Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Cells ; 13(2)2024 01 10.
Article in English | MEDLINE | ID: mdl-38247817

ABSTRACT

The membrane (M) glycoprotein of coronaviruses (CoVs) serves as the nidus for virion assembly. Using a yeast two-hybrid screen, we identified the interaction of the cytosolic tail of Murine Hepatitis Virus (MHV-CoV) M protein with Myosin Vb (MYO5B), specifically with the alternative splice variant of cellular MYO5B including exon D (MYO5B+D), which mediates interaction with Rab10. When co-expressed in human lung epithelial A549 and canine kidney epithelial MDCK cells, MYO5B+D co-localized with the MHV-CoV M protein, as well as with the M proteins from Porcine Epidemic Diarrhea Virus (PEDV-CoV), Middle East Respiratory Syndrome (MERS-CoV) and Severe Acute Respiratory Syndrome 2 (SARS-CoV-2). Co-expressed M proteins and MYO5B+D co-localized with endogenous Rab10 and Rab11a. We identified point mutations in MHV-CoV M that blocked the interaction with MYO5B+D in yeast 2-hybrid assays. One of these point mutations (E121K) was previously shown to block MHV-CoV virion assembly and its interaction with MYO5B+D. The E to K mutation at homologous positions in PEDV-CoV, MERS-CoV and SARS-CoV-2 M proteins also blocked colocalization with MYO5B+D. The knockdown of Rab10 blocked the co-localization of M proteins with MYO5B+D and was rescued by re-expression of CFP-Rab10. Our results suggest that CoV M proteins traffic through Rab10-containing systems, in association with MYO5B+D.


Subject(s)
Coronavirus M Proteins , Animals , Dogs , Humans , Madin Darby Canine Kidney Cells/metabolism , Madin Darby Canine Kidney Cells/virology , Middle East Respiratory Syndrome Coronavirus , Myosins , rab GTP-Binding Proteins/genetics , Saccharomyces cerevisiae , Swine , Viral Matrix Proteins , SARS-CoV-2/metabolism , Murine hepatitis virus/metabolism , A549 Cells/metabolism , A549 Cells/virology , Porcine epidemic diarrhea virus/metabolism
2.
Eur J Med Chem ; 227: 113929, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34700269

ABSTRACT

Annual unpredictable efficacy of vaccines, coupled with emerging drug resistance, underlines the development of new antiviral drugs to treat influenza infections. The N-terminal domain of the PA (PAN) endonuclease is both highly conserved across influenza strains and serotypes and is indispensable for the viral lifecycle, making it an attractive target for new antiviral therapies. Here, we describe the discovery of a new class of PAN inhibitors derived from recently identified, highly active hits for PAN endonuclease inhibition. By use of structure-guided design and systematic SAR exploration, the hits were elaborated through a fragment growing strategy, giving rise to a series of 1, 3-cis-2-substituted-1-(3, 4-dihydroxybenzyl)-6, 7-dihydroxy-1, 2, 3, 4-tetrahydroisoquinoline-3-carboxylic acid derivatives as potent PAN inhibitors. This approach ultimately resulted in the development of a new lead compound 13e, which exhibited an EC50 value of 4.50 µM against H1N1 influenza virus in MDCK cells.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery , Endonucleases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Tetrahydroisoquinolines/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cell Survival/drug effects , Dogs , Dose-Response Relationship, Drug , Endonucleases/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Tetrahydroisoquinolines/chemical synthesis , Tetrahydroisoquinolines/chemistry
3.
Front Immunol ; 12: 747654, 2021.
Article in English | MEDLINE | ID: mdl-34956182

ABSTRACT

The complement system is designed to recognise and eliminate invading pathogens via activation of classical, alternative and lectin pathways. Human properdin stabilises the alternative pathway C3 convertase, resulting in an amplification loop that leads to the formation of C5 convertase, thereby acting as a positive regulator of the alternative pathway. It has been noted that human properdin on its own can operate as a pattern recognition receptor and exert immune functions outside its involvement in complement activation. Properdin can bind directly to microbial targets via DNA, sulfatides and glycosaminoglycans, apoptotic cells, nanoparticles, and well-known viral virulence factors. This study was aimed at investigating the complement-independent role of properdin against Influenza A virus infection. As one of the first immune cells to arrive at the site of IAV infection, we show here that IAV challenged neutrophils released properdin in a time-dependent manner. Properdin was found to directly interact with haemagglutinin, neuraminidase and matrix 1 protein Influenza A virus proteins in ELISA and western blot. Furthermore, modelling studies revealed that properdin could bind HA and NA of the H1N1 subtype with higher affinity compared to that of H3N2 due to the presence of an HA cleavage site in H1N1. In an infection assay using A549 cells, properdin suppressed viral replication in pH1N1 subtype while promoting replication of H3N2 subtype, as revealed by qPCR analysis of M1 transcripts. Properdin treatment triggered an anti-inflammatory response in H1N1-challenged A549 cells and a pro-inflammatory response in H3N2-infected cells, as evident from differential mRNA expression of TNF-α, NF-κB, IFN-α, IFN-ß, IL-6, IL-12 and RANTES. Properdin treatment also reduced luciferase reporter activity in MDCK cells transduced with H1N1 pseudotyped lentiviral particles; however, it was increased in the case of pseudotyped H3N2 particles. Collectively, we conclude that infiltrating neutrophils at the site of IAV infection can release properdin, which then acts as an entry inhibitor for pandemic H1N1 subtype while suppressing viral replication and inducing an anti-inflammatory response. H3N2 subtype can escape this immune restriction due to altered haemagglutinin and neuraminindase, leading to enhanced viral entry, replication and pro-inflammatory response. Thus, depending on the subtype, properdin can either limit or aggravate IAV infection in the host.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/immunology , Neutrophils/immunology , Properdin/immunology , Animals , Dogs , Humans , Madin Darby Canine Kidney Cells/immunology , Madin Darby Canine Kidney Cells/virology
4.
Eur J Med Chem ; 225: 113762, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34411893

ABSTRACT

Neuraminidase (NA) is an important target for the treatment of influenza. In this study, a new lead NA inhibitor, 4 (ZINC01121127), was discovered by pharmacophore-based virtual screening and molecular dynamic (MD) simulation. Some novel NA inhibitors containing thiophene ring were synthesized by optimizing the skeleton of the lead compound 4. Compound 4b had the most potent inhibitory activity against NA (IC50 = 0.03 µM), which was better than the positive control oseltamivir carboxylate (IC50 = 0.06 µM). 4b (EC50 = 1.59 µM) also exhibits excellent antiviral activity against A/chicken/Hubei/327/2004 (H5N1-DW), which is superior to the reference drug OSC (EC50 = 5.97 µM). Molecular docking study shows that the thiophene moiety plays an essential role in compound 4b, which can bind well to the active site of NA. The good activity of 4b may be also ascribed to the extending of quinoline ring into the 150-cavity. The results of this study may provide an insightful help for the development of new NA inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Influenza A Virus, H5N1 Subtype/drug effects , Neuraminidase/antagonists & inhibitors , Thiophenes/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dogs , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Influenza A Virus, H5N1 Subtype/enzymology , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Neuraminidase/metabolism , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry
5.
FEBS J ; 288(16): 4869-4891, 2021 08.
Article in English | MEDLINE | ID: mdl-33629527

ABSTRACT

Glycosylation is considered as a critical quality attribute for the production of recombinant biopharmaceuticals such as hormones, blood clotting factors, or monoclonal antibodies. In contrast, glycan patterns of immunogenic viral proteins, which differ significantly between the various expression systems, are hardly analyzed yet. The influenza A virus (IAV) proteins hemagglutinin (HA) and neuraminidase (NA) have multiple N-glycosylation sites, and alteration of N-glycan micro- and macroheterogeneity can have strong effects on virulence and immunogenicity. Here, we present a versatile and powerful glycoanalytical workflow that enables a comprehensive N-glycosylation analysis of IAV glycoproteins. We challenged our workflow with IAV (A/PR/8/34 H1N1) propagated in two closely related Madin-Darby canine kidney (MDCK) cell lines, namely an adherent MDCK cell line and its corresponding suspension cell line. As expected, N-glycan patterns of HA and NA from virus particles produced in both MDCK cell lines were similar. Detailed analysis of the HA N-glycan microheterogeneity showed an increasing variability and a higher complexity for N-glycosylation sites located closer to the head region of the molecule. In contrast, NA was found to be exclusively N-glycosylated at site N73. Almost all N-glycan structures were fucosylated. Furthermore, HA and NA N-glycan structures were exclusively hybrid- and complex-type structures, to some extent terminated with alpha-linked galactose(s) but also with blood group H type 2 and blood group A epitopes. In contrast to the similarity of the overall glycan pattern, differences in the relative abundance of individual structures were identified. This concerned, in particular, oligomannose-type, alpha-linked galactose, and multiantennary complex-type N-glycans.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A virus/chemistry , Madin Darby Canine Kidney Cells/metabolism , Neuraminidase/metabolism , Animals , Dogs , Glycosylation , Hemagglutinin Glycoproteins, Influenza Virus/analysis , Influenza A virus/metabolism , Madin Darby Canine Kidney Cells/virology , Neuraminidase/analysis
6.
ACS Appl Bio Mater ; 4(3): 2044-2051, 2021 03 15.
Article in English | MEDLINE | ID: mdl-35014330

ABSTRACT

Influenza A virus (IAV) is internalized into its host cells by endocytosis, which involves many cellular proteins and molecules. In this study, we focus on the function of calcium ion (Ca2+) in IAV endocytosis. We have found that IAV infection is accompanied by the increase in concentration of cytosolic Ca2+, which is mainly attributed to the influx of extracellular Ca2+. When Ca2+ influx is abolished, IAV internalization will be markedly suppressed, but the virus attachment to its host cells will be unaffected. Extracellular Ca2+ influx is essential to the clathrin-mediated endocytosis (CME) of IAVs but dispensable to the clathrin-independent endocytosis of the virus and is dispensable to the CME of transferrin or low-density lipoprotein as a control. Ca2+ influx might participate in the dynamin-promoted membrane fission in the CME of IAVs. Our study highlights that IAVs enter host cells via extracellular Ca2+ influx-involved clathrin- and dynamin-dependent endocytosis, which will facilitate better understanding of IAV infection and development of anti-influenza drugs.


Subject(s)
Biocompatible Materials/chemistry , Calcium/metabolism , Clathrin/metabolism , Influenza A virus/metabolism , Madin Darby Canine Kidney Cells/metabolism , Animals , Dogs , Endocytosis , Madin Darby Canine Kidney Cells/virology , Materials Testing , Particle Size
7.
Bioorg Chem ; 102: 104106, 2020 09.
Article in English | MEDLINE | ID: mdl-32739481

ABSTRACT

Parasitic characteristics, mutations and resistance of influenza A virus make it difficult for current influenza antiviral drugs to maintain long-term effectiveness. Currently, to design non-adamantane compounds targeting the S31N mutant of M2 proton channel is a promising direction for the development of novel anti-influenza drugs. In our previous research, a pinanamine-based antiviral M090 was discovered to target hemagglutinin instead of M2, with its structure being highly similar to reported M2-S31N inhibitors. Herein, a series of pinane oxime derivatives were designed from scratch and evaluated for anti-influenza activity and their cytotoxicity in vitro. Utilizing a combination of structure-activity relationship analysis, electrophysiological assay and molecular docking, the most potent compound 11h, as a M2-S31N blocker, exhibited excellent activity with EC50 value at the low micromolar level against both H3N2 and H1N1. No significant toxicity of 11h was observed. In addition, compound 11h was located tightly in the pore of the drug-binding site with the thiophene moiety facing down toward the C-terminus, and did not adopt a similar position and orientation as the reference inhibitor.


Subject(s)
Antiviral Agents/pharmacology , Bicyclic Monoterpenes/pharmacology , Drug Design , Influenza A virus/drug effects , Oximes/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Bicyclic Monoterpenes/chemical synthesis , Bicyclic Monoterpenes/chemistry , Dogs , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Molecular Structure , Oximes/chemical synthesis , Oximes/chemistry , Structure-Activity Relationship
8.
Int J Mol Sci ; 21(17)2020 Aug 25.
Article in English | MEDLINE | ID: mdl-32854295

ABSTRACT

Phenotypic variation in cultured mammalian cell lines is known to be induced by passaging and culture conditions. Yet, the effect these variations have on the production of viral vectors has been overlooked. In this work we evaluated the impact of using Madin-Darby canine kidney (MDCK) parental cells from American Type Culture Collection (ATCC) or European Collection of Authenticated Cell Cultures (ECACC) cell bank repositories in both adherent and suspension cultures for the production of canine adenoviral vectors type 2 (CAV-2). To further explore the differences between cells, we conducted whole-genome transcriptome analysis. ECACC's MDCK showed to be a less heterogeneous population, more difficult to adapt to suspension and serum-free culture conditions, but more permissive to CAV-2 replication progression, enabling higher yields. Transcriptome data indicated that this increased permissiveness is due to a general down-regulation of biological networks of innate immunity in ECACC cells, including apoptosis and death receptor signaling, Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling, toll-like receptors signaling and the canonical pathway of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling. These results show the impact of MDCK source on the outcome of viral-based production processes further elucidating transcriptome signatures underlying enhanced adenoviral replication. Following functional validation, the genes and networks identified herein can be targeted in future engineering approaches aiming at improving the production of CAV-2 gene therapy vectors.


Subject(s)
Adenoviruses, Canine/growth & development , Gene Expression Profiling/methods , Madin Darby Canine Kidney Cells/cytology , Virus Cultivation/methods , Animals , Biological Specimen Banks , Cell Adhesion , Culture Media, Serum-Free , Dogs , Gene Expression Regulation , Gene Regulatory Networks , Madin Darby Canine Kidney Cells/classification , Madin Darby Canine Kidney Cells/virology , Virus Replication , Exome Sequencing
9.
Viruses ; 12(7)2020 07 20.
Article in English | MEDLINE | ID: mdl-32698456

ABSTRACT

The influenza A virus (IAV) matrix-2 (M2) protein is an antigenically conserved viral envelope protein that plays an important role in virus budding together with another envelope protein, hemagglutinin (HA). An M2-specific mouse monoclonal IgG antibody, rM2ss23, which binds to the ectodomain of the M2 protein, has been shown to be a non-neutralizing antibody, but inhibits plaque formation of IAV strains. In this study, we generated chimeric rM2ss23 (ch-rM2ss23) IgG and IgA antibodies with the same variable region and compared their antiviral activities. Using gel chromatography, ch-rM2ss23 IgA were divided into three antibody subsets: monomeric IgA (m-IgA), dimeric IgA (d-IgA), and trimeric and tetrameric IgA (t/q-IgA). We found that t/q-IgA had a significantly higher capacity to reduce the plaque size of IAVs than IgG and m-IgA, most likely due to the decreased number of progeny virus particles produced from infected cells. Interestingly, HA-M2 colocalization was remarkably reduced on the infected cell surface in the presence of ch-rM2ss23 antibodies. These results indicate that anti-M2 polymeric IgA restricts IAV budding more efficiently than IgG and suggest a role of anti-M2 IgA in cross-protective immunity to IAVs.


Subject(s)
Immunoglobulin A/immunology , Immunoglobulin G/immunology , Influenza A virus/immunology , Viral Matrix Proteins/immunology , Animals , Blotting, Western , Cross Reactions/immunology , Dogs , Enzyme-Linked Immunosorbent Assay , Humans , Influenza A virus/ultrastructure , Madin Darby Canine Kidney Cells/virology , Mice , Microscopy, Electron, Transmission , Neutralization Tests , Real-Time Polymerase Chain Reaction , Recombinant Proteins , Surface Plasmon Resonance , Viral Plaque Assay
10.
Viruses ; 12(7)2020 07 16.
Article in English | MEDLINE | ID: mdl-32708703

ABSTRACT

Canine adenoviruses (CAdVs) are divided into pathotypes CAdV1 and CAdV2, which cause infectious hepatitis and laryngotracheitis in canid animals, respectively. They can be the backbones of viral vectors that could be applied in recombinant vaccines or for gene transfer in dogs and in serologically naïve humans. Although conventional plasmid-based reverse genetics systems can be used to construct CAdV vectors, their large genome size creates technical difficulties in gene cloning and manipulation. In this study, we established an improved reverse genetics system for CAdVs using bacterial artificial chromosomes (BACs), in which genetic modifications can be efficiently and simply made through BAC recombineering. To validate the utility of this system, we used it to generate CAdV2 with the early region 1 gene deleted. This mutant was robustly generated and attenuated in cell culture. The results suggest that our established BAC-based reverse genetics system for CAdVs would be a useful and powerful tool for basic and advanced practical studies with these viruses.


Subject(s)
Adenoviruses, Canine/genetics , Chromosomes, Artificial, Bacterial/genetics , Reverse Genetics/methods , Adenoviridae Infections/veterinary , Adenoviridae Infections/virology , Animals , Cloning, Molecular , Dogs , Genome, Viral/genetics , Hepatitis, Infectious Canine/virology , High-Throughput Nucleotide Sequencing , Madin Darby Canine Kidney Cells/virology
11.
Viruses ; 12(7)2020 07 17.
Article in English | MEDLINE | ID: mdl-32709116

ABSTRACT

The complex overlap in waterfowl migratory pathways across the world has established numerous occurrences of genetic reassortment and intercontinental spread of avian influenza virus (AIV) over long distances, thereby calling for huge efforts and targeted surveillance for infection control. During annual surveillance in South Korea in 2018, a novel avian influenza H6N5 (K6) subtype was isolated from the fecal sample of wild bird. Genomic characterization using a phylogenetic tree indicated the K6 virus to be of North American-origin, with partial homology to an H6N5 strain, A/Aix galericulata/South Korea/K17-1638-5/2017 (K17). A monobasic residue at the HA cleavage site and absence of a notable mutation at the HA receptor-binding site suggested the isolate to be of low pathogenicity. However, molecular analysis revealed the E119V mutation in the NA gene and a human host marker mutation E382D in the polymerase acidic (PA) gene, implying their susceptibility to neuraminidase inhibitors and potential infectivity in humans, respectively. For comparison, K6 and K17 were found to be dissimilar for various mutations, such as A274T of PB2, S375N/T of PB1, or V105M of NP, each concerning the increased virulence of K6 in mammalian system. Moreover, kinetic data presented the highest viral titer of this H6N5 isolate at 106.37 log10TCID50 after 48 h of infection, thus proving efficient adaptability for replication in a mammalian system in vitro. The mouse virus challenge study showed insignificant influence on the total body weight, while viral load shedding in lungs peaked at 1.88 ± 0.21 log10 TICD50/mL, six days post infection. The intercontinental transmission of viruses from North America may continuously be present in Korea, thereby providing constant opportunities for virus reassortment with local resident AIVs; these results hint at the increased potential risk of host jumping capabilities of the new isolates. Our findings reinforce the demand for regular surveillance, not only in Korea but also along the flyways in Alaska.


Subject(s)
Geese/virology , Genome, Viral/genetics , Influenza A virus/genetics , Influenza in Birds/virology , Poultry Diseases/virology , Animals , Dogs , Female , High-Throughput Nucleotide Sequencing , Influenza A virus/isolation & purification , Influenza A virus/physiology , Influenza in Birds/epidemiology , Madin Darby Canine Kidney Cells/virology , Mice , Mice, Inbred BALB C , North America/epidemiology , Orthomyxoviridae Infections/virology , Phylogeny , Poultry Diseases/epidemiology , Republic of Korea/epidemiology , Sequence Homology , Virus Replication
12.
Eur J Med Chem ; 199: 112357, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32428793

ABSTRACT

We describe the preparation of thiosialoside-modified poly (methyl vinyl ether-alt-maleic anhydride) as second-generation polymeric conjugates for the inhibition of influenza virus infection. These synthetic glycopolymers show significantly enhanced neuraminidase inhibitory and antiviral activity in enzyme and cellular levels, respectively. The polyvalent thiosialosides also exhibit comparable inhibitory activity to the first-line anti-influenza drugs Zanamivir® and Oseltamivir® against the PR8 influenza virus strain in virus growth inhibition assays, which may be attributed to multivalent binding to neuraminidase on the virion particles, leading to the virion aggregation and further inhibiting the attaching/fusion and releasing steps in the influenza virus life-cycle. These findings suggest that attaching monomeric sialoside with neuraminidase inhibitory activity to a polymeric scaffold will synergistically disturb both the early and late stages of influenza virus infection, and provides a basis for the development of efficacious anti-viral agents against both wild-type and drug-resistant mutant strains.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/drug effects , Orthomyxoviridae Infections/drug therapy , Polymers/pharmacology , Sialic Acids/pharmacology , Thioglycosides/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cells, Cultured , Dogs , Dose-Response Relationship, Drug , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Molecular Structure , Polymers/chemical synthesis , Polymers/chemistry , Sialic Acids/chemical synthesis , Sialic Acids/chemistry , Structure-Activity Relationship , Thioglycosides/chemical synthesis , Thioglycosides/chemistry
13.
Angew Chem Int Ed Engl ; 59(36): 15532-15536, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32421225

ABSTRACT

In this study, we demonstrate the concept of "topology-matching design" for virus inhibitors. With the current knowledge of influenza A virus (IAV), we designed a nanoparticle-based inhibitor (nano-inhibitor) that has a matched nanotopology to IAV virions and shows heteromultivalent inhibitory effects on hemagglutinin and neuraminidase. The synthesized nano-inhibitor can neutralize the viral particle extracellularly and block its attachment and entry to the host cells. The virus replication was significantly reduced by 6 orders of magnitude in the presence of the reverse designed nano-inhibitors. Even when used 24 hours after the infection, more than 99.999 % inhibition is still achieved, which indicates such a nano-inhibitor might be a potent antiviral for the treatment of influenza infection.


Subject(s)
Antiviral Agents/pharmacology , Drug Design , Influenza A virus/drug effects , Influenza, Human/drug therapy , Nanoparticles/chemistry , Zanamivir/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dogs , Glycerol/chemistry , Glycerol/pharmacology , Humans , Lactose/analogs & derivatives , Lactose/chemistry , Lactose/pharmacology , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Molecular Structure , Particle Size , Polymers/chemistry , Polymers/pharmacology , Sialic Acids/chemistry , Sialic Acids/pharmacology , Surface Properties , Virus Replication/drug effects , Zanamivir/chemical synthesis , Zanamivir/chemistry
14.
Arch Pharm Res ; 43(5): 489-502, 2020 May.
Article in English | MEDLINE | ID: mdl-32248350

ABSTRACT

Curcumin derivatives have been shown to inhibit replication of human influenza A viruses (IAVs). However, it is not clear whether curcumin and its derivatives can inhibit neuraminidase (NA) of influenza virus. In this study, a meaningful 3D quantitative structure-activity relationship model (comparative molecular field analysis R2 = 0.997, q2 = 0.527, s = 0.064, F = 282.663) was built to understand the chemical-biological interactions between their activities and neuraminidase. Molecular docking was used to predict binding models between curcumin derivatives and neuraminidase. Real-time polymerase chain reactions showed that the five active curcumin derivatives might have direct effects on viral particle infectivity in H1N1-infected lung epithelial (MDCK) cells. Neuraminidase activation assay showed that five active curcumin derivatives decreased H1N1-induced neuraminidase activation in MDCK cells. Indirect immunofluorescence assay indicated that two active curcumin derivatives (tetramethylcurcumin and curcumin) down-regulated the nucleoprotein expression. Curcumin inhibited IAV in vivo. The therapeutic mechanism of curcumin in the treatment of influenza viral pneumonia is related to improving the immune function of infected mice and regulating secretion of tumor necrosis-α, interleukin-6, and interferon-γ. These results indicate that curcumin derivatives inhibit IAV by blocking neuraminidase in the cellular model and curcumin also has anti-IAV activity in the animal model.


Subject(s)
Antiviral Agents/pharmacology , Curcumin/pharmacology , Enzyme Inhibitors/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Neuraminidase/antagonists & inhibitors , Quantitative Structure-Activity Relationship , Animals , Antiviral Agents/chemistry , Cell Survival/drug effects , Curcumin/chemistry , Dogs , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Influenza A Virus, H1N1 Subtype/enzymology , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Neuraminidase/metabolism
15.
Bioorg Chem ; 98: 103733, 2020 05.
Article in English | MEDLINE | ID: mdl-32171985

ABSTRACT

Seasonal or pandemic influenza virus infections are a worldwide health problem requiring antiviral therapy. Since virus resistance to the established neuraminidase inhibitors and novel polymerase inhibitors is growing, new drug targets are needed. Heat shock protein 90 (Hsp90) is associated with several aspects of the influenza virus life cycle, and is considered a relevant host cell target. We report here on a series of benzo[d]thiazole and 4,5,6,7-tetrahydrobenzo[d]thiazole derivatives with robust and selective activities against influenza A (H1N1, H3N2) and influenza B viruses. Two compounds, 1 and 4, have low micromolar EC50 values and show high binding affinities for Hsp90, which suggests that inhibition of Hsp90 is the mechanism underlying their antiviral effects. These compounds represent suitable scaffolds for designing novel Hsp90 inhibitors with favourable activities against influenza virus.


Subject(s)
Antiviral Agents/pharmacology , Benzothiazoles/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Influenza B virus/drug effects , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Benzothiazoles/chemical synthesis , Benzothiazoles/chemistry , Cells, Cultured , Dogs , Dose-Response Relationship, Drug , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship
16.
Eur J Med Chem ; 194: 112223, 2020 May 15.
Article in English | MEDLINE | ID: mdl-32220685

ABSTRACT

The influenza virus hemagglutinin (HA) is an attractive target for antiviral therapy due to its essential role in mediating virus entry into the host cell. We here report the identification of a class of N-benzyl-4,4,-disubstituted piperidines as influenza A virus fusion inhibitors with specific activity against the H1N1 subtype. Using the highly efficient one-step Ugi four-component reaction, diverse library of piperidine-based analogues was synthesized and evaluated to explore the structure-activity relationships (SAR). Mechanistic studies, including resistance selection with the most active compound (2) demonstrated that it acts as an inhibitor of the low pH-induced HA-mediated membrane fusion process. Computational studies identified an as yet unrecognized fusion inhibitor binding site, which is located at the bottom of the HA2 stem in close proximity to the fusion peptide. A direct π-stacking interaction between the N-benzylpiperidine moiety of 2 and F9HA2 of the fusion peptide, reinforced with an additional π-stacking interaction with Y119HA2, and a salt bridge of the protonated piperidine nitrogen with E120HA2, were identified as important interactions to mediate ligand binding. This site rationalized the observed SAR and provided a structural explanation for the H1N1-specific activity of our inhibitors. Furthermore, the HA1-S326V mutation resulting in resistance to 2 is close to the proposed new binding pocket. Our findings point to the N-benzyl-4,4,-disubstituted piperidines as an interesting class of influenza virus inhibitors, representing the first example of fusion peptide binders with great potential for anti-influenza drug development.


Subject(s)
Antiviral Agents/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H1N1 Subtype/drug effects , Piperidines/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dogs , Dose-Response Relationship, Drug , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Humans , Influenza A Virus, H1N1 Subtype/metabolism , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Piperidines/chemical synthesis , Piperidines/chemistry , Structure-Activity Relationship
17.
Influenza Other Respir Viruses ; 14(2): 204-209, 2020 03.
Article in English | MEDLINE | ID: mdl-31651085

ABSTRACT

BACKGROUND: Cell-based influenza vaccines can solve the problem of the frequent occurrence of egg adaptation-associated antigenic changes observed in egg-based vaccines. Seed viruses for cell-based vaccines can be prepared from clinical specimens by cell culture; however, clinical samples risk harboring respiratory viruses other than influenza virus. Therefore, it is necessary to investigate the patterns of co-infection in clinical samples and explore whether cell culture technology can selectively propagate influenza viruses from samples containing other respiratory viruses. METHODS: A total of 341 clinical specimens were collected from patients with influenza or influenza-like illness and analyzed by ResPlex II assay to detect 18 respiratory viruses. The patterns of co-infection were statistically analyzed with Fisher's exact test. The samples with double or triple infections were passaged in suspension MDCK cells (MDCK-S), adherent MDCK cells (MDCK-A), and LLC-MK2D cells. Cell-passaged samples were analyzed by ResPlex II assay again to investigate whether each cell line could amplify influenza viruses and eliminate other respiratory viruses. RESULTS: Double infections were detected in 8.5% and triple infections in 0.9% of the collected clinical specimens. We identified four pairs of viruses with significant correlation. For all samples with double and triple infection, MDCK-S and MDCK-A could selectively propagate influenza viruses, while eliminating all contaminating viruses. In contrast, LLC-MK2D showed lower isolation efficiency for influenza virus and higher isolation efficiency for coxsackievirus/echovirus than MDCK-S and MDCK-A. CONCLUSIONS: Both MDCK-S and MDCK-A are considered suitable for the preparation of influenza vaccine seed viruses without adventitious agents or egg-adaptation mutations.


Subject(s)
Madin Darby Canine Kidney Cells/virology , Orthomyxoviridae/isolation & purification , Virus Cultivation/methods , Animals , Cell Line , Dogs , Humans , Orthomyxoviridae/growth & development , Viral Vaccines
18.
Biochem Biophys Res Commun ; 523(1): 183-189, 2020 02 26.
Article in English | MEDLINE | ID: mdl-31843192

ABSTRACT

Canarium album (Lour.) Raeusch (C. album) as a normally medicinal and edible plant has been used widely in Asian countries and is considered a source of phytochemicals that are beneficial to human health. Here, we showed at the first time isocorilagin, a polyphenolic compound isolated from C. album, displayed antiviral activity against diverse strains of influenza A virus (IAV), including A/Puerto Rico/8/34 (H1N1), A/Aichi/2/68 (H3N2) and NA-H274Y (H1N1) with IC50 value of 9.19 ±â€¯1.99, 23.72 ±â€¯2.51 and 4.64 ±â€¯3.01 µM, respectively. Further mechanistic studies revealed that it clearly inhibited neuraminidase activity of IAV and directly influenced the virus release. The molecular docking studies presented isocorilagin could bind to the highly conserved residues in the active sites of NA, implying that isocorilagin may be effective against various influenza strains and not susceptible to produce drug resistance. Taken together, the results strongly suggest that isocorilagin has potential to be an effective, safe and affordable neuraminidase inhibitor against a diverse panel of IAV strains. More importantly, our work expands the biological activities of C. album extracts and provide a new option for the development of anti-influenza drug.


Subject(s)
Antiviral Agents/pharmacology , Burseraceae/chemistry , Enzyme Inhibitors/pharmacology , Influenza A virus/drug effects , Neuraminidase/antagonists & inhibitors , Tannins/pharmacology , Animals , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Cell Survival/drug effects , Dogs , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Neuraminidase/metabolism , Structure-Activity Relationship , Tannins/chemistry , Tannins/isolation & purification
19.
Bioorg Med Chem ; 28(1): 115130, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31753804

ABSTRACT

The influenza virus hemagglutinin (HA) mediates membrane fusion after viral entry by endocytosis. The fusion process requires drastic low pH-induced HA refolding and is prevented by arbidol and tert-butylhydroquinone (TBHQ). We here report a class of superior inhibitors with indole-substituted spirothiazolidinone structure. The most active analogue 5f has an EC50 value against influenza A/H3N2 virus of 1 nM and selectivity index of almost 2000. Resistance data and in silico modeling indicate that 5f combines optimized fitting in the TBHQ/arbidol HA binding pocket with a capability for endosomal accumulation. Both criteria appear relevant to achieve superior inhibitors of HA-mediated fusion.


Subject(s)
Antiviral Agents/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Indoles/pharmacology , Influenza A Virus, H3N2 Subtype/drug effects , Influenza, Human/drug therapy , Spiro Compounds/pharmacology , Thiazolidines/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dogs , Dose-Response Relationship, Drug , Humans , Hydrogen-Ion Concentration , Indoles/chemistry , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Protein Refolding/drug effects , Spiro Compounds/chemistry , Structure-Activity Relationship , Thiazolidines/chemistry
20.
Curr Top Med Chem ; 20(2): 111-120, 2020.
Article in English | MEDLINE | ID: mdl-31854280

ABSTRACT

BACKGROUND: Neuraminidase inhibitors (NAIs) are the only class of antivirals in clinical use against influenza virus approved worldwide. However, approximately 1-3% of circulating strains present resistance mutations to oseltamivir (OST), the most used NAI. Therefore, it is important to catalogue new molecules to inhibit influenza virus, especially OST-resistant strains. Natural products from tropical plants used for human consumption represent a worthy class of substances. Their use could be stimulated in resource-limited setting where the access to expensive antiviral therapies is restricted. METHODS: We evaluated the anti-influenza virus activity of agathisflavone derived from Anacardium occidentale L. RESULTS: The neuraminidase (NA) activity of wild-type and OST-resistant influenza virus was inhibited by agathisflavone, with IC50 values ranging from 20 to 2.0 µM, respectively. Agathisflavone inhibited influenza virus replication with EC50 of 1.3 µM. Sequential passages of the virus in the presence of agathisflavone revealed the emergence of mutation R249S, A250S and R253Q in the NA gene. These changes are outside the OST binding region, meaning that agathisflavone targets this viral enzyme at a region different than conventional NAIs. CONCLUSION: Altogether our data suggest that agathisflavone has a promising chemical structure for the development of anti-influenza drugs.


Subject(s)
Anacardium/chemistry , Biflavonoids/pharmacology , Enzyme Inhibitors/pharmacology , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae/drug effects , Phytochemicals/pharmacology , Animals , Biflavonoids/chemistry , Biflavonoids/isolation & purification , Cell Survival/drug effects , Dogs , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Molecular Structure , Neuraminidase/metabolism , Orthomyxoviridae/enzymology , Phytochemicals/chemistry , Phytochemicals/isolation & purification , Structure-Activity Relationship , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...