane Blythe C Dy
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Jason Girkin
- College of Health, Medicine and Wellbeing, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Antonella Marrocco
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Adam Collison
- College of Health, Medicine and Wellbeing, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Chimwemwe Mwase
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Michael J O'Sullivan
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Thien-Khoi N Phung
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Joerg Mattes
- College of Health, Medicine and Wellbeing, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Australia
| | | | - James E Gern
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Yury A Bochkov
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Nathan W Bartlett
- College of Health, Medicine and Wellbeing, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Jin-Ah Park
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA.
| |
Collapse
|
5
|
Swartz AR, Shieh Y, Gulasarian A, Curtis E, Hofmann CF, Baker JB, Templeton N, Olson JW. Glutathione affinity chromatography for the scalable purification of an oncolytic virus immunotherapy from microcarrier cell culture. Front Bioeng Biotechnol 2023; 11:1193454. [PMID: 37397964 PMCID: PMC10310922 DOI: 10.3389/fbioe.2023.1193454] [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: 03/24/2023] [Accepted: 05/31/2023] [Indexed: 07/04/2023] Open
Abstract
Therapeutic viral vectors are an emerging technology with several clinical applications in gene therapy, vaccines, and immunotherapy. Increased demand has required the redevelopment of conventional, low-throughput cell culture and purification manufacturing methods such as static cell stacks and ultracentrifugation. In this work, scalable methods were investigated for the manufacture of an oncolytic virus immunotherapy application consisting of a prototype strain of coxsackievirus A21 (CVA21) produced in adherent MRC-5 cells. Cell culture was established in stirred-tank microcarrier bioreactors, and an efficient affinity chromatography method was developed for the purification of harvested CVA21 through binding of the viral capsids to an immobilized glutathione (GSH) ligand. Bioreactor temperature during infection was investigated to maximize titer, and a decrease in temperature from 37°C to 34°C yielded a two-three-fold increase in infectivity. After purification of the 34°C harvests, the GSH affinity chromatography elution not only maintained a >two-fold increase in infectivity and viral genomes but also increased the proportion of empty capsids compared to 37°C harvests. Using material generated from both infection temperature setpoints, chromatographic parameters and mobile phase compositions were studied at the laboratory scale to maximize infectious particle yields and cell culture impurity clearance. Empty capsids that co-eluted with full capsids from 34°C infection temperature harvests were poorly resolved across the conditions tested, but subsequent polishing anion exchange and cation exchange chromatography steps were developed to clear residual empty capsids and other impurities. Oncolytic CVA21 production was scaled-up 75-fold from the laboratory scale and demonstrated across seven batches in 250 L single-use microcarrier bioreactors and purified with customized, prepacked, single-use 1.5 L GSH affinity chromatography columns. The large-scale bioreactors controlled at 34°C during infection maintained a three-fold increase in productivity in the GSH elution, and excellent clearance of host cell and media impurities was observed across all batches. This study presents a robust method for the manufacture of an oncolytic virus immunotherapy application that may be implemented for the scalable production of other viruses and viral vectors which interact with glutathione.
Collapse
Affiliation(s)
- Andrew R. Swartz
- Process Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| | - Yvonne Shieh
- Process Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| | - Amanda Gulasarian
- Process Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| | - Erik Curtis
- Process Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| | - Carl F. Hofmann
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| | - Jack B. Baker
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| | - Neil Templeton
- Process Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| | - Jessica W. Olson
- Process Research and Development, Merck & Co., Inc., Rahway, NJ, United States
| |
Collapse
|
6
|
Yuan XH, Pang LL, Yang J, Jin Y. Comparison of immune response to human rhinovirus C and respiratory syncytial virus in highly differentiated human airway epithelial cells. Virol J 2022; 19:81. [PMID: 35570279 PMCID: PMC9107719 DOI: 10.1186/s12985-022-01805-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 04/27/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Human rhinovirus C (HRV-C) accounts for a large proportion of HRV-related illnesses, but the immune response to HRV-C infection has not been elucidated. Our objective was to assess the effect of HRV-C on cytokine secretion in human bronchial epithelial (HBE) cells grown at air–liquid interface (ALI) and compare it with that of respiratory syncytial virus (RSV).
Methods
HBE cells were differentiated at ALI culture and the full-length cDNA clones of HRV-C651 and HRV-C15, clinical isolates of HRV-C79 and HRV-C101, and two RSV isolates were inoculated in the HBE cells. The effect of HRV-C on cytokine secretion was assessed and compared with that of RSV.
Results
HRV-Cs infect and propagate in fully differentiated HBE cells and significantly increase the secretion of IFN-λ1, CCL5, IP10, IL-6, IL-8, and MCP-1. The virus loads positively correlated with the levels of the cytokines. HRV-C induced lower secretion of CCL5 (P = 0.048), IL-6 (P = 0.016), MCP-1 (P = 0.008), and IL-8 (P = 0.032), and similar secretion of IP10 (P = 0.214) and IFN-λ1 (P = 0.214) when compared with RSV.
Conclusion
HBE ALI culture system supported HRV-C infection and propagation and HRV-C induced relatively weaker cytokine expression than RSV.
Collapse
|
7
|
Bhat T, Cao A, Yin J. Virus-like Particles: Measures and Biological Functions. Viruses 2022; 14:383. [PMID: 35215979 PMCID: PMC8877645 DOI: 10.3390/v14020383] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/10/2022] [Accepted: 02/10/2022] [Indexed: 12/25/2022] Open
Abstract
Virus-like particles resemble infectious virus particles in size, shape, and molecular composition; however, they fail to productively infect host cells. Historically, the presence of virus-like particles has been inferred from total particle counts by microscopy, and infectious particle counts or plaque-forming-units (PFUs) by plaque assay; the resulting ratio of particles-to-PFUs is often greater than one, easily 10 or 100, indicating that most particles are non-infectious. Despite their inability to hijack cells for their reproduction, virus-like particles and the defective genomes they carry can exhibit a broad range of behaviors: interference with normal virus growth during co-infections, cell killing, and activation or inhibition of innate immune signaling. In addition, some virus-like particles become productive as their multiplicities of infection increase, a sign of cooperation between particles. Here, we review established and emerging methods to count virus-like particles and characterize their biological functions. We take a critical look at evidence for defective interfering virus genomes in natural and clinical isolates, and we review their potential as antiviral therapeutics. In short, we highlight an urgent need to better understand how virus-like genomes and particles interact with intact functional viruses during co-infection of their hosts, and their impacts on the transmission, severity, and persistence of virus-associated diseases.
Collapse
Affiliation(s)
| | | | - John Yin
- Department of Chemical and Biological Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, 330 N. Orchard Street, Madison, WI 53715, USA; (T.B.); (A.C.)
| |
Collapse
|
8
|
Assadi I, Guesmi A, Baaloudj O, Zeghioud H, Elfalleh W, Benhammadi N, Khezami L, Assadi AA. Review on inactivation of airborne viruses using non-thermal plasma technologies: from MS2 to coronavirus. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:4880-4892. [PMID: 34796437 PMCID: PMC8601095 DOI: 10.1007/s11356-021-17486-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/08/2021] [Indexed: 04/12/2023]
Abstract
Although several non-thermal plasmas (NTPs) technologies have been widely investigated in air treatment, very few studies have focused on the inactivation mechanism of viruses by NTPs. Due to its efficiency and environmental compatibility, non-thermal plasma could be considered a promising virus-inactivation technology. Plasma is a partly or fully ionized gas including some species (i.e., electrons, free radicals, ions, and neutral molecules) to oxidize pollutants or inactivate harmful organisms. Non-thermal plasmas are made using less energy and have an active electron at a much higher temperature than bulk gas molecules. This review describes NTPs for virus inactivation in indoor air. The different application processes of plasma for microorganism inactivation at both laboratory and pilot-scale was also reviewed This paper reports on recent advances in this exciting area of viral inactivation identifying applications and mechanisms of inactivation, and summarizing the results of the latest experiments in the literature. Moreover, special attention was paid to the mechanism of virus inactivation. Finally, the paper suggests research directions in the field of airborne virus inactivation using non-thermal plasma.
Collapse
Affiliation(s)
- Imen Assadi
- Laboratoire Energie, Eau, Environnement Et Procèdes, ENIG, Université de Gabès, LR18ES356072, Gabès, Tunisia
| | - Ahlem Guesmi
- Department of Chemistry, Imam Mohammad Ibn Saud Islamic University (IMSIU), P.O. Box 5701, 11432, Riyadh, Saudi Arabia
| | - Oussama Baaloudj
- Laboratory of Reaction Engineering, USTHB, BP 32, 16111, Algiers, Algeria
| | - Hichem Zeghioud
- Department of Process Engineering, Badji Mokhtar University, P.O. Box 12, 23000, Annaba, Algeria
| | - Walid Elfalleh
- Laboratoire Energie, Eau, Environnement Et Procèdes, ENIG, Université de Gabès, LR18ES356072, Gabès, Tunisia
| | - Naoufel Benhammadi
- Department of Chemistry, Imam Mohammad Ibn Saud Islamic University (IMSIU), P.O. Box 5701, 11432, Riyadh, Saudi Arabia
| | - Lotfi Khezami
- Department of Chemistry, Imam Mohammad Ibn Saud Islamic University (IMSIU), P.O. Box 5701, 11432, Riyadh, Saudi Arabia
| | | |
Collapse
|
9
|
Gagliardi TB, Goldstein ME, Song D, Gray KM, Jung JW, Ignacio MA, Stroka KM, Duncan GA, Scull MA. Rhinovirus C replication is associated with the endoplasmic reticulum and triggers cytopathic effects in an in vitro model of human airway epithelium. PLoS Pathog 2022; 18:e1010159. [PMID: 34995322 PMCID: PMC8741012 DOI: 10.1371/journal.ppat.1010159] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 11/29/2021] [Indexed: 12/21/2022] Open
Abstract
The clinical impact of rhinovirus C (RV-C) is well-documented; yet, the viral life cycle remains poorly defined. Thus, we characterized RV-C15 replication at the single-cell level and its impact on the human airway epithelium (HAE) using a physiologically-relevant in vitro model. RV-C15 replication was restricted to ciliated cells where viral RNA levels peaked at 12 hours post-infection (hpi), correlating with elevated titers in the apical compartment at 24hpi. Notably, infection was associated with a loss of polarized expression of the RV-C receptor, cadherin-related family member 3. Visualization of double-stranded RNA (dsRNA) during RV-C15 replication revealed two distinct replication complex arrangements within the cell, likely corresponding to different time points in infection. To further define RV-C15 replication sites, we analyzed the expression and colocalization of giantin, phosphatidylinositol-4-phosphate, and calnexin with dsRNA. Despite observing Golgi fragmentation by immunofluorescence during RV-C15 infection as previously reported for other RVs, a high ratio of calnexin-dsRNA colocalization implicated the endoplasmic reticulum as the primary site for RV-C15 replication in HAE. RV-C15 infection was also associated with elevated stimulator of interferon genes (STING) expression and the induction of incomplete autophagy, a mechanism used by other RVs to facilitate non-lytic release of progeny virions. Notably, genetic depletion of STING in HAE attenuated RV-C15 and -A16 (but not -B14) replication, corroborating a previously proposed proviral role for STING in some RV infections. Finally, RV-C15 infection resulted in a temporary loss in epithelial barrier integrity and the translocation of tight junction proteins while a reduction in mucociliary clearance indicated cytopathic effects on epithelial function. Together, our findings identify both shared and unique features of RV-C replication compared to related rhinoviruses and define the impact of RV-C on both epithelial cell organization and tissue functionality–aspects of infection that may contribute to pathogenesis in vivo. Rhinovirus C has a global distribution and significant clinical impact–especially in those with underlying lung disease. Although RV-C is genetically, structurally, and biologically distinct from RV-A and -B viruses, our understanding of the RV-C life cycle has been largely inferred from these and other related viruses. Here, we performed a detailed analysis of RV-C15 replication in a physiologically-relevant model of human airway epithelium. Our single-cell, microscopy-based approach revealed that–unlike other RVs–the endoplasmic reticulum is the primary site for RV-C15 replication. RV-C15 replication also stimulated STING expression, which was proviral, and triggered dramatic changes in cellular organization, including altered virus receptor distribution, fragmented Golgi stacks, and the induction of incomplete autophagy. Additionally, we observed a loss of epithelial barrier function and a decrease in mucociliary clearance, a major defense mechanism in the lung, during RV-C15 infection. Together, these data reveal novel insight into RV-C15 replication dynamics and resulting cytopathic effects in the primary target cells for infection, thereby furthering our understanding of the pathogenesis of RV-C. Our work highlights similar, as well as unique, aspects of RV-C15 replication compared to related pathogens, which will help guide future studies on the molecular mechanisms of RV-C infection.
