51
|
Zhu F, Teng Z, Zhou X, Xu R, Bing X, Shi L, Guo N, Wang M, Liu C, Xia M. H1N1 Influenza Virus-Infected Nasal Mucosal Epithelial Progenitor Cells Promote Dendritic Cell Recruitment and Maturation. Front Immunol 2022; 13:879575. [PMID: 35572503 PMCID: PMC9095954 DOI: 10.3389/fimmu.2022.879575] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/07/2022] [Indexed: 12/24/2022] Open
Abstract
The barrier function of nasal mucosal epithelial cells plays an irreplaceable role in the spread and expansion of viruses in the body. This study found that influenza A virus H1N1 could induce apoptosis of nasal mucosal epithelial progenitor cells, cause an inflammatory response, and trigger the maturation and recruitment of nasal submucosal dendritic cells (DCs), but the mechanism remained unclear. Therefore, we used RNA sequencing and high-resolution untargeted metabolomics to sequence and perform combined bioinformatic analysis of H1N1 virus-infected nasal mucosal epithelial cells from 6 different patients. The abnormal arginine metabolism signaling pathway caused by H1N1 virus infection was screened out, and arginase inhibitors were used to interfere with the abnormal arginine metabolism and the maturation and recruitment of submucosal DCs caused by the H1N1 virus in vitro and in vivo. We conclude that H1N1 influenza virus promotes the recruitment and maturation of submucosal DCs by causing abnormal arginine metabolism in nasal mucosal epithelial cells, thereby triggering respiratory mucosal immunity.
Collapse
Affiliation(s)
- Fangyuan Zhu
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhenxiao Teng
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Department of Otolaryngology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuanchen Zhou
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Runtong Xu
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xin Bing
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Department of Otolaryngology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Shi
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Na Guo
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Min Wang
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Chengcheng Liu
- Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ming Xia
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Department of Otolaryngology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
52
|
Zhang C, Cui H, Li E, Guo Z, Wang T, Yan F, Liu L, Li Y, Chen D, Meng K, Li N, Qin C, Liu J, Gao Y, Zhang C. The SARS-CoV-2 B.1.351 Variant Can Transmit in Rats But Not in Mice. Front Immunol 2022; 13:869809. [PMID: 35572504 PMCID: PMC9095975 DOI: 10.3389/fimmu.2022.869809] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 04/05/2022] [Indexed: 11/23/2022] Open
Abstract
Previous studies have shown that B.1.351 and other variants have extended the host range of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to mice. Sustained transmission is a prerequisite for viral maintenance in a population. However, no evidence of natural transmission of SARS-CoV-2 in wild mice has been documented to date. Here, we evaluated the replication and contact transmission of the B.1.351 variant in mice and rats. The B.1.351 variant could infect and replicate efficiently in the airways of mice and rats. Furthermore, the B.1.351 variant could not be transmitted in BALB/c or C57BL/6 mice but could be transmitted with moderate efficiency in rats by direct contact. Additionally, the B.1.351 variant did not transmit from inoculated Syrian hamsters to BALB/c mice. Moreover, the mouse-adapted SARS-CoV-2 strain C57MA14 did not transmit in mice. In summary, the risk of B.1.351 variant transmission in mice is extremely low, but the transmission risk in rats should not be neglected. We should pay more attention to the potential natural transmission of SARS-CoV-2 variants in rats and their possible spillback to humans.
Collapse
Affiliation(s)
- Cheng Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Huan Cui
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,College of Veterinary Medicine, Jilin University, Changchun, China
| | - Entao Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zhendong Guo
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Tiecheng Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Fang Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Lina Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yuanguo Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Di Chen
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Keyin Meng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Nan Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Chengfeng Qin
- Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Juxiang Liu
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Yuwei Gao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Chunmao Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| |
Collapse
|
53
|
Vimalajeewa D, Balasubramaniam S, Berry DP, Barry G. Virus particle propagation and infectivity along the respiratory tract and a case study for SARS-CoV-2. Sci Rep 2022; 12:7666. [PMID: 35538182 PMCID: PMC9088735 DOI: 10.1038/s41598-022-11816-2] [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: 04/30/2021] [Accepted: 04/22/2022] [Indexed: 11/09/2022] Open
Abstract
Respiratory viruses including Respiratory Syncytial Virus, influenza virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) cause serious and sometimes fatal disease in thousands of people annually. Understanding virus propagation dynamics within the respiratory system is critical because new insights will increase our understanding of virus pathogenesis and enable infection patterns to be more predictable in vivo, which will enhance our ability to target vaccine and drug delivery. This study presents a computational model of virus propagation within the respiratory tract network. The model includes the generation network branch structure of the respiratory tract, biophysical and infectivity properties of the virus, as well as air flow models that aid the circulation of the virus particles. As a proof of principle, the model was applied to SARS-CoV-2 by integrating data about its replication-cycle, as well as the density of Angiotensin Converting Enzyme expressing cells along the respiratory tract network. Using real-world physiological data associated with factors such as the respiratory rate, the immune response and virus load that is inhaled, the model can improve our understanding of the concentration and spatiotemporal dynamics of the virus. We collected experimental data from a number of studies and integrated them with the model in order to show in silico how the virus load propagates along the respiratory network branches.
Collapse
Affiliation(s)
- Dixon Vimalajeewa
- Department of Statistics, Texas A & M University, College Station, TX, USA.
| | | | - Donagh P Berry
- Teagasc, Animal & Grassland Research and Innovation Center, Moorepark, Cork, Ireland
| | - Gerald Barry
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
54
|
Vadlamudi G, Thirumalaikumaran SK, Chakravortty D, Saha A, Basu S. Penetration and aerosolization of cough droplet spray through face masks: A unique pathway of transmission of infection. PHYSICS OF FLUIDS 2022; 34. [DOI: 10.1063/5.0093297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
The advent of the COVID-19 pandemic has necessitated the use of face masks, making them an integral part of the daily routine. Face masks occlude the infectious droplets during any respiratory event contributing to source control. In the current study, spray impingement experiments were conducted on porous surfaces like masks having a different porosity, pore size, and thickness. The spray mimics actual cough or a mild sneeze with respect to the droplet size distribution (20–500 μm) and velocity scale (0–14 m/s), which makes the experimental findings physiologically realistic. The penetration dynamics through the mask showed that droplets of all sizes beyond a critical velocity penetrate through the mask fabric and atomize into daughter droplets in the aerosolization range, leading to harmful effects due to the extended airborne lifetime of aerosols. By incorporating spray characteristics along with surface tension and viscous dissipation of the fluid passing through the mask, multi-step penetration criteria have been formulated. The daughter droplet size and velocity distribution after atomizing through multi-layered masks and its effects have been discussed. Moreover, the virus-emulating particle-laden surrogate respiratory droplets are used in impingement experiments to study the filtration and entrapment of virus-like nanoparticles in the mask. Furthermore, the efficacy of the mask from the perspective of a susceptible person has been investigated.
Collapse
Affiliation(s)
- Gautham Vadlamudi
- Department of Mechanical Engineering, Indian Institute of Science 1 , Bangalore, Karnataka 560012, India
| | - S. K. Thirumalaikumaran
- Department of Mechanical Engineering, Indian Institute of Science 1 , Bangalore, Karnataka 560012, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of Science 2 , Bangalore, Karnataka 560012, India
- Center of Biosystems Science and Engineering, Indian Institute of Science 3 , Bangalore, Karnataka 560012, India
| | - Abhishek Saha
- Department of Mechanical and Aerospace Engineering, University of California San Diego 4 , La Jolla, California 92093, USA
| | - Saptarshi Basu
- Department of Mechanical Engineering, Indian Institute of Science 1 , Bangalore, Karnataka 560012, India
- Interdisciplinary Centre for Energy Research (ICER), Indian Institute of Science 5 , Bangalore, Karnataka 560012, India
| |
Collapse
|
55
|
Innate and Adaptive Immune Responses in the Upper Respiratory Tract and the Infectivity of SARS-CoV-2. Viruses 2022; 14:v14050933. [PMID: 35632675 PMCID: PMC9143801 DOI: 10.3390/v14050933] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/20/2022] [Accepted: 04/26/2022] [Indexed: 02/01/2023] Open
Abstract
Increasing evidence shows the nasal epithelium to be the initial site of SARS-CoV-2 infection, and that early and effective immune responses in the upper respiratory tract (URT) limit and eliminate the infection in the URT, thereby preventing infection of the lower respiratory tract and the development of severe COVID-19. SARS-CoV-2 interferes with innate immunity signaling and evolves mutants that can reduce antibody-mediated immunity in the URT. Recent genetic and immunological advances in understanding innate immunity to SARS-CoV-2 in the URT, and the ability of prior infections as well as currently available injectable and potential intranasal COVID-19 vaccines to generate anamnestic adaptive immunity in the URT, are reviewed. It is suggested that the more detailed investigation of URT immune responses to all types of COVID-19 vaccines, and the development of safe and effective COVID-19 vaccines for intranasal administration, are important needs.
Collapse
|
56
|
Mtambo SE, Ugbaja SC, Mushebenge AG, Abubakar BH, Ntuli ML, Kumalo HM. Intermolecular Mechanism and Dynamic Investigation of Avian Influenza H7N9 Virus' Susceptibility to E119V-Substituted Peramivir-Neuraminidase Complex. Molecules 2022; 27:1640. [PMID: 35268741 PMCID: PMC8911867 DOI: 10.3390/molecules27051640] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/14/2022] [Accepted: 01/14/2022] [Indexed: 11/24/2022] Open
Abstract
The H7N9 virus attaches itself to the human cell receptor protein containing the polysaccharide that terminates with sialic acid. The mutation of neuraminidase at residue E119 has been explored experimentally. However, there is no adequate information on the substitution with E119V in peramivir at the intermolecular level. Therefore, a good knowledge of the interatomic interactions is a prerequisite in understanding its transmission mode and subsequent effective inhibitions of the sialic acid receptor cleavage by neuraminidase. Herein, we investigated the mechanism and dynamism on the susceptibility of the E119V mutation on the peramivir-neuraminidase complex relative to the wildtype complex at the intermolecular level. This study aims to investigate the impact of the 119V substitution on the neuraminidase-peramivir complex and unveil the residues responsible for the complex conformations. We employed molecular dynamic (MD) simulations and extensive post-MD analyses in the study. These extensive computational investigations were carried out on the wildtype and the E119V mutant complex of the protein for holistic insights in unveiling the effects of this mutation on the binding affinity and the conformational terrain of peramivir-neuraminidase E119V mutation. The calculated total binding energy (ΔGbind) for the peramivir wildtype is -49.09 ± 0.13 kcal/mol, while the E119V mutant is -58.55 ± 0.15 kcal/mol. The increase in binding energy (9.46 kcal/mol) is consistent with other post-MD analyses results, confirming that E119V substitution confers a higher degree of stability on the protein complex. This study promises to proffer contributory insight and additional knowledge that would enhance future drug designs and help in the fight targeted at controlling the avian influenza H7N9 virus. Therefore, we suggest that experimentalists collaborate with computational chemists for all investigations of this topic, as we have done in our previous studies.
Collapse
Affiliation(s)
- Sphamandla E. Mtambo
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (S.E.M.); (A.G.M.)
| | - Samuel C. Ugbaja
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (S.E.M.); (A.G.M.)
| | - Aganze G. Mushebenge
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (S.E.M.); (A.G.M.)
| | - Bahijjahtu H. Abubakar
- Renewable Energy Programme, Federal Ministry of Environment, 444 Aguiyi Ironsi Way, Maitama, Abuja 904101, Nigeria;
| | - Mthobisi L. Ntuli
- Department of Mathematics, Faculty of Applied Science, Durban University of Technology, Durban 4001, South Africa;
| | - Hezekiel M. Kumalo
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (S.E.M.); (A.G.M.)
| |
Collapse
|
57
|
Xie J, Tian S, Liu J, Cao R, Yue P, Cai X, Shang Q, Yang M, Han L, Zhang DK. Dual role of the nasal microbiota in neurological diseases—An unignorable risk factor or a potential therapy carrier. Pharmacol Res 2022; 179:106189. [DOI: 10.1016/j.phrs.2022.106189] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/06/2022] [Accepted: 03/17/2022] [Indexed: 12/11/2022]
|
58
|
Contribution of neuraminidase to the efficacy of seasonal split influenza vaccines in the ferret model. J Virol 2022; 96:e0195921. [PMID: 35107371 PMCID: PMC8941921 DOI: 10.1128/jvi.01959-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Seasonal influenza vaccination takes into account primarily hemagglutinin (HA)-specific neutralizing antibody responses. However, the accumulation of substitutions in the antigenic regions of HA (i.e., antigenic drift) occasionally results in a mismatch between the vaccine and circulating strains. To prevent poor vaccine performance, we investigated whether an antigenically matched neuraminidase (NA) may compensate for reduced vaccine efficacy due to a mismatched HA. Ferrets were vaccinated twice with adjuvanted split inactivated influenza vaccines containing homologous HA and NA (vacH3N2), only homologous HA (vacH3N1), only homologous NA (vacH1N2), heterologous HA and NA (vacH1N1), or phosphate-buffered saline (vacPBS), followed by challenge with H3N2 virus (A/Netherlands/16190/1968). Ferrets vaccinated with homologous HA (vacH3N2 and vacH3N1) displayed minimum fever and weight loss compared to vacH1N1 and vacPBS ferrets, while ferrets vaccinated with NA-matched vacH1N2 displayed intermediate fever and weight loss. Vaccination with vacH1N2 further led to a reduction in virus shedding from the nose and undetectable virus titers in the lower respiratory tract, similarly to when the homologous vacH3N2 was used. Some protection was observed upon vacH1N1 vaccination, but this was not comparable to that observed for vacH1N2, again highlighting the important role of NA in vaccine-induced protection. These results illustrate that NA antibodies can prevent severe disease caused by influenza virus infection and that an antigenically matched NA in seasonal vaccines might prevent lower respiratory tract complications. This underlines the importance of considering NA during the yearly vaccine strain selection process, which may be particularly beneficial in seasons when the HA component of the vaccine is mismatched. IMPORTANCE Despite the availability of vaccines, influenza virus infections continue to cause substantial morbidity and mortality in humans. Currently available influenza vaccines take primarily the hemagglutinin (HA) into account, but the highly variable nature of this protein as a result of antigenic drift has led to a recurrent decline in vaccine effectiveness. While the protective effect of neuraminidase (NA) antibodies has been highlighted by several studies, there are no requirements with regard to quantity or quality of NA in licensed vaccines, and NA immunity remains largely unexploited. Since antigenic changes in HA and NA are thought to occur asynchronously, NA immunity could compensate for reduced vaccine efficacy when drift in HA occurs. By matching and mismatching the HA and NA components of monovalent split inactivated vaccines, we demonstrated the potential of NA immunity to protect against disease, virus replication in the lower respiratory tract, and virus shedding in the ferret model.
