Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Minireviews
    • JVI Classic Spotlights
    • Archive
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About JVI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
Journal of Virology
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Minireviews
    • JVI Classic Spotlights
    • Archive
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About JVI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
Genome Replication and Regulation of Viral Gene Expression

TFII-I Regulates Induction of Chromosomally Integrated Human Immunodeficiency Virus Type 1 Long Terminal Repeat in Cooperation with USF

Jiguo Chen, Tom Malcolm, Mario C. Estable, Robert G. Roeder, Ivan Sadowski
Jiguo Chen
1Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Tom Malcolm
1Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Mario C. Estable
2Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
3Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, New York
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Robert G. Roeder
3Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, New York
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ivan Sadowski
1Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: sadowski@interchange.ubc.ca
DOI: 10.1128/JVI.79.7.4396-4406.2005
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) replication is coupled to T-cell activation through its dependence on host cell transcription factors. Despite the enormous sequence variability of these factors, several cis elements for host factors are highly conserved within the 5′ long terminal repeats (LTRs) of viruses from AIDS patients; among these is the RBEIII upstream element for the Ras response element binding factor 2 (RBF-2). Here we show that RBF-2 is comprised of a USF1/USF2 heterodimer and TFII-I, which bind cooperatively to RBEIII. Recombinant USF1/USF2 binds to the RBEIII core sequence 160-fold less efficiently than it binds to an E box element, but the interaction with RBEIII is stimulated by TFII-I. Chromosomally integrated HIV-1 LTRs bearing an RBEIII mutation have slightly elevated basal transcription in unstimulated Jurkat cells but are unresponsive to cross-linking of the T-cell receptor or stimulation with phorbol myristate acetate (PMA) and ionomycin. Induction is inhibited by dominant interfering USF and TFII-I but not by the dominant negative I-κB protein. USF1, USF2, and TFII-I bind to the integrated wild-type LTR in unstimulated cells and become phosphorylated during the induction of transcription upon stimulation with PMA. These results demonstrate that USF1/USF2 and TFII-I interact cooperatively at the upstream RBEIII element and are necessary for the induction of latent HIV-1 in response to T-cell activation signals.

Transcription from the human immunodeficiency virus (HIV) long terminal repeat (LTR) is tightly coupled to T-cell activation signals. The dependence of HIV-1 replication on T-cell signaling may contribute to the development of a population of latently infected cells with chromosomally integrated proviral DNA, and conversely, causes a reactivation of replication in cells that are stimulated by exposure to an antigen (25). Expression of provirus in unstimulated T cells prior to expression of the viral transactivator TAT is completely dependent upon host cell regulatory factors (30). The functional significance of many host factors with respect to latency is unknown because most experiments have been performed with transiently transfected HIV-1 LTR templates, which are not properly assembled into chromatin (1). Furthermore, the high rate of viral turnover in infected individuals generates extensive sequence polymorphisms, and relatively few binding sites for transcription factors are highly conserved among isolates from HIV-infected persons (Table 1 and Fig. 1A) (19).

  • Open in new tab
  • Download powerpoint
  • Open in new tab
  • Download powerpoint
FIG. 1.

Purified RBF-2 contains USF1, USF2, and TFII-I. (A) Locations of binding sites for USF, RBF-2, NF-κB (κB), SP1, and TATA binding protein (IID) on the HIV-1 LTR. (B) Samples from purification of RBF-2 were analyzed by EMSA with the P3 RBEIII oligonucleotide. Jurkat T-cell nuclear extracts (lane 1) were fractionated by chromatography on heparin agarose (lane 2), Mono-Q (lane 3), poly(dI-dC) (not shown), RBEIII mutant oligonucleotide (not shown), and WT RBEIII oligonucleotide affinity columns (flowthrough, lanes 4 and 5; 100 mM KCl wash, lane 6; 200 mM KCl wash, lane 7; 400 mM KCl elution, lane 8). (C) Fractions from panel B were analyzed by immunoblotting with antibodies against the indicated proteins.

View this table:
  • View inline
  • View popup
TABLE 1.

Conservation of cis elements in the HIV-1 LTR

One of the most highly conserved sequences within the 5′ LTRs of viral isolates (Table 1) and within LTRs amplified from patient samples (13, 19, 26) represents a binding site for the Ras response element binding factor 2 (RBF-2) (Fig. 1A). RBF-2 was identified as a nuclear factor that binds elements which are necessary for the responsiveness of the LTR to activated protein tyrosine kinases and v-Ras (2, 12). Ras is essential but not sufficient for T-cell activation and acts in parallel with pathways regulated by calcineurin and protein kinase Cθ (18). RBF-2 appeared to be unrelated to factors that were previously shown to mediate Ras-responsive transcription (12) and is notable because its upstream cis element (RBEIII) is duplicated within the most frequently occurring length polymorphism of the HIV-1 LTR in up to 38% of infected patients (13, 22). In this work, we show that RBF-2 is comprised of USF1 and USF2, whose interaction with the highly conserved RBEIII requires TFII-I as a cofactor.

USF1 and USF2 are related basic-region helix-loop-helix-leucine zipper DNA binding factors that are constitutively expressed in most cell types and that bind E box (CACGTG) cis elements predominately as heterodimers (43). The USFs can exert positive or negative effects on transcription, depending on the promoter context and cell type specificity (24). Previous reports implicating a role for USF in the regulation of HIV-1 transcription have focused on the significance of an upstream E box (−160) (Fig. 1A) (11, 28, 40). This element is not particularly well conserved in viral isolates (Table 1) or within LTRs amplified from patient samples (13, 26). Nonetheless, overexpression of USF activates transcription from all HIV-1 LTR subtypes in T cells, independent of the upstream E box, but causes an E-box-dependent inhibition of transcription in HeLa cells (28). These observations demonstrate the context-dependent potential of USF function and also suggest that HIV-1 LTRs universally contain additional information conferring responsiveness to USF.

Part of the E-box-independent function of USF for HIV-1 transcription may involve its ability to activate transcription from initiator elements in cooperation with TFII-I (11). TFII-I was identified as an initiator-binding protein, but more recently has been found in complexes bound to upstream enhancer elements in association with a variety of transcription factors, including USF1, SRF, Phox1, NF-κB, Myc, STAT1, STAT3, and YY1 (35). TFII-I is phosphorylated at serine/threonine and tyrosine residues in response to cell signaling (6, 20, 21). In addition to functioning as a signal-responsive activator, recent experiments indicate that TFII-I might also contribute to the repression of transcription through interactions with histone deacetylases (49). The regulation of TFII-I by multiple signaling pathways, in combination with its ability to interact cooperatively with additional factors to cause a repression or activation of transcription, positions this factor as an important coordinator of cellular responses to a variety of stimuli.

Consistent with its defined role in regulating signal-responsive expression in fibroblasts and B cells, in this work we show that the expression of chromosomally integrated HIV-1 in response to T-cell activation signals requires TFII-I in combination with USF1 and -2. This function is mediated through binding to the stringently conserved upstream cis element RBEIII. Therefore, these observations indicate an important role for a TFII-I/USF1/USF2 complex in the pathology of AIDS as a key regulator of the reactivation of replication from latency upon T-cell activation.

MATERIALS AND METHODS

Recombinant DNA molecules.Details of plasmids for the expression of proteins in insect and mammalian cells and of the reporter genes will be described elsewhere. Plasmids expressing USF-2 (pCDNA-USF2) and the dominant negative USF (pCMV566-A-USF), TFII-I (pEB-P70), and I-κB (pSVK3-IKBα2N) proteins were described previously (7, 23, 33).

Cell culture, transfections, metabolic labeling, and immunoprecipitation.T-lymphoblastoid Jurkat cells were grown in RPMI 1640, and COS-1 cells were grown in Dulbecco's modified Eagle medium supplemented with 10% heat-inactivated fetal bovine serum at 37°C in a 5% CO2 atmosphere. Jurkat cell lines with stably integrated wild-type (WT) and RBEIII mutant HIV-1 LTR-luciferase reporters were established by transfection of the pGLneo-WT or pGLneo-M3 plasmid by use of the SuperFect transfection reagent (QIAGEN). The cells were washed with phosphate-buffered saline 48 h later, transferred to medium containing 800 μg of G-418 sulfate (Promega) per ml, and maintained with selection for 1 month. Cells were then grown as pools (WP and MP) in medium containing 100 μg of G-418 per ml or were cloned by limiting dilution. Jurkat cells stably expressing USF1-Flag were generated similarly by transfection with pcDNA-USF1. Transient transfections of Jurkat and COS-1 cells were performed by use of the SuperFect transfection reagent, and transfection efficiencies were normalized by cotransfection of an internal control plasmid, pCMV-galactosidase; activities were measured by use of a luciferase assay system (Promega), a luminescent β-galactosidase detection kit II (Clontech), and a microplate luminometer (Turner Designs) from a minimum of three separate transfections. To enrich for cells expressing dominant negative proteins, we cotransfected cells with the relevant expression plasmid and pEGFP (Clontech) at a 10:1 molar ratio. At 48 h posttransfection, green fluorescent protein (GFP)-expressing cells were isolated by fluorescence-activated cell sorting (FACS), and the luciferase activity was measured after treatment with phorbol-12-myristate-13-acetate (PMA; Sigma) and ionomycin (Sigma). Cells were stimulated with PMA at 25 ng/ml, ionomycin at 1 μM, tumor necrosis factor alpha (TNF-α; Sigma) at 10 ng/ml, and trichostatin A (TSA; Sigma) at 50 ng/ml. Stimulation by T-cell receptor cross-linking was performed as described previously (48) by use of the monoclonal antibody C305 against CD3. Cells were stimulated for 24 h before harvesting and measurement of luciferase activity. T-cell activation was monitored by FACS analysis with an anti-human CD69-phycoerythrin (PE) conjugate (BD Biosciences) (48).

For metabolic labeling, 7 × 106 Jurkat cells were washed in phosphate-depleted medium (Gibco) and incubated in the same medium for 2 h prior to the addition of 0.1 mCi of [32P]orthophosphate (ICN) per ml. Cells were labeled for a total of 3 h, with the addition of PMA 1 h prior to harvesting. The cells were lysed in RIPA buffer containing phosphatase inhibitors and protease inhibitor cocktail (Sigma), and USF1, USF2, and TFII-I were immunoprecipitated and analyzed by tryptic phosphopeptide analysis (39).

Protein expression and purification.Nuclear extraction from Jurkat cells and the purification of RBF-2 were performed as described previously (2, 14). USF proteins were produced by in vitro transcription and translation with the TNT T7 system (Promega), using template DNAs amplified from pFastbac-USF vectors with the primers indicated in Table 2. For the production of recombinant proteins in insect cells, SF9 cells were infected at a multiplicity of infection of 10 and harvested 4 days later. The cells were washed in phosphate-buffered saline and then lysed in buffer C (10 mM HEPES [pH 7.9], 20% [vol/vol] glycerol, 1.5 mM MgCl2, 420 mM KCl, 0.2 mM EDTA, 0.5 mM dithiothreitol, 0.5 mM phenylmethylsulfonyl fluoride) by sonication. An equal volume of buffer D (10 mM HEPES [pH 7.9], 20% [vol/vol] glycerol, 0.2 mM EDTA, 0.5 mM dithiothreitol, 0.5 mM phenylmethylsulfonyl fluoride) was added, and the extracts were clarified by centrifugation.

View this table:
  • View inline
  • View popup
TABLE 2.

Oligonucleotides used for this study

Immunoblotting, electrophoretic mobility shift, and chromatin immunoprecipitation assays.Rabbit polyclonal antibodies against USF1, USF2, c-fos, JunB, Myc-1, YY-1, ERK-1, Non-O, PSF, STAT-1, SRF, XBP-1, NF-κB, β-actin, and LexA and a mouse monoclonal antibody against Myc-1 were purchased from Santa Cruz Biotech. Polyclonal antibodies against TFII-I were produced against a glutathione S-transferase--TFII-I fusion protein (36) in mice. Electrophoretic mobility shift assay (EMSA) binding reactions were performed as described previously (2) and typically contained 2 pmol of labeled oligonucleotide probe, 2 μg of poly(dI-dC) (Sigma), 2 mg of bovine serum albumin, 10 mM HEPES [pH 7.9], 100 mM KCl, 5 mM MgCl2, 5% glycerol, and 2 to 5 μg of Jurkat nuclear extract, 0.1 to 1 μg of purified protein fractions, 4 μl of in vitro translation mix, or SF9 extract, as indicated, in a total volume of 20 μl. Labeled oligonucleotide probes were typically added to binding reactions after a 20-min preincubation of nuclear or insect cell extracts with the reaction components plus competitor oligonucleotides or specific antibodies. The sequences of the WT and mutant RBEIII-containing oligonucleotides P3 and C1 are shown in Fig. 2A. The E-box-containing oligonucleotide oJC033 (5′-CTAGAAAGACACGTGACATCGAGCTTTCTCCAAG-3′) was used as a probe and competitor for the experiment shown in Fig. 4. Chromatin immunoprecipitations were performed with the W26 and M2 Jurkat cell lines bearing WT or mutant HIV-1 LTR-luciferase reporters as described previously (3). Primers for the amplification of the HIV-1 LTR upstream region and β-globin promoter are indicated in Table 2 and produce 220- and 321-bp products, respectively.

FIG. 2.
  • Open in new tab
  • Download powerpoint
FIG. 2.

Antibodies against USF1, USF2, and TFII-I inhibit DNA binding of RBF-2. (A) Sequences of oligonucleotides used for EMSAs. The location of the RBEIII element is boxed. Nucleotide substitutions in the CM1 and P3M mutant oligonucleotides are indicated in lowercase bold letters. (B) Purified RBF-2 was analyzed by EMSA with the P3 oligonucleotide probe. The unlabeled competitor P3 (lane 2) or P3M (lane 3) oligonucleotide was added at a 100-fold molar excess. Antibodies against the indicated proteins (top) were added to the binding reactions (lanes 4 to 15). The migration of RBF-2 is indicated (left). (C) A Jurkat nuclear extract was analyzed by EMSA with the P3 oligonucleotide probe. The unlabeled competitor C1 (lane 2), CM1 (lane 3), P3 (lane 4), or P3M (lane 5) oligonucleotide was added at a 100-fold molar excess. Antibodies against the indicated proteins (top) were added to the binding reactions (lanes 6 to 13). In addition to RBF-2, YY1 bound to the 3′ end of the P3 oligonucleotide from Jurkat nuclear extracts (indicated with arrows).

RESULTS

RBF-2 is comprised of USF1, USF2, and TFII-I.RBF-2 was identified as a nuclear factor that binds specifically to the highly conserved RBEIII cis element (ACTGCTGA) (Fig. 1A), which was shown to be necessary for the responsiveness of HIV-1 to oncogenic protein tyrosine kinases and v-Ras (2, 12). Purified RBF-2 (14) bound specifically to RBEIII oligonucleotides in EMSAs (Fig. 1B, lane 8, and Fig. 2B, lane 1). Mutation of the core RBEIII sequence to ACTGCACT prevents the interaction of purified RBF-2 with RBEIII, as an oligonucleotide containing this mutation (P3M) (Fig. 2A) was unable to compete for binding with a WT probe in EMSAs (Fig. 2B, compare lanes 2 and 3). The P3M mutation impairs the responsiveness of transiently transfected LTR reporter templates to v-Ras (2, 12), consistent with the hypothesis that RBF-2 is a downstream target of tyrosine kinase and Ras signaling.

A fortuitous observation that an RBEIII oligonucleotide could compete with the binding of nuclear extract USF and purified recombinant USF-1 to an E-box element, along with the presence of RBF-2 preparations of a polypeptide equivalent in size to the USF-interacting TFII-I (14, 36, 37), suggested to us that RBF-2 might be composed, at least in part, of USF-1/USF-2 and TFII-I. Indeed, an immunoblot revealed this to be the case (Fig. 1C, lane 8). We observed SRF, STAT1, and Myc, proteins which are known to interact with TFII-I (35), in Jurkat T-cell nuclear extracts (Fig. 1C, lane 1) and RBF-2-containing fractions eluted from heparin agarose (lane 2), but STAT1 and SRF were lost upon purification by Mono-Q chromatography (lane 3), and Myc was not retained specifically by RBEIII oligonucleotide affinity (lanes 4 to 8).

Consistent with the observation that USF1, USF2, and TFII-I can be detected in purified fractions by immunoblotting, we also found that antibodies recognizing these factors prevented the binding of purified RBF-2 to an RBEIII oligonucleotide in EMSAs (Fig. 2B, lanes 4 to 6). In contrast, antibodies against a variety of other factors had no effect on the binding of purified RBF-2 (lanes 7 to 15). Additionally, the binding of RBF-2 from Jurkat nuclear extracts to RBEIII oligonucleotides was prevented by antibodies against USF1, USF2, and TFII-I (Fig. 2C, lanes 6 to 8). Taken together, these results demonstrate that RBF-2 is comprised, minimally, of USF1, USF2, and TFII-I.

RBF-2 can be produced in vitro as a USF1/USF2 heterodimer.We noticed that RBEIII is weakly related to an E box within the core sequence (ACTGCTGA). USF1 and USF2 produced by in vitro transcription and translation were each capable of binding to the RBEIII P3 oligonucleotide in an EMSA (Fig. 3A, lanes 2 and 4). USF1 and -2 exist predominately as heterodimers in most cell types (42), and accordingly, USF1/USF2 heterodimers were also capable of binding RBEIII in vitro (Fig. 3A, lane 6). When cotranslated, USF1 and USF2 produced a complex with an intermediate mobility (lane 6) compared to those of USF1 and USF2 produced individually (lanes 2 and 4). The mobility of the complex formed by in vitro-translated USF1/USF2 heterodimers was identical to that of the complex formed by RBF-2 from Jurkat nuclear extracts (Fig. 3A, compare lanes 6 and 10). Furthermore, the binding of both recombinant USF1/2 (see below) and RBF-2 could be competed by a WT RBEIII oligonucleotide (Fig. 3A, lane 11) but not by the mutant P3M oligonucleotide (lane 12). Importantly, however, the binding of RBF-2 to RBEIII was sensitive to antibodies against TFII-I (Fig. 3A, lane 15, and Fig. 2B and C) in addition to antibodies against USF1 and -2 (Fig. 3A, lanes 13 and 14), whereas in vitro-translated USF1/2 was sensitive to antibodies against USF1 and -2 (Fig. 3A, lanes 7 and 8) but not to antibodies against TFII-I (lane 9). We demonstrate below that the requirement for TFII-I is necessitated by the weak affinity of USF1/2 for the RBEIII element.

FIG. 3.
  • Open in new tab
  • Download powerpoint
FIG. 3.

Recombinant USF1 and USF2 bind RBEIII in vitro. (A) RBF-2 can be produced as a USF1/USF2 heterodimer. USF1 and USF2 were produced by translation in vitro, either separately (lanes 2 and 3 and lanes 4 and 5, respectively) or by cotranslation (lanes 6 to 9), and were assayed for binding to a labeled P3 oligonucleotide. For comparison, Jurkat nuclear extracts were analyzed with the same probe (lanes 10 to 15). Binding reaction mixtures contained a 100-fold molar excess of unlabeled P3 (lane 11) or P3M (lane 12) oligonucleotide or antibodies against USF1 (lanes 3, 7, and 13), USF2 (lanes 5, 8, and 14), or TFII-I (lanes 9 and 15). Migration of USF1 and USF2 homodimers, USF1/USF2 heterodimers, and RBF-2 is indicated. Lanes 6 to 9 and 10 to 13 are identical gels but were exposed for 5 or 1 h, respectively. (B) USF1/USF2 heterodimers produced in insect cells bind specifically to RBEIII in vitro. USF1 and USF2 coexpressed in insect cells were assayed for binding to the RBEIII P3 oligonucleotide probe. Binding reaction mixtures contained 0 to 100 pM total USF protein (lanes 1 to 8) or a 100 pM concentration of total USF protein (lanes 9 to 12) or a lysate from uninfected SF9 cells (lane 13). Antibodies against USF1 (lane 9), USF2 (lane 10), and the competitor oligonucleotide P3 (lane 11) or P3M (lane 12) were added to the binding reactions.

RBEIII is a low-affinity binding site for recombinant USF1 and USF2 in vitro.The coexpression of USF1 and USF2 in insect cells produced homodimers as well as USF1/2 heterodimers capable of binding RBEIII in EMSAs (Fig. 3B, lanes 1 to 8). However, this interaction was considerably weaker than binding to a consensus E box, as an unlabeled E box competitor oligonucleotide was found to compete for binding of USF proteins to the RBEIII element more avidly than the WT RBEIII oligonucleotide (Fig. 4A, compare lanes 4 and 5 to lanes 7 to 9). Conversely, an unlabeled RBEIII oligonucleotide was not as efficient at competing for binding to an E box probe (Fig. 4A, lanes 16 to 18). Competition titration experiments indicated that USF binds approximately 160-fold less efficiently to an RBEIII oligonucleotide than to a consensus E-box-containing oligonucleotide (Fig. 4B). Thus, although recombinant USF proteins are capable of binding the RBEIII element in vitro, their affinity for this element is significantly less than that for a canonical E box element.

FIG. 4.
  • Open in new tab
  • Download powerpoint
FIG. 4.

RBEIII is a low-affinity binding site for USF in vitro. (A) EMSAs were performed with 50 pM USF1 and USF2, produced by coinfection of SF9 cells, and a labeled oligonucleotide P3 probe (lanes 1 to 9) or E box probe (lanes 10 to 18). An unlabeled E box competitor oligonucleotide was added at a 12.5- or 25-fold (lanes 4 and 5) or 25-, 50-, or 100-fold molar excess (lanes 13 to 15). The RBEIII oligonucleotide P3 competitor was added in a 25-, 50-, or 100-fold molar excess (lanes 7 to 9 and 16 to 18). Antibodies against USF2 or USF1 were included in lanes 1 and 2, respectively. (B) Competition experiments by EMSA with coexpressed USF1 and USF2 and a labeled RBEIII oligonucleotide P3 probe. Binding reactions contained 50 pM USF1/USF2 and an unlabeled RBEIII P3 competitor (○, □, and ▵) or E box competitor (•, ▪, ▴) oligonucleotide at the indicated molar excess (x axis). The amounts of probe bound to USF1 (▪ and □), USF2 (▴ and ▵), and the USF1/USF2 heterodimer (• and ○) were quantitated from phosphorimaging scans and presented as the proportions of probe bound in the sample without competitor. Inset, expanded view of data for E box competitor from 0 to 0.8-fold molar excess.

TFII-I promotes interaction of USF1/USF2 with RBEIII in vitro.TFII-I is capable of binding DNA on its own to initiator and E box consensus elements (36). We found that recombinant TFII-I is also capable of binding to RBEIII in EMSAs (not shown). Moreover, as indicated above, TFII-I was specifically retained on an RBEIII oligonucleotide affinity column, although significant amounts were detected in the flowthrough and early wash fractions (Fig. 1B, lanes 4 to 6). Although capable of a weak interaction with RBEIII on its own, recombinant TFII-I promoted the binding of USF1, as well as USF1/USF2 heterodimers, to the RBEIII oligonucleotide in vitro (Fig. 5). EMSA reaction mixtures containing 5 pmol of USF1 produced a barely detectable complex with the labeled P3 oligonucleotide (Fig. 5A, lane 1), whereas recombinant TFII-I showed a dose-dependent stimulation of binding of USF1 to RBEIII (Fig. 5A, lanes 2 to 9, and D). In contrast, the addition of TFII-I to EMSA reaction mixtures containing USF2 did not cause an increase in binding to the P3 RBEIII oligonucleotide (Fig. 5B, lanes 2 to 9, and D). The addition of TFII-I to coexpressed USF1/USF2 enhanced the formation of USF1 homodimer, USF1/USF2 heterodimer, and to a lesser extent, USF2 homodimer complexes with the P3 oligonucleotide (Fig. 5C, lanes 2 to 7, and D). Thus, the stimulatory effect of TFII-I on USF binding seems to be mediated predominately through USF1. This observation may be consistent with the finding that an interaction between USF1 and TFII-I, but not between USF2 and TFII-I, can be detected in Jurkat cells by coimmunoprecipitation (see below; also data not shown). In EMSA binding reactions in which TFII-I was limiting, the inclusion of antibodies against TFII-I inhibited the binding of all USF species to RBEIII (Fig. 5C, lane 10). These results are consistent with the observation that the binding of purified RBF-2 and RBF-2 present in Jurkat nuclear extracts to RBEIII can be inhibited by TFII-I antibodies (Fig. 2 and 3A), and they demonstrate that TFII-I can promote the binding of USF1 and USF1/USF2 heterodimers to the RBEIII element in vitro.

FIG. 5.
  • Open in new tab
  • Download powerpoint
FIG. 5.

TFII-I promotes binding of USF1 and USF1/USF2 to RBEIII in vitro. (A) EMSA binding reactions with labeled RBEIII oligonucleotide P3 were performed with 5 pM USF1-Flag and TFII-I at increasing concentrations (0 to 10 pM, lanes 1 to 9) or with 10 pM TFII-I (lanes 11 to 14). Binding reactions contained antibodies against USF1 (lane 10), the Flag epitope (lane 11), or TFII-I (lane 12) or an unlabeled P3 (lane 13) or P3M (lane 14) oligonucleotide. (B) EMSA binding reactions with RBEIII oligonucleotide P3 probe were performed with 5 pM USF2 and either 0 to 10 pM (lanes 1 to 9) or 10 pM (lanes 10 to 14) TFII-I. Binding reactions contained antibodies against USF2 (lane 10) or TFII-I (lane 11) or an unlabeled P3 (lane 12) or P3M (lane 13) oligonucleotide. (C) EMSA reactions with 10 pM coexpressed USF1 and USF2 and the RBEIII oligonucleotide P3 probe contained 0 to 10 pM TFII-I (lanes 1 to 6) or 5 pM TFII-I (lanes 7 to 10). The USF1/USF2 heterodimer complex and TFII-I complex migrated identically in the EMSA (indicated with an asterisk). Binding reactions contained antibodies against USF1 (lane 8), USF2 (lane 9), or TFII-I (lane 10). (D) EMSA reactions were performed as described for panels A to C. The relative amounts of RBEIII probe bound to USF1/2 heterodimers (less the contribution of signal from the binding of TFII-I [○]), USF1(□ and ▪), and USF2 (▵ and ▴) were quantitated from phosphorimager scans. Reactions contained the indicated molar ratios of TFII-I/total USF protein (x axis) and 10 pM coexpressed USF1/USF2 (○, □, and ▵) or 5 pM USF1 (▪) or USF2 (▴).

RBEIII is necessary for the response of integrated LTRs to T-cell activation.To determine the role of RBEIII in regulating the LTR in the context of an integrated provirus, we produced Jurkat cell lines with stably integrated WT or RBEIII mutant LTR-luciferase reporter genes (Fig. 6A). In general, stably transfected cloned Jurkat lines with RBEIII mutant LTR reporters had slightly higher basal expression than clones with WT LTR-luciferase reporter genes in unstimulated cells but were relatively unresponsive in cells stimulated with PMA and ionomycin in combination with the histone deacetylase inhibitor TSA (Fig. 6A). Jurkat clones with stably integrated WT LTR-luciferase reporters were typically induced 20- to 40-fold, but the mutant LTRs were induced only 2- to 3-fold (Fig. 6A). We chose representative WT (W26) and RBEIII mutant (M2) integrated LTR clones for further analysis (Fig. 6B). The activation of T cells through engagement of the T-cell receptor causes an induction of the WT integrated HIV-1 LTR through a combination of signaling mechanisms (18). We found that the RBEIII mutation significantly impairs induction of the integrated LTR in cells stimulated by cross-linking of the T-cell receptor (TCR) (Fig. 6B). Cell lines bearing the wild-type and mutant integrated LTR reporter genes were both stimulated by TCR cross-linking, as indicated by the expression of CD69, an early T-cell activation marker (Fig. 6C), but expression of the RBEIII mutant LTR was induced only approximately 4-fold, in contrast to a 25-fold induction for the WT integrated LTR (Fig. 6B). The RBEIII mutation also slightly impaired induction in cells stimulated with PMA alone (Fig. 6B). In contrast, the wild-type and RBEIII mutant integrated LTRs were induced to similar levels by treatment with TNF-α, alone or in combination with PMA (Fig. 6B). TNF-α activates NF-κB through IKK (29, 41), and thus this result demonstrates that the RBEIII mutation selectively impairs the activation of the LTR by RBF-2 but not by NF-κB.

FIG. 6.
  • Open in new tab
  • Download powerpoint
FIG. 6.

RBEIII is required for response of integrated HIV-1 LTR to T-cell activation signals. (A) Luciferase activities were measured from stable clones of Jurkat cells transfected with WT LTR-luciferase (W1, W2, W7, W14, W24, and W26) or RBEIII mutant LTR-luciferase (M1, M2, M3, M4, M10, and M20) or from pools of cells with stably transfected wild-type (WP) or RBEIII mutant (MP) reporters. Cells were untreated or were stimulated with PMA, TSA, and ionomycin. (B) Luciferase activities was measured in representative clones of stably transfected Jurkat cells bearing WT LTR-luciferase (W26, closed bars) or RBEIII mutant LTR-luciferase (M2, open bars) following treatment with TSA, TNF-α, PMA, PMA-TSA-ionomycin, T cell receptor cross-linking (TCR), or TNF-α--PMA. The results are presented as fold activities relative to untreated samples. (C) Stably transfected Jurkat clones bearing WT LTR-luciferase (W26, WT) or RBEIII mutant LTR-luciferase (M2, Mut.) were analyzed by FACS with anti-CD69 antibodies. Cells were unstimulated or were treated with PMA-TSA-ionomycin (PMA) or by cross-linking of the T-cell receptor (TCR).

USF and TFII-I are necessary for induction of the integrated HIV LTR in T cells.To address whether USF1, USF2, and TFII-I can function through the RBEIII element in vivo, we used luciferase reporter genes with four tandem repeats of the wild-type (pTA-P3WT) or mutant (pTA-P3mut) RBEIII element upstream of the minimal herpes simplex virus thymidine kinase core promoter. The WT RBEIII reporter had a significantly higher basal level of expression than the RBEIII mutant in transient transfections of COS-1 cells (Fig. 7A, vector). Cotransfection of a USF1 expression vector, alone or in combination with a USF2 expression vector, caused significantly elevated expression from the WT, but not the mutant, RBEIII reporter, indicating that USF1 can function in vivo through the RBEIII element.

FIG. 7.
  • Open in new tab
  • Download powerpoint
FIG. 7.

USF and TFII-I are required for induction of the integrated HIV-1 LTR. (A) Luciferase activities were measured from COS-1 cells cotransfected with minimal TK-luciferase reporter constructs bearing four upstream direct tandem repeats of the WT RBEIII oligonucleotide P3 (pTA-P3WT, closed bars) or the RBEIII mutant oligonucleotide P3M (pTA-P3mut, open bars) and plasmids expressing USF1, USF2, TFII-I, both USF1 and USF2, both USF1 and TFII-I, or a vector control (vector). (B) Jurkat clone W26 bearing the stably integrated WT LTR-luciferase reporter gene was cotransfected with a vector control or a plasmid expressing dominant negative USF (DN-USF), TFII-I (DN-TFII-I), or I-κb (DN-IκB) and a GFP expression plasmid at a 10:1 molar ratio. Transfected cells were enriched by sorting for GFP fluorescence, and luciferase activities were measured from unstimulated cells or following treatment with PMA-TSA-ionomycin. Activities are presented as fold stimulations relative to the unstimulated vector control.

Consistent with the observation that RBEIII is necessary for the response of the integrated LTR to T-cell activation signals, we found that the expression of dominant negative forms of USF and TFII-I prevented the stimulation of the LTR in cells treated with PMA, TSA, and ionomycin (Fig. 7B). In contrast, expression of the dominant negative IκB, which was previously shown to prevent the activation of NF-κB in Jurkat cells by TCR cross-linking (23, 46), had no effect (Fig. 7B). These results demonstrate that RBF-2 is necessary for activation of the integrated LTR by PMA and T-cell receptor engagement, but not for induction by TNF-α through the activation of NF-κB.

RBF-2 components are phosphorylated in PMA-stimulated Jurkat cells.USF1, USF2, and TFII-I are components of RBF-2 and are required for induction of the integrated HIV-1 LTR in response to PMA treatment. Accordingly, we observed the phosphorylation of USF1 and TFII-I in response to PMA treatment in Jurkat cells, as determined by metabolic labeling (Fig. 8A, lane 2, USF1 32P and TFII-I 32P). An analysis of the labeled proteins from PMA-treated cells indicated that both proteins are phosphorylated at three independent sites (Fig. 8C). USF2 is phosphorylated in unstimulated Jurkat cells (Fig. 8A, lane 1, USF2 32P) and is hyperphosphorylated in response to PMA treatment (lane 2, USF2 32P). Phosphopeptide analysis indicated that USF2 is phosphorylated at two different sites and that PMA treatment does not cause the appearance of novel phosphopeptides (not shown). We observed an additional labeled protein of approximately 120 kDa in immunoprecipitates of USF1 from PMA-treated Jurkat cells (Fig. 8A, lane 2, “*”). This protein appears to be TFII-I, as it generated an identical two-dimensional phosphopeptide fingerprint to that of TFII-I (data not shown; Fig. 8C, right panel). Also, we observed an interaction of TFII-I with USF1 in Jurkat cells by coimmunoprecipitation with Flag-tagged USF1 (Fig. 8B). Under similar conditions, we were not able to observe an interaction between USF2 and TFII-I (data not shown; Fig. 8A). Thus, all of the factors identified as components of RBF-2 become phosphorylated in response to the stimulation of Jurkat T cells with PMA, which is consistent with a role in regulating the response of HIV-1 to T-cell activation signals.

FIG. 8.
  • Open in new tab
  • Download powerpoint
FIG. 8.

USF1, USF2, and TFII-I are phosphorylated in PMA-stimulated T cells. (A) Jurkat cells were labeled with [32P]orthophosphate, and USF1 (top), USF2 (center), or TFII-I (bottom) was recovered by immunoprecipitation from unstimulated cells (lane 1) or cells treated with PMA (lane 2). The migration of labeled USF1 (USF1 32P), USF2 (USF2 32P), and TFII-I (TFII-I 32P) is indicated with arrows. The band indicated with an asterisk from USF1 immunoprecipitates was found to represent labeled TFII-I (not shown). Parallel unlabeled samples were analyzed by immunoblotting with antibodies against USF1 (α-USF1 WB), USF2 (α-USF2 WB), TFII-I (α-TFII-I WB), or actin (α-Actin WB). (B) In vivo interaction between USF1 and TFII-I. Lysates from Jurkat cells (lanes 1 and 3) or Jurkat cells stably expressing USF-Flag (lanes 2 and 4) were immunoprecipitated with anti-Flag antibodies, and the samples were analyzed by immunoblotting with antibodies against USF1 (lanes 1 and 2) or TFII-I (lanes 3 and 4). (C) Labeled USF1 (left) and TFII-I (right) from PMA-stimulated Jurkat cells were digested with trypsin, and phosphopeptides were analyzed by 2D fingerprinting. The origin is labeled (○), and phosphopeptides observed in PMA-treated cells are numbered (1 to 3).

TFII-I and USF1/2 are bound to the RBEIII region of the LTR in unstimulated cells.Using chromatin immunoprecipitation, we found that USF1, USF2, and TFII-I are bound to the upstream region (−220 to −1) (Fig. 1A) of the WT LTR in both unstimulated cells and cells treated with PMA (Fig. 9A, panels iii and iv). In contrast, TFII-I and USF2 were not observed on the RBEIII mutant LTR under either condition, and only a small amount of USF1 was observed in stimulated cells, likely reflecting an interaction with the upstream E box (Fig. 9A, panel ii). This demonstrates that RBEIII is necessary for the interaction of USF1, USF2, and TFII-I with the upstream LTR region in vivo. Furthermore, since USF1, TFII-I, and USF2 were bound in unstimulated cells, these factors must be available for recruiting complexes that are necessary for induction, concomitant with their modification by phosphorylation. Unlike the USFs and TFII-I, we only observed bound NF-κB in PMA-treated cells (Fig. 9A, panels iii and iv), and furthermore, the RBEIII mutation did not affect the interaction with the LTR in vivo (Fig. 9A, panels i and ii). Thus, although the RBEIII mutation impaired the binding of the RBF-2 components in vitro and in vivo, as well as induction by PMA and ionomycin, it did not prevent the binding of NF-κB. This observation supports the view that TFII-I, in combination with USF1 and USF2, plays an important role in the LTR response to T-cell activation signals independently of NF-κB.

FIG. 9.
  • Open in new tab
  • Download powerpoint
FIG. 9.

USF1, USF2, and TFII-I are bound to RBEIII on the integrated WT HIV-1 LTR in unstimulated cells. (A) Chromatin immunoprecipitation was performed with formaldehyde cross-linked stable Jurkat clones bearing the RBEIII mutant LTR-luciferase (i and ii) or WT LTR-luciferase (iii and iv) reporter by the use of antibodies against USF1 (lane 3), USF2 (lane 4), TFII-I (lane 5), or NF-κB (lane 6). Samples were analyzed by PCR with oligonucleotides specific for the HIV-1 LTR flanking the RBEIII region (−220 to −1) and the β-globin promoter. A 100-bp ladder is shown in lane 1, and the input samples are shown in lane 2. Cells were untreated (i and iii) or stimulated with PMA-TSA-ionomycin (ii and iv).(B) The results from panel A were quantitated from a phosphorimager scan and normalized relative to the input sample and a β-globin internal control.

DISCUSSION

Replication of HIV-1 produces enormous sequence diversity, primarily because of error-prone reverse transcription (9). This variability underscores the importance of highly conserved cis elements within the LTR, including the binding site for RBF-2 (Table 1). Here we showed that RBEIII is a low-affinity binding site for USF and that RBF-2 is comprised of a USF1/USF2 heterodimer whose binding to RBEIII is dependent upon TFII-I. Mutation of RBEIII prevents induction of the integrated HIV-1 LTR by PMA and ionomycin and cross-linking of the TCR. Furthermore, the expression of dominant interfering USF and TFII-I proteins prevents induction of the wild-type integrated HIV-1 LTR in response to PMA and ionomycin. USF1, USF2, and TFII-I are also phosphorylated concomitant with induction of the LTR in PMA-treated Jurkat cells. These observations demonstrate a central role for these factors in regulating the induction of HIV-1 expression from latency.

With the understanding that RBF-2 is comprised of a USF1/USF2 heterodimer, it is interesting that a low-affinity binding site for USF is so stringently conserved in LTRs from AIDS patients, whereas an upstream high-affinity site is present in only 31% of viral sequences (Table 1). This suggests that cooperativity between USF and TFII-I at RBEIII is essential for HIV-1 replication. Cooperative interactions with cis elements are commonly employed for developmental decisions in both prokaryotes and eukaryotes (32). A requirement for cooperativity provides a mechanism for the integration of multiple signals at the RBEIII site, which might be important for the coordination of HIV-1 transcription with T-cell physiology. Additionally, USF1 and -2 are members of the basic-region helix-loop-helix-zip family of transcription factors, which also includes Myc, Max, and TFE3, all of which are E-box-binding proteins. Many of these factors are ubiquitously expressed, and consequently additional sequence features of their cis elements must be required for gene- and cell-type-specific regulation. In several instances, USF has been shown to activate transcription from E box elements cooperatively with factors bound to adjacent cis elements, including HNF-4 on the ApoA-II promoter (34) and C/EBPα on its own promoter (45). Additionally, activation of the HIV-1 LTR by USF from the upstream E box (Fig. 1A) requires a cooperative interaction with Ets (40). Several recent reports have demonstrated the regulation of genes by USF through noncanonical E box elements, including the polymeric immunoglobulin receptor (PIGR) (5), vasopressin (10), the FcεRI-α chain (44), fibroblast growth factor binding protein (17), and lama3 genes (47). In these cases, it has not been clearly defined whether this reflects preferential binding of USF to the noncanonical sequence or if binding involves a cooperative interaction with another factor.

Recombinant TFII-I is capable of stimulating the binding of USF1 and USF1/USF2 heterodimers to the RBEIII element in vitro (Fig. 5). This observation is consistent with previous results showing that TFII-I can stimulate the binding of USF1 to the adenovirus ML E box and initiator elements (36, 37). Recombinant USF1/USF2 heterodimers produce a complex with an identical mobility in EMSAs as RBF-2 from nuclear extracts (Fig. 3), but RBF-2 purified by specific oligonucleotide affinity chromatography from nuclear extracts contains TFII-I in addition to the USFs (Fig. 1). This supports the view that RBF-2 consists of a USF1/USF2 heterodimer that requires associated TFII-I for binding to RBEIII. Consistent with this model, antibodies against TFII-I prevent the binding of RBF-2 from Jurkat cells to RBEIII (Fig. 2), and they also inhibit the loading of USF onto RBEIII by recombinant TFII-I in vitro (Fig. 5). Because we did not observe a ternary complex containing TFII-I and USF by EMSA with recombinant proteins or with nuclear extracts, the mechanism for the stimulation of USF binding by TFII-I is not completely clear. TFII-I is capable of a weak interaction with RBEIII in vitro on its own (Fig. 5) (data not shown), and considering previous observations, this interaction is also likely to be stimulated by USF1 (36). The stimulation of USF binding by TFII-I likely results from a direct interaction between these proteins, as we observed an interaction between USF1 and TFII-I by coimmunoprecipitation (Fig. 8B). It is possible that a ternary complex formed by the cooperative interaction between these proteins at RBEIII is not sufficiently stable to be detected by EMSA.

HIV-1 replication is induced from latency by antigen-driven T-cell activation (31). Engagement of the T-cell receptor causes the activation of parallel pathways through the generation of the second messengers inositol triphosphate and diacylglycerol (18). Inositol triphosphate causes activation of the protein phosphatase calcineurin through the release of intracellular calcium, while in T cells, DAG activates protein kinase Cθ and the Ras-Raf-MEK-ERK pathway through RasGRP. T-cell activation is mediated by downstream transcription factors which are responsive to these parallel pathways (18). Mutations of RBEIII inhibit the activation of the LTR in response to T-cell receptor cross-linking and also in cells stimulated with PMA and ionomycin. Furthermore, induction of the wild-type integrated LTR by PMA and ionomycin is inhibited in cells expressing dominant negative forms of USF or TFII-I. We also observed phosphorylation of USF1 and TFII-I in PMA-treated T cells, along with hyperphosphorylation of USF2. These observations are consistent with the view that TFII-I and USF are downstream targets for signaling from the T-cell receptor and are necessary for the activation of transcription from latency. TFII-I is known to be regulated by Bruton’s tyrosine kinase-mediated phosphorylation in B cells (38) and by c-Src, JAK2, and ERK1 in response to growth factor stimulation in fibroblasts (6, 20, 21). Similarly, USF1 was shown to be phosphorylated by stress-activated protein kinase in melanocytes and fibroblasts (15) and by cdc2/cyclin B1 in HeLa cells (8). We have not determined whether the phosphorylation observed in T cells corresponds to these modifications, but considering that all three factors are bound to the wild-type LTR in unstimulated cells, we expect that phosphorylation will be involved in regulating their activities.

Most studies examining the induction of HIV-1 by T-cell signaling have focused on NF-κB, which activates transcription in T cells in response to a variety of stimuli, including phorbol esters (27), interleukin-1 (4), and TNF (16). The NF-κB binding sites within the enhancer region are highly conserved (Table 1), but viral isolates with deletions of this region have been reported (50). In our studies, the mutation of RBEIII severely impairs its responsiveness to T-cell receptor cross-linking and treatment with PMA and ionomycin, but not to stimulation by TNF-α alone or in combination with PMA (Fig. 6B). These observations demonstrate that latent integrated HIV-1 LTRs can be activated independently by RBF-2 and NF-κB. RBF-2 appears to be synergistically responsive to activation of the calcineurin and ERK pathways, as a treatment with both PMA and ionomycin caused significantly more stimulation of the WT LTR than did PMA alone (Fig. 6B). For RBF-2, the synergistic effect of PMA and ionomycin might be mediated through the cooperative interaction of USF and TFII-I, as has been demonstrated for the adenovirus ML promoter (36).

Our results demonstrate that TFII-I, in cooperation with USF1 and USF2 through their interaction with the upstream element RBEIII, contributes an essential function for the transition of integrated HIV-1 proviruses from a repressed latent state to active transcription in response to T-cell activation signals. These factors are bound to the LTR in unstimulated cells, presumably at RBEIII, where they might contribute to transcriptional repression. Upon T-cell activation by antigen presentation, TFII-I and the USFs become phosphorylated and thus allow transcriptional activation. The mechanisms regulating TFII-I and USF will be of interest for understanding the control of the establishment of latency and reactivation of HIV-1 replication in T cells, and these factors may represent potential targets for novel AIDS therapies.

ACKNOWLEDGMENTS

This work was supported by funds from the Canadian Institute for Health Research (to I.S.) and by a grant (AI37327) from the NIH (to R.G.R.). J.C. is a postdoctoral fellow of the Natural Sciences and Engineering Research Council of Canada (NSERC) and the Michael Smith Foundation for Health Research.

M.E. was a Postdoctoral Fellow of the Canadian Institute for Health Research. We thank A. Roy, M. Sawadogo, J. Hiscott, and C. Vinson for generous gifts of plasmid expression constructs, David Mitchell for comments on the manuscript, and Mojgan Naghavi for help with the original USF-oligonucleotide binding assays at The Rockefeller University.

FOOTNOTES

    • Received 25 September 2004.
    • Accepted 11 November 2004.
  • Copyright © 2005 American Society for Microbiology

REFERENCES

  1. 1.↵
    Archer, T. K., E. Zaniewski, M. L. Moyer, and S. K. Nordeen. 1994. The differential capacity of glucocorticoids and progestins to alter chromatin structure and induce gene expression in human breast cancer cells. Mol. Endocrinol.8:1154-1162.
    OpenUrlCrossRefPubMedWeb of Science
  2. 2.↵
    Bell, B., and I. Sadowski. 1996. Ras-responsiveness of the HIV-1 LTR requires RBF-1 and RBF-2 binding sites. Oncogene13:2687-2697.
    OpenUrlPubMedWeb of Science
  3. 3.↵
    Bell, B., E. Scheer, and L. Tora. 2001. Identification of hTAF(II)80 delta links apoptotic signaling pathways to transcription factor TFIID function. Mol. Cell8:591-600.
    OpenUrlCrossRefPubMed
  4. 4.↵
    Bird, T. A., K. Schooley, S. K. Dower, H. Hagen, and G. D. Virca. 1997. Activation of nuclear transcription factor NF-kappaB by interleukin-1 is accompanied by casein kinase II-mediated phosphorylation of the p65 subunit. J. Biol. Chem.272:32606-32612.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    Bruno, M. E., R. B. West, T. A. Schneeman, E. H. Bresnick, and C. S. Kaetzel. 2004. Upstream stimulatory factor but not c-Myc enhances transcription of the human polymeric immunoglobulin receptor gene. Mol. Immunol.40:695-708.
    OpenUrlCrossRefPubMedWeb of Science
  6. 6.↵
    Cheriyath, V., Z. P. Desgranges, and A. L. Roy. 2002. c-Src-dependent transcriptional activation of TFII-I. J. Biol. Chem.277:22798-22805.
    OpenUrlAbstract/FREE Full Text
  7. 7.↵
    Cheriyath, V., C. D. Novina, and A. L. Roy. 1998. TFII-I regulates Vbeta promoter activity through an initiator element. Mol. Cell. Biol.18:4444-4454.
    OpenUrlAbstract/FREE Full Text
  8. 8.↵
    Cheung, E., P. Mayr, F. Coda-Zabetta, P. G. Woodman, and D. S. Boam. 1999. DNA-binding activity of the transcription factor upstream stimulatory factor 1 (USF-1) is regulated by cyclin-dependent phosphorylation. Biochem. J.344:145-152.
    OpenUrlAbstract/FREE Full Text
  9. 9.↵
    Coffin, J. M. 1995. HIV population dynamics in vivo: implications for genetic variation, pathogenesis and therapy. Science267:483-489.
    OpenUrlAbstract/FREE Full Text
  10. 10.↵
    Coulson, J. M., J. L. Edgson, Z. V. Marshall-Jones, R. Mulgrew, J. P. Quinn, and P. J. Woll. 2003. Upstream stimulatory factor activates the vasopressin promoter via multiple motifs, including a non-canonical E-box. Biochem. J.369:549-561.
    OpenUrlCrossRefPubMed
  11. 11.↵
    Du, H., A. L. Roy, and R. G. Roeder. 1993. Human transcription factor USF stimulates transcription through the initiator elements of the HIV-1 and the Ad-ML promoters. EMBO J.12:501-511.
    OpenUrlPubMedWeb of Science
  12. 12.↵
    Estable, M. C., B. Bell, M. Hirst, and I. Sadowski. 1998. Naturally occurring human immunodeficiency virus type 1 long terminal repeats have a frequently observed duplication that binds RBF-2 and represses transcription. J. Virol.72:6465-6474.
    OpenUrlAbstract/FREE Full Text
  13. 13.↵
    Estable, M. C., B. Bell, A. Merzouki, J. S. Montaner, M. V. O'Shaughnessy, and I. J. Sadowski. 1996. Human immunodeficiency virus type 1 long terminal repeat variants from 42 patients representing all stages of infection display a wide range of sequence polymorphism and transcription activity. J. Virol.70:4053-4062.
    OpenUrlAbstract/FREE Full Text
  14. 14.↵
    Estable, M. C., M. Hirst, B. Bell, M. V. O'Shaughnessy, and I. Sadowski. 1999. Purification of RBF-2, a transcription factor with specificity for the most conserved cis-element of naturally occurring HIV-1 LTRs. J. Biomed. Sci.6:320-332.
    OpenUrlPubMed
  15. 15.↵
    Galibert, M. D., S. Carreira, and C. R. Goding. 2001. The Usf-1 transcription factor is a novel target for the stress-responsive p38 kinase and mediates UV-induced tyrosinase expression. EMBO J.20:5022-5031.
    OpenUrlAbstract
  16. 16.↵
    Griffin, G. E., K. Leung, T. M. Folks, S. Kunkel, and G. J. Nabel. 1989. Activation of HIV gene expression during monocyte differentiation by induction of NF-κB. Nature339:70-73.
    OpenUrlCrossRefPubMed
  17. 17.↵
    Harris, V. K., C. M. Coticchia, H. J. List, A. Wellstein, and A. T. Riegel. 2000. Mitogen-induced expression of the fibroblast growth factor-binding protein is transcriptionally repressed through a non-canonical E-box element. J. Biol. Chem.275:28539-28548.
    OpenUrlAbstract/FREE Full Text
  18. 18.↵
    Huang, Y., and R. L. Wange. 2004. T cell receptor signaling: beyond complex complexes. J. Biol. Chem.279:28827-28830.
    OpenUrlFREE Full Text
  19. 19.↵
    Jeeninga, R. E., M. Hoogenkamp, M. Armand-Ugon, M. de Baar, K. Verhoef, and B. Berkhout. 2000. Functional differences between the long terminal repeat transcriptional promoters of human immunodeficiency virus type 1 subtypes A through G. J. Virol.74:3740-3751.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    Kim, D. W., and B. H. Cochran. 2000. Extracellular signal-regulated kinase binds to TFII-I and regulates its activation of the c-fos promoter. Mol. Cell. Biol.20:1140-1148.
    OpenUrlAbstract/FREE Full Text
  21. 21.↵
    Kim, D. W., and B. H. Cochran. 2001. JAK2 activates TFII-I and regulates its interaction with extracellular signal-regulated kinase. Mol. Cell. Biol.21:3387-3397.
    OpenUrlAbstract/FREE Full Text
  22. 22.↵
    Koken, S. E., J. L. van Wamel, J. Goudsmit, B. Berkhout, and J. L. Geelen. 1992. Natural variants of the HIV-1 long terminal repeat: analysis of promoters with duplicated DNA regulatory motifs. Virology191:968-972.
    OpenUrlCrossRefPubMed
  23. 23.↵
    Kwon, H., N. Pelletier, C. DeLuca, P. Genin, S. Cisternas, R. Lin, M. A. Wainberg, and J. Hiscott. 1998. Inducible expression of IkappaBalpha repressor mutants interferes with NF-kappaB activity and HIV-1 replication in Jurkat T cells. J. Biol. Chem.273:7431-7440.
    OpenUrlAbstract/FREE Full Text
  24. 24.↵
    Luo, X., and M. Sawadogo. 1996. Functional domains of the transcription factor USF2: atypical nuclear localization signals and context-dependent transcriptional activation domains. Mol. Cell. Biol.16:1367-1375.
    OpenUrlAbstract/FREE Full Text
  25. 25.↵
    Lusic, M., A. Marcello, A. Cereseto, and M. Giacca. 2003. Regulation of HIV-1 gene expression by histone acetylation and factor recruitment at the LTR promoter. EMBO J.22:6550-6561.
    OpenUrlAbstract
  26. 26.↵
    Michael, N. L., L. D'Arcy, P. K. Ehrenberg, and R. R. Redfield. 1994. Naturally occurring genotypes of the human immunodeficiency virus type 1 long terminal repeat display a wide range of basal and Tat-induced transcriptional activities. J. Virol.68:3163-3174.
    OpenUrlAbstract/FREE Full Text
  27. 27.↵
    Nabel, G., and D. Baltimore. 1987. An inducible transcription factor activates expression of human immunodeficiency virus in T cells. Nature326:711-713.
    OpenUrlCrossRefPubMedWeb of Science
  28. 28.↵
    Naghavi, M. H., M. C. Estable, S. Schwartz, R. G. Roeder, and A. Vahlne. 2001. Upstream stimulating factor affects human immunodeficiency virus type 1 (HIV-1) long terminal repeat-directed transcription in a cell-specific manner, independently of the HIV-1 subtype and the core-negative regulatory element. J. Gen. Virol.82:547-559.
    OpenUrlPubMed
  29. 29.↵
    Ozes, O. N., L. D. Mayo, J. A. Gustin, S. R. Pfeffer, L. M. Pfeffer, and D. B. Donner. 1999. NF-kappaB activation by tumour necrosis factor requires the Akt serine-threonine kinase. Nature401:82-85.
    OpenUrlCrossRefPubMedWeb of Science
  30. 30.↵
    Pereira, L. A., K. Bentley, A. Peeters, M. J. Churchill, and N. J. Deacon. 2000. A compilation of cellular transcription factor interactions with the HIV-1 LTR promoter. Nucleic Acids Res.28:663-668.
    OpenUrlCrossRefPubMedWeb of Science
  31. 31.↵
    Persaud, D., Y. Zhou, J. M. Siliciano, and R. F. Siliciano. 2003. Latency in human immunodeficiency virus type 1 infection: no easy answers. J. Virol.77:1659-1665.
    OpenUrlFREE Full Text
  32. 32.↵
    Ptashne, M. 2003. Regulated recruitment and cooperativity in the design of biological regulatory systems. Philos. Trans. Ser. A361:1223-1234.
    OpenUrlCrossRefPubMed
  33. 33.↵
    Qyang, Y., X. Luo, T. Lu, P. M. Ismail, D. Krylov, C. Vinson, and M. Sawadogo. 1999. Cell-type-dependent activity of the ubiquitous transcription factor USF in cellular proliferation and transcriptional activation. Mol. Cell. Biol.19:1508-1517.
    OpenUrlAbstract/FREE Full Text
  34. 34.↵
    Ribeiro, A., D. Pastier, D. Kardassis, J. Chambaz, and P. Cardot. 1999. Cooperative binding of upstream stimulatory factor and hepatic nuclear factor 4 drives the transcription of the human apolipoprotein A-II gene. J. Biol. Chem.274:1216-1225.
    OpenUrlAbstract/FREE Full Text
  35. 35.↵
    Roy, A. L. 2001. Biochemistry and biology of the inducible multifunctional transcription factor TFII-I. Gene274:1-13.
    OpenUrlCrossRefPubMedWeb of Science
  36. 36.↵
    Roy, A. L., H. Du, P. D. Gregor, C. D. Novina, E. Martinez, and R. G. Roeder. 1997. Cloning of an inr- and E-box-binding protein, TFII-I, that interacts physically and functionally with USF1. EMBO J.16:7091-7104.
    OpenUrlAbstract
  37. 37.↵
    Roy, A. L., M. Meisterernst, P. Pognonec, and R. G. Roeder. 1991. Cooperative interaction of an initiator-binding transcription initiation factor and the helix-loop-helix activator USF. Nature354:245-248.
    OpenUrlCrossRefPubMedWeb of Science
  38. 38.↵
    Sacristan, C., M. I. Tussie-Luna, S. M. Logan, and A. L. Roy. 2004. Mechanism of Bruton's tyrosine kinase-mediated recruitment and regulation of TFII-I. J. Biol. Chem.279:7147-7158.
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    Sadowski, I., J. C. Stone, and T. Pawson. 1986. A noncatalytic domain conserved among cytoplasmic protein-tyrosine kinases modifies the kinase function and transforming activity of Fujinami sarcoma virus P130gag-fps. Mol. Cell. Biol.6:4396-4408.
    OpenUrlAbstract/FREE Full Text
  40. 40.↵
    Sieweke, M. H., H. Tekotte, U. Jarosch, and T. Graf. 1998. Cooperative interaction of ets-1 with USF-1 required for HIV-1 enhancer activity in T cells. EMBO J.17:1728-1739.
    OpenUrlAbstract
  41. 41.↵
    Sigala, J. L., V. Bottero, D. B. Young, A. Shevchenko, F. Mercurio, and I. M. Verma. 2004. Activation of transcription factor NF-kappaB requires ELKS, an IkappaB kinase regulatory subunit. Science304:1963-1967.
    OpenUrlAbstract/FREE Full Text
  42. 42.↵
    Sirito, M., Q. Lin, J. M. Deng, R. R. Behringer, and M. Sawadogo. 1998. Overlapping roles and asymmetrical cross-regulation of the USF proteins in mice. Proc. Natl. Acad. Sci. USA95:3758-3763.
    OpenUrlAbstract/FREE Full Text
  43. 43.↵
    Sirito, M., Q. Lin, T. Maity, and M. Sawadogo. 1994. Ubiquitous expression of the 43- and 44-kDa forms of transcription factor USF in mammalian cells. Nucleic Acids Res.22:427-433.
    OpenUrlCrossRefPubMedWeb of Science
  44. 44.↵
    Takahashi, K., C. Nishiyama, M. Nishiyama, K. Okumura, C. Ra, Y. Ohtake, and T. Yokota. 2001. A complex composed of USF1 and USF2 activates the human FcepsilonRI alpha chain expression via a CAGCTG element in the first intron. Eur. J. Immunol.31:590-599.
    OpenUrlCrossRefPubMedWeb of Science
  45. 45.↵
    Timchenko, N., D. R. Wilson, L. R. Taylor, S. Abdelsayed, M. Wilde, M. Sawadogo, and G. J. Darlington. 1995. Autoregulation of the human C/EBP alpha gene by stimulation of upstream stimulatory factor binding. Mol. Cell. Biol.15:1192-1202.
    OpenUrlAbstract/FREE Full Text
  46. 46.↵
    Van Antwerp, D. J., S. J. Martin, T. Kafri, D. R. Green, and I. M. Verma. 1996. Suppression of TNF-alpha-induced apoptosis by NF-kappaB. Science274:787-789.
    OpenUrlAbstract/FREE Full Text
  47. 47.↵
    Virolle, T., C. Coraux, O. Ferrigno, L. Cailleteau, J. P. Ortonne, P. Pognonec, and D. Aberdam. 2002. Binding of USF to a non-canonical E-box following stress results in a cell-specific derepression of the lama3 gene. Nucleic Acids Res.30:1789-1798.
    OpenUrlCrossRefPubMed
  48. 48.↵
    Weiss, A., and J. D. Stobo. 1984. Requirement for the coexpression of T3 and the T cell antigen receptor on a malignant human T cell line. J. Exp. Med.160:1284-1299.
    OpenUrlAbstract/FREE Full Text
  49. 49.↵
    Wen, Y. D., W. D. Cress, A. L. Roy, and E. Seto. 2003. Histone deacetylase 3 binds to and regulates the multifunctional transcription factor TFII-I. J. Biol. Chem.278:1841-1847.
    OpenUrlAbstract/FREE Full Text
  50. 50.↵
    Zhang, L., Y. Huang, H. Yuan, B. K. Chen, J. Ip, and D. D. Ho. 1997. Identification of a replication-competent pathogenic human immunodeficiency virus type 1 with a duplication in the TCF-1alpha region but lacking NF-kappaB binding sites. J. Virol.71:1651-1656.
    OpenUrlAbstract/FREE Full Text
View Abstract
PreviousNext
Back to top
Download PDF
Citation Tools
TFII-I Regulates Induction of Chromosomally Integrated Human Immunodeficiency Virus Type 1 Long Terminal Repeat in Cooperation with USF
Jiguo Chen, Tom Malcolm, Mario C. Estable, Robert G. Roeder, Ivan Sadowski
Journal of Virology Mar 2005, 79 (7) 4396-4406; DOI: 10.1128/JVI.79.7.4396-4406.2005

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this Journal of Virology article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
TFII-I Regulates Induction of Chromosomally Integrated Human Immunodeficiency Virus Type 1 Long Terminal Repeat in Cooperation with USF
(Your Name) has forwarded a page to you from Journal of Virology
(Your Name) thought you would be interested in this article in Journal of Virology.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
TFII-I Regulates Induction of Chromosomally Integrated Human Immunodeficiency Virus Type 1 Long Terminal Repeat in Cooperation with USF
Jiguo Chen, Tom Malcolm, Mario C. Estable, Robert G. Roeder, Ivan Sadowski
Journal of Virology Mar 2005, 79 (7) 4396-4406; DOI: 10.1128/JVI.79.7.4396-4406.2005
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

DNA-binding proteins
Gene Expression Regulation, Viral
HIV Long Terminal Repeat
HIV-1
T-Lymphocytes
transcription factors
Transcription Factors, TFII
virus integration
virus replication

Related Articles

Cited By...

About

  • About JVI
  • Editor in Chief
  • Editorial Board
  • Policies
  • For Reviewers
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Article Types
  • Ethics
  • Contact Us

Follow #Jvirology

@ASMicrobiology

       

 

JVI in collaboration with

American Society for Virology

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

 

American Society for Microbiology
1752 N St. NW
Washington, DC 20036
Phone: (202) 737-3600

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Print ISSN: 0022-538X; Online ISSN: 1098-5514