Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 180
Filter
1.
Bioorg Med Chem Lett ; 105: 129743, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38608962

ABSTRACT

Neuraminidase (NA) serves as a promising target for the exploration and development of anti-influenza drugs. In this work, lead compound 5 was discovered through pharmacophore-based virtual screening and molecular dynamics simulation, and 14 new compounds were obtained by modifying the lead compound 5 based on pharmacophore features. The biological activity test shows that 5n (IC50 = 0.13 µM) has a better inhibitory effect on wild-type NA (H5N1), while 5i (IC50 = 0.44 µM) has a prominent inhibitory effect on mutant NA (H5N1-H274Y), both of them are better than the positive control oseltamivir carboxylate (OSC). The analysis of docking results indicate that the good activities of compounds 5n and 5i may be attributed to the thiophene ring in 5n can stretch into the 150-cavity of NA, whereas the thiophene moiety in 5i can extend to the 430-cavity of NA. The findings of this study may be helpful for the discovery of new NA inhibitors.


Subject(s)
Antiviral Agents , Enzyme Inhibitors , Neuraminidase , Neuraminidase/antagonists & inhibitors , Neuraminidase/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Structure-Activity Relationship , Hydrazones/chemistry , Hydrazones/pharmacology , Hydrazones/chemical synthesis , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/enzymology , Drug Discovery , Molecular Docking Simulation , Molecular Structure , Humans , Molecular Dynamics Simulation , Dose-Response Relationship, Drug
2.
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
3.
Molecules ; 26(11)2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34199752

ABSTRACT

Coffee has been studied for its health benefits, including prevention of several chronic diseases, such as type 2 diabetes mellitus, cancer, Parkinson's, and liver diseases. Chlorogenic acid (CGA), an important component in coffee beans, was shown to possess antiviral activity against viruses. However, the presence of caffeine in coffee beans may also cause insomnia and stomach irritation, and increase heart rate and respiration rate. These unwanted effects may be reduced by decaffeination of green bean Arabica coffee (GBAC) by treatment with dichloromethane, followed by solid-phase extraction using methanol. In this study, the caffeine and chlorogenic acid (CGA) level in the coffee bean from three different areas in West Java, before and after decaffeination, was determined and validated using HPLC. The results showed that the levels of caffeine were reduced significantly, with an order as follows: Tasikmalaya (2.28% to 0.097% (97 ppm), Pangalengan (1.57% to 0.049% (495 ppm), and Garut (1.45% to 0.00002% (0.2 ppm). The CGA levels in the GBAC were also reduced as follows: Tasikmalaya (0.54% to 0.001% (118 ppm), Pangalengan (0.97% to 0.0047% (388 ppm)), and Garut (0.81% to 0.029% (282 ppm). The decaffeinated samples were then subjected to the H5N1 neuraminidase (NA) binding assay to determine its bioactivity as an anti-influenza agent. The results show that samples from Tasikmalaya, Pangalengan, and Garut possess NA inhibitory activity with IC50 of 69.70, 75.23, and 55.74 µg/mL, respectively. The low level of caffeine with a higher level of CGA correlates with their higher levels of NA inhibitory, as shown in the Garut samples. Therefore, the level of caffeine and CGA influenced the level of NA inhibitory activity. This is supported by the validation of CGA-NA binding interaction via molecular docking and pharmacophore modeling; hence, CGA could potentially serve as a bioactive compound for neuraminidase activity in GBAC.


Subject(s)
Caffeine/analysis , Chlorogenic Acid/analysis , Coffea/chemistry , Influenza A Virus, H5N1 Subtype/enzymology , Methylene Chloride/pharmacology , Neuraminidase/antagonists & inhibitors , Caffeine/adverse effects , Caffeine/pharmacology , Chlorogenic Acid/chemistry , Chlorogenic Acid/pharmacology , Chromatography, High Pressure Liquid , Coffea/drug effects , Food Handling , Humans , Influenza A Virus, H5N1 Subtype/chemistry , Influenza A Virus, H5N1 Subtype/drug effects , Inhibitory Concentration 50 , Models, Molecular , Molecular Docking Simulation , Protein Binding , Solid Phase Extraction , Viral Proteins/antagonists & inhibitors
4.
J Virol ; 95(17): e0075921, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34160258

ABSTRACT

The hemagglutinin (HA) surface protein is the primary immune target for most influenza vaccines. The neuraminidase (NA) surface protein is often a secondary target for vaccine designs. In this study, computationally optimized broadly reactive antigen (COBRA) methodology was used to generate the N1-I NA vaccine antigen that was designed to cross-react with avian, swine, and human influenza viruses of the N1 NA subtype. The elicited antibodies bound to NA proteins derived from A/California/07/2009 (H1N1)pdm09, A/Brisbane/59/2007 (H1N1), A/Swine/North Carolina/154074/2015 (H1N1), and A/Viet Nam/1203/2004 (H5N1) influenza viruses, with NA-neutralizing activity against a broad panel of HXN1 influenza strains. Mice vaccinated with the N1-I COBRA NA vaccine were protected from mortality and viral lung titers were lower when challenged with four different viral challenges (A/California/07/2009, A/Brisbane/59/2007, A/Swine/North Carolina/154074/2015, and A/Viet Nam/1203/2004). Vaccinated mice had little to no weight loss against both homologous, but also cross-NA, genetic clade challenges. Lung viral titers were lower than the mock-vaccinated mice and, at times, equivalent to the homologous control. Thus, the N1-I COBRA NA antigen has the potential to be a complementary component in a multiantigen universal influenza virus vaccine formulation that also contains HA antigens. IMPORTANCE The development and distribution of a universal influenza vaccine would alleviate global economic and public health stress from annual influenza virus outbreaks. The influenza virus NA vaccine antigen allows for protection from multiple HA subtypes and virus host origins, but it has not been the focus of vaccine development. The N1-I NA antigen described here protected mice from direct challenge of four distinct influenza viruses and inhibited the enzymatic activity of an N1 influenza virus panel. The use of the NA antigen in combination with the HA antigen widens the breadth of protection against various virus strains. Therefore, this research opens the door to the development of a longer-lasting vaccine with increased protective breadth.


Subject(s)
Immunity/immunology , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/enzymology , Influenza Vaccines/administration & dosage , Neuraminidase/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antigens, Viral/immunology , Cross Protection , Female , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Swine , Vaccination
5.
J Virol ; 95(1)2020 12 09.
Article in English | MEDLINE | ID: mdl-33028722

ABSTRACT

Adaptive mutations and/or reassortments in avian influenza virus polymerase subunits PA, PB1, and PB2 are one of the major factors enabling the virus to overcome the species barrier to infect humans. The majority of human adaptation polymerase mutations have been identified in PB2; fewer adaptation mutations have been characterized in PA and PB1. Clade 2.2.1 avian influenza viruses (H5N1) are unique to Egypt and generally carry the human adaptation PB2-E627K substitution during their dissemination in nature. In this study, we identified other human adaptation polymerase mutations by analyzing phylogeny-associated PA mutations that H5N1 clade 2.2.1 viruses have accumulated during their evolution in the field. This analysis identified several PA mutations that produced increased replication by contemporary clade 2.2.1.2 viruses in vitro in human cells and in vivo in mice compared to ancestral clade 2.2.1 viruses. The PA mutations acted cooperatively to increase viral polymerase activity and replication in both avian and human cells, with the effect being more prominent in human cells at 33°C than at 37°C. These results indicated that PA mutations have a role in establishing contemporary clade 2.2.1.2 virus infections in poultry and in adaptation to infect mammals. Our study provided data on the mechanism for PA mutations to accumulate during avian influenza virus evolution and extend the viral host range.IMPORTANCE Clade 2.2.1 avian influenza viruses (H5N1) are unique to Egypt and have caused the highest number of human H5N1 influenza cases worldwide, presenting a serious global public health threat. These viruses may have the greatest evolutionary potential for adaptation from avian hosts to human hosts. Using a comprehensive phylogenetic approach, we identified several novel clade 2.2.1 virus polymerase mutations that increased viral replication in vitro in human cells and in vivo in mice. These mutations were in the polymerase PA subunit and acted cooperatively with the E627K mutation in the PB2 polymerase subunit to provide higher replication in contemporary clade 2.2.1.2 viruses than in ancestral clade 2.2.1 viruses. These data indicated that ongoing clade 2.2.1 dissemination in the field has driven PA mutations to modify viral replication to enable host range expansion, with a higher public health risk for humans.


