151
|
Xu X, Wu L, Sheng Y, Liu J, Xu Z, Kong W, Tang L, Chen Z. Airway microbiota in children with bronchial mucus plugs caused by Mycoplasma pneumoniae pneumonia. Respir Med 2020; 170:105902. [PMID: 32843185 DOI: 10.1016/j.rmed.2020.105902] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 01/29/2020] [Accepted: 02/14/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND There is increasing evidence for a role of lung microbiota in the pathogenesis of Mycoplasma pneumoniae pneumonia (MPP). However, the alterations of lung microbiota in MPP with bronchial mucus plugs and its role in disease pathogenesis remain poorly understood. METHODS In this prospective observational study, we performed a longitudinal 16S rRNA-based microbiome survey on bronchoalveolar lavage (BAL) samples collected from 31 MPP with bronchial mucus plugs and 52 MPP without mucus plugs. RESULTS Our study showed a clear difference in airway microbiota between MPP children with and without bronchial mucus plugs. The MPP children with mucus plugs had lower abundances of Sphingomonas and Elizabethkingia, and a high abundance of Mycoplasma compared with MPP children without mucus plugs, subsequently contributing to increased ratios of Mycoplasma to Sphingomonas and Mycoplasma to Elizabethkingia. Children's age, fever time and serum cytokine levels were associated with airway microbiota alteration. Furthermore, significant correlations between bacterial genus abundances were found in MPP children with mucus plugs. CONCLUSIONS Our results suggest an impact of airway microbiota on the clinical course of MPP in children, deserving further investigations.
Collapse
Affiliation(s)
- Xuefeng Xu
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, PR China; Department of Rheumatology Immunology & Allergy, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, PR China
| | - Lei Wu
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, PR China
| | - Yuanjian Sheng
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, PR China
| | - Jinling Liu
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, PR China
| | - Zhufei Xu
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, PR China
| | - Weixing Kong
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, PR China
| | - Lanfang Tang
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, PR China
| | - Zhimin Chen
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, PR China.
| |
Collapse
|
152
|
Blundell PA, Lu D, Dell A, Haslam S, Pleass RJ. Choice of Host Cell Line Is Essential for the Functional Glycosylation of the Fc Region of Human IgG1 Inhibitors of Influenza B Viruses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:1022-1034. [PMID: 31907284 PMCID: PMC6994840 DOI: 10.4049/jimmunol.1901145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022]
Abstract
Abs are glycoproteins that carry a conserved N-linked carbohydrate attached to the Fc whose presence and fine structure profoundly impacts on their in vivo immunogenicity, pharmacokinetics, and functional attributes. The host cell line used to produce IgG plays a major role in this glycosylation, as different systems express different glycosylation enzymes and transporters that contribute to the specificity and heterogeneity of the final IgG-Fc glycosylation profile. In this study, we compare two panels of glycan-adapted IgG1-Fc mutants expressed in either the human endothelial kidney 293-F or Chinese hamster ovary-K1 systems. We show that the types of N-linked glycans between matched pairs of Fc mutants vary greatly and in particular, with respect, to sialylation. These cell line effects on glycosylation profoundly influence the ability of the engineered Fcs to interact with either human or pathogen receptors. For example, we describe Fc mutants that potently disrupted influenza B-mediated agglutination of human erythrocytes when expressed in Chinese hamster ovary-K1, but not in human endothelial kidney 293-F cells.
Collapse
Affiliation(s)
- Patricia A Blundell
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, United Kingdom; and
| | - Dongli Lu
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Anne Dell
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Stuart Haslam
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Richard J Pleass
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, United Kingdom; and
| |
Collapse
|
153
|
LeMessurier KS, Tiwary M, Morin NP, Samarasinghe AE. Respiratory Barrier as a Safeguard and Regulator of Defense Against Influenza A Virus and Streptococcus pneumoniae. Front Immunol 2020; 11:3. [PMID: 32117216 PMCID: PMC7011736 DOI: 10.3389/fimmu.2020.00003] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/03/2020] [Indexed: 12/27/2022] Open
Abstract
The primary function of the respiratory system of gas exchange renders it vulnerable to environmental pathogens that circulate in the air. Physical and cellular barriers of the respiratory tract mucosal surface utilize a variety of strategies to obstruct microbe entry. Physical barrier defenses including the surface fluid replete with antimicrobials, neutralizing immunoglobulins, mucus, and the epithelial cell layer with rapidly beating cilia form a near impenetrable wall that separates the external environment from the internal soft tissue of the host. Resident leukocytes, primarily of the innate immune branch, also maintain airway integrity by constant surveillance and the maintenance of homeostasis through the release of cytokines and growth factors. Unfortunately, pathogens such as influenza virus and Streptococcus pneumoniae require hosts for their replication and dissemination, and prey on the respiratory tract as an ideal environment causing severe damage to the host during their invasion. In this review, we outline the host-pathogen interactions during influenza and post-influenza bacterial pneumonia with a focus on inter- and intra-cellular crosstalk important in pulmonary immune responses.
Collapse
Affiliation(s)
- Kim S LeMessurier
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| | - Meenakshi Tiwary
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| | - Nicholas P Morin
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Critical Care Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Amali E Samarasinghe
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| |
Collapse
|
154
|
Kida Y, Yamamoto T, Kuwano K. SdsA1, a secreted sulfatase, contributes to the in vivo virulence of Pseudomonas aeruginosa in mice. Microbiol Immunol 2020; 64:280-295. [PMID: 31907968 DOI: 10.1111/1348-0421.12772] [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: 09/26/2019] [Revised: 12/10/2019] [Accepted: 01/05/2020] [Indexed: 01/08/2023]
Abstract
Mucin is a glycoprotein that is the primary component of the mucus overlaying the epithelial tissues. Because mucin functions as a first line of the innate immune system, Pseudomonas aeruginosa appears to require interaction with mucin to establish infection in the host. However, the interactions between P. aeruginosa and mucin have been poorly understood. In this study, using in vivo expression technology (IVET), we attempted to identify mucin-inducible promoters that are likely to be involved in the establishment of P. aeruginosa infection. The IVET analysis revealed that the genes encoding glycosidases, sulfatases, and peptidases that are thought to be required for the utilization of mucin as a nutrient are present in 13 genes downstream of the identified promoters. Our results indicated that, among them, sdsA1 encoding a secreted sulfatase plays a central role in the degradation of mucin. It was then demonstrated that disruption of sdsA1 leads to a decreased release of sulfate from mucin and sulfated sugars. Furthermore, the sdsA1 mutant showed a reduction in the ability of mucin gel penetration and an attenuation of virulence in leukopenic mice compared with the wild-type strain. Collectively, these results suggest that SdsA1 plays an important role as a virulence factor of P. aeruginosa.
Collapse
Affiliation(s)
- Yutaka Kida
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Takeshi Yamamoto
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Koichi Kuwano
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, Fukuoka, Japan
| |
Collapse
|
155
|
Hamming PHE, Overeem NJ, Huskens J. Influenza as a molecular walker. Chem Sci 2020; 11:27-36. [PMID: 32153750 PMCID: PMC7021193 DOI: 10.1039/c9sc05149j] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022] Open
Abstract
The surface of the influenza virus is decorated with the receptor-binding protein hemagglutinin (HA) and the receptor-cleaving enzyme neuraminidase (NA). HA is responsible for host cell recognition, while NA prevents aggregation and entrapment, but the intricate mechanism of how the functions of these glycoproteins cooperate and how they are regulated by mutational responses to environmental pressures remains unclear. Recently, several groups have described the motion of influenza over surfaces and reported that this motion is inhibited by NA inhibitors. We argue that the motion of influenza resembles the motility of artificial receptor-cleaving particles called "molecular spiders". The cleaving of receptors by this type of molecular walkers leads to self-avoiding motion across a surface. When the binding and cleaving rates of molecular spiders are balanced, they move both rapidly and efficiently. The studies of molecular spiders offer new insights into the functional balance of HA and NA, but they do not address the asymmetric distribution of HA and NA on the surface of influenza. We propose that receptor-cleaving molecular walkers could play an important role in the further investigation of the motility of influenza viruses.
Collapse
Affiliation(s)
- P H Erik Hamming
- Molecular Nanofabrication Group , MESA + Institute for Nanotechnology , Faculty of Science and Technology , University of Twente , P.O. Box 217 , 7500 AE Enschede , The Netherlands .
| | - Nico J Overeem
- Molecular Nanofabrication Group , MESA + Institute for Nanotechnology , Faculty of Science and Technology , University of Twente , P.O. Box 217 , 7500 AE Enschede , The Netherlands .
| | - Jurriaan Huskens
- Molecular Nanofabrication Group , MESA + Institute for Nanotechnology , Faculty of Science and Technology , University of Twente , P.O. Box 217 , 7500 AE Enschede , The Netherlands .
| |
Collapse
|
156
|
de Vries E, Du W, Guo H, de Haan CA. Influenza A Virus Hemagglutinin-Neuraminidase-Receptor Balance: Preserving Virus Motility. Trends Microbiol 2020; 28:57-67. [PMID: 31629602 PMCID: PMC7172302 DOI: 10.1016/j.tim.2019.08.010] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/29/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022]
Abstract
Influenza A viruses (IAVs) occasionally cross the species barrier and adapt to novel host species. This requires readjustment of the functional balance of the sialic acid receptor-binding hemagglutinin (HA) and the receptor-destroying neuraminidase (NA) to the sialoglycan-receptor repertoire of the new host. Novel techniques have revealed mechanistic details of this HA-NA-receptor balance, emphasizing a previously underappreciated crucial role for NA in driving the motility of receptor-associated IAV particles. Motility enables virion penetration of the sialylated mucus layer as well as attachment to, and uptake into, underlying epithelial cells. As IAVs are essentially irreversibly bound in the absence of NA activity, the fine-tuning of the HA-NA-receptor balance rather than the binding avidity of IAV particles per se is an important factor in determining host species tropism.
Collapse
Affiliation(s)
- Erik de Vries
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands.
| | - Wenjuan Du
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Hongbo Guo
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands
| | - Cornelis A.M. de Haan
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, the Netherlands,Correspondence:
| |
Collapse
|
157
|
Is pallial mucus involved in Ostrea edulis defenses against the parasite Bonamia ostreae? J Invertebr Pathol 2019; 169:107259. [PMID: 31805287 DOI: 10.1016/j.jip.2019.107259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 10/10/2019] [Accepted: 10/12/2019] [Indexed: 02/04/2023]
Abstract
Bonamia ostreae is an intrahemocytic parasite that has been responsible for severe mortalities in the flat oyster Ostrea edulis since the 1970́s. The Pacific oyster Crassostrea gigas is considered to be resistant to the disease and appears to have mechanisms to avoid infection. Most studies carried out on the invertebrate immune system focus on the role of hemolymph, although mucus, which covers the body surface of molluscs, could also act as a barrier against pathogens. In this study, the in vitro effect of mucus from the oyster species Ostrea edulis and C. gigas on B. ostreae was investigated using flow cytometry. Results showed an increase in esterase activities and mortality rate of parasites exposed to mucus from both oyster species. In order to better understand the potential role of mucus in the defense of the oyster against parasites such as B. ostreae, liquid chromatography and tandem mass spectrometry were used to describe and compare mucus protein composition from both species. In all oyster species, pallial mucus contains a high level of proteins; however, O. edulis mucus produced a variety of proteins that could be involved in the immune response against the parasite, including Cu/Zn extracellular superoxide dismutase, thioxiredoxin, peroxiredon VI, heat shock protein 90 as well as several hydrolases. Conversely, a different set of antioxidant proteins, hydrolases and stress related proteins were identified in mucus from C. gigas. Our results suggest an innate immunity adaptation of oysters to develop a specific response against their respective pathogens. The mucosal protein composition also provides new insights for further investigations into the immune response in oysters.
Collapse
|
158
|
Barnard KN, Wasik BR, LaClair JR, Buchholz DW, Weichert WS, Alford-Lawrence BK, Aguilar HC, Parrish CR. Expression of 9- O- and 7,9- O-Acetyl Modified Sialic Acid in Cells and Their Effects on Influenza Viruses. mBio 2019; 10:e02490-19. [PMID: 31796537 PMCID: PMC6890989 DOI: 10.1128/mbio.02490-19] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022] Open
Abstract
Sialic acids (Sia) are widely displayed on the surfaces of cells and tissues. Sia come in a variety of chemically modified forms, including those with acetyl modifications at the C-7, C-8, and C-9 positions. Here, we analyzed the distribution and amounts of these acetyl modifications in different human and canine cells. Since Sia or their variant forms are receptors for influenza A, B, C, and D viruses, we examined the effects of these modifications on virus infections. We confirmed that 9-O-acetyl and 7,9-O-acetyl modified Sia are widely but variably expressed across cell lines from both humans and canines. Although they were expressed on the cell surfaces of canine MDCK cell lines, they were located primarily within the Golgi compartment of human HEK-293 and A549 cells. The O-acetyl modified Sia were expressed at low levels of 1 to 2% of total Sia in these cell lines. We knocked out and overexpressed the sialate O-acetyltransferase gene (CasD1) and knocked out the sialate O-acetylesterase gene (SIAE) using CRISPR/Cas9 editing. Knocking out CasD1 removed 7,9-O- and 9-O-acetyl Sia expression, confirming previous reports. However, overexpression of CasD1 and knockout of SIAE gave only modest increases in 9-O-acetyl levels in cells and no change in 7,9-O-acetyl levels, indicating that there are complex regulations of these modifications. These modifications were essential for influenza C and D infection but had no obvious effect on influenza A and B infection.IMPORTANCE Sialic acids are key glycans that are involved in many different normal cellular functions, as well as being receptors for many pathogens. However, Sia come in diverse chemically modified forms. Here, we examined and manipulated the expression of 7,9-O- and 9-O-acetyl modified Sia on cells commonly used in influenza virus and other research by engineering the enzymes that produce or remove the acetyl groups.
Collapse
Affiliation(s)
- Karen N Barnard
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Brian R Wasik
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Justin R LaClair
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - David W Buchholz
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Wendy S Weichert
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Brynn K Alford-Lawrence
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Hector C Aguilar
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Colin R Parrish
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
159
|
Wasik BR, Voorhees IEH, Barnard KN, Alford-Lawrence BK, Weichert WS, Hood G, Nogales A, Martínez-Sobrido L, Holmes EC, Parrish CR. Influenza Viruses in Mice: Deep Sequencing Analysis of Serial Passage and Effects of Sialic Acid Structural Variation. J Virol 2019; 93:e01039-19. [PMID: 31511393 PMCID: PMC6854484 DOI: 10.1128/jvi.01039-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/09/2019] [Indexed: 12/19/2022] Open
Abstract
Influenza A viruses have regularly jumped to new host species to cause epidemics or pandemics, an evolutionary process that involves variation in the viral traits necessary to overcome host barriers and facilitate transmission. Mice are not a natural host for influenza virus but are frequently used as models in studies of pathogenesis, often after multiple passages to achieve higher viral titers that result in clinical disease such as weight loss or death. Here, we examine the processes of influenza A virus infection and evolution in mice by comparing single nucleotide variations of a human H1N1 pandemic virus, a seasonal H3N2 virus, and an H3N2 canine influenza virus during experimental passage. We also compared replication and sequence variation in wild-type mice expressing N-glycolylneuraminic acid (Neu5Gc) with those seen in mice expressing only N-acetylneuraminic acid (Neu5Ac). Viruses derived from plasmids were propagated in MDCK cells and then passaged in mice up to four times. Full-genome deep sequencing of the plasmids, cultured viruses, and viruses from mice at various passages revealed only small numbers of mutational changes. The H3N2 canine influenza virus showed increases in frequency of sporadic mutations in the PB2, PA, and NA segments. The H1N1 pandemic virus grew well in mice, and while it exhibited the maintenance of some minority mutations, there was no clear evidence for adaptive evolution. The H3N2 seasonal virus did not establish in the mice. Finally, there were no clear sequence differences associated with the presence or absence of Neu5Gc.IMPORTANCE Mice are commonly used as a model to study the growth and virulence of influenza A viruses in mammals but are not a natural host and have distinct sialic acid receptor profiles compared to humans. Using experimental infections with different subtypes of influenza A virus derived from different hosts, we found that evolution of influenza A virus in mice did not necessarily proceed through the linear accumulation of host-adaptive mutations, that there was variation in the patterns of mutations detected in each repetition, and that the mutation dynamics depended on the virus examined. In addition, variation in the viral receptor, sialic acid, did not affect influenza virus evolution in this model. Overall, our results show that while mice provide a useful animal model for influenza virus pathology, host passage evolution will vary depending on the specific virus tested.
Collapse
Affiliation(s)
- Brian R Wasik
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Ian E H Voorhees
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Karen N Barnard
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Brynn K Alford-Lawrence
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Wendy S Weichert
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Grace Hood
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
- College of Veterinary Medicine, University of Queensland, Gatton, Queensland, Australia
| | - Aitor Nogales
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Edward C Holmes
- Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, School of Biological Sciences and Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Colin R Parrish
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
160
|
Marczynski M, Rickert CA, Semerdzhiev SA, van Dijk WR, Segers-Nolten IMJ, Claessens MMAE, Lieleg O. α-Synuclein Penetrates Mucin Hydrogels Despite Its Mucoadhesive Properties. Biomacromolecules 2019; 20:4332-4344. [DOI: 10.1021/acs.biomac.9b00905] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Matthias Marczynski
- Department of Mechanical Engineering and Munich School of Bioengineering, Technical University of Munich, Garching 85748, Germany
| | - Carolin A. Rickert
- Department of Mechanical Engineering and Munich School of Bioengineering, Technical University of Munich, Garching 85748, Germany
| | - Slav A. Semerdzhiev
- Nanobiophysics, Faculty of Science and Technology, University of Twente, Enschede 7522NB, The Netherlands
| | - Wouter R. van Dijk
- Nanobiophysics, Faculty of Science and Technology, University of Twente, Enschede 7522NB, The Netherlands
| | - Ine M. J. Segers-Nolten
- Nanobiophysics, Faculty of Science and Technology, University of Twente, Enschede 7522NB, The Netherlands
| | | | - Oliver Lieleg
- Department of Mechanical Engineering and Munich School of Bioengineering, Technical University of Munich, Garching 85748, Germany
| |
Collapse
|
161
|
Abstract
This article discusses key concepts important for mucosal immunity. The mucosa is the largest immune organ of the body. The mucosal barrier (the tight junctions and the "kill zone") along with the mucosa epithelial cells maintaining an anti-inflammatory state are essential for the mucosal firewall. The microbiome (the microorganisms that are in the gastrointestinal, respiratory, and reproductive tract) is essential for immune development, homeostasis, immune response, and maximizing animal productivity. Mucosal vaccination provides an opportunity to protect animals from most infectious diseases because oral, gastrointestinal, respiratory, and reproductive mucosa are the main portals of entry for infectious disease.
Collapse
Affiliation(s)
- Christopher Chase
- Department of Veterinary and Biomedical Sciences, South Dakota State University, PO Box 2175, SAR Room 125, North Campus Drive, Brookings, SD 57007, USA.
| | - Radhey S Kaushik
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57007, USA
| |
Collapse
|
162
|
McAllister CS, Ansaldi D, Growcott EJ, Zhong Y, Quackenbush D, Wolff KC, Chen Z, Tanaseichuk O, Lelais G, Barnes SW, Federe GC, Luna F, Walker JR, Zhou Y, Kuhen KL. Dexamethasone inhibits respiratory syncytial virus-driven mucus production while increasing viral replication without altering antiviral interferon signaling. Virology 2019; 540:195-206. [PMID: 31929001 DOI: 10.1016/j.virol.2019.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/23/2019] [Accepted: 10/18/2019] [Indexed: 01/03/2023]
Abstract
Respiratory syncytial virus (RSV) infection can cause mucus overproduction and bronchiolitis in infants leading to severe disease and hospitalization. As a therapeutic strategy, immune modulatory agents may help prevent RSV-driven immune responses that cause severe airway disease. We developed a high throughput screen to identify compounds that reduced RSV-driven mucin 5AC (Muc5AC) expression and identified dexamethasone. Despite leading to a pronounced reduction in RSV-driven Muc5AC, dexamethasone increased RSV infection in vitro and delayed viral clearance in mice. This correlated with reduced expression of a subset of immune response genes and reduced lymphocyte infiltration in vivo. Interestingly, dexamethasone increased RSV infection levels without altering antiviral interferon signaling. In summary, the immunosuppressive activities of dexamethasone had favorable inhibitory effects on RSV-driven mucus production yet prevented immune defense activities that limit RSV infection in vitro and in vivo. These findings offer an explanation for the lack of efficacy of glucocorticoids in RSV-infected patients.
Collapse
Affiliation(s)
| | - Dan Ansaldi
- Novartis Institute for BioMedical Research, Emeryville, CA, USA
| | | | - Yang Zhong
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Doug Quackenbush
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Karen C Wolff
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Zhong Chen
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Olga Tanaseichuk
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Gerald Lelais
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - S Whitney Barnes
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Glenn C Federe
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Fabio Luna
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - John R Walker
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Yingyao Zhou
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Kelli L Kuhen
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA
| |
Collapse
|
163
|
Huang S, Constant S, De Servi B, Meloni M, Culig J, Bertini M, Saaid A. In vitro safety and performance evaluation of a seawater solution enriched with copper, hyaluronic acid, and eucalyptus for nasal lavage. MEDICAL DEVICES-EVIDENCE AND RESEARCH 2019; 12:399-410. [PMID: 31576180 PMCID: PMC6766585 DOI: 10.2147/mder.s209644] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/20/2019] [Indexed: 01/02/2023] Open
Abstract
Background The common cold is a viral infectious disease with symptoms such as runny nose, sore throat, and mainly, nasal congestion. State-of-the-art therapeutic approaches focus on alleviating the symptoms of this disease by non-invasive and simple-to-use methods. Nasal irrigation is one of the most accepted approaches to ease nasal congestion which, if left untreated, has a negative impact on the quality of life of patients. Purpose In this study, the safety and efficacy of a novel hypertonic seawater solution for nasal lavage enriched with hyaluronic acids, eucalyptus oil, copper, and manganese salts (Stérimar Stop & Protect Cold and Flu; SSPCF) have been investigated in vitro. Methods An in vitro 3D reconstituted human nasal epithelium tissue model, MucilAir™, has been used in this study to investigate the safety of SSPCF on nasal epithelium by measuring transepithelial electrical resistance (TEER), lactate dehydrogenase (LDH), and interleukin-8 (IL-8) secretion. The efficacy of SSPCF was measured by mucociliary clearance (MCC), ATP release, Alcian blue and aquaporin (AQP3) stainings. Results SSPCF treatment respected nasal epithelium tissue integrity and enhanced barrier function without inducing a cytotoxic response. Secreted LDH and IL-8 levels were similar to untreated controls. MCC rate was increased 2.5-fold and ATP release decreased 87% upon SSPCF treatment, indicating improved decongestion activity. SSPCF treatment after hypotonic stress helped recover cellular organization, as shown by Alcian blue and AQP3 staining assays. Conclusion SSPCF appears as a safe and effective nasal irrigation formula that may alleviate the symptoms associated with common cold such as nasal congestion.
Collapse
Affiliation(s)
| | | | - Barbara De Servi
- Department of in Vitro Research, VitroScreen, Milan, 20149, Italy
| | - Marisa Meloni
- Department of in Vitro Research, VitroScreen, Milan, 20149, Italy
| | - Josip Culig
- Department of Pharmacology, University of Applied Health Sciences, Zagreb, 10000, Croatia
| | - Marco Bertini
- R&D Department, Laboratori Baldacci SpA, Pisa, Italy
| | - Amina Saaid
- Department of R&D and Innovation, Laboratoire Fumouze, Levallois-Perret, 92686, France
| |
Collapse
|
164
|
Costa-Hurtado M, Garcia-Rodriguez L, Lopez-Serrano S, Aragon V. Haemophilus parasuis VtaA2 is involved in adhesion to extracellular proteins. Vet Res 2019; 50:69. [PMID: 31547880 PMCID: PMC6755704 DOI: 10.1186/s13567-019-0687-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/23/2019] [Indexed: 12/11/2022] Open
Abstract
Haemophilus parasuis is part of the microbiota of the upper respiratory tract in swine. However, virulent strains can cause a systemic disease known as Glässer’s disease. Several virulence factors have been described in H. parasuis including the virulence-associated trimeric autotransporters (VtaAs). VtaA2 is up-regulated during infection and is only found in virulent strains. In order to determine its biological function, the vtaA2 gene was cloned with its native promotor region in pACYC184, and the transformed Escherichia coli was used to perform functional in vitro assays. VtaA2 was found to have a role in attachment to plastic, mucin, BSA, fibronectin and collagen. As other VtaAs from H. parasuis, the passenger domain of VtaA2 contains collagen domains. In order to examine the contribution of the collagen repeats to VtaA2 function, a recombinant vtaA2 without the central collagen domains was obtained and named vtaA2OL. VtaA2OL showed similar capacity than VtaA2 to adhere to plastic, mucin, BSA, fibronectin and plasma but a reduced capacity to adhere to collagen, suggesting that the collagen domains of VtaA2 are involved in collagen attachment. No function in cell adhesion and invasion to epithelial alveolar cell line A549 or unspecific binding to primary alveolar macrophages was found. Likewise VtaA2 had no role in serum or phagocytosis resistance. We propose that VtaA2 mediates adherence to the host by binding to the mucin, found in the upper respiratory tract mucus, and to the extracellular matrix proteins, present in the connective tissue of systemic sites, such as the serosa.
Collapse
Affiliation(s)
- Mar Costa-Hurtado
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
| | - Laura Garcia-Rodriguez
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Sergi Lopez-Serrano
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Virginia Aragon
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| |
Collapse
|
165
|
Saku A, Hirose K, Ito T, Iwata A, Sato T, Kaji H, Tamachi T, Suto A, Goto Y, Domino SE, Narimatsu H, Kiyono H, Nakajima H. Fucosyltransferase 2 induces lung epithelial fucosylation and exacerbates house dust mite-induced airway inflammation. J Allergy Clin Immunol 2019; 144:698-709.e9. [PMID: 31125592 DOI: 10.1016/j.jaci.2019.05.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/30/2019] [Accepted: 05/06/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND One of the pathognomonic features of asthma is epithelial hyperproduction of mucus, which is composed of a series of glycoproteins; however, it remains unclear how glycosylation is induced in lung epithelial cells from asthmatic patients and how glycan residues play a role in the pathogenesis of asthma. OBJECTIVE The objective of this study was to explore comprehensive epithelial glycosylation status induced by allergic inflammation and reveal its possible role in the pathogenesis of asthma. METHODS We evaluated the glycosylation status of lung epithelium using a lectin microarray. We next searched for molecular mechanisms underlying epithelial glycosylation. We also examined whether epithelial glycosylation is involved in induction of allergic inflammation. RESULTS On allergen inhalation, lung epithelial cells were heavily α(1,2)fucosylated by fucosyltransferase 2 (Fut2), which was induced by the IL-13-signal transducer and activator of transcription 6 pathway. Importantly, Fut2-deficient (Fut2-/-) mice, which lacked lung epithelial fucosylation, showed significantly attenuated eosinophilic inflammation and airway hyperresponsiveness in house dust mite (HDM)-induced asthma models. Proteome analyses and immunostaining of the HDM-challenged lung identified that complement C3 was accumulated in fucosylated areas. Indeed, Fut2-/- mice showed significantly reduced levels of C3a and impaired accumulation of C3a receptor-expressing monocyte-derived dendritic cells in the lung on HDM challenge. CONCLUSION Fut2 induces epithelial fucosylation and exacerbates airway inflammation in asthmatic patients in part through C3a production and monocyte-derived dendritic cell accumulation in the lung.
Collapse
Affiliation(s)
- Aiko Saku
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Koichi Hirose
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan; Department of Rheumatology, School of Medicine, International University of Health and Welfare, Chiba, Japan.
| | - Takashi Ito
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Arifumi Iwata
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takashi Sato
- Glycoscience and Glycotechnology Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Ibaraki, Japan
| | - Hiroyuki Kaji
- Glycoscience and Glycotechnology Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Ibaraki, Japan
| | - Tomohiro Tamachi
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akira Suto
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yoshiyuki Goto
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan; International Research and Development Center for Mucosal Vaccines, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Steven E Domino
- Department of Obstetrics and Gynecology, Cellular and Molecular Biology Program, University of Michigan Medical Center, Ann Arbor, Mich
| | - Hisashi Narimatsu
- Glycoscience and Glycotechnology Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Ibaraki, Japan
| | - Hiroshi Kiyono
- International Research and Development Center for Mucosal Vaccines, Institute of Medical Science, University of Tokyo, Tokyo, Japan; Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan; Division of Mucosal Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
166
|
Bui CHT, Chan RWY, Ng MMT, Cheung MC, Ng KC, Chan MPK, Chan LLY, Fong JHM, Nicholls JM, Peiris JSM, Chan MCW. Tropism of influenza B viruses in human respiratory tract explants and airway organoids. Eur Respir J 2019; 54:13993003.00008-2019. [PMID: 31097520 DOI: 10.1183/13993003.00008-2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/07/2019] [Indexed: 11/05/2022]
Abstract
Despite causing regular seasonal epidemics with substantial morbidity, mortality and socioeconomic burden, there is still a lack of research into influenza B viruses (IBVs). In this study, we provide for the first time a systematic investigation on the tropism, replication kinetics and pathogenesis of IBVs in the human respiratory tract.Physiologically relevant ex vivo explant cultures of human bronchus and lung, human airway organoids, and in vitro cultures of differentiated primary human bronchial epithelial cells and type-I-like alveolar epithelial cells were used to study the cellular and tissue tropism, replication competence and induced innate immune response of 16 IBV strains isolated from 1940 to 2012 in comparison with human seasonal influenza A viruses (IAVs), H1N1 and H3N2. IBVs from the diverged Yamagata- and Victoria-like lineages and the earlier undiverged period were included.The majority of IBVs replicated productively in human bronchus and lung with similar competence to seasonal IAVs. IBVs infected a variety of cell types, including ciliated cells, club cells, goblet cells and basal cells, in human airway organoids. Like seasonal IAVs, IBVs are low inducers of pro-inflammatory cytokines and chemokines. Most results suggested a higher preference for the conducting airway than the lower lung and strain-specific rather than lineage-specific pathogenicity of IBVs.Our results highlighted the non-negligible virulence of IBVs which require more attention and further investigation to alleviate the disease burden, especially when treatment options are limited.
Collapse
Affiliation(s)
- Christine H T Bui
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.,Joint first authors
| | - Renee W Y Chan
- Dept of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,Joint first authors
| | - Mandy M T Ng
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - M-C Cheung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ka-Chun Ng
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Megan P K Chan
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Louisa L Y Chan
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.,Dept of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Joanne H M Fong
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - J M Nicholls
- Dept of Pathology, Queen Mary Hospital, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - J S Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Michael C W Chan
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
167
|
Dutta D, Sailapu SK, Simon AT, Ghosh SS, Chattopadhyay A. Gold-Nanocluster-Embedded Mucin Nanoparticles for Photodynamic Therapy and Bioimaging. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:10475-10483. [PMID: 31291114 DOI: 10.1021/acs.langmuir.9b00998] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Effective delivery of a photosensitizer with the ability to trace its eventual progress forms an important aspect in photodynamic therapy (PDT). Further, the delivery mechanism might require possessing the ability to traverse through the complex mucus barrier that offers retention of therapeutic molecules. In this work, gold nanocluster (Au NC)-embedded mucin nanoparticles were synthesized by a rapid green synthetic procedure for application as nanocarriers and to achieve image-guided PDT. The mucin-based nanocarrier exhibited excellent biocompatibility toward normal cells (HEK 293T). The photosensitizer methylene blue (MB) was loaded onto these Au NC-mucin nanoparticles (NPs). HeLa cancer cells were treated with MB-loaded Au NC-mucin nanoparticles under irradiation of 640 nm light. The cell viability assay revealed that the viability of HeLa cells was reduced to 50% after treatment with MB-loaded Au NC-mucin NPs under 640 nm irradiation. The luminescence exhibited by Au NCs in the nanocarrier was applied for tracking the delivery of MB inside the HeLa cells using confocal microscopy. The flow cytometry assays elucidated the mechanism of cell death.
Collapse
|
168
|
Sathyamoorthy R, Maoz A, Pasternak Z, Im H, Huppert A, Kadouri D, Jurkevitch E. Bacterial predation under changing viscosities. Environ Microbiol 2019; 21:2997-3010. [DOI: 10.1111/1462-2920.14696] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 03/23/2019] [Accepted: 05/24/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Rajesh Sathyamoorthy
- Department of Plant Pathology and Microbiology, Faculty of Agriculture, Food and Environment The Hebrew University of Jerusalem Rehovot Israel
| | - Anat Maoz
- Bio‐statistical Unit, The Gertner Institute for Epidemiology and Health Policy Research Chaim Sheba Medical Center Tel Hashomer Israel
| | - Zohar Pasternak
- Department of Plant Pathology and Microbiology, Faculty of Agriculture, Food and Environment The Hebrew University of Jerusalem Rehovot Israel
| | - Hansol Im
- School of Life Sciences Ulsan National Institute of Science & Technology 50 UNIST‐gil Ulju‐gun, Ulsan 44919 Republic of Korea
| | - Amit Huppert
- Bio‐statistical Unit, The Gertner Institute for Epidemiology and Health Policy Research Chaim Sheba Medical Center Tel Hashomer Israel
| | - Daniel Kadouri
- Department of Oral Biology Rutgers School of Dental Medicine Newark NJ USA
| | - Edouard Jurkevitch
- Department of Plant Pathology and Microbiology, Faculty of Agriculture, Food and Environment The Hebrew University of Jerusalem Rehovot Israel
| |
Collapse
|
169
|
Butnarasu C, Barbero N, Pacheco D, Petrini P, Visentin S. Mucin binding to therapeutic molecules: The case of antimicrobial agents used in cystic fibrosis. Int J Pharm 2019; 564:136-144. [DOI: 10.1016/j.ijpharm.2019.04.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/04/2019] [Accepted: 04/10/2019] [Indexed: 11/15/2022]
|
170
|
Heinonen S, Rodriguez-Fernandez R, Diaz A, Oliva Rodriguez-Pastor S, Ramilo O, Mejias A. Infant Immune Response to Respiratory Viral Infections. Immunol Allergy Clin North Am 2019; 39:361-376. [PMID: 31284926 DOI: 10.1016/j.iac.2019.03.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Of all respiratory viruses that affect infants, respiratory syncytial virus (RSV) and rhinovirus (RV) represent the leading pathogens causing acute disease (bronchiolitis) and are associated with the development of recurrent wheezing and asthma. The immune system in infants is still developing, and several factors contribute to their increased susceptibility to viral infections. These factors include differences in pathogen detection, weaker interferon responses, lack of immunologic memory toward the invading pathogen, and T-cell responses that are balanced to promote tolerance and restrain inflammation. These aspects are reviewed here with a focus on RSV and RV infections.
Collapse
Affiliation(s)
- Santtu Heinonen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, PO Box 347, Helsinki 00029 HUS, Finland
| | - Rosa Rodriguez-Fernandez
- Department of Pediatrics, Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Hospital Materno-Infantil Gregorio Marañón, Madrid 28009, Spain; Section of General Pediatrics, Hospital Gregorio Marañón, Madrid, Spain
| | - Alejandro Diaz
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, The Ohio State Collage of Medicine, 700 Children's Drive, Columbus, OH 43205, USA; Division of Infectious Diseases, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State Collage of Medicine, 700 Children's Drive, Columbus, OH 43205, USA
| | - Silvia Oliva Rodriguez-Pastor
- Division of Pediatric Emergency Medicine and Critical Care, Hospital Regional Universitario de Malaga, Malaga 29001, Spain; Department of Pharmacology and Pediatrics, Malaga Medical Shool, Malaga University (UMA), Malaga, Spain
| | - Octavio Ramilo
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, The Ohio State Collage of Medicine, 700 Children's Drive, Columbus, OH 43205, USA; Division of Infectious Diseases, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State Collage of Medicine, 700 Children's Drive, Columbus, OH 43205, USA
| | - Asuncion Mejias
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, The Ohio State Collage of Medicine, 700 Children's Drive, Columbus, OH 43205, USA; Division of Infectious Diseases, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State Collage of Medicine, 700 Children's Drive, Columbus, OH 43205, USA; Department of Pharmacology and Pediatrics, Malaga Medical Shool, Malaga University (UMA), Malaga, Spain.
| |
Collapse
|
171
|
Vahey MD, Fletcher DA. Influenza A virus surface proteins are organized to help penetrate host mucus. eLife 2019; 8:43764. [PMID: 31084711 PMCID: PMC6516830 DOI: 10.7554/elife.43764] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 04/26/2019] [Indexed: 12/13/2022] Open
Abstract
Influenza A virus (IAV) enters cells by binding to sialic acid on the cell surface. To accomplish this while avoiding immobilization by sialic acid in host mucus, viruses rely on a balance between the receptor-binding protein hemagglutinin (HA) and the receptor-cleaving protein neuraminidase (NA). Although genetic aspects of this balance are well-characterized, little is known about how the spatial organization of these proteins in the viral envelope may contribute. Using site-specific fluorescent labeling and super-resolution microscopy, we show that HA and NA are asymmetrically distributed on the surface of filamentous viruses, creating a spatial organization of binding and cleaving activities that causes viruses to step consistently away from their NA-rich pole. This Brownian ratchet-like diffusion produces persistent directional mobility that resolves the virus’s conflicting needs to both penetrate mucus and stably attach to the underlying cells, potentially contributing to the prevalence of the filamentous phenotype in clinical isolates of IAV.
Collapse
Affiliation(s)
- Michael D Vahey
- Department of Bioengineering, University of California, Berkeley, Berkeley, United States.,Biophysics Program, University of California, Berkeley, Berkeley, United States
| | - Daniel A Fletcher
- Department of Bioengineering, University of California, Berkeley, Berkeley, United States.,Biological Systems & Engineering, Lawrence Berkeley National Laboratory, Berkeley, United States.,Chan Zuckerberg Biohub, San Francisco, United States
| |
Collapse
|
172
|
Tropism and Infectivity of a Seasonal A(H1N1) and a Highly Pathogenic Avian A(H5N1) Influenza Virus in Primary Differentiated Ferret Nasal Epithelial Cell Cultures. J Virol 2019; 93:JVI.00080-19. [PMID: 30814288 DOI: 10.1128/jvi.00080-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 02/21/2019] [Indexed: 12/25/2022] Open
Abstract
Ferrets represent an invaluable animal model to study influenza virus pathogenesis and transmission. To further characterize this model, we developed a differentiated primary ferret nasal epithelial cell (FNEC) culture model for investigation of influenza A virus infection and virus-host interactions. This well-differentiated culture consists of various cell types, a mucociliary clearance system, and tight junctions, representing the nasal ciliated pseudostratified respiratory epithelium. Both α2,6-linked and α2,3-linked sialic acid (SA) receptors, which preferentially bind the hemagglutinin (HA) of human and avian influenza viruses, respectively, were detected on the apical surface of the culture with different cellular tropisms. In accordance with the distribution of SA receptors, we observed that a pre-2009 seasonal A(H1N1) virus infected both ciliated and nonciliated cells, whereas a highly pathogenic avian influenza (HPAI) A(H5N1) virus primarily infected nonciliated cells. Transmission electron microscopy revealed that virions were released from or associated with the apical membranes of ciliated, nonciliated, and mucin-secretory goblet cells. Upon infection, the HPAI A(H5N1) virus replicated to titers higher than those of the human A(H1N1) virus at 37°C; however, replication of the A(H5N1) virus was significantly attenuated at 33°C. Furthermore, we found that infection with the A(H5N1) virus induced higher expression levels of immune mediator genes and resulted in more cell damage/loss than with the human A(H1N1) virus. This primary differentiated FNEC culture model, recapitulating the structure of the nasal epithelium, provides a useful model to bridge in vivo and in vitro studies of cellular tropism, infectivity, and pathogenesis of influenza viruses during the initial stages of infection.IMPORTANCE Although ferrets serve as an important model of influenza virus infection, much remains unknown about virus-host interactions in this species at the cellular level. The development of differentiated primary cultures of ferret nasal epithelial cells is an important step toward understanding cellular tropism and the mechanisms of influenza virus infection and replication in the airway milieu of this model. Using lectin staining and microscopy techniques, we characterized the sialic acid receptor distribution and the cellular composition of the culture model. We then evaluated the replication of and immune response to human and avian influenza viruses at relevant physiological temperatures. Our findings offer significant insight into this first line of defense against influenza virus infection and provide a model for the evaluation of emerging influenza viruses in a well-controlled in vitro environmental setting.
Collapse
|
173
|
Hong S, Shi Y, Wu NC, Grande G, Douthit L, Wang H, Zhou W, Sharpless KB, Wilson IA, Xie J, Wu P. Bacterial glycosyltransferase-mediated cell-surface chemoenzymatic glycan modification. Nat Commun 2019; 10:1799. [PMID: 30996301 PMCID: PMC6470217 DOI: 10.1038/s41467-019-09608-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 02/26/2019] [Indexed: 12/14/2022] Open
Abstract
Chemoenzymatic modification of cell-surface glycan structures has emerged as a complementary approach to metabolic oligosaccharide engineering. Here, we identify Pasteurella multocida α2-3-sialyltransferase M144D mutant, Photobacterium damsela α2-6-sialyltransferase, and Helicobacter mustelae α1-2-fucosyltransferase, as efficient tools for live-cell glycan modification. Combining these enzymes with Helicobacter pylori α1-3-fucosyltransferase, we develop a host-cell-based assay to probe glycan-mediated influenza A virus (IAV) infection including wild-type and mutant strains of H1N1 and H3N2 subtypes. At high NeuAcα2-6-Gal levels, the IAV-induced host-cell death is positively correlated with haemagglutinin (HA) binding affinity to NeuAcα2-6-Gal. Remarkably, an increment of host-cell-surface sialyl Lewis X (sLeX) exacerbates the killing by several wild-type IAV strains and a previously engineered mutant HK68-MTA. Structural alignment of HAs from HK68 and HK68-MTA suggests formation of a putative hydrogen bond between Trp222 of HA-HK68-MTA and the C-4 hydroxyl group of the α1-3-linked fucose of sLeX, which may account for the enhanced host cell killing of that mutant.
Collapse
Affiliation(s)
- Senlian Hong
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Yujie Shi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Nicholas C Wu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Geramie Grande
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Lacey Douthit
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Hua Wang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Wen Zhou
- College of Chemistry and Molecular Engineering, Peking University, 100871, Beijing, China
| | - K Barry Sharpless
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jia Xie
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| | - Peng Wu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
174
|
Spolski R, West EE, Li P, Veenbergen S, Yung S, Kazemian M, Oh J, Yu ZX, Freeman AF, Holland SM, Murphy PM, Leonard WJ. IL-21/type I interferon interplay regulates neutrophil-dependent innate immune responses to Staphylococcus aureus. eLife 2019; 8:45501. [PMID: 30969166 PMCID: PMC6504231 DOI: 10.7554/elife.45501] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/09/2019] [Indexed: 12/24/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a major hospital- and community-acquired pathogen, but the mechanisms underlying host-defense to MRSA remain poorly understood. Here, we investigated the role of IL-21 in this process. When administered intra-tracheally into wild-type mice, IL-21 induced granzymes and augmented clearance of pulmonary MRSA but not when neutrophils were depleted or a granzyme B inhibitor was added. Correspondingly, IL-21 induced MRSA killing by human peripheral blood neutrophils. Unexpectedly, however, basal MRSA clearance was also enhanced when IL-21 signaling was blocked, both in Il21r KO mice and in wild-type mice injected with IL-21R-Fc fusion-protein. This correlated with increased type I interferon and an IFN-related gene signature, and indeed anti-IFNAR1 treatment diminished MRSA clearance in these animals. Moreover, we found that IFNβ induced granzyme B and promoted MRSA clearance in a granzyme B-dependent fashion. These results reveal an interplay between IL-21 and type I IFN in the innate immune response to MRSA.
Collapse
Affiliation(s)
- Rosanne Spolski
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States.,Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Erin E West
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States.,Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Peng Li
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States.,Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Sharon Veenbergen
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Sunny Yung
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States
| | - Majid Kazemian
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States.,Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Jangsuk Oh
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States.,Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Zu-Xi Yu
- The Pathology Core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States
| | - Stephen M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States
| | - Philip M Murphy
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States
| | - Warren J Leonard
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States.,Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| |
Collapse
|
175
|
Paz C, Suárez E, Vence J, Cabarcos A. Analysis of the volume of fluid (VOF) method for the simulation of the mucus clearance process with CFD. Comput Methods Biomech Biomed Engin 2019; 22:547-566. [PMID: 30773045 DOI: 10.1080/10255842.2019.1569637] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The clearance of mucus through coughing is a complex, multiphase process, which is affected principally by mucus viscosity and airflow velocity; however, it is also critically affected by the thickness of the two layers of mucus-the serous and gel layers-and oscillation level. The present study examines the effects of the latter parameters more closely. To do so, the mucus clearance process is simulated with a transient 3D volume of fluid (VOF) multiphase model in ANSYS Fluent. The model includes mucus' bilayer properties and a wide range of boundary conditions were tested. The model was analysed in both a straight tube and a realistic trachea. Ultimately, the model was able to both capture air-mucus interface wave evolution and predict the overall behaviour of the clearance process. The results were consistent with experimental clearance data and numerical airflow simulations, which indicates our methodology is appropriate for future studies. Ultimately, the mere presence of the serous layer was found to increase mucus clearance by more than 15 percent. An oscillating flow enhanced clearance by up to 5 percent. Interestingly, interface wave steepness was found to be inversely correlated with mucus thickness, but directly with mucus velocity, which suggests it will be an interesting parameter for further study.
Collapse
Affiliation(s)
- Concepción Paz
- a School of Industrial Engineering , University of Vigo , Vigo , Spain.,b Biofluids Research Group, Galicia Sur Heath Research Institute (IIS Galicia Sur) , SERGAS-UVIGO , Vigo , Spain
| | - Eduardo Suárez
- a School of Industrial Engineering , University of Vigo , Vigo , Spain.,b Biofluids Research Group, Galicia Sur Heath Research Institute (IIS Galicia Sur) , SERGAS-UVIGO , Vigo , Spain
| | - Jesús Vence
- a School of Industrial Engineering , University of Vigo , Vigo , Spain
| | - Adrián Cabarcos
- a School of Industrial Engineering , University of Vigo , Vigo , Spain
| |
Collapse
|
176
|
Nieto A, Vasilijevic J, Santos NB, Zamarreño N, López P, Amorim MJ, Falcon A. Mutation S110L of H1N1 Influenza Virus Hemagglutinin: A Potent Determinant of Attenuation in the Mouse Model. Front Immunol 2019; 10:132. [PMID: 30787926 PMCID: PMC6372558 DOI: 10.3389/fimmu.2019.00132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 01/16/2019] [Indexed: 01/27/2023] Open
Abstract
Characterization of a pandemic 2009 H1N1 influenza virus isolated from a fatal case patient (F-IAV), showed the presence of three different mutations; potential determinants of its high pathogenicity that were located in the polymerase subunits (PB2 A221T and PA D529N) and the hemagglutinin (HA S110L). Recombinant viruses containing individually or in combination the polymerase mutations in the backbone of A/California/04/09 (CAL) showed that PA D529N was clearly involved in the increased pathogenicity of the F-IAV virus. Here, we have evaluated the contribution of HA S110L to F-IAV pathogenicity, through introduction of this point mutation in CAL recombinant virus (HA mut). The HA S110L protein has similar pH stability, comparable mobility, and entry properties both in human and mouse cultured cells that wild type HA. The change HA S110L leads to a non-significant trend to reduce the replication capacity of influenza virus in tissue culture, and HA mut is better neutralized than CAL virus by monoclonal and polyclonal antibodies against HA from CAL strain. In addition, recombinant viruses containing HA S110L alone or in combination with polymerase mutations considerably increased the LD50 in infected mice. Characterization of the lungs of HA mut infected animals showed reduced lung damage and inflammation compared with CAL infected mice. Accordingly, lower virus replication, decreased presence in bronchioli and parenchyma and lower leukocytes and epithelial infected cells were found in the lungs of HA mut-infected animals. Our results indicate that, mutation HA S110L constitutes a determinant of attenuation and suggest that its interaction with components of the respiratory tract mucus and lectins, that play an important role on influenza virus outcome, may constitute a physical barrier impeding the infection of the target cells, thus compromising the infection outcome.
Collapse
Affiliation(s)
- Amelia Nieto
- National Center for Biotechnology (CNB-CSIC), Madrid, Spain.,Center for Biomedical Research (CIBER), Madrid, Spain
| | | | - Nuno Brito Santos
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Noelia Zamarreño
- National Center for Biotechnology (CNB-CSIC), Madrid, Spain.,Center for Biomedical Research (CIBER), Madrid, Spain
| | - Pablo López
- National Center for Biotechnology (CNB-CSIC), Madrid, Spain
| | - Maria Joao Amorim
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Ana Falcon
- National Center for Biotechnology (CNB-CSIC), Madrid, Spain.,Center for Biomedical Research (CIBER), Madrid, Spain
| |
Collapse
|
177
|
Jang SJ, Park MH, Lee TK, Choi SH. Healing Effect of Platelet-rich Plasma on Decellularized Tracheal Allotransplantation in Rabbits. In Vivo 2018; 32:1443-1447. [PMID: 30348699 DOI: 10.21873/invivo.11397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/19/2018] [Accepted: 09/21/2018] [Indexed: 01/30/2023]
Abstract
BACKGROUND/AIM The purpose of this study was to explore the effect of platelet-rich plasma (PRP) on enhancing healing of trachea allotransplantation and confirm the effect via parallel histological and tracheoscopic examinations in seven adult New Zealand White rabbits. MATERIALS AND METHODS Harvested trachea was inserted into recipients with end-to-end anastomosis by a simple interrupted suture. PRP-treated rabbits were treated with 0.5 ml of PRP at the trachea grafts, while control rabbit allografts were treated with 0.5 ml of saline. RESULTS Tracheoscopy of tracheal allografts treated with PRP revealed that the trachea was well healed with no stenosis. The healing effect in the PRP-treated rabbits increased tracheal activity and produced faster trachea regeneration compared to that in control rabbits. There was a good correlation between the subjective symptom of noisy breathing and the objective grading of tracheal stenosis. The tracheal allografts with suture materials appeared slightly pale and looked more like mucosa erosion than normal mucosa at four weeks post-surgery. Contact of trachea-to-transplanted grafts in PRP-treated rabbits was intimate with the surface of the transplanted region and showed high-density epithelialization. After 8 weeks, blood vessels were observed in the transplanted graft in PRP-treated rabbits. Normal epithelium was present in grafts at 8 weeks after allotransplantation. No CD20+ cells were detected in grafts but a few CD3+ cells were observed under the epithelium. CONCLUSION The results of this study show that it is possible to perform tracheal reconstruction in rabbits treated with PRP after tracheal transplantation.
Collapse
Affiliation(s)
- Seok Jin Jang
- Department of Veterinary Surgery, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Min-Ho Park
- Department of Veterinary Surgery, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Tae-Ki Lee
- Department of Veterinary Surgery, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Seok Hwa Choi
- Department of Veterinary Surgery, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
178
|
González-Parra G, Dobrovolny HM. A quantitative assessment of dynamical differences of RSV infections in vitro and in vivo. Virology 2018; 523:129-139. [PMID: 30144786 DOI: 10.1016/j.virol.2018.07.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 07/24/2018] [Accepted: 07/25/2018] [Indexed: 10/28/2022]
Abstract
Experimental results in vitro and in animal models are used to guide researchers in testing vaccines or treatment in humans. However, viral kinetics are different in vitro, in animals, and in humans, so it is sometimes difficult to translate results from one system to another. In this study, we use a mathematical model to fit experimental data from multiple cycle respiratory syncytial virus (RSV) infections in vitro, in african green monkey (AGM), and in humans in order to quantitatively compare viral kinetics in the different systems. We find that there are differences in viral clearance rate, productively infectious cell lifespan, and eclipse phase duration between in vitro and in vivo systems and among different in vivo systems. We show that these differences in viral kinetics lead to different estimates of drug effectiveness of fusion inhibitors in vitro and in AGM than in humans.
Collapse
Affiliation(s)
| | - Hana M Dobrovolny
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, United States.
| |
Collapse
|
179
|
Lee CY, An SH, Choi JG, Lee YJ, Kim JH, Kwon HJ. Acquisition of Innate Inhibitor Resistance and Mammalian Pathogenicity During Egg Adaptation by the H9N2 Avian Influenza Virus. Front Microbiol 2018; 9:1939. [PMID: 30186261 PMCID: PMC6110911 DOI: 10.3389/fmicb.2018.01939] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/31/2018] [Indexed: 01/04/2023] Open
Abstract
An H9N2 avian influenza A virus (AIV), A/chicken/Korea/01310/2001 (01310-CE20), was established after 20 passages of influenza A/chicken/Korea/01310/2001 (01310-CE2) virus through embryonated chicken eggs (ECEs). As a result of this process, the virus developed highly replicative and pathogenic traits within the ECEs through adaptive mutations in hemagglutinin (HA: T133N, V216G, and E439D) and neuraminidase (NA: 18-amino acid deletion and E54D). Here, we also established that 01310-CE20 acquired resistance to innate inhibitors present in the egg white during these passages. To investigate the role of egg-adapted mutations in resistance to innate inhibitors, we generated four PR8-derived recombinant viruses using various gene combinations of HA and NA from 01310-CE2 and 01310-CE20 (rH2N2, rH2N20, rH20N2, and rH20N20). As expected, rH20N20 showed significantly higher replication efficiency in MDCK cells and mouse lungs, and demonstrated greater pathogenicity in mice. In addition, rH20N20 showed higher resistance to innate inhibitors than the other viruses. By using a loss-of-function mutant and receptor-binding assay, we demonstrated that a T133N site directed mutation created an additional N-glycosite at position 133 in rH20N20. Further, this mutation played a crucial role in viral replication and resistance to innate inhibitors by modulating the binding affinities to avian-like and mammalian-like receptors on the host cells and inhibitors. Thus, egg-adapted HA and NA may exacerbate the mammalian pathogenicity of AIVs by defying host innate inhibitors as well as by increasing replication efficiency in mammalian cells.
Collapse
Affiliation(s)
- Chung-Young Lee
- Laboratory of Avian Diseases, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Se-Hee An
- Laboratory of Avian Diseases, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Jun-Gu Choi
- Avian Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, South Korea
| | - Youn-Jeong Lee
- Avian Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, South Korea
| | - Jae-Hong Kim
- Laboratory of Avian Diseases, College of Veterinary Medicine, Seoul National University, Seoul, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyuk-Joon Kwon
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea.,Department of Farm Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, South Korea.,Farm Animal Clinical Training and Research Center, Institutes of Green-bio Science & Technology, Seoul National University, Gangwon-do, South Korea
| |
Collapse
|
180
|
Staphylococcus aureus Biofilm Growth on Cystic Fibrosis Airway Epithelial Cells Is Enhanced during Respiratory Syncytial Virus Coinfection. mSphere 2018; 3:3/4/e00341-18. [PMID: 30111629 PMCID: PMC6094059 DOI: 10.1128/msphere.00341-18] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The airways of individuals with cystic fibrosis (CF) are commonly chronically infected, and Staphylococcus aureus is the dominant bacterial respiratory pathogen in CF children. CF patients also experience frequent respiratory virus infections, and it has been hypothesized that virus coinfection increases the severity of S. aureus lung infections in CF. We investigated the relationship between S. aureus and the CF airway epithelium and observed that coinfection with respiratory syncytial virus (RSV) enhances S. aureus biofilm growth. However, iron, which was previously found to be a significant factor influencing Pseudomonas aeruginosa biofilms during virus coinfection, plays a minor role in S. aureus coinfections. Transcriptomic analyses provided new insight into how bacterial and viral pathogens alter host defense and suggest potential pathways by which dampening of host responses to one pathogen may favor persistence of another in the CF airways, highlighting complex interactions occurring between bacteria, viruses, and the host during polymicrobial infections. Staphylococcus aureus is a major cause of chronic respiratory infection in patients with cystic fibrosis (CF). We recently showed that Pseudomonas aeruginosa exhibits enhanced biofilm formation during respiratory syncytial virus (RSV) coinfection on human CF airway epithelial cells (AECs). The impact of respiratory viruses on other bacterial pathogens during polymicrobial infections in CF remains largely unknown. To investigate if S. aureus biofilm growth in the CF airways is impacted by virus coinfection, we evaluated S. aureus growth on CF AECs. Initial studies showed an increase in S. aureus growth over 24 h, and microscopy revealed biofilm-like clusters of bacteria on CF AECs. Biofilm growth was enhanced when CF AECs were coinfected with RSV, and this observation was confirmed with S. aureus CF clinical isolates. Apical conditioned medium from RSV-infected cells promoted S. aureus biofilms in the absence of the host epithelium, suggesting that a secreted factor produced during virus infection benefits S. aureus biofilms. Exogenous iron addition did not significantly alter biofilm formation, suggesting that it is not likely the secreted factor. We further characterized S. aureus-RSV coinfection in our model using dual host-pathogen RNA sequencing, allowing us to observe specific contributions of S. aureus and RSV to the host response during coinfection. Using the dual host-pathogen RNA sequencing approach, we observed increased availability of nutrients from the host and upregulation of S. aureus genes involved in growth, protein translation and export, and amino acid metabolism during RSV coinfection. IMPORTANCE The airways of individuals with cystic fibrosis (CF) are commonly chronically infected, and Staphylococcus aureus is the dominant bacterial respiratory pathogen in CF children. CF patients also experience frequent respiratory virus infections, and it has been hypothesized that virus coinfection increases the severity of S. aureus lung infections in CF. We investigated the relationship between S. aureus and the CF airway epithelium and observed that coinfection with respiratory syncytial virus (RSV) enhances S. aureus biofilm growth. However, iron, which was previously found to be a significant factor influencing Pseudomonas aeruginosa biofilms during virus coinfection, plays a minor role in S. aureus coinfections. Transcriptomic analyses provided new insight into how bacterial and viral pathogens alter host defense and suggest potential pathways by which dampening of host responses to one pathogen may favor persistence of another in the CF airways, highlighting complex interactions occurring between bacteria, viruses, and the host during polymicrobial infections.
Collapse
|
181
|
Huong TN, Yan Y, Jumat MR, Lui J, Tan BH, Wang DY, Sugrue RJ. A sustained antiviral host response in respiratory syncytial virus infected human nasal epithelium does not prevent progeny virus production. Virology 2018; 521:20-32. [PMID: 29870884 DOI: 10.1016/j.virol.2018.05.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/09/2018] [Accepted: 05/18/2018] [Indexed: 12/01/2022]
Abstract
Respiratory syncytial virus infection was examined using a human nasal epithelial cell model. Maximum levels of shed-virus were produced at between 3 and 5 days post-infection (dpi), and the infectivity of the shed-virus was stable up to 10 dpi. The highest levels of interferon signalling were recorded at 2dpi, and infection induced a widespread antivirus response in the nasal epithelium, involving both infected cells and non-infected cells. Although these cellular responses were associated with reduced levels of progeny virus production and restricted virus spread, they did not inhibit the infectivity virus that is shed early in infection. In the clinical context these data suggest that although the host cell response in the nasal epithelium may restrict the levels of progeny virus particles produced, the stability of the shed-virus in the nasal mucosa may be an important factor in both disease progression and virus transmission.
Collapse
Affiliation(s)
- Tra Nguyen Huong
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Yan Yan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore 119228, Republic of Singapore
| | - Muhammad Raihan Jumat
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Jing Lui
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore 119228, Republic of Singapore
| | - Boon Huan Tan
- Detection and Diagnostics Laboratory, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Republic of Singapore
| | - De Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore 119228, Republic of Singapore
| | - Richard J Sugrue
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Republic of Singapore.
| |
Collapse
|
182
|
Lee LYY, Izzard L, Hurt AC. A Review of DNA Vaccines Against Influenza. Front Immunol 2018; 9:1568. [PMID: 30038621 PMCID: PMC6046547 DOI: 10.3389/fimmu.2018.01568] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/25/2018] [Indexed: 01/07/2023] Open
Abstract
The challenges of effective vaccination against influenza are gaining more mainstream attention, as recent influenza seasons have reported low efficacy in annual vaccination programs worldwide. Combined with the potential emergence of novel influenza viruses resulting in a pandemic, the need for effective alternatives to egg-produced conventional vaccines has been made increasingly clear. DNA vaccines against influenza have been in development since the 1990s, but the initial excitement over success in murine model trials has been tempered by comparatively poor performance in larger animal models. In the intervening years, much progress has been made to refine the DNA vaccine platform-the rational design of antigens and expression vectors, the development of novel vaccine adjuvants, and the employment of innovative gene delivery methods. This review discusses how these advances have been applied in recent efforts to develop an effective influenza DNA vaccine.
Collapse
|
183
|
Bakshani CR, Morales-Garcia AL, Althaus M, Wilcox MD, Pearson JP, Bythell JC, Burgess JG. Evolutionary conservation of the antimicrobial function of mucus: a first defence against infection. NPJ Biofilms Microbiomes 2018; 4:14. [PMID: 30002868 PMCID: PMC6031612 DOI: 10.1038/s41522-018-0057-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/05/2018] [Accepted: 06/06/2018] [Indexed: 02/07/2023] Open
Abstract
Mucus layers often provide a unique and multi-functional hydrogel interface between the epithelial cells of organisms and their external environment. Mucus has exceptional properties including elasticity, changeable rheology and an ability to self-repair by re-annealing, and is therefore an ideal medium for trapping and immobilising pathogens and serving as a barrier to microbial infection. The ability to produce a functional surface mucosa was an important evolutionary step, which evolved first in the Cnidaria, which includes corals, and the Ctenophora. This allowed the exclusion of non-commensal microbes and the subsequent development of the mucus-lined digestive cavity seen in higher metazoans. The fundamental architecture of the constituent glycoprotein mucins is also evolutionarily conserved. Although an understanding of the biochemical interactions between bacteria and the mucus layer are important to the goal of developing new antimicrobial strategies, they remain relatively poorly understood. This review summarises the physicochemical properties and evolutionary importance of mucus, which make it so successful in the prevention of bacterial infection. In addition, the strategies developed by bacteria to counteract the mucus layer are also explored.
Collapse
Affiliation(s)
- Cassie R Bakshani
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Ana L Morales-Garcia
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mike Althaus
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Matthew D Wilcox
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Jeffrey P Pearson
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - John C Bythell
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - J Grant Burgess
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
184
|
Borges LGDA, Giongo A, Pereira LDM, Trindade FJ, Gregianini TS, Campos FS, Ghedin E, da Veiga ABG. Comparison of the nasopharynx microbiome between influenza and non-influenza cases of severe acute respiratory infections: A pilot study. Health Sci Rep 2018; 1:e47. [PMID: 30623080 PMCID: PMC6266421 DOI: 10.1002/hsr2.47] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 03/21/2018] [Accepted: 04/06/2018] [Indexed: 12/23/2022] Open
Abstract
AIMS Influenza A virus (IAV) can cause severe acute respiratory infection (SARI), and disease outcome may be associated with changes in the microbiome of the nasopharynx. This is a pilot study to characterize the microbiome of the nasopharynx in patients hospitalized with SARI, infected and not infected by IAV. METHODS AND RESULTS Using target sequencing of the 16S rRNA gene, we assessed the bacterial community of nasopharyngeal aspirate samples and compared the microbiome of patients infected with IAV with the microbiome of patients who were negative for IAV. We observed differences in the relative abundance of Proteobacteria and Firmicutes between SARI patients, with Streptococcus being enriched and Pseudomonas underrepresented in IAV patients compared with patients who were not infected with IAV. CONCLUSION Pseudomonas taxon seems to be in high frequency on the nasopharynx of SARI patients with non-IAV infection and might present a negative association with Streptococcus taxon. Microbial profile appears to be different between SARI patients infected or not infected with IAV.
Collapse
Affiliation(s)
- Luiz Gustavo dos Anjos Borges
- Laboratório de Biologia Molecular, Programa de Pós‐Graduação em PatologiaUniversidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA)Porto AlegreRSBrazil
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Adriana Giongo
- Instituto do Petróleo e dos Recursos Naturais (IPR)Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS)Porto AlegreRSBrazil
| | - Leandro de Mattos Pereira
- Faculdade de BiociênciasPontifícia Universidade Católica do Rio Grande do Sul (PUCRS)Porto AlegreRSBrazil
| | - Fernanda J. Trindade
- Faculdade de BiociênciasPontifícia Universidade Católica do Rio Grande do Sul (PUCRS)Porto AlegreRSBrazil
| | - Tatiana Schäffer Gregianini
- Laboratório Central de Saúde Pública da Secretaria de Saúde do Estado do Rio Grande do Sul (LACEN/SES‐RS)Porto AlegreRSBrazil
| | - Fabrício Souza Campos
- College of Veterinary Medicine and AgronomyUniversity of Brasília, Darcy Ribeiro University Campus, ICCAsa Norte, CEP 70.910-970 BrasíliaDFBrazil
| | - Elodie Ghedin
- Center for Genomics and Systems Biology, Department of BiologyNew York UniversityNew YorkNYUSA
- Department of Epidemiology, College of Global Public HealthNew York UniversityNew YorkNYUSA
| | - Ana Beatriz Gorini da Veiga
- Laboratório de Biologia Molecular, Programa de Pós‐Graduação em PatologiaUniversidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA)Porto AlegreRSBrazil
| |
Collapse
|
185
|
Hobbs JK, Pluvinage B, Boraston AB. Glycan-metabolizing enzymes in microbe-host interactions: the Streptococcus pneumoniae paradigm. FEBS Lett 2018; 592:3865-3897. [PMID: 29608212 DOI: 10.1002/1873-3468.13045] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 12/31/2022]
Abstract
Streptococcus pneumoniae is a frequent colonizer of the upper airways; however, it is also an accomplished pathogen capable of causing life-threatening diseases. To colonize and cause invasive disease, this bacterium relies on a complex array of factors to mediate the host-bacterium interaction. The respiratory tract is rich in functionally important glycoconjugates that display a vast range of glycans, and, thus, a key component of the pneumococcus-host interaction involves an arsenal of bacterial carbohydrate-active enzymes to depolymerize these glycans and carbohydrate transporters to import the products. Through the destruction of host glycans, the glycan-specific metabolic machinery deployed by S. pneumoniae plays a variety of roles in the host-pathogen interaction. Here, we review the processing and metabolism of the major host-derived glycans, including N- and O-linked glycans, Lewis and blood group antigens, proteoglycans, and glycogen, as well as some dietary glycans. We discuss the role of these metabolic pathways in the S. pneumoniae-host interaction, speculate on the potential of key enzymes within these pathways as therapeutic targets, and relate S. pneumoniae as a model system to glycan processing in other microbial pathogens.
Collapse
Affiliation(s)
- Joanne K Hobbs
- Department of Biochemistry and Microbiology, University of Victoria, British Columbia, Canada
| | - Benjamin Pluvinage
- Department of Biochemistry and Microbiology, University of Victoria, British Columbia, Canada
| | - Alisdair B Boraston
- Department of Biochemistry and Microbiology, University of Victoria, British Columbia, Canada
| |
Collapse
|
186
|
Infection and Replication of Influenza Virus at the Ocular Surface. J Virol 2018; 92:JVI.02192-17. [PMID: 29321303 DOI: 10.1128/jvi.02192-17] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 12/28/2017] [Indexed: 01/02/2023] Open
Abstract
Although influenza viruses typically cause respiratory tract disease, some viruses, particularly those with an H7 hemagglutinin, have been isolated from the eyes of conjunctivitis cases. Previous work has shown that isolates of multiple subtypes from both ocular and respiratory infections are capable of replication in human ex vivo ocular tissues and corneal or conjunctival cell monolayers, leaving the determinants of ocular tropism unclear. Here, we evaluated the effect of several variables on tropism for ocular cells cultured in vitro and examined the potential effect of the tear film on viral infectivity. All viruses tested were able to replicate in primary human corneal epithelial cell monolayers subjected to aerosol inoculation. The temperature at which cells were cultured postinoculation minimally affected infectivity. Replication efficiency, in contrast, was reduced at 33°C relative to that at 37°C, and this effect was slightly greater for the conjunctivitis isolates than for the respiratory ones. With the exception of a seasonal H3N2 virus, the subset of viruses studied in multilayer corneal tissue constructs also replicated productively after either aerosol or liquid inoculation. Human tears significantly inhibited the hemagglutination of both ocular and nonocular isolates, but the effect on viral infectivity was more variable, with tears reducing the infectivity of nonocular isolates more than ocular isolates. These data suggest that most influenza viruses may be capable of establishing infection if they reach the surface of ocular cells but that this is more likely for ocular-tropic viruses, as they are better able to maintain their infectivity during passage through the tear film.IMPORTANCE The potential spread of zoonotic influenza viruses to humans represents an important threat to public health. Unfortunately, despite the importance of cellular and tissue tropism to pathogenesis, determinants of influenza virus tropism have yet to be fully elucidated. Here, we sought to identify factors that limit the ability of most influenza viruses to cause ocular infection. Although ocular symptoms in humans caused by avian influenza viruses tend to be relatively mild, these infections are concerning due to the potential of the ocular surface to serve as a portal of entry for viruses that go on to establish respiratory infections. Furthermore, a better understanding of the factors that influence infection and replication in this noncanonical site may point toward novel determinants of tropism in the respiratory tract.
Collapse
|
187
|
Colombo C, Podlipnik Č, Lo Presti L, Niikura M, Bennet AJ, Bernardi A. Design and synthesis of constrained bicyclic molecules as candidate inhibitors of influenza A neuraminidase. PLoS One 2018; 13:e0193623. [PMID: 29489903 PMCID: PMC5831633 DOI: 10.1371/journal.pone.0193623] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/14/2018] [Indexed: 11/19/2022] Open
Abstract
The rise of drug-resistant influenza A virus strains motivates the development of new antiviral drugs, with different structural motifs and substitution. Recently, we explored the use of a bicyclic (bicyclo[3.1.0]hexane) analogue of sialic acid that was designed to mimic the conformation adopted during enzymatic cleavage within the neuraminidase (NA; sialidase) active site. Given that our first series of compounds were at least four orders of magnitude less active than available drugs, we hypothesized that the new carbon skeleton did not elicit the same interactions as the cyclohexene frameworks used previously. Herein, we tried to address this critical point with the aid of molecular modeling and we proposed new structures with different functionalization, such as the introduction of free ammonium and guanidinium groups and ether side chains other than the 3-pentyl side chain, the characteristic side chain in Oseltamivir. A highly simplified synthetic route was developed, starting from the cyclopropanation of cyclopentenone and followed by an aziridination and further functionalization of the five-member ring. This allowed the efficient preparation of a small library of new bicyclic ligands that were characterized by enzyme inhibition assays against influenza A neuraminidases N1, its H274Y mutant, and N2. The results show that none of the new structural variants synthesized, including those containing guanidinium groups rather than free ammonium ions, displayed activity against influenza A neuraminidases at concentrations less than 2 mM. We conclude that the choice and positioning of functional groups on the bicyclo[3.1.0]hexyl system still need to be properly tuned for producing complementary interactions within the catalytic site.
Collapse
Affiliation(s)
- Cinzia Colombo
- Università degli Studi di Milano, Dipartimento di Chimica, Milano, Italy
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- * E-mail:
| | - Črtomir Podlipnik
- University of Ljubljana, Faculty of Chemistry and Chemical Technology, Ljubljana, Slovenia
| | - Leonardo Lo Presti
- Università degli Studi di Milano, Dipartimento di Chimica, Milano, Italy
| | - Masahiro Niikura
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Andrew J. Bennet
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Anna Bernardi
- Università degli Studi di Milano, Dipartimento di Chimica, Milano, Italy
| |
Collapse
|
188
|
García-Díaz M, Birch D, Wan F, Nielsen HM. The role of mucus as an invisible cloak to transepithelial drug delivery by nanoparticles. Adv Drug Deliv Rev 2018; 124:107-124. [PMID: 29117511 DOI: 10.1016/j.addr.2017.11.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/03/2017] [Accepted: 11/01/2017] [Indexed: 01/05/2023]
Abstract
Mucosal administration of drugs and drug delivery systems has gained increasing interest. However, nanoparticles intended to protect and deliver drugs to epithelial surfaces require transport through the surface-lining mucus. Translation from bench to bedside is particularly challenging for mucosal administration since a variety of parameters will influence the specific barrier properties of the mucus including the luminal fluids, the microbiota, the mucus composition and clearance rate, and the condition of the underlying epithelia. Besides, after administration, nanoparticles interact with the mucosal components, forming a biomolecular corona that modulates their behavior and fate after mucosal administration. These interactions are greatly influenced by the nanoparticle properties, and therefore different designs and surface-engineering strategies have been proposed. Overall, it is essential to evaluate these biomolecule-nanoparticle interactions by complementary techniques using complex and relevant mucus barrier matrices.
Collapse
Affiliation(s)
- María García-Díaz
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Ditlev Birch
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Feng Wan
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Hanne Mørck Nielsen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
189
|
Wasik BR, Barnard KN, Ossiboff RJ, Khedri Z, Feng KH, Yu H, Chen X, Perez DR, Varki A, Parrish CR. Distribution of O-Acetylated Sialic Acids among Target Host Tissues for Influenza Virus. mSphere 2017; 2:e00379-16. [PMID: 28904995 PMCID: PMC5588038 DOI: 10.1128/msphere.00379-16] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 08/17/2017] [Indexed: 12/30/2022] Open
Abstract
Sialic acids (Sias) are important glycans displayed on the cells and tissues of many different animals and are frequent targets for binding and modification by pathogens, including influenza viruses. Influenza virus hemagglutinins bind Sias during the infection of their normal hosts, while the encoded neuraminidases and/or esterases remove or modify the Sia to allow virion release or to prevent rebinding. Sias naturally occur in a variety of modified forms, and modified Sias can alter influenza virus host tropisms through their altered interactions with the viral glycoproteins. However, the distribution of modified Sia forms and their effects on pathogen-host interactions are still poorly understood. Here we used probes developed from viral Sia-binding proteins to detect O-acetylated (4-O-acetyl, 9-O-acetyl, and 7,9-O-acetyl) Sias displayed on the tissues of some natural or experimental hosts for influenza viruses. These modified Sias showed highly variable displays between the hosts and tissues examined. The 9-O-acetyl (and 7,9-) modified Sia forms were found on cells and tissues of many hosts, including mice, humans, ferrets, guinea pigs, pigs, horses, dogs, as well as in those of ducks and embryonated chicken egg tissues and membranes, although in variable amounts. The 4-O-acetyl Sias were found in the respiratory tissues of fewer animals, being primarily displayed in the horse and guinea pig, but were not detected in humans or pigs. The results suggest that these Sia variants may influence virus tropisms by altering and selecting their cell interactions. IMPORTANCE Sialic acids (Sias) are key glycans that control or modulate many normal cell and tissue functions while also interacting with a variety of pathogens, including many different viruses. Sias are naturally displayed in a variety of different forms, with modifications at several positions that can alter their functional interactions with pathogens. In addition, Sias are often modified or removed by enzymes such as host or pathogen esterases or sialidases (neuraminidases), and Sia modifications can alter those enzymatic activities to impact pathogen infections. Sia chemical diversity in different hosts and tissues likely alters the pathogen-host interactions and influences the outcome of infection. Here we explored the display of 4-O-acetyl, 9-O-acetyl, and 7,9-O-acetyl modified Sia forms in some target tissues for influenza virus infection in mice, humans, birds, guinea pigs, ferrets, swine, horses, and dogs, which encompass many natural and laboratory hosts of those viruses.
Collapse
Affiliation(s)
- Brian R. Wasik
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Karen N. Barnard
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Robert J. Ossiboff
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Zahra Khedri
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, USA
| | - Kurtis H. Feng
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Hai Yu
- Department of Chemistry, University of California, Davis, Davis, California, USA
| | - Xi Chen
- Department of Chemistry, University of California, Davis, Davis, California, USA
| | - Daniel R. Perez
- Department of Population Health, Poultry Diagnostic and Research Center, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Ajit Varki
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, USA
| | - Colin R. Parrish
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
190
|
Zhu W, Wang C, Wang BZ. From Variation of Influenza Viral Proteins to Vaccine Development. Int J Mol Sci 2017; 18:ijms18071554. [PMID: 28718801 PMCID: PMC5536042 DOI: 10.3390/ijms18071554] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/10/2017] [Accepted: 07/14/2017] [Indexed: 11/19/2022] Open
Abstract
Recurrent influenza epidemics and occasional pandemics are one of the most important global public health concerns and are major causes of human morbidity and mortality. Influenza viruses can evolve through antigen drift and shift to overcome the barriers of human immunity, leading to host adaption and transmission. Mechanisms underlying this viral evolution are gradually being elucidated. Vaccination is an effective method for the prevention of influenza virus infection. However, the emergence of novel viruses, including the 2009 pandemic influenza A (H1N1), the avian influenza A virus (H7N9), and the highly pathogenic avian influenza A virus (HPAI H5N1), that have infected human populations frequently in recent years reveals the tremendous challenges to the current influenza vaccine strategy. A better vaccine that provides protection against a wide spectrum of various influenza viruses and long-lasting immunity is urgently required. Here, we review the evolutionary changes of several important influenza proteins and the influence of these changes on viral antigenicity, host adaption, and viral pathogenicity. Furthermore, we discuss the development of a potent universal influenza vaccine based on this knowledge.
Collapse
Affiliation(s)
- Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA 30303, USA.
| | - Chao Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA 30303, USA.
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA 30303, USA.
| |
Collapse
|
191
|
Ruch TR, Engel JN. Targeting the Mucosal Barrier: How Pathogens Modulate the Cellular Polarity Network. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a027953. [PMID: 28193722 DOI: 10.1101/cshperspect.a027953] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The mucosal barrier is composed of polarized epithelial cells with distinct apical and basolateral surfaces separated by tight junctions and serves as both a physical and immunological barrier to incoming pathogens. Specialized polarity proteins are critical for establishment and maintenance of polarity. Many human pathogens have evolved virulence mechanisms that target the polarity network to enhance binding, create replication niches, move through the barrier by transcytosis, or bypass the barrier by disrupting cell-cell junctions. This review summarizes recent advances and compares and contrasts how three important human pathogens that colonize mucosal surfaces, Pseudomonas aeruginosa, Helicobacter pylori, and Neisseria meningitidis, subvert the host cell polarization machinery during infection.
Collapse
Affiliation(s)
- Travis R Ruch
- Department of Medicine, University of California, San Francisco, San Francisco, California 94143
| | - Joanne N Engel
- Department of Medicine, University of California, San Francisco, San Francisco, California 94143.,Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
192
|
Openshaw PJ, Chiu C, Culley FJ, Johansson C. Protective and Harmful Immunity to RSV Infection. Annu Rev Immunol 2017; 35:501-532. [DOI: 10.1146/annurev-immunol-051116-052206] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Peter J.M. Openshaw
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, London W2 1PG, United Kingdom
| | - Chris Chiu
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, London W2 1PG, United Kingdom
| | - Fiona J. Culley
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, London W2 1PG, United Kingdom
| | - Cecilia Johansson
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, London W2 1PG, United Kingdom
| |
Collapse
|
193
|
Abstract
Landscape ecology examines the relationships between the spatial arrangement of different landforms and the processes that give rise to spatial and temporal patterns in local community structure. The spatial ecology of the microbial communities that inhabit the human body-in particular, those of the nose, mouth, and throat-deserves greater attention. Important questions include what defines the size of a population (i.e., "patch") in a given body site, what defines the boundaries of distinct patches within a single body site, and where and over what spatial scales within a body site are gradients detected. This Review looks at the landscape ecology of the upper respiratory tract and mouth and seeks greater clarity about the physiological factors-whether immunological, chemical, or physical-that govern microbial community composition and function and the ecological traits that underlie health and disease.
Collapse
Affiliation(s)
- Diana M Proctor
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David A Relman
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Infectious Diseases Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA.
| |
Collapse
|
194
|
Solano MI, Woolfitt AR, Williams TL, Pierce CL, Gubareva LV, Mishin V, Barr JR. Quantification of Influenza Neuraminidase Activity by Ultra-High Performance Liquid Chromatography and Isotope Dilution Mass Spectrometry. Anal Chem 2017; 89:3130-3137. [PMID: 28192976 DOI: 10.1021/acs.analchem.6b04902] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mounting evidence suggests that neuraminidase's functionality extends beyond its classical role in influenza virus infection and that antineuraminidase antibodies offer protective immunity. Therefore, a renewed interest in the development of neuraminidase (NA)-specific methods to characterize the glycoprotein and evaluate potential advantages for NA standardization in influenza vaccines has emerged. NA displays sialidase activity by cleaving off the terminal N-acetylneuraminic acid on α-2,3 or α-2,6 sialic acid containing receptors of host cells. The type and distribution of these sialic acid containing receptors is considered to be an important factor in transmission efficiency of influenza viruses between and among host species. Changes in hemagglutinin (HA) binding and NA specificity in reassortant viruses may be related to the emergence of new and potentially dangerous strains of influenza. Current methods to investigate neuraminidase activity use small derivatized sugars that are poor models for natural glycoprotein receptors and do not provide information on the linkage specificity. Here, a novel approach for rapid and accurate quantification of influenza neuraminidase activity is achieved utilizing ultra-high performance liquid chromatography (UPLC) and isotope dilution mass spectrometry (IDMS). Direct LC-MS/MS quantification of NA-released sialic acid provides precise measurement of influenza neuraminidase activity over a range of substrates. The method provides exceptional sensitivity and specificity with a limit of detection of 0.38 μM for sialic acid and the capacity to obtain accurate measurements of specific enzyme activity preference toward α-2,3-sialyllactose linkages, α-2,6-sialyllactose linkages, or whole glycosylated proteins such as fetuin.
Collapse
Affiliation(s)
- Maria I Solano
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention , 4770 Buford Highway, Atlanta, Georgia 30341, United States
| | - Adrian R Woolfitt
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention , 4770 Buford Highway, Atlanta, Georgia 30341, United States
| | - Tracie L Williams
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention , 4770 Buford Highway, Atlanta, Georgia 30341, United States
| | - Carrie L Pierce
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention , 4770 Buford Highway, Atlanta, Georgia 30341, United States
| | - Larisa V Gubareva
- Influenza Division, National Center for Immunization and Respiratory Infections, Centers for Disease Control and Prevention , Atlanta, Georgia 30329, United States
| | - Vasiliy Mishin
- Influenza Division, National Center for Immunization and Respiratory Infections, Centers for Disease Control and Prevention , Atlanta, Georgia 30329, United States
| | - John R Barr
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention , 4770 Buford Highway, Atlanta, Georgia 30341, United States
| |
Collapse
|
195
|
Thibault PA, Watkinson RE, Moreira-Soto A, Drexler JF, Lee B. Zoonotic Potential of Emerging Paramyxoviruses: Knowns and Unknowns. Adv Virus Res 2017; 98:1-55. [PMID: 28433050 PMCID: PMC5894875 DOI: 10.1016/bs.aivir.2016.12.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The risk of spillover of enzootic paramyxoviruses and the susceptibility of recipient human and domestic animal populations are defined by a broad collection of ecological and molecular factors that interact in ways that are not yet fully understood. Nipah and Hendra viruses were the first highly lethal zoonotic paramyxoviruses discovered in modern times, but other paramyxoviruses from multiple genera are present in bats and other reservoirs that have unknown potential to spillover into humans. We outline our current understanding of paramyxovirus reservoir hosts and the ecological factors that may drive spillover, and we explore the molecular barriers to spillover that emergent paramyxoviruses may encounter. By outlining what is known about enzootic paramyxovirus receptor usage, mechanisms of innate immune evasion, and other host-specific interactions, we highlight the breadth of unexplored avenues that may be important in understanding paramyxovirus emergence.
Collapse
Affiliation(s)
| | - Ruth E Watkinson
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Jan F Drexler
- Institute of Virology, University of Bonn Medical Centre, Bonn, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Benhur Lee
- Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
196
|
Abstract
Respiratory syncytial virus (RSV) is a common cause of upper respiratory tract infection in children and adults. However, infection with this virus sometimes leads to severe lower respiratory disease and is the major cause of infant hospitalisations in the developed world. Several risk factors such as baby prematurity and congenital heart disease are known to predispose towards severe disease but previously healthy, full-term infants can also develop bronchiolitis and viral pneumonia during RSV infection. The causes of severe disease are not fully understood but may include dysregulation of the immune response to the virus, resulting in excessive recruitment and activation of innate and adaptive immune cells that can cause damage. This review highlights recent discoveries on the balancing act of immune-mediated virus clearance versus immunopathology during RSV infection.
Collapse
Affiliation(s)
- Cecilia Johansson
- Respiratory Infections Section, St Mary's campus, National Heart and Lung Institute, Imperial College London, London, W2 1PG, UK
| |
Collapse
|
197
|
Gardner JK, Herbst-Kralovetz MM. Three-Dimensional Rotating Wall Vessel-Derived Cell Culture Models for Studying Virus-Host Interactions. Viruses 2016; 8:v8110304. [PMID: 27834891 PMCID: PMC5127018 DOI: 10.3390/v8110304] [Citation(s) in RCA: 34] [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: 08/13/2016] [Revised: 10/18/2016] [Accepted: 10/31/2016] [Indexed: 12/31/2022] Open
Abstract
The key to better understanding complex virus-host interactions is the utilization of robust three-dimensional (3D) human cell cultures that effectively recapitulate native tissue architecture and model the microenvironment. A lack of physiologically-relevant animal models for many viruses has limited the elucidation of factors that influence viral pathogenesis and of complex host immune mechanisms. Conventional monolayer cell cultures may support viral infection, but are unable to form the tissue structures and complex microenvironments that mimic host physiology and, therefore, limiting their translational utility. The rotating wall vessel (RWV) bioreactor was designed by the National Aeronautics and Space Administration (NASA) to model microgravity and was later found to more accurately reproduce features of human tissue in vivo. Cells grown in RWV bioreactors develop in a low fluid-shear environment, which enables cells to form complex 3D tissue-like aggregates. A wide variety of human tissues (from neuronal to vaginal tissue) have been grown in RWV bioreactors and have been shown to support productive viral infection and physiological meaningful host responses. The in vivo-like characteristics and cellular features of the human 3D RWV-derived aggregates make them ideal model systems to effectively recapitulate pathophysiology and host responses necessary to conduct rigorous basic science, preclinical and translational studies.
Collapse
Affiliation(s)
- Jameson K Gardner
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ 85004, USA.
| | - Melissa M Herbst-Kralovetz
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ 85004, USA.
| |
Collapse
|