Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 98(22): 12718-23, 2001 Oct 23.
Article in English | MEDLINE | ID: mdl-11606733

ABSTRACT

We describe here the identification and properties of SCH-C (SCH 351125), a small molecule inhibitor of HIV-1 entry via the CCR5 coreceptor. SCH-C, an oxime-piperidine compound, is a specific CCR5 antagonist as determined in multiple receptor binding and signal transduction assays. This compound specifically inhibits HIV-1 infection mediated by CCR5 in U-87 astroglioma cells but has no effect on infection of CXCR4-expressing cells. SCH-C has broad and potent antiviral activity in vitro against primary HIV-1 isolates that use CCR5 as their entry coreceptor, with mean 50% inhibitory concentrations ranging between 0.4 and 9 nM. Moreover, SCH-C strongly inhibits the replication of an R5-using HIV-1 isolate in SCID-hu Thy/Liv mice. SCH-C has a favorable pharmacokinetic profile in rodents and primates with an oral bioavailability of 50-60% and a serum half-life of 5-6 h. On the basis of its novel mechanism of action, potent antiviral activity, and in vivo pharmacokinetic profile, SCH-C is a promising new candidate for therapeutic intervention of HIV infection.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Anti-HIV Agents/pharmacology , CCR5 Receptor Antagonists , Cyclic N-Oxides/pharmacology , HIV-1/drug effects , Piperidines , Pyridines/pharmacology , Animals , Chemokine CCL5/antagonists & inhibitors , Cyclic N-Oxides/pharmacokinetics , Cyclic N-Oxides/therapeutic use , Humans , Macaca fascicularis , Male , Mice , Mice, SCID , Oximes , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley
2.
J Med Chem ; 44(21): 3343-6, 2001 Oct 11.
Article in English | MEDLINE | ID: mdl-11585438

ABSTRACT

Truncation of the original piperidino-2(S)-methyl piperazine lead structure 2, from a family of muscarinic antagonists, gave compound 8 which has improved selectivity for the HIV-1 co-receptor CCR5 over muscarinic receptors. Further optimization for pharmacokinetic properties afforded Sch-350634 (1), a prototypical piperazine-based CCR5 antagonist, which is a potent inhibitor of HIV-1 entry and replication in PBMCs. The title compound (1) has excellent oral bioavailability in rat, dog, and monkey.


Subject(s)
Anti-HIV Agents/chemical synthesis , CCR5 Receptor Antagonists , Cyclic N-Oxides/chemical synthesis , Piperazines/chemical synthesis , Administration, Oral , Animals , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/pharmacology , Biological Availability , Cell Line , Cyclic N-Oxides/chemistry , Cyclic N-Oxides/pharmacokinetics , Cyclic N-Oxides/pharmacology , Dogs , HIV-1/drug effects , In Vitro Techniques , Leukocytes, Mononuclear/virology , Macaca fascicularis , Piperazines/chemistry , Piperazines/pharmacokinetics , Piperazines/pharmacology , Rats , Structure-Activity Relationship
3.
Am J Pathol ; 151(4): 1035-42, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9327737

ABSTRACT

Entry of the type 1 human immunodeficiency virus into most cells requires the presence of the CD4 protein in combination with one of several recently described co-receptors. CXCR-4 (fusin) was the first identified, and it serves as co-receptor for T-cell-line tropic (T-tropic) HIV-1 isolates. To determine the expression of CXCR-4 in the brain, a major target of HIV pathology, we used immunohistochemistry and reverse transcriptase polymerase chain reaction with CXCR-4-specific antibodies and probes. We found that CXCR-4 was expressed in several cell types in brain, but notably in neurons and microglia, a finding that was replicated in tissue culture. The study of the expression of CXCR-4 in the brain, which may be one of many chemokine receptors in the central nervous system, may provide further insight into the interactions between brain cells, pathogens, and the immune system, and help understand the pathogenesis of HIV dementia.


Subject(s)
AIDS Dementia Complex/metabolism , Brain/metabolism , HIV-1 , Microglia/metabolism , Neurons/metabolism , Receptors, CXCR4/biosynthesis , AIDS Dementia Complex/pathology , Adult , Aged , Antibodies, Monoclonal , Brain/pathology , Cells, Cultured , DNA Primers/chemistry , Female , Humans , Immunohistochemistry , In Situ Hybridization , Male , Microglia/pathology , Middle Aged , Neurons/pathology , Polymerase Chain Reaction , RNA, Messenger/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology
4.
J Virol ; 71(7): 5678-83, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9188648

ABSTRACT

We used a monoclonal antibody (12G5) directed against an extracellular domain of CXCR-4 to investigate the role of this receptor in infection of immortalized lymphoid cell lines, peripheral blood mononuclear cells (PBMCs), and primary brain microglia with a dual-tropic strain of human immunodeficiency virus (HIV-1(89.6)) and a T-tropic strain (HIV-1(IIIB)). Addition of antibody 12G5 to cells prior to and during infection with HIV-1(89.6) inhibited p24 production 100- to 10,000-fold in CEMx174 and 174-CD4 cells and about 10-fold in PBMC cultures but had no activity against infection of either monocyte-derived macrophages or brain microglia. In contrast, 12G5 had little or no effect on infection of CEMx174 cells with HIV-1(IIIB) or HIV-1(HxB). To identify the region of the HIV-1(89.6) envelope that confers sensitivity to 12G5, we used chimeric molecular clones. Chimeras containing the V3 loop region of HIV-1(89.6) were inhibited by 12G5 to the same degree as wild-type HIV-1(89.6) whereas replication of those viruses containing the V3 loop of HIV-1(HxB) was not inhibited by the antibody. A similar pattern was seen in infections of a U87 glioblastoma line that coexpresses CD4 and CXCR-4. Antibody 12G5 was also able to block fusion between HeLa-CD4 cells and CEMx174 cells chronically infected with HIV-1(89.6) but had no effect on fusion mediated by cells chronically infected with HIV-1(IIIB). Taken together, these results suggest that different strains of HIV-1 may interact with different sites on CXCR-4 or may have different binding affinities for the coreceptor.


Subject(s)
HIV-1/metabolism , Membrane Proteins/metabolism , Receptors, HIV/metabolism , Antibodies, Monoclonal/metabolism , CD4 Antigens/metabolism , Cell Fusion , Cell Line , Cells, Cultured , HIV Core Protein p24/analysis , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/metabolism , HIV-1/isolation & purification , HeLa Cells , Humans , Microglia/cytology , Microglia/virology , Peptide Fragments/genetics , Peptide Fragments/metabolism , Receptors, CXCR4 , T-Lymphocytes/cytology , T-Lymphocytes/virology , Tumor Cells, Cultured
6.
J Virol ; 70(11): 7654-62, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8892885

ABSTRACT

To ascertain whether viruses present at the time of primary viremia can infect the central nervous system and to determine if microglial tropism is distinct from tropism for monocyte-derived macrophages (MDM), 27 human immunodeficiency virus type 1 (HIV-1) isolates obtained from acutely infected individuals, as well as laboratory strains, were assayed for their ability to replicate in primary adult microglial cultures and in MDM. Most of the isolates replicated equally well in both microglia and MDM, but several isolates replicated preferentially in one of the two cell types, differing by as much as 40-fold in p24gag production. This indicated that while MDM and microglial tropism overlap, a subset of isolates is particularly tropic for one of the two cell types. One isolate was further adapted to microglia by 15 sequential passages, raising the peak p24 concentration produced by 1,000-fold. In addition, the passaged virus induced marked cytopathologic changes (vacuolization and syncytium formation) in infected microglial cultures. Sequence comparison of the V3 loop of unpassaged and multiply passaged virus revealed amino acid changes shown to be associated with isolates from patients with HIV dementia. Our data support the hypothesis that HIV-1 infection can be established in the central nervous system by viruses present early in HIV infection, that some of these viruses are particularly tropic for microglia, and that adaptation in this cell type can result in the selection of a pool of predominantly microglia-tropic (neurotropic) viruses.


Subject(s)
HIV Infections/virology , HIV-1/metabolism , Macrophages/virology , Microglia/virology , Monocytes/virology , Adult , Amino Acid Sequence , Cells, Cultured , Cytopathogenic Effect, Viral , HIV Core Protein p24/metabolism , HIV Envelope Protein gp120/genetics , HIV-1/genetics , HIV-1/isolation & purification , Humans , Macrophages/cytology , Microglia/cytology , Molecular Sequence Data , Monocytes/cytology , Peptide Fragments/genetics , Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology, Amino Acid
7.
Virus Res ; 43(1): 69-75, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8822635

ABSTRACT

We have used restriction endonuclease digestion analysis of polymerase chain reaction (PCR)-amplified gene regions to rapidly examine individual structural gene relationships among field isolates of eastern equine encephalitis (EEE) virus. The E1+ (E1 gene plus 292 nucleotides 3' of the coding region), E2, and C gene regions from North American (NA) variety viruses and the E1 and C gene regions of South American (SA) variety viruses were successfully amplified by RT-PCR using a single primer set for each locus. The products were then digested with a panel of restriction endonucleases and the resulting DNA fragments electrophoretically compared. Our findings revealed marked similarity among the E1+ and the E2 gene restriction patterns, respectively, of most NA strains. In contrast, the restriction patterns exhibited by the E1+ gene of SA strains differed substantially from those of NA strains and also appeared more heterogeneous. The digestion patterns of the C gene were generally similar for all strains of the virus examined. These results thus demonstrate that EEE viral E1+ and C structural gene sequences can be amplified from an assortment of both NA and SA varieties of the virus by RT-PCR using a single primer set per locus, and that both varietal and individual isolate distinctions can be identified by comparison of subsequent restriction digestion patterns. This technique should prove useful as an epidemiological tool for rapid identification of EEE isolates from clinical and field specimens, and as a rapid screen for alterations within structural gene regions.


Subject(s)
Encephalitis Virus, Eastern Equine/genetics , Genes, Viral , Polymerase Chain Reaction/methods , Restriction Mapping/methods , Viral Structural Proteins/genetics , Animals , Encephalitis Virus, Eastern Equine/isolation & purification
8.
Am J Trop Med Hyg ; 53(5): 564-70, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7485719

ABSTRACT

Eastern equine encephalitis (EEE) virus is a mosquito-borne alphavirus that can produce a severe and often fatal acute encephalitis in humans, with significant neurologic sequelae in survivors. Due to the serious nature of the disease, an investigational inactivated EEE vaccine (PE-6) is available to individuals at risk for infection. Both serologic and recent molecular analyses of EEE viruses have demonstrated marked differences between the two antigenic varieties of EEE virus, designated North American (NA) and South American (SA). In view of these findings, we have examined the reactivity of sera from three individuals immunized with the EEE vaccine, derived from an NA isolate, with field strains of EEE virus. Anti-EEE serum antibodies from vaccinees reacted strongly in Western blot assays with both of the envelope (E1 and E2) glycoproteins of each NA strain examined, while reactivities with the glycoproteins of SA strains were substantially weaker and variable and dependent upon both the immune response of the vaccinee and the virus isolate assayed. Most striking was the modest to virtual lack of reactivity with the E2 protein of SA strains. Antigenic differences among the glycoproteins of EEE viruses were not as pronounced in immunoprecipitation analysis. Most significantly, although human immune sera displayed high neutralizing titers against each of the NA isolates examined, only negligible neutralizing titers were obtained against SA isolates. These data suggest that immunized individuals would mount an effective antibody response against infection with NA strains of EEE virus, but that further investigation is clearly warranted to fully assess the protective capability of the vaccine against infection with SA strains.


Subject(s)
Antibodies, Viral/blood , Encephalitis Virus, Eastern Equine/immunology , Encephalomyelitis, Equine/prevention & control , Viral Vaccines/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Base Sequence , Blotting, Western , Encephalitis Virus, Eastern Equine/classification , Encephalitis Virus, Eastern Equine/genetics , Encephalomyelitis, Equine/immunology , Humans , Immune Sera/immunology , Molecular Sequence Data , Neutralization Tests , Precipitin Tests , RNA, Messenger/chemistry , RNA, Viral/chemistry , Vaccination , Viral Proteins/immunology , Virion/immunology
9.
J Gen Virol ; 75 ( Pt 11): 2897-909, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7964601

ABSTRACT

We have re-evaluated the relationships among the polypeptides of eastern equine encephalitis (EEE) viruses using SDS-PAGE and peptide mapping of individual virion proteins. Four to five distinct polypeptide bands were detected upon SDS-PAGE analysis of viruses: the E1, E2 and C proteins normally associated with alphavirus virions, as well as an additional more rapidly-migrating E2-associated protein and a high M(r) (HMW) protein. In contrast with previous findings by others, the electrophoretic profiles of the virion proteins of EEE viruses displayed a marked correlation with serotype. The protein profiles of the 33 North American (NA)-serotype viruses examined were remarkably homogeneous, with variation detected only in the E1 protein of two isolates. In contrast, considerable heterogeneity was observed in the migration profiles of both the E1 and E2 glycoproteins of the 13 South American (SA)-type viruses examined. Peptide mapping of individual virion proteins using limited proteolysis with Staphylococcus aureus V8 protease confirmed that, in addition to the homogeneity evident among NA-type viruses and relative heterogeneity among SA-type viruses, the E1 and E2 proteins of NA- and SA-serotype viruses exhibited serotype-specific structural variation. The C protein was highly conserved among isolates of both virus serotypes. Endoglycosidase analyses of intact virions did not reveal substantial glycosylation differences between the glycoproteins of NA- and SA-serotype viruses. Both the HMW protein and the E2 protein (doublet) of EEE virus appeared to contain, at least in part, high-mannose type N-linked oligosaccharides. No evidence of O-linked glycans was found on either the E1 or the E2 glycoprotein. Despite the observed structural differences between proteins of NA- and SA-type viruses, Western blot analyses utilizing polyclonal antibodies indicated that immunoreactive epitopes appeared to be conserved.


Subject(s)
Encephalitis Virus, Eastern Equine/classification , Animals , Birds/virology , Culicidae/virology , Dogs/virology , Electrophoresis, Polyacrylamide Gel , Encephalitis Virus, Eastern Equine/isolation & purification , Glycoproteins/analysis , Glycoproteins/isolation & purification , Horses/virology , Humans , Peptide Mapping , Serotyping , South America , United States , Viral Proteins/analysis , Viral Proteins/isolation & purification , Virion/chemistry
10.
J Gen Virol ; 75 ( Pt 5): 1177-81, 1994 May.
Article in English | MEDLINE | ID: mdl-7513744

ABSTRACT

The carbohydrate epitope Gal alpha 1-3Gal beta 1-4GlcNAc-R (alpha-galactosyl) is abundantly expressed on cells of non-primate mammals, prosimians and New World monkeys, where it is synthesized by the enzyme alpha 1,3-galactosyltransferase (alpha 1,3GT). Old World monkeys, apes and humans lack alpha 1,3GT and hence do not synthesize alpha-galactosyl epitopes. Instead, these species produce a natural antibody, anti-Gal, which interacts specifically with alpha-galactosyl epitopes and which constitutes up to 1% of circulating immunoglobulins in humans. We have used eastern equine encephalitis (EEE) virus as a model to examine the differential expression of alpha-galactosyl epitopes on the glycoproteins of virus propagated in cells that either produce or lack alpha 1,3GT. As predicted, virus propagated in Vero cells (derived from the African green monkey, an Old World monkey) did not express alpha-galactosyl epitopes. In contrast, virus propagated in mouse 3T3 cells (EEE3T3) expressed approximately 80 alpha-galactosyl epitopes per virion on both the E1 and the E2 envelope glycoproteins. Thus, expression of the alpha-galactosyl epitope on virions paralleled that on host cells. The binding of anti-Gal antibody to these epitopes on EEE3T3 virions partially neutralized virus infectivity, raising the possibility that anti-Gal production in hosts may influence the initial infectious stage of viruses expressing alpha-galactosyl epitopes.


Subject(s)
Antigens, Viral/immunology , Encephalitis Virus, Eastern Equine/immunology , Epitopes/immunology , Galactosides/immunology , Viral Structural Proteins/immunology , Animals , Carbohydrate Sequence , Cells, Cultured , Chlorocebus aethiops , Galactosyltransferases/metabolism , Mice , Molecular Sequence Data , Oligosaccharides/immunology , Radioimmunoassay , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...