Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem ; 28(1): 115130, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31753804

ABSTRACT

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


Subject(s)
Antiviral Agents/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Indoles/pharmacology , Influenza A Virus, H3N2 Subtype/drug effects , Influenza, Human/drug therapy , Spiro Compounds/pharmacology , Thiazolidines/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dogs , Dose-Response Relationship, Drug , Humans , Hydrogen-Ion Concentration , Indoles/chemistry , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Protein Refolding/drug effects , Spiro Compounds/chemistry , Structure-Activity Relationship , Thiazolidines/chemistry
2.
Biochim Biophys Acta Biomembr ; 1860(3): 784-791, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29229526

ABSTRACT

BACKGROUND AND OBJECTIVES: Recently influenza pandemic outbreaks were caused by emerging H5N1, H7N9 and H1N1 viruses. However, virucidal disinfectants are mainly unspecific and toxic. It is tactical to discover specific virucidal compounds. METHODS: The inhibitory potency was determined in H5N1 pseudovirus system; Interactions of compounds with hemagglutinin (HA) were detected with surface plasmon resonance (SPR) and further calculated with molecular docking. Virucidal effect was also estimated in influenza virus A/Puerto Rico/8/34(H1N1). Prevention efficacy was further estimated in mice model. RESULTS: Oligothiophene compound 4sc was potently virucidal against H5N1 pseudovirus with selective index>1169 (IC50=0.17±0.01µM). Pseudovirus assay revealed 4sc may interact with HA. However, HA inhibition test indicated 4sc did not interact with receptor pocket in HA. SPR detection revealed 4sc interacted directly with HA and its HA2 subunits. Molecular docking analysis revealed that 4sc interacted with the cavity of HA2 stem region and HA1-HA2 interface which consist of 7 residues: L22, K262, G472 and F1102 in HA2; M241, E251 and N271 in HA1. 4sc also potently and irreversibly neutralized PR8 (H1N1) virus, causing 105.06±0.26 fold decrease of virus titer after exposure for 10min. 4sc blocked PR8 transmission to MDCK cells. Amazingly, virucidal effect of 4sc was not significantly reduced even at 4°C. Furthermore, 4sc blocked viral transmission to mice. CONCLUSION: Oligothiophene compound 4sc is a novel selective virucide of influenza virus, which blocks entry by interfering viral hemagglutinin. Due to promising safety profile and stable virucidal effect at 4°C, 4sc may be useful in disinfecting H5N1 and H1N1 influenza virus.


Subject(s)
Antiviral Agents/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/drug effects , Animals , Antiviral Agents/chemical synthesis , Dogs , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Molecular Structure , Specific Pathogen-Free Organisms , Virus Internalization/drug effects
3.
Curr Opin Virol ; 24: 60-69, 2017 06.
Article in English | MEDLINE | ID: mdl-28527859

ABSTRACT

Monoclonal antibodies have revolutionized the treatment of several human diseases, including cancer, autoimmunity and inflammatory conditions and represent a new frontier for the treatment of infectious diseases. In the last decade, new methods have allowed the efficient interrogation of the human antibody repertoire from influenza immune individuals and the isolation of several monoclonal antibodies capable of dealing with the high variability of influenza viruses. Here, we will provide a comprehensive overview of the specificity, antiviral and immunological mechanisms of action and development into the clinic of broadly reactive monoclonal antibodies against influenza A and B viruses.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Influenza, Human/immunology , Influenza, Human/therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/chemistry , Antiviral Agents/immunology , Antiviral Agents/therapeutic use , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A virus/immunology , Influenza A virus/physiology , Influenza B virus/immunology , Influenza B virus/physiology , Influenza, Human/virology , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/therapy , Orthomyxoviridae Infections/virology
4.
Adv Exp Med Biol ; 966: 37-54, 2017.
Article in English | MEDLINE | ID: mdl-27966108

ABSTRACT

The influenza virus is a major health concern associated with an estimated 5000 to 30,000 deaths every year (Reed et al. 2015) and a significant economic impact with the development of treatments, vaccinations and research (Molinari et al. 2007). The entirety of the influenza genome is comprised of only eleven coding genes. An enormous degree of variation in non-conserved regions leads to significant challenges in the development of inclusive inhibitors for treatment. The fusion peptide domain of the influenza A hemagglutinin (HA) is a promising candidate for treatment since it is one of the most highly conserved sequences in the influenza genome (Heiny et al. 2007), and it is vital to the viral life cycle. Hemagglutinin is a class I viral fusion protein that catalyzes the membrane fusion process during cellular entry and infection. Impediment of the hemagglutinin's function, either through incomplete post-translational processing (Klenk et al. 1975; Lazarowitz and Choppin 1975) or through mutations (Cross et al. 2001), leads to non-infective virus particles. This review will investigate current research on the role of hemagglutinin in the virus life cycle, its structural biology and mechanism as well as the central role of the hemagglutinin fusion peptide (HAfp) to influenza membrane fusion and infection.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A virus/metabolism , Influenza, Human/virology , Viral Fusion Proteins/metabolism , Virus Internalization , Animals , Drug Design , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Host-Pathogen Interactions , Humans , Influenza A virus/drug effects , Influenza A virus/pathogenicity , Influenza, Human/drug therapy , Models, Molecular , Protein Domains , Structure-Activity Relationship , Viral Fusion Protein Inhibitors/chemistry , Viral Fusion Protein Inhibitors/therapeutic use , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/drug effects , Virulence , Virus Internalization/drug effects
5.
J Am Chem Soc ; 136(2): 783-8, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24377426

ABSTRACT

Influenza virus attaches itself to sialic acids on the surface of epithelial cells of the upper respiratory tract of the host using its own protein hemagglutinin. Species specificity of influenza virus is determined by the linkages of the sialic acids. Birds and humans have α2-3 and α2-6 linked sialic acids, respectively. Viral hemagglutinin is a homotrimeric receptor, and thus, tri- or oligovalent ligands should have a high binding affinity. We describe the in silico design, chemical synthesis and binding analysis of a trivalent glycopeptide mimetic. This compound binds to hemagglutinin H5 of avian influenza with a dissociation constant of K(D) = 446 nM and an inhibitory constant of K(I) = 15 µM. In silico modeling shows that the ligand should also bind to hemagglutinin H7 of the virus that causes the current influenza outbreak in China. The trivalent glycopeptide mimetic and analogues have the potential to block many different influenza viruses.


Subject(s)
Glycopeptides/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Drug Design , Glycopeptides/chemical synthesis , Glycopeptides/chemistry , Ligands , Models, Molecular , Molecular Structure , Structure-Activity Relationship
6.
FEBS J ; 280(22): 5829-40, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24034558

ABSTRACT

Curcumin (Cur) is a commonly used colouring agent and spice in food. Previously, we reported that Cur inhibits type A influenza virus (IAV) infection by interfering with viral haemagglutination (HA) activity. To search for a stable Cur analogue with potent anti-IAV activity and to investigate the structure contributing to its anti-IAV activity, a comparative analysis of structural and functional analogues of Cur, such as tetrahydrocurcumin (THC) and petasiphenol (Pet), was performed. The result of time-of-drug addition tests indicated that these curcuminoids were able to inhibit IAV production in cell cultures. Noticeably, Pet and THC inhibit IAV to a lesser extent than Cur, which is in line with their effect on reducing plaque formation when IAV was treated with Cur analogues before infection. Unexpectedly, both THC and Pet did not harbour any HA inhibitory effect. It should be noted that the structure of Pet and THC differs from Cur with respect to the number of double bonds present in the central seven-carbon chain, and structure modelling of Cur analogues indicates that the conformations of THC and Pet are distinct from that of Cur. Moreover, simulation docking of Cur with the HA structure revealed that Cur binds to the region constituting sialic acid anchoring residues, supporting the results obtained by the inhibition of HA activity assay. Collectively, structure-activity relationship analyses indicate that the presence of the double bonds in the central seven-carbon chain enhanced the Cur -dependent anti-IAV activity and also that Cur might interfere with IAV entry by its interaction with the receptor binding region of viral HA protein.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Curcumin/analogs & derivatives , Influenza A virus/drug effects , Animals , Binding Sites , Caffeic Acids/chemistry , Caffeic Acids/pharmacology , Curcumin/chemistry , Curcumin/pharmacology , Dogs , Glutathione/pharmacology , Glutathione Disulfide/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Humans , Madin Darby Canine Kidney Cells , Models, Molecular , Molecular Conformation , Protein Conformation , Structure-Activity Relationship , Viral Plaque Assay
7.
Expert Opin Ther Pat ; 23(11): 1517-24, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23967861

ABSTRACT

A series of compounds incorporating two aromatic heterocycles were prepared as inhibitors of influenza virus replication in the patent. Some of them presented potent activity against influenza virus in Madin-Darby canine kidney (MDCK) cells and in influenza therapeutic mouse model. These compounds in the patent were also defined to be pharmaceutically acceptable salts and pharmaceutical compositions that were claimed to be useful for treating influenza. In view of the threat of influenza pandemic, it is necessary to discover new anti-influenza drugs. Although there is a lack of essential biological data and the molecular mechanisms are not clear, these compounds with potent antiviral activity stand for a new type of anti-influenza agents and deserve further studies.


Subject(s)
Antiviral Agents/pharmacology , Influenza, Human/drug therapy , Orthomyxoviridae/drug effects , Animals , Cell Line , Drug Design , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Humans , Influenza Vaccines/therapeutic use , Influenza, Human/virology , Mice , Nucleic Acid Synthesis Inhibitors , Patents as Topic , Virus Replication/drug effects
8.
Antivir Chem Chemother ; 23(2): 59-65, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23425865

ABSTRACT

BACKGROUND: The purpose of this study was to develop a new class of influenza A virus haemagglutinin (HA) blockers by tethering thiosialoside molecules to metal nanoparticles and producing glycoclusters that enhance the affinity of HA binding by N-acetylneuraminic acid. METHODS: Oxygen of the glycoside bond of sialoside was replaced with sulfur to prevent hydrolytic digestion of the N-acetylneuraminic acid residue by viral neuraminidase. Two novel thiosialosides, α-2-S-[p-(N-levulinyl)aminophenyl]-5-N-acetylneuraminic acid (Neu5Ac-S-Lev) and α-2-S-[m-(N-levulinyl)aminobenzyl]-5-N-acetylneuraminic acid (Neu5Ac-S-CH2-Lev), were tethered onto the surface of metal nanoparticles via an aminooxy functionalized thiol linker in a glycoblotting reaction. Gold (Au) and silver (Ag) nanoparticles were coated simultaneously with 11-mercaptoundecyl phosphorylcholine to reduce non-specific adsorption of proteins. Phosphorylcholine self-assembled monolayer-coated metals displaying clustered Neu5Ac (Neu5Ac-PCSAM-Au and Neu5Ac-PCSAM-Ag) were subjected to haemagglutination inhibition (HI) assays using the influenza A virus strain A/PR/8/1934 (H1N1). RESULTS: Glyconanoparticles with thiosialosides had potent HI activities. In particular, Neu5Ac-PCSAM-Au with a diameter of 20 nm corresponding to 9.8 µM monosaccharide Neu5Ac was the most potent HA inhibitor. The versatility of this strategy was demonstrated by similar submicromolar HI activities of Neu5Ac-PCSAM-Ag with diameters of 50 nm and 150 nm. CONCLUSIONS: Glycosylated metal nanoparticles were designed and synthesized as potent influenza A virus HA blockers. This study may contribute to the acceleration of the discovery of a new class of nanoparticle anti-influenza drugs.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Influenza A virus/drug effects , Metal Nanoparticles , Sialic Acids/pharmacology , Animals , Cells, Cultured , Influenza A virus/metabolism , Sialic Acids/chemistry
9.
Proc Natl Acad Sci U S A ; 109(50): 20385-90, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23185023

ABSTRACT

Covalently conjugating multiple copies of the drug zanamivir (ZA; the active ingredient in Relenza) via a flexible linker to poly-l-glutamine (PGN) enhances the anti-influenza virus activity by orders of magnitude. In this study, we investigated the mechanisms of this phenomenon. Like ZA itself, the PGN-attached drug (PGN-ZA) binds specifically to viral neuraminidase and inhibits both its enzymatic activity and the release of newly synthesized virions from infected cells. Unlike monomeric ZA, however, PGN-ZA also synergistically inhibits early stages of influenza virus infection, thus contributing to the markedly increased antiviral potency. This inhibition is not caused by a direct virucidal effect, aggregation of viruses, or inhibition of viral attachment to target cells and the subsequent endocytosis; rather, it is a result of interference with intracellular trafficking of the endocytosed viruses and the subsequent virus-endosome fusion. These findings both rationalize the great anti-influenza potency of PGN-ZA and reveal that attaching ZA to a polymeric chain confers a unique mechanism of antiviral action potentially useful for minimizing drug resistance.


Subject(s)
Antiviral Agents/administration & dosage , Influenza A Virus, H1N1 Subtype/drug effects , Influenza, Human/drug therapy , Influenza, Human/virology , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Zanamivir/analogs & derivatives , Animals , Antiviral Agents/chemistry , Dogs , Drug Synergism , Endocytosis/drug effects , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H1N1 Subtype/ultrastructure , Madin Darby Canine Kidney Cells , Microscopy, Electron, Transmission , Neuraminidase/antagonists & inhibitors , Peptides/chemistry , Zanamivir/administration & dosage , Zanamivir/chemistry
10.
J Gen Virol ; 93(Pt 12): 2558-2563, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22933663

ABSTRACT

Airborne influenza virus infection of mice can be prevented by gaseous chlorine dioxide (ClO(2)). This study demonstrated that ClO(2) abolished the function of the haemagglutinin (HA) of influenza A virus (H1N1) in a concentration-, time- and temperature-dependent manner. The IC(50) during a 2 min reaction with ClO(2) at 25 °C was 13.7 µM, and the half-life time of HA with 100 µM ClO(2) at 25 °C was 19.5 s. Peptides generated from a tryptic digest of ClO(2)-treated virus were analysed by mass spectrometry. An HA fragment, (150)NLLWLTGK(157) was identified in which the tryptophan residue (W153) was 32 mass units greater than expected. The W153 residue of this peptide, which is derived from the central region of the receptor-binding site of HA, is highly conserved. It was shown that W153 was oxidized to N-formylkynurenine in ClO(2)-treated virus. It was concluded that the inactivation of influenza virus by ClO(2) is caused by oxidation of W153 in HA, thereby abolishing its receptor-binding ability.


Subject(s)
Chlorine Compounds/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Oxides/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Chickens , Conserved Sequence , Hemagglutination Tests , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , In Vitro Techniques , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/metabolism , Mice , Oxidation-Reduction , Receptors, Virus/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Tryptophan/chemistry
11.
Biochim Biophys Acta ; 1808(2): 547-53, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20599692

ABSTRACT

The recent outbreaks of avian flu in Southeast Asia and swine flu in Mexico City painfully exemplify the ability of the influenza virus to rapidly mutate and develop resistance to modern medicines. This review seeks to detail the molecular mechanism by which the influenza virus has obtained resistance to amino-adamantyls, one of only two classes of drugs that combat the flu. Amino-adamantyls target the viral M2 H(+) channel and have become largely ineffective due to mutations in the transmembrane domain of the protein. Herein we describe these resistance rendering mutations and the compounded effects they have upon the protein's function and resulting virus viability.


Subject(s)
Adamantane/pharmacology , Influenza, Human/drug therapy , Amantadine/pharmacology , Amino Acid Sequence , Drug Resistance, Viral/genetics , Genes, Viral , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A virus/chemistry , Influenza A virus/drug effects , Influenza A virus/genetics , Influenza A virus/pathogenicity , Influenza, Human/virology , Ion Channels/chemistry , Ion Channels/drug effects , Ion Channels/genetics , Models, Molecular , Molecular Sequence Data , Mutation , Protein Stability , Protein Structure, Tertiary , Rimantadine/pharmacology , Sequence Homology, Amino Acid , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/drug effects , Viral Matrix Proteins/genetics
12.
Bioorg Med Chem Lett ; 20(12): 3772-6, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20488701

ABSTRACT

We designed and synthesized novel trivalent anti-influenza reagents. Sialyllactose was located at the terminal of each valence which aimed to block each receptor-binding site of the hemagglutinin (HA) trimer on the surface of the virus. Structural analyses were carried out with a model which was constructed with a computer simulation. A previously reported cyclic glycopeptide blocker [Ohta, T.; Miura, N.; Fujitani, N.; Nakajima, F.; Niikura, K.; Sadamoto, R.; Guo, C.-T.; Suzuki, T.; Suzuki, Y.; Monde, K.; Nishimura, S.-I. Angew. Chem. Int. Ed., 2003, 42, 5186] bound to the HA in the model. The analyses suggest that the glutamine residue in the cyclic peptide bearing Neu5Acalpha2,3Galbeta1,4Glc trisaccharide via a linker interacts with the Gln189 in HA through hydrogen bonding. The present anti-influenza reagents likely interact with a glutamine residue included in the vicinity of Gln189. A plague reduction assay of the influenza virus, A/PR/8/1934 (H1N1), was performed in MDCK cells to evaluate for the synthesized compounds to inhibit viral replication. One of the compounds showed approximately 85% inhibition at the concentration of 400 microM at 4 degrees C.


Subject(s)
Antiviral Agents/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Peptides, Cyclic/pharmacology , Animals , Antiviral Agents/pharmacology , Cell Line , Dogs , Drug Design , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza, Human/drug therapy , Models, Molecular , Peptides, Cyclic/chemistry , Receptors, Virus , Viral Plaque Assay , Virus Replication/drug effects
13.
Bioorg Med Chem Lett ; 20(12): 3507-10, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20494579

ABSTRACT

A novel class of HA inhibitors (4a) was identified based on ligand similarity search of known HA inhibitors. Parallel synthesis and further structural modifications resulted in 1-phenyl-cyclopentanecarboxylic acid (4-cyano-phenyl)-methyl-amide 4t as a potent and selective inhibitor to phylogenetic H1 influenza viruses with an EC(50) of 98 nM against H1N1 A/Weiss/43 strain and over 1000-fold selectivity against host MDCK cells.


Subject(s)
Cycloparaffins/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Influenza A Virus, H1N1 Subtype/drug effects , Urea/pharmacology , Animals , Cell Line , Cycloparaffins/chemistry , Cycloparaffins/therapeutic use , Dogs , Drug Discovery , Humans , Influenza, Human/drug therapy , Structure-Activity Relationship , Urea/chemistry , Urea/therapeutic use
15.
Antivir Chem Chemother ; 14(2): 75-80, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12856918

ABSTRACT

The pavine alkaloid (-)-thalimonine (Thl), isolated from the Mongolian plant Thalictrum simplex inhibited markedly the reproduction of influenza virus A/Germany/27, str. Weybridge (H7N7) and A/Germany/34, str. Rostock (H7N1) in cell cultures of chicken embryo fibroblasts. In a number of assays at a non-toxic concentration range of 0.1-6.4 microM the alkaloid inhibited viral reproduction in a selective and specific way (selectivity index = 640, 106.6, respectively). Expression of viral glycoproteins haemagglutinin (HA), neuraminidase (NA) and nucleoprotein (NP) on the surface of infected cells, virus-induced cytopathic effect, infectious virus yields, HA production and virus-specific protein synthesis were all reduced. The inhibition was dose-related and depended on virus inoculum. The time of addition experiments indicated that viral reproduction was markedly inhibited when Thl was added at 4-5 h of infection. No inactivating effect on extracellular virus was found.


Subject(s)
Alkaloids/pharmacology , Antiviral Agents/pharmacology , Benzylisoquinolines/pharmacology , Influenza A virus/drug effects , Thalictrum/chemistry , Alkaloids/isolation & purification , Animals , Benzylisoquinolines/isolation & purification , Chick Embryo , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/virology , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A virus/metabolism , Neuraminidase/antagonists & inhibitors , Neuraminidase/metabolism , Nucleoproteins/drug effects , Nucleoproteins/metabolism , Plant Extracts/pharmacology , Time Factors
16.
Traffic ; 3(11): 822-32, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12383348

ABSTRACT

Golgi Microtubule-Associated Protein (GMAP)-210 is a peripheral coiled-coil protein associated with the cis-Golgi network that interacts with microtubule minus ends. GMAP-210 overexpression has previously been shown to perturb the microtubule network and to induce a dramatic enlargement and fragmentation of the Golgi apparatus (Infante C, Ramos-Morales F, Fedriani C, Bornens M, Rios RM. J Cell Biol 1999; 145: 83-98). We now report that overexpressing GMAP-210 blocks the anterograde transport of both a soluble form of alkaline phosphatase and the hemagglutinin protein of influenza virus, an integral membrane protein, between the endoplasmic reticulum and the cis/medial (mannosidase II-positive) Golgi compartment. Retrograde transport of the Shiga toxin B-subunit is also blocked between the Golgi apparatus and the endoplasmic reticulum. As a consequence, the B-subunit accumulates in compartments positive for GMAP-210. Ultrastructural analysis revealed that, under these conditions, the Golgi complex is totally disassembled and Golgi proteins as well as proteins of the intermediate compartment are found in vesicle clusters distributed throughout the cell. The role of GMAP-210 on membrane processes at the interface between the endoplasmic reticulum and the Golgi apparatus is discussed in the light of the property of this protein to bind CGN membranes and microtubules.


Subject(s)
Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Microtubule-Associated Proteins/metabolism , Protein Transport , Alkaline Phosphatase/metabolism , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/ultrastructure , Cytoskeletal Proteins , Drug Resistance , Endopeptidases/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/ultrastructure , Golgi Apparatus/drug effects , Golgi Apparatus/ultrastructure , HeLa Cells , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Kinetics , Nuclear Proteins , Protein Subunits/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Shiga Toxin/chemistry , Shiga Toxin/metabolism , Tumor Cells, Cultured
17.
Vopr Virusol ; 47(5): 21-5, 2002.
Article in Russian | MEDLINE | ID: mdl-12522964

ABSTRACT

The antineuraminidase activity of 5 inactivated split and subunit influenza vaccines (IIV) was studied in individuals aged above 65 years. Postvaccinal antibody titers were determined in the lectin test. All the vaccines were shown to have a high antigenicity, by providing high titers of neuraminidase antibodies in most vaccinated persons: The mean geometric titers (MGT) to influenza viruses A(H1N1) and A(H3N2), and B were 7.4-8.0, 8.5-9.2, and 6.8-8.5 log2, respectively. Neuraminidases showed a higher activity in Vaxigrip-vaccinated persons in terms of both the rate of MGT increases and seroconversion that was 78-84%. The parameters of the anti-neuraminidase (anti-NA) and anti-hemagglutinating (anti-HA) activities of IIV are summarized in the paper. There was a high coincidence of the results of both tests. At the same time it was shown that the postvaccinal humoral immune response might be directed only to one of the surface influenza virus proteins. The highest rate of seroconversions as to the surface antigens of all three influenza viruses was observed in Vaxigrip-vaccinated persons. The paper presents the results of comparison of the levels of MGT of antibodies to both viral surface proteins for all vaccinated with IIV, for those vaccinated who were infected during an epidemic season and a place group. The findings confirmed the value of higher titers of postvaccinal antibodies against influenza infection and illness.


Subject(s)
Influenza Vaccines/pharmacology , Neuraminidase/antagonists & inhibitors , Vaccines, Inactivated/pharmacology , Aged , Antibodies, Viral/biosynthesis , Antibodies, Viral/blood , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Humans , Neuraminidase/immunology
18.
Prog Drug Res ; Spec No: 35-77, 2001.
Article in English | MEDLINE | ID: mdl-11548210

ABSTRACT

Each year, influenza viruses are responsible for considerable illness, complications and mortality. An effective treatment will have a major impact on the severe personal and economic burden that this disease incurs. There are several points in the influenza life cycle that may be potentially inhibited. One critical point is the release of newly synthesized virions from the host cell surface. Viral neuraminidase (NA) cleaves the virus from host cell sialic acid residues allowing infection of other host cells. Rationally designed NA inhibitors that block the viral life cycle are now in the clinic and these molecules are effective and safe for the treatment of influenza. Compared with other anti-influenza agents the NA inhibitors are well tolerated, effective against all influenza types and there has been little evidence of the emergence of viral resistance. NA inhibitors provide an important new therapeutic weapon for the management of influenza infection.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae/drug effects , Animals , Drug Design , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Humans , IMP Dehydrogenase/antagonists & inhibitors , Influenza, Human/drug therapy , Viral Fusion Proteins/antagonists & inhibitors , Virus Replication/drug effects
19.
Arch Virol ; 144(5): 865-78, 1999.
Article in English | MEDLINE | ID: mdl-10416371

ABSTRACT

Stachyflin is a novel compound having H1 and H2 subtype-specific anti-influenza A virus activity. Stachyflin has no inhibition on H3 subtype influenza A or influenza B viruses. The susceptibility of the reassortant viruses between H1 and H3 subtype influenza A viruses to Stachyflin indicated that its target was virus-encoded hemagglutinin (HA). The results of the timing of Stachyflin addition against in vitro virus infection and virus-mediated hemolysis assay suggested that the drug inhibited the HA-mediated virus-cell fusion process. More directly, Stachyflin interfered with HA conformational change induced by low pH or heat treatment. The effect of Stachyflin could not be eliminated by washing of the Stachyflin-treated virus, which caused very specific virucidal effect. This is a remarkable property among small molecules which inhibit low-pH induced HA conformational change. From these findings, we concluded that the mechanism of Stachyflin action is to inhibit HA conformational change which is necessary for virus-cell membrane fusion. Stachyflin may be used as a tool for a study of molecular mechanism of low-pH induced HA conformational change, and offers potential as a pharmaceutical agent.


Subject(s)
Antiviral Agents/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinins, Viral/chemistry , Influenza A virus/drug effects , Influenza B virus/drug effects , Sesquiterpenes/pharmacology , Animals , Cell Line , Chickens , Cricetinae , Dogs , Giant Cells/drug effects , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Hemagglutinins, Viral/drug effects , Hemolysis/drug effects , Humans , Influenza A virus/classification , Influenza A virus/physiology , Influenza B virus/classification , Influenza B virus/physiology , Membrane Fusion/drug effects , Protein Conformation/drug effects
20.
J Cell Biol ; 140(4): 795-806, 1998 Feb 23.
Article in English | MEDLINE | ID: mdl-9472032

ABSTRACT

We have studied the biosynthesis and transport of the endogenous caveolins in MDCK cells. We show that in addition to homooligomers of caveolin-1, heterooligomeric complexes of caveolin-1 and -2 are formed in the ER. The oligomers become larger, increasingly detergent insoluble, and phosphorylated on caveolin-2 during transport to the cell surface. In the TGN caveolin-1/-2 heterooligomers are sorted into basolateral vesicles, whereas larger caveolin-1 homooligomers are targeted to the apical side. Caveolin-1 is present on both the apical and basolateral plasma membrane, whereas caveolin-2 is enriched on the basolateral surface where caveolae are present. This suggests that caveolin-1 and -2 heterooligomers are involved in caveolar biogenesis in the basolateral plasma membrane. Anti-caveolin-1 antibodies inhibit the apical delivery of influenza virus hemagglutinin without affecting basolateral transport of vesicular stomatitis virus G protein. Thus, we suggest that caveolin-1 homooligomers play a role in apical transport.


Subject(s)
Caveolins , Exocytosis/physiology , Membrane Proteins/genetics , Amino Acid Sequence , Animals , Antibodies/analysis , Antibodies/pharmacology , Base Sequence , Biological Transport , Caveolin 1 , Cell Line/chemistry , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Cloning, Molecular , Cytosol/chemistry , Cytosol/ultrastructure , DNA, Complementary/genetics , Exocytosis/genetics , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Membrane Proteins/chemistry , Membrane Proteins/physiology , Molecular Sequence Data , Polymers , Precipitin Tests , Proteins/chemistry , Sheep , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...