Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters











Publication year range
1.
J Virol ; 75(13): 5998-6006, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11390601

ABSTRACT

The envelope glycoprotein of human immunodeficiency virus type 1 (HIV-1) is extensively glycosylated, containing approximately 23 asparagine (N)-linked glycosylation sites on its gp120 subunit. In this study, specific glycosylation sites on gp120 of a dualtropic primary HIV-1 isolate, DH12, were eliminated by site-directed mutagenesis and the properties of the resulting mutant envelopes were evaluated using a recombinant vaccinia virus-based cell-to-cell fusion assay alone or in the context of viral infections. Of the glycosylation sites that were evaluated, those proximal to the V1/V2 loops (N135, N141, N156, N160) and the V3 loops (N301) of gp120 were functionally critical. The glycosylation site mutations near the V1/V2 loop compromised the use of CCR5 and CXCR4 equally. In contrast, a mutation within the V3 loop preferentially inhibited the usage of CCR5; although this mutant protein completely lost its CCR5-dependent fusion activity, it retained 50% of the wild-type fusion activity with CXCR4. The replication of a virus containing this mutation was severely compromised in peripheral blood mononuclear cells, MT-4 cells, and primary monocyte-derived macrophages. A revertant virus, which acquired second site changes in the V3 loop that resulted in an increase in net positive charge, was isolated. The revertant virus fully recovered the usage of CXCR4 but not of CCR5, thereby altering the tropism of the parental virus from dualtropic to T-tropic. These results suggest that carbohydrate moieties near the V1/V2 and the V3 loops play critical roles in maintaining proper conformation of the variable loops for optimal interaction with receptors. Our results, combined with those of previously reported studies, further demonstrate that the function of individual glycans may be virus isolate dependent.


Subject(s)
HIV Envelope Protein gp120/chemistry , HIV-1/physiology , Receptors, CCR5/physiology , Receptors, CXCR4/physiology , Base Sequence , Glycosylation , HIV-1/chemistry , Humans , Molecular Sequence Data , Virus Replication
2.
J Virol ; 75(14): 6645-59, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11413332

ABSTRACT

Previous studies identified three molecular clones of the acutely pathogenic SIVsmPBj strain that varied in terms of relative in vivo pathogenicity. One clone, SIVsmPBj6.6, reproducibly induced a rapidly fatal disease in pigtailed macaques. In contrast, a highly related clone (SIVsmPBj6.9) was only minimally pathogenic in macaques. PBj6.6 and PBj6.9 shared a tyrosine substitution at position 17 in the Nef protein that is a major determinant of virulence but differed at one residue in Vpx (C89R), three residues within the envelope (D119G, R871G, G872R), and a single residue in Nef (F252L). SIVsmPBj6.9 was less efficient in inducing proliferation of resting macaque peripheral blood mononuclear cells in vitro than SIVsmPBj6.6 and exhibited a marked reduction in infectivity relative to SIVsmPBj6.6. Chimeric viruses for each of these variable residues were constructed, and their biologic properties were compared to those of the parental strains. Differences in Vpx and Nef did not alter the basic biologic phenotype of the chimeras. However, the D119G substitution in the envelope of SIVsmPBj6.9 was associated with a marked reduction in the infectivity of this virus relative to SIVsmPBj6.6. An associated processing defect in gp160 of SIVsmPBj6.9 and chimeras expressing the D119G substitution suggests that a reduction in virion envelope incorporation is the mechanistic basis for reduced virion infectivity. In vivo studies revealed that substitution of the PBj6.9 amino acid into PBj6.6 (D119) abrogated the pathogenicity of this previously pathogenic virus. Introduction of the PBj6.9 G119, however, did not confer full virulence to the parental PBj6.9 virus, implicating one or all of the other four substitutions in the virulence of SIVsmPBj6.6.


Subject(s)
Acquired Immunodeficiency Syndrome/virology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity , Viral Envelope Proteins/genetics , Viral Envelope Proteins/physiology , Amino Acid Substitution , Animals , Chimera , Macaca nemestrina , Simian Immunodeficiency Virus/genetics , Virulence
3.
J Virol ; 75(11): 5151-8, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11333896

ABSTRACT

Since cytotoxic T lymphocytes (CTLs) are critical for controlling human immunodeficiency virus type 1 (HIV-1) replication in infected individuals, candidate HIV-1 vaccines should elicit virus-specific CTL responses. In this report, we study the immune responses elicited in rhesus monkeys by a recombinant poxvirus vaccine and the degree of protection afforded against a pathogenic simian-human immunodeficiency virus SHIV-89.6P challenge. Immunization with recombinant modified vaccinia virus Ankara (MVA) vectors expressing SIVmac239 gag-pol and HIV-1 89.6 env elicited potent Gag-specific CTL responses but no detectable SHIV-specific neutralizing antibody (NAb) responses. Following intravenous SHIV-89.6P challenge, sham-vaccinated monkeys developed low-frequency CTL responses, low-titer NAb responses, rapid loss of CD4+ T lymphocytes, high-setpoint viral RNA levels, and significant clinical disease progression and death in half of the animals by day 168 postchallenge. In contrast, the recombinant MVA-vaccinated monkeys demonstrated high-frequency secondary CTL responses, high-titer secondary SHIV-89.6-specific NAb responses, rapid emergence of SHIV-89.6P-specific NAb responses, partial preservation of CD4+ T lymphocytes, reduced setpoint viral RNA levels, and no evidence of clinical disease or mortality by day 168 postchallenge. There was a statistically significant correlation between levels of vaccine-elicited CTL responses prior to challenge and the control of viremia following challenge. These results demonstrate that immune responses elicited by live recombinant vectors, although unable to provide sterilizing immunity, can control viremia and prevent disease progression following a highly pathogenic AIDS virus challenge.


Subject(s)
HIV-1/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Vaccines, Synthetic/administration & dosage , Vaccinia virus/genetics , Animals , Antibodies, Viral/analysis , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , Disease Progression , Gene Products, env/immunology , Gene Products, gag/immunology , Gene Products, pol/immunology , HIV-1/genetics , Humans , Macaca mulatta , RNA, Viral/blood , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , T-Lymphocytes, Cytotoxic/immunology , Time Factors , Vaccines, Synthetic/immunology , Vaccinia virus/immunology
4.
J Virol ; 75(5): 2224-34, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11160726

ABSTRACT

The great difficulty in eliciting broadly cross-reactive neutralizing antibodies (NAbs) against human immunodeficiency virus type 1 (HIV-1) isolates has been attributed to several intrinsic properties of their viral envelope glycoprotein, including its complex quaternary structure, extensive glycosylation, and marked genetic variability. Most previously evaluated vaccine candidates have utilized envelope glycoprotein from a single virus isolate. Here we compare the breadth of NAb and protective immune response following vaccination of pigtailed macaques with envelope protein(s) derived from either single or multiple viral isolates. Animals were challenged with Simian/human immunodeficiency virus strain DH12 (SHIV(DH12)) following priming with recombinant vaccinia virus(es) expressing gp160(s) and boosting with gp120 protein(s) from (i) LAI, RF, 89.6, AD8, and Bal (Polyvalent); (ii) LAI, RF, 89.6, AD8, Bal, and DH12 (Polyvalent-DH12); (iii) 89.6 (Monovalent-89.6); and (iv) DH12 (Monovalent-DH12). Animals in the two polyvalent vaccine groups developed NAbs against more HIV-1 isolates than those in the two monovalent vaccine groups (P = 0.0054). However, the increased breadth of response was directed almost entirely against the vaccine strains. Resistance to SHIV(DH12) strongly correlated with the level of NAbs directed against the virus on the day of challenge (P = 0.0008). Accordingly, the animals in the Monovalent-DH12 and Polyvalent-DH12 vaccine groups were more resistant to the SHIV(DH12) challenge than the macaques immunized with preparations lacking a DH12 component (viz. Polyvalent and Monovalent-89.6) (P = 0.039). Despite the absence of any detectable NAb, animals in the Polyvalent vaccine group, but not those immunized with Monovalent-89.6, exhibited markedly lower levels of plasma virus than those in the control group, suggesting a superior cell-mediated immune response induced by the polyvalent vaccine.


Subject(s)
AIDS Vaccines/immunology , HIV Antibodies/blood , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp160/immunology , HIV Infections/prevention & control , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Animals , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp160/genetics , HIV-1/classification , HIV-1/genetics , HIV-1/immunology , HIV-1/metabolism , Macaca nemestrina , Neutralization Tests , RNA, Viral/blood , Simian Immunodeficiency Virus/pathogenicity , Vaccination , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology
5.
Proc Natl Acad Sci U S A ; 98(2): 658-63, 2001 Jan 16.
Article in English | MEDLINE | ID: mdl-11136236

ABSTRACT

The highly pathogenic simian immunodeficiency virus/HIV type 1 (SHIV) chimeric virus SHIV(DH12R) induces a systemic depletion of CD4(+) T lymphocytes in rhesus monkeys during the initial 3-4 weeks of infection. Nonetheless, high levels of viral RNA production continue unabated for an additional 2-5 months. In situ hybridization and immunohistochemical analyses revealed that tissue macrophage in the lymph nodes, spleen, gastrointestinal tract, liver, and kidney sustain high plasma virus loads in the absence of CD4(+) T cells. Quantitative confocal immunofluorescence analysis indicated that greater than 95% of the virus-producing cells in these tissues are macrophage and less than 2% are T lymphocytes. Interestingly, the administration of a potent reverse transcriptase inhibitor blocked virus production during the early T cell phase but not during the later macrophage phase of the SHIV(DH12R) infection. When interpreted in the context of HIV-1 infections, these results implicate tissue macrophage as an important reservoir of virus in vivo. They become infected during the acute infection, gradually increase in number over time, and can be a major contributor to total body virus burden during the symptomatic phase of the human infection.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Lentivirus Infections/virology , Macaca mulatta/virology , Macrophages/virology , Simian Immunodeficiency Virus/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Anti-HIV Agents/pharmacology , CD4 Lymphocyte Count , Chimera , Digestive System/pathology , Digestive System/virology , Disease Progression , Drug Resistance, Microbial , HIV-1/drug effects , HIV-1/genetics , Lentivirus Infections/pathology , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Organ Specificity , Organophosphonates/pharmacology , RNA, Viral/blood , Reverse Transcriptase Inhibitors/pharmacology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/genetics , Tenofovir , Viral Load , Virus Replication
6.
J Virol ; 74(24): 11744-53, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11090174

ABSTRACT

The distribution and levels of simian immunodeficiency virus (SIV) in tissues and plasma were assessed in naturally infected African green monkeys (AGM) of the vervet subspecies (Chlorocebus pygerythrus) by limiting-dilution coculture, quantitative PCR for viral DNA and RNA, and in situ hybridization for SIV expression in tissues. A wide range of SIV RNA levels in plasma was observed among these animals (<1,000 to 800,000 copies per ml), and the levels appeared to be stable over long periods of time. The relative numbers of SIV-expressing cells in tissues of two monkeys correlated with the extent of plasma viremia. SIV expression was observed in lymphoid tissues and was not associated with immunopathology. Virus-expressing cells were observed in the lamina propria and lymphoid tissue of the gastrointestinal tract, as well as within alveolar macrophages in the lung tissue of one AGM. The range of plasma viremia in naturally infected AGM was greater than that reported in naturally infected sooty mangabeys. However, the degree of viremia in some AGM was similar to that observed during progression to AIDS in human immunodeficiency virus-infected individuals. Therefore, containment of viremia is an unlikely explanation for the lack of pathogenicity of SIVagm in its natural host species, AGM.


Subject(s)
Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/isolation & purification , Viral Load , Animals , Base Sequence , Chlorocebus aethiops , Humans , Molecular Sequence Data , Viremia
7.
J Virol ; 74(15): 6935-45, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10888632

ABSTRACT

A highly pathogenic simian/human immunodeficiency virus (SHIV), SHIV(DH12R), isolated from a rhesus macaque that had been treated with anti-human CD8 monoclonal antibody at the time of primary infection with the nonpathogenic, molecularly cloned SHIV(DH12), induced marked and rapid CD4(+) T cell loss in all rhesus macaques intravenously inoculated with 1.0 50% tissue culture infective dose (TCID(50)) to 4.1 x 10(5) TCID(50)s of virus. Animals inoculated with 650 TCID(50)s of SHIV(DH12R) or more experienced irreversible CD4(+) T lymphocyte depletion and developed clinical disease requiring euthanasia between weeks 12 and 23 postinfection. In contrast, the CD4(+) T-cell numbers in four of five monkeys receiving 25 TCID(50)s of SHIV(DH12R) or less stabilized at low levels, and these surviving animals produced antibodies capable of neutralizing SHIV(DH12R). In the fifth monkey, no recovery from the CD4(+) T cell decline occurred, and the animal had to be euthanized. Viral RNA levels, subsequent to the initial peak of infection but not at peak viremia, correlated with the virus inoculum size and the eventual clinical course. Both initial infection rate constants, k, and decay constants, d, were determined, but only the latter were statistically correlated to clinical outcome. The attenuating effects of reduced inoculum size were also observed when virus was inoculated by the mucosal route. Because the uncloned SHIV(DH12R) stock possessed the genetic properties of a lentivirus quasispecies, we were able to assess the evolution of the input virus swarm in animals surviving the acute infection by monitoring the emergence of neutralization escape viral variants.


Subject(s)
HIV Infections/virology , HIV-1/pathogenicity , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity , Amino Acid Substitution , Animals , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , Evolution, Molecular , HIV Antibodies/blood , HIV Infections/immunology , HIV-1/classification , HIV-1/genetics , Macaca mulatta , Neutralization Tests , RNA, Viral/blood , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/classification , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Viral Load
8.
Proc Natl Acad Sci U S A ; 96(24): 14049-54, 1999 Nov 23.
Article in English | MEDLINE | ID: mdl-10570196

ABSTRACT

Although simian/human immunodeficiency virus (SHIV) strain DH12 replicates to high titers and causes immunodeficiency in pig-tailed macaques, virus loads measured in SHIV(DH12)-infected rhesus monkeys are consistently 100-fold lower and none of 22 inoculated animals have developed disease. We previously reported that the administration of anti-human CD8 mAb to rhesus macaques at the time of primary SHIV(DH12) infection resulted in marked elevations of virus loads. One of the treated animals experienced rapid and profound depletions of circulating CD4(+) T lymphocytes. Although the CD4(+) T cell number partially recovered, this monkey subsequently suffered significant weight loss and was euthanized. A tissue culture virus stock derived from this animal, designated SHIV(DH12R), induced marked and rapid CD4(+) cell loss after i.v. inoculation of rhesus monkeys. Retrospective analyses of clinical specimens, collected during the emergence of SHIV(DH12R) indicated: (i) the input cloned SHIV remained the predominant virus during the first 5-7 months of infection; (ii) variants bearing only a few of the SHIV(DH12R) consensus changes first appeared 7 months after the administration of anti-CD8 mAb; (iii) high titers of neutralizing antibody directed against the input SHIV were detected by week 10 and persisted throughout the infection; and (iv) no neutralizing antibody against SHIV(DH12R) ever developed.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD8 Antigens/immunology , HIV/pathogenicity , Simian Immunodeficiency Virus/pathogenicity , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , CD4 Lymphocyte Count , Gene Products, nef/genetics , Gene Products, vpr/genetics , HIV/genetics , HIV/immunology , HIV Core Protein p24/immunology , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp41/genetics , HIV Envelope Protein gp41/immunology , Humans , Macaca mulatta , Molecular Sequence Data , Recombination, Genetic , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Tumor Cells, Cultured , Viral Load , nef Gene Products, Human Immunodeficiency Virus , vpr Gene Products, Human Immunodeficiency Virus
9.
Nat Med ; 5(2): 204-10, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9930869

ABSTRACT

Virus-specific antibodies protect individuals against a wide variety of viral infections. To assess whether human immunodeficiency virus type 1 (HIV-1) envelope-specific antibodies confer resistance against primate lentivirus infections, we purified immunoglobulin (IgG) from chimpanzees infected with several different HIV-1 isolates, and used this for passive immunization of pig-tailed macaques. These monkeys were subsequently challenged intravenously with a chimeric simian-human immunodeficiency virus (SHIV) bearing an envelope glycoprotein derived form HIV-1DH12, a dual-tropic primary virus isolate. Here we show that anti-SHIV neutralizing activity, determined in vitro using an assay measuring loss of infectivity, is the absolute requirement for antibody-mediated protection in vivo. Using an assay that measures 100% neutralization, the titer in plasma for complete protection of the SHIV-challenged macaques was in the range of 1:5-1:8. The HIV-1-specific neutralizing antibodies studied are able to bind to native gp120 present on infectious virus particles. Administration of non-neutralizing anti-HIV IgG neither inhibited nor enhanced a subsequent SHIV infection.


Subject(s)
Gene Products, env/immunology , HIV Infections/prevention & control , HIV-1 , Simian Acquired Immunodeficiency Syndrome/prevention & control , Animals , HIV Antibodies/biosynthesis , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/metabolism , Humans , Immunization, Passive , Macaca , Macaca nemestrina , Neutralization Tests , Pan troglodytes , Reassortant Viruses , Viral Load
10.
Nucleic Acids Res ; 26(10): 2505-7, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9580708

ABSTRACT

We report here a simple method of directly visualizing in automated DNA sequencing chromatograms DNA methylations of different types including cytosine methylations in Hpa II and dcm sites as well as adenine methylations in dam sites. This is made possible by the observation that the extent of incorporation of fluorescently labeled dideoxynucleotides is influenced by the methylated bases in template DNA. This simple approach involves routine automated DNA sequencing without any prior treatment of DNA specific for detecting DNA methylation.


Subject(s)
DNA Methylation , Sequence Analysis, DNA/methods , Adenine/chemistry , Cytosine/chemistry , DNA, Viral/chemistry , DNA-Cytosine Methylases , Escherichia coli , Escherichia coli Proteins , Plasmids/chemistry , Simian virus 40 , Site-Specific DNA-Methyltransferase (Adenine-Specific)
11.
Mamm Genome ; 9(12): 1049-55, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9880676

ABSTRACT

Alleles at the Fv1 gene of inbred mice confer resistance to infection and spread of vertically or horizontally transmitted murine leukemia viruses (MuLV). The nucleotide sequence of Fv1 bears similarity to the gag of a human endogenous retrovirus, HERV-L, but is more closely related to the gag-coding sequence of a newly described class of HERV-L-related mouse endogenous retroviruses designated MuERV-L. Both observations suggest an origin of Fv1 from endogenous gag sequences. The molecular definition of Fv1 provided an opportunity to determine the phylogeny of the gene among wild mice and its relation to MuERV-L. PCR primers, chosen to include most of the coding region of Fv1 for both the n and b alleles, were used to amplify sequences from animals of the genus Mus, which were then sequenced. Closely related products were obtained from almost all animals examined that evolved after the separation from Rattus, in which the homologous gene was shown to be absent. A phylogenetic tree generated with Fv1 sequence data differs noticeably from that developed with sequence data from other genes. In addition, non-synonymous changes were found to be present twice as frequently as synonymous changes, a fact that departs from the standard behavior of a structural gene. These observations suggest that the Fv1 gene may have been subjected to possible horizontal transfers as well as to positive Darwinian selection.


Subject(s)
Cell Cycle Proteins , Neoplasm Proteins , Proteins/genetics , Amino Acid Sequence , Animals , DNA/analysis , DNA/chemistry , DNA/genetics , Evolution, Molecular , Genetic Variation , Mice , Mice, Inbred Strains , Molecular Sequence Data , Muridae , Phylogeny , Rats , Sequence Alignment , Sequence Homology, Amino Acid
12.
J Med Virol ; 53(1): 85-95, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9298738

ABSTRACT

Immunologic reagents and methodology are essential to develop further the woodchuck and woodchuck hepatitis virus (WHV) as a model of immune response, inflammation, and immunotherapy in hepatitis B virus (HBV) infection. Partial cDNA clones for the woodchuck CD3epsilon marker of T cells (536 bp) and for selected woodchuck cytokines were developed, including IL-1beta (332 bp), IL-2 (249 bp), IL-4 (205 bp), IL-10 (476 bp), IFN-gamma (476 bp), and TNF-alpha (381 bp). This panel of markers includes sets to measure RNAs for T cells (CD3epsilon), immune response induction (IL-1beta, IL-2), TH subsets (TH1, IL-2/IFN-gamma vs. TH2, IL-4/IL-10), and effector molecules that regulate hepadnavirus replication and liver injury (IFN-gamma, TNF-alpha). Primers representing highly conserved segments of genes from other species were used to derive the partial cDNA clones. Target RNA was obtained from woodchuck peripheral blood mononuclear cells (PBMC) that were stimulated in vitro with ConA, LPS, and human rIL-2. The cDNA clones were validated by 1) comparison with other species for homologies in the nucleotide and predicted amino acid sequences and 2) a first generation assay demonstrating induction of the respective RT-PCR products in stimulated woodchuck PBMC. The corresponding RNAs were also detectable in most cases in the total RNA from the livers of uninfected and WHV-infected woodchucks and differential expression of IFN-gamma and TNF-alpha RNAs was suggested. Second generation, semi-quantitative assays for the RNAs were validated using RT-PCR and dot-blot hybridization with 32P-oligomers derived from the internal sequences of the respective clones. Continued study of the woodchuck immune response to WHV infection using these assays will provide insight into the kinetics and immune mechanisms that initiate and maintain chronic hepadnavirus infection and, hence, enable development of improved immunotherapies for established chronic HBV infection.


Subject(s)
CD3 Complex/genetics , Cytokines/genetics , DNA, Complementary/genetics , Marmota/genetics , Marmota/immunology , RNA/genetics , RNA/isolation & purification , Amino Acid Sequence , Animals , Base Sequence , CD3 Complex/biosynthesis , Cloning, Molecular , Cytokines/biosynthesis , DNA Primers/genetics , Disease Models, Animal , Hepatitis B/immunology , Hepatitis B/virology , Hepatitis B Virus, Woodchuck , Humans , Liver/immunology , Marmota/virology , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Homology, Amino Acid
13.
Mol Med ; 3(1): 23-36, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9132277

ABSTRACT

BACKGROUND: CC chemokine receptor 5 (CCR5) is a cell entry cofactor for macrophage-tropic isolates of human immunodeficiency virus-1 (HIV-1). Recently, an inactive CCR5 allele (designated here as CCR5-2) was identified that confers resistance to HIV-1 infection in homozygotes and slows the rate of progression to AIDS in heterozygotes. The reports conflict on the effect of heterozygous CCR5-2 on HIV-1 susceptibility, and race and risk levels have not yet been fully analyzed. Here we report our independent identification of CCR5-2 and test its effects on HIV-1 pathogenesis in individuals with contrasting clinical outcomes, defined race, and quantified risk. MATERIALS AND METHODS: Mutant CCR5 alleles were sought by directed heteroduplex analysis of genomic DNA from random blood donors. Genotypic frequencies were then determined in (1) random blood donors from North America, Asia, and Africa; (2) HIV-1+ individuals; and (3) highly exposed-seronegative homosexuals with quantified risk. RESULTS: CCR5-2 was the only mutant allele found. It was common in Caucasians, less common in other North American racial groups, and not detected in West Africans or Tamil Indians. Homozygous CCR5-2 frequencies differed reciprocally in highly exposed-seronegative (4.5%, n = 111) and HIV-1-seropositive (0%, n = 614) Caucasians relative to Caucasian random blood donors (0.8%, n = 387). This difference was highly significant (p < 0.0001). By contrast, heterozygous CCR5-2 frequencies did not differ significantly in the same three groups (21.6, 22.6, and 21.7%, respectively). A 55% increase in the frequency of heterozygous CCR5-2 was observed in both of two cohorts of Caucasian homosexual male, long-term nonprogressors compared with other HIV-1+ Caucasian homosexuals (p = 0.006) and compared with Caucasian random blood donors. Moreover, Kaplan-Meier estimates indicated that CCR5-2 heterozygous seroconvertors had a 52.6% lower risk of developing AIDS than homozygous wild-type seroconvertors. CONCLUSIONS: The data suggest that homozygous CCR5-2 is an HIV-1 resistance factor in Caucasians with complete penetrance, and that heterozygous CCR5-2 slows the rate of disease progression in infected Caucasian homosexuals. Since the majority (approximately 96%) of highly exposed-seronegative individuals tested are not homozygous for CCR5-2, other resistance factors must exist. Since CCR5-2 homozygotes have no obvious clinical problems, CCR5 may be a good target for the development of novel antiretroviral therapy.


Subject(s)
Gene Frequency , HIV Infections , HIV Seronegativity/genetics , HIV-1 , Receptors, Cytokine/genetics , Receptors, HIV/genetics , Adult , Black People/genetics , Cloning, Molecular , Disease Progression , Disease Susceptibility , Frameshift Mutation/genetics , HeLa Cells , Heterozygote , Homosexuality, Male , Homozygote , Humans , Male , Membrane Fusion , Middle Aged , Molecular Sequence Data , Phenotype , Polymorphism, Restriction Fragment Length , Racial Groups/genetics , Receptors, CCR5 , Receptors, Cytokine/physiology , Receptors, HIV/physiology , Risk Factors
15.
J Virol ; 69(7): 4453-62, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7769705

ABSTRACT

Fresh human immunodeficiency virus type 1 (HIV-1) isolates from patients with AIDS were screened for infectivity in chimpanzee peripheral blood mononuclear cells (PBMC) to identify strains potentially able to generate high virus loads in an inoculated animal. Only 3 of 23 isolates obtained were infectious in chimpanzee cells. Of these three, only one (HIV-1DH12) was able to initiate a productive infection in PBMC samples from all 25 chimpanzees tested. HIV-1DH12 tissue culture infections were characterized by extremely rapid replication kinetics, profound cytopathicity, and tropism for chimp and human PBMC, primary human macrophage, and several human T-cell lines. An infection was established within 1 week of inoculating a chimpanzee with 50 50% tissue culture infective doses of HIV-1DH12; cell-free virus was recovered from the plasma at weeks 1, 2, and 4 and was associated with the development of lymphadenopathy. Virus loads during the primary infection and at 6 months postinoculation were comparable to those reported in HIV-1-seropositive individuals.


Subject(s)
HIV-1/pathogenicity , Macrophages/virology , T-Lymphocytes/virology , Acquired Immunodeficiency Syndrome/virology , Adult , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA, Viral/genetics , HIV-1/isolation & purification , Humans , Male , Molecular Sequence Data , Pan troglodytes
16.
J Virol ; 68(8): 5311-20, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8035531

ABSTRACT

The matrix protein of human immunodeficiency virus type 1 is encoded by the amino-terminal portion of the Gag precursor and is postulated to be involved in a variety of functions in the virus life cycle. To define domains and specific amino acid residues of the matrix protein that are involved in virus particle assembly, we introduced 35 amino acid substitution mutations in the human immunodeficiency virus type 1 matrix protein. Using reverse transcriptase and radioimmunoprecipitation analyses and transmission electron microscopy, we assessed the mutants for their ability to form virus particles and to function in the infection process. This study has identified several domains of the matrix protein in which single amino acid substitutions dramatically reduce the efficiency of virus particle production. These domains include the six amino-terminal residues of matrix, the region of matrix between amino acids 55 and 59, and the region between amino acids 84 and 95. Single amino acid substitutions in one of these domains (between matrix amino acids 84 and 88) result in a redirection of the majority of virus particle formation to sites within cytoplasmic vacuoles.


Subject(s)
Gene Products, gag/chemistry , HIV Antigens/chemistry , HIV-1/physiology , Viral Proteins , Amino Acid Sequence , Cell Line , Gene Products, gag/physiology , HIV Antigens/physiology , HIV-1/chemistry , HIV-1/ultrastructure , HeLa Cells , Humans , Microscopy, Electron , Molecular Sequence Data , Mutagenesis, Insertional , Protein Precursors/chemistry , Radioimmunoprecipitation Assay , Virus Replication/physiology , gag Gene Products, Human Immunodeficiency Virus
17.
J Virol ; 68(2): 1207-12, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8289353

ABSTRACT

CD4 is an integral membrane glycoprotein which functions as the human immunodeficiency virus receptor for infection of human host cells. We have recently demonstrated that Vpu, a human immunodeficiency virus type 1-encoded integral membrane phosphoprotein, induces rapid degradation of CD4 in the endoplasmic reticulum. Using an in vitro model system, we demonstrated that Vpu targets specific sequences in the cytoplasmic domain of CD4 to promote its degradation. In this report, we have further delineated regions within CD4 which are required for susceptibility to Vpu. Transfer of the CD4 cytoplasmic region into a heterologous protein, CD8, rendered the chimeric protein sensitive to Vpu-dependent degradation. In contrast, substitution of the CD8 transmembrane domain with the analogous region from CD4 did not confer sensitivity to Vpu. Finally, mutant forms of the CD4 protein containing the extracellular region alone or the extracellular and transmembrane regions linked to a heterologous cytoplasmic domain were not targeted by Vpu. Thus, sequences present in the cytoplasmic domain of CD4 are necessary and sufficient to confer sensitivity to Vpu.


Subject(s)
CD4 Antigens/metabolism , HIV-1/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Amino Acid Sequence , Base Sequence , CD4 Antigens/genetics , CD8 Antigens/genetics , CD8 Antigens/metabolism , HeLa Cells , Human Immunodeficiency Virus Proteins , Humans , Molecular Sequence Data , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Structure-Activity Relationship , Transfection
18.
J Virol ; 65(8): 4350-8, 1991 Aug.
Article in English | MEDLINE | ID: mdl-2072454

ABSTRACT

Starting with a replication-incompetent molecular clone of human immunodeficiency virus type 1, lacking all the NF-kappa B and Sp1 binding sites present in the native long terminal repeat (LTR), proviruses containing reconstructed LTRs with individual or combinations of NF-kappa B and Sp1 elements were generated and evaluated for their capacity to produce virus progeny following transfection-cocultivation. Virus stocks obtained from these experiments exhibited a continuum of replicative capacities in different human T-cell types depending on which element(s) was present in the LTR. For example, in experiments involving proviral clones with LTRs containing one or two NF-kappa B elements (and no Sp1 binding sites), a hierarchy of cellular permissivity to virus replication (peripheral blood lymphocytes = MT4 greater than H9 greater than CEM greater than Jurkat) was observed. Of note was the associated emergence of second-site LTR revertants which involved an alteration of the TATA box. These results suggest that the human immunodeficiency virus type 1 LTR possesses functional redundancy which ensures virus replication in different T-cell types and is capable of changing depending on the particular combination of transcriptional factors present.


Subject(s)
DNA, Viral/chemistry , HIV-1/physiology , NF-kappa B/genetics , Repetitive Sequences, Nucleic Acid , Sp1 Transcription Factor/genetics , T-Lymphocytes/microbiology , Virus Replication , Base Sequence , Cell Line , DNA Replication , DNA, Viral/biosynthesis , Enhancer Elements, Genetic , HIV-1/genetics , HIV-1/growth & development , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , NF-kappa B/metabolism , Polymerase Chain Reaction , Proviruses/genetics , RNA, Viral/chemistry , Sp1 Transcription Factor/metabolism , Transfection
19.
Science ; 251(5001): 1597-600, 1991 Mar 29.
Article in English | MEDLINE | ID: mdl-2011739

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) gene expression is activated by Tat, a virally encoded protein. Tat trans-activation requires viral (trans-activation--responsive; TAR) RNA sequences located in the R region of the long terminal repeat (LTR). Existing evidence suggests that Tat probably cooperates with cellular factors that bind to TAR RNA in the overall trans-activation process. A HeLa complementary DNA was isolated and characterized that encodes a TAR RNA-binding protein (TRBP). TRBP activated the HIV-1 LTR and was synergistic with Tat function.


Subject(s)
Carrier Proteins/genetics , Escherichia coli Proteins , HIV Long Terminal Repeat , HIV-1/genetics , RNA-Binding Proteins , Amino Acid Sequence , Base Sequence , Binding Sites , Endoribonucleases/genetics , Escherichia coli/enzymology , Gene Products, tat/metabolism , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Plasmids , RNA, Viral/genetics , Ribonuclease III , Sequence Homology, Nucleic Acid , tat Gene Products, Human Immunodeficiency Virus
20.
J Virol ; 65(3): 1414-9, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1995951

ABSTRACT

The long terminal repeat (LTR) of the human immunodeficiency virus (HIV) contains three binding sites for the transcriptional factor Sp1. In order to investigate the role that the Sp1-binding sites play in regulation of HIV replication, we have introduced a deletion of all three Sp1-binding sites into the LTR of an infectious molecular clone of HIV. Viral stocks have been prepared from this mutant virus, designated dl-Sp, and these stocks have been used to study its replicative ability in human T cells. The dl-Sp virus replicated efficiently in MT4 cells and in phytohemagglutinin-stimulated human peripheral blood lymphocytes, but it replicated poorly and with delayed kinetics in A3.01 (CEM) T cells unless those cells had been treated with the cytokine tumor necrosis factor alpha. Gel retardation assays to study the levels of DNA-binding proteins present in these cells showed that NF-kappa B activity could be detected in the nuclei of MT4 cells but not in A3.01 cells unless they had been treated with tumor necrosis factor alpha. Thus, the presence of NF-kappa B activity appeared to be required for efficient replication of an HIV whose LTR Sp1-binding sites had been deleted. This suggests that NF-kappa B can functionally compensate for Sp1 in activating HIV replication. The HIV LTR is therefore similar to the promoter-enhancer units of other viruses in that it is composed of multiple functional elements that may contribute differently to viral replication depending on the levels of DNA-binding proteins present in the target cells.


Subject(s)
HIV Long Terminal Repeat , HIV/physiology , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Virus Replication , Animals , Base Sequence , Binding Sites , Cell Line , Chromosome Deletion , HIV/genetics , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , NF-kappa B/metabolism , Restriction Mapping , T-Lymphocytes
SELECTION OF CITATIONS
SEARCH DETAIL