1
|
Tang J, Chen S, Zhong Y, Deng Y, Huang D, Liu J, Zheng Y, Xu J, Xue B, Wang F, Zhou Y, Wang H, Yang Q, Chen X. Development of a reporter HBoV1 strain for antiviral drug screening and life cycle studies. Virol Sin 2025; 40:275-283. [PMID: 40147635 DOI: 10.1016/j.virs.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/21/2025] [Indexed: 03/29/2025] Open
Abstract
Human bocavirus 1 (HBoV1; family: Parvoviridae) causes a wide spectrum of respiratory diseases in children and gastroenteritis in adults. A lack of sensitive cell lines and efficient animal models hinders research on HBoV, including the development of anti-HBoV drugs or vaccines. Although the construction of a wild-type HBoV1 infectious clone has been reported, generating HBoV1 infectious clone carrying foreign reporter genes with suitable insertion sites in its genome while retaining replicative ability remains challenging. Here, HBoV1 infectious clones harboring the 11-amino-acid HiBiT tag at five distinct insertion sites were constructed and evaluated. Only the recombinant HBoV1 carrying the HiBiT tag in the N-terminus of the NS1 protein (HBoV1-HiBiTNS1) displayed comparable characteristics to wild-type HBoV1 as determined via the analysis of viral DNA copy number, NanoLuc activity, viral protein expression, and the formation of replication intermediates. Notably, the replication kinetics of HBoV1-HiBiTNS1 could be examined by monitoring NanoLuc activity, which was noted to be correlated with the viral DNA level. Additionally, we successfully applied HiBiT-tagged HBoV1 for the evaluation of antiviral drug activity and identified ivermectin (EC50 = 2.27 μM) as a potent anti-HBoV1 replication drug. Overall, our study demonstrated that the HBoV1-HiBiTNS1 reporter can serve as a convenient platform for screening candidate drugs targeting HBoV1 replication and may also be useful for investigating the life cycle of the virus.
Collapse
Affiliation(s)
- Jielin Tang
- Guangzhou National Laboratory, Guangzhou 510005, China; State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 511436, China.
| | - Sijie Chen
- Guangzhou National Laboratory, Guangzhou 510005, China; State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 511436, China
| | - Yi Zhong
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Yijun Deng
- Guangzhou National Laboratory, Guangzhou 510005, China; State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 511436, China
| | - Dan Huang
- Guangzhou National Laboratory, Guangzhou 510005, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Junjun Liu
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Yi Zheng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Jiyuan Xu
- Guangzhou National Laboratory, Guangzhou 510005, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Bao Xue
- Guangzhou National Laboratory, Guangzhou 510005, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Fan Wang
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yuan Zhou
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Hanzhong Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Qi Yang
- Guangzhou National Laboratory, Guangzhou 510005, China; State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 511436, China.
| | - Xinwen Chen
- Guangzhou National Laboratory, Guangzhou 510005, China; State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
2
|
Tang J, Chen S, Deng Y, Liu J, Huang D, Fu M, Xue B, Liu C, Wu C, Wang F, Zhou Y, Yang Q, Chen X. MA104 cell line is permissive for human bocavirus 1 infection. J Virol 2025; 99:e0153924. [PMID: 39846742 PMCID: PMC11852709 DOI: 10.1128/jvi.01539-24] [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: 09/14/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025] Open
Abstract
Human bocavirus 1 (HBoV1) has appeared as an emerging pathogen, causing mild to life-threatening respiratory tract infections, acute otitis media, and encephalitis in young children and immunocompromised individuals. The lack of cell lines suitable for culturing replicative viruses hinders research on HBoV1. Here, we characterized the susceptibility to HBoV1 of 29 human and 7 animal cell lines, and identified a permissive cell line, MA104. The complete HBoV1 life cycle was achieved in MA104 cells, including viral entry, complete replication, and infectious progeny virion production. Additionally, the suppression of the interferon pathway facilitated the viral genome replication in MA104 cells. RNA-sequencing showed that innate immunity, inflammation, the PI3K-Akt and MAPK signaling pathways, and the cellular membrane system were mobilized in response to HBoV1 infection. Overall, our study is the first to identify a cell line, MA104, that supports the complete HBoV1 life cycle, which will promote research on HBoV1 virology and pathogenesis and benefit drug and vaccine development.IMPORTANCEHBoV1 is an emerging pathogen that mainly causes respiratory tract infections, while the lack of cell lines suitable for culture replicative viruses hindered research on HBoV1. Here, we identify a permissive cell line for HBoV1 infection, MA104, and reveal that the complete life cycle of HBoV1 was supported in MA104 cells. Our findings provide a suitable cell model for the study of HBoV1 and explore its application for antiviral drug evaluation, which is vital for research on HBoV1 virology and pathogenesis, as well as for drug and vaccine development.
Collapse
Affiliation(s)
- Jielin Tang
- Guangzhou National Laboratory, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
| | - Sijie Chen
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yijun Deng
- Guangzhou National Laboratory, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
| | - Junjun Liu
- Guangzhou National Laboratory, Guangzhou, China
| | - Dan Huang
- Guangzhou National Laboratory, Guangzhou, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Muqing Fu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Bao Xue
- Guangzhou National Laboratory, Guangzhou, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Canyu Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Chunchen Wu
- Department of Laboratory Medicine, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Wang
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yuan Zhou
- Guangzhou National Laboratory, Guangzhou, China
| | - Qi Yang
- Guangzhou National Laboratory, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
| | - Xinwen Chen
- Guangzhou National Laboratory, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
3
|
Li C, Yu Y, Wan Z, Chiu MC, Huang J, Zhang S, Zhu X, Lan Q, Deng Y, Zhou Y, Xue W, Yue M, Cai JP, Yip CCY, Wong KKY, Liu X, Yu Y, Huang L, Chu H, Chan JFW, Clevers H, Yuen KY, Zhou J. Human respiratory organoids sustained reproducible propagation of human rhinovirus C and elucidation of virus-host interaction. Nat Commun 2024; 15:10772. [PMID: 39738014 PMCID: PMC11686133 DOI: 10.1038/s41467-024-55076-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 11/26/2024] [Indexed: 01/01/2025] Open
Abstract
The lack of a robust system to reproducibly propagate HRV-C, a family of viruses refractory to cultivation in standard cell lines, has substantially hindered our understanding of this common respiratory pathogen. We sought to develop an organoid-based system to reproducibly propagate HRV-C, and characterize virus-host interaction using respiratory organoids. We demonstrate that airway organoids sustain serial virus passage with the aid of CYT387-mediated immunosuppression, whereas nasal organoids that more closely simulate the upper airway achieve this without any intervention. Nasal organoids are more susceptible to HRV-C than airway organoids. Intriguingly, upon HRV-C infection, we observe an innate immune response that is stronger in airway organoids than in nasal organoids, which is reproduced in a Poly(I:C) stimulation assay. Treatment with α-CDHR3 and antivirals significantly reduces HRV-C viral growth in airway and nasal organoids. Additionally, an organoid-based immunofluorescence assay is established to titrate HRV-C infectious particles. Collectively, we develop an organoid-based system to reproducibly propagate the poorly cultivable HRV-C, followed by a comprehensive characterization of HRV-C infection and innate immunity in physiologically active respiratory organoids. The organoid-based HRV-C infection model can be extended for developing antiviral strategies. More importantly, our study has opened an avenue for propagating and studying other uncultivable human and animal viruses.
Collapse
Affiliation(s)
- Cun Li
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yifei Yu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Zhixin Wan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Man Chun Chiu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Jingjing Huang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Shuxin Zhang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xiaoxin Zhu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Qiaoshuai Lan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Yanlin Deng
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Ying Zhou
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Wei Xue
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ming Yue
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jian-Piao Cai
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Cyril Chik-Yan Yip
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kenneth Kak-Yuen Wong
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, and Queen Mary Hospital, Hong Kong, China
| | - Xiaojuan Liu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Yang Yu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | | | - Hin Chu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Jasper Fuk-Woo Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), and University Medical Center (UMC) Utrecht, Utrecht, the Netherlands
- Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Kwok Yung Yuen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jie Zhou
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China.
- BiomOrgan Ltd, Hong Kong, China.
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
4
|
Ning K, Zhao J, Feng Z, Park SY, McFarlin S, Cheng F, Yan Z, Wang J, Qiu J. N6-methyladenosine modification of a parvovirus-encoded small noncoding RNA facilitates viral DNA replication through recruiting Y-family DNA polymerases. Proc Natl Acad Sci U S A 2024; 121:e2320782121. [PMID: 38875150 PMCID: PMC11194592 DOI: 10.1073/pnas.2320782121] [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: 11/27/2023] [Accepted: 05/14/2024] [Indexed: 06/16/2024] Open
Abstract
Human bocavirus 1 (HBoV1) is a human parvovirus that causes lower respiratory tract infections in young children. It contains a single-stranded (ss) DNA genome of ~5.5 kb that encodes a small noncoding RNA of 140 nucleotides known as bocavirus-encoded small RNA (BocaSR), in addition to viral proteins. Here, we determined the secondary structure of BocaSR in vivo by using DMS-MaPseq. Our findings reveal that BocaSR undergoes N6-methyladenosine (m6A) modification at multiple sites, which is critical for viral DNA replication in both dividing HEK293 cells and nondividing cells of the human airway epithelium. Mechanistically, we found that m6A-modified BocaSR serves as a mediator for recruiting Y-family DNA repair DNA polymerase (Pol) η and Pol κ likely through a direct interaction between BocaSR and the viral DNA replication origin at the right terminus of the viral genome. Thus, this report represents direct involvement of a viral small noncoding RNA in viral DNA replication through m6A modification.
Collapse
Affiliation(s)
- Kang Ning
- Department of Microbiology, Molecular Genetics and Immunology, University of KansasMedical Center, Kansas City, KS66160
| | - Junxing Zhao
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS66045
- Section of Genetic Medicine, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL60637
| | - Zehua Feng
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA52242
| | - Soo Yeun Park
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA52242
| | - Shane McFarlin
- Department of Microbiology, Molecular Genetics and Immunology, University of KansasMedical Center, Kansas City, KS66160
| | - Fang Cheng
- Department of Microbiology, Molecular Genetics and Immunology, University of KansasMedical Center, Kansas City, KS66160
| | - Ziying Yan
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA52242
| | - Jingxin Wang
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS66045
- Section of Genetic Medicine, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL60637
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of KansasMedical Center, Kansas City, KS66160
| |
Collapse
|
5
|
Karaaslan C, Wirz O, Tan G, Globinska A, Boonpiyathad T, Hedman K, Vaselek S, Venermo MS, Jartti T, Akdis M, Akdis CA. B cell immune response to human bocaviruses. Clin Exp Allergy 2024; 54:388-401. [PMID: 38321724 DOI: 10.1111/cea.14453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 01/03/2024] [Accepted: 01/15/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Human bocaviruses (HBoVs) have been demonstrated in respiratory and gastrointestinal infections; however, the immune response to them has not been studied in detail. In this study, we investigated the B cell immune responses to HBoV1 and HBoV2, representing two different species of bocaviruses in humans. METHODS We analyzed the effects of stimulations with HBoV1 and 2 virus-like particles (VLPs) and of co-stimulation with HBoV1-rhinovirus (RV) on cells of the immune system by flow cytometry, transcriptomics, and luminometric immune assays. RESULTS Human B cells, and particularly B regulatory cells (Breg cells), showed an increased immune response to HBoV1-VLPs stimulation. These immune responses were also supported by increased IL-1RA and PDL1 expressions in IL-10+ B cells from peripheral blood mononuclear cells (PBMCs) stimulated with HBoV1-VLPs. In addition, increased levels of IL-10 and IL-1RA were determined in the supernatants of PBMCs following HBoV1-VLPs stimulation. HBoV1-VLPs and RV co-stimulation increased the IL-10+ B cell population. Transcriptome analysis by next-generation RNA sequencing showed an increased expression of IL-10 signalling and Breg cell markers in PBMCs stimulated with HBoV1-VLPs. Furthermore, TGF-β and chemoattractants MIP-1α, MIP-1β and IP10 protein levels were high in the supernatants of PBMCs stimulated with HBoV1-VLPs. CONCLUSIONS The findings demonstrate that in Breg cells, IL-10 signalling pathways, and anti-inflammatory activity are induced by HBoV1, which can explain the often mild nature of the disease. In addition, the immune regulatory response induced by HBoV1-VLPs may indicate a potential immunomodulatory role of HBoV1 on the immune system and may represent an immune regulatory strategy.
Collapse
Affiliation(s)
- Cagatay Karaaslan
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Molecular Biology Section, Biology Department, Faculty of Science, Hacettepe University, Ankara, Turkey
| | - Oliver Wirz
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ge Tan
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Anna Globinska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Tadech Boonpiyathad
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Klaus Hedman
- Department of Virology, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital Diagnostics Center, Helsinki, Finland
| | - Slavica Vaselek
- Molecular Biology Section, Biology Department, Faculty of Science, Hacettepe University, Ankara, Turkey
| | | | - Tuomas Jartti
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
6
|
Soldwedel S, Demuth S, Schildgen O. T84 Monolayer Cell Cultures Support Productive HBoV and HSV-1 Replication and Enable In Vitro Co-Infection Studies. Viruses 2024; 16:773. [PMID: 38793654 PMCID: PMC11125666 DOI: 10.3390/v16050773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Based on several clinical observations it was hypothesized that herpesviruses may influence the replication of human bocaviruses, the second known parvoviruses that have been confirmed as human pathogens. While several cell lines support the growth of HSV-1, HBoV-1 was exclusively cultivated on air-liquid interface cultures, the latter being a rather complicated, slow, and low throughput system. One of the cell lines are T84 cells, which are derived from the lung metastasis of a colorectal tumor. In this study, we provide evidence that T84 also supports HBoV replication when cultivated as monolayers, while simultaneously being permissive for HSV-1. The cell culture model thus would enable co-infection studies of both viruses and is worth being optimized for high throughput studies with HBoV-1. Additionally, the study provides evidence for a supporting effect of HSV-1 on the replication and packaging of HBoV-1 progeny DNA into DNase-resistant viral particles.
Collapse
Affiliation(s)
- Swen Soldwedel
- Kliniken der Stadt Köln, Institut für Pathologie, 51109 Köln/Cologne, Germany
| | - Sabrina Demuth
- Kliniken der Stadt Köln, Institut für Pathologie, 51109 Köln/Cologne, Germany
| | - Oliver Schildgen
- Institut für Pathologie, Klinikum der Privaten Universität Witten/Herdecke, Ostmerheimer Str. 200, 51109 Köln/Cologne, Germany
| |
Collapse
|
7
|
Peng L, Yang F, Shi J, Pan L, Liu Y, Mao D, Luo Y. Molecular characterization of human bocavirus in municipal wastewaters using amplicon target sequencing. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 918:170674. [PMID: 38316309 DOI: 10.1016/j.scitotenv.2024.170674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/22/2024] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
Human bocavirus (HBoV) is an emerging health concern worldwide, associated with range of clinical manifestations, including gastroenteritis and respiratory infections. Therefore, it is crucial to comprehend and minimize their prevalence in different systems. In this study, we conducted regular sampling throughout the year in two different sizes and work processes of wastewater treatment plants (WWTPs) in Tianjin, China. Our objective was to investigate the occurrence, prevalence, and endurance of HBoV in wastewater, while also evaluating the efficacy of amplicon target sequencing in directly detecting HBoV in wastewater. At two WWTPs, HBoV2 (45.51 %-45.67 %) and HBoV3 (38.30 %-40.25 %) were the most common genotypes identified, and the mean concentration range of HBoV was 2.54-7.40 log10 equivalent copies/l as determined by multiplex real-time quantitative PCR assay. A positive rate of HBoV was found in 96.6 % (29/30) samples of A-WWTP, and 96.6 % (26/27) samples of B-WWTP. The phylogenetic analysis indicated that the nucleotide similarity between the HBoV DNA sequences to the reference HBoV sequences published globally ranged from 90.14 %-100 %. A significant variation in the read abundance of HBoV2 and HBoV3 in two wastewater treatment plants (p < 0.05) was detected, specifically in the Winter and Summer seasons. The findings revealed a strong correlation between the genotypes detected in wastewater and the clinical data across various regions in China. In addition, it is worth mentioning that HBoV4 was exclusively detected in wastewater and not found in the clinical samples from patients. This study highlights the high prevalence of human bocavirus in municipal wastewater. This finding illustrates that amplicon target sequencing can amplify a wide variety of viruses, enabling the identification of newly discovered viruses.
Collapse
Affiliation(s)
- Liang Peng
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin 300071, China
| | - Fengxia Yang
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin 300191, China.
| | - Jingliang Shi
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin 300071, China
| | - Liuzhu Pan
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Yixin Liu
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Daqing Mao
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Yi Luo
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin 300071, China; State Key Laboratory of Pollution Control and Resource reuse, School of the Environment, Nanjing university, Nanjing 210093, China.
| |
Collapse
|
8
|
Cerato JA, da Silva EF, Porto BN. Breaking Bad: Inflammasome Activation by Respiratory Viruses. BIOLOGY 2023; 12:943. [PMID: 37508374 PMCID: PMC10376673 DOI: 10.3390/biology12070943] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023]
Abstract
The nucleotide-binding domain leucine-rich repeat-containing receptor (NLR) family is a group of intracellular sensors activated in response to harmful stimuli, such as invading pathogens. Some NLR family members form large multiprotein complexes known as inflammasomes, acting as a platform for activating the caspase-1-induced canonical inflammatory pathway. The canonical inflammasome pathway triggers the secretion of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18 by the rapid rupture of the plasma cell membrane, subsequently causing an inflammatory cell death program known as pyroptosis, thereby halting viral replication and removing infected cells. Recent studies have highlighted the importance of inflammasome activation in the response against respiratory viral infections, such as influenza and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While inflammasome activity can contribute to the resolution of respiratory virus infections, dysregulated inflammasome activity can also exacerbate immunopathology, leading to tissue damage and hyperinflammation. In this review, we summarize how different respiratory viruses trigger inflammasome pathways and what harmful effects the inflammasome exerts along with its antiviral immune response during viral infection in the lungs. By understanding the crosstalk between invading pathogens and inflammasome regulation, new therapeutic strategies can be exploited to improve the outcomes of respiratory viral infections.
Collapse
Affiliation(s)
- Julia A. Cerato
- Department of Medical Microbiology and Infectious Diseases, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (J.A.C.); (E.F.d.S.)
| | - Emanuelle F. da Silva
- Department of Medical Microbiology and Infectious Diseases, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (J.A.C.); (E.F.d.S.)
| | - Barbara N. Porto
- Department of Medical Microbiology and Infectious Diseases, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (J.A.C.); (E.F.d.S.)
- Biology of Breathing Group, Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
9
|
De R, Zhang KX, Wang F, Zhou YT, Sun Y, Chen DM, Zhu RN, Guo Q, Liu S, Qu D, Qian Y, Zhao LQ. Human bocavirus 1 is a genuine pathogen for acute respiratory tract infection in pediatric patients determined by nucleic acid, antigen, and serology tests. Front Microbiol 2022; 13:932858. [PMID: 35966673 PMCID: PMC9372409 DOI: 10.3389/fmicb.2022.932858] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/28/2022] [Indexed: 11/24/2022] Open
Abstract
Background Human bocavirus 1 (HBoV1), first discovered in 2005, was positive in symptomatic and healthy children and co-detected with other respiratory viruses. It is a long journey to decisively demonstrate the unique viral pathogenic function of acute respiratory tract infection (ARTI) in pediatric patients. Methods Respiratory specimens collected from pediatric patients with ARTI from January 2017 to December 2021 were screened by a capillary electrophoresis-based multiplex PCR (CEMP) assay, then genotyped by PCR and sequencing for HBoV1. For the antigen test, a part of HBoV1 DNA positive nasopharyngeal aspirates (NPAs) was used as an antigen, while a rabbit anti-HBoV1 DR2 specific to HBoV1 was used as an antibody in the indirect-immunofluorescence assay (IFA). Finally, the levels of IgG specific to HBoV1 in acute and convalescent sera selected retrospectively from only HBoV1 DNA-positive patients were evaluated by IFA. Results Among 9,899 specimens, 681 were positive for HBoV1 DNA (6.88%, 681/9899), which included 336 positives only for HBoV1 (49.34%, 336/681) and 345 (50.66%, 345/681) positives also for other pathogens. In the antigen test, there were 37 among 47 NPAs determined as HBoV1 antigen-positive (78.72%, 37/47), including 18 (48.65%, 18/37) positives solely for HBoV1 DNA. Among 4 pediatric patients with both acute and convalescent sera, there was one positive for HBoV1 antigen (D8873) and 2 lack the antigen results (D1474 and D10792), which showed seroconversion with a ≥ 4-fold increase in IgG levels. Conclusions The combination results of nucleic acid, antigen, and serology tests answered that HBoV1 is a genuine pathogen for ARTI in pediatric patients.
Collapse
Affiliation(s)
- Ri De
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Ke-Xiang Zhang
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Fang Wang
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, China
| | - Yu-Tong Zhou
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, China
| | - Yu Sun
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, China
| | - Dong-Mei Chen
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, China
| | - Ru-Nan Zhu
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, China
| | - Qi Guo
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, China
| | - Shuang Liu
- Department of Intensive Care Unit, Affiliated Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Dong Qu
- Department of Intensive Care Unit, Affiliated Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Yuan Qian
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, China
| | - Lin-Qing Zhao
- Laboratory of Virology, Beijing Key Laboratory of Etiology of Viral Diseases in Children, Capital Institute of Pediatrics, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
- *Correspondence: Lin-Qing Zhao
| |
Collapse
|
10
|
Chitcharoen S, Sivapornnukul P, Payungporn S. Revolutionized virome research using systems microbiology approaches. Exp Biol Med (Maywood) 2022; 247:1135-1147. [PMID: 35723062 PMCID: PMC9335507 DOI: 10.1177/15353702221102895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Currently, both pathogenic and commensal viruses are continuously being discovered and acknowledged as ubiquitous components of microbial communities. The advancements of systems microbiological approaches have changed the face of virome research. Here, we focus on viral metagenomic approach to study virus community and their interactions with other microbial members as well as their hosts. This review also summarizes challenges, limitations, and benefits of the current virome approaches. Potentially, the studies of virome can be further applied in various biological and clinical fields.
Collapse
Affiliation(s)
- Suwalak Chitcharoen
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand,Research Unit of Systems Microbiology, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pavaret Sivapornnukul
- Research Unit of Systems Microbiology, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand,Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sunchai Payungporn
- Research Unit of Systems Microbiology, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand,Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand,Sunchai Payungporn.
| |
Collapse
|
11
|
Mostafa-Hedeab G, Allayeh AK, Elhady HA, Eledrdery AY, Mraheil MA, Mostafa A. Viral Eco-Genomic Tools: Development and Implementation for Aquatic Biomonitoring. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:7707. [PMID: 35805367 PMCID: PMC9265447 DOI: 10.3390/ijerph19137707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 12/17/2022]
Abstract
Enteric viruses (EVs) occurrence within aquatic environments varies and leads to significant risk on public health of humans, animals, and diversity of aquatic taxa. Early and efficacious recognition of cultivable and fastidious EVs in aquatic systems are important to ensure the sanitary level of aquatic water and implement required treatment strategies. Herein, we provided a comprehensive overview of the conventional and up-to-date eco-genomic tools for aquatic biomonitoring of EVs, aiming to develop better water pollution monitoring tools. In combination with bioinformatics techniques, genetic tools including cloning sequencing analysis, DNA microarray, next-generation sequencing (NGS), and metagenomic sequencing technologies are implemented to make informed decisions about the global burden of waterborne EVs-associated diseases. The data presented in this review are helpful to recommend that: (1) Each viral pollution detection method has its own merits and demerits; therefore, it would be advantageous for viral pollution evaluation to be integrated as a complementary platform. (2) The total viral genome pool extracted from aquatic environmental samples is a real reflection of pollution status of the aquatic eco-systems; therefore, it is recommended to conduct regular sampling through the year to establish an updated monitoring system for EVs, and quantify viral peak concentrations, viral typing, and genotyping. (3) Despite that conventional detection methods are cheaper, it is highly recommended to implement molecular-based technologies to complement aquatic ecosystems biomonitoring due to numerous advantages including high-throughput capability. (4) Continuous implementation of the eco-genetic detection tools for monitoring the EVs in aquatic ecosystems is recommended.
Collapse
Affiliation(s)
- Gomaa Mostafa-Hedeab
- Pharmacology Department and Health Research Unit, Medical College, Jouf University, Skaka 11564, Saudi Arabia
| | - Abdou Kamal Allayeh
- Water Pollution Department, Virology Laboratory, National Research Centre, Dokki, Giza 12622, Egypt;
| | | | - Abozer Y. Eledrdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 11564, Saudi Arabia;
| | - Mobarak Abu Mraheil
- German Center for Infection Research (DZIF), Institute of Medical Microbiology, Justus-Liebig University, 35392 Giessen, Germany
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| |
Collapse
|
12
|
The small nonstructural protein NP1 of human bocavirus 1 directly interacts with Ku70 and RPA70 and facilitates viral DNA replication. PLoS Pathog 2022; 18:e1010578. [PMID: 35653410 PMCID: PMC9197078 DOI: 10.1371/journal.ppat.1010578] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/14/2022] [Accepted: 05/10/2022] [Indexed: 11/19/2022] Open
Abstract
Human bocavirus 1 (HBoV1), a member of the genus Bocaparvovirus of the family Parvoviridae, causes acute respiratory tract infections in young children. Well-differentiated pseudostratified human airway epithelium cultured at an air-liquid interface (HAE-ALI) is an ideal in vitro culture model to study HBoV1 infection. Unique to other parvoviruses, bocaparvoviruses express a small nonstructured protein NP1 of ~25 kDa from an open reading frame (ORF) in the center of the viral genome. NP1 plays an important role in viral DNA replication and pre-mRNA processing. In this study, we performed an affinity purification assay to identify HBoV1 NP1-inteacting proteins. We identified that Ku70 and RPA70 directly interact with the NP1 at a high binding affinity, characterized with an equilibrium dissociation constant (KD) of 95 nM and 122 nM, respectively. Furthermore, we mapped the key NP1-interacting domains of Ku70 at aa266-439 and of RPA70 at aa181-422. Following a dominant negative strategy, we revealed that the interactions of Ku70 and RPA70 with NP1 play a significant role in HBoV1 DNA replication not only in an in vitro viral DNA replication assay but also in HBoV1-infected HAE-ALI cultures. Collectively, our study revealed a novel mechanism by which HBoV1 NP1 enhances viral DNA replication through its direct interactions with Ku70 and RPA70.
Collapse
|
13
|
Shao L, Ning K, Wang J, Cheng F, Wang S, Qiu J. The Large Nonstructural Protein (NS1) of Human Bocavirus 1 Directly Interacts with Ku70, Which Plays an Important Role in Virus Replication in Human Airway Epithelia. J Virol 2022; 96:e0184021. [PMID: 34878919 PMCID: PMC8865542 DOI: 10.1128/jvi.01840-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/22/2021] [Indexed: 12/16/2022] Open
Abstract
Human bocavirus 1 (HBoV1), an autonomous human parvovirus, causes acute respiratory tract infections in young children. HBoV1 infects well-differentiated (polarized) human airway epithelium cultured at an air-liquid interface (HAE-ALI). HBoV1 expresses a large nonstructural protein, NS1, that is essential for viral DNA replication. HBoV1 infection of polarized human airway epithelial cells induces a DNA damage response (DDR) that is critical to viral DNA replication involving DNA repair with error-free Y-family DNA polymerases. HBoV1 NS1 or the isoform NS1-70 per se induces a DDR. In this study, using the second-generation proximity-dependent biotin identification (BioID2) approach, we identified that Ku70 is associated with the NS1-BioID2 pulldown complex through a direct interaction with NS1. Biolayer interferometry (BLI) assay determined a high binding affinity of NS1 with Ku70, which has an equilibrium dissociation constant (KD) value of 0.16 μM and processes the strongest interaction at the C-terminal domain. The association of Ku70 with NS1 was also revealed during HBoV1 infection of HAE-ALI. Knockdown of Ku70 and overexpression of the C-terminal domain of Ku70 significantly decreased HBoV1 replication in HAE-ALI. Thus, our study provides, for the first time, a direct interaction of parvovirus large nonstructural protein NS1 with Ku70. IMPORTANCE Parvovirus infection induces a DNA damage response (DDR) that plays a pivotal role in viral DNA replication. The DDR includes activation of ATM (ataxia telangiectasia mutated), ATR (ATM- and RAD3-related), and DNA-PKcs (DNA-dependent protein kinase catalytic subunit). The large nonstructural protein (NS1) often plays a role in the induction of DDR; however, how the DDR is induced during parvovirus infection or simply by the NS1 is not well studied. Activation of DNA-PKcs has been shown as one of the key DDR pathways in DNA replication of HBoV1. We identified that HBoV1 NS1 directly interacts with Ku70, but not Ku80, of the Ku70/Ku80 heterodimer at high affinity. This interaction is also important for HBoV1 replication in HAE-ALI. We propose that the interaction of NS1 with Ku70 recruits the Ku70/Ku80 complex to the viral DNA replication center, which activates DNA-PKcs and facilitates viral DNA replication.
Collapse
Affiliation(s)
- Liting Shao
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Kang Ning
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jianke Wang
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Fang Cheng
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Shengqi Wang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
14
|
Thirteen Nearly Complete Genome Sequences of Human Bocavirus 1 Isolated from Pediatric Inpatients in Fukushima, Japan. Microbiol Resour Announc 2022; 11:e0102721. [PMID: 35049344 PMCID: PMC8772594 DOI: 10.1128/mra.01027-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We report 13 genomic sequences of human bocavirus 1 isolated from pediatric inpatients in Fukushima, Japan, using an air-liquid interface culture of human bronchial tracheal epithelial cells. This work suggests the endemic circulation of a human bocavirus variant with a unique amino acid signature in Fukushima.
Collapse
|
15
|
Abstract
Pandemics caused by respiratory viruses have impacted millions of lives and caused massive destruction to global infrastructure. With their emergence, it has become a priority to develop platforms to rapidly dissect host/pathogen interactions, develop diagnostics, and evaluate therapeutics. Traditional viral culture methods do not faithfully recapitulate key aspects of infection. Tissue engineering as a discipline has developed techniques to produce three-dimensional human tissues which can serve as platforms to study respiratory viruses in vitro. In this chapter, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has been used as a representative respiratory virus motivating the use of tissue engineering to generate in vitro culture models. SARS-CoV-2 pathophysiology, traditional cell culture, tissue engineering-based cell culture, and future directions for the field are highlighted.
Collapse
|
16
|
Sang Y, Miller LC, Nelli RK, Giménez-Lirola LG. Harness Organoid Models for Virological Studies in Animals: A Cross-Species Perspective. Front Microbiol 2021; 12:725074. [PMID: 34603253 PMCID: PMC8481363 DOI: 10.3389/fmicb.2021.725074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/27/2021] [Indexed: 02/02/2023] Open
Abstract
Animal models and cell culture in vitro are primarily used in virus and antiviral immune research. Whereas the limitation of these models to recapitulate the viral pathogenesis in humans has been made well aware, it is imperative to introduce more efficient systems to validate emerging viruses in both domestic and wild animals. Organoids ascribe to representative miniatures of organs (i.e., mini-organs), which are derived from three-dimensional culture of stem cells under respective differential conditions mimicking endogenous organogenetic niches. Organoids have broadened virological studies in the human context, particularly in recent uses for COVID19 research. This review examines the status and potential for cross-species applied organotypic culture in validating emerging animal, particularly zoonotic, viruses in domestic and wild animals.
Collapse
Affiliation(s)
- Yongming Sang
- Department of Agricultural and Environmental Sciences, College of Agriculture, Tennessee State University, Nashville, TN, United States
| | - Laura C Miller
- Virus and Prion Research Unit, National Animal Disease Center, United States Department of Agriculture, Agricultural Research Service, Ames, IA, United States
| | - Rahul K Nelli
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Luis Gabriel Giménez-Lirola
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
17
|
Hairpin transfer-independent Parvovirus DNA Replication Produces Infectious Virus. J Virol 2021; 95:e0110821. [PMID: 34346761 DOI: 10.1128/jvi.01108-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Parvoviruses package a linear single-stranded DNA genome with hairpin structures at both ends. It has been thought that terminal hairpin sequences are indispensable for viral DNA replication. Here, we provide evidence that the hairpin-deleted duplex genomes of human bocavirus 1 (HBoV1) replicate in human embryonic kidney (HEK) 293 cells. We propose an alternative model for HBoV1 DNA replication in which the leading strand can initiate strand-displacement without "hairpin-transfer." The transfection of the HBoV1 duplex genomes that retain a minimal replication origin at the right-end (OriR), but with extensive deletions in the right-end hairpin (REH), generated viruses in HEK293 cells at a level 10-20 times lower than the wild-type (WT) duplex genome. Importantly, these viruses that have a genome with various deletions after the OriR, but not the one retaining only the OriR, replicated in polarized human airway epithelia. We discovered that the 18-nt sequence (nt 5,403-5,420) beyond the OriR was sufficient to confer virus replication in polarized human airway epithelia, although its progeny virus production was ∼5 times lower than that of the WT virus. Thus, our study demonstrates that hairpin transfer-independent productive parvovirus DNA replication can occur. Importance Hairpin transfer-independent parvovirus replication was modeled with human bocavirus 1 (HBoV1) duplex genomes whose 5' hairpin structure was ablated by various deletions. In HEK293 cells, these duplex viral genomes with ablated 5'/hairpin sequence replicated efficiently and generated viruses that productively infected polarized human airway epithelium. Thus, for the first time, we reveal a previously unknown phenomenon that the productive parvovirus DNA replication does not depend on the hairpin sequence at REH to initiate "rolling hairpin" DNA replication. Notably, the intermediates of viral DNA replication, as revealed two-dimensional electrophoresis, from transfections of hairpin sequence-deleted duplex genome and full-length genome in HEK293 cells, as well as from virus infection of polarized human airway epithelia are similar. Thus, the establishment of the hairpin transfer-independent parvoviral DNA replication deepens our understanding in viral DNA replication and may have implications in development of parvovirus-based viral vectors with alternative properties.
Collapse
|
18
|
Clementi N, Ghosh S, De Santis M, Castelli M, Criscuolo E, Zanoni I, Clementi M, Mancini N. Viral Respiratory Pathogens and Lung Injury. Clin Microbiol Rev 2021; 34:e00103-20. [PMID: 33789928 PMCID: PMC8142519 DOI: 10.1128/cmr.00103-20] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Several viruses target the human respiratory tract, causing different clinical manifestations spanning from mild upper airway involvement to life-threatening acute respiratory distress syndrome (ARDS). As dramatically evident in the ongoing SARS-CoV-2 pandemic, the clinical picture is not always easily predictable due to the combined effect of direct viral and indirect patient-specific immune-mediated damage. In this review, we discuss the main RNA (orthomyxoviruses, paramyxoviruses, and coronaviruses) and DNA (adenoviruses, herpesviruses, and bocaviruses) viruses with respiratory tropism and their mechanisms of direct and indirect cell damage. We analyze the thin line existing between a protective immune response, capable of limiting viral replication, and an unbalanced, dysregulated immune activation often leading to the most severe complication. Our comprehension of the molecular mechanisms involved is increasing and this should pave the way for the development and clinical use of new tailored immune-based antiviral strategies.
Collapse
Affiliation(s)
- Nicola Clementi
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
- Laboratory of Microbiology and Virology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sreya Ghosh
- Harvard Medical School, Boston Children's Hospital, Division of Immunology, Boston, Massachusetts, USA
| | - Maria De Santis
- Department of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| | - Matteo Castelli
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
| | - Elena Criscuolo
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
| | - Ivan Zanoni
- Harvard Medical School, Boston Children's Hospital, Division of Immunology, Boston, Massachusetts, USA
- Harvard Medical School, Boston Children's Hospital, Division of Gastroenterology, Boston, Massachusetts, USA
| | - Massimo Clementi
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
- Laboratory of Microbiology and Virology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicasio Mancini
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
- Laboratory of Microbiology and Virology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
19
|
Shao L, Shen W, Wang S, Qiu J. Recent Advances in Molecular Biology of Human Bocavirus 1 and Its Applications. Front Microbiol 2021; 12:696604. [PMID: 34220786 PMCID: PMC8242256 DOI: 10.3389/fmicb.2021.696604] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 05/12/2021] [Indexed: 11/25/2022] Open
Abstract
Human bocavirus 1 (HBoV1) was discovered in human nasopharyngeal specimens in 2005. It is an autonomous human parvovirus and causes acute respiratory tract infections in young children. HBoV1 infects well differentiated or polarized human airway epithelial cells in vitro. Unique among all parvoviruses, HBoV1 expresses 6 non-structural proteins, NS1, NS1-70, NS2, NS3, NS4, and NP1, and a viral non-coding RNA (BocaSR), and three structural proteins VP1, VP2, and VP3. The BocaSR is the first identified RNA polymerase III (Pol III) transcribed viral non-coding RNA in small DNA viruses. It plays an important role in regulation of viral gene expression and a direct role in viral DNA replication in the nucleus. HBoV1 genome replication in the polarized/non-dividing airway epithelial cells depends on the DNA damage and DNA repair pathways and involves error-free Y-family DNA repair DNA polymerase (Pol) η and Pol κ. Importantly, HBoV1 is a helper virus for the replication of dependoparvovirus, adeno-associated virus (AAV), in polarized human airway epithelial cells, and HBoV1 gene products support wild-type AAV replication and recombinant AAV (rAAV) production in human embryonic kidney (HEK) 293 cells. More importantly, the HBoV1 capsid is able to pseudopackage an rAAV2 or rHBoV1 genome, producing the rAAV2/HBoV1 or rHBoV1 vector. The HBoV1 capsid based rAAV vector has a high tropism for human airway epithelia. A deeper understanding in HBoV1 replication and gene expression will help find a better way to produce the rAAV vector and to increase the efficacy of gene delivery using the rAAV2/HBoV1 or rHBoV1 vector, in particular, to human airways. This review summarizes the recent advances in gene expression and replication of HBoV1, as well as the use of HBoV1 as a parvoviral vector for gene delivery.
Collapse
Affiliation(s)
- Liting Shao
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Weiran Shen
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Shengqi Wang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
20
|
Yan Z, Deng X, Qiu J. Human Bocavirus 1 Infection of Well-Differentiated Human Airway Epithelium. ACTA ACUST UNITED AC 2021; 58:e107. [PMID: 32639683 DOI: 10.1002/cpmc.107] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Human bocavirus 1 (HBoV1) is a small DNA virus that belongs to the Bocaparvovirus genus of the Parvoviridae family. HBoV1 is a common respiratory pathogen that causes mild to life-threatening acute respiratory tract infections in children and immunocompromised individuals, infecting both the upper and lower respiratory tracts. HBoV1 infection causes death of airway epithelial cells, resulting in airway injury and inflammation. In vitro, HBoV1 only infects well-differentiated (polarized) human airway epithelium cultured at an air-liquid interface (HAE-ALI), but not any dividing human cells. A full-length HBoV1 genome of 5543 nucleotides has been cloned from DNA extracted from a human nasopharyngeal swab into a plasmid called HBoV1 infectious clone pIHBoV1. Transfection of pIHBoV1 replicates efficiently in human embryonic kidney 293 (HEK293) cells and produces virions that are highly infectious. This article describes protocols for production of HBoV1 in HEK293 cells, generation of HAE-ALI cultures, and infection with HBoV1 in HAE-ALI. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Human bocavirus 1 production in HEK293 cells Support Protocol 1: HEK293 cell culture and transfection Support Protocol 2: Quantification of human bocavirus 1 using real-time quantitative PCR Basic Protocol 2: Differentiation of human airway cells at an air-liquid interface Support Protocol 3: Expansion of human airway epithelial cell line CuFi-8 Support Protocol 4: Expansion of human airway basal cells Support Protocol 5: Coating of plastic dishes and permeable membranes of inserts Support Protocol 6: Transepithelial electrical resistance measurement Basic Protocol 3: Human bocavirus 1 infection in human airway epithelium cultured at an air-liquid interface Support Protocol 7: Isolation of infected human airway epithelium cells from inserts Basic Protocol 4: Transduction of airway basal cells with lentiviral vector.
Collapse
Affiliation(s)
- Ziying Yan
- Department of Anatomy, University of Iowa, Iowa City, Iowa
| | - Xuefeng Deng
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
21
|
Yu JC, Mietzsch M, Singh A, Jimenez Ybargollin A, Kailasan S, Chipman P, Bhattacharya N, Fakhiri J, Grimm D, Kapoor A, Kučinskaitė-Kodzė I, Žvirblienė A, Söderlund-Venermo M, McKenna R, Agbandje-McKenna M. Characterization of the GBoV1 Capsid and Its Antibody Interactions. Viruses 2021; 13:v13020330. [PMID: 33672786 PMCID: PMC7924616 DOI: 10.3390/v13020330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 12/14/2022] Open
Abstract
Human bocavirus 1 (HBoV1) has gained attention as a gene delivery vector with its ability to infect polarized human airway epithelia and 5.5 kb genome packaging capacity. Gorilla bocavirus 1 (GBoV1) VP3 shares 86% amino acid sequence identity with HBoV1 but has better transduction efficiency in several human cell types. Here, we report the capsid structure of GBoV1 determined to 2.76 Å resolution using cryo-electron microscopy (cryo-EM) and its interaction with mouse monoclonal antibodies (mAbs) and human sera. GBoV1 shares capsid surface morphologies with other parvoviruses, with a channel at the 5-fold symmetry axis, protrusions surrounding the 3-fold axis and a depression at the 2-fold axis. A 2/5-fold wall separates the 2-fold and 5-fold axes. Compared to HBoV1, differences are localized to the 3-fold protrusions. Consistently, native dot immunoblots and cryo-EM showed cross-reactivity and binding, respectively, by a 5-fold targeted HBoV1 mAb, 15C6. Surprisingly, recognition was observed for one out of three 3-fold targeted mAbs, 12C1, indicating some structural similarity at this region. In addition, GBoV1, tested against 40 human sera, showed the similar rates of seropositivity as HBoV1. Immunogenic reactivity against parvoviral vectors is a significant barrier to efficient gene delivery. This study is a step towards optimizing bocaparvovirus vectors with antibody escape properties.
Collapse
Affiliation(s)
- Jennifer Chun Yu
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.C.Y.); (M.M.); (A.S.); (A.J.Y.); (S.K.); (P.C.); (R.M.)
| | - Mario Mietzsch
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.C.Y.); (M.M.); (A.S.); (A.J.Y.); (S.K.); (P.C.); (R.M.)
| | - Amriti Singh
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.C.Y.); (M.M.); (A.S.); (A.J.Y.); (S.K.); (P.C.); (R.M.)
| | - Alberto Jimenez Ybargollin
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.C.Y.); (M.M.); (A.S.); (A.J.Y.); (S.K.); (P.C.); (R.M.)
| | - Shweta Kailasan
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.C.Y.); (M.M.); (A.S.); (A.J.Y.); (S.K.); (P.C.); (R.M.)
| | - Paul Chipman
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.C.Y.); (M.M.); (A.S.); (A.J.Y.); (S.K.); (P.C.); (R.M.)
| | - Nilakshee Bhattacharya
- Biological Science Imaging Resource, Department of Biological Sciences, Florida State University, Tallahassee, FL 32306, USA;
| | - Julia Fakhiri
- Department of Infectious Diseases/Virology, Medical Faculty, BioQuant, University of Heidelberg, 69120 Heidelberg, Germany; (J.F.); (D.G.)
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Medical Faculty, BioQuant, University of Heidelberg, 69120 Heidelberg, Germany; (J.F.); (D.G.)
| | - Amit Kapoor
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43220, USA;
| | - Indrė Kučinskaitė-Kodzė
- Department of Immunology and Cell Biology of the Institute of Biotechnology of Vilnius University, 10257 Vilnius, Lithuania; (I.K.-K.); (A.Ž.)
| | - Aurelija Žvirblienė
- Department of Immunology and Cell Biology of the Institute of Biotechnology of Vilnius University, 10257 Vilnius, Lithuania; (I.K.-K.); (A.Ž.)
| | | | - Robert McKenna
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.C.Y.); (M.M.); (A.S.); (A.J.Y.); (S.K.); (P.C.); (R.M.)
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.C.Y.); (M.M.); (A.S.); (A.J.Y.); (S.K.); (P.C.); (R.M.)
- Correspondence:
| |
Collapse
|
22
|
Xu M, Perdomo MF, Mattola S, Pyöriä L, Toppinen M, Qiu J, Vihinen-Ranta M, Hedman K, Nokso-Koivisto J, Aaltonen LM, Söderlund-Venermo M. Persistence of Human Bocavirus 1 in Tonsillar Germinal Centers and Antibody-Dependent Enhancement of Infection. mBio 2021; 12:e03132-20. [PMID: 33531399 PMCID: PMC7858059 DOI: 10.1128/mbio.03132-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/08/2020] [Indexed: 12/11/2022] Open
Abstract
Human bocavirus 1 (HBoV1), a nonenveloped single-stranded DNA parvovirus, causes mild to life-threatening respiratory tract infections, acute otitis media, and encephalitis in young children. HBoV1 often persists in nasopharyngeal secretions for months, hampering diagnosis. It has also been shown to persist in pediatric palatine and adenoid tonsils, which suggests that lymphoid organs are reservoirs for virus spread; however, the tissue site and host cells remain unknown. Our aim was to determine, in healthy nonviremic children with preexisting HBoV1 immunity, the adenotonsillar persistence site(s), host cell types, and virus activity. We discovered that HBoV1 DNA persists in lymphoid germinal centers (GCs), but not in the corresponding tonsillar epithelium, and that the cell types harboring the virus are mainly naive, activated, and memory B cells and monocytes. Both viral DNA strands and both sides of the genome were detected, as well as infrequent mRNA. Moreover, we showed, in B-cell and monocyte cultures and ex vivo tonsillar B cells, that the cellular uptake of HBoV1 occurs via the Fc receptor (FcγRII) through antibody-dependent enhancement (ADE). This resulted in viral mRNA transcription, known to occur exclusively from double-stranded DNA in the nucleus, however, with no detectable productive replication. Confocal imaging with fluorescent virus-like particles moreover disclosed endocytosis. To which extent the active HBoV1 GC persistence has a role in chronic inflammation or B-cell maturation disturbances, and whether the virus can be reactivated, will be interesting topics for forthcoming studies.IMPORTANCE Human bocavirus 1 (HBoV1), a common pediatric respiratory pathogen, can persist in airway secretions for months hampering diagnosis. It also persists in tonsils, providing potential reservoirs for airway shedding, with the exact location, host cell types, and virus activity unknown. Our study provides new insights into tonsillar HBoV1 persistence. We observed HBoV1 persistence exclusively in germinal centers where immune maturation occurs, and the main host cells were B cells and monocytes. In cultured cell lines and primary tonsillar B cells, we showed the virus uptake to be significantly enhanced by HBoV1-specific antibodies, mediated by the cellular IgG receptor, leading to viral mRNA synthesis, but without detectable productive replication. Possible implications of such active viral persistence could be tonsillar inflammation, disturbances in immune maturation, reactivation, or cell death with release of virus DNA, explaining the long-lasting HBoV1 airway shedding.
Collapse
Affiliation(s)
- Man Xu
- Department of Virology, University of Helsinki, Helsinki, Finland
| | | | - Salla Mattola
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
- Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Lari Pyöriä
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Mari Toppinen
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Maija Vihinen-Ranta
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
- Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Klaus Hedman
- Department of Virology, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital, Helsinki, Finland
| | - Johanna Nokso-Koivisto
- Department of Otorhinolaryngology-Head and Neck Surgery, Helsinki University Hospital, Helsinki, Finland
- University of Helsinki, Helsinki, Finland
| | - Leena-Maija Aaltonen
- Department of Otorhinolaryngology-Head and Neck Surgery, Helsinki University Hospital, Helsinki, Finland
- University of Helsinki, Helsinki, Finland
| | | |
Collapse
|
23
|
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
|
24
|
Sridhar A, Simmini S, Ribeiro CMS, Tapparel C, Evers MM, Pajkrt D, Wolthers K. A Perspective on Organoids for Virology Research. Viruses 2020; 12:E1341. [PMID: 33238561 PMCID: PMC7700289 DOI: 10.3390/v12111341] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/12/2020] [Accepted: 11/22/2020] [Indexed: 12/27/2022] Open
Abstract
Animal models and cell lines are invaluable for virology research and host-pathogen interaction studies. However, it is increasingly evident that these models are not sufficient to fully understand human viral diseases. With the advent of three-dimensional organotypic cultures, it is now possible to study viral infections in the human context. This perspective explores the potential of these organotypic cultures, also known as organoids, for virology research, antiviral testing, and shaping the virology landscape.
Collapse
Affiliation(s)
- Adithya Sridhar
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1100 AZ Amsterdam, The Netherlands; (A.S.); (D.P.)
- Department of Pediatric Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1100 AZ Amsterdam, The Netherlands
| | - Salvatore Simmini
- Gastrointestinal Biology Group, STEMCELL Technologies UK Ltd., Cambridge CB28 9TL, UK;
| | - Carla M. S. Ribeiro
- Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, 1100 AZ Amsterdam, The Netherlands;
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
- Division of Infectious Diseases, Geneva University Hospital, 1205 Geneva, Switzerland
| | - Melvin M. Evers
- Department of Research and Development, uniQure Biopharma B.V., 1105 BE Amsterdam, The Netherlands;
| | - Dasja Pajkrt
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1100 AZ Amsterdam, The Netherlands; (A.S.); (D.P.)
- Department of Pediatric Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1100 AZ Amsterdam, The Netherlands
| | - Katja Wolthers
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1100 AZ Amsterdam, The Netherlands; (A.S.); (D.P.)
| |
Collapse
|
25
|
Cao X, Coyle JP, Xiong R, Wang Y, Heflich RH, Ren B, Gwinn WM, Hayden P, Rojanasakul L. Invited review: human air-liquid-interface organotypic airway tissue models derived from primary tracheobronchial epithelial cells-overview and perspectives. In Vitro Cell Dev Biol Anim 2020; 57:104-132. [PMID: 33175307 PMCID: PMC7657088 DOI: 10.1007/s11626-020-00517-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023]
Abstract
The lung is an organ that is directly exposed to the external environment. Given the large surface area and extensive ventilation of the lung, it is prone to exposure to airborne substances, such as pathogens, allergens, chemicals, and particulate matter. Highly elaborate and effective mechanisms have evolved to protect and maintain homeostasis in the lung. Despite these sophisticated defense mechanisms, the respiratory system remains highly susceptible to environmental challenges. Because of the impact of respiratory exposure on human health and disease, there has been considerable interest in developing reliable and predictive in vitro model systems for respiratory toxicology and basic research. Human air-liquid-interface (ALI) organotypic airway tissue models derived from primary tracheobronchial epithelial cells have in vivo–like structure and functions when they are fully differentiated. The presence of the air-facing surface allows conducting in vitro exposures that mimic human respiratory exposures. Exposures can be conducted using particulates, aerosols, gases, vapors generated from volatile and semi-volatile substances, and respiratory pathogens. Toxicity data have been generated using nanomaterials, cigarette smoke, e-cigarette vapors, environmental airborne chemicals, drugs given by inhalation, and respiratory viruses and bacteria. Although toxicity evaluations using human airway ALI models require further standardization and validation, this approach shows promise in supplementing or replacing in vivo animal models for conducting research on respiratory toxicants and pathogens.
Collapse
Affiliation(s)
- Xuefei Cao
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR Rd., AR, Jefferson, USA.
| | - Jayme P Coyle
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - Rui Xiong
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR Rd., AR, Jefferson, USA
| | - Yiying Wang
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR Rd., AR, Jefferson, USA
| | - Robert H Heflich
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR Rd., AR, Jefferson, USA
| | - Baiping Ren
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR Rd., AR, Jefferson, USA
| | - William M Gwinn
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Durham, NC, USA
| | | | - Liying Rojanasakul
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| |
Collapse
|
26
|
Liu X, Mondal AM. Conditional cell reprogramming for modeling host-virus interactions and human viral diseases. J Med Virol 2020; 92:2440-2452. [PMID: 32478897 PMCID: PMC7586785 DOI: 10.1002/jmv.26093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 01/08/2023]
Abstract
Conventional cancer and transformed cell lines are widely used in cancer biology and other fields within biology. These cells usually have abnormalities from the original tumor itself, but may also develop abnormalities due to genetic manipulation, or genetic and epigenetic changes during long-term passages. Primary cultures may maintain lineage functions as the original tissue types, yet they have a very limited life span or population doubling time because of the nature of cellular senescence. Primary cultures usually have very low yields, and the high variability from any original tissue specimens, largely limiting their applications in research. Animal models are often used for studies of virus infections, disease modeling, development of antiviral drugs, and vaccines. Human viruses often need a series of passages in vivo to adapt to the host environment because of variable receptors on the cell surface and may have intracellular restrictions from the cell types or host species. Here, we describe a long-term cell culture system, conditionally reprogrammed cells (CRCs), and its applications in modeling human viral diseases and drug discovery. Using feeder layer coculture in presence of Y-27632 (conditional reprogramming, CR), CRCs can be obtained and rapidly propagated from surgical specimens, core or needle biopsies, and other minimally invasive or noninvasive specimens, for example, nasal cavity brushing. CRCs preserve their lineage functions and provide biologically relevant and physiological conditions, which are suitable for studies of viral entry and replication, innate immune responses of host cells, and discovery of antiviral drugs. In this review, we summarize the applications of CR technology in modeling host-virus interactions and human viral diseases including severe acute respiratory syndrome coronavirus-2 and coronavirus disease-2019, and antiviral discovery.
Collapse
Affiliation(s)
- Xuefeng Liu
- Department of Pathology, Center for Cell ReprogrammingGeorgetown University Medical CenterWashingtonDC
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| | - Abdul M. Mondal
- Department of Pathology, Center for Cell ReprogrammingGeorgetown University Medical CenterWashingtonDC
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| |
Collapse
|
27
|
Bagasi AA, Howson-Wells HC, Clark G, Tarr AW, Soo S, Irving WL, McClure CP. Human Bocavirus infection and respiratory tract disease identified in a UK patient cohort. J Clin Virol 2020; 129:104453. [PMID: 32534437 PMCID: PMC7240277 DOI: 10.1016/j.jcv.2020.104453] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 12/20/2022]
Abstract
Human Bocavirus 1 (HBoV1) was commonly detected in a survey of circa 13,000 UK respiratory samples between 2015 and 2019. Co-infection with other viruses was observed in approximately three quarters of samples. However, mono-infection was also prevalent, and associated with clinically relevant disease. Intensive care was required in 31% of HBoV1 mono-infections and ventilation in 17%. Fatal multi-organ failure was observed in an apparently HBoV1 mono-infected and otherwise healthy child.
Background Since its first isolation in 2005, Human Bocavirus (HBoV) has been repeatedly associated with acute respiratory tract infections, although its role in pathogenicity remains unclear due to high co-infection rates. Objectives To assess HBoV prevalence and associated disease in a cohort of respiratory patients in the East Midlands, UK between 2015 and 2019. Study design We initially investigated the undiagnosed burden of HBoV in a retrospective paediatric cohort sampled between 2015 and 2017 using an in-house PCR assay. HBoV was subsequently incorporated into the standard respiratory diagnostic pathway and we audited a calendar year of HBoV positive results between 2018 and 2019. Results Our retrospective PCR screening of previously routine diagnostic-negative samples from juvenile patients identified a 9% (n = 30) prevalence of HBoV type 1. These apparent HBoV1 mono-infections were frequently associated with respiratory tract symptoms, often severe requiring ventilation, oxygen and steroid intervention with 31% (n = 9) of individuals requiring intensive care. When HBoV screening was subsequently adopted into the routine respiratory diagnostic pathway, year-round infections were observed in both children and adults peaking in February. 185 of 9098 (2.03%) individuals were found to be HBoV positive with children aged 12–24 months the principally infected group. However, HBoV infection was also observed in patients aged over 60, predominantly as a mono-infection. 23% of the 185 unique patients were HBoV monoinfected and persistent low-level DNA positivity was observed in 15 individuals up to 6-months after initial presentation. Conclusion HBoV1 is a prevalent respiratory infection in the UK capable of causing serious monoinfections.
Collapse
Affiliation(s)
- Arwa A Bagasi
- King Saud University, Riyadh, 11451, Saudi Arabia; NIHR Nottingham Digestive Diseases Biomedical Research Centre and School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Hannah C Howson-Wells
- Clinical Microbiology, Nottingham University Hospitals NHS Trust, Nottingham, NG7 2UH, UK
| | - Gemma Clark
- Clinical Microbiology, Nottingham University Hospitals NHS Trust, Nottingham, NG7 2UH, UK
| | - Alexander W Tarr
- NIHR Nottingham Digestive Diseases Biomedical Research Centre and School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Shiu Soo
- Clinical Microbiology, Nottingham University Hospitals NHS Trust, Nottingham, NG7 2UH, UK
| | - William L Irving
- NIHR Nottingham Digestive Diseases Biomedical Research Centre and School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK; Clinical Microbiology, Nottingham University Hospitals NHS Trust, Nottingham, NG7 2UH, UK
| | - C Patrick McClure
- NIHR Nottingham Digestive Diseases Biomedical Research Centre and School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK.
| |
Collapse
|
28
|
Establishment of a Recombinant AAV2/HBoV1 Vector Production System in Insect Cells. Genes (Basel) 2020; 11:genes11040439. [PMID: 32316599 PMCID: PMC7231168 DOI: 10.3390/genes11040439] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/10/2020] [Accepted: 04/15/2020] [Indexed: 12/13/2022] Open
Abstract
We have previously developed an rAAV2/HBoV1 vector in which a recombinant adeno-associated virus 2 (rAAV2) genome is pseudopackaged into a human bocavirus 1 (HBoV1) capsid. Recently, the production of rAAV2/HBoV1 in human embryonic kidney (HEK) 293 cells has been greatly improved in the absence of any HBoV1 nonstructural proteins (NS). This NS-free production system yields over 16-fold more vectors than the original production system that necessitates NS expression. The production of rAAV with infection of baculovirus expression vector (BEV) in the suspension culture of Sf9 insect cells is highly efficient and scalable. Since the replication of the rAAV2 genome in the BEV system is well established, we aimed to develop a BEV system to produce the rAAV2/HBoV1 vector in Sf9 cells. We optimized the usage of translation initiation signals of the HBoV1 capsid proteins (Cap), and constructed a BEV Bac-AAV2Rep-HBoV1Cap, which expresses the AAV2 Rep78 and Rep52 as well as the HBoV1 VP1, VP2, and VP3 at the appropriate ratios. We found that it is sufficient as a trans helper to the production of rAAV2/HBoV1 in Sf9 cells that were co-infected with the transfer Bac-AAV2ITR-GFP-luc that carried a 5.4-kb oversized rAAV2 genome with dual reporters. Further study found that incorporation of an HBoV1 small NS, NP1, in the system maximized the viral DNA replication and thus the rAAV2/HBoV1 vector production at a level similar to that of the rAAV2 vector in Sf9 cells. However, the transduction potency of the rAAV2/HBoV1 vector produced from BEV-infected Sf9 cells was 5-7-fold lower in polarized human airway epithelia than that packaged in HEK293 cells. Transmission electron microscopy analysis found that the vector produced in Sf9 cells had a high percentage of empty capsids, suggesting the pseudopackage of the rAAV2 genome in HBoV1 capsid is not as efficient as in the capsids of AAV2. Nevertheless, our study demonstrated that the rAAV2/HBoV1 can be produced in insect cells with BEVs at a comparable yield to rAAV, and that the highly efficient expression of the HBoV1 capsid proteins warrants further optimization.
Collapse
|
29
|
|
30
|
Cellular Cleavage and Polyadenylation Specificity Factor 6 (CPSF6) Mediates Nuclear Import of Human Bocavirus 1 NP1 Protein and Modulates Viral Capsid Protein Expression. J Virol 2020; 94:JVI.01444-19. [PMID: 31666379 DOI: 10.1128/jvi.01444-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 10/18/2019] [Indexed: 12/13/2022] Open
Abstract
Human bocavirus 1 (HBoV1), which belongs to the genus Bocaparvovirus of the Parvoviridae family, causes acute respiratory tract infections in young children. In vitro, HBoV1 infects polarized primary human airway epithelium (HAE) cultured at an air-liquid interface (HAE-ALI). HBoV1 encodes a small nonstructural protein, nuclear protein 1 (NP1), that plays an essential role in the maturation of capsid protein (VP)-encoding mRNAs and viral DNA replication. In this study, we determined the broad interactome of NP1 using the proximity-dependent biotin identification (BioID) assay combined with mass spectrometry (MS). We confirmed that two host mRNA processing factors, DEAH-box helicase 15 (DHX15) and cleavage and polyadenylation specificity factor 6 (CPSF6; also known as CFIm68), a subunit of the cleavage factor Im complex (CFIm), interact with HBoV1 NP1 independently of any DNA or mRNAs. Knockdown of CPSF6 significantly decreased the expression of capsid protein but not that of DHX15. We further demonstrated that NP1 directly interacts with CPSF6 in vitro and colocalizes within the virus replication centers. Importantly, we revealed a novel role of CPSF6 in the nuclear import of NP1, in addition to the critical role of CPSF6 in NP1-facilitated maturation of VP-encoding mRNAs. Thus, our study suggests that CPSF6 interacts with NP1 to escort NP1 imported into the nucleus for its function in the modulation of viral mRNA processing and viral DNA replication.IMPORTANCE Human bocavirus 1 (HBoV1) is one of the significant pathogens causing acute respiratory tract infections in young children worldwide. HBoV1 encodes a small nonstructural protein (NP1) that plays an important role in the maturation of viral mRNAs encoding capsid proteins as well as in viral DNA replication. Here, we identified a critical host factor, CPSF6, that directly interacts with NP1, mediates the nuclear import of NP1, and plays a role in the maturation of capsid protein-encoding mRNAs in the nucleus. The identification of the direct interaction between viral NP1 and host CPSF6 provides new insights into the mechanism by which a viral small nonstructural protein facilitates the multiple regulation of viral gene expression and replication and reveals a novel target for potent antiviral drug development.
Collapse
|
31
|
Verbeke V, Reynders M, Floré K, Vandewal W, Debulpaep S, Sauer K, Cardoen F, Padalko E. Human bocavirus infection in Belgian children with respiratory tract disease. Arch Virol 2019; 164:2919-2930. [PMID: 31520220 PMCID: PMC7087345 DOI: 10.1007/s00705-019-04396-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 08/05/2019] [Indexed: 02/07/2023]
Abstract
Human bocavirus (HBoV) has been detected primarily in children with acute lower respiratory tract disease (LRTD), but its occurrence, clinical profile, and role as a causative agent of RTD are not clear. The aim of this study was to investigate the prevalence and the potential clinical relevance of HBoV. Using molecular tests, we tested 1352 nasopharyngeal samples obtained between October 1, 2017 and April 30, 2018 from children up to the age of 16 with RTD for the presence of HBoV DNA and 20 other respiratory pathogens at three different hospitals in Belgium. HBoV was detected in 77 children with a median age of 10.6 months. Consecutive samples were available for 15 HBoV-positive children and showed persistent HBoV positivity in four of them. Monoinfection was observed in six infants. Four of them were born prematurely and were infected during hospitalization at the neonatal intensive care unit (NICU). Only one of these six monoinfected children was diagnosed with recurrent wheezing due to HBoV. This child was carried to term and had a high viral load. Coinfections, most frequently with rhinovirus (52.1%) and adenovirus (49.3%), were observed in 72 patients. In seventeen of them in which HBoV was present at high viral load or higher viral load than its copathogens, bronchi(oli)tis (n = 8), recurrent wheezing (n = 8) or episodic wheezing (n = 1) were diagnosed. Our results suggest that HBoV infection at high viral load in infants is associated with wheezing (P = 0.013, Cramer's V = 0.613).
Collapse
Affiliation(s)
- Vanessa Verbeke
- Department of Medical Microbiology, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | | | | | - Wouter Vandewal
- AZ Sint-Lucas Bruges, Sint-Lucaslaan 29, 8310 Brugge, Belgium
| | - Sara Debulpaep
- Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Kate Sauer
- AZ Sint-Jan Bruges, Ruddershove 10, 8000 Brugge, Belgium
| | | | - Elizaveta Padalko
- Department of Medical Microbiology, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| |
Collapse
|
32
|
Di Martino B, Di Profio F, Melegari I, Marsilio F. Feline Virome-A Review of Novel Enteric Viruses Detected in Cats. Viruses 2019; 11:v11100908. [PMID: 31575055 PMCID: PMC6832874 DOI: 10.3390/v11100908] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/28/2019] [Accepted: 09/28/2019] [Indexed: 12/13/2022] Open
Abstract
Recent advances in the diagnostic and metagenomic investigations of the feline enteric environment have allowed the identification of several novel viruses that have been associated with gastroenteritis in cats. In the last few years, noroviruses, kobuviruses, and novel parvoviruses have been repetitively detected in diarrheic cats as alone or in mixed infections with other pathogens, raising a number of questions, with particular regards to their pathogenic attitude and clinical impact. In the present article, the current available literature on novel potential feline enteric viruses is reviewed, providing a meaningful update on the etiology, epidemiologic, pathogenetic, clinical, and diagnostic aspects of the infections caused by these pathogens.
Collapse
Affiliation(s)
- Barbara Di Martino
- Laboratory of Infectious Diseases, Faculty of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy.
| | - Federica Di Profio
- Laboratory of Infectious Diseases, Faculty of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy.
| | - Irene Melegari
- Laboratory of Infectious Diseases, Faculty of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy.
| | - Fulvio Marsilio
- Laboratory of Infectious Diseases, Faculty of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy.
| |
Collapse
|
33
|
Jonsdottir HR, Marti S, Geerts D, Rodriguez R, Thiel V, Dijkman R. Establishment of Primary Transgenic Human Airway Epithelial Cell Cultures to Study Respiratory Virus-Host Interactions. Viruses 2019; 11:v11080747. [PMID: 31412613 PMCID: PMC6723040 DOI: 10.3390/v11080747] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/09/2019] [Accepted: 08/09/2019] [Indexed: 12/21/2022] Open
Abstract
Primary human airway epithelial cell (hAEC) cultures represent a universal platform to propagate respiratory viruses and characterize their host interactions in authentic target cells. To further elucidate specific interactions between human respiratory viruses and important host factors in the airway epithelium, it is important to make hAEC cultures amenable to genetic modification. However, the short and finite lifespan of primary cells in cell culture creates a bottleneck for the genetic modification of these cultures. In the current study, we show that the incorporation of the Rho-associated protein kinase (ROCK) inhibitor (Y-27632) during cell propagation extends the life span of primary human cells in vitro and thereby facilitates the incorporation of lentivirus-based expression systems. Using fluorescent reporters for fluorescence-activated cell sorting (FACS)-based sorting, we generated homogenously fluorescent hAEC cultures that differentiate normally after lentiviral transduction. As a proof-of-principle, we demonstrate that host gene expression can be modulated post-differentiation via inducible short hairpin (sh)RNA-mediated knockdown. Importantly, functional characterization of these transgenic hAEC cultures with exogenous poly (I:C), as a proxy for virus infection, demonstrates that such modifications do not influence the host innate immune response. Moreover, the propagation kinetics of both human coronavirus 229E (HCoV-229E) and human respiratory syncytial virus (hRSV) were not affected. Combined, these results validate our newly established protocol for the genetic modification of hAEC cultures, thereby unlocking a unique potential for detailed molecular characterization of virus–host interactions in human respiratory epithelium.
Collapse
Affiliation(s)
- Hulda R Jonsdottir
- Institute of Virology and Immunology, 3012 Bern & 3147 Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Sabrina Marti
- Institute of Virology and Immunology, 3012 Bern & 3147 Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Dirk Geerts
- Department of Medical Biology, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Regulo Rodriguez
- Institute of Pathology, Cantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
| | - Volker Thiel
- Institute of Virology and Immunology, 3012 Bern & 3147 Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Ronald Dijkman
- Institute of Virology and Immunology, 3012 Bern & 3147 Mittelhäusern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
- Institute for Infectious Diseases, University of Bern, 3010 Bern, Switzerland.
| |
Collapse
|
34
|
Holwerda M, Kelly J, Laloli L, Stürmer I, Portmann J, Stalder H, Dijkman R. Determining the Replication Kinetics and Cellular Tropism of Influenza D Virus on Primary Well-Differentiated Human Airway Epithelial Cells. Viruses 2019; 11:v11040377. [PMID: 31022887 PMCID: PMC6521319 DOI: 10.3390/v11040377] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 12/19/2022] Open
Abstract
Influenza viruses are notorious pathogens that frequently cross the species barrier with often severe consequences for both animal and human health. In 2011, a novel member of the Orthomyxoviridae family, Influenza D virus (IDV), was identified in the respiratory tract of swine. Epidemiological surveys revealed that IDV is distributed worldwide among livestock and that IDV-directed antibodies are detected in humans with occupational exposure to livestock. To identify the transmission capability of IDV to humans, we determined the viral replication kinetics and cell tropism using an in vitro respiratory epithelium model of humans. The inoculation of IDV revealed efficient replication kinetics and apical progeny virus release at different body temperatures. Intriguingly, the replication characteristics of IDV revealed higher replication kinetics compared to Influenza C virus, despite sharing the cell tropism preference for ciliated cells. Collectively, these results might indicate why IDV-directed antibodies are detected among humans with occupational exposure to livestock.
Collapse
Affiliation(s)
- Melle Holwerda
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland.
- Department of Infectious diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland.
| | - Jenna Kelly
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland.
- Department of Infectious diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
| | - Laura Laloli
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland.
- Department of Infectious diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland.
| | - Isabel Stürmer
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland.
- Department of Infectious diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland.
| | - Jasmine Portmann
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland.
- Department of Infectious diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
| | - Hanspeter Stalder
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland.
- Department of Infectious diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
| | - Ronald Dijkman
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland.
- Department of Infectious diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
| |
Collapse
|
35
|
Chen AJ, Dong J, Yuan XH, Bo H, Li SZ, Wang C, Duan ZJ, Zheng LS. Anti-H7N9 avian influenza A virus activity of interferon in pseudostratified human airway epithelium cell cultures. Virol J 2019; 16:44. [PMID: 30944006 PMCID: PMC6448296 DOI: 10.1186/s12985-019-1146-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Since H7N9 influenza A virus (H7N9) was first reported in 2013, five waves of outbreaks have occurred, posing a huge threat to human health. In preparation for a potential H7N9 epidemic, it is essential to evaluate the efficacy of anti-H7N9 drugs with an appropriate model. METHODS Well-differentiated pseudostratified human airway epithelium (HAE) cells were grown at the air-liquid interface, and the H7N9 cell tropism and cytopathic effect were detected by immunostaining and hematoxylin-eosin (HE) staining. The H7N9 replication kinetics and anti-H7N9 effect of recombinant human α2b (rhIFN-α2b) and rhIFN-λ1 were compared with different cell lines. The H7N9 viral load and interferon-stimulated gene (ISG) expression were quantified by real-time PCR assays. RESULTS H7N9 could infect both ciliated and non-ciliated cells within the three-dimensional (3D) HAE cell culture, which reduced the number of cilia and damaged the airways. The H7N9 replication kinetics differed between traditional cells and 3D HAE cells. Interferon had antiviral activity against H7N9 and alleviated epithelial cell lesions; the antiviral activity of rhIFN-α2b was slightly better than that of rhIFN-λ1. In normal cells, rhIFN-α2b induced a greater amount of ISG expression (MX1, OAS1, IFITM3, and ISG15) compared with rhIFN-λ1, but in 3D HAE cells, this trend was reversed. CONCLUSIONS Both rhIFN-α2b and rhIFN-λ1 had antiviral activity against H7N9, and this protection was related to the induction of ISGs. The 3D cell culture model is suitable for evaluating interferon antiviral activity because it can demonstrate realistic in vivo-like effects.
Collapse
Affiliation(s)
- Ai-jun Chen
- National Institute for Viral Disease Control and Prevention, China CDC, Key Laboratory for Medical Virology National Health Commission, 100 Ying-Xin St., Xi-Cheng District, Beijing, 100052 China
| | - Jie Dong
- National Institute for Viral Disease Control and Prevention, China CDC, Key Laboratory for Medical Virology National Health Commission, 100 Ying-Xin St., Xi-Cheng District, Beijing, 100052 China
| | - Xin-hui Yuan
- The First Hospital of Lanzhou University, Lanzhou, 730000 China
| | - Hong Bo
- National Institute for Viral Disease Control and Prevention, China CDC, Key Laboratory for Medical Virology National Health Commission, 100 Ying-Xin St., Xi-Cheng District, Beijing, 100052 China
| | - Shu-zhen Li
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, 210008 China
| | - Chao Wang
- National Institute for Viral Disease Control and Prevention, China CDC, Key Laboratory for Medical Virology National Health Commission, 100 Ying-Xin St., Xi-Cheng District, Beijing, 100052 China
| | - Zhao-jun Duan
- National Institute for Viral Disease Control and Prevention, China CDC, NHC Key Laboratory of Medical Virology and Viral Diseases, 100 Ying-Xin St., Xi-Cheng District, Beijing, 100052 China
| | - Li-shu Zheng
- National Institute for Viral Disease Control and Prevention, China CDC, Key Laboratory for Medical Virology National Health Commission, 100 Ying-Xin St., Xi-Cheng District, Beijing, 100052 China
| |
Collapse
|
36
|
Christensen A, Kesti O, Elenius V, Eskola AL, Døllner H, Altunbulakli C, Akdis CA, Söderlund-Venermo M, Jartti T. Human bocaviruses and paediatric infections. THE LANCET CHILD & ADOLESCENT HEALTH 2019; 3:418-426. [PMID: 30948251 DOI: 10.1016/s2352-4642(19)30057-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/14/2019] [Accepted: 02/15/2019] [Indexed: 12/16/2022]
Abstract
Human bocavirus 1 (HBoV1), belonging to the Parvoviridae family, was discovered in 2005, in nasopharyngeal samples from children with respiratory tract infections. Three additional bocaviruses, HBoV2-4, were discovered in 2009-10. These viruses have mainly been found in faecal samples and their role in human diseases is still uncertain. HBoV1 causes a wide spectrum of respiratory diseases in children, including common cold, acute otitis media, pneumonia, bronchiolitis, and asthma exacerbations. HBoV1 DNA can persist in airway secretions for months after an acute infection. Consequently, acute HBoV1 infection cannot be diagnosed with standard DNA PCR; quantitative PCR and serology are better diagnostic approaches. Because of their high clinical specificity, diagnostic developments such as HBoV1 mRNA and antigen detection have shown promising results. This Review summarises the knowledge on human bocaviruses, with a special focus on HBoV1.
Collapse
Affiliation(s)
- Andreas Christensen
- Department of Medical Microbiology, St Olavs Hospital, Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Olli Kesti
- Department of Paediatrics, Turku University Hospital and University of Turku, Turku, Finland
| | - Varpu Elenius
- Department of Paediatrics, Turku University Hospital and University of Turku, Turku, Finland
| | - Anna L Eskola
- Department of Education, University of Turku, Turku, Finland
| | - Henrik Døllner
- Department of Pediatrics, St Olavs Hospital, Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Can Altunbulakli
- Swiss Institute of Allergy and Asthma Research, University of Zürich and Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research, University of Zürich and Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | | | - Tuomas Jartti
- Department of Paediatrics, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
37
|
Zou W, Xiong M, Deng X, Engelhardt JF, Yan Z, Qiu J. A Comprehensive RNA-seq Analysis of Human Bocavirus 1 Transcripts in Infected Human Airway Epithelium. Viruses 2019; 11:v11010033. [PMID: 30621044 PMCID: PMC6357044 DOI: 10.3390/v11010033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 12/30/2018] [Accepted: 01/02/2019] [Indexed: 02/07/2023] Open
Abstract
Human bocavirus 1 (HBoV1) infects well-differentiated (polarized) human airway epithelium (HAE) cultured at an air-liquid interface (ALI). In the present study, we applied next-generation RNA sequencing to investigate the genome-wide transcription profile of HBoV1, including viral mRNA and small RNA transcripts, in HBoV1-infected HAE cells. We identified novel transcription start and termination sites and confirmed the previously identified splicing events. Importantly, an additional proximal polyadenylation site (pA)p2 and a new distal polyadenylation site (pA)dREH lying on the right-hand hairpin (REH) of the HBoV1 genome were identified in processing viral pre-mRNA. Of note, all viral nonstructural proteins-encoding mRNA transcripts use both the proximal polyadenylation sites [(pA)p1 and (pA)p2] and distal polyadenylation sites [(pA)d1 and (pA)dREH] for termination. However, capsid proteins-encoding transcripts only use the distal polyadenylation sites. While the (pA)p1 and (pA)p2 sites were utilized at roughly equal efficiency for proximal polyadenylation of HBoV1 mRNA transcripts, the (pA)d1 site was more preferred for distal polyadenylation. Additionally, small RNA-seq analysis confirmed there is only one viral noncoding RNA (BocaSR) transcribed from nt 5199⁻5340 of the HBoV1 genome. Thus, our study provides a systematic and unbiased transcription profile, including both mRNA and small RNA transcripts, of HBoV1 in HBoV1-infected HAE-ALI cultures.
Collapse
Affiliation(s)
- Wei Zou
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Min Xiong
- The Children's Mercy Hospital, School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA.
| | - Xuefeng Deng
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA.
| | - Ziying Yan
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA.
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
38
|
The Human Bocavirus 1 NP1 Protein Is a Multifunctional Regulator of Viral RNA Processing. J Virol 2018; 92:JVI.01187-18. [PMID: 30135129 DOI: 10.1128/jvi.01187-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/16/2018] [Indexed: 12/12/2022] Open
Abstract
Human bocavirus 1 (HBoV1) encodes a genus-specific protein, NP1, which regulates viral alternative pre-mRNA processing. Similar to NP1 of the related bocavirus minute virus of canine (MVC), HBoV1 NP1 suppressed cleavage and polyadenylation of RNAs at the viral internal polyadenylation site (pA)p. HBoV1 (pA)p is a complex region. It contains 5 significant cleavage and polyadenylation sites, and NP1 was found to regulate only the three of these sites that are governed by canonical AAUAAA hexamer signals. HBoV1 NP1 also facilitated splicing of the upstream intron adjacent to (pA)p. Alternative polyadenylation and splicing of the upstream intron were independent of each other, functioned efficiently within an isolated transcription unit, and were responsive independent of NP1. Characterization of HBoV1 NP1 generalizes its function within the genus Bocaparvovirus, uncovers important differences, and provides important comparisons with MVC NP1 for mechanistic and evolutionary considerations.IMPORTANCE The Parvovirinae are small nonenveloped icosahedral viruses that are important pathogens in many animal species, including humans. The NP1 protein of human bocavirus 1 (HBoV1), similar to NP1 of the bocavirus minute virus of canine (MVC), regulates viral alternative RNA processing by both suppressing polyadenylation at an internal site, (pA)p, and facilitating splicing of an upstream adjacent intron. These effects allow both extension into the capsid gene and splicing of the viral pre-mRNA that correctly registers the capsid gene open reading frame. Characterization of HBoV1 NP1 generalizes this central mode of parvovirus gene regulation to another member of the bocavirus genus and uncovers both important similarities and differences in function compared to MVC NP1 that will be important for future comparative studies.
Collapse
|
39
|
Hansen M, Brockmann M, Schildgen V, Schildgen O. Human bocavirus is detected in human placenta and aborted tissues. Influenza Other Respir Viruses 2018; 13:106-109. [PMID: 30198208 PMCID: PMC6304315 DOI: 10.1111/irv.12609] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/17/2018] [Accepted: 08/07/2018] [Indexed: 12/19/2022] Open
Abstract
Background To date, four human bocaviruses (HBoV) have been described. The most closely related viruses (bovine and canine parvoviruses) are associated with miscarriage in their hosts. The objective of this retrospective study was to determine the frequency of HBoV DNA in miscarriage. Study Design Tissue samples from 172 patients, in which miscarriage occurred, were included and tested with a published qPCR protocol. Positive PCRs were mutually confirmed by sequencing. Results 43 patients (25%) were positive for HBoV DNA. Of those, the majority of HBoV‐positive samples were tissues from miscarriage (placenta: 6; aborted tissue products of conception: 37 specimens). The samples were not paired; either placental or aborted tissue was available. Conclusions The results show that, as long as no animal model is available, the role of HBoV in the occurrence of miscarriage requires additional prospective studies in order to investigate its significance and causal involvements of this pathogen.
Collapse
Affiliation(s)
- Max Hansen
- Institut für Pathologie, Kliniken der Stadt Köln gGmbH, Klinikum der Privaten Universität Witten/Herdecke mit Sitz in Köln, Cologne, Germany
| | - Michael Brockmann
- Institut für Pathologie, Kliniken der Stadt Köln gGmbH, Klinikum der Privaten Universität Witten/Herdecke mit Sitz in Köln, Cologne, Germany
| | - Verena Schildgen
- Institut für Pathologie, Kliniken der Stadt Köln gGmbH, Klinikum der Privaten Universität Witten/Herdecke mit Sitz in Köln, Cologne, Germany
| | - Oliver Schildgen
- Institut für Pathologie, Kliniken der Stadt Köln gGmbH, Klinikum der Privaten Universität Witten/Herdecke mit Sitz in Köln, Cologne, Germany
| |
Collapse
|
40
|
Niemeyer D, Mösbauer K, Klein EM, Sieberg A, Mettelman RC, Mielech AM, Dijkman R, Baker SC, Drosten C, Müller MA. The papain-like protease determines a virulence trait that varies among members of the SARS-coronavirus species. PLoS Pathog 2018; 14:e1007296. [PMID: 30248143 PMCID: PMC6171950 DOI: 10.1371/journal.ppat.1007296] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 10/04/2018] [Accepted: 08/26/2018] [Indexed: 12/22/2022] Open
Abstract
SARS-coronavirus (CoV) is a zoonotic agent derived from rhinolophid bats, in which a plethora of SARS-related, conspecific viral lineages exist. Whereas the variability of virulence among reservoir-borne viruses is unknown, it is generally assumed that the emergence of epidemic viruses from animal reservoirs requires human adaptation. To understand the influence of a viral factor in relation to interspecies spillover, we studied the papain-like protease (PLP) of SARS-CoV. This key enzyme drives the early stages of infection as it cleaves the viral polyprotein, deubiquitinates viral and cellular proteins, and antagonizes the interferon (IFN) response. We identified a bat SARS-CoV PLP, which shared 86% amino acid identity with SARS-CoV PLP, and used reverse genetics to insert it into the SARS-CoV genome. The resulting virus replicated like SARS-CoV in Vero cells but was suppressed in IFN competent MA-104 (3.7-fold), Calu-3 (2.6-fold) and human airway epithelial cells (10.3-fold). Using ectopically-expressed PLP variants as well as full SARS-CoV infectious clones chimerized for PLP, we found that a protease-independent, anti-IFN function exists in SARS-CoV, but not in a SARS-related, bat-borne virus. This PLP-mediated anti-IFN difference was seen in primate, human as well as bat cells, thus independent of the host context. The results of this study revealed that coronavirus PLP confers a variable virulence trait among members of the species SARS-CoV, and that a SARS-CoV lineage with virulent PLPs may have pre-existed in the reservoir before onset of the epidemic.
Collapse
Affiliation(s)
- Daniela Niemeyer
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Institute of Virology, University of Bonn Medical Centre, Bonn, Germany
- German Centre for Infection Research, associated partner Charité, Berlin, Germany
| | - Kirstin Mösbauer
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Eva M. Klein
- Institute of Virology, University of Bonn Medical Centre, Bonn, Germany
| | - Andrea Sieberg
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Institute of Virology, University of Bonn Medical Centre, Bonn, Germany
| | - Robert C. Mettelman
- Department of Microbiology and Immunology, Loyola University of Chicago, Maywood, IL, United States of America
| | - Anna M. Mielech
- Department of Microbiology and Immunology, Loyola University of Chicago, Maywood, IL, United States of America
| | - Ronald Dijkman
- Institute of Virology and Immunology, Bern & Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, University of Bern, Bern, Switzerland
| | - Susan C. Baker
- Department of Microbiology and Immunology, Loyola University of Chicago, Maywood, IL, United States of America
| | - Christian Drosten
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Institute of Virology, University of Bonn Medical Centre, Bonn, Germany
- German Centre for Infection Research, associated partner Charité, Berlin, Germany
| | - Marcel A. Müller
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Institute of Virology, University of Bonn Medical Centre, Bonn, Germany
- German Centre for Infection Research, associated partner Charité, Berlin, Germany
| |
Collapse
|
41
|
Karuppannan AK, Opriessnig T. Possible risks posed by single-stranded DNA viruses of pigs associated with xenotransplantation. Xenotransplantation 2018; 25:e12453. [PMID: 30264878 PMCID: PMC6120555 DOI: 10.1111/xen.12453] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/12/2018] [Accepted: 07/20/2018] [Indexed: 12/11/2022]
Abstract
Routine large-scale xenotransplantation from pigs to humans is getting closer to clinical reality owing to several state-of-the-art technologies, especially the ability to rapidly engineer genetically defined pigs. However, using pig organs in humans poses risks including unwanted cross-species transfer of viruses and adaption of these pig viruses to the human organ recipient. Recent developments in the field of virology, including the advent of metagenomic techniques to characterize entire viromes, have led to the identification of a plethora of viruses in many niches. Single-stranded DNA (ssDNA) viruses are the largest group prevalent in virome studies in mammals. Specifically, the ssDNA viral genomes are characterized by a high rate of nucleotide substitution, which confers a proclivity to adapt to new hosts and cross-species barriers. Pig-associated ssDNA viruses include torque teno sus viruses (TTSuV) in the Anelloviridae family, porcine parvoviruses (PPV), and porcine bocaviruses (PBoV) both in the family of Parvoviridae, and porcine circoviruses (PCV) in the Circoviridae family, some of which have been confirmed to be pathogenic to pigs. The risks of these viruses for the human recipient during xenotransplantation procedures are relatively unknown. Based on the scant knowledge available on the prevalence, predilection, and pathogenicity of pig-associated ssDNA viruses, careful screening and monitoring are required. In the case of positive identification, risk assessments and strategies to eliminate these viruses in xenotransplantation pig stock may be needed.
Collapse
Affiliation(s)
- Anbu K. Karuppannan
- Department of Veterinary Diagnostic and Production Animal MedicineCollege of Veterinary MedicineIowa State UniversityAmesIowa
| | - Tanja Opriessnig
- Department of Veterinary Diagnostic and Production Animal MedicineCollege of Veterinary MedicineIowa State UniversityAmesIowa
- The Roslin Institute and The Royal (Dick) School of Veterinary StudiesUniversity of EdinburghRoslinMidlothianUK
| |
Collapse
|
42
|
Schildgen V, Longo Y, Pieper M, Schildgen O. T84 air-liquid interface cultures enable isolation of human bocavirus. Influenza Other Respir Viruses 2018; 12:667-668. [PMID: 29676538 PMCID: PMC6086856 DOI: 10.1111/irv.12567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Verena Schildgen
- Kliniken der Stadt Köln gGmbH, Klinikum der Privaten Universität Witten/Herdecke, Institut für Pathologie, Köln, Germany
| | - Ylenia Longo
- Kliniken der Stadt Köln gGmbH, Klinikum der Privaten Universität Witten/Herdecke, Institut für Pathologie, Köln, Germany
| | - Monika Pieper
- Kliniken der Stadt Köln gGmbH, Klinikum der Privaten Universität Witten/Herdecke, Institut für Pathologie, Köln, Germany
| | - Oliver Schildgen
- Kliniken der Stadt Köln gGmbH, Klinikum der Privaten Universität Witten/Herdecke, Institut für Pathologie, Köln, Germany
| |
Collapse
|
43
|
Abstract
Human bocaviruses (HBoVs) have been detected in human gastrointestinal infections worldwide. In 2005, HBoV was also discovered in infants and children with infections of the lower respiratory tract. Recently, several genotypes of this parvovirus, including HBoV genotype 2 (HBoV2), genotype 3 (HBoV3) and genotype 4 (HBoV4), were discovered and found to be closely related to HBoV. HBoV2 was first detected in stool samples from children in Pakistan, followed by detection in other countries. HBoV3 was detected in Australia and HBoV4 was identified in stool samples from Nigeria, Tunisia and the USA. Recently, HBoV infection has been on the rise throughout the world, particularly in countries neighbouring South Korea; however, there have been very few studies on Korean strains. In this study, we characterised the whole genome and determined the phylogenetic position of CUK-BC20, a new clinical HBoV strain isolated in South Korea. The CUK-BC20 genome of 5184 nucleotides (nt) contains three open-reading frames (ORFs). The genotype of CUK-BC20 is HBoV2, and 98.77% of its nt sequence is identical with those of other HBoVs, namely Rus-Nsc10-N386. Especially, the ORF3 amino acid sequences from positions 212-213 and 454 corresponding to a variable region (VR)1 and VR5, respectively, showed genotype-specific substitutions that distinguished the four HBoV genotypes. As the first whole-genome sequence analysis of HBoV in South Korea, this information will provide a valuable reference for the detection of recombination, tracking of epidemics and development of diagnosis methods for HBoV.
Collapse
|
44
|
Tang YW, Stratton CW. The Role of the Human Bocavirus (HBoV) in Respiratory Infections. ADVANCED TECHNIQUES IN DIAGNOSTIC MICROBIOLOGY 2018. [PMCID: PMC7120174 DOI: 10.1007/978-3-319-95111-9_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The human bocavirus is one of the most common respiratory viruses and occurs in all age groups. Because Koch’s postulates have been fulfilled unintendedly, it is currently accepted that the virus is a real pathogen associated with upper and lower respiratory tract infections causing clinical symptoms ranging from a mild common cold to life-threatening respiratory diseases. In order to exclude a viremia, serological analysis should be included during laboratory diagnostics, as acute and chronic infections cannot be differentiated by detection of viral nucleic acids in respiratory specimen alone due to prolonged viral shedding. Besides its ability to persist, the virus appears to trigger chronic lung disease and increases clinical symptoms by causing fibrotic lung diseases. Due to the lack of an animal model, clinical trials remain the major method for studying the long-term effects of HBoV infections.
Collapse
Affiliation(s)
- Yi-Wei Tang
- Departments of Laboratory Medicine and Internal Medicine, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Charles W. Stratton
- Department of Pathology, Microbiology and Immunology and Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| |
Collapse
|
45
|
Human Parvovirus Infection of Human Airway Epithelia Induces Pyroptotic Cell Death by Inhibiting Apoptosis. J Virol 2017; 91:JVI.01533-17. [PMID: 29021400 DOI: 10.1128/jvi.01533-17] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/03/2017] [Indexed: 02/06/2023] Open
Abstract
Human bocavirus 1 (HBoV1) is a human parvovirus that causes acute respiratory tract infections in young children. In this study, we confirmed that, when polarized/well-differentiated human airway epithelia are infected with HBoV1 in vitro, they develop damage characterized by barrier function disruption and cell hypotrophy. Cell death mechanism analyses indicated that the infection induced pyroptotic cell death characterized by caspase-1 activation. Unlike infections with other parvoviruses, HBoV1 infection did not activate the apoptotic or necroptotic cell death pathway. When the NLRP3-ASC-caspase-1 inflammasome-induced pathway was inhibited by short hairpin RNA (shRNA), HBoV1-induced cell death dropped significantly; thus, NLRP3 mediated by ASC appears to be the pattern recognition receptor driving HBoV1 infection-induced pyroptosis. HBoV1 infection induced steady increases in the expression of interleukin 1α (IL-1α) and IL-18. HBoV1 infection was also associated with the marked expression of the antiapoptotic genes BIRC5 and IFI6 When the expression of BIRC5 and/or IFI6 was inhibited by shRNA, the infected cells underwent apoptosis rather than pyroptosis, as indicated by increased cleaved caspase-3 levels and the absence of caspase-1. BIRC5 and/or IFI6 gene inhibition also significantly reduced HBoV1 replication. Thus, HBoV1 infection of human airway epithelial cells activates antiapoptotic proteins that suppress apoptosis and promote pyroptosis. This response may have evolved to confer a replicative advantage, thus allowing HBoV1 to establish a persistent airway epithelial infection. This is the first report of pyroptosis in airway epithelia infected by a respiratory virus.IMPORTANCE Microbial infection of immune cells often induces pyroptosis, which is mediated by a cytosolic protein complex called the inflammasome that senses microbial pathogens and then activates the proinflammatory cytokines IL-1 and IL-18. While virus-infected airway epithelia often activate NLRP3 inflammasomes, studies to date suggest that these viruses kill the airway epithelial cells via the apoptotic or necrotic pathway; involvement of the pyroptosis pathway has not been reported previously. Here, we show for the first time that virus infection of human airway epithelia can also induce pyroptosis. Human bocavirus 1 (HBoV1), a human parvovirus, causes lower respiratory tract infections in young children. This study indicates that HBoV1 kills airway epithelial cells by activating genes that suppress apoptosis and thereby promote pyroptosis. This strategy appears to promote HBoV1 replication and may have evolved to allow HBoV1 to establish persistent infection of human airway epithelia.
Collapse
|
46
|
Adaptation of influenza A (H7N9) virus in primary human airway epithelial cells. Sci Rep 2017; 7:11300. [PMID: 28900138 PMCID: PMC5595892 DOI: 10.1038/s41598-017-10749-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/14/2017] [Indexed: 01/10/2023] Open
Abstract
Influenza A (H7N9) is an emerging zoonotic pathogen with pandemic potential. To understand its adaptation capability, we examined the genetic changes and cellular responses following serial infections of A (H7N9) in primary human airway epithelial cells (hAECs). After 35 serial passages, six amino acid mutations were found, i.e. HA (R54G, T160A, Q226L, H3 numbering), NA (K289R, or K292R for N2 numbering), NP (V363V/I) and PB2 (L/R332R). The mutations in HA enabled A(H7N9) virus to bind with higher affinity (from 39.2% to 53.4%) to sialic acid α2,6-galactose (SAα2,6-Gal) linked receptors. A greater production of proinflammatory cytokines in hAECs was elicited at later passages together with earlier peaking at 24 hours post infection of IL-6, MIP-1α, and MCP-1 levels. Viral replication capacity in hAECs maintained at similar levels throughout the 35 passages. In conclusion, during the serial infections of hAECs by influenza A(H7N9) virus, enhanced binding of virion to cell receptors with subsequent stronger innate cell response were noted, but no enhancement of viral replication could be observed. This indicates the existence of possible evolutional hurdle for influenza A(H7N9) virus to transmit efficiently from human to human.
Collapse
|
47
|
Hamza H, Leifels M, Wilhelm M, Hamza IA. Relative Abundance of Human Bocaviruses in Urban Sewage in Greater Cairo, Egypt. FOOD AND ENVIRONMENTAL VIROLOGY 2017; 9:304-313. [PMID: 28233174 DOI: 10.1007/s12560-017-9287-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/15/2017] [Indexed: 05/18/2023]
Abstract
Human bocavirus (HBoV) is predominantly found in the respiratory tract infections and in the stool of patients with gastroenteritis symptoms. However, data on the prevalence of HBoV genotypes in environmental samples are limited. Here we addressed the prevalence of HBoV in sewage collected from three different wastewater treatment plants in Egypt. HBoV-1, HBoV-2, and HBoV-3 were detected, whereas none of the samples were positive for HBoV-4. The median concentration of HBoV in influent samples was 8.5 × 103 GC/l for HBoV-1, 3.0 × 104 GC/l for HBoV-2, and 2.5 × 104 GC/l for HBoV-3. The concentration was reduced but not completely removed in the effluent samples. The median concentration in the outlet samples was 2.9 × 103 GC/l for HBoV-1, 4.1 × 103 GC/l for HBoV-2, and 2.1 × 103 GC/l for HBoV-3. Moreover, no seasonality pattern of HBoVs was found. The high incidence of HBoV in sewage samples provided an evidence of its circulation in the local population. Although the role of HBoV in respiratory or gastro-intestinal infections still remains to be fully elucidated, the risk of infection via contaminated water should be taken into consideration.
Collapse
Affiliation(s)
- Hazem Hamza
- Environmental Virology Laboratory, Department of Water Pollution Research, National Research Centre, Cairo, Egypt
- Department of Hygiene, Social- and Environmental Medicine, Ruhr-University Bochum, Bochum, Germany
| | - Mats Leifels
- Department of Hygiene, Social- and Environmental Medicine, Ruhr-University Bochum, Bochum, Germany
| | - Michael Wilhelm
- Department of Hygiene, Social- and Environmental Medicine, Ruhr-University Bochum, Bochum, Germany
| | - Ibrahim Ahmed Hamza
- Environmental Virology Laboratory, Department of Water Pollution Research, National Research Centre, Cairo, Egypt.
| |
Collapse
|
48
|
Human Bocavirus 1 Is a Novel Helper for Adeno-associated Virus Replication. J Virol 2017; 91:JVI.00710-17. [PMID: 28659483 DOI: 10.1128/jvi.00710-17] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/22/2017] [Indexed: 12/11/2022] Open
Abstract
Human bocavirus 1 (HBoV1) is an autonomous parvovirus that infects well-differentiated primary human airway epithelia (HAE) in vitro In human embryonic kidney HEK293 cells, the transfection of a duplex HBoV1 genome initiates viral DNA replication and produces progeny virions that are infectious in HAE. HBoV1 takes advantage of signaling pathways in the DNA damage response for efficient genome amplification in both well-differentiated (nondividing) HAE and dividing HEK293 cells. On the other hand, adeno-associated virus 2 (AAV2) is a helper-dependent dependoparvovirus, and productive AAV2 replication requires coinfection with a helper virus (e.g., adenovirus or herpesvirus) or treatment with genotoxic agents. Here, we report that HBoV1 is a novel helper virus for AAV2 replication. Coinfection by HBoV1 and AAV2 rescued AAV2 replication in HAE cells. The helper function of HBoV1 for AAV2 is not limited to HAE cells but also includes HEK293 and HeLa cells. Importantly, the helper function of HBoV1 for AAV2 relies on neither HBoV1 replication nor the DNA damage response. Following transfection of HEK293 cells, the minimal requirements for the replication of the AAV2 duplex DNA genome and the production of progeny virions included the HBoV1 NP1 and NS4 proteins and a newly identified viral long noncoding RNA (BocaSR). However, following infection of HEK293 and HeLa cells with AAV2 virions, HBoV1 NS2 (but not NS4), NP1, and BocaSR were required for AAV2 DNA replication and progeny virion formation. These new methods for packaging the AAV2 genome may be useful for generating recombinant AAV-packaging cell lines and the directed evolution of AAV capsids.IMPORTANCE We first report that an autonomous parvovirus, HBoV1, helps the replication of a dependoparvovirus, AAV2, in differentiated human airway epithelia. We identified the minimal sets of HBoV1 genes required to facilitate the replication of the AAV2 duplex genome and for AAV2 infection. Notably, together with the expression of the NP1 and BocaSR genes, HBoV1 NS2 is required for the productive infection of HEK293 and HeLa cells by AAV2, whereas NS4 is sufficient for viral DNA replication of an AAV2 duplex genome. The identification of HBoV1 as a helper virus for AAV2 replication has implications for the improvement of recombinant AAV production in HEK293 cells and cell types that do not express the adenovirus E1 gene as well as for the rescue of wild-type AAV genomes from tissues during directed evolution in the absence of wild-type adenovirus. A further understanding of the mechanism underlying HBoV1 helper-dependent AAV2 replication may also provide insights into its functions in HBoV1 replication.
Collapse
|
49
|
Zhang J, Bai Y, Zhu B, Hao S, Chen Z, Wang H, Guan W. Mutations in the C-terminus of HBoV NS1 affect the function of NP1. Sci Rep 2017; 7:7407. [PMID: 28785044 PMCID: PMC5547040 DOI: 10.1038/s41598-017-06513-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/13/2017] [Indexed: 02/07/2023] Open
Abstract
Human bocavirus 1 (HBoV1) is an autonomous parvovirus in the Bocaparvovirus genus. The multifunctional nuclear protein NP1 is involved in viral replication. In the present study, we found that the mutations in the C-terminus of NS1 affected NP1 function in viral replication. Knocking out NP1 expression in the recombinant infectious clone, on which the C-terminus of NS1 was mutated based on the clinical samples from nasopharyngeal aspirates, resulted in different degrees of decreased replication. The result suggested that NP1 facilitated the replication of viral genome but was not necessary, which is different from the minute virus of canines, where NP1 is essential for viral replication. Further studies showed that clinical mutations in the NP1 region did not affect viral genome replication, and UP1 promoted viral DNA replication. Our results suggested that the C-terminus of NS1 is important for viral replication and may be a target for regulating the replication of the viral genome.
Collapse
Affiliation(s)
- Junmei Zhang
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yan Bai
- Pediatric department of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing Zhu
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou, 510120, P. R. China
| | - Sujuan Hao
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhen Chen
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Hanzhong Wang
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Wuxiang Guan
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China.
| |
Collapse
|
50
|
Ghietto LM, Toigo D'Angelo AP, Viale FA, Adamo MP. Human bocavirus 1 infection of CACO-2 cell line cultures. Virology 2017; 510:273-280. [PMID: 28777951 PMCID: PMC7172243 DOI: 10.1016/j.virol.2017.07.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/27/2017] [Accepted: 07/28/2017] [Indexed: 02/07/2023]
Abstract
Human bocavirus 1 (HBoV1) is a parvovirus associated with pneumonia in infants. It has been detected in different tissues, including colorectal tumors. In this study, we investigated whether Caco-2 cell line, derived from human colon cancer, can be utilized as a model for HBoV1 replication. We demonstrate HBoV1 replication in Caco-2 cultures supplemented with DEAE-dextran after inoculation with respiratory material from infected patients presenting with acute respiratory infection. A viral cycle of rapid development is displayed. However, in spite of HBoV1 DNA 4-fold increment in the supernatants and monolayers by day 1, evidencing that the system allows the virus genome replication after the entry occurred, infectious progeny particles were not produced. These results are consistent with an infection that is limited to a single growth cycle, which can be associated to mutations in the NS1 and VP1/VP2 regions of HBoV1 genome. Further research will contribute to fully elucidate these observations. HBoV1 replicates within 24 h in standard and differentiated Caco-2 cells. DNA is detected in attached cell and supernatant medium. Immunofluorescence tests evidences HBoV1 infection. DNA replication occurred but infectious progeny particles were not produced. The viral genome presents deleterious mutations at NS1 and VP1/VP2 regions.
Collapse
Affiliation(s)
- Lucía María Ghietto
- Instituto de Virología "Dr. J. M. Vanella", Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina
| | - Ana Paola Toigo D'Angelo
- Instituto de Virología "Dr. J. M. Vanella", Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina
| | - Franco Agustin Viale
- Instituto de Virología "Dr. J. M. Vanella", Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina
| | - María Pilar Adamo
- Instituto de Virología "Dr. J. M. Vanella", Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina.
| |
Collapse
|