101
|
Chan Y, Ng SW, Singh SK, Gulati M, Gupta G, Chaudhary SK, Hing GB, Collet T, MacLoughlin R, Löbenberg R, Oliver BG, Chellappan DK, Dua K. Revolutionizing polymer-based nanoparticle-linked vaccines for targeting respiratory viruses: A perspective. Life Sci 2021; 280:119744. [PMID: 34174324 PMCID: PMC8223024 DOI: 10.1016/j.lfs.2021.119744] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022]
Abstract
Viral respiratory tract infections have significantly impacted global health as well as socio-economic growth. Respiratory viruses such as the influenza virus, respiratory syncytial virus (RSV), and the recent SARS-CoV-2 infection (COVID-19) typically infect the upper respiratory tract by entry through the respiratory mucosa before reaching the lower respiratory tract, resulting in respiratory disease. Generally, vaccination is the primary method in preventing virus pathogenicity and it has been shown to remarkably reduce the burden of various infectious diseases. Nevertheless, the efficacy of conventional vaccines may be hindered by certain limitations, prompting the need to develop novel vaccine delivery vehicles to immunize against various strains of respiratory viruses and to mitigate the risk of a pandemic. In this review, we provide an insight into how polymer-based nanoparticles can be integrated with the development of vaccines to effectively enhance immune responses for combating viral respiratory tract infections.
Collapse
Affiliation(s)
- Yinghan Chan
- School of Pharmacy, International Medical University (IMU), Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Sin Wi Ng
- School of Pharmacy, International Medical University (IMU), Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Jaipur, India
| | - Sushil Kumar Chaudhary
- Faculty of Pharmacy, DIT University, Mussoorie-Diversion Road, Makkawala, Dehradun 248 009, Uttarakhand, India
| | - Goh Bey Hing
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Trudi Collet
- Innovative Medicines Group, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Ronan MacLoughlin
- Aerogen, IDA Business Park, Dangan, H91 HE94 Galway, Ireland; School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
| | - Raimar Löbenberg
- University of Alberta, Faculty of Pharmacy and Pharmaceutical Sciences, Edmonton, AB T6G 2N8, Canada
| | - Brian G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Bukit Jalil 57000, Kuala Lumpur, Malaysia.
| | - Kamal Dua
- University of Alberta, Faculty of Pharmacy and Pharmaceutical Sciences, Edmonton, AB T6G 2N8, Canada; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia.
| |
Collapse
|
102
|
Tsai YY, Vazquez CI, Shiu RF, Garcia AK, Le C, Patel P, Sadqi M, Chin WC. Effects of Rock Dust Particles on Airway Mucus Viscosity. BIOTECHNOL BIOPROC E 2021. [DOI: 10.1007/s12257-020-0236-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
103
|
Posch W, Vosper J, Zaderer V, Noureen A, Constant S, Bellmann-Weiler R, Lass-Flörl C, Wilflingseder D. ColdZyme Maintains Integrity in SARS-CoV-2-Infected Airway Epithelia. mBio 2021; 12:e00904-21. [PMID: 33906927 PMCID: PMC8092264 DOI: 10.1128/mbio.00904-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 04/01/2021] [Indexed: 01/04/2023] Open
Abstract
SARS-CoV-2 infection causing the COVID-19 pandemic calls for immediate interventions to avoid viral transmission, disease progression, and subsequent excessive inflammation and tissue destruction. Primary normal human bronchial epithelial cells are among the first targets of SARS-CoV-2 infection. Here, we show that ColdZyme medical device mouth spray efficiently protected against virus entry, excessive inflammation, and tissue damage. Applying ColdZyme to fully differentiated, polarized human epithelium cultured at an air-liquid interphase (ALI) completely blocked binding of SARS-CoV-2 and increased local complement activation mediated by the virus as well as productive infection of the tissue model. While SARS-CoV-2 infection resulted in exaggerated intracellular complement activation immediately following infection and a drop in transepithelial resistance, these parameters were bypassed by single pretreatment of the tissues with ColdZyme mouth spray. Crucially, our study highlights the importance of testing already evaluated and safe drugs such as ColdZyme mouth spray for maintaining epithelial integrity and hindering SARS-CoV-2 entry within standardized three-dimensional (3D) in vitro models mimicking the in vivo human airway epithelium.IMPORTANCE Although our understanding of COVID-19 continuously progresses, essential questions regarding prophylaxis and treatment remain open. A hallmark of severe SARS-CoV-2 infection is a hitherto-undescribed mechanism leading to excessive inflammation and tissue destruction associated with enhanced pathogenicity and mortality. To tackle the problem at the source, transfer of SARS-CoV-2, subsequent binding, infection, and inflammatory responses have to be avoided. In this study, we used fully differentiated, mucus-producing, and ciliated human airway epithelial cultures to test the efficacy of ColdZyme medical device mouth spray in terms of protection from SARS-CoV-2 infection. Importantly, we found that pretreatment of the in vitro airway cultures using ColdZyme mouth spray resulted in significantly shielding the epithelial integrity, hindering virus binding and infection, and blocking excessive intrinsic complement activation within the airway cultures. Our in vitro data suggest that ColdZyme mouth spray may have an impact in prevention of COVID-19.
Collapse
Affiliation(s)
- W Posch
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - J Vosper
- Institute of Medical Biochemistry, Geneva, Switzerland
| | - V Zaderer
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - A Noureen
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - R Bellmann-Weiler
- University Hospital of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - C Lass-Flörl
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - D Wilflingseder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
104
|
Wallace LE, Liu M, van Kuppeveld FJM, de Vries E, de Haan CAM. Respiratory mucus as a virus-host range determinant. Trends Microbiol 2021; 29:983-992. [PMID: 33875348 PMCID: PMC8503944 DOI: 10.1016/j.tim.2021.03.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 11/19/2022]
Abstract
Efficient penetration of the mucus layer is needed for respiratory viruses to avoid mucociliary clearance prior to infection. Many respiratory viruses bind to glycans on the heavily glycosylated mucins that give mucus its gel-like characteristics. Influenza viruses, some paramyxoviruses, and coronaviruses avoid becoming trapped in the mucus by releasing themselves by means of their envelope-embedded enzymes that destroy glycan receptors. For efficient infection, receptor binding and destruction need to be in balance with the host receptor repertoire. Establishment in a novel host species requires resetting of the balance to adapt to the different glycan repertoire encountered. Growing understanding of species-specific mucosal glycosylation patterns and the dynamic interaction with respiratory viruses identifies the mucus layer as a major host-range determinant and barrier for zoonotic transfer.
Collapse
Affiliation(s)
- Louisa E Wallace
- Section Virology, Division Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, The Netherlands
| | - Mengying Liu
- Section Virology, Division Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, The Netherlands
| | - Frank J M van Kuppeveld
- Section Virology, Division Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, The Netherlands
| | - Erik de Vries
- Section Virology, Division Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, The Netherlands.
| | - Cornelis A M de Haan
- Section Virology, Division Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, The Netherlands.
| |
Collapse
|
105
|
Chu AWH, Yip CCY, Chan WM, Ng ACK, Chan DLS, Siu RHP, Chung CYT, Ng JPL, Kittur H, Mosley GL, Poon RWS, Chiu RYT, To KKW. Evaluation of an Automated High-Throughput Liquid-Based RNA Extraction Platform on Pooled Nasopharyngeal or Saliva Specimens for SARS-CoV-2 RT-PCR. Viruses 2021; 13:v13040615. [PMID: 33918447 PMCID: PMC8067048 DOI: 10.3390/v13040615] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/25/2021] [Accepted: 03/31/2021] [Indexed: 11/30/2022] Open
Abstract
SARS-CoV-2 RT-PCR with pooled specimens has been implemented during the COVID-19 pandemic as a cost- and manpower-saving strategy for large-scale testing. However, there is a paucity of data on the efficiency of different nucleic acid extraction platforms on pooled specimens. This study compared a novel automated high-throughput liquid-based RNA extraction (LRE) platform (PHASIFY™) with a widely used magnetic bead-based total nucleic acid extraction (MBTE) platform (NucliSENS® easyMAG®). A total of 60 pools of nasopharyngeal swab and 60 pools of posterior oropharyngeal saliva specimens, each consisting of 1 SARS-CoV-2 positive and 9 SARS-CoV-2 negative specimens, were included for the comparison. Real-time RT-PCR targeting the SARS-CoV-2 RdRp/Hel gene was performed, and GAPDH RT-PCR was used to detect RT-PCR inhibitors. No significant differences were observed in the Ct values and overall RT-PCR positive rates between LRE and MBTE platforms (92.5% (111/120] vs. 90% (108/120]), but there was a slightly higher positive rate for LRE (88.3% (53/60]) than MBTE (81.7% (49/60]) among pooled saliva. The automated LRE method is comparable to a standard MBTE method for the detection of SAR-CoV-2 in pooled specimens, providing a suitable alternative automated extraction platform. Furthermore, LRE may be better suited for pooled saliva specimens due to more efficient removal of RT-PCR inhibitors.
Collapse
Affiliation(s)
- Allen Wing-Ho Chu
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; (A.W.-H.C.); (W.-M.C.); (A.C.-K.N.)
| | - Cyril Chik-Yan Yip
- Department of Microbiology, Queen Mary Hospital, Pok Fu Lam, Hong Kong; (C.C.-Y.Y.); (R.W.-S.P.)
| | - Wan-Mui Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; (A.W.-H.C.); (W.-M.C.); (A.C.-K.N.)
| | - Anthony Chin-Ki Ng
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; (A.W.-H.C.); (W.-M.C.); (A.C.-K.N.)
| | - Dream Lok-Sze Chan
- PHASE Scientific International Limited, Kowloon, Hong Kong; (D.L.-S.C.); (R.H.-P.S.); (C.Y.T.C.); (J.P.-L.N.); (H.K.); (G.L.M.); (R.Y.-T.C.)
| | - Ryan Ho-Ping Siu
- PHASE Scientific International Limited, Kowloon, Hong Kong; (D.L.-S.C.); (R.H.-P.S.); (C.Y.T.C.); (J.P.-L.N.); (H.K.); (G.L.M.); (R.Y.-T.C.)
| | - Cheuk Yiu Tenny Chung
- PHASE Scientific International Limited, Kowloon, Hong Kong; (D.L.-S.C.); (R.H.-P.S.); (C.Y.T.C.); (J.P.-L.N.); (H.K.); (G.L.M.); (R.Y.-T.C.)
| | - Jessica Pui-Ling Ng
- PHASE Scientific International Limited, Kowloon, Hong Kong; (D.L.-S.C.); (R.H.-P.S.); (C.Y.T.C.); (J.P.-L.N.); (H.K.); (G.L.M.); (R.Y.-T.C.)
| | - Harsha Kittur
- PHASE Scientific International Limited, Kowloon, Hong Kong; (D.L.-S.C.); (R.H.-P.S.); (C.Y.T.C.); (J.P.-L.N.); (H.K.); (G.L.M.); (R.Y.-T.C.)
| | - Garrett Lee Mosley
- PHASE Scientific International Limited, Kowloon, Hong Kong; (D.L.-S.C.); (R.H.-P.S.); (C.Y.T.C.); (J.P.-L.N.); (H.K.); (G.L.M.); (R.Y.-T.C.)
| | - Rosana Wing-Shan Poon
- Department of Microbiology, Queen Mary Hospital, Pok Fu Lam, Hong Kong; (C.C.-Y.Y.); (R.W.-S.P.)
| | - Ricky Yin-To Chiu
- PHASE Scientific International Limited, Kowloon, Hong Kong; (D.L.-S.C.); (R.H.-P.S.); (C.Y.T.C.); (J.P.-L.N.); (H.K.); (G.L.M.); (R.Y.-T.C.)
| | - Kelvin Kai-Wang To
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; (A.W.-H.C.); (W.-M.C.); (A.C.-K.N.)
- Department of Microbiology, Queen Mary Hospital, Pok Fu Lam, Hong Kong; (C.C.-Y.Y.); (R.W.-S.P.)
- Correspondence: ; Tel.: +(852)-2255-2413; Fax: +(852)-2855-1241
| |
Collapse
|
106
|
Ge T, Cheng S, Tg. Physicochemical properties of respiratory droplets and their role in COVID-19 pandemics: a critical review. BIOMATERIALS TRANSLATIONAL 2021; 2:10-18. [PMID: 35837254 PMCID: PMC9255823 DOI: 10.3877/cma.j.issn.2096-112x.2021.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/24/2020] [Accepted: 12/29/2020] [Indexed: 01/10/2023]
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic is a serious challenge faced by the global community. Physical scientists can help medical workers and biomedical scientists, engineers, and practitioners, who are working on the front line, to slow down and eventually contain the spread of the COVID-19 virus. This review is focused on the physicochemical characteristics, including composition, aerodynamics, and drying behavior of respiratory droplets as a complex and multicomponent soft matter system, which are the main carrier of the virus for interpersonal transmission. The distribution and dynamics of virus particles within a droplet are also discussed. Understanding the characteristics of virus-laden respiratory droplets can lead to better design of personal protective equipment, frequently touched surfaces such as door knobs and touchscreens, and filtering equipment for indoor air circulation. Such an understanding also provides the scientific basis of public policy, including social distancing rules and public hygiene guidelines, implemented by governments around the world.
Collapse
Affiliation(s)
- Ting Ge
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA,Corresponding authors: Ting Ge, ; Shengfeng Cheng,
| | - Shengfeng Cheng
- Department of Physics, Department of Mechanical Engineering, Center for Soft Matter and Biological Physics, and Macromolecules Innovation Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA,Corresponding authors: Ting Ge, ; Shengfeng Cheng,
| | | |
Collapse
|
107
|
Öner D, Drysdale SB, McPherson C, Lin GL, Janet S, Broad J, Pollard AJ, Aerssens J. Biomarkers for Disease Severity in Children Infected With Respiratory Syncytial Virus: A Systematic Literature Review. J Infect Dis 2021; 222:S648-S657. [PMID: 32794555 DOI: 10.1093/infdis/jiaa208] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Clinical manifestations of respiratory syncytial virus (RSV) infection vary widely from mild, self-limiting illness to severe life-threatening disease. There are gaps in knowledge of biomarkers to objectively define severe disease and predict clinical outcomes. METHODS A systematic search was performed, 1945-March 2019 in databases Ovid Medline, Embase, Global health, Scopus, and Web of Science. Risk of bias was assessed using the Cochrane tool. RESULTS A total of 25 132 abstracts were screened and studies were assessed for quality, risk of bias, and extracted data; 111 studies met the inclusion criteria. RSV severity was correlated with antibody titers, reduced T and B cells, dysregulated innate immunity, neutrophil mobilization to the lungs and blood, decreased Th1 response, and Th2 weighted shift. Microbial exposures in respiratory tract may contribute to neutrophil mobilization to the lungs of the infants with severe RSV compared with mild RSV disease. CONCLUSIONS Although a wide range of biomarkers have been associated with RSV disease severity, robust validated biomarkers are lacking. This review illustrates the broad heterogeneity of study designs and high variability in the definition of severe RSV disease. Prospective studies are required to validate biomarkers. Additional research investigating epigenetics, metabolomics, and microbiome holds promise for novel biomarkers.
Collapse
Affiliation(s)
- Deniz Öner
- Infectious Diseases Translational Biomarkers, Janssen Pharmaceutica, Beerse, Belgium
| | - Simon B Drysdale
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford, United Kingdom.,Paediatric Infectious Diseases Research Group, Institute of Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Calum McPherson
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Gu-Lung Lin
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Sophie Janet
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Jonathan Broad
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Jeroen Aerssens
- Infectious Diseases Translational Biomarkers, Janssen Pharmaceutica, Beerse, Belgium
| | | |
Collapse
|
108
|
Thorn CR, Thomas N, Boyd BJ, Prestidge CA. Nano-fats for bugs: the benefits of lipid nanoparticles for antimicrobial therapy. Drug Deliv Transl Res 2021; 11:1598-1624. [PMID: 33675007 DOI: 10.1007/s13346-021-00921-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2021] [Indexed: 12/24/2022]
Abstract
Bacterial infections are an imminent global healthcare threat evolving from rapidly advancing bacterial defence mechanisms that antibiotics fail to overcome. Antibiotics have been designed for systemic administration to target planktonic bacteria, leading to difficulties in reaching the site of localized bacterial infection and an inability to overcome the biological, chemical and physical barriers of bacteria, including biofilms, intracellular infections and antimicrobial resistance. The amphiphilic, biomimetic and antimicrobial properties of lipids provide a promising toolbox to innovate and advance antimicrobial therapies, overcoming the barriers presented by bacteria in order to directly and effectively treat recalcitrant infections. Nanoparticulate lipid-based drug delivery systems can enhance antibiotic permeation through the chemical and physical barriers of bacterial infections, as well as fuse with bacterial cell membranes, release antibiotics in response to bacteria and act synergistically with loaded antibiotics to enhance the total antimicrobial efficacy. This review explores the barriers presented by bacterial infections that pose bio-pharmaceutical challenges to antibiotics and how different structural and functional mechanisms of lipids can enhance antimicrobial therapies. Different nanoparticulate lipid-based systems are presented as valuable drug delivery systems to advance the efficacy of antibiotics, including liposomes, liquid crystalline nanoparticles, solid lipid nanoparticles, nanostructured lipid carriers and lipid nanocarriers. In summary, liquid crystalline nanoparticles are emerging with the greatest potential for clinical applications and commercial success as an "all-rounder" advanced lipid-based antimicrobial therapy that overcomes the multiple biological, chemical and physical barriers of bacteria.
Collapse
Affiliation(s)
- Chelsea R Thorn
- Clinical and Health Science, University of South Australia, City East Campus, Adelaide, SA, 5000, Australia.,The Basil Hetzel Institute for Translational Health Research, Woodville, SA, 5011, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, University of South Australia, SA, 5000, Adelaide, Australia
| | - Nicky Thomas
- Clinical and Health Science, University of South Australia, City East Campus, Adelaide, SA, 5000, Australia.,The Basil Hetzel Institute for Translational Health Research, Woodville, SA, 5011, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, University of South Australia, SA, 5000, Adelaide, Australia
| | - Ben J Boyd
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, University of South Australia, SA, 5000, Adelaide, Australia.,Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Pde, Parkville, VIC, 3052, Australia
| | - Clive A Prestidge
- Clinical and Health Science, University of South Australia, City East Campus, Adelaide, SA, 5000, Australia. .,ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, University of South Australia, SA, 5000, Adelaide, Australia.
| |
Collapse
|
109
|
Božič A, Kanduč M. Relative humidity in droplet and airborne transmission of disease. J Biol Phys 2021; 47:1-29. [PMID: 33564965 PMCID: PMC7872882 DOI: 10.1007/s10867-020-09562-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
A large number of infectious diseases are transmitted by respiratory droplets. How long these droplets persist in the air, how far they can travel, and how long the pathogens they might carry survive are all decisive factors for the spread of droplet-borne diseases. The subject is extremely multifaceted and its aspects range across different disciplines, yet most of them have only seldom been considered in the physics community. In this review, we discuss the physical principles that govern the fate of respiratory droplets and any viruses trapped inside them, with a focus on the role of relative humidity. Importantly, low relative humidity-as encountered, for instance, indoors during winter and inside aircraft-facilitates evaporation and keeps even initially large droplets suspended in air as aerosol for extended periods of time. What is more, relative humidity affects the stability of viruses in aerosol through several physical mechanisms such as efflorescence and inactivation at the air-water interface, whose role in virus inactivation nonetheless remains poorly understood. Elucidating the role of relative humidity in the droplet spread of disease would permit us to design preventive measures that could aid in reducing the chance of transmission, particularly in indoor environment.
Collapse
Affiliation(s)
- Anže Božič
- Department of Theoretical Physics, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Matej Kanduč
- Department of Theoretical Physics, Jožef Stefan Institute, Ljubljana, Slovenia
| |
Collapse
|
110
|
Eosinophil Responses at the Airway Epithelial Barrier during the Early Phase of Influenza A Virus Infection in C57BL/6 Mice. Cells 2021; 10:cells10030509. [PMID: 33673645 PMCID: PMC7997358 DOI: 10.3390/cells10030509] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/09/2021] [Accepted: 02/12/2021] [Indexed: 02/07/2023] Open
Abstract
Eosinophils, previously considered terminally differentiated effector cells, have multifaceted functions in tissues. We previously found that allergic mice with eosinophil-rich inflammation were protected from severe influenza and discovered specialized antiviral effector functions for eosinophils including promoting cellular immunity during influenza. In this study, we hypothesized that eosinophil responses during the early phase of influenza contribute to host protection. Using in vitro and in vivo models, we found that eosinophils were rapidly and dynamically regulated upon influenza A virus (IAV) exposure to gain migratory capabilities to traffic to lymphoid organs after pulmonary infection. Eosinophils were capable of neutralizing virus upon contact and combinations of eosinophil granule proteins reduced virus infectivity through hemagglutinin inactivation. Bi-directional crosstalk between IAV-exposed epithelial cells and eosinophils occurred after IAV infection and cross-regulation promoted barrier responses to improve antiviral defenses in airway epithelial cells. Direct interactions between eosinophils and airway epithelial cells after IAV infection prevented virus-induced cytopathology in airway epithelial cells in vitro, and eosinophil recipient IAV-infected mice also maintained normal airway epithelial cell morphology. Our data suggest that eosinophils are important in the early phase of IAV infection providing immediate protection to the epithelial barrier until adaptive immune responses are deployed during influenza.
Collapse
|
111
|
Bonser LR, Eckalbar WL, Rodriguez L, Shen J, Koh KD, Zlock LT, Christenson S, Woodruff PG, Finkbeiner WE, Erle DJ. The type 2 asthma mediator IL-13 inhibits SARS-CoV-2 infection of bronchial epithelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.02.25.432762. [PMID: 33655249 PMCID: PMC7924269 DOI: 10.1101/2021.02.25.432762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
RATIONALE Asthma is associated with chronic changes in the airway epithelium, a key target of SARS-CoV-2. Many epithelial changes are driven by the type 2 cytokine IL-13, but the effects of IL-13 on SARS-CoV-2 infection are unknown. OBJECTIVES We sought to discover how IL-13 and other cytokines affect expression of genes encoding SARS-CoV-2-associated host proteins in human bronchial epithelial cells (HBECs) and determine whether IL-13 stimulation alters susceptibility to SARS-CoV-2 infection. METHODS We used bulk and single cell RNA-seq to identify cytokine-induced changes in SARS-CoV-2-associated gene expression in HBECs. We related these to gene expression changes in airway epithelium from individuals with mild-moderate asthma and chronic obstructive pulmonary disease (COPD). We analyzed effects of IL-13 on SARS-CoV-2 infection of HBECs. MEASUREMENTS AND MAIN RESULTS Transcripts encoding 332 of 342 (97%) SARS-CoV-2-associated proteins were detected in HBECs (≥1 RPM in 50% samples). 41 (12%) of these mRNAs were regulated by IL-13 (>1.5-fold change, FDR < 0.05). Many IL-13-regulated SARS-CoV-2-associated genes were also altered in type 2 high asthma and COPD. IL-13 pretreatment reduced viral RNA recovered from SARS-CoV-2 infected cells and decreased dsRNA, a marker of viral replication, to below the limit of detection in our assay. Mucus also inhibited viral infection. CONCLUSIONS IL-13 markedly reduces susceptibility of HBECs to SARS-CoV-2 infection through mechanisms that likely differ from those activated by type I interferons. Our findings may help explain reports of relatively low prevalence of asthma in patients diagnosed with COVID-19 and could lead to new strategies for reducing SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Luke R. Bonser
- Lung Biology Center, Critical Care, Allergy and Sleep Medicine, Department of Medicine; University of California, San Francisco
| | - Walter L. Eckalbar
- Lung Biology Center, Critical Care, Allergy and Sleep Medicine, Department of Medicine; University of California, San Francisco
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine; University of California, San Francisco
- UCSF CoLabs; University of California, San Francisco
| | | | - Jiangshan Shen
- Lung Biology Center, Critical Care, Allergy and Sleep Medicine, Department of Medicine; University of California, San Francisco
| | - Kyung Duk Koh
- Lung Biology Center, Critical Care, Allergy and Sleep Medicine, Department of Medicine; University of California, San Francisco
| | - Lorna T. Zlock
- Department of Pathology; University of California, San Francisco
| | - Stephanie Christenson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine; University of California, San Francisco
| | - Prescott G. Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine; University of California, San Francisco
- Cardiovascular Research Institute; University of California, San Francisco
- ImmunoX Initiative; University of California, San Francisco
| | | | - David J. Erle
- Lung Biology Center, Critical Care, Allergy and Sleep Medicine, Department of Medicine; University of California, San Francisco
- UCSF CoLabs; University of California, San Francisco
- Cardiovascular Research Institute; University of California, San Francisco
- ImmunoX Initiative; University of California, San Francisco
| |
Collapse
|
112
|
Yang B, Liu C, Pan X, Fu W, Fan Z, Jin Y, Bai F, Cheng Z, Wu W. Identification of Novel PhoP-PhoQ Regulated Genes That Contribute to Polymyxin B Tolerance in Pseudomonas aeruginosa. Microorganisms 2021; 9:microorganisms9020344. [PMID: 33572426 PMCID: PMC7916210 DOI: 10.3390/microorganisms9020344] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
Polymyxin B and E (colistin) are the last resorts to treat multidrug-resistant Gram-negative pathogens. Pseudomonas aeruginosa is intrinsically resistant to a variety of antibiotics. The PhoP-PhoQ two-component regulatory system contributes to the resistance to polymyxins by regulating an arnBCADTEF-pmrE operon that encodes lipopolysaccharide modification enzymes. To identify additional PhoP-regulated genes that contribute to the tolerance to polymyxin B, we performed a chromatin immunoprecipitation sequencing (ChIP-Seq) assay and found novel PhoP binding sites on the chromosome. We further verified that PhoP directly controls the expression of PA14_46900, PA14_50740 and PA14_52340, and the operons of PA14_11970-PA14_11960 and PA14_52350-PA14_52370. Our results demonstrated that mutation of PA14_46900 increased the bacterial binding and susceptibility to polymyxin B. Meanwhile, mutation of PA14_11960 (papP), PA14_11970 (mpl), PA14_50740 (slyB), PA14_52350 (ppgS), and PA14_52370 (ppgH) reduced the bacterial survival rates and increased ethidium bromide influx under polymyxin B or Sodium dodecyl sulfate (SDS) treatment, indicating roles of these genes in maintaining membrane integrity in response to the stresses. By 1-N-phenylnaphthylamine (NPN) and propidium iodide (PI) staining assay, we found that papP and slyB are involved in maintaining outer membrane integrity, and mpl and ppgS-ppgH are involved in maintaining inner membrane integrity. Overall, our results reveal novel PhoP-PhoQ regulated genes that contribute to polymyxin B tolerance.
Collapse
|
113
|
Liu S, Cao Y, Du T, Zhi Y. Prevalence of Comorbid Asthma and Related Outcomes in COVID-19: A Systematic Review and Meta-Analysis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2021; 9:693-701. [PMID: 33309934 PMCID: PMC7725230 DOI: 10.1016/j.jaip.2020.11.054] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/07/2020] [Accepted: 11/25/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND The impact of asthma on coronavirus disease 2019 (COVID-19) remains largely unknown. OBJECTIVE To investigate the asthma prevalence among patients with COVID-19 and compare outcomes between patients with and without asthma. METHODS In this systematic review and meta-analysis, we searched PubMed, Embase, Web of Science, bioRxiv, and medRxiv for studies reporting asthma prevalence in general patients with COVID-19 or comparing outcomes between patients with and without asthma, and excluded duplicate publications, reviews, editorials, comments, single case reports, or small case series (<10 cases). We determined the pooled estimates of effect using random-effect model. RESULTS On the basis of 131 studies (410,382 patients), we found great variability in the prevalence of comorbid asthma among patients with COVID-19 in different countries or regions ranging from 1.1% to 16.9%. No significant difference in asthma prevalence was found between hospitalized and nonhospitalized (risk ratio [RR], 1.15; 95% CI, 0.92-1.43), severe and nonsevere (RR, 1.21; 95% CI, 0.92-1.57), intensive care unit and non-intensive care unit (RR, 1.19; 95% CI, 0.92-1.54), dead and survived (RR, 0.90; 95% CI, 0.73-1.11), intubated/mechanically ventilated and nonintubated/mechanically ventilated (RR, 0.91; 95% CI, 0.71-1.17) patients with COVID-19. Patients with asthma have a lower risk of death compared with patients without asthma (RR, 0.65; 95% CI, 0.43-0.98). Asthma is not associated with a higher risk of intubation or mechanical ventilation (RR, 1.03; 95% CI, 0.72-1.46). CONCLUSIONS There is great variability in asthma prevalence among patients with COVID-19 in different countries or regions. Asthma is not associated with higher COVID-19 severity or worse prognosis, and patients with asthma are found to have a lower risk of death compared with patients without asthma.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Allergy & Clinical Immunology, National Clinical Research Center for Immunologic Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China; School of Clinical Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yang Cao
- Department of Allergy & Clinical Immunology, National Clinical Research Center for Immunologic Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China; School of Clinical Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tian Du
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuxiang Zhi
- Department of Allergy & Clinical Immunology, National Clinical Research Center for Immunologic Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
114
|
Woodby B, Arnold MM, Valacchi G. SARS-CoV-2 infection, COVID-19 pathogenesis, and exposure to air pollution: What is the connection? Ann N Y Acad Sci 2021; 1486:15-38. [PMID: 33022781 PMCID: PMC7675684 DOI: 10.1111/nyas.14512] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 12/20/2022]
Abstract
Exposure to air pollutants has been previously associated with respiratory viral infections, including influenza, measles, mumps, rhinovirus, and respiratory syncytial virus. Epidemiological studies have also suggested that air pollution exposure is associated with increased cases of SARS-CoV-2 infection and COVID-19-associated mortality, although the molecular mechanisms by which pollutant exposure affects viral infection and pathogenesis of COVID-19 remain unknown. In this review, we suggest potential molecular mechanisms that could account for this association. We have focused on the potential effect of exposure to nitrogen dioxide (NO2 ), ozone (O3 ), and particulate matter (PM) since there are studies investigating how exposure to these pollutants affects the life cycle of other viruses. We have concluded that pollutant exposure may affect different stages of the viral life cycle, including inhibition of mucociliary clearance, alteration of viral receptors and proteases required for entry, changes to antiviral interferon production and viral replication, changes in viral assembly mediated by autophagy, prevention of uptake by macrophages, and promotion of viral spread by increasing epithelial permeability. We believe that exposure to pollutants skews adaptive immune responses toward bacterial/allergic immune responses, as opposed to antiviral responses. Exposure to air pollutants could also predispose exposed populations toward developing COIVD-19-associated immunopathology, enhancing virus-induced tissue inflammation and damage.
Collapse
Affiliation(s)
- Brittany Woodby
- Animal Science DepartmentPlants for Human Health Institute, N.C. Research Campus, North Carolina State UniversityKannapolisNorth Carolina
| | - Michelle M. Arnold
- Department of Microbiology and ImmunologyCenter for Molecular and Tumor VirologyLouisiana State University Health Sciences CenterShreveportLouisiana
| | - Giuseppe Valacchi
- Animal Science DepartmentPlants for Human Health Institute, N.C. Research Campus, North Carolina State UniversityKannapolisNorth Carolina
- Department of Life Sciences and BiotechnologyUniversity of FerraraFerraraItaly
- Department of Food and NutritionKyung Hee UniversitySeoulSouth Korea
| |
Collapse
|
115
|
Khan MA, Khan ZA, Charles M, Pratap P, Naeem A, Siddiqui Z, Naqvi N, Srivastava S. Cytokine Storm and Mucus Hypersecretion in COVID-19: Review of Mechanisms. J Inflamm Res 2021; 14:175-189. [PMID: 33519225 PMCID: PMC7838037 DOI: 10.2147/jir.s271292] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/08/2020] [Indexed: 12/18/2022] Open
Abstract
Mucus is an integral part of the respiratory physiology. It protects the respiratory tract by acting as a physical barrier against inhaled particles and microbes. Excessive inflammation in conditions such as COVID-19 can result in over-production of mucus which obstructs the airway. Build-up of mucus can also contribute to recurrent airway infection, causing further obstruction. This article summarizes the current understanding and knowledge of respiratory mucus production and proposes the role of cytokine storm in inducing sudden mucus hypersecretion in COVID-19. Based on these cascades, the active constituents that inhibit or activate several potential targets are outlined for further research. These may be explored for the discovery and design of drugs to combat cytokine storm and its ensuing complications.
Collapse
Affiliation(s)
- Mohsin Ali Khan
- Reseach & Development Department, Era's Lucknow Medical College & Hospital, Lucknow, Uttar Pradesh, India
| | - Zaw Ali Khan
- Reseach & Development Department, Era's Lucknow Medical College & Hospital, Lucknow, Uttar Pradesh, India
| | - Mark Charles
- Metabolic Research Unit, Era's Lucknow Medical College & Hospital, Lucknow, Uttar Pradesh, India
| | - Pushpendra Pratap
- Metabolic Research Unit, Era's Lucknow Medical College & Hospital, Lucknow, Uttar Pradesh, India
| | - Abdul Naeem
- Metabolic Research Unit, Era's Lucknow Medical College & Hospital, Lucknow, Uttar Pradesh, India
| | - Zainab Siddiqui
- Department of Pathology, Era's Lucknow Medical College & Hospital, Lucknow, Uttar Pradesh, India
| | - Nigar Naqvi
- Department of Nutrition, Era's Lucknow Medical College & Hospital, Lucknow, Uttar Pradesh, India
| | - Shikha Srivastava
- Department of Nutrition, Era's Lucknow Medical College & Hospital, Lucknow, Uttar Pradesh, India
| |
Collapse
|
116
|
Wang BX, Wheeler KM, Cady KC, Lehoux S, Cummings RD, Laub MT, Ribbeck K. Mucin Glycans Signal through the Sensor Kinase RetS to Inhibit Virulence-Associated Traits in Pseudomonas aeruginosa. Curr Biol 2021; 31:90-102.e7. [DOI: 10.1016/j.cub.2020.09.088] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/29/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022]
|
117
|
Pleass RJ. The therapeutic potential of sialylated Fc domains of human IgG. MAbs 2021; 13:1953220. [PMID: 34288809 PMCID: PMC8296966 DOI: 10.1080/19420862.2021.1953220] [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] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/08/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Pathogens frequently use multivalent binding to sialic acid to infect cells or to modulate immunity through interactions with human sialic acid-binding immunoglobulin-type lectins (Siglecs). Molecules that interfere with these interactions could be of interest as diagnostics, anti-infectives or as immune modulators. This review describes the development of molecular scaffolds based on the crystallizable fragment (Fc) region of immunoglobulin (Ig) G that deliver high-avidity binding to innate immune receptors, including sialic acid-dependent receptors. The ways in which the sialylated Fc may be engineered as immune modulators that mimic the anti-inflammatory properties of intravenous polyclonal Ig or as blockers of sialic-acid-dependent infectivity by viruses are also discussed.
Collapse
Affiliation(s)
- Richard J. Pleass
- Department of Tropical Disease Biology, Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
118
|
Morphogenetic (Mucin Expression) as Well as Potential Anti-Corona Viral Activity of the Marine Secondary Metabolite Polyphosphate on A549 Cells. Mar Drugs 2020; 18:md18120639. [PMID: 33327522 PMCID: PMC7764923 DOI: 10.3390/md18120639] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/04/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022] Open
Abstract
The mucus layer of the nasopharynx and bronchial epithelium has a barrier function against inhaled pathogens such as the coronavirus SARS-CoV-2. We recently found that inorganic polyphosphate (polyP), a physiological, metabolic energy (ATP)-providing polymer released from blood platelets, blocks the binding of the receptor binding domain (RBD) to the cellular ACE2 receptor in vitro. PolyP is a marine natural product and is abundantly present in marine bacteria. Now, we have approached the in vivo situation by studying the effect of polyP on the human alveolar basal epithelial A549 cells in a mucus-like mucin environment. These cells express mucins as well as the ectoenzymes alkaline phosphatase (ALP) and adenylate kinase (ADK), which are involved in the extracellular production of ATP from polyP. Mucin, integrated into a collagen-based hydrogel, stimulated cell growth and attachment. The addition of polyP to the hydrogel significantly increased cell attachment and also the expression of the membrane-tethered mucin MUC1 and the secreted mucin MUC5AC. The increased synthesis of MUC1 was also confirmed by immunostaining. This morphogenetic effect of polyP was associated with a rise in extracellular ATP level. We conclude that the nontoxic and non-immunogenic polymer polyP could possibly also exert a protective effect against SARS-CoV-2-cell attachment; first, by stimulating the innate antiviral response by strengthening the mucin barrier with its antimicrobial proteins, and second, by inhibiting virus attachment to the cells, as deduced from the reduction in the strength of binding between the viral RBD and the cellular ACE2 receptor.
Collapse
|
119
|
Iverson E, Kaler L, Agostino EL, Song D, Duncan GA, Scull MA. Leveraging 3D Model Systems to Understand Viral Interactions with the Respiratory Mucosa. Viruses 2020; 12:E1425. [PMID: 33322395 PMCID: PMC7763686 DOI: 10.3390/v12121425] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Respiratory viruses remain a significant cause of morbidity and mortality in the human population, underscoring the importance of ongoing basic research into virus-host interactions. However, many critical aspects of infection are difficult, if not impossible, to probe using standard cell lines, 2D culture formats, or even animal models. In vitro systems such as airway epithelial cultures at air-liquid interface, organoids, or 'on-chip' technologies allow interrogation in human cells and recapitulate emergent properties of the airway epithelium-the primary target for respiratory virus infection. While some of these models have been used for over thirty years, ongoing advancements in both culture techniques and analytical tools continue to provide new opportunities to investigate airway epithelial biology and viral infection phenotypes in both normal and diseased host backgrounds. Here we review these models and their application to studying respiratory viruses. Furthermore, given the ability of these systems to recapitulate the extracellular microenvironment, we evaluate their potential to serve as a platform for studies specifically addressing viral interactions at the mucosal surface and detail techniques that can be employed to expand our understanding.
Collapse
Affiliation(s)
- Ethan Iverson
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA; (E.I.); (E.L.A.)
| | - Logan Kaler
- Biophysics Program, University of Maryland, College Park, MD 20742, USA; (L.K.); (G.A.D.)
| | - Eva L. Agostino
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA; (E.I.); (E.L.A.)
| | - Daniel Song
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA;
| | - Gregg A. Duncan
- Biophysics Program, University of Maryland, College Park, MD 20742, USA; (L.K.); (G.A.D.)
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA;
| | - Margaret A. Scull
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA; (E.I.); (E.L.A.)
| |
Collapse
|
120
|
Gale P. Thermodynamic equilibrium dose-response models for MERS-CoV infection reveal a potential protective role of human lung mucus but not for SARS-CoV-2. MICROBIAL RISK ANALYSIS 2020; 16:100140. [PMID: 32984489 PMCID: PMC7501778 DOI: 10.1016/j.mran.2020.100140] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/09/2020] [Accepted: 09/11/2020] [Indexed: 05/12/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Middle East respiratory syndrome coronavirus (MERS-CoV) infect the human respiratory tract. A prototype thermodynamic equilibrium model is presented here for the probability of the virions getting through the mucus barrier and infecting epithelial cells based on the binding affinity (Kmucin) of the virions to mucin molecules in the mucus and parameters for binding and infection of the epithelial cell. Both MERS-CoV and SARS-CoV-2 bind strongly to their cellular receptors, DDP4 and ACE2, respectively, and infect very efficiently both bronchus and lung ex vivo cell cultures which are not protected by a mucus barrier. According to the model, mucin binding could reduce the infectivity for MERS-CoV compared to SARS-CoV-2 by at least 100-fold depending on the magnitude of Kmucin. Specifically Kmucin values up to 106 M-1 have little protective effect and thus the mucus barrier would not remove SARS-CoV-2 which does not bind to sialic acids (SA) and hence would have a very low Kmucin. Depending on the viability of individual virions, the ID50 for SARS-CoV-2 is estimated to be ~500 virions (viral RNA genomic copies) representing 1 to 2 pfu. In contrast MERS-CoV binds both SA and human mucin and a Kmucin of 5 × 109 M-1 as reported for lectins would mop up 99.83% of the virus according to the model with the ID50 for MERS-CoV estimated to be ~295,000 virions (viral RNA genomic copies) representing 819 pfu. This could in part explain why MERS-CoV is poorly transmitted from human to human compared to SARS-CoV-2. Some coronaviruses use an esterase to escape the mucin, although MERS-CoV does not. Instead, it is shown here that "clustering" of virions into single aerosol particles as recently reported for rotavirus in extracellular vesicles could provide a co-operative mechanism whereby MERS-CoV could theoretically overcome the mucin barrier locally and a small proportion of 10 μm diameter aerosol particles could contain ~70 virions based on reported maximum levels in saliva. Although recent evidence suggests SARS-CoV-2 initiates infection in the nasal epithelium, the thermodynamic equilibrium models presented here could complement published approaches for modelling the physical entry of pathogens to the lung based on the fate and transport of the pathogen particles (as for anthrax spores) to develop a dose-response model for aerosol exposure to respiratory viruses. This would enable the infectivity through aerosols to be defined based on molecular parameters as well as physical parameters. The role of the spike proteins of MERS-CoV and SARS-CoV-2 binding to SA and heparan sulphate, respectively, may be to aid non-specific attachment to the host cell. It is proposed that a high Kmucin is the cost for subsequent binding of MERS-CoV to SAs on the cell surface to partially overcome the unfavourable entropy of immobilisation as the virus adopts the correct orientation for spike protein interactions with its protein cellular receptor DPP4.
Collapse
Affiliation(s)
- Paul Gale
- Independent Scientist, 15 Weare Close, Portland, Dorset, DT5 1JP, UK
| |
Collapse
|
121
|
Observations of, and Insights into, Cystic Fibrosis Mucus Heterogeneity in the Pre-Modulator Era: Sputum Characteristics, DNA and Glycoprotein Content, and Solubilization Time. JOURNAL OF RESPIRATION 2020. [DOI: 10.3390/jor1010002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] Open
Abstract
Airway obstruction with chronic inflammation and infection are major contributors to the lung damage and mortality of cystic fibrosis (CF). A better understanding of the congested milieu of CF airways will aid in improving therapeutic strategies. This article retrospectively reports our observations, and discusses insights gained in the handling and analysis of CF sputa. CF and non-CF mucus samples were surveyed for morphological features by electron microscopy and analyzed for the macromolecular dry weight (MDW), total protein, lipid, carbohydrate, and DNA. Mucus character was investigated with chemical solubilization time as a comparative tool. CF mucus appeared distinctly thick, viscous, and heterogeneous, with neutrophils as the dominant immune cell. CF sputum DNA content varied markedly for and between individuals (~1–10% MDW), as did solubilization times (~1–20 h). CF Sputum DNA up to 7.1% MDW correlated positively with solubilization time, whereas DNA >7.1% MDW correlated negatively. 3D analysis of CF sputa DNA, GP, and solubilization times revealed a dynamic and predictive relationship. Reflecting on the heterogeneous content and character of CF mucus, and the possible interplay in space and time in the respiratory tract of polymeric DNA and mucous glycoproteins, we highlight it’s potential to affect infection-related airway pathologies and the success of therapeutic interventions.
Collapse
|
122
|
Bueckert M, Gupta R, Gupta A, Garg M, Mazumder A. Infectivity of SARS-CoV-2 and Other Coronaviruses on Dry Surfaces: Potential for Indirect Transmission. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E5211. [PMID: 33218120 PMCID: PMC7698891 DOI: 10.3390/ma13225211] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/09/2020] [Accepted: 11/16/2020] [Indexed: 01/03/2023]
Abstract
The unwavering spread of COVID-19 has taken the world by storm. Preventive measures like social distancing and mask usage have been taken all around the globe but still, as of September 2020, the number of cases continues to rise in many countries. Evidently, these measures are insufficient. Although decreases in population density and surges in the public's usage of personal protective equipment can mitigate direct transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), indirect transmission of the virus is still probable. By summarizing the current state of knowledge on the stability of coronaviruses on dry materials, this review uncovers the high potential for SARS-CoV-2 transmission through contaminated surfaces (i.e., fomites) and prompts future research. Fully contextualized data on coronavirus persistence are presented. The methods and limitations to testing the stability of coronaviruses are explored, and the SARS-CoV-2 representativeness of different coronaviruses is analyzed. The factors which dictate the persistence of coronaviruses on surfaces (media, environmental conditions, and material-type) are investigated, and the review is concluded by encouraging material innovation to combat the current pandemic. To summarize, SARS-CoV-2 remains viable on the timescale of days on hard surfaces under ambient indoor conditions. Similarly, the virus is stable on human skin, signifying the necessity of hand hygiene amidst the current pandemic. There is an inverse relationship between SARS-CoV-2 surface persistence and temperature/humidity, and the virus is well suited to air-conditioned environments (room temperature, ~ 40% relative humidity). Sunlight may rapidly inactivate the virus, suggesting that indirect transmission predominantly occurs indoors. The development of antiviral materials and surface coatings would be an extremely effective method to mitigate the spread of COVID-19. To obtain applicable data on the persistence of coronaviruses and the efficiency of virucidal materials, future researchers should understand the common experimental limitations outlined in this review and plan their studies accordingly.
Collapse
Affiliation(s)
- Max Bueckert
- Department of Biochemistry & Microbiology, University of Victoria, 3800 Finnerty Road, Victoria, BC V8P 5C2, Canada
| | - Rishi Gupta
- Department of Civil Engineering, University of Victoria, 3800 Finnerty Road, Victoria, BC V8P 5C2, Canada;
| | - Aditi Gupta
- Mearns Centre for Learning—McPherson Library, University of Victoria, 3800 Finnerty Road, Victoria, BC V8P 5C2, Canada;
| | - Mohit Garg
- Department of Civil Engineering, University of Victoria, 3800 Finnerty Road, Victoria, BC V8P 5C2, Canada;
| | - Asit Mazumder
- Department of Biology, University of Victoria, 3800 Finnerty Road, Victoria, BC V8P 5C2, Canada;
| |
Collapse
|
123
|
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: 65] [Impact Index Per Article: 13.0] [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
|
124
|
Chen S, Kasper B, Zhang B, Lashua LP, Ross TM, Ghedin E, Mahal LK. Age-Dependent Glycomic Response to the 2009 Pandemic H1N1 Influenza Virus and Its Association with Disease Severity. J Proteome Res 2020; 19:4486-4495. [PMID: 32981324 PMCID: PMC7640967 DOI: 10.1021/acs.jproteome.0c00455] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Indexed: 01/05/2023]
Abstract
Influenza A viruses cause a spectrum of responses, from mild coldlike symptoms to severe respiratory illness and death. Intrinsic host factors, such as age, can influence disease severity. Glycosylation plays a critical role in influenza pathogenesis; however, the molecular drivers of influenza outcomes remain unknown. In this work, we characterized the host glycomic response to the H1N1 2009 pandemic influenza A virus (H1N1pdm09) as a function of age-dependent severity in a ferret model. Using our dual-color lectin microarray technology, we examined baseline glycosylation and glycomic response to infection in newly weaned and aged animals, models for young children and the elderly, respectively. Compared to adult uninfected ferrets, we observed higher levels of α-2,6-sialosides, the receptor for H1N1pdm09, in newly weaned and aged animals. We also observed age-dependent loss of O-linked α-2,3-sialosides. The loss of these highly charged groups may impact viral clearance by mucins, which corresponds to the lower clearance rates observed in aged animals. Upon infection, we observed dramatic changes in the glycomes of aged animals, a population severely impacted by the virus. In contrast, no significant alterations were observed in the newly weaned animals, which show mild to moderate responses to the H1N1pdm09. High mannose, a glycan recently identified as a marker of severity in adult animals, increased with severity in the aged population. However, the response was delayed, in line with the delayed development of pneumonia observed. Overall, our results may help explain the differential susceptibility to influenza A infection and severity observed as a function of age.
Collapse
Affiliation(s)
- Shuhui Chen
- Biomedical Research Institute, Department of Chemistry, New York University, NY, 10003, USA
| | - Brian Kasper
- Biomedical Research Institute, Department of Chemistry, New York University, NY, 10003, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Lauren P. Lashua
- Center for Genomics & Systems Biology, Department of Biology, New York University, NY, 10003, USA
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, GA, 30602, USA
| | - Elodie Ghedin
- Center for Genomics & Systems Biology, Department of Biology, New York University, NY, 10003, USA
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID/NIH, Bethesda, MD, 20894, USA
| | - Lara K. Mahal
- Biomedical Research Institute, Department of Chemistry, New York University, NY, 10003, USA
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, CANADA
| |
Collapse
|
125
|
Bhattacharjee S, Brayden DJ. Addressing the challenges to increase the efficiency of translating nanomedicine formulations to patients. Expert Opin Drug Discov 2020; 16:235-254. [PMID: 33108229 DOI: 10.1080/17460441.2021.1826434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Nanotechnology is in a growth phase for drug delivery and medical imaging. Nanomaterials with unique properties present opportunities for encapsulation of therapeutics and imaging agents, along with conjugation to ligands for targeting. Favorable chemistry of nanomaterials can create formulations that address critical challenges for therapeutics, such as insolubility and a low capacity to cross the blood-brain-barrier (BBB) and intestinal wall. AREAS COVERED The authors investigate challenges faced during translation of nanomedicines while suggesting reasons as to why some nanoformulations have under-performed in clinical trials. They assess physiological barriers such as the BBB and gut mucus that nanomedicines must overcome to deliver cargos. They also provide an overview with examples of how nanomedicines can be designed to improve localization and site-specific delivery (e.g., encapsulation, bioconjugation, and triggered-release). EXPERT OPINION There are examples where nanomedicines have demonstrated improved efficacy of payload in humans; however, most of the advantages conferred were in improved pharmacokinetics and reduced toxicity. Problematic data show susceptibility of nanoformulations against natural protective mechanisms present in the body, including distribution impediment by physiological barriers and activation of the reticuloendothelial system. Further initiatives should address current challenges while expanding the scope of nanomedicine into advanced biomedical imaging and antibiotic delivery.
Collapse
Affiliation(s)
- Sourav Bhattacharjee
- School of Veterinary Medicine, University College Dublin (UCD), Belfield, Dublin, Ireland
| | - David J Brayden
- School of Veterinary Medicine, University College Dublin (UCD), Belfield, Dublin, Ireland.,Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Belfield, Dublin, Ireland
| |
Collapse
|
126
|
Gudmundsdottir Á, Scheving R, Lindberg F, Stefansson B. Inactivation of SARS-CoV-2 and HCoV-229E in vitro by ColdZyme® a medical device mouth spray against the common cold. J Med Virol 2020; 93:1792-1795. [PMID: 32975843 PMCID: PMC7537187 DOI: 10.1002/jmv.26554] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022]
Abstract
Background The coronavirus disease 2019 (COVID‐19) pandemic calls for effective and safe treatments. Severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) causing COVID‐19 actively replicates in the throat, unlike SARS‐CoV, and shows high pharyngeal viral shedding even in patients with mild symptoms of the disease. HCoV‐229E is one of four coronaviruses causing the common cold. In this study, the efficacy of ColdZyme® (CZ‐MD), a medical device mouth spray, was tested against SARS‐CoV‐2 and HCoV‐229E in vitro. The CZ‐MD provides a protective glycerol barrier containing cod trypsin as an ancillary component. Combined, these ingredients can inactivate common cold viruses in the throat and mouth. The CZ‐MD is believed to act on the viral surface proteins that would perturb their entry pathway into cells. The efficacy and safety of the CZ‐MD have been demonstrated in clinical trials on the common cold. Method of Study The ability of the CZ‐MD to inactivate SARS‐CoV‐2 and HCoV‐229E was tested using an in vitro virucidal suspension test (ASTM E1052). Results CZ‐MD inactivated SARS‐CoV‐2 by 98.3% and HCoV‐229E by 99.9%. Conclusion CZ‐MD mouth spray can inactivate the respiratory coronaviruses SARS‐CoV‐2 and HCoV‐229E in vitro. Although the in vitro results presented cannot be directly translated into clinical efficacy, the study indicates that CZ‐MD might offer a protective barrier against SARS‐CoV‐2 and a decreased risk of COVID‐19 transmission. The ability of ColdZyme® (CZ‐MD), a medical device mouth spray, to inactivate coronaviruses (SARS‐CoV‐2 and HCoV‐229E) was tested using an in vitro virucidal suspension test (ASTM E1052). CZ‐MD mouth spray inactivated SARS‐CoV‐2 and HCoV‐229E in vitro by 98.3% and 99.9% respectively. The study indicates that CZ‐MD might offer a protective barrier against SARS‐CoV‐2 and a decreased risk of COVID‐19 transmission.
Collapse
Affiliation(s)
- Ágústa Gudmundsdottir
- Faculty of Food Science and Nutrition, School of Health Sciences, University of Iceland, Reykjavík, Iceland.,Zymetech, Reykjavík, Iceland
| | | | - Fredrik Lindberg
- Enzymatica AB, Lund, Sweden.,Medical Faculty, Lund University, Lund, Sweden
| | | |
Collapse
|
127
|
The Small RNA ErsA Plays a Role in the Regulatory Network of Pseudomonas aeruginosa Pathogenicity in Airway Infections. mSphere 2020; 5:5/5/e00909-20. [PMID: 33055260 PMCID: PMC7565897 DOI: 10.1128/msphere.00909-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bacterial small RNAs play a remarkable role in the regulation of functions involved in host-pathogen interaction. ErsA is a small RNA of Pseudomonas aeruginosa that contributes to the regulation of bacterial virulence traits such as biofilm formation and motility. Shown to take part in a regulatory circuit under the control of the envelope stress response sigma factor σ22, ErsA targets posttranscriptionally the key virulence-associated gene algC Moreover, ErsA contributes to biofilm development and motility through the posttranscriptional modulation of the transcription factor AmrZ. Intending to evaluate the regulatory relevance of ErsA in the pathogenesis of respiratory infections, we analyzed the impact of ErsA-mediated regulation on the virulence potential of P. aeruginosa and the stimulation of the inflammatory response during the infection of bronchial epithelial cells and a murine model. Furthermore, we assessed ErsA expression in a collection of P. aeruginosa clinical pulmonary isolates and investigated the link of ErsA with acquired antibiotic resistance by generating an ersA gene deletion mutant in a multidrug-resistant P. aeruginosa strain which has long been adapted in the airways of a cystic fibrosis (CF) patient. Our results show that the ErsA-mediated regulation is relevant for the P. aeruginosa pathogenicity during acute infection and contributes to the stimulation of the host inflammatory response. Besides, ErsA was able to be subjected to selective pressure for P. aeruginosa pathoadaptation and acquirement of resistance to antibiotics commonly used in clinical practice during chronic CF infections. Our findings establish the role of ErsA as an important regulatory element in the host-pathogen interaction.IMPORTANCE Pseudomonas aeruginosa is one of the most critical multidrug-resistant opportunistic pathogens in humans, able to cause both lethal acute and chronic lung infections. Thorough knowledge of the regulatory mechanisms involved in the establishment and persistence of the airways infections by P. aeruginosa remains elusive. Emerging candidates as molecular regulators of pathogenesis in P. aeruginosa are small RNAs, which act posttranscriptionally as signal transducers of host cues. Known for being involved in the regulation of biofilm formation and responsive to envelope stress response, we show that the small RNA ErsA can play regulatory roles in acute infection, stimulation of host inflammatory response, and mechanisms of acquirement of antibiotic resistance and adaptation during the chronic lung infections of cystic fibrosis patients. Elucidating the complexity of the networks regulating host-pathogen interactions is crucial to identify novel targets for future therapeutic applications.
Collapse
|
128
|
Habibi MS, Thwaites RS, Chang M, Jozwik A, Paras A, Kirsebom F, Varese A, Owen A, Cuthbertson L, James P, Tunstall T, Nickle D, Hansel TT, Moffatt MF, Johansson C, Chiu C, Openshaw PJM. Neutrophilic inflammation in the respiratory mucosa predisposes to RSV infection. Science 2020; 370:eaba9301. [PMID: 33033192 PMCID: PMC7613218 DOI: 10.1126/science.aba9301] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/30/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
The variable outcome of viral exposure is only partially explained by known factors. We administered respiratory syncytial virus (RSV) to 58 volunteers, of whom 57% became infected. Mucosal neutrophil activation before exposure was highly predictive of symptomatic RSV disease. This was associated with a rapid, presymptomatic decline in mucosal interleukin-17A (IL-17A) and other mediators. Conversely, those who resisted infection showed presymptomatic activation of IL-17- and tumor necrosis factor-related pathways. Vulnerability to infection was not associated with baseline microbiome but was reproduced in mice by preinfection chemokine-driven airway recruitment of neutrophils, which caused enhanced disease mediated by pulmonary CD8+ T cell infiltration. Thus, mucosal neutrophilic inflammation at the time of RSV exposure enhances susceptibility, revealing dynamic, time-dependent local immune responses before symptom onset and explaining the as-yet unpredictable outcomes of pathogen exposure.
Collapse
Affiliation(s)
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Agnieszka Jozwik
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Allan Paras
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Freja Kirsebom
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Augusto Varese
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Amber Owen
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Leah Cuthbertson
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Phillip James
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - David Nickle
- Genetics & Pharmacogenomics, Department of Translational Medicine, Merck & Co., Inc., Boston, MA, USA
| | - Trevor T Hansel
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Miriam F Moffatt
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Cecilia Johansson
- National Heart and Lung Institute, Imperial College London, London, UK.
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK.
| | | |
Collapse
|
129
|
Immunopathogenesis of infectious bronchitis virus Q1 in specific pathogen free chicks. Microb Pathog 2020; 149:104535. [PMID: 32980469 DOI: 10.1016/j.micpath.2020.104535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/17/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
The immunopathogenesis of avian coronavirus, infectious bronchitis virus (IBV) Q1, was investigated in specific pathogen free chicks. Following infection, chicks exhibited respiratory clinical signs and reduced body weight. Oropharyngeal (OP) and cloacal (CL) swabs were collected at intervals and found to be RT-PCR positive, with a greater number of partial-S1 amino acid changes noted in CL swabs compared to OP swabs. In tissue samples, IBV viral load peaked 9 days post infection (dpi) in the trachea and kidneys, and 14 dpi in the proventriculus. At 28 dpi, ELISA data showed that 63% of infected chicks seroconverted. There was significantly higher mRNA up-regulation of IFN-α, TLR3, MDA5, LITAF, IL-1β and IL-6 in the trachea compared to the kidneys. Findings presented here demonstrate that this Q1 isolate induces greater lesions and host innate immune responses in chickens' tracheas compared to the kidneys.
Collapse
|
130
|
Poon WCK, Brown AT, Direito SOL, Hodgson DJM, Le Nagard L, Lips A, MacPhee CE, Marenduzzo D, Royer JR, Silva AF, Thijssen JHJ, Titmuss S. Soft matter science and the COVID-19 pandemic. SOFT MATTER 2020; 16:8310-8324. [PMID: 32909024 DOI: 10.1039/d0sm01223h] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Much of the science underpinning the global response to the COVID-19 pandemic lies in the soft matter domain. Coronaviruses are composite particles with a core of nucleic acids complexed to proteins surrounded by a protein-studded lipid bilayer shell. A dominant route for transmission is via air-borne aerosols and droplets. Viral interaction with polymeric body fluids, particularly mucus, and cell membranes controls their infectivity, while their interaction with skin and artificial surfaces underpins cleaning and disinfection and the efficacy of masks and other personal protective equipment. The global response to COVID-19 has highlighted gaps in the soft matter knowledge base. We survey these gaps, especially as pertaining to the transmission of the disease, and suggest questions that can (and need to) be tackled, both in response to COVID-19 and to better prepare for future viral pandemics.
Collapse
Affiliation(s)
- Wilson C K Poon
- Edinburgh Complex Fluids Partnership (ECFP), SUPA and School of Physics & Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| | - Aidan T Brown
- Edinburgh Complex Fluids Partnership (ECFP), SUPA and School of Physics & Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| | - Susana O L Direito
- Edinburgh Complex Fluids Partnership (ECFP), SUPA and School of Physics & Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| | - Daniel J M Hodgson
- Edinburgh Complex Fluids Partnership (ECFP), SUPA and School of Physics & Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| | - Lucas Le Nagard
- Edinburgh Complex Fluids Partnership (ECFP), SUPA and School of Physics & Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| | - Alex Lips
- Edinburgh Complex Fluids Partnership (ECFP), SUPA and School of Physics & Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| | - Cait E MacPhee
- Edinburgh Complex Fluids Partnership (ECFP), SUPA and School of Physics & Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| | - Davide Marenduzzo
- Edinburgh Complex Fluids Partnership (ECFP), SUPA and School of Physics & Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| | - John R Royer
- Edinburgh Complex Fluids Partnership (ECFP), SUPA and School of Physics & Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| | - Andreia F Silva
- Edinburgh Complex Fluids Partnership (ECFP), SUPA and School of Physics & Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| | - Job H J Thijssen
- Edinburgh Complex Fluids Partnership (ECFP), SUPA and School of Physics & Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| | - Simon Titmuss
- Edinburgh Complex Fluids Partnership (ECFP), SUPA and School of Physics & Astronomy, The University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK.
| |
Collapse
|
131
|
Induction of the Antiviral Immune Response and Its Circumvention by Coronaviruses. Viruses 2020; 12:v12091039. [PMID: 32961897 PMCID: PMC7551260 DOI: 10.3390/v12091039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/12/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022] Open
Abstract
Some coronaviruses are zoonotic viruses of human and veterinary medical importance. The novel coronavirus, severe acute respiratory symptoms coronavirus 2 (SARS-CoV-2), associated with the current global pandemic, is characterized by pneumonia, lymphopenia, and a cytokine storm in humans that has caused catastrophic impacts on public health worldwide. Coronaviruses are known for their ability to evade innate immune surveillance exerted by the host during the early phase of infection. It is important to comprehensively investigate the interaction between highly pathogenic coronaviruses and their hosts. In this review, we summarize the existing knowledge about coronaviruses with a focus on antiviral immune responses in the respiratory and intestinal tracts to infection with severe coronaviruses that have caused epidemic diseases in humans and domestic animals. We emphasize, in particular, the strategies used by these coronaviruses to circumvent host immune surveillance, mainly including the hijack of antigen-presenting cells, shielding RNA intermediates in replication organelles, 2′-O-methylation modification for the evasion of RNA sensors, and blocking of interferon signaling cascades. We also provide information about the potential development of coronavirus vaccines and antiviral drugs.
Collapse
|
132
|
Du X, Yang Y, Xiao G, Yang M, Yuan L, Qin L, He R, Wang L, Wu M, Wu S, Feng J, Xiang Y, Qu X, Liu H, Qin X, Liu C. Respiratory syncytial virus infection-induced mucus secretion by down-regulation of miR-34b/c-5p expression in airway epithelial cells. J Cell Mol Med 2020; 24:12694-12705. [PMID: 32939938 PMCID: PMC7687004 DOI: 10.1111/jcmm.15845] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/15/2020] [Accepted: 08/19/2020] [Indexed: 12/21/2022] Open
Abstract
Severe RSV infection is the main cause of hospitalization to children under the age of five. The regulation of miRNAs on the severity of RSV infection is unclear. The aim of the study was to identify the critical differential expression miRNAs (DE miRNAs) that can regulate the pathological response in RSV‐infected airway epithelial cells. In this study, miRNA and mRNA chips of RSV‐infected airway epithelia from Gene Expression Omnibus (GEO) were screened and analysed, separately. DE miRNAs‐targeted genes were performed for further pathway and process enrichment analysis. DE miRNA‐targeted gene functional network was constructed on the basis of miRNA‐mRNA interaction. The screened critical miRNA was also investigated by bioinformatics analysis. Then, RSV‐infected human bronchial epithelial cells (HBECs) were constructed to verify the expression of the DE miRNAs. Finally, specific synthetic DE miRNAs mimics were used to confirm the effect of DE miRNAs on the RSV‐infected HBECs. 45 DE miRNAs were identified from GEO62306 dataset. Our results showed that hsa‐mir‐34b‐5p and hsa‐mir‐34c‐5p decreased significantly in HBECs after RSV infection. Consistent with the biometric analysis, hsa‐mir‐34b/c‐5p is involved in the regulation of mucin expression gene MUC5AC. In RSV‐infected HBECs, the inducement of MUC5AC production by decreased hsa‐mir‐34b/c‐5p was partly mediated through activation of c‐Jun. These findings provide new insights into the mechanism of mucus obstruction after RSV infection and represent valuable targets for RSV infection and airway obstruction treatment.
Collapse
Affiliation(s)
- Xizi Du
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China.,Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China.,Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, China
| | - Yu Yang
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ming Yang
- Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, Australia.,School of Basic Medical Sciences & Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Lin Yuan
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Ling Qin
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China.,Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, China
| | - Ruoxi He
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China.,Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, China
| | - Leyuan Wang
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Mengping Wu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - ShuangYan Wu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Juntao Feng
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China.,Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, China
| | - Yang Xiang
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Xiangping Qu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Huijun Liu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoqun Qin
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Chi Liu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China.,Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China.,Research Center of China-Africa Infectious Diseases, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
133
|
Decoding Susceptibility to Respiratory Viral Infections and Asthma Inception in Children. Int J Mol Sci 2020; 21:ijms21176372. [PMID: 32887352 PMCID: PMC7503410 DOI: 10.3390/ijms21176372] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 01/19/2023] Open
Abstract
Human Respiratory Syncytial Virus and Human Rhinovirus are the most frequent cause of respiratory tract infections in infants and children and are major triggers of acute viral bronchiolitis, wheezing and asthma exacerbations. Here, we will discuss the application of the powerful tools of systems biology to decode the molecular mechanisms that determine risk for infection and subsequent asthma. An important conceptual advance is the understanding that the innate immune system is governed by a Bow-tie architecture, where diverse input signals converge onto a few core pathways (e.g., IRF7), which in turn generate diverse outputs that orchestrate effector and regulatory functions. Molecular profiling studies in children with severe exacerbations of asthma/wheeze have identified two major immunological phenotypes. The IRF7hi phenotype is characterised by robust upregulation of antiviral response networks, and the IRF7lo phenotype is characterised by upregulation of markers of TGFβ signalling and type 2 inflammation. Similar phenotypes have been identified in infants and children with severe viral bronchiolitis. Notably, genome-wide association studies supported by experimental validation have identified key pathways that increase susceptibility to HRV infection (ORMDL3 and CHDR3) and modulate TGFβ signalling (GSDMB, TGFBR1, and SMAD3). Moreover, functional deficiencies in the activation of type I and III interferon responses are already evident at birth in children at risk of developing febrile lower respiratory tract infections and persistent asthma/wheeze, suggesting that the trajectory to asthma begins at birth or in utero. Finally, exposure to microbes and their products reprograms innate immunity and provides protection from the development of allergies and asthma in children, and therefore microbial products are logical candidates for the primary prevention of asthma.
Collapse
|
134
|
Esam Z. Protective potential of expectorants against COVID-19. Med Hypotheses 2020; 142:109844. [PMID: 32425306 PMCID: PMC7229712 DOI: 10.1016/j.mehy.2020.109844] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/26/2020] [Accepted: 05/12/2020] [Indexed: 12/03/2022]
Affiliation(s)
- Zohreh Esam
- Pharmaceutical Sciences Research Center, Department of Medicinal Chemistry, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Ayatollah Amoli Branch, Islamic Azad University, Amol, Iran
| |
Collapse
|
135
|
Alkhatib A. Antiviral Functional Foods and Exercise Lifestyle Prevention of Coronavirus. Nutrients 2020; 12:E2633. [PMID: 32872374 PMCID: PMC7551447 DOI: 10.3390/nu12092633] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/19/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
Novel coronavirus (COVID-19) is causing global mortality and lockdown burdens. A compromised immune system is a known risk factor for all viral influenza infections. Functional foods optimize the immune system capacity to prevent and control pathogenic viral infections, while physical activity augments such protective benefits. Exercise enhances innate and adaptive immune systems through acute, transient, and long-term adaptations to physical activity in a dose-response relationship. Functional foods prevention of non-communicable disease can be translated into protecting against respiratory viral infections and COVID-19. Functional foods and nutraceuticals within popular diets contain immune-boosting nutraceuticals, polyphenols, terpenoids, flavonoids, alkaloids, sterols, pigments, unsaturated fatty-acids, micronutrient vitamins and minerals, including vitamin A, B6, B12, C, D, E, and folate, and trace elements, including zinc, iron, selenium, magnesium, and copper. Foods with antiviral properties include fruits, vegetables, fermented foods and probiotics, olive oil, fish, nuts and seeds, herbs, roots, fungi, amino acids, peptides, and cyclotides. Regular moderate exercise may contribute to reduce viral risk and enhance sleep quality during quarantine, in combination with appropriate dietary habits and functional foods. Lifestyle and appropriate nutrition with functional compounds may offer further antiviral approaches for public health.
Collapse
Affiliation(s)
- Ahmad Alkhatib
- School of Health and Life sciences, Teesside University, Tees Valley, Middlesbrough TS1 3BX, UK
| |
Collapse
|
136
|
Debnath SK, Srivastava R, Omri A. Emerging therapeutics for the management of COVID 19. Expert Opin Emerg Drugs 2020; 25:337-351. [PMID: 32799566 DOI: 10.1080/14728214.2020.1810663] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION The coronavirus-19 (COVID-19) disease pandemic can be characterized as the most critical and changeable hazard to healthcare systems in eras. The high fatality rate associated with coronavirus infection underlines the urgent need for an effective treatment to reduce disease severity and mortality. AREAS COVERED A detailed search for treatments related to severe acute respiratory syndrome coronavirus-2 (SARS-CoV2) was carried out using PubMed. Components of the virus relevant to the infectious mechanism were identified. We have highlighted all the latest emerging and repurposed drugs that were found to be active against this novel coronavirus and classified these drugs according to their category. Different drug targets are discussed in order to identify new molecules or new combinations as candidates to manage SARS-CoV2/COVID-19 infections. EXPERT OPINION The development of novel molecules and vaccines has been a challenge during this urgent crisis. Nucleoside analogs and IL-6 receptor antagonists have been identified as the best candidates for treatment of this disease. Multi-drug therapy by targeting different pathways will need to be corroborated and then confirmed through clinical trials. Until a vaccine is available, an alternative drug regimen needs to be adopted by clinicians in the management of coronavirus symptoms.
Collapse
Affiliation(s)
- Sujit Kumar Debnath
- Nanobios Lab, Department of Biosciences and Bioengineering, IIT Bombay , Mumbai, India
| | - Rohit Srivastava
- Nanobios Lab, Department of Biosciences and Bioengineering, IIT Bombay , Mumbai, India
| | - Abdelwahab Omri
- The Novel Drug and Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University , Sudbury, Ontario, Canada
| |
Collapse
|
137
|
Sapkota D, Søland TM, Galtung HK, Sand LP, Giannecchini S, To KKW, Mendes-Correa MC, Giglio D, Hasséus B, Braz-Silva PH. COVID-19 salivary signature: diagnostic and research opportunities. J Clin Pathol 2020; 74:jclinpath-2020-206834. [PMID: 32769214 DOI: 10.1136/jclinpath-2020-206834] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/16/2020] [Accepted: 07/21/2020] [Indexed: 01/05/2023]
Abstract
The COVID-19 (caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)) epidemic started in Wuhan (Hubei Province, China) in mid-December 2019 and quickly spread across the world as a pandemic. As a key to tracing the disease and to implement strategies aimed at breaking the chain of disease transmission, extensive testing for SARS-CoV-2 was suggested. Although nasopharyngeal/oropharyngeal swabs are the most commonly used biological samples for SARS-CoV-2 diagnosis, they have a number of limitations related to sample collection and healthcare personnel safety. In this context, saliva is emerging as a promising alternative to nasopharyngeal/oropharyngeal swabs for COVID-19 diagnosis and monitoring. Saliva collection, being a non-invasive approach with possibility for self-collection, circumvents to a great extent the limitations associated with the use of nasopharyngeal/oropharyngeal swabs. In addition, various salivary biomarkers including the salivary metabolomics offer a high promise to be useful for better understanding of COVID-19 and possibly in the identification of patients with various degrees of severity, including asymptomatic carriers. This review summarises the clinical and scientific basis for the potential use of saliva for COVID-19 diagnosis and disease monitoring. Additionally, we discuss saliva-based biomarkers and their potential clinical and research applications related to COVID-19.
Collapse
Affiliation(s)
- Dipak Sapkota
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Tine Merete Søland
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
- Department of Pathology, Rikshospitalet University Hospital, Oslo, Norway
| | - Hilde Kanli Galtung
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Lars Peter Sand
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Simone Giannecchini
- Department of Experimental and Clinical Medicine, Universita degli Studi di Firenze, Firenze, Toscana, Italy
| | - Kelvin K W To
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, Li KaShing Faculty of Medicine of the University of Hong Kong, Hong Kong, China
- Department of Microbiology, Queen Mary Hospital, Hong Kong, China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong - Shenzhen Hospital, Shenzhen, China
| | - Maria Cassia Mendes-Correa
- Laboratory of Virology (LIM-52), Institute of Tropical Medicine of São Paulo, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Daniel Giglio
- Department of Clinical Oncology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Bengt Hasséus
- Department of Oral Medicine and Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinic of Oral Medicine, Region Västra Götaland, Gothenburg, Sweden
| | - Paulo Henrique Braz-Silva
- Laboratory of Virology (LIM-52), Institute of Tropical Medicine of São Paulo, School of Medicine, University of São Paulo, São Paulo, Brazil
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
138
|
Xing L, Chang X, Shen L, Zhang C, Fan Y, Cho C, Zhang Z, Jiang H. Progress in drug delivery system for fibrosis therapy. Asian J Pharm Sci 2020; 16:47-61. [PMID: 33613729 PMCID: PMC7878446 DOI: 10.1016/j.ajps.2020.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/22/2020] [Accepted: 06/22/2020] [Indexed: 12/18/2022] Open
Abstract
Fibrosis is a necessary process in the progression of chronic disease to cirrhosis or even cancer, which is a serious disease threatening human health. Recent studies have shown that the early treatment of fibrosis is turning point and particularly important. Therefore, how to reverse fibrosis has become the focus and research hotspot in recent years. So far, the considerable progress has been made in the development of effective anti-fibrosis drugs and targeted drug delivery. Moreover, the existing research results will lay the foundation for more breakthrough delivery systems to achieve better anti-fibrosis effects. Herein, this review summaries anti-fibrosis delivery systems focused on three major organ fibrotic diseases such as liver, pulmonary, and renal fibrosis accompanied by the elaboration of relevant pathological mechanisms, which will provide inspiration and guidance for the design of fibrosis drugs and therapeutic systems in the future.
Collapse
Affiliation(s)
- Lei Xing
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Xin Chang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Lijun Shen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Chenglu Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yatong Fan
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Chongsu Cho
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
- Corresponding authors.
| | - Zhiqi Zhang
- Department of General Surgery, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200081 China
- Corresponding authors.
| | - Hulin Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
- Corresponding authors.
| |
Collapse
|
139
|
Urine Levels of γ-Aminobutyric Acid Are Associated with the Severity of Respiratory Syncytial Virus Infection in Infancy. Ann Am Thorac Soc 2020; 17:1489-1493. [PMID: 32649834 DOI: 10.1513/annalsats.201910-738rl] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
140
|
Affiliation(s)
- Kimberly A Prather
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA 92037, USA.
| | - Chia C Wang
- Department of Chemistry, National Sun Yat-sen University, Kaohsiung, Taiwan 804, Republic of China
- Aerosol Science Research Center, National Sun Yat-Sen University, Kaohsiung, Taiwan 804, Republic of China
| | - Robert T Schooley
- Department of Medicine, Division of Infectious Diseases and Global Public Health, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
141
|
Host-Pathogen Responses to Pandemic Influenza H1N1pdm09 in a Human Respiratory Airway Model. Viruses 2020; 12:v12060679. [PMID: 32599823 PMCID: PMC7354428 DOI: 10.3390/v12060679] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
The respiratory Influenza A Viruses (IAVs) and emerging zoonotic viruses such as Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) pose a significant threat to human health. To accelerate our understanding of the host–pathogen response to respiratory viruses, the use of more complex in vitro systems such as normal human bronchial epithelial (NHBE) cell culture models has gained prominence as an alternative to animal models. NHBE cells were differentiated under air-liquid interface (ALI) conditions to form an in vitro pseudostratified epithelium. The responses of well-differentiated (wd) NHBE cells were examined following infection with the 2009 pandemic Influenza A/H1N1pdm09 strain or following challenge with the dsRNA mimic, poly(I:C). At 30 h postinfection with H1N1pdm09, the integrity of the airway epithelium was severely impaired and apical junction complex damage was exhibited by the disassembly of zona occludens-1 (ZO-1) from the cell cytoskeleton. wdNHBE cells produced an innate immune response to IAV-infection with increased transcription of pro- and anti-inflammatory cytokines and chemokines and the antiviral viperin but reduced expression of the mucin-encoding MUC5B, which may impair mucociliary clearance. Poly(I:C) produced similar responses to IAV, with the exception of MUC5B expression which was more than 3-fold higher than for control cells. This study demonstrates that wdNHBE cells are an appropriate ex-vivo model system to investigate the pathogenesis of respiratory viruses.
Collapse
|
142
|
Powell H, Pekosz A. Neuraminidase antigenic drift of H3N2 clade 3c.2a viruses alters virus replication, enzymatic activity and inhibitory antibody binding. PLoS Pathog 2020; 16:e1008411. [PMID: 32598381 PMCID: PMC7351227 DOI: 10.1371/journal.ppat.1008411] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 07/10/2020] [Accepted: 05/11/2020] [Indexed: 11/19/2022] Open
Abstract
In the 2014-2015 influenza season a novel neuraminidase (NA) genotype was detected in global human influenza A surveillance. This novel genotype encoded an N-linked glycosylation site at position 245-247 in the NA protein from clade 3c.2a H3N2 viruses. In the years following the 2014-2015 season, this novel NA glycosylation genotype quickly dominated the human H3N2 population of viruses. To assess the effect this novel N-linked glycan has on virus fitness and antibody binding, recombinant viruses with (NA Gly+) or without (NA Gly-) the 245 NA glycan were created. Viruses with the 245 NA Gly+ genotype grew to a significantly lower infectious virus titer on primary, differentiated human nasal epithelial cells (hNEC) compared to viruses with the 245 NA Gly- genotype, but growth was similar on immortalized cells. The 245 NA Gly+ blocked human and rabbit monoclonal antibodies that target the enzymatic site from binding to their epitope. Additionally, viruses with the 245 NA Gly+ genotype had significantly lower enzymatic activity compared to viruses with the 245 NA Gly- genotype. Human monoclonal antibodies that target residues near the 245 NA glycan were less effective at inhibiting NA enzymatic activity and virus replication of viruses encoding an NA Gly+ protein compared to ones encoding NA Gly- protein. Additionally, a recombinant H6N2 virus with the 245 NA Gly+ protein was more resistant to enzymatic inhibition from convalescent serum from H3N2-infected humans compared to viruses with the 245 NA Gly- genotype. Finally, the 245 NA Gly+ protected from NA antibody mediated virus neutralization. These results suggest that while the 245 NA Gly+ decreases virus replication in hNECs and decreases enzymatic activity, the 245 NA glycan blocks the binding of monoclonal and human serum NA specific antibodies that would otherwise inhibit enzymatic activity and virus replication.
Collapse
Affiliation(s)
- Harrison Powell
- Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Andrew Pekosz
- Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
143
|
Das P, Choudhuri T. Decoding the global outbreak of COVID-19: the nature is behind the scene. Virusdisease 2020; 31:106-112. [PMID: 32656307 PMCID: PMC7306562 DOI: 10.1007/s13337-020-00605-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 05/24/2020] [Indexed: 12/13/2022] Open
Abstract
The sudden emergence of SARS-CoV-2 causing the global pandemic is a major public health concern. Though the virus is considered as a novel entity, it is not a completely new member. It is just a new version of previously emerged human SARS corona virus. The rapid evolving nature by changing host body environment and extreme environmental stability, collectively makes SARS-CoV-2 into an extremely virulent genetic variant. The evolution of the virus has been occurred by the continuous process of molecular genetic manipulation, through mutation, deletion and genetic recombinationevents. Different host body environment acts as the supportive system for the pathogen which creates extreme selective pressure. By the process of genetic evolution the pathogen developes new characters. Then the new version of the virus has been naturally selected by susceptible human host and adapt itself inside the host body causing deadly effect. Moreover, extreme environmental stability helps in the process of viral survival outside the host and its transmission. Thus both the host body or internal environment and the external environment performs equally as a source, responsible for shaping the genetic evolution of the SARS-CoV-2 towards theCOVID-19 disease fitness in nature in a pandemic form.
Collapse
Affiliation(s)
- Piyanki Das
- Department of Biotechnology, Siksha-Bhavana, Visva-Bharati, Santiniketan, West Bengal 731235 India
| | - Tathagata Choudhuri
- Department of Biotechnology, Siksha-Bhavana, Visva-Bharati, Santiniketan, West Bengal 731235 India
| |
Collapse
|
144
|
Eccles R. Respiratory mucus and persistence of virus on surfaces. J Hosp Infect 2020; 105:350. [PMID: 32243948 PMCID: PMC7270553 DOI: 10.1016/j.jhin.2020.03.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 11/23/2022]
|
145
|
Abstract
Introduction:
The common cold syndrome of acute upper respiratory tract viral infection is the most common disease among mankind and is an extremely common illness in children. There is a great need for a safe and effective antiviral treatment with minimal side effects. The challenge in developing a treatment is the numerous and varied respiratory viruses that cause this common illness and the need for a treatment with good tolerability and safety.
Explanation:
All respiratory viruses must reach the cell surface by passing through respiratory fluid and mucus, and this common feature may allow for the development of antivirals that capture viruses during this transit.
This article discusses how large polyanionic molecules such as iota-carrageenan may trap positively charged respiratory viruses. Iota-carrageenan is a large polysaccharide molecule which is neither absorbed from the respiratory tract nor metabolised. It, therefore, does not have any pharmacological properties. Iota-carrageenan nasal spray has been shown to reduce the titres of respiratory viruses and to reduce the severity of symptoms in placebo-controlled clinical trials, including children and adults. The results of four clinical trials are presented.
Conclusion:
Iota-carrageenan is a good candidate as a safe and effective non-specific antiviral treatment for common cold, and more research is justified on polyanionic molecules like carrageenans as antivirals.
Collapse
|
146
|
Abstract
INTRODUCTION Otitis media (OM) is a spectrum of infectious and inflammatory diseases that involve the middle ear. It includes acute otitis media (AOM), otitis media with effusion (OME) and chronic suppurative otitis media (CSOM). AREAS COVERED This manuscript discusses some of the emerging and unsolved problems regarding OM, and some of the newly developed prophylactic and therapeutic medical measures. EXPERT OPINION In recent years, considerable progress in the knowledge of OM physiopathology has been made. However, although extremely common, diseases included under OM have not been adequately studied, and many areas of development, evolution and possible treatments of these pathologies are not defined. It is necessary that these deficiencies be quickly overcome if we want to reduce the total burden of a group of diseases that still have extremely high medical, social and economic relevance.
Collapse
Affiliation(s)
- Nicola Principi
- Emeritus of Pediatrics, Università Degli Studi Di Milano , Milan, Italy
| | - Susanna Esposito
- Pediatric Clinic, Pietro Barilla Children's Hospital, Department of Medicine and Surgery, University of Parma , Parma, Italy
| |
Collapse
|
147
|
Cilia and centrosomes: Ultrastructural and mechanical perspectives. Semin Cell Dev Biol 2020; 110:61-69. [PMID: 32307225 DOI: 10.1016/j.semcdb.2020.03.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/12/2020] [Accepted: 03/21/2020] [Indexed: 11/20/2022]
Abstract
Cilia and centrosomes of eukaryotic cells play important roles in cell movement, fluid transport, extracellular sensing, and chromosome division. The physiological functions of cilia and centrosomes are generated by their dynamics, motions, and forces controlled by the physical, chemical, and biological environments. How an individual cilium achieves its beat pattern and induces fluid flow is governed by its ultrastructure as well as the coordination of associated molecular motors. Thus, a bottom-up understanding of the physiological functions of cilia and centrosomes from the molecular to tissue levels is required. Correlations between the structure and motion can be understood in terms of mechanics. This review first focuses on cilia and centrosomes at the molecular level, introducing their ultrastructure. We then shift to the organelle level and introduce the kinematics and mechanics of cilia and centrosomes. Next, at the tissue level, we introduce nodal ciliary dynamics and nodal flow, which play crucial roles in the organogenetic process of left-right asymmetry. We also introduce respiratory ciliary dynamics and mucous flow, which are critical for protecting the epithelium from drying and exposure to harmful particles and viruses, i.e., respiratory clearance function. Finally, we discuss the future research directions in this field.
Collapse
|
148
|
Gellatly S, Pavelka N, Crue T, Schweitzer KS, Day BJ, Min E, Numata M, Voelker DR, Scruggs A, Petrache I, Chu HW. Nicotine-Free e-Cigarette Vapor Exposure Stimulates IL6 and Mucin Production in Human Primary Small Airway Epithelial Cells. J Inflamm Res 2020; 13:175-185. [PMID: 32368126 PMCID: PMC7170627 DOI: 10.2147/jir.s244434] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 03/19/2020] [Indexed: 01/08/2023] Open
Abstract
PURPOSE Electronic cigarettes (e-cigs) are relatively new devices that allow the user to inhale a heated and aerosolized solution. At present, little is known about their health effects in the human lung, particularly in the small airways (<2 mm in diameter), a key site of airway obstruction and destruction in chronic obstructive pulmonary disease and other acute and chronic lung conditions. The aim of this study was to investigate the effect of e-cigarettes on human distal airway inflammation and remodeling. METHODS We isolated primary small airway epithelial cells from donor lungs without known lung disease. Small airway epithelial cells were cultured at air-liquid interface and exposed to 15 puffs vapor obtained by heating a commercially available e-cigarette solution (e-vapor) with or without nicotine. After 24 hrs of e-vapor exposure, basolateral and apical media as well as cell lysates were collected to measure the pleiotropic cytokine interleukin 6 (IL6) and MUC5AC, one of the major components in mucus. RESULTS Unlike the nicotine-containing e-vapor, nicotine-free e-vapor significantly increased the amount of IL6, which was coupled with increased levels of intracellular MUC5AC protein. Importantly, a neutralizing IL6 antibody (vs an IgG isotype control) significantly inhibited the production of MUC5AC induced by nicotine-free e-vapor. CONCLUSION Our results suggest that human small airway epithelial cells exposed to nicotine-free e-vapor increase the inflammatory response and mucin production, which may contribute to distal lung airflow limitation and airway obstruction.
Collapse
Affiliation(s)
- Shaan Gellatly
- Department of Medicine, National Jewish Health, Denver, CO80206, USA
| | - Nicole Pavelka
- Department of Medicine, National Jewish Health, Denver, CO80206, USA
| | - Taylor Crue
- Department of Medicine, National Jewish Health, Denver, CO80206, USA
| | | | - Brian J Day
- Department of Medicine, National Jewish Health, Denver, CO80206, USA
| | - Elysia Min
- Department of Medicine, National Jewish Health, Denver, CO80206, USA
| | - Mari Numata
- Department of Medicine, National Jewish Health, Denver, CO80206, USA
| | - Dennis R Voelker
- Department of Medicine, National Jewish Health, Denver, CO80206, USA
| | - April Scruggs
- Department of Medicine, National Jewish Health, Denver, CO80206, USA
| | - Irina Petrache
- Department of Medicine, National Jewish Health, Denver, CO80206, USA
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, CO80206, USA
| |
Collapse
|
149
|
Host Mucin Is Exploited by Pseudomonas aeruginosa To Provide Monosaccharides Required for a Successful Infection. mBio 2020; 11:mBio.00060-20. [PMID: 32127446 PMCID: PMC7064748 DOI: 10.1128/mbio.00060-20] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
One of the primary functions of the mucosal barrier, found lining epithelial cells, is to serve as a first-line of defense against microbial pathogens. The major structural components of mucus are heavily glycosylated proteins called mucins. Mucins are key components of the innate immune system as they aid in the clearance of pathogens and can decrease pathogen virulence. It has also been recently reported that individual mucins and derived glycans can attenuate the virulence of the human pathogen Pseudomonas aeruginosa Here, we show data indicating that mucins not only play a role in host defense but that they can also be subverted by P. aeruginosa to cause disease. We found that the mucin MUL-1 and mucin-derived monosaccharides N-acetyl-galactosamine and N-acetylglucosamine are required for P. aeruginosa killing of Caenorhabditis elegans We also found that the defective adhesion of P. aeruginosa to human lung alveolar epithelial cells, deficient in the mucin MUC1, can be reversed by the addition of individual monosaccharides. The monosaccharides identified in this study are found in a wide range of organisms where they act as host factors required for bacterial pathogenesis. While mucins in C. elegans lack sialic acid caps, which makes their monosaccharides readily available, they are capped in other species. Pathogens such as P. aeruginosa that lack sialidases may rely on enzymes from other bacteria to utilize mucin-derived monosaccharides.IMPORTANCE One of the first lines of defense present at mucosal epithelial tissues is mucus, which is a highly viscous material formed by mucin glycoproteins. Mucins serve various functions, but importantly they aid in the clearance of pathogens and debris from epithelial barriers and serve as innate immune factors. In this study, we describe a requirement of host monosaccharides, likely derived from host mucins, for the ability of Pseudomonas aeruginosa to colonize the intestine and ultimately cause death in Caenorhabditis elegans We also demonstrate that monosaccharides alter the ability of bacteria to bind to both Caenorhabditis elegans intestinal cells and human lung alveolar epithelial cells, suggesting that there are conserved mechanisms underlying host-pathogen interactions in a range of organisms. By gaining a better understanding of pathogen-mucin interactions, we can develop better approaches to protect against pathogen infection.
Collapse
|
150
|
Abstract
Mucus in the gastrointestinal (GI) tract is the primary point-of-interaction between humans and their gut microbiota. This intimates that mucus not only ensures protection against endogenous and exogenous opportunists but also provisions for the human microbiota to reside and flourish. With the emergence of living therapeutics, engineered microbes can deliver and produce increasingly complex medicine, and controlling the mucoadhesive properties of different microbial chassis can dictate dose-response in a patient. Here we present a redesigned, in vitro, plate-based assay to measure the mucus adhesion of various probiotics. Cell-mucus interactions were isolated by immobilizing mucus to the plate surface. Binding parameters were derived for each probiotic strain by measuring cell adhesion over a wide range of cell concentrations, providing dose-dependent adhesion metrics. Surface proteins and cell components known to influence mucoadhesion were then heterologously expressed or altered in Lactococcus lactis MG1363 and Escherichia coli Nissle 1917 to control mucus-binding capacity, avidity, and cooperativity.
Collapse
Affiliation(s)
- Zachary J. S. Mays
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Todd C. Chappell
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Nikhil U. Nair
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|