Collapse
Affiliation(s)
- Talita B. Gagliardi
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, United States of America
| | - Monty E. Goldstein
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, United States of America
| | - Daniel Song
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States of America
| | - Kelsey M. Gray
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States of America
| | - Jae W. Jung
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States of America
| | - Maxinne A. Ignacio
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, United States of America
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States of America
- Biophysics Program, University of Maryland, College Park, Maryland, United States of America
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, Maryland, United States of America
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland, United States of America
| | - Gregg A. Duncan
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States of America
| | - Margaret A. Scull
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
10
|
Bird G, Braithwaite I, Harper J, Koorevaar I, van den Berg M, Maijers I, Kearns N, Dilcher M, Jennings L, Fingleton J, Shortt N, Weatherall M, Beasley R. Rhinothermy delivered by nasal high flow therapy in the treatment of the common cold: a randomised controlled trial. BMJ Open 2021; 11:e047760. [PMID: 34848508 PMCID: PMC8634207 DOI: 10.1136/bmjopen-2020-047760] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND The common cold is the most common infectious disease affecting humans and has a substantial economic impact on society. Human rhinoviruses, which cause almost two-thirds of colds, have demonstrated temperature-dependent replication which is optimal between 33°C and 35°C. METHODS This randomised, single-blind, parallel-group trial completed at a single-centre in New Zealand, recruited 170 participants aged 18-75 years (mean age 27.5 years) who were within 48 hours of common cold symptom onset and had a symptom score (the Modified Jackson Score (MJS)) ≥7 and a negative point-of-care test for influenza. Participants were blinded to the intervention and randomised (1:1) to 5 days of either nasal high flow rhinothermy (rNHF) (100% humidified air delivered at 35 L/min and 41°C for 2 hours daily) (n=85) or 'sham' rhinothermy (100% humidified air delivered at 10 L/min and 31°C for 10 min daily) (n=85) and completed daily symptom diaries, which included the MJS, for 14 days, to investigate whether rNHF reduced common cold symptom severity and duration compared with 'sham' rhinothermy. RESULTS An intention-to-treat superiority analysis included all randomised participants and showed no difference between treatment groups for the primary outcome, the day 4 MJS analysed by analysis of covariance: mean (SD) 6.33 (3.97) for rNHF vs 5.8 (3.15) for 'sham'; estimated difference (95% CI) 0.37 (-0.69 to 1.42), p=0.49. There was no difference in time until resolution of symptoms: mean (SD) 5.96 (4.47) days for rNHF vs 6.42 (4.09) days for 'sham'; estimated difference (95% CI) 1.02 (0.75 to 1.38), p=0.91. There were no serious adverse events related to the study treatments. CONCLUSIONS This well-powered, single-blind randomised controlled trial does not provide evidence that 5 days of rNHF (100% humidified air heated to 41°C delivered at 35 L/min for 2 hours daily) reduces common cold symptom severity or duration. However, investigation of rNHF in the treatment of influenza is warranted. TRIAL REGISTRATION NUMBER ACTRN12617001340325.
Collapse
Affiliation(s)
- Grace Bird
- Medical Research Institute of New Zealand, Wellington, New Zealand
| | | | - James Harper
- Medical Research Institute of New Zealand, Wellington, New Zealand
| | - Iris Koorevaar
- Medical Research Institute of New Zealand, Wellington, New Zealand
| | | | - Ingrid Maijers
- Medical Research Institute of New Zealand, Wellington, New Zealand
| | - Nethmi Kearns
- Medical Research Institute of New Zealand, Wellington, New Zealand
| | - Meik Dilcher
- Canterbury Health Laboratories, Christchurch, New Zealand
| | - Lance Jennings
- Canterbury Health Laboratories, Christchurch, New Zealand
| | - James Fingleton
- Medical Research Institute of New Zealand, Wellington, New Zealand
| | - Nick Shortt
- Medical Research Institute of New Zealand, Wellington, New Zealand
| | | | - Richard Beasley
- Medical Research Institute of New Zealand, Wellington, New Zealand
| |
Collapse
|
11
|
Baldassi D, Gabold B, Merkel O. Air-liquid interface cultures of the healthy and diseased human respiratory tract: promises, challenges and future directions. ADVANCED NANOBIOMED RESEARCH 2021; 1:2000111. [PMID: 34345878 PMCID: PMC7611446 DOI: 10.1002/anbr.202000111] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Air-liquid interface (ALI) culture models currently represent a valid instrument to recreate the typical aspects of the respiratory tract in vitro in both healthy and diseased state. They can help reducing the number of animal experiments, therefore, supporting the 3R principle. This review discusses ALI cultures and co-cultures derived from immortalized as well as primary cells, which are used to study the most common disorders of the respiratory tract, in terms of both pathophysiology and drug screening. The article displays ALI models used to simulate inflammatory lung diseases such as chronic obstructive pulmonary disease (COPD), asthma, cystic fibrosis, lung cancer, and viral infections. It also includes a focus on ALI cultures described in literature studying respiratory viruses such as SARS-CoV-2 causing the global Covid-19 pandemic at the time of writing this review. Additionally, commercially available models of ALI cultures are presented. Ultimately, the aim of this review is to provide a detailed overview of ALI models currently available and to critically discuss them in the context of the most prevalent diseases of the respiratory tract.
Collapse
Affiliation(s)
- Domizia Baldassi
- Pharmaceutical Technology and Biopharmacy, LMU Munich Butenandtstr. 5-13, 81377 Munich, Germany
| | - Bettina Gabold
- Pharmaceutical Technology and Biopharmacy, LMU Munich Butenandtstr. 5-13, 81377 Munich, Germany
| | - Olivia Merkel
- Pharmaceutical Technology and Biopharmacy, LMU Munich Butenandtstr. 5-13, 81377 Munich, Germany
| |
Collapse
|
12
|
Iverson E, Kaler L, Agostino EL, Song D, Duncan GA, Scull MA. Leveraging 3D Model Systems to Understand Viral Interactions with the Respiratory Mucosa. Viruses 2020; 12:E1425. [PMID: 33322395 PMCID: PMC7763686 DOI: 10.3390/v12121425] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Respiratory viruses remain a significant cause of morbidity and mortality in the human population, underscoring the importance of ongoing basic research into virus-host interactions. However, many critical aspects of infection are difficult, if not impossible, to probe using standard cell lines, 2D culture formats, or even animal models. In vitro systems such as airway epithelial cultures at air-liquid interface, organoids, or 'on-chip' technologies allow interrogation in human cells and recapitulate emergent properties of the airway epithelium-the primary target for respiratory virus infection. While some of these models have been used for over thirty years, ongoing advancements in both culture techniques and analytical tools continue to provide new opportunities to investigate airway epithelial biology and viral infection phenotypes in both normal and diseased host backgrounds. Here we review these models and their application to studying respiratory viruses. Furthermore, given the ability of these systems to recapitulate the extracellular microenvironment, we evaluate their potential to serve as a platform for studies specifically addressing viral interactions at the mucosal surface and detail techniques that can be employed to expand our understanding.
Collapse
Affiliation(s)
- Ethan Iverson
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA; (E.I.); (E.L.A.)
| | - Logan Kaler
- Biophysics Program, University of Maryland, College Park, MD 20742, USA; (L.K.); (G.A.D.)
| | - Eva L. Agostino
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA; (E.I.); (E.L.A.)
| | - Daniel Song
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA;
| | - Gregg A. Duncan
- Biophysics Program, University of Maryland, College Park, MD 20742, USA; (L.K.); (G.A.D.)
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA;
| | - Margaret A. Scull
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA; (E.I.); (E.L.A.)
| |
Collapse
|
13
|
Xi Y, Upham JW. Plasmacytoid dendritic cells and asthma: a review of current knowledge. Expert Rev Respir Med 2020; 14:1095-1106. [PMID: 32726181 DOI: 10.1080/17476348.2020.1803741] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION While medications are available to treat asthma symptoms and control inflammation, no treatments can cure asthma, and efforts to develop primary prevention strategies or improved exacerbation management are limited by incomplete knowledge of the mechanisms responsible for asthma development and progression. Plasmacytoid dendritic cells (pDC) are involved in anti-viral host defense and immune regulation, and increasing evidence suggests a role for pDC in asthma pathogenesis. AREAS COVERED We undertook a literature search using PubMed for articles including the phrase 'plasmacytoid dendritic cells and asthma' published from 2015 to 2020. We reviewed the remarkable progress made over the past 5 years in understanding the role of pDC in asthma pathogenesis and how pDC regulate anti-viral immune function. This review highlights key recent findings in asthma pathogenesis and virus-triggered asthma exacerbations; pDC biology and functionality; how pDC regulate the immune response; and pDC function in asthma. EXPERT OPTION A deeper understanding of pDC function provides an important foundation for future pDC-targeted therapies that might prevent and treat asthma.
Collapse
Affiliation(s)
- Yang Xi
- The Lung and Allergy Research Centre, the University of Queensland Diamantina Institute, Translational Research Institute , Brisbane, QLD, Australia
| | - John W Upham
- The Lung and Allergy Research Centre, the University of Queensland Diamantina Institute, Translational Research Institute , Brisbane, QLD, Australia.,Department of Respiratory Medicine, Princess Alexandra Hospital , Brisbane, QLD, Australia
| |
Collapse
|
14
|
Parikh V, Scala J, Patel R, Corbi C, Lo D, Bochkov YA, Kennedy JL, Kurten RC, Liggett SB, Gern JE, Koziol-White CJ. Rhinovirus C15 Induces Airway Hyperresponsiveness via Calcium Mobilization in Airway Smooth Muscle. Am J Respir Cell Mol Biol 2020; 62:310-318. [PMID: 31533004 PMCID: PMC7055698 DOI: 10.1165/rcmb.2019-0004oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 09/18/2019] [Indexed: 12/18/2022] Open
Abstract
Rhinovirus (RV) exposure evokes exacerbations of asthma that markedly impact morbidity and mortality worldwide. The mechanisms by which RV induces airway hyperresponsiveness (AHR) or by which specific RV serotypes differentially evoke AHR remain unknown. We posit that RV infection evokes AHR and inflammatory mediator release, which correlate with degrees of RV infection. Furthermore, we posit that rhinovirus C-induced AHR requires paracrine or autocrine mediator release from epithelium that modulates agonist-induced calcium mobilization in human airway smooth muscle. In these studies, we used an ex vivo model to measure bronchoconstriction and mediator release from infected airways in human precision cut lung slices to understand how RV exposure alters airway constriction. We found that rhinovirus C15 (RV-C15) infection augmented carbachol-induced airway narrowing and significantly increased release of IP-10 (IFN-γ-induced protein 10) and MIP-1β (macrophage inflammatory protein-1β) but not IL-6. RV-C15 infection of human airway epithelial cells augmented agonist-induced intracellular calcium flux and phosphorylation of myosin light chain in co-cultured human airway smooth muscle to carbachol, but not after histamine stimulation. Our data suggest that RV-C15-induced structural cell inflammatory responses are associated with viral load but that inflammatory responses and alterations in agonist-mediated constriction of human small airways are uncoupled from viral load of the tissue.
Collapse
Affiliation(s)
- Vishal Parikh
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Jacqueline Scala
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Riva Patel
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Corinne Corbi
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Dennis Lo
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Yury A. Bochkov
- Department of Pediatrics, University of Wisconsin–Madison, Madison, Wisconsin
| | | | - Richard C. Kurten
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas; and
| | - Stephen B. Liggett
- Department of Molecular Pharmacology and Physiology and
- Department of Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - James E. Gern
- Department of Pediatrics, University of Wisconsin–Madison, Madison, Wisconsin
| | | |
Collapse
|
15
|
Basharat U, Aiche MM, Kim MM, Sohal M, Chang EH. Are rhinoviruses implicated in the pathogenesis of sinusitis and chronic rhinosinusitis exacerbations? A comprehensive review. Int Forum Allergy Rhinol 2019; 9:1159-1188. [PMID: 31430424 DOI: 10.1002/alr.22403] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/16/2019] [Indexed: 01/23/2023]
Abstract
BACKGROUND Rhinovirus (RV) infections are the most common cause of viral upper respiratory infections (URIs), and in the majority of persons they are self-limiting. However, in others, viral URIs can progress to bacterial sinusitis and induce chronic rhinosinusitis (CRS) exacerbations. METHODS We conducted a comprehensive Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) review through April 2018 based on MEDLINE, EMBASE, Web of Science-Science Citation Index (SCI), and Conference Proceedings Citation Index- Science (CPCI-S) using keywords: RV, respiratory virus, sinusitis, and airway epithelial cells. The goal of this systematic review was to: (1) determine the prevalence between RV and CRS, (2) study the changes that occur after experimental RV inoculation, (3) investigate the pathophysiologic mechanisms by which RV induces sinonasal inflammation, and (4) explore the treatment options available for RV-associated sinusitis. Data regarding study design, research question, intervention, subjects, outcomes, and biases was extracted. RESULTS The initial search yielded 2395 unique abstracts, of which 614 were selected for full-text review; 147 were included in the final review. We determined that (1) the prevalence of RV infections is increased in those with CRS, (2) humans challenged in vivo with RV secrete local inflammatory mediators with radiographic mucosal thickening, (3) RV species RV-A and RV-C challenges in vitro to sinonasal epithelia produce robust cytokine responses and differential gene changes, and (4) no current therapies have produced consistent and significant resolution of disease. CONCLUSION RV infections are common in persons with CRS, and incite inflammatory reactions that may result in CRS exacerbations and progression of disease. Further studies assessing RV species, and the host-virome response are required to develop new strategies targeting RV-induced CRS.
Collapse
Affiliation(s)
- Usmaan Basharat
- Department of Otolaryngology-Head and Neck Surgery, University of Arizona College of Medicine, Tucson, AZ
| | - Mazen M Aiche
- Department of Otolaryngology-Head and Neck Surgery, University of Arizona College of Medicine, Tucson, AZ
| | - Marianne M Kim
- Department of Otolaryngology-Head and Neck Surgery, University of Arizona College of Medicine, Tucson, AZ
| | - Maheep Sohal
- Department of Otolaryngology-Head and Neck Surgery, University of Arizona College of Medicine, Tucson, AZ
| | - Eugene H Chang
- Department of Otolaryngology-Head and Neck Surgery, University of Arizona College of Medicine, Tucson, AZ
| |
Collapse
|
16
|
Nakauchi M, Nagata N, Takayama I, Saito S, Kubo H, Kaida A, Oba K, Odagiri T, Kageyama T. Propagation of Rhinovirus C in Differentiated Immortalized Human Airway HBEC3-KT Epithelial Cells. Viruses 2019; 11:v11030216. [PMID: 30836639 PMCID: PMC6466094 DOI: 10.3390/v11030216] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/18/2019] [Accepted: 02/27/2019] [Indexed: 01/13/2023] Open
Abstract
Rhinoviruses (RVs) are classified into three species: RV-A, B, and C. Unlike RV-A and -B, RV-C cannot be propagated using standard cell culture systems. In order to isolate RV-Cs from clinical specimens and gain a better understanding of their biological properties and pathogenesis, we established air–liquid-interface (ALI) culture methods using HBEC3-KT and HSAEC1-KT immortalized human airway epithelial cells. HBEC3- and HSAEC1-ALI cultures morphologically resembled pseudostratified epithelia with cilia and goblet cells. Two fully sequenced clinical RV-C isolates, RV-C9 and -C53, were propagated in HBEC3-ALI cultures, and increases in viral RNA ranging from 1.71 log10 to 7.06 log10 copies were observed. However, this propagation did not occur in HSAEC1-ALI cultures. Using the HBEC3-ALI culture system, 11 clinical strains of RV-C were isolated from 23 clinical specimens, and of them, nine were passaged and re-propagated. The 11 clinical isolates were classified as RV-C2, -C6, -C9, -C12, -C18, -C23, -C40, and -C53 types according to their VP1 sequences. Our stable HBEC3-ALI culture system is the first cultivable cell model that supports the growth of multiple RV-C virus types from clinical specimens. Thus, the HBEC3-ALI culture system provides a cheap and easy-to-use alternative to existing cell models for isolating and investigating RV-Cs.
Collapse
Affiliation(s)
- Mina Nakauchi
- Influenza Virus Research Center, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo 208-0011, Japan.
| | - Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo 208-0011, Japan.
| | - Ikuyo Takayama
- Influenza Virus Research Center, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo 208-0011, Japan.
| | - Shinji Saito
- Influenza Virus Research Center, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo 208-0011, Japan.
| | - Hideyuki Kubo
- Division of Microbiology, Osaka Institute of Public Health, 8-34 Tojo-cho, Tennoji-ku, Osaka 543-0026, Japan.
| | - Atsushi Kaida
- Division of Microbiology, Osaka Institute of Public Health, 8-34 Tojo-cho, Tennoji-ku, Osaka 543-0026, Japan.
| | - Kunihiro Oba
- Department of Pediatrics, Showa General Hospital, 8-1-1 Hanakoganei, Kodaira-shi, Tokyo 187-0002, Japan.
| | - Takato Odagiri
- Influenza Virus Research Center, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo 208-0011, Japan.
| | - Tsutomu Kageyama
- Influenza Virus Research Center, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo 208-0011, Japan.
| |
Collapse
|
17
|
Watters K, Palmenberg AC. CDHR3 extracellular domains EC1-3 mediate rhinovirus C interaction with cells and as recombinant derivatives, are inhibitory to virus infection. PLoS Pathog 2018; 14:e1007477. [PMID: 30532249 PMCID: PMC6301718 DOI: 10.1371/journal.ppat.1007477] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 12/20/2018] [Accepted: 11/20/2018] [Indexed: 01/04/2023] Open
Abstract
Viruses in the rhinovirus C species (RV-C) are more likely to cause severe wheezing illnesses and asthma exacerbations in children than related isolates of the RV-A or RV-B. The RV-C capsid is structurally distinct from other rhinoviruses and does not bind ICAM-1 or LDL receptors. The RV-C receptor is instead, human cadherin-related family member 3 (CDHR3), a protein unique to the airway epithelium. A single nucleotide polymorphism (rs6967330, encoding C529Y) in CDHR3 regulates the display density of CDHR3 on cell surfaces and is among the strongest known genetic correlates for childhood virus-induced asthma susceptibility. CDHR3 immunoprecipitations from transfected or transduced cell lysates were used to characterize the RV-C interaction requirements. The C529 and Y529 variations in extracellular repeat domain 5 (EC5), bound equivalently to virus. Glycosylase treatment followed by mass spectrometry mapped 3 extracellular N-linked modification sites, and further detected surface-dependent, α2-6 sialyation unique to the Y529 format. None of these modifications were required for RV-C recognition, but removal or even dilution of structurally stabilizing calcium ions from the EC junctions irreversibly abrogated virus binding. CDHR3 deletions expressed in HeLa cells or as bacterial recombinant proteins, mapped the amino-terminal EC1 unit as the required virus contact. Derivatives containing the EC1 domain, could not only recapitulate virus:receptor interactions in vitro, but also directly inhibit RV-C infection of susceptible cells for several virus genotypes (C02, C15, C41, and C45). We propose that all RV-C use the same EC1 landing pad, interacting with putative EC3-mediated multimerization formats of CDHR3.
Collapse
Affiliation(s)
- Kelly Watters
- Institute for Molecular Virology, Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ann C. Palmenberg
- Institute for Molecular Virology, Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
18
|
Schagen J, Sly PD, Fantino E. Characterizing well-differentiated culture of primary human nasal epithelial cells for use in wound healing assays. J Transl Med 2018; 98:1478-1486. [PMID: 30089850 DOI: 10.1038/s41374-018-0100-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/31/2018] [Accepted: 06/05/2018] [Indexed: 12/18/2022] Open
Abstract
The nasal epithelium is the initial contact between the external environment and the respiratory tract and how it responds to noxious stimuli and repairs epithelial damage is important. Growing airway epithelial cells in culture at air-liquid interface allows for a physiologically relevant model of the human upper airways. The aim of the present study was to characterize human primary nasal epithelial cells grown at the air-liquid interface and establish a model for use in wound healing assays. This study determined the time required for full differentiation of nasal epithelial cells in an air-liquid interface culture to be at least 7 weeks using the standardized B-ALI media. Also, a model was established that studied the response to wounding and the effect of EGFR inhibition on this process. Nasal epithelial cultures from healthy subjects were differentiated at air-liquid interface and manually wounded. Wounds were monitored over time to complete closure using a time lapse imaging microscope with cultures identified to have a rate of wound healing above 2.5%/h independent of initial wound size. EGFR inhibition caused the rate of wound healing to drop a significant 4.6%/h with there being no closure of the wound after 48 h. The robust model established in this study will be essential for studying factors influencing wound healing, including host disease status and environmental exposures in the future.
Collapse
Affiliation(s)
- Johanna Schagen
- Children's Lung, Environment and Asthma Research Team, Centre for Children's Health Research, The University of Queensland, Brisbane, Australia
| | - Peter D Sly
- Children's Lung, Environment and Asthma Research Team, Centre for Children's Health Research, The University of Queensland, Brisbane, Australia.
| | - Emmanuelle Fantino
- Children's Lung, Environment and Asthma Research Team, Centre for Children's Health Research, The University of Queensland, Brisbane, Australia
| |
Collapse
|
19
|
Cox DW, Khoo SK, Zhang G, Lindsay K, Keil AD, Knight G, Gern JE, Laing IA, Bizzintino J, Le Souëf PN. Rhinovirus is the most common virus and rhinovirus-C is the most common species in paediatric intensive care respiratory admissions. Eur Respir J 2018; 52:13993003.00207-2018. [PMID: 29976655 DOI: 10.1183/13993003.00207-2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/13/2018] [Indexed: 11/05/2022]
Affiliation(s)
- Desmond W Cox
- Respiratory Dept, Our Lady's Children's Hospital, Dublin, Ireland.,School of Paediatrics and Child Health, University of Western Australia, Perth, Australia
| | - Siew-Kim Khoo
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia.,Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Guicheng Zhang
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia.,School of Public Health, Curtin University, Centre for Genetic Origins of Health and Disease, Curtin University and the University of Western Australia, Perth, Australia
| | - Katie Lindsay
- Microbiology Dept, PathWest Laboratory Medicine, Perth, Australia
| | - Anthony D Keil
- Microbiology Dept, PathWest Laboratory Medicine, Perth, Australia
| | - Geoff Knight
- Paediatric Intensive Care, Princess Margaret Hospital, Perth, Australia
| | - James E Gern
- Dept of Paediatrics, University of Wisconsin-Madison, Madison, WI, USA
| | - Ingrid A Laing
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia.,Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Joelene Bizzintino
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia.,Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Peter N Le Souëf
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia
| |
Collapse
|
20
|
Martinovich KM, Iosifidis T, Buckley AG, Looi K, Ling KM, Sutanto EN, Kicic-Starcevich E, Garratt LW, Shaw NC, Montgomery S, Lannigan FJ, Knight DA, Kicic A, Stick SM. Conditionally reprogrammed primary airway epithelial cells maintain morphology, lineage and disease specific functional characteristics. Sci Rep 2017; 7:17971. [PMID: 29269735 PMCID: PMC5740081 DOI: 10.1038/s41598-017-17952-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 12/04/2017] [Indexed: 01/19/2023] Open
Abstract
Current limitations to primary cell expansion led us to test whether airway epithelial cells derived from healthy children and those with asthma and cystic fibrosis (CF), co-cultured with an irradiated fibroblast feeder cell in F-medium containing 10 µM ROCK inhibitor could maintain their lineage during expansion and whether this is influenced by underlying disease status. Here, we show that conditionally reprogrammed airway epithelial cells (CRAECs) can be established from both healthy and diseased phenotypes. CRAECs can be expanded, cryopreserved and maintain phenotypes over at least 5 passages. Population doublings of CRAEC cultures were significantly greater than standard cultures, but maintained their lineage characteristics. CRAECs from all phenotypes were also capable of fully differentiating at air-liquid interface (ALI) and maintained disease specific characteristics including; defective CFTR channel function cultures and the inability to repair wounds. Our findings indicate that CRAECs derived from children maintain lineage, phenotypic and importantly disease-specific functional characteristics over a specified passage range.
Collapse
Affiliation(s)
- Kelly M Martinovich
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Thomas Iosifidis
- School of Paediatrics and Child Health, The University of Western Australia, Crawley, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Alysia G Buckley
- Centre of Microscopy, Characterisation and Analysis, The University of Western Australia, Crawley, Western Australia, Australia
| | - Kevin Looi
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Kak-Ming Ling
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Erika N Sutanto
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Elizabeth Kicic-Starcevich
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Luke W Garratt
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Nicole C Shaw
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Samuel Montgomery
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Francis J Lannigan
- School of Paediatrics and Child Health, The University of Western Australia, Crawley, Western Australia, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Anthony Kicic
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia. .,School of Paediatrics and Child Health, The University of Western Australia, Crawley, Western Australia, Australia. .,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia, Australia. .,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia. .,Occupation and Environment, School of Public Health, Curtin University, Perth, Western Australia, Australia.
| | - Stephen M Stick
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia.,School of Paediatrics and Child Health, The University of Western Australia, Crawley, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia
| |
Collapse
|
21
|
Stobart CC, Nosek JM, Moore ML. Rhinovirus Biology, Antigenic Diversity, and Advancements in the Design of a Human Rhinovirus Vaccine. Front Microbiol 2017; 8:2412. [PMID: 29259600 PMCID: PMC5723287 DOI: 10.3389/fmicb.2017.02412] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/21/2017] [Indexed: 01/06/2023] Open
Abstract
Human rhinovirus (HRV) remains a leading cause of several human diseases including the common cold. Despite considerable research over the last 60 years, development of an effective vaccine to HRV has been viewed by many as unfeasible due, in part, to the antigenic diversity of circulating HRVs in nature. Over 150 antigenically distinct types of HRV are currently known which span three species: HRV A, HRV B, and HRV C. Early attempts to develop a rhinovirus vaccine have shown that inactivated HRV is capable of serving as a strong immunogen and inducing neutralizing antibodies. Yet, limitations to virus preparation and recovery, continued identification of antigenic variants of HRV, and logistical challenges pertaining to preparing a polyvalent preparation of the magnitude required for true efficacy against circulating rhinoviruses continue to prove a daunting challenge. In this review, we describe HRV biology, antigenic diversity, and past and present advances in HRV vaccine design.
Collapse
Affiliation(s)
- Christopher C Stobart
- Department of Biological Sciences, Butler University, Indianapolis, IN, United States
| | - Jenna M Nosek
- Department of Biological Sciences, Butler University, Indianapolis, IN, United States
| | - Martin L Moore
- Department of Pediatrics, Emory University, Atlanta, GA, United States.,Children's Healthcare of Atlanta, Atlanta, GA, United States
| |
Collapse
|
22
|
Patel MC, Pletneva LM, Boukhvalova MS, Vogel SN, Kajon AE, Blanco JCG. Immunization with Live Human Rhinovirus (HRV) 16 Induces Protection in Cotton Rats against HRV14 Infection. Front Microbiol 2017; 8:1646. [PMID: 28912760 PMCID: PMC5583225 DOI: 10.3389/fmicb.2017.01646] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/15/2017] [Indexed: 11/13/2022] Open
Abstract
Human rhinoviruses (HRVs) are the main cause of cold-like illnesses, and currently no vaccine or antiviral therapies against HRVs are available to prevent or mitigate HRV infection. There are more than 150 antigenically heterogeneous HRV serotypes, with ∼90 HRVs belonging to major group species A and B. Development of small animal models that are susceptible to infection with major group HRVs would be beneficial for vaccine research. Previously, we showed that the cotton rat (Sigmodon hispidus) is semi-permissive to HRV16 (major group, species HRV-A virus) infection, replicating in the upper and lower respiratory tracts with measurable pathology, mucus production, and expression of inflammatory mediators. Herein, we report that intranasal infection of cotton rats with HRV14 (major group, species HRV-B virus) results in isolation of infectious virus from the nose and lung. Similar to HRV16, intramuscular immunization with live HRV14 induces homologous protection that correlated with high levels of serum neutralizing antibodies. Vaccination and challenge experiments with HRV14 and HRV16 to evaluate the development of cross-protective immunity demonstrate that intramuscular immunization with live HRV16 significantly protects animals against HRV14 challenge. Determination of the immunological mechanisms involved in heterologous protection and further characterization of infection with other major HRV serotypes in the cotton rat could enhance the robustness of the model to define heterotypic relationships between this diverse group of viruses and thereby increase its potential for development of a multi-serotype HRV vaccine.
Collapse
Affiliation(s)
- Mira C Patel
- Sigmovir Biosystems, Inc., RockvilleMD, United States
| | | | | | - Stefanie N Vogel
- University of Maryland School of Medicine, BaltimoreMD, United States
| | - Adriana E Kajon
- Infectious Disease Program, Lovelace Respiratory Research Institute, AlbuquerqueNM, United States
| | | |
Collapse
|
23
|
Griggs TF, Bochkov YA, Basnet S, Pasic TR, Brockman-Schneider RA, Palmenberg AC, Gern JE. Rhinovirus C targets ciliated airway epithelial cells. Respir Res 2017; 18:84. [PMID: 28472984 PMCID: PMC5418766 DOI: 10.1186/s12931-017-0567-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/27/2017] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The Rhinovirus C (RV-C), first identified in 2006, produce high symptom burdens in children and asthmatics, however, their primary target host cell in the airways remains unknown. Our primary hypotheses were that RV-C target ciliated airway epithelial cells (AECs), and that cell specificity is determined by restricted and high expression of the only known RV-C cell-entry factor, cadherin related family member 3 (CDHR3). METHODS RV-C15 (C15) infection in differentiated human bronchial epithelial cell (HBEC) cultures was assessed using immunofluorescent and time-lapse epifluorescent imaging. Morphology of C15-infected differentiated AECs was assessed by immunohistochemistry. RESULTS C15 produced a scattered pattern of infection, and infected cells were shed from the epithelium. The percentage of cells infected with C15 varied from 1.4 to 14.7% depending on cell culture conditions. Infected cells had increased staining for markers of ciliated cells (acetylated-alpha-tubulin [aat], p < 0.001) but not markers of goblet cells (wheat germ agglutinin or Muc5AC, p = ns). CDHR3 expression was increased on ciliated epithelial cells, but not other epithelial cells (p < 0.01). C15 infection caused a 27.4% reduction of ciliated cells expressing CDHR3 (p < 0.01). During differentiation of AECs, CDHR3 expression progressively increased and correlated with both RV-C binding and replication. CONCLUSIONS The RV-C only replicate in ciliated AECs in vitro, leading to infected cell shedding. CDHR3 expression positively correlates with RV-C binding and replication, and is largely confined to ciliated AECs. Our data imply that factors regulating differentiation and CDHR3 production may be important determinants of RV-C illness severity.
Collapse
Affiliation(s)
- Theodor F Griggs
- Department of Pediatrics, School of Medicine and Public Health, CSC K4/945, 600 Highland Ave, Madison, 53792, WI, USA.
- Cellular & Molecular Pathology Graduate Program, Madison, WI, USA.
- Medical Scientist Training Program, Madison, WI, USA.
| | - Yury A Bochkov
- Department of Pediatrics, School of Medicine and Public Health, CSC K4/945, 600 Highland Ave, Madison, 53792, WI, USA
| | - Sarmila Basnet
- Department of Pediatrics, School of Medicine and Public Health, CSC K4/945, 600 Highland Ave, Madison, 53792, WI, USA
| | - Thomas R Pasic
- Department of Surgery, School of Medicine and Public Health, Madison, WI, USA
| | - Rebecca A Brockman-Schneider
- Department of Pediatrics, School of Medicine and Public Health, CSC K4/945, 600 Highland Ave, Madison, 53792, WI, USA
| | - Ann C Palmenberg
- Institute for Molecular Virology, University of Wisconsin, Madison, WI, USA
| | - James E Gern
- Department of Pediatrics, School of Medicine and Public Health, CSC K4/945, 600 Highland Ave, Madison, 53792, WI, USA
- Cellular & Molecular Pathology Graduate Program, Madison, WI, USA
| |
Collapse
|
24
|
Guibas GV, Papadopoulos NG. Viral Upper Respiratory Tract Infections. VIRAL INFECTIONS IN CHILDREN, VOLUME II 2017. [PMCID: PMC7121526 DOI: 10.1007/978-3-319-54093-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The upper respiratory system is one of the most common sites of infection for adults, but even more so for children. Several viruses, from variable families, cause upper respiratory infections which, although generally underestimated due to their typically self-limiting nature, underlie enormous healthcare resource utilization and financial burden. Such, otherwise “benign” infections, can have very significant sequelae both in the form of bringing about local complications but also inducing asthma attacks, thus greatly increasing morbidity. Their enormous prevalence also indicates that rigorous research should be undertaken in order to tackle them, in both the prevention and treatment field.
Collapse
|
25
|
Abstract
Allergy and viral respiratory infections have long been recognized as two of the most important risk factors for exacerbations of asthma. These observations have raised questions regarding potential interactions between these two important risk factors. For example, does allergy diminish the antiviral response, thereby promoting exacerbations of asthma? Alternately, do viral respiratory infections potentiate ongoing allergic inflammation in the airway? The answers to these questions are likely to have implications regarding the prevention and treatment of exacerbations of asthma. This article reviews that clinical evidence linking viral infections and allergy to exacerbations of asthma, reviews potential interactions between these two risk factors, and discusses possible application of new insights in virus/allergen interactions to the prevention and treatment of exacerbations of asthma.
Collapse
|
26
|
Fawkner-Corbett DW, Khoo SK, Duarte CM, Bezerra PGM, Bochkov YA, Gern JE, Le Souef PN, McNamara PS. Rhinovirus-C detection in children presenting with acute respiratory infection to hospital in Brazil. J Med Virol 2016; 88:58-63. [PMID: 26100591 PMCID: PMC4682890 DOI: 10.1002/jmv.24300] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2015] [Indexed: 01/28/2023]
Abstract
Human rhinovirus (RV) is a common cause of acute respiratory infection (ARI) in children. We aimed to characterize the clinical and demographic features associated with different RV species detected in children attending hospital with ARI, from low‐income families in North‐east Brazil. Nasopharyngeal aspirates were collected from 630 children <5 years with ARI. Clinical diagnosis and disease severity were also recorded. Samples were analyzed by multiplex PCR for 18 viral and atypical bacterial pathogens; RV positive samples underwent partial sequencing to determine species and type. RV was the fourth commonest pathogen accounting for 18.7% of pathogens detected. RV was commonly detected in children with bronchiolitis, pneumonia, and asthma/episodic viral wheeze (EVW). Species and type were assigned in 112 cases (73% RV‐A; 27% RV‐C; 0% RV‐B). Generally, there were no differences in clinical or demographic characteristics between those infected with RV‐A and RV‐C. However, in children with asthma/EVW, RV‐C was detected relatively more frequently than RV‐A (23% vs. 5%; P = 0.04). Our findings highlight RV as a potentially important pathogen in this setting. Generally, clinical and demographic features were similar in children in whom RV‐A and C species were detected. However, RV‐C was more frequently found in children with asthma/EVW than RV‐A. J. Med. Virol. 88:58–63, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Siew Kim Khoo
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia
| | - Carminha M Duarte
- Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
| | | | - Yury A Bochkov
- Department of Paediatrics and Medicine, University of Wisconsin School of Medicine and Public Health, Wisconsin
| | - James E Gern
- Department of Paediatrics and Medicine, University of Wisconsin School of Medicine and Public Health, Wisconsin
| | - Peter N Le Souef
- School of Paediatrics and Child Health, University of Western Australia, Perth, Australia
| | - Paul S McNamara
- Institute of Child Health, Alder Hey Children's Hospital, Liverpool, United Kingdom
| |
Collapse
|
27
|
Jackson DJ, Gern JE, Lemanske RF. The contributions of allergic sensitization and respiratory pathogens to asthma inception. J Allergy Clin Immunol 2016; 137:659-65; quiz 666. [PMID: 26947979 PMCID: PMC4782609 DOI: 10.1016/j.jaci.2016.01.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/06/2016] [Accepted: 01/13/2016] [Indexed: 01/10/2023]
Abstract
Of the chronic diseases affecting grade-school children, asthma is the most common and accounts for the greatest number of school days missed. Moreover, it can influence family dynamics and function in other ways, and unfortunately, it can also be associated with mortality, particularly in the inner-city environments of the United States. Thus understanding factors that lead to its development in early life is essential in developing strategies aimed at primary prevention. Two risk factors that have been identified by a number of investigators include the development of allergic sensitization and wheezing respiratory tract illnesses caused by viruses and bacteria, either alone or in combination. Both of these factors appear to exert their influences within the first few years of life, such that asthma becomes established before the child enters grade school at age 5 to 6 years. Therefore, because both allergic sensitization and viral and bacterial illnesses can occur in children who do not have asthma, it is paramount to identify genetic and environmental factors that activate, interact with, and/or direct the immune system and components of the respiratory tract along pathways that allow asthma to become established and expressed clinically.
Collapse
Affiliation(s)
- Daniel J Jackson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wis.
| | - James E Gern
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wis; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Robert F Lemanske
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wis; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| |
Collapse
|
28
|
Bochkov YA, Gern JE. Rhinoviruses and Their Receptors: Implications for Allergic Disease. Curr Allergy Asthma Rep 2016; 16:30. [PMID: 26960297 DOI: 10.1007/s11882-016-0608-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Human rhinoviruses (RVs) are picornaviruses that can cause a variety of illnesses including the common cold, lower respiratory tract illnesses such as bronchitis and pneumonia, and exacerbations of asthma. RVs are classified into three species, RV-A, B, and C, which include over 160 types. They utilize three major types of cellular membrane glycoproteins to gain entry into the host cell: intercellular adhesion molecule 1 (ICAM-1) (the majority of RV-A and all RV-B), low-density lipoprotein receptor (LDLR) family members (12 RV-A types), and cadherin-related family member 3 (CDHR3) (RV-C). CDHR3 is a member of cadherin superfamily of transmembrane proteins with yet unknown biological function, and there is relatively little information available about the mechanisms of RV-C interaction with CDHR3. A coding single nucleotide polymorphism (rs6967330) in CDHR3 could promote RV-C infections and illnesses in infancy, which could in turn adversely affect the developing lung to increase the risk of asthma. Further studies are needed to determine how RV infections contribute to pathogenesis of asthma and to develop the optimal treatment approach to control asthma exacerbations.
Collapse
Affiliation(s)
- Yury A Bochkov
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave, Madison, WI, 53792, USA.
| | - James E Gern
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave, Madison, WI, 53792, USA.,Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave, Madison, WI, 53792, USA
| |
Collapse
|
29
|
Jackson DJ, Lemanske RF, Gern JE. Infections and Asthma. PEDIATRIC ALLERGY: PRINCIPLES AND PRACTICE 2016. [PMCID: PMC7173469 DOI: 10.1016/b978-0-323-29875-9.00031-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Wheezing viral respiratory illnesses are the most common initial presentation of childhood asthma. Once asthma is established, viral infections, most notably rhinovirus (RV), are the most frequent trigger of severe asthma exacerbations. RV-C appears to be a particularly pathogenic virus in children with asthma. Evidence has recently emerged to suggest that bacterial pathogens in the lower airway may contribute to the expression of asthma. Ongoing studies are critical to our understanding of the role of the airway microbiome in asthma inception and exacerbation. Synergistic interactions between underlying allergy and virus infections play an important mechanistic role in asthma inception and exacerbation, and are an important therapeutic target. Novel therapies are needed to prevent and treat virus-induced wheezing and asthma exacerbations.
Collapse
|
30
|
Ljungman P, Snydman D, Boeckh M. Rhinovirus, Coronavirus, Enterovirus, and Bocavirus After Hematopoietic Cell Transplantation or Solid Organ Transplantation. TRANSPLANT INFECTIONS 2016. [PMCID: PMC7123292 DOI: 10.1007/978-3-319-28797-3_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Respiratory viral infections represent a significant cause of morbidity and mortality in immunocompromised hosts. Newer molecular detection assays have allowed for the characterization of several respiratory viruses not previously recognized as having significant clinical impact in the immunocompromised population. Human rhinoviruses are the most common respiratory viruses detected in the upper respiratory tract of hematopoietic cell transplant and lung transplant recipients, and evidence on the impact on clinical outcomes is mounting. Other respiratory viruses including enteroviruses (EVs), coronaviruses (CoVs), and bocavirus may also contribute to pulmonary disease; however, data is limited in the immunocompromised population. Further studies are needed to define the epidemiology, risk factors, and clinical outcomes of these infections; this data will help inform decisions regarding development of antiviral therapy and infection prevention strategies.
Collapse
Affiliation(s)
- Per Ljungman
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - David Snydman
- Tufts University School of Medicine Tufts Medical Center, Boston, Massachusetts USA
| | - Michael Boeckh
- University of Washington Fred Hutchinson Cancer Research Center, Seattle, Washington USA
| |
Collapse
|
31
|
Viral-Induced Wheeze and Asthma Development. ALLERGY, IMMUNITY AND TOLERANCE IN EARLY CHILDHOOD 2016. [PMCID: PMC7173475 DOI: 10.1016/b978-0-12-420226-9.00005-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
32
|
Lee WM, Chen Y, Wang W, Mosser A. Infectivity assays of human rhinovirus-A and -B serotypes. Methods Mol Biol 2015; 1221:71-81. [PMID: 25261308 DOI: 10.1007/978-1-4939-1571-2_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Infectivity is a fundamental property of viral pathogens such as human rhinoviruses (HRVs). This chapter describes two methods for measuring the infectivity of HRV-A and -B serotypes: end point dilution (TCID50) assay and plaque assay. End point dilution assay is a quantal, not quantitative, assay that determines the dilution of the sample at which 50 % of the aliquots have infectious virus. It can be used for all the HRV-A and -B serotypes and related clinical isolates that grow in cell culture and induce cytopathic effect (CPE), degenerative changes in cells that are visible under a microscope. Plaque assay is a quantitative assay that determines the number of infectious units of a virus in a sample. After an infectious unit of virus infects one cell, the infected cell produces progeny viruses that then infect and kill a circle of adjacent cells. This circle of dead cells detaches from the dish and thus leaves a clear hole in a cell monolayer. Plaque assay works only for HeLa-adapted HRV-A and -B serotypes that can make visible plaques on the cell monolayer. Currently the end point dilution assay and plaque assay have not been developed for the newly discovered HRV-C.
Collapse
Affiliation(s)
- Wai-Ming Lee
- Biological Mimetics Inc., 124 Byte Drive, Frederick, MD, 21702, USA,
| | | | | | | |
Collapse
|
33
|
Cadherin-related family member 3, a childhood asthma susceptibility gene product, mediates rhinovirus C binding and replication. Proc Natl Acad Sci U S A 2015; 112:5485-90. [PMID: 25848009 DOI: 10.1073/pnas.1421178112] [Citation(s) in RCA: 298] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Members of rhinovirus C (RV-C) species are more likely to cause wheezing illnesses and asthma exacerbations compared with other rhinoviruses. The cellular receptor for these viruses was heretofore unknown. We report here that expression of human cadherin-related family member 3 (CDHR3) enables the cells normally unsusceptible to RV-C infection to support both virus binding and replication. A coding single nucleotide polymorphism (rs6967330, C529Y) was previously linked to greater cell-surface expression of CDHR3 protein, and an increased risk of wheezing illnesses and hospitalizations for childhood asthma. Compared with wild-type CDHR3, cells transfected with the CDHR3-Y529 variant had about 10-fold increases in RV-C binding and progeny yields. We developed a transduced HeLa cell line (HeLa-E8) stably expressing CDHR3-Y529 that supports RV-C propagation in vitro. Modeling of CDHR3 structure identified potential binding sites that could impact the virus surface in regions that are highly conserved among all RV-C types. Our findings identify that the asthma susceptibility gene product CDHR3 mediates RV-C entry into host cells, and suggest that rs6967330 mutation could be a risk factor for RV-C wheezing illnesses.
Collapse
|
34
|
Glanville N, Johnston SL. Challenges in developing a cross-serotype rhinovirus vaccine. Curr Opin Virol 2015; 11:83-8. [PMID: 25829255 DOI: 10.1016/j.coviro.2015.03.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 02/13/2015] [Accepted: 03/09/2015] [Indexed: 12/20/2022]
Abstract
A great burden of disease is attributable to human rhinovirus (HRV) infections which are the major cause of the common cold, exacerbations of both asthma and chronic obstructive pulmonary disease (COPD), and are associated with asthma development. Despite this there is currently no vaccine for HRV. The first vaccine studies showed some promise in terms of serotype-specific protection against cold symptoms, but antigenic heterogeneity amongst the >150 HRVs has been regarded as a major barrier to effective vaccine development and has resulted in little progress over 50 years. Here we review those vaccine studies conducted to date, discuss the difficulties posed by antigenic heterogeneity and describe some recent advances in generating cross-reactive antibodies and T cell responses using peptide immunogens.
Collapse
Affiliation(s)
- Nicholas Glanville
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College London, United Kingdom; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Sebastian L Johnston
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College London, United Kingdom; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom.
| |
Collapse
|
35
|
Griggs TF, Bochkov YA, Nakagome K, Palmenberg AC, Gern JE. Production, purification, and capsid stability of rhinovirus C types. J Virol Methods 2015; 217:18-23. [PMID: 25724434 DOI: 10.1016/j.jviromet.2015.02.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 12/09/2014] [Accepted: 02/17/2015] [Indexed: 10/23/2022]
Abstract
The rhinovirus C (RV-C) were discovered in 2006 and these agents are an important cause of respiratory morbidity. Little is known about their biology. RV-C15 (C15) can be produced by transfection of recombinant viral RNA into cells and subsequent purification over a 30% sucrose cushion, even though yields and infectivity of other RV-C genotypes with this protocol are low. The goal of this study was to determine whether poor RV-C yields were due to capsid instability, and moreover, to develop a robust protocol suitable for the purification of many RV-C types. Capsid stability assays indicated that virions of RV-C41 (refractory to purification) have similar tolerance for osmotic and temperature stress as RV-A16 (purified readily), although C41 is more sensitive to low pH. Modification to the purification protocol by removing detergent increased the yield of RV-C. Addition of nonfat dry milk to the sucrose cushion increased the virus yield but sacrificed purity of the viral suspension. Analysis of virus distribution following centrifugation indicated that the majority of detectable viral RNA (vRNA) was found in pellets refractory to resuspension. Reduction of the centrifugal force with commiserate increase in spin-time improved the recovery of RV-C for both C41 and C2. Transfection of primary lung fibroblasts (WisL cells) followed by the modified purification protocol further improved yields of infectious C41 and C2. Described herein is a higher yield purification protocol suitable for RV-C types refractory to the standard purification procedure. The findings suggest that aggregation-adhesion problems rather than capsid instability influence RV-C yield during purification.
Collapse
Affiliation(s)
- Theodor F Griggs
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, United States; Cellular & Molecular Pathology Graduate Program, University of Wisconsin, Madison, United States; Medical Scientist Training Program, University of Wisconsin, Madison, United States.
| | - Yury A Bochkov
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, United States
| | - Kazuyuki Nakagome
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, United States
| | - Ann C Palmenberg
- Institute for Molecular Virology, University of Wisconsin, Madison, United States
| | - James E Gern
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, United States; Cellular & Molecular Pathology Graduate Program, University of Wisconsin, Madison, United States
| |
Collapse
|
36
|
Phenotypic responses of differentiated asthmatic human airway epithelial cultures to rhinovirus. PLoS One 2015; 10:e0118286. [PMID: 25706956 PMCID: PMC4338293 DOI: 10.1371/journal.pone.0118286] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 01/12/2015] [Indexed: 12/19/2022] Open
Abstract
Objectives Human airway epithelial cells are the principal target of human rhinovirus (HRV), a common cold pathogen that triggers the majority of asthma exacerbations. The objectives of this study were 1) to evaluate an in vitro air liquid interface cultured human airway epithelial cell model for HRV infection, and 2) to identify gene expression patterns associated with asthma intrinsically and/or after HRV infection using this model. Methods Air-liquid interface (ALI) human airway epithelial cell cultures were prepared from 6 asthmatic and 6 non-asthmatic donors. The effects of rhinovirus RV-A16 on ALI cultures were compared. Genome-wide gene expression changes in ALI cultures following HRV infection at 24 hours post exposure were further analyzed using RNA-seq technology. Cellular gene expression and cytokine/chemokine secretion were further evaluated by qPCR and a Luminex-based protein assay, respectively. Main Results ALI cultures were readily infected by HRV. RNA-seq analysis of HRV infected ALI cultures identified sets of genes associated with asthma specific viral responses. These genes are related to inflammatory pathways, epithelial structure and remodeling and cilium assembly and function, including those described previously (e.g. CCL5, CXCL10 and CX3CL1, MUC5AC, CDHR3), and novel ones that were identified for the first time in this study (e.g. CCRL1). Conclusions ALI-cultured human airway epithelial cells challenged with HRV are a useful translational model for the study of HRV-induced responses in airway epithelial cells, given that gene expression profile using this model largely recapitulates some important patterns of gene responses in patients during clinical HRV infection. Furthermore, our data emphasize that both abnormal airway epithelial structure and inflammatory signaling are two important asthma signatures, which can be further exacerbated by HRV infection.
