Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.235
Filter
1.
Sci Rep ; 14(1): 12876, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38834612

ABSTRACT

This study investigates quercetin complexes as potential synergistic agents against the important respiratory pathogen Streptococcus pneumoniae. Six quercetin complexes (QCX1-6) were synthesized by reacting quercetin with various metal salts and boronic acids and characterized using FTIR spectroscopy. Their antibacterial activity alone and in synergism with antibiotics was evaluated against S. pneumoniae ATCC 49619 using disc diffusion screening, broth microdilution MIC determination, and checkerboard assays. Complexes QCX-3 and QCX-4 demonstrated synergy when combined with levofloxacin via fractional inhibitory concentration indices ≤ 0.5 as confirmed by time-kill kinetics. Molecular docking elucidated interactions of these combinations with virulence enzymes sortase A and sialidase. A biofilm inhibition assay found the synergistic combinations more potently reduced biofilm formation versus monotherapy. Additionally, gene-gene interaction networks, biological activity predictions and in-silico toxicity profiling provided insights into potential mechanisms of action and safety.


Subject(s)
Anti-Bacterial Agents , Biofilms , Microbial Sensitivity Tests , Molecular Docking Simulation , Quercetin , Streptococcus pneumoniae , Streptococcus pneumoniae/drug effects , Quercetin/pharmacology , Quercetin/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Biofilms/drug effects , Drug Synergism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/antagonists & inhibitors , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/chemistry , Aminoacyltransferases/antagonists & inhibitors , Aminoacyltransferases/metabolism , Neuraminidase/antagonists & inhibitors , Neuraminidase/metabolism
2.
PLoS One ; 19(5): e0302865, 2024.
Article in English | MEDLINE | ID: mdl-38723016

ABSTRACT

Influenza A viruses (IAVs) continue to pose a huge threat to public health, and their prevention and treatment remain major international issues. Neuraminidase (NA) is the second most abundant surface glycoprotein on influenza viruses, and antibodies to NA have been shown to be effective against influenza infection. In this study, we generated a monoclonal antibody (mAb), named FNA1, directed toward N1 NAs. FNA1 reacted with H1N1 and H5N1 NA, but failed to react with the NA proteins of H3N2 and H7N9. In vitro, FNA1 displayed potent antiviral activity that mediated both NA inhibition (NI) and blocking of pseudovirus release. Moreover, residues 219, 254, 358, and 388 in the NA protein were critical for FNA1 binding to H1N1 NA. However, further validation is necessary to confirm whether FNA1 mAb is indeed a good inhibitor against NA for application against H1N1 and H5N1 viruses.


Subject(s)
Antibodies, Monoclonal , Influenza A Virus, H1N1 Subtype , Neuraminidase , Neuraminidase/immunology , Neuraminidase/metabolism , Neuraminidase/antagonists & inhibitors , Antibodies, Monoclonal/immunology , Influenza A Virus, H1N1 Subtype/immunology , Humans , Animals , Antibodies, Viral/immunology , Mice , Influenza A Virus, H5N1 Subtype/immunology , Mice, Inbred BALB C , Antiviral Agents/pharmacology , Viral Proteins/immunology , Viral Proteins/metabolism , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H7N9 Subtype/immunology
3.
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
4.
Bioorg Chem ; 147: 107353, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615475

ABSTRACT

Dialkyl/aryl aminophosphonates, 3a-g and 4a-e were synthesized using the LiClO4 catalyzed Kabachnic Fields-type reaction straightforwardly and efficiently. The synthesized phosphonates structures were characterized using elemental analyses, FT-IR, 1H NMR, 13C NMR, and MS spectroscopy. The new compounds were subjected to in-silico molecular docking simulations to evaluate their potential inhibition against Influenza A Neuraminidase and RNA-dependent RNA polymerase of human coronavirus 229E. Subsequently, the compounds were further tested in vitro using a cytopathic inhibition assay to assess their antiviral activity against both human Influenza (H1N1) and human coronavirus (HCoV-229E). Diphenyl ((2-(5-cyano-6-oxo-4-phenyl-1,6-dihydropyrimidin-2-yl) hydrazinyl) (furan-2-yl) methyl) phosphonate (3f) and diethyl ((2-(5-cyano-6-oxo-4-phenyl-1,6-dihydropyrimidin-2-yl) hydrazinyl) (1,5-dimethyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazol-4-yl) methyl) phosphonate (4e) were demonstrated direct inhibition activity against Influenza A Neuraminidase and RNA-dependent RNA polymerase. This was supported by their highly favorable binding energies in-silico, with top-ranked values of -12.5 kcal/mol and -14.2 kcal/mol for compound (3f), and -13.5 kcal/mol and -9.89 kcal/mol for compound (4e). Moreover, they also displayed notable antiviral efficacy in vitro against both viruses. These compounds demonstrated significant antiviral activity, as evidenced by selectivity indices (SI) of 101.7 and 51.8, respectively against H1N1, and 24.5 and 5.1 against HCoV-229E, respectively.


Subject(s)
Antiviral Agents , Coronavirus 229E, Human , Drug Design , Influenza A Virus, H1N1 Subtype , Molecular Docking Simulation , Organophosphonates , Pyrimidinones , Antiviral Agents/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Influenza A Virus, H1N1 Subtype/drug effects , Humans , Pyrimidinones/pharmacology , Pyrimidinones/chemical synthesis , Pyrimidinones/chemistry , Structure-Activity Relationship , Organophosphonates/pharmacology , Organophosphonates/chemistry , Organophosphonates/chemical synthesis , Coronavirus 229E, Human/drug effects , Molecular Structure , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Neuraminidase/antagonists & inhibitors , Neuraminidase/metabolism , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/metabolism
5.
J Virol Methods ; 323: 114838, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37914041

ABSTRACT

In influenza A virus-infected cells, newly synthesized viral neuraminidases (NAs) transiently localize at the host cell Golgi due to glycosylation, before their expression on the cell surface. It remains unproven whether Golgi-localized intracellular NAs exhibit sialidase activity. We have developed a sialidase imaging probe, [2-(benzothiazol-2-yl)-5-(non-1-yn-1-yl) phenyl]-α-D-N-acetylneuraminic acid (BTP9-Neu5Ac). This probe is designed to be cleaved by sialidase activity, resulting in the release of a hydrophobic fluorescent compound, 2-(benzothiazol-2-yl)-5-(non-1-yn-1-yl) phenol (BTP9). BTP9-Neu5Ac makes the location of sialidase activity visually detectable by the BTP9 fluorescence that results from the action of sialidase activity. In this study, we established a protocol to visualize the sialidase activity of intracellular NA at the Golgi of influenza A virus-infected cells using BTP9-Neu5Ac. Furthermore, we employed this fluorescence imaging protocol to elucidate the intracellular inhibition of laninamivir octanoate, an anti-influenza drug. At approximately 7 h after infection, newly synthesized viral NAs localized at the Golgi. Using our developed protocol, we successfully histochemically stained the sialidase activity of intracellular viral NAs localized at the Golgi. Importantly, we observed that laninamivir octanoate effectively inhibited the intracellular viral NA, in contrast to drugs like zanamivir or laninamivir. Our study establishes a visualization protocol for intracellular viral NA sialidase activity and visualizes the inhibitory effect of laninamivir octanoate on Golgi-localized intracellular viral NA in infected cells.


Subject(s)
Antiviral Agents , Enzyme Inhibitors , Influenza A virus , Neuraminidase , Viral Proteins , Humans , Antiviral Agents/pharmacology , Influenza A virus/drug effects , Influenza A virus/enzymology , Neuraminidase/analysis , Neuraminidase/antagonists & inhibitors , Optical Imaging/methods , Zanamivir/pharmacology , Viral Proteins/analysis , Viral Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology
6.
J Virol ; 97(10): e0060223, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37754760

ABSTRACT

IMPORTANCE: Influenza A viruses (IAVs) contain hemagglutinin (HA) proteins involved in sialoglycan receptor binding and neuraminidase (NA) proteins that cleave sialic acids. While the importance of the NA protein in virion egress is well established, its role in virus entry remains to be fully elucidated. NA activity is needed for the release of virions from mucus decoy receptors, but conflicting results have been reported on the importance of NA activity in virus entry in the absence of decoy receptors. We now show that inhibition of NA activity affects virus entry depending on the receptor-binding properties of HA and the receptor repertoire present on cells. Inhibition of entry by the presence of mucus correlated with the importance of NA activity for virus entry, with the strongest inhibition being observed when mucus and OsC were combined. These results shed light on the importance in virus entry of the NA protein, an important antiviral drug target.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus , Influenza A virus , Neuraminidase , Receptors, Virus , Viral Proteins , Virus Internalization , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A virus/enzymology , Influenza A virus/metabolism , Influenza, Human/enzymology , Influenza, Human/metabolism , Neuraminidase/antagonists & inhibitors , Neuraminidase/metabolism , Protein Binding , Receptors, Virus/metabolism , Substrate Specificity , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism , Cell Line , Mucus
7.
Curr Pharm Des ; 29(16): 1266-1273, 2023.
Article in English | MEDLINE | ID: mdl-37190805

ABSTRACT

BACKGROUND: Neuraminidase is a pathogenic protein of the avian influenza virus. Previous studies have shown that silibinin has the potential to inhibit neuraminidase activity. OBJECTIVE: This study aims to explore the interaction between silibinin and neuraminidase and the effect of silibinin on the structure and activity of neuraminidase. METHODS: In this study, two-dimensional fluorescence spectrum, three-dimensional fluorescence spectrometry, Uv-vis spectroscopy, and circular dichroism analysis were used. RESULTS: Silibinin alters the secondary structure of neuraminidase and inhibits the activity of neuraminidase. CONCLUSION: Silibinin can interact with neuraminidase and inhibit its activity.


Subject(s)
Antiviral Agents , Influenza A virus , Neuraminidase , Silybin , Animals , Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Influenza A virus/drug effects , Neuraminidase/antagonists & inhibitors , Silybin/pharmacology
8.
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
9.
Anal Sci ; 39(4): 547-556, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36617368

ABSTRACT

A new analytical method for rapid screening of influenza virus neuraminidase inhibitors was established. The method is based on the principle that, given a certain amount of neuraminidase, the sample and the neuraminidase act in the microplate for a period of time, and the active neuraminidase that is not inhibited by the sample can generate a fluorescence value at a specific wavelength after binding to the substrate, and the rate of inhibition of neuraminidase by the sample can be calculated based on the actual detected fluorescence value. This newly developed method was used to screen and evaluate the in vitro anti-neuraminidase activity of 39 high-purity compounds contained in three traditional Chinese herbal medicines, and finally 25 compounds with strong activity were obtained. The newly established neuraminidase inhibitor analytical method has these advantages of practicality, rapidity, high sensitivity and low cost, and has a good value for promotion and application. This article newly establishes a rapid, sensitive, simple and practical screening method for influenza virus neuraminidase inhibitors, which is a great complement to the existing methods and has a good promotion and application value.


Subject(s)
Influenza, Human , Neuraminidase , Orthomyxoviridae , Humans , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Fluorescence , Influenza, Human/drug therapy , Influenza, Human/metabolism , Neuraminidase/antagonists & inhibitors
10.
Antiviral Res ; 210: 105499, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36567025

ABSTRACT

Effective antivirals provide crucial benefits during the early phase of an influenza pandemic, when vaccines are still being developed and manufactured. Currently, two classes of viral protein-targeting drugs, neuraminidase inhibitors and polymerase inhibitors, are approved for influenza treatment and post-exposure prophylaxis. Resistance to both classes has been documented, highlighting the need to develop novel antiviral options that may include both viral and host-targeted inhibitors. Such efforts will form the basis of management of seasonal influenza infections and of strategic planning for future influenza pandemics. This review focuses on the two classes of approved antivirals, their drawbacks, and ongoing work to characterize novel agents or combination therapy approaches to address these shortcomings. The importance of these topics in the ongoing process of influenza pandemic planning is also discussed.


Subject(s)
Antiviral Agents , Influenza, Human , Humans , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Drug Resistance, Viral , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Influenza, Human/drug therapy , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Neuraminidase/antagonists & inhibitors , Oseltamivir/pharmacology , Pandemics/prevention & control
11.
Molecules ; 27(17)2022 Sep 05.
Article in English | MEDLINE | ID: mdl-36080488

ABSTRACT

Ganlanye (GLY), the leaf of Canarium album (Lour.) DC., is a traditional Chinese medicinal herb for warm disease treatment. We found that its aqueous extract could inhibit the influenza A virus. To find and characterize anti-influenza virus phytochemicals from GLY, we performed (1) bioassay-guided isolation, (2) a cell and animal assay, and (3) a mechanism study. Bioassay-guided isolation was used to identify the effective components. Influenza virus-infected MDCK cell and BALB/c mouse models were employed to evaluate the anti-influenza virus activities. A MUNANA assay was performed to find the NA inhibitory effect. As a result, urolithin M5 was obtained from the crude extract of GLY. It inhibited influenza virus activities in vitro and in vivo by suppressing the viral NA activity. In the MDCK cell model, urolithin M5 could inhibit an oseltamivir-resistant strain. In a PR8-infected mouse model, 200 mg/kg/d urolithin M5 protected 50% of mice from death and improved lung edema conditions. GLY was recorded as a major traditional herb for warm disease treatment. Our study identified GLY as a potent anti-influenza herb and showed urolithin M5 as the active component. We first report the in vivo activity of urolithin M5 and support the anti-influenza application of GLY.


Subject(s)
Antiviral Agents , Burseraceae , Influenza A Virus, H1N1 Subtype , Neuraminidase , Animals , Antiviral Agents/chemistry , Burseraceae/chemistry , Dogs , Influenza A Virus, H1N1 Subtype/drug effects , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Neuraminidase/antagonists & inhibitors , Oseltamivir/pharmacology , Plant Leaves/chemistry
12.
J Med Virol ; 94(9): 4406-4416, 2022 09.
Article in English | MEDLINE | ID: mdl-35585032

ABSTRACT

H1N1 influenza has brought serious threats to people's health and a high socioeconomic burden to society. Oseltamivir, a kind of neuraminidase (NA) inhibitor, is the second-generation specific drug that is broadly used currently. However, H1N1 influenza viruses have exhibited oseltamivir resistance in the past decades, which might be a hidden danger. To understand the frequency and distribution laws of oseltamivir-resistant viruses, we conducted a thorough and deep analysis of the available NA protein sequences of H1N1 influenza viruses worldwide from 1918 to 2020. The differences and similarities before and after 2009 were also considered since the dominant viruses changed in this period. Results showed that 3.76% of H1N1 viruses harbored oseltamivir resistance currently. Among various significative mutations, H274Y had the highest frequency of 3.30%, while the frequencies of the other mutations were far below this whether before or after 2009. The oseltamivir resistance was mainly found in three hosts, humans, swine, and avian. Different mutation sites could exhibit different distributions in each host. Our results showed that the resistance level reached a peak during the 2007-2008 influenza season and then quickly decreased in 2009. The resistance also displayed a global distribution. The densely populated countries usually had a high resistance level. However, frequent significative mutations were also found in some small countries. Our findings indicated the necessity of monitoring oseltamivir resistance around the world. The study could provide a unique perspective toward the cognition of viruses and facilitate the future study of both pandemic and drug development.


Subject(s)
Drug Resistance, Viral , Influenza A Virus, H1N1 Subtype , Influenza, Human , Oseltamivir , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Drug Resistance, Viral/genetics , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/drug therapy , Influenza, Human/epidemiology , Mutation , Neuraminidase/antagonists & inhibitors , Neuraminidase/genetics , Oseltamivir/pharmacology , Oseltamivir/therapeutic use , Swine , Viral Proteins/genetics
13.
Eur Heart J Cardiovasc Pharmacother ; 8(4): 392-401, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35294004

ABSTRACT

AIMS: The aim of this study was to investigate the effects of Neuraminidase inhibitors (NI) on COVID-19 in a retrospective study. METHODS AND RESULTS: The study included an overall COVID-19 patients (n = 3267) and a 1:1 propensity score-matched patients (n = 972). The levels of plasma N-acetylneuraminic acid and neuraminidase expression were further evaluated in a panel of hospitalized and 1-month post-infection recovered COVID-19 subjects. The mortality rate in the overall patients was 9.6% (313/3267) and 9.2% (89/972) in the propensity-score matched patients. The NI treatment lowered the mortality rate (5.7% vs. 10.3%) and the critically ill conversion rate (14.1% vs. 19.7%) compare to those in the non-NI group in the overall patients and evaluated in the propensity score-matched patients when applying the multivariate Cox model for adjusting imbalanced confounding factors. Furthermore, NI treatment was associated with attenuated cytokine storm levels and acute heart injury but not liver or kidney injuries. Further analysis in a small panel of patients found the levels of N-acetylneuraminic acid and neuraminidase (dominantly the NEU3 isoform) were elevated in the hospitalized COVID-19 subjects and recovered at the 1-month post-infection stage, suggesting increasing desialylation in COVID-19 patients. CONCLUSION: These results suggest that NI treatment is associated with decreased mortality in COVID-19 subjects, especially for those subjects with acute heart injury.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , COVID-19 , Neuraminidase , Antiviral Agents/therapeutic use , COVID-19/mortality , Cardiovascular Diseases/virology , Humans , N-Acetylneuraminic Acid , Neuraminidase/antagonists & inhibitors , Retrospective Studies
14.
Bioorg Med Chem ; 57: 116647, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35121400

ABSTRACT

Neuraminidase (NA) is an important target in the development of anti-influenza virus drugs. Compounds containing 1,3, 4-oxadiazole heterocycles have good biological activity and have been proved to have wide applications in antibacterial and antiviral drugs. In this paper, a series of novel 1, 3, 4-oxadiazole neuraminidase inhibitors (6a-6l) were designed and synthesized and their inhibitory activities of NA was tested in vitro. The results displayed that compound 6d exerts the best inhibitory activity (IC50 = 0.027 µM), which was obviously lower than that of oseltamivir carboxylate (OSC) (IC50 = 0.082 µM). Molecular docking analysis showed that the 1, 3, 4-oxadiazole heterocycle plays crucial part in compound 6d, and it can interact with the key arginine triad (Arg118, Arg292 and Arg 371) at the NA S1 site. The good efficacy of 6d may also be attributed to the extension of the substituted aniline ring to the 150-cavitiy. The theoretical and experimental results may provide reference for development of new anti-influenza drugs.


Subject(s)
Antiviral Agents/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae/drug effects , Oxadiazoles/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Neuraminidase/metabolism , Oxadiazoles/chemical synthesis , Oxadiazoles/chemistry , Structure-Activity Relationship
15.
Bioorg Med Chem Lett ; 61: 128590, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35108623

ABSTRACT

Neuraminidase (NA) is an ideal target for the development of anti-influenza drugs. In this paper, ZINC06057848 was screened out as a hit compound by docking-based virtual screening and molecular dynamics (MD) simulation. The modification and optimization of hit ZINC06057848 resulted in the discovery of a series of novel 1,3,4-triazole-containing NA inhibitors (5a-5j). Compound 5c exerts the best inhibitory activity (IC50 = 0.11 µM) against NA, which is comparable to the positive control oseltamivir carboxylate (OSC) (IC50 = 0.10 µM). Molecular docking analysis indicates that the good efficacy of inhibitor 5c may be attributed to the furan and triazole rings extending into 430-cavity and the ethylbenzene part occupying the active site. The results of this work may help in the development of new NA inhibitors.


Subject(s)
Acetamides/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Neuraminidase/antagonists & inhibitors , Triazoles/pharmacology , Acetamides/chemical synthesis , Acetamides/chemistry , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Neuraminidase/metabolism , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry
16.
J Med Chem ; 65(4): 3002-3025, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35170942

ABSTRACT

This Perspective describes the classification, structures, substrates, mechanisms of action, and implications of human neuraminidases (hNEUs) in various pathologies. Some inhibitors have been developed for each isoform, leading to more precise interactions with hNEUs. Although crystal structure data are available for NEU2, most of the findings are based on NEU1 inhibition, and limited information is available for other hNEUs. Therefore, the synthesis of new compounds would facilitate the enrichment of the arsenal of inhibitors to better understand the roles of hNEUs and their mechanisms of action. Nevertheless, due to the already known inhibitors of human neuraminidase enzymes, a structure-activity relationship is presented along with different approaches to inhibit these enzymes for the development of potent and selective inhibitors. Among the different emerging strategies, one is the inhibition of the dimerization of NEU1 or NEU3, and the second is the inhibition of certain receptors located close to hNEU.


Subject(s)
Enzyme Inhibitors/pharmacology , Neuraminidase/antagonists & inhibitors , Neuraminidase/chemistry , Amino Acid Sequence , Humans , Models, Molecular , Neuraminidase/classification , Stereoisomerism , Structure-Activity Relationship
17.
Molecules ; 27(3)2022 Jan 30.
Article in English | MEDLINE | ID: mdl-35164214

ABSTRACT

Neuraminidase (NA) is an enzyme that prevents virions from aggregating within the host cell and promotes cell-to-cell spread by cleaving glycosidic linkages to sialic acid. The best-known neuraminidase is the viral neuraminidase, which present in the influenza virus. Thus, the development of anti-influenza drugs that inhibit NA has emerged as an important and intriguing approach in the treatment of influenza. Garcinia atroviridis L. (GA) dried fruits (GAF) are used commercially as seasoning and in beverages. The main objective of this study was to identify a new potential neuraminidase inhibitor from GA. A bioassay-guided fractionation method was applied to obtain the bioactive compounds leading to the identification of garcinia acid and naringenin. In an enzyme inhibition study, garcinia acid demonstrated the highest activity when compared to naringenin. Garcinia acid had the highest activity, with an IC50 of 17.34-17.53 µg/mL or 91.22-92.21 µM against Clostridium perfringens-NA, and 56.71-57.85 µg/mL or 298.32-304.31 µM against H1N1-NA. Based on molecular docking results, garcinia acid interacted with the triad arginine residues (Arg118, Arg292, and Arg371) of the viral neuraminidase, implying that this compound has the potential to act as a NA enzyme inhibitor.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Fruit/chemistry , Garcinia/chemistry , Neuraminidase/antagonists & inhibitors , Plant Extracts/pharmacology , Plant Leaves/chemistry , Humans , Viral Proteins/antagonists & inhibitors
18.
Molecules ; 27(2)2022 Jan 12.
Article in English | MEDLINE | ID: mdl-35056785

ABSTRACT

Sub-Saharan Africa is profoundly challenged with African Animal Trypanosomiasis and the available trypanocides are faced with drawbacks, necessitating the search for novel agents. Herein, the chemotherapeutic potential of phloroglucinol on T. congolense infection and its inhibitory effects on the partially purified T. congolense sialidase and phospholipase A2 (PLA2) were investigated. Treatment with phloroglucinol for 14 days significantly (p < 0.05) suppressed T. congolense proliferation, increased animal survival and ameliorated anemia induced by the parasite. Using biochemical and histopathological analyses, phloroglucinol was found to prevent renal damages and splenomegaly, besides its protection against T. congolense-associated increase in free serum sialic acids in infected animals. Moreover, the compound inhibited bloodstream T. congolense sialidase via mixed inhibition pattern with inhibition binding constant (Ki) of 0.181 µM, but a very low uncompetitive inhibitory effects against PLA2 (Ki > 9000 µM) was recorded. Molecular docking studies revealed binding energies of -4.9 and -5.3 kcal/mol between phloroglucinol with modeled sialidase and PLA2 respectively, while a 50 ns molecular dynamics simulation using GROMACS revealed the sialidase-phloroglucinol complex to be more compact and stable with higher free binding energy (-67.84 ± 0.50 kJ/mol) than PLA2-phloroglucinol complex (-77.17 ± 0.52 kJ/mol), based on MM-PBSA analysis. The sialidase-phloroglucinol complex had a single hydrogen bond interaction with Ser453 while none was observed for the PLA2-phloroglucinol complex. In conclusion, phloroglucinol showed moderate trypanostatic activity with great potential in ameliorating some of the parasite-induced pathologies and its anti-anemic effects might be linked to inhibition of sialidase rather than PLA2.


Subject(s)
Phloroglucinol/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma congolense/drug effects , Trypanosomiasis, African/drug therapy , Anemia/complications , Anemia/drug therapy , Animals , Female , Kidney/drug effects , Kidney/parasitology , Kidney/pathology , Liver/drug effects , Liver/parasitology , Liver/pathology , Male , Molecular Docking Simulation , Molecular Dynamics Simulation , Neuraminidase/antagonists & inhibitors , Neuraminidase/chemistry , Organ Size/drug effects , Phloroglucinol/chemistry , Phloroglucinol/therapeutic use , Phospholipases A2/chemistry , Phospholipases A2/metabolism , Rats, Wistar , Survival Analysis , Trypanocidal Agents/chemistry , Trypanocidal Agents/therapeutic use , Trypanosoma congolense/parasitology , Trypanosomiasis, African/blood , Trypanosomiasis, African/complications , Trypanosomiasis, African/parasitology
19.
J Mol Model ; 28(2): 36, 2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35024968

ABSTRACT

The influenza virus is an important respiratory pathogen that causes many incidences of diseases and even death each year. One of the primary factors of this virus is the Neuraminidase surface protein, which causes the virus to leave the host cell and spread to new target cells. The main antiviral medication for influenza is designed as a protein inhibitor ligand that prevents further spread of the disease, and eventually relieves the emerged symptoms. The effectiveness of such inhibitory drugs is highly associated with their binding affinity. In this paper, the binding affinity of an herbal ligand of Capsaicin bound to Neuraminidase of the influenza virus is investigated using steered molecular dynamics (SMD) simulation. Since mutations of the virus directly impact the binding affinity of the inhibitory drugs, different mutations were generated by using Mutagenesis module. The rapid spread of infection during the avian influenza A/H5N1 epidemic has raised concerns about far more dangerous consequences if the virus becomes resistant to current drugs. Currently, oseltamivir (Tamiflu), zanamivir (Relenza), pramivir (Rapivab), and laninamivir (Inavir) are increasingly used to treat the flu. However, with the rapid evolution of the virus, some drug-resistant strains are emerging. Therefore, it is very important to seek alternative therapies and identify the roots of drug resistance. Obtained results demonstrated a reduced binding affinity for the applied mutations. This reduction in binding affinity will cause the virus mutation to become resistant to the drug, which will spread the disease and make it more difficult to treat. From a molecular prospect, this decrease in binding affinity is due to the loss of a number of effective bonds between the ligand and the receptor, which occurs with mutations of the wild-type (WT) species. The results of the present study can be used in the rational design of novel drugs that are compatible with specific mutations.


Subject(s)
Capsaicin/chemistry , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutant Proteins/chemistry , Neuraminidase/chemistry , Amino Acid Substitution , Binding Sites , Capsaicin/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Ligands , Molecular Conformation , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/genetics , Neuraminidase/antagonists & inhibitors , Neuraminidase/genetics , Protein Binding , Structure-Activity Relationship
20.
Chem Biol Interact ; 351: 109757, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34848165

ABSTRACT

New thiosemicarbazone-based zinc(II) complexes were synthesized to study their cytotoxicity on A375 malignant melanoma cells. The complexes containing salicylidene (Zn1a), 3-methoxy-salicylidene (Zn1b) or 4-methoxy-salicylidene (Zn1c) moiety were characterized by analytical and spectroscopic methods. Anticancer potential of the complexes was determined by MTT test and HUVEC endothelial cells line was used to comprehend the effect on normal cells. Zn1b with an IC50 of 13 µM was found to be highly cytotoxic against A375 cancer cells, more effective than cisplatin (IC50: 37 µM). Zn1a and Zn1c did not have a negative effect on cell viability in the normal cells and gave the impression that they are more advantageous than cisplatin in this respect. Further, the ability of Zn1a-c to inhibit neuraminidase enzyme and its role in cytotoxicity was discussed. The test revealed that the Zn1b with 3-methoxy substituent exhibited higher inhibition activity against the neuraminidase than the Zn1a and Zn1c as analogical to the cytotoxicity results. In neuraminidase inhibition, IC50 values of Zn1b and Zn1c were 14 and 66 µM, respectively. These concentrations were very close to the cytotoxicity concentrations for Zn1b and Zn1c. The findings may indicate the role of neuraminidase enzyme inhibition in cell death for Zn1b and Zn1c.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Coordination Complexes/pharmacology , Enzyme Inhibitors/pharmacology , Neuraminidase/antagonists & inhibitors , Thiosemicarbazones/pharmacology , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Human Umbilical Vein Endothelial Cells , Humans , Molecular Structure , Structure-Activity Relationship , Thiosemicarbazones/chemical synthesis , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...