Collapse
|
59
|
Port JR, Yinda CK, Avanzato VA, Schulz JE, Holbrook MG, van Doremalen N, Shaia C, Fischer RJ, Munster VJ. Increased small particle aerosol transmission of B.1.1.7 compared with SARS-CoV-2 lineage A in vivo. Nat Microbiol 2022; 7:213-223. [PMID: 35017676 PMCID: PMC11218742 DOI: 10.1038/s41564-021-01047-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022]
Abstract
The major transmission route for SARS-CoV-2 is airborne. However, previous studies could not elucidate the contribution between large droplets and aerosol transmission of SARS-CoV-2 and its variants. Here, we designed and validated an optimized transmission caging setup, which allows for the assessment of aerosol transmission efficiency at various distances. At a distance of 2 m, only particles of <5 μm traversed between cages. Using this setup, we investigated the relative efficiency of aerosol transmission between the SARS-CoV-2 Alpha variant (B.1.1.7) and lineage A in Syrian hamsters. Aerosol transmission of both variants was confirmed in all sentinels after 24 h of exposure as demonstrated by respiratory virus shedding and seroconversion. Productive transmission also occurred after 1 h of exposure, highlighting the efficiency of this transmission route. Interestingly, after donors were infected with a mix of both variants, the Alpha variant outcompeted the lineage A variant in an airborne transmission chain. Overall, these data indicate that a lower infectious dose of the Alpha variant, compared to lineage A, could be sufficient for successful transmission. This highlights the continuous need to assess emerging variants and the development for pre-emptive transmission mitigation strategies.
Collapse
Affiliation(s)
- Julia R Port
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Claude Kwe Yinda
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Victoria A Avanzato
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jonathan E Schulz
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Myndi G Holbrook
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Neeltje van Doremalen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Carl Shaia
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Robert J Fischer
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Vincent J Munster
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
60
|
Das SK, Alam JE, Plumari S, Greco V. Airborne virus transmission under different weather conditions. AIP ADVANCES 2022; 12:015019. [PMID: 35070489 PMCID: PMC8759630 DOI: 10.1063/5.0082017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 12/22/2021] [Indexed: 05/07/2023]
Abstract
The COVID19 infection is known to disseminate through droplets ejected by infected individuals during coughing, sneezing, speaking, and breathing. The spread of the infection and hence its menace depend on how the virus-loaded droplets evolve in space and time with changing environmental conditions. In view of this, we investigate the evolution of the droplets within the purview of the Brownian motion of the evaporating droplets in the air with varying weather conditions under the action of gravity. We track the movement of the droplets until either they gravitationally settle on the ground or evaporate to aerosols of size 2 μm or less. Droplets with radii 2 μm or less may continue to diffuse and remain suspended in the air for a long time. The effects of relative humidity and temperature on the evaporation are found to be significant. We note that under strong flowing conditions, droplets travel large distances. It is found that the bigger droplets fall on the ground due to the dominance of gravity over the diffusive force despite the loss of mass due to evaporation. The smaller evaporating droplets may not settle on the ground but remain suspended in the air due to the dominance of the diffusive force. The fate of the intermediate size droplets depends on the weather conditions and plays crucial roles in the spread of the infection. These environment dependent effects indicate that the maintenance of physical separation to evade the virus is not corroborated, making the use of face masks indispensable.
Collapse
Affiliation(s)
- Santosh K. Das
- School of Physical Sciences, Indian Institute of Technology Goa, Ponda 403401, Goa, India
| | | | | | | |
Collapse
|
61
|
Biswas R, Pal A, Pal R, Sarkar S, Mukhopadhyay A. Risk assessment of COVID infection by respiratory droplets from cough for various ventilation scenarios inside an elevator: An OpenFOAM-based computational fluid dynamics analysis. PHYSICS OF FLUIDS (WOODBURY, N.Y. : 1994) 2022; 34:013318. [PMID: 35340680 PMCID: PMC8939552 DOI: 10.1063/5.0073694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/30/2021] [Indexed: 05/15/2023]
Abstract
Respiratory droplets-which may contain disease spreading virus-exhaled during speaking, coughing, or sneezing are one of the significant causes for the spread of the ongoing COVID-19 pandemic. The droplet dispersion depends on the surrounding air velocity, ambient temperature, and relative humidity. In a confined space like an elevator, the risk of transmission becomes higher when there is an infected person inside the elevator with other individuals. In this work, a numerical investigation is carried out in a three-dimensional domain resembling an elevator using OpenFoam. Three different modes of air ventilation, viz., quiescent, axial exhaust draft, and exhaust fan, have been considered to investigate the effect of ventilation on droplet transmission for two different climatic conditions (30 °C , 50% relative humidity and 10 °C , 90% relative humidity). The risk assessment is quantified using a risk factor based on the time-averaged droplet count present near the passenger's hand to head region (risky height zone). The risk factor drops from 40% in a quiescent scenario to 0% in an exhaust fan ventilation condition in a hot dry environment. In general, cold humid conditions are safer than hot dry conditions as the droplets settle down quickly below the risky height zone owing to their larger masses maintained by negligible evaporation. However, an exhaust fan renders the domain in a hot dry ambience completely safe (risk factor, 0%) in 5.5 s whereas it takes 7.48 s for a cold humid ambience.
Collapse
Affiliation(s)
- Riddhideep Biswas
- Department of Mechanical Engineering, Jadavpur University, Kolkata-700032, India
| | - Anish Pal
- Department of Mechanical Engineering, Jadavpur University, Kolkata-700032, India
| | - Ritam Pal
- Department of Mechanical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Sourav Sarkar
- Department of Mechanical Engineering, Jadavpur University, Kolkata-700032, India
- Author to whom correspondence should be addressed:
| | | |
Collapse
|
62
|
Patra SS, Nath J, Panda S, Das T, Ramasamy B. Evaluating the filtration efficiency of commercial facemasks' materials against respiratory aerosol droplets. JOURNAL OF THE AIR & WASTE MANAGEMENT ASSOCIATION (1995) 2022; 72:3-9. [PMID: 34170783 DOI: 10.1080/10962247.2021.1948459] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/11/2021] [Accepted: 06/14/2021] [Indexed: 06/13/2023]
Abstract
Respiratory droplets serve as a viable transmission mechanism for many viruses and other pathogens. Facemasks are commonly used to minimize the risk of this transmission. However, information on the size-resolved filtration efficiency of commonly available commercial facemasks is not readily available in the literature. To fill this gap, the current study performs aerosolized chamber experiments to evaluate the filtration efficiencies of commonly available commercial facemasks' materials in a size range of 0.3-10 µm. Results rank the performance of filtration through commercial facemasks' materials as follows (values in brackets indicate the average filtration efficiencies across 0.3-10 µm): 6-Layer N95 mask (0.918) > N95 mask - without valve (0.88) > KN95 mask (0.84) > N95 mask -with valve (0.834) > Heavy knitted cotton mask (0.808) > Surgical mask (0.778) > Cotton mask-2 layers (0.744) > Nylon fabric mask-2 layers (0.740) > T-shirt fabric mask-2 layers (0.708) > T-shirt fabric mask-1 layer (0.648). The size-resolved filtration efficiencies through the material across the evaluated commercial facemasks ranged from 38-83% in the size range of 0.3-0.5 µm, 55-88% in the size range of 0.5-1 µm, 69-93% in the size range of 1-2.5 µm, 76-96% in the size range of 2.5-5 µm, and 86-99% in the size range of 5-10 µm. Subsequently, the filtration efficiencies of materials post washing (with detergent in warm water and allowing to dry completely) were also evaluated. The average reduction in filtration efficiencies post washing are as follows: 6-Layer N95 mask: 3%, N95 mask - without valve: 2%, KN95 mask: 4%, N95 mask -with valve: 3%, Heavy knitted cotton mask: 4%, Surgical mask: 18%, Cotton mask-2 layers: 11%, Nylon fabric mask-2 layers: 6%, T-shirt fabric mask-2 layers: 6%, T-shirt fabric mask-1 layer: 8%. This decrease in the filtration efficiency was more pronounced for the sub-micron particles than the super-micron ones.Implications: Facemasks are commonly used to minimize the risk of pathogens through ambient air transmission. However, information on the size-resolved filtration efficiency of commonly available commercial facemasks materials is not readily available in the literature. To fill this gap, the current study performs aerosolized chamber experiments to evaluate the filtration efficiencies of commonly available commercial facemasks materials in a size range of 0.3-10 µm. The performance of the commercial facemasks materials as follows in the order of (values in brackets indicate the average filtration efficiencies across 0.3-10 µm): 6-Layer N95 mask (0.918) > N95 mask - without valve (0.88) > KN95 mask (0.84) > N95 mask -with valve (0.834) > Heavy knitted cotton mask (0.808) > Surgical mask (0.778) > Cotton mask-2 layers (0.744) > Nylon fabric mask-2 layers (0.740) > T-shirt fabric mask-2 layers (0.708) > T-shirt fabric mask-1 layer (0.648). The choice of facemask is greatly driven by the size of viable respiratory droplets that need to be eliminated. If droplets with particle size less than 0.5 µm are required to be filtered, N95 masks without the valve or more layers are preferred. If the primary objective is to filter particles between 0.5-1 µm, then N95 (both with or without valves) or KN95 masks are recommended. Surgical masks and heavy knitted cotton masks may also be used for this purpose, but with caution.
Collapse
Affiliation(s)
- Satya S Patra
- Lyles School of Civil Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Jyotishree Nath
- Environment & Sustainability Department, CSIR-Institute of Minerals and Materials Technology (CSIR-IMMT), Bhubaneswar, India
- Academy of Scientific and Innovative Research (Acsir), CSIR-Institute of Minerals and Materials Technology (CSIR-IMMT), Bhubaneswar, India
| | - Subhasmita Panda
- Environment & Sustainability Department, CSIR-Institute of Minerals and Materials Technology (CSIR-IMMT), Bhubaneswar, India
- Academy of Scientific and Innovative Research (Acsir), CSIR-Institute of Minerals and Materials Technology (CSIR-IMMT), Bhubaneswar, India
| | - Trupti Das
- Environment & Sustainability Department, CSIR-Institute of Minerals and Materials Technology (CSIR-IMMT), Bhubaneswar, India
- Academy of Scientific and Innovative Research (Acsir), CSIR-Institute of Minerals and Materials Technology (CSIR-IMMT), Bhubaneswar, India
| | - Boopathy Ramasamy
- Environment & Sustainability Department, CSIR-Institute of Minerals and Materials Technology (CSIR-IMMT), Bhubaneswar, India
- Academy of Scientific and Innovative Research (Acsir), CSIR-Institute of Minerals and Materials Technology (CSIR-IMMT), Bhubaneswar, India
| |
Collapse
|
63
|
Lopez-Gordo E, Orlowski A, Wang A, Weinberg A, Sahoo S, Weber T. Hydroxylation of N-acetylneuraminic Acid Influences the in vivo Tropism of N-linked Sialic Acid-Binding Adeno-Associated Viruses AAV1, AAV5, and AAV6. Front Med (Lausanne) 2021; 8:732095. [PMID: 35036407 PMCID: PMC8757481 DOI: 10.3389/fmed.2021.732095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Adeno-associated virus (AAV) vectors are promising candidates for gene therapy. However, a number of recent preclinical large animal studies failed to translate into the clinic. This illustrates the formidable challenge of choosing the animal models that promise the best chance of a successful translation into the clinic. Several of the most common AAV serotypes use sialic acid (SIA) as their primary receptor. However, in contrast to most mammals, humans lack the enzyme CMAH, which hydroxylates cytidine monophosphate-N-acetylneuraminic acid (CMP-Neu5Ac) into cytidine monophosphate-N-glycolylneuraminic acid (CMP-Neu5Gc). As a result, human glycans only contain Neu5Ac and not Neu5Gc. Here, we investigate the tropism of AAV1, 5, 6 and 9 in wild-type C57BL/6J (WT) and CMAH knock-out (CMAH−/−) mice. All N-linked SIA-binding serotypes (AAV1, 5 and 6) showed significantly lower transduction of the heart in CMAH−/− when compared to WT mice (5–5.8-fold) and, strikingly, skeletal muscle transduction by AAV5 was almost 30-fold higher in CMAH−/− compared to WT mice. Importantly, the AAV tropism or distribution of expression among different organs was also affected. For AAV1, AAV5 and AAV6, expression in the heart compared to the liver was 4.6–8-fold higher in WT than in CMAH−/− mice, and for AAV5 the expression in the heart compared to the skeletal muscle was 57.3-fold higher in WT than in CMAH−/− mice. These data thus strongly suggest that the relative abundance of Neu5Ac and Neu5Gc plays a role in AAV tropism, and that results obtained in commonly used animal models might not translate into the clinic.
Collapse
Affiliation(s)
- Estrella Lopez-Gordo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Alejandro Orlowski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Arthur Wang
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Alan Weinberg
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Thomas Weber
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
- *Correspondence: Thomas Weber
| |
Collapse
|
64
|
McKimm-Breschkin JL, Hay AJ, Cao B, Cox RJ, Dunning J, Moen AC, Olson D, Pizzorno A, Hayden FG. COVID-19, Influenza and RSV: Surveillance-informed prevention and treatment - Meeting report from an isirv-WHO virtual conference. Antiviral Res 2021; 197:105227. [PMID: 34933044 PMCID: PMC8684224 DOI: 10.1016/j.antiviral.2021.105227] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 12/19/2022]
Abstract
The International Society for Influenza and other Respiratory Virus Diseases (isirv) and the WHO held a joint virtual conference from 19th-21st October 2021. While there was a major focus on the global response to the SARS-CoV-2 pandemic, including antivirals, vaccines and surveillance strategies, papers were also presented on treatment and prevention of influenza and respiratory syncytial virus (RSV). Potential therapeutics for SARS-CoV-2 included host-targeted therapies baricitinib, a JAK inhibitor, tocilizumab, an IL-6R inhibitor, verdinexor and direct acting antivirals ensovibep, S-217622, AT-527, and monoclonal antibodies casirivimab and imdevimab, directed against the spike protein. Data from trials of nirsevimab, a monoclonal antibody with a prolonged half-life which binds to the RSV F-protein, and an Ad26.RSV pre-F vaccine were also presented. The expanded role of the WHO Global Influenza Surveillance and Response System to address the SARS-CoV-2 pandemic was also discussed. This report summarizes the oral presentations given at this meeting for the benefit of the broader medical and scientific community involved in surveillance, treatment and prevention of respiratory virus diseases.
Collapse
Affiliation(s)
- Jennifer L McKimm-Breschkin
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia.
| | - Alan J Hay
- The Francis Crick Institute, London, UK.
| | - Bin Cao
- China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, China.
| | - Rebecca J Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway.
| | - Jake Dunning
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK.
| | - Ann C Moen
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Daniel Olson
- University of Colorado School of Medicine and Colorado School of Public Health, Anschutz Medical Campus, Aurora, CO, USA.
| | - Andrés Pizzorno
- International Center for Research in Infectious Diseases, University of Lyon, Lyon, France.
| | - Frederick G Hayden
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
65
|
Shah AU, Li Y, Ouyang W, Wang Z, Zuo J, Shi S, Yu Q, Lin J, Yang Q. From nasal to basal: single-cell sequencing of the bursa of Fabricius highlights the IBDV infection mechanism in chickens. Cell Biosci 2021; 11:212. [PMID: 34915931 PMCID: PMC8675306 DOI: 10.1186/s13578-021-00728-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 12/07/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chickens, important food animals and model organisms, are susceptible to many RNA viruses that invade via the nasal cavity. To determine the nasal entry site of the virus and clarify why avians are susceptible to RNA viruses, infectious bursal disease virus (IBDV) was selected because it is a typical avian RNA virus that infects chickens mainly via the nasal route. RESULTS First, we found that IBDV infected the posterior part of the nasal cavity in chickens, which is rich in lymphoid tissue and allows the virus to be easily transferred to the blood. Via the blood circulation, IBDV infected peripheral blood mononuclear cells (PBMCs) and was transferred to the bursa of Fabricius to damage the IgM + B lymphocyte population. Subsequently, the single-cell RNA sequencing (scRNA-seq) results suggested the more detailed response of different bursal cell populations (B cells, epithelial cells, dendritic cells, and fibroblasts) to IBDV. Regarding B cells, IBDV infection greatly decreased the IgM + B cell population but increased the IgA + B cell population in the bursal follicles. In contrast to B cells, bursal epithelial cells, especially basal cells, accumulated a large number of IBDV particles. Furthermore, we found that both innate RNA sensors and interferon-stimulated genes (ISGs) were highly expressed in the IBDV-infected groups, while dicer and ago2 expression was largely blocked by IBDV infection. This result suggests that dicer-related RNA interference (RNAi) might be an effective antiviral strategy for IBDV infection in avian. CONCLUSION Our study not only comprehensively elaborates on the transmission of airborne IBDV via the intranasal route and establishes the main target cell types for productive IBDV infection but also provides sufficient evidence to explain the cellular antiviral mechanism against IBDV infection.
Collapse
Affiliation(s)
- Abid Ullah Shah
- College of Life Sciences, Nanjing Agricultural University, Wei gang 1, Nanjing, Jiangsu, 210095, People's Republic of China.,College of Veterinary Medicine, Nanjing Agricultural University, Wei gang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Yuchen Li
- College of Veterinary Medicine, Nanjing Agricultural University, Wei gang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Wei Ouyang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences/Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, People's Republic of China
| | - Zhisheng Wang
- Institute of Veterinary Immunology and Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, People's Republic of China
| | - Jinjiao Zuo
- College of Life Sciences, Nanjing Agricultural University, Wei gang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Song Shi
- Shanghai OE Biotech. Co., Ltd, Shanghai, 201114, People's Republic of China
| | - Qinghua Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Wei gang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Jian Lin
- College of Life Sciences, Nanjing Agricultural University, Wei gang 1, Nanjing, Jiangsu, 210095, People's Republic of China.
| | - Qian Yang
- College of Veterinary Medicine, Nanjing Agricultural University, Wei gang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| |
Collapse
|
66
|
Nakayama T, Lee IT, Jiang S, Matter MS, Yan CH, Overdevest JB, Wu CT, Goltsev Y, Shih LC, Liao CK, Zhu B, Bai Y, Lidsky P, Xiao Y, Zarabanda D, Yang A, Easwaran M, Schürch CM, Chu P, Chen H, Stalder AK, McIlwain DR, Borchard NA, Gall PA, Dholakia SS, Le W, Xu L, Tai CJ, Yeh TH, Erickson-Direnzo E, Duran JM, Mertz KD, Hwang PH, Haslbauer JD, Jackson PK, Menter T, Andino R, Canoll PD, DeConde AS, Patel ZM, Tzankov A, Nolan GP, Nayak JV. Determinants of SARS-CoV-2 entry and replication in airway mucosal tissue and susceptibility in smokers. Cell Rep Med 2021; 2:100421. [PMID: 34604819 PMCID: PMC8479532 DOI: 10.1016/j.xcrm.2021.100421] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/21/2021] [Accepted: 09/22/2021] [Indexed: 02/07/2023]
Abstract
Understanding viral tropism is an essential step toward reducing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) transmission, decreasing mortality from coronavirus disease 2019 (COVID-19) and limiting opportunities for mutant strains to arise. Currently, little is known about the extent to which distinct tissue sites in the human head and neck region and proximal respiratory tract selectively permit SARS-CoV-2 infection and replication. In this translational study, we discover key variabilities in expression of angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2), essential SARS-CoV-2 entry factors, among the mucosal tissues of the human proximal airways. We show that SARS-CoV-2 infection is present in all examined head and neck tissues, with a notable tropism for the nasal cavity and tracheal mucosa. Finally, we uncover an association between smoking and higher SARS-CoV-2 viral infection in the human proximal airway, which may explain the increased susceptibility of smokers to developing severe COVID-19. This is at least partially explained by differences in interferon (IFN)-β1 levels between smokers and non-smokers.
Collapse
Affiliation(s)
- Tsuguhisa Nakayama
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Road, Stanford, CA, USA
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Ivan T. Lee
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Road, Stanford, CA, USA
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sizun Jiang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Matthias S. Matter
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Carol H. Yan
- Division of Otolaryngology – Head and Neck Surgery, Department of Surgery, University of California San Diego School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jonathan B. Overdevest
- Department of Otolaryngology–Head and Neck Surgery, Columbia University School of Medicine, New York, NY, USA
| | - Chien-Ting Wu
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yury Goltsev
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Liang-Chun Shih
- Department of Otorhinolaryngology, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Terry Fox Cancer Research Laboratory, Translational Medicine Center, China Medical University Hospital, Taichung, Taiwan
| | - Chun-Kang Liao
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Bokai Zhu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yunhao Bai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Peter Lidsky
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Yinghong Xiao
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - David Zarabanda
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Road, Stanford, CA, USA
| | - Angela Yang
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Road, Stanford, CA, USA
| | - Meena Easwaran
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Road, Stanford, CA, USA
| | - Christian M. Schürch
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Pauline Chu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Han Chen
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anna K. Stalder
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - David R. McIlwain
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Nicole A. Borchard
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Road, Stanford, CA, USA
| | - Phillip A. Gall
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Road, Stanford, CA, USA
| | - Sachi S. Dholakia
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Road, Stanford, CA, USA
| | - Wei Le
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Road, Stanford, CA, USA
| | - Le Xu
- Department of Pediatrics, University of California San Diego School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Chih-Jaan Tai
- Department of Otorhinolaryngology, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Te-Huei Yeh
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Otolaryngology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Elizabeth Erickson-Direnzo
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Road, Stanford, CA, USA
| | - Jason M. Duran
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Kirsten D. Mertz
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Peter H. Hwang
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Road, Stanford, CA, USA
| | - Jasmin D. Haslbauer
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Peter K. Jackson
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas Menter
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Raul Andino
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Peter D. Canoll
- Department of Pathology and Cell Biology, Columbia University Medical Center, Irving Cancer Research Center, New York, NY, USA
| | - Adam S. DeConde
- Division of Otolaryngology – Head and Neck Surgery, Department of Surgery, University of California San Diego School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Zara M. Patel
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Road, Stanford, CA, USA
| | - Alexandar Tzankov
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Garry P. Nolan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jayakar V. Nayak
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Road, Stanford, CA, USA
- Department of Otolaryngology – Head and Neck Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
67
|
Wang T, Wei F, Liu L, Sun Y, Song J, Wang M, Yang J, Li C, Liu J. Recombinant HA1-ΔfliC enhances adherence to respiratory epithelial cells and promotes the superiorly protective immune responses against H9N2 influenza virus in chickens. Vet Microbiol 2021; 262:109238. [PMID: 34560407 DOI: 10.1016/j.vetmic.2021.109238] [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] [Received: 06/16/2021] [Accepted: 09/11/2021] [Indexed: 01/17/2023]
Abstract
H9N2 subtype avian influenza virus (AIV) is an ongoing threat causing substantial loss to the poultry industry and thus necessitating the development of safe and effective vaccines against AIV. Given that inactivated vaccines are less effective in activating the mucosal immune system, we aimed to generate a vaccine that can actively engage the mucosal immunity which is the front line of the immune system. We generated a group of flagellin-based hemagglutinin globular head (HA1) fusion proteins and characterized their immunogenicity and efficacy. We found that Salmonella typhimurium flagellin (fliC) lacking the hypervariable domain (called herein as HA1-ΔfliC) was recognized by TLR5 and induced a moderate innate immune response compared to N-terminus of fliC (HA1-fliC) and C-terminus of fliC (fliC-HA1). The HA1-ΔfliC protein had increased adherence to the nasal cavity and trachea than HA1-fliC and fliC-HA1 and significantly increased the HA-specific sIgA titers. Our in vivo results revealed that chickens treated with HA1-ΔfliC had a significantly reduced level of viral loads in the cloaca and throat compared with chickens treated with inactivated vaccine. Overall, these results revealed that HA1-ΔfliC can protect chickens against H9N2 AIV by eliciting the efficient mucosal immune responses.
Collapse
Affiliation(s)
- Tong Wang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100094, China
| | - Fanhua Wei
- College of Agriculture, Ningxia University, Yinchuan, 750021, China
| | - Litao Liu
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100094, China
| | - Yan Sun
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100094, China
| | - Jingwei Song
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100094, China
| | - Mingyang Wang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100094, China
| | - Jizhe Yang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100094, China
| | - Chengye Li
- College of Agriculture, Ningxia University, Yinchuan, 750021, China
| | - Jinhua Liu
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100094, China.
| |
Collapse
|
68
|
The potential of neuraminidase as an antigen for nasal vaccines to increase cross-protection against influenza viruses. J Virol 2021; 95:e0118021. [PMID: 34379511 DOI: 10.1128/jvi.01180-21] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Despite the availability of vaccines that efficiently reduce the severity of clinical symptoms, influenza viruses still cause substantial morbidity and mortality worldwide. In this regard, nasal influenza vaccines-because they induce virus-specific IgA-may be more effective than traditional parenteral formulations in preventing infection of the upper respiratory tract. In addition, the neuraminidase (NA) of influenza virus has shown promise as a vaccine antigen to confer broad cross-protection, in contrast to hemagglutinin (HA), the target of most current vaccines, which undergoes frequent antigenic changes leading to vaccine ineffectiveness against mismatched heterologous strains. However, the usefulness of NA as an antigen for nasal vaccines is unclear. Here, we compared NA and HA as antigens for nasal vaccines in mice. Intranasal immunization with recombinant NA (rNA) plus adjuvant protected mice against not only homologous but also heterologous virus challenge in the upper respiratory tract, whereas intranasal immunization with rHA failed to protect against heterologous challenge. In addition, intranasal immunization with rNA, but not rHA, conferred cross-protection even in the absence of adjuvant in virus infection-experienced mice; this strong cross-protection was due to the broader binding capacity of NA-specific antibodies to heterologous virus. Furthermore, the NA-specific IgA in the upper respiratory tract that was induced through rNA intranasal immunization recognized more epitopes than did the NA-specific IgG and IgA in plasma, again increasing cross-protection. Together, our findings suggest the potential of NA as an antigen for nasal vaccines to provide broad cross-protection against both homologous and heterologous influenza viruses. IMPORTANCE Because mismatch between vaccine strains and epidemic strains cannot always be avoided, the development of influenza vaccines that induce broad cross-protection against antigenically mismatched heterologous strains is needed. Although the importance of NA-specific antibodies to cross-protection in humans and experimental animals is becoming clear, the potential of NA as an antigen for providing cross-protection through nasal vaccines is unknown. We show here that intranasal immunization with NA confers broad cross-protection in the upper respiratory tract, where virus transmission is initiated, by inducing NA-specific IgA that recognizes a wide range of epitopes. These data shed new light on NA-based nasal vaccines as powerful anti-influenza tools that confer broad cross-protection.
Collapse
|
69
|
Port J, Yinda CK, Avanzato V, Schulz J, Holbrook M, van Doremalen N, Shaia C, Fischer R, Munster V. Increased aerosol transmission for B.1.1.7 (alpha variant) over lineage A variant of SARS-CoV-2. RESEARCH SQUARE 2021:rs.3.rs-753550. [PMID: 34401871 PMCID: PMC8366800 DOI: 10.21203/rs.3.rs-753550/v1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Airborne transmission, a term combining both large droplet and aerosol transmission, is thought to be the main transmission route of SARS-CoV-2. Here we investigated the relative efficiency of aerosol transmission of two variants of SARS-CoV-2, B.1.1.7 (alpha) and lineage A, in the Syrian hamster. A novel transmission caging setup was designed and validated, which allowed the assessment of transmission efficiency at various distances. At 2 meters distance, only particles <5 µm traversed between cages. In this setup, aerosol transmission was confirmed in 8 out of 8 (N = 4 for each variant) sentinels after 24 hours of exposure as demonstrated by respiratory shedding and seroconversion. Successful transmission occurred even when exposure time was limited to one hour, highlighting the efficiency of this transmission route. Interestingly, the B.1.1.7 variant outcompeted the lineage A variant in an airborne transmission chain after mixed infection of donors. Combined, this data indicates that the infectious dose of B.1.1.7 required for successful transmission may be lower than that of lineage A virus. The experimental proof for true aerosol transmission and the increase in the aerosol transmission potential of B.1.1.7 underscore the continuous need for assessment of novel variants and the development or preemptive transmission mitigation strategies.
Collapse
Affiliation(s)
- Julia Port
- National Institute of Allergy and Infectious Diseases
| | | | | | | | | | | | - Carl Shaia
- National Institute of Allergy and Infectious Diseases
| | | | | |
Collapse
|
70
|
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) emerged as a virus with a pathogenicity closer to Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and Middle East Respiratory Syndrome Coronavirus (MERS-CoV) and a transmissibility similar to common cold coronaviruses (CoVs). In this review, we briefly discuss the features of the receptor-binding domain (RBD) and protease cleavage of the SARS-CoV-2 spike protein that enable SARS-CoV-2 to be a pandemic virus.
Collapse
Affiliation(s)
- Michelle N. Vu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Vineet D. Menachery
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
71
|
Abstract
Human respiratory virus infections lead to a spectrum of respiratory symptoms and disease severity, contributing to substantial morbidity, mortality and economic losses worldwide, as seen in the COVID-19 pandemic. Belonging to diverse families, respiratory viruses differ in how easy they spread (transmissibility) and the mechanism (modes) of transmission. Transmissibility as estimated by the basic reproduction number (R0) or secondary attack rate is heterogeneous for the same virus. Respiratory viruses can be transmitted via four major modes of transmission: direct (physical) contact, indirect contact (fomite), (large) droplets and (fine) aerosols. We know little about the relative contribution of each mode to the transmission of a particular virus in different settings, and how its variation affects transmissibility and transmission dynamics. Discussion on the particle size threshold between droplets and aerosols and the importance of aerosol transmission for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza virus is ongoing. Mechanistic evidence supports the efficacies of non-pharmaceutical interventions with regard to virus reduction; however, more data are needed on their effectiveness in reducing transmission. Understanding the relative contribution of different modes to transmission is crucial to inform the effectiveness of non-pharmaceutical interventions in the population. Intervening against multiple modes of transmission should be more effective than acting on a single mode.
Collapse
Affiliation(s)
- Nancy H L Leung
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
72
|
Port JR, Yinda CK, Avanzato VA, Schulz JE, Holbrook MG, van Doremalen N, Shaia C, Fischer RJ, Munster VJ. Increased aerosol transmission for B.1.1.7 (alpha variant) over lineage A variant of SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.07.26.453518. [PMID: 34341792 PMCID: PMC8328059 DOI: 10.1101/2021.07.26.453518] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Airborne transmission, a term combining both large droplet and aerosol transmission, is thought to be the main transmission route of SARS-CoV-2. Here we investigated the relative efficiency of aerosol transmission of two variants of SARS-CoV-2, B.1.1.7 (alpha) and lineage A, in the Syrian hamster. A novel transmission caging setup was designed and validated, which allowed the assessment of transmission efficiency at various distances. At 2 meters distance, only particles <5 µm traversed between cages. In this setup, aerosol transmission was confirmed in 8 out of 8 (N = 4 for each variant) sentinels after 24 hours of exposure as demonstrated by respiratory shedding and seroconversion. Successful transmission occurred even when exposure time was limited to one hour, highlighting the efficiency of this transmission route. Interestingly, the B.1.1.7 variant outcompeted the lineage A variant in an airborne transmission chain after mixed infection of donors. Combined, this data indicates that the infectious dose of B.1.1.7 required for successful transmission may be lower than that of lineage A virus. The experimental proof for true aerosol transmission and the increase in the aerosol transmission potential of B.1.1.7 underscore the continuous need for assessment of novel variants and the development or preemptive transmission mitigation strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Vincent J. Munster
- Materials and Correspondence: All material requests should be sent to Vincent J. Munster,
| |
Collapse
|
73
|
Perspective of the Relationship between the Susceptibility to Initial SARS-CoV-2 Infectivity and Optimal Nasal Conditioning of Inhaled Air. Int J Mol Sci 2021; 22:ijms22157919. [PMID: 34360686 PMCID: PMC8348706 DOI: 10.3390/ijms22157919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), as with the influenza virus, has been shown to spread more rapidly during winter. Severe coronavirus disease 2019 (COVID-19), which can follow SARS-CoV-2 infection, disproportionately affects older persons and males as well as people living in temperate zone countries with a tropical ancestry. Recent evidence on the importance of adequately warming and humidifying (conditioning) inhaled air in the nasal cavity for reducing SARS-CoV-2 infectivity in the upper respiratory tract (URT) is discussed, with particular reference to: (i) the relevance of air-borne SARS-CoV-2 transmission, (ii) the nasal epithelium as the initial site of SARS-CoV-2 infection, (iii) the roles of type 1 and 3 interferons for preventing viral infection of URT epithelial cells, (iv) weaker innate immune responses to respiratory viral infections in URT epithelial cells at suboptimal temperature and humidity, and (v) early innate immune responses in the URT for limiting and eliminating SARS-CoV-2 infections. The available data are consistent with optimal nasal air conditioning reducing SARS-CoV-2 infectivity of the URT and, as a consequence, severe COVID-19. Further studies on SARS-CoV-2 infection rates and viral loads in the nasal cavity and nasopharynx in relation to inhaled air temperature, humidity, age, gender, and genetic background are needed in this context. Face masks used for reducing air-borne virus transmission can also promote better nasal air conditioning in cold weather. Masks can, thereby, minimise SARS-CoV-2 infectivity and are particularly relevant for protecting more vulnerable persons from severe COVID-19.
Collapse
|
74
|
Kim T, Lee JS, Ju YS. Experimental Models for SARS-CoV-2 Infection. Mol Cells 2021; 44:377-383. [PMID: 34187969 PMCID: PMC8245318 DOI: 10.14348/molcells.2021.0094] [Citation(s) in RCA: 7] [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: 04/14/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) is a novel virus that causes coronavirus disease 2019 (COVID-19). To understand the identity, functional characteristics and therapeutic targets of the virus and the diseases, appropriate infection models that recapitulate the in vivo pathophysiology of the viral infection are necessary. This article reviews the various infection models, including Vero cells, human cell lines, organoids, and animal models, and discusses their advantages and disadvantages. This knowledge will be helpful for establishing an efficient system for defense against emerging infectious diseases.
Collapse
Affiliation(s)
- Taewoo Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | | | - Young Seok Ju
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- GENOME INSIGHT Inc., Daejeon 34051, Korea
| |
Collapse
|
75
|
Ma Y, Mao G, Wu G, Chen M, Qin F, Zheng L, Zhang XE. Dual-Fluorescence Labeling Pseudovirus for Real-Time Imaging of Single SARS-CoV-2 Entry in Respiratory Epithelial Cells. ACS APPLIED MATERIALS & INTERFACES 2021; 13:24477-24486. [PMID: 33961399 PMCID: PMC8117782 DOI: 10.1021/acsami.1c03897] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/25/2021] [Indexed: 05/12/2023]
Abstract
The pseudovirus strategy makes studies of highly pathogenic viruses feasible without the restriction of high-level biosafety facility, thus greatly contributing to virology and is used in the research studies of SARS-CoV-2. Here, we generated a dual-color pseudo-SARS-CoV-2 virus using a human immunodeficiency virus-1 pseudovirus production system and the SARS-CoV-2 spike (S) glycoprotein, of which the membrane was labeled with a lipophilic dye (DiO) and the genomic RNA-related viral protein R (Vpr) of the viral core was fused with mCherry. With this dual-color labeling strategy, not only the movement of the whole virus but also the fate of the labeled components can be traced. The pseudovirions were applied to track the viral entry at a single-particle level in four types of the human respiratory cells: nasal epithelial cells (HNEpC), pulmonary alveolar epithelial cells (HPAEpiC), bronchial epithelial cells (BEP-2D), and oral epithelial cells (HOEC). Pseudo-SARS-CoV-2 entered into the host cell and released the viral core into the cytoplasm, which clearly indicates that the host entry mainly occurred through endocytosis. The infection efficiency was found to be correlated with the expression of the known receptor of SARS-CoV-2, angiotensin-converting 2 (ACE2) on the host cell surface. We believe that the dual-color fluorescently labeled pseudovirus system created in this study can be applied as a useful tool for many purposes in SARS-CoV-2/COVID-19.
Collapse
Affiliation(s)
- Yingxin Ma
- CAS Key Laboratory of Quantitative Engineering
Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced
Technology, Chinese Academy of Sciences, Shenzhen 518055,
China
| | - Guobin Mao
- CAS Key Laboratory of Quantitative Engineering
Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced
Technology, Chinese Academy of Sciences, Shenzhen 518055,
China
| | - Guoqiang Wu
- CAS Key Laboratory of Quantitative Engineering
Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced
Technology, Chinese Academy of Sciences, Shenzhen 518055,
China
| | - Minghai Chen
- CAS Key Laboratory of Quantitative Engineering
Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced
Technology, Chinese Academy of Sciences, Shenzhen 518055,
China
| | - Fujun Qin
- CAS Key Laboratory of Quantitative Engineering
Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced
Technology, Chinese Academy of Sciences, Shenzhen 518055,
China
| | - Luping Zheng
- CAS Key Laboratory of Quantitative Engineering
Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced
Technology, Chinese Academy of Sciences, Shenzhen 518055,
China
| | - Xian-En Zhang
- CAS Key Laboratory of Quantitative Engineering
Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced
Technology, Chinese Academy of Sciences, Shenzhen 518055,
China
- National Key Laboratory of Biomacromolecules, CAS
Center for Biological Macromolecules, Institute of Biophysics, Chinese
Academy of Sciences, Beijing 100101, China
- Faculty of Synthetic Biology, Shenzhen Institutes of
Advanced Technology, Chinese Academy of Sciences, Shenzhen
518055, China
| |
Collapse
|
76
|
Mather MW, Jardine L, Talks B, Gardner L, Haniffa M. Complexity of immune responses in COVID-19. Semin Immunol 2021; 55:101545. [PMID: 34865933 PMCID: PMC8626289 DOI: 10.1016/j.smim.2021.101545] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022]
Abstract
The global COVID-19 pandemic has caused substantial morbidity and mortality to humanity. Remarkable progress has been made in understanding both the innate and adaptive mechanisms involved in the host response to the causative SARS-CoV-2 virus, but much remains to be discovered. Robust upper airway defenses are critical in restricting SARS-CoV-2 replication and propagation. Further, the nasal abundance of viral uptake receptor, ACE2, and the host epithelial transcriptional landscape, are associated with differential disease outcomes across different patient cohorts. The adaptive host response to systemic COVID-19 is heterogeneous and complex. Blunted responses to interferon and robust cytokine generation are hallmarks of the disease, particularly at the advanced stages. Excessive immune cell influx into tissues can lead to substantial collateral damage to the host akin to sepsis. This review offers a contemporary summary of these mechanisms of disease and highlights potential avenues for diagnostic and therapeutic development. These include improved disease stratification, targeting effectors of immune-mediated tissue damage, and blunting of immune cell-mediated tissue damage.
Collapse
Affiliation(s)
- Michael William Mather
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; Department of Otolaryngology, Freeman Hospital, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, NE7 7DN, UK
| | - Laura Jardine
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; Haematology Department, Freeman Hospital, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, NE7 7DN, UK
| | - Ben Talks
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; Department of Otolaryngology, Freeman Hospital, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, NE7 7DN, UK
| | - Louis Gardner
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE2 4LP, UK
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE2 4LP, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
| |
Collapse
|
77
|
Nguyen TQ, Rollon R, Choi YK. Animal Models for Influenza Research: Strengths and Weaknesses. Viruses 2021; 13:1011. [PMID: 34071367 PMCID: PMC8228315 DOI: 10.3390/v13061011] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/16/2022] Open
Abstract
Influenza remains one of the most significant public health threats due to its ability to cause high morbidity and mortality worldwide. Although understanding of influenza viruses has greatly increased in recent years, shortcomings remain. Additionally, the continuous mutation of influenza viruses through genetic reassortment and selection of variants that escape host immune responses can render current influenza vaccines ineffective at controlling seasonal epidemics and potential pandemics. Thus, there is a knowledge gap in the understanding of influenza viruses and a corresponding need to develop novel universal vaccines and therapeutic treatments. Investigation of viral pathogenesis, transmission mechanisms, and efficacy of influenza vaccine candidates requires animal models that can recapitulate the disease. Furthermore, the choice of animal model for each research question is crucial in order for researchers to acquire a better knowledge of influenza viruses. Herein, we reviewed the advantages and limitations of each animal model-including mice, ferrets, guinea pigs, swine, felines, canines, and non-human primates-for elucidating influenza viral pathogenesis and transmission and for evaluating therapeutic agents and vaccine efficacy.
Collapse
Affiliation(s)
- Thi-Quyen Nguyen
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju 28644, Korea; (T.-Q.N.); (R.R.)
| | - Rare Rollon
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju 28644, Korea; (T.-Q.N.); (R.R.)
| | - Young-Ki Choi
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju 28644, Korea; (T.-Q.N.); (R.R.)
- Zoonotic Infectious Diseases Research Center, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
78
|
Xie C, Su W, Sia SF, Choy KT, Morrell S, Zhou J, Peiris M, Bloom J, Yen HL. A(H1N1)pdm09 influenza viruses replicating in ferret upper or lower respiratory tract differed in onward transmission potential by air. J Infect Dis 2021; 225:65-74. [PMID: 34036370 DOI: 10.1093/infdis/jiab286] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/21/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND A(H1N1)pdm09 influenza viruses replicate efficiently in respiratory epithelia and are transmitted via respiratory droplets and aerosols expelled by infected hosts. The relative onward transmission potential of influenza viruses replicating in the upper and lower respiratory epithelial cells has not been fully defined. METHODS Wild-type and barcoded A(H1N1)pdm09 viruses that differed by 2 synonymous mutations per gene segment were inoculated into ferrets via intra-nasal and intra-tracheal routes. Naïve recipients were exposed to the exhaled breath of inoculated donors for 8 hours on day 2 post-inoculation. Onward transmission potential of wild-type and barcoded genotypes were monitored by next generation sequencing. RESULTS Transmissible airborne particles were respired from the upper but not the lower respiratory epithelial cells of donor ferrets. There was limited mixing of viral populations replicating in the upper and lower respiratory tissues. CONCLUSIONS The ferret upper respiratory epithelium was mapped as the anatomic site that generated influenza virus-laden particles mediating onward transmission by air. Our results suggest that vaccines and antivirals should aim to reduce viral loads in the upper respiratory tract for prevention of influenza transmission.
Collapse
Affiliation(s)
- Chenyi Xie
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Wen Su
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Sin Fun Sia
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Ka-Tim Choy
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Steven Morrell
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Jie Zhou
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Jesse Bloom
- Basic Sciences and Computational Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Howard Hughes Medical Institutes, Seattle, WA, USA
| | - Hui-Ling Yen
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
79
|
Hein MD, Arora P, Marichal-Gallardo P, Winkler M, Genzel Y, Pöhlmann S, Schughart K, Kupke SY, Reichl U. Cell culture-based production and in vivo characterization of purely clonal defective interfering influenza virus particles. BMC Biol 2021; 19:91. [PMID: 33941189 PMCID: PMC8091782 DOI: 10.1186/s12915-021-01020-5] [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: 08/20/2020] [Accepted: 04/01/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Infections with influenza A virus (IAV) cause high morbidity and mortality in humans. Additional to vaccination, antiviral drugs are a treatment option. Besides FDA-approved drugs such as oseltamivir or zanamivir, virus-derived defective interfering (DI) particles (DIPs) are considered promising new agents. IAV DIPs typically contain a large internal deletion in one of their eight genomic viral RNA (vRNA) segments. Consequently, DIPs miss the genetic information necessary for replication and can usually only propagate by co-infection with infectious standard virus (STV), compensating for their defect. In such a co-infection scenario, DIPs interfere with and suppress STV replication, which constitutes their antiviral potential. RESULTS In the present study, we generated a genetically engineered MDCK suspension cell line for production of a purely clonal DIP preparation that has a large deletion in its segment 1 (DI244) and is not contaminated with infectious STV as egg-derived material. First, the impact of the multiplicity of DIP (MODIP) per cell on DI244 yield was investigated in batch cultivations in shake flasks. Here, the highest interfering efficacy was observed for material produced at a MODIP of 1E-2 using an in vitro interference assay. Results of RT-PCR suggested that DI244 material produced was hardly contaminated with other defective particles. Next, the process was successfully transferred to a stirred tank bioreactor (500 mL working volume) with a yield of 6.0E+8 PFU/mL determined in genetically modified adherent MDCK cells. The produced material was purified and concentrated about 40-fold by membrane-based steric exclusion chromatography (SXC). The DI244 yield was 92.3% with a host cell DNA clearance of 97.1% (99.95% with nuclease digestion prior to SXC) and a total protein reduction of 97.2%. Finally, the DIP material was tested in animal experiments in D2(B6).A2G-Mx1r/r mice. Mice infected with a lethal dose of IAV and treated with DIP material showed a reduced body weight loss and all animals survived. CONCLUSION In summary, experiments not only demonstrated that purely clonal influenza virus DIP preparations can be obtained with high titers from animal cell cultures but confirmed the potential of cell culture-derived DIPs as an antiviral agent.
Collapse
Affiliation(s)
- Marc D Hein
- Otto-von-Guericke-University Magdeburg, Chair of Bioprocess Engineering, Magdeburg, Germany
| | - Prerna Arora
- German Primate Center-Leibniz Institute for Primate Research, Infection Biology Unit, Göttingen, Germany.,University Göttingen, Faculty of Biology and Psychology, Göttingen, Germany
| | - Pavel Marichal-Gallardo
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Michael Winkler
- German Primate Center-Leibniz Institute for Primate Research, Infection Biology Unit, Göttingen, Germany.,University Göttingen, Faculty of Biology and Psychology, Göttingen, Germany
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Stefan Pöhlmann
- German Primate Center-Leibniz Institute for Primate Research, Infection Biology Unit, Göttingen, Germany.,University Göttingen, Faculty of Biology and Psychology, Göttingen, Germany
| | - Klaus Schughart
- Helmholtz Centre for Infection Research, Department of Infection Genetics, Braunschweig, Germany.,University of Veterinary Medicine Hannover, Hannover, Germany.,University of Tennessee Health Science Center, Department of Microbiology, Immunology and Biochemistry, Memphis, TN, USA
| | - Sascha Y Kupke
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany.
| | - Udo Reichl
- Otto-von-Guericke-University Magdeburg, Chair of Bioprocess Engineering, Magdeburg, Germany.,Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| |
Collapse
|
80
|
Glezer I, Bruni‐Cardoso A, Schechtman D, Malnic B. Viral infection and smell loss: The case of COVID-19. J Neurochem 2021; 157:930-943. [PMID: 32970861 PMCID: PMC7537178 DOI: 10.1111/jnc.15197] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/19/2022]
Abstract
Olfactory disorders have been increasingly reported in individuals infected with SARS-CoV-2, the virus causing the coronavirus disease 2019 (COVID-19). Losing the sense of smell has a strong impact on the quality of life, since it may lead to malnutrition, weight loss, food poisoning, depression, and exposure to dangerous chemicals. Individuals who suffer from anosmia (inability to smell) also cannot sense the flavor of food, which is a combination of taste and smell. Interestingly, infected individuals have reported sudden loss of smell with no congested nose, as is frequently observed in common colds or other upper respiratory tract infections. These observations suggest that SARS-CoV-2 infection leads to olfactory loss through a distinct mechanism, which is still unclear. This article provides an overview of olfactory loss and the recent findings relating to COVID-19. Possible mechanisms of SARS-CoV-2-induced olfactory loss are also discussed.
Collapse
Affiliation(s)
- Isaias Glezer
- Department of BiochemistryUNIFESPEscola Paulista de MedicinaUniversidade Federal de São PauloRua Tres de MaioSão PauloBrazil
| | | | | | - Bettina Malnic
- Department of BiochemistryUniversity of São PauloSão PauloBrazil
| |
Collapse
|
81
|
Ziegler P, Tian Y, Bai Y, Abrahamsson S, Bäckerholm A, Reznik AS, Green A, Moore JA, Lee SE, Myerburg MM, Park HJ, Tang KW, Shair KHY. A primary nasopharyngeal three-dimensional air-liquid interface cell culture model of the pseudostratified epithelium reveals differential donor- and cell type-specific susceptibility to Epstein-Barr virus infection. PLoS Pathog 2021; 17:e1009041. [PMID: 33914843 PMCID: PMC8112674 DOI: 10.1371/journal.ppat.1009041] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 05/11/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023] Open
Abstract
Epstein-Barr virus (EBV) is a ubiquitous γ-herpesvirus with latent and lytic cycles. EBV replicates in the stratified epithelium but the nasopharynx is also composed of pseudostratified epithelium with distinct cell types. Latent infection is associated with nasopharyngeal carcinoma (NPC). Here, we show with nasopharyngeal conditionally reprogrammed cells cultured at the air-liquid interface that pseudostratified epithelial cells are susceptible to EBV infection. Donors varied in susceptibility to de novo EBV infection, but susceptible cultures also displayed differences with respect to pathogenesis. The cultures from one donor yielded lytic infection but cells from two other donors were positive for EBV-encoded EBERs and negative for other lytic infection markers. All cultures stained positive for the pseudostratified markers CK7, MUC5AC, α-tubulin in cilia, and the EBV epithelial cell receptor Ephrin receptor A2. To define EBV transcriptional programs by cell type and to elucidate latent/lytic infection-differential changes, we performed single cell RNA-sequencing on one EBV-infected culture that resulted in alignment with many EBV transcripts. EBV transcripts represented a small portion of the total transcriptome (~0.17%). All cell types in the pseudostratified epithelium had detectable EBV transcripts with suprabasal cells showing the highest number of reads aligning to many EBV genes. Several restriction factors (IRF1, MX1, STAT1, C18orf25) known to limit lytic infection were expressed at lower levels in the lytic subcluster. A third of the differentially-expressed genes in NPC tumors compared to an uninfected pseudostratified ALI culture overlapped with the differentially-expressed genes in the latent subcluster. A third of these commonly perturbed genes were specific to EBV infection and changed in the same direction. Collectively, these findings suggest that the pseudostratified epithelium could harbor EBV infection and that the pseudostratified infection model mirrors many of the transcriptional changes imposed by EBV infection in NPC. It has been known for over 50 years that EBV infection is associated with NPC. Despite many advances from studies in 2-dimensional cell culture, many aspects of EBV molecular pathogenesis in the nasopharynx remain undefined because the cell types and the biology of the nasopharyngeal epithelium can only be faithfully captured in 3-dimensional cell culture. In the stratified epithelium, cellular differentiation triggers lytic infection but it is not clear to what degree the pseudostratified epithelium is involved. The pseudostratified epithelium is abundant in the lateral wall where the lymphoid-rich fossa of Rosenmüller is located and is a site where NPC tumors most often arises. While the oral epithelium is a site of EBV replication, whether the nasopharyngeal epithelium is a major source of EBV shedding in the nasopharynx is not well defined. Here, we present a 3-dimensional organoid model of the nasopharyngeal pseudostratified epithelium showing that such cells can be infected with EBV in some donor cultures, with examples of both latent and lytic infection. We propose that the cell types of the pseudostratified epithelium should be considered a component of EBV pathogenesis in the nasopharynx and that the difference in donor susceptibility and latent/lytic infection could influence EBV’s fitness in the nasopharynx.
Collapse
Affiliation(s)
- Phillip Ziegler
- Cancer Virology Program, University of Pittsburgh Medical Center (UPMC), University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Yarong Tian
- Wallenberg Centre for Molecular and Translational Medicine, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Yulong Bai
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Sanna Abrahamsson
- Wallenberg Centre for Molecular and Translational Medicine, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alan Bäckerholm
- Wallenberg Centre for Molecular and Translational Medicine, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alex S. Reznik
- Cancer Virology Program, University of Pittsburgh Medical Center (UPMC), University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Anthony Green
- University of Pittsburgh Research Histology Services, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - John A. Moore
- UPMC Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stella E. Lee
- UPMC Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Michael M. Myerburg
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Hyun Jung Park
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Ka-Wei Tang
- Wallenberg Centre for Molecular and Translational Medicine, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Microbiology, Gothenburg, Sweden
| | - Kathy Ho Yen Shair
- Cancer Virology Program, University of Pittsburgh Medical Center (UPMC), University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
82
|
Russell CJ. Hemagglutinin Stability and Its Impact on Influenza A Virus Infectivity, Pathogenicity, and Transmissibility in Avians, Mice, Swine, Seals, Ferrets, and Humans. Viruses 2021; 13:746. [PMID: 33923198 PMCID: PMC8145662 DOI: 10.3390/v13050746] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
Genetically diverse influenza A viruses (IAVs) circulate in wild aquatic birds. From this reservoir, IAVs sporadically cause outbreaks, epidemics, and pandemics in wild and domestic avians, wild land and sea mammals, horses, canines, felines, swine, humans, and other species. One molecular trait shown to modulate IAV host range is the stability of the hemagglutinin (HA) surface glycoprotein. The HA protein is the major antigen and during virus entry, this trimeric envelope glycoprotein binds sialic acid-containing receptors before being triggered by endosomal low pH to undergo irreversible structural changes that cause membrane fusion. The HA proteins from different IAV isolates can vary in the pH at which HA protein structural changes are triggered, the protein causes membrane fusion, or outside the cell the virion becomes inactivated. HA activation pH values generally range from pH 4.8 to 6.2. Human-adapted HA proteins tend to have relatively stable HA proteins activated at pH 5.5 or below. Here, studies are reviewed that report HA stability values and investigate the biological impact of variations in HA stability on replication, pathogenicity, and transmissibility in experimental animal models. Overall, a stabilized HA protein appears to be necessary for human pandemic potential and should be considered when assessing human pandemic risk.
Collapse
Affiliation(s)
- Charles J Russell
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105-3678, USA
| |
Collapse
|
83
|
Schepler H, Wang X, Neufurth M, Wang S, Schröder HC, Müller WEG. The therapeutic potential of inorganic polyphosphate: A versatile physiological polymer to control coronavirus disease (COVID-19). Theranostics 2021; 11:6193-6213. [PMID: 33995653 PMCID: PMC8120197 DOI: 10.7150/thno.59535] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/19/2021] [Indexed: 12/15/2022] Open
Abstract
Rationale: The pandemic caused by the novel coronavirus SARS-CoV-2 is advancing rapidly. In particular, the number of severe courses of the disease is still dramatically high. An efficient drug therapy that helps to improve significantly the fatal combination of damages in the airway epithelia, in the extensive pulmonary microvascularization and finally multiorgan failure, is missing. The physiological, inorganic polymer, polyphosphate (polyP) is a molecule which could prevent the initial phase of the virus life cycle, the attachment of the virus to the target cells, and improve the epithelial integrity as well as the mucus barrier. Results: Surprisingly, polyP matches perfectly with the cationic groove on the RBD. Subsequent binding studies disclosed that polyP, with a physiological chain length of 40 phosphate residues, abolishes the binding propensity of the RBD to the ACE2 receptor. In addition to this first mode of action of polyP, this polymer causes in epithelial cells an increased gene expression of the major mucins in the airways, of MUC5AC and MUC1, as well as a subsequent glycoprotein production. MUC5AC forms a gel-like mucus layer trapping inhaled particles which are then transported out of the airways, while MUC1 constitutes the periciliary liquid layer and supports ciliary beating. As a third mode of action, polyP undergoes enzymatic hydrolysis of the anhydride bonds in the airway system by alkaline phosphatase, releasing metabolic energy. Conclusions: This review summarizes the state of the art of the biotherapeutic potential of the polymer polyP and the findings from basic research and outlines future biomedical applications.
Collapse
Affiliation(s)
- Hadrian Schepler
- Department of Dermatology, University Clinic Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Xiaohong Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, 55128 Mainz, Germany
| | - Meik Neufurth
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, 55128 Mainz, Germany
| | - Shunfeng Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, 55128 Mainz, Germany
| | - Heinz C. Schröder
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, 55128 Mainz, Germany
| | - Werner E. G. Müller
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, 55128 Mainz, Germany
| |
Collapse
|
84
|
Overeem NJ, van der Vries E, Huskens J. A Dynamic, Supramolecular View on the Multivalent Interaction between Influenza Virus and Host Cell. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2007214. [PMID: 33682339 DOI: 10.1002/smll.202007214] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/11/2021] [Indexed: 06/12/2023]
Abstract
Understanding how influenza viruses traverse the mucus and recognize host cells is critical for evaluating their zoonotic potential, and for prevention and treatment of the disease. The surface of the influenza A virus is covered with the receptor-binding protein hemagglutinin and the receptor-cleaving enzyme neuraminidase, which jointly control the interactions between the virus and the host cell. These proteins are organized in closely spaced trimers and tetramers to facilitate multivalent interactions with sialic acid-terminated glycans. This review shows that the individually weak multivalent interactions of influenza viruses allow superselective binding, virus-induced recruitment of receptors, and the formation of dynamic complexes that facilitate molecular walking. Techniques to measure the avidity and receptor specificity of influenza viruses are reviewed, and the pivotal role of multivalent interactions with their emergent properties in crossing the mucus and entering host cells is discussed. A model is proposed for the initiation of cell entry through virus-induced receptor clustering. The multivalent interactions of influenza viruses are maintained in a dynamic regime by a functional balance between binding and cleaving.
Collapse
Affiliation(s)
- Nico J Overeem
- Department of Molecules and Materials, MESA+ Institute for Nanotechnology, Faculty of Science and Technology, University of Twente, P.O. Box 217, Enschede, 7500 AE, The Netherlands
| | - Erhard van der Vries
- Royal GD, Arnsbergstraat 7, Deventer, 7418 EZ, The Netherlands
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Jurriaan Huskens
- Department of Molecules and Materials, MESA+ Institute for Nanotechnology, Faculty of Science and Technology, University of Twente, P.O. Box 217, Enschede, 7500 AE, The Netherlands
| |
Collapse
|
85
|
Plante JA, Liu Y, Liu J, Xia H, Johnson BA, Lokugamage KG, Zhang X, Muruato AE, Zou J, Fontes-Garfias CR, Mirchandani D, Scharton D, Bilello JP, Ku Z, An Z, Kalveram B, Freiberg AN, Menachery VD, Xie X, Plante KS, Weaver SC, Shi PY. Spike mutation D614G alters SARS-CoV-2 fitness. Nature 2021; 592:116-121. [PMID: 33106671 PMCID: PMC8158177 DOI: 10.1038/s41586-020-2895-3] [Citation(s) in RCA: 1146] [Impact Index Per Article: 286.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/20/2020] [Indexed: 11/09/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein substitution D614G became dominant during the coronavirus disease 2019 (COVID-19) pandemic1,2. However, the effect of this variant on viral spread and vaccine efficacy remains to be defined. Here we engineered the spike D614G substitution in the USA-WA1/2020 SARS-CoV-2 strain, and found that it enhances viral replication in human lung epithelial cells and primary human airway tissues by increasing the infectivity and stability of virions. Hamsters infected with SARS-CoV-2 expressing spike(D614G) (G614 virus) produced higher infectious titres in nasal washes and the trachea, but not in the lungs, supporting clinical evidence showing that the mutation enhances viral loads in the upper respiratory tract of COVID-19 patients and may increase transmission. Sera from hamsters infected with D614 virus exhibit modestly higher neutralization titres against G614 virus than against D614 virus, suggesting that the mutation is unlikely to reduce the ability of vaccines in clinical trials to protect against COVID-19, and that therapeutic antibodies should be tested against the circulating G614 virus. Together with clinical findings, our work underscores the importance of this variant in viral spread and its implications for vaccine efficacy and antibody therapy.
Collapse
Affiliation(s)
- Jessica A Plante
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yang Liu
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jianying Liu
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Hongjie Xia
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Bryan A Johnson
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Kumari G Lokugamage
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Xianwen Zhang
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Antonio E Muruato
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jing Zou
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Camila R Fontes-Garfias
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Divya Mirchandani
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Dionna Scharton
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Birte Kalveram
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Alexander N Freiberg
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Vineet D Menachery
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA.
| | - Kenneth S Plante
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA.
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| | - Scott C Weaver
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA.
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA.
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA.
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX, USA.
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA.
| | - Pei-Yong Shi
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA.
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA.
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA.
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX, USA.
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
86
|
Shibuya M, Tamiya S, Kawai A, Yoshioka Y. Nasal-subcutaneous prime-boost regimen for inactivated whole-virus influenza vaccine efficiently protects mice against both upper and lower respiratory tract infections. Biochem Biophys Res Commun 2021; 554:166-172. [PMID: 33798943 DOI: 10.1016/j.bbrc.2021.03.099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/18/2021] [Indexed: 10/21/2022]
Abstract
Although influenza vaccines are effective for reducing viral transmission and the severity of clinical symptoms, influenza viruses still induce considerable morbidity and mortality worldwide. Seasonal influenza viruses infect the upper respiratory tract initially but then often induce severe pulmonary complications in the lower respiratory tract. Therefore, influenza vaccines that prevent viral infection at both the upper and lower respiratory tracts are highly anticipated. Here, we examined whether using different vaccination routes for priming and boosting achieved protection in both regions of the respiratory tract. To this end, we used inactivated whole-virion influenza vaccines to immunize mice either subcutaneously or intranasally for both priming and boosting. Regardless of the route used for boosting, the levels of virus-specific IgG in plasma were higher in mice primed subcutaneously than those in control mice, which received PBS only. In addition, intranasal priming followed by subcutaneous boosting induced higher levels of virus-specific IgG in plasma than those in control mice. The levels of virus-specific nasal IgA were higher in mice that were primed intranasally than in control mice or in mice primed subcutaneously. Furthermore, intranasal priming but not subcutaneous priming provided protection against viral challenge in the upper respiratory tract. In addition, when coupled with subcutaneous boosting, both subcutaneous and intranasal priming protected against viral challenge in the lower respiratory tract. These results indicate that intranasal priming followed by subcutaneous boosting induces both virus-specific IgG in plasma and IgA in nasal washes and protects against virus challenge in both the upper and lower respiratory tracts. Our results will help to develop novel vaccines against influenza viruses and other respiratory viruses.
Collapse
Affiliation(s)
- Meito Shibuya
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shigeyuki Tamiya
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Atsushi Kawai
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yasuo Yoshioka
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; The Research Foundation for Microbial Diseases of Osaka University (BIKEN), 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
87
|
Kutter JS, de Meulder D, Bestebroer TM, Lexmond P, Mulders A, Richard M, Fouchier RAM, Herfst S. SARS-CoV and SARS-CoV-2 are transmitted through the air between ferrets over more than one meter distance. Nat Commun 2021; 12:1653. [PMID: 33712573 PMCID: PMC7955093 DOI: 10.1038/s41467-021-21918-6] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/19/2021] [Indexed: 02/08/2023] Open
Abstract
SARS-CoV-2 emerged in late 2019 and caused a pandemic, whereas the closely related SARS-CoV was contained rapidly in 2003. Here, an experimental set-up is used to study transmission of SARS-CoV and SARS-CoV-2 through the air between ferrets over more than a meter distance. Both viruses cause a robust productive respiratory tract infection resulting in transmission of SARS-CoV-2 to two of four indirect recipient ferrets and SARS-CoV to all four. A control pandemic A/H1N1 influenza virus also transmits efficiently. Serological assays confirm all virus transmission events. Although the experiments do not discriminate between transmission via small aerosols, large droplets and fomites, these results demonstrate that SARS-CoV and SARS-CoV-2 can remain infectious while traveling through the air. Efficient virus transmission between ferrets is in agreement with frequent SARS-CoV-2 outbreaks in mink farms. Although the evidence for virus transmission via the air between humans under natural conditions is absent or weak for SARS-CoV and SARS-CoV-2, ferrets may represent a sensitive model to study interventions aimed at preventing virus transmission.
Collapse
Affiliation(s)
- Jasmin S Kutter
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dennis de Meulder
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Theo M Bestebroer
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Pascal Lexmond
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ard Mulders
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mathilde Richard
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ron A M Fouchier
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sander Herfst
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
88
|
Deliyannis G, Wong CY, McQuilten HA, Bachem A, Clarke M, Jia X, Horrocks K, Zeng W, Girkin J, Scott NE, Londrigan SL, Reading PC, Bartlett NW, Kedzierska K, Brown LE, Mercuri F, Demaison C, Jackson DC, Chua BY. TLR2-mediated activation of innate responses in the upper airways confers antiviral protection of the lungs. JCI Insight 2021; 6:140267. [PMID: 33561017 PMCID: PMC8021123 DOI: 10.1172/jci.insight.140267] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 02/03/2021] [Indexed: 12/21/2022] Open
Abstract
The impact of respiratory virus infections on global health is felt not just during a pandemic, but endemic seasonal infections pose an equal and ongoing risk of severe disease. Moreover, vaccines and antiviral drugs are not always effective or available for many respiratory viruses. We investigated how induction of effective and appropriate antigen-independent innate immunity in the upper airways can prevent the spread of respiratory virus infection to the vulnerable lower airways. Activation of TLR2, when restricted to the nasal turbinates, resulted in prompt induction of innate immune-driven antiviral responses through action of cytokines, chemokines, and cellular activity in the upper but not the lower airways. We have defined how nasal epithelial cells and recruitment of macrophages work in concert and play pivotal roles to limit progression of influenza virus to the lungs and sustain protection for up to 7 days. These results reveal underlying mechanisms of how control of viral infection in the upper airways can occur and support the implementation of strategies that can activate TLR2 in nasal passages to provide rapid protection, especially for at-risk populations, against severe respiratory infection when vaccines and antiviral drugs are not always effective or available.
Collapse
Affiliation(s)
- Georgia Deliyannis
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Chinn Yi Wong
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hayley A. McQuilten
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Annabell Bachem
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Michele Clarke
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Xiaoxiao Jia
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Kylie Horrocks
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Weiguang Zeng
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jason Girkin
- Viral Immunology and Respiratory Disease group, School of Biomedical Science and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Nichollas E. Scott
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sarah L. Londrigan
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Patrick C. Reading
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Nathan W. Bartlett
- Viral Immunology and Respiratory Disease group, School of Biomedical Science and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lorena E. Brown
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | | | | | - David C. Jackson
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Brendon Y. Chua
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
89
|
Sharma S, Pinto R, Saha A, Chaudhuri S, Basu S. On secondary atomization and blockage of surrogate cough droplets in single- and multilayer face masks. SCIENCE ADVANCES 2021; 7:eabf0452. [PMID: 33674314 PMCID: PMC7935372 DOI: 10.1126/sciadv.abf0452] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/21/2021] [Indexed: 05/18/2023]
Abstract
Face masks prevent transmission of infectious respiratory diseases by blocking large droplets and aerosols during exhalation or inhalation. While three-layer masks are generally advised, many commonly available or makeshift masks contain single or double layers. Using carefully designed experiments involving high-speed imaging along with physics-based analysis, we show that high-momentum, large-sized (>250 micrometer) surrogate cough droplets can penetrate single- or double-layer mask material to a significant extent. The penetrated droplets can atomize into numerous much smaller (<100 micrometer) droplets, which could remain airborne for a significant time. The possibility of secondary atomization of high-momentum cough droplets by hydrodynamic focusing and extrusion through the microscale pores in the fibrous network of the single/double-layer mask material needs to be considered in determining mask efficacy. Three-layer masks can effectively block these droplets and thus could be ubiquitously used as a key tool against COVID-19 or similar respiratory diseases.
Collapse
Affiliation(s)
- Shubham Sharma
- Department of Mechanical Engineering, Indian Institute of Science, Bengaluru, KA 560012, India
| | - Roven Pinto
- Department of Mechanical Engineering, Indian Institute of Science, Bengaluru, KA 560012, India
| | - Abhishek Saha
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Swetaprovo Chaudhuri
- Institute for Aerospace Studies, University of Toronto, Toronto, ON M3H 5T6, Canada
| | - Saptarshi Basu
- Department of Mechanical Engineering, Indian Institute of Science, Bengaluru, KA 560012, India.
| |
Collapse
|
90
|
Yuan C, Jin Y, Li Y, Zhang E, Zhang P, Yang Q. PEDV infection in neonatal piglets through the nasal cavity is mediated by subepithelial CD3 + T cells. Vet Res 2021; 52:26. [PMID: 33597007 PMCID: PMC7888150 DOI: 10.1186/s13567-020-00883-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/16/2020] [Indexed: 01/18/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) primarily infects neonatal piglets causing catastrophic effects on the global pig farming industry. PEDV infects piglets through the nasal cavity, a process in which dendritic cells (DCs) play an important role. However, neonatal piglets have fewer nasal DCs. This study found that subepithelial CD3+ T cells mediated PEDV invasion through the nasal cavity in neonatal piglets. PEDV could replicate in the nasal epithelial cells (NECs) isolated from the nasal cavity of neonatal piglets. Infection of NECs with PEDV could induce antiviral and inflammatory cytokines at the late stage. The infected NECs mediated transfer of virus to CD3+ T cells distributed in the subepithelial of the nasal cavity via cell-to-cell contact. The infected CD3+ T cells could migrate to the intestine via blood circulation, causing intestinal infection in neonatal piglets. Thus, the findings of this study indicate the importance of CD3+T cells in the dissemination of PEDV from the nasal cavity to the intestinal mucosa in neonatal piglets.
Collapse
Affiliation(s)
- Chen Yuan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, China
| | - Yuxin Jin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, China
| | - Yuchen Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, China
| | - En Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, China
| | - Penghao Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, China
| | - Qian Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, China.
| |
Collapse
|
91
|
Wu L, Liu X, Yao F, Chen Y. Numerical study of virus transmission through droplets from sneezing in a cafeteria. PHYSICS OF FLUIDS (WOODBURY, N.Y. : 1994) 2021; 33:023311. [PMID: 33746490 PMCID: PMC7976044 DOI: 10.1063/5.0040803] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/20/2021] [Indexed: 05/04/2023]
Abstract
To provide a comprehensive understanding of virus transmission inside small indoor spaces, numerical simulation of sneezing droplets spreading in a cafeteria is conducted through computational fluid dynamics. The numerical results show that dining face to face is extremely vulnerable to direct infection by others' respiratory droplets. Different heights of droplet sources are compared, which indicates that sneezing from a standing person results in a longer survival time of droplets in the air. Scenarios with fewer customers without face to face seating and turning off the horizontal supplying air conditioner are examined as well. Various surfaces are still detected with droplets in 300 s after sneezing. The horizontal supplying air conditioner causes increment in the velocities of the droplets and leads to further spreading of the droplets. It is essential to sanitize all surfaces in a cafeteria including the walls, floor, ceiling, and tables that are not occupied by any customer. Keeping a safe distance in small indoor spaces such as cafeterias does not offer sufficient protection for activities without wearing a face mask. It is recommended that cafeterias and canteens only accept take-away orders.
Collapse
Affiliation(s)
- Liangyu Wu
- College of Electrical, Energy and Power Engineering, Yangzhou University, Yangzhou 225009, People's Republic of China
| | - Xiangdong Liu
- College of Electrical, Energy and Power Engineering, Yangzhou University, Yangzhou 225009, People's Republic of China
| | - Feng Yao
- Jiangsu Key Laboratory of Micro and Nano Heat Fluid Flow Technology and Energy Application, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, People's Republic of China
| | | |
Collapse
|
92
|
Bui CHT, Yeung HW, Ho JCW, Leung CYH, Hui KPY, Perera RAPM, Webby RJ, Schultz-Cherry SL, Nicholls JM, Peiris JSM, Chan MCW. Tropism of SARS-CoV-2, SARS-CoV and influenza virus in canine tissue explants. J Infect Dis 2021; 224:821-830. [PMID: 33395484 PMCID: PMC7799041 DOI: 10.1093/infdis/jiab002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/04/2021] [Indexed: 01/04/2023] Open
Abstract
Background Human spillovers of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to dogs and the emergence of a highly contagious avian-origin H3N2 canine influenza virus have raised concerns on the role of dogs in the spread of SARS-CoV-2 and their susceptibility to existing human and avian influenza viruses, which might result in further reassortment. Methods We systematically studied the replication kinetics of SARS-CoV-2, SARS-CoV, influenza A viruses of H1, H3, H5, H7, and H9 subtypes, and influenza B viruses of Yamagata-like and Victoria-like lineages in ex vivo canine nasal cavity, soft palate, trachea, and lung tissue explant cultures and examined ACE2 and sialic acid (SA) receptor distribution in these tissues. Results There was limited productive replication of SARS-CoV-2 in canine nasal cavity and SARS-CoV in canine nasal cavity, soft palate, and lung, with unexpectedly high ACE2 levels in canine nasal cavity and soft palate. Canine tissues were susceptible to a wide range of human and avian influenza viruses, which matched with the abundance of both human and avian SA receptors. Conclusions Existence of suitable receptors and tropism for the same tissue foster virus adaptation and reassortment. Continuous surveillance in dog populations should be conducted given the many chances for spillover during outbreaks.
Collapse
Affiliation(s)
- Christine H T Bui
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - H W Yeung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - John C W Ho
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Connie Y H Leung
- Centre for Comparative Medicine Research, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kenrie P Y Hui
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ranawaka A P M Perera
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Richard J Webby
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Stacey L Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - John M Nicholls
- Department of Pathology, Queen Mary Hospital, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - J S Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Michael C W Chan
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
93
|
Dbouk T, Drikakis D. On airborne virus transmission in elevators and confined spaces. PHYSICS OF FLUIDS (WOODBURY, N.Y. : 1994) 2021; 33:011905. [PMID: 33790526 PMCID: PMC7984422 DOI: 10.1063/5.0038180] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/14/2020] [Indexed: 05/04/2023]
Abstract
The impact of air ventilation systems on airborne virus transmission (AVT), and aerosols in general, in confined spaces is not yet understood. The recent pandemic has made it crucial to understand the limitations of ventilation systems regarding AVT. We consider an elevator as a prototypical example of a confined space and show how ventilation designs alone, regardless of cooling or heating, contribute to AVT. Air circulation effects are investigated through multiphase computational fluid dynamics, and the performance of an air purifier in an elevator for reducing AVT is assessed. We have investigated three different flow scenarios regarding the position and operation of inlets and outlets in the elevator and a fourth scenario that includes the operation of the air purifier. The position of the inlets and outlets significantly influences the flow circulation and droplet dispersion. An air purifier does not eliminate airborne transmission. The droplet dispersion is reduced when a pair of an inlet and an outlet is implemented. The overall practical conclusion is that the placement and design of the air purifier and ventilation systems significantly affect the droplet dispersion and AVT. Thus, engineering designs of such systems must take into account the flow dynamics in the confined space the systems will be installed.
Collapse
|
94
|
Chowdhury MA, Shuvho MBA, Shahid MA, Haque AKMM, Kashem MA, Lam SS, Ong HC, Uddin MA, Mofijur M. Prospect of biobased antiviral face mask to limit the coronavirus outbreak. ENVIRONMENTAL RESEARCH 2021; 192:110294. [PMID: 33022215 PMCID: PMC7532771 DOI: 10.1016/j.envres.2020.110294] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 05/19/2023]
Abstract
The rapid spread of COVID-19 has led to nationwide lockdowns in many countries. The COVID-19 pandemic has played serious havoc on economic activities throughout the world. Researchers are immensely curious about how to give the best protection to people before a vaccine becomes available. The coronavirus spreads principally through saliva droplets. Thus, it would be a great opportunity if the virus spread could be controlled at an early stage. The face mask can limit virus spread from both inside and outside the mask. This is the first study that has endeavoured to explore the design and fabrication of an antiviral face mask using licorice root extract, which has antimicrobial properties due to glycyrrhetinic acid (GA) and glycyrrhizin (GL). An electrospinning process was utilized to fabricate nanofibrous membrane and virus deactivation mechanisms discussed. The nanofiber mask material was characterized by SEM and airflow rate testing. SEM results indicated that the nanofibers from electrospinning are about 15-30 μm in diameter with random porosity and orientation which have the potential to capture and kill the virus. Theoretical estimation signifies that an 85 L/min rate of airflow through the face mask is possible which ensures good breathability over an extensive range of pressure drops and pore sizes. Finally, it can be concluded that licorice root membrane may be used to produce a biobased face mask to control COVID-19 spread.
Collapse
Affiliation(s)
| | - Md Bengir Ahmed Shuvho
- Department of Industrial and Production Engineering, National Institute of Textile Engineering and Research (NITER), Savar, Dhaka, 1350, Bangladesh
| | - Md Abdus Shahid
- Department of Textile Engineering, Dhaka University of Engineering and Technology (DUET), Gazipur, 1707, Bangladesh
| | - A K M Monjurul Haque
- Department of Industrial and Production Engineering, National Institute of Textile Engineering and Research (NITER), Savar, Dhaka, 1350, Bangladesh
| | - Mohammod Abul Kashem
- Department of Computer Science and Engineering, Dhaka University of Engineering and Technology (DUET), Gazipur, 1707, Bangladesh
| | - Su Shiung Lam
- Pyrolysis Technology Research Group, Institute of Tropical Aquaculture and Fisheries (Akuatrop), Universiti Malaysia Terengganu, 21030, Kuala Nerus, Terengganu, Malaysia
| | - Hwai Chyuan Ong
- School of Information, Systems and Modelling, Faculty of Engineering and IT, University of Technology Sydney, NSW, 2007, Australia
| | - Md Alhaz Uddin
- Department of Civil Engineering, College of Engineering, Jouf University, Sakaka, Saudi Arabia
| | - M Mofijur
- School of Information, Systems and Modelling, Faculty of Engineering and IT, University of Technology Sydney, NSW, 2007, Australia; Mechanical Engineering Department, Prince Mohammad Bin Fahd University, Al Khobar, 31952, Saudi Arabia
| |
Collapse
|
95
|
Proud PC, Tsitoura D, Watson RJ, Chua BY, Aram MJ, Bewley KR, Cavell BE, Cobb R, Dowall S, Fotheringham SA, Ho CMK, Lucas V, Ngabo D, Rayner E, Ryan KA, Slack GS, Thomas S, Wand NI, Yeates P, Demaison C, Zeng W, Holmes I, Jackson DC, Bartlett NW, Mercuri F, Carroll MW. Prophylactic intranasal administration of a TLR2/6 agonist reduces upper respiratory tract viral shedding in a SARS-CoV-2 challenge ferret model. EBioMedicine 2021; 63:103153. [PMID: 33279857 PMCID: PMC7711201 DOI: 10.1016/j.ebiom.2020.103153] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/16/2020] [Accepted: 11/13/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The novel human coronavirus SARS-CoV-2 is a major ongoing global threat with huge economic burden. Like all respiratory viruses, SARS-CoV-2 initiates infection in the upper respiratory tract (URT). Infected individuals are often asymptomatic, yet highly infectious and readily transmit virus. A therapy that restricts initial replication in the URT has the potential to prevent progression of severe lower respiratory tract disease as well as limiting person-to-person transmission. METHODS SARS-CoV-2 Victoria/01/2020 was passaged in Vero/hSLAM cells and virus titre determined by plaque assay. Challenge virus was delivered by intranasal instillation to female ferrets at 5.0 × 106 pfu/ml. Treatment groups received intranasal INNA-051, developed by Ena Respiratory. SARS-CoV-2 RNA was detected using the 2019-nCoV CDC RUO Kit and QuantStudio™ 7 Flex Real-Time PCR System. Histopathological analysis was performed using cut tissues stained with haematoxylin and eosin (H&E). FINDINGS We show that prophylactic intra-nasal administration of the TLR2/6 agonist INNA-051 in a SARS-CoV-2 ferret infection model effectively reduces levels of viral RNA in the nose and throat. After 5 days post-exposure to SARS-CoV-2, INNA-051 significantly reduced virus in throat swabs (p=<0.0001) by up to a 24 fold (96% reduction) and in nasal wash (p=0.0107) up to a 15 fold (93% reduction) in comparison to untreated animals. INTERPRETATION The results of our study support clinical development of a therapy based on prophylactic TLR2/6 innate immune activation in the URT, to reduce SARS-CoV-2 transmission and provide protection against COVID-19. FUNDING This work was funded by Ena Respiratory, Melbourne, Australia.
Collapse
Affiliation(s)
- Pamela C Proud
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Daphne Tsitoura
- Ena Respiratory, Level 9, 31 Queen St, Melbourne, Victoria, 3000, Australia
| | - Robert J Watson
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Brendon Y Chua
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne, Victoria 3000, Australia
| | - Marilyn J Aram
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Kevin R Bewley
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Breeze E Cavell
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Rebecca Cobb
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Stuart Dowall
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Susan A Fotheringham
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Catherine M K Ho
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Vanessa Lucas
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Didier Ngabo
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Emma Rayner
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Kathryn A Ryan
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Gillian S Slack
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Stephen Thomas
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Nadina I Wand
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Paul Yeates
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | | | - Weiguang Zeng
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne, Victoria 3000, Australia
| | - Ian Holmes
- Ena Respiratory, Level 9, 31 Queen St, Melbourne, Victoria, 3000, Australia
| | - David C Jackson
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne, Victoria 3000, Australia
| | - Nathan W Bartlett
- Viral Immunology and Respiratory Disease group and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Francesca Mercuri
- Ena Respiratory, Level 9, 31 Queen St, Melbourne, Victoria, 3000, Australia.
| | - Miles W Carroll
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG; Nuffield Dept of Medicine, Oxford University, Oxford, UK.
| |
Collapse
|
96
|
Overeem NJ, Hamming PHE, Grant OC, Di Iorio D, Tieke M, Bertolino MC, Li Z, Vos G, de Vries RP, Woods RJ, Tito NB, Boons GJPH, van der Vries E, Huskens J. Hierarchical Multivalent Effects Control Influenza Host Specificity. ACS CENTRAL SCIENCE 2020; 6:2311-2318. [PMID: 33376792 PMCID: PMC7760459 DOI: 10.1021/acscentsci.0c01175] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Indexed: 05/15/2023]
Abstract
Understanding how emerging influenza viruses recognize host cells is critical in evaluating their zoonotic potential, pathogenicity, and transmissibility between humans. The surface of the influenza virus is covered with hemagglutinin (HA) proteins that can form multiple interactions with sialic acid-terminated glycans on the host cell surface. This multivalent binding affects the selectivity of the virus in ways that cannot be predicted from the individual receptor-ligand interactions alone. Here, we show that the intrinsic structural and energetic differences between the interactions of avian- or human-type receptors with influenza HA translate from individual site affinity and orientation through receptor length and density on the surface into virus avidity and specificity. We introduce a method to measure virus avidity using receptor density gradients. We found that influenza viruses attached stably to a surface at receptor densities that correspond to a minimum number of approximately 8 HA-glycan interactions, but more interactions were required if the receptors were short and human-type. Thus, the avidity and specificity of influenza viruses for a host cell depend not on the sialic acid linkage alone but on a combination of linkage and the length and density of receptors on the cell surface. Our findings suggest that threshold receptor densities play a key role in virus tropism, which is a predicting factor for both their virulence and zoonotic potential.
Collapse
Affiliation(s)
- Nico J. Overeem
- Molecular
Nanofabrication Group, MESA+ Institute for Nanotechnology, Faculty
of Science and Technology, University of
Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - P. H. Erik Hamming
- Molecular
Nanofabrication Group, MESA+ Institute for Nanotechnology, Faculty
of Science and Technology, University of
Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Oliver C. Grant
- Complex
Carbohydrate Research Center, University
of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United
States
| | - Daniele Di Iorio
- Molecular
Nanofabrication Group, MESA+ Institute for Nanotechnology, Faculty
of Science and Technology, University of
Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Malte Tieke
- Division
of Virology, Department of Infectious Diseases and Immunology, Faculty
of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - M. Candelaria Bertolino
- Molecular
Nanofabrication Group, MESA+ Institute for Nanotechnology, Faculty
of Science and Technology, University of
Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Zeshi Li
- Department
of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Gaël Vos
- Department
of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Robert P. de Vries
- Department
of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Robert J. Woods
- Complex
Carbohydrate Research Center, University
of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United
States
- E-mail:
| | - Nicholas B. Tito
- Electric
Ant Lab, Science Park
106, 1098 XG Amsterdam, The Netherlands
| | - Geert-Jan P. H. Boons
- Complex
Carbohydrate Research Center, University
of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United
States
- Department
of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
- Bijvoet Center
for Biomolecular Research, Utrecht University, 3584 CH Utrecht, The Netherlands
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
- E-mail:
| | - Erhard van der Vries
- Division
of Virology, Department of Infectious Diseases and Immunology, Faculty
of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
- Royal
GD, Arnsbergstraat 7, 7418 EZ Deventer, The Netherlands
- Department
of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
- E-mail:
| | - Jurriaan Huskens
- Molecular
Nanofabrication Group, MESA+ Institute for Nanotechnology, Faculty
of Science and Technology, University of
Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
- E-mail:
| |
Collapse
|
97
|
Temporal and spatial heterogeneity of host response to SARS-CoV-2 pulmonary infection. Nat Commun 2020; 11:6319. [PMID: 33298930 PMCID: PMC7725958 DOI: 10.1038/s41467-020-20139-7] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/13/2020] [Indexed: 12/15/2022] Open
Abstract
The relationship of SARS-CoV-2 pulmonary infection and severity of disease is not fully understood. Here we show analysis of autopsy specimens from 24 patients who succumbed to SARS-CoV-2 infection using a combination of different RNA and protein analytical platforms to characterize inter-patient and intra-patient heterogeneity of pulmonary virus infection. There is a spectrum of high and low virus cases associated with duration of disease. High viral cases have high activation of interferon pathway genes and a predominant M1-like macrophage infiltrate. Low viral cases are more heterogeneous likely reflecting inherent patient differences in the evolution of host response, but there is consistent indication of pulmonary epithelial cell recovery based on napsin A immunohistochemistry and RNA expression of surfactant and mucin genes. Using a digital spatial profiling platform, we find the virus corresponds to distinct spatial expression of interferon response genes demonstrating the intra-pulmonary heterogeneity of SARS-CoV-2 infection.
Collapse
|
98
|
Chatterjee M, van Putten JPM, Strijbis K. Defensive Properties of Mucin Glycoproteins during Respiratory Infections-Relevance for SARS-CoV-2. mBio 2020; 11:e02374-20. [PMID: 33184103 PMCID: PMC7663010 DOI: 10.1128/mbio.02374-20] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mucus plays a pivotal role in protecting the respiratory tract against microbial infections. It acts as a primary contact site to entrap microbes and facilitates their removal from the respiratory tract via the coordinated beating of motile cilia. The major components of airway mucus are heavily O-glycosylated mucin glycoproteins, divided into gel-forming mucins and transmembrane mucins. The gel-forming mucins MUC5AC and MUC5B are the primary structural components of airway mucus, and they enable efficient clearance of pathogens by mucociliary clearance. MUC5B is constitutively expressed in the healthy airway, whereas MUC5AC is upregulated in response to inflammatory challenge. MUC1, MUC4, and MUC16 are the three major transmembrane mucins of the respiratory tracts which prevent microbial invasion, can act as releasable decoy receptors, and activate intracellular signal transduction pathways. Pathogens have evolved virulence factors such as adhesins that facilitate interaction with specific mucins and mucin glycans, for example, terminal sialic acids. Mucin expression and glycosylation are dependent on the inflammatory state of the respiratory tract and are directly regulated by proinflammatory cytokines and microbial ligands. Gender and age also impact mucin glycosylation and expression through the female sex hormone estradiol and age-related downregulation of mucin production. Here, we discuss what is currently known about the role of respiratory mucins and their glycans during bacterial and viral infections of the airways and their relevance for the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Understanding the impact of microbe-mucin interaction in the respiratory tract could inspire the development of novel therapies to boost mucosal defense and combat respiratory infections.
Collapse
Affiliation(s)
- Maitrayee Chatterjee
- Department Biomolecular Health Sciences, Division Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jos P M van Putten
- Department Biomolecular Health Sciences, Division Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Karin Strijbis
- Department Biomolecular Health Sciences, Division Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
99
|
Frank S, Brown SM, Capriotti JA, Westover JB, Pelletier JS, Tessema B. In Vitro Efficacy of a Povidone-Iodine Nasal Antiseptic for Rapid Inactivation of SARS-CoV-2. JAMA Otolaryngol Head Neck Surg 2020; 146:1054-1058. [PMID: 32940656 PMCID: PMC7499242 DOI: 10.1001/jamaoto.2020.3053] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/29/2020] [Indexed: 12/13/2022]
Abstract
Importance Research is needed to demonstrate the efficacy of nasal povidone-iodine (PVP-I) against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Objective To evaluate the in vitro efficacy of PVP-I nasal antiseptic for the inactivation of SARS-CoV-2 at clinically significant contact times of 15 and 30 seconds. Interventions The SARS-CoV-2, USA-WA1/2020 strain, virus stock was tested against nasal antiseptic solutions consisting of aqueous PVP-I as the sole active ingredient. Povidone-iodine was tested at diluted concentrations of 0.5%, 1.25%, and 2.5% and compared with controls. The test solutions and virus were incubated at mean (SD) room temperature of 22 (2) °C for time periods of 15 and 30 seconds. Design and Setting This controlled in vitro laboratory research study used 3 different concentrations of study solution and ethanol, 70%, as a positive control on test media infected with SARS-CoV-2. Test media without virus were added to 2 tubes of the compounds to serve as toxicity and neutralization controls. Ethanol, 70%, was tested in parallel as a positive control and water only as a negative control. Main Outcomes and Measures The primary study outcome measurement was the log reduction value after 15 seconds and 30 seconds of given treatment. Surviving virus from each sample was quantified by standard end point dilution assay, and the log reduction value of each compound was compared with the negative (water) control. Results Povidone-iodine nasal antiseptics at concentrations (0.5%, 1.25%, and 2.5%) completely inactivated SARS-CoV-2 within 15 seconds of contact as measured by log reduction value of greater than 3 log10 of the 50% cell culture infectious dose of the virus. The ethanol, 70%, positive control did not completely inactivate SARS-CoV-2 after 15 seconds of contact. The nasal antiseptics tested performed better than the standard positive control routinely used for in vitro assessment of anti-SARS-CoV-2 agents at a contact time of 15 seconds. No cytotoxic effects on cells were observed after contact with each of the nasal antiseptics tested. Conclusions and Relevance Povidone-iodine nasal antiseptic solutions at concentrations as low as 0.5% rapidly inactivate SARS-CoV-2 at contact times as short as 15 seconds. Intranasal use of PVP-I has demonstrated safety at concentrations of 1.25% and below and may play an adjunctive role in mitigating viral transmission beyond personal protective equipment.
Collapse
Affiliation(s)
- Samantha Frank
- University of Connecticut School of Medicine, Farmington
| | - Seth M. Brown
- University of Connecticut School of Medicine, Farmington
- ProHealth, Ear, Nose and Throat, Farmington, Connecticut
| | | | - Jonna B. Westover
- The Institute for Antiviral Research at Utah State University, Logan
| | | | - Belachew Tessema
- University of Connecticut School of Medicine, Farmington
- ProHealth, Ear, Nose and Throat, Farmington, Connecticut
| |
Collapse
|
100
|
DHANASEKARAN S, PUSHPARAJ SELVADOSS P, SOLOMON SM, CHANDRAMOULISWARAN RB, KAGITHAKARA VAJRAVELU L, VENKATESALU V. Public Health Awareness and Knowledge Dissemination Essentially Becomes a Need of the Hour in Combating COVID-19. IRANIAN JOURNAL OF PUBLIC HEALTH 2020; 49:145-146. [PMID: 34268228 PMCID: PMC8266001 DOI: 10.18502/ijph.v49is1.3692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 03/20/2020] [Indexed: 11/24/2022]
Abstract
This article is a Letter to the Editor and does not include an Abstract.
Collapse
Affiliation(s)
- Sivaraman DHANASEKARAN
- Department of Pharmacology and Toxicology, Centre for Laboratory Animal Technology and Research, Sathyabama Institute of Science and Technology, Jeppiaar Nagar, Chennai, Tamil Nadu 600119, India
| | - Pradeep PUSHPARAJ SELVADOSS
- Department of Pharmacology and Toxicology, Centre for Laboratory Animal Technology and Research, Sathyabama Institute of Science and Technology, Jeppiaar Nagar, Chennai, Tamil Nadu 600119, India
| | - Sundar Manoharan SOLOMON
- School of Technology, Pandit Deendayal Petroleum University, Gandhi Nagar-382 007, Gujarat, India
| | | | | | - V.Venugopal VENKATESALU
- Department of Internal Medicine, Sundaram Health Center, Sholinghur, Tamil Nadu 631102, India
| |
Collapse
|