Skip to main content
Journal of Virology logoLink to Journal of Virology
. 2009 Jun 17;83(17):8646–8654. doi: 10.1128/JVI.00020-09

Cytoplasmic APOBEC3G Restricts Incoming Vif-Positive Human Immunodeficiency Virus Type 1 and Increases Two-Long Terminal Repeat Circle Formation in Activated T-Helper-Subtype Cells

Michael L Vetter 1, Richard T D'Aquila 1,2,*
PMCID: PMC2738200  PMID: 19535442

Abstract

Cytoplasmic APOBEC3G has been reported to block wild-type human immunodeficiency virus type 1 (HIV-1) infection in some primary cells. It is not known whether cytoplasmic APOBEC3G has residual activity in activated T cells, even though virion-packaged APOBEC3G does restrict HIV-1 in activated T cells. Because we found that APOBEC3G expression is greater in activated CD4+ T-helper type 1 (Th1) lymphocytes than in T-helper type 2 (Th2) lymphocytes, we hypothesized that residual target cell restriction of incoming Vif-positive virions that lack APOBEC3G, if present, would be greater in Th1 than Th2 lymphocytes. Infection of activated Th1 cells with APOBEC3-negative virions did result in decreased amounts of early and late reverse transcription products and integrated virus relative to infection of activated Th2 cells. Two-long terminal repeat (2-LTR) circles, which are formed in the nucleus when reverse transcripts do not integrate, were increased after APOBEC3-negative virus infection of activated Th1 cells relative to infection of activated Th2 cells. In contrast, 2-LTR circle forms were decreased after infection of APOBEC3G-negative cells with APOBEC3G-containing virions relative to APOBEC3G-negative virions and with Th1 cell-produced virions relative to Th2 cell-produced virions. Increasing APOBEC3G in Th2 cells and decreasing APOBEC3G in Th1 cells modulated the target cell phenotypes, indicating causation by APOBEC3G. The comparison between activated Th1 and Th2 cells indicates that cytoplasmic APOBEC3G in activated Th1 cells partially restricts reverse transcription and integration of incoming Vif-positive, APOBEC3G-negative HIV-1. The differing effects of cytoplasmic and virion-packaged APOBEC3G on 2-LTR circle formation indicate a difference in their antiviral mechanisms.


Human APOBEC3G (A3G) is a cytidine deaminase that limits endogenous retrotransposition, as well as replication of multiple retroviruses including human immunodeficiency virus type 1 (HIV-1) (15, 23, 32). Two separate A3G mechanisms that restrict HIV-1 have been hypothesized. A3G packaged into virions released from a producer cell exerts antiviral effects during both reverse transcription and integration in the subsequent target cell (30, 41). This A3G activity is antagonized by HIV-1 Vif limiting virion packaging in the producer cell (42). Endogenous cytoplasmic A3G in resting CD4+ T cells, monocytes, and mature dendritic cells can also restrict the replication of incoming A3G-free HIV-1 virions (10). Two studies reported that small interfering RNA (siRNA) knockdown of this endogenous A3G in resting CD4+ T cells and dendritic cells, respectively, removed this restriction (9, 27). However, another group reported that the same siRNAs did knock down A3G but did not remove the block to HIV infection of resting CD4+ T cells (17). Cellular activation, or specific cytokine signaling, moved A3G from a “low-molecular-mass” form to a “high-molecular-mass” complex in T cells, as did differentiation of monocytes, and abrogated the antiviral effect (10, 34). When immature dendritic cells were matured in vitro, A3G levels increased and were predominately in the “low-molecular-mass” form; dendritic cell maturation also decreased permissiveness to incoming HIV infection (24, 29). Our goal was to compare whether early replication of vif-positive HIV (e.g., lacking virion A3G) varied in activated T-helper cells that harbored different amounts of cytoplasmic A3G.

The CD4+ T-helper cell is a major target of HIV-1. There are several subtypes of CD4+ T-helper cells including T-helper type 1 (Th1) cells, T-helper type 2 (Th2) cells, regulatory T cells, and T-helper 17 cells (29). These subtypes are defined by effector functions following activation. In particular, Th1 cells express gamma interferon (IFN-γ) to stimulate cell-mediated responses to an invading pathogen, whereas Th2 cells predominantly express interleukin 4 (IL-4) to induce a humoral immune response (1, 22, 27). In a previous study, we demonstrated that these two CD4+ T-helper cell subtypes differ in the level of expression of A3G and in the infectivity/A3G content of HIV-1 virions produced from them (38). Given this difference in cellular A3G expression, we tested whether Th1 target cells would better restrict replication of incoming HIV-1 than would Th2 cells. The comparison indicates that endogenous A3G can also play a role in partially restricting HIV-1 infection in some activated CD4+ T cells.

MATERIALS AND METHODS

Human subjects.

Peripheral blood samples were obtained with informed consent from healthy volunteers under a protocol approved by the Vanderbilt University Institutional Review Board.

Cells.

Peripheral blood mononuclear cells were isolated from blood using Ficoll-Hypaque (Amersham Biosciences) gradients. CD4+ cells were isolated by negative selection through magnetic separation using autoMacs (Miltenyi Biotec, Auburn, CA) or Robosep (StemCell Technologies, Vancouver, British Columbia, Canada). Naïve CD4+ T cells were stained with fluorescein isothiocyanate-labeled anti-CD45RO antibodies (anti-CD45RO-FITC) and phycoerythrin (PE)-labeled anti-CD25 antibodies (anti-CD25-PE) (BD Pharmingen, San Jose, CA) and then sorted (FACSAria; Becton Dickinson, San Jose, CA). Sorted naïve CD4+ T cells were differentiated to Th1 cells using anti-CD3 (OKT3; ATCC)-coated plates in RPMI 1640 medium supplemented with anti-CD28 antibodies (BD Biosciences Pharmingen), 0.5 μg/ml neutralizing anti-IL-4 antibody, and 30 ng/ml recombinant IL-12. For Th2 cell differentiation, sorted naïve cells were cultured on plates coated with anti-CD3 in medium supplemented with anti-CD28 antibodies (BD Biosciences Pharmingen), 2.5 μg/ml neutralizing anti-IFN-γ antibody, and 50 ng/ml recombinant IL-4. Cytokines and neutralizing antibodies were obtained from R&D Systems, Minneapolis, MN. In vitro-differentiated Th1 and Th2 cells were removed from the anti-CD3-coated plates after 48 h and were subsequently expanded for 10 days in the medium described above. Differentiation was confirmed by intracellular cytokine staining for IL-4-PE and IFN-γ-allophycocyanin (IFN-γ-APC) (BD Pharmingen), as well as surface staining for CXCR3-PE and CRTh2-APC (BD Pharmingen) (5, 12, 35). To increase APOBEC3G expression in cytokine-polarized Th2 cells, differentiating cultures were transduced with an APOBEC3G-expressing, HIV-derived lentiviral vector at the time of activation. The vector expressed APOBEC3G, and human serum albumin was used as a marker of transduction (37, 38). To reduce APOBEC3G expression in cytokine-polarized Th1 cells, fully differentiated Th1 cells were activated for 48 h with anti-CD3/CD28-coated beads (Invitrogen) in the presence of 10 μg anti-IFN-γ antibody (38) (R&D Systems).

Viruses and infection.

Vesicular stomatitis virus glycoprotein (VSV-G)-pseudotyped, green fluorescent protein (GFP)-expressing HIV-1 was produced by polyethylenimine transfection (13) of 293T cells with a NL4-3 recombinant with GFP replacing nef as well as phCMV-VSV-G. Prior to infection, viral supernatants were treated with 100 U DNase I (Bio-Rad) for 1 hour. In vitro-differentiated Th1 and Th2 cells (1 × 106 cells of each type) were used for infections. Subsequent to the expansion described above in the absence of anti-CD3/CD28 antibody, each cell type was reactivated for 48 h with anti-CD3/CD28 antibody-coated beads (Invitrogen) and then inoculated with virus (400 ng of p24) for 4 h. After infection, the cells were washed and resuspended in fresh RPMI 1640 medium with 10% fetal bovine serum, 1% penicillin/streptomycin, and 50 U/ml IL-2. Cells were fixed in 2% paraformaldehyde 48 h postinfection and analyzed for the percentage of cells expressing GFP on a FACSAria cell sorter/cytometer (Becton Dickinson).

Virions with or without packaged A3G were produced by polyethylenimine transfection of 293T cells with a vif-deleted NL4-3 provirus construct (15 μg of DNA) in combination with an empty control plasmid (A3G-negative virions) or an A3G expression plasmid (3 μg) (A3G-positive virions). Five nanograms of the resulting viral supernatants was used for infection. Th1- and Th2-produced virions were generated as previously described (38), and 5 ng of collected supernatant was used for infection.

PCRs.

Infected Th cells were collected at 2, 4, 18, 24, and 48 h after infection and washed in phosphate-buffered saline. Cellular DNA was isolated using the DNeasy kit (Qiagen). DNA was quantified by spectrophotometry on a GeneQuant Pro (Amersham Biosciences) and normalized prior to use in quantitative PCR (qPCR) assays on an ABI Prism 7000 (Applied Biosystems). To detect early reverse transcription (RT) products, the primers used were 5′-GTGCCCGTCTGTTGTGTGAC-3′ and 5′-GGCGCCACTGCTAGAGATTT-3′, in conjunction with a probe 5′-FAM-CTAGAGATCCCTCAGACCCTTTTAGTCAGTGTGG-TAMRA-3′, where FAM is 6-carboxyfluorescein and TAMRA is 6-carboxytetramethylrhodamine. Late RT products were detected using primers 5′-TGTGTGCCCGTCTGTTGTGT-3′ and 5′-GAGTCCTGCGTCGAGAGAGC-3′ with probe 5′-FAM-CAGTGGCGCCCGAACAGGGA-TAMRA-3′ (7, 24). For two-long terminal repeat (2-LTR) circle quantification, DNA isolated at 48 h after infection was normalized and used in qPCR. The forward primer was MH535 (5′-AACTAGGGAACCCACTGCTTAAG-3′), the reverse primer was MH536 (5′-TCCACAGATCAAGGATATCTTGTC-3′), and the probe was MH603 (5′-FAM-ACACTACTTGAAGCACTCAAGGCAAGCTTT-TAMRA-3′) (7). For quantification of GFP DNA following reverse transcription, the forward primer used was 5′-AAGCTGACCCTGAAGTTCATCTG-3′, the reverse primer was 5′-TTGAAGAAGTCGTGCTGCTTCAT-3′, and the probe was 5′-FAM-ACCGGCAAGCTGC-MGB NFQ-3′ (19), where MGB NFQ is minor groove binder-nonfluorescent quencher.

Cellular fractionation.

In vitro-differentiated Th1 and Th2 cells (expanded as described above in the absence of an activating stimulus) were either restimulated by anti-CD3/CD28 antibody for 48 h (activated) or not stimulated in IL-2-containing medium for 72 h (nonactivated). Activated and nonactivated cells were stained with CD4-APC-Cy7 and CD25-FITC antibodies (BD Pharmingen, San Jose, CA) to determine differences in this activation marker. Cells were then lysed in 50 mM HEPES (pH 7.4), 125 mM NaCl, 0.2% NP-40, 0.1 mM phenylmethylsulfonyl fluoride, and EDTA-free protease inhibitor cocktail (CalBiochem, San Diego, CA) and subjected to ultracentrifugation as previously described (125,000 × g, TLA 55 rotor [Beckman Coulter]) (28). Equal volumes of supernatant and pellet were subjected to Western blotting using polyclonal anti-APOBEC3G antibody (33), with a goat anti-rabbit secondary antibody conjugated to Alexa Fluor 680 (Invitrogen Molecular Probes, Carlsbad, CA). Band intensity was quantified using the Odyssey infrared imaging system (Li-Cor Biosciences, Lincoln, NE). Values are expressed as the fraction of relative light units (RLUs) of the supernatant band relative to the total RLUs for both supernatant and pellet.

Cellular lysates of activated Th1 and Th2 cells were also subjected to centrifugation through a 4% to 40% sucrose gradient as previously described (39). Gradients were centrifuged overnight at 32,000 rpm in an SW-41 rotor (Beckman), and then 12 1-milliliter fractions were collected, precipitated by trichloroacetic acid, resuspended in sodium dodecyl sulfate sample buffer, and subjected to Western blotting. Values are presented as the percent RLUs in each fraction relative to the total RLUs for each sample.

RESULTS

HIV-1 infection of, and integration into, CD4+ Th1 cells relative to Th2 cells.

Following 48 h of T-cell receptor activation by anti-CD3/CD28 antibody-coated beads, cytokine-polarized Th1 and Th2 cell cultures were infected with VSV-G-pseudotyped, vif-competent HIV-1 expressing GFP (2, 6, 9). Forty-eight hours after infection, the percentage of cells with integrated HIV genomes was determined by GFP expression using fluorescence-activated cell sorting analysis. In cells from each of three donors, infection of Th2 cells yielded greater GFP signal than did infection of Th1 cells (Fig. 1A).

FIG. 1.

FIG. 1.

HIV-1 infection/integration is decreased in activated CD4+ Th1 lymphocytes relative to Th2 lymphocytes, and these cells differ slightly in the predominant form of cytoplasmic A3G. (A) Cytokine-polarized Th1 and Th2 cells derived from naïve cells from three individual donors were activated with anti-CD3/CD28 antibody for 48 h and then infected with VSV-G-pseudotyped, vif-positive, GFP-expressing HIV-1. Virus was washed off after 4 h, and the number of GFP-positive cells, reflecting cells with integrated provirus, was counted 48 h postinfection. A smaller percentage of the Th1 cell population is GFP positive (GFP +) than the Th2 cell population. (B) Th1 and Th2 cells were activated with anti-CD3/CD28 antibody for 48 h and then lysed and subjected to sucrose gradient separation as described in Materials and Methods. Collected fractions (fractions 4 to 12) were subjected to Western blotting for A3G. (C) The intensities of the A3G bands for each fraction (designated by arrows to the right of panel B) were quantified by the Li-Cor Odyssey infrared imaging system. The percent of total RLU per fraction is shown in the graph ([RLU of fraction/total RLU of all fractions] × 100). Subsequent to activation, some A3G still remains in lower-density fractions (fractions 4, 5, and 6); this is a smaller proportion of the total A3G in activated Th2 cells (32%) than in activated Th1 cells (39%).

Since the block to reverse transcription in some primary cells has been attributed to the “low-molecular-mass” form of A3G (10), we separated A3G forms in Th subtype cells by two methods. Sucrose gradient density centrifugation was validated previously to separate A3G forms (39). Density gradients of activated Th1 cell lysates confirmed that 39% of the total A3G was in the two lower-density fractions (fractions 5 and 6) and that 23% was in the two highest-density fractions (fractions 11 and 12). This compared to 32% of the total A3G in the two lower-density fractions and 28% in the two highest-density fractions, respectively, in activated Th2 cells (Fig. 1B and C).

A second method of characterizing A3G forms was also used. “Low-molecular-mass” A3G has previously been shown to remain in the supernatant after ultracentrifugation (28). Following expansion, Th1 and Th2 cells were either placed in fresh medium with no activation signal (nonactivated) or restimulated with anti-CD3/CD28 antibody-coated beads (activated) (Fig. 2A) and then subjected to ultracentrifugation. After ultracentrifugation, activated Th1 cell lysates had less supernatant A3G than nonactivated lysates did (Fig. 2B and C). A lower amount of supernatant A3G was also seen in activated Th2 cells relative to nonactivated cells. A substantial proportion of A3G remained in the supernatant of activated lysates from both Th1 and Th2 cells after ultracentrifugation. The amount of total and supernatant A3G was lower in the Th2 cell lysates than in Th1 cell lysates (Fig. 2B and C).

FIG. 2.

FIG. 2.

Low-density APOBEC3G remains in ultracentrifuged lysate supernatants after Th1 and Th2 cell activation. (A) Nonactivated or anti-CD3/CD28 antibody-activated Th1 and Th2 cells were stained with CD4 and CD25 antibodies to evaluate the extent of activation. The nonactivated Th1 population was 1% CD25 high, and the Th2 population was 22% CD25 high. After activation, Th1 cells were 89% CD25 high and Th2 cells were 91% CD25 high after activation. Cells were lysed and subjected to ultracentifugation separation as described in Materials and Methods. (B) The pelleted (P) and supernatant (S) fractions were subjected to Western blotting for A3G. (C) The fraction of supernatant A3G ([supernatant RLU/pellet RLU] × total RLU) after ultracentrifugation of nonactivated (N.A.) and activated (A.) Th1 and Th2 cells from three individual donors was determined by quantification of band intensity (using Li-Cor Odyssey). A shift from the supernatant to pellet fraction is seen after activation, although some A3G remains in the supernatant fraction in each activated cell type.

Reverse transcription in CD4+ Th1 lymphocytes relative to Th2 lymphocytes.

Given the reduced amount of GFP from integrated virus in Th1 cells, we next determined at which step of infection this restriction was occurring relative to Th2 cells. A qPCR assay for early and late reverse transcription products showed that the two cell types differed in their ability to support reverse transcription (Fig. 3). Figures 3A and B indicate that infected Th1 cells lag behind Th2 cells from one donor in the amount of both early and late RT products formed. There was a significant difference at a single time point (18 h after infection) for multiple donors in both early and late RT products (Fig. 3C and D), as well as a significant difference between Th1 and Th2 early and late RT products from three replicate infections of cells from a single donor (Fig. 3E).

FIG. 3.

FIG. 3.

Reverse transcription is decreased in CD4+ Th1 lymphocytes relative to Th2 lymphocytes. (A and B) After the cells were infected with VSV-G-pseudotyped, vif-positive, GFP-expressing HIV-1 (HIV-GFP) DNA from Th1 and Th2 cells was isolated and assayed by qPCR for early (A) and late (B) RT products. DNA was collected at 2, 4, 18, 24 and 48 h postinfection from the cells from a single donor. Th1 cells produced fewer reverse transcription products over time than the Th2 cells did. (C and D) Reverse transcription products were also measured after VSV-G, vif-positive HIV-GFP infection of Th1 and Th2 cells derived from naïve cells from multiple donors at a single time point,18 h postinfection. Early RT products (C) and late RT products (D) are shown. Medians and interquartile ranges are indicated. Values that are significantly different (P ≤ 0.02) from the value for Th1 cells by the Mann-Whitney U test are indicated by an asterisk. Th1 cell RT products were decreased relative to Th2 cell RT products. (E) Early and late RT products were also assayed after triplicate VSV-G, vif-positive HIV-GFP infections of Th1 and Th2 cells derived from naïve cells from a single donor at 18 h postinfection (mean plus standard error [error bar] are indicated). Th1 cells had fewer early and late reverse transcription products at 18 h following infection. Values that are significantly different (P ≤ 0.02) from the value for Th1 cells by the Mann-Whitney U test are indicated by an asterisk.

2-LTR circle formation in CD4+ Th1 lymphocytes relative to Th2 lymphocytes.

The measurement of 2-LTR circles formed following HIV-1 infection serves as a marker for nuclear entry of reverse transcripts and abortive integration events (11). We studied whether 2-LTR circle formation differed after A3G-free virion infection of activated Th1 versus Th2 cells. Infection of activated Th1 cells with A3G-free virions led to significantly more 2-LTR circles than did infection of activated Th2 cells (Fig. 4A). This is consistent with relatively more reverse transcripts not integrating and forming abortive 2-LTR circle forms in activated Th1 cells than in Th2 cells. Previous studies reported, however, that A3G packaged in the virion reduced 2-LTR circle formation as well as decreasing integration, consistent with impaired host cell-mediated 2-LTR circle formation and integration (3, 21). Therefore, we also evaluated the effects of virion-packaged A3G on 2-LTR circle formation.

FIG. 4.

FIG. 4.

2-LTR circle formation is increased in CD4+ Th1 lymphocytes relative to Th2 lymphocytes after vif-positive HIV-1 infection. Increased cytoplasmic A3G led to increased 2-LTR circle formation (A), while increased virion-packaged A3G caused decreased 2-LTR circle formation (B and C). (A) Th1 and Th2 cells derived from four individual donors were infected with VSV-G-pseudotyped, vif-positive, GFP-expressing HIV-1. Forty-eight hours after infection, DNA from infected Th1 and Th2 cells was assayed by qPCR for 2-LTR circles, which were found to be increased in Th1 cells relative to Th2 cells. Means and standard errors for cells from the four donors are shown. The value that was significantly different (P < 0.03) from the value for Th1 cells by the Mann-Whitney U test is indicated by an asterisk. (B) TZM-bl cells, which lack APOBEC3G, were infected with vif-deleted (vif−) virus with or without packaged A3G. Forty-eight hours after infection, DNA was isolated and assayed for 2-LTR circle formation by qPCR. A Western blot of viral lysates demonstrates the amount of A3G packaged in the virions. Multiple separate experiments have shown that this A3G-containing virus is ∼50-fold-less infectious than the A3G-free virus. Virions containing A3G produced fewer 2-LTR circles. Means and standard errors (error bars) of duplicate experiments are shown. (C) TZM-bl cells were infected with vif-deleted HIV-1 produced from Th1 or Th2 cells. A Western blot of viral lysates demonstrates the amount of A3G packaged in the virions. Previous data demonstrated that the Th1 cell-produced virions were approximately fivefold-less infectious than Th2 cell-produced virions were (38). Th1 cell-produced virions led to fewer 2-LTR circles.

To study virion packaged A3G, TZM-bl cells, which do not contain A3G, were infected with vif-deleted virions that contained A3G or did not contain A3G. These virions were produced by transfection of 293T cells with or without a cotransfected A3G expression plasmid. Infection with vif-deleted viruses containing A3G led to fewer 2-LTR circles relative to TZM-bl cells infected with A3G-free virions (Fig. 4B). Fewer 2-LTR circles were also seen in TZM-bl cells infected with Th1 cell-produced virions than in those infected with Th2 cell-produced virions (Fig. 4C), consistent with the previously documented greater packaging of A3G in Th1 cell-produced virions (38). Thus, virion-packaged A3G and cytoplasmic A3G have different effects on 2-LTR circle formation.

Changes in APOBEC3G expression modulate reverse transcription, 2-LTR circle formation, and integration.

To determine whether the difference in cytoplasmic A3G between Th1 and Th2 lymphocytes causes the observed differences in reverse transcription, 2-LTR circle formation, and integration between these cells when infected, the expression level of A3G was altered in each cell type. Retroviral transduction of A3G was used to increase A3G expression in Th2 cells as previously reported (38). Since the A3G-transducing vector was derived from HIV and the virus used for subsequent infection expresses GFP in place of nef, GFP DNA was quantified by PCR rather than using the early and late HIV-1 reverse transcription product primers. The amount of reverse-transcribed GFP DNA was reduced in Th2 cells overexpressing A3G relative to control vector-transduced Th2 cells (Fig. 5A and B). The formation of 2-LTR circles was also increased after infection in Th2 cells overexpressing A3G relative to control vector-transduced cells (Fig. 5C). A3G overexpression in Th2 cells also reduced the amount of integrated genomes as measured by GFP production (Fig. 5D).

FIG. 5.

FIG. 5.

Overexpression of cytoplasmic A3G in Th2 cells reduces reverse transcription products and increases 2-LTR circle formation. Th2 cells were transduced with an A3G-expressing vector (Th2-A3G) or control retrovirus vector (Th2-Empty), activated for 48 h, and then infected with VSV-G-pseudotyped, vif-positive GFP-expressing HIV-1. The increased expression of A3G decreases the formation of reverse transcription products, increases the formation of 2-LTR circles, and reduces the amount of integrated proviruses as measured by GFP expression. (A) DNA was isolated at 18 h after infection and used in qPCR assays to analyze GFP reverse transcription product formation in triplicate from the cells from one donor. Overexpression of A3G in Th2 cells decreased reverse transcription products measured by amplifying the GFP sequences that replaced the nef open reading frame. Means plus standard errors (error bars) are shown. The value that was significantly different (P ≤ 0.02) from the value for Th2 cells transduced by empty vector by the Mann-Whitney U test is indicated by an asterisk. (B) GFP-labeled reverse transcription products measured as in panel A from matched A3G- and control (Th2-empty) vector-transduced Th2 cells derived from naïve cells from four different donors. (C) DNA was isolated 48 h after infection to analyze 2-LTR circle formation. Overexpression of A3G in Th2 cells increased the number of 2-LTR circles. Means and interquartile ranges are shown. The value that was significantly different (P ≤ 0.02) from the value for Th2 cells transduced by empty vector by the Mann-Whitney U test is indicated by an asterisk. (D) Cells were also analyzed 48 h postinfection for GFP expression. A3G overexpression in Th2 cells decreased GFP expression from integrated provirus. Means and standard errors are shown. The value that was significantly different (P ≤ 0.02) from the value for Th2 cells transduced by empty vector by the Mann-Whitney U test is indicated by an asterisk. (E) Western blot of cell lysates from Th1 and Th2 cells and Th2 cells transduced with an A3G-expressing vector.

As further confirmation of the role of endogenous cytoplasmic A3G in modulating reverse transcription, 2-LTR circle formation, and integration, Th1 cells were incubated with a neutralizing anti-IFN-γ antibody previously documented to reduce A3G expression in Th1 cells (38) or with an isotype control antibody. Infection of Th1 cells treated with the neutralizing anti-IFN-γ antibody with GFP-expressing HIV-1 led to increased early and late RT products (Fig. 6A), reduced 2-LTR circle formation (Fig. 6B), and an increase in integrated genomes based on increased GFP expression (Fig. 6C) compared to the cells treated with the isotype antibody control.

FIG. 6.

FIG. 6.

Reduction of cytoplasmic A3G in Th1 cells increases reverse transcription products and decreases 2-LTR circle formation. Th1 cells were activated for 48 h in conjunction with neutralizing anti-IFN-γ antibody, which decreases A3G expression, or with an isotype antibody control. The cells were infected with VSV-G-pseudotyped, vif-positive GFP-expressing HIV-1, and DNA was isolated at 18 or 48 h postinfection. Decreased expression of A3G in Th1 cells leads to increased reverse transcription products, decreased 2-LTR circles, and increased integration relative to the cells activated with the isotype antibody control. (A) Early and late RT products were determined 18 h postinfection. Both early and late RT products were increased in the presence of the A3G-reducing anti-IFN-γ antibody. Means and standard errors (error bars) are shown. Values that were significantly different (P ≤ 0.02) from the value for Th1 cells activated with anti-IFN-γ antibody by the Mann-Whitney U test are indicated by an asterisk. (B) 2-LTR circles were quantified 48 h postinfection. Two-LTR circles were decreased in the presence of the A3G-reducing anti-IFN-γ antibody. Means and standard errors (error bars) are shown. The value that was significantly different (P ≤ 0.05) from the value for Th1 cells activated with anti-IFN-γ antibody by the Mann-Whitney U test is indicated by an asterisk. (C) GFP expression from integrated proviruses in live cells was analyzed 48 h postinfection. Integrated copies of HIV were increased in the presence of the A3G-reducing anti-IFN-γ antibody. Means and standard errors (error bars) are shown. The value that was significantly different (P ≤ 0.02) from the value for Th1 cells activated with A3G-reducing anti-IFN-γ antibody by the Mann-Whitney U test is indicated by an asterisk. GFP +, GFP positive.

DISCUSSION

Comparison of HIV-1 infection of activated Th1 and Th2 lymphocytes in the present study provides some of the first documentation that cytoplasmic A3G partially restricts incoming A3G-negative HIV-1 virions at the level of reverse transcription and integration in activated T lymphocytes. The effects of cytoplasmic A3G on incoming A3G-negative virions, the “target cell effects” of A3G, have previously been characterized in resting T cells, monocytes, and mature dendritic cells (10, 14, 28). T-cell activation, as well as monocyte differentiation to macrophages, has been hypothesized to completely abrogate this target cell block. There are conflicting data about whether siRNA specific for A3G abrogates this restriction in total CD4+ T cells isolated from blood (10, 17). However, the present comparison of different types of activated T-helper cells has now revealed relatively less reverse transcription, less integration, and more 2-LTR circle formation in activated Th1 than in activated Th2 cells. Modulating the levels of expression of A3G in each of these two cell types alters each of these phenotypes and strongly supports the causal role for A3G in the relative restriction in Th1 cells observed here. When Th2 cells that express relatively lower levels of A3G were transduced with a vector that overexpresses A3G, they become more restrictive to reverse transcription and integration. When A3G expression was reduced in Th1 cells, they were rendered less restrictive to reverse transcription and integration. In addition to supporting that A3G is responsible for the observed reduction in replication of vif-positive, A3G-negative virus in activated Th1 cells noted here, these results add to prior data indicating that A3G activity in CD4+ T lymphocytes is regulated by IFN-γ (38) as well as other cytokines (34).

Cytoplasmic A3G unexpectedly differed from virion-packaged A3G in its effect on 2-LTR circle formation, an indicator of abortive integration. Earlier studies observed that virion-packaged A3G decreased 2-LTR circle formation (3, 21). We confirmed this in a comparison of A3G-positive and A3G-negative virion infections of A3G-negative cells, as well as in a comparison of infections of A3G-negative cells with virions produced from activated Th1 cells (e.g., with greater virion A3G content) and virions produced from activated Th2 cells (e.g., containing relatively less virion A3G). In sharp contrast, A3G-negative virion infection of activated Th1 cells led to increased 2-LTR circle formation compared to infection of activated Th2 cells. This opposite effect on 2-LTR circle formation indicates that A3G packaged in the virion has a different, additional effect than does cytoplasmic A3G.

Earlier work has strongly suggested that virion A3G affects multiple steps of HIV replication. Virion A3G decreases early RT product formation potentially by interfering with tRNA primer initiation of reverse transcription (16, 40) and late RT product formation perhaps by impairing tRNA primer removal or otherwise limiting plus-strand DNA transfer (18, 21), 2-LTR circle formation, and provirus integration. Each of these inhibitory effects progressively increases, suggesting a culmination of effects on multiple targets (3). Integrase inhibition, which increases 2-LTR circles in wild-type, vif-positive HIV-1 infection, did not increase the amount of 2-LTR circles in cells infected with virions containing A3G (3). It has been suggested that nuclear nonhomologous end-joining cell repair proteins responsible for 2-LTR circle formation cannot ligate RT products made when virion A3G is present, as the virion A3G alters the ends of viral cDNA substrates needed for both provirus integration and 2-LTR circle formation (21); however, it is not yet clear whether the extent to which cDNA ends are altered completely explains the observed decrease in 2-LTR circles. A second possibility is that virion A3G interferes with nuclear import of preintegration complexes (PICs). Several lines of evidence suggest that virion A3G interacts with HIV reverse transcription complexes and/or PICs (8, 20, 21, 31).

On the basis of our data and these earlier studies analyzing virion-packaged A3G, we hypothesize that virion-packaged A3G has a different or additional association with one or more components of the HIV reverse transcription complexes, PICs, or another cellular protein than does cytoplasmic A3G. Indeed, the results of the present study argue against the possibility that the mechanism for decreased 2-LTR formation by virion A3G is solely that virion A3G decreases 2-LTR circles just by decreasing the cDNA substrate for both 2-LTR circles and provirus formation. (3). Since cytoplasmic A3G decreased both late RT products and provirus integration, the difference noted here in 2-LTR circle formation by cytoplasmic versus virion A3G strongly suggests that virion A3G has an additional effect that diminishes 2-LTR circle formation beyond its effects on reverse transcription. This is an important area for further study, as it may lead to identification of a new cellular target that could be inhibited for antiretroviral effect, as well as more fully defining the multiple mechanisms of A3G restriction activity.

Prior studies have implicated a “low-molecular-mass” form of A3G in mediating the block to incoming A3G-negative virions in resting T cells (10). Other studies have looked at the effects of monocyte differentiation and dendritic cell maturation on the A3G-dependent block to incoming infection (28, 34). Monocyte differentiation to macrophages relieved the block and dendritic cell maturation increased the block. Our study analyzed CD4+ T-helper subtypes but focused on the restriction of HIV infection in activated T cells; these are different cells activated in different ways than in the other studies cited. The results of the present study indicate that some A3G persists in activated Th1 and Th2 cell lysate supernatants after ultracentrifugation and in lower-density fractions of sucrose density gradients. We hypothesize that the relatively greater remnant “low-molecular-mass” forms mediate this relatively greater restriction we have identified in activated Th1 cells compared to Th2 cells. Further work will be needed to confirm or refute this hypothesis by further characterizing, and selectively affecting, the remnant “low-molecular-mass” A3G in activated Th1 cells. If this hypothesis is supported, it will also be of interest for future studies to determine the minimum concentration of “low-molecular-mass” A3G required for restriction.

The results reported here indicate that wild-type HIV-1 virions that infect Th2 cells are more likely to successfully replicate than those infecting Th1 cells. Our earlier observations also indicated that Th2 cells that become infected will subsequently produce virions that are more infectious due to reduced virion packaging of A3G. These data may explain earlier studies demonstrating better spread of HIV in cultured Th2 cells (4, 25, 26, 36) and suggest new work to characterize the role of Th2 cells during HIV pathogenesis. This study also adds further support to the important role of A3G in HIV pathogenesis through multiple different restricting mechanisms. Further characterization of these mechanisms may lead to novel approaches to improving A3G effects as a potential therapeutic or preventive strategy.

Acknowledgments

This work was supported by NIH T32 AI 060571and NIH R01 AI 29193. This work was facilitated by the infrastructure and programs of the Vanderbilt Meharry Center for AIDS Research (NIH P30 AI 54999).

We thank Megan Johnson for her assistance with ultracentifugation experiments, Chisu Song and John Donahue for Western blotting assistance and advice, Louise Barnett for expert flow cytometry support, as well as Chris Aiken and his laboratory members for NL4-3 and phCMV-VSV-G and for helpful advice.

Footnotes

Published ahead of print on 17 June 2009.

REFERENCES

  • 1.Abbas, A. K., K. M. Murphy, and A. Sher. 1996. Functional diversity of helper T lymphocytes. Nature 383787-793. [DOI] [PubMed] [Google Scholar]
  • 2.Aiken, C. 1997. Pseudotyping human immunodeficiency virus type 1 (HIV-1) by the glycoprotein of vesicular stomatitis virus targets HIV-1 entry to an endocytic pathway and suppresses both the requirement for Nef and the sensitivity to cyclosporin A. J. Virol. 715871-5877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Anderson, J. L., and T. J. Hope. 2008. APOBEC3G restricts early HIV-1 replication in the cytoplasm of target cells. Virology 3751-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Annunziato, F., G. Galli, F. Nappi, L. Cosmi, R. Manetti, E. Maggi, B. Ensoli, and S. Romagnani. 2000. Limited expression of R5-tropic HIV-1 in CCR5-positive type 1-polarized T cells explained by their ability to produce RANTES, MIP-1alpha, and MIP-1beta. Blood 951167-1174. [PubMed] [Google Scholar]
  • 5.Bahbouhi, B., A. Landay, A. Tenorio, and L. Al-Harthi. 2007. HIV infection of primary CD4+ Th2 cells, defined by expression of the chemoattractant receptor-homologous (CRTH2), induces a Th0 phenotype. AIDS Res. Hum. Retrovir. 23269-277. [DOI] [PubMed] [Google Scholar]
  • 6.Burns, J. C., T. Friedmann, W. Driever, M. Burrascano, and J. K. Yee. 1993. Vesicular stomatitis virus G glycoprotein pseudotyped retroviral vectors: concentration to very high titer and efficient gene transfer into mammalian and nonmammalian cells. Proc. Natl. Acad. Sci. USA 908033-8037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Butler, S. L., E. P. Johnson, and F. D. Bushman. 2002. Human immunodeficiency virus cDNA metabolism: notable stability of two-long terminal repeat circles. J. Virol. 763739-3747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Carr, J. M., A. J. Davis, C. Coolen, K. Cheney, C. J. Burrell, and P. Li. 2006. Vif-deficient HIV reverse transcription complexes (RTCs) are subject to structural changes and mutation of RTC-associated reverse transcription products. Virology 35180-91. [DOI] [PubMed] [Google Scholar]
  • 9.Chazal, N., G. Singer, C. Aiken, M. L. Hammarskjold, and D. Rekosh. 2001. Human immunodeficiency virus type 1 particles pseudotyped with envelope proteins that fuse at low pH no longer require Nef for optimal infectivity. J. Virol. 754014-4018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Chiu, Y. L., V. B. Soros, J. F. Kreisberg, K. Stopak, W. Yonemoto, and W. C. Greene. 2005. Cellular APOBEC3G restricts HIV-1 infection in resting CD4+ T cells. Nature 435108-114. [DOI] [PubMed] [Google Scholar]
  • 11.Coffin, J. M., S. H. Hughes, and H. E. Varmus (ed.). 1997. Retroviruses. Cold Spring Harbor Laboratory Press, Plainview, NY. [PubMed]
  • 12.Cosmi, L., F. Annunziato, M. I. G. Galli, R. M. E. Maggi, K. Nagata, and S. Romagnani. 2000. CRTH2 is the most reliable marker for the detection of circulating human type 2 Th and type 2 T cytotoxic cells in health and disease. Eur. J. Immunol. 302972-2979. [DOI] [PubMed] [Google Scholar]
  • 13.Durocher, Y., S. Perret, and A. Kamen. 2002. High-level and high-throughput recombinant protein production by transient transfection of suspension-growing human 293-EBNA1 cells. Nucleic Acids Res. 30E9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ellery, P. J., E. Tippett, Y. L. Chiu, G. Paukovics, P. U. Cameron, A. Solomon, S. R. Lewin, P. R. Gorry, A. Jaworowski, W. C. Greene, S. Sonza, and S. M. Crowe. 2007. The CD16+ monocyte subset is more permissive to infection and preferentially harbors HIV-1 in vivo. J. Immunol. 1786581-6589. [DOI] [PubMed] [Google Scholar]
  • 15.Esnault, C., O. Heidmann, F. Delebecque, M. Dewannieux, D. Ribet, A. J. Hance, T. Heidmann, and O. Schwartz. 2005. APOBEC3G cytidine deaminase inhibits retrotransposition of endogenous retroviruses. Nature 433430-433. [DOI] [PubMed] [Google Scholar]
  • 16.Guo, F., S. Cen, M. Niu, J. Saadatmand, and L. Kleiman. 2006. Inhibition of formula-primed reverse transcription by human APOBEC3G during human immunodeficiency virus type 1 replication. J. Virol. 8011710-11722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kamata, M., Y. Nagaoka, and I. S. Chen. 2009. Reassessing the role of APOBEC3G in human immunodeficiency virus type 1 infection of quiescent CD4+ T-cells. PLoS Pathog. 5e1000342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Klarmann, G. J., X. Chen, T. W. North, and B. D. Preston. 2003. Incorporation of uracil into minus strand DNA affects the specificity of plus strand synthesis initiation during lentiviral reverse transcription. J. Biol. Chem. 2787902-7909. [DOI] [PubMed] [Google Scholar]
  • 19.Laham-Karam, N., and E. Bacharach. 2007. Transduction of human immunodeficiency virus type 1 vectors lacking encapsidation and dimerization signals. J. Virol. 8110687-10698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Luo, K., T. Wang, B. Liu, C. Tian, Z. Xiao, J. Kappes, and X. F. Yu. 2007. Cytidine deaminases APOBEC3G and APOBEC3F interact with human immunodeficiency virus type 1 integrase and inhibit proviral DNA formation. J. Virol. 817238-7248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mbisa, J. L., R. Barr, J. A. Thomas, N. Vandegraaff, I. J. Dorweiler, E. S. Svarovskaia, W. L. Brown, L. M. Mansky, R. J. Gorelick, R. S. Harris, A. Engelman, and V. K. Pathak. 2007. Human immunodeficiency virus type 1 cDNAs produced in the presence of APOBEC3G exhibit defects in plus-strand DNA transfer and integration. J. Virol. 817099-7110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Mosmann, T. R., and S. Sad. 1996. The expanding universe of T-cell subsets: Th1, Th2 and more. Immunol. Today 17138-146. [DOI] [PubMed] [Google Scholar]
