Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurovirol ; 7(3): 235-49, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11517398

ABSTRACT

Recent studies have shown that two CAAT/enhancer binding protein (C/EBP) sites are critically important for efficient human immunodeficiency virus (HIV) type 1 (HIV-1) replication within cells of the monocyte/macrophage lineage, a primary cell type infected by HIV-1 and a potentially important vehicle for transport of virus to the central nervous system (CNS). Given the relevance of HIV-1 LTR sequence variation with respect to HIV-1 replication within monocyte populations and the important role that monocyte tropism likely plays in HIV-1 infection of the brain, C/EBP site sequence variation was examined within peripheral blood- and brain-derived LTR populations. Brain-derived LTRs commonly possessed a C/EBP site I configuration (6G, comprised of a thymidine to guanosine substitution with respect to the clade B consensus sequence at position 6 of C/EBP site I) that leads to enhanced binding of C/EBP proteins over that observed with the HIV-1 clade B consensus sequence at this site. In contrast, the 6GC/EBP site I configuration appeared infrequently within sequenced peripheral blood-derived LTRs. In addition, C/EBP site II was even more highly conserved in brain-derived HIV-1 LTR populations than site I. This was not the case with peripheral blood-derived LTR C/EBP site II sequences. The high degree of C/EBP site II conservation in brain-derived LTRs was likely important in LTR regulation since the clade B consensus sequence conserved at C/EBP site II recruited high amounts of C/EBP family members. Transient transfection analyses indicated that conservation of the strong C/EBP site II in brain-derived LTRs was likely due to important interactions with Tat. Overall, brain-derived HIV-1 LTRs preferentially contained two highly reactive C/EBP binding sites, which may suggest that these sites play important roles in LTR-directed transcription during invasion and maintenance of HIV-1 in the central nervous system.


Subject(s)
AIDS Dementia Complex/virology , Brain/virology , CCAAT-Enhancer-Binding Proteins/metabolism , HIV Long Terminal Repeat/physiology , HIV-1/growth & development , Proteasome Endopeptidase Complex , AIDS Dementia Complex/metabolism , ATPases Associated with Diverse Cellular Activities , Base Sequence , Binding Sites/physiology , Brain/metabolism , Cells, Cultured , Consensus Sequence , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral/physiology , Humans , Monocytes/cytology , Monocytes/virology , Transcription, Genetic/physiology , Virus Replication
2.
Virology ; 274(2): 262-77, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10964770

ABSTRACT

It has been widely demonstrated that the human immunodeficiency virus type 1 (HIV-1) envelope, specifically the V3 loop of the gp120 spike, evolves to facilitate adaptation to different cellular populations within an infected host. Less energy has been directed at determining whether the viral promoter, designated the long terminal repeat (LTR), also exhibits this adaptive quality. Because of the unique nature of the cell populations infected during the course of HIV-1 infection, one might expect the opportunity for such adaptation to exist. This would permit select viral species to take advantage of the different array of conditions and factors influencing transcription within a given cell type. To investigate this hypothesis, the function of natural variants of the NF-kappaB-proximal Sp element (Sp site III) was examined in human cell line models of the two major cell types infected during the natural course of HIV-1 infection, T cells and monocytes. Utilizing the HIV-1 LAI molecular clone, which naturally contains a high-affinity Sp site III, substitution of low-affinity Sp sites in place of the natural site III element markedly decreased viral replication in Jurkat T cells. However, these substitutions had relatively small effects on viral replication in U-937 monocytic cells. Transient transfections of HIV-1 LAI-based LTR-luciferase constructs into these cell lines suggest that the large reduction in viral replication in Jurkat T cells, caused by low-affinity Sp site III variants, may result from reduced basal as well as Vpr- and Tat-activated LTR activities in Jurkat T cells compared to those in U-937 monocytic cells. When the function of Sp site III was examined in the context of HIV-1 YU-2-based LTR-luciferase constructs, substitution of a high-affinity element in place of the natural low-affinity element resulted in increased basal YU-2 LTR activity in Jurkat T cells and reduced activity in U-937 monocytic cells. These observations suggest that recruitment of Sp family members to Sp site III is of greater importance to the function of the viral promoter in the Jurkat T cell line as compared to the U-937 monocytic cell line. These observations also suggest that other regions of the LTR may compensate for Sp recruitment defects in specific cell populations.


Subject(s)
Gene Expression Regulation, Viral , HIV Long Terminal Repeat/genetics , HIV-1/genetics , NF-kappa B/metabolism , Sp1 Transcription Factor/metabolism , Virus Replication , Base Sequence , Binding, Competitive , Cells, Cultured , Cloning, Molecular , Consensus Sequence/genetics , DNA/genetics , DNA/metabolism , Gene Products, tat/metabolism , Gene Products, vpr/metabolism , Genes, Reporter , Genetic Variation/genetics , HIV-1/physiology , Humans , Jurkat Cells , Leukocytes, Mononuclear/virology , Mutation/genetics , Organ Specificity , Promoter Regions, Genetic/genetics , Response Elements/genetics , Transcriptional Activation , U937 Cells , tat Gene Products, Human Immunodeficiency Virus , vpr Gene Products, Human Immunodeficiency Virus
3.
J Neurovirol ; 6(1): 6-24, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10786993

ABSTRACT

Herpes simplex virus (HSV) type 1 (HSV-1) enters nerve endings during a primary infection, is transported to sensory ganglia, and establishes latency within nuclei of a subpopulation of neurons. The latent state is characterized by absence of detectable HSV-1 antigen, minimal transcription of productive cycle genes, and high expression of 1.5- and 2-kb viral transcripts, termed the major latency-associated transcripts (LATs), within nuclei of a subpopulation of infected neurons. Transcription within the HSV-1 LAT genomic locus has been reported to both facilitate the establishment of latency in additional neurons and to increase the frequency of spontaneous and induced viral reactivation in animal model systems. More recent evidence suggests some possible mechanisms that may explain the relationship between LATs and both reactivation and establishment of latency. This review summarizes general aspects of latency, but focuses on the structure, expression, and function of LATs, and the interaction between host transcriptional regulators and viral gene expression that may impact latency and reactivation. A model that incorporates evidence from a number of experimental studies is proposed that summarizes the involvement of the LAT locus in the biology of HSV-1 latency.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 1, Human/physiology , Neurons/metabolism , Neurons/virology , Virus Activation/genetics , Virus Latency/genetics , Animals , Disease Models, Animal , Humans , Mice , Models, Biological , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Transcription, Genetic/genetics , Virus Replication/genetics
4.
J Biomed Sci ; 5(6): 451-64, 1998.
Article in English | MEDLINE | ID: mdl-9845850

ABSTRACT

The herpes simplex virus type 1 (HSV-1) latency-associated transcript (LAT) promoter 1 (LP1) is an inducible and cell type-specific promoter involved in regulating the production of an 8.3-kb primary LAT transcript during acute and latent infection of peripheral sensory neurons and during subsequent virus reactivation. A number of cis-acting regulatory elements have been identified in LP1, including two cyclic-AMP (cAMP) response element (CRE)-like sequences, designated CRE-1 and CRE-2. CRE-1 has previously been shown to confer cAMP responsiveness to LP1 and to regulate reactivation of HSV-1 from latency in vivo. A role for CRE-2 in modulating inducible activity is not yet as clear; however, it has been shown to support basal expression in neuronal cells in vitro. Electrophoretic mobility shift (EMS) analyses demonstrate that the LP1 CRE-like elements interact with distinct subsets of neuronal ATF/CREB and Jun/Fos proteins including CREB-1, CREB-2, ATF-1, and JunD. The factor-binding properties of each LP1 CRE element distinguish them from each other and from a highly related canonical CRE binding site and the TPA response element (TRE). LP1 CRE-1 shares binding characteristics of both a canonical CRE and a TRE. LP1 CRE-2 is more unusual in that it shares more features of a canonical CRE site than a TRE with two notable exceptions: it does not bind CREB-1 very well and it binds CREB-2 better than the canonical CRE. Interestingly, a substantial proportion of the C1300 neuroblastoma factors that bind to CRE-1 and CRE-2 have been shown to be immunologically related to JunD, suggesting that the AP-1 family of transcription factors may be important in regulating CRE-dependent LP1 transcriptional activity. In addition, we have demonstrated the two HSV-1 LP1 CRE sites to be unique with respect to their ability to bind neuronal AP1-related factors that are regulated by cAMP. These studies suggest that both factor binding and activation of bound factors may be involved in cAMP regulation of HSV-1 LP1 through the CRE elements, and indicate the necessity of investigating the expression and posttranslational modification of a variety of ATF/CREB and AP-1 factors during latency and reactivation.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation, Viral , Herpesvirus 1, Human/physiology , Peptide Fragments , Protein Precursors , Proteins/genetics , Transcription Factor AP-1/genetics , Transcription Factors/genetics , Virus Latency/genetics , Activating Transcription Factor 1 , Cell Line , Humans , Promoter Regions, Genetic , Transforming Growth Factor beta1 , Virus Replication/genetics
5.
J Neurovirol ; 4(3): 312-23, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9639074

ABSTRACT

The HIV-1 LTR promoter proximal G/C box array has been demonstrated to function by interacting with the Sp1 transcription factor family whose members can act as either activators or repressors of transcription. In this regard, we have examined the interaction of the HIV-1 Sp binding sites with nuclear factors that are present in cell types that support HIV-1 replication, including those of lymphocytic, monocytic, and astrocytic origin. As determined by electrophoretic mobility shift (EMS) competition analyses using oligonucleotides containing the sequences of each of the Sp1 sites of HIV-1 strain LAI, the NF-kappaB-proximal Sp site (site III) displayed the highest binding activity compared to sites I and II with regard to Sp1 and related factors present in lymphocytic (Jurkat) and astrocytic (U-373 MG) nuclear extracts. Sp1 and two Sp3 isoforms were detected as the primary cellular constituents of DNA-protein complexes formed with the NF-kappaB-proximal site. Only modest differences in Sp1:Sp3 binding ratios were observed when this site was reacted with either astrocytic or lymphocytic nuclear extract. However, when nuclear extracts derived from two monocytic cell types that differ in the degree of differentiation were reacted with the HIV-1 LAI Sp site III, a large difference in Sp1 and Sp3 binding was observed. To determine if naturally occurring and replication-competent strains of HIV-1 contain base pair alterations within the Sp elements that affect the ability of the site to interact with Sp1 and related factors, a series of Sp site III variants were constructed and examined by EMS analyses. One of these sites, obtained from the published sequence of the YU-2 strain (a brain-derived macrophage tropic strain of HIV-1), displayed almost no Sp1 or Sp3 binding activity as a result of a single base pair alteration in Sp site III. This base-pair alteration, when placed in the context of an HIV-1 LAI LTR-luciferase construct, resulted in a 40-50% reduction in LTR activity in transiently transfected Jurkat and U-373 MG cells. Overall, these results suggest that specific G/C box sequence alterations present in the brain-derived HIV-1 variant YU-2, or possibly other brain-derived variants, may exhibit altered replication properties as a result of the low affinity of the NF-kappaB-proximal G/C box for members of the Sp transcription factor family.


Subject(s)
HIV Long Terminal Repeat/genetics , HIV-1/genetics , NF-kappa B/genetics , Promoter Regions, Genetic/physiology , Sp1 Transcription Factor/genetics , Astrocytes/cytology , Astrocytes/virology , Base Sequence , Brain/virology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/virology , Cell Nucleus/metabolism , Cells, Cultured , DNA, Viral/analysis , HIV-1/classification , HIV-1/growth & development , Humans , Jurkat Cells/virology , Mutagenesis, Site-Directed , NF-kappa B/metabolism , Protein Binding/genetics , Sp1 Transcription Factor/metabolism
6.
Virology ; 230(2): 381-91, 1997 Apr 14.
Article in English | MEDLINE | ID: mdl-9143295

ABSTRACT

The herpes simplex virus type 1 (HSV-1) latency-associated transcript (LAT) promoter 1 (LP1) is the only viral promoter that exhibits detectable transcriptional activity during a latent HSV infection. The LAT promoter-binding factor (LPBF) regulatory sequence (nucleotides -65 to -72 relative to the transcriptional start site of the 8.3-kb primary transcript) closely resembles the core recognition sequence required for binding members of the upstream stimulatory factor (USF)/major late transcription factor (MLTF) family. In this analysis, we demonstrate that oligonucleotides containing either the LPBF recognition sequence or the USF/MLTF recognition sequences from previously described promoters bind cellular factors which exhibit very similar mobilities in electrophoretic mobility shift (EMS) analyses. We also observe a high degree of similarity in competition profiles obtained in competition EMS analyses utilizing oligonucleotides containing recognition sequences for either LPBF or USF/MLTF. Furthermore, antibody supershift EMS analyses have demonstrated that the factors binding the LPBF or USF/MLTF recognition sites in these oligonucleotides are antigenically related, if not identical, and that greater than 90% of the LPBF-binding activity is antigenically related to USF. In addition, we demonstrate that both forms of in vitro translated USF proteins (43 and 44 kDa) bind to the LPBF recognition sequence within HSV-1 LP1. Taken together, these data indicate that USF is capable of binding to the HSV-1 LPBF recognition sequence and that USF is a major LPBF-binding activity in cells of neuronal and nonneuronal lineage. These data further support the hypothesis that USF may indeed play a significant role in the transcriptional activity of HSV-1 LP1.


Subject(s)
DNA-Binding Proteins , Herpesvirus 1, Human/genetics , Promoter Regions, Genetic , Transcription Factors/metabolism , Binding Sites , Herpesvirus 1, Human/physiology , Humans , Transcription, Genetic , Upstream Stimulatory Factors , Virus Latency
7.
Virology ; 216(1): 284-7, 1996 Feb 01.
Article in English | MEDLINE | ID: mdl-8615004

ABSTRACT

The tat gene product of the human immunodeficiency virus type 1 (HIV-1) strongly induces the transcription directed by the viral long terminal repeat (LTR). Tat acts by interacting with a target RNA element located immediately downstream of the initiation site. In addition, the action of Tat appears to be assisted by the upstream DNA enhancer elements, including the binding sites for the NF-kappa B/Rel family of host transcription factors. In the present study, we demonstrate that Tat transactivation of the HIV-1 LTR is markedly inhibited by several cytoplasmic inhibitors of NF-kappa B/Rel, suggesting the critical involvement of these host transcription factors in the function of the viral Tat protein. Furthermore, the various NF-kappa B inhibitors appear to have differential effects on Tat. While I kappa B alpha, I kappa B beta, and p100 potently inhibit Tat-mediated transactivation, p105 fails to inhibit, but even moderately synergizes, the action of Tat. We further demonstrate that the action of these NF-kappa B/Rel inhibitors on Tat correlates with their inhibitory activities on the RelA subunit of NF-kappa B. Finally, we show that a degradation-resistant I kappa B alpha mutant is able to potently inhibit Tat-mediated activation of the HIV-1 LTR in both untreated and tumor necrosis factor alpha-stimulated T cells, thus suggesting that such an I kappa B alpha mutant may serve as a constitutive repressor of HIV-1 LTR.


Subject(s)
Gene Products, tat/metabolism , HIV Long Terminal Repeat/genetics , HIV-1/genetics , Proto-Oncogene Proteins/metabolism , Transcription Factors , Transcriptional Activation , Cell Line , Humans , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Transcription Factor RelB , tat Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...