Collapse
|
37
|
Chimeric rhinoviruses obtained via genetic engineering or artificially induced recombination are viable only if the polyprotein coding sequence derives from the same species. J Virol 2015; 89:4470-80. [PMID: 25653446 DOI: 10.1128/jvi.03668-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
UNLABELLED Recombination is a widespread phenomenon that ensures both the stability and variation of RNA viruses. This phenomenon occurs with different frequencies within species of the Enterovirus genus. Intraspecies recombination is described frequently among non-rhinovirus enteroviruses but appears to be sporadic in rhinoviruses. Interspecies recombination is even rarer for rhinoviruses and mostly is related to ancient events which contributed to the speciation of these viruses. We reported that artificially engineered 5' untranslated region (UTR) interspecies rhinovirus/rhinovirus or rhinovirus/non-rhinovirus enterovirus recombinants are fully viable. Using a similar approach, we demonstrated in this study that exchanges of the P1-2A polyprotein region between members of the same rhinovirus species, but not between members of different species, give rise to competent chimeras. To further assess the rhinovirus intra- and interspecies recombination potential, we used artificially induced recombination by cotransfection of 5'-end-deleted and 3'-end-deleted and replication-deficient genomes. In this system, intraspecies recombination also resulted in viable viruses with high frequency, whereas no interspecies rhinovirus recombinants could be recovered. Mapping intraspecies recombination sites within the polyprotein highlighted recombinant hotspots in nonstructural genes and at gene boundaries. Notably, all recombinants occurring at gene junctions presented in-frame sequence duplications, whereas most intragenic recombinants were homologous. Taken together, our results suggest that only intraspecies recombination gives rise to viable rhinovirus chimeras in the polyprotein coding region and that recombination hotspots map to nonstructural genes with in-frame duplications at gene boundaries. These data provide new insights regarding the mechanism and limitations of rhinovirus recombination. IMPORTANCE Recombination represents a means to ensure both the stability and the variation of RNA viruses. While intraspecies recombination is described frequently among non-rhinovirus enteroviruses, it seems to occur more rarely in rhinoviruses. Interspecies recombination is even rarer in this virus group and is mostly related to ancient events, which contributed to its speciation. We used engineered chimeric genomes and artificially induced RNA recombination to study experimentally the recombination potential of rhinoviruses and analyze recombination sites. Our results suggest that only intraspecies recombination gives rise to viable chimeras in the polyprotein coding region. Furthermore, characterization of intraspecies chimeras provides new insight into putative recombination hotspots within the polyprotein. In summary, we applied two powerful and complementary experimental approaches to improve current knowledge on rhinovirus recombination.
Collapse
|
38
|
Del Vecchio AM, Branigan PJ, Barnathan ES, Flavin SK, Silkoff PE, Turner RB. Utility of animal and in vivo experimental infection of humans with rhinoviruses in the development of therapeutic agents for viral exacerbations of asthma and chronic obstructive pulmonary disease. Pulm Pharmacol Ther 2015; 30:32-43. [PMID: 25445932 PMCID: PMC7110859 DOI: 10.1016/j.pupt.2014.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/24/2014] [Accepted: 10/29/2014] [Indexed: 12/16/2022]
Abstract
There is an association with acute viral infection of the respiratory tract and exacerbations of asthma and chronic obstructive pulmonary disease (COPD). Although these exacerbations are associated with several types of viruses, human rhinoviruses (HRVs) are associated with the vast majority of disease exacerbations. Due to the lack of an animal species that is naturally permissive for HRVs to use as a facile model system, and the limitations associated with animal models of asthma and COPD, studies of controlled experimental infection of humans with HRVs have been used and conducted safely for decades. This review discusses how these experimental infection studies with HRVs have provided a means of understanding the pathophysiology underlying virus-induced exacerbations of asthma and COPD with the goal of developing agents for their prevention and treatment.
Collapse
Affiliation(s)
- Alfred M Del Vecchio
- Janssen Research and Development, Immunology Clinical Research and Development, Welsh and McKean Roads, Spring House, PA 19477, USA
| | - Patrick J Branigan
- Janssen Research and Development, Immunology Clinical Research and Development, Welsh and McKean Roads, Spring House, PA 19477, USA
| | - Elliot S Barnathan
- Janssen Research and Development, Immunology Clinical Research and Development, Welsh and McKean Roads, Spring House, PA 19477, USA
| | - Susan K Flavin
- Janssen Research and Development, Immunology Clinical Research and Development, Welsh and McKean Roads, Spring House, PA 19477, USA
| | - Philip E Silkoff
- Janssen Research and Development, Immunology Clinical Research and Development, Welsh and McKean Roads, Spring House, PA 19477, USA.
| | - Ronald B Turner
- University of Virginia, School of Medicine, Charlottesville, VA 22908, USA
| |
Collapse
|
39
|
Jazaeri Farsani SM, Oude Munnink BB, Canuti M, Deijs M, Cotten M, Jebbink MF, Verhoeven J, Kellam P, Loens K, Goossens H, Ieven M, van der Hoek L. Identification of a novel human rhinovirus C type by antibody capture VIDISCA-454. Viruses 2015; 7:239-51. [PMID: 25606972 PMCID: PMC4306836 DOI: 10.3390/v7010239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 01/08/2015] [Accepted: 01/13/2015] [Indexed: 12/31/2022] Open
Abstract
Causative agents for more than 30 percent of respiratory infections remain unidentified, suggesting that unknown respiratory pathogens might be involved. In this study, antibody capture VIDISCA-454 (virus discovery cDNA-AFLP combined with Roche 454 high-throughput sequencing) resulted in the discovery of a novel type of rhinovirus C (RV-C). The virus has an RNA genome of at least 7054 nt and carries the characteristics of rhinovirus C species. The gene encoding viral protein 1, which is used for typing, has only 81% nucleotide sequence identity with the closest known RV-C type, and, therefore, the virus represents the first member of a novel type, named RV-C54.
Collapse
Affiliation(s)
- Seyed Mohammad Jazaeri Farsani
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam 1105 AZ, the Netherlands.
| | - Bas B Oude Munnink
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam 1105 AZ, the Netherlands.
| | - Marta Canuti
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam 1105 AZ, the Netherlands.
| | - Martin Deijs
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam 1105 AZ, the Netherlands.
| | - Matthew Cotten
- Department of Virus Genomics, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK.
| | - Maarten F Jebbink
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam 1105 AZ, the Netherlands.
| | - Joost Verhoeven
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam 1105 AZ, the Netherlands.
| | - Paul Kellam
- Department of Virus Genomics, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK.
| | - Katherine Loens
- Department of Medical Microbiology, Vaccine and Infectious Disease Institute, University Hospital, Antwerp B-2650, Belgium.
| | - Herman Goossens
- Department of Medical Microbiology, Vaccine and Infectious Disease Institute, University Hospital, Antwerp B-2650, Belgium.
| | - Margareta Ieven
- Department of Medical Microbiology, Vaccine and Infectious Disease Institute, University Hospital, Antwerp B-2650, Belgium.
| | - Lia van der Hoek
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam 1105 AZ, the Netherlands.
| |
Collapse
|
40
|
Temperature-dependent innate defense against the common cold virus limits viral replication at warm temperature in mouse airway cells. Proc Natl Acad Sci U S A 2015; 112:827-32. [PMID: 25561542 DOI: 10.1073/pnas.1411030112] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Most isolates of human rhinovirus, the common cold virus, replicate more robustly at the cool temperatures found in the nasal cavity (33-35 °C) than at core body temperature (37 °C). To gain insight into the mechanism of temperature-dependent growth, we compared the transcriptional response of primary mouse airway epithelial cells infected with rhinovirus at 33 °C vs. 37 °C. Mouse airway cells infected with mouse-adapted rhinovirus 1B exhibited a striking enrichment in expression of antiviral defense response genes at 37 °C relative to 33 °C, which correlated with significantly higher expression levels of type I and type III IFN genes and IFN-stimulated genes (ISGs) at 37 °C. Temperature-dependent IFN induction in response to rhinovirus was dependent on the MAVS protein, a key signaling adaptor of the RIG-I-like receptors (RLRs). Stimulation of primary airway cells with the synthetic RLR ligand poly I:C led to greater IFN induction at 37 °C relative to 33 °C at early time points poststimulation and to a sustained increase in the induction of ISGs at 37 °C relative to 33 °C. Recombinant type I IFN also stimulated more robust induction of ISGs at 37 °C than at 33 °C. Genetic deficiency of MAVS or the type I IFN receptor in infected airway cells permitted higher levels of viral replication, particularly at 37 °C, and partially rescued the temperature-dependent growth phenotype. These findings demonstrate that in mouse airway cells, rhinovirus replicates preferentially at nasal cavity temperature due, in part, to a less efficient antiviral defense response of infected cells at cool temperature.
Collapse
|
41
|
Lee WM, Chen Y, Wang W, Mosser A. Growth of human rhinovirus in H1-HeLa cell suspension culture and purification of virions. Methods Mol Biol 2015; 1221:49-61. [PMID: 25261306 DOI: 10.1007/978-1-4939-1571-2_5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
HeLa cell culture is the most widely used system for in vitro studies of the basic biology of human rhinovirus (HRV). It is also useful for making sufficient quantities of virus for experiments that require highly concentrated and purified virus. This chapter describes the protocols for producing a large amount of HeLa cells in suspension culture, using these cells to grow a large quantity of virus of HeLa-adapted HRV-A and -B serotypes, and making highly concentrated virus stock and highly purified virions. These purified HRV virions are free of cellular components and suitable for experiments that are sensitive to cellular contaminations.
Collapse
Affiliation(s)
- Wai-Ming Lee
- Biological Mimetics Inc., 124 Byte Drive, Frederick, MD, 21702, USA
| | | | | | | |
Collapse
|
42
|
Ashraf S, Brockman-Schneider R, Gern JE. Propagation of rhinovirus-C strains in human airway epithelial cells differentiated at air-liquid interface. Methods Mol Biol 2015; 1221:63-70. [PMID: 25261307 DOI: 10.1007/978-1-4939-1571-2_6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Rhinovirus-C (RV-C) were discovered recently using molecular methods. Classical methods failed to detect them since they could not grow in standard cell culture. The complete genome sequences of several RV-C strains are now available but there is little information about their biological characteristics. HRV-C were first grown in organ culture, and more recently, we developed a system for culturing RV-C strains in differentiated epithelial cells of human airway at air-liquid interface (ALI). These cultures supported efficient replication of RV-C strains as determined by quantitative RT-PCR. This system has enabled study of the biological characteristics of RV-C strains, including quantitative research.
Collapse
Affiliation(s)
- Shamaila Ashraf
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA,
| | | | | |
Collapse
|
43
|
Abstract
The historical classification of human rhinoviruses (RV) by serotyping has been replaced by a logical system of comparative sequencing. Given that strains must diverge within their capsid sequenced by a reasonable degree (>12-13 % pairwise base identities) before becoming immunologically distinct, the new nomenclature system makes allowances for the addition of new, future types, without compromising historical designations. Currently, three species, the RV-A, RV-B, and RV-C, are recognized. Of these, the RV-C, discovered in 2006, are the most unusual in terms of capsid structure, receptor use, and association with severe disease in children.
Collapse
Affiliation(s)
- Ann C Palmenberg
- Institute for Molecular Virology, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI, 53706, USA,
| | | |
Collapse
|
44
|
Affiliation(s)
- J. E. Gern
- Pediatrics and Medicine; University of Wisconsin-Madison; Madison WI USA
| |
Collapse
|
45
|
Abstract
Human rhinovirus (HRV) contains a 7.2 kb messenger-sense RNA genome which is the template for reproducing progeny viruses after it enters the cytoplasm of a host cell. Reverse genetics refers to the regeneration of progeny viruses from an artificial cDNA copy of the RNA genome of an RNA virus. It has been a powerful molecular genetic tool for studying HRV and other RNA viruses because the artificial DNA stage makes it practical to introduce specific mutations into the viral RNA genome. This chapter uses HRV-16 as the model virus to illustrate the strategy and methods for constructing and cloning the artificial cDNA copy of a full-length HRV genome, identifying the infectious cDNA clone isolates, and selecting the most vigorous cDNA clone isolate to serve as the standard parental clone for future molecular genetic study of the virus.
Collapse
|
46
|
Abstract
Human rhinovirus (HRV) infections are now widely accepted as the commonest cause of acute respiratory illnesses (ARIs) in children. Advanced PCR techniques have enabled HRV infections to be identified as causative agents in most common ARIs in childhood including bronchiolitis, acute asthma, pneumonia and croup. However, the long-term implications of rhinovirus infections are less clear. The aim of this review is to examine the relationship between rhinovirus infections and disorders of the lower airways in childhood.
Collapse
Affiliation(s)
- D W Cox
- School of Paediatrics and Child Health, University of Western Australia, Perth, WA, Australia; Respiratory Department, Our Lady's Children's Hospital, Crumlin, Dublin 12, Ireland
| | - P N Le Souëf
- School of Paediatrics and Child Health, University of Western Australia, Perth, WA, Australia; Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| |
Collapse
|
47
|
Parker LC, Stokes CA, Sabroe I. Rhinoviral infection and asthma: the detection and management of rhinoviruses by airway epithelial cells. Clin Exp Allergy 2014; 44:20-8. [PMID: 24355017 DOI: 10.1111/cea.12182] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 07/02/2013] [Accepted: 07/22/2013] [Indexed: 11/28/2022]
Abstract
Human rhinoviruses (HRV) have been linked to the development of childhood asthma and recurrent acute asthma exacerbations throughout life, and contribute considerably to the healthcare and economic burden of this disease. However, the ability of HRV infections to trigger exacerbations, and the link between allergic status and HRV responsiveness, remains incompletely understood. Whilst the receptors on human airway cells that detect and are utilized by most HRV group A and B, but not C serotypes are known, how endosomal pattern recognition receptors (PRRs) detect HRV replication products that are generated within the cytoplasm remains somewhat of an enigma. In this article, we explore a role for autophagy, a cellular homeostatic process that allows the cell to encapsulate its own cytosolic constituents, as the crucial mechanism controlling this process and regulating the innate immune response of airway epithelial cells to viral infection. We will also briefly describe some of the recent insights into the immune responses of the airway to HRV, focusing on neutrophilic inflammation that is a potentially unwanted feature of the acute response to viral infection, and the roles of IL-1 and Pellinos in the regulation of responses to HRV.
Collapse
Affiliation(s)
- L C Parker
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK
| | | | | |
Collapse
|
48
|
Karta MR, Gavala ML, Curran CS, Wickert LE, Keely PJ, Gern JE, Bertics PJ. LPS modulates rhinovirus-induced chemokine secretion in monocytes and macrophages. Am J Respir Cell Mol Biol 2014; 51:125-34. [PMID: 24498897 PMCID: PMC4091859 DOI: 10.1165/rcmb.2013-0404oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 01/30/2014] [Indexed: 01/01/2023] Open
Abstract
Recent studies suggest that both bacteria and rhinoviruses (RVs) contribute to asthma exacerbations. We hypothesized that bacteria might alter antiviral responses early in the course of infection by modifying monocyte-lineage chemokine responses to RV infection. To test this hypothesis, human blood monocytes or bronchoalveolar lavage (BAL) macrophages were treated with RV types A016, B014, A001, and/or A002 in the presence or absence of LPS, and secretion of chemokines (CXCL10, CXCL11, CCL2, and CCL8) and IFN-α was measured by ELISA. Treatment with RV alone induced blood monocytes and BAL macrophages to secrete CXCL10, CXCL11, CCL2, and CCL8. Pretreatment with LPS significantly attenuated RV-induced CXCL10, CXCL11, and CCL8 secretion by 68-99.9% on average (P < 0.0001, P < 0.004, and P < 0.002, respectively), but did not inhibit RV-induced CCL2 from blood monocytes. Similarly, LPS inhibited RV-induced CXCL10 and CXCL11 secretion by over 88% on average from BAL macrophages (P < 0.002 and P < 0.0001, respectively). Furthermore, LPS inhibited RV-induced signal transducer and activator of transcription 1 phosphorylation (P < 0.05), as determined by immunoblotting, yet augmented RV-induced IFN-α secretion (P < 0.05), and did not diminish expression of RV target receptors, as measured by flow cytometry. In summary, major and minor group RVs strongly induce chemokine expression and IFN-α from monocytic cells. The bacterial product, LPS, specifically inhibits monocyte and macrophage secretion of RV-induced CXCL10 and CXCL11, but not other highly induced chemokines or IFN-α. These effects suggest that airway bacteria could modulate the pattern of virus-induced cell recruitment and inflammation in the airways.
Collapse
Affiliation(s)
- Maya R. Karta
- Molecular and Cellular Pharmacology Graduate Program, and
- Departments of Biomolecular Chemistry
| | | | | | | | - Patricia J. Keely
- Molecular and Cellular Pharmacology Graduate Program, and
- Cellular and Regenerative Biology, and
| | - James E. Gern
- Pediatrics and Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | | |
Collapse
|
49
|
Nakagome K, Bochkov YA, Ashraf S, Brockman-Schneider RA, Evans MD, Pasic TR, Gern JE. Effects of rhinovirus species on viral replication and cytokine production. J Allergy Clin Immunol 2014; 134:332-41. [PMID: 24636084 DOI: 10.1016/j.jaci.2014.01.029] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 01/27/2014] [Accepted: 01/29/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND Epidemiologic studies provide evidence of differential virulence of rhinovirus species (RV). We recently reported that RV-A and RV-C induced more severe illnesses than RV-B, which suggests that the biology of RV-B might be different from RV-A or RV-C. OBJECTIVE To test the hypothesis that RV-B has lower replication and induces lesser cytokine responses than RV-A or RV-C. METHODS We cloned full-length cDNA of RV-A16, A36, B52, B72, C2, C15, and C41 from clinical samples and grew clinical isolates of RV-A7 and RV-B6 in cultured cells. Sinus epithelial cells were differentiated at the air-liquid interface. We tested for differences in viral replication in epithelial cells after infection with purified viruses (10(8) RNA copies) and measured virus load by quantitative RT-PCR. We measured lactate dehydrogenase (LDH) concentration as a marker of cellular cytotoxicity, and cytokine and/or chemokine secretion by multiplex ELISA. RESULTS At 24 hours after infection, the virus load of RV-B (RV-B52, RV-B72, or RV-B6) in adherent cells was lower than that of RV-A or RV-C. The growth kinetics of infection indicated that RV-B types replicate more slowly. Furthermore, RV-B released less LDH than RV-A or RV-C, and induced lower levels of cytokines and chemokines such as CXCL10, even after correction for viral replication. RV-B replicates to lower levels also in primary bronchial epithelial cells. CONCLUSIONS Our results indicate that RV-B types have lower and slower replication, and lower cellular cytotoxicity and cytokine and/or chemokine production compared with RV-A or RV-C. These characteristics may contribute to reduced severity of illnesses that has been observed with RV-B infections.
Collapse
Affiliation(s)
- Kazuyuki Nakagome
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, Wis
| | - Yury A Bochkov
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, Wis
| | - Shamaila Ashraf
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, Wis
| | | | - Michael D Evans
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, Wis
| | - Thomas R Pasic
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, Wis
| | - James E Gern
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, Wis; Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wis.
| |
Collapse
|
50
|
Multiple classes of antiviral agents exhibit in vitro activity against human rhinovirus type C. Antimicrob Agents Chemother 2013; 58:1546-55. [PMID: 24366736 DOI: 10.1128/aac.01746-13] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human rhinovirus type C (HRV-C) is a newly discovered enterovirus species frequently associated with exacerbation of asthma and other acute respiratory conditions. Until recently, HRV-C could not be propagated in vitro, hampering in-depth characterization of the virus replication cycle and preventing efficient testing of antiviral agents. Herein we describe several subgenomic RNA replicon systems and a cell culture infectious model for HRV-C that can be used for antiviral screening. The replicon constructs consist of genome sequences from HRVc15, HRVc11, HRVc24, and HRVc25 strains, with the P1 capsid region replaced by a Renilla luciferase coding sequence. Following transfection of the replicon RNA into HeLa cells, the constructs produced time-dependent increases in luciferase signal that can be inhibited in a dose-dependent manner by known inhibitors of HRV replication, including the 3C protease inhibitor rupintrivir, the nucleoside analog inhibitor MK-0608, and the phosphatidylinositol 4-kinase IIIβ (PI4K-IIIβ) kinase inhibitor PIK93. Furthermore, with the exception of pleconaril and pirodavir, the other tested classes of HRV inhibitors blocked the replication of full-length HRVc15 and HRVc11 in human airway epithelial cells (HAEs) that were differentiated in the air-liquid interface, exhibiting antiviral activities similar to those observed with HRV-16. In summary, this study is the first comprehensive profiling of multiple classes of antivirals against HRV-C, and the set of newly developed quantitative HRV-C antiviral assays represent indispensable tools for the identification and evaluation of novel panserotype HRV inhibitors.
Collapse
|