Subject(s)
Evolution, Molecular , Influenza A Virus, H5N1 Subtype/physiology , Orthomyxoviridae Infections/virology , RNA-Dependent RNA Polymerase/genetics , Viral Nonstructural Proteins/genetics , Adaptation, Physiological , Animals , Cell Line , Chickens , Egypt/epidemiology , Host Specificity , Humans , Influenza A Virus, H5N1 Subtype/classification , Influenza A Virus, H5N1 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/genetics , Mice , Models, Molecular , Mutation , Phylogeny , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Virus Replication/genetics
6.
Eur J Med Chem ; 200: 112423, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32512482

ABSTRACT

The most of potent neuraminidase inhibitors as zwitterions with poor lipophilicity suffered from the poor oral bioavailability. Herein, we describe a rational journey to discover a non-zwitterionic neuraminidase inhibitor 24a containing urea. It showed potent inhibitions against neuraminidases from group 1(H5N1 and H1N1) and group 2 (H3N2) subtypes and exhibited more strong inhibitory activities against neuraminidases from H274Y mutants than oseltamivir carboxylate. Whether administrated by orally or intravenous injection, the pharmacokinetic profile of compound 24a in SD rats were improved compared to oseltamivir carboxylate.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Neuraminidase/antagonists & inhibitors , Oseltamivir/pharmacology , Viral Proteins/antagonists & inhibitors , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/enzymology , Microbial Sensitivity Tests , Molecular Structure , Neuraminidase/metabolism , Oseltamivir/chemical synthesis , Oseltamivir/chemistry , Structure-Activity Relationship , Viral Proteins/metabolism
7.
Biochemistry (Mosc) ; 85(4): 490-498, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32569556

ABSTRACT

Using classical molecular dynamics, constant-pH molecular dynamics simulation, metadynamics, and combined quantum mechanical and molecular mechanical approach, we identified an alternative pathway of glycosyl-enzyme intermediate formation during oligosaccharide substrate conversion by the influenza H5N1 neuraminidase. The Asp151 residue located in the enzyme mobile loop plays a key role in catalysis within a wide pH range due to the formation of a network of interactions with water molecules. Considering that propagation of influenza virus takes place in the digestive tract of birds at low pH values and in the human respiratory tract at pH values close to neutral, the existence of alternative reaction pathways functioning at different medium pH can explain the dual tropism of the virus and circulation of H5N1 viral strains capable of transmission from birds to humans.


Subject(s)
Influenza A Virus, H5N1 Subtype/enzymology , Influenza in Birds/virology , Influenza, Human/virology , Molecular Dynamics Simulation , Neuraminidase/metabolism , Oligosaccharides/chemistry , Protein Conformation , Animals , Birds , Catalysis , Humans , Hydrogen-Ion Concentration , Influenza in Birds/genetics , Influenza in Birds/metabolism , Influenza, Human/genetics , Influenza, Human/metabolism , Models, Molecular
8.
Eur J Med Chem ; 185: 111841, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31708183

ABSTRACT

Influenza A neuraminidase plays an indispensable role in the process of replication and transmission of influenza, so the neuraminidase inhibition can prevent the reproduction of the viruses therefore achieve the effect of treatment of influenza. However, drug resistance of neuraminidase inhibitors such as oseltamivir highlights the need to develop novel structural neuraminidase inhibitors. Here we explored a series of oseltamivir derivatives bearing pyridyl group. Among them, compound 23b exhibiting potent inhibitory activity against neuraminidase from H5N1 subtype was comparable to oseltamivir carboxylate. Cytopathic effect inhibition assay in MDCK cells indicated that compound 23b exerted powerful inhibitions on influenza viruses. And compound 23b were nontoxic to MDCK cells. Meanwhile, compound 23b showed high stability towards rat liver microsomes, human liver microsomes and human plasma. This research enriched the structural type of neuraminidase inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Influenza A Virus, H5N1 Subtype/drug effects , Neuraminidase/antagonists & inhibitors , Oseltamivir/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dogs , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Influenza A Virus, H5N1 Subtype/enzymology , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/microbiology , Microbial Sensitivity Tests , Molecular Structure , Neuraminidase/metabolism , Oseltamivir/chemical synthesis , Oseltamivir/chemistry , Structure-Activity Relationship
9.
Nature ; 573(7773): 287-290, 2019 09.
Article in English | MEDLINE | ID: mdl-31485076

ABSTRACT

Influenza A viruses are responsible for seasonal epidemics, and pandemics can arise from the transmission of novel zoonotic influenza A viruses to humans1,2. Influenza A viruses contain a segmented negative-sense RNA genome, which is transcribed and replicated by the viral-RNA-dependent RNA polymerase (FluPolA) composed of PB1, PB2 and PA subunits3-5. Although the high-resolution crystal structure of FluPolA of bat influenza A virus has previously been reported6, there are no complete structures available for human and avian FluPolA. Furthermore, the molecular mechanisms of genomic viral RNA (vRNA) replication-which proceeds through a complementary RNA (cRNA) replicative intermediate, and requires oligomerization of the polymerase7-10-remain largely unknown. Here, using crystallography and cryo-electron microscopy, we determine the structures of FluPolA from human influenza A/NT/60/1968 (H3N2) and avian influenza A/duck/Fujian/01/2002 (H5N1) viruses at a resolution of 3.0-4.3 Å, in the presence or absence of a cRNA or vRNA template. In solution, FluPolA forms dimers of heterotrimers through the C-terminal domain of the PA subunit, the thumb subdomain of PB1 and the N1 subdomain of PB2. The cryo-electron microscopy structure of monomeric FluPolA bound to the cRNA template reveals a binding site for the 3' cRNA at the dimer interface. We use a combination of cell-based and in vitro assays to show that the interface of the FluPolA dimer is required for vRNA synthesis during replication of the viral genome. We also show that a nanobody (a single-domain antibody) that interferes with FluPolA dimerization inhibits the synthesis of vRNA and, consequently, inhibits virus replication in infected cells. Our study provides high-resolution structures of medically relevant FluPolA, as well as insights into the replication mechanisms of the viral RNA genome. In addition, our work identifies sites in FluPolA that could be targeted in the development of antiviral drugs.


Subject(s)
Genome, Viral/genetics , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/enzymology , Models, Molecular , RNA-Dependent RNA Polymerase/chemistry , Cryoelectron Microscopy , Crystallization , Protein Structure, Tertiary , Single-Domain Antibodies/metabolism , Virus Replication
10.
Viruses ; 11(3)2019 03 22.
Article in English | MEDLINE | ID: mdl-30909490

ABSTRACT

Significantly higher numbers of human infections with H5N1 virus have occurred in Indonesia and Egypt, compared with other affected areas, and it is speculated that there are specific viral factors for human infection with avian H5N1 viruses in these locations. We previously showed PB2-K526R is present in 80% of Indonesian H5N1 human isolates, which lack the more common PB2-E627K substitution. Testing the hypothesis that this mutation may prime avian H5N1 virus for human infection, we showed that: (1) K526R is rarely found in avian influenza viruses but was identified in H5N1 viruses 2⁻3 years after the virus emerged in Indonesia, coincident with the emergence of H5N1 human infections in Indonesia; (2) K526R is required for efficient replication of Indonesia H5N1 virus in mammalian cells in vitro and in vivo and reverse substitution to 526K in human isolates abolishes this ability; (3) Indonesian H5N1 virus, which contains K526R-PB2, is stable and does not further acquire E627K following replication in infected mice; and (4) virus containing K526R-PB2 shows no fitness deficit in avian species. These findings illustrate an important mechanism in which a host adaptive mutation that predisposes avian H5N1 virus towards infecting humans has arisen with the virus becoming prevalent in avian species prior to human infections occurring. A similar mechanism is observed in the Qinghai-lineage H5N1 viruses that have caused many human cases in Egypt; here, E627K predisposes towards human infections. Surveillance should focus on the detection of adaptation markers in avian strains that prime for human infection.


Subject(s)
Host-Pathogen Interactions/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/transmission , Mutation, Missense , Viral Proteins/genetics , Adaptation, Physiological , Amino Acid Substitution , Animals , Birds , Egypt , Humans , Indonesia , Influenza A Virus, H5N1 Subtype/enzymology , Influenza in Birds/virology , Influenza, Human/virology , Mice , Mice, Inbred BALB C , Virus Replication
11.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 2): 89-97, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30713159

ABSTRACT

The influenza neuraminidase (NA) is a homotetramer with head, stalk, transmembrane and cytoplasmic regions. The structure of the NA head with a stalk has never been determined. The NA head from an N9 subtype influenza A virus, A/tern/Australia/G70C/1975 (H1N9), was expressed with an artificial stalk derived from the tetrabrachion (TB) tetramerization domain from Staphylothermus marinus. The NA was successfully crystallized both with and without the TB stalk, and the structures were determined to 2.6 and 2.3 Šresolution, respectively. Comparisons of the two NAs with the native N9 NA structure from egg-grown virus showed that the artificial TB stalk maintained the native NA head structure, supporting previous biological observations.


Subject(s)
Bacterial Proteins/chemistry , Influenza A Virus, H5N1 Subtype/enzymology , Neuraminidase/chemistry , Amino Acid Sequence , Bacterial Proteins/metabolism , Crystallization , Crystallography, X-Ray , Desulfurococcaceae/metabolism , Humans , Influenza, Human/virology , Models, Molecular , Neuraminidase/metabolism , Protein Conformation , Protein Domains
12.
J Biomol Struct Dyn ; 37(13): 3354-3365, 2019 08.
Article in English | MEDLINE | ID: mdl-30126341

ABSTRACT

Two important glycoproteins on the influenza virus membrane, hemagglutinin (HA) and neuraminidase (NA), are relevant to virus replication. As previously reported, HA has a substrate specificity towards SIA-2,3-GAL-1,4-NAG (3SL) and SIA-2,6-GAL-1,4-NAG (6SL) glycans, while NA can cleave both types of linkages. However, the substrate binding into NA and its preference are not well understood. In this work, the glycan binding and specificity of human and avian NAs were evaluated by classical molecular dynamics (MD) simulations, whilst the conformational diversity of 3SL avian and 6SL human glycans in an unbound state was investigated by replica exchange MD simulations. The results indicated that the 3SL avian receptor fits well in the binding cavity of all NAs and does not require a conformational change for such binding compared to the flexible shape of the 6SL human receptor. From the QM/MM-GBSA binding free energy and decomposition free energy data, 6SL showed a much stronger binding towards human NAs (H1N1, H2N2 and H3N2) than to avian NAs (H5N1 and H7N9). This suggests that influenza NAs have a substrate specificity corresponding to their HA, indicating the functional balance between the two important glycoproteins. Both linkages show distinct glycan topologies when complexed with NAs, while the flexibility of torsion angles between GAL and NAG in 6SL results in the various shapes of glycan and different binding patterns. Lower conformational diversities of both glycans when bound to NA compared to the unbound state were found, and were required in order to be accommodated within the NA cavity. Communicated by Ramaswamy H. Sarma.


Subject(s)
Molecular Dynamics Simulation , Neuraminidase/metabolism , Polysaccharides/metabolism , Receptors, Virus/metabolism , Binding Sites , Humans , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/enzymology , Influenza A Virus, H7N9 Subtype/enzymology , Influenza, Human/virology , Neuraminidase/chemistry , Protein Binding , Protein Conformation , Receptors, Virus/chemistry , Substrate Specificity , Virus Replication
13.
J Virol ; 92(20)2018 10 15.
Article in English | MEDLINE | ID: mdl-30089692

ABSTRACT

The influenza A virus (IAV) neuraminidase (NA) protein plays an essential role in the release of virus particles from cells and decoy receptors. The NA enzymatic activity presumably needs to match the activity of the IAV hemagglutinin (HA) attachment protein and the host sialic acid (SIA) receptor repertoire. We analyzed the enzymatic activities of N1 NA proteins derived from avian (H5N1) and human (H1N1) IAVs and analyzed the role of the second SIA-binding site, located adjacent to the conserved catalytic site, therein. SIA contact residues in the second SIA-binding site of NA are highly conserved in avian, but not human, IAVs. All N1 proteins preferred cleaving α2,3- over α2,6-linked SIAs even when their corresponding HA proteins displayed a strict preference for α2,6-linked SIAs, indicating that the specificity of the NA protein does not need to fully match that of the corresponding HA protein. NA activity was affected by substitutions in the second SIA-binding site that are observed in avian and human IAVs, at least when multivalent rather than monovalent substrates were used. These mutations included both SIA contact residues and residues that do not directly interact with SIA in all three loops of the second SIA-binding site. Substrate binding via the second SIA-binding site enhanced the catalytic activity of N1. Mutation of the second SIA-binding site was also shown to affect virus replication in vitro Our results indicate an important role for the N1 second SIA-binding site in binding to and cleavage of multivalent substrates.IMPORTANCE Avian and human influenza A viruses (IAVs) preferentially bind α2,3- and α2,6-linked sialic acids (SIAs), respectively. A functional balance between the hemagglutinin (HA) attachment and neuraminidase (NA) proteins is thought to be important for host tropism. What this balance entails at the molecular level is, however, not well understood. We now show that N1 proteins of both avian and human viruses prefer cleaving avian- over human-type receptors although human viruses were relatively better in cleavage of the human-type receptors. In addition, we show that substitutions at different positions in the second SIA-binding site found in NA proteins of human IAVs have a profound effect on binding and cleavage of multivalent, but not monovalent, receptors and affect virus replication. Our results indicate that the HA-NA balance can be tuned via modification of substrate binding via this site and suggest an important role of the second SIA-binding site in host tropism.


Subject(s)
Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/enzymology , N-Acetylneuraminic Acid/metabolism , Neuraminidase/metabolism , Amino Acid Substitution , Binding Sites , DNA Mutational Analysis , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/physiology , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neuraminidase/genetics , Substrate Specificity , Virus Replication
14.
Arch Virol ; 163(10): 2775-2786, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29974255

ABSTRACT

Polymerase acidic (PA) protein is a multifunctional regulator of influenza A virus (IAV) replication and pathogenesis. In a previous study, we reported that nucleolin (NCL) is a novel PA-interacting host protein. In this study, we further explored the role of NCL during highly pathogenic H5N1 avian influenza virus infection. We found that depletion of endogenous NCL in mammalian cells by siRNA targeting during H5N1 infection resulted in significantly increased viral polymerase activity, elevated viral mRNA, cRNA and vRNA synthesis, accelerated viral replication, and enhanced apoptosis and necrosis. Moreover, siRNA silencing of NCL significantly exacerbated the inflammatory response, resulting in increased secretion of IL-6, TNF-α, TNF-ß, CCL-4, CCL-8, IFN-α, IFN-ß and IFN-γ. Conversely, overexpression of NCL significantly decreased IAV replication. Collectively, these data show that NCL acts as a novel potential antiviral factor during H5N1 infection. Further studies exploring the antiviral mechanisms of NCL may accelerate the development of new anti-influenza drugs.


Subject(s)
Influenza A Virus, H5N1 Subtype/enzymology , Influenza in Birds/metabolism , Influenza, Human/metabolism , Phosphoproteins/metabolism , Poultry Diseases/metabolism , RNA-Binding Proteins/metabolism , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Animals , Chemokine CCL8/genetics , Chemokine CCL8/metabolism , Chickens , Host-Pathogen Interactions , Humans , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/genetics , Influenza in Birds/virology , Influenza, Human/genetics , Influenza, Human/virology , Interferon-alpha/genetics , Interferon-alpha/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Phosphoproteins/genetics , Poultry Diseases/genetics , Poultry Diseases/virology , RNA-Binding Proteins/genetics , RNA-Dependent RNA Polymerase/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Viral Proteins/genetics , Virulence , Nucleolin
15.
Virus Res ; 253: 20-27, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29859234

ABSTRACT

The reassortment of two highly pathogenic avian influenza (HPAI) H5N1 and H7N9 viruses presents a potential challenge to human health. The hemagglutinins (HAs) and neuraminidases (NAs) of these simultaneously circulating avian influenza viruses were evaluated using the pseudoparticle (pp) system. Native and mismatched virus pps were generated to investigate their biological characteristics. The HAs and NAs of the two viruses reassorted successfully to generate infectious viral particles. H7 was demonstrated to have the ability to reassort with NA from the H5N1 viruses, resulting in the generation of virions that were highly infectious to bronchial epithelial cells. Although the Anhui H5+Anhui N9 combination showed an moderate infectivity to the four cell lines, it was most sensitive to oseltamivir. The H7 in the pps was found to be predominantly HA0. Further, H5 in the pps primarily presented as HA1, owing to the particular mechanisms underlying its maturation. All NAs predominantly existed in monomer form. In our study, HAs/NAs, in all combinations, were functional and able to perform their corresponding function in the viral life cycle. Our data suggest that HAs/NAs from the (HPAI) H5N1 and H7N9 viruses are capable of assembly into infectious virions, posing a threat topublic health.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H5N1 Subtype/metabolism , Influenza A Virus, H7N9 Subtype/metabolism , Influenza, Human/virology , Neuraminidase/metabolism , Reassortant Viruses/metabolism , Virion/metabolism , Animals , Chickens , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H5N1 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H7N9 Subtype/enzymology , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza in Birds/virology , Neuraminidase/genetics , Poultry Diseases/virology , Reassortant Viruses/enzymology , Reassortant Viruses/genetics , Recombination, Genetic , Virion/enzymology , Virion/genetics , Virion/pathogenicity , Virulence
16.
PLoS One ; 13(2): e0193623, 2018.
Article in English | MEDLINE | ID: mdl-29489903

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.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Neuraminidase/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Catalytic Domain , Chemistry Techniques, Synthetic , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/enzymology , Influenza A Virus, H9N2 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/enzymology , Molecular Docking Simulation , Neuraminidase/chemistry , Neuraminidase/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism
17.
J Biomol Struct Dyn ; 36(1): 233-242, 2018 01.
Article in English | MEDLINE | ID: mdl-28013578

ABSTRACT

A series of dimeric naphthoquinones containing natural 2-hydroxy-1-4-naphthoquinone moiety was designed, synthesized, and evaluated against neuraminidase of H5N1 virus. p-hydroxy derivatives showed higher inhibition when compared to p-halogenated compounds. Molecular docking studies conducted with H5N1 neuraminidase clearly demonstrated different binding modes of the most active compound onto the open and closed conformations of loop 150. The results thus provide not only evidences of a novel scaffold evaluated as inhibitor, but also a rational explanation involving molecular modeling and the role of loop 150 in the binding.


Subject(s)
Influenza A Virus, H5N1 Subtype/enzymology , Molecular Docking Simulation , Naphthoquinones/chemistry , Neuraminidase/chemistry , Binding Sites , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Molecular Structure , Naphthoquinones/metabolism , Naphthoquinones/pharmacology , Neuraminidase/antagonists & inhibitors , Neuraminidase/metabolism , Protein Domains
18.
Chem Biol Drug Des ; 91(1): 105-115, 2018 01.
Article in English | MEDLINE | ID: mdl-28646621

ABSTRACT

Neuraminidase, which plays a critical role in the influenza virus life cycle, is a target for new therapeutic agents. The study of structure-activity relationships revealed that the C-5 position amino group of oseltamivir was pointed to 150-cavity of the neuraminidase in group 1. This cavity is important for selectivity of inhibitors against N1 versus N2 NA. A serial of influenza neuraminidase inhibitors with the oseltamivir scaffold containing lipophilic side chains at the C-5 position have been synthesized and evaluated for their influenza neuraminidase inhibitory activity and selectivity. The results indicated that compound 13o (H5N1 IC50  = 0.1 ± 0.04 µm, H3N2 IC50  = 0.26 ± 0.18 µm) showed better inhibitory activity and selectivity against the group 1 neuraminidase. This study may provide a clue to design of better group 1 neuraminidase inhibitors.


Subject(s)
Enzyme Inhibitors/metabolism , Neuraminidase/antagonists & inhibitors , Oseltamivir/metabolism , Binding Sites , Catalytic Domain , Enzyme Inhibitors/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/enzymology , Inhibitory Concentration 50 , Molecular Docking Simulation , Neuraminidase/metabolism , Oseltamivir/chemistry , Structure-Activity Relationship
19.
Eur J Med Chem ; 141: 648-656, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29107426

ABSTRACT

Neuraminidase inhibitors can deter nascent viruses from infecting intact cells by preventing their release from host cells. Herein, a neuraminidase inhibitor 11b absent of basic moieties was discovered in the process of searching for inhibitors targeting 150 cavity. It exhibited potent inhibitions against wild-type neuraminidases from group 1 (H5N1 and H1N1) and group 2 (H7N9) subtypes with IC50 values similar to those of oseltamivir carboxylate. Moreover, 11b showed moderate inhibitions against mutant neuraminidases from H5N1-H274Y and H1N1-H274Y with IC50 values of 2075 nM and 1382 nM, which were inferior to those of oseltamivir carboxylate (6095 nM and 4071 nM). The results were not consistent with the recognized SARs that a basic moiety was an indispensable part of a potent inhibitor.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neuraminidase/antagonists & inhibitors , Oseltamivir/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/enzymology , Influenza A Virus, H7N9 Subtype/drug effects , Influenza A Virus, H7N9 Subtype/enzymology , Microbial Sensitivity Tests , Molecular Structure , Neuraminidase/metabolism , Oseltamivir/chemical synthesis , Oseltamivir/chemistry , Structure-Activity Relationship
20.
Antiviral Res ; 139: 41-48, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28012921

ABSTRACT

High morbidity and mortality associated with human cases of highly pathogenic avian influenza (HPAI) viruses, including H5N1 influenza virus, have been reported. The purpose of the present study was to evaluate the antiviral effects of peramivir against HPAI viruses. In neuraminidase (NA) inhibition and virus replication inhibition assays, peramivir showed strong inhibitory activity against H5N1, H7N1 and H7N7 HPAI viruses with sub-nanomolar activity in enzyme assays. In H5N1 viruses containing the NA H275Y mutation, the antiviral activity of peramivir against the variant was lower than that against the wild-type. Evaluation of the in vivo antiviral activity showed that a single intravenous treatment of peramivir (10 mg/kg) prevented lethality in mice infected with wild-type H5N1 virus and also following infection with H5N1 virus with the H275Y mutation after a 5 day administration of peramivir (30 mg/kg). Furthermore, mice injected with peramivir showed low viral titers and low levels of proinflammatory cytokines in the lungs. These results suggest that peramivir has therapeutic activity against HPAI viruses even if the virus harbors the NA H275Y mutation.


Subject(s)
Antiviral Agents/therapeutic use , Cyclopentanes/therapeutic use , Guanidines/therapeutic use , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/genetics , Neuraminidase/genetics , Orthomyxoviridae Infections/drug therapy , Acids, Carbocyclic , Animals , Antiviral Agents/administration & dosage , Cyclopentanes/administration & dosage , Cytokines/immunology , Disease Models, Animal , Guanidines/administration & dosage , Humans , Influenza A Virus, H5N1 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H7N1 Subtype/drug effects , Influenza A Virus, H7N1 Subtype/enzymology , Influenza A Virus, H7N7 Subtype/drug effects , Influenza A Virus, H7N7 Subtype/enzymology , Influenza, Human/drug therapy , Lung/immunology , Lung/virology , Mice , Mutation , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/virology , Viral Load/drug effects , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...