  • 23.Muckenfuss, H., M. Hamdorf, U. Held, M. Perkovic, J. Lower, K. Cichutek, E. Flory, G. G. Schumann, and C. Munk. 2006. APOBEC3 proteins inhibit human LINE-1 retrotransposition. J. Biol. Chem. 28122161-22172. [DOI] [PubMed] [Google Scholar]
  • 24.Munk, C., S. M. Brandt, G. Lucero, and N. R. Landau. 2002. A dominant block to HIV-1 replication at reverse transcription in simian cells. Proc. Natl. Acad. Sci. USA 9913843-13848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ofori, H., and P. P. Jagodzinski. 2004. Increased in vitro replication of CC chemokine receptor 5-restricted human immunodeficiency virus type 1 primary isolates in Th2 lymphocytes may correlate with AIDS progression. Scand. J. Infect. Dis. 3646-51. [DOI] [PubMed] [Google Scholar]
  • 26.Ofori, H., J. Prokop, and P. P. Jagodzinski. 2003. Replication at the level of reverse transcription of HIV-1SF2 strain in Th1 and Th2 cells. Biomed. Pharmacother. 5715-19. [DOI] [PubMed] [Google Scholar]
  • 27.O'Garra, A. 1998. Cytokines induce the development of functionally heterogeneous T helper cell subsets. Immunity 8275-283. [DOI] [PubMed] [Google Scholar]
  • 28.Pion, M., A. Granelli-Piperno, B. Mangeat, R. Stalder, R. Correa, R. M. Steinman, and V. Piguet. 2006. APOBEC3G/3F mediates intrinsic resistance of monocyte-derived dendritic cells to HIV-1 infection. J. Exp. Med. 2032887-2893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Romagnani, S. 2006. Regulation of the T cell response. Clin. Exp. Allergy 361357-1366. [DOI] [PubMed] [Google Scholar]
  • 30.Sheehy, A. M., N. C. Gaddis, J. D. Choi, and M. H. Malim. 2002. Isolation of a human gene that inhibits HIV-1 infection and is suppressed by the viral Vif protein. Nature 418646-650. [DOI] [PubMed] [Google Scholar]
  • 31.Soros, V. B., W. Yonemoto, and W. C. Greene. 2007. Newly synthesized APOBEC3G is incorporated into HIV virions, inhibited by HIV RNA, and subsequently activated by RNase H. PLoS Pathog. 3e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Stenglein, M. D., and R. S. Harris. 2006. APOBEC3B and APOBEC3F inhibit L1 retrotransposition by a DNA deamination-independent mechanism. J. Biol. Chem. 28116837-16841. [DOI] [PubMed] [Google Scholar]
  • 33.Stopak, K., C. de Noronha, W. Yonemoto, and W. C. Greene. 2003. HIV-1 Vif blocks the antiviral activity of APOBEC3G by impairing both its translation and intracellular stability. Mol. Cell 12591-601. [DOI] [PubMed] [Google Scholar]
  • 34.Stopak, K. S., Y. L. Chiu, J. Kropp, R. M. Grant, and W. C. Greene. 2007. Distinct patterns of cytokine regulation of APOBEC3G expression and activity in primary lymphocytes, macrophages, and dendritic cells. J. Biol. Chem. 2823539-3546. [DOI] [PubMed] [Google Scholar]
  • 35.Sundrud, M. S., S. M. Grill, D. Ni, K. Nagata, S. S. Alkan, A. Subramaniam, and D. Unutmaz. 2003. Genetic reprogramming of primary human T cells reveals functional plasticity in Th cell differentiation. J. Immunol. 1713542-3549. [DOI] [PubMed] [Google Scholar]
  • 36.Tanaka, Y., Y. Koyanagi, R. Tanaka, Y. Kumazawa, T. Nishimura, and N. Yamamoto. 1997. Productive and lytic infection of human CD4+ type 1 helper T cells with macrophage-tropic human immunodeficiency virus type 1. J. Virol. 71465-470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Unutmaz, D., V. N. KewalRamani, S. Marmon, and D. R. Littman. 1999. Cytokine signals are sufficient for HIV-1 infection of resting human T lymphocytes. J. Exp. Med. 1891735-1746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Vetter, M. L., M. E. Johnson, A. K. Antons, D. Unutmaz, and R. T. D'Aquila. 2009. Differences in APOBEC3G expression in CD4+ T helper lymphocyte subtypes modulate HIV-1 infectivity. PLoS Pathog. 5e1000292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Wang, X., P. T. Dolan, Y. Dang, and Y. H. Zheng. 2007. Biochemical differentiation of APOBEC3F and APOBEC3G proteins associated with HIV-1 life cycle. J. Biol. Chem. 2821585-1594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Yang, Y., F. Guo, S. Cen, and L. Kleiman. 2007. Inhibition of initiation of reverse transcription in HIV-1 by human APOBEC3F. Virology 36592-100. [DOI] [PubMed] [Google Scholar]
  • 41.Yu, Q., R. Konig, S. Pillai, K. Chiles, M. Kearney, S. Palmer, D. Richman, J. M. Coffin, and N. R. Landau. 2004. Single-strand specificity of APOBEC3G accounts for minus-strand deamination of the HIV genome. Nat. Struct. Mol. Biol. 11435-442. [DOI] [PubMed] [Google Scholar]
  • 42.Yu, X., Y. Yu, B. Liu, K. Luo, W. Kong, P. Mao, and X. F. Yu. 2003. Induction of APOBEC3G ubiquitination and degradation by an HIV-1 Vif-Cul5-SCF complex. Science 3021056-1060. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Virology are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES