Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Antibiot (Tokyo) ; 60(8): 524-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17827664

ABSTRACT

A novel fungal secondary metabolite, Sch 213766 was isolated from the fungal fermentation broth of Chaetomium globosum as the chemokine receptor CCR-5 inhibitor and shown to be the methyl ester of the previously described tetramic acid Sch 210972 on the basis of UV, MS and NMR spectral data analyses. Sch213766 exhibited an IC(50) value of 8.6 muM in the CCR-5 receptor in vitro binding assay.


Subject(s)
CCR5 Receptor Antagonists , Chaetomium/metabolism , Anti-HIV Agents/chemistry , Anti-HIV Agents/isolation & purification , Anti-HIV Agents/metabolism , Chaetomium/growth & development , Culture Media, Conditioned/metabolism , Fermentation , Methyl Ethers/chemistry , Methyl Ethers/isolation & purification , Methyl Ethers/metabolism , Methyl Ethers/pharmacology
2.
J Antimicrob Chemother ; 59(1): 51-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17151003

ABSTRACT

BACKGROUND: Current hepatitis C virus (HCV) therapies may cure approximately 60% of infections. They are often contraindicated or poorly tolerated, underscoring the need for safer and more effective drugs. A novel, alpha-ketoamide-derived, substrate-based inhibitor of the HCV serine protease (SCH446211) was developed. Compared with earlier reported inhibitors of similar chemical class, it has a P1'-P2' extension which provides extended interaction with the protease active site. The aim of this study was to evaluate the in vitro antiviral activity of SCH446211. METHODS: Binding constant of SCH446211 to HCV NS3 protease was measured with the chromogenic substrate in vitro cleavage assay. Cell-based activity of SCH446211 was evaluated in replicon cells, which are Huh-7 hepatoma cells stably transfected with a subgenomic HCV RNA as reported previously. After 72 h of incubation with SCH446211, viral transcription and protein expression were measured by real-time RT-PCR (TaqMan), quantitative in situ hybridization, immunoblot and indirect immunofluorescence. RESULTS: The binding constant of SCH446211 to HCV NS3 protease was 3.8 +/- 0.4 nM. HCV replication and protein expression were inhibited by SCH446211 in replicon cells as consistently shown by four techniques. In particular, based on quantitative real-time RT-PCR measurements, the IC50 and IC90 of SCH446211 were estimated to be 40 +/- 20 and 100 +/- 20 nM (n = 17), respectively. Long-term culture of replicon cells with SCH446211 reduced replicon RNA to <0.1 copy per cell. SCH446211 did not show cellular toxicity at concentrations up to 50 microM. CONCLUSIONS: SCH446211 is a potent inhibitor of HCV protease in vitro. Its extended interaction with the HCV NS3 protease active site is associated with potent in vitro antiviral activity. This observation is potentially a useful guide for development of future potent inhibitors against HCV NS3 protease.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Oligopeptides/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Hepacivirus/genetics , RNA, Viral/analysis , Replicon
3.
J Med Chem ; 49(20): 6074-86, 2006 Oct 05.
Article in English | MEDLINE | ID: mdl-17004721

ABSTRACT

Hepatitis C virus (HCV) infection is the major cause of chronic liver disease, leading to cirrhosis and hepatocellular carcinoma, which affects more than 170 million people worldwide. Currently the only therapeutic regimens are subcutaneous interferon-alpha or polyethylene glycol (PEG)-interferon-alpha alone or in combination with oral ribavirin. Although combination therapy is reasonably successful with the majority of genotypes, its efficacy against the predominant genotype (genotype 1) is moderate at best, with only about 40% of the patients showing sustained virological response. Herein, the SAR leading to the discovery of 70 (SCH 503034), a novel, potent, selective, orally bioavailable NS3 protease inhibitor that has been advanced to clinical trials in human beings for the treatment of hepatitis C viral infections is described. X-ray structure of inhibitor 70 complexed with the NS3 protease and biological data are also discussed.


Subject(s)
Antiviral Agents/chemical synthesis , Hepacivirus/enzymology , Proline/analogs & derivatives , Viral Nonstructural Proteins/antagonists & inhibitors , Administration, Oral , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Area Under Curve , Binding Sites , Biological Availability , Crystallography, X-Ray , Dogs , Haplorhini , Molecular Structure , Proline/chemical synthesis , Proline/chemistry , Proline/pharmacokinetics , Rats , Structure-Activity Relationship , Tissue Distribution , Viral Nonstructural Proteins/chemistry
4.
J Nat Prod ; 69(7): 1025-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16872138

ABSTRACT

Two novel chemokine receptor CCR-5 inhibitors, Sch 210971 (1) and Sch 210972 (2), were isolated from the fungal fermentation broth of Chaetomium globosum by normal- and reversed-phase HPLC purifications. The structure determination of 1 and 2 was accomplished on the basis of UV, MS, and NMR spectral data analyses including COSY, NOESY, HMQC, and HMBC experiments. The structure and relative configuration of 2 were determined unequivocally by X-ray crystallographic analysis. The major component 2 demonstrated a potent inhibitory activity of IC(50) = 79 nM in the CCR-5 receptor in vitro binding assay.


Subject(s)
Anti-HIV Agents/pharmacology , CCR5 Receptor Antagonists , Chaetomium/chemistry , Heterocyclic Compounds, 4 or More Rings/pharmacology , Anti-HIV Agents/chemistry , Anti-HIV Agents/isolation & purification , Arizona , Crystallography, X-Ray , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/isolation & purification , Humans , Inhibitory Concentration 50 , Molecular Structure , Plant Leaves/chemistry , Spectrometry, Mass, Electrospray Ionization
5.
J Med Chem ; 49(9): 2750-7, 2006 May 04.
Article in English | MEDLINE | ID: mdl-16640336

ABSTRACT

Introduction of various modified prolines at P(2) and optimization of the P(1) side chain led to the discovery of SCH6 (24, Table 2), a potent ketoamide inhibitor of the HCV NS3 serine protease. In addition to excellent enzyme potency (K(i)*= 3.8 nM), 24 was also found to be a potent inhibitor of HCV subgenomic RNA replication with IC(50) and IC(90) of 40 and 100 nM, respectively. Recently, antiviral activity of 24 was demonstrated with inhibition of the full-length genotype 2a HCV genome. In addition, 24 was found to restore the responsiveness of the interferon regulatory factor 3 (IRF-3) in cells containing HCV RNA replicons.


Subject(s)
Amides/chemistry , Amides/pharmacology , Genome, Viral/genetics , Hepacivirus/drug effects , Oligopeptides/chemistry , Oligopeptides/pharmacology , Serine Endopeptidases/metabolism , Animals , Haplorhini , Hepacivirus/enzymology , Hepacivirus/genetics , Models, Molecular , Molecular Structure , RNA, Viral/genetics , Rats , Serine Endopeptidases/chemistry , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism
6.
Virology ; 349(1): 41-54, 2006 May 25.
Article in English | MEDLINE | ID: mdl-16494916

ABSTRACT

The CC-chemokine receptor 5 (CCR5) is the major coreceptor for macrophage-tropic (R5) HIV-1 strains. Several small molecule inhibitors of CCR5 that block chemokine binding and HIV-1 entry are being evaluated as drug candidates. Here we define how CCR5 antagonists TAK-779, AD101 (SCH-350581) and SCH-C (SCH-351125), which inhibit HIV-1 entry, interact with CCR5. Using a mutagenesis approach in combination with a viral entry assay to provide a direct functional read out, we tested predictions based on a homology model of CCR5 and analyzed the functions of more than 30 amino acid residues. We find that a key set of aromatic and aliphatic residues serves as a hydrophobic core for the ligand binding pocket, while E283 is critical for high affinity interaction, most likely by acting as the counterion for a positively charged nitrogen atom common to all three inhibitors. These results provide a structural basis for understanding how specific antagonists interact with CCR5, and may be useful for the rational design of new, improved CCR5 ligands.


Subject(s)
HIV Fusion Inhibitors/metabolism , Receptors, CCR5/metabolism , Amides/metabolism , Binding Sites/genetics , Cell Line , Cyclic N-Oxides/metabolism , HIV-1/growth & development , Humans , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , Oximes , Piperidines/metabolism , Protein Structure, Secondary , Pyridines/metabolism , Quaternary Ammonium Compounds/metabolism , Receptors, CCR5/genetics
8.
J Interferon Cytokine Res ; 26(1): 40-52, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16426147

ABSTRACT

Most type I interferons (IFNs) are expressed by the majority of cell types in response to viral infection. In contrast, IFN-kappa has been reported to have a cellular distribution limited to keratinocytes and certain lymphoid cell populations. Recombinant expressed IFN-kappa has been shown previously to possess weak antiviral activity when directly compared with IFN-beta. In order to expand on the antiviral potential of IFN-kappa, we transiently transfected human cell lines to circumvent the need to purify recombinant proteins and to avoid the possible loss of biologic activity by the purification process. We evaluated the transcriptional signaling and antiviral activity of IFN-kappa in parallel with IFN-alpha2b with mammalian expression vectors to express each protein transiently. Both IFN-kappa and IFN-alpha2b exhibited comparable transcriptional and antiviral activities. However, in contrast to IFN-alpha2b transcriptional signaling and antiviral activity, IFN-kappa activity was not detectable in conditioned cell culture medium. Subsequent experiments revealed there was a direct relationship between IFN-kappa-expressing cells and antiviral activity. These results were confirmed in immunocytochemical studies. Furthermore, IFN-kappa exhibited cell-associated antiviral activity against a hepatitis C virus (HCV) replicon cell line. This novel IFN signaling strategy may represent an important distinct and divergent mechanism for limiting viral infections.


Subject(s)
Antiviral Agents/immunology , Interferon Type I/immunology , Animals , Chickens , Culture Media, Conditioned , HeLa Cells , Humans , Interferon Type I/genetics , Interferon alpha-2 , Interferon-alpha/genetics , Interferon-alpha/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Signal Transduction/physiology , Transcription, Genetic , Transfection
9.
Antimicrob Agents Chemother ; 49(12): 4911-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16304152

ABSTRACT

Inhibiting human immunodeficiency virus type 1 (HIV-1) infection by blocking the host cell coreceptors CCR5 and CXCR4 is an emerging strategy for antiretroviral therapy. Currently, several novel coreceptor inhibitors are being developed in the clinic, and early results have proven promising. In this report, we describe a novel CCR5 antagonist, vicriviroc (formerly SCH-D or SCH 417690), with improved antiviral activity and pharmacokinetic properties compared to those of SCH-C, a previously described CCR5 antagonist. Like SCH-C, vicriviroc binds specifically to the CCR5 receptor and prevents infection of target cells by CCR5-tropic HIV-1 isolates. In antiviral assays, vicriviroc showed potent, broad-spectrum activity against genetically diverse and drug-resistant HIV-1 isolates and was consistently more active than SCH-C in inhibiting viral replication. This compound demonstrated synergistic anti-HIV activity in combination with drugs from all other classes of approved antiretrovirals. Competition binding assays revealed that vicriviroc binds with higher affinity to CCR5 than SCH-C. Functional assays, including inhibition of calcium flux, guanosine 5'-[35S]triphosphate exchange, and chemotaxis, confirmed that vicriviroc acts as a receptor antagonist by inhibiting signaling of CCR5 by chemokines. Finally, vicriviroc demonstrated diminished affinity for the human ether a-go-go related gene transcript ion channel compared to SCH-C, suggesting a reduced potential for cardiac effects. Vicriviroc represents a promising new candidate for the treatment of HIV-1 infection.


Subject(s)
Anti-HIV Agents/pharmacology , CCR5 Receptor Antagonists , HIV-1/drug effects , Piperazines/pharmacology , Pyrimidines/pharmacology , Humans , Leukocytes, Mononuclear
10.
Virology ; 338(1): 182-99, 2005 Jul 20.
Article in English | MEDLINE | ID: mdl-15935415

ABSTRACT

We describe the generation of two genetically related human immunodeficiency virus type 1 (HIV-1) isolates highly (>20,000-fold) resistant to the small molecule CCR5 inhibitor, SCH-417690 (formerly SCH-D). Both viruses were cross-resistant to other small molecules targeting entry via CCR5, but they were inhibited by some MAbs against the same coreceptor on primary CD4+ T-cells. The resistant isolates remained sensitive to inhibitors of other stages of virus entry, and to replication inhibitors acting post-entry. Neither escape mutant could replicate detectably in peripheral blood mononuclear cells (PBMC) from two donors homozygous for the CCR5-Delta32 allele and both were insensitive to the CXCR4-specific inhibitor, AMD3100. Hence, the SCH-D escape mutants retained the R5 phenotype. One of the resistant isolates was, however, capable of replication in U87.CD4.CXCR4 cells and, after expansion in those cells, was sensitive to AMD3100 in primary CD4+ T-cells. Hence, some X4 variants may be present in this escape mutant swarm. A notable observation was that the SCH-D escape mutants were also cross-resistant to PSC-RANTES and AOP-RANTES, chemokine derivatives that are reported to down-regulate cell surface CCR5 almost completely. However, the extent to which CCR5 is down-regulated was dependent upon the detection MAb. Hence, the escape mutants may be using a CCR5 configuration that is only detected by some anti-CCR5 MAbs. Finally, two SCH-D-resistant clonal viruses revealed no amino acid changes in the gp120 V3 region relative to the parental viruses, in marked contrast to clones resistant to the AD101 small molecule CCR5 inhibitor that possess 4 such sequence changes. Several sequence changes elsewhere in gp120 (V2, C3 and V4) were present in the SCH-D-resistant clones. Their influence on the resistant phenotype remains to be determined.


Subject(s)
CCR5 Receptor Antagonists , HIV-1/drug effects , Piperazines/pharmacology , Pyrimidines/pharmacology , Alleles , Amino Acid Sequence , Anti-HIV Agents/pharmacology , Antibodies, Monoclonal , Base Sequence , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cell Line , Chemokine CCL5/analogs & derivatives , Chemokine CCL5/pharmacology , DNA, Viral/genetics , Drug Resistance, Viral/genetics , Gene Expression , Gene Products, env/genetics , Genes, env , HIV Envelope Protein gp120/genetics , HIV-1/genetics , HIV-1/isolation & purification , HIV-1/physiology , Humans , In Vitro Techniques , Molecular Sequence Data , Mutation , Peptide Fragments/genetics , Phenotype , Receptors, CCR5/genetics , Receptors, CCR5/immunology , Virus Replication/drug effects , Virus Replication/genetics
11.
J Med Chem ; 47(10): 2405-8, 2004 May 06.
Article in English | MEDLINE | ID: mdl-15115380

ABSTRACT

The nature and the size of the benzylic substituent are shown to be the key to controlling receptor selectivity (CCR5 vs M1, M2) and potency in the title compounds. Optimization of the lead benzylic methyl compound 3 led to the methoxymethyl analogue 30, which had excellent receptor selectivity and oral bioavailability in rats and monkeys. Compound 30 (Sch-417690/Sch-D), a potent inhibitor of HIV-1 entry into target cells, is currently in clinical trials.


Subject(s)
Anti-HIV Agents/chemical synthesis , CCR5 Receptor Antagonists , HIV-1/drug effects , Piperazines/chemical synthesis , Piperidines/chemical synthesis , Potassium Channels, Voltage-Gated , Pyrimidines/chemical synthesis , Administration, Oral , Animals , Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacology , Biological Availability , Brain/drug effects , Cation Transport Proteins/drug effects , Digestive System/drug effects , Ether-A-Go-Go Potassium Channels , HIV-1/isolation & purification , Humans , In Vitro Techniques , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Macaca fascicularis , Piperazines/adverse effects , Piperazines/chemistry , Piperazines/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Potassium Channels/drug effects , Pyrimidines/adverse effects , Pyrimidines/chemistry , Pyrimidines/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship
12.
J Virol ; 78(8): 4134-44, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15047829

ABSTRACT

AD101 and SCH-C are two chemically related small molecules that inhibit the entry of human immunodeficiency virus type 1 (HIV-1) via human CCR5. AD101 also inhibits HIV-1 entry via rhesus macaque CCR5, but SCH-C does not. Among the eight residues that differ between the human and macaque versions of the coreceptor, only one, methionine-198, accounts for the insensitivity of macaque CCR5 to inhibition by SCH-C. Thus, the macaque coreceptor engineered to contain the natural human CCR5 residue (isoleucine) at position 198 is sensitive to HIV-1 entry inhibition by SCH-C, whereas a human CCR5 mutant containing the corresponding macaque residue (methionine) is resistant. Position 198 is in CCR5 transmembrane (TM) helix 5 and is not located within the previously defined binding site for AD101 and SCH-C, which involves residues in TM helices 1, 2, 3, and 7. SCH-C binds to human CCR5 whether residue 198 is isoleucine or methionine, and it also binds to macaque CCR5. However, the binding of a conformation-dependent monoclonal antibody to human CCR5 is inhibited by SCH-C only when residue 198 is isoleucine. These observations, taken together, suggest that the antiviral effects of SCH-C and AD101 involve stabilization, or induction, of a CCR5 conformation that is not compatible with HIV-1 infection. However, SCH-C is unable to exert this effect on CCR5 conformation when residue 198 is methionine. The region of CCR5 near residue 198 has, therefore, an important influence on the conformational state of this receptor.


Subject(s)
CCR5 Receptor Antagonists , HIV-1/drug effects , Piperidines , Receptors, CCR5/genetics , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Cell Line , Chemokine CCL5/antagonists & inhibitors , Cyclic N-Oxides/pharmacology , HIV-1/pathogenicity , Humans , Macaca mulatta , Models, Biological , Models, Molecular , Mutagenesis, Site-Directed , Oximes , Protein Conformation , Protein Structure, Tertiary , Pyridines/pharmacology , Receptors, CCR5/chemistry , Signal Transduction/drug effects , Species Specificity
13.
J Virol ; 78(6): 2790-807, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14990699

ABSTRACT

We have described previously the generation of an escape variant of human immunodeficiency virus type 1 (HIV-1), under the selection pressure of AD101, a small molecule inhibitor that binds the CCR5 coreceptor (A. Trkola, S. E. Kuhmann, J. M. Strizki, E. Maxwell, T. Ketas, T. Morgan, P. Pugach, S. X. L. Wojcik, J. Tagat, A. Palani, S. Shapiro, J. W. Clader, S. McCombie, G. R. Reyes, B. M. Baroudy, and J. P. Moore, Proc. Natl. Acad. Sci. USA 99:395-400, 2002). The escape mutant, CC101.19, continued to use CCR5 for entry, but it was at least 20,000-fold more resistant to AD101 than the parental virus, CC1/85. We have now cloned the env genes from the the parental and escape mutant isolates and made chimeric infectious molecular clones that fully recapitulate the phenotypes of the corresponding isolates. Sequence analysis of the evolution of the escape mutants suggested that the most relevant changes were likely to be in the V3 loop of the gp120 glycoprotein. We therefore made a series of mutant viruses and found that full AD101 resistance was conferred by four amino acid changes in V3. Each change individually caused partial resistance when they were introduced into the V3 loop of a CC1/85 clone, but their impact was dependent on the gp120 context in which they were made. We assume that these amino acid changes alter how the HIV-1 Env complex interacts with CCR5. Perhaps unexpectedly, given the complete dependence of the escape mutant on CCR5 for entry, monomeric gp120 proteins expressed from clones of the fully resistant isolate failed to bind to CCR5 on the surface of L1.2-CCR5 cells under conditions where gp120 proteins from the parental virus and a partially AD101-resistant virus bound strongly. Hence, the full impact of the V3 substitutions may only be apparent at the level of the native Env complex.


Subject(s)
Anti-HIV Agents/pharmacology , CCR5 Receptor Antagonists , Drug Resistance, Viral , HIV-1/drug effects , HIV-1/genetics , Amino Acid Sequence , Genotype , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp160/chemistry , HIV Envelope Protein gp160/genetics , HIV Infections/virology , HIV-1/classification , HIV-1/metabolism , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/genetics , Phenotype , Receptors, CCR5/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment
14.
Virology ; 310(2): 333-42, 2003 Jun 05.
Article in English | MEDLINE | ID: mdl-12781720

ABSTRACT

The recent development of in vitro hepatitis C virus (HCV) RNA replication systems has provided useful tools for studying the intracellular anti-HCV activity of ribavirin. Ribavirin has been shown to: (1) induce "error catastrophe" in poliovirus, Proc. Natl. Acad. Sci. USA 98, 6895-6900), (2) be a pseudo-substrate of the HCV RNA-dependent RNA polymerase (RdRp) in vitro, J. Biol. Chem. 276, 46094-46098), and (3) increase mutations in HCV RNA in the binary T7 polymerase/HCV cDNA replication system, J. Virol. 76, 8505-8517). These findings have led to the hypothesis that ribavirin may also induce error catastrophe in HCV. However, the functional relevance of ribavirin-induced HCV RNA mutagenesis is unclear. By use of a colony formation assay, in which RNA is isolated from the HCV subgenomic replicon system following treatment, the impact of ribavirin, inosine-5'-monophosphate dehydrogenase (IMPDH) inhibitors, and the combination was assessed. Ribavirin reduced HCV replicon colony-forming efficiency (CFE) in a dose-dependent fashion, suggesting that ribavirin may be misincorporated into replicon RNA and result in an anti-replicon effect analogous to error catastrophe. This effect was markedly suppressed by addition of exogenous guanosine. Combination treatment with ribavirin and mycophenolic acid (MPA) or VX-497, both potent, nonnucleoside IMPDH inhibitors, led to a greatly enhanced anti-replicon effect. This enhancement was reversed by inclusion of guanosine with the treatment. In contrast, MPA or VX-497 alone had only marginal effects on both the quantity and quality (CFE) of replicon RNA, suggesting that although IMPDH inhibition is an important contributing factor to the overall ribavirin anti-HCV replicon activity, IMPDH inhibition by itself is not sufficient to exert an anti-HCV effect. Sequencing data targeting the neo gene segment of the HCV replicon indicated that ribavirin together with MPA or VX-497 increased the replicon error rate by about two-fold. Taken together these results further suggest that lethal mutagenesis may be an effective anti-HCV strategy. The colony formation assay provides a useful tool for evaluating mutagenic nucleoside analogs for HCV therapy. Finally, the data from combination treatment indicate potential therapeutic value for an enhanced anti-HCV effect when using ribavirin in combination with IMPDH inhibition.


Subject(s)
Antimetabolites/pharmacology , Carbamates/pharmacology , Enzyme Inhibitors/pharmacology , Genome, Viral , Hepacivirus/genetics , IMP Dehydrogenase/pharmacology , Mycophenolic Acid/pharmacology , Phenylurea Compounds/pharmacology , Replicon/drug effects , Ribavirin/pharmacology , Cell Line, Transformed , Guanosine , Hepacivirus/drug effects , Hepacivirus/physiology , Humans , IMP Dehydrogenase/antagonists & inhibitors , Mutation , RNA/biosynthesis , RNA, Viral/biosynthesis , Replicon/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Virus Replication/drug effects
15.
J Immunol ; 170(11): 5748-55, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12759458

ABSTRACT

IFN-kappa belongs to a recently identified subclass of type I IFNs. In this study, we report the cloning and preliminary characterization of the murine homologue of IFN-kappa. The gene encodes a 200-aa protein which is 38.5% homologous to human IFN-kappa. Murine IFN-kappa contains four cysteines in analogous positions to those observed in the IFN-alpha and an additional fifth unique cysteine, C174. The murine gene is located on chromosome 4, where other type I murine IFN genes, IFN-alpha and IFN-beta, are clustered. This region is syntenic with human chromosome 9 where the gene encoding IFN-kappa and the type I IFN gene cluster are found. Mouse IFN-kappa is expressed at low levels in peritoneal macrophages and its expression is up-regulated by dsRNA and IFN-gamma. Similar to previously reported transgenic mice carrying type I and type II IFNs, transgenic mice overexpressing murine IFN-kappa in the beta cells of the pancreas develop overt diabetes with hyperglycemia. Histological characterization of pancreatic islets from these transgenic mice showed inflammatory infiltrates with corresponding destruction of beta cells.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Interferon Type I/biosynthesis , Interferon Type I/genetics , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Humans , Interferon Type I/isolation & purification , Interferon-gamma/pharmacology , Islets of Langerhans/pathology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Molecular Sequence Data , RNA, Double-Stranded/pharmacology , Sequence Alignment , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
16.
J Virol ; 77(9): 5201-8, 2003 May.
Article in English | MEDLINE | ID: mdl-12692222

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) entry is mediated by the consecutive interaction of the envelope glycoprotein gp120 with CD4 and a coreceptor such as CCR5 or CXCR4. The CCR5 coreceptor is used by the most commonly transmitted HIV-1 strains that often persist throughout the course of infection. Compounds targeting CCR5-mediated entry are a novel class of drugs being developed to treat HIV-1 infection. In this study, we have identified the mechanism of action of two inhibitors of CCR5 function, SCH-350581 (AD101) and SCH-351125 (SCH-C). AD101 is more potent than SCH-C at inhibiting HIV-1 replication in primary lymphocytes, as well as viral entry and gp120 binding to cell lines. Both molecules also block the binding of several anti-CCR5 monoclonal antibodies that recognize epitopes in the second extracellular loop of CCR5. Alanine mutagenesis of the transmembrane domain of CCR5 suggests that AD101 and SCH-C bind to overlapping but nonidentical sites within a putative ligand-binding cavity formed by transmembrane helices 1, 2, 3, and 7. We propose that the binding of small molecules to the transmembrane domain of CCR5 may disrupt the conformation of its extracellular domain, thereby inhibiting ligand binding to CCR5.


Subject(s)
CCR5 Receptor Antagonists , Cyclic N-Oxides/pharmacology , HIV-1/drug effects , HIV-1/pathogenicity , Piperidines , Pyridines/pharmacology , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , HIV Envelope Protein gp120/metabolism , Humans , Membrane Fusion , Models, Molecular , Molecular Sequence Data , Oximes , Pyridines/chemistry , Receptors, CCR5/chemistry , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Virus Replication
17.
AIDS Res Hum Retroviruses ; 19(3): 177-86, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12689409

ABSTRACT

The small-molecule CCR5 antagonist SCH-C (SCH 351125) was tested for its ability to inhibit HIV-1 replication in peripheral blood mononuclear cells (PBMCs), cord blood mononuclear cells, immature dendritic cells (DCs), and macrophages. Inhibition of infection of PBMCs by virus associated with mature DC in trans was also studied. For comparison, the peptide-based fusion inhibitor T-20 and the CC-chemokine RANTES were also evaluated. Although some cell type-dependent differences in potency were observed, each of the three entry inhibitors was active against the replication of three different CCR5-using primary isolates in each cell type. CCR5-dependent HIV-1 infectivity, whether DC associated or not, is thus vulnerable to inhibitors that block the virus-cell fusion process by different mechanisms. Together, these results suggest that SCH-C and other entry inhibitors should be evaluated for their clinical potential as inhibitors of HIV-1 replication in several settings, including the prevention of maternal-infant transmission and the prevention of sexual transmission by topical application as a microbicide.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/drug effects , Piperidines , Receptors, CCR5/immunology , Virus Replication/drug effects , CCR5 Receptor Antagonists , Cell Compartmentation , Chemokine CCL5/pharmacology , Cyclic N-Oxides/pharmacology , Dendritic Cells/virology , Drug Interactions , Enfuvirtide , Fetal Blood/virology , HIV Envelope Protein gp41/pharmacology , HIV-1/physiology , Humans , Leukocytes, Mononuclear/virology , Macrophages/virology , Oximes , Peptide Fragments/pharmacology , Pyridines/pharmacology , Receptors, CCR5/physiology , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...