Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Nature ; 618(7965): 590-597, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37258672

ABSTRACT

Rapidly evolving influenza A viruses (IAVs) and influenza B viruses (IBVs) are major causes of recurrent lower respiratory tract infections. Current influenza vaccines elicit antibodies predominantly to the highly variable head region of haemagglutinin and their effectiveness is limited by viral drift1 and suboptimal immune responses2. Here we describe a neuraminidase-targeting monoclonal antibody, FNI9, that potently inhibits the enzymatic activity of all group 1 and group 2 IAVs, as well as Victoria/2/87-like, Yamagata/16/88-like and ancestral IBVs. FNI9 broadly neutralizes seasonal IAVs and IBVs, including the immune-evading H3N2 strains bearing an N-glycan at position 245, and shows synergistic activity when combined with anti-haemagglutinin stem-directed antibodies. Structural analysis reveals that D107 in the FNI9 heavy chain complementarity-determinant region 3 mimics the interaction of the sialic acid carboxyl group with the three highly conserved arginine residues (R118, R292 and R371) of the neuraminidase catalytic site. FNI9 demonstrates potent prophylactic activity against lethal IAV and IBV infections in mice. The unprecedented breadth and potency of the FNI9 monoclonal antibody supports its development for the prevention of influenza illness by seasonal and pandemic viruses.


Subject(s)
Antibodies, Viral , Antibody Specificity , Influenza A virus , Influenza B virus , Influenza Vaccines , Influenza, Human , Molecular Mimicry , Neuraminidase , Animals , Humans , Mice , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Antibodies, Viral/therapeutic use , Antibody Specificity/immunology , Arginine/chemistry , Catalytic Domain , Hemagglutinins, Viral/immunology , Influenza A virus/classification , Influenza A virus/enzymology , Influenza A virus/immunology , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H3N2 Subtype/immunology , Influenza B virus/classification , Influenza B virus/enzymology , Influenza B virus/immunology , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Influenza Vaccines/therapeutic use , Influenza, Human/immunology , Influenza, Human/prevention & control , Neuraminidase/antagonists & inhibitors , Neuraminidase/chemistry , Neuraminidase/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Seasons , Sialic Acids/chemistry
2.
J Med Virol ; 93(6): 3465-3472, 2021 06.
Article in English | MEDLINE | ID: mdl-32827314

ABSTRACT

Influenza A virus (IAV) causes great morbidity and mortality worldwide every year. However, there are only a limited number of drugs clinically available against IAV infection. Further, emergence of drug-resistant strains can render those drugs ineffective. Thus there is an unmet medical need to develop new anti-influenza agents. In this study, we show that punicalagin from plants possesses strong anti-influenza activity with a low micromolar IC50 value in tissue culture. Using a battery of bioassays such as single-cycle replication assay, neuraminidase (NA) inhibition assay, and virus yield reduction assay, we demonstrate that the primary mechanism of action (MOA) of punicalagin is the NA-mediated viral release. Moreover, punicalagin can inhibit replication of different strains of influenza A and B viruses, including oseltamivir-resistant virus (NA/H274Y), indicating that punicalagin is a broad spectrum antiviral against both IAV and IBV. Further, although punicalagin targets NA like oseltamivir, it has a different MOA. These results suggest that punicalagin is an influenza NA inhibitor that may be further developed as a novel antiviral against influenza viruses.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hydrolyzable Tannins/pharmacology , Influenza A virus/drug effects , Influenza B virus/drug effects , Neuraminidase/antagonists & inhibitors , Plant Extracts/pharmacology , Animals , Dogs , Influenza A virus/enzymology , Inhibitory Concentration 50 , Madin Darby Canine Kidney Cells , Virus Replication/drug effects
3.
Eur J Med Chem ; 208: 112754, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32883638

ABSTRACT

The biological effects of flavonoids on mammal cells are diverse, ranging from scavenging free radicals and anti-cancer activity to anti-influenza activity. Despite appreciable effort to understand the anti-influenza activity of flavonoids, there is no clear consensus about their precise mode-of-action at a cellular level. Here, we report the development and validation of a screening assay based on AlphaScreen technology and illustrate its application for determination of the inhibitory potency of a large set of polyols against PA N-terminal domain (PA-Nter) of influenza RNA-dependent RNA polymerase featuring endonuclease activity. The most potent inhibitors we identified were luteolin with an IC50 of 72 ± 2 nM and its 8-C-glucoside orientin with an IC50 of 43 ± 2 nM. Submicromolar inhibitors were also evaluated by an in vitro endonuclease activity assay using single-stranded DNA, and the results were in full agreement with data from the competitive AlphaScreen assay. Using X-ray crystallography, we analyzed structures of the PA-Nter in complex with luteolin at 2.0 Å resolution and quambalarine B at 2.5 Å resolution, which clearly revealed the binding pose of these polyols coordinated to two manganese ions in the endonuclease active site. Using two distinct assays along with the structural work, we have presumably identified and characterized the molecular mode-of-action of flavonoids in influenza-infected cells.


Subject(s)
Antiviral Agents/chemistry , Endonucleases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Flavonoids/chemistry , Influenza A virus/enzymology , Viral Proteins/antagonists & inhibitors , Antiviral Agents/metabolism , Crystallography, X-Ray , Drug Evaluation, Preclinical , Endonucleases/chemistry , Endonucleases/metabolism , Enzyme Assays/methods , Enzyme Inhibitors/metabolism , Flavonoids/metabolism , Microbial Sensitivity Tests , Molecular Structure , Protein Binding , Protein Domains , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Structure-Activity Relationship , Viral Proteins/chemistry , Viral Proteins/metabolism
4.
Comput Biol Chem ; 85: 107241, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32120300

ABSTRACT

The resistances of matrix protein 2 (M2) protein inhibitors and neuraminidase inhibitors for influenza virus have attracted much attention and there is an urgent need for new drug. The antiviral drugs that selectively act on RNA polymerase are less prone to resistance and possess fewer side effects on the patient. Therefore, there is increased interest in screening compounds that can inhibit influenza virus RNA polymerase. Three natural compounds were found by using molecular docking-based virtual screening, which could bind tightly within the polymerase acidic protein-polymerase basic protein 1 (PA-PB1) subunit of influenza virus polymerase. Firstly, their drug likeness properties were evaluated, which showed that the hepatotoxicity values of all the three compounds indicating they had less or no hepatotoxicity, and did not have the plasma protein biding (PPB) ability, the three compounds needed to be modified in some aspects, like bulky molecular size. The stability of the complexes of PA-hits was validated through molecular dynamics (MD) simulation, revealing compound 2 could form more stable complex with PA subunit. The torsional conformations of each rotatable bond of the ligands in PA subunit were also monitored, to investigate variation in the ligand properties during the simulation, compound 3 had fewer rotatable bonds, indicating that the molecule had stronger rigidity. The bar charts of protein-ligand contacts and contacts over the course of trajectory showed that four key residues (Glu623, Lys643, Asn703 and Trp706) of PA subunit that participated in hydrogen-bond, water bridge and hydrophobic interactions with the hit compounds. Finally, the binding free energy and contributed energies were calculated by using MM-GBSA method. Out of the three compounds, compound 1 showed the lowest total binding free energy. Among all the interactions, the contribution of the covalent binding and the van der Waals energy were more than other items, compound 1 formed more stable hydrogen bonds with the residues of PA subunit binding pocket. This study smoothed the path for the development of novel lead compounds with improved binding properties, high drug likeness, and low toxicity to humans for the treatment of influenza, which provided a good basis for further research on novel and effective influenza virus PA-PB1 interaction inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Biological Products/pharmacology , DNA-Directed RNA Polymerases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Influenza A virus/drug effects , Thermodynamics , Antiviral Agents/chemistry , Biological Products/chemistry , Computational Biology , DNA-Directed RNA Polymerases/metabolism , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Humans , Influenza A virus/enzymology , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure
5.
Viruses ; 11(9)2019 09 05.
Article in English | MEDLINE | ID: mdl-31491939

ABSTRACT

Influenza A virus infections cause significant morbidity and mortality, and novel antivirals are urgently needed. Influenza RNA-dependent RNA polymerase (RdRp) activity has been acknowledged as a promising target for novel antivirals. In this study, a phenotypic versus target-based screening strategy was established to identify the influenza A virus inhibitors targeting the virus RNA transcription/replication steps by sequentially using an RdRp-targeted screen and a replication-competent reporter virus-based approach using the same compounds. To demonstrate the utility of this approach, a pilot screen of a library of 891 compounds derived from natural products was carried out. Quality control analysis indicates that the primary screen was robust for identification of influenza A virus inhibitors targeting RdRp activity. Finally, two hit candidates were identified, and one was validated as a putative RdRp inhibitor. This strategy can greatly reduce the number of false positives and improve the accuracy and efficacy of primary screening, thereby providing a powerful tool for antiviral discovery.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/drug effects , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Cell Line , Drug Evaluation, Preclinical , Humans , Influenza A virus/enzymology , Influenza A virus/genetics , Influenza, Human/virology , Pilot Projects , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
6.
Biochem J ; 475(23): 3847-3860, 2018 12 10.
Article in English | MEDLINE | ID: mdl-30404922

ABSTRACT

Influenza neuraminidase is responsible for the escape of new viral particles from the infected cell surface. Several neuraminidase inhibitors are used clinically to treat patients or stockpiled for emergencies. However, the increasing development of viral resistance against approved inhibitors has underscored the need for the development of new antivirals effective against resistant influenza strains. A facile, sensitive, and inexpensive screening method would help achieve this goal. Recently, we described a multiwell plate-based DNA-linked inhibitor antibody assay (DIANA). This highly sensitive method can quantify femtomolar concentrations of enzymes. DIANA also has been applied to high-throughput enzyme inhibitor screening, allowing the evaluation of inhibition constants from a single inhibitor concentration. Here, we report the design, synthesis, and structural characterization of a tamiphosphor derivative linked to a reporter DNA oligonucleotide for the development of a DIANA-type assay to screen potential influenza neuraminidase inhibitors. The neuraminidase is first captured by an immobilized antibody, and the test compound competes for binding to the enzyme with the oligo-linked detection probe, which is then quantified by qPCR. We validated this novel assay by comparing it with the standard fluorometric assay and demonstrated its usefulness for sensitive neuraminidase detection as well as high-throughput screening of potential new neuraminidase inhibitors.


Subject(s)
DNA/chemistry , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Influenza A virus/drug effects , Oseltamivir/analogs & derivatives , Phosphorous Acids/chemistry , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Enzyme Inhibitors/chemistry , Humans , Influenza A virus/enzymology , Influenza A virus/physiology , Influenza, Human/drug therapy , Influenza, Human/enzymology , Influenza, Human/virology , Neuraminidase/antagonists & inhibitors , Neuraminidase/metabolism , Oseltamivir/chemistry , Reproducibility of Results , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism
7.
Sci Rep ; 8(1): 2280, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29396435

ABSTRACT

Currently, many strains of influenza A virus have developed resistance against anti-influenza drugs, and it is essential to find new chemicals to combat this virus. The influenza polymerase with three proteins, PA, PB1 and PB2, is a crucial component of the viral ribonucleoprotein (RNP) complex. Here, we report the identification of a hit compound 221 by surface plasmon resonance (SPR) direct binding screening on the C-terminal of PA (PAC). Compound 221 can subdue influenza RNP activities and attenuate influenza virus replication. Its analogs were subsequently investigated and twelve of them could attenuate RNP activities. One of the analogs, compound 312, impeded influenza A virus replication in Madin-Darby canine kidney cells with IC50 of 27.0 ± 16.8 µM. In vitro interaction assays showed that compound 312 bound directly to PAC with Kd of about 40 µM. Overall, the identification of novel PAC-targeting compounds provides new ground for drug design against influenza virus in the future.


Subject(s)
Antiviral Agents/isolation & purification , Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , Influenza A virus/enzymology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Surface Plasmon Resonance , Viral Proteins/antagonists & inhibitors , Animals , DNA-Directed RNA Polymerases/analysis , Dogs , Influenza A virus/drug effects , Influenza A virus/physiology , Inhibitory Concentration 50 , Madin Darby Canine Kidney Cells , Protein Binding , Virus Replication/drug effects
8.
Antiviral Res ; 144: 186-195, 2017 08.
Article in English | MEDLINE | ID: mdl-28629986

ABSTRACT

Influenza A virus is the major cause of epidemics and pandemics worldwide. In this study, virtual screening was used to identify compounds interacting with influenza A polymerase PB2 cap-binding domain (CBD). With a database of 21,351 small molecules, 28 candidate compounds were tested and one compound (225) was identified as hit compound. Compound 225 and three of its analogs (225D1, 426 and 426Br) were found to bind directly to PB2 CBD by surface plasmon resonance (SPR). The evaluation of compounds 426Br and 225 indicated that they could bind to PB2 CBD and inhibit influenza virus at low micromolar concentration. They were predicted to bind the cap binding site of the protein by molecular modeling and were confirmed by SPR assay using PB2 CBD mutants. These two compounds have novel scaffolds and could be further developed into lead compound for influenza virus inhibition.


Subject(s)
Antiviral Agents/isolation & purification , Drug Evaluation, Preclinical , Enzyme Inhibitors/isolation & purification , Influenza A virus/enzymology , Viral Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemistry , Dogs , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Microbial Sensitivity Tests , Protein Binding , Surface Plasmon Resonance
9.
J Virol ; 91(5)2017 03 01.
Article in English | MEDLINE | ID: mdl-28031371

ABSTRACT

Influenza virus RNA-dependent RNA polymerase consists of three viral protein subunits: PA, PB1, and PB2. Protein-protein interactions (PPIs) of these subunits play pivotal roles in assembling the functional polymerase complex, which is essential for the replication and transcription of influenza virus RNA. Here we developed a highly specific and robust bimolecular luminescence complementation (BiLC) reporter system to facilitate the investigation of influenza virus polymerase complex formation. Furthermore, by combining computational modeling and the BiLC reporter assay, we identified several novel small-molecule compounds that selectively inhibited PB1-PB2 interaction. Function of one such lead compound was confirmed by its activity in suppressing influenza virus replication. In addition, our studies also revealed that PA plays a critical role in enhancing interactions between PB1 and PB2, which could be important in targeting sites for anti-influenza intervention. Collectively, these findings not only aid the development of novel inhibitors targeting the formation of influenza virus polymerase complex but also present a new tool to investigate the exquisite mechanism of PPIs. IMPORTANCE Formation of the functional influenza virus polymerase involves complex protein-protein interactions (PPIs) of PA, PB1, and PB2 subunits. In this work, we developed a novel BiLC assay system which is sensitive and specific to quantify both strong and weak PPIs between influenza virus polymerase subunits. More importantly, by combining in silico modeling and our BiLC assay, we identified a small molecule that can suppress influenza virus replication by disrupting the polymerase assembly. Thus, we developed an innovative method to investigate PPIs of multisubunit complexes effectively and to identify new molecules inhibiting influenza virus polymerase assembly.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/enzymology , Viral Nonstructural Proteins/metabolism , A549 Cells , Animals , Dogs , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Influenza A virus/drug effects , Influenza, Human/drug therapy , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Protein Interaction Mapping , Protein Multimerization/drug effects
10.
Molecules ; 21(9)2016 Aug 26.
Article in English | MEDLINE | ID: mdl-27571059

ABSTRACT

Paeonia delavayi, an endemic species in southwestern China, has been widely used as a traditional remedy for cardiovascular, extravasated blood, stagnated blood and female diseases in traditional Chinese medicine (TCM). However, there are no reports on the anti-influenza virus activity of this species. Here, the anti-influenza virus activity of P. delavayi root extracts was first evaluated by an influenza virus neuraminidase (NA) inhibition assay. Meantime, constituents in the active extracts were identified using ultra-high performance liquid coupled with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS) and seven major identified constituents were used to further evaluate the NA inhibitory activity. The results showed that the ethyl acetate fraction (EA) and the ethanol fraction (E) of P. delavayi both presented strong NA inhibitory activity with IC50 values of 75.932 µg/mL and 83.550 µg/mL, respectively. Twenty-seven constituents were characterized in these two active extracts by UPLC-Q-TOF-MS analysis, and seven major identified constituents exhibited high activity against the influenza virus. Among them, Benzoylpaeoniflorin (IC50 = 143.701 µM) and pentagalloylglucose (IC50 = 62.671 µM) exhibited the highest activity against the influenza virus, even far stronger than oseltamivir acid (IC50 = 281.308 µM). This study indicated that P. delavayi was a strong NA inhibitor, but cell-based inhibition, anti-influenza virus activity in vivo and anti-influenza virus mechanism still need to be tested and explored.


Subject(s)
Antiviral Agents , Drugs, Chinese Herbal , Enzyme Inhibitors , Influenza A virus/enzymology , Neuraminidase/antagonists & inhibitors , Paeonia/chemistry , Viral Proteins/antagonists & inhibitors , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Neuraminidase/chemistry , Viral Proteins/chemistry
11.
Sci Rep ; 6: 27156, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27257160

ABSTRACT

Influenza virus neuraminidase (NA) is the primary target for influenza therapeutics. Severe complications are often related to secondary pneumonia caused by Streptococcus pneumoniae (pneumococci), which also express NAs. Recently, a NA-mediated lethal synergism between influenza A viruses and pneumococci was described. Therefore, dual inhibitors of both viral and bacterial NAs are expected to be advantageous for the treatment of influenza. We investigated the traditional Chinese herbal drug sang bái pí (mulberry root bark) as source for anti-infectives. Two prenylated flavonoid derivatives, sanggenon G (4) and sanggenol A (5) inhibited influenza A viral and pneumococcal NAs and, in contrast to the approved NA inhibitor oseltamivir, also planktonic growth and biofilm formation of pneumococci. Evaluation of 27 congeners of 5 revealed a correlation between the degree of prenylation and bioactivity. Abyssinone-V 4'-methyl ether (27) inhibited pneumococcal NA with IC50 = 2.18 µM, pneumococcal growth with MIC = 5.63 µM, and biofilm formation with MBIC = 4.21 µM, without harming lung epithelial cells. Compounds 5 and 27 also disrupt the synergism between influenza A virus and pneumococcal NA in vitro, hence functioning as dual-acting anti-infectives. The results warrant further studies on whether the observed disruption of this synergism is transferable to in vivo systems.


Subject(s)
Benzofurans/pharmacology , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Flavanones/pharmacology , Influenza A virus/drug effects , Neuraminidase/antagonists & inhibitors , Streptococcus pneumoniae/drug effects , Bacterial Proteins/antagonists & inhibitors , Benzofurans/chemistry , Biofilms/drug effects , Chromones/chemistry , Enzyme Inhibitors/chemistry , Flavanones/chemistry , Flavonoids/chemistry , Flavonoids/pharmacology , Influenza A virus/enzymology , Microbial Sensitivity Tests , Molecular Structure , Morus/chemistry , Plankton/drug effects , Plant Roots/chemistry , Prenylation , Streptococcus pneumoniae/enzymology , Streptococcus pneumoniae/physiology , Viral Proteins/antagonists & inhibitors
12.
Sci Rep ; 6: 22880, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26956222

ABSTRACT

The RNA-dependent RNA polymerase of influenza A virus comprises conserved and independently-folded subdomains with defined functionalities. The N-terminal domain of the PA subunit (PA(N)) harbors the endonuclease function so that it can serve as a desired target for drug discovery. To identify a class of anti-influenza inhibitors that impedes PA(N) endonuclease activity, a screening approach that integrated the fluorescence resonance energy transfer based endonuclease inhibitory assay with the DNA gel-based endonuclease inhibitory assay was conducted, followed by the evaluation of antiviral efficacies and potential cytotoxicity of the primary hits in vitro and in vivo. A small-molecule compound ANA-0 was identified as a potent inhibitor against the replication of multiple subtypes of influenza A virus, including H1N1, H3N2, H5N1, H7N7, H7N9 and H9N2, in cell cultures. Combinational treatment of zanamivir and ANA-0 exerted synergistic anti-influenza effect in vitro. Intranasal administration of ANA-0 protected mice from lethal challenge and reduced lung viral loads in H1N1 virus infected BALB/c mice. In summary, ANA-0 shows potential to be developed to novel anti-influenza agents.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/drug effects , Influenza A virus/enzymology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Virus Replication/drug effects , Administration, Intranasal , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/isolation & purification , Drug Evaluation, Preclinical , Drug Synergism , Female , Influenza A virus/physiology , Lung/virology , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Survival Analysis , Treatment Outcome , Viral Load
13.
Drug Discov Ther ; 10(2): 109-13, 2016.
Article in English | MEDLINE | ID: mdl-26902927

ABSTRACT

To find a novel influenza inhibitor targeting the endonuclease activity of influenza A virus polymerase acidic protein (PA), which is essential for the acquisition of primers for viral mRNA transcription, seven Kampo extracts were tested in vitro for their ability to inhibit endonuclease activity of the recombinant PA protein that was expressed and purified from Escherichia coli. The Kampo medicines Kakkonto, Shosaikoto, Saikokeishito, Keishito, Maobushisaishinto, and Maoto, but not Chikujountanto, inhibited PA endonuclease activity in a dose-dependent manner. Our results indicate that Kampo medicines are good sources providing a structural lead for optimization of an influenza endonuclease inhibitor.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Enzyme Inhibitors/pharmacology , Medicine, Kampo , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Dose-Response Relationship, Drug , Escherichia coli/metabolism , Influenza A virus/enzymology , Plasmids/genetics , RNA, Messenger/drug effects , RNA-Dependent RNA Polymerase/genetics , Recombinant Proteins/chemistry , Viral Proteins/genetics
14.
Assay Drug Dev Technol ; 13(8): 488-506, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26461433

ABSTRACT

Novel antiviral drugs, which are less prone to resistance development, are desirable alternatives to the currently approved drugs for the treatment of potentially serious influenza virus infections. The viral polymerase is highly conserved and serves as an attractive target for antiviral drugs since potent inhibitors would directly stop viral replication at an early stage. Recent structural studies on the functional domains of the heterotrimeric influenza polymerase, which comprises subunits PA, PB1, and PB2, opened the way to a structure-based approach for optimizing inhibitors of viral replication. These strategies, however, are limited by the use of isolated protein fragments instead of employing the entire ribonucleoprotein complex (RNP), which represents the functional form of the influenza polymerase in infected cells. In this study, we have established a screening assay for efficient and reliable analysis of potential influenza polymerase inhibitors of various molecular targets such as monoselective polymerase inhibitors targeting the endonuclease site, the cap-binding domain, and the polymerase active site, respectively. By utilizing whole viral RNPs and a radioactivity-free endpoint detection with the capability for efficient compound screening while offering high-content information on potential inhibitors to drive medicinal chemistry program in a reliable manner, this biochemical assay provides significant advantages over the currently available conventional assays. We propose that this assay can eventually be adapted for coinstantaneous analysis and subsequent optimization of two or more different chemical scaffold classes targeting multiple active sites within the polymerase complex, thus enabling the evaluation of drug combinations and characterization of molecules with dual functionality.


Subject(s)
Antiviral Agents/analysis , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/analysis , Influenza A virus/enzymology , Ribonucleoproteins/analysis , Antiviral Agents/pharmacology , DNA-Directed RNA Polymerases/genetics , Drug Evaluation, Preclinical/methods , Humans , Influenza A virus/drug effects , Ribonucleoproteins/genetics , Ribonucleoproteins/pharmacology , Virus Replication/drug effects , Virus Replication/physiology
15.
Bioorg Med Chem Lett ; 23(20): 5575-7, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24012120

ABSTRACT

The identification of a novel hit compound inhibitor of the protein-protein interaction between the influenza RNA-polymerase PA and PB1 subunits has been accomplished by means of high-throughput screening. A small family of structurally related molecules has been synthesized and biologically evaluated with most of the compounds showing micromolar potency of inhibition against viral replication.


Subject(s)
Antiviral Agents/toxicity , Benzoxazoles/chemistry , DNA-Directed RNA Polymerases/metabolism , Enzyme Inhibitors/chemical synthesis , Influenza A virus/drug effects , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Benzoxazoles/chemical synthesis , Benzoxazoles/toxicity , DNA-Directed RNA Polymerases/chemistry , Dogs , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Influenza A virus/enzymology , Madin Darby Canine Kidney Cells , Protein Interaction Domains and Motifs/drug effects , Protein Subunits/chemistry , Protein Subunits/metabolism , Structure-Activity Relationship
16.
Antivir Ther ; 18(5): 651-61, 2013.
Article in English | MEDLINE | ID: mdl-23111657

ABSTRACT

BACKGROUND: Seasonal interpandemic influenza causes >200,000 annual hospitalizations in the United States. Optimal antiviral treatment in hospitalized patients is not established. METHODS: During three interpandemic influenza seasons, 137 patients hospitalized with suspected acute influenza were randomized to 5-day treatment with intravenous peramivir 400 mg or 200 mg once daily or oral oseltamivir 75 mg twice daily. Time to clinical stability and quantitative changes in viral titres from nasopharyngeal specimens were primary and key secondary end points, respectively. RESULTS: Infection was confirmed in 122 patients with influenza A (H1N1), influenza A (H3N2) or influenza B. Median times (95% CI) to clinical stability were 37.0 h (22.0, 48.7) with peramivir 400 mg, 23.7 h (16.0, 38.9) with peramivir 200 mg and 28.1 h (22.0, 37.0) with oseltamivir (P=0.306). Patients (n=97) who were clinically unstable at enrolment had median times (95% CI) to clinical stability of 24.3 h (21.2, 47.5) with peramivir 400 mg, 31.0 h (17.2, 47.7) with peramivir 200 mg and 35.5 h (23.3, 37.9) with oseltamivir (P=0.541). Titres of influenza A viruses in nasopharyngeal specimens decreased similarly across treatments, but more rapid decreases in titres of influenza B occurred with peramivir treatment. There were no deaths among patients with confirmed influenza and the incidence of adverse events was low and generally similar among treatment groups. CONCLUSIONS: Treatment of acute seasonal influenza in hospitalized adults with either peramivir or oseltamivir resulted in generally similar clinical outcomes. Treatment with peramivir was generally safe and well tolerated and could be of benefit in this population.


Subject(s)
Antiviral Agents/therapeutic use , Cyclopentanes/therapeutic use , Guanidines/therapeutic use , Hospitalization , Influenza A virus , Influenza, Human/drug therapy , Oseltamivir/therapeutic use , Acids, Carbocyclic , Administration, Intravenous , Administration, Oral , Adult , Aged , Aged, 80 and over , Antiviral Agents/pharmacology , Cyclopentanes/pharmacology , Female , Guanidines/pharmacology , Humans , Influenza A virus/drug effects , Influenza A virus/enzymology , Influenza, Human/virology , Inhibitory Concentration 50 , Male , Microbial Sensitivity Tests , Middle Aged , Oseltamivir/pharmacology , Risk Factors , Seasons , Treatment Outcome , Viral Load
17.
Biol Pharm Bull ; 35(5): 786-90, 2012.
Article in English | MEDLINE | ID: mdl-22687418

ABSTRACT

In this study, twelve neuraminidase (NA) inhibitory compounds 1-12 were isolated from heartwood of Caesalpinia sappan on the basis of their biological activities against three types of viral NAs. Of isolated homoisoflavonoids, sappanone A (2) showed the most potent NAs inhibitory activities with IC(50) values of 0.7 µM [H1N1], 1.1 µM [H3N2], and 1.0 µM [H9N2], respectively, whereas saturated homoisoflavonoid (3) did not show significantly inhibition. This result revealed that α,ß-unsaturated carbonyl group in A-ring was the key requirements for viral NAs inhibitory activity. In our enzyme kinetic study, all NA inhibitors screened were found to be reversible noncompetitive types.


Subject(s)
Antiviral Agents/pharmacology , Caesalpinia/chemistry , Flavonoids/pharmacology , Influenza A virus/drug effects , Neuraminidase/antagonists & inhibitors , Plant Extracts/pharmacology , Viral Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Cell Line , Dogs , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Flavonoids/chemistry , Flavonoids/isolation & purification , 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, H9N2 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/enzymology , Influenza A virus/enzymology , Inhibitory Concentration 50 , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Phytotherapy , Plant Extracts/chemistry , Plant Extracts/therapeutic use , Wood
18.
J Asian Nat Prod Res ; 13(6): 575-80, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21623524

ABSTRACT

Three new phenolic compounds, eurycorymboside A (1), eurycorymboside B (6), and eurycorymbic acid (8), were isolated from the stem part of Eurycorymbus cavaleriei (Sapindaceae) along with five known phenolic compounds, glucosyringic acid (2), vanillic acid 4-O-ß-d-glucoside (3), koaburaside (4), tachioside (5), and 4-hydroxy-3,5-bis(3-methyl-2-butenyl)benzaldehyde (7). The structures were established on the basis of spectral analysis. The antioxidant activities of compounds 1-6 were evaluated by the 1,1-diphenyl-2-picrylhydrazyl-free radical scavenging assay. Compound 4 exhibited antioxidant activity with an IC(50) value of 9.0 µM. Compound 4 also showed weak inhibitory activity against influenza A neuraminidase.


Subject(s)
Antioxidants/isolation & purification , Antiviral Agents/isolation & purification , Drugs, Chinese Herbal/isolation & purification , Glycosides/isolation & purification , Phenols/isolation & purification , Sapindaceae/chemistry , Antioxidants/chemistry , Antioxidants/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Biphenyl Compounds/pharmacology , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Glycosides/chemistry , Glycosides/pharmacology , Influenza A virus/drug effects , Influenza A virus/enzymology , Neuraminidase/antagonists & inhibitors , Phenols/chemistry , Phenols/pharmacology , Picrates/pharmacology , Plant Stems/chemistry
19.
Yao Xue Xue Bao ; 44(2): 162-6, 2009 Feb.
Article in Chinese | MEDLINE | ID: mdl-19408687

ABSTRACT

Radix Isatidis (Banlangen in Chinese) is a traditional Chinese medicinal (TCM) herb, and is frequently used for treating influenza. However, the current quality control method for Radix Isatidis should be developed since it has little correlation to the pharmacodynamic action. In this paper, the in vitro inhibitory action of Radix Isatidis on neuraminidase (NA) was investigated by fluorometric assay with 4-methylumbelliferyl-D-N-acetylneuraminate (FL-MU-NANA) method. Based on the method, the experimental condition was optimized and a bioassay statistic method was established according to the reaction type and the regularity of "parallel lines of qualitative effect". Then the bioassay method of Radix Isatidis was established. This study indicated that Radix Isatidis had obvious in vitro inhibitory activity on NA with IC50 = (0.90 +/- 0.20) mg x mL(-1) (herb). The correlation between logarithmic dose and reaction rate showed an "S" shape--is quite similar to Tamiflu's reaction curve, which hinted that Radix Isatidis had the same inhibitory function on NA as Tamiflu. The established bioassay method of "parallel lines of qualitative effect" had a good reproducibility (RSD = 5.78%). The results of potency determination of Radix Isatidis were reliable (reliability test: deviation from straight line P > 0.05, deviation from parallel line P > 0.05) and well regular. As a conclusion, this bioassay method is suitable to control and evaluate the quality of Radix Isatidis.


Subject(s)
Antiviral Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Influenza A virus/enzymology , Isatis/chemistry , Neuraminidase/metabolism , Animals , Antiviral Agents/isolation & purification , Biological Assay , Cell Line , Dogs , Drugs, Chinese Herbal/isolation & purification , Inhibitory Concentration 50 , Kidney/cytology , Kidney/enzymology , Kidney/virology , Oseltamivir/pharmacology , Plant Roots/chemistry , Plants, Medicinal/chemistry , Quality Control , Reproducibility of Results , Structure-Activity Relationship
20.
Virol J ; 5: 109, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18822145

ABSTRACT

BACKGROUND: The emergence of influenza strains that are resistant to commonly used antivirals has highlighted the need to develop new compounds that target viral gene products or host mechanisms that are essential for effective virus replication. Existing assays to identify potential antiviral compounds often use high throughput screening assays that target specific viral replication steps. To broaden the search for antivirals, cell-based replication assays can be performed, but these are often labor intensive and have limited throughput. RESULTS: We have adapted a traditional virus neutralization assay to develop a practical, cell-based, high throughput screening assay. This assay uses viral neuraminidase (NA) as a read-out to quantify influenza replication, thereby offering an assay that is both rapid and sensitive. In addition to identification of inhibitors that target either viral or host factors, the assay allows simultaneous evaluation of drug toxicity. Antiviral activity was demonstrated for a number of known influenza inhibitors including amantadine that targets the M2 ion channel, zanamivir that targets NA, ribavirin that targets IMP dehydrogenase, and bis-indolyl maleimide that targets protein kinase A/C. Amantadine-resistant strains were identified by comparing IC50 with that of the wild-type virus. CONCLUSION: Antivirals with specificity for a broad range of targets are easily identified in an accelerated viral inhibition assay that uses NA as a read-out of replication. This assay is suitable for high throughput screening to identify potential antivirals or can be used to identify drug-resistant influenza strains.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Influenza A virus/drug effects , Influenza B virus/drug effects , Influenza, Human/drug therapy , Neuraminidase/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Animals , Chick Embryo , Drug Evaluation, Preclinical , Drug Resistance, Viral , Ducks , Humans , Influenza A virus/enzymology , Influenza B virus/enzymology , Influenza, Human/virology , Neuraminidase/genetics , Neuraminidase/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL