Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
PLoS Pathog ; 18(6): e1010590, 2022 06.
Article in English | MEDLINE | ID: mdl-35700214

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been transmitted across all over the world, in contrast to the limited epidemic of genetically- and virologically-related SARS-CoV. However, the molecular basis explaining the difference in the virological characteristics among SARS-CoV-2 and SARS-CoV has been poorly defined. Here we identified that host sialoglycans play a significant role in the efficient spread of SARS-CoV-2 infection, while this was not the case with SARS-CoV. SARS-CoV-2 infection was significantly inhibited by α2-6-linked sialic acid-containing compounds, but not by α2-3 analog, in VeroE6/TMPRSS2 cells. The α2-6-linked compound bound to SARS-CoV-2 spike S1 subunit to competitively inhibit SARS-CoV-2 attachment to cells. Enzymatic removal of cell surface sialic acids impaired the interaction between SARS-CoV-2 spike and angiotensin-converting enzyme 2 (ACE2), and suppressed the efficient spread of SARS-CoV-2 infection over time, in contrast to its least effect on SARS-CoV spread. Our study provides a novel molecular basis of SARS-CoV-2 infection which illustrates the distinctive characteristics from SARS-CoV.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Peptidyl-Dipeptidase A/metabolism , Polysaccharides/metabolism , Protein Binding , Spike Glycoprotein, Coronavirus/metabolism
2.
Biol Pharm Bull ; 46(9): 1231-1239, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37357386

ABSTRACT

Personal protective equipment (PPE), including medical masks, should be worn for preventing the transmission of respiratory pathogens via infective droplets and aerosols. In medical masks, the key layer is the filter layer, and the melt-blown nonwoven fabric (NWF) is the most used fabric. However, the NWF filter layer cannot kill or inactivate the pathogens spread via droplets and aerosols. Povidone-iodine (PVP-I) has been used as an antiseptic solution given its potent broad-spectrum activity against pathogens. To develop PPE (e.g., medical masks) with anti-pathogenic activity, we integrated PVP-I into nylon-66 NWF. We then evaluated its antiviral activity against influenza A viruses by examining the viability of Madin-Darby canine kidney (MDCK) cells after inoculation with the virus strains exposed to the PVP-I-integrated nylon-66 NWF. The PVP-I nylon-66 NWF protected the MDCK cells from viral infection in a PVP-I concentration-dependent manner. Subsequently, we found to integrate PVP-I into nylon-66 and polyurethane materials among various materials. These PVP-I materials were also effective against influenza virus infection, and treatment with PVP-I nylon-66 NWF showed the highest cell survival among all the tested materials. PVP-I showed anti-influenza A virus activity when used in conjunction with PPE materials. Moreover, nylon-66 NWF integrated with PVP-I was found to be the best material to ensure anti-influenza activity. Therefore, PVP-I-integrated masks could have the potential to inhibit respiratory virus infection. Our results provide new information for developing multi-functional PPEs with anti-viral activity by integrating them with PVP-I to prevent the potential transmission of respiratory viruses.


Subject(s)
Influenza, Human , Orthomyxoviridae , Animals , Dogs , Humans , Povidone-Iodine/pharmacology , Povidone-Iodine/therapeutic use , Nylons , Respiratory Aerosols and Droplets , Influenza, Human/prevention & control
3.
J Virol ; 94(18)2020 08 31.
Article in English | MEDLINE | ID: mdl-32641475

ABSTRACT

Some avian influenza (AI) viruses have a deletion of up to 20 to 30 amino acids in their neuraminidase (NA) stalk. This has been associated with changes in virus replication and host range. Currently prevalent H9N2 AI viruses have only a 2- or 3-amino-acid deletion, and such deletions were detected in G1 and Y280 lineage viruses, respectively. The effect of an NA deletion on the H9N2 phenotype has not been fully elucidated. In this study, we isolated G1 mutants that carried an 8-amino-acid deletion in their NA stalk. To systematically analyze the effect of NA stalk length and concomitant (de)glycosylation on G1 replication and host range, we generated G1 viruses that had various NA stalk lengths and that were either glycosylated or not glycosylated. The stalk length was correlated with NA sialidase activity, using low-molecular-weight substrates, and with virus elution efficacy from erythrocytes. G1 virus replication in avian cells and eggs was positively correlated with the NA stalk length but was negatively correlated in human cells and mice. NA stalk length modulated G1 virus entry into host cells, with shorter stalks enabling more efficient G1 entry into human cells. However, with a hemagglutinin (HA) with a higher α2,6-linked sialylglycan affinity, the effect of NA stalk length on G1 virus infection was reversed, with shorter NA stalks reducing virus entry into human cells. These results indicate that a balance between HA binding affinity and NA sialidase activity, modulated by NA stalk length, is required for optimal G1 virus entry into human airway cells.IMPORTANCE H9N2 avian influenza (AI) virus, one of the most prevalent AI viruses, has caused repeated poultry and human infections, posing a huge public health risk. The H9N2 virus has diversified into multiple lineages, with the G1 lineage being the most prevalent worldwide. In this study, we isolated G1 variants carrying an 8-amino-acid deletion in their NA stalk, which is, to our knowledge, the longest deletion found in H9N2 viruses in the field. The NA stalk length was found to modulate G1 virus entry into host cells, with the effects being species specific and dependent on the corresponding HA binding affinity. Our results suggest that, in nature, H9N2 G1 viruses balance their HA and NA functions by the NA stalk length, leading to the possible association of host range and virulence in poultry and mammals during the evolution of G1 lineage viruses.


Subject(s)
Gene Expression Regulation, Viral , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H9N2 Subtype/genetics , Influenza in Birds/virology , Neuraminidase/genetics , Orthomyxoviridae Infections/virology , Amino Acid Sequence , Animals , Chickens , Genotype , Glycosylation , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hemagglutinins , Host Specificity , Host-Pathogen Interactions/genetics , Humans , Influenza A Virus, H9N2 Subtype/metabolism , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza in Birds/genetics , Influenza in Birds/metabolism , Influenza in Birds/pathology , Mice , Neuraminidase/metabolism , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Phenotype , Phylogeny , Receptors, Virus , Sequence Deletion , Structure-Activity Relationship , Virulence , Virus Internalization , Virus Replication
4.
J Virol ; 92(11)2018 06 01.
Article in English | MEDLINE | ID: mdl-29563293

ABSTRACT

Transmission of avian influenza (AI) viruses to mammals involves phylogenetic bottlenecks that select small numbers of variants for transmission to new host species. However, little is known about the AI virus quasispecies diversity that produces variants for virus adaptation to humans. Here, we analyzed the hemagglutinin (HA) genetic diversity produced during AI H5N1 single-virus infection of primary human airway cells and characterized the phenotypes of these variants. During single-virus infection, HA variants emerged with increased fitness to infect human cells. These variants generally had decreased HA thermostability, an indicator of decreased transmissibility, that appeared to compensate for their increase in α2,6-linked sialic acid (α2,6 Sia) binding specificity and/or in the membrane fusion pH threshold, each of which is an advantageous mutational change for viral infection of human airway epithelia. An HA variant with increased HA thermostability also emerged but could not outcompete variants with less HA thermostability. These results provided data on HA quasispecies diversity in human airway cells.IMPORTANCE The diversity of the influenza virus quasispecies that emerges from a single infection is the starting point for viral adaptation to new hosts. A few studies have investigated AI virus quasispecies diversity during human adaptation using clinical samples. However, those studies could be appreciably affected by individual variability and multifactorial respiratory factors, which complicate identification of quasispecies diversity produced by selective pressure for increased adaptation to infect human airway cells. Here, we found that detectable HA genetic diversity was produced by H5N1 single-virus infection of human airway cells. Most of the HA variants had increased fitness to infect human airway cells but incurred a fitness cost of less HA stability. To our knowledge, this is the first report to characterize the adaptive changes of AI virus quasispecies produced by infection of human airway cells. These results provide a better perspective on AI virus adaptation to infect humans.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza, Human/transmission , Quasispecies/genetics , Receptors, Virus/metabolism , Respiratory Mucosa/cytology , Animals , Cell Line , Chlorocebus aethiops , Dogs , Genetic Variation/genetics , HEK293 Cells , Humans , Influenza A Virus, H5N1 Subtype/classification , Influenza, Human/pathology , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Receptors, Virus/genetics , Respiratory Mucosa/virology , Respiratory System/virology , Sialic Acids/metabolism , Vero Cells , Virus Attachment
5.
Molecules ; 24(14)2019 Jul 17.
Article in English | MEDLINE | ID: mdl-31319565

ABSTRACT

Influenza A viruses leading to infectious respiratory diseases cause seasonal epidemics and sometimes periodic global pandemics. Viral polymerase is an attractive target in inhibiting viral replication, and 4'-ethynyladenosine, which has been reported as a highly potent anti-human immunodeficiency virus (HIV) nucleoside derivative, can work as an anti-influenza agent. Herein, we designed and synthesized a 4'-ethynyl-2'-deoxyadenosine 5'-monophosphate analog called EdAP (5). EdAP exhibited potent inhibition against influenza virus multiplication in Madin-Darby canine kidney (MDCK) cells transfected with human α2-6-sialyltransferase (SIAT1) cDNA and did not show any toxicity toward the cells. Surprisingly, this DNA-type nucleic acid analog (5) inhibited the multiplication of influenza A virus, although influenza virus is an RNA virus that does not generate DNA.


Subject(s)
Antiviral Agents/pharmacology , Deoxyadenine Nucleotides/pharmacology , Deoxyadenosines/chemical synthesis , Influenza, Human/drug therapy , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Deoxyadenine Nucleotides/chemical synthesis , Deoxyadenine Nucleotides/chemistry , Deoxyadenosines/chemistry , Deoxyadenosines/pharmacology , Dogs , HEK293 Cells , Humans , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Virus Replication/drug effects
6.
Bioorg Med Chem ; 26(1): 17-24, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29198893

ABSTRACT

We are interested in new non-natural glycosides with sialic acid conjugates and their biological activities. We report the synthesis of eleven non-natural occurring glycosides, which are triterpene (glycyrrhetinic acid and its derivatives)-sialic acid conjugates, and their inhibitory activities against influenza virus sialidases and influenza virus multiplication in MDCK host cells. Deoxoglycyrrhetol-sialic acid conjugates (6d and 6e) and oleanolic acid-sialic acid conjugates (7d and 7e) showed strong inhibitory activities against three subtypes of influenza virus sialidases. These four compounds (6d, 6e, 7d and 7e) showed clear inhibition to influenza virus multiplication but not to MDCK host cell survival.


Subject(s)
Antiviral Agents/pharmacology , N-Acetylneuraminic Acid/pharmacology , Orthomyxoviridae/drug effects , Triterpenes/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cell Survival/drug effects , Chickens , Dogs , Dose-Response Relationship, Drug , Madin Darby Canine Kidney Cells , Microbial Sensitivity Tests , Molecular Structure , N-Acetylneuraminic Acid/chemistry , Structure-Activity Relationship , Triterpenes/chemistry
7.
J Antimicrob Chemother ; 70(10): 2797-809, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26169554

ABSTRACT

OBJECTIVES: The purpose of this study was to develop a new compound to overcome influenza epidemics and pandemics as well as drug resistance. METHODS: We synthesized a new compound carrying: (i) Neu5Acα2-6Galß1-4GlcNAc (6SLN) for targeting immutable haemagglutinins (HAs) unless switched from human-type receptor preference; (ii) an acyl chain (lipo) for locking the compound with the viral HA via hydrophobic interactions; and (iii) a flexible poly-α-L-glutamic acid (PGA) for enhancing the compound solubility and for coating the viral surface, precluding accessibility of the PGA-coated virus to the negatively charged sialic acid on the host cell surface. RESULTS: 6SLN-lipo PGA appears to subvert binding of pandemic H1 and seasonal H3 HAs to receptors, as assessed by using guinea pig erythrocytes, which is critical for virus entry into host cells for multiplication. It shows high potency with IC50 values in the range of 300-500 nM against multiplication of both influenza pandemic H1N1/2009 and seasonal H3N2/2004 viruses in cell culture. It acts in synergism with either of the two FDA-approved neuraminidase inhibitor (NAI) clinical drugs, zanamivir (Relenza(®)) and oseltamivir carboxylate (active form of Tamiflu(®)), and it has the potential to aid NAI drugs to achieve complete clearance of the virus from the culture. CONCLUSIONS: 6SLN-lipo PGA is a new potential candidate drug for influenza control and is an attractive candidate for use in combination with an NAI drug for minimized toxicity, delayed development of resistance, prevention and treatment with the potential for eradication of human influenza.


Subject(s)
Antiviral Agents/pharmacology , Glycolipids/pharmacology , Influenza A virus/drug effects , Neuraminidase/antagonists & inhibitors , Polyglutamic Acid/analogs & derivatives , Viral Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cell Line , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Inhibitors/pharmacology , Erythrocytes/drug effects , Erythrocytes/virology , Glycolipids/chemical synthesis , Glycolipids/chemistry , Humans , Influenza A virus/physiology , Inhibitory Concentration 50 , Polyglutamic Acid/chemical synthesis , Polyglutamic Acid/chemistry , Polyglutamic Acid/pharmacology , Protein Binding , Receptors, Virus/metabolism , Virus Attachment/drug effects
8.
J Virol ; 88(8): 3953-64, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24501418

ABSTRACT

UNLABELLED: Influenza viruses of the H6 subtype have been isolated from wild and domestic aquatic and terrestrial avian species throughout the world since their first detection in a turkey in Massachusetts in 1965. Since 1997, H6 viruses with different neuraminidase (NA) subtypes have been detected frequently in the live poultry markets of southern China. Although sequence information has been gathered over the last few years, the H6 viruses have not been fully biologically characterized. To investigate the potential risk posed by H6 viruses to humans, here we assessed the receptor-binding preference, replication, and transmissibility in mammals of a series of H6 viruses isolated from live poultry markets in southern China from 2008 to 2011. Among the 257 H6 strains tested, 87 viruses recognized the human type receptor. Genome sequence analysis of 38 representative H6 viruses revealed 30 different genotypes, indicating that these viruses are actively circulating and reassorting in nature. Thirty-seven of 38 viruses tested in mice replicated efficiently in the lungs and some caused mild disease; none, however, were lethal. We also tested the direct contact transmission of 10 H6 viruses in guinea pigs and found that 5 viruses did not transmit to the contact animals, 3 viruses transmitted to one of the three contact animals, and 2 viruses transmitted to all three contact animals. Our study demonstrates that the H6 avian influenza viruses pose a clear threat to human health and emphasizes the need for continued surveillance and evaluation of the H6 influenza viruses circulating in nature. IMPORTANCE: Avian influenza viruses continue to present a challenge to human health. Research and pandemic preparedness have largely focused on the H5 and H7 subtype influenza viruses in recent years. Influenza viruses of the H6 subtype have been isolated from wild and domestic aquatic and terrestrial avian species throughout the world since their first detection in the United States in 1965. Since 1997, H6 viruses have been detected frequently in the live poultry markets of southern China; however, the biological characterization of these viruses is very limited. Here, we assessed the receptor-binding preference, replication, and transmissibility in mammals of a series of H6 viruses isolated from live poultry markets in southern China and found that 34% of the viruses are able to bind human type receptors and that some of them are able to transmit efficiently to contact animals. Our study demonstrates that the H6 viruses pose a clear threat to human health.


Subject(s)
Influenza A virus/isolation & purification , Influenza A virus/pathogenicity , Influenza in Birds/virology , Influenza, Human/virology , Poultry Diseases/virology , Animals , Chickens , Ducks , Female , Guinea Pigs , Humans , Influenza A virus/genetics , Influenza A virus/physiology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Phylogeny , Turkeys , Virulence
9.
Article in English | MEDLINE | ID: mdl-22728439

ABSTRACT

Influenza virus hemagglutinin (HA) contains antigenic sites recognized by the host immune system, cleavage sites cleaved by host proteases, receptor binding sites attaching to sialyl receptors on the target cell, and fusion peptides mediating membrane fusion. Change in an amino acid(s) in these sites may affect the potential of virus infection and spread within and between hosts. Influenza viruses with H1 HA infect birds, pigs and humans and have caused two of the four pandemics in the past 100 years: 1918 pandemic that killed 21-50 million people and 2009 pandemic that caused more than 18,000 deaths. Understanding the relationship between antigenic structure and immune specificity, the receptor binding specificity in virus transmission, how the cleavage site controls pathogenicity, and how the fusion peptide causes membrane fusion for the entry of influenza virus into the host cell should provide information to find more effective ways to prevent and control influenza.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Orthomyxoviridae/immunology , Amino Acid Sequence , Animals , Antigens, Viral/immunology , Humans , Molecular Sequence Data , Receptors, Virus/metabolism , Structure-Activity Relationship , Virus Internalization
10.
Methods Mol Biol ; 2556: 205-242, 2022.
Article in English | MEDLINE | ID: mdl-36175637

ABSTRACT

The large variation of influenza A viruses (IAVs) in various susceptible hosts and their rapid evolution, which allows host/tissue switching, host immune escape, vaccine escape, and drug resistance, are difficult challenges for influenza control in all countries worldwide. Access and binding of the IAV to actual receptors at endocytic sites is critical for the establishment of influenza infection. In this chapter, the progress in identification of and roles of glycans and non-glycans on the epithelium and in the immune system in H1-H18 IAV infections are reviewed. The first part of the review is on current knowledge of H1-H16 IAV receptors on the epithelium including sialyl glycans, other negatively charged glycans, and annexins. The second part of the review focuses on H1-H16 IAV receptors in the immune system including acidic surfactant phospholipids, Sia on surfactant proteins, the carbohydrate recognition domain (CRD) of surfactant proteins, Sia on mucins, Sia and C-type lectins on macrophages and dendritic cells, and Sia on NK cells. The third part of the review is about a possible H17-H18 IAV receptor. Binding of these receptors to IAVs may result in inhibition or enhancement of IAV infection depending on their location, host cell type, and IAV strain. Among these receptors, host sialyl glycans are key determinants of viral hemagglutinin (HA) lectins for H1-H16 infections. HA must acquire mutations to bind to sialyl glycans that are dominant on a new target tissue when switching to a new host for efficient transmission and to bind to long sialyl glycans found in the case of seasonal HAs with multiple glycosylation sites as a consequence of immune evasion. Although sialyl receptors/C-type lectins on immune cells are decoy receptors/pathogen recognition receptors for capturing viral HA lectin/glycans protecting HA antigenic sites, some IAV strains do not escape, such as by release with neuraminidase, but hijack these molecules to gain entry and replication in immune cells. An understanding of the virus-host battle tactics at the receptor level might lead to the establishment of novel strategies for effective control of influenza.


Subject(s)
Influenza A virus , Influenza Vaccines , Influenza, Human , Epithelium , Hemagglutinins, Viral , Humans , Lectins, C-Type , Macrophages , Mucins , Neuraminidase , Polysaccharides
11.
Methods Mol Biol ; 2556: 243-271, 2022.
Article in English | MEDLINE | ID: mdl-36175638

ABSTRACT

Ongoing seasonal HCoV-OC43 and HCoV-HKU1 (common cold), an ongoing zoonotic infection of highly lethal MERS-CoV in humans (MERS disease), and an ongoing pandemic SARS-CoV-2 (COVID-19) with high mutability giving some variants causing severe illness and death have been reported to attach to sialyl receptors via their spike (S) glycoproteins and via additional short spikes, hemagglutinin-esterase (HE) glycoproteins, for HCoV-OC43 and HCoV-HKU1. There is lack of zoonotic viruses that are origins of HCoV-HKU1 and the first recorded pandemic CoV (SARS-CoV-2) for studies. In this chapter, we review current knowledge of the roles of sialyl glycans in infections with these viruses in distinct infection stages. Determination of the similarities and differences in roles of sialyl glycans in infections with these viruses could lead to a better understanding of the pathogenesis and transmission that is essential for combating infections with CoVs that recognize sialyl glycans.


Subject(s)
COVID-19 , Coronavirus OC43, Human , Middle East Respiratory Syndrome Coronavirus , Esterases , Hemagglutinins , Humans , Polysaccharides , SARS-CoV-2
12.
Methods Mol Biol ; 2556: 303-320, 2022.
Article in English | MEDLINE | ID: mdl-36175641

ABSTRACT

Methods to synthesize influenza virus inhibitors with fluoro, phosphono, and/or sulfo functional groups are described. The resulting sialic acid analogues are produced from the natural substrate N-acetylneuraminic acid as starting material. Fluorescent assay methods for inhibition of influenza neuraminidase and virus proliferation are also provided.


Subject(s)
Influenza, Human , N-Acetylneuraminic Acid , Coloring Agents , Humans , Influenza, Human/drug therapy , N-Acetylneuraminic Acid/pharmacology , Neuraminidase
13.
Methods Mol Biol ; 2556: 321-353, 2022.
Article in English | MEDLINE | ID: mdl-36175642

ABSTRACT

Depending on the strain, influenza A virus causes animal, zoonotic, pandemic, or seasonal influenza with varying degrees of severity. Two surface glycoprotein spikes, hemagglutinin (HA) and neuraminidase (NA), are the most important influenza A virus antigens. NA plays an important role in the propagation of influenza virus by removing terminal sialic acid from sialyl decoy receptors and thereby facilitating the release of viruses from traps such as in mucus and on infected cells. Some NA inhibitors have become widely used drugs for treatment of influenza. However, attempts to develop effective and safe NA inhibitors that can be used for treatment of anti-NA drugs-resistant influenza viruses have continued. In this chapter, we describe the following updates on influenza A NA inhibitor development: (i) N-acetylneuraminic acid (Neu5Ac)-based derivatives, (ii) covalent NA inhibitors, (iii) sulfo-sialic acid analogs, (iv) N-acetyl-6-sulfo-ß-D-glucosaminide-based inhibitors, (v) inhibitors targeting the 150-loop of group 1 NAs, (vi) conjugation inhibitors, (vii) acylhydrazone derivatives, (viii) monoclonal antibodies, (ix) PVP-I, and (x) natural products. Finally, we provide future perspectives on the next-generation anti-NA drugs.


Subject(s)
Biological Products , Influenza A virus , Influenza, Human , Animals , Antibodies, Monoclonal , Antiviral Agents/pharmacology , Hemagglutinins , Humans , N-Acetylneuraminic Acid , Neuraminidase , Povidone-Iodine
14.
J Gen Virol ; 91(Pt 4): 938-48, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20007353

ABSTRACT

Alterations of the receptor-binding properties of swine influenza A viruses (SIVs) during their isolation in embryonated chicken eggs have not been well studied. In this study, the receptor-binding properties of classical H1 SIVs isolated solely in eggs or Madin-Darby canine kidney (MDCK) cells were examined. Sequencing analysis revealed substitutions of D190V/N or D225G in the haemagglutinin (HA) proteins in egg isolates, whereas MDCK isolates retained HA genes identical to those of the original viruses present in the clinical samples. Egg isolates with substitution of either D190V/N or D225G had increased haemagglutinating activity for mouse and sheep erythrocytes, but reduced activity for rabbit erythrocytes. Additionally, egg isolates with D225G had increased haemagglutination activity for chicken erythrocytes. A direct binding assay using a sialyl glycopolymer that possessed either a 5-N-acetylneuraminic acid (Neu5Ac) alpha2,6galactose (Gal) or a Neu5Acalpha2,3Gal linkage revealed that the egg isolates used in this study showed higher binding activity to the Neu5Acalpha2,3Gal receptor than MDCK isolates. Increased binding activity of the egg isolates to the Neu5Acalpha2,3Gal receptor was also confirmed by haemagglutination assay with resialylated chicken erythrocytes by Galbeta1,3/4GlcNAcalpha2,3-sialyltransferase. These observations were reinforced by flow-cytometric and N-glycan analyses of the erythrocytes. The alpha2,3-linked sialic acids were expressed predominantly on the surface of mouse and sheep erythrocytes. Chicken erythrocytes expressed Neu5Acalpha2,3Gal more abundantly than Neu5Acalpha2,6Gal, and rabbit erythrocytes expressed both 5-N-glycolylneuraminic acid (Neu5Gc) alpha2,6Gal and Neu5Acalpha2,6Gal. Our results demonstrate clearly that classical H1 SIVs undergo alterations in receptor-binding activity associated with an amino acid substitution in the HA protein during isolation and propagation in embryonated chicken eggs.


Subject(s)
Influenza A Virus, H1N1 Subtype/physiology , Receptors, Virus/physiology , Amino Acid Substitution , Animals , Cell Line , Chick Embryo/virology , Dogs , Erythrocytes/chemistry , Hemagglutination Tests , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N1 Subtype/isolation & purification , Mice , N-Acetylneuraminic Acid/blood , N-Acetylneuraminic Acid/chemistry , Rats , Sheep , Swine
15.
Methods Mol Biol ; 2132: C1, 2020.
Article in English | MEDLINE | ID: mdl-32990934

ABSTRACT

The inadvertently published below contents have been corrected.

16.
Methods Mol Biol ; 2132: 547-565, 2020.
Article in English | MEDLINE | ID: mdl-32306356

ABSTRACT

Infections by H1-H16 influenza A viruses require sufficient binding of viral hemagglutinins (HAs) to specific target receptors, glycoconjugates bearing sialyl sugar chains, on the host cell surface. Synthesized sialyl sugar chains targeting sialyl sugar-binding sites in HAs that are immutable as long as the virus does not switch to a different host species might therefore be highly effective candidate drugs for inhibition of the initial required step of virus entry. In this chapter, we describe the following aspects of updated sialyl sugar chains as influenza A virus HA inhibitors (HAIs): (1) mode of terminal sialyl-galactose linkage, (2) molecular length and structure of sialyl glycan receptors, (3) multivalent sialyl sugar chain dimension, (4) clustering of sialyl sugar chains on macromolecular scaffolds, and (5) enhancement of the stability of sialyl sugar chain HA inhibitors. We also discuss about the use of HAI-based combinations that should be considered for future influenza therapy.


Subject(s)
Antiviral Agents/chemical synthesis , Glycoconjugates/metabolism , Hemagglutinins, Viral/metabolism , Influenza A virus/physiology , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Binding Sites/drug effects , Drug Development , Drug Therapy, Combination , Glycoconjugates/chemistry , Hemagglutinins, Viral/chemistry , Humans , Influenza A virus/drug effects , N-Acetylneuraminic Acid/chemistry , Protein Binding/drug effects , Virus Internalization/drug effects
17.
Methods Mol Biol ; 2132: 483-545, 2020.
Article in English | MEDLINE | ID: mdl-32306355

ABSTRACT

On the cell sur "face", sialoglycoconjugates act as receptionists that have an important role in the first step of various cellular processes that bridge communication between the cell and its environment. Loss of Sia production can cause the developmental of defects and lethality in most animals; hence, animal cells are less prone to evolution of resistance to interactions by rapidly evolved Sia-binding viruses. Obligative intracellular viruses mostly have rapid evolution that allows escape from host immunity, leading to an epidemic variant, and that allows emergence of a novel strain, occasionally leading to pandemics that cause health-social-economic problems. Recently, much attention has been given to the mutual recognition systems via sialosugar chains between viruses and their host cells and there has been rapid growth of the research field "sialoglycovirology." In this chapter, the structural diversity of sialoglycoconjugates is overviewed, and enveloped and non-enveloped viruses that bind to Sia are reviewed. Also, interactions of viral lectins-host Sia receptors, which determine viral transmission, host range, and pathogenesis, are presented. The future direction of new therapeutic routes targeting viral lectins, development of easy-to-use detection methods for diagnosis and monitoring changes in virus binding specificity, and challenges in the development of suitable viruses to use in virus-based therapies for genetic disorders and cancer are discussed.


Subject(s)
Lectins/metabolism , N-Acetylneuraminic Acid/chemistry , Receptors, Virus/physiology , Viruses/metabolism , Binding Sites , Host Microbial Interactions , Lectins/chemistry , Receptors, Virus/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Attachment , Virus Physiological Phenomena
18.
Vaccines (Basel) ; 8(4)2020 Oct 06.
Article in English | MEDLINE | ID: mdl-33036202

ABSTRACT

Among the four genera of influenza viruses (IVs) and the four genera of coronaviruses (CoVs), zoonotic αIV and ßCoV have occasionally caused airborne epidemic outbreaks in humans, who are immunologically naïve, and the outbreaks have resulted in high fatality rates as well as social and economic disruption and losses. The most devasting influenza A virus (IAV) in αIV, pandemic H1N1 in 1918, which caused at least 40 million deaths from about 500 million cases of infection, was the first recorded emergence of IAVs in humans. Usually, a novel human-adapted virus replaces the preexisting human-adapted virus. Interestingly, two IAV subtypes, A/H3N2/1968 and A/H1N1/2009 variants, and two lineages of influenza B viruses (IBV) in ßIV, B/Yamagata and B/Victoria lineage-like viruses, remain seasonally detectable in humans. Both influenza C viruses (ICVs) in γIV and four human CoVs, HCoV-229E and HCoV-NL63 in αCoV and HCoV-OC43 and HCoV-HKU1 in ßCoV, usually cause mild respiratory infections. Much attention has been given to CoVs since the global epidemic outbreaks of ßSARS-CoV in 2002-2004 and ßMERS-CoV from 2012 to present. ßSARS-CoV-2, which is causing the ongoing COVID-19 pandemic that has resulted in 890,392 deaths from about 27 million cases of infection as of 8 September 2020, has provoked worldwide investigations of CoVs. With the aim of developing efficient strategies for controlling virus outbreaks and recurrences of seasonal virus variants, here we overview the structures, diversities, host ranges and host receptors of all IVs and CoVs and critically review current knowledge of receptor binding specificity of spike glycoproteins, which mediates infection, of IVs and of zoonotic, pandemic and seasonal CoVs.

19.
Methods Mol Biol ; 2132: 567-583, 2020.
Article in English | MEDLINE | ID: mdl-32306357

ABSTRACT

We describe a method to detect influenza virus using an evanescent-field-activated fluorescence scanner type glycan array and ELISA system. Neoglycoprotein was prepared by combination of organic chemistry and biomaterial preparation. These ligands were spotted on a glass plate or plastic well to make a glycan array and ELISA plate. We detected cultured influenza virus using glycan array and ELISA. Then, we showed that the neoglycoprotein binds to Cy3-labeled hemagglutinins (H1 and H5), a NeuAcα2,6LacNAc or NeuAcα2,3LacNAc recognized protein, as detected.


Subject(s)
Glycoproteins/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A virus/isolation & purification , Animals , Chemistry, Organic , Dogs , Enzyme-Linked Immunosorbent Assay , Humans , Madin Darby Canine Kidney Cells , Protein Array Analysis
20.
Glycoconj J ; 26(4): 433-43, 2009 May.
Article in English | MEDLINE | ID: mdl-18853253

ABSTRACT

The initial step essential in influenza virus infection is specific binding of viral hemagglutinin to host cell-surface glycan receptors. Influenza A virus specificity for the host is mediated by viral envelope hemagglutinin, that binds to receptors containing glycans with terminal sialic acids. Human viruses preferentially bind to alpha2-->6 linked sialic acids on receptors of host cells, whereas avian viruses are specific for the alpha2-->3 linkage on the target cells. Human influenza virus isolates more efficiently infect amniotic membrane (AM) cells than chorioallantoic membrane (CAM) cells. N-glycans were isolated from AM and CAM cells of 10-day-old chicken embryonated eggs and their structures were analyzed by multi-dimensional HPLC mapping and MALDI-TOF-MS techniques. Terminal N-acetylneuraminic acid contents in the two cell types were similar. However, molar percents of alpha2-->3 linkage preferentially bound by avian influenza virus were 27.2 in CAM cells and 15.4 in AM cells, whereas those of alpha2-->6 linkage favored by human influenza virus were 8.3 (CAM) and 14.2 (AM). Molar percents of sulfated glycans, recognized by human influenza virus, in CAM and AM cells were 3.8 and 12.7, respectively. These results have revealed structures and molar percents of N-glycans in CAM and AM cells important in determining human and avian influenza virus infection and viral adaptation.


Subject(s)
Adaptation, Biological , Amnion/cytology , Birds/virology , Chorioallantoic Membrane/cytology , Influenza A virus/metabolism , Ovum/cytology , Polysaccharides/analysis , Amnion/metabolism , Animals , Carbohydrate Conformation , Carbohydrate Sequence , Chick Embryo , Chorioallantoic Membrane/metabolism , Chromatography, High Pressure Liquid , Humans , Molecular Sequence Data , Polysaccharides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL