Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Nat Prod ; 64(7): 874-82, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11473415

ABSTRACT

From the screening of a microbial extract library, isocomplestatin (1), a new axial-chiral isomer of complestatin (2) which is a known rigid bicyclic hexapeptide, was identified as a potent natural product inhibitor of HIV-1 integrase, a unique enzyme responsible for viral replication. Isocomplestatin showed inhibitory activities (IC(50)) in coupled 3'-end processing/strand transfer (200 nM), strand transfer (4 microM), and HIV-1 replication (200 nM) in virus-infected cells. Attempted large-scale isolation of 1 by the literature method, used for the isolation of complestatin, led to lower yield and limited availability. We have developed several new, two-step, high-yielding absorption/elution methods of isolation based on reverse-phase chromatography at pH 8 that are applicable to scales from one gram to potential industrial quantities. We have also discovered and determined the structure of two new congeners of 1, namely, complestatins A (4) and B (5), with almost equal HIV-1 integrase activity. They differ from 1 at C2' and C3' of the tryptophan moiety (residue F). Selective acid hydrolysis of chloropeptin I (3), itself a known acid-catalyzed rearranged isomer of 1 and 2 (8'- vs 7'-substitution in tryptophan residue F, respectively), an isomer of complestatin, and isocomplestatin resulted in a number of fragments (6-10) with retention of most of the HIV-1 integrase activity. The structure-activity relationship as revealed by these compounds could possibly lead to the design of better inhibitors or understanding of the HIV-1 integrase target.


Subject(s)
Anti-HIV Agents/isolation & purification , Chlorophenols/isolation & purification , HIV Integrase Inhibitors/isolation & purification , HIV Integrase/metabolism , HIV-1/enzymology , Oligopeptides/isolation & purification , Peptides, Cyclic , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Chlorophenols/chemistry , Chlorophenols/pharmacology , Chromatography, High Pressure Liquid , Gas Chromatography-Mass Spectrometry , HIV Envelope Protein gp120/metabolism , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , HIV-1/metabolism , Leukocyte Common Antigens/metabolism , Magnetic Resonance Spectroscopy , Models, Chemical , Molecular Structure , Oligopeptides/chemistry , Oligopeptides/pharmacology , Stereoisomerism , Streptomyces/chemistry , Streptomyces/metabolism , Structure-Activity Relationship
2.
Proc Natl Acad Sci U S A ; 97(21): 11244-9, 2000 Oct 10.
Article in English | MEDLINE | ID: mdl-11016953

ABSTRACT

Diketo acids such as L-731,988 are potent inhibitors of HIV-1 integrase that inhibit integration and viral replication in cells. These compounds exhibit the unique ability to inhibit the strand transfer activity of integrase in the absence of an effect on 3' end processing. To understand the reasons for this distinct inhibitory profile, we developed a scintillation proximity assay that permits analysis of radiolabeled inhibitor binding and integrase function. High-affinity binding of L-731,988 is shown to require the assembly of a specific complex on the HIV-1 long terminal repeat. The interaction of L-731,988 with the complex and the efficacy of L-731, 988 in strand transfer can be abrogated by the interaction with target substrates, suggesting competition between the inhibitor and the target DNA. The L-731,988 binding site and that of the target substrate are thus distinct from that of the donor substrate and are defined by a conformation of integrase that is only adopted after assembly with the viral end. These results elucidate the basis for diketo acid inhibition of strand transfer and have implications for integrase-directed HIV-1 drug discovery efforts.


Subject(s)
Acetoacetates/pharmacology , DNA, Viral/metabolism , HIV Integrase Inhibitors/pharmacology , HIV Integrase/chemistry , HIV-1/enzymology , Pyrroles/pharmacology , Base Sequence , Catalysis , DNA Primers , Epitopes/metabolism , HIV Integrase/metabolism , HIV-1/genetics , Substrate Specificity
3.
Bioorg Med Chem Lett ; 10(3): 235-8, 2000 Feb 07.
Article in English | MEDLINE | ID: mdl-10698443

ABSTRACT

Integric acid (1), an acyl eremophilane sesquiterpenoid, was identified as an inhibitor of HIV-1 integrase, the enzyme responsible for provirus entry into the host cell nucleus and integration in to the host genome. Chemical and enzymatic modification of integric acid led to the preparation of several selective chemical derivatives of integric acid. Preparation, HIV-1 inhibitory activity, and the structure-activity relationship against coupled and strand transfer assays are described. It appears that most of the groups present in the natural product are required for inhibition of HIV-1 integrase strand transfer activity. In contrast, inhibition of 3' processing activity is less stringent suggesting distinct SAR for the two integrase reactions.


Subject(s)
Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , HIV-1/enzymology , Naphthalenes/chemistry , Naphthalenes/pharmacology , Amides/chemistry , Hydrolysis , Structure-Activity Relationship
4.
J Mol Biol ; 296(2): 521-33, 2000 Feb 18.
Article in English | MEDLINE | ID: mdl-10669606

ABSTRACT

The crystal structure of simian immunodeficiency virus (SIV) integrase that contains in a single polypeptide the core and the C-terminal deoxyoligonucleotide binding domain has been determined at 3 A resolution with an R-value of 0.203 in the space group P2(1)2(1)2(1). Four integrase core domains and one C-terminal domain are found to be well defined in the asymmetric unit. The segment extending from residues 114 to 121 assumes the same position as seen in the integrase core domain of avian sarcoma virus as well as human immunodeficiency virus type-1 (HIV-1) crystallized in the absence of sodium cacodylate. The flexible loop in the active site, composed of residues 141-151, remains incompletely defined, but the location of the essential Glu152 residue is unambiguous. The residues from 210-218 that link the core and C-terminal domains can be traced as an extension from the core with a short gap at residues 214-215. The C(alpha) folding of the C-terminal domain is similar to the solution structure of this domain from HIV-1 integrase. However, the dimeric form seen in the NMR structure cannot exist as related by the non-crystallographic symmetry in the SIV integrase crystal. The two flexible loops of the C-terminal domain, residues 228-236 and residues 244-249, are much better fixed in the crystal structure than in the NMR structure with the former in the immediate vicinity of the flexible loop of the core domain. The interface between the two domains encompasses a solvent-exclusion area of 1500 A(2). Residues from both domains purportedly involved in DNA binding are narrowly distributed on the same face of the molecule. They include Asp64, Asp116, Glu152 and Lys159 from the core and Arg231, Leu234, Arg262, Arg263 and Lys264 from the C-terminal domain. A model for DNA binding is proposed to bridge the two domains by tethering the 228-236 loop of the C-terminal domain and the flexible loop of the core.


Subject(s)
Catalytic Domain , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Integrases/chemistry , Integrases/metabolism , Simian Immunodeficiency Virus/enzymology , Amino Acid Sequence , Avian Sarcoma Viruses/enzymology , Binding Sites , Crystallization , Crystallography, X-Ray , DNA/genetics , DNA/metabolism , Dimerization , Glutamine/chemistry , Glutamine/metabolism , HIV Integrase/chemistry , Models, Molecular , Molecular Sequence Data , Protein Structure, Quaternary , Protein Structure, Tertiary , Solutions
5.
Science ; 287(5453): 646-50, 2000 Jan 28.
Article in English | MEDLINE | ID: mdl-10649997

ABSTRACT

Integrase is essential for human immunodeficiency virus-type 1 (HIV-1) replication; however, potent inhibition of the isolated enzyme in biochemical assays has not readily translated into antiviral activity in a manner consistent with inhibition of integration. In this report, we describe diketo acid inhibitors of HIV-1 integrase that manifest antiviral activity as a consequence of their effect on integration. The antiviral activity of these compounds is due exclusively to inhibition of one of the two catalytic functions of integrase, strand transfer.


Subject(s)
Acetoacetates/pharmacology , Anti-HIV Agents/pharmacology , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV-1/drug effects , Pyrroles/pharmacology , Virus Integration/drug effects , Acetoacetates/chemistry , Acetoacetates/metabolism , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Catalysis/drug effects , Coculture Techniques , DNA, Circular/biosynthesis , DNA, Circular/metabolism , DNA, Viral/biosynthesis , DNA, Viral/metabolism , Drug Resistance, Microbial , HIV Integrase/genetics , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/metabolism , HIV Long Terminal Repeat/drug effects , HIV-1/enzymology , HIV-1/genetics , HIV-1/physiology , Humans , Mutation , Pyrroles/chemistry , Pyrroles/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , T-Lymphocytes/virology , Transcription, Genetic , Tumor Cells, Cultured , Virus Replication/drug effects
7.
Acta Crystallogr D Biol Crystallogr ; 55(Pt 11): 1906-10, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10531491

ABSTRACT

The C-terminal two-thirds segment of integrase derived from the simian immunodeficiency virus has been cloned, expressed in Escherichia coli, and purified to greater than 95% homogeneity. The protein encompasses amino-acid residues 50-293 and contains a F185H substitution to enhance solubility. In dilute solutions at concentrations below 1 mg ml(-1), the enzyme is predominantly dimeric. At the higher concentrations (>10 mg ml(-1)) required to enable crystallization, the enzyme self-associates to form species with molecular weights greater than 200 kDa. Despite the apparent high aggregation in solution, the enzyme crystallizes from a 8%(v/v) polyethylene glycol (molecular weight 6000) solution in a form suitable for X-ray diffraction studies. The resulting single crystals belong to the space group P2(1)2(1)2(1), with unit-cell parameters a = 79.76, b = 99.98, c = 150.2 A, alpha = beta = gamma = 90 degrees and Z = 4. Under X-ray irradiation generated with a rotating-anode generator, the crystals diffract to 2.8 A resolution and allow collection of a native 3 A resolution diffraction data set.


Subject(s)
Integrases/chemistry , Simian Immunodeficiency Virus/enzymology , Cloning, Molecular , Crystallization , Dimerization , Escherichia coli , Integrases/genetics , Integrases/isolation & purification , Mutation , Polyethylene Glycols , Protein Conformation , Recombinant Proteins/isolation & purification , Software , Solubility , Ultracentrifugation , X-Ray Diffraction
8.
J Virol ; 73(10): 8104-11, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10482559

ABSTRACT

Full-site integration by recombinant wild-type and mutant simian immunodeficiency virus (SIV) integrase (IN) was investigated with linear retrovirus-like DNA (469 bp) as a donor substrate and circular DNA (2,867 bp) as a target substrate. Under optimized conditions, recombinant SIV IN produced donor-target products consistent with full-site (two donor ends) and half-site (one donor end) reactions with equivalent frequency. Restriction enzyme analysis of the 3.8-kbp full-site reaction products confirmed the concerted insertion of two termini from separate donors into a single target molecule. Donor ends carrying the viral U5 termini were preferred over U3 termini for producing both half-site and full-site products. Bacterial genetic selection was used to isolate individual donor-target recombinants, and the donor-target junctions of the cloned products were characterized by sequencing. Analysis of 149 recombinants demonstrated approximately 84% fidelity for the appropriate simian retrovirus 5-bp host duplication. As seen previously in similar reactions with human immunodeficiency virus type 1 (HIV-1) IN from lysed virions, approximately 8% of the donor-target recombinants generated with recombinant SIV IN incurred specific 17- to 18- or 27- to 29-bp deletions. The efficiency and fidelity of the full-site integration reaction mediated by the purified, recombinant SIV IN is comparable to that of HIV-1 IN from virions. These observations suggest that a purified recombinant lentivirus IN is itself sufficient to recapitulate the full-site integration process.


Subject(s)
Integrases/genetics , Simian Immunodeficiency Virus/physiology , Virus Integration/genetics , Escherichia coli , Humans , Recombinant Proteins/genetics , Substrate Specificity
9.
Antivir Chem Chemother ; 10(2): 63-70, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10335400

ABSTRACT

We have identified a series of novel inhibitors of human immunodeficiency virus type 1 (HIV-1) integrase by randomly screening natural product extracts using an in vitro biochemical assay designed to identify inhibitors of integrase-catalysed strand transfer. Equisetin recovered from the fungus Fusarium heterosporum and a novel enantiomeric homologue of equisetin from Phoma sp. were isolated as inhibitors of HIV-1 integrase in vitro. Two additional analogues, a novel decalin derivative, integric acid, and oteromycin were also discovered to be inhibitors of integrase. Equisetin and related compounds inhibit 3' end-processing and strand transfer as well as disintegration catalysed by either the full-length enzyme or the truncated integrase core domain (amino acids 50-212). These compounds also inhibit strand transfer reactions catalysed by stable complexes assembled in vitro and integration reactions catalysed by pre-integration complexes isolated from HIV-1-infected cells. The compounds described in this report are structurally novel and mechanistically distinct from many previously described inhibitors of HIV-1 integrase. These results demonstrate the utility of using an appropriately configured assay to identify compounds that are effective post-assembly and the potential of isolating novel integrase inhibitors from complex natural product extracts.


Subject(s)
Carboxylic Acids/isolation & purification , Fusarium/metabolism , HIV Integrase Inhibitors/isolation & purification , Naphthalenes/isolation & purification , Pyrroles/isolation & purification , Pyrrolidines/isolation & purification , Tetrahydronaphthalenes , Base Sequence , Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , Catalysis , DNA Primers , Fusarium/chemistry , HIV Integrase/chemistry , HIV Integrase/isolation & purification , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , Naphthalenes/chemistry , Naphthalenes/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , Pyrrolidinones/chemistry , Pyrrolidinones/isolation & purification , Pyrrolidinones/pharmacology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
10.
J Virol ; 71(9): 7005-11, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9261430

ABSTRACT

Previous in vitro analyses have shown that the human immunodeficiency virus type 1 (HIV-1) integrase uses either manganese or magnesium to assemble as a stable complex on the donor substrate and to catalyze strand transfer. We now demonstrate that subsequent to assembly, catalysis of both 3' end processing and strand transfer requires a divalent cation cofactor and that the divalent cation requirements for assembly and catalysis can be functionally distinguished based on the ability to utilize calcium and cobalt, respectively. The different divalent cation requirements manifest by these processes are exploited to uncouple assembly and catalysis, thus staging the reaction. Staged 3' end processing and strand transfer assays are then used in conjunction with exonuclease III protection analysis to investigate the effects of integrase inhibitors on each step in the reaction. Analysis of a series of related inhibitors demonstrates that these types of compounds affect assembly and not either catalytic process, therefore reconciling the apparent disparate results obtained for such inhibitors in assays using isolated preintegration complexes. These studies provide evidence for a distinct role of the divalent cation cofactor in assembly and catalysis and have implications for both the identification and characterization of integrase inhibitors.


Subject(s)
Cations, Divalent/metabolism , HIV Integrase/metabolism , HIV-1/enzymology , Virus Assembly , Calcium/metabolism , Catalysis , Cobalt/metabolism , HIV Integrase Inhibitors , HIV-1/physiology , Humans , Magnesium/metabolism , Manganese/metabolism , Substrate Specificity
11.
Drug Des Discov ; 15(1): 17-24, 1997 May.
Article in English | MEDLINE | ID: mdl-9332828

ABSTRACT

An essential step in the replication of retroviruses is the integration of a DNA copy of the viral genome into the genome of the host cell. Integration encompasses a series of ordered endonucleolytic and DNA strand transfer reactions catalyzed by the viral enzyme, integrase. The requirement for integrase activity in the propagation of HIV-1 in cell culture defines the enzyme as a potential target for chemotherapeutic intervention. We have therefore developed a non-radioisotopic microtiter plate assay which can be used to identify novel inhibitors of integrase from random chemical screens and for the bioassay driven isolation of inhibitors from natural products. This assay uncouples various steps in the reaction pathway and therefore can be exploited to characterize inhibitors. In this monograph we describe a series of modifications to the method which facilitate such mechanistic studies using as an example a series of previously described integrase inhibitors.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , Cations, Divalent/pharmacology , Cell-Free System , DNA, Viral/metabolism , Drug Evaluation , HIV-1/drug effects , HIV-1/physiology , Humans , Methods , Virus Assembly/drug effects , Virus Assembly/physiology , Virus Replication/drug effects
12.
J Virol ; 71(1): 807-11, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8985421

ABSTRACT

In vitro assay systems which use recombinant retroviral integrase (IN) and short DNA oligonucleotides fail to recapitulate the full-site integration reaction as it is known to occur in vivo. The relevance of using such circumscribed in vitro assays to define inhibitors of retroviral integration has not been formerly demonstrated. Therefore, we analyzed a series of structurally diverse inhibitors with respect to inhibition of both half-site and full-site strand transfer reactions with either recombinant or virion-produced IN. Half-site and full-site reactions catalyzed by avian myeloblastosis virus and human immunodeficiency virus type 1 (HIV-1) IN from virions are shown to be equivalently sensitive to inhibition by compounds which inhibit half-site reactions catalyzed by the recombinant HIV-1 IN. These studies therefore support the utility of using in vitro assays employing either recombinant or virion-derived IN to identify inhibitors of integration.


Subject(s)
Avian Myeloblastosis Virus/enzymology , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV-1/enzymology , Integrase Inhibitors/pharmacology , Integrases/metabolism , Virus Integration , Animals , HIV Integrase Inhibitors/chemistry , Humans , Integrase Inhibitors/chemistry , Magnesium , Manganese , Molecular Structure , Oligodeoxyribonucleotides , Recombinant Proteins/antagonists & inhibitors
13.
J Virol ; 70(3): 1424-32, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8627659

ABSTRACT

The integration of a DNA copy of the viral genome into the genome of the host cell is an essential step in the replication of all retroviruses. Integration requires two discrete biochemical reactions; specific processing of each viral long terminal repeat terminus or donor substrate, and a DNA strand transfer step wherein the processed donor substrate is joined to a nonspecific target DNA. Both reactions are catalyzed by a virally encoded enzyme, integrase. A microtiter assay for the strand transfer activity of human immunodeficiency virus type 1 integrase which uses an immobilized oligonucleotide as the donor substrate was previously published (D. J. Hazuda, J. C. Hastings, A. L. Wolfe, and E. A. Emini, Nucleic Acids Res. 22;1121-1122, 1994). We now describe a series of modifications to the method which facilitate study of both the nature and the dynamics of the interaction between integrase and the donor DNA. The enzyme which binds to the immobilized donor is shown to be sufficient to catalyze strand transfer with target DNA substrates added subsequent to assembly; in the absence of the target substrate, the complex was retained on the donor in an enzymatically competent state. Assembly required high concentrations of divalent cation, with optimal activity achieved at 25 mM MnCl2. In contrast, preassembled complexes catalyzed strand transfer equally efficiently in either 1 or 25 mM MnCl2, indicating mechanistically distinct functions for the divalent cation in assembly and catalysis, respectively. Prior incubation of the enzyme in 25 mM MnCl2 was shown to promote the multimerization of integrase in the absence of a DNA substrate and alleviate the requirement for high concentrations of divalent cation during assembly. The superphysiological requirement for MnCl2 may, therefore, reflect an insufficiency for functional self-assembly in vitro. Subunits were observed to exchange during the assembly reaction, suggesting that multimerization can occur either before or coincident with but not after donor binding. These studies both validate and illustrate the utility of this novel methodology and suggest that the approach may be generally useful in characterizing other details of this biochemical reaction.


Subject(s)
Chlorides/metabolism , DNA Nucleotidyltransferases/metabolism , HIV Long Terminal Repeat/physiology , HIV-1/enzymology , Manganese Compounds/metabolism , Base Sequence , Catalysis , Cations, Divalent , DNA/metabolism , DNA, Viral , Humans , Integrases , Molecular Sequence Data , Substrate Specificity , Time Factors , Virus Integration/physiology
14.
Acta Crystallogr D Biol Crystallogr ; 51(Pt 4): 496-503, 1995 Jul 01.
Article in English | MEDLINE | ID: mdl-15299836

ABSTRACT

Four novel antiviral WIN compounds, that contain a methyl tetrazole ring as well as isoxazole, pyridazine or acetylfuran rings, have had their structures determined in human rhinovirus serotype 14 at 2.9 A resolution. These compounds bind in the VP1 hydrophobic pocket, but are shifted significantly towards the pocket pore when compared to previously examined WIN compounds. A putative water network at the pocket pore is positioned to hydrogen bond with these four WIN compounds, and this network can account for potency differences seen in structurally similar WIN compounds.

15.
J Mol Biol ; 210(1): 91-111, 1989 Nov 05.
Article in English | MEDLINE | ID: mdl-2555523

ABSTRACT

The structure of human rhinovirus 1A (HRV1A) has been determined to 3.2 A resolution using phase refinement and extension by symmetry averaging starting with phases at 5 A resolution calculated from the known human rhinovirus 14 (HRV14) structure. The polypeptide backbone structures of HRV1A and HRV14 are similar, but the exposed surfaces are rather different. Differential charge distribution of amino acid residues in the "canyon", the putative receptor binding site, provides a possible explanation for the difference in minor versus major receptor group specificities, represented by HRV1A and HRV14, respectively. The hydrophobic pocket in VP1, into which antiviral compounds bind, is in an "open" conformation similar to that observed in drug-bound HRV14. Drug binding in HRV1A does not induce extensive conformational changes, in contrast to the case of HRV14.


Subject(s)
Rhinovirus/ultrastructure , Amino Acid Sequence , Antiviral Agents/metabolism , Capsid , Crystallization , Humans , Models, Biological , Molecular Sequence Data , Molecular Structure , Receptors, Drug/metabolism , Receptors, Drug/ultrastructure , Rhinovirus/immunology , Serotyping
16.
J Virol ; 63(5): 2002-7, 1989 May.
Article in English | MEDLINE | ID: mdl-2539499

ABSTRACT

A series of eight antiviral compounds complexed with human rhinovirus 14 (HRV-14) were previously shown to displace segments of polypeptide chains in the floor of the "canyon" by as much as 0.45 nm in C-alpha positions from the native conformation (J. Badger, I. Minor, M. J. Kremer, M. A. Oliveira, T. J. Smith, J. P. Griffith, D. M. A. Guerin, S. Krishnaswamy, M. Luo, M. G. Rossman, M. A. McKinlay, G. D. Diana, F. J. Dutko, M. Fancher, R. R. Rueckert, and B. A. Heinz, Proc. Natl. Acad. Sci. USA 85:3304-3308, 1988). Because the canyon is thought to serve as the viral receptor-binding site (M. G. Rossmann, E. Arnold, J. W. Erickson, E. A. Frankenberger, J. P. Griffith, H. J. Hecht, J. E. Johnson, G. Kamer, M. Luo, A. G. Mosser, R. R. Rueckert, B. Sherry, and G. Vriend, Nature [London] 317:145-153, 1985; M. G. Rossmann and R. R. Rueckert, Microbiol. Sci. 4:206-214, 1987), these compounds were assessed for their ability to block adsorption of HRV-14 to HeLa cell membrane receptors. In parallel experiments, the compounds were assessed directly for antiviral activity in an in vitro plaque reduction assay in intact HeLa cells. All eight compounds blocked the adsorption of 50% of HRV-14 at approximately the same concentration required to reduce the number of visible plaques by 50% (MIC). A structurally related compound which was inactive in the plaque reduction assay had no effect on HRV-14 binding. A drug-resistant mutant of HRV-14 (Leu-1188), which was less sensitive to the eight compounds in plaque reduction assays was similarly less sensitive in the adsorption assay. We propose that the conformational changes in the floor of the HRV-14 canyon induced by these compounds substantially decrease adsorption of the virion to its receptor. These results provide further evidence for the role of the HRV canyon in receptor binding.


Subject(s)
Antiviral Agents/pharmacology , Receptors, Virus/metabolism , Rhinovirus/ultrastructure , Virus Replication , Adsorption , Cell Membrane/metabolism , HeLa Cells , Humans , Models, Molecular , Rhinovirus/drug effects , Rhinovirus/metabolism , Structure-Activity Relationship , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...