Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
J Virol ; 80(15): 7676-87, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16840346

ABSTRACT

While modified vaccinia virus Ankara (MVA) is currently in clinical development as a safe vaccine against smallpox and heterologous infectious diseases, its immunogenicity is likely limited due to the inability of the virus to replicate productively in mammalian hosts. In light of recent data demonstrating that vaccinia viruses, including MVA, preferentially infect antigen-presenting cells (APCs) that play crucial roles in generating antiviral immunity, we hypothesized that expression of specific cytokines and chemokines that mediate APC recruitment and activation from recombinant MVA (rMVA) vectors would enhance the immunogenicity of these vectors. To test this hypothesis, we generated rMVAs that express murine granulocyte-macrophage colony-stimulating factor (mGM-CSF), human CCL20/human macrophage inflammatory protein 3alpha (hCCL20/hMIP-3alpha), or human fms-like tyrosine kinase 3 ligand (hFlt3-L), factors predicted to increase levels of dendritic cells (DCs), to recruit DCs to sites of immunization, or to promote maturation of DCs in vivo, respectively. These rMVAs also coexpress the well-characterized, immunodominant lymphocytic choriomeningitis virus nucleoprotein (NP) antigen that enabled sensitive and quantitative assessment of antigen-specific CD8(+) T-cell responses following immunization of BALB/c mice. Our results demonstrate that immunization of mice with rMVAs expressing mGM-CSF or hCCL20, but not hFlt3-L, results in two- to fourfold increases of cellular immune responses directed against vector-encoded antigens and 6- to 17-fold enhancements of MVA-specific antibody titers, compared to those responses elicited by nonadjuvanted rMVA. Of note, cytokine augmentation of cellular immune responses occurs when rMVAs are given as primary immunizations but not when they are used as booster immunizations, suggesting that these APC-modulating proteins, when used as poxvirus-encoded adjuvants, are more effective at stimulating naïve T-cell responses than in promoting recall of preexisting memory T-cell responses. Our results demonstrate that a strategy to express specific genetic adjuvants from rMVA vectors can be successfully applied to enhance the immunogenicity of MVA-based vaccines.


Subject(s)
Chemokines, CC/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Macrophage Inflammatory Proteins/metabolism , Membrane Proteins/metabolism , Vaccinia virus/immunology , Animals , Antibodies, Viral/immunology , Antibody Formation , Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Chemokine CCL20 , Chemokines, CC/genetics , Chemokines, CC/immunology , Defective Viruses , Dendritic Cells/immunology , Dendritic Cells/virology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunity, Cellular , Immunization , Immunoblotting , Lymphocytic choriomeningitis virus/immunology , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/immunology , Mice , Mice, Inbred BALB C , Nucleoproteins/genetics , Nucleoproteins/immunology
2.
Retrovirology ; 3: 19, 2006 Mar 20.
Article in English | MEDLINE | ID: mdl-16549011

ABSTRACT

BACKGROUND: The ability of emerging pathogens to infect new species is likely related to the diversity of pathogen variants present in existing reservoirs and their degree of genomic plasticity, which determines their ability to adapt to new environments. Certain simian immunodeficiency viruses (SIVcpz, SIVsm) have demonstrated tremendous success in infecting new species, including humans, resulting in the HIV-1 and HIV-2 epidemics. Although SIV diversification has been studied on a population level, the essential substrates for cross-species transmission, namely SIV sequence diversity and the types and extent of viral diversification present in individual reservoir animals have not been elucidated. To characterize this intra-host SIV diversity, we performed sequence analyses of clonal viral envelope (env) V1V2 and gag p27 variants present in individual SIVsm-infected sooty mangabeys over time. RESULTS: SIVsm demonstrated extensive intra-animal V1V2 length variation and amino acid diversity (le38%), and continual variation in V1V2 N-linked glycosylation consensus sequence frequency and location. Positive selection was the predominant evolutionary force. Temporal sequence shifts suggested continual selection, likely due to evolving antibody responses. In contrast, gag p27 was predominantly under purifying selection. SIVsm V1V2 sequence diversification is at least as great as that in HIV-1 infected humans, indicating that extensive viral diversification in and of itself does not inevitably lead to AIDS. CONCLUSION: Positive diversifying selection in this natural reservoir host is the engine that has driven the evolution of the uniquely adaptable SIV/HIV envelope protein. These studies emphasize the importance of retroviral diversification within individual host reservoir animals as a critical substrate in facilitating cross-species transmission.


Subject(s)
Evolution, Molecular , Simian Immunodeficiency Virus/genetics , Viral Envelope Proteins/genetics , Animals , Base Sequence , Cloning, Molecular , DNA Primers , DNA, Viral/genetics , Genetic Variation , HIV-1/genetics , Humans , Phylogeny , Primates/virology , Reverse Transcriptase Polymerase Chain Reaction , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/classification , Simian Immunodeficiency Virus/isolation & purification , Zoonoses
3.
Methods ; 31(2): 120-6, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12957569

ABSTRACT

Cytotoxic T lymphocytes (CTLs) play a major role in the immune response against viruses and other intracellular pathogens. In addition, CTLs are implicated in the control of tumor cells in certain settings. Accurate measures of CTL function are of critical importance to study the pathogenesis of infectious diseases and to evaluate the efficacy of new vaccines and immunotherapies. To this end, we have recently developed a flow cytometry-based CTL (FCC) assay that measures the CTL-induced caspase activation within target cells using cell permeable fluorogenic caspase substrates. This novel assay reliably detects, by flow cytometry or fluorescence/confocal microscopy, antigen-specific CTLs in a wide variety of human and murine systems, and is safer and more informative than the standard 51Cr-release assay. In addition, the flow cytometric CTL (FCC) assay provides an alternative method that is often more sensitive and physiologically informative when compared to previously described FCC assays, as it measures a biological indicator of apoptosis within the target cell. The FCC assay may thus represent a useful tool to further understand the molecular and cellular mechanisms that underlie CTL-mediated killing during tumorigenesis or following infection with viruses or other intracellular pathogens.


Subject(s)
Caspases/metabolism , Flow Cytometry/methods , Microscopy, Fluorescence/methods , T-Lymphocytes, Cytotoxic/immunology , Animals , Cytotoxicity, Immunologic , Fluorescent Dyes , Humans , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/metabolism
4.
Eur J Immunol ; 31(11): 3207-17, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11745337

ABSTRACT

Although CTL escape has been well documented in pathogenic simian immunodeficiency virus (SIV) infection, there is no information on CTL escape in nonpathogenic SIV infection in nonhuman primate hosts like the sooty mangabeys. CTL responses and sequence variation in the SIV nef gene were evaluated in one sooty mangabey and one rhesus macaque inoculated together with the same stock of cloned SIVmac239. Each animal developed an immunodominant response to a distinct CTL epitope in Nef, aa 157-167 in the macaque and aa 20-28 in the mangabey. Nonsynonymous mutations in their respective epitopes were observed in both animals and resulted in loss of CTL recognition. These mutations were present in the majority of proviral DNA sequences at 16 weeks post infection in the macaque and >2 years post infection in the mangabey. These results document the occurrence of CTL escape in a host that does not develop AIDS, and adds to the growing body of evidence that CTL exert significant selective pressure in SIV infection.


Subject(s)
Mutation , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/genetics , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Cercocebus atys , Epitopes, T-Lymphocyte , Macaca mulatta , Molecular Sequence Data , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Viral Load , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/immunology
5.
J Virol ; 75(5): 2262-75, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11160730

ABSTRACT

African green monkeys can maintain long-term persistent infection with simian immunodeficiency viruses (SIVagm) without developing AIDS and thus provide an important model for understanding mechanisms of natural host resistance to disease. This study assessed the levels and anatomic distribution of SIVagm in healthy, naturally infected monkeys. Quantitative competitive reverse transcriptase PCR assays developed to measure SIVagm from two African green monkey subspecies demonstrated high levels of SIV RNA in plasma (>6 x 10(6) RNA copies/ml) in sabaeus and vervet monkeys. Infectious virus was readily recovered from plasma and peripheral blood mononuclear cells and shown to be highly cytopathic in human cell lines and macrophages. SIVagm DNA levels were highest in the gastrointestinal tract, suggesting that the gut is a major site for SIVagm replication in vivo. Appreciable levels of virus were also found within the brain parenchyma and the cerebrospinal fluid (CSF), with lower levels detected in peripheral blood cells and lymph nodes. Virus isolates from the CSF and brain parenchyma readily infected macrophages in culture, whereas lymph node isolates were more restricted to growth in human T-cell lines. Comparison of env V2-C4 sequences showed extensive amino acid diversity between SIVagm recovered from the central nervous system and that recovered from lymphoid tissues. Homology between brain and CSF viruses, macrophage tropism, and active replication suggest compartmentalization in the central nervous system without associated neuropathology in naturally infected monkeys. These studies provide evidence that the nonpathogenic nature of SIVagm in the natural host can be attributed neither to more effective host control over viral replication nor to differences in the tissue and cell tropism from those for human immunodeficiency virus type 1-infected humans or SIV-infected macaques.


Subject(s)
Immunity, Innate , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/pathogenicity , Amino Acid Sequence , Animals , Brain/virology , Cerebrospinal Fluid/virology , Chlorocebus aethiops , Gene Products, env/chemistry , Gene Products, env/genetics , Humans , Lymphoid Tissue/virology , Macrophages/virology , Molecular Sequence Data , RNA, Viral/analysis , RNA, Viral/blood , Sequence Analysis, DNA , Simian Acquired Immunodeficiency Syndrome/physiopathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/isolation & purification , Simian Immunodeficiency Virus/physiology , Virus Replication
6.
J Infect Dis ; 182(2): 435-41, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10915073

ABSTRACT

Bacteriophage phiX174 immunization was used to measure CD4(+) T cell function in vivo in human immunodeficiency virus (HIV)-infected patients across all disease stages. Function was evaluated by measuring the ability of T cells to provide help to B cells in antibody production, amplification, and isotype switching. A total of 33 patients and 10 controls received 3 bacteriophage phiX174 immunizations 6 weeks apart. The patients' responses regarding bacteriophage-specific total antibody titers and IgG titers were quantitatively and qualitatively inferior to the controls' responses. Overall, 7 of 33 patients had normal T cell function. Baseline CD4 counts provided the strongest correlation with total antibody and IgG titers. HIV RNA had a weaker association with responses but had some predictive power among patients with a CD4 count >200 cells/microL. Bacteriophage phiX174 immunization seems to be a useful tool for measuring immune function in vivo, which suggests that most HIV-infected patients may have abnormal CD4(+) T cell function despite adequate antiretroviral treatment.


Subject(s)
HIV Infections/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adolescent , Adult , Antibody Formation , B-Lymphocytes/immunology , Bacteriophage phi X 174/immunology , Female , HIV Long-Term Survivors , Humans , Immunoglobulin Class Switching , Immunologic Memory , Male , T-Lymphocyte Subsets , Vaccination
7.
Eur J Immunol ; 30(4): 1145-53, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10760804

ABSTRACT

The thymus is the primary organ responsible for the production of mature TCR alpha / beta T cells. Quantification of a DNA excision circle that is produced during TCR rearrangement, termed a signal joint TCR rearrangement excision circle (sjTREC) can be used as a measure of thymic function. Here sjTREC measurement has been applied to two monkey species used as animal models of human disease, rhesus macaques (Asian origin) and sooty mangabeys (African origin). Initial PCR analysis determined that the TCR deltaRec-PsiJalpha rearrangement leading to sjTREC formation occurs in both species. Primers to a DNA sequence conserved in macaques, mangabeys and humans were used in a quantitative competitive PCR assay to quantify sjTREC. We found that as in humans, sjTREC in these two monkey species decline with age. sjTREC are first generated in thymocytes during the early stages of TCR rearrangement. Lymph node CD4(+) and CD8(+) T cells contain more sjTREC than peripheral blood T cell populations, suggesting that recent thymic emigrants home to the lymphoid tissues. The sjTREC level is significantly higher within the peripheral blood CD4(+) and CD8(+) T cells of mangabeys compared to macaques. Removal of the thymus in four macaques led to a profound decrease in peripheral blood sjTREC level by 1 year post-thymectomy, indicating the lack of a significant extra-thymic source of peripheral naive T cells in macaques. Our results indicate that production, trafficking, and proliferation of recent thymic emigrants in these two monkey species represents a useful animal model system for understanding human immunological disorders.


Subject(s)
Cercocebus atys/immunology , Gene Rearrangement, T-Lymphocyte/genetics , Lymphoid Tissue/immunology , Macaca mulatta/immunology , Receptors, Antigen, T-Cell/genetics , Thymus Gland/immunology , Aging/immunology , Aging/metabolism , Animals , Base Sequence , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cercocebus atys/genetics , Conserved Sequence/genetics , DNA Primers/genetics , Disease Models, Animal , Flow Cytometry , Gene Rearrangement, T-Lymphocyte/immunology , Humans , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Macaca mulatta/genetics , Molecular Sequence Data , Polymerase Chain Reaction , Receptors, Antigen, T-Cell/immunology , Thymectomy , Thymus Gland/cytology , Thymus Gland/metabolism
9.
J Virol ; 73(6): 4829-39, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10233944

ABSTRACT

To elucidate the relationship between early viral infection events and immunodeficiency virus disease progression, quantitative-competitive and branched-DNA methods of simian immunodeficiency virus (SIV) RNA quantitation were cross-validated and used to measure viremia following infection of rhesus macaques with the pathogenic SIVmac251 virus isolate. Excellent correlation between the methods suggests that both accurately approximate SIV copy number. Plasma viremia was evident 4 days postinfection, and rapid viral expansion led to peak viremia levels of 10(7) to 10(9) SIV RNA copies/ml by days 8 to 17. Limited resolution of primary viremia was accompanied by relatively short, though variable, times to the development of AIDS (81 to 630 days). The persistent high-level viremia observed following intravenous inoculation of SIVmac251 explains the aggressive disease course in this model. Survival analyses demonstrated that the disease course is established 8 to 17 days postinfection, when peak viremia is observed. The most significant predictor of disease progression was the extent of viral decline following peak viremia; larger decrements in viremia were associated with both lower steady-state viremia (P = 0.0005) and a reduced hazard of AIDS (P = 0.004). The data also unexpectedly suggested that following SIVmac251 infection, animals with the highest peak viremia were better able to control virus replication rather than more rapidly developing disease. Analysis of early viral replication dynamics should help define host responses that protect from disease progression and should provide quantitative measures to assess the extent to which protective responses may be induced by prophylactic vaccination.


Subject(s)
Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Viremia/virology , Virus Replication , Animals , Macaca mulatta , Polymerase Chain Reaction , RNA, Viral/blood
10.
Proc Natl Acad Sci U S A ; 96(9): 5169-72, 1999 Apr 27.
Article in English | MEDLINE | ID: mdl-10220437

ABSTRACT

Over the course of HIV-1 infection, the lymphoid follicles where the humoral immune response is generated initially increase in size and number and then progressively involute. In advanced disease, the network of the processes of follicular dendritic cells (FDCs) that serve as antigen repositories and anatomical substrate for B and T cells and antigen to interact is destroyed, contributing to the breakdown of the immune system. Because destruction of FDCs is associated with deposition of HIV-1, and much of the virus can be cleared from the network with antiretroviral therapy, we investigated the reversibility of damage. We measured the immunohistochemically stainable FDC compartment by quantitative image analysis, and we documented changes in this compartment at different stages of disease. We show that treatment, initiated even at advanced stages of HIV-1 disease, can slowly reverse pathological changes in the FDC network.


Subject(s)
Dendritic Cells/pathology , HIV Infections/pathology , HIV-1/isolation & purification , Lymphoid Tissue/pathology , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Infections/immunology , Humans , Lymphoid Tissue/drug effects
11.
Proc Natl Acad Sci U S A ; 95(11): 6314-9, 1998 May 26.
Article in English | MEDLINE | ID: mdl-9600962

ABSTRACT

CD4(+) T cell activation, required for virus replication in these cells, occurs in local microenvironmental domains in transient bursts. Thus, although most HIV originates from short-lived virus-producing cells, it is unlikely that chronic infection is generally sustained in rapid continuous cycles of productive infection as has been proposed. Such continuity of productive infection cycles would depend on efficient long-range transmission of HIV from one set of domains to another, in turn requiring the maintenance of sufficiently high concentrations of cell-free virus across lymphoid tissues at all times. By contrast, long-lived cellular sources of HIV maintain the capacity to infect newly activated cells at close range despite the temporal and spatial discontinuities of activation events. Such proximal activation and transmission (PAT) involving chronically and latently infected cells may be responsible for sustained infection, particularly when viral loads are low. Once CD4 cells are productively infected through PAT, they can infect other activated cells in their immediate vicinity. Such events propagate locally but generally do not spread systemically, unlike in the acute phase of the infection, because of the early establishment of protective anergy. Importantly, antiretroviral drug treatment is likely to differentially impact long-range transmission and PAT.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , HIV Infections/virology , HIV-1/physiology , Virus Replication , Humans , Lymphocyte Activation
12.
Nature ; 396(6712): 690-5, 1998 Dec 17.
Article in English | MEDLINE | ID: mdl-9872319

ABSTRACT

The thymus represents the major site of the production and generation of T cells expressing alphabeta-type T-cell antigen receptors. Age-related involution may affect the ability of the thymus to reconstitute T cells expressing CD4 cell-surface antigens that are lost during HIV infection; this effect has been seen after chemotherapy and bone-marrow transplantation. Adult HIV-infected patients treated with highly active antiretroviral therapy (HAART) show a progressive increase in their number of naive CD4-positive T cells. These cells could arise through expansion of existing naive T cells in the periphery or through thymic production of new naive T cells. Here we quantify thymic output by measuring the excisional DNA products of TCR-gene rearrangement. We find that, although thymic function declines with age, substantial output is maintained into late adulthood. HIV infection leads to a decrease in thymic function that can be measured in the peripheral blood and lymphoid tissues. In adults treated with HAART, there is a rapid and sustained increase in thymic output in most subjects. These results indicate that the adult thymus can contribute to immune reconstitution following HAART.


Subject(s)
Aging/physiology , HIV Infections/immunology , Thymus Gland/physiology , Adolescent , Adult , Aged , Aging/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Case-Control Studies , Child , Child, Preschool , Cohort Studies , Gene Rearrangement, T-Lymphocyte , HIV Infections/drug therapy , Humans , Infant , Infant, Newborn , Leukopoiesis , Middle Aged , Polymerase Chain Reaction , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Thymus Gland/cytology , Thymus Gland/drug effects
13.
AIDS Res Hum Retroviruses ; 13(13): 1077-81, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9282811

ABSTRACT

We constructed a recombinant human immunodeficiency virus type 1 (HIV-1) provirus called R7-GFP that expresses a modified form of a green fluorescent protein (GFP) from the jellyfish Aequorea victoria by substituting GFP-coding sequences for Nef-coding sequences. Alanine was substituted for serine at amino acid position 65 in the modified GFP, resulting in markedly increased fluorescence at an excitation wavelength of 488 nm as compared to wild-type GFP. The replication kinetics of R7-GFP were identical to that measured with an isogenic, nef-negative strain lacking GFP. Expression of GFP by replication-competent HIV-1 allowed simultaneous quantitation of viral infection and cell surface CD4 levels, revealing rapid and nearly complete CD4 downregulation on R7-GFP-infected PBMCs.


Subject(s)
CD4 Antigens/biosynthesis , HIV-1/growth & development , Leukocytes, Mononuclear/virology , Luminescent Proteins , Cells, Cultured , Down-Regulation , Flow Cytometry , Green Fluorescent Proteins , HIV-1/genetics , Humans , Leukocytes, Mononuclear/cytology , Luminescent Proteins/genetics , Luminescent Proteins/isolation & purification , Proviruses/genetics , Proviruses/growth & development , Virus Cultivation/methods
14.
J Infect Dis ; 176(2): 514-7, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9237721

ABSTRACT

Previous studies have shown that CD4-positive T cells vary in a predictable manner over 24 h. This diurnal variance has significant clinical implications. Recently, viral RNA measurements have been increasingly used as a standard marker in the management of human immunodeficiency virus (HIV)-infected patients. Little detailed analysis of the variability of this marker has been conducted. To define the variance of plasma HIV-1 RNA levels within days, 11 clinically stable patients with established HIV infection and a baseline viral RNA level >40,000 copies/mL were studied. Following the patients' admission to an inpatient research unit, plasma samples were obtained frequently over 48 h and analyzed for HIV-1 RNA levels by use of a quantitative branched chain DNA assay (bDNA). No diurnal pattern was detected. In these clinically stable patients, viral RNA levels exhibited a variance of approximately 0.4 log.


Subject(s)
Circadian Rhythm , HIV Infections/virology , HIV-1/isolation & purification , RNA, Viral/blood , Viral Load/methods , Adult , Analysis of Variance , Cohort Studies , Female , Genetic Techniques , HIV-1/genetics , Humans , Male , Middle Aged , Sample Size
15.
J Virol ; 71(7): 5495-504, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9188623

ABSTRACT

The dependence of human immunodeficiency virus type 1 (HIV-1) on its NF-kappaB binding sites (kappaB sites) for replication in transformed and primary T-cell targets was examined by infecting cells with HIV-1 reporter viruses containing kappaB site enhancer mutations. Viral transcription was measured either with luciferase-expressing HIV-1 that infects for a single round or by flow cytometric analyses with HIV-1 expressing placental alkaline phosphatase (PLAP) or green-fluorescent protein (GFP). Both PLAP- and GFP-expressing viruses spread from cell to cell and allowed analysis of viral gene expression patterns in single cells. Infection of a panel of T-cell lines with different basal levels of NF-kappaB demonstrated a direct correlation between the amount of constitutive nuclear NF-kappaB and the degree to which a wild-type virus outperformed kappaB site mutants. One T-cell line with a constitutively high level of nuclear NF-kappaB, PM1, showed a 20-fold decrease in transcription when its kappaB sites were mutated. In contrast, in a T-cell line with a low basal level of NF-kappaB, SupT1, mutation of the kappaB site in the enhancer had no effect on viral transcription or growth rate. Phytohemagglutinin-activated peripheral blood mononuclear cells showed a large dependence on the kappaB sites for optimal virus growth. Viruses without marker genes corroborated the finding that mutations to the kappaB sites impair virus production in cells with a high basal level of NF-kappaB. These data show that in T cells, HIV-1 can use NF-kappaB to enhance its growth but the virus is clearly able to grow in its absence.


Subject(s)
HIV Long Terminal Repeat , HIV-1/genetics , NF-kappa B/metabolism , Base Sequence , DNA, Viral , HIV Core Protein p24/analysis , HIV-1/growth & development , HIV-1/metabolism , HIV-1/physiology , Humans , Jurkat Cells , Molecular Sequence Data , Transcription, Genetic , Tumor Cells, Cultured , Virus Replication
16.
J Virol ; 71(5): 3641-51, 1997 May.
Article in English | MEDLINE | ID: mdl-9094638

ABSTRACT

Adult rhesus macaques infected with nef-defective simian immunodeficiency virus (SIV) exhibit extremely low levels of steady-state virus replication, do not succumb to immunodeficiency disease, and are protected from experimental challenge with pathogenic isolates of SIV. Similarly, rare humans found to be infected with nef-defective human immunodeficiency virus type 1 (HIV-1) variants display exceptionally low viral burdens and do not show evidence of disease progression after many years of infection. HIV-1 Nef induces the rapid endocytosis and lysosomal degradation of cell surface CD4 and enhances virus infectivity in primary human T cells and macrophages. Although expression of SIV Nef also leads to down-modulation of cell surface CD4 levels, no evidence for SIV Nef-induced enhancement of virus infectivity was observed in earlier studies. Thus, it remains unclear whether fundamental differences exist between the activities of HIV-1 and SIV Nef. To establish more clearly whether the SIV and HIV-1 nef gene products are functionally analogous, we compared the replication kinetics and infectivity of variants of SIVmac239 that either do (SIVnef+) or do not (SIV delta nef) encode intact nef gene products. SIVnef+ replicates more rapidly than nef-defective viruses in both human and rhesus peripheral blood mononuclear cells (PBMCs). As previously described for HIV-1 Nef, SIV Nef also enhances virus infectivity within each cycle of virus replication. As a strategy for evaluating the in vivo contribution of HIV-1 nef alleles and long terminal repeat regulatory sequences to the pathogenesis of immunodeficiency disease, we constructed SIV-HIV chimeras in which the nef coding and U3 regulatory regions of SIVmac239 were replaced by the corresponding regions from HIV-1/R73 (SIVR7nef+). SIVR7nef+ displays enhanced infectivity and accelerated replication kinetics in primary human and rhesus PBMC infections compared to its nef-defective counterpart. Converse chimeras, containing SIV Nef in an HIV-1 background (R7SIVnef+) also exhibit greater infectivity than matched nef-defective viruses (R7SIV delta nef). These data indicate that SIV Nef, like that of HIV-1, does enhance virus replication in primary cells in tissue culture and that HIV-1 and SIV Nef are functionally interchangeable in the context of both HIV-1 and SIV.


Subject(s)
Gene Products, nef/physiology , HIV/physiology , Simian Immunodeficiency Virus/physiology , Animals , Base Sequence , Cell Line , Humans , Macaca mulatta , Molecular Sequence Data , Virus Replication , nef Gene Products, Human Immunodeficiency Virus
17.
J Virol ; 71(5): 3776-87, 1997 May.
Article in English | MEDLINE | ID: mdl-9094653

ABSTRACT

We have developed an in vitro model to study the influence that human immunodeficiency virus type 1 (HIV-1) may have on the ability of T cells to respond to antigenic challenge. We have examined consequences of HIV-1 gene expression on T-cell activation in antigen-dependent T cells that have stably integrated copies of replication-defective proviral HIV-1. Virus production by HIV-infected, antigen-dependent T cells was induced in response to antigenic stimulation and then decreased as infected cells returned to a state of quiescence. Contrary to the predictions of models proposing that Nef alters signal transduction pathways in T lymphocytes and thereby alters cellular activation, Nef expression in antigen-dependent T-cell clones did not influence their proliferative responses to low or intermediate concentrations of antigen and did not affect other measures of T-cell activation, such as induction of interleukin 2 receptor alpha-chain expression and cytokine production. In addition, we found no evidence for alteration of T-cell responsiveness to antigen by the gag, pol, vif, tat, or rev gene of HIV-1.


Subject(s)
Genes, nef/physiology , HIV-1/genetics , Lymphocyte Activation , T-Lymphocytes/immunology , Antigens/immunology , Cells, Cultured , Cytokines/biosynthesis , Gene Expression , Hemagglutinins, Viral/physiology , Humans , Sensitivity and Specificity
18.
Curr Biol ; 7(3): R136-40, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9162473
20.
Lancet ; 348(9022): 239-46, 1996 Jul 27.
Article in English | MEDLINE | ID: mdl-8684203

ABSTRACT

Our understanding of the pathogenesis of AIDS has advanced considerably since the disease was first reported 15 years ago. We now know that the primary damage inflicted by HIV-1 is mainly brought about by active virus replication. With the advent of sensitive tools for monitoring HIV replication in vivo, an individual's risk of disease progression can be assessed early in the course of the infection and the efficacy of antiviral therapies can now be determined accurately and expeditiously. When used appropriately, potent combinations of antiviral drugs seem to be able to circumvent the inherent tendency of HIV-1 to generate drug-resistant viruses, the main reason for failure of all antiviral therapies, and are significantly more effective than earlier approaches. For the first time, rational approaches to contain and perhaps eliminate HIV-1 infection can be pursued.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Antiviral Agents/therapeutic use , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , Disease Progression , HIV Infections/drug therapy , HIV Infections/immunology , Humans , RNA, Viral/